You are on page 1of 1187

Luca Saba

Eytan Raz 
Editors

Neurovascular
Imaging
From Basics to
Advanced Concepts

1 3Reference
Neurovascular Imaging
Luca Saba • Eytan Raz
Editors

Neurovascular Imaging
From Basics to Advanced Concepts

With 799 Figures and 102 Tables


Editors
Luca Saba Eytan Raz
Department of Radiology Neurointerventional Radiology Section
AOU of Cagliari Department of Radiology
Monserrato, Cagliari NYU Langone Medical Center
Italy New York, NY
USA

ISBN 978-1-4614-9028-9 ISBN 978-1-4614-9029-6 (eBook)


ISBN 978-1-4614-9030-2 (print and electronic bundle)
DOI 10.1007/978-1-4614-9029-6
Library of Congress Control Number: 2015956531

# Springer Science+Business Media New York 2016


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software, or
by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are exempt
from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this
book are believed to be true and accurate at the date of publication. Neither the publisher nor the
authors or the editors give a warranty, express or implied, with respect to the material contained
herein or for any errors or omissions that may have been made.

Printed on acid-free paper

This Springer imprint is published by SpringerNature


The registered company is Springer Science+Business Media LLC New York
Luca: This book is dedicated to Tiziana
Eytan: To the women of my life, my mother Daniela and
my wife Lia
Preface

Vascular neurology, vascular neurosurgery, and interventional neuroradiology


are independent fields with dedicated training programs. Neuroimaging, and in
particular what we call “neurovascular imaging,” is a unifying factor which
can be considered the intersection of these three medical specialties. With this
book, we aim to cover thoroughly the imaging techniques, potentialities, and
present and future applications as applied to the vascular diseases of the central
nervous system from the imaging point of view.
In recent years, several advances in imaging technology have dramatically
expanded the frontiers of human investigation in the neurovascular field. We
hope that this book is a timely and complete addition to the growing body of
literature on this topic.
Our aim is to cover all aspects of neurovascular disease in a comprehensive
way. The illustrations and high quality of the images will be the key to success.
This is the first collection to be published dedicated to the field of vascular
neuroimaging.

Cagliari, Italy Luca Saba


New York, USA Eytan Raz
December 2015

vii
Quotes

Do not go where the path may lead, go instead where there is no path and
leave a trail.
Ralph Waldo Emerson (1803–1882)

The foundation of every state is the education of its youth.


Diogenes (412 BC–323 BC)

Life is like riding a bicycle. To keep your balance you must keep moving.
Albert Einstein (1879–1955)

Live as if you were to die tomorrow; learn as if you were to live forever.
Mahatma Gandhi (1869–1948)

ix
Acknowledgments

It is not possible to overstate my gratitude to the many individuals who helped


to produce this book; their enthusiasm and dedication was unbelievable. In
particular, I would like to thank Professor Mario Piga for his help and advice. I
am deeply appreciative of the support offered by my colleagues at the Azienda
Ospedaliero-Universitaria di Cagliari. My gratitude goes to all authors who
have contributed to this project. Without their incredible work, this would not
have been possible.
I would like to express my appreciation to Springer for their professional-
ism in handling this project and for the patience. Most particularly, I would like
to thank Sandra Fabiani, Sunaina Dadhwal, Neha Thapa, and Gregory Baer for
their tireless dedication and advice during each stage in the production of this
book. Your help was wonderful and made producing this project an enjoyable
and worthwhile experience.
Finally, again, I would like to acknowledge Tiziana! Thank you, for
everything.

Cagliari, Italy Luca Saba

First and foremost, I want to thank Luca Saba for inviting me to help him in this
project and for his efforts in making this project going along the way. Luca,
you are one of the most hardworking persons I have ever met!
I want to thank Springer for the patience and enthusiasm that they applied to
this project, especially Neha Thapa and Sunaina Dadwhal. Thank you very
much!
I also wanted to thank all the teachers I had in my medical career so far,
especially those teachers that did that without clamor, those that taught ethics
before medicine, and those that taught me a method, a way of living, and
secondarily a way of working.

New York, USA Eytan Raz

xi
Contents

Volume 1

Part I The Basics: Embryology, Pathological Basis of


Vascular Diseases, and Imaging Techniques . . . . . . . . . . . . . . . . . . 1

1 Cerebrovascular Development and Evolution . . . . . . . . . . . . 3


Maksim Shapiro and Eytan Raz
2 Atherosclerosis: Biological and Pathological Basis . . . . . . . . 33
Michele Porcu, Eytan Raz, and Luca Saba
3 Nonatherosclerotic Vascular Disease: Biological and
Pathological Basis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
Pierleone Lucatelli, Beatrice Sacconi, and Carlo Catalano
4 Essentials of Transcranial Doppler Ultrasound . . . . . . . . . . . 47
Jonathan D. Kirsch
5 DSA Imaging Protocol and Technique . . . . . . . . . . . . . . . . . . 67
Eytan Raz

Part II Carotid and Vertebral Artery: Anatomy, Diseases,


and Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85

6 Anatomy of the Neck Arteries . . . . . . . . . . . . . . . . . . . . . . . . . 87


Nasim Sheikh-Bahaei, Tomasz Matys, and Jonathan H. Gillard
7 Imaging of Carotid Atherosclerosis . . . . . . . . . . . . . . . . . . . . 95
Jie Sun, Niranjan Balu, and Chun Yuan
8 Carotid Artery Dissection . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
Olivier Naggara, Myriam Edjlali-Goujon, Eric Bodiguel,
Marie Pierre Gobin-Metteil, Denis Trystram, Christine
Rodriguez-Regent, Jean-Louis Mas, Jean Francois Meder, and
Catherine Oppenheim
9 Nonatherosclerotic Nondissection Diseases of
Carotid Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
Santosh Kumar Kannath and T.R. Kapilamoorthy

xiii
xiv Contents

10 Imaging of the Pathology of the Vertebral Arteries . . . . . . . . 163


David Chiao and Max Wintermark

11 Carotid Artery Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191


Roberto Montisci and Luca Saba

Part III The Basics of Intracranial Arterial Circulation . . . . . . . . 203

12 Intracranial Atherosclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . 205


Xinyi Leng and David S. Liebeskind

13 Imaging of Spontaneous Nontraumatic Intracerebral


Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233
Stephen Quinet and Patrick Turski

14 ICA–ECA Collaterals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293


Maxim Mokin and Adnan H. Siddiqui

15 Imaging Biomarkers of Angiogenesis and the


Microvascular Environment in Cerebral Tumors . . . . . . . . . 303
Alan Jackson, Ibrahim Djoukhadar, and David J. Coope

Part IV Stroke Imaging .................................... 327

16 Patterns of Ischemic Stroke: From Lacunar to Territorial


to Multiple Embolic to Watershed Hypotensive . . . . . . . . . . 329
Jessica Rotman and Robert Zimmerman

17 Territorial Strokes as a Tool to Learn Vascular


Territories . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363
Behroze Adi Vachha and Pamela Whitney Schaefer

18 Hemorrhagic Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 383


Julius Griauzde, Elliot Dickerson, and Joseph J. Gemmete

19 Medical Therapy of Acute Stroke . . . . . . . . . . . . . . . . . . . . . . 413


Keith G. DeSousa and Albert S. Favate

20 Endovascular Treatment of Stroke . . . . . . . . . . . . . . . . . . . . . 425


Paolo Machi and Paolo Garofalo

21 Sickle Cell Disease and Stroke . . . . . . . . . . . . . . . . . . . . . . . . 439


Akifumi Fujita, Chie Asai, Yu-Ming Chang, Nadja Kadom,
Martin H. Steinberg, Naoko Saito, and Osamu Sakai

22 Hypoxic–Ischemic Encephalopathy (Preterm, Term,


and Adult) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 469
Mauricio Castillo and Francisco Chiang
Contents xv

Part V Aneurysms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495

23 Aneurysmal Subarachnoid Hemorrhage . . . . . . . . . . . . . . . . 497


Frédéric Clarençon, Nader-Antoine Sourour, Vincent Degos,
Aurélien Nouet, Federico Di Maria, Eimad Shotar,
Joseph Gabrieli, Lise Le Jean, and Jacques Chiras
24 Blister Aneurysms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 521
James V. Byrne and Svein Harald Mørkve
25 Surgical Treatment of Aneurysms . . . . . . . . . . . . . . . . . . . . . 535
Saul F. Morales-Valero, Shanna Fang, and Giuseppe Lanzino
26 Endovascular Treatment of Brain Aneurysms . . . . . . . . . . . 551
Luca Quilici and Edoardo Boccardi
27 Postop Evaluation of Aneurysms (Including Vasospasm) . . . 583
Michael Mayich, Brian P. Walcott, Christopher
J. Stapleton, and Daniel Thomas Ginat

Volume 2

Part VI Other CNS Vascular Diseases . . . . . . . . . . . . . . . . . . . . . . . . 603

28 Brain Arteriovenous Malformations . . . . . . . . . . . . . . . . . . . 605


Daniel Sahlein and Nathan Manning
29 Dural AVF and Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . 641
Pierre Guedin
30 Carotid Cavernous Fistula . . . . . . . . . . . . . . . . . . . . . . . . . . . 657
Timothy R. Miller, Ravishankar Shivashankar,
Gaurav Jindal, and Dheeraj Gandhi
31 Imaging of the Neurovasculitides . . . . . . . . . . . . . . . . . . . . . . 683
Adam J. Davis
32 Miscellaneous Vascular Malformations (Cavernous
Malformations, Developmental Venous Anomaly,
Capillary Telangiectasia, Sinus Pericranii) . . . . . . . . . . . . . . 719
Luiz Celso Hygino da Cruz and Cintia Elias Pires
33 Miscellaneous Vascular Diseases . . . . . . . . . . . . . . . . . . . . . . . 751
Matylda H. Machnowska and Richard I. Aviv
34 Vascular Loop Syndromes . . . . . . . . . . . . . . . . . . . . . . . . . . . 781
Divyata R. Hingwala and Kesavadas Chandrasekharan
35 Migraine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 791
Mark Kruit
xvi Contents

36 Moyamoya Disease (Spontaneous Occlusion


of the Circle of Willis) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 817
Akira Yamamoto, Tomohisa Okada, and
Jun C. Takahashi
37 Brain Death Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 865
M. Sawicki, Joanna Wojczal, Bozena Birkenfeld, and
Lech Cyrylowski

Part VII Veins Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 897

38 Jugular Vein Thrombosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . 899


Nicholas A. Koontz, Richard H. Wiggins III, and
Lubdha M. Shah
39 Imaging of Cerebral Venous and Sinus Thrombosis . . . . . . . 935
Jennifer Linn
40 The Role of Diagnostic Imaging Techniques for Detection
of Extracranial Venous System Abnormalities Associated
with Central Nervous System Disorders . . . . . . . . . . . . . . . . 953
Kresimir Dolic and Robert Zivadinov

Part VIII Pediatrics ........................................ 981

41 Pediatric Vascular Malformations . . . . . . . . . . . . . . . . . . . . . 983


Sachin K. Pandey and Darren B. Orbach
42 Pediatric Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1009
Daniel Tibussek, Gabrielle deVeber, and Manohar Shroff
43 Congenital Vascular Syndromes and Diseases . . . . . . . . . . . . 1041
Sarah Milla, Jennifer Vaughn, and Nilesh K. Desai

Part IX Spine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1061

44 Spinal Vascular Imaging: Technique . . . . . . . . . . . . . . . . . . . 1063


Maria Isabel Vargas, Fabrice Bing, Joanna Gariani, and
Jean-Louis Dietemann
45 Spinal Vascular Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1095
Tibor Becske and Eytan Raz
46 Spinal Vascular Malformations and Treatment . . . . . . . . . . . 1105
Srinivasan Paramasivam
47 Spinal Cord Infarction and Differential Diagnosis . . . . . . . . 1125
Srikanth R. Boddu, Alessandro Cianfoni, Kyung-Wha Kim,
Mohammad Amin Banihashemi, Emanuele Pravatà,
Y. Pierre Gobin, and Athos Patsalides
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1185
About the Editors

Prof. Luca Saba received his M.D. from the University of Cagliari, Italy, in
2002. Today, he works in the A.O.U. of Cagliari. Professor Saba’s research
fields are focused on multi-detector-row computed tomography, magnetic
resonance, ultrasound, neuroradiology, and diagnostic in vascular sciences.
His works, as lead author, are more than 170 publications in high-impact-
factor, peer-reviewed journals such as American Journal of Neuroradiology,
Atherosclerosis, European Radiology, European Journal of Radiology, Acta
Radiologica, Cardiovascular and Interventional Radiology, Journal of
Computer Assisted Tomography, American Journal of Roentgenology, Neuro-
radiology, Clinical Radiology, Journal of Cardiovascular Surgery, Cerebro-
vascular Diseases, Brain Pathology, Medical Physics, and Atherosclerosis.
He is a well-known speaker and has spoken over 45 times at national and
international level conferences.
Dr. Saba has won 15 scientific and extracurricular awards during his career.
Dr. Saba has presented more than 500 papers and posters in national and
international congresses (RSNA, ESGAR, ECR, ISR, AOCR, AINR, JRS,
SIRM, AINR). He has written 21 book chapters and is Editor of 10 books in
the field of computed tomography, cardiovascular, plastic surgery, gynecolog-
ical imaging, and neurodegenerative imaging.
He is a member of the Italian Society of Radiology (SIRM), European
Society of Radiology (ESR), Radiological Society of North America (RSNA),
American Roentgen Ray Society (ARRS), and European Society of Neurora-
diology (ESNR) and serves as reviewer of 40 scientific journals.

xvii
xviii About the Editors

Eytan Raz currently works at NYU Langone Medical Center in New York
City, where he is an Assistant Professor working for the Neuroradiology and
Interventional Neuroradiology Sections. He graduated from Sapienza
University of Rome School of Medicine in 2006, where he also obtained his
Radiology Specialty in 2011. He completed his fellowships in Diagnostic
Neuroradiology and Interventional Neuroradiology at NYU Langone
Medical Center. He is the author of more than 60 publications in
high-impact-factor, peer-reviewed journals such as Radiology, AJNR, AJR,
JNIS, Neurology, and Brain Pathology. His fields of expertise include
neurovascular diseases, traumatic brain injury, and multiple sclerosis. In
2012, he was appointed Editor for the Fellows’ Portal of AJNR. He is a
reviewer for more than 10 journals and has presented more than 70 abstracts
at international conferences. He is a member of the Radiological Society of
North America (RSNA), the American Society of Neuroradiology (ASNR),
and the Italian Society of Neuroradiology (AINR).
Contributors

Chie Asai Departments of Radiology, Boston University, Boston Medical


Center, Boston, MA, USA

Richard I. Aviv Division of Neuroradiology, Department of Medical Imag-


ing, School of Medicine, Sunnybrook Health Sciences Centre, University of
Toronto, Toronto, ON, Canada

Niranjan Balu Department of Radiology, University of Washington, Seattle,


WA, USA

Mohammad Amin Banihashemi Department of Neurological Surgery,


Weill Cornell Medical Center, New York, NY, USA

Tibor Becske Neurointerventional Radiology Section, Department of


Radiology, NYU Langone Medical Center, New York, NY, USA

Fabrice Bing Department of Radiology, University Hospital of Strasbourg,


Strasbourg, France

Bozena Birkenfeld Department of Nuclear Medicine, Pomeranian Medical


University, Szczecin, Poland

Edoardo Boccardi Ospedale Niguarda – Ca’ Granda, Milan, Italy

Srikanth R. Boddu Department of Neurological Surgery, Weill Cornell


Medical Center, New York, NY, USA

Eric Bodiguel Service de Neurologie, Faculté de Médecine, Centre


Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité,
Paris, France

James V. Byrne Nuffield Department of Surgical Sciences, Oxford Univer-


sity, John Radcliffe Hospital, Oxford, UK

Mauricio Castillo Division of Neuroradiology, Department of Radiology,


University of North Carolina, Chapel Hill, NC, USA

Carlo Catalano Department of Radiological, Oncological and Anatomo-


Pathological Sciences, Sapienza University of Rome, Rome, Italy

xix
xx Contributors

Kesavadas Chandrasekharan NM Medical Centre, Mumbai, Maharashtra,


India
Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivan-
drum, Kerela, India
Yu-Ming Chang Departments of Radiology, Boston University, Boston
Medical Center, Boston, MA, USA
Francisco Chiang Division of Neuroradiology, Department of Radiology,
University of North Carolina, Chapel Hill, NC, USA
David Chiao Department of Radiology, School of Medicine, University of
Virginia, Charlottesville, VA, USA
Jacques Chiras Department of Interventional Neuroradiology,
Pitié-Salpêtrière Hospital, Paris VI University, Paris, France
Alessandro Cianfoni Department of Radiology, Medical University of South
Carolina, Charleston, SC, USA
Frédéric Clarençon Department of Interventional Neuroradiology,
Pitié-Salpêtrière Hospital, Paris VI University, Paris, France
David J. Coope Wolfson Molecular Imaging Centre, University of Manchester,
Withington, Manchester, UK
Lech Cyrylowski CT and MRI Clinic, “Nasz Doktor” Health Care Institu-
tion, Szczecin, Poland
MRI Clinic, “Euromedic” Health Care Institution, Szczecin, Poland
Luiz Celso Hygino da Cruz Department of Radiology, Clinics CDPI/DASA
and IRM, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
Adam J. Davis Neuroradiology, Department of Radiology, NYU Langone
Medical Center, New York, NY, USA
Vincent Degos Department of Anesthesia and Critical Care, Neuro-intensive
Care Unit, Pitié- Salpêtrière Hospital, Paris VI University, Paris, France
Nilesh K. Desai Department of Radiology, Texas Children’s Hospital, Baylor
College of Medicine, Houston, TX, USA
Keith G. DeSousa Department of Neurology, NYU Langone Medical Cen-
ter, New York, NY, USA
Vascular Neurology, University of Miami, Miami, FL, USA
Gabrielle deVeber Children’s Stroke Program, Division of Neurology,
The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
Federico Di Maria Department of Interventional Neuroradiology, Pitié-
Salpêtrière Hospital, Paris VI University, Paris, France
Elliot Dickerson Department of Radiology, Radiology Resident, University
of Michigan, Ann Arbor, MI, USA
Contributors xxi

Jean-Louis Dietemann Department of Radiology, University Hospital of


Strasbourg, Strasbourg, France
Ibrahim Djoukhadar Wolfson Molecular Imaging Centre, University of
Manchester, Withington, Manchester, UK
Kresimir Dolic Buffalo Neuroimaging Analysis Center, Department of
Neurology, University at Buffalo, State University of New York, Buffalo,
NY, USA
Clinical Department of Diagnostic and Interventional Radiology, Clinical
Hospital Center Split, University of Split, Split, Croatia
Myriam Edjlali-Goujon Service de Neuroradiologie, Centre Hospitalier
Sainte-Anne, INSERM UMR894, Université Paris Descartes, Sorbonne
Paris Cité, Paris, France
Shanna Fang Mayo Medical School, Mayo Clinic, Rochester, MN, USA
Albert S. Favate NYU Langone Medical Center, New York, NY, USA
Akifumi Fujita Departments of Radiology, Boston University, Boston
Medical Center, Boston, MA, USA
Joseph Gabrieli Department of Interventional Neuroradiology,
Pitié-Salpêtrière Hospital, Paris VI University, Paris, France
Dheeraj Gandhi Department of Diagnostic Radiology, Neuroradiology,
University of Maryland Medical Center, Baltimore, MD, USA
Joanna Gariani Department of Radiology, Geneva University Hospitals,
Geneva, Switzerland
Paolo Garofalo Medicine School, University of Parma, Parma, Italy
Joseph J. Gemmete Department of Radiology, Neurosurgery, and Otolaryn-
gology, University of Michigan, Ann Arbor, MI, USA
Jonathan H. Gillard Department of Radiology, School of Clinical Medicine,
University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
Daniel Thomas Ginat Department of Radiology, University of Chicago,
Chicago, IL, USA
Y. Pierre Gobin Department of Neurological Surgery, Weill Cornell Medical
Center, New York, NY, USA
Marie Pierre Gobin-Metteil Service de Neuroradiologie, Centre Hospitalier
Sainte-Anne, INSERM UMR894, Université Paris Descartes, Sorbonne Paris
Cité, Paris, France
Julius Griauzde Department of Radiology, Radiology Resident, University
of Michigan, Ann Arbor, MI, USA
Pierre Guedin Department of Diagnostic and Therapeutic Neuroradiology,
Hôpital Foch, Suresnes, France
Divyata R. Hingwala NM Medical Centre, Mumbai, Maharashtra, India
xxii Contributors

Alan Jackson Wolfson Molecular Imaging Centre, University of Manchester,


Withington, Manchester, UK

Gaurav Jindal Department of Diagnostic Radiology, Neuroradiology,


University of Maryland Medical Center, Baltimore, MD, USA

Nadja Kadom Departments of Radiology, Boston University, Boston Med-


ical Center, Boston, MA, USA

Santosh Kumar Kannath Neurointervention Center, Department of Imag-


ing Sciences and Interventional Radiology, Neurointervention Center, Sree
Chitra Institute of Medical Sciences and Technology, Trivandrum, Kerala,
India

T. R. Kapilamoorthy Department of Imaging Sciences and Interventional


Radiology, Sree Chitra Institute of Medical Sciences and Technology, Trivan-
drum, Kerala, India

Kyung-Wha Kim New York Presbyterian Hospital, Weill Cornell Medical


College of Cornell University, New York, NY, USA

Jonathan D. Kirsch Department of Diagnostic Radiology, Yale University


School of Medicine, New Haven, CT, USA

Nicholas A. Koontz Department of Radiology, University of Utah Health


Sciences Center, Salt Lake City, UT, USA

Mark Kruit Department of Radiology, Leiden University Medical Center,


Leiden, The Netherlands

Giuseppe Lanzino Department of Neurologic Surgery, Mayo Clinic, Roch-


ester, MN, USA

Lise Le Jean Department of Anesthesia and Critical Care, Neuro-intensive


Care Unit, Pitié- Salpêtrière Hospital, Paris VI University, Paris, France

Xinyi Leng Department of Medicine and Therapeutics, The Chinese Univer-


sity of Hong Kong, Prince of Wales Hospital, Hong Kong, SAR, China

David S. Liebeskind UCLA Department of Neurology, UCLA, Los Angeles,


CA, USA

Jennifer Linn Institute of Neuroradiology, University Hospital Carl Gustav


Carus, Dresden, Germany

Pierleone Lucatelli Department of Radiological, Oncological and Anatomo-


Pathological Sciences, Sapienza University of Rome, Rome, Italy

Paolo Machi Department of Neuroradiology Unit, CHU Gui de Chauliac,


Montpellier University Hospital, Montpellier, France

Matylda H. Machnowska Division of Neuroradiology, Department of


Medical Imaging, School of Medicine, Sunnybrook Health Sciences Centre,
University of Toronto, Toronto, ON, Canada
Contributors xxiii

Nathan Manning Department of Neurological Surgery, Columbia Univer-


sity Medical Centre, New York, NY, USA
Jean-Louis Mas Service de Neurologie, Faculté de Médecine, Centre
Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité,
Paris, France
Tomasz Matys Department of Radiology, School of Clinical Medicine,
University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
Michael Mayich Department of Radiology, University of Chicago, Chicago,
IL, USA
Jean Francois Meder Service de Neuroradiologie, Centre Hospitalier
Sainte-Anne, INSERM UMR894, Université Paris Descartes, Sorbonne
Paris Cité, Paris, France
Sarah Milla Department of Radiology, Emory University School of Medi-
cine Egleston Hospital, Children’s Healthcare of Atlanta, Atlanta, GA, USA
Timothy R. Miller Department of Diagnostic Radiology, Neuroradiology,
University of Maryland Medical Center, Baltimore, MD, USA
Maxim Mokin Department of Neurosurgery, School of Medicine and
Biomedical Sciences, University at Buffalo, State University of New York,
Buffalo, NY, USA
Department of Neurosurgery, Gates Vascular Institute/Kaleida Health,
Buffalo, NY, USA
Roberto Montisci Department of Vascular Surgery, AOU of Cagliari,
Monserrato, Cagliari, Italy
Saul F. Morales-Valero Department of Neurologic Surgery, Mayo Clinic,
Rochester, MN, USA
Svein Harald Mørkve Department of Neurosurgery, Haukeland University
Hospital, Bergen, Norway
Olivier Naggara Service de Neuroradiologie, Centre Hospitalier Sainte-
Anne, INSERM UMR894, Université Paris Descartes, Sorbonne Paris Cité,
Paris, France
Service de Radiologie pédiatrique, Faculté de Médecine, Hôpital Necker
Enfants Malades, Université Paris Descartes, Sorbonne Paris Cité, Paris,
France
Aurélien Nouet Department of Neurosurgery, Pitié-Salpêtrière Hospital,
Paris VI University, Paris, France
Tomohisa Okada Department of Diagnostic Imaging and Nuclear Medicine,
Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
Catherine Oppenheim Service de Neuroradiologie, Centre Hospitalier
Sainte-Anne, INSERM UMR894, Université Paris Descartes, Sorbonne
Paris Cité, Paris, France
xxiv Contributors

Darren B. Orbach Neurointerventional Radiology, Boston Children’s


Hospital, Harvard Medical School, Boston, MA, USA

Sachin K. Pandey Neurointerventional Radiology, Brigham and Women’s


Hospital, Boston, MA, USA
Srinivasan Paramasivam Mount Sinai Roosevelt Hospital, Hyman New-
man Institute for Neurology and Neurosurgery, Centre for Endovascular
Surgery, New York, NY, USA

Athos Patsalides Department of Neurological Surgery, Weill Cornell Medi-


cal Center, New York, NY, USA

Cintia Elias Pires Department of Radiology, Clinics CDPI/DASA, National


Institute of Cardiology, Federal University of Rio de Janeiro, Rio de Janeiro,
Brazil
Michele Porcu Department of Radiology, AOU of Cagliari, Monserrato,
Cagliari, Italy

Emanuele Pravatà Neuroradiology Department, Neurocenter of Italian


Switzerland, Lugano, Switzerland
Luca Quilici Ospedale Niguarda – Ca’ Granda, Milan, Italy

Stephen Quinet Department of Radiology, School of Medicine and


Public Health, Clinical Science Center, University of Wisconsin, Madison,
WI, USA

Eytan Raz Neurointerventional Radiology Section, Department of


Radiology, NYU Langone Medical Center, New York, NY, USA

Christine Rodriguez-Regent Service de Neuroradiologie, Centre


Hospitalier Sainte-Anne, INSERM UMR894, Université Paris Descartes,
Sorbonne Paris Cité, Paris, France

Jessica Rotman PGY-3 Diagnostic Radiology Resident, New York Presby-


terian Hospital, Weill Cornell Medical Center, New York, NY, USA

Luca Saba Department of Radiology, AOU of Cagliari, Monserrato,


Cagliari, Italy

Beatrice Sacconi Department of Radiological, Oncological and Anatomo-


Pathological Sciences, Sapienza University of Rome, Rome, Italy

Daniel Sahlein Goodman Campbell Brain and Spine, Indianapolis, IN, USA

Naoko Saito Department of Diagnostic Radiology, Saitama Medical Univer-


sity International Medical Center, Saitama, Japan

Osamu Sakai Departments of Radiology, Boston University, Boston Medi-


cal Center, Boston, MA, USA
Department of Otolaryngology – Head and Neck Surgery, Boston University,
Boston Medical Center, Boston, MA, USA
Contributors xxv

Department of Radiation Oncology, Boston University, Boston Medical Cen-


ter, Boston, MA, USA
M. Sawicki Department of Diagnostic Imaging and Interventional Radiol-
ogy, Pomeranian Medical University, Szczecin, Poland
Pamela Whitney Schaefer Neuroradiology, Massachusetts General Hospital,
Boston, MA, USA
Lubdha M. Shah Department of Radiology, University of Utah Health
Sciences Center, Salt Lake City, UT, USA
Maksim Shapiro Neurointerventional Radiology Section, Department of
Radiology, NYU Langone Medical Center, New York, NY, USA
Nasim Sheikh-Bahaei Department of Radiology, School of Clinical
Medicine, University of Cambridge, Cambridge Biomedical Campus,
Cambridge, UK
Ravishankar Shivashankar Department of Diagnostic Radiology, Neurora-
diology, University of Maryland Medical Center, Baltimore, MD, USA
Eimad Shotar Department of Interventional Neuroradiology, Pitié-
Salpêtrière Hospital, Paris VI University, Paris, France
Manohar Shroff Department of Diagnostic Imaging, The Hospital for Sick
Children, University of Toronto, Toronto, ON, Canada
Adnan H. Siddiqui Department of Neurosurgery, School of Medicine and
Biomedical Sciences, University at Buffalo, State University of New York,
Buffalo, NY, USA
Department of Neurosurgery, Gates Vascular Institute/Kaleida Health,
Buffalo, NY, USA
Department of Radiology, School of Medicine and Biomedical Sciences,
University at Buffalo, State University of New York, Buffalo, NY, USA
Toshiba Stroke and Vascular Research Center, University at Buffalo, State
University of New York, Buffalo, NY, USA
Jacobs Institute, Buffalo, NY, USA
Nader-Antoine Sourour Department of Interventional Neuroradiology,
Pitié-Salpêtrière Hospital, Paris VI University, Paris, France
Christopher J. Stapleton Department of Neurosurgery, Massachusetts
General Hospital, Boston, MA, USA
Martin H. Steinberg Departments of Medicine, Pediatrics, and Pathology
and Laboratory Medicine, Boston University, Boston Medical Center, Boston,
MA, USA
Jie Sun Department of Radiology, University of Washington, Seattle,
WA, USA
Jun C. Takahashi Department of Neurosurgery, National Cerebral and
Cardiovascular Center, Suita City, Osaka, Japan
xxvi Contributors

Daniel Tibussek Children’s Stroke Program, Division of Neurology, The


Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
Department of General Pediatrics, Neonatalogy and Pediatric Cardiology,
University Children’s Hospital, Heinrich-Heine University, D€usseldorf,
Germany
Denis Trystram Service de Neuroradiologie, Centre Hospitalier Sainte-
Anne, INSERM UMR894, Université Paris Descartes, Sorbonne Paris Cité,
Paris, France
Patrick Turski Department of Radiology, School of Medicine and Public
Health, Clinical Science Center, University of Wisconsin, Madison, WI, USA
Behroze Adi Vachha Neuroradiology, Massachusetts General Hospital,
Boston, MA, USA
Maria Isabel Vargas Department of Neuroradiology, Geneva University
Hospitals, Geneva, Switzerland
Jennifer Vaughn Department of Radiology, Boston Children’s Hospital,
Boston, MA, USA
Brian P. Walcott Department of Neurosurgery, Massachusetts General
Hospital, Boston, MA, USA
Richard H. Wiggins III Department of Radiology, University of Utah
Health Sciences Center, Salt Lake City, UT, USA
Max Wintermark Department of Radiology, School of Medicine, Univer-
sity of Virginia, Charlottesville, VA, USA
Joanna Wojczal Department of Neurology, Medical University of Lublin,
Lublin, Poland
Akira Yamamoto Department of Diagnostic Imaging and Nuclear Medicine,
Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
Chun Yuan Department of Radiology, University of Washington, Seattle,
WA, USA
Robert Zimmerman Diagnostic Radiology Department, New York Presby-
terian Hospital, Weill Cornell Medical Center, New York, NY, USA
Robert Zivadinov Buffalo Neuroimaging Analysis Center, Department
of Neurology, University at Buffalo, State University of New York, Buffalo,
NY, USA
MRI Clinical Translational Research Center, University at Buffalo, State
University of New York, Buffalo, NY, USA
Part I
The Basics: Embryology, Pathological
Basis of Vascular Diseases, and Imaging
Techniques
Cerebrovascular Development and
Evolution 1
Maksim Shapiro and Eytan Raz

Contents Abstract
Vascular Neurophylogeny . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 The comprehensive appreciation of the embryo-
The Spinal Cord . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 logical events is paramount to develop a good
The Brain: From Fishes to Humans . . . . . . . . . . . . . . . . . . 6 understanding of the cerebrovasculature in all its
Vascular Neuroembryology . . . . . . . . . . . . . . . . . . . . . . . . . 12 aspects, from the anatomy to the physiology and
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 hence the pathology. Vascularization of the brain
Stage I (Prechoroidal Stage): and spinal cord has certain fundamental similar-
Approximately 4 Weeks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Stage II (Choroidal Stage):
ities. The evolution of brain circulation attests to
Approximately 5 Weeks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15 progressive recruitment of already existing vas-
Stage III: 6 Weeks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16 cular networks to supply emergent cortical terri-
Stage IV: Approximately 7 Weeks . . . . . . . . . . . . . . . . . . . . 16 tories, rather than development of de novo
Stage V: Approximately 9 Weeks . . . . . . . . . . . . . . . . . . . . . 17
Stage VI: Approximately Adult Pattern . . . . . . . . . . . . . . 17
arterial solutions. This principle can be applied
to many concepts in evolutionary developmental
Practical Aspects of Neurophylogeny biology. And, since, in many aspects, “ontogeny
and Neuroembryology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Fetal Posterior Cerebral Artery . . . . . . . . . . . . . . . . . . . . . . . 18 recapitulates phylogeny” to quote Ernst Haeckel,
Accessory MCA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 father of the embryological parallelism law, the
Dominant Anterior Choroidal Artery . . . . . . . . . . . . . . . . . 19 phylogeny of the brain and spinal cord is a key
Persistent Trigeminal Artery . . . . . . . . . . . . . . . . . . . . . . . . . . 19 concept to understand in order to develop a mind
Persistent Hypoglossal Artery . . . . . . . . . . . . . . . . . . . . . . . . 19
Proatlantal Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20 framework which becomes then useful for the
Variations in Fusion Patterns of the Basilar Artery . . . 20 approach of embryology.
Duplications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Anterior Cerebral Artery Variations . . . . . . . . . . . . . . . . . . 25 Part of the text, the concept, and the figures in this
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26 chapter are used with permission from Maksim
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 Shapiro, from the website www.neuroangio.org.

Vascular Neurophylogeny

M. Shapiro (*) • E. Raz The Spinal Cord


Neurointerventional Radiology Section, Department of
Radiology, NYU Langone Medical Center, New York, The neural tube and the spinal cord can be thought
NY, USA
e-mail: Maksim.Shapiro@nyumc.org; of, simplistically, as a cylinder (Fig. 1). After
eytan.raz@gmail.com; eytan.raz@nyumc.org closure of the neural tube, nutritional support
# Springer Science+Business Media New York 2016 3
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_15
4 M. Shapiro and E. Raz

can be provided by simple diffusion, but, at a coming from the two dorsal aortae (Fig. 2). These
certain point of body growth, dedicated vascular vessels are also called metameric: in biology,
supply to the spinal cord and adjacent tissues is metamerism is the phenomenon of having a linear
needed and is established using segmental vessels series of similar body segments; each segment
may be called somite or metamer. Each
metameric vessel supports its own block of tissue,
including the endodermal, ectodermal, and meso-
dermal tissue (Fig. 3). As diagrammatically shown
in Fig. 4, between segmental vessels longitudinal
anastomoses extending craniocaudally along the
length of the cord are then formed (Fig. 4),
establishing a primordial matrix of the adult vascu-
Spinal Cord lature. Once the longitudinal connection between
the metamers is established, there is no need for a
segmental vessel at each single level, and most
segmental vessels shrink to supply only the region
of the nerve root of the same level (Figs. 5 and 6).
The spinal cord is now supplied by an anterior
spinal artery and posteriorly by two spinal vessels
that will become the posterior spinal arteries. The
Fig. 1
segmental vessels that supply flow to the anterior
spinal artery are called radiculomedullary arteries,
while the ones that supply only the nerve root are
called radicular branches. Sometimes, one segmen-
Dorsal tal vessel supplies two nerve roots via longitudinal
Aorta anastomoses (Fig. 7). As the spinal cord continues
Folded Neural Tube to enlarge, sulco-commissural branches are
formed, which can be matched with the brain per-
forator arteries (Fig. 8).
In Figs. 9 and 10, the spinal angiographic cor-
relation can be visualized in vivo with subtracted
and unsubtracted views which demonstrate a
radiculomedullary vessel giving rise to an anterior
Fig. 2 spinal artery.

Fig. 3
To ribs,
muscle,
Dorsal etc.
Aorta

Segmental (a.k.a Metameric)


Vessels
1 Cerebrovascular Development and Evolution 5

Fig. 4
Devloping To ribs,
Longitudinal muscle,
Anastomoses etc.

Segmental (a.k.a. Metamaric)


Vessels

Fig. 5
To ribs,
muscle,
etc.
Vertebral
artery-like
vessel

Future Anterior Spinal Artery


Segmental Vessels

Fig. 6

Longitudinal vessel, future


Anterior Spinal artery

Surface pial vessel

Most segmental vessels regress and remain


purely radicular; several dominant
radiculomedullary ones remain
6 M. Shapiro and E. Raz

Fig. 7
Obviously, only To ribs,
Radiculomedullary
one aorta muscle,
artery
remains in the Anterior etc.
adult, from Spinal
which left and Artery
right segmental
vessels emerge.
Two are shown
here for
diagrammatic
purpose.

Radicular artery
Dominant radicular artery
supplying two levels

variations. The fact that newly formed vessels


Anterior are the exception, while existing pathways are
Spinal usually utilized and enlarged to accommodate
Artery
tissue needs, was recognized since the early twen-
tieth century.

Fishes
The earliest vertebrates still existing today are the
jawless fishes, cyclostomes, and hagfish. The
brains of these animals possess all of the features
of vertebrates, including five divisions of the
brain: medulla, pons, midbrain, diencephalon,
Sulco-comissural Arteries and telencephalon (Fig. 11). Even though some
earlier fishes lack a cerebellum (lampreys and
Fig. 8 hagfish), its presence becomes a regular factor in
later fish evolution. Other than a primitive cere-
bellum, in fishes we see a small but effective
olfactory lobe and a primitive three-layer cortex,
The Brain: From Fishes to Humans the forerunner of the hippocampus. The “higher
center” of movement is controlled by what will
When thinking about the brain neuroembryology eventually become the present-day basal ganglia
and neurophylogeny, it is useful to conceptualize (Fig. 11). The spinal cord will remain essentially
its structure as a complex spinal cord, supplied by unchanged, supplied by segmental radiculo-
segmental vessels. Once, together with higher medullary arteries. The vertebral arteries form as
organism complexity, more complicated anatom- a result of longitudinal anastomoses between
ical structures are added on, we will see a more metameric branches of the cervical somites
sophisticated vasculature pattern that is better (Fig. 12).
understood keeping in mind the very simple In the brain two large longitudinal vessels – the
basic segmental arrangement of the spinal cord. carotids – travel up to the neck and enter the
We can think of the developing brain as a progres- cranial cavity, in a way similar to that of segmental
sively enlarging segment of the cord, requiring radiculomedullary vessels in the cord (Fig. 12).
larger surface and deep perforator vessels as key Inside the skull, the internal carotid artery gives
to understanding evolution and vascular rise to paired longitudinal arteries: one that goes
1 Cerebrovascular Development and Evolution 7

Fig. 9 Frontal view from a spinal angiogram, subtracted spinal artery (black arrow). In this particular case, the
and unsubtracted views, demonstrating a radiculo- radiculomedullary vessel arises from the T9 intercostal
medullary vessel (blue arrow) giving rise to an anterior artery (white arrow)

Fig. 10 Frontal view from


a spinal angiogram,
subtracted view. Anterior
spinal artery (red arrows)
originating from a
radiculomedullary branch
(orange arrow) arising from
the costocervical trunk
(purple arrow). This
conformation attests to the
metameric nature of the
system, where the spinal
artery can arise from
longitudinal vessels other
than the vertebral artery. A
tumor blush (yellow arrow)
from T1 hepatoma
metastasis is present, along
with several coil masses
(black arrows) deposited
after embolization of the
left-sided tumor soft tissue
component
8 M. Shapiro and E. Raz

up, the “cranial ramus,” and one extending cau- the forerunner of the PComm and of a small part
dally, the “caudal ramus” (Fig. 12). The “cranial of the basilar artery, which, at this point, is already
ramus” is the forerunner of the anterior cerebral fused at midline. At this point of development, a
and middle cerebral arteries. The caudal ramus is functionally significant anastomosis between the
caudal ramus and the posterior circulation system
is not activated yet, and the entire intracranial
CNS (including the brainstem and the cerebellum)
is supplied by the carotids.
Olfactory
Lobe
The cranial ramus has two notable branches:
the medial olfactory branch, which is the future
Primitive ACA, and the lateral olfactory branch, which is
Cortex the forerunner of the Heubner and of the anterior
(mostly choroidal artery. The caudal ramus gives off a
forerunner of tectal artery which supplies the posterosuperior
hippocampus) aspect of the midbrain; from this vessel comes
off a cerebellar artery supplying the primitive
cerebellum – forerunner of the superior cerebellar
Forebrain artery.
Segmental anastomoses also exist, usually in
Midbrain very small form, between the carotid and vertebral
systems. In the human embryo, these pathways
are quite functional at early stages while they
Cerebellum regress as the vertebrobasilar system. Occasional
persistence of these channels leads to the presence
of anomalous vessels such as the persistent tri-
Cord geminal artery, the persistent hypoglossal artery,
or the proatlantal arteries, depending on the seg-
ment that failed to develop.
Fig. 11

Fig. 12 Medical Olfactory

Lateral Olfactory

Cranial Ramus

Caudal Ramus
Carotid Tectal
Cerebellar
Fused Caudal Ramus (Basilar)

Segmental Inferior Cerebellar (PICA)


Carotido- Vertebral
vertebral
Anastomoses
1 Cerebrovascular Development and Evolution 9

Fishes Amphibians
Medial Olfactory
Lateral Olfactory
Lateral Striate
Posterior Telencephalic
(future Ant. Choroidal)
Cranial Ramus
Caudal Ramus
Posterior Choroidal
Tectal
Cerebellar
Fused Caudal Ramus (Basilar)
Inferior Cerebellar (PICA)

Vertebral
Segmental Carotido -
vertebral Anastomoses

Carotid

Fig. 13

Amphibians cranial ramus arise multiple “perforators” supply-


The changes seen with the shift to amphibians are ing the cerebral hemispheres (Fig. 14). These
schematized in Fig. 13. The brain continues to multiple perforators will eventually become the
grow, with a larger primitive hippocampus and MCA. We understand now the concept that the
larger basal ganglia. This larger size of the very large human MCA is a relatively late
brain leads to enlargement of the supplying, phylogenetical acquisition and represents a
already existing branches and to the fusion of hypertrophied perforator branch of the anterior
smaller feeders into larger arteries (Fig. 13). cerebral artery (Fig. 14). From another perspec-
The lateral olfactory branch has enlarged with tive we can also understand how the MCA can be
additional named vessels and now consists of embryologically and phylogenetically considered
two main branches – the lateral striate artery, a branch of the ACA.
forerunner of the Heubner, of the Middle cerebral The “posterior telencephalic branch” of the
artery (MCA), and of the MCA perforators, and ICA, forerunner of the anterior choroidal, supplies
the “posterior telencephalic branch,” forerunner the posterior telencephalic structures and is
of the anterior choroidal artery (Fig. 13). On the connected anteriorly with the olfactory artery;
caudal ramus, the tectal/cerebellar complex this function will continue to be fulfilled until
assumes supply of a developing choroid plexus very late in mammals’ evolution, when the poste-
with a posterior choroidal branch. The anastomo- rior brain territory will be annexed by the “poste-
sis with the posterior circulation is still absent in rior cerebral artery.” Other important steps in
these animals. reptiles are the formation of an anterior anastomo-
sis between the medial olfactory arteries, which is
Reptiles the future Acomm, and the enlargement of the
The changes seen with the shift from amphibians cerebellum, particularly of the more caudal ver-
to reptiles are schematized in Fig. 14. From the mis, which is supplied by the distal “basilar”
lateral striate (Heubner forerunner) branch of the system.
10 M. Shapiro and E. Raz

Fig. 14 Amphibians Reptiles


Medial Olfactory
ACOM
Lateral Olfactory
Lateral Striate
Lat. Striate perforators (MCA)

Posterior Telencephalic
(future Ant. Choroidal)
Posterior Choroidal
Tectal
Cerebellar

Inferior Cerebellar (PICA)


Vertebral
Segmental Carotido-
vertebral Anastomoses
Carotid

Fig. 15 Reptiles Birds


Medial Olfactory (ACA)
Lateral Olfactory
(Heubner)

Lat. Striate perforators


MCA
Posterior Telencephalic
(future Ant. Choroidal)

Posterior Choroidal
Tectal
Cerebellar

Inferior Cerebellar (PICA)


Vertebral
Segmental Carotido -
vertebral Anastomoses

Carotid

Birds of its territory (Fig. 15). This large telencephalic


In the birds a discrete MCA emerges as a domi- territory of the anterior choroidal will be lost at the
nant perforator from among a collection of lateral early stages of embryogenesis. In the birds, the
striate vessels, driven by rapidly enlarging cortical vertebral system remains confined to the cord or in
volume (Fig. 15). The posterior cerebral artery some cases lower medulla/vermian region
(PCA) of birds, which is in fact the anterior cho- (Fig. 15).
roidal, continues as the dominant parieto-occipital
supply source, while the tectal artery (which is the Mammals
real PCA) extends its coverage and forms a func- The biggest change in mammals regards the
tional anastomosis with the anterior choroidal development of the caudal ramus, since the
system, setting the ground for future annexation enlargement of the cerebellum and of the posterior
1 Cerebrovascular Development and Evolution 11

Fig. 16 Birds Mammals


ACA
(Heubner)
Lat. Striate perforators

MCA
Ant. Choroidal
Posterior Choroidal

Tectal (PCA)
Cerebellar
AICA
Inferior Cerebellar (PICA)

Segmental Carotido -
vertebral Anastomoses

Fig. 17 Mammals Human


ACA
(Heubner)
Lat. Striate perforators
MCA
Ant. Choroidal
Posterior Choroidal
Tectal (PCA)
Superior Cerebellar

AICA
Inferior Cerebellar (PICA)

Segmental Carotido-
vertebral Anastomoses

telencephalon place increasing demand on the portions of basilar territory, resulting in reversal
carotid system. Notwithstanding, also in some of craniopetal basilar flow to the craniofugal form
mammals such as the sheep, the vertebral system observed in monkeys, apes, and humans (Figs. 16
does not contribute to brain supply, and the basilar and 17).
artery is continuing to demonstrate craniopetal The phylogenetically novel participation of the
flow from the internal cerebral system. However, vertebral system in brain supply is maximized in
in higher mammals the carotid system appears to the human, where the posterior cerebral artery,
reach its functional limit, leading to progressive previously known as the tectal artery in the
annexation of the brainstem and cerebellar terri- “lower” species, is now functionally acquired by
tory by the vertebrobasilar system (Fig. 16). At the posterior circulation, even though develop-
first, this is confined to the PICA region. Progres- mentally it belongs to the carotids (Fig. 17).
sively, however, the vertebral artery annexes Even as such, frequent occurrence of “fetal”
12 M. Shapiro and E. Raz

Pcomm disposition attests to this relatively recent The way the vessels are arranged, i.e., trans-
phylogenetic acquisition (see embryology sec- verse vessels connected by longitudinal ones
tion). Recent territory shifts in this region (Fig. 18), is called metameric, because they ulti-
are borne out by marked variability in PICA- mately relate to the metamers, somites, segments,
AICA dominance, whereas the phylogenetically levels, etc., that are the basic building blocks of
older superior cerebellar arteries are much less our anatomy, similar to the arrangement found in
variable. the lower species.
The early arrangement centers on the six aortic
arches, which relate to the branchial arches
Vascular Neuroembryology (Fig. 19). The aortic arches consist of the dorsal
and ventral aortae, connected by longitudinal
Introduction anastomoses – the arches themselves. There are
a few adult arrangements which are almost
At about 4 weeks of gestation, the neural plate has constant:
just closed, and the brain is a tube with a central
canal of fluid, similar to the spinal cord. A series • The left-sided fourth aortic arch will become
of small vessels wrap around the brain and nourish the adult aortic arch.
it by simple diffusion in a similar way to that of the • The third arch will become the proximal inter-
spinal cord (Fig. 18). nal carotid artery.

Fig. 18
1 Cerebrovascular Development and Evolution 13

Fig. 19

• The dorsal aorta between the third and fourth Stage I (Prechoroidal Stage):
arch regresses, while homologous ventral aorta Approximately 4 Weeks
becomes the common carotid.
• The ventral aorta rostral to the third arch will There is a large vessel arising from the fourth
become the external carotid artery. aortic arch (Fig. 21). Keeping in mind that this is
a simplification, we will call it the common
Other than the dorsal and ventral aortae, there carotid. The place of origin of the internal carotid
is an additional longitudinal system, called the is shown, but again the external carotid is not
longitudinal neural system, related more displayed. The carotid system appears to feed
directly to the developing brain and spinal cord the entire brain, which is in fact true of the early
(Fig. 20), which consists of a series of metameric human embryology; the posterior circulation does
vessels with longitudinal connections. This longi- not exist yet. From this carotid vessel, a series of
tudinal neural system will eventually mature small arteries wrap around the brain and nourish it
into the vertebrobasilar system. We will go by simple diffusion (Fig. 21).
through six stages: the prechoroidal stage At this point, the carotid divides in two rami –
(stage 1), the choroidal stage (stage 2), the telen- the cranial and the caudal. The cranial ramus is
cephalic primitive stage (stage 3), the telence- simply a larger feeding vessel among the many
phalic intermediate stage (stage 4), the smaller vessels that wrap around the brain. It is the
telencephalic final stage (stage 5), and the adult forerunner of the anterior cerebral artery, which is
disposition (stage 6). the phylogenetically oldest telencephalic vessel.
14 M. Shapiro and E. Raz

Fig. 20

The caudal ramus feeds the midbrain and hind- These transverse arteries are in hemodynamic
brain and is the forerunner of the future Pcomm, balance with the caudal ramus of the carotid artery
P1, and part of the basilar. These adult segments in supply of the brainstem. As the longitudinal
therefore all belong embryologically to the ante- neural system matures, the future vertebral artery
rior circulation; this is a quite important concept to begins to meet to demands of the brainstem and
bear in mind in order to understand the vascular cerebellum, and, as a consequence, the anasto-
variation and pathology. Blood flow in the caudal motic transverse arteries typically regress, though
ramus at this point is craniocaudal. never fully disappear. Rarely, one of these vessels
Four transverse vessels are known to anasto- remains dominant and thus continues to supply
mose the caudal ramus with the ICA, the brainstem, for example, in the so-called per-
corresponding to the segmental arrangement men- sistent trigeminal artery. As was mentioned, the
tioned above. These segmental vessels are named posterior circulation does not exist as such in
trigeminal, hypoglossal, and proatlantal type embryological terms. Now look again at the
1 and 2 arteries (Fig. 22). They are very similar small vessels surrounding the brain. From
to the abovementioned transverse metameric among many of these feeders, a dominant vessel
arteries; they receive special attention because of eventually arises to wrap around the brain and
their relationship with branchial arteries and sprout its own small branches. The concept of
nerves. For example, the trigeminal artery sup- selection for a dominant vessel among many is
plies the trigeminal nerve, which is the nerve of very important, because it explains most “vari-
the first arch. ants” seen in the adult. From this arrangement it
1 Cerebrovascular Development and Evolution 15

Fig. 21

is clear that if two of these small vessels continue telencephalon. The choroid plexus of the dien-
to be in hemodynamic balance instead of one cephalon and midbrain is supplied by the artery
vessel being dominant, an apparent duplication called “posterior choroidal” (Fig. 24). The con-
may ensue. nection between telencephalic and diencephalic
central tube – where in the future the lateral and
third ventricles communicate through the foramen
Stage II (Choroidal Stage): of Monro – is important as the area of hemody-
Approximately 5 Weeks namic anterior choroidal/posterior choroidal bal-
ance. The choroid of the medulla (future 4th
The brain continues to be supplied entirely by the ventricle) is being supplied by a dominant feeder
anterior circulation, and the choroid (purple stuff that will become the PICA. Variations of posterior
over the blue tube in Fig. 23) develops as an fossa supply are common, but the 4th ventricle
invagination of the ependymal cells into the cen- choroid plexus is almost always supplied by a
tral canal from the “roof” of the brain and helps PICA-like vessel even when an AICA-PICA var-
supply nutrients to the brain from the inside of the iant is present.
neural tube. The choroid plexus is quite metabol- The early cerebellum is fed by a ramus that
ically active and promotes development of its own belongs to the anterior circulation, the future supe-
vascular supply. At this stage, a vessel is seen rior cerebellar artery. As a rule, the older the
coming off the cranial ramus to feed the plexus vessel, the more constant its presence. And this
of the telencephalon – this is the future anterior is particularly true for the SCA, which is essen-
choroidal artery (Fig. 23). It is a hemodynamic tially always present, while variations involving
balance with the distal cranial ramus (the ACA) the AICA and PICA are quite common (Fig. 25).
and is the earliest feeder of the posterior When a case of agenesis of these early vessels is
16 M. Shapiro and E. Raz

Fig. 22

considered carefully, the vessel is in fact found to the homologue of the future anterior spinal artery.
exist under an incorrect name. For example, ante- The one more laterally related to the vertebral
rior choroidal agenesis in fact represents an bodies will become the future vertebral artery.
enlarged anterior choroidal that supplies the In the anterior circulation, the growing cere-
PCA territory, while the latter is hypoplastic. bellum is going to annex the brainstem forerun-
This is confirmed by the observation that in ners of the AICA and PICA, and the branches of
most mammals the flow in the vertebrobasilar the cranial ramus develop branches of their own,
system is directed caudally and the reversal of some of which will become important adult struc-
flow of the posterior circulation is a very recent tures, such as the Heubner or the MCA, but at this
phenomenon. The important changes taking place point the ACA and the anterior choroidal continue
in the longitudinal neural system (LNS) are to dominate the supply of the telencephalon.
responsible for this and will be illustrated in the
next stage.
Stage IV: Approximately 7 Weeks

Stage III: 6 Weeks The LNS continues to “mature” in an interesting


way: the two longitudinal neural arteries that are
What happens is that the discontinuous LNS the forerunners of the anterior spinal artery and the
plexus coalesces to form actual distinct longitudi- basilar artery fuse into one vessel for most of the
nal vessels (Fig. 20). The one next to the cord is length of the cord and the brainstem. The whole
known as the “longitudinal spinal artery” and is vasculature is starting to resemble the adult
1 Cerebrovascular Development and Evolution 17

Fig. 23

pattern. Still, at this point the flow in the new change in flow direction seems to trigger some
basilar remains caudally directed. The PICA con- of the final events of embryogenesis. The relief of
tinues to expand to cover the cerebellum and the anterior circulation from the responsibility to
acquires its two basic divisions – choroidal and supply the posterior brain and the posterior
parenchymal. The trigeminal, hypoglossal, and fossa allows for continued growth of the PCA
proatlantal vessels continue to regress as the ver- and for development of the true MCA from sev-
tebral axis matures. eral ACA perforators. Indeed, conceptually and
As regards the anterior circulation, the most embryologically, the MCA is simply a very large
important change concerns the development of perforator with cortical territory, similar to the
the future “PCA,” which takes over the territory Heubner. For example, the accessory MCA vari-
previously supplied by the anterior choroidal ant is simply a case of two perforators being in
artery (Fig. 26). The growth of the PCA parallels relative balance and thus developing into
the development of the vertebrobasilar system, by MCA-like vessels.
which it will be annexed. But it is not known
whether it takes place after the flow in the basilar
has been reversed or before. Stage VI: Approximately Adult Pattern

One of the particular curiosities of the late vascu-


Stage V: Approximately 9 Weeks lar embryogenesis is the seemingly sudden
appearance of the Acomm (Fig. 28). Conceptu-
The posterior circulation has assumed a more or ally, it is easy to think of it as a short segment
less adult pattern (Fig. 27). At some point about fusion of the ACA in a fashion homologous to the
this time, the flow becomes craniocaudal. The basilar.
18 M. Shapiro and E. Raz

Fig. 24

To conclude, it is noteworthy to state how this A unilateral fetal posterior cerebral artery is seen
section only illustrates the basic framework of the in 25 % of cases, bilateral in 5 % cases (Figs. 29,
cerebral circulation, but the development of 30, and 31). In Figs. 30 and 32 examples of fetal
smaller vessels, countless branches of branches, Pcomm strokes are demonstrated. The existence
and of the penetrating vasculature continues well of a “duplicated Pcomm” is not possible – as may
after birth, as the brain matures and this level of be confused on occasion with a larger anterior
complexity is quite beyond our scope. choroidal artery.

Practical Aspects of Neurophylogeny Accessory MCA


and Neuroembryology
The MCA (purple in Fig. 33) arises as a dominant
Fetal Posterior Cerebral Artery vessel from a number of lateral striate perforators
of the anterior cerebral artery in the reptile. It may
This configuration is the most common “variant” happen in the case where two perforators remain
of the cerebral circulation. The fetal Pcomm, or dominant, resulting in the “accessory MCA”
fetal origin of the posterior cerebral artery, occurs (Fig. 33). The accessory vessel is, in fact, either
when the Pcomm is larger than the P1 segment a hypertrophied recurrent artery of Heubner – a
and supplies the bulk of the posterior cerebral medial ACA perforator – or another perforator-
artery territory. This reflects the effective persis- like vessel. To qualify as an MCA, the vessel must
tence in carotid supply of the PCA, a reminder of have cortical territory. In the cartoon in Fig. 33,
the phylogeny and embryology of this vessel. the schematic on the left side shows the accessory
1 Cerebrovascular Development and Evolution 19

Fig. 25

MCA configuration where both branches appear cortex remaining under anterior choroidal control
to originate proximal to the A1 complex (which is while PCA contribution is correspondingly
here defined as segment past the more “distal” reduced. This is sometimes erroneously consid-
MCA branch). These are known as “Manelfe ered a “duplicated PCA” (Fig. 36). Mistaking the
type 1” or “type 2” – depending on which branch choroidal artery for the Pcomm can have disas-
is larger. The important feature however is to note trous surgical consequences.
from which vessel the perforators originate and
whether they are medial or lateral. The schematic
on the right side shows the Heubner-type aMCA, Persistent Trigeminal Artery
known as “Manelfe type 3.” Check out the exam-
ples in Figs. 34 and 35. This most common embryonic carotid-
vertebrobasilar anastomosis at the mid-basilar
level represents persistence of segmental
Dominant Anterior Choroidal Artery ICA-basilar connection along the trigeminal
nerve (Fig. 37). The posterior communicating
The anterior choroidal artery, until late in evolu- artery is usually small in these cases.
tion, supplies a substantial portion of cortical ter-
ritory, which in the human is annexed by the PCA.
This annexation is determined by the proximity of Persistent Hypoglossal Artery
the two vessels in the subarachnoid space around
the midbrain. On occasion, the degree of annexa- More caudal than the persistent trigeminal artery,
tion is incomplete, with large portions of the the persistent hypoglossal artery enters the cranial
20 M. Shapiro and E. Raz

Fig. 26

vault through the hypoglossal canal, connecting Variations in Fusion Patterns


the distal vertebral and the proximal internal of the Basilar Artery
carotid arteries. It is, like the trigeminal and
proatlantal vessels, a segmental artery. Examples The basilar artery is formed via coalescence of
in Figs. 38 and 39. multiple channels belonging to the longitudinal
neural system, following development of the ante-
rior circulation. This process sets the stage for
Proatlantal Artery multiple basilar artery variations, which can be
considered from the standpoint of (1) extent of
An embryonic carotido-vertebral anastomosis, fusion and (2) completeness of fusion (i.e.,
the proatlantal artery is, in fact, the fenestrations).
occipital artery. A C1 segmental connection It is conceptually helpful to think of the basilar
between the occipital and vertebral artery is the artery as being “zipped” in the middle, with ver-
proatlantal type I; C2 level connection is tebral and PCA segments being “unzipped.”
proatlantal type II (Fig. 40). Usually, the vertebral Thus, both length and the integrity of the zipper
artery proximal to the proatlantal segment is determine the final configuration of the artery. As
hypoplastic. such, the artery may be:
1 Cerebrovascular Development and Evolution 21

Fig. 27

1. “Short” – corresponding to relative lack of aberration but instead a “deficiency” in basilar


fusion of the caudal segment fusion (Fig. 42).
2. Unfused at the top – corresponding to P1 origin of the superior cerebellar artery. The
“unzipping” at the basilar tip, less common SCA (blue) on the right arises from the P1 seg-
but much more confusing ment. Further “zipping” up would elongate the
3. Fenestrated – broken zipper in the middle basilar trunk, foreshorten both P1 segments, and
shift the SCA onto the basilar (Fig. 43). When
The first one, short basilar, is in fact a contin- especially prominent, basilar tip “unzipping” can
uum in terms of fusion extent (Fig. 41). There is lead to some confusion, as in this patient reported
no “number” to guide what short or long is, but to have a basilar tip aneurysm on CTA. Catheter
some basilar arteries are clearly quite angiogram shows an unfused basilar tip with tor-
foreshortened (the tortuous basilar of the aged tuous P1 vessels simulating aneurysm, further
hypertensive is a different matter). The second complicated by the presence of a fetal Pcomm
variation, involving lack of basilar tip fusion, on the right.
can generate a lot of confusion (Fig. 42). Effec- The third, basilar fenestration (Fig. 44), is quite
tively, the top of the basilar is split in two, so that common and usually of little clinical significance,
one or both superior cerebellar arteries originate except when it is so short as to mimic a dissection
from the P1 segment. This variant is not, there- (Fig. 45). Occasional species, including some
fore, a primary superior cerebellar artery monkeys, do not have a basilar artery at all. The
22 M. Shapiro and E. Raz

Fig. 28

Fig. 29 Bilateral fetal Pcomm (blue arrows) which reflect effective persistence in carotid supply of the PCA. The P1
segment of the PCA (brown) is hypoplastic
1 Cerebrovascular Development and Evolution 23

Fig. 30 CTA reconstruction demonstrating a right fetal related to a clot in both the fetal Pcomm and the MCA with
Pcomm (black arrows). This disposition was particularly a very large infarct. The AChoA territory was preserved
unfavorable in this patient when he/she developed a stroke

Fig. 31 Fetal Pcomm on a cerebral angiogram marked in Notice other unusual arrangements in this case, including a
red on left ICA injection lateral projection. AP vertebral somewhat low origin of the left superior cerebellar artery
artery injection shows a small left P1 segment (orange), as (yellow) and a basilar fenestration (brown)
compared with normal course of the right PCA (green).
24 M. Shapiro and E. Raz

Fig. 32 PCA territory infarction in a case of “fetal the right ICA. Typical diminutive appearance of the basilar
Pcomm.” Time to Peak and rCBV maps (top) demonstrate artery in a patient with bilateral fetal Pcomms is seen on
a completed infarction in right PCA territory. The embolus bottom right T2 image
(right lateral ICA injection on bottom left) originated from

caudal rami are unfused throughout, mimicking identical one, since these branches actually
the anterior cerebral disposition. supply separate territories. Understanding that
arteries in essence are dominant collectors arising
from a number of potential homologous candi-
Duplications dates clarifies this occurrence. The most com-
monly duplicated vessel is the superior
As in the case of accessory MCA, essentially any cerebellar artery. Duplicated AICA is quite rare
vessel on occasion is gifted with a twin, not an (Fig. 46).
1 Cerebrovascular Development and Evolution 25

Anterior Cerebral Artery Variations

The number of potential ACA variations may


seem mind-boggling. Phylogeny and embryology,
however, help make sense of frustration and con-
fusion. The appearance of the ACA system can be
conceptualized in terms of two factors – extent of
fusion and pattern of branch takeoff.
On the fusion side, you can expect anything
from no fusion – that is, no Acomm, various short
segment patterns of fusion (duplicated Acomm),
or elongated segments of fusion (unpaired or azy-
gos arrangements). The unpaired type is one
where a long segment of fusion finally breaks up
into two branches, each of which gives rise to
Fig. 33
individual cortical branches to its respective hemi-
sphere. The azygos type is where the ACAs are
fused for the entire length, with individual
branches to both hemispheres arising from a com-
mon trunk. Thinking of unpaired/azygos arrange-
ments as a kind of “basilar” is helpful and may not
be conceptually incorrect. Many species, such as
dogs, have a “normal” azygos arrangement with
“variants” of unpaired or “human-type” disposi-
tion. In the human, unpaired cases have a much
higher incidence of aneurysm formation at the
eventual branch point, – not a favorable evolu-
tionary development.
On the takeoff pattern side, you can have any
takeoff pattern imaginable, but of particular
importance is the arrangement at the Acomm
region. The frontopolar branch may arise from
the Acomm complex. More importantly, the
callosomarginal-pericallosal split may occur any-
where along the length of ACA from Acomm to
the callosum genu. Particularly when this split
occurs at the Acomm region, a triplicated appear-
ance may result with pericallosal and
callosomarginal trunks arising from one side and
regular A2 from the other, for a total of three
Fig. 34 Catheter angiogram of a patient with type I/II arteries (Fig. 47). Ultimately, ACA analysis in
accessory MCA. Notice how well MCA perforators can terms of some combination of fusion extent and
be seen, originating from the smaller and more distal of the branch takeoff can clarify any ACA arrangement.
two MCA vessels
26 M. Shapiro and E. Raz

Fig. 35 Type III (Heubner-


like) aMCA originating
from the Acomm complex

Fig. 36 The anterior choroidal artery (black), until late in large portions of the cortex remaining under anterior cho-
evolution, supplies a substantial portion of cortical terri- roidal control (blue), while PCA contribution is corre-
tory, which in the human is annexed by the PCA (brown). spondingly reduced (green). On MRA images, the
On occasion, the degree of annexation is incomplete, with Pcomm is labeled in red and choroidal in yellow

anatomy, the physiology, and the pathology of


Conclusion the central nervous system. Countless are the
cases in which this knowledge comes in handy
The deep understanding of the phylogenesis and in the real life of a neuro doctor, radiologist,
embryogenesis of the brain and spine vasculature clinician, or surgeon.
is paramount to develop a good perception of
1 Cerebrovascular Development and Evolution 27

Fig. 37 This diagram


demonstrates the
persistence of the trigeminal
artery. The angiographic
images illustrate a persistent
trigeminal, an accessory
MCA, and an associated
aneurysm treated with coils.
Notice the classic tau shape
of the trigeminal artery in
the lateral view

Fig. 38 More caudal than


the persistent trigeminal
artery (green arrow on the
diagram), the persistent
hypoglossal artery enters
the cranial vault through the
hypoglossal canal,
connecting the distal
vertebral and the proximal
internal carotid arteries. It
is, like the trigeminal and
the proatlantal vessels, a
segmental artery
28 M. Shapiro and E. Raz

Fig. 39 Different patient. Source and reconstructed MRA vertebrals and Pcomms are hypoplastic. Notice the flow
demonstrating a large vessel traversing through the hypo- void on the axial T2-weighted image
glossal canal and becoming the basilar artery. Both

Fig. 40 Stereo AP and lateral views of left ECA injection, (red). This is the proatlantal type I; the proximal occipital artery
opacifying the vertebrobasilar system (yellow) via the C1 (red) is the proatlantal. Occipital artery distal to the vertebral
segmental artery (purple) connection to the occipital artery anastomosis is blue, and internal maxillary artery is black
1 Cerebrovascular Development and Evolution 29

Fig. 41

Fig. 42

Fig. 43 P1 origin of the


superior cerebellar artery.
The SCA (blue) on the right
arises from the P1 segment.
Further “zipping” up would
elongate the basilar trunk,
foreshorten both P1
segments, and shift the SCA
onto the basilar
30 M. Shapiro and E. Raz

Fig. 44

Fig. 45

Fig. 46 Rare case of


duplicated AICA (brown) is
shown on diagram and
frontal view of left vertebral
artery injection
1 Cerebrovascular Development and Evolution 31

Fig. 47 Lateral projection angiogram with early misleading term since not three but two ACA arteries are
callosomarginal takeoff (purple) and pericallosal in red. present. It is the A2 segment on the right that is absent
Corresponding MRA image with left A2 shown in yellow because of early bifurcation into callosomarginal and
and Acomm in dark blue. This is the appearance of a pericallosal
“triplicated ACA” which is discussed above; it is a

8. Lasjaunias P, Manelfe C (1979) Arterial supply for the


References upper cervical nerves and the cervicocarotid anasto-
motic channels: systematization of radiological anat-
1. Abanou A, Lasjaunias P, Manelfe C, Lopez-Ibor L omy. Neuroradiology 18(3):125–131
(1984) The accessory middle cerebral artery 9. Lasjaunias P, Moret J, Manelfe C, Théron J, Hasso T,
(AMCA). Diagnostic and therapeutic consequences. Seeger J (1977) Arterial anomalies at the base of the
Anat Clin 6(4):305–309 skull. Neuroradiology 13(5):267–272
2. Butler AB, Hodos W (2005) Comparative vertebrate neu- 10. Lasjaunias P, Santoyo-Vazquez A (1984) Segmental
roanatomy: evolution and adaptation. Wiley, Hoboken agenesis of the internal carotid artery: angiographic
3. Campos C, Churojana A, Rodesch G, Alvarez H, aspects with embryological discussion. Anat Clin
Lasjaunias P (1998) Basilar tip aneurysms and basilar 6(2):133–141
tip anatomy. Interv Neuroradiol 4(2):121–125 11. Lasjaunias P, ter Brugge KG, Berenstein A (2007)
4. Dimmick SJ, Faulder KC (2009) Normal variants of Surgical neuroangiography: vol. 3: clinical and inter-
the cerebral circulation at multidetector CT angiogra- ventional aspects in children, vol 3. Springer Science &
phy. Radiographics 29(4):1027–1043 Business Media, Berlin
5. Gore AV, Monzo K, Cha YR, Pan W, Weinstein BM 12. Meckel S, Spittau B, McAuliffe W (2013) The persis-
(2012) Vascular development in the zebrafish. Cold tent trigeminal artery: development, imaging anatomy,
Spring Harb Perspect Med 2(5):a006684 variants, and associated vascular pathologies. Neuro-
6. Krings T, Baccin CE, Alvarez H, Ozanne A, Stracke P, radiology 55(1):5–16
Lasjaunias PL (2007) Segmental unfused basilar artery 13. Siqueira M, Piske R, Ono M, Marino Júnior R (1993)
with kissing aneurysms: report of three cases and liter- Cerebellar arteries originating from the internal
ature review. Acta Neurochir (Wien) 149(6):567–574; carotid artery. AJNR Am J Neuroradiol
discussion 574 14(5):1229–1235
7. Lasjaunias P, Braun JP, Hasso AN, Moret J, Manelfe C 14. Vignaud J, Hasso AN, Lasjaunias P, Clay C (1974)
(1980) True and false fenestration of the vertebral Orbital vascular anatomy and embryology. Radiology
artery. J Neuroradiol 7(3):157–166 111(3):617–626
Atherosclerosis: Biological and
Pathological Basis 2
Michele Porcu, Eytan Raz, and Luca Saba

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33 Atherosclerosis is a extremely complicated and
still well not perfectly understood pathologic
Arteries: Classification and Anatomical
Structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
process that involves the arteries of the human
body. It is a multi-step process that starts with
Normal Physiological Aspects . . . . . . . . . . . . . . . . . . . . . . . 34 the endothelial damage and can be influenced
Pathogenesis of Atherosclerosis . . . . . . . . . . . . . . . . . . . . . 34 by many factors, modifiable or not. The defi-
Endothelial Injury and Dysfunction . . . . . . . . . . . . . . . . . . 35 nition of the grade of stenosis determined by an
Plaque Growth and Evolution . . . . . . . . . . . . . . . . . . . . . . . . 35
Types of Plaque and Possible Complications . . . . . . . . . 36
atherosclerotic plaque and the identification of
vulnerable plaques are the main goals for diag-
Imaging Evaluation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
nosis, staging and therapies.
Therapeutic Options . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38 Keywords
Pathogenesis • Risk factors • Nitric Oxide
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
(NO) • Vulnerable plaque

Introduction

Atherosclerosis can be defined as a multifactorial


chronic inflammatory disease of the large (elastic)
and medium (muscular) arteries, characterized by
endothelial dysfunction, vascular inflammation,
and deposition of fibro-fatty particles in the con-
text of the vessel wall [1, 2]. This pathological
M. Porcu (*) • L. Saba
Department of Radiology, AOU of Cagliari, Monserrato, process is on the bases of a huge amount of com-
Cagliari, Italy mon clinical conditions, including myocardial
e-mail: micheleporcu87@gmail.com; lucasabamd@gmail. ischemia and infarction and cerebrovascular and
com; lucasaba@tiscali.it
peripheral vascular disease.
E. Raz In this chapter, we will briefly analyze the
Neurointerventional Radiology Section, Department of
biological and pathological basis of atherosclero-
Radiology, NYU Langone Medical Center, New York,
NY, USA sis, focalizing our attention in particular on the
e-mail: eytan.raz@gmail.com formation process of the atherosclerotic plaques.
# Springer Science+Business Media New York 2016 33
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_48
34 M. Porcu et al.

Arteries: Classification and Anatomical Table 1 Principal mediator produced by the endothelium
Structure Molecule Function
Nitric oxide (NO) Vasodilator
The arteries are those tubular shape structures of Prostacyclin (PGI2) Vasodilator
the human in which the blood flows from the heart Endothelin-1 (ET-1) Vasoconstrictor
to the periphery. Endothelium-derived contracting Vasoconstrictor
According to the caliber of the vessel, we can factors (EDCF)
recognize large arteries (caliber between 30 and
7 mm) and medium and small (caliber between are present: these small vessels are fundamen-
7 and 0.2 mm) arteries [3]. Because of the differ- tal for the nutrition of the tunica media and
ent characteristics of the tunica media (see below), could be an important site for
the large arteries are called even elastic arteries, neoangiogenesis and inflammatory
whereas those of medium and small caliber are processes [1].
called muscular arteries.
The vessel wall can be divided into three dif-
ferent layers, which are from the inner to the outer
(1, 2, 3, 4): Normal Physiological Aspects

(a) Tunica intima: it is the innermost, and it An artery is not a simple “conduct” for the blood;
consists of a single layer of endothelial cells by the opposite, it has to be considered a proper
(endothelium) that lie on their basement organ, with proper physiological functions
membrane. The endothelial cells have an (in particular the endothelium).
important role in the production and secretion The endothelial cells can produce and secrete
of several molecules that play a key role in the different molecules after mechanical or chemical
regulation of the blood flux, of the hemostasis, stimulation or the interaction with molecules
and of the inflammatory process. transported in the blood, such as the mediators
(b) Tunica media: this layer is the thickest of the of inflammation [1, 2, 4]: some of the most impor-
three, and it is composed of vascular smooth tant molecules produced by the endothelium are
muscle cells (VSMC), elastin fibers, and reported in Table 1.
extracellular matrix. The elastin fibers are The activity of the VSMC can be regulated
more represented in the tunica media of large directly by the sympathetic and parasympathetic
caliber arteries, whereas the tunica media of system, by the circulating catecholamines (adren-
medium and small arteries consists mainly of aline and noradrenaline) and other molecules such
VSMC. The VSMC are arranged on a circular as angiotensin II, antidiuretic hormone (ADH),
or spiral way. and corticosteroid [4].
The function of this layer in the elastic
arteries is to accumulate the kinetic energy
generated during the systolic phase of the
cardiac cycle, releasing it during the diastolic Pathogenesis of Atherosclerosis
phase in order to facilitate the blood flow to
the periphery; in the muscular arteries, it rules The pathogenesis of atherosclerosis is a compli-
the tone of the arteries and so the amount of cated and still not well-known multistep process.
blood which reaches the organs. Most of these processes are chained one to each
(c) Tunica adventitia: it is the outermost layer other, and a huge number of factors are involved.
and consists of connective tissue in continuity In this paragraph, we will try to explain the
with the perivascular connective tissues. In main elements and mechanisms which lead to the
the context of this layer, the vasa vasorum formation of the atherosclerotic plaque.
2 Atherosclerosis: Biological and Pathological Basis 35

Endothelial Injury and Dysfunction Table 2 Principal modifiable risk factors of


atherosclerosis
In paragraph two, we have seen how the endo- Principal modifiable risk factors
thelium has to be considered not just an internal Hypertension
smooth cover of the artery but an active tissue Dyslipidemia and hypercholesterolemia
important for many functions. On the surface of Obesity
the endothelium, a great number of receptors are Diabetes
present, for example, insulin, serotonin, hista- Smoking
mine, serum lipoproteins, and adhesion mole-
cules more expressed during the inflammatory
status. endothelial damage induced by the blood flow
The endothelium is also able to produce many (together with the other risk factors we have seen
chemical substances, including nitric oxide before).
(NO) via the endothelial isoform of the nitric
oxide synthases (eNOS) [1, 2, 5]. This chemical
mediator has a very simple chemical structure; it is Plaque Growth and Evolution
unstable, with a very short life (very low seconds),
and once it is produced, it acts on the VSMC Once the endothelial damage has started, the pro-
inducing vasodilation and slowing their prolifera- cess continues with the formation of the athero-
tion and reduces the expression of adhesion mol- sclerotic plaque.
ecules for macrophages on the endothelial surface The first lesions evident in the arteries are the
(see below) [6, 7]. so-called fatty streaks, which are linear lesions
The first step of the pathogenesis of the athero- well evident in particular in younger patients.
sclerosis is the endothelium damage [1, 2, 7]. These kinds of lesions tend to modify during the
This process occurs more frequently in those arte- years, according to various factors:
rial regions in which the blood flow is non-laminar
or turbulent (e.g., the bifurcations or the origin of • Non-modifiable factors (genetic, familiarity)
collateral vessels) [1], and it is favored by “risk • Modifiable factors
factors” such as hypertension, smoking, and
hypercholesterolemia through mechanisms like The modifiable factors include the “risk fac-
free radical oxidation and the activation of multi- tors” mentioned before, i.e., hypertension,
ple genetic pathways [2, 7]. dyslipidemia and hypercholesterolemia, obesity,
The endothelial damage is characterized by the diabetes, and smoke (Table 2): the presence of
expression of pro-inflammatory receptors on their these factors accelerates the process and forma-
surface, the reduction of the production of the NO, tion of the atherosclerotic plaques, increasing the
and the rearrangement of the junction and cyto- probabilities of minor and major cardiovascular
skeletal proteins [2, 8, 9]. The final results are an and cerebrovascular events and pathologies of the
increased endothelial cell turnover, an increased peripheral vascular system [10].
permeability of the endothelium with the recruit- At a microscopic level during this phase, the
ment of inflammatory cells (mainly scavenger macrophages, the T cells, and the VSMC are
macrophages), and the accumulation of lipopro- the most important cellular elements involved;
teins (in particular LDL; see below) beneath the they operate inside the growing lesion in an
tunica intima [2]. This process tends to have a inflammatory context, interacting in particular
positive feedback: the more the permeability, the with the low-density lipoproteins (LDLs).
more the inflammation, the more the lipid accu- LDLs are lipoproteins with a very high rich
mulation, the more the reduction of the caliber of content in cholesterol. These lipoproteins circu-
the lumen of the vessel, and the more the late free in the blood. When the endothelium is
36 M. Porcu et al.

damaged, the LDLs can infiltrate beneath the stiffening of the arterial walls, the physiological
tunica intima and accumulate into the growing mechanisms of narrowing and dilation of the arte-
plaque. At this level, they are susceptible to the rial walls in response to vasoactive molecules and
oxidative stress of various enzymes such as neurovegetative regulation are damaged. Even
myeloperoxidases and lipoxygenases (produced this condition further promotes the atherosclerotic
and released normally during an inflammatory plaque growing process.
process by the immunity cells). The oxidized The continuous growth of the plaque can
LDLs (oxLDLs) thanks to their cytotoxic action extend inside the lumen of the vessel (negative
promote the recruitment of monocytes from the remodeling) or on the external side (positive
blood circulation upregulating the expression of remodeling).
adhesion molecules on the surface of the endothe- The negative remodeling is important because
lial cells [2, 11]. Once they have penetrated inside it is responsible for the reduction of the blood flow
the plaque, they differentiate into resident macro- distally to the plaque, especially during stress
phages [1]. OxLDLs phagocytized by the macro- conditions. For example, a plaque that obstructs
phages are more resistant to degradation of their the lumen of the coronary artery limits the flux of
lysosome, and the macrophages continue to accu- blood to the territories supplied by that vessel: in
mulate cholesterol, becoming foam cells [2]. The the early stages, when the obstruction is not crit-
death of the foam cells led to the formation of the ical (up to 70 % of reduction of the vessel caliber),
necrotic lipidic core inside the plaque [1]. this condition could be compensated during rest,
The macrophages continue to feed this inflam- but during stress condition such as intensive phys-
matory reaction producing chemokines such as ical activity, the presence of the plaque does not
monocyte chemoattractant protein-1 (MCP-1), allow an adequate dilation of the coronary artery
interferon-γ (INF-γ), and macrophage colony- and an adequate blood flow for the heart segments
stimulating factor (MCFS), which increase supplied by the vessel, inducing myocardial
recruitment of other inflammatory cells [2]. ischemia which manifests with pain and
Some of these chemokines attract in the grow- neurovegetative symptoms.
ing plaque and stimulate the proliferation of the On the other hand, the positive remodeling is
VSMC [1]. These cells produce extracellular pro- responsible for the dilation of the vessel involved
teins such as collagen and elastin, producing a in the atherosclerotic process and can lead to the
fibrous cap over the plaque and contributing to formation of aneurysm. The evolution of the
the stability of itself; other molecules produced, aneurismatic lesions is highly variable, even if
the proteoglycans and fibronectin, create a reticu- the most terrible is the rupture of the vessel and
lar web that contribute to trap LDLs inside the the consequent hemorrhage, often fatal.
lesion [12]. Other molecules produced are the
matrix metalloproteinases (MMPs), whose role
in the degradation of the extracellular matrix is Types of Plaque and Possible
very important in the macrophage migration and Complications
destabilization of the plaque [13, 14].
At a microscopic level, it has been seen that as The natural evolution of the atherosclerotic plaque
the plaques increase in complexity, mineralization is highly variable, according to many factors,
processes can develop inside them, leading to the modifiable or not (see before). In particular, we
formation of calcifications [2]. In particular medi- can recognize two types of plaques, non-vulner-
ators of the bone metabolism have been evidenced able and vulnerable.
inside the plaque, close to the calcification The non-vulnerable plaques are characterized
centers [15]. by the presence of a small necrotic lipidic core and
Of course because of the involvement of the a thick fibrous cap and are less prone to rupture.
smooth musculature of the media layer in the The vulnerable plaques are those more prone to
atherosclerotic process and the subsequent rupture and thrombosis [16]. From a histological
2 Atherosclerosis: Biological and Pathological Basis 37

point of view, the vulnerable plaques are usually stenosis of the vessels but even to evaluate the
characterized by the presence of a large fatty composition of the plaques [19].
necrotic core, covered by a very thin fibrous cap The new techniques introduced in magnetic
[17]. The cap consists of molecules (in particular resonance imaging (MRI) are opening the way
collagen, elastin, and proteoglycans) produced for the characterization of the vulnerable
largely by the VSMC and protects the core of plaque [20].
the plaque from the direct contact with the blood Among the invasive methods of imaging, ves-
[16]. The necrotic lipid core is rich in sel angiography is still the gold standard method
prothrombotic molecules; after the fissuring of to evaluate the grade of occlusion of the vessel in
the plaque, the necrotic lipid core is exposed to some fields (e.g., the study of coronary arteries)
the blood flow, unleashing the coagulation cas- and allows even to operate directly at the level of
cade and the formation of thrombus; the thrombus the vessel-operating angioplasty, using stent in
can completely occlude the vessel determining order to make again patent the vessel or injecting
ischemia and then infarction of the tissues sup- thrombolytic drugs intra-arteriously in case of
plied by the vessels and/or can shatter and recent thrombosis. The intravascular ultrasound
embolize peripherally. A vulnerable atheroscle- (IVUS) is another promising technique that
rotic lesion could even fissure after an acute allows assessing the composition of the plaque
intra-plaque hemorrhage because of the rupture with a very small US probe placed at the tip of
of the thin-walled vasa vasorum [18]. an angiographic catheter [21].

Therapeutic Options
Imaging Evaluation
The most important concept to remember is that
Nowadays, we have many diagnostic imaging atherosclerosis is not a local disease, but it must be
methods to evaluate the gravity of the atheroscle- considered a systemic disease of the body; a local-
rotic disease (in particular trying to identify the ization of disease in the carotid arteries could be
vulnerable plaques, more prone to rupture) associated often with the presence of plaques at
suspected on the clinical and laboratory data. In the level of the coronary artery and vice versa.
this paragraph, we will briefly expose the main According to the localization, the characteris-
characteristics of these examinations. tics, and the dimensions of the atherosclerotic
Ultrasound (US) usually is the first imaging plaques, we have different therapeutic options,
technique used; it is safe, repeatable, and very pre- surgical (open surgery or endovascular treat-
cise in particular for the evaluation of the superficial ments) and nonsurgical.
vessels, such as supra-aortic trunks located in the Among the nonsurgical options, the most
neck, and the arteries of the upper and lower limbs. important are the removal and the treatment of
Multidetector computed tomography (MDCT) the modifiable risk factors; in particular, in case
is more accurate for the evaluation of atheroscle- of high levels of LDL, the use of statins could be
rotic plaque; it uses ionizing radiation, and an indicated. These drugs block the cholesterol syn-
intravenous iodinate contrast medium administra- thesis acting on the 3-hydroxy-3-methylglutaryl
tion is required in order to enhance the vessels and coenzyme A reductase at the hepatic level, but it
to evaluate the composition of the plaque and the has also been evidenced that they act directly at
grade of stenosis of the vessel. For example, the the level of the plaque inhibiting some inflamma-
evaluation of coronary arteries with a coronary tory processes, diminishing the recruitment of
computed tomography angiography (CTA) is not inflammatory cells inside the lesion, and increas-
an invasive method for the study of patients with ing the production of NO [22].
low to intermediate risk of coronary artery disease The surgical options are reserved for those
and allows not only to evaluate the grade of cases in which the other therapies have been
38 M. Porcu et al.

resulted insufficient; it is not the goal of this 9. Sima AV, Stancu CS, Simionescu M (2009) Vascular
paragraph to expose in detail the huge number of endothelium in atherosclerosis. Cell Tissue Res
335:191e203
surgical procedures at our disposal, and we invite 10. Whayne TF Jr (2011) Atherosclerosis: current status of
the gentle readers to deepen in the next chapters of prevention and treatment. Int J Angiol 20(4):213–222.
this book and in other specialist texts. doi:10.1055/s-0031-1295520
11. Tsimikas S, Miller YI (2011) Oxidative modification of
lipoproteins: mechanisms, role in inflammation and
potential clinical applications in cardiovascular
Summary disease. Curr Pharm Des 17:27e37
12. Doran AC, Meller N, McNamara CA (2008) Role of
In these pages we have seen how much compli- smooth muscle cells in the initiation and early progres-
sion of atherosclerosis. Arterioscler Thromb Vasc Biol
cated and variable is the pathogenesis of athero- 28:812e9
sclerosis, in which a central role is played by the 13. Heissig B, Hattori K, Friedrich M, Rafii S, Werb Z
endothelium damage and its subsequent dysfunc- (2003) Angiogenesis: vascular remodeling of the extra-
tion. Many factors, modifiable or not, influence its cellular matrix involves metalloproteinases. Curr Opin
Hematol 10:136e41
evolution. The imaging techniques are fundamen- 14. Visse R, Nagase H (2003) Matrix metalloproteinases
tal for the evaluation of the plaque and of the and tissue inhibitors of metalloproteinases:
vessel stenosis, leading the clinicians and the sur- structure, function, and biochemistry. Circ Res
geons to the correct therapeutic approach for the 92:827e39
15. Dhore CR, Cleutjens JP, Lutgens E et al (2001) Differ-
patient. ential expression of bone matrix regulatory proteins in
human atherosclerotic plaques. Arterioscler Thromb
Vasc Biol 21:1998e2003
References 16. Mishra TK et al (2013) An approach to the classifica-
tion, diagnosis and management of vulnerable plaque.
1. Douglas G et al (2010) The pathogenesis of atheroscle- J Indian College Cardiol 3:57–66
rosis. Medicine 42:9; 38(8):397–402 17. Virmani R, Burke AP, Kolodgie FD, Farb A (2002)
2. Tao W et al (2012) Atherosclerosis: pathogenesis and Vulnerable plaque: the pathology of unstable coronary
pathology. Diagn Histopathol 18(11):461–492 lesions. J Interv Cardiol 15:439e446
3. Balboni GC et al (2004) Anatomia Umana (terza edizione, 18. Depre C, Havaux X, Wijns W (1996) Neovascu-
tomo I, chapter 3). Edi.Ermes s.r.l., Milano larization in human coronary atherosclerotic lesions.
4. Rindi G et al (2005) Fisiologia Umana (IX edizione). Cathet Cardiovasc Diagn 39:215e220
UTET S.p.A. Divisione scienze mediche, Corso 19. Sun Z et al (2012) Coronary CT angiography: current
Raffaello, Torino status and continuing challenges. Br J Radiol
5. Hopkins PN (2013) Molecular biology of atheroscle- 85:495–510
rosis. Physiol Rev 93:1317–1542. doi:10.1152/ 20. Yang J et al (2013) MR and targeted molecular MRI of
physrev.00004.2012 vulnerable plaques. Interv Neurol 1(3–4):124–131.
6. Ignarro LJ et al (1987) Endothelium-derived relaxing doi:10.1159/000346767
factor produced and released from artery and vein is nitric 21. Kovárník T et al (2014) Current status of intravascular
oxide. Proc Natl Acad Sci U S A 84(24):9265–9269 ultrasound in interventional cardiology. Vnitr Lek
7. Vanhoutte PM (2009) Endothelial dysfunction; the first 60(12):1062–1067
step toward coronary arteriosclerosis. Circ J 73:595e601 22. Biasucci LM, Biasillo G, Stefanelli A (2010) Inflam-
8. Deanfield JE, Halcox JP, Rabelink TJ (2007) Endothe- matory markers, cholesterol and statins: pathophysio-
lial function and dysfunction: testing and clinical rele- logical role and clinical importance. Clin Chem Lab
vance. Circulation 115:1285e95 Med 48:1685e91
Nonatherosclerotic Vascular Disease:
Biological and Pathological Basis 3
Pierleone Lucatelli, Beatrice Sacconi, and Carlo Catalano

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39 Atherosclerosis is the main cause of vascular
disease in most cases; in about 10 % of cases,
Takayasu Arteritis (TA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
carotid artery disease is related to
Giant Cell Arteritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41 nonatherosclerotic causes, including several
Fibromuscular Dysplasia (FMD) . . . . . . . . . . . . . . . . . . . 42 unfrequent pathologies such as Takayasu arter-
itis, giant cell arteritis, fibromuscular disease,
Moyamoya Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
moyamoya syndrome, arterial dissection and
Extracranial Internal Carotid Artery extracranial carotid aneurysm. These entities
Aneurysms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
are discussed in the present chapter, with a
Craniocervical Arterial Dissection . . . . . . . . . . . . . . . . . 44 special focus on pathogenesis. Indeed, the
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45 aetiology of these diseases is in most cases
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
not completely known, since related to several
factors (genetic, immune and infectious). Early
diagnosis, usually leading to a good patient’s
outcome, is usually achieved after clinical
examination and imaging tests.

Keywords
Arterial dissection • Carotid artery disease •
Extracranial carotid aneurysm • Fibromuscular
dysplasia • Giant cell arteritis • Moyamoya
syndrome • Nonatherosclerotic disease •
Takayasu arteritis

Introduction

P. Lucatelli (*) • B. Sacconi • C. Catalano Atherosclerosis is identified as the main cause of


Department of Radiological, Oncological and Anatomo- vascular disease in around 90 % of cases; in the
Pathological Sciences, Sapienza University of Rome,
remaining 10 % of cases, nonatherosclerotic
Rome, Italy
e-mail: pierleone.lucatelli@gmail.com; beatrice. causes include several less common entities,
sacconi@fastwebnet.it; carlo.catalano@uniroma1.it such as Takayasu arteritis, giant cell arteritis,
# Springer Science+Business Media New York 2016 39
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_49
40 P. Lucatelli et al.

fibromuscular disease, moyamoya syndrome, mainly involves the media and adventitia, usually
arterial dissection, and extracranial carotid aneu- characterized by three stages: [5, 6].
rysm. The pathogenesis of these diseases is in
most cases unclear or even related to several fac- 1. A systemic stage, characterized by signs and
tors, such as genetic, immune, and infectious, symptoms of an acute inflammatory condition,
often associated with triggering events. The cor- such as fever, arthralgia, anemia, and increased
rect diagnosis is currently achieved after clinical erythrocyte sedimentation rate
examination and imaging tests; although US, CT, 2. A vascular inflammatory stage, when vascular
and MRA are useful, catheter angiography repre- stenosis and less frequently aneurysms occur,
sents the gold standard in diagnosing most of with corresponding signs and symptoms
these diseases. The majority of patients have a (stroke, transitory ischemic attacks, hypoten-
good outcome if the specific disease is diagnosed sive ischemic retinopathy with visual symp-
early. toms, vertebrobasilar ischemia, hypertensive
encephalopathy)
3. A burned-out stage, when fibrosis sets, usually
Takayasu Arteritis (TA) associated with remission According to the
American College of Rheumatology (ACR)
Takayasu arteritis (TA) is a granulomatous arteri- [7], the criteria for assessing the diagnosis are:
tis affecting the aorta and its branches [1]. The first
case was reported by Takayasu in 1905 [2]; lately, • Age at disease onset <40 years
the disease was more comprehensively described • Claudication of extremities
as “pulseless disease” by Shimizu and Sano in • Decreased brachial artery pulse
1951 [3]. It generally affects women in the first • BP difference >10 mmHg
fourth decades of life (nine females/one male), • Bruit over subclavian arteries or aorta
with a general incidence of 2.6 cases per million • Arteriogram abnormality, represented by arte-
per year in the USA and major prevalence in riographic narrowing or occlusion of the entire
patients of Asiatic origin [4]. aorta, its primary branches, or large arteries in
The etiology of TA is still unclear; the under- the proximal upper or lower extremities, not
lying pathologic process is inflammatory, with due to arteriosclerosis, fibromuscular dyspla-
several etiologic factors, either infective or auto- sia, or similar causes (changes usually focal or
immune, having been proposed. The most likely segmental)
hypothesis is that an unknown stimulus triggers
the expression of the 65 kDa heat-shock protein in A patient shall be said to have TA if at least
the aortic tissue which induces the major histo- three of these six criteria are present.
compatibility class I chain-related A (MICA) on According to the classification proposed by
vascular cells. The T cells and NK cells recognize Hata et al. [8], TA can be divided into six types
MICA on vascular smooth muscle cells and based on angiographic involvement:
release perforin, resulting in acute vascular
inflammation; pro-inflammatory cytokines are • Type I – branches of the aortic arch
also released and increase the recruitment of • Type IIa – ascending aorta, aortic arch, and its
mononuclear cells within the vascular wall. Th1 branches
lymphocytes drive the formation of giant cells • Type IIb – Type IIa region plus thoracic
through the production of interferon-γ and acti- descending aorta
vate macrophages with release of VEGF resulting • Type III – thoracic descending aorta, abdomi-
in increased neovascularization and PDGF, nal aorta, renal arteries, or a combination
resulting in smooth muscle migration and intimal • Type IV – abdominal aorta, renal arteries, or both
proliferation. The inflammatory cellular infiltrate • Type V – entire aorta and its branches
3 Nonatherosclerotic Vascular Disease: Biological and Pathological Basis 41

Hence, only types I, IIa, IIb, and V usually leading to vascular complications. In the majority
involve the carotid arteries, whereas the other of patients, a systemic inflammatory syndrome is
types more commonly involve the thoracic also present. The systemic and the vascular com-
descending and abdominal aorta and the renal ponents of giant cell arteritis seem to have differ-
arteries. ent underlying pathogenetic mechanisms: the
Angiography is the gold standard for diagno- vascular inflammation results from abnormal
sis, even though color Doppler imaging can be adaptive immune responses, whereas the systemic
useful in the first part of the diagnostic process, inflammation likely depends on an excessively
showing the mural thickening of the common activated innate immune system [13]. Despite
carotid arteries (hypoechoic in the early stages increased understanding of the inflammatory cas-
and then hyperechoic after the development of cade responsible for the disease process, the initial
fibrosis). CT, and especially MRA due to the event triggering the cascade remains uncertain;
young age of these patients, is also extremely the adventitia is more likely considered as the
helpful, even more during the follow-up [9]. If site of initial immunologic injury [14].
possible, whole-body MRI technique should be According to the American College of Rheu-
employed in order to exclude other localizations matology [15], the criteria for assessing the diag-
of disease [10]. nosis are:
TA is a chronic relapsing and remitting disor-
der. The overall 10-year survival rate is approxi- • Age at disease onset >=50 years
mately 90 %, this rate being reduced in case of • New headache
major complications, such as stroke, intracranial • Temporal artery abnormality (temporal artery
hemorrhage, and graft stenosis and/or occlusion. tenderness to palpation or decreased pulsation,
For this reason, strict management of traditional unrelated to arteriosclerosis of cervical
cardiovascular risk factors is mandatory in order arteries)
to minimize secondary cardiovascular complica- • Elevated erythrocyte sedimentation rate
tions. Approximately 20 % of patients have a • Abnormal artery biopsy (biopsy specimen with
monophasic and self-limited disease; in the artery showing vasculitis characterized by a
remaining 80 % of patients, TA requires immuno- predominance of mononuclear cell infiltration
suppressive treatment, resulting in remission in or granulomatous inflammation, usually with
around 60 % of cases [11]. multinucleated giant cells)

A patient shall be said to have GCA if at least


Giant Cell Arteritis three of these five criteria are present.
The disease usually has an acute or subacute
Giant cell arteritis (GCA), also known as temporal start, characterized by symptoms related to the
arteritis, is the most common form of vasculitis acute inflammatory status (fever, night sweats)
occurring in adults, with a prevalence of persons and headache, jaw pain, and blurred or double
with active or remitted GCA of 200 cases per vision; if the disease is not early diagnosed, com-
100,000 population aged 50 years or older. The plications may occur, such as blindness and less
female-to-male ratio is 3.7:1 [12]. It is a granulo- frequently stroke [9].
matous arteritis affecting large- or medium-sized The diagnostic process starts with clinical
vessels, usually the terminal branches of carotid examination and imaging studies. US findings
arteries (more frequently the temporal and oph- can show swelling of the vessel’s wall as a
thalmic arteries). The clinical manifestations of hypoechoic dark halo around the color-coded
giant cell arteritis result from two different pro- flow in the artery; the disease is segmental; there-
cesses. Inflammatory cells infiltrate the arterial fore, its visualization is suitable for localization of
wall and cause structural damage, eventually the biopsy [16]. CT and MR imaging can show an
42 P. Lucatelli et al.

abnormal wall enhancement of the affected arter- epidemiological evidence for the role of female
ies after contrast media injection in the acute hormones beyond the sex and age distribution of
phase, whereas vascular stenosis and aneurysm FMD has not been described yet. Although no
can be observed in the subacute-chronic phase. etiologic genes for FMD have been identified,
In the chronic phase, the differential diagnosis evidence supports a genetic basis for susceptibil-
between GCA and atherosclerotic disease may ity to FMD; a few authors reported the potential
not be easy, but the different localization of the inheritance pattern to be autosomal dominant
abnormalities (temporal and ophthalmic arteries with variable penetrance. However, several fac-
for GCA, ubiquitarious for atherosclerosis) can tors have limited the identification and character-
lead to the correct diagnosis. Standard test for ization of genes contributing to FMD, such as
definitive diagnosis is biopsy of the temporal disease rarity, variable phenotype, and gene-
artery, being more samples needed because the environment interactions [19]. FMD is usually
inflammation usually does not involve all parts described in terms of the affected arterial layer
of the artery [10]. and the composition of the lesions; depending
The prognosis for patients with untreated on the type of FMD, the narrowing (stenosis) of
GCA is extremely poor; these patients may suffer the artery is caused by an excess of either
blindness or death from myocardial infarction, the fibrous or muscular components of the arte-
stroke, or dissecting aortic aneurysm. On the rial wall [18].
contrary, with prompt and adequate therapy, full Many people with FMD do not have any symp-
recovery is the rule. Symptoms from temporal toms; symptoms can occur if the stenosis is severe
arteritis usually improve within days of treatment enough to restrict blood flow through the affected
with corticosteroids, except for those symptoms artery or if dissection occurs. Symptoms of FMD
related to an effective vision damage that in the carotid artery include headaches, ringing or
occurred before initiation of therapy, which are “swishing” noise in the ears, or light-headedness;
often irreversible. The mean duration of treat- advanced cases of FMD can cause stroke or a
ment is 2 years [17]. transient ischemic attack [9].
FMD is often accidentally diagnosed when the
beaded appearance in the arteries is observed dur-
Fibromuscular Dysplasia (FMD) ing examinations performed for other reasons.
Noninvasive imaging studies such as duplex ultra-
Fibromuscular dysplasia (FMD) is an angiopathy sound, MRA, and CTA can be used to confirm the
affecting medium-sized arteries, more frequently diagnosis of FMD and determine the extent of the
in young women of childbearing age. Among lesions. In general, angiographic studies are
patients with identified FMD, renal involvement performed only when the diagnosis is not clear
occurs in 60–75 % and cerebrovascular involve- or if the patient requires a therapeutic procedure
ment in 25–30 %; involvement of visceral arteries such as a balloon angioplasty [10].
and arteries of the limbs is less commonly The most common type of FMD is the medial
observed (about 9 % and 5 %, respectively); in fibrodysplasia (75–80 % of cases), affecting the
the case of cerebrovascular localization, the inter- tunica media; it is characterized by areas of vessel
nal carotid artery is more frequently affected stenosis alternating with areas of ectasia, resulting
(C2 segment) [9, 18]. in a classic “beads on a string” appearance on
The etiology of FMD is not known, even angiograms. Intimal and perimedial fibroplasias
though several factors have been considered as are less common (10 % of cases, respectively),
involved in its pathogenesis. More in detail, hor- caused by collagen deposits in the intima and in
monal factors such as estrogen have been pro- the outer portion of the tunica media; in the inti-
posed; however, although in the US Registry mal type, a concentric, smooth, narrowing (with-
91 % of registrants were female, clear supporting out beads) appearance of arteries can be observed,
3 Nonatherosclerotic Vascular Disease: Biological and Pathological Basis 43

whereas the perimedial type has a “beads on a According to the Classification of the Japanese
string” appearance, but with “beads showing a Health Ministry, there are four clinical forms of
smaller diameter in comparison to the normal the moyamoya disease: ischemic, hemorrhagic,
vessel” [20]. epileptic, and “other.” Clinical presentation in
When a patient is diagnosed with FMD in the children is usually ischemic, related to occlusion
carotid arteries, additional imaging studies may be of internal carotid artery or one of the branches of
obtained to evaluate the other blood vessels, espe- the Willis circle, resulting in paresis, sensory
cially vertebral arteries, Willis circle, and renal impairment, involuntary movements, headaches,
arteries. dizziness, or seizures; mental retardation is often
When FMD is present without any symptoms, present. In adults, the hemorrhagic form, espe-
it usually does not require intervention; risk fac- cially with subarachnoid hemorrhage, is more fre-
tors for vascular disease, such as high blood pres- quently observed as a result of hemorrhage of
sure, diabetes, and high cholesterol, should be fragile vessels [9, 26].
evaluated and treated, and imaging studies should In the affected cerebral vessels, pathological
be performed at regular intervals to evaluate the examinations do not show atherosclerotic or
disease progression. Angioplasty is recommended inflammatory lesions. Currently, the major pro-
for patients with FMD of the internal carotid teins believed to be involved in the pathogenesis
artery who experience TIAs or stroke related to are vascular endothelial growth factor (VEGF),
severe arterial narrowing. Stenting is rarely nec- basic fibroblast growth factor (bFGF), hepatocyte
essary only, in the case of carotid or vertebral growth factor (HGF), transforming growth factor-
artery dissection or carotid aneurysm. Surgery β (TGF-β), and granulocyte colony-stimulating
depends upon the location and the extent of dis- factor (G-CSF) [27]. The cause of stenosis is the
ease and consists in removing or bypassing the overgrowth of the smooth muscle layer, with
affected portion of the artery to restore normal thrombotic changes. The disease leads to different
blood flow; reconstructive surgery may be degrees of stenosis and occlusions of arteries of
recommended for patients with an aneurysm of the anterior part of the Willis circle and to the
the internal carotid or vertebral arteries [19, 21]. development of the collateral vasculature. The
vessels of the collateral circulation are formed as
a result of the widening of the existing vessels
Moyamoya Syndrome or development of new perforating arteries. There
are three main pathways of collateral circulation –
Moyamoya is a disease causing intimal thickening parenchymal, meningeal, and transdural. In the
of the walls and stenosis of the terminal branches vessels of the collateral circulation, there may
of the internal carotid arteries bilaterally; the term appear thrombotic changes, which are the cause
“moyamoya” (Japanese for “puff of smoke”) of ischemic symptoms. An increased blood flow
refers to the angiographic appearance of abnormal through thin collateral walls during stress, as well
vascular collateral networks that develop adjacent as the presence of microaneurysms, is the proba-
to the stenotic vessels [22, 23]. Moyamoya dis- ble cause of intracranial hemorrhages [28].
ease occurs primarily in Asians; the female-to- Before CTA or MRA were introduced to a wide
male ratio of moyamoya disease is 1.8:1. The clinical practice, the final diagnosis of the vascular
disease has two peaks of incidence in the first changes was based on cerebral angiography.
and fourth decades of life [8, 24]. The cause of Although CT examination is sufficient to diagnose
moyamoya disease is not clear. The disease is ischemic or hemorrhagic stroke in the course of the
believed to be hereditary; a few authors suggested disease, MRA can be considered as the first imaging
that the transmission may be autosomal dominant technique for definitive diagnosis, considering the
with incomplete penetrance based on age and young age of the patients [29]. To support the diag-
genomic imprinting factors [25]. nosis, the following findings should be observed:
44 P. Lucatelli et al.

• Stenosis or occlusion at the terminal portion of involved with the aneurysm and performing a
the internal carotid artery or the proximal por- bypass from the normal artery below the aneu-
tion of the anterior or middle cerebral arteries rysm to the normal artery above the aneurysm.
• Abnormal vascular networks in the vicinity of Stroke and cranial nerve injuries (particularly XII
the occlusive or stenotic areas pair) are potential risks for surgical treatment of
• Bilaterality of the findings (although some aneurysms with a diameter of more than 3 cm
patients may initially present with unilateral and/or extending to the skull base; endovascular
involvement) treatment may be preferred in appropriate cases as
an alternative to surgical therapy; since there is no
Mortality rates from moyamoya disease are risk of cranial nerve injury, it allows to treat
around 10 % in adults and 4 % in children. lesions that are hard to reach surgically, and
About 50–60 % of affected patients experience a there is no need for general anesthesia [31].
gradual deterioration of cognitive function, likely
due to recurrent strokes. The outcome of the dis-
ease depends on the severity of the hemorrhage, Craniocervical Arterial Dissection
the prognosis on recurrent attacks. Cases of mild
clinical course are normally treated conserva- Craniocervical artery dissection (CCAD) is one of
tively. In severe cases, surgery is indicated, the major causes of ischemic symptoms in young
including direct anastomoses, indirect procedures, adults. It has a prevalence of 2.6 cases per
and combined therapies [30]. 100, 000 persons per year; vertebral artery dissec-
tions are less common than carotid artery dissec-
tions. Genetic factors such as constitutional
Extracranial Internal Carotid Artery weakness of the arterial wall (such as in FMD or
Aneurysms in monogenic connective tissue disease, mainly
Ehlers-Danlos syndrome or Marfan’s syndrome)
Aneurysms of the extracranial internal carotid artery might have a role in the pathophysiology of
are extremely rare, being more commonly observed CCAD; environmental factors such as minor
in males (prevalence ranging from 0.1 % to 3.7 %); trauma act as a trigger [32]. Arterial dissections
most of them are of atherosclerotic origin. In case of begin with a tear in the intima or media resulting
nonatherosclerotic etiology, they can be congenital in bleeding in the arterial wall; expansion of the
(due to collagen disorders, FMD), infectious (TBC, wall by intramural blood causes compression and
HIV, mycotic), and posttraumatic. True aneurysms narrowing of the lumen, contributing to the for-
must be differentiated from pseudoaneurysms, mation of an intraluminal thrombus. The intramu-
which are usually iatrogenic (due to carotid punc- ral hematoma can create a false lumen that might
ture or endarterectomy). For a definitive diagnosis reconnect with the true lumen and forms parallel
of aneurysm, the diameter of the ectatic artery must flow; alternatively, wall rupture through the
increase of at least 50 %, if compared to the normal adventitia causes extraluminal bleeding. CCAD
vessel diameter [9, 10]. can be asymptomatic and discovered during rou-
Most of these aneurysms are asymptomatic, tine examinations. The most common symptom is
whereas TIA or stroke is the most common pain in the head and neck and in the region of
cause of hospital admission, related to emboliza- dissections, usually following a minor trauma. If
tion from the aneurysmal contents. Spontaneous the dissection compromises the arterial lumen or
rupture or bleeding is very rare, but fatal compli- causes thrombus formation, clinical symptoms are
cations are seen particularly in cases with mycotic related to ischemia; the closer the dissection to the
aneurysms [31]. brain is, the higher the possibility of brain infarc-
The mainstay of treatment of extracranial tion is present. On the contrary, if the dissection is
carotid artery aneurysms is surgical repair; it con- more extracranial, the probability of local symp-
sists in resecting the portion of the carotid artery toms from space-occupying lesions is higher;
3 Nonatherosclerotic Vascular Disease: Biological and Pathological Basis 45

bleeding in the subadventitial wall results in com- extracranial carotid aneurysm must be taken into
pression of the adjacent structures, such as lower account. These less common diseases have some
cranial nerves. Patients with subadventitial intra- similarities, such as an unclear pathogenesis and a
cranial dissections often present with subarach- good outcome if the specific disease is diagnosed
noid hemorrhage [32, 33]. early.
CCAD can be noninvasively diagnosed by
performing MRA and CTA [34]; color Doppler
flow imaging showed good results in visualization
of dissections, even though the main limitation is
References
represented by intracranial dissection (which is 1. Jennette JC (1994) Nomenclature of systemic vasculi-
the most common site of localization). The tradi- tis. Arthritis Rheum 37:187–192
tional method for CCAD diagnosis is catheter 2. Numano F et al (1996) Takayasu arteritis-five doctor in
angiography that may show irregular luminal the history of Takayasu arteritis. Int J Cardiol
54:S1–S10
narrowing occlusion, pseudoaneurysm, intimal 3. Shimizu K, Sano K (1951) Pulseless disease.
flap, double lumen, or distal branch J Neuropathol Clin Neurol 1.7–1.47
occlusion [35]. 4. Hall S et al (1985) Takayasu arteritis. A study of
There is no general consensus regarding opti- 32 North American patients. Medicine 64:89–99
5. Arnaud L, Haroche J, Mathian A (2011) Pathogenesis
mal management of internal carotid artery dissec- of Takayasu’s arteritis: a 2011 update. Autoimmun Rev
tion, but the choice among medical, endovascular, 11(1):61–67
and surgical options may depend on the type of 6. Matsunaga N, Hayashi K, Sakamoto I et al (1997)
and anatomic location of dissection; while surgery Takayasu arteritis: protean radiologic manifestations
and diagnosis. Radiographics 17:579–594
has a limited role, candidates for angioplasty and 7. Arend WP et al (1990) The American College of Rheu-
stent placement include patients with persistent matology 1990 criteria for the classification of
ischemic symptoms despite adequate Takayasu arteritis. Arthritis Rheum 33:1129–1134
anticoagulation, patients with a contraindication 8. Hata A, Noda M, Moriwaky R et al (1996) Angio-
graphic findings of Takayasu arteritis: new classifica-
to anticoagulant therapy, and patients with signif- tion. Int J Cardiol 54(suppl):S155–S163
icantly compromised cerebral blood flow [36, 37]. 9. Bartels E, Bartels S, Poppert H (eds) (2012) New trends
Overall, the prognosis for spontaneous internal in neurosonology and cerebral hemodynamics-an
carotid artery dissection is favorable, with about update. Perspect Med 1:244–249
10. Catalano C, Anzidei M, Napoli A (editors): (2013)
75 % of patients making a good recovery; the Cardiovascular CT and MR imaging. Springer-Verlag
reported mortality is less than 5 %. The functional Mailand Italia Srl; ISBN 978-88-470-2868-5.
outcome is also generally good, and recurrence of 11. Phillip R, Luqmani R (2008) Mortality in systemic
cerebral ischemia and carotid artery dissection is vasculitis: a systematic review. Clin Exp Rheumatol
26(5 Suppl 51):S94–S104
rare; the risk of recurrence is highest in the first 12. Salvarani C, Gabriel SE, O’Fallon WM, Hunder GG
month and then remains around 1 % per year (1995) The incidence of giant cell arteritis in Olmsted
for about a decade. Headache may persist for a County, Minnesota: apparent fluctuations in a cyclic
long time, in some cases for years, after the pattern. Ann Intern Med 123(3):192–194
13. Weyand CM, Goronzy JJ (2003) Giant-cell arteritis and
dissection [38]. polymyalgia rheumatica. Ann Intern Med
139:505–515
14. Weyand CM, Goronzy JJ (2000) Pathogenic principles
Summary in giant cell arteritis. Int J Cardiol 75(Suppl 1):S9–S15,
discussion S17–S9
15. Hunder GG, Bloch DA, Michel BA, Stevens MB,
Atherosclerosis is the main cause of vascular dis- Arend WP, Calabrese LH et al (1990) The American
ease, being responsible in around 90 % of cases of College of Rheumatology 1990 criteria for the classi-
carotid disease; in the remaining cases, fication of giant cell arteritis. Arthritis Rheum
33:1122–1128
nonatherosclerotic causes such as Takayasu arter- 16. Schmidt WA, Kraft HE, Vorpahl K (1997) Color
itis, giant cell arteritis, fibromuscular disease, duplex ultrasonography in the diagnosis of temporal
moyamoya syndrome, arterial dissection and arteritis. N Engl J Med 337:1336–1342
46 P. Lucatelli et al.

17. Borchers AT, Gershwin ME (2012) Giant cell arteritis: 28. Lim M, Cheshier S, Steinberg G (2006) New vessel
a review of classification, pathophysiology, formation in the central nervous system during tumor
geoepidemiology and treatment. Autoimmun Rev growth, vascular malformations, and Moyamoya. Curr
11(6–7):A544–A554 Neurovasc Res 3(3):237–245
18. Luscher TF, Lie JT, Stanson AW et al (1987) Arterial 29. Tarasów E, Kułakowska A, Łukasiewicz A et al (2011)
fibromuscular dysplasia. Mayo Clin Proc Moyamoya disease: diagnostic imaging. Pol J Radiol
62(10):931–952 76(1):73–79
19. Olin JW, Gornik HL, Bacharach JM et al (2014) 30. Burke GM, Burke AM, Sherma AK et al (2009)
Fibromuscular dysplasia: state of the science and crit- Moyamoya disease: a summary. Neurosurg Focus
ical unanswered questions: a scientific statement from 26(4):E11
the American Heart Association. Circulation 31. Yetkin U, Y€ urekli, G€
urb€
uz A (2006) Extracranial inter-
129(9):1048–1078 nal carotid artery aneurysms. Internet J Thorac
20. Begelman SM, Olin JW (2000) Fibromuscular dyspla- Cardiovasc Surg 10(2)
sia. Curr Opin Rheumatol 12(1):41–47 32. Debette S, Leys D (2009) Cervical artery dissections:
21. Olin JW, Pierce M (2008) Contemporary management predisposing factors, diagnosis and outcome. Lancet
of fibromuscular dysplasia. Curr Opin Cardiol Neurol 8:668–678
23(6):527–536 33. Caplan LR (2008) Dissections of brain-supplying
22. Kuroda S, Houkin K (2008) Moyamoya disease: cur- arteries. Nat Clin Pract Neurol 4:34–42
rent concepts and future perspectives. Lancet Neurol 34. Debette S, Provenzale JM (2009) MRI and MRA for
7(11):1056–1066 evaluation of dissection of craniocerebral arteries: les-
23. Scott R, Smith ER (2009) Moyamoya disease and sons from the medical literature. Emerg Radiol
moyamoya syndrome. N Engl J Med 360:1226–1237 16:185–193
24. Uchino K, Johnson A, Claiborne S, Tirschwell DL 35. Houser OW, Mokri B, Sundt TM et al (1984) Sponta-
(2005) Moyamoya disease in Washington State and neous cervical cephalic arterial dissection and its resid-
California. Neurology 65:956–958 uum: angiographic spectrum. Am J Neuroradiol
25. Mineharu Y, Takenaka K, Yamakawa H et al (2006) 5:27–34
Inheritance pattern of familial moyamoya disease: 36. Baumgartner RW (2010) Management of spontaneous
autosomal dominant mode and genomic imprinting. dissection of the cervical carotid artery. Acta Neurochir
J Neurol Neurosurg Psychiatry 77(9):1025–1029 Suppl 107:57–61
26. Shamim S, Kumar J, Jamalvi SW et al (2008) Moya 37. Xianjun H, Zhiming Z (2013) A systematic review of
Moya disease in a child. J Coll Physicians Surg Pak endovascular management of internal carotid artery
18(4):252–253 dissections. Interv Neurol 1(3–4):164–170
27. Weinberg DG, Arnaout OM, Rahme RJ et al (2011) 38. Schievink WI (2001) Spontaneous dissection of the
Moyamoya disease: a review of histopathology, bio- carotid and vertebral arteries. N Engl J Med
chemistry, and genetics. Neurosurg Focus 30(6):E20 344(12):898–906
Essentials of Transcranial Doppler
Ultrasound 4
Jonathan D. Kirsch

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47 Transcranial Doppler (TCD) ultrasound imag-
ing is a noninvasive, portable imaging tech-
Anatomy and Technique . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
nique that can be used to evaluate the
Physics of Doppler Ultrasound . . . . . . . . . . . . . . . . . . . . . 49 intracerebral arteries. TCD ultrasound has
Applications of Transcranial Doppler become an important modality in the monitor-
Ultrasound . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54 ing and evaluation of vasospasm of the intra-
Vasospasm . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54 cerebral arteries in patients with subarachnoid
Sickle Cell Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
Brain Death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
hemorrhage due to aneurysmal rupture.
Internal Carotid Artery Occlusion . . . . . . . . . . . . . . . . . . . . 58 Advances in transcranial ultrasound imaging
Other Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60 utilizing gray-scale, color Doppler flow, and
Ultrasound Biosafety . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63 spectral Doppler allow for direct visualization
and flow velocity measurements within the
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
vessels in real time. This is a significant
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65 improvement over the older “blind” technique
which utilized a range-gated Doppler probe
that inferred the vessels being interrogated by
a complex set of parameters including vessel
depth, flow direction, and probe orientation.
TCD has also been found to be useful in the
evaluation of patients with sickle cell disease
who may be at the risk of stroke and determi-
nation of brain death, internal carotid artery
(ICA) occlusion and collateral pathways, ste-
nosis, arteriovenous malformations, and intra-
cardiac right-to-left shunts.

Introduction
J.D. Kirsch (*)
Transcranial Doppler (TCD) ultrasound has
Department of Diagnostic Radiology, Yale University
School of Medicine, New Haven, CT, USA emerged as an important technique for imaging
e-mail: jonathan.kirsch@yale.edu of the intracranial vasculature. Originally
# Springer Science+Business Media New York 2016 47
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_36
48 J.D. Kirsch

developed as an indirect “blind technique” involv- internal carotid arteries (ICA) which bifurcate
ing range-gated spectral Doppler, the technique into the middle and anterior cerebral arteries
has advanced in recent years with the ability to near the skull base (MCA, ACA). The anterior
image and visualize the vessels directly utilizing cerebral arteries communicate via the anterior
gray-scale, color Doppler, and spectral Doppler communicating artery (AComm). Posteriorly,
imaging. TCD has found its greatest application the vertebral arteries join to form the basilar
and utility in the neurosurgical intensive care unit artery which then divides into the posterior
in monitoring for vasospasm in patients with cerebral arteries (PCA). The posterior communi-
intracranial bleeds due to ruptured aneurysms. cating arteries complete the circle joining the
However, the technique has also found promising internal carotid artery system anteriorly with
and potential uses in the evaluation of stenosis, the posterior circulation.
internal carotid artery occlusion, brain death, Transcranial Doppler imaging can be utilized
intracranial arteriovenous malformations, intra- to visualize the circle of Willis, the ophthalmic
cardiac right-to-left shunts, and sickle cell artery, and the cephalad portions of the vertebral
disease [1]. and basilar arteries at the skull base.
Although the intracranial vessels can be Imaging of the circle of Willis can be achieved
imaged by other techniques such as conventional by transcranial Doppler imaging. Before the
angiography, CT angiography (CTA), or MR advent of high-resolution gray-scale and color
angiography (MRA), these techniques require Doppler flow imaging, Aaslid et al. demonstrated
transport of the patient to the diagnostic radiology that cerebral arterial flow velocities could be mea-
department. Conventional angiography is inva- sured using pulsed Doppler US through the tem-
sive in nature with the potential risk of vessel poral bone or the foramen magnum [2]. The
damage or stroke. All three techniques involve technique was “blind” in that the vessels them-
the use of contrast agents which carry the risk of selves were not directly visualized. The identity of
anaphylactoid reaction for MR and CT contrast the vessel being interrogated was determined by
agents, nephrotoxicity for CT contrast, and the depth of the vessel from the skull, its flow
nephrogenic systemic fibrosis with MR contrast. direction, and the transducer orientation in rela-
CTA and conventional angiography, in addition, tion to the skull [3]. Using this complex approach,
require exposure to ionizing radiation. Aaslid was able to measure and determine normal
Transcranial Doppler ultrasound, on the other mean flow velocity (MFV) ranges of the intracra-
hand, is a noninvasive imaging modality that can nial arteries (Table 1). This technique had obvious
be performed portably, if necessary, in the ICU limitations related to lack of visualization of the
setting. TCD does not require the use of contrast vessels and the inability to angle correct the mea-
or ionizing radiation and serial follow-up exami- surements of flow velocity (see section “Physics
nations can be easily performed. This chapter will of Doppler Ultrasound”).
review the relevant anatomy, physics, and tech- With advances in ultrasound equipment and
niques of transcranial Doppler ultrasound and the the utilization of gray-scale, color Doppler flow,
interpretation of the findings. and spectral Doppler imaging, direct visualization
of the vessel and more accurate flow velocity
measurements are now possible. Imaging of the
Anatomy and Technique circle of Willis is performed via a transtemporal
approach utilizing a 5–1 MHz sector-array trans-
The circle of Willis is an anastomotic ring of ducer with most of the imaging being performed
arteries that sits at the base of the brain encircling at the lower frequencies (1–2 MHz) to allow pen-
the sella turcica and pituitary gland. It provides etration of the temporal bone. The transducer is
communication between the internal carotid placed on the temporal bone either above the
arteries and the vertebrobasilar system. The ante- zygomatic arch and anterior to the external audi-
rior portion of the circle is comprised of the tory canal or slightly more cephalad and posterior
4 Essentials of Transcranial Doppler Ultrasound 49

Table 1 Parameters for evaluation of the intracranial vessels with range-gated spectral Doppler. Before the advent
of transcranial Doppler utilizing direct visualization of the vessels with gray-scale and color Doppler flow imaging, the
cerebral vessel being interrogated by spectral Doppler had to be inferred from multiple parameters as noted in the above
table. The vessel being measured was inferred from its depth from the skull, its flow direction with respect to the
transducer, and the transducer orientation to the vessel and skull. Mean flow velocity (MFV) ranges shown are those
determined by Aaslid et al. [2]
Flow Transducer
Artery Depth (mm) Window direction orientation MFV (cm/s)
Ophthalmic artery 40–50 Orbital Toward Slightly medial 16–26
Middle cerebral artery 35–60 Temporal Toward En face 46–86
(MCA)
Anterior cerebral artery 60–75 Temporal Away Anterior 41–76
(ACA)
Posterior cerebral artery 60–75 Temporal Toward Posterior 33–64
(PCA)
Vertebral artery 45–75 Transforaminal Away Superior and 27–55
oblique
Basilar artery 70–120 Transforaminal Away Superior 30–57

over the earlobe. Imaging is performed from the intracerebral vessels. High-frequency (5–10
left and right temporal bones with color Doppler MHz) linear, curved, or sector transducers can be
flow imaging utilized to visualize the vessels. used depending on the size of the child. Utilizing
Flow velocity measurements (both MFV and the anterior fontanelle, the ICA, MCA, ACA, and
peak systolic velocity) are obtained. With the anterior communicating artery (AComm) can be
transtemporal approach, visualization of the visualized in the coronal plane (Fig. 3). Midline
MCA, ACA, PCA, and terminal ICA is possible. sagittal imaging allows visualization and flow
At our institution, spectral Doppler waveforms velocity measurements from the ACA,
and flow velocities are obtained from the proxi- pericallosal, callosomarginal, and frontopolar
mal, mid, and distal MCA and single measure- arteries in addition to the vein of Galen (Fig. 4).
ments are obtained in the visualized portion of the Patency of the superior sagittal sinus can be dem-
ACA, PCA, and terminal ICA. onstrated through the anterior fontanelle or an
The basilar and distal vertebral arteries can be open sagittal suture in the coronal or sagittal
visualized via a transforaminal approach. The ver- planes (Fig. 5). The posterior circulation can be
tebral arteries enter the skull base through the imaged through a transforaminal approach, as in
foramen magnum and then join to form the basilar the adult, or through an open posterolateral fonta-
artery at the base of the medulla oblongata. The nelle just posterior to the mastoid process.
basilar artery courses cephalad, anterior to the
medulla and pons, in the prepontine cistern. To
visualize these vessels utilizing the transforaminal Physics of Doppler Ultrasound
window, the transducer is placed in the midline
below the occiput and angled cephalad (Fig. 1). An understanding of the basic underlying physical
The ophthalmic artery and carotid siphon of principles related to spectral Doppler ultrasound is
the ICA can be visualized via the transorbital essential for the interpretation of transcranial
window. The probe is lightly placed on the closed Doppler measurements and results. All vascular
eyelid with a small amount of gel applied to the studies utilizing spectral Doppler make use of two
closed eyelid to aid in acoustic transmission well-known physical principles when measuring
(Fig. 2). flow velocities and evaluating for stenosis and/or
The open fontanelles in infants provide an vasospasm: the Doppler effect and Bernoulli’s
excellent acoustic window for visualizing the principle.
50 J.D. Kirsch

Fig. 1 Transtemporal and transforaminal acoustic win- Doppler flow imaging, the circle of Willis can be visualized
dows (a). The transducer is placed superior to the zygo- (b). The transforaminal approach allows visualization of
matic arch and anterior to the external auditory canal or the basilar and vertebral arteries (c). ACA anterior cerebral
more cephalad above the earlobe for the transtemporal artery, ICA terminal internal carotid artery, MCA middle
approach. For the transforaminal approach, the transducer cerebral artery, PCA posterior cerebral artery, PComm pos-
is placed in the midline below the occiput and angled terior communicating artery
cephalad. Utilizing the transtemporal approach and color

The Doppler effect, first described in 1842 by transducer at a certain frequency, fo, and is then
the physicist Christian Doppler, relates the change reflected back to the transducer from the moving
in frequency of a sound wave observed (or other red blood cells at a different frequency, fr. This
periodic event) if the source of the wave is moving frequency shift ( fd = fr  fo) and its relation to
relative to the observer. This effect is commonly velocity can be expressed by the following
seen in everyday life when an automobile with a equation:
siren passes by. The sound waves increase in
frequency as the car approaches the observer and c  fd

decreases in frequency as the car passes by 2 cos θ  fo
resulting in a frequency shift in the sound of the
siren heard by the observer. For flowing blood, a where v is the velocity of the flowing blood, c is
similar shift in frequency is noted when interro- the speed of the sound wave, and θ is the angle
gated by the sound beam from an ultrasound between the incident beam of the sound wave
transducer. The sound wave is emitted from the arising from the transducer and the direction of
4 Essentials of Transcranial Doppler Ultrasound 51

Fig. 2 Transorbital acoustic window: The transducer is carotid siphon can be visualized by color Doppler flow
placed lightly on the closed eyelid (a) and utilizing the imaging (b). OA ophthalmic artery (Image used with per-
globe as an acoustic window, the ophthalmic artery and mission from Kirsch et al. [1])

Fig. 3 Coronal color Doppler flow imaging through the anterior cerebral artery (ACA), M1 segments of the middle
circle of Willis (a, b), utilizing the anterior fontanelle as an cerebral arteries (MCA), and the intracranial internal
acoustic window, demonstrates the A1 segments of the carotid artery (ICA)
52 J.D. Kirsch

blood flow within the vessel (Fig. 6). From the its direction. A shift to a higher frequency is a
equation, it becomes apparent that the most accu- positive Doppler shift and indicates flow toward
rate flow velocities would be obtained with the the transducer. A shift to a lower frequency is a
transducer parallel to the direction of blood flow negative Doppler shift and indicates flow away
as θ = 0 and cos θ = 1. The greatest error in from the transducer. This frequency shift and
measurement and loss of signal would occur as directionality can be color-coded to indicate flow
the angle approached 90 and cos θ approaches direction. Conventionally with color Doppler
zero. It is recommended that velocity measure- flow imaging, red is used to indicate flow toward
ments be obtained with θ < 60 to obtain the the transducer and blue for away; however, this
most accurate measurements. color designation is arbitrary and can be switched
The Doppler equation allows one to not only so attention must be paid to the color bar on the
calculate the velocity of the flowing blood but also image. The color on the top of the bar is toward
the transducer and the color below is away
(Fig. 7).
Bernoulli’s principle, as applied to flowing liq-
uids, can be utilized for the diagnosis of areas of
stenosis and vasospasm. According to the princi-
ple, as fluid flows through conduits of differing
diameters, the velocity and pressure associated
with the fluid will change. As fluid flows from a
conduit (or blood vessel) of greater diameter to
one of smaller diameter, the flow velocity must
increase to allow the same volume of blood to
flow through the narrower area (this is analogous
to one putting one’s thumb over the opening of a
garden hose as water is flowing out resulting in a
high-velocity jet of water being formed).
Fig. 4 Sagittal midline color Doppler flow imaging Bernoulli’s principle and the law of conservation
through the anterior fontanelle demonstrates the of energy also state that the pressure in the region
pericallosal artery, the callosomarginal artery (CM), the
anterior cerebral artery (ACA), the vein of Galen, and the of increased velocity will decrease (Fig. 8). The
corpus callosum (CC) increase in velocity noted in areas of narrowing is

Fig. 5 Coronal gray-scale (a) and sagittal color Doppler flow imaging (b) through the anterior fontanelle demonstrate the
superior sagittal sinus (arrows)
4 Essentials of Transcranial Doppler Ultrasound 53

a mainstay for diagnosing stenosis or vasospasm


in blood vessels. As described above, these flow
velocities can be easily calculated by the
Doppler equation. As elucidated by Spencer and
Reid [4], there is a fairly linear relationship
between flow velocity and the degree of stenosis
(up to a critical stenosis at which point flow veloc-
ities decline due to extreme narrowing of the
vessel). In general, the higher the velocity of the
blood flow, the greater the degree of stenosis or
vasospasm.
Spectral Doppler allows measurements of flow
velocity in a specified area of a vessel (the sample
volume) over time and displays these flow veloc-
ities as a time/velocity curve throughout the car-
Fig. 6 Doppler effect. The velocity of blood flow can be diac cycle. From this waveform, peak systolic
calculated utilizing the Doppler effect. The ultrasound trans-
ducer emits a sound wave pulse at a certain frequency, fo, velocity (PSV), the velocity of the flowing blood
which is reflected off the moving red blood cells back to the at the peak of systole on the curve, and
transducer at a different frequency, fr. This resulting fre- end-diastolic velocity (EDV), the velocity just
quency shift ( fd = fr  fo) can be used in the Doppler before the next systolic upstroke, can be mea-
equation to calculate the velocity of the flowing blood. The
angle, θ, is the angle between the incident sound wave beam sured. The spectral waveform also indicates direc-
and the direction of the flowing blood. The most accurate tionality of flow. Conventionally, a waveform
flow velocity measurements are obtained with θ < 60 above the baseline (positive) indicates flow
(Image used with permission from Kirsch et al. [1]) toward the transducer and a waveform below the
baseline (negative) indicates flow away. Again,
close attention should be paid to the scale as this
can be inverted on the screen. The spectral line
and the area underneath it (the spectral envelope)
can indicate the type of flow present. A narrow
spectral line with a clear envelope beneath it indi-
cates fairly uniform (laminar) flow with a narrow
range of velocities. Filling in of the spectral win-
dow indicates a broader range of flow velocities
within the sample volume consistent with turbu-
lent flow seen with stenosis or narrowing (Fig. 9).
The amount of flow during the different portions
of the cardiac cycle provides information as to
whether the vessel is feeding a high-resistance
system such as a muscle bed or extremity or a
low-resistance system such as an end organ. High-
Fig. 7 Color Doppler flow imaging allows one to visual- resistance waveforms will have little to no flow
ize vessels more easily and also encode flow direction by
during diastole, whereas low-resistance wave-
the color map. The color bar in the upper right-hand corner
(thick arrow) denotes flow direction. Although the color forms will have more abundant forward flow
designation is arbitrary and can be reversed, the colors on during diastole.
the top half of the bar codes are for flow toward the The resistive index (RI) is a measure of
transducer (red) and the colors on the bottom, away
the resistance to the flow of blood into an
(blue). In this example, the right MCA is flowing toward
the transducer and the left MCA and PCAs are organ and is calculated utilizing the following
flowing away formula:
54 J.D. Kirsch

Fig. 8 Bernoulli’s principle. As fluid (blood) flows the area of narrowing and its pressure will decrease. This
through conduits of differing diameters, the velocity and principle, along with the Doppler effect, allows for ultra-
pressure of the fluid will change. As blood flows from a sonographic detection of areas of vasospasm or stenosis
vessel of greater to narrowed diameter (either due to vaso- (Image used with permission from Kirsch et al. [1])
spasm or stenosis), the velocity of the blood will increase in

Mean flow velocity (MFV) can be calculated


from the spectral Doppler waveform and is essen-
tially the average flow velocity over the time/
velocity waveform. Thresholds for PSV and
MFV have been determined that correlate with
varying degrees of vasospasm (see section
“Vasospasm”).

Applications of Transcranial Doppler


Ultrasound
Fig. 9 Spectral Doppler waveform. The spectral Doppler
waveform is a time/velocity curve that demonstrates the Vasospasm
blood flow velocities over time within a defined sample
window in the vessel being evaluated. The area below the Transcranial Doppler ultrasound has found its
curve or spectral line is called the spectral envelope or
greatest application in the neurosurgical intensive
window. With turbulent blood flow, this window will “fill
in” representing the myriad flow velocities associated with care unit screening patients for vasospasm sec-
turbulent flow (as seen in this example). The spectral ondary to subarachnoid hemorrhage (SAH) from
Doppler also can be used to measure peak systolic velocity aneurysmal rupture. Vasospasm is used to
(thick arrow) and end-diastolic velocity (thin arrow) from
describe both the clinical picture of delayed-
which the mean flow velocity and the resistive index can be
calculated (table to left of CDF image). A waveform above onset neurological deficits following SAH and
the baseline (positive) indicates flow toward the transducer the imaging findings of vessel narrowing. Vaso-
and a waveform below indicates flow away from the spasm from SAH has been associated with a
transducer
15–20 % risk of stroke or death [5]. About half
of those affected will demonstrate clinical signs
ðPSV  EDVÞ and symptoms. It has been reported that vaso-
RI ¼ spasm doubles the risk for mortality in SAH
PSV
[6–9].
In low-resistance waveforms, EDV will be fairly Vasospasm following SAH is generally absent
high as there is forward flow of blood into the in the first 72 h post bleed. Vasospasm generally
organ during diastole and the ratio will, therefore, occurs by day 3, peaking between days 6 and
be lower. In organs or systems that have a high 12, and then resolving 15–20 days after the
resistance to blood flow, EDV will fall toward bleed [10]. The clinical findings associated with
zero and the ratio will approach 1. vasospasm usually manifest themselves 4–5 days
4 Essentials of Transcranial Doppler Ultrasound 55

post hemorrhage. Insidious onset of confusion Table 2 MCA evaluation for vasospasm: Mean flow
and decreasing levels of consciousness are the velocity, peak systolic velocity, and Lindegaard ratio cut-
offs used at our institution for evaluation of varying
initial presenting signs which may progress to degrees of vasospasm in the middle cerebral arteries
focal neurologic defects, infarction, coma, and
Severity of MFV PSV Lindegaard
death. The severity and extent of vessel narrowing vasospasm (cm/s) (cm/s) ratio
has been seen to correlate with the clinical Mild 120–150 200–250 3.0–4.5
findings. vasospasm
The pathogenesis of cerebral vasospasm is Moderate 150–200 250–300 4.5–6.0
poorly understood but results primarily from vasospasm
prolonged smooth muscle contraction. Data sug- Severe >200 >300 >6.0
vasospasm
gests that calcium-dependent and calcium-
independent vasoconstriction is taking place.
Breakdown products from the blood in the sub-
arachnoid space may also be playing a direct or
indirect role. Other factors such as an imbalance Table 3 ACA and PCA evaluation for vasospasm:
between vasoconstrictors and vasodilators includ- Mean flow velocity and peak systolic velocity parameters
ing nitric oxide, prostaglandins, prostacyclin, free used at our institution for evaluation of the presence of
vasospasm in the anterior and posterior cerebral arteries.
radicals, and endothelin may also be involved in The Sloan ratio is calculated by dividing the MFV of the
the pathogenesis of vasospasm following ACA by the MFV of the distal extracranial ICA (NA not
SAH [11]. applicable)
Screening of patients at risk for vasospasm is MFV PSV Sloan
essential as patients cannot be placed prophylac- Vessel (cm/s) (cm/s) ratio
tically on treatment for potential vasospasm. Med- Anterior cerebral >80 >120 >4.0
artery
ical treatment for vasospasm consists of “triple-H
Posterior cerebral >85 >120 NA
therapy”: induced hypertension, hemodilution,
artery
and hypervolemia which are all aimed at improv-
ing cerebral perfusion. Calcium channel blockers,
such as nimodipine, have also been recommended the velocity, the greater is the degree of
but should be used cautiously to avoid the delete- vasospasm.
rious effects of hypotension [7]. Unfortunately, For intracranial vessels, cutoff values for PSV
these treatments carry their own inherent risks and MFV have been established that correlate
and potential complications including renewed with various degrees of vasospasm in the MCA
bleeding from the aneurysm site or into areas of and for the presence of vasospasm in the ACA or
infarction, increased cerebral edema, myocardial PCA (Tables 2 and 3). For the MCA, MFV in the
infarction, and congestive heart failure. If medical 120–150 cm/s range correlates with mild vaso-
therapy is ineffective, catheterization of the cere- spasm, whereas MFV in the 150–200 cm/s and
bral vessels can be performed with a vasodilating 200–250 cm/s ranges correlate with moderate and
agent injected. TCDI allows for quick and nonin- severe vasospasm, respectively [2, 4, 12].
vasive screening and monitoring of the patients in Torbey et al. have shown that the age of the
the ICU setting for vasospasm to determine if patient does play a role in the incidence and diag-
medical or interventional treatment is necessary. nosis of cerebral vasospasm. Older patients (>68
Vasospasm results in a narrowing of the vessel years) had a lower incidence of symptomatic cere-
lumen, and as noted above, the velocity of the bral vasospasm and a shorter window in which
blood flow within a vessel is inversely propor- vasospasm may occur (up to 10 days post bleed
tional to its diameter until a critical narrowing for older patients as compared to younger patients
(usually greater than 95 %) is reached at which that demonstrated symptomatic vasospasm as late
point blood flow through the extremely narrowed as 16 days following SAH). With age, cerebral
vessel actually decreases. In general, the higher blood flow velocities decrease and older patients
56 J.D. Kirsch

were seen to develop symptomatic vasospasm at more indicative of an etiology other than vaso-
lower MFV than younger patients [13]. spasm as the cause of the flow velocity elevation
Physiologic processes such as autoregulation [14, 15]. An accurate measurement of the extracra-
and hyperemia and medically induced conditions nial distal ICA MFV is important as slight varia-
including hypertension and hypervolemia may also tions in this measurement can result in over- or
result in elevated flow velocities within the intra- underestimating the presence of vasospasm. The
cranial vessels. To help distinguish between these velocity should be obtained as close to the skull
different etiologies for elevated flow velocity in the base as possible (Fig. 10).
MCA, Lindegaard et al. developed a ratio utilizing The Sloan ratio (MFV of the ACA divided by
the MFV of the MCA and the MFV of the distal the MFV of the extracranial ipsilateral ICA) can
extracranial ipsilateral ICA (VMCA/VICA). aid in the detection of vasospasm in the ACA. A
Lindegaard ratios in the range of 3.0–6.0 are indic- Sloan ratio greater than 4.0 is considered indica-
ative of mild to moderate vasospasm and ratios tive of vasospasm.
>6.0 are indicative of severe stenosis. Elevated Baseline and serial examinations are important
flow velocities with a Lindegaard ratio <3.0 is to obtain as elevations of flow velocities within a

Fig. 10 (a) Noncontrast CT demonstrates an aneurysmal respectively). The Lindegaard ratio, however, was mark-
SAH. Left frontal craniectomy has been performed to help edly elevated (11.5) consistent with severe vasospasm. (b)
relieve intracranial pressure (*). Transcranial Doppler 3D reconstruction from a CT angiography performed after
imaging (TCDI) demonstrates mild to moderately elevated the TCD revealed severe vasospasm of the left MCA
peak systolic velocity and mean flow velocities in the left (arrow, c) (Images used with permission from Kirsch
middle cerebral artery (280 cm/s and 150 cm/s, et al. [1])
4 Essentials of Transcranial Doppler Ultrasound 57

Fig. 11 (a) Initial transcranial Doppler of the right ante- showed a normal caliber vessel (arrow). TCDI obtained
rior cerebral artery (ACA) in a patient with SAH demon- 10 days later demonstrates the PSV and MFV to have more
strates peak systolic and mean flow velocities in the normal than doubled in the same vessel. (c) Repeat CT angiogram
range (67 cm/s and 32 cm/s, respectively). (b) 3D recon- now demonstrates vasospasm of the right ACA (d, arrow)
struction form a CT angiogram performed at this time (Images used with permission from Kirsch et al. [1])

vessel between subsequent exams may be indica- vertebral arteries and dividing this value into the
tive of vasospasm. Mean flow velocity increases highest MFV obtained in the basilar artery. A
of greater than 50 % or absolute increases in BA/VA MFV ratio of 2.0–3.0 plus a basilar mean
velocity of greater than 50 cm/s over a 24 h period flow velocity greater than 85 cm/s was found to be
may indicate vasospasm in the vessel (Fig. 11). indicative of mild to moderate vasospasm. A ratio
Through the transforaminal window, the distal >3.0 was found to be highly accurate in diagnosing
vertebral arteries (VA) and basilar artery (BA) can severe vasospasm [16].
be visualized and evaluated for vasospasm involv-
ing the posterior circulation. Sviri et al. demon-
strated good sensitivity and specificity utilizing a Sickle Cell Disease
ratio of the MFV of the extracranial vertebral artery
and the basilar artery in conjunction with MFV of Sickle cell disease is the result of a coding muta-
the basilar artery. The ratio was calculated using the tion in the beta chain gene for hemoglobin. The
average of the mean flow velocities of both mutation results in substituting thymine for
58 J.D. Kirsch

adenine, GAG to GTG, in the sixth codon. The subarachnoid hemorrhage are the chief causes of
resulting abnormal hemoglobin (HbS) under deoxy brain death in adults, whereas in children, abuse,
conditions forms polymers and demonstrates motor vehicle accidents, and asphyxia are more
increased viscosity and decreased solubility. This common etiologies [22, 23]. Clinically, the deter-
polymer formation in red blood cells (RBCs) mination of brain death includes documentation
which are homozygous for HbS results in the clas- of coma, absence of brain stem reflexes, and
sic sickled shape of the RBCs. Factors other than apnea. Confirmatory tests include cerebral angi-
local tissue hypoxia that can precipitate the sickling ography which should demonstrate no filling of
process include dehydration, infection, acidosis, the intracerebral vessels from their point of entry
and cold weather. Recurrent episodes of sickling into the skull, electroencephalography which
result in membrane damage and loss of the ability should demonstrate absence of electrical activity
of the RBCs to reattain their normal shape. These and lack of reactivity to intense somatosensory or
properties of HbS and the resulting physiological audiovisual stimuli, and cerebral scintigraphy
changes in RBCs result in the clinical and physical which demonstrate absence of radioisotope within
findings of the disease including hemolytic anemia, the brain [12]. TCDI has been found to have a
vaso-occlusive crisis, and multiple organ damage high sensitivity and specificity for the evaluation
due to microinfarcts [17]. of brain death and can be utilized to evaluate the
In children, the most common cerebrovascular intracranial vessels utilizing the transtemporal
manifestation of sickle cell disease is cerebral approach [24].
infarction due to occlusive vasculopathy. An Due to increased intracranial pressures,
approximately 11 % incidence of stroke is seen transcranial Doppler waveforms will demon-
in patients with sickle cell disease by the age of strate initially decreased forward diastolic flow.
20. Incidence of stroke is highest in the first As pressure continues to increase, reversed dia-
decade [18]. Fibrous proliferation of the intima, stolic flow will be noted resulting in a reverber-
usually within the intracranial ICA, proximal ating or oscillating flow pattern. Reversed
MCA, and ACA, leads to vessel narrowing, occlu- diastolic flow, however, is not pathognomonic
sion, stroke, and infarction. In stroke-free chil- for brain death as other entities such as mass-
dren, those with high cerebral blood flow occupying lesions, large strokes, edema, or her-
velocities in the distal ICA or proximal MCA niation may also result in increased intracranial
had a significantly increased risk of stroke [19]. pressure with resulting loss of diastolic flow.
Children with sickle cell disease and mean Small, low-velocity systolic peaks or spikes
flow velocities below 170 cm/s were considered may be noted (Fig. 12). Care must be taken in
at low risk for stroke in the 60 months post TCD. the interpretation of apparent lack or cessation of
Patients with MFV in the 171–199 cm/s range flow in the vessels as technical factors such as
were recommended to have more frequent TCD poor skull penetration by the ultrasound beam
testing. Adams et al. have shown that children may result in false-positive exams.
with mean flow velocities greater than 200 cm/s
have a significantly increased risk of stroke and
showed significant decrease in the risk of stroke Internal Carotid Artery Occlusion
when treated with blood transfusions that
decreased the HbS concentration to less than Atherosclerotic plaques consisting of necrotic
30 % of the total hemoglobin [20, 21]. cells, lipids, and cholesterol crystals can result in
stenosis of the internal carotid artery which, in
conjunction with superimposed thrombosis, can
Brain Death result in complete occlusion of the vessel. Emboli,
originating either from a cardiac source or ather-
Transcranial Doppler imaging can aid in the diag- oma from the aortic arch, are the other most fre-
nosis of brain death. Traumatic brain injury and quent cause of occlusion. The proximal 2 cm of
4 Essentials of Transcranial Doppler Ultrasound 59

Fig. 12 Brain death: (a–c) Transcranial Doppler demon- lack of intracranial arterial flow with an abrupt cutoff of
strates reversed diastolic flow (thin arrows) in the right and activity at the skull base because increased intracranial
left middle cerebral arteries and low-velocity, systolic pressure exceeds cerebral perfusion pressure. Note also
spikes in the anterior cerebral artery (thick arrow), wave- the “hot nose” sign which is related to the decreased flow
forms typical for brain death. (d) Noncontrast CT demon- within the internal carotid arteries compared to the
strates diffuse cerebral edema with loss of gray-white increased flow in the external carotid circulation (e)
matter differentiation. Nuclear scintigraphy demonstrates
60 J.D. Kirsch

the origin of the artery and the carotid siphon are stenoses greater than 50 % were elevations in flow
the two most common sites for occlusion. The velocities greater than 150 % at the site of stenosis
occlusion may be clinically silent due to a collat- compared to proximal or distal flow velocities in
eral pathway that forms from the external carotid the vessel or flow velocity ratios >2.5. Detection
artery to the ophthalmic artery which reverses its of turbulent blood flow, aliasing, visualization of
flow to feed the intracranial ICA/MCA. Duplex narrowing on color Doppler flow imaging, and
and transcranial Doppler imaging can demon- distal tardus-parvus waveforms have also aided
strate this collateral pathway. Ultrasonogra- in detection of stenosis [25, 26].
phically, the occluded internal carotid artery will Although not approved by the Food and Drug
be filled with echogenic material and will demon- Administration for clinical use in the United
strate absence of flow by color Doppler flow States, ultrasound contrast-enhancing agents
imaging and spectral Doppler. The common such as Levovist (Schering AG) can help aid in
carotid artery waveform, which has normally the detection of vessels and areas of stenoses
low resistance with forward diastolic flow, will [17]. Intracranial vessels may be difficult to see
frequently convert to a high-resistance waveform due to an insufficient temporal bone window, poor
due to the occlusion. If the collateral pathway insonation angle, or low flow velocities or vol-
develops, the common carotid artery may main- umes. Imaging after intravenous injection of
tain its normal low-resistance pattern. The ipsilat- Levovist, galactose-based microbubbles (<5 μm
eral external carotid artery, which normally has a in diameter) coupled with palmitic acid, has been
high-resistance waveform (little forward diastolic found to increase significantly the ability to detect
flow), will become “internalized” with its wave- vessels and obtain spectral Doppler waveforms
form changing from a high-resistance pattern to a compared to pre-contrast examinations. Detection
low-resistance pattern. The ipsilateral ophthalmic of collateral flow through the anterior and poste-
artery can be visualized via the transorbital rior communicating arteries in patients with high-
approach and will demonstrate flow reversal grade ICA stenosis or occlusion was also signifi-
away from the transducer and globe (Fig. 13). cantly improved with the use of an echo-contrast
Other collateral pathways are possible including agent [27, 28].
feeding of the anterior circulation from the poste- Transcranial power Doppler imaging has been
rior vertebrobasilar system via the posterior com- useful in the detection of intracranial aneurysm.
municating arteries or filling of the ipsilateral The detection of aneurysm by TCDI is dependent
MCA via the contralateral MCA and anterior com- on the size of the aneurysm, its location, and its
municating artery. morphology. Detection rates of 47–73 % have
been reported in the literature [29, 30]. Aneurysms
in the anterior communicating and basilar arteries
Other Applications had the highest detection rates (>50 %) as did
those greater than 10 mm in diameter. Aneurysms
TCDI has found a limited role in detection of in the posterior communicating and pericallosal
stenosis, aneurysms, arteriovenous malformations, arteries had the lowest detection rate. MCA and
and intracardiac right-to- left shunts. ICA aneurysms had detection rates of about 50 %.
For stenosis of intracranial vessels, TCDI has Morphologically, spherical and multi-loculated
been found to be useful but not definitive for the aneurysms were more easily detected than elon-
diagnosis. Confirmatory studies such as CTA or gated ones. The administration of IV echo-
conventional angiography are often necessary. An contrast agent significantly increased the detec-
elevation in flow velocity of 50–150 % (ratio of tion rate for aneurysms [31]. TCDI has also
1.5–2.5) at the site of stenosis as compared to the shown promise in the long-term surveillance of
flow velocity just proximal or distal to the stenosis aneurysms treated by detachable coils. High sen-
has been used by Roubec et al. as criteria for the sitivity and specificity was seen in the ability of
detection of stenoses less than 50 %. Criteria for TCDI to detect and distinguish aneurysms that
4 Essentials of Transcranial Doppler Ultrasound 61

Fig. 13 (continued)
62 J.D. Kirsch

Fig. 13 (continued)
4 Essentials of Transcranial Doppler Ultrasound 63

Fig. 13 Internal carotid artery (ICA) occlusion: Gray- The ipsilateral external carotid artery normally has a high-
scale ultrasound demonstrates echogenic material filling resistance waveform with little diastolic flow but has
the lumen of the right internal carotid artery (a, arrows). become “internalized” and demonstrates increased forward
Power and spectral Doppler demonstrates no flow within diastolic flow (arrow, e). The right ophthalmic demon-
the vessel consistent with occlusion (b, c). The common strates flow reversal (f) away from the transducer (wave-
carotid artery (CCA) has maintained its normal form below baseline), away from the globe, and toward the
low-resistance (d) pattern as a collateral pathway has brain. (g) Normal waveform and flow direction in the left
formed to the brain via the ipsilateral ophthalmic artery. ophthalmic artery for comparison (OA ophthalmic artery)

had been successfully treated demonstrating com-


plete occlusion from those with significant resid- Ultrasound Biosafety
ual flow [32].
In patients with patent foramen ovale (PFO) Although generally considered a safe imaging
and right-to-left intracardiac shunting, TCDI has modality, diagnostic ultrasound does carry poten-
shown high sensitivity and specificity for tial biosafety concerns. As such, diagnostic ultra-
establishing the diagnosis. Patent foramen ovale, sound imaging should be performed only by
a remnant of the fetal circulation, is a flap-like personnel who have an understanding of the
opening in the atrial septum primum and potential biohazards related to ultrasound imaging
secundum that persists into adulthood. It is esti- and who are fully trained in its safe and
mated to be present in 20–25 % of the general proper use.
population and can cause paradoxical embolic Ultrasound waves are mechanical compression
events resulting in stroke, migraine or migraine- waves through a given medium. The waves prop-
like symptoms, systemic embolism, and neuro- agate in a longitudinal fashion with alternating
logic decompression sickness [33, 34]. The pres- areas of compression and expansion. The term
ence of a PFO and right-to-left shunt is usually “ultrasonic” refers to sound waves above the audi-
diagnosed by transesophageal echo (TEE) ble range and generally is considered to be sound
[35]. TCDI offers a less-invasive option for diag- waves above a frequency of 20,000 Hz (0.02
nosing PFO by intravenously injecting 1 cm3 of MHz). Medical ultrasound utilizes frequencies of
microbubbles and then evaluating for the presence sound generally in the range of 1–15 MHz (1–15
of bubbles in the MCA by TCDI via a  106 Hz).
transtemporal window indicating a right-to-left Ultrasonic sound waves and the resultant
shunt is present. The technique is showing prom- imaging results in the deposition of energy into
ise as being as accurate as TEE for the the body that can potentially cause damage to the
diagnosis [36]. tissues secondary to thermal and mechanical
64 J.D. Kirsch

(nonthermal) mechanisms. Attenuation of the heat from the bone to the soft tissues. Exposure to
ultrasonic sound wave as it passes through the the eye by ultrasound should also be kept to a
soft tissues or bone results in loss of energy due minimum due to low perfusion in the eye, espe-
to absorption and scatter. Absorption of the sound cially the lens, which results in decreased capabil-
wave results in the wave’s energy being converted ity for heat dissipation [41].
to heat. Spectral Doppler has a greater heating Two indices have been devised, the thermal
potential than other modes of ultrasound (gray- index (TI) and the mechanical index (MI) which,
scale and color Doppler flow imaging). Since through an on-screen display, give an indication to
sound waves are mechanical energy being com- the sonographer of the potential for ultrasound-
posed of alternating areas of compression and induced bio-effects.
expansion, nonthermal injury can also result Three types of thermal indices have been
from direct interaction of the sound wave with devised. The thermal index for the soft tissue
the soft tissue. Rarefaction from the passing ultra- (TIS) is utilized when the ultrasound beam only
sound wave can result in cavitation of the tissue. insonates the soft tissue; the TIB (thermal index
The presence of contrast agents containing for the bone) is used when the ultrasound beam
microbubbles can increase the probability of cav- insonates the bone at or near its focal zone; and the
itation and nonthermal effects [37, 38]. TIC (thermal index for cranial bone) is used for
To limit the potential for biosafety effects, the transcranial Doppler imaging when the probes are
key principle in the safe use of ultrasound includes adjacent to the bone (the skull) during scanning.
the ALARA (as low as reasonably achievable) The TI is an indicator of the relative potential
principle. The principle, as applied to medical for tissue temperature rise and biological effect. It
diagnostic ultrasound exams, consists of using is defined as the ratio of the emitted acoustic
the lowest output power and the shortest scan power to the power required to raise the tempera-
time reasonably possible to obtain the necessary ture of the tissue by 1  C [41]. The TI is intended
diagnostic information to formulate an accurate to give an approximate guide to the likely maxi-
diagnosis. It is acknowledged that it may be nec- mum temperature rise that might be incurred after
essary to use longer scan times or higher outputs if a long exposure to ultrasound. A greater TI repre-
that is what is required to obtain accurate diagnos- sents a higher heating potential and a higher risk
tic information [39]. for tissue injury. The MI is a rough guide as to the
As noted above, insonation of the soft tissue likelihood of cavitation occurring. In general, the
with ultrasound can result in a rise in temperature risk of cavitation increases with an increasing
of the tissue due to energy deposition causing MI. Various recommendations are present in the
potential thermal injury. The extent of damage literature for suggested thresholds for TI and MI
depends on the degree of temperature rise, the depending on the age of the patient, the body part
duration of exposure to the elevated temperature, being scanned, and the duration of exposure to the
the type of tissue exposed, its cellular proliferation ultrasound beam [39–41].
rate, and its potential for regeneration. Tempera-
tures of 39–43  C have been shown to cause
detrimental effects in vitro. Temperatures above Summary
44  C can result in denaturation of proteins
[40]. Self-heating of the ultrasound transducers Transcranial Doppler ultrasound is an extremely
(probes) can occur but is more an issue with useful imaging modality for evaluating the intra-
endo-cavitary transducers than for transducers cerebral arterial system. It has found its greatest
placed on the skin where heat dissipation is utility in evaluating and monitoring patients in the
greater. The presence of the bone (e.g., skull) in neurosurgical intensive care unit for vasospasm
the ultrasound beam increases the likelihood of following subarachnoid hemorrhage due to aneu-
temperature rise due to direct absorption of the rysmal rupture. The technology has also found
beam by the bone and subsequent conduction of clinical utility in evaluating and screening
4 Essentials of Transcranial Doppler Ultrasound 65

pediatric patients with sickle cell disease who may Council, American Heart Association. Stroke
be at risk of stroke. In conjunction with clinical 40(3):994–1025
10. Rasulo FA, De Peri E, Lavinio A (2008) Transcranial
findings, electroencephalography, and nuclear Doppler ultrasound in intensive care. Eur J
medicine imaging, TCDI has also been shown to Anaesthesiol Suppl 42:167–173
be a useful adjunct in determining brain death. 11. Kolias AG, Sen J, Belli A (2009) Pathogenesis of
Other possible uses for the imaging modality cerebral vasospasm following aneurysmal subarach-
noid hemorrhage: putative mechanisms and novel
include detection of stenosis, arteriovenous approaches. J Neurosci Res 87:1–11
malformations, determination of collateral path- 12. Wozniak MA, Sloan MA, Rothman MI et al (1996)
ways with occlusion of the extracranial internal Detection of vasospasm by transcranial
carotid artery, and right-to-left intracardiac shunts Doppler sonography: the challenges of the anterior
and posterior cerebral arteries. J Neuroimaging
due to patent foramen ovale. 6(2):87–93
Biosafety concerns are of paramount impor- 13. Torbey MT, Till-Karsten H, Bhardwaj A et al (2001)
tance. Although ultrasound is considered a very Effect of age on cerebral blood flow velocity and inci-
safe imaging modality, the practicing sonographer dence of vasospasm after aneurysmal subarachnoid
hemorrhage. Stroke 32:2005–2011
should be aware and cognizant of the potential 14. Lindegaard KF, Nornes H, Bakke SJ et al (1989) Cere-
injuries that could result to soft tissue and bone bral vasospasm diagnosis by means of angiography
from potential thermal and mechanical injury. and blood velocity measurements. Acta Neurochir
100:12–24
15. White H, Venkatesh B (2006) Intensive applications of
transcranial Doppler in the ICU: a review. Intensive
References Care Med 32(7):981–994
16. Sviri GE, Britz GW, Douville CM et al (2006)
1. Kirsch JD, Mathur M, Johnson MH et al (2013) Transcranial Doppler grading criteria for basilar artery
Advances in transcranial Doppler US: imaging ahead. vasospasm. Neurosurgery 59(2):360–366
Radiographics 33:E1–E14 17. Maakaon JE, Taher AT, Besa EC (2014) Sickle cell
2. Aaslid R, Markwalder TM, Nornes H (1982) Noninva- anemia. Medscape reference: emedicine.medscape.
sive transcranial Doppler ultrasound recording of flow com/article/205926-overview
velocity in basal cerebral arteries. J Neurosurg 18. Frempong KO (1991) Stroke in sickle cell disease:
57(6):769–774 demographic, clinical and therapeutic considerations.
3. Lupetin AR, Davis DA, Beckman I et al (1995) Semin Hematol 28:213–219
Transcranial Doppler sonography. Part 1. Principles, 19. Adams RJ, MCvie V, Nichols FT et al (1992) The use
technique, and normal appearances. Radiographics of transcranial ultrasonography to predict stroke in
15:179–191 sickle cell disease. NEJM 326:605–610
4. Spencer MP, Reid JM (1979) Quantitation of carotid 20. Adams RJ, McKie VC, Carl EM et al (1997) Long
stenosis with continuous-wave (C-W) Doppler ultra- term stroke risk in children with sickle cell disease
sound. Stroke 10(3):326–330 screened with transcranial Doppler. Ann Neurol
5. Marshall SA, Nyquist P, Ziai WC (2010) The role of 42:699–704
transcranial Doppler ultrasonography in the diagnosis 21. Adams RJ, McKie VC, Hsu L et al (1998) Prevention
and management of vasospasm after aneurysmal sub- of a first stroke by transfusions in children with sickle
arachnoid hemorrhage. Neurosurg Clin N Am cell anemia and abnormal results on transcranial Dopp-
21(2):291–303 ler ultrasonography. N Engl J Med 339:5–11
6. Keyrouz SG, Diringer MN (2007) Clinical review: 22. Wijdicks EFM (2001) The diagnosis of brain death.
prevention and therapy of vasospasm in subarachnoid N Engl J Med 344(16):1215–1221
hemorrhage. Crit Care 11(4):220 23. Ropper AH, Kehne SM, Wechsler L (1987)
7. Condette-Auliac S, Bracard S, Anxionnat E et al (2001) Transcranial Doppler in brain death. Neurology
Vasospasm after subarachnoid hemorrhage: interest in 37:1733–1735
diffusion-weighted MR imaging. Stroke 24. Petty GW, Mohr JP, Pedley TA et al (1990) The role of
32:1818–1824 transcranial Doppler in confirming brain death: sensi-
8. Awasthi D (1997) Cerebral Vasospasm: current think- tivity, specificity, and suggestions for performance and
ing and future trends. www.medschool.lsuhsc.edu/neu interpretation. Neurology 40:300–303
rosurgery/nervecenter/spasm.html 25. Roubec M, Kuliha M, Jonszta T et al (2011) Detection
9. Bederson JB, Connolly ES Jr, Batjer HH et al (2009) of intracranial arterial stenosis using transcranial color-
Guidelines for the management of aneurysmal sub- coded duplex sonography, computed tomographic
arachnoid hemorrhage: a statement for healthcare pro- angiography, and digital subtraction angiography.
fessionals from a special writing group of the Stroke J Ultrasound Med 30:1069–1075
66 J.D. Kirsch

26. Feldman E, Wilterdink JL, Kosinski A et al (2007) The 34. Schwerzman M, Nedeltchev K et al (2005) Prevalence
stroke outcomes and neuroimaging on intracranial in size of directly detected patent foramen ovale in
atherosclerosis trial. Neurology 68:2099–2116 migraine with aura. Neurology 65:1415–1418
27. Hansberg T, Wong KS, Droste DW et al (2002) Effects 35. Schneider B, Zienkiewicz T, Jansen V et al (1996)
of the ultrasound contrast-enhancing agent Levovist on Diagnosis of patent foramen ovale by transesophageal
the detection of intracranial arteries and stenoses in echocardiography and correlation with autopsy find-
Chinese by transcranial Doppler ultrasound. ings. Am J Cardiol 77:1202–1209
Cerebrovasc Dis 14(2):105–108 36. Jauss M, Kaps M, Keberle M et al (1994) A compari-
28. Droste DW, Jurgens R, Weber S et al (2000) Benefit of son of transesophageal echocardiography and
echocontrast-enhanced transcranial color-coded transcranial Doppler sonography with contrast medium
duplex ultrasound in the assessment of intracranial for detection of patent foramen ovale. Stroke
collateral pathways. Stroke 31(4):920–923 25:1265–1267
29. Becker G, Greiner K, Kaune B et al (1991) Diagnosis 37. Miller DL, Averkiou MA, Brayman AA et al (2008)
and monitoring of subarachnoid hemorrhage by Bioeffects considerations for diagnostic ultrasound
transcranial color-coded real-time sonography. Neuro- contrast agents. J Ultrasound Med 27(special
surgery 28:814–820 issue):611–632
30. Klotzsch Z, Nahser HC, Fischer B et al (1996) Visual- 38. Bioeffects Committee of the American Institute of
ization of intracranial aneurysms by transcranial Ultrasound in Medicine (2008) American Institute of
duplex sonography. Neuroradiology 38:555–559 Ultrasound in Medicine consensus report on potential
31. Turner CL, Kirkpatrick PJ (2000) Detection of intra- bioeffects of diagnostic ultrasound. J Ultrasound Med
cranial aneurysms with unenhanced and echo contrast 27:503–515
enhanced transcranial power Doppler. J Neurol 39. The British Medical Ultrasound Society (2009) Guide-
Neurosurg Psychiatry 68:489–495 lines for the safe use of diagnostic medical ultrasound
32. Turner CL, Higgins JN, Kirkpatrick PJ (2003) Assess- equipment
ment of transcranial color-coded duplex sonography 40. O’Brien WD, Deng CX, Harris GR et al (2008) The
for the surveillance of intracranial aneurysms treated risk of exposure to diagnostic ultrasound in postnatal
with Guglielmi detachable coils. Neurosurgery subjects: thermal effects. J Ultrasound Med
53(4):866–871 27:517–535
33. Hagen PT, Scholz DG, Edwards WD (1984) Incidence 41. Nelson TR, Fowlkes B, Abramowicz JS et al (2009)
and size of patent foramen ovale during the first Ultrasound biosafety considerations for the practicing
10 decades of life: an autopsy study of 965 normal sonographer and sonologist. J Ultrasound Med
hearts. Mayo Clin Proc 59:17–20 28:139–150
DSA Imaging Protocol and Technique
5
Eytan Raz

Contents Abstract
Preprocedural Assessment . . . . . . . . . . . . . . . . . . . . . . . . . . 68 Currently, digital subtraction cervico-cerebral
angiography (DSA) is seldom the first-line
Patient Preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
study to evaluate the vasculature of the neck
Tray . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68 or of the head and is usually performed after
Procedure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68 duplex ultrasound, CT angiography, or MR
angiography, in cases where additional details
Vascular Access . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
or assessment is needed. Many times, DSA is
Catheter and Wire . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69 obtained in the same setting just before the
Cervico-Cerebral Angiography . . . . . . . . . . . . . . . . . . . . . 70 interventional endovascular treatment.
Projections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
Special Situations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
Keywords
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83 Cerebral angiogram • DSA • catheter • wire •
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84 vascular access

Obviously, this approach is justified when the


same information can be obtained with a nonin-
vasive approach, but it is the belief and the expe-
rience of the author that cervico-cerebral
angiography may add many details and informa-
tion compared to other techniques and has to be
kept in the armamentarium to use in all cases in
which vascular questions remains open after non-
invasive studies are performed. Multiple such
examples will be presented in the current chapter.
Another introductory point that needs to be
pointed out is that DSA is an operator-dependent
technique, and as such, a wide spectrum of quality
of exam and spectrum of complications incidence
E. Raz (*) will result: in good hands, a DSA can have as low
Neurointerventional Radiology Section, Department of
as 0.03 % incidence of stroke.
Radiology, NYU Langone Medical Center, New York,
NY, USA
e-mail: eytan.raz@gmail.com; eytan.raz@nyumc.org

# Springer Science+Business Media New York 2016 67


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_50
68 E. Raz

Preprocedural Assessment order to take the routine morning medications.


Once the patient arrives in the department, a
Visiting the patient before the angiogram is man- 20–22 G IV is placed by the nursing team,
datory. This evaluation should focus on the clini- connected to an intravenous line that will serve
cal history, previous surgical history, history of as a means to administer fluids and medications.
medications, and allergies. Many important This peripheral IV line is also a lifeline, in the
changes in the procedure are usually tailored event of an emergency. After the tray is set up, the
based on all the findings of the preprocedural patient is brought to the angiography suite, appro-
assessment. priately positioned on the table, and connected to
For elective angiograms, a basic thorough continuous EKG, oxygen saturation monitoring,
evaluation, other than medical clearance, includes and intermittent blood pressure monitoring. Pubic
blood work (CBC with differential, PT, PTT, hairs close to the groin are removed, after which
BMP, and, for female patients, hCG), urinalysis, the groins can be prepped and the patient draped.
chest x-ray, and EKG. The reason to obtain these The patient’s head is held with a tape and aligned
studies is to evaluate the general medical risk, to carefully along the long axis of the detector.
evaluate for renal insufficiency or renal failure
(worsened by use of iodine contrast), and to eval-
uate for ongoing infections and for bleeding diath-
esis. If the patient is pregnant – and the procedure Tray
can be postponed – that is usually the best option.
The authors perform angiograms on The basic tray should have the following items:
anticoagulated patients knowing that a micro-
puncture kit and a groin closure device will prob- • Femoral drape
ably be used. • Skin prep tray
During the preprocedural assessment, the • Needle box
neurointerventionalist will educate the patient • Sterile labels
about the exam. Focusing on the sensations that • Telfa
the patient will experience during the procedure • 18 G 22 G–25 G needles
will ultimately improve the collaboration and, • Disposable scalpel no. 11
consequently, improve the quality of the study • Five 12 cc syringes
and decrease the rate of complications. The most • Light handle cover
important points to discuss with the patient are: • 3-way and 1-way stopcocks
the sensations in the groin when the local anes- • Torque device
thetic is injected, the warming sensation when the • Small J wire 0.038
contrast is injected, and the possible dizziness and • Terumo 0.038/150 cm
appearance of flashes behind the eyes. Likewise, • Heparin 4,000 units/1 L bag
also exemplifying the usual commands that are • Lidocaine 1 %
given prior to the injection is needed. A written • 4 F or 5 F micropuncture kit
informed consent needs to be signed by the patient • 4 F or 5 F vertebral catheter
before the procedure.

Patient Preparation Procedure

In elective angiograms, the autors request the Before starting the procedure, a time-out should
patient to be NPO since the midnight before. be conducted before the incision. The time-out
The only exception is usually a sip of water in should involve the entire operative team, use
5 DSA Imaging Protocol and Technique 69

active communication, be briefly documented, continuous flushing technique is preferable; both


and use a checklist which should include: (1) cor- pros and cons can be found in other excellent
rect patient identity using at least name and date of reviews. Double flushing consists of aspiration
birth, (2) allergies, pregnancy, or any other patient of the contents of the catheter with one 10 ml
specific issue, and (3) agreement on the procedure syringe followed by partial aspiration and irriga-
to be done. tion with a second syringe, while continuous
flushing technique consists in connecting the cath-
eter hub with a Y-connector plus three-way stop-
cock connected to a heparinized pressurized
Vascular Access saline bag. On average, a catheter should be
flushed every 90–120 s and after each wire place-
A successful angiography begins with the groin ment. The tip of the syringe should be oriented
puncture. Before puncturing, localization of the inferiorly so that the air will rise upward toward
femoral head with x-ray is suggested, especially the plunger and away from the catheter. As a
for obese patients, in which the fat folds may syringe is connected to the catheter or stopcock,
masquerade the anatomic landmarks. The right it is best to allow a meniscus of blood to form by
common femoral artery, which is in the vast allowing slight backflow into the hub of the cath-
majority of cases the aimed vessel for the access, eter: this meniscus is then brought together
is in fact located in the medial upper quadrant of against the syringe meniscus in a meniscus-to-
the femoral head. Local anesthesia is performed meniscus technique which reduces the likelihood
using 1 % lidocaine using two different needles: of air being trapped between the tip of the syringe
first superficial injection of 2 cc using a 25 G and the hub. After the syringe has been removed
needle, wait 30 s and then injection of approxi- from the end of the catheter, the hub should be
mately 8 cc deep to the artery with a 22 G needle, flushed and the best way to do it is using a
after careful evaluation of no blood return to avoid so-called Jabbie in angio suite jargon, which
injection in the bloodstream. Because of its pH, as essentially is a syringe with a dilator attached to
the lidocaine is being injected, it can be painful. the tip. The Jabbie fits easily into the hub and
Prior to puncturing, a small skin incision (few allows a jet of flush to be injected for cleaning
millimeters long) is made using a no. 11 scalpel the hub.
parallel to the skin fold and dissected using a
hemostat in order to facilitate the following pas-
sage of needle and catheter and to prevent the
subcutaneous forming of hematoma, creating a Catheter and Wire
tract with the surface. For discussion of the vari-
ous techniques to obtain access in the femoral The kind of catheter chosen is a personal decision.
artery, please report to excellent discussions else- In the authors’ experience, they start each angio-
where such as in Harrigan and Deveikis book. gram with a vertebral catheter leaving the option
Once the access is obtained, two main options to exchange for another shape in case of difficult
exist: pushing the catheter over the wire without a access. The vertebral catheter is a simple curve
sheath in place, or placing a sheath in order to catheter, meaning that it has only a primary curve
facilitate the possible catheter exchange. Both and does not need to be formed. The catheter is
pros and cons can be detailed for either technique. advanced over the wire in the ascending aorta, and
The main advantage of not placing the sheath is the wire is then pulled out. In a standard angio-
that a smaller hole is present in the artery. gram, the first vessel to be catheterized is the right
The catheter is then advanced under fluoro- common carotid artery. In order to select this
scopic guidance in the descending aorta. Another vessel, once it is in the ascending aorta, the cath-
option to face at this point is if a double flushing or eter needs to be pulled and rotated
70 E. Raz

counterclockwise. Once in the ostium of the ves- Table 1 Flow rate for different vessels
sel, additional selective catheterization can be Injector
obtained getting a new roadmap, reinserting the settings
wire and readvancing the catheter over the wire. Vessel injected ml/s ml
Every time the wire is removed and the catheter is Aortic arch 20 25
in position for injection, a flow check injecting Common carotid artery (CCA) 8 12
1–2 cc of contrast under fluoroscopy should be Subclavian artery 6 15
performed in order to confirm the appropriate Internal carotid artery (ICA) 6 8
External carotid artery (ECA) 3 6
catheter positioning and exclude iatrogenic dis-
Vertebral artery 6 8
section during the catheter advancement.
3D angiogram in internal carotid artery 3 20
A bifurcation run in both frontal and lateral
projections may be obtained if necessary; other-
wise, a roadmap to access the ICA may be
enough. Advancing the catheter in the ICA with If the automatic injection is used, the following
attention to have the vertebral catheter curve par- are the volumes and rates used – keep in mind
allel to the bloodstream and not against the vessel that, in jargon, the flow rate followed by the total
wall is important. This is usually obtained by volume is indicated in the angio suite (e.g., 5 by
rotating the catheter slightly counterclockwise in 7 means 7 cc of contrast to be injected with a rate
the right ICA and clockwise in the left ICA. of 5 cc/s) (Table 1).
If difficulties are encountered to access a cer- Another feature to keep in mind is the rise: a
vical vessels, other techniques can be considered, “rate rise” in cerebral angiography refers to a
such as the breath-holding technique or exchang- progressive acceleration of contrast over the first
ing the catheter for a double curved one such as second of the injection. The linear rate rise feature
the Simmons – please consult other excellent is especially useful when a catheter is near the
resources to good use of these catheters and how origin of a vessel and a sudden injection of con-
to form and unform. Keep in mind that the more trast may result in recoil of the catheter proximal
complex the vasculature and the more catheters outside of the desired vessel. The PSI setting of
are used, the higher the incidence of the injector refers to the maximum pressure
complications. (pounds per square inch) that the injector will
In some cases, an aortogram is needed, for generate while injecting the contrast.
example, in case an arch study was not obtained
with MRA or CTA, in case of possible vasculitis
or in patients with trauma to exclude an aortic tear.
If the aortogram is needed, a groin sheath has to be Cervico-Cerebral Angiography
placed. The catheter used for aortogram is a pigtail
catheter which is placed in the proximal ascending Biplane angiography is nowadays the standard of
aorta, proximal to the innominate artery. care for cervico-cerebral angiograms and allows
The contrast is diluted in a 2:1 solution with for two planes to be acquired simultaneously with
heparinized saline. This ratio allows excellent a single contrast injection, limiting the total
opacification of the bloodstream with a relatively amount of time and contrast needed to complete
limited contrast load. The injection of contrast the study.
(iopromide, which is a nonionic agent with
300 mg of iodine/ml) is different to obtain
roadmap or to obtain angiographic images. For Projections
roadmaps smooth continuous injection is
performed, while for angiography, fast, strong Standard projections are illustrated in the figures;
injection with appropriate handling is needed. the x-ray detector has a limited space, and as such,
5 DSA Imaging Protocol and Technique 71

the space needs to be optimized. Standard Most cerebral angiographic runs are obtained
arch projection is a left anterior oblique that with a rate of two frames per second (FPS). A
opens the view of the aortic arch. To better variable frame rate can also be obtained limiting
visualize the origin of the vertebral artery, a the rate for venous phase. Usually, an imaging
Towne’s view is necessary, given the origin of sequence lasts approximately 10–12 s in order to
the artery in the superoposterior aspect of the visualize the arterial, capillary, and venous
subclavian artery. The Towne’s view is obtained phases.
angulating the detector 30–40 cranially aligning Modern angiography makes use of three-
the petrous ridges below the superior rim of the dimensional images in various occasions. In
orbits. order to obtain the source 3D images, only a
Standard neck arteries views are the straight single plane is needed, which is rotated around
frontal and lateral projections (Fig. 1). If a single the head of the patient before and during the
plane is used, an ipsilateral oblique frontal view is injection of contrast in a CT-like manner
preferable. (Fig. 6). The volumetric images thus obtained
Before obtaining the cranial images, careful can be postprocessed in different ways and
evaluation of the alignment of the head along the reconstructed in the preferred views (Fig. 7). An
long axis of the detector should be checked. In the obvious advantage of the 3D images is that they
standard frontal (PA) view of the internal carotid can further guide the best 2D projections useful
injection, the petrous ridges are aligned with the that can be selected based on the 3D reconstruc-
lower third of the orbits (Figs. 2a–e and 3a–f). In tion (Fig. 8).
the lateral projection, careful evaluation of the After the angiogram, careful evaluation of the
alignment of the orbital roof and of the external images obtained is done before pulling the cathe-
auditory canals is needed (Figs. 2f–j and 3g–j). ter out of the body. This suggestion has two
While for a CCA injection, the outer skull has to purposes: first, to do a thorough evaluation in
be included on the field of view, for an ICA case additional images are needed and, second,
injection the positioning of the detector has to be carefully evaluate that no evidence of complica-
adjusted just to cover the inner skull. For vertebral tions such as dissection or thromboembolism is
injection intracranial views, both a frontal and present. Once these things are ascertained,
Towne’s views are needed, while for the lateral the management of arteriotomy can start,
projection, the C1 vertebra coverage needs to be preferably accomplished with manual compres-
checked (Figs. 4 and 5). Special views are sion, with pressure applied approximately
obtained in particular situations: for example, to 1–2 cm above the skin incision for 15–20 min,
better see the ICA cavernous segment, the com- or longer if oozing is still noted after that time.
municating segment, or the middle cerebral artery In certain situations, a closure device can be
branches, a Haughton view may be obtained; in used for a mechanical closure and multiple
order to position for this view, the x-ray tube devices for this purpose are available in the
needs to be tilted caudally towards the shoulder market.
of the side of interest. To better visualize the ICA After the angiogram, they request that the
bifurcation or the anterior communicating artery patient lies flat on bed for 6 h with the accessed
complex, an ipsilateral transorbital oblique is leg extended and with head of bed at <30 . Vital
recommended. To memorize all the useful pro- signs need to be checked q1h. Nursing
jections for the different vessels can be cumber- team should check the groin puncture as
some; the best way to obtain the best views is to follows: q15min for 1 h, q30min for 2 h, and
create a 3D virtual map of the cerebral vasculature then q1h for 3 h. The patient may restart the
in thier mind and use this virtual map to guide the usual diet after the angiogram with encourage
best projection based on the orientation of the water intake. The IV can be discontinued before
vessel of interest. discharge.
72 E. Raz

Fig. 1 Standard angiographic views for the CCA bifurcation: lateral, subtracted and unsubtracted (a, b) and straight
frontal subtracted and unsubtracted (c, d)
5 DSA Imaging Protocol and Technique 73

Fig. 2 (continued)
74 E. Raz

Fig. 2 Right ICA injection. In the standard frontal view of late venous (e) views. (f–j) Lateral projection in early
the internal carotid injection, petrous ridges are aligned arterial – subtracted (f) and unsubtracted (g) –, capillary
with the lower third of the orbits. (a–e) Frontal projection (h), late capillary/early venous (i), and early venous (j)
of right ICA in early arterial – subtracted (a) and views
unsubtracted (b), – capillary (c), early venous (d), and

Special Situations For evaluation of intracranial aneurysms, a


complete four-vessel angiogram should be
For evaluation of atherosclerotic disease, an performed: in 15 % of patients with SAH, multi-
aortogram is suggested as well as routine neck ple aneurysms are present. If surgery is an option,
views of both common carotid and vertebral an external carotid injection is also performed in
arteries. order to have a map of the vessels for possible
5 DSA Imaging Protocol and Technique 75

Fig. 3 (continued)
76 E. Raz

Fig. 3 Left ICA injection. In the standard frontal view of early venous (e), and late venous (f) views. In the lateral
the internal carotid injection, petrous ridges are aligned projection (g–j) the alignment follows the orbital roof and
with the lower third of the orbits. (a–e) Frontal projection the external auditory canals: early arterial – subtracted (g)
of left ICA in early arterial – subtracted (a) and and unsubtracted (h) – capillary (i), and late venous (j)
unsubtracted (b) – early capillary (c), late capillary (d), views

intracranial arterial bypass (superficial temporal obtain the best view to show the neck aneurysm.
artery, internal maxillary artery, etc.). When an Sometimes, cross-compression may be necessary
aneurysm is identified, the mere diagnosis is not to visualize the anterior communicating artery by
enough: for thorough planning, the anatomy of the compressing the contralateral carotid and
aneurysm has to be defined, especially trying to allowing crossflow through the Acomm. In a
5 DSA Imaging Protocol and Technique 77

Fig. 4 (continued)
78 E. Raz

Fig. 4 Left vertebral artery injection. In the standard capillary (d), and late venous (e) views. In the lateral
frontal view of the internal carotid injection, petrous ridges projection (f–k), the alignment follows the orbital roof
are aligned with the lower third of the orbits. (a–e) Townes and the external auditory canals: early arterial – subtracted
projection of left vertebral artery in early arterial – (f) and unsubtracted (g) – late arterial (h), capillary (i),
subtracted (a) and unsubtracted (b) – late arterial (c), early venous (j), and late venous (k) views
5 DSA Imaging Protocol and Technique 79

Fig. 5 Additional vertebral artery injection views. Additional injection with frontal projection (a, b) and double oblique
(c) views

similar fashion, the carotid artery can be com- understand the subjacent anatomy. Angiograms of
pressed during vertebral artery injection to better other branches are often the most important runs
visualize the Pcomm. in cases of a high-flow AVM because partial
In cases of arteriovenous malformations, high- opacification is visualized with higher details
rate runs are obtained (3–5 FPS), which help to (Fig. 9). Another important point when imaging a
80 E. Raz

Fig. 6 Source images from 3D injection. These are the are similar to tomographic images obtained from a CTA,
source images acquired during injection (in this case, left and can be reconstructed in a similar fashion
ICA injection) that are used to create the 3D views. They
5 DSA Imaging Protocol and Technique 81

Fig. 7 (a–c) The volumetric images obtained can be postprocessed in different ways and reconstructed in the preferred
views using different color algorithms

patient with AVM is that, in order to see the normal also the ascending and deep cervical arteries may
brain venous drainage, a longer imaging sequence is need to be separately imaged according to the
necessary. In AVM ECA injection should be location.
obtained in order to ascertain ECA feeders. What to look for on an angiogram:
For evaluation of dural arteriovenous fistulas, The neuroradiologist should be familiar with
the ECA, some of its branches, more commonly the evaluation of angiographic images. Multiple
the ascending pharyngeal and the occipital, but notions should be kept in mind when looking at an
82 E. Raz

angiogram. Differently from a CTA, the contrast


is injected inside a single vessel and hemody-
namic consequences may be noted such as the
inflow of nonopacified blood from other
vessels (Fig. 10). The angiography is a dynamic
technique and all the phases have to be
evaluated, early arterial, late arterial, capillary,
early venous, and late venous. In order to
better understand the anatomy, or in order to iden-
tify some branches (especially the ECA
branches), the unsubtracted images are very help-
ful (Fig. 11).
In the arterial phase, the vessel contour and size
are evaluated. The arterial anatomy is visualized
with identification of anatomical variants or
nonvisualization of expected vessels; careful eval-
uation of early draining veins is accomplished
Fig. 8 After analysis of 3D images, the best view to
during this phase. On the capillary phase, the
evaluate the neck of the aneurysm is selected and the 2D
acquisition is then performed. Compare with Fig. 7a blush of physiologic or pathologic structures is

Fig. 9 (a, b) In cases of arteriovenous malformations, opacification is visualized with higher details. In this
angiograms of other branches are often the most important example of a right frontal AVM, better details of the
runs in cases of high-flow AVM because partial nidus are obtained during vertebral artery injection
5 DSA Imaging Protocol and Technique 83

identified: in order to better see it, hard Patency of the veins is visualized. Abnormal per-
windowing may be helpful. On the venous sistent arterial opacification in this phase should
phase, dominance of different veins is identified, prompt to think of possible thromboembolism.
which can help to better tailor an intervention.

Conclusion

Even though noninvasive methods to depict the


vessels of the neck and head are nowadays avail-
able, catheter-based angiography is still the gold
standard to visualize the vasculature. This proce-
dure needs to be performed following strict
criteria in order to decrease as much as possible
the incidence rate and increase as much as possi-
ble the diagnostic yield.

Fig. 10 Different from a CTA, the contrast is injected


inside a single vessel and hemodynamic consequences
may be noted such as the inflow of nonopacified blood
from other vessels

Fig. 11 (a, b) External carotid artery injection. In order to better understand the anatomy, or in order to identify some
branches (especially the ECA branches), the unsubtracted images are very helpful
84 E. Raz

References 4. Björkesten G, Halonen V (1965) Incidence of intracra-


nial vascular lesions in patients with subarachnoid
1. Fifi JT, Meyers PM, Lavine SD, Cox V, Silverberg L, hemorrhage investigated by four-vessel angiography.
Mangla S, Pile-Spellman J (2009) Complications of J Neurosurg 23(1):29–32
modern diagnostic cerebral angiography in an academic 5. Lin JP, Kricheff II (1972) Angiographic investigation
medical center. J Vasc Interv Radiol 20(4):442–447 of cerebral aneurysms. Technical aspects. Radiology
2. Thiex R, Norbash AM, Frerichs KU (2010) The safety 105(1):69–76
of dedicated-team catheter-based diagnostic cerebral 6. Harrigan MR, Deveikis JP (2012) Handbook of cerebro-
angiography in the era of advanced noninvasive imag- vascular disease and neurointerventional technique.
ing. AJNR Am J Neuroradiol 31(2):230–234 Springer Science & Business Media. New York, NY,
3. Willinsky RA, Taylor SM, TerBrugge K, Farb RI, USA, 850 p
Tomlinson G, Montanera W (2003) Neurologic compli- 7. Osborn AG (1999) Diagnostic cerebral angiography.
cations of cerebral angiography: prospective analysis of Lippincott Williams & Wilkins, Philadelphia, 462 p
2,899 procedures and review of the literature. Radiology 8. Morris P (2013) Practical neuroangiography. Lippincott
227(2):522–528 Williams & Wilkins, Philadelphia, 528 p
Part II
Carotid and Vertebral Artery: Anatomy,
Diseases, and Treatment
Anatomy of the Neck Arteries
6
Nasim Sheikh-Bahaei, Tomasz Matys, and Jonathan H. Gillard

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88 The anatomy of neck arteries, normal varia-
tions, and anastomoses between different arter-
Aortic Arch . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
ies is discussed in this chapter. All major
Common Carotid Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89 arteries of the neck originate from the aortic
Cervical Internal Carotid Artery . . . . . . . . . . . . . . . . . . . 89 arch via three main vessels: the
brachiocephalic trunk, left common carotid
External Carotid Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
(CCA), and left subclavian arteries. The CCA
External Carotid Artery Anastomoses . . . . . . . . . . . . . 92 courses superiorly in the neck, anteromedial to
Vertebral Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92 the jugular vein and alongside the vagus nerve.
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93 CCA typically divides at the level of C3 or C4
vertebral body into internal and external
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
carotid arteries. The cervical segment of the
internal carotid artery (ICA) usually does not
have branches in the neck, unless there is rem-
nant of carotid-basilar anastomoses such as the
hypoglossal artery or proatlantal artery type I.
The external carotid artery (ECA) is the
smaller branch of the CCA and runs
anteromedial to ICA. It has six branches in
the neck before entering the parotid gland and
divides into two terminal branches: superficial
temporal artery and internal maxillary artery.
The ECA supplies most of the neck structures,
scalp, and meninges. The vertebral artery is
usually the first branch of the subclavian artery,
running superiorly in the transverse foramen of
C6 to C1 with no cervical branches. It then
N. Sheikh-Bahaei (*) • T. Matys • J.H. Gillard courses between the C1 and foramen magnum
Department of Radiology, School of Clinical Medicine, and enters intracranial space. There are exten-
University of Cambridge, Cambridge Biomedical Campus,
sive anastomoses between carotid and verte-
Cambridge, UK
e-mail: n.sh.bahaei@doctors.org.uk; ns548@cam.ac.uk; bral arteries, the knowledge of which is
tomasz.matys@me.com; jhg21@cam.ac.uk

# Springer Science+Business Media New York 2016 87


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_2
88 N. Sheikh-Bahaei et al.

essential to prevent disastrous complications scope of this chapter; in particular, developmental


during interventions. anomalies leading to the formation of right or
double aortic arches will be omitted here. The
Keywords most common anatomical variants, knowledge
Aortic arch • Common carotid artery • Internal of which is of paramount importance for
carotid artery • External carotid artery • Verte- performing safe catheter angiography, deserve a
bral artery • External-internal carotid artery brief mention however.
anastomoses • Embryonic carotid-basilar sys- The most frequent anatomical variants are the
tem anastomoses • Superior thyroid artery • common origin of the brachiocephalic trunk and
Ascending pharyngeal artery • Lingual artery the left common carotid artery seen on average in
• Facial artery • Occipital artery • Posterior 13 % of population and the origin of the left
auricular artery • Superficial temporal artery • common carotid artery from the brachiocephalic
Internal maxillary artery trunk encountered in 9 % of patients [3, 4]. Both
of these variants are more common in blacks
(25 % and 10 %, respectively). Instead of arising
Introduction separately, the left common carotid artery shares
the aortic arch origin with the brachiocephalic
In this chapter, the anatomy of the aortic arch and trunk (Fig. 1b) or arises from the brachiocephalic
neck arteries is described. The aortic arch config- trunk slightly more cranially (1–2.5 cm distal to its
uration and its common variations are reviewed as ostium) (Fig. 1c). As a result, only two major
well as the anatomy of CCA, ECA, cervical ICA, vessels ascend from the aortic arch. The common
and vertebral artery. The normal variations and trunk variant, or sometimes both of the above
anastomoses between different arteries, the variants, has been referred to as “bovine” arch
knowledge of which is crucial for performing implying high frequency of such configuration in
safe catheter angiography, are also discussed. cattle. In fact, the aortic arch in cows more often
The aim of this chapter is to illustrate the applica- gives rise to a single common vessel, and these
tion of anatomical knowledge in clinical practice. branching patterns are not specific on any species.
Thus, “bovine-type” aortic arch should be
avoided as a misnomer and replaced with an ana-
Aortic Arch tomical terminology [3, 4].
The arch origin of the left vertebral artery as a
All major arteries of the neck originate directly or fourth separate arch vessel is seen in 0.5 % of
indirectly from the aortic arch. In the standard patients; in such cases the left vertebral artery
configuration, the aortic arch gives rise to three most commonly arises between the left common
great vessels: the brachiocephalic trunk (also carotid and the left subclavian artery (Fig. 1d) and
termed brachiocephalic or innominate artery), sometimes distal to the left subclavian artery. Rare
the left common carotid artery, and the left sub- variants include the bi-innominate artery (sym-
clavian artery. The brachiocephalic trunk then metrical right and left innominate arteries each
bifurcates at the level of the right sternoclavicular bifurcating into the common carotid and subcla-
joint into the right common carotid and the right vian vessels, Fig. 1e) and bicarotid trunk (bifur-
subclavian arteries (Fig. 1a). This standard cating into the right and left common carotid
branching pattern occurs in approximately 70 % vessels, with separate origins of subclavian arter-
of patients [1, 2]. ies, Fig. 1f). Different combinations in which the
There are many possible anatomical configu- neck vessels arise separately from the aortic arch
rations of the aortic arch and its branches. are also encountered, in extreme case giving rise
Description of all these variations is beyond the to six aortic arch branches (separate right and left
6 Anatomy of the Neck Arteries 89

Fig. 1 Branching patterns of the aortic arch. (a) Standard (e) Bi-innominate artery. (f) Bicarotid trunk. (g) Separate
configuration. (b) Common origin of the brachiocephalic origins of subclavian, common carotid, and vertebral arter-
trunk and the left common carotid artery. (c) Origin of ies. (h) Aberrant right subclavian artery. R/L-SA right/left
the left common carotid artery from the brachiocephalic subclavian artery, R/L-VA right/left vertebral artery, R/L-
trunk. (d) Arch origin of the left vertebral artery. CCA right/left common carotid artery

subclavian, common carotid, and vertebral arter- the vagus nerve. The common carotid artery typ-
ies, Fig. 1g) [1, 2]. ically divides at the level of C3 or C4 vertebral
The most frequent anomaly of the aortic arch body into the external carotid artery (ECA) and
seen in 0.4–2 % of population is the aberrant right the internal carotid artery (ICA); this corresponds
subclavian artery (Fig. 1h) arising distal to the left approximately to the superior border of the thy-
subclavian artery and passing to the right side of roid cartilage. Bifurcation can occur as low as T2
the mediastinum behind the esophagus causing its and as high as C2. Normally CCA does not have
typical posterior indentation and dysphagia any named cervical branches, but occasionally the
lusoria [2]. vertebral artery or proximal ECA branches can
arise from it (Fig. 2) [1].

Common Carotid Artery


Cervical Internal Carotid Artery
The right and left common carotid arteries (CCA)
course superiorly on both sides of the neck lying The cervical segment of the ICA (C1) extends
within the respective carotid space, anteromedial from the carotid bifurcation to the skull base
to the internal jugular veins and accompanied by where the artery enters the carotid canal
90 N. Sheikh-Bahaei et al.

Fig. 2 (a) Volume rendering demonstrating the cervical The vertebral artery (VA) enters the transverse canal at C6
arteries. Common carotid artery (CCA) bifurcates into the level. Note a dominant left vertebral artery with hypoplas-
internal (ICA) and external (ECA) carotid artery at the tic right vertebral artery. (b) Posterolateral view. Distal to
level of C4 vertebral body. Note the variant origin of the the CCA bifurcation, the ICA lies posterior and lateral to
left common carotid artery from the brachiocephalic trunk. the ECA

transitioning into the petrous (C2) segment. At the basilar artery) and persistent proatlantal artery
bifurcation, the ICA most frequently arises type I (proatlantal intersegmental artery, joining
posterolaterally to ECA; its medial origin from the vertebrobasilar system after passing through
CCA is seen in 10–15 % of the population. The the foramen magnum) [1].
most proximal part of the ICA shows a focal
dilatation termed the carotid bulb or carotid
sinus. Reversal of flow and turbulent flow pattern External Carotid Artery
in this region predispose it to atherosclerotic
plaque formation. Ascending cranially, the ICA The ECA is the second branch of the CCA, which
passes medial to the ECA and lies medial to the unlike the ICA has multiple branches in the neck
internal jugular vein and anterior to the transverse and supplies most of the neck structures. The ECA
processes of the upper cervical vertebrae. Usually extends from the carotid bifurcation, usually at the
there are no major named branches of the C1 level of the upper border of the thyroid cartilage, and
segment of the ICA. Important exceptions include curves anteriorly before inclining backward behind
remnants of the carotid-basilar system anastomo- the neck of the mandible. The ECA is anteromedial
ses such as persistent hypoglossal artery (arising to the ICA [1]. It gives six main branches in the neck
from the cervical ICA between C1 and C3 and and, after entering the parotid gland, divides into its
passing through the hypoglossal canal to join the two terminal branches. In comparison to ICA, it has
6 Anatomy of the Neck Arteries 91

smaller caliber and rapidly tapers along its course as 3. The lingual artery is the second anterior branch
it gives multiple branches. of the ECA. Initially it runs obliquely upward
The following structures cross the ECA anteri- to the hyoid bone. Then it curves downward
orly: the hypoglossal nerve (CN XII), posterior and forward, forming a loop, which is crossed
belly of digastric muscle, stylohyoid muscle and by the hypoglossal nerve [1]. It passes beneath
ligament, and facial nerve (CN VII) within the the digastric and stylohyoid muscles and
parotid gland, while the pharyngeal wall, superior ascends and courses forward underneath the
laryngeal branch of the vagus nerve (CN X), and tongue. It supplies the tongue, oral cavity, and
deep lobe of the parotid gland are posterior to submandibular gland and has four branches:
ECA. The pharyngeal branch of the vagus nerve suprahyoid, dorsal lingual, deep lingual, and
(CN X) and glossopharyngeal nerve (CN IX) are sublingual arteries. The lingual artery may
between the ECA and ICA [5]. have common origin with the facial artery
(10–20 %) [5].
Branches of ECA 4. The facial artery originates above the lingual
1. The superior thyroid artery is the first branch artery. It is sheltered by the ramus of the man-
of ECA, which arises anteriorly and descends dible and courses above the submandibular
to the apex of the thyroid. It supplies the larynx gland, then curves upward over the body of
and superior thyroid and anastomoses with the the mandible, and passes anterosuperiorly
inferior thyroid artery which is a branch of the across the cheek to the angle of the mouth
thyrocervical trunk. The superior thyroid artery and nasolabial fold. It then runs toward the
may arise from CCA bifurcation (20 %) or inner canthus of the eye and terminates as the
have common origin with the lingual artery as angular artery. The facial artery is a very tortu-
thyrolingual trunk (2.5 %) or with both lingual ous vessel and supplies the face, lip, palate, and
and facial arteries as the thyrolinguofacial superficial neck [1, 5]. Its cervical branches are
trunk (2.5 %). Its main branches are the hyoid the ascending palatine artery, tonsillar,
artery, sternocleidomastoid branches, superior submental, and glandular branches. Branches
laryngeal artery, and cricothyroid branch [1, 5]. in the face include inferior and superior labial
2. The ascending pharyngeal artery is the arteries, inferior alar artery, and lateral nasal
smallest branch of the ECA which arises pos- branch [8]. The facial artery has anastomoses
teriorly near the carotid bifurcation. It is a long with the ophthalmic artery, branch of the ICA,
vessel that ascends superiorly between the ICA and other ECA branches [7, 8].
and ECA deeply in the neck [5]. After a short 5. The occipital artery originates from the poste-
common trunk, it divides into two major rior aspect of the ECA opposite to the facial
branches: anteriorly, the pharyngeal trunk, artery, at the level of the posterior belly of the
which is extracranial, and, posteriorly, the digastric muscle [1, 5]. It ascends posterosu-
neuromeningeal trunk, which is intracranial. periorly between the occiput and C1 in the
The other branches are inferior tympanic and occipital groove toward the superior nuchal
musculospinal arteries. The pharyngeal trunk line and then becomes subgaleal and extends
supplies the nasopharynx, oropharynx, and up to the vertex. It supplies the scalp, upper
eustachian tube. The neuromeningeal branches cervical musculature, and posterior fossa
supply the dura, CN IX–XII, and first, second, meninges. Branches of the occipital artery are
and third cervical roots. Inferior tympanic and sternocleidomastoid, auricular, mastoid,
musculospinal branches supply the middle ear descending, and occipital [9]. The occipital
and prevertebral muscles, respectively [6]. The artery is a remnant of the embryologic type I
ascending pharyngeal artery has several impor- and type II proatlantal arteries. It corresponds
tant and potentially dangerous anastomoses to C1 and C2 segmental arteries and, therefore,
with internal carotid, internal maxillary, verte- retains extensive connection to the vertebral
bral, and occipital arteries [6, 7]. artery in adulthood [10]. It is critical to
92 N. Sheikh-Bahaei et al.

understand the type and pattern of anastomoses anterior to the external pterygoid muscle and
between occipital and vertebral arteries to enters the pterygopalatine fossa. It terminates
avoid any disastrous complications during by sending off six branches to deep facial
embolization. structures: the posterior superior alveolar
6. The posterior auricular artery arises from the artery, infraorbital artery, artery of pterygoid
posterior ECA above the occipital artery and canal, pharyngeal artery, greater palatine
opposite to the apex of the styloid process artery, and sphenopalatine artery [12].
[1]. It ascends posteriorly beneath the parotid
and along the styloid process toward the ear. It There are several potential anastomoses
supplies the pinna, scalp, external auditory between the internal maxillary artery and ICA: in
canal, and chorda tympani. Common origin the orbital region, potential collateral route from
of occipital and posterior auricular arteries is both proximal and distal internal maxillary arteries
seen in 12.5 % [5]. to the ophthalmic branch of ICA. In the cavernous-
7. The superficial temporal artery is the smaller petrous region, the major anastomotic route is
of the two terminal branches of the ECA. It between the middle and accessory meningeal arter-
arises in the parotid gland behind the neck of ies and inferolateral trunk of the cavernous ICA [7].
the mandible and runs superiorly between the
deep and superficial lobes of the parotid gland
[1]. The transverse facial artery comes off the External Carotid Artery Anastomoses
superficial temporal artery before it leaves the
parotid gland. Then the superficial temporal There are three main regions of anastomoses
artery passes superficially over the zygomatic between the ECA and ICA and vertebral arteries:
process and divides into two branches: frontal
and parietal. It supplies the scalp and some 1. Anterior or orbital region: ECA anastomoses
parts of the face [11]. through internal maxillary and facial arteries
8. The internal maxillary artery arises within the with ophthalmic branch of ICA.
substance of the parotid gland behind the man- 2. Middle or petrocavernous: ECA connects with
dibular neck. It then runs forward between the petrocavernous branches of the ICA via inter-
ramus of the mandible and sphenomandibular nal maxillary and ascending pharyngeal
ligament and courses along the pterygoid mus- branches.
cle to the pterygopalatine fossa [5]. The inter- 3. Posterior or cervical: anastomoses between the
nal maxillary artery is divided into three major ECA and vertebral artery via occipital or
parts based on its relationship to the external ascending pharyngeal arteries [7]. Persistent
(lateral) pterygoid muscle. The first (mandibu- embryonic connection between the ECA and
lar) section is posterior to the external ptery- vertebrobasilar system is called type II
goid muscle and courses along its lower border proatlantal artery [1, 10].
giving rise to five branches: the deep auricular
artery, anterior tympanic artery, middle menin-
geal artery, accessory meningeal artery, and Vertebral Artery
inferior alveolar artery. The second (pterygoid
or muscular) section is within the pterygoid The vertebral artery usually arises as the first
muscle and runs obliquely forward and upward branch of the subclavian artery, proximal to the
as well as medially through the infratemporal thyrocervical trunk. Less frequent anatomical var-
fossa. Branches arising from this section are iations include the vertebral artery originating
entirely muscular including: anterior and pos- together with the thyrocervical trunk, from the
terior deep temporal branches, pterygoid thyrocervical trunk, from the subclavian artery
branches, masseteric artery, and buccinator distal to the thyrocervical trunk, or from the com-
artery. The third section (pterygopalatine) is mon carotid artery. In its typical course, the
6 Anatomy of the Neck Arteries 93

vertebral artery courses superiorly and medially to The level at which the vertebral artery enters
enter the transverse foramen of C6; this part of the the transverse foramen is variable and dependent
vertebral artery is termed an extraosseous segment on which embryonic intersegmental artery per-
(V1). The foraminal (V2) segment of the vertebral sists to form the vertebral artery during develop-
artery ascends through the transverse foramina of ment. Vertebral arteries often demonstrate
C6 to C1 vertebrae. At the level of the axis, the variation in size with unilateral (usually left
artery makes a sharp superolateral turn to pass sided) dominance [1]. Cervical branches of the
through its obliquely positioned transverse fora- vertebral artery include segmental muscular and
men. In the extraspinal (V3) segment between the spinal branches, often with a single branch to
C1 transverse foramen and foramen magnum, the brachial widening of the cervical cord (which
artery courses medially and posteriorly lying can also arise from the thyrocervical or
superior to the arch of C1. In the last intracranial costocervical trunk or superior thyroid artery). In
segment (V4), the artery pierces the dura and the presence of carotid-basilar anastomoses (per-
courses superomedially over the clivus to unite sistent hypoglossal artery or proatlantal artery
with its contralateral counterpart into the basilar type I from the ICA or proatlantal artery type II
artery (Fig. 3) [1]. from the ECA), the vertebral artery proximal to
anastomosis may be hypoplastic or absent.

Summary

All major neck arteries originate from the aortic


arch. In the standard configuration the aortic arch
gives rise to three great vessels: the
brachiocephalic trunk, the left common carotid
artery, and the left subclavian artery. The most
frequent anatomical variants are the common ori-
gin of the brachiocephalic trunk and the left com-
mon carotid artery (13 %). The common carotid
arteries course superiorly on both sides of the neck
and divide at the level of C3 or C4 vertebral body
into the ECA and ICA. The cervical segment of
ICA (C1) extends from the carotid bifurcation to
the skull base. At the bifurcation, the ICA most
frequently arises posterolaterally to the ECA and
then passes medially. Usually there are no major
named branches of the C1 segment of the ICA.
Important exceptions are persistent hypoglossal
artery and proatlantal artery type I as the remnants
of the carotid-basilar system anastomoses. The
ECA is the smaller branch of the CCA which has
six branches in the neck before entering the
Fig. 3 Volume rendering demonstrating the typical course
of the vertebral artery. The segment proximal to the trans- parotid gland and divides into two terminal
verse foramen of C6 is termed V1; the segment coursing branches: superficial temporal artery and internal
through the transverse canal from C6 to C1 is termed V2; maxillary artery. The ECA supplies most of the
note a sharp superolateral turn at the C2 transverse fora-
neck structures, scalp, and meninges. The verte-
men. Extraspinal V3 segment stretches from the transverse
foramen of C1 to the site at which the artery pierces the bral artery is usually the first branch of the sub-
dura and enters the cranium clavian artery, running superiorly in the transverse
94 N. Sheikh-Bahaei et al.

foramen of C6 to C1 with no cervical branches. It clarification of a common misnomer. AJNR Am J


then courses between the C1 and foramen mag- Neuroradiol 27(7):1541–1542
5. Standring S (2011) Gray’s anatomy, 39th edn. Living-
num and enters intracranial space. stone, Churchill
There are three main regions of anastomoses 6. Hacein-Bey L, Daniels DL, Ulmer JL, Mark LP et al
between the ECA with ICA and vertebral artery: (2002) The ascending pharyngeal artery: branches,
(1) anterior or orbital region, (2) middle or anastomoses, and clinical significance. Am J
Neuroradiol 23:1246–1256
petrocavernous, and (3) posterior or cervical. Per- 7. Geibprasert S, Pongpech S, Armstrong D, Krings T
sistent embryonic connection between the ECA (2009) Dangerous extracranial–intracranial anastomo-
and vertebrobasilar system is called type II ses and supply to the cranial nerves: vessels the
proatlantal artery. It is very crucial to identify neurointerventionalist needs to know. Am J
Neuroradiol 30:1459–1468
these anastomoses or embryological remnants to 8. Lohn JWG, Penn JW, Norton J, Butler PEM (2011)
prevent disastrous complications during The course and variation of the facial artery and vein,
interventions. implications for facial transplantation and facial sur-
gery. Ann Plast Surg 67:184–188
9. Alvernia JE, Fraser K, Lanzino G (2006) The occipital
artery: a microanatomical study. Neurosurgery
References 58(1 Suppl):114–122
10. Chen PR, Siddiqui AH, Chen PR (2011) Intracranial
1. Harnsberger HR, Macdonald AJ (2006) Diagnostic and arterial collateralization: relevance in neuro-
surgical imaging anatomy, 1st edn, Brain, head and endovascular procedures In: Peres JFP (ed)
neck, spine. Amirsys, Salt Lake City Neuroimaging for clinicians- combining research and
2. Lippert H, Pabst R (1985) Arterial variations in man: practice. InTech Europe, Rijeka, p 175–202
classification and frequency. J F Bergmann, Munchen 11. Pinar YA, Govsa F (2006) Anatomy of superficial
3. Katz JC, Chakravarti S, Ko HH, Lytrivi ID, temporal artery and its branches: its importance for
Srivastava S, Lai WW et al (2006) Common origin of surgery. Surg Radiol Anat 28:248–253
the innominate and carotid arteries: prevalence, 12. Kashiwagi N, Nakanishi K, Kozuka T, Sato Y
nomenclature, and surgical implications. J Am Soc et al (2010) Vascular supply with angio-CT for
Echocardiogr 19(12):1446–1448 superselective intra-arterial chemotherapy in advanced
4. Layton KF, Kallmes DF, Cloft HJ, Lindell EP, Cox VS maxillary sinus cancer. Brit J Radiol 83:171–178
(2006) Bovine aortic arch variant in humans:
Imaging of Carotid Atherosclerosis
7
Jie Sun, Niranjan Balu, and Chun Yuan

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96 Growing interest in the in vivo characterization
of plaque pathology has furthered the develop-
Pathology and Implications for Imaging . . . . . . . . . 96
Pathogenesis of Carotid Atherosclerosis . . . . . . . . . . . . 97
ment of vessel wall imaging approaches
Histopathological Characteristics . . . . . . . . . . . . . . . . . . . . 97 beyond traditional techniques of luminal imag-
Plaque Morphology and Clinical Presentation . . . . . . 98 ing. By leveraging histopathological informa-
MR Imaging Techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . 99 tion from carotid endarterectomy specimens,
Field Strength . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99 magnetic resonance imaging has been proven
Coils . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99 capable of providing qualitative and quantita-
Multi-slice Black-Blood MRI . . . . . . . . . . . . . . . . . . . . . . . 100 tive information on a number of morphological
3D Black-Blood MRI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
Multi-Contrast MRI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102 and pathological features of carotid atheroscle-
Lipid-Rich Necrotic Core . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102 rosis, including fibrous cap and necrotic core,
Intraplaque Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103 intraplaque hemorrhage, plaque neovascu-
Calcification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104 larization, and inflammation. These technical
Loose Matrix . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
Neovasculature and Inflammation . . . . . . . . . . . . . . . . . . . 105 advancements present new opportunities to
Measurement of Plaque Morphology expand the understanding of the pathophysiol-
and Composition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106 ogy of ischemic stroke and devise more effi-
Current Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106 cient diagnostic and prognostic tools to address
Imaging-Based Patient Stratification . . . . . . . . . . . . . . . . 106 contemporary clinical problems in the manage-
Carotid Artery Stenting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108 ment of carotid atherosclerosis. From a clinical
Cryptogenic Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
In Vivo Insights on Pathobiological Mechanisms . . . 109
perspective, this article introduces the various
Efficacy Markers for Testing Therapeutic magnetic resonance techniques for carotid ath-
Strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110 erosclerosis imaging. The background and
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112 pathological basis of direct plaque imaging in
carotid arteries are discussed, followed by
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112
available solutions and key technical consider-
ations, as well as promising applications that
have arisen from these techniques.

J. Sun • N. Balu • C. Yuan (*) Keywords


Department of Radiology, University of Washington,
Carotid artery • Atherosclerosis • Magnetic
Seattle, WA, USA
e-mail: sunjie@u.washington.edu; ninja@u.washington. resonance imaging • Plaque rupture • Black-
edu; cyuan@u.washington.edu

# Springer Science+Business Media New York 2016 95


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_34
96 J. Sun et al.

blood • Multi-contrast • Ischemic stroke • morphology and clinical complications has


Carotid endarterectomy • Carotid artery shifted the focus of imaging of carotid atheroscle-
stenting • Cryptogenic stroke rosis from luminal narrowing to plaque instability,
i.e., the risk of acute plaque rupture and thrombo-
sis. Magnetic resonance imaging (MR, MRI) has
Introduction emerged as a promising imaging modality for
in vivo assessment of plaque instability. Over the
Atherosclerosis is a systemic condition that years, many validation studies have leveraged
affects large- and medium-sized arteries, such as carotid endarterectomy specimens to correlate
the aorta and its branches. Carotid atherosclerosis, in vivo imaging findings with histopathological
especially atherosclerotic plaques at the carotid features. Carotid MRI has been shown capable of
bifurcation, is one of the most prevalent manifes- characterizing a number of pathological features
tations of systemic atherosclerosis. It has been and biological activities of atherosclerotic plaques
estimated from pooled population-based data that are implicated in plaque rupture, including
that the prevalence of moderate carotid stenosis fibrous cap and necrotic core, intraplaque hemor-
(>=50 %) is 4.8 % and 2.2 % for men and women rhage, plaque neovascularization, and inflamma-
less than 70 years old, respectively. The numbers tion [2–5]. As such, direct MR imaging of carotid
increase further still in those aged more than atherosclerosis presents a unique opportunity to
70 years to 12.5 % and 6.9 % [1]. develop more effective diagnostic and prognostic
Although the majority of carotid atheroscle- approaches for clinical research and practice.
rotic plaques have a benign clinical course, a
portion of them result in ischemic stroke, a leading
cause of death and disability worldwide. A pri- Pathology and Implications
mary goal of clinical imaging of carotid athero- for Imaging
sclerosis is therefore to identify lesions that will
lead to ischemic stroke despite control of risk Atherosclerotic plaques are small but complex
factors. Atherosclerotic plaques typically become pathological structures with considerable mor-
flow-limiting as they progress. Unfortunately, the phological variations (Fig. 1). For imaging scien-
simple assessment of luminal narrowing has tists and radiologists to develop effective imaging
proven to be of limited value in predicting the protocols and properly interpret image findings, a
risk of ischemic stroke. Expanded knowledge on basic understanding of the pathology of carotid
the relationship between atherosclerotic plaque atherosclerosis is essential.

Fig. 1 Atherosclerosis in the carotid bulb from three (b) The plaque is mainly fibrotic with small areas of lipid
endarterectomy specimens showing morphological var- accumulation (white arrows) and calcification. (c) Pieces
iations. (a) A mature necrotic core with intraplaque hem- of dense calcification comprise the main portion of the
orrhage (asterisk) is the main component of the plaque. plaque (hollow arrow)
The overlying fibrous cap is an inhomogeneous thickness.
7 Imaging of Carotid Atherosclerosis 97

Pathogenesis of Carotid microvasculature present in large- and medium-


Atherosclerosis sized arteries, with the physiological function of
supplying oxygen and nutrients to the outer por-
Atherogenesis is now appreciated as a multifacto- tion of the medial layer). Initially considered as a
rial process in which the arterial wall interacts compensatory mechanism to alleviate arterial wall
with a number of systemic and local factors. hypoxia as wall thickness increases, accumulating
The disturbed flow conditions at carotid bifurca- evidence from recent studies suggests that plaque
tion make the carotid bulb a predilection site of neovascularization has an active role in
extracranial carotid artery for atherosclerosis. atherothrombosis by contributing to the develop-
Nonetheless, more extensive disease, detectable ment, progression, and destabilization of athero-
by improved imaging approaches, is not uncom- sclerotic plaques. Leaky microvessels may
mon, particularly in the common carotid artery. facilitate the entry of inflammatory cells as well
Individual variations in the geometry of carotid as plasma proteins. Furthermore, it is thought that
bulb and the unknown extent of outward episodic ruptures of plaque microvessels are
remodeling make it difficult to discern the true likely the origin of intraplaque hemorrhage, a
plaque burden by angiography or similar common finding in advanced human lesions.
techniques. Improvements in research approaches, for both
The accumulation of extracellular lipids and histopathology and imaging, have recently
cell debris, the migration and proliferation of enabled a better appreciation of the prominent
smooth muscle cells, and extracellular matrix pro- role of intraplaque hemorrhage in atherogenesis
duction are recognized processes in the formation as well as the pathophysiology of clinical mani-
of plaques with well-developed necrotic core festations. However, mechanistic insights about
(s) and an overlying fibrous cap, or fibroatheroma intraplaque hemorrhage are still limited at this
as described by Virmani et al. in coronary athero- moment, as the phenomenon is rarely seen in
sclerosis [6]. Structural changes are paralleled by prevailing animal models.
the recruitment and activation of immune cells
that respond to local insult and modulate the
inflammatory process. A variety of immune cells Histopathological Characteristics
including monocytes, lymphocytes, and, more
recently, granulocytes have been shown to play a To date, much of the understanding of carotid
role in the pathogenesis and prognosis of athero- atherosclerotic plaque morphology is based on
sclerosis. Furthermore, it has been suggested histopathological examination of carotid endarter-
through dedicated experimental and human ectomy specimens, which do not cover the full
autopsy studies that immune cells can be highly spectrum of lesion severity as compared to human
adaptive in response to microenvironmental sig- autopsy. The peripheral sections of advanced
nals. In accord, functionally indistinct monocytes carotid specimens are presumed to give a glimpse
may differentiate into polarized macrophage sub- of early or intermediate lesions, which frequently
populations in the atherogenic milieu, taking on show proteoglycan accumulation, lipid retention,
heterogeneous biological functions that may sta- or early stage necrotic cores. Based on these
bilize or destabilize the plaque [7]. Therefore, the observations, it appears sensible to describe
biological complexity of plaque inflammation has carotid plaque morphology with nomenclature
placed a higher demand on molecular imaging by proposed for classifying coronary atherosclerosis.
emphasizing the activities rather than the mere Yet caution should be exercised when making an
presence of immune cells. Another morphological analogy for a specific feature with regards to its
change that initiates early in the disease process is role in pathogenesis and prognosis, given the dis-
plaque neovascularization, resulting from the pen- tinct differences in vessel diameter and flow con-
etration of the medial layer (i.e., tunica media) by ditions between coronary circulation and
hyperplastic vasa vasorum (i.e., the nutritional extracranial carotid circulation.
98 J. Sun et al.

Few studies have attempted to compare plaque mural thrombus constituted of platelets or fibrin
morphologies between coronary and carotid ath- on plaque surface, which was associated with
erosclerosis, yet notable differences seem to be either plaque rupture (90.1 %) or erosion (9.9 %)
present. The fibrous cap overlying necrotic core(s) in patients with stroke [9]. Furthermore, fresh
is at risk of rupture in both coronary and carotid thrombus was more frequently observed when
lesions. The high mechanical forces in the carotid carotid endarterectomy was performed closer to
circulation may render the fibrous cap of carotid symptom onset. With carotid plaques collected
atherosclerosis more vulnerable to rupture, as it has from participants of the North American Symp-
been reported that ruptured carotid plaques have tomatic Carotid Endarterectomy Trial (NASCET)
much thicker fibrous cap and fewer macrophage and the Asymptomatic Carotid Atherosclerosis
infiltration than ruptured coronary plaques [8]. Study (ACAS) trials (71 symptomatic, 170 asymp-
Thrombotic activity typically ensues, yet the high tomatic), Fisher et al. noted that thrombus was
flow velocity is likely a limiting factor for the associated with plaque ulceration, both of which
propagation of mural thrombus and the healing of were more prevalent in symptomatic patients than
fibrous cap rupture. In line with the hypothesis, in asymptomatic patients [10]. Another study by
ulceration is reported more frequently in carotid Redgrave et al. examined 570 consecutive symp-
atherosclerosis than in coronary atherosclerosis, tomatic carotid plaques and demonstrated a con-
whereas total thrombotic occlusion of carotid ste- tinued post-event decline of plaque vulnerability
nosis is not as common as in coronary atheroscle- features including cap rupture and plaque inflam-
rosis. Fibrous cap erosion and calcified nodules are mation [11]. These studies have provided impor-
the other two types of surface disruption reported in tant insights onto the pathogenesis of ischemic
the coronary artery, which may be accountable for events associated with carotid atherosclerosis. A
over a third of atherothrombotic events in coronary typical plaque responsible for a recent ischemic
atherosclerosis [6]. However, in the carotid artery, stroke can be described as one with active throm-
plaque rupture is predominant, while plaque ero- bosis on the luminal surface, triggered by plaque
sion does not seem to be a major cause for rupture and exposure of thrombogenic materials
atherothrombosis [9]. Although calcified nodules to the flowing blood. Subsequently, acute plaque
are more frequently seen in carotid atherosclerosis rupture and thrombosis may lead to artery-to-
in comparison to coronary atherosclerosis, its role artery embolism or thrombotic occlusion, which
in atherothrombosis in the carotid artery has yet to is in line with clinical observations.
be determined. Mechanisms of plaque rupture are not yet fully
understood. In the study by Redgrave et al. [11],
two morphological features demonstrated inde-
Plaque Morphology and Clinical pendent association with cap rupture – intraplaque
Presentation hemorrhage and inflammatory cell infiltration of
fibrous cap. Other studies based on data from
Morphological and pathological features of coronary atherosclerosis have suggested that
carotid atherosclerosis have been shown to be fibrous cap thickness and necrotic core size are
associated with downstream neurological ische- good discriminators of plaques at risk for rupture
mic events. Despite the longtime focus of clinical from those that are stable [12]. Of note, the degree
imaging on luminal stenosis, chronic stenosis or of stenosis could fit in here as one of the risk
occlusion from carotid atherosclerosis rarely leads factors for plaque rupture, considering the
to ischemic stroke and is therefore not considered increased mechanical forces associated with
the pathological substrate of ischemic stroke. In a local narrowing. Some of these risk factors may
study of 269 carotid plaques obtained en bloc have a more dynamic natural course, such as cap
from surgical endarterectomy (96 ischemic stroke, inflammation. It is plausible that in vivo charac-
91 transient ischemic attack, 82 asymptomatic), terization of major risk factors of cap rupture may
Spagnoli et al. observed a high prevalence of fresh enable accurate estimation of the short-term or
7 Imaging of Carotid Atherosclerosis 99

long-term risk of neurological ischemic events in core, intraplaque hemorrhage, etc. Indeed, a
patients with carotid atherosclerosis. unique advantage of MR plaque imaging is to
employ more than one contrast weightings to
help differentiate various atherosclerotic
MR Imaging Techniques components.
Thus, carotid vessel wall imaging places spe-
In contrast to luminal imaging using bright blood cial technical constraints on field strength, coils,
MR angiography, vessel wall visualization and and pulse sequences, which are required for accu-
the identification of components of atherosclero- rate identification of pathology. Carotid MRI pro-
sis require suppression of signal from intraluminal tocols need to consider all these aspects while
blood. Thus, MRI sequences commonly referred being tailored for each study. This section outlines
to as black-blood MRI are essential for vessel wall the established carotid MRI sequences and emerg-
imaging. Normal extracranial carotids have thin ing trends in carotid atherosclerosis imaging.
pulsating walls and are often situated in deep
tissue planes in patients with a large neck. Carotid
bifurcation, the most frequent site of carotid ath- Field Strength
erosclerosis, has a complex flow pattern with
recirculating flow. Thus, complete flow suppres- Initial development of carotid vessel wall MRI
sion in carotid bifurcation requires special consid- took place on clinical 1.5-T scanners. Moving to
eration of the flow conditions at carotid higher field strengths can increase the available
bifurcation. SNR. Initial studies comparing 1.5 and 3-T carotid
In addition to blood suppression, consider- MRI found an improvement of wall SNR and
ations of spatial resolution, signal-to-noise ratio lumen/wall CNR by 1.5-fold for T1-WI imaging
(SNR), motion suppression, etc., are also impor- and 1.7–1.8-fold for proton-density-weighted/T2-
tant to obtaining diagnostic quality vessel wall WI imaging [13]. With optimization of sequences
images. Atherosclerotic plaque components are for change in field strength, plaque components
small but complicated structures that require can be identified with similar repeatability at 3-T.
high spatial resolution for identification and quan- However, quantification of some components,
tification. Of note, the requirement of high spatial such as calcification and intraplaque hemorrhage,
resolution must be achieved with good SNR may differ due to higher susceptibility effects at
within a clinically acceptable scan time. Another 3-T. Additionally, the specific absorption rate
reason to avoid long scan times is to reduce (SAR) is higher at 3-T and should be accounted
motion artifacts. Due to the unique location of for in protocol optimization. Carotid MRI at
carotid arteries, patient swallowing has been one higher field strengths is challenging due to inho-
of the major causes for degraded image quality of mogeneous excitation, suboptimal fat suppres-
carotid MRI, whereas it will be less of a problem if sion, and higher susceptibility. Despite these
the scanning time is short. disadvantages, carotid imaging is moving into
Contrast-to-noise ratio (CNR) is equally 7-T due to the advantages of higher SNR.
important in order to characterize vessel wall
pathology. Delineation of the lumen requires
good CNR with adequate blood suppression, Coils
while delineation of the outer wall requires good
fat suppression. This allows plaque boundaries to At 1.5 and 3-T, body coil transmission is used for
be clearly identified and disease burden to be homogenous excitation, which is also useful for
accurately estimated. Furthermore, MRI many blood suppression sequences that use
sequences with special contrast mechanisms are nonselective excitation. However, body coil
required for identification of various plaque com- reception does not provide a high enough SNR
ponents such as calcification, lipid-rich necrotic for carotid vessel wall MRI within clinically
100 J. Sun et al.

acceptable scan times. Thus, phased array surface flows through the imaging slice and produces
coils with a small diameter and applied close to a black-blood effect. This method can be used
the neck provide improved SNR and are often for short TE sequences such as T1-WI con-
used for vessel wall MRI. Phased array coils trast but is prone to flow artifacts depending
with four, six, and eight coil elements have been on the thickness of the saturation slab and its
designed for carotid imaging. Compared to a four- distance from the imaging slice. A traveling
element design, an eight-element design was saturation band is used to ensure that the
shown to provide 1.7-fold higher SNR and larger distance to all slices remains the same and a
coverage along the axis of the artery [14]. Some of short delay time is used so that blood signal
these coil designs are available commercially on does not recover before being imaged.
all major scanner vendor platforms. MRI at 7-T Advantages of inflow saturation include low
also requires custom coil design, but these are SAR, wide availability, and selectivity in
currently available for research purposes only. suppressing arteries and/or veins.
(c) Double inversion-recovery (DIR) and variants:
DIR preparation consists of two inversion
Multi-slice Black-Blood MRI pulses followed by a delay before image acqui-
sition. A nonselective inversion pulse is
Traditionally, the carotid vessel wall is scanned followed by a slice-selective re-inversion of
using 2 mm thick slices with higher in-plane res- the imaging slice. Blood with inverted magne-
olution of 0.5–0.6 mm. A stack of 12–16 slices tization proximal and distal to the imaging slice
centered on the carotid bifurcation covering a flows into the imaging slice while simulta-
3 cm region is used in a multi-slice black-blood neously undergoing T1 relaxation. The inver-
MRI protocol. Blood suppression can be achieved sion delay time (TI) is calculated such that
by black-blood preparation of the magnetization blood magnetization is zero during image
prior to gradient or spin-echo acquisition. The acquisition, thereby causing a black-blood
sequences used for black-blood imaging can be effect. Both arterial and venous blood are there-
briefly described as below: fore nulled by DIR. The long TI time allows
sufficient time for blood inflow and ensures
(a) Spin-echo acquisition: Direct spin-echo MRI good blood suppression. However, artifacts
without any magnetization preparation can may still occur due to slow or recirculating
provide black-blood contrast with proper flow since the non-inverted blood within the
choice of echo time (TE). Only spins imaging slice may not have time for outflow
experiencing both the excitation and from the imaging slice. Increasing imaging
refocusing spins will produce signals. If after slice or slab thickness tends to worsen flow
experiencing the 90 pulse blood flows artifacts with DIR. DIR has better performance
through the slice during TE and does not than inflow suppression and is widely available
experience the refocusing pulse, it will pro- on all vendor scanner platforms. Accurate
duce a black-blood effect in the acquired echo. lumen identification in vessel wall imaging
Long TE spin-echo or fast spin-echo requires at least a DIR preparation. The pri-
sequences such as T2-WI images exhibit this mary disadvantage of DIR for carotid imaging
effect. However, slow flow and recirculating is that it is a single slice method. Multi-slice
flow are not suppressed by this method. DIR (MDIR) preparations have been proposed
Increasing slice thickness will also lead to to reduce scan time associated with DIR
more flow artifacts. [15]. The current most efficient method uses a
(b) Inflow saturation: In this method, the signal slab-selective re-inversion rather than a slice-
from a thick slab proximal to the carotid bifur- selective inversion, thus allowing up to eight
cation is completely dephased every repeti- slices to be acquired with good flow suppres-
tion time (TR). The dephased blood then sion within a single TR [16].
7 Imaging of Carotid Atherosclerosis 101

(d) Motion sensitization: All the above methods 3D Black-Blood MRI


perform best with laminar blood flow with
artifacts arising in cases of slow flow, turbu- MRI studies of atherosclerotic plaque have gen-
lent flow, and recirculating flow. Flow in the erally used 2–3 mm thick axial slices, since black-
normal carotid bifurcation can be blood techniques such as inflow suppression or
recirculating or turbulent, causing plaque- DIR rely on inflow or outflow for efficient blood
mimicking artifacts on black-blood MRI. suppression. Compared to the traditional
Use of motion-sensitized-driven equilibrium two-dimensional (2D) sequences, 3D black-
preparation (MSDE) can effectively suppress blood MRI can improve the accuracy and the
artifacts from recirculating or turbulent flow repeatability of plaque measurements due to
[17]. MSDE with two refocusing pulses is less higher spatial resolution in the z direction and
affected by B1 inhomogeneity and eddy cur- less partial volume averaging. However, this
rent effects than MSDE with one refocusing requires blood suppression in a relatively thick
pulse [18]. MSDE is not dependent on inflow slab, and therefore use of DIR can result in
of blood into the imaging slice. Hence, it can increased flow artifacts. Since MSDE flow sup-
be used for non-axial slice acquisition. Addi- pression does not depend on flow, 3D black-blood
tionally, MSDE is also compatible with acqui- MRI can be achieved with high isotropic spatial
sition of thick slabs and is therefore used for resolution with this technique. A 3D MSDE pre-
three-dimensional (3D) black-blood MRI. An pared rapid gradient echo (3D-MERGE) vessel
extension of motion sensitization uses a train wall imaging technique can provide
of delay alternating with nutation for tailored 0.7  0.7  0.7 mm3 isotropic resolution within
excitation (DANTE) subpulses with inter- a 2-min scan time with good blood suppression
spersed gradient dephasers for suppression (Fig. 2) [20]. Since 3D-MERGE datasets are iso-
of blood flow [19]. DANTE with high ampli- tropic, interactive reformatting of multiple image
tude dephasing gradients can also be used for planes can be used for plaque visualization. Spin-
3D MRI. echo-based sequences with variable flip angle

Fig. 2 3D isotropic high-


resolution
(0.7  0.7  0.7 mm3)
carotid artery images
acquired with
3D-MERGE. Two major
advantages offered by 3D
isotropic black-blood vessel
wall imaging are shortened
scan times for clinical utility
and flexible reformation
capability for improved
plaque visualization. Note
that vessel wall boundaries
are clearly visible on all
reformats (Reprinted with
permission from Balu
et al. [20])
102 J. Sun et al.

Table 1 Multi-contrast carotid MRI protocol for 3-T


Pre/post- Proton-
contrast Time-of- density 3D-MERGE
T1-WI flight T2-WI -weighted SNAP sequence sequence
Sequence TSE TFE TSE TSE PSIR TFE
Image mode 2D 3D 2D 2D 3D 3D
Scan plane Axial Axial Axial Axial Coronal Coronal
TR (ms) 800 20 4,800 4,800 13 10
TE (ms) 10 4.6 50 10 4 5
Flip angle ( ) 90 50 90 90 11 6
FOV (cm) 14  14 14  14 14  14 14  14 14  14  3.2
Resolution 0.55  0.55 0.55  0.55 0.55  0.55 0.55  0.55 0.7  0.7  0.7 0.7  0.7  0.7
(mm)
Slice 2 1 2 2 Not applicable Not applicable
thickness (interpolated)
(mm)
Blood QIR/MSDE Venous DIR/MSDE DIR/MSDE Not applicable MSDE
suppression inflow
suppression
Fat Yes No Yes Yes Yes Yes
suppression
3D-MERGE 3-dimensional MSDE prepared rapid gradient echo, DIR double inversion-recovery, FOV field of view,
MSDE motion-sensitized-driven equilibrium preparation, PSIR phase-sensitive inversion-recovery, QIR quadruple
inversion-recovery, SNAP simultaneous non-contrast angiography and intraplaque hemorrhage, TFE turbo field echo,
TSE turbo spin echo

long echo trains have also been suggested for complementary fashion is essential for compre-
imaging plaque burden [21]. Further studies on hensive evaluation of atherosclerotic plaques.
the use of 3D sequences for identification of Table 1 shows a typical carotid MRI protocol
plaque components are required before 3D MRI with traditional 2D multi-contrast MRI in com-
can be used in a similar way to current 2D MRI parison to newer isotropic 3D MRI sequences.
protocols. Single contrast weightings may be used when
only a subset of plaque components is of interest.
Sequences of interest for detecting important
Multi-Contrast MRI plaque components are briefly described. Table 2
shows the appearance of plaque components on
Atherosclerotic plaques may range from simple each of the different contrast weightings.
wall thickening to complicated plaques with mul-
tiple plaque components. By varying pulse
sequence parameters, image contrast may be Lipid-Rich Necrotic Core
manipulated such that specific plaque components
are visible in each contrast weighting. For exam- Lipid accumulation in plaques evolves from lipid
ple, a highly T1-WI fat-suppressed sequence will pools to necrotic cores containing cell debris with
show intraplaque hemorrhage with high signal progression of atherosclerosis. Fat suppression
intensity compared to fibrous tissue or muscle. used for plaque imaging suppresses signal from
Thus, a multi-contrast MRI protocol where multi- triglycerides in subcutaneous fat but does not
ple contrast weightings are used in a suppress signal from necrotic core, where the
7 Imaging of Carotid Atherosclerosis 103

Table 2 Plaque component classification using multi-contrast MRI


Time-of-flight T1-WI Proton-density-weighted T2-WI Post-contrast T1-WI
Lipid-rich necrotic core o o/+ o/+ /o
Intraplaque hemorrhage + + /o /o /+
Calcification
Loose extracellular matrix o /o + + +
Dense fibrous tissue o o o +
Signal intensities are relative to adjacent muscle
+ hyperintense, o isointense, hypointense

signal mainly originates from free cholesterols Intraplaque Hemorrhage


and cholesteryl esters. Necrotic core is isointense
on T1-WI sequences, but can be seen on T2-WI Bleeding into the plaque may come from either
sequences as a hypointense region due to its short the lumen secondary to surface rupture or from the
T2 components. Thus, a non-contrast MRI proto- vasa vasorum originating from the adventitia.
col requires a T2-WI sequence for detection of Hemoglobin in the red blood cells then passes
necrotic core. through several stages of degradation. One of the
Sensitivity and specificity for detection of intermediate stages is the formation of highly
necrotic core can be improved by administration paramagnetic methemoglobin. Methemoglobin is
of gadolinium contrast agent such as gadopentetic visible as hyperintense regions on T1-WI images
acid (Gd-DTPA) [2]. Due to necrotic contents, such as T1-WI fast spin echo or time-of-flight
gadolinium uptake into necrotic core is negligible images. However, the sensitivity and specificity
compared to surrounding fibrous tissue. Post- for detection of intraplaque hemorrhage can be
contrast difference between necrotic core and improved by use of highly T1-WI sequences [23].
fibrous tissue is maximal 5–10 min after contrast Magnetization-prepared rapid gradient echo
injection. Therefore, necrotic core can be readily (MPRAGE) is often used for detection of
identified by comparing pre-contrast and post- intraplaque hemorrhage [3]. The MPRAGE
contrast black-blood T1-WI images. However, sequence employs an inversion recovery preced-
adequate blood-suppression post-contrast is chal- ing a gradient echo acquisition. Fat suppression is
lenging. The T1 of blood decreases sharply after also employed to reduce signals from subcutane-
contrast administration, but the change in blood ous fat. MPRAGE has been shown to have higher
relaxivity is not predictable since it is dependent sensitivity and specificity for intraplaque hemor-
on distribution volume – an unknown patient- rhage detection than T1-WI fast spin echo or time-
dependent factor. Thus, while black-blood tech- of-flight. However, blood suppression in
niques such as DIR provide good blood- MPRAGE can be challenging since TI has to be
suppression pre-contrast, adjustment of TI of optimized simultaneously for both blood suppres-
DIR post-contrast is empirical and does not pro- sion and visualization of intraplaque hemorrhage.
vide adequate blood suppression in all subjects. A A better approach combines MPRAGE acquisi-
quadruple inversion-recovery (QIR) preparation tion with phase-sensitive reconstruction (PSR).
has been introduced for this purpose [22]. QIR While PSR requires the additional acquisition of
consists of two DIR modules in tandem and con- a reference image, it provides additional benefits
sists of two TIs that are adjusted such that blood over blood suppression. PSR is used in the simul-
suppression is achieved for the set repetition time taneous non-contrast angiography and intraplaque
(TR) of the sequence. QIR blood suppression is hemorrhage (SNAP) MRI sequence where the
T1 insensitive, thus enabling the same sequence nonselective inversion pulse of MPRAGE is
parameters to be used for both pre- and post- replaced with a slab-selective inversion [24]. The
contrast T1-WI MRI. TI of SNAP is adjusted such that blood flowing
104 J. Sun et al.

Fig. 3 SNAP imaging and histopathology of a compli- Axial reformats of the bifurcation are shown in (d) with an
cated plaque with intraplaque hemorrhage and ulcera- ulcer (arrows) shown as a dark depression and intraplaque
tion. Two image portions are simultaneously acquired. The hemorrhage as hyperintense signals. Imaging findings are
portion of non-contrast MR angiography (a) shows a high- confirmed on matched histological cross sections
grade stenosis in the proximal external carotid artery (Masson’s trichrome). Note that the good blood suppres-
(arrowhead) and ulcerated plaque in the proximal internal sion in (d) allows clear distinction of intraplaque hemor-
carotid artery (ICA) (arrow). The intraplaque hemorrhage rhage from lumen (Reprinted with permission from Wang
portion (b) can be fused with the angiogram portion (c). et al. [24])

into the imaging plane is inverted only a single calcification in multi-contrast plaque MRI. How-
time compared to the blood in MPRAGE that may ever, two issues need to be noted. First, it is
be inverted multiple times. During image acquisi- common to witness calcifications that are either
tion at TI, flowing blood which has a long T1 has close to luminal surface or even in direct contact
negative longitudinal magnetization, while short T1 with flowing blood in carotid atherosclerosis. In
components such as intraplaque hemorrhage have a such situations, it could be challenging to separate
positive longitudinal magnetization. PSR of SNAP calcification from lumen based on black-blood
takes advantage of this difference in sign of longi- images alone. As such, accurate identification of
tudinal magnetization to provide two images from calcification and therefore overall plaque size
the same acquisition: negative part provides an depend on the combined use of bright-blood and
angiogram, while the positive part provides signal black-blood sequences. Most multi-contrast pro-
for intraplaque hemorrhage. Figure 3 shows an tocols used in previous clinical studies have
ulcer on the SNAP angiogram with corresponding involved at least 3D time-of-flight imaging to
intraplaque hemorrhage in the plaque. Since both overcome the problem. Second, the quantification
MR angiography and intraplaque hemorrhage of calcification should be mainly performed on
imaging are obtained from the same SNAP acqui- spin echo sequences since their size tends to be
sition, they are naturally registered together. overestimated on gradient echo sequences due to
increased susceptibility of calcification. Use of
very short TE sequences called ultrashort echo
Calcification time (UTE) sequences renders calcification bright
and is less affected by susceptibility. UTE
Calcification is hypointense on all contrast sequences require special hardware and spiral or
weightings due to its very short T2 relaxation radial readouts in addition to dual echo acquisition
time. This forms the basis for identification of for the identification of calcifications.
7 Imaging of Carotid Atherosclerosis 105

Loose Matrix as gadolinium contrast is injected. Additionally, a


bright blood technique is preferred to measure the
Loose matrix refers to the category of plaque arterial input function. Therefore, a dynamic
tissue rich in proteoglycans, which can be encoun- contrast-enhancement (DCE) MRI sequence is
tered frequently in carotid atherosclerosis of var- started prior to gadolinium injection. In practice,
ious stages. Loose matrix tissue is rich in water this is a T1-WI bright-blood gradient echo
and shows up as hyperintense signals on T2-WI sequence with inflow saturation of both arterial
images. In addition, loose matrix may be perme- and venous flow. After two dynamics are
able to gadolinium and show up as areas of high acquired, half dose of gadolinium contrast bolus
contrast uptake on post-contrast black-blood is injected at a rate of 1 cc/s. Since the T1 relax-
MRI. However, their clinical implications when ation time of blood is drastically reduced, a bright
encountered in advanced lesions are uncertain. blood effect is created post-contrast. About
20 dynamic scans with scan times of 15 s per
dynamic scan are collected. Kinetic modeling
Neovasculature and Inflammation using the Patlak model provides two quantitative
measurements: fractional plasma volume (vp) and
Unlike the previous components, neovasculature permeability rate constant (Ktrans). Both vp and
and inflammation detection require specialized Ktrans have been found to be correlated with his-
modeling of gadolinium contrast uptake for detec- tologically measured neovasculature and macro-
tion. In contrast to post-contrast T1-WI black- phages in a validation study using carotid
blood MRI used for identification of lipid core, endarterectomy specimens [4]. Figure 4 shows a
neovasculature and inflammation detection pseudo-color image with vp in red and Ktrans in
requires fast and continuous acquisition of images green.

Fig. 4 Vasa vasorum


imaging for evaluating
plaque revascularization
and inflammation. Cross-
sectional images of right
carotid bifurcation on
pre-contrast T1-WI QIR (a),
post-contrast T1-WI QIR
(b), and corresponding vasa
vasorum image (c) obtained
from contrast kinetic
modeling of DCE-MRI.
Red areas correspond to
partial volume of blood (vp)
and green areas correspond
to transfer constant (Ktrans).
Asterisk indicates lumen;
circle, jugular vein; arrow
head, lipid-rich necrotic
core (Reprinted with
permission from Balu
et al. [51])
106 J. Sun et al.

Ultrasmall super paramagnetic particles of iron analysis software, CASCADE, developed at the
oxides (USPIO) are a class of iron oxide magnetic University of Washington, has processing rou-
particles that are also used for imaging plaque tines for multi-contrast image registration and
inflammation [25]. After systemic injection, semiautomated lumen and wall detection [27]. In
USPIOs are concentrated in macrophages and order to assist readers in measurement of plaque
can be detected by their strong magnetic components, a morphology-enhanced probabilis-
susceptibility. tic plaque segmentation (MEPPS) algorithm can
Long TE gradient echo sequences can be used be used to automatically segment plaque compo-
to detect the T2 or T2* effect of USPIOs. Similar nents [28]. The component boundaries are reader
to gadolinium contrast injection, areas of plaque correctable if required. Following the analysis,
with macrophages can be identified by comparing total volume of plaque components as well as
pre and post USPIO injection. In contrast to gad- percent volumes relative to plaque volume can
olinium, areas of macrophage concentration are be calculated. A 3D distribution of plaque com-
identified by loss of signal, and the signal differ- ponents and plaque burden can also be used for
ence is maximized at 36 h as compared to assessment as shown in Fig. 5.
5–10 min for gadolinium. Use of positive suscep-
tibility contrast techniques may improve detection
of USPIOs [26]. Current Applications

Imaging-Based Patient Stratification


Measurement of Plaque Morphology
and Composition Patients with carotid atherosclerosis are managed
with stringent risk factor control that covers life-
Several important measurements can be derived style changes and medications. Nonetheless,
from a multi-contrast plaque MRI protocol. Out- revascularization could be highly beneficial to
lines of the lumen and outer wall on black-blood those with a high risk of future stroke. However,
MRI provide a measure of the plaque burden. The this potential benefit needs to be evaluated against
wall area defined as the difference between the non-negligible surgical risk for complications
two outlines provides a first measurement of such as perioperative myocardial infarction and
plaque in the vessel wall. Plaque area normalized distal embolism on an individual basis. In the
for vessel size by the outer wall area provides the NASCET study which compared carotid endarter-
normalized wall index (NWI), which is similar to ectomy with medical treatment alone in patients
percent atheroma volume used in intravascular with transient ischemic attack or nondisabling ische-
ultrasound imaging of coronary atherosclerosis. mic stroke within the previous 6 months, there
While plaque burden can be measured on a single was an absolute risk reduction of 17  3.5 % at
contrast weighting such as pre-contrast black- 2 years favoring surgery in patients with ipsilateral
blood T1-WI sequence, outlining plaque bound- high-grade stenosis (70–99 %) [29]. In contrast,
aries on multi-contrast MRI is more robust for patients with mild stenosis (<50 %) did not benefit
advanced lesions in view of superficial calcifica- from surgery, although the remaining risk with med-
tion and plaque-mimicking flow artifacts. ical therapy was still considerable at 18.7 % over
Measurement of plaque composition requires a 5 years. Risk reduction with carotid endarterectomy
series of steps for multi-contrast image review. in asymptomatic patients was more moderate, even
These typically include multi-contrast image reg- in the presence of high-grade stenosis, as shown in
istration and segmentation of plaque component ACAS and the Asymptomatic Carotid Stenosis
boundaries. Due to the small components and Trial (ACST), which estimated that about 17 patients
complex nature of plaque constituents, special need to be treated with carotid endarterectomy to
algorithms targeted at atherosclerotic plaque anal- prevent one stroke in 5 years [30, 31]. Improvement
ysis are typically used. For example, the plaque in cost-effectiveness of carotid endarterectomy
7 Imaging of Carotid Atherosclerosis 107

Fig. 5 3D volume rendering of vessel wall morphology plaque burden and components. Orange indicates
and plaque components. Segmentation results of vessel intraplaque hemorrhage (arrow); blue, calcification; yel-
wall morphology and plaque components can be displayed low, lipid-rich necrotic core
using 3D volume rendering to illustrate distribution of

in asymptomatic patients is urgent in light of the MRI-detected plaque characteristics to increase


much larger patient population with asymptomatic survival benefits from carotid endarterectomy.
carotid atherosclerosis. In fact, 76.7 % of the In a study comparing symptomatic and asymp-
795,000 strokes that happen each year are first tomatic carotid plaques that cause at least 50 %
attacks. stenosis, multi-contrast MRI was used to compre-
The unmet need to identify who is at risk of hensively characterize plaque morphological and
future stroke and therefore would benefit from compositional features [32]. Fibrous cap rupture
surgical intervention has highlighted the impor- as well as large necrotic core and intraplaque
tance of imaging-based patient stratification in hemorrhage were more prevalent in symptomatic
clinical decision making. The aforementioned plaques, whereas no difference was found in lumi-
clinical trials were performed at a time when the nal stenosis or plaque volumes. As such, plaque
assessment of carotid atherosclerosis was largely compositional features appear to further differen-
based on angiography for measuring luminal tiate symptomatic plaques from those with a
narrowing as the sole criterion to inform surgeons. benign natural history among plaques causing
Since then, the understanding of carotid athero- severe stenosis. The convenience of using a single
sclerosis biology has advanced to appreciate the T1-WI imaging sequence to detect intraplaque
fundamental role of plaque rupture and thrombo- hemorrhage has led to studies that specifically
sis in the pathophysiology of neurological ische- examined the association of intraplaque hemor-
mic events. In the new paradigm built on plaque rhage with cerebrovascular events in broad
instability, luminal stenosis is more appropriately populations. Yamada et al. studied 392 carotid
viewed as one of the many risk factors for plaque arteries from 222 patients with suspected or con-
rupture rather than the primary cause of stroke. A firmed carotid atherosclerosis using the MPRAGE
number of morphological and pathological fea- sequence [33]. Carotid arteries were categorized
tures that differentiate ruptured from as mild (0–29 %), moderate (30–69 %), or severe
non-ruptured plaques can now be characterized (70–99 %) stenosis measured on contrast-
by carotid MRI, which holds the promise to be enhanced MR angiography. They noted that
used in prospective clinical trials that use carotid plaques with high signal intensity,
108 J. Sun et al.

indicative of intraplaque hemorrhage, were more Carotid Artery Stenting


likely to have had ipsilateral ischemic events within
the previous 6 months. The association between Carotid artery stenting has become a reasonable
ischemic events and MRI-detected ipsilateral alternative to carotid endarterectomy when
intraplaque hemorrhage was strongest in the patients are considered for revascularization due
group with moderate stenosis, implying that infor- to its less invasive nature. A few randomized trials
mation on plaque characteristics may be particu- have compared the two approaches in terms of
larly helpful in selecting high-risk patients where clinical outcomes including perioperative compli-
assessment of luminal stenosis is not able to cations and long-term event rates. Although long-
do. Howarth et al. compared ten symptomatic term event rates are comparable between the two,
patients and ten asymptomatic patients with a it has been noted that carotid artery stenting has a
focus on plaque inflammation measured with higher perioperative risk of embolic stroke com-
USPIO-enhanced MRI [25]. More regions with pared to carotid endarterectomy [39]. This has
signal drop, indicative of more extensive macro- raised concerns for carotid artery stenting, as
phage infiltration, were observed in symptomatic those perioperative microembolizations adversely
patients. These retrospective imaging studies affect patients’ cognitive function and has been
have reaffirmed the role of plaque instability in shown to lower the quality of life after procedure.
the pathophysiology of ischemic stroke as noted Plaque composition and thrombogenicity could
in histopathological studies and have extended the be a major determinant of the risk of distal embo-
finding to carotid atherosclerosis without severe lization in the setting of carotid artery stenting.
stenosis. Yoshimura et al. tested the hypothesis in a study
The prognostic value of plaque characteristics involving 112 patients undergoing carotid artery
has been demonstrated in prospective studies of stenting [40]. Diffusion-weighted imaging was
both symptomatic and asymptomatic patient performed before and after stenting to detect
cohorts. Takaya et al. followed 154 consecutive new ischemic lesions due to intervention.
patients with asymptomatic 50–79 % stenosis. Pre-operation time-of-flight MR angiography
Baseline plaque characteristics, including thin/ images were used to identify ipsilateral plaques
ruptured fibrous cap, intraplaque hemorrhage, with intraplaque hemorrhage. Both new ischemic
larger hemorrhage area, large necrotic core, and lesions and perioperative symptoms were found to
maximal wall thickness, were predictive of subse- be more frequent in patients with intraplaque
quent ischemic events [34]. Most of these features hemorrhage. Carotid MRI performed prior to sur-
appear to predict recurrent strokes as well, as gical intervention therefore may contribute to an
shown by Kwee et al. in a study involving accurate evaluation of the risk of perioperative
126 symptomatic patients with 30–69 % stenosis microembolization, which can be translated into
[35]. In particular, MRI detection of intraplaque improved clinical outcomes of both carotid artery
hemorrhage has been shown in various clinical stenting and carotid endarterectomy.
populations to be associated with increased
atherothrombotic risk. Three independent meta-
analyses were recently published which estimated Cryptogenic Stroke
the predictive value of MRI-detected intraplaque
hemorrhage for planning future observational As many as a third of ischemic strokes are con-
studies and clinical trials [36–38]. Intraplaque sidered cryptogenic, which means that no definite
hemorrhage alone appears to be associated with etiology can be determined after clinical workup.
a more than tenfold increased risk of neurological Imaging carotid atherosclerosis with MRI may
ischemic events in symptomatic patients. enable visualization of telltale features of culprit
Leveraging such prognostic information assess- lesions, which will facilitate targeted secondary
able via carotid MRI could prove to be invaluable prevention strategies for individual patients by
in future clinical practice. revealing or excluding large artery atherosclerosis
7 Imaging of Carotid Atherosclerosis 109

as the underlying etiology. Freilinger classification based on plaque morphology and


et al. performed a preliminary study in a series of composition. Several studies have utilized MR
32 consecutive patients with clinically diagnosed imaging to look into the association of traditional
cryptogenic stroke and nonstenosing (<50 %) and novel atherosclerotic risk factors with plaque
carotid atherosclerosis [41]. A multi-contrast composition, such as calcification, necrotic core,
MRI protocol detected complicated lesions, and intraplaque hemorrhage, in population-based
including fibrous cap rupture, intraplaque hemor- study samples. Wasserman et al. studied 214 sub-
rhage, and mural thrombus, in 12 (37.5 %) jects with carotid atherosclerosis enrolled in the
plaques ipsilateral to the cryptogenic stroke, Multi-Ethnic Study of Atherosclerosis (MESA)
whereas none was found on the contralateral side study [42]. After adjustment for potential
which showed MR signals consistent with confounding factors, subjects in the middle and
fibroatheroma and pathological intimal thickening highest tertile of total plasma cholesterol had a
as the predominant lesion type. higher likelihood of showing a lipid core com-
For the purpose of differential diagnosis in pared to the lowest tertile (odds ratio: 2.76 and
patients presenting with cryptogenic stroke, it is 4.63, respectively). The MRI study of the
preferable to use a protocol that affords extended Rotterdam study participants found that
coverage as patients can have multiple tandem intraplaque hemorrhage was associated with a
lesions and the location of culprit features, if number of clinical risk factors including age,
any, is not known a priori. The newly developed sex, hypertension, and smoking, whereas necrotic
3D gradient echo-based sequences are showing core was associated with sex and
capability to cover both extracranial and intracra- hypercholesterolemia [43].
nial carotid arteries, which may be a viable solu- Lesions in animal models are usually in the
tion in the near future. early stage of atherosclerosis and lack important
advanced features seen in human lesions. This has
led to a significant knowledge gap in understand-
In Vivo Insights on Pathobiological ing how atherosclerosis progresses from subclin-
Mechanisms ical lesions to clinically overt, culprit lesions.
Asymptomatic carotid atherosclerosis coupled
As an increasing number of biological targets with in vivo imaging offers a unique opportunity
implicated in the pathogenesis of atherosclerosis for clinical researchers to advance understanding
can now be successfully imaged with advances in of important pathological events leading to clini-
MR sequence design and post-processing, carotid cal events. In a case-control study comparing
MRI has become a powerful tool for studying carotid plaques with and without intraplaque hem-
disease mechanisms. The advantages of carotid orrhage, an 18-month imaging follow-up demon-
MRI as a research tool for clinical studies include strated distinctively different progression patterns
its noninvasive nature, absence of ionizing radia- between the two groups [44]. While plaques with-
tion, high reproducibility facilitating serial imag- out intraplaque hemorrhage tended to be station-
ing design, and the opportunity of combining ary over the study period, those with intraplaque
different contrast weightings for better plaque hemorrhage showed a larger increase in wall vol-
characterization. ume and necrotic core volume, which was trans-
Epidemiological studies have traditionally lated mainly into luminal narrowing. The ability
used carotid ultrasound or coronary calcium scan of MRI to serially track changes of carotid athero-
to measure subclinical atherosclerosis. Recent sclerosis was shown in a recent study, where mul-
studies have attempted to introduce MR plaque tiple MRI scans with a constant time interval were
imaging to epidemiological studies. This is a retrospectively retrieved from subjects with
novel concept as MR plaque imaging not only intraplaque hemorrhage to examine the immediate
provides measurement of subclinical atheroscle- and long-term effects of intraplaque hemorrhage
rotic burden but also allows phenotypic on plaque progression [45]. Images spanning 4–5
110 J. Sun et al.

years successfully captured how plaques Efficacy Markers for Testing


progressed from the stage without intraplaque Therapeutic Strategies
hemorrhage to the stage with intraplaque hemor-
rhage. Rather than a transient mass effect on The ability of MRI to track morphological and
plaque volume, the development of intraplaque compositional changes of atherosclerotic plaques
hemorrhage was found to have gradually altered has also led to research efforts using quantitative
the trajectory of plaque progression by posing an MRI measurements of carotid atherosclerosis
immediate and long-lasting promoting effect on as efficacy markers for testing therapeutic
plaque progression. Given that animal models do strategies that aim to stabilize atherosclerotic
not mimic advanced, clinical relevant plaques, plaques (Fig. 6). As a surrogate for systemic ath-
knowledge so-obtained from imaging human erosclerosis, information on carotid atherosclero-
carotid atherosclerosis is invaluable to the under- sis should give clues to the underlying coronary
standing of the natural history of high-risk atherosclerosis, in which vessel wall information
plaques, which may point to promising directions currently obtainable is still limited due to
for reducing residual cardiovascular risk in con- technical constraints. This may be particularly
temporary medicine. important in the current era due to several reasons.

Fig. 6 Serial MR scans showing plaque stabilization same plaque, consistent with plaque stabilization. The
under pharmacotherapy. Top panel: three consecutive distinct lumen (L ) and outer wall boundaries (long arrows)
cross sections from the baseline scan show a plaque with suggest a reduction in plaque volume. However, the most
a big necrotic core, which is delineated as non-enhanced dramatic change is the decrease in non-enhanced areas,
areas on contrast-enhanced T1-WI (CE-T1) images. Bot- indicative of smaller necrotic core size, at follow-up. A
tom panel: Matched cross sections from the follow-up scan region with thin fibrous cap at baseline had increased cap
show morphological and compositional changes of the thickness at follow-up (hollow arrows)
7 Imaging of Carotid Atherosclerosis 111

First, the prevalence of atherosclerosis is high, rosuvastatin on carotid plaque morphology and
but the incidence of atherothrombotic complica- composition in more advanced lesions causing
tions of any given population is low, especially in 16–79 % stenosis [48]. A significant reduction in
the primary prevention setting. Consequently, necrotic core size was noted at 2 years, but not in
while a high volume of myocardial infarction wall volume, which suggests plaque lipid deple-
and ischemic strokes are seen clinically, the tion, and thus stability, may be one of the under-
large number of subjects needed to power a lying reasons for clinical benefits with statin
clinical trial has been daunting. Thus, early sig- therapy. The time course of plaque lipid depletion
nals from surrogate markers that can display under lipid-lowering treatment was later
the efficacy of experimental agents are highly addressed by Zhao et al. [49]. In 33 subjects
desirable. Second, novel surrogate markers, with documented necrotic core at baseline,
to certain extent, target biological activities or necrotic core size gradually decreased over
pathways in atherogenesis. Characterizing 3 years. While a significant reduction in necrotic
changes in those biomarkers will help clarify core could be seen in year 1, reduction in wall
the mechanisms of actions exerted by the volume was not apparent until year 2, lagging
experimental agent, which often deviate behind the change in necrotic core. When plaque
from what are known from preclinical studies. inflammation is targeted, previous studies have
Third, it is common for patients with different shown a reduction in imaging signals within a
demographics or clinical conditions to have het- few weeks [50]. Tang et al. compared two doses
erogeneous responses to the same treatment, of atorvastatin treatment in 47 patients with over
which has led to the common practice of 40 % carotid stenosis and USPIO uptake at base-
predefined subgroup analysis in clinical line. The high-dose group showed a significant
trials. Given the dramatic morphological and reduction in USPIO-defined inflammation at
pathological differences, plaques at different 6 weeks [50].
stages or with different phenotypes are likely to The aforementioned studies are all single cen-
respond heterogeneously to the same medication, ter studies. However, in cases of large-scale
which, however, is less understood. Intraplaque recruitment, short recruitment period, or clinical
hemorrhage appears to be one such factor conditions with low prevalence, multicenter
influencing therapeutic response [46]. In this design is usually the norm. In view of the substan-
regard, plaque characteristics may not only tial variations in technique instrumentation
serve as efficacy markers but also enable a between imaging sites, the recent completion of
deeper understanding of the efficacy of a few multicenter carotid MRI studies represents a
antiatherogenic medications in terms of individual solid step toward the standardization of MRI
responses. protocol and quantitative image analysis. A mul-
A number of studies have been conducted ticenter clinical trial involving 13 imaging
using MRI-based plaque measurements as end sites and two vendor platforms at 3-T was carried
points, predominantly using statins. Corti out to examine change in necrotic core size of
et al. studied 32 carotid plaques under the carotid atherosclerosis over a short 6-month
treatment of simvastatin [47]. Reduction in wall period [46]. Under current standard-of-care to
area but not lumen area was seen at 1 year. When medically control modifiable cardiovascular risk
follow-up was extended to 2 years, there was a factors, it was shown that plaques without
further decrease in wall area and a slight intraplaque hemorrhage had a small but signifi-
increase in lumen area. These findings indicate cant reduction in necrotic core size. In contrast,
that carotid atherosclerosis is subject to rapid plaques with intraplaque hemorrhage had an
progression or regression. Change in wall area increase in necrotic core size and wall volume
appeared to precede change in lumen area. and a decrease in lumen volume – a progression
Another study by Underhill et al. used pattern consistent with previous natural history
multi-contrast MRI to examine the effect of studies.
112 J. Sun et al.

Summary References

Carotid atherosclerosis is a common clinical find- 1. de Weerd M, Greving JP, de Jong AW, Buskens E, Bots
ML (2009) Prevalence of asymptomatic carotid artery
ing, which almost always involves the bifurcation stenosis according to age and sex: systematic review
segment where recirculating flow is most and metaregression analysis. Stroke 40:1105–1113
severe. Vessel wall MRI, with high soft tissue 2. Cai JM, Hatsukami TS, Ferguson MS, Kerwin WS,
contrast and flexibility of sequence design, affords Saam T, Chu BC, Takaya N, Polissar NL, Yuan C
(2005) In vivo quantitative measurement of intact
inherent advantages for imaging carotid
fibrous cap and lipid-rich necrotic core size in athero-
atherosclerosis, yet several key technical points sclerotic carotid plaque: comparison of high-
must be addressed to achieve satisfactory visual- resolution, contrast-enhanced magnetic resonance
ization of vessel wall pathology, including flow imaging and histology. Circulation 112:3437–3444
3. Moody AR, Murphy RE, Morgan PS, Martel AL,
and fat suppression, submillimeter spatial resolu-
Delay GS, Allder S, MacSweeney ST, Tennant WG,
tion, and sufficient SNR within clinically applica- Gladman J, Lowe J, Hunt BJ (2003) Characterization
ble scan times. Traditional 2D imaging has gained of complicated carotid plaque with magnetic resonance
success in providing clinically relevant informa- direct thrombus imaging in patients with cerebral
ischemia. Circulation 107:3047–3052
tion of atherosclerosis, demonstrated by a broad 4. Kerwin WS, O’Brien KD, Ferguson MS, Polissar N,
spectrum of applications in previous literature. Hatsukami TS, Yuan C (2006) Inflammation in carotid
But newly developed 3D sequences hold the atherosclerotic plaque: a dynamic contrast-enhanced
promise for future clinical practice due to short- MR imaging study. Radiology 241:459–468
5. Trivedi RA, Mallawarachi C, U-King-Im JM, Graves
ened scan times and increased scan coverage.
MJ, Horsley J, Goddard MJ, Brown A, Wang L,
Atherosclerotic plaques display considerable Kirkpatrick PJ, Brown J, Gillard JH (2006) Identifying
morphological and pathological variations. inflamed carotid plaques using in vivo USPIO-
Appropriate interpretation of image findings may enhanced MR imaging to label plaque macrophages.
Arterioscler Thromb Vasc Biol 26:1601–1606
depend on a sensible understanding of both
6. Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz
pathology and the capability and limitations SM (2000) Lessons from sudden coronary death – a
of MRI. comprehensive morphological classification scheme
With the changing landscape in the field of for atherosclerotic lesions. Arterioscler Thromb Vasc
Biol 20:1262–1275
atherosclerosis management including pharmaco-
7. Leitinger N, Schulman IG (2013) Phenotypic polariza-
therapy and surgical intervention, clinicians are tion of macrophages in atherosclerosis. Arterioscler
facing either a shortage of data or data that are Thromb Vasc Biol 33:1120–1126
not generalizable to current practice, when 8. Virmani R, Ladich ER, Burke AP, Kolodgie FD (2006)
Histopathology of carotid atherosclerotic disease. Neu-
selecting between medical treatment, carotid end-
rosurgery 59:S219–S227, S3-S13
arterectomy, and carotid artery stenting. Further- 9. Spagnoli LG, Mauriello A, Sangiorgi G, Fratoni S,
more, the limitations of luminal stenosis in Bonanno E, Schwartz RS, Piepgras DG, Pistolese R,
predicting patients’ future risk of ischemic stroke Ippoliti A, Holmes DR (2004) Extracranial
thrombotically active carotid plaque as a risk factor
are increasingly recognized. New opportunities
for ischemic stroke. JAMA 292:1845–1852
have emerged that leverage technical develop- 10. Fisher M, Paganini-Hill A, Martin A, Cosgrove M,
ments in MRI of carotid atherosclerosis to assist Toole JF, Barnett HJ, Norris J (2005) Carotid plaque
clinical decision making. As such, management of pathology: thrombosis, ulceration, and stroke patho-
genesis. Stroke 36:253–257
patients with symptomatic or asymptomatic
11. Redgrave J, Lovett JK, Gallagher PJ, Rothwell PM
carotid atherosclerosis is awaiting significant par- (2006) Histological assessment of 526 symptomatic
adigm shift. With continued efforts in clinical carotid plaques in relation to the nature and timing of
studies using direct MR imaging of carotid ath- ischemic symptoms – the Oxford plaque study. Circu-
lation 113:2320–2328
erosclerosis, the clinical outcomes of carotid ath-
12. Narula J, Nakano M, Virmani R, Kolodgie FD,
erosclerosis may eventually be improved to a new Petersen R, Newcomb R, Malik S, Fuster V, Finn AV
level. (2013) Histopathologic characteristics of
7 Imaging of Carotid Atherosclerosis 113

atherosclerotic coronary disease and implications of atherosclerotic disease evaluation. Magn Reson Med
the findings for the invasive and noninvasive detection 69:337–345
of vulnerable plaques. J Am Coll Cardiol 25. Howarth SP, Tang TY, Trivedi R, Weerakkody R,
61:1041–1051 U-King-Im J, Gaunt ME, Boyle JR, Li ZY, Miller SR,
13. Yarnykh VL, Terashima M, Hayes CE, Shimakawa A, Graves MJ, Gillard JH (2009) Utility of USPIO-
Takaya N, Nguyen PK, Brittain JH, McConnell MV, enhanced MR imaging to identify inflammation and
Yuan C (2006) Multicontrast black-blood MRI of the fibrous cap: a comparison of symptomatic and
carotid arteries: comparison between 1.5 and 3 Tesla asymptomatic individuals. Eur J Radiol 70:555–560
magnetic field strengths. J Magn Reson Imaging 26. Stuber M, Gilson WD, Schar M, Kedziorek DA, Hof-
23:691–698 mann LV, Shah S, Vonken EJ, Bulte JW, Kraitchman
14. Balu N, Yarnykh VL, Scholnick J, Chu B, Yuan C, DL (2007) Positive contrast visualization of iron oxide-
Hayes C (2009) Improvements in carotid plaque imag- labeled stem cells using inversion-recovery with
ing using a new eight-element phased array coil at 3 T. ON-resonant water suppression (IRON). Magn Reson
J Magn Reson Imaging 30:1209–1214 Med 58:1072–1077
15. Parker DL, Goodrich KC, Masiker M, Tsuruda JS, 27. Kerwin WS, Xu D, Liu F, Saam T, Underhill HR,
Katzman GL (2002) Improved efficiency in double- Takaya N, Chu BC, Hatsukami TS, Yuan C (2007)
inversion fast spin-echo imaging. Magn Reson Med Magnetic resonance imaging of carotid atherosclerosis:
47:1017–1021 plaque analysis. Top Magn Reson Imaging
16. Yarnykh VL, Yuan C (2003) Multislice double 18:371–378
inversion-recovery black-blood imaging with simulta- 28. Liu F, Xu DX, Ferguson MS, Chu BC, Saam T,
neous slice reinversion. J Magn Reson Imaging Takaya N, Hatsukami TS, Yuan C, Kerwin WS
17:478–483 (2006) Automated in vivo segmentation of carotid
17. Wang J, Yarnykh VL, Hatsukami T, Chu B, Balu N, plaque MRI with morphology-enhanced probability
Yuan C (2007) Improved suppression of plaque- maps. Magn Reson Med 55:659–668
mimicking artifacts in black-blood carotid atheroscle- 29. Barnett HJ, Taylor DW, Eliasziw M, Fox AJ, Ferguson
rosis imaging using a multislice motion-sensitized GG, Haynes RB, Rankin RN, Clagett GP, Hachinski
driven-equilibrium (MSDE) turbo spin-echo (TSE) VC, Sackett DL, Thorpe KE, Meldrum HE, Spence JD
sequence. Magn Reson Med 58:973–981 (1998) Benefit of carotid endarterectomy in patients
18. Wang J, Yarnykh VL, Yuan C (2010) Enhanced image with symptomatic moderate or severe stenosis. North
quality in black-blood MRI using the improved American Symptomatic Carotid Endarterectomy Trial
motion-sensitized driven-equilibrium (iMSDE) Collaborators. N Engl J Med 339:1415–1425
sequence. J Magn Reson Imaging 31:1256–1263 30. Walker MD, Marler JR, Goldstein M, Grady PA, Toole
19. Li L, Miller KL, Jezzard P (2012) DANTE-prepared JF, Baker WH, Castaldo JE, Chambless LE, Moore
pulse trains: a novel approach to motion-sensitized and WS, Robertson JT, Young B, Howard VJ, Purvis S,
motion-suppressed quantitative magnetic resonance Vernon DD, Needham K, Beck P, Celani VJ,
imaging. Magn Reson Med 68:1423–1438 Sauerbeck L, von Rajcs JA, Atkins D (1995) Endarter-
20. Balu N, Yarnykh VL, Chu B, Wang J, Hatsukami T, ectomy for asymptomatic carotid artery stenosis.
Yuan C (2011) Carotid plaque assessment using fast 3D JAMA 273:1421–1428
isotropic resolution black-blood MRI. Magn Reson 31. Halliday A, Mansfield A, Marro J, Peto C, Peto R,
Med 65:627–637 Potter J, Thomas D (2004) Prevention of disabling
21. Fan Z, Zhang Z, Chung YC, Weale P, Zuehlsdorff S, and fatal strokes by successful carotid endarterectomy
Carr J, Li D (2010) Carotid arterial wall MRI at 3T in patients without recent neurological symptoms:
using 3D variable-flip-angle turbo spin-echo (TSE) randomised controlled trial. Lancet 363:1491–1502
with flow-sensitive dephasing (FSD). J Magn Reson 32. U-King-Im JM, Tang TY, Patterson A, Graves MJ,
Imaging 31:645–654 Howarth S, Li ZY, Trivedi R, Bowden D, Kirkpatrick
22. Yarnykh VL, Yuan C (2002) T-1-insensitive flow sup- PJ, Gaunt ME, Warburton EA, Antoun NM, Gillard JH
pression using quadruple inversion-recovery. Magn (2008) Characterisation of carotid atheroma in symp-
Reson Med 48:899–905 tomatic and asymptomatic patients using high resolu-
23. Ota H, Yarnykh VL, Ferguson MS, Underhill HR, tion MRI. J Neurol Neurosurg Psychiatry 79:905–912
DeMarco JK, Zhu DC, Oikawa M, Dong L, Zhao 33. Yamada N, Higashi M, Otsubo R, Sakuma T,
XH, Collar A, Hatsukami TS, Yuan C (2010) Carotid Oyama N, Tanaka R, Iihara K, Naritomi H,
intraplaque hemorrhage imaging at 3.0-T MR imaging: Minematsu K, Naito H (2007) Association between
comparison of the diagnostic performance of three signal hyperintensity on T1-weighted MR imaging of
T1-weighted sequences. Radiology 254:551–563 carotid plaques and ipsilateral ischemic events. AJNR
24. Wang J, Bornert P, Zhao H, Hippe DS, Zhao X, Balu N, Am J Neuroradiol 28:287–292
Ferguson MS, Hatsukami TS, Xu J, Yuan C, Kerwin 34. Takaya N, Yuan C, Chu BC, Saam T, Underhill H, Cai
WS (2013) Simultaneous noncontrast angiography and JM, Tran N, Polissar NL, Isaac C, Ferguson MS, Gar-
intraPlaque hemorrhage (SNAP) imaging for carotid den GA, Cramer SC, Maravilla KR, Hashimoto B,
114 J. Sun et al.

Hatsukami TS (2006) Association between carotid Determinants of magnetic resonance imaging detected
plaque characteristics and subsequent ischemic cere- carotid plaque components: the Rotterdam study. Eur
brovascular events: a prospective assessment with MRI Heart J 33:221–229
– initial results. Stroke 37:818–823 44. Takaya N, Yuan C, Chu BC, Saam T, Polissar NL,
35. Kwee RM, van Oostenbrugge RJ, Mess WH, Prins Jarvik GP, Isaac C, McDonough J, Natiello C,
MH, van der Geest RJ, Ter Berg JW, Franke CL, Small R, Ferguson MS, Hatsukami TS (2005) Presence
Korten AG, Meems BJ, van Engelshoven JM, of intraplaque hemorrhage stimulates progression of
Wildberger JE, Kooi ME (2013) MRI of carotid ath- carotid atherosclerotic plaques: a high-resolution mag-
erosclerosis to identify TIA and stroke patients who are netic resonance imaging study. Circulation
at risk of a recurrence. J Magn Reson Imaging 111:2768–2775
37:1189–1194 45. Sun J, Underhill HR, Hippe DS, Xue Y, Yuan C,
36. Saam T, Hetterich H, Hoffmann V, Yuan C, Hatsukami TS (2012) Sustained acceleration in carotid
Dichgans M, Poppert H, Koeppel T, Hoffmann U, atherosclerotic plaque progression with intraplaque
Reiser MF, Bamberg F (2013) Meta-analysis and sys- hemorrhage: a long-term time course study. J Am
tematic review of the predictive value of carotid plaque Coll Cardiol Img 5:798–804
hemorrhage on cerebrovascular events by magnetic 46. Sun J, Balu N, Hippe DS, Xue Y, Dong L, Zhao X, Li F,
resonance imaging. J Am Coll Cardiol 62:1081–1091 Xu D, Hatsukami TS, Yuan C (2013) Subclinical
37. Hosseini AA, Kandiyil N, Macsweeney ST, Altaf N, carotid atherosclerosis: short-term natural history of
Auer DP (2013) Carotid plaque hemorrhage on mag- lipid-rich necrotic core–a multicenter study with MR
netic resonance imaging strongly predicts recurrent imaging. Radiology 268:61–68
ischemia and stroke. Ann Neurol 73:774–784 47. Corti R, Fuster V, Fayad ZA, Worthley SG, Helft G,
38. Gupta A, Baradaran H, Schweitzer AD, Kamel H, Smith D, Weinberger J, Wentzel J, Mizsei G,
Pandya A, Delgado D, Dunning A, Mushlin AI, Sanelli Mercuri M, Badimon JJ (2002) Lipid lowering by
PC (2013) Carotid plaque MRI and stroke risk: a sys- simvastatin induces regression of human atheroscle-
tematic review and meta-analysis. Stroke rotic lesions: two years’ follow-up by high-resolution
44:3071–3077 noninvasive magnetic resonance imaging. Circulation
39. Brott TG, Hobson RW, Howard G, Roubin GS, Clark 106:2884–2887
WM, Brooks W, Mackey A, Hill MD, Leimgruber PP, 48. Underhill HR, Yuan C, Zhao XQ, Kraiss LW, Parker
Sheffet AJ, Howard VJ, Moore WS, Voeks JH, Hop- DL, Saam T, Chu B, Takaya N, Liu F, Polissar NL,
kins LN, Cutlip DE, Cohen DJ, Popma JJ, Ferguson Neradilek B, Raichlen JS, Cain VA, Waterton JC,
RD, Cohen SN, Blackshear JL, Silver FL, Mohr JP, Lal Hamar W, Hatsukami TS (2008) Effect of rosuvastatin
BK, Meschia JF (2010) Stenting versus endarterec- therapy on carotid plaque morphology and composi-
tomy for treatment of carotid-artery stenosis. N Engl J tion in moderately hypercholesterolemic patients: a
Med 363:11–23 high-resolution magnetic resonance imaging trial. Am
40. Yoshimura S, Yamada K, Kawasaki M, Asano T, Heart J 155:581–584
Kanematsu M, Takamatsu M, Hara A, Iwama T 49. Zhao XQ, Dong L, Hatsukami T, Phan BA, Chu B,
(2011) High-intensity signal on time-of-flight magnetic Moore A, Lane T, Neradilek MB, Polissar N,
resonance angiography indicates carotid plaques at Monick D, Lee C, Underhill H, Yuan C (2011) MR
high risk for cerebral embolism during stenting. Stroke imaging of carotid plaque composition during
42:3132–3137 lipid-lowering therapy: a prospective assessment of
41. Freilinger TM, Schindler A, Schmidt C, Grimm J, effect and time course. J Am Coll Cardiol Img
Cyran C, Schwarz F, Bamberg F, Linn J, Reiser M, 4:977–986
Yuan C, Nikolaou K, Dichgans M, Saam T (2012) 50. Tang TY, Howarth SP, Miller SR, Graves MJ, Patterson
Prevalence of nonstenosing, complicated atheroscle- AJ, U-King-Im JM, Li ZY, Walsh SR, Brown AP,
rotic plaques in cryptogenic stroke. J Am Coll Cardiol Kirkpatrick PJ, Warburton EA, Hayes PD, Varty K,
Img 5:397–405 Boyle JR, Gaunt ME, Zalewski A, Gillard JH (2009)
42. Wasserman BA, Sharrett AR, Lai S, Gomes AS, The ATHEROMA (Atorvastatin Therapy: Effects on
Cushman M, Folsom AR, Bild DE, Kronmal RA, Reduction of Macrophage Activity) study. Evaluation
Sinha S, Bluemke DA (2008) Risk factor associations using ultrasmall superparamagnetic iron oxide-
with the presence of a lipid core in carotid plaque of enhanced magnetic resonance imaging in carotid dis-
asymptomatic individuals using high-resolution MRI: ease. J Am Coll Cardiol 53:2039–2050
the multi-ethnic study of atherosclerosis (MESA). 51. Balu N, Wang J, Dong L, Baluyot F, Chen H, Yuan C
Stroke 39:329–335 (2009) Current techniques for MR imaging of
43. van den Bouwhuijsen QJ, Vernooij MW, Hofman A, atherosclerosis. Top Magn Reson Imaging
Krestin GP, van der Lugt A, Witteman JC (2012) 20(4):203–215
Carotid Artery Dissection
8
Olivier Naggara, Myriam Edjlali-Goujon, Eric Bodiguel,
Marie Pierre Gobin-Metteil, Denis Trystram,
Christine Rodriguez-Regent, Jean-Louis Mas,
Jean Francois Meder, and Catherine Oppenheim

Contents Mural Hematoma Consequences . . . . . . . . . . . . . . . . . . . . 119


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116 Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Local Symptoms and Signs . . . . . . . . . . . . . . . . . . . . . . . . . . 120
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116 Ischemic Symptoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117 Outcome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Traumatic Carotid Artery Dissection . . . . . . . . . . . . . . . . 117 Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
Spontaneous Carotid Artery Dissection . . . . . . . . . . . . . 117 Digital Subtraction Angiography . . . . . . . . . . . . . . . . . . . . 122
Insights on the Mural Hematoma . . . . . . . . . . . . . . . . . 118 Ultrasound . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
Mural Hematoma Constitution . . . . . . . . . . . . . . . . . . . . . . 118 Cross-Sectional Imaging: CT or MRI? . . . . . . . . . . . . . . 123
Mural Hematoma Location . . . . . . . . . . . . . . . . . . . . . . . . . . 119 Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Medical Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Mechanical Revascularization Strategies . . . . . . . . . . . . 135

O. Naggara (*) Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135


Service de Neuroradiologie, Centre Hospitalier Sainte- References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Anne, INSERM UMR894, Université Paris Descartes,
Sorbonne Paris Cité, Paris, France
Service de Radiologie pédiatrique, Faculté de Médecine,
Hôpital Necker Enfants Malades, Université Paris
Descartes, Sorbonne Paris Cité, Paris, France
e-mail: o.naggara@ch-sainte-anne.fr
M. Edjlali-Goujon • M.P. Gobin-Metteil • D. Trystram •
C. Rodriguez-Regent • J.F. Meder • C. Oppenheim
Service de Neuroradiologie, Centre Hospitalier Sainte-
Anne, INSERM UMR894, Université Paris Descartes,
Sorbonne Paris Cité, Paris, France
e-mail: m.edjlali@ch-sainte-anne.fr;
gobin-metteil@ch-sainte-anne.fr;
d.trystram@ch-sainte-anne.fr;
c.rodriguez@ch-sainte-anne.fr;
jf.meder@ch-sainte-anne.fr;
c.oppenheim@ch-sainte-anne.fr
E. Bodiguel • J.-L. Mas
Service de Neurologie, Faculté de Médecine, Centre
Hospitalier Sainte-Anne, Université Paris Descartes,
Sorbonne Paris Cité, Paris, France
e-mail: e.bodiguel@ch-sainte-anne.fr;
jl.mas@ch-sainte-anne.fr

# Springer Science+Business Media New York 2016 115


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_16
116 O. Naggara et al.

Abstract Keywords
Carotid artery dissections (CAD) are a leading Carotid artery dissection • Mural hematoma •
cause of nonatherosclerotic stroke in young High resolution MRI • Stroke
adults, responsible for up to 25 % of ischemic
strokes. The cornerstone of CAD pathophysiol-
ogy and diagnosis is the presence of a mural Introduction
hematoma of unknown etiology, possibly
caused by an intimal tear, a primary rupture of Carotid artery dissection (CAD) is caused by the
vasa vasorum, or an underlying arteriopathy formation of a mural hematoma, of the subpetrous
impairing vasomotion. This mural hematoma segment of the internal carotid artery (ICA).
occurs within the media layer extending distally Although the clinical presentation of CAD can
and circumferentially. The diagnosis of CAD is be benign, cervical dissections are a major cause
generally based on a suggestive clinical presen- of ischemic stroke in middle-aged adults [1]. The
tation, exclusion of atherosclerosis, and support- increased diagnosis of this condition in the last
ive radiologic evidence. An early and reliable decade can be attributed to the availability of
diagnosis is important as anticoagulation or diagnostic imaging techniques, such as color
antithrombotic treatment is recommended in Doppler ultrasound (US), computed tomography
order to reduce the risk of an early thromboem- (CT), magnetic resonance (MR) imaging, and,
bolic event. The optimal imaging method for the more recently, high-resolution MR imaging of
diagnosis of CAD is still debated. Doppler ultra- the arterial wall (HR-MRI). Although several
sound examination (DUS), the first-line screen- comprehensive reviews have focused on different
ing tool, may show normal findings whenever aspects of CAD such as risk factors or treatment,
the mural hematoma results in subtle lumen the present review proposes a global overview of
alterations or is located in a segment that cannot the epidemiology, pathophysiology, and
readily be displayed by DUS. Despite the ability predisposing factors, clinical and imaging charac-
of CT angiography to reveal imaging findings of teristics, and therapeutic options. Additionally,
CAD, brain CT is known to be poorly sensitive published data on the outcome of CAD, including
for the detection of ischemic lesions. Thus, cer- mortality rates and recurrences, were summa-
vical contrast-enhanced MR angiography rized. This review deliberately focuses on sponta-
(CE-MRA) coupled with T1-WI fat-suppressed neous (nontraumatic) CADs in adults because
axial sequences has gradually replaced conven- these are managed by stroke physicians, whereas
tional angiography for the diagnosis of cervical traumatic ICAD and cases in children are more
artery dissection. However, the diagnosis of commonly handled by surgeons and pediatricians
CAD often remains challenging, and it is not and can require different management strategies.
rare for the diagnosis of CAD to remain pre- Moreover, purely intracranial dissections or aortic
sumptive despite an extensive imaging work- dissections extending to the cervical arteries are
up. The diagnosis can be retrospectively con- outside the field of this review because their
firmed on imaging follow-up by monitoring the mechanisms, symptoms, and management are
lumen healing or progression in response to different.
treatment. High-resolution MRI (HR-MRI) of
the arterial wall is a noninvasive imaging tech-
nique that provides a delineation of the lumen Epidemiology
and arterial wall. This technique, available at 1.5
and 3-T, which requires standard or dedicated The incidence of CAD in the general population is
surface coils, has been extensively used for low, around 2,6 (95 % CI 1.9–3.3) per 100,000
carotid atherosclerosis and, more recently, for inhabitants per year [2]. Vertebral artery dissec-
carotid dissection. tions (VAD) are less common than CAD, although
8 Carotid Artery Dissection 117

this difference has been reduced by the increased The risk of ICAD is increased after major thoracic
availability of MRI, which enables an accurate injuries, whereas the risk of vertebral artery dis-
diagnosis of vertebral artery dissection [3]. sections, over the scope of this review, is
In the North American study, CAD occurred in increased in case of cervical spine fractures [6].
the community at a mean age of 45, 8 years, which
is similar to reports from two large published Minor Traumatism or Mechanical Trigger
multicenter series of ICAD in European Event
populations (44,0 years [459 patients] and 45,3 A dissection is termed spontaneous whenever
years [696 patients]) [2, 4]. there is no history of major trauma. Major pene-
A slight predominance in men was reported in trating or nonpenetrating trauma to the artery is
the European multicenter hospital-based series often lacking, but precipitating events involving
(53–57 %), whereas 0–52 % of the patients were hyperextension, rotation, or lateroversion of the
women in the North American population-based neck are often reported by patients with ICAD
study [2, 4]. CADs seem to occur more frequently [7]. These include whiplash injury, chiropractic
in men and at an older age (47,0 vs. 43,4 years) manipulation, various sporting activities, neck
than do vertebral artery dissections [2, 4]. These movements, and severe coughing. The exact role
age and gender differences are difficult to explain. of these precipitating events in the pathophysiol-
However, it is possible that risk factors or trigger- ogy of ICAD is unclear but suggests a mechanical
ing events, such as traumas in young patients, are contribution. The styloid process and hyoid bone,
more involved in the pathogenesis of VAD both in the vicinity to the ICA, have been impli-
than ICAD. cated in the pathogenesis of CAD with shorter
distances between the styloid process and hyoid
bone and dissected ICA than in nondissection
Pathogenesis carotid pathology [8, 9].
However, such traumas are recurrent and result
The pathophysiology of CAD remains controver- in ICAD in only a few individuals. If anatomic
sial. Patients with ICAD may have a constitu- factors contribute so strongly to CAD risk, one
tional, at least in part, genetically determined could wonder why ICAD recurrence is rare. If
weakness of the vessel wall. Environmental fac- prior cervical trauma seems to be an important
tors such as minor trauma or acute infection could environmental determinant of CAD, it is not an
act as triggers [5]. ICADs are usually classified as independent outcome predictor [7]. Because of
either traumatic in case of major trauma preceding the characteristics of most prior cervical trauma,
the onset of clinical symptoms or spontaneous, the term mechanical trigger event rather than
otherwise. trauma may be more appropriate [7].

Traumatic Carotid Artery Dissection Spontaneous Carotid Artery Dissection

Major Traumatism In the absence of a traumatic trigger, the etiology


Traumatic ICADs can occur as a result of major often remains undefined, likely to be multifactorial,
penetrating or nonpenetrating traumas. The most including infections, environmental exposures, and
common cause of a traumatic dissection is blunt collagen abnormalities of the arterial wall.
(severe hyperextension) or penetrating (stab
wound) injury to the neck. CAD has been shown Infection
to occur in about 1–2 % of patients who have had An association of ICAD with history of recent
blunt trauma, and the risk is also increased with infection has been reported [10, 11], potentially
trauma-associated injuries, such as facial or skull- predisposing individuals to CAD via endothelial
base fractures, and traumatic brain injury [6]. damage or prothrombotic mechanisms [10–14].
118 O. Naggara et al.

A possible causal role of recent infection is Transient Arteriopathy


supported by a seasonal peak of CAD in autumn. The epidemiology of CAD with an incidence peak
This pattern has been attributed to an increased in autumn, the clinical observation that some
occurrence of infection and weather-related patients with CAD develop multiple consecutive
changes in blood pressure, coagulation parame- dissections within weeks [18, 19], and the associ-
ters, and physical activity [13]. The failure to ation with hereditary connective tissue diseases,
prove a specific infective agent led to the hypoth- as well as ultrastructural abnormalities of connec-
esis that the inflammatory response rather than a tive tissue in skin biopsy specimens, support the
specific infective agent might be responsible for a hypothesis that a general susceptibility state is
transient arteriopathy nonspecific inflammation- involved in the pathogenesis of CAD.
mediated injury of the arterial wall linked to The presence of an underlying vasculopathy is
CAD [15]. Reports of elevated C-reactive protein also suggested by the fact that CAD patients com-
(CRP) and peripheral leukocyte counts in CAD monly present with concomitant arterial abnor-
support this hypothesis [16]. malities, such as fibromuscular dysplasia, aortic-
root dilation, hyper-distensibility [20], and
Genetic Risk Factors increased stiffness of the arterial wall, as well as
Genetic factors might also have a role in the impaired spontaneous and endothelial-dependent
pathophysiology of CAD, mainly as part of a vasodilation [21]. About 50 % of patients with
multifactorial predisposition. The prevailing the- CAD have connective tissue aberrations in their
ory is that CAD is a multifactorial disease caused skin, including composite collagen fibrils and
by several genetic variants and environmental fragmentation of elastic fibers [22]. In addition,
factors, each probably having a modest and signs of tissue weakening along the tunica media
potentially synergistic effect. The high preva- and the tunica adventitia junction in superficial
lence of multiple dissection events and of long- temporal artery biopsy specimens of patients
term (>1 year) recurrent dissections in patients with CAD but not of controls suggest the presence
with a familial history of CAD indicates that a of a generalized arteriopathy leading to impair-
specific predisposition for familial cervical dis- ment of the stability of the arterial wall in patients
sections may exist [15]. Less often (<2 % of with CAD [23]. Association of CAD with visceral
cases), ICAD has been associated with a known artery dissection, or reversible cerebral vasocon-
monogenic connective tissue disease, such as striction syndrome [24], strengthens this inflamma-
vascular Ehlers-Danlos syndrome type IV [15], tory hypothesis [5, 15]. This latter hypothesis is
or, more rarely, in patients with Marfan’s further reinforced by high-resolution MRI findings
syndrome or other known monogenic connective (see “High-Resolution MRI” section) [25, 26].
tissue disorders, such as osteogenesis imperfect
[15]. About half of patients with cervical dissec-
tions have abnormalities in skin connective tissue Insights on the Mural Hematoma
that follow an autosomal-dominant inheritance
pattern [15]. Mural Hematoma Constitution
Although these findings suggest that genetic
factors play a role in CAD [15], all published As in aortic or coronary dissections, CAD is the
genetic association studies on ICAD have been mere consequence of a wall hematoma, either
underpowered. More accurate data on genetic secondary to an intimal tear or to direct bleeding
risk factors for CAD can only be obtained from within the arterial wall caused by ruptured vasa
much larger multicenter genetic association stud- vasorum. Current insight into the pathogenesis of
ies, such as the ongoing genome-wide associa- coronary arteries and thoracic aorta dissection
tion study within the Cervical Artery Dissections suggests that dissecting aneurysms are disorders
and Ischemic Stroke Patients (CADISP) of the outer arterial layers, involving the vasa
consortium [17]. vasorum. Histological evidences of pathological
8 Carotid Artery Dissection 119

alterations of the tunica adventitia and the tunica temporal bone [29]. Since intracranial arteries
media suggest that CAD is due to an “outside-in” have no external elastic limitans and have a thin-
process starting in the outer arterial layers rather ner media and adventitia compared with cervical
than an “inside-out” injury starting from the endo- arteries, intracranial extension of a CAD has a
thelium leading to endothelial rupture as the pri- high risk of intracranial rupture.
mary event [27].
Regardless of its mechanism, the mural hema-
toma can expand toward the intima or the adven- Mural Hematoma Consequences
titia, resulting in a stenosis or pseudoaneurysm
formation. Mural blood then dissects longitudi- Dissections can be divided into two groups. The
nally. Dissections can tear through the intima, first group is mainly subintimal, and the dissec-
with the consequence for mural blood to enter tions compromise the arterial lumen or cause
the lumen of the artery. Increased thickness of thrombus to enter the lumen. The second group
the arterial wall by mural blood accumulation is subadventitial, and these dissections either per-
causes compression of the lumen, causes pertur- turb structures adjacent to the outer arterial wall.
bation of the blood flow stream and of the vascular Extracranial subintimal CAD causes symptoms
endothelium, and, finally, causes activation of primarily through luminal compromise and pres-
platelets and the coagulation cascade. These ence of luminal clot. Ischemic symptoms and
changes contribute to the formation of a luminal infarction are caused by reduced perfusion in the
thrombus. CAD may start from the luminal side at brain territory supplied by the artery or embolism.
the intimal surface and, through an intimal flap, Subadventitial cervical CAD can perturb the sym-
dissect into the media. The mural hematoma can pathetic fibers located along the dilated artery and
create a false lumen that might reconnect with the might compress or cause ischemia to the lower
true lumen, creating parallel circulatory channels, cranial nerves (IX–XII) that exit near the
separated by the intimal flap [28]. This false skull base.
lumen may either remain patent, resolve
completely, or thrombose and cause narrowing
of the true lumen. The mural hematoma some- Clinical Features
times extends between the media and the adven-
titia, leading to the formation of an aneurysmal The consequences of the mural hematoma are
lesion. local symptoms and signs including headache
and neck pain (supposedly attributable to a dis-
tension of the artery by the mural hematoma stim-
Mural Hematoma Location ulating pain-sensitive receptors), Horner’s
syndrome, or cranial-nerve palsies resulting from
The commonest location for a spontaneous CAD the stretching of sympathetic-nerve and cranial-
is the cervical segment 2–3 cm distal to the carotid nerve fibers by an enlarged carotid artery. The
bulb, an area that is probably subject to most of the mechanism by which CAD leads to cerebral or
stretch during extension or rotation of the neck. retinal ischemia is thought to be embolic, from
Although several studies have described the prox- intraluminal thrombi forming at the site of the
imal anatomical location of spontaneous CAD, intimal tear. Hemodynamic infarcts are also pos-
data on the cranial extension of the mural hema- sible. Occasionally, CAD can lead to subarach-
toma are scarce. One study reported that the mural noid hemorrhage when the dissection extends
hematoma extended in the cranial direction, intracranially.
involving the petrous segment of the ICA in The classical triad of presentation of a CAD is
nearly 75 % of CAD [29]. If CADs extend for a ipsilateral headache, facial or neck pain, and
variable length cranially, they do not usually past Horner’s syndrome, followed hours or days later
the point of entry of the ICA into the petrous by cerebral or retinal ischemia. This triad of
120 O. Naggara et al.

symptoms is found in less than one third of attributable to other factors. Although sometimes
patients, but the presence of two elements of this inaugural, it is most often preceded by local symp-
triad strongly suggests the diagnosis [1, 5]. toms that started shortly before the ischemic
event, a finding that is highly suggestive of CAD.
Cerebral infarction and TIA are inaugural in
Local Symptoms and Signs respectively 10–15 % and 10–20 % of cases
[30]. Middle cerebral artery territory is involved
The following symptoms and signs can occur in more than 80 % of cases [30]. In rare instances,
separately or in combination: [2, 19, 30] Horner’s carotid artery dissection can (MCA) lead to retinal
syndrome, unusual neck pain or headache, ischemia [19, 32].
cranial-nerve palsy, tinnitus, and, rarely, The literature provides support for several con-
cervical-root injury. Horner’s syndrome and cepts, regarding the primary cervical lesion and
cranial-nerve palsy occur in carotid artery dissec- the secondary intracranial lesion in CAD. An
tions. Sudden-onset Horner’s syndrome, particu- autopsy case has provided the only direct demon-
larly if associated with headache or neck pain or stration of secondary intracranial emboli
with an ipsilateral ischemic stroke in the carotid [33]. Three studies described indirect signs of
territory, can be considered to be specific to CAD emboli on transcranial Doppler monitoring stud-
and should lead to urgent imaging of the cervical ies [34–36], but the sample size of these studies
arteries. limits the significance of these results. According
The characteristics of pain associated with to current concepts relating to mechanism and
CAD are not specific and can sometimes mimic stroke pattern, strokes in pial or perforating artery
migraine or even cluster headache. CAD with territory are more likely to be embolic, whereas
isolated pain might be more common than border-zone infarcts are more likely to be hemo-
expected [31] and is more often caused by extra- dynamic. Using this simplification scheme, sev-
cranial vertebral artery dissection but can also be eral authors discussed the mechanism of
caused by carotid artery dissections. The onset infarction in CAD patients, with conflicting
type ranged from thunderclap headache to results [37, 38]. Based on the stroke pattern on
progressive pain. CT, one group demonstrated an embolic mecha-
Cranial-nerve palsies are rare, representing less nism in 92.2 % of 65 patients [39], whereas others
than 7 % of CAD cases in large hospital-based reported a similar frequency of embolic and
series. The hypoglossal nerve is the most com- hemodynamic infarcts in 11 patients
monly affected, followed by the CN IX and CN X [38]. Recently, the occlusive intracranial throm-
cranial nerves, which are topographically close to bus in vivo was directly demonstrated using T2*
the carotid artery in its cervical trajectory. The sequence. Other findings that substantiate the con-
most likely mechanism is compression of the cept of artery-to-artery embolism in the pathogen-
nerves by an enlarged carotid artery. Cranial- esis of stroke in CAD were the multiple acute
nerve ischemia is another potential mechanism, DWI lesions in the majority of patients and the
particularly in very rare cases of upper cranial- occurrence of pial artery or perforating artery ter-
nerve palsy. ritory stroke in 96 % of patients [40–43].
If the simplification of the relationship between
the mechanism and stroke pattern provides an
Ischemic Symptoms easy approach to the presumed mechanisms of
cerebral infarction in most patients, one should
Stroke (transient ischemic attack [TIA] or cerebral bear in mind that, even at the individual level,
infarction) is the most frequent and severe mani- both mechanisms may be at play and may rein-
festation of extracranial CAD. The clinical pre- force each other in causing the ischemic lesion.
sentation of cerebral ischemia caused by CAD Border-zone infarcts might result from mixed
does not differ from that of cerebral ischemia mechanisms by impaired clearance of emboli in
8 Carotid Artery Dissection 121

hypoperfused regions. Indeed, during the consti- accounted for in published series. The rate of
tution of the mural hematoma, only the embolic ischemic recurrences ranges from 0 % to 13,3 %
mechanism may explain the occurrence of an at 1 year, with the largest recurrence rates
infarction, whereas the hemodynamic mechanism observed when recurrent events that occurred
may appear progressively with the narrowing of before the diagnosis of CAD are taken into
the lumen due to the growth of the mural hema- account [19, 44]. Recurrent ischemic events usu-
toma. Once the lumen is severely stenosed, hemo- ally occur during the first weeks after the dissec-
dynamic impairment may encourage the tion. Factors associated with an increased risk of
formation of secondary clots in border-zone recurrent ischemic events are multiple dissections
regions, impede the clearance of distal clots, and and a history of hypertension [19]. Although iso-
increase the impact on cerebral tissues. The fact lated cases of ischemic strokes caused by chronic
that border-zone infarction rarely occurred in iso- dissecting aneurysms of the carotid artery have
lation but mainly occurred in association with pial been reported, two prospective series of aneurys-
or perforating artery territory infarction supports mal CAD found no ischemic events after about
the hypothesis of an impaired clearance of small 3 years of follow-up [44]. Similar rates of ische-
emboli [40]. mic recurrences have been reported in 46 patients
with CAD with a transient stenosis or occlusion
and in 46 patients with CAD with a permanent
Outcome stenosis or occlusion [45]. However, in another
series of 130 consecutive patients with CAD,
Clinical Outcome ischemic recurrences were attributed to a worsen-
Mortality rates in the acute phase of CAD are ing carotid stenosis in five of six cases [46].
generally low (<5 %) [2, 19], although higher
rates of up to 23 % have been reported in previous Arterial Lesion Evolution
series of subsets of patients with severe CAD. In The proportion of patients with complete resolu-
general, mortality after CAD might be tion of arterial abnormalities varies between stud-
underestimated, as patients with severe forms of ies: 46 % for stenoses, 33 % for occlusions, and
CAD sometimes die before arterial imaging and 12 % for dissecting aneurysms in the general
etiological diagnosis can be done. In support of population [46–48]. Complete resolution or stable
this possibility, data from studies on patients with residual luminal irregularity was documented
acute complete MCA infarction have suggested after a median duration of 0,29 years and in
that CAD could be a major cause of “malignant” 82 % of cases within the first year [49]. The like-
cerebral infarction. Functional outcome is good in lihood of complete recanalization seems higher in
about three-quarters of patients with CAD, but patients with CAD who present with only local
impact on quality of life and socioprofessional symptoms and signs [50]. Occlusions can lead to
integration can be important [1, 32]. The func- residual stenoses, and residual aneurysms can
tional outcome after CAD does not seem to be appear after the acute phase in initially stenotic
better than in other types of ischemic strokes in or occluded arteries [44].
young individuals. Factors associated with a poor
functional outcome are presence of cerebral ische- Recurrences of Dissections
mia, arterial occlusion, carotid location, older age, Recurrences of dissections are defined by a new
and a severe National Institutes of Health stroke mural hematoma, remote the initial lesion. Early
scale score at onset. recurrences of dissection in the days or weeks
after the initial event could be a manifestation of
Recurrent Ischemic Events a unique transient disorder, whereas late recur-
Recurrent ischemic events seem to be rare, rences occurring several months or years later
although early recurrences (before the patient is could indicate an underlying connective tissue
discharged from the stroke unit) are not always weakness. Both early and late recurrences are
122 O. Naggara et al.

rare; they seem to be most frequent within the first since it does not allow direct visualization of the
2 months after the initial event [19], but some vessel wall, whenever the false lumen does not
dissections that were diagnosed as multiple opacify with contrast medium or when the lumen
might have occurred sequentially within a short contains thrombus. It has been replaced by non-
time frame. Late recurrences are possibly invasive imaging modalities, both for the primary
underestimated as studies with long-term follow- diagnosis and the follow-up of dissections.
up are scarce [51]. Recurrent dissections were not Pathognomonic signs are double lumen and inti-
reported in the only population-based series with mal flap. These are detected in less than 10 % of
follow-up data of 48 patients with CAD (mean cases [52], while the following patterns are found
duration of follow-up: 7–8 years). In hospital- more commonly. The string sign, the angiographic
based series, the rate of recurrent dissections hallmark of CAD, is a long, tapered, usually eccen-
ranged between 0,6 % and 25,0 %. The recurrence tric, and irregular stenosis that begins beyond the
rate was probably overestimated in tertiary refer- ICA bulb and might extend to the skull base
ral series. Risk factors for recurrent CAD are [52, 53]. The “string and pearl” sign corresponds
younger age [18], a familial history of CAD, to a focal narrowing with a distal dilatation. The
vascular Ehlers-Danlos syndrome, and flame-shaped tapering of the lumen, a tapered
fibromuscular dysplasia [32]. The prognosis of occlusion that spares the carotid bulb, is suggestive
recurrent CAD has been described as benign [51]. of dissection. Although these patterns are indicative
of dissection, they are not specific. DSA findings of
spontaneous CAD may also include a dissecting
Imaging aneurysm, alone or associated with a tapered steno-
sis [52, 53], with changing lumen patterns over
Dissection of the craniocervical arteries, which time, often returning to a normal caliber.
was once considered as an uncommon diagnosis,
has become increasingly recognized as a cause of
stroke in young and middle-aged patients. In part, Ultrasound
this increased recognition is due to increased use
of noninvasive imaging studies. The latter has Noninvasive imaging is required for the detection
allowed the screening of larger numbers of of possible CAD. Ultrasound imaging provides
patients. In some populations, the rate of dissec- direct visualization of pathological findings that
tion of the craniocervical arteries has been diag- may be related to CAD and demonstrates
nosed at rates 3–10 times greater than that impaired lumen patency [54]. Pathognomonic
determined before the use of MRI, MR angiogra- findings on gray-scale US include mural hema-
phy (MRA), and, more recently, CT angiography toma, an intimal flap, or coexistence of a true and
(CTA) studies. a false lumen (Fig. 1) [55]. Unlike DSA, US is
able to demonstrate a false lumen even if this
lumen is thrombosed.
Digital Subtraction Angiography Spectral Doppler US waveforms in CAD can
be normal or may demonstrate nonspecific fea-
In the past, the diagnosis of arterial dissection tures also occurring in high-grade stenosis and
relied on standard catheter angiography (DSA). occlusions (damped spectral waveform with
As some authors have pointed out, DSA is an lower amplitude and biphasic pattern, high-
imperfect reference standard because of question- resistance spectral waveform, or absence of flow
able interobserver variability. Indeed, a number of with no spectral Doppler waveform) [54].
entities can mimic arterial dissection and DSA is The diagnostic sensitivity of US decreased if
an invasive procedure. In addition, if DSA may CAD results in a low-grade stenosis [54].
detect vessel wall irregularities, such as Sturzenegger et al. showed high accuracy in
fibromuscular dysplasia, it may miss dissections, detecting a dissection with US in case of ICA
8 Carotid Artery Dissection 123

Fig. 1 Longitudinal color


(a, b) and power (c)
Doppler ultrasound image
demonstrating a
hypoechoic thickened wall,
a finding consistent with a
mural hematoma (a)
narrowing and displacing
the true lumen (b) of the
ICA with enlargement of its
external diameter to 7 mm
(normal 5 mm). An
abnormal high-resistance
spectral Doppler US
waveform is demonstrated
in the dissected lumen (d)

occlusion or high-grade stenosis but a 20 % sen- Magnetic Resonance Imaging


sitivity in low-grade stenosis [49]. Another limi- Dissections of the craniocervical arteries are best
tation is that US is less reliable for CAD outside assessed with cross-sectional fat-suppressed T1
the “window” of the US examination such as sequence with cranial and caudal saturation pulses
dissections located in the subpetrous segment and with MRA techniques. The former can pro-
and the carotid canal or in case of small mural vide direct visualization of the mural hematoma,
hematoma [54, 55]. which is the pathological hallmark of a dissection,
and the latter allows noninvasive visualization of
arteries.
Cross-Sectional Imaging: CT or MRI?
Lumen Imaging: MR Angiography
MRI, including MR angiography (MRA) and high- Contrast-enhanced MRA (CE-MRA) is the tech-
resolution MRI (HR-MRI), and CT angiography nique of choice, thanks to recent improvements in
(CTA) are the main noninvasive cross-sectional MR gradients and software, which allow rapid
imaging tools used for assessing CAD. The use of acquisitions with centric k-space filling and auto-
MRI and CTA is twofold: (1) demonstration of a matic bolus triggering. Like DSA, CE-MRA can
mural hematoma and (2) evaluation of the conse- demonstrate luminal irregularity; long narrowing
quence of the mural hematoma on the lumen of the vessel, the so-called string sign; or occlu-
patency. MRI is currently the recommended imag- sion and pseudoaneurysm formation (Fig. 2).
ing technique [5], thanks to its higher ability to CE-MRA also has the advantage of being easily
demonstrate arterial wall abnormalities and its bet- preceded with fat-suppressed T1-WI axial images
ter specificity as compared with CTA. CTA can be through narrowed segments to confirm or refute a
performed as a first-line screening tool, particularly suspected dissection. The combination of cross-
in case of trauma. sectional MRI and MRA is not only a powerful
124 O. Naggara et al.

Fig. 2 Luminal patterns of carotid artery dissection on (arrow) that begins beyond the ICA bulb, with a distal
contrast-enhanced MR angiography (CE-MRA). (a, b) dilatation, the “string and pearl” sign. (c) Occlusive pattern
Stenotic type of CAD. In (a) CE-MRA demonstrates a of CAD. Flame-shaped tapering followed by an occlusion
long, tapered, eccentric, and irregular stenosis (arrows) of the lumen that spares the carotid bulb. (d)
that begins beyond the ICA bulb and that extends to the Pseudoaneurysmal pattern of CAD (arrow) associated
skull base, the “string” sign. In (b) focal eccentric stenosis with a distal long-tapered stenosis (double arrows)

diagnostic modality but can also be used effec- hematoma after 6 months, as it becomes isointense
tively for noninvasive follow-up of CAD to mon- with the surrounding soft tissue. These patterns are,
itor lumen patency evolution, resolution of a however, not specific for a dissection as fresh
mural hematoma, or development of complica- thrombus due to an atheromatous or embolic occlu-
tions (Figs. 3, 4, and 5). sion that may exhibit a similar evolution of MR
signal intensity. It may, indeed, be impossible to
Mural Hematoma Imaging distinguish an intraluminal clot from an occlusive
Carotid artery dissection is characterized, on mural hematoma. In such cases, a crescentic thick-
fat-suppressed T1 sequence, by a narrowed eccen- ening with eccentric signal void proximal to com-
tric flow void, which is surrounded by a crescent- plete occlusion should suggest dissection rather
shaped, hyperintense area expanding the vessel than atheromatous occlusion (Fig. 9).
diameter (Fig. 6) [53]. MR signal of the mural At least four causes of false-positive MRI or
hematoma has a similar temporal evolution than MRA findings have been reported [57].
intracerebral counterpart. [56] An acute mural Atheromatous plaques causing subtotal occlu-
hematoma can be hypointense on T2- and T1-WI sion can be hyperintense on T2-WI images and
images and therefore be difficult to distinguish mimic a CAD [57]. This rim is, however, typically
from an area of flow void. The mural hematoma isointense on T1-WI images, whereas the
may therefore be missed on MRI within the first periarterial rim in subacute CAD is usually
48 h after CAD [53]. Thereafter, the hematoma hyperintense on T1-WI images [56].
becomes of intermediate signal intensity on Structures adjacent to arteries can simulate a
T1-WI images and hyperintense on proton density periarterial rim of hyperintense signal which can
and T2-WI images (Fig. 7). After a few days, the mimic a mural hematoma or a pseudoaneurysm.
hematoma remains hyperintense on T1-WI images For instance, carotid arteries are surrounded
for approximately 2 months (Figs. 7 and 8). Sub- by fat. Systematic fat-saturation imaging is the
sequent loss of signal intensity can mask the best solution.
8 Carotid Artery Dissection 125

Fig. 3 Evolution of a pseudoaneurysm type of carotid the stenosis associated with a progression of the
artery dissection. The pseudoaneurysm (arrow) was initially pseudoaneurysm 2 months after onset of the CAD (b).
associated with a focal-tapered stenosis (a). Resolution of Progressive normalization seen at 6 and 12 months (c, d)

Fig. 4 Evolution of a stenotic type of carotid artery dissection. The long, irregular stenosis (arrow) improves with
progressive normalization seen at 12 months

Turbulent or slow flow on MRI and MRA can can produce hyperintense slow flow distally,
be another confounding factor for CAD by pro- which can be manifested by loss of part or all of
ducing hyperintense signal in the periphery of the the intracranial flow void. This signal can be mis-
arterial flow void, along the surface of the arterial taken for mural hematoma. Inflow phenomena
lumen. For example, marked narrowing of the generally produce signal abnormalities that are
extracranial segment of the internal carotid artery located centrally within the flow void rather than
126 O. Naggara et al.

Fig. 5 Twelve-month evolution of a pseudoaneurysm type of carotid artery dissection with true and false lumen
remaining patent

Fig. 6 Right carotid artery


dissection on contrast-
enhanced MR angiography
(CE-MRA) (a) and axial
fat-suppressed T1-WI
image (b). Carotid artery
dissection is characterized,
on fat-suppressed T1-WI by
a narrowed eccentric flow
void, which is surrounded
by a crescent-shaped,
hyperintense mural
hematoma expanding the
outer vessel diameter

peripherally. Homogeneity of the hyperintense mistaken for dissection. The recognition that the
signal on all slices associated with vessel expan- image is obtained at the extreme of the imaging
sion supports dissection rather than slow flow. volume (i.e., first or second image in a stack of
Flow-related enhancement in arteries and veins images) and correlation with the signal of the
can simulate mural hematoma. Bright signal in arteries on other imaging sequences resolve the
arteries at the extreme of an imaging volume due issue.
to entry of unsaturated spins (i.e., the so-called A false-negative diagnosis of dissection on
entry slice phenomenon) has long been recog- MR is most likely at the very early stage, when
nized as a cause of abnormal arterial signal on the mural hematoma is not yet hyperintense.
selected images. This phenomenon can simulate Indeed, at this stage, the blood products within
mural abnormal signal or complete absence of the the hematoma have not yet progressed to the stage
expected flow void within an artery and be of methemoglobin. In this case, the signal
8 Carotid Artery Dissection 127

Fig. 7 Temporal evolution


of MR signal of the mural
hematoma on T1 and T2
sequences. An acute mural
hematoma, e.g., within
2 days after onset, is
isointense on T2- and
T1-WI images. The
hematoma becomes of
intermediate signal
intensity on T1-WI images
and hyperintense on T2-WI
images after day 2. The
hematoma remains
hyperintense on both T2-
and T1-WI images for
approximately 2 months.
Subsequent loss of signal
intensity can mask the
hematoma after 6 months,
as it becomes isointense
with the surrounding soft
tissue (not shown)

Fig. 8 Temporal evolution of MR signal of the mural isointense to the surrounding tissue with bright heteroge-
hematoma on T1 sequence. Contrast-enhanced MR angi- neous signal of its peripheral part (b). It becomes homo-
ography (a) demonstrated a long irregular stenosis sparing geneously hyperintense at day 3 (c) and was no longer
the right ICA bulb. The mural hematoma was initially visible on fat-suppressed T1-WI image at day 60 (d)
128 O. Naggara et al.

Fig. 9 Occlusive cases of


carotid artery dissection. In
the first patient (a, b),
CE-MRA demonstrated
right ICA occlusion and
long stenosis of the left
ICA. On fat-suppressed
T1-WI image (b), it was
difficult in the right ICA
lumen to distinguish an
intraluminal clot from an
occlusive mural hematoma
(arrow). CAD diagnosis
was done, thanks to left ICA
mural hematoma (double
arrows). In the second
patient (c, d), CE-MRA
demonstrated left ICA
occlusion (arrow). On
fat-suppressed T1-WI
image (d), it was also
difficult distinguishing an
intraluminal clot from an
occlusive mural hematoma.
In such a case, the
crescentic hyperintense
thickening associated with
increase outer external
diameter of the occluded
ICA suggested dissection
rather than atheromatous
occlusion

intensity of the hematoma does not differ from The mural hematoma frequently extends cra-
that of background fat-suppressed tissue. Other nially to the petrous carotid segment in the case of
signs, such as the presence of a pseudoaneurysm, carotid artery dissection. This location is within
or an increase in external diameter may help in the limits of the field of view of standard brain
these difficult cases. MRI. Consequently, apart from dedicated cervical
In addition, whenever a CAD produces arterial fat-suppressed T1 sequences, brain MRI may be
occlusion (as opposed to stenosis), the mural useful to demonstrate CAD. More than three-
hematoma may not be conspicuous against the quarters of such acute CAD can be diagnosed
background of the hyperintense signal abnormal- using stroke brain MRI protocol only, including
ity of an occluded arterial lumen on DWI, FLAIR, T2* sequences, and time-of-flight
fat-suppressed T1-WI images. In such cases, MRA (Fig. 10) [29]. This result might have clin-
although the arterial occlusion is recognized, the ical implications for patients who are not initially
cause may not be discernible. suspected of having CAD based on available
8 Carotid Artery Dissection 129

Fig. 10 Illustration of carotid artery dissection on brain circle of Willis (d). Sagittal T1-WI imaging (e). Increased
MRI sequences in five patients. Axial gradient-recalled external diameter, crescentic mural thickening (arrows),
echo T2 (a), diffusion-weighted imaging (b), FLAIR (c), and eccentric lumen (arrows) were clearly seen in all
and native slices of 3D time-of-flight angiography of the patients

history at the time of protocol and at institutions visualization of the cervical arterial mural hema-
where cervical CE-MRA is not coupled with MR toma on both carotid and vertebral arteries [59].
brain imaging. The interpretation of direct signs of
CAD on brain MRI is a learnable skill and High-Resolution MRI Using Surface Coils
requires little specialized training and can also be at 1.5-T
achieved in regular clinical practice. The aim of MRI of the arterial wall of cervical
arteries is to obtain in plane, submillimetric
High-Resolution MRI voxels. The loss of signal to noise induced by
High-resolution magnetic resonance imaging reducing the voxel size (500  500 μm  2–3
(HR-MRI) is an attractive tool for the assessment mm) is compensated for by the use of phased-
of carotid artery diseases. Using dedicated surface array surface coils, which increases the signal-to-
radiofrequency coils at 1.5-T or standard coils at noise ratio by 40 %. The so-called HR-MRI cor-
3-T, it provides high-resolution images of the responds to images with submillimetric voxels
arterial wall in vivo. Optimal image quality relies that imply the use of dedicated surface coils.
on correct patient’s preparation, on adequate coil These coils are commercially available and com-
positioning, and on the use of pulse sequences patible with most MR manufacturers. They con-
with signal suppression of the flowing blood. sist of a pair of multichannel coils. The coils,
HR-MRI has initially been applied for the made of flexible material, are placed bilaterally
in vivo analysis of carotid atherosclerosis with around the neck. The carotids, being superficial
now solid evidence that HR-MRI can identify structures, are well suited for surface coil imaging.
the major components of atherosclerotic plaque, To explore the patient with suspected CAD,
i.e., lipid core, hemorrhage, calcifications, as well the radiologist should ensure that the coil is at the
as the fibrous cap [58]. Beyond atherosclerosis, level of the carotid-presumed lesion. A multicentric
HR-MRI can be used in case of suspicion of study reports a rate of 90 % of interpretable MRI
cervical artery dissection, providing an excellent exams performed for the purpose of carotid
130 O. Naggara et al.

bifurcation analysis. For a given magnetic field examination. The surface coils should be posi-
when using the same surface coil, image quality tioned anteriorly and laterally, facing the mandib-
seems stable across different platforms [60]. ular bone for the petrous portion of internal
The MR protocol comprises multiple carotid artery, whereas it should be placed poste-
sequences in the axial plane, covering about rior and under the mastoid for the V3 segment;
3 cm in height. Consequently, the slices must be (2) acquire an oblique scout view (3D phase con-
focused on the index lesion or artery. trast, 2D time of flight, 1 min) of the entire index
As in atherosclerotic plaque exploration, it is artery so as to visualize the plaque distribution and
currently recommended to combine multiple con- the level of the carotid lesion; (3) acquire HR
trasts to fully characterize potential mural hema- images (voxel <500  500 μm in plane); (4) use
toma. Pulse sequences designed for vascular multicontrasts, by combining black and bright
imaging are classed as “bright blood” or “black blood sequences, to identify the mural hematoma
blood,” depending upon the flowing blood signal. presence and age; (5) keep the total acquisition
Both of these sequences provide a good contrast time to less than 30 min. Indicative parameters at
between the lumen and the arterial wall. Bright 1.5-T are detailed in Table 1 [58].
blood sequence corresponds to 3D TOF gradient
echo sequences used for MR angiography. On this High-Resolution MRI at 3-T
sequence, the flowing blood signal is enhanced, Although most of the HR-MRI studies have been
and the lumen is brighter than the vessel’s wall. performed on 1.5-T MR units, 3-T units are now
The lack of 180 refocusing pulse creates T2*- more widely available. Surface coils for carotid
sensitive signal. The aim of black blood imaging are commercially available for 3-T MR
(BB) sequences is to cancel the signal of the units. These are slightly different from those used
flowing blood so as to increase the contrast on 1.5-T units (tuned to a different frequency and
between the lumen (in black) and the wall. For adapted to the 3-T connectors and/or interfacing).
that reason, the acquisition rate is synchronized to They provide significant improvement in signal-
the heartbeat and is preceded by a double- to-noise and contrast-to-noise ratios [61],
inversion-recovery (IR) magnetization preparation allowing reduction in both the acquisition time
pulse. This process maximizes flow suppression and the in-plane resolution. For a given acquisi-
due to outflow and minimizes artifacts due to vessel tion time, thinner slices or larger anatomic cover-
motion. The time of inversion (TI) for the double- age in the z-axis can be obtained. At 3-T, longer
IR preparatory pulse is fixed as close to the null repetition times are advised for PD- and T2-WI
point of the blood signal. The double-IR prepara- images. Double-IR blood suppression techniques
tion and rapid SE acquisition are typically repeated are available at 3-T, but they require an adjustment
using 2RR cardiac gating until all the rapid SE of the TI for the blood T1 at 3-T, i.e., slightly
shots are acquired. Black blood sequence can be T2 longer TI for 3-T compared to that used for
or proton density (PD) weighted (2RR) or T1-WI 1.5-T [61]. Demonstration of strong agreement
(1RR). By applying the double-IR preparation between 3 and 1.5-T HR-MRI of the carotid
just after the cardiac trigger, the slice-selective plaques, particularly in the identification of ath-
re-inversion pulse is placed in the “end-diastolic” erosclerotic plaque composition, including hem-
part of the cardiac cycle. Because the TI is usually orrhage, supports the translation of histologically
close to 650 ms, the rapid SE acquisition is also validated 1.5-T criteria to 3-T carotid images
played out during diastole. A third inversion pulse [62]. However, there are differences in the mea-
can be added for fat suppression. sured size of arterial wall hemorrhage measures,
Several principles should be followed when larger at 1.5-T than at 3-T, suggesting that patients
designing an MR protocol for imaging cervical enrolled in serial prospective trials of hematoma
arterial wall in CAD: (1) the coil positioning is of measurement in CAD should be imaged at the
importance and should be guided by lumen abnor- same field strength in order to minimize interstudy
malities seen on CE-MRA or ultrasonographic errors during analysis [62].
8 Carotid Artery Dissection 131

Table 1 Indicative imaging parameters for 1.5-T high-resolution plaque MR imaging using dedicated surface coils [60]
Parameters 3D TOF T1 BB DP BB T2 BB
TR (ms) 30 1 RR 2 RR 2 RR
TE (ms) 6.9 9 16–20 50
FOV (mm) 130 130 130 130
Slice thickness (mm) 2.2 3 3 3
Matrix 288  224 352  256 256  512 256  512
NEX 2 3 2 2
Synchronization – Cardiac Cardiac Cardiac
Number of slices 20 8 8 8
Resolution (μm)a 254  254 254  254 254  254 254  254
Scan time (min)b 4 4–5b 4–6b 4–6b
PD indicates proton density, TR repetition time, TE echo time, TI inversion time, NEX no. of excitations, BB black blood
sequence with double-inversion-recovery preparation pulse
a
After zero filling interpolation
b
Reported scan times are based on a heart rate of 70 beats/min. Times shorten with faster rates

Fig. 11 1.5-T high-


resolution MRI using
surface coils. Axial
fat-suppressed T1-WI
images showing the mural
hematoma (a) and an
intimal flap (b) creating two
parallel circulatory
channels. Although most
often seen under CTA, the
intimal flap with the double
lumen sign can be seen on
HR-MR images

High-Resolution MRI Findings in CAD excellent depiction of the structural characteristics


Using standard MRI, an early and reliable identi- of the vessel wall and the vessel lumen in acute
fication of acute CAD might be impaired by the CAD. The eccentric vessel lumen (occlusion, ste-
limited spatial resolution, the tortuous course of nosis, luminal thrombus) and the vessel wall
the arteries, the great variability in normal vessel (crescent mural hematoma, pseudoaneurysm,
caliber, and the presence of the thick bony cover- double lumen, intimal tear) of the dissected artery
ing and adjacent veins. This is the case for dissec- can then be exquisitely analyzed using HR-MRI
tion of the upper portions of the vertebral arteries (Fig. 11) [63–65]. The mural hematoma is well
(V2 and V3 segments), outside the field of this defined between intimal and adventitial wall
review, and for dissection of petrous ICA. In some boundary. The signal intensity of mural hematoma
cases, the diagnosis of CAD remains presumptive, varies with the time elapsed since the onset of
further confirmed by sequential follow-up MR symptoms [56]. More specifically, the age of the
showing signs of healing or progression in mural hematoma may be classified into acute,
response to treatment. early subacute, late subacute, and chronic hema-
1.5-T HR-MRI has demonstrated its usefulness tomas according to its signal intensities on the T1-
for the assessment of CAD [59] and permits an and T2-WI sequences. One study suggested that
132 O. Naggara et al.

the MR signal changes of mural hematoma had correspond to a localized inflammation of the
only moderate agreement with the dynamics seen arterial wall. Spontaneous CAD is more fre-
in cerebral hematomas, even if it can be difficult to quently associated with the presence of PAE com-
determine the exact delay between the occurrence pared to traumatic CAD [25]. Another study
of CAD and the HR-MRI scan. Initially, the mural demonstrated that a subset of patients with spon-
hematoma is isointense to the sternocleidomastoid taneous CAD showed signs of a generalized tran-
muscle on T1-WI and hypointense on T2 and TOF sient inflammatory arteriopathy in contrast-
imaging. At a mean delay of 5,8 days, mural enhanced HR-MRI and [18F]-
hematoma may appear hyperintense on all fluorodeoxyglucose positron emission tomogra-
sequences (Figs. 7 and 8). Progressively, the sig- phy CT (PET-CT) [26]. This subset of patients
nal drops on all pulse sequences [56]. may be more prone to multiple dissections. Inter-
HR-MRI can help in distinguishing between estingly, such HR-MRI pattern is similar to those
dissection and atherosclerosis in case of cervical observed in inflammatory diseases, such as
artery occlusion [66]. The signal of the mural Takayasu’s disease and giant cell arteritis. PAE
hematoma is usually homogeneous in CAD and inflammation could simply result from com-
while being heterogeneous in hemorrhagic ath- pression by the mural hematoma. However, such
erosclerotic plaques. In addition, unequivocal dis- HR-MRI pattern was more frequently found in
tinction between mural hematoma and thrombus spontaneous CAD and rarely observed in trau-
is allowed by HR-MRI. matic CAD despite a larger hematoma in this latter
Recent studies have shown the presence of group in one study [25]. This minimizes the like-
perivascular soft tissue signal changes, related to lihood of periarterial abnormalities simply
periarterial edema (PAE) on 3-T HR-MRI in resulting from a mechanical compression. How-
patients with recent CAD. PAE can be detected ever, we cannot exclude the possibility that
on a 1.5-T MR unit (Fig. 12), with a high preva- inflammatory HR-MRI pattern could be associ-
lence (83.3 %) of PAE in the spontaneous CAD ated with thrombogenicity after CAD rather than
group including 18 patients. These changes may with the CAD mechanism itself.
Because of the limited accessibility of HR-MRI
using dedicated surface coils and the long scanning
time inherent to the technique, the idea is not to
perform HR-MRI in all patients suspected of CAD
but rather to improve our imaging protocols to get
supportive imaging evidence in most patients. This
is already feasible using 3-T MR magnets with
standard multichannel head and neck coils. The
3-T’s increase signal-to-noise ratio can be targeted
at improving spatial resolution without the need of
dedicated surface coil. In daily practice, 3-T
fat-suppressed T1- and T2-WI sequences with
3 mm slice thickness challenge the high resolution
obtained using dedicated surface coil with 1.5-T
MR unit to image the cervical artery wall.
Recent technological innovations, including
parallel imaging, optimized k-space trajectories,
Fig. 12 1.5-T high-resolution MRI using surface coils. and variable flip angles, allow the acquisition, at
Periarterial edema in spontaneous carotid artery dissection.
High-resolution fat-suppressed T2-WI MR image. Note
3-T, of fat-suppressed tridimensional T1-WI
the signal changes in the soft tissues (arrows) surrounding sequences (3D T1), with neck and head coverage,
the right dissected ICA submillimetric voxels (spatial resolution of
8 Carotid Artery Dissection 133

Fig. 13 3-T fat-suppressed 3D FSE T1 sequence, with of contrast-enhanced MR angiography and 3D FSE T1
luminal black blood effect. Axial (a), coronal (b), and images on coronal (d, f) and axial (e) views: the
sagittal views (c) showing a bright T1 hematoma and its hyperintense mural hematoma is facing luminal
extension. Fusion images combining a volume rendering irregularities

0.8  0.8  1 mm [3] before and 0.4  0.4  submillimetric quasi isotropic voxels, allowing
0.5 mm [3] after interpolation), and acceptable scan for multiplanar reformations; (5) a large field of
duration for clinical practice (total scan time in view in the z-axis, allowing whole neck coverage.
between 5 and 7 min). In a preliminary study, it This potentially allows 3D T1 to be used as a first-
was demonstrated that fat-suppressed 3D T1 line diagnostic tool, when the CAD location is
sequence can demonstrate the mural hematoma unknown, as opposed to HR-MRI using dedicated
[64]. The 3D T1 sequence may offer several advan- surface coil sequences that are centered on the
tages over standard axial T1 sequence: (1) absence abnormal segment of the dissected artery because
of inflow artifacts, thanks to a 3D acquisition and to of a limited number of axial sections in clinically
an outer volume suppression pulse; (2) a compatible acquisition time (Fig. 13). Another
noncardiac gated black blood effect due to the group confirmed the added value of the 3D T1
presence of different intravoxel velocities in the sequence, compared to the fat-suppressed 2D T1
blood vessels, causing a phase dispersion, which sequence, in patients with acute or subacute carotid
results in signal loss. This phenomenon is ampli- or vertebral artery dissection using a standard
fied with the use of long echo trains and low multi-array surface coil at 1.5-T, with the limitation
refocusing flip angles; (3) homogeneous fat sup- of incomplete fat saturation in the lower cervical
pression; (4) a tridimensional acquisition with region [67].
134 O. Naggara et al.

CT Angiography present is from a systematic meta-analysis of non-


Multisection CT angiography (CTA) provides randomized studies that reported on outcomes
high-resolution and high-contrast images of the with antiplatelets versus anticoagulants [70].
arterial lumen and vessel wall. Axial source There was no significant difference in the rates
images provide excellent visualization of the of death or disability between both treatment
craniocervical arteries as a consequence of the groups. Thus, at present, empirical results are
contrast difference between the enhanced arterial used to choose between anticoagulants and
lumen and the surrounding structures. antiplatelet drugs, in a case-by-case-based deci-
Postprocessing of the source data as MIPs or sion. Anticoagulation is sometimes preferred over
curved multiplanar reformats MPR produces antiplatelet drugs when there is a severe stenosis,
images that closely resemble those obtained with an occlusion (with a risk of embolization before
DSA or MRA, and voxel-rendering techniques recanalization), or a pseudoaneurysm, provided
allow a 3D display of the vessels and surrounding that the infarct is not too large. Anticoagulants
soft tissues. CTA of CAD has shown excellent might also be preferred whenever a thrombus is
agreement with DSA [68]. A 100 % sensitivity seen in the arterial lumen or when the presence of
and specificity was achieved using the presence of multiple ischemic lesions in the same arterial terri-
a narrowed eccentric lumen in association with tory suggests an embolic mechanism. Conversely,
enlargement of the overall vessel diameter as a antiplatelet drugs are commonly favored when
criterion for acute carotid dissection. Other signs there is a general contraindication to anticoagulants
of extracranial carotid artery dissection on CT or when CAD is associated with a large infarct,
include stenosis, mural thickening, occlusion, exposing the individual to a high risk of hemor-
aneurysm formation, and thin annular contrast rhagic transformation. Whether antiplatelets
enhancement [69]. should be preferred in cases of local symptoms
Another advantage of CTA examinations over only is still a matter of debate. Antiplatelet drugs
MRI resides in identification of the rare but patho- might also be preferred to anticoagulants in the rare
gnomonic intimal flap. The higher sensitivity of case of intracranial extension of CAD that could
CTA is likely related to both the faster acquisition theorically lead to a subarachnoid hemorrhage.
time and higher spatial resolution. The information of evolution of the mural
hematoma during the first week after initiation of
the treatment may be important. Since CAD is
Treatment characterized by a mural accumulation of blood,
the dissecting hematoma may enlarge under
Medical Treatment anticoagulation, with subsequent lumen
narrowing. It was recently demonstrated, using
Anticoagulants or antiplatelets are usually HR-MRI, that heparin did not promote enlarge-
recommended in the acute phase of CAD to pre- ment of the mural hematoma, progression of lumi-
vent primary or recurrent stroke. However, no nal narrowing, or dissection recurrence much
randomized trial has successfully compared the more than aspirin [71].
efficacy of these treatments. The Cervical Artery The currently available observational studies
Dissection in Stroke Study (CADISS) is an ongo- suggest that thrombolytic therapy should not be
ing randomized multicentric prospective study withheld in patients in whom CAD is associated
comparing antiplatelet therapy with with an acute ischemic stroke. However, the lack
anticoagulation for patients with carotid and ver- of any trend toward a benefit of thrombolysis in
tebral artery dissection. Randomization must be such patients explains the research for more effi-
within 7 days of onset of symptoms. In the cient treatment options including mechanical
absence of randomized data, the best evidence at revascularization strategies.
8 Carotid Artery Dissection 135

Mechanical Revascularization with standard multichannel head and neck coils


Strategies and volumic fat-suppressed T1-WI sequence.

Recently, stent-supported angioplasty has been


used to treat CAD in case of neurological symp- References
toms while on anticoagulation or as an alternative
to the traditionally accepted use of anticoagulation 1. Schievink WI (2001) Spontaneous dissection of the
carotid and vertebral arteries. N Engl J Med
[72]. Stenting permitted resolution of the stenosis
344:898–906
with immediate recanalization of the artery. In a 2. Lee VH, Brown RD Jr, Mandrekar JN, Mokri B (2006)
recent systematic review of 140 patients Incidence and outcome of cervical artery dissection: a
(153 CAD), including traumatic (48 %, n = 64), population-based study. Neurology 67:1809–1812
3. von Babo M, De Marchis GM, Sarikaya H, Stapf C,
spontaneous (37 %, n = 49), and iatrogenic (16 %,
Buffon F, Fischer U, Heldner MR, Gralla J, Jung S,
n = 21) CAD, the technical success rate of Simonetti BG, Mattle HP, Baumgartner RW, Bousser
stenting was 99 %, and the procedural complica- MG, Arnold M (2013) Differences and similarities
tion rate was 1.3 %. At a mean angiographic between spontaneous dissections of the internal carotid
artery and the vertebral artery. Stroke 44:1537–1542
follow-up of 12.8 months, in-stent stenosis or
4. Touze E, Oppenheim C, Zuber M, Meary E, Meder JF,
occlusion was encountered in 2 % of patients, Mas JL (2003) Early asymptomatic recurrence of cer-
and at a mean clinical follow-up of 17.7 months, vical artery dissection: three cases. Neurology
neurological events were seen in 1.4 % of patients 61:572–574
5. Debette S, Leys D (2009) Cervical-artery dissections:
[73]. Further trials comparing stenting with med-
predisposing factors, diagnosis, and outcome. Lancet
ical management are needed to elucidate the role Neurol 8:668–678
of stenting in selected situations. 6. Majidi S, Hassan AE, Adil MM, Jadhav V, Qureshi AI
(2014) Incidence and outcome of vertebral artery dis-
section in trauma setting: analysis of national trauma
data base. Neurocrit Care 21(2):253–258
Summary 7. Engelter ST, Grond-Ginsbach C, Metso TM, Metso AJ,
Kloss M, Debette S, Leys D, Grau A, Dallongeville J,
An early and reliable identification of CAD is of Bodenant M, Samson Y, Caso V, Pezzini A, Bonati LH,
Thijs V, Gensicke H, Martin JJ, Bersano A, Touze E,
importance in order to prevent ischemic brain
Tatlisumak T, Lyrer PA, Brandt T, Cervical Artery D,
lesion. The diagnosis of carotid artery dissection Ischemic Stroke Patients Study G (2013) Cervical
is currently based on (1) a suggestive clinical artery dissection: trauma and other potential mechani-
presentation, (2) exclusion of atherosclerosis, cal trigger events. Neurology 80:1950–1957
8. Renard D, Azakri S, Arquizan C, Swinnen B,
and (3) supportive radiologic evidence. Obtaining
Labauge P, Thijs V (2013) Styloid and hyoid bone
radiological evidence of CAD may be a real chal- proximity is a risk factor for cervical carotid artery
lenge. The emergence of high-resolution, rapid dissection. Stroke 44:2475–2479
imaging methods has enabled MRI to noninva- 9. Raser JM, Mullen MT, Kasner SE, Cucchiara BL,
Messe SR (2011) Cervical carotid artery dissection is
sively image the fine internal structure of the
associated with styloid process length. Neurology
cervical arterial walls. HR-MRI can be used in 77:2061–2066
case of suspicion of cervical artery dissection, 10. Grau AJ, Brandt T, Buggle F, Orberk E, Mytilineos J,
providing an excellent visualization of the cervi- Werle E, Conradt, Krause M, Winter R, Hacke W
(1999) Association of cervical artery dissection with
cal arterial mural hematoma. Because of the lim-
recent infection. Arch Neurol 56:851–856
ited accessibility of 1.5-T HR-MRI and the long 11. Guillon B, Berthet K, Benslamia L, Bertrand M,
scanning time inherent to the technique, the idea is Bousser MG, Tzourio C (2003) Infection and the risk
not to perform HR-MRI in all patients with CAD of spontaneous cervical artery dissection: a case-
control study. Stroke 34:e79–e81
but rather to improve our imaging protocols to get
12. Grau AJ, Brandt T, Forsting M, Winter R, Hacke W
supportive imaging evidence in most patients. (1997) Infection-associated cervical artery dissection.
This is already feasible using 3-T MR magnets Three cases. Stroke 28:453–455
136 O. Naggara et al.

13. Lindsberg PJ, Grau AJ (2003) Inflammation and infec- Nikolaou K, Bartenstein P, Reiser M, Hacker M,
tions as risk factors for ischemic stroke. Stroke Dichgans M (2011) Vessel wall inflammation in spon-
34:2518–2532 taneous cervical artery dissection: a prospective, obser-
14. Rubinstein SM, Peerdeman SM, van Tulder MW, vational positron emission tomography, computed
Riphagen I, Haldeman S (2005) A systematic review tomography, and magnetic resonance imaging study.
of the risk factors for cervical artery dissection. Stroke Stroke 42:1563–1568
36:1575–1580 27. Volker W, Dittrich R, Grewe S, Nassenstein I, Csiba L,
15. Debette S, Markus HS (2009) The genetics of cervical Herczeg L, Borsay BA, Robenek H, Kuhlenbaumer G,
artery dissection: a systematic review. Stroke 40: Ringelstein EB (2011) The outer arterial wall layers are
e459–e466 primarily affected in spontaneous cervical artery dis-
16. Genius J, Dong-Si T, Grau AP, Lichy C (2005) section. Neurology 76:1463–1471
Postacute c-reactive protein levels are elevated in cer- 28. Caplan LR (2008) Dissections of brain-supplying
vical artery dissection. Stroke 36:e42–e44 arteries. Nat Clin Pract Neurol 4:34–42
17. Debette S, Metso TM, Pezzini A, Engelter ST, Leys D, 29. Naggara O, Soares F, Touze E, Roy D, Leclerc X,
Lyrer P, Metso AJ, Brandt T, Kloss M, Lichy C, Pruvo JP, Mas JL, Meder JF, Oppenheim C (2011) Is
Hausser I, Touze E, Markus HS, Abboud S, Caso V, it possible to recognize cervical artery dissection on
Bersano A, Grau A, Altintas A, Amouyel P, stroke brain mr imaging? A matched case-control
Tatlisumak T, Dallongeville J, Grond-Ginsbach C, study. AJNR Am J Neuroradiol 32:869–873
Group C (2009) Cadisp-genetics: an international pro- 30. Biousse V, D’Anglejan-Chatillon J, Touboul PJ,
ject searching for genetic risk factors of cervical artery Amarenco P, Bousser MG (1995) Time course of
dissections. Int J Stroke 4:224–230 symptoms in extracranial carotid artery dissections. A
18. Schievink WI, Mokri B, O’Fallon WM (1994) Recur- series of 80 patients. Stroke 26:235–239
rent spontaneous cervical-artery dissection. N Engl J 31. Biousse V, Woimant F, Amarenco P, Touboul PJ,
Med 330:393–397 Bousser MG (1992) Pain as the only manifestation of
19. Touze E, Gauvrit JY, Moulin T, Meder JF, Bracard S, internal carotid artery dissection. Cephalalgia
Mas JL (2003) Risk of stroke and recurrent dissection 12:314–317
after a cervical artery dissection: a multicenter study. 32. Leys D, Bandu L, Henon H, Lucas C, Mounier-Vehier-
Neurology 61:1347–1351 F, Rondepierre P, Godefroy O (2002) Clinical outcome
20. Calvet D, Boutouyrie P, Touze E, Laloux B, Mas JL, in 287 consecutive young adults (15 to 45 years) with
Laurent S (2004) Increased stiffness of the carotid wall ischemic stroke. Neurology 59:26–33
material in patients with spontaneous cervical artery 33. Steinke W, Schwartz A, Hennerici M (1996) Topogra-
dissection. Stroke 35:2078–2082 phy of cerebral infarction associated with carotid artery
21. Baumgartner RW, Lienhardt B, Mosso M, Gandjour J, dissection. J Neurol 243:323–328
Michael N, Georgiadis D (2007) Spontaneous and 34. Molina CA, Alvarez-Sabin J, Schonewille W,
endothelial-independent vasodilation are impaired in Montaner J, Rovira A, Abilleira S, Codina A (2000)
patients with spontaneous carotid dissection: a case- Cerebral microembolism in acute spontaneous internal
control study. Stroke 38:405–406 carotid artery dissection. Neurology 55:1738–1740
22. Brandt T, Orberk E, Weber R, Werner I, Busse O, 35. Srinivasan J, Newell DW, Sturzenegger M, Mayberg
Muller BT, Wigger F, Grau A, Grond-Ginsbach C, MR, Winn HR (1996) Transcranial doppler in the eval-
Hausser I (2001) Pathogenesis of cervical artery dis- uation of internal carotid artery dissection. Stroke
sections: association with connective tissue abnormal- 27:1226–1230
ities. Neurology 57:24–30 36. Droste DW, Junker K, Stogbauer F, Lowens S,
23. Volker W, Besselmann M, Dittrich R, Nabavi D, Besselmann M, Braun B, Ringelstein EB (2001) Clin-
Konrad C, Dziewas R, Evers S, Grewe S, Kramer SC, ically silent circulating microemboli in 20 patients with
Bachmann R, Stogbauer F, Ringelstein EB, carotid or vertebral artery dissection. Cerebrovasc Dis
Kuhlenbaumer G (2005) Generalized arteriopathy in 12:181–185
patients with cervical artery dissection. Neurology 37. Bonati LH, Wetzel SG, Gandjour J, Baumgartner RW,
64:1508–1513 Lyrer PA, Engelter ST (2008) Diffusion-weighted
24. Mawet J, Boukobza M, Franc J, Sarov M, Arnold M, imaging in stroke attributable to internal carotid artery
Bousser MG, Ducros A (2013) Reversible cerebral dissection: the significance of vessel patency. Stroke
vasoconstriction syndrome and cervical artery dissec- 39:483–485
tion in 20 patients. Neurology 81:821–824 38. Weiller C, Mullges W, Ringelstein EB, Buell U, Reiche
25. Naggara O, Touze E, Marsico R, Leclerc X, Nguyen T, W (1991) Patterns of brain infarctions in internal
Mas JL, Pruvo JP, Meder JF, Oppenheim C (2009) carotid artery dissections. Neurosurg Rev 14:111–113
High-resolution mr imaging of periarterial edema asso- 39. Lucas C, Moulin T, Deplanque D, Tatu L, Chavot D
ciated with biological inflammation in spontaneous (1998) Stroke patterns of internal carotid artery dissec-
carotid dissection. Eur Radiol 19(9):2255–2260 tion in 40 patients. Stroke 29:2646–2648
26. Pfefferkorn T, Saam T, Rominger A, Habs M, Gerdes 40. Morel A, Naggara O, Touze E, Raymond J, Mas JL,
LA, Schmidt C, Cyran C, Straube A, Linn J, Meder JF, Oppenheim C (2012) Mechanism of
8 Carotid Artery Dissection 137

ischemic infarct in spontaneous cervical artery dissec- 54. Benninger DH, Georgiadis D, Gandjour J,
tion. Stroke 43:1354–1361 Baumgartner RW (2006) Accuracy of color duplex
41. Naggara O, Morel A, Touze E, Raymond J, Mas JL, ultrasound diagnosis of spontaneous carotid dissection
Meder JF, Oppenheim C (2012) Stroke occurrence and causing ischemia. Stroke 37:377–381
patterns are not influenced by the degree of stenosis in 55. Dittrich R, Dziewas R, Ritter MA, Kloska SP,
cervical artery dissection. Stroke 43:1150–1152 Bachmann R, Nassenstein I, Kuhlenbaumer G,
42. Naggara O, Touze E, Seiller N, Gobin-Metteil MP, Mas Heindel W, Ringelstein EB, Nabavi DG (2006) Nega-
JL, Meder JF, Oppenheim C (2008) Asymmetry of tive ultrasound findings in patients with cervical artery
intracranial internal carotid artery on 3d tof mr angiog- dissection: negative ultrasound in cad. J Neurol
raphy: a sign of unilateral extracranial stenosis. Eur 253:424–433
Radiol 18:1038–1042 56. Habs M, Pfefferkorn T, Cyran CC, Grimm J,
43. Iancu-Gontard D, Oppenheim C, Touze E, Meary E, Rominger A, Hacker M, Opherk C, Reiser MF,
Zuber M, Mas JL, Fredy D, Meder JF (2003) Evalua- Nikolaou K, Saam T (2011) Age determination of
tion of hyperintense vessels on flair mri for the diagno- vessel wall hematoma in spontaneous cervical artery
sis of multiple intracerebral arterial stenoses. Stroke dissection: a multi-sequence 3t cardiovascular mag-
34:1886–1891 netic resonance study. J Cardiovasc Magn Reson 13:76
44. Touze E, Randoux B, Meary E, Arquizan C, Meder JF, 57. Provenzale JM, Sarikaya B, Hacein-Bey L, Wintermark
Mas JL (2001) Aneurysmal forms of cervical artery M (2011) Causes of misinterpretation of cross-sectional
dissection: associated factors and outcome. Stroke imaging studies for dissection of the craniocervical arter-
32:418–423 ies. AJR Am J Roentgenol 196:45–52
45. Kremer C, Mosso M, Georgiadis D, Stockli E, 58. Oppenheim C, Naggara O, Touze E, Lacour JC,
Benninger D, Arnold M, Baumgartner RW (2003) Schmitt E, Bonneville F, Crozier S, Guegan-
Carotid dissection with permanent and transient occlu- Massardier E, Gerardin E, Leclerc X, Neau JP,
sion or severe stenosis: long-term outcome. Neurology Sirol M, Toussaint JF, Mas JL, Meder JF (2009)
60:271–275 High-resolution mr imaging of the cervical arterial
46. Arauz A, Hoyos L, Espinoza C, Cantu C, wall: what the radiologist needs to know. Radio-
Barinagarrementeria F, Roman G (2006) Dissection graphics 29:1413–1431
of cervical arteries: long-term follow-up study of 59. Bachmann R, Nassenstein I, Kooijman H, Dittrich R,
130 consecutive cases. Cerebrovasc Dis 22:150–154 Kugel H, Niederstadt T, Kuhlenbaumer G, Ringelstein
47. Charbonneau F, Gauvrit JY, Touze E, Moulin T, EB, Kramer S, Heindel W (2006) Spontaneous acute
Bracard S, Leclerc X, Mas JL, Meder JF (2005) Diag- dissection of the internal carotid artery: high-resolution
nosis and follow-up of cervical arterial magnetic resonance imaging at 3.0 Tesla with a dedi-
dissections–results of the sfnv-sfnr study. J cated surface coil. Invest Radiol 41:105–111
Neuroradiol 32:255–257 60. Touze E, Toussaint JF, Coste J, Schmitt E,
48. Leclerc X, Lucas C, Godefroy O, Nicol L, Moretti A, Bonneville F, Vandermarcq P, Gauvrit JY, Douvrin F,
Leys D, Pruvo JP (1999) Preliminary experience using Meder JF, Mas JL, Oppenheim C (2007) Reproducibil-
contrast-enhanced mr angiography to assess vertebral ity of high-resolution mri for the identification and the
artery structure for the follow-up of suspected dissec- quantification of carotid atherosclerotic plaque compo-
tion. AJNR Am J Neuroradiol 20:1482–1490 nents: consequences for prognosis studies and thera-
49. Sturzenegger M, Mattle HP, Rivoir A, Baumgartner peutic trials. Stroke 38:1812–1819
RW (1995) Ultrasound findings in carotid artery dis- 61. Yarnykh VL, Terashima M, Hayes CE, Shimakawa A,
section: analysis of 43 patients. Neurology 45:691–698 Takaya N, Nguyen PK, Brittain JH, McConnell MV,
50. Nedeltchev K, Bickel S, Arnold M, Sarikaya H, Yuan C (2006) Multicontrast black-blood mri of
Georgiadis D, Sturzenegger M, Mattle HP, carotid arteries: comparison between 1.5 and 3 Tesla
Baumgartner RW (2009) R2-recanalization of sponta- magnetic field strengths. J Magn Reson Imaging
neous carotid artery dissection. Stroke 40:499–504 23:691–698
51. Dittrich R, Nassenstein I, Bachmann R, Maintz D, 62. Underhill HR, Yarnykh VL, Hatsukami TS, Wang J,
Nabavi DG, Heindel W, Kuhlenbaumer G, Ringelstein Balu N, Hayes CE, Oikawa M, Yu W, Xu D, Chu B,
EB (2007) Polyarterial clustered recurrence of cervical Wyman BT, Polissar NL, Yuan C (2008) Carotid
artery dissection seems to be the rule. Neurology plaque morphology and composition: initial compari-
69:180–186 son between 1.5- and 3.0-t magnetic field strengths.
52. Houser OW, Mokri B, Sundt TM Jr, Baker HL Jr, Radiology 248:550–560
Reese DF (1984) Spontaneous cervical cephalic arte- 63. Naggara O, Louillet F, Touze E, Roy D, Leclerc X, Mas
rial dissection and its residuum: angiographic spec- JL, Pruvo JP, Meder JF, Oppenheim C (2010) Added
trum. AJNR Am J Neuroradiol 5:27–34 value of high-resolution mr imaging in the diagnosis of
53. Provenzale JM, Morgenlander JC, Gress D (1996) vertebral artery dissection. AJNR Am J Neuroradiol
Spontaneous vertebral dissection: clinical, conven- 31:1707–1712
tional angiographic, ct, and mr findings. J Comput 64. Edjlali M, Roca P, Rabrait C, Naggara O, Oppenheim C
Assist Tomogr 20:185–193 (2012) 3d fast spin-echo t1 black-blood imaging for the
138 O. Naggara et al.

diagnosis of cervical artery dissection. AJNR Am J imaging of cervical artery dissection. AJNR Am J
Neuroradiol 34(9):E103–E106 Neuroradiol 29:1753–1760
65. Naggara O, Oppenheim C, Toussaint JF, Calvet D, 70. Kennedy F, Lanfranconi S, Hicks C, Reid J,
Touze E, Mas JL, Meder JF (2007) Asymptomatic Gompertz P, Price C, Kerry S, Norris J, Markus HS,
spontaneous acute vertebral artery dissection: diagno- Investigators C (2012) Antiplatelets vs anticoagulation
sis by high-resolution magnetic resonance images with for dissection: cadiss nonrandomized arm and meta-
a dedicated surface coil. Eur Radiol 17:2434–2435 analysis. Neurology 79:686–689
66. Bachmann R, Nassenstein I, Kooijman H, Dittrich R, 71. Machet A, Fonseca AC, Oppenheim C, Touze E,
Stehling C, Kugel H, Niederstadt T, Kuhlenbaumer G, Meder JF, Mas JL, Naggara O (2013) Does
Ringelstein EB, Kramer S, Heindel W (2007) High- anticoagulation promote mural hematoma growth or
resolution magnetic resonance imaging (mri) at 3.0 delayed occlusion in spontaneous cervical artery dis-
Tesla in the short-term follow-up of patients with proven sections? Cerebrovasc Dis 35:175–181
cervical artery dissection. Invest Radiol 42:460–466 72. Lavallee PC, Mazighi M, Saint-Maurice JP,
67. Cuvinciuc V, Viallon M, Momjian-Mayor I, Sztajzel R, Meseguer E, Abboud H, Klein IF, Houdart E,
Pereira VM, Lovblad KO, Vargas MI (2013) 3d Amarenco P (2007) Stent-assisted endovascular throm-
fat-saturated t1 space sequence for the diagnosis of bolysis versus intravenous thrombolysis in internal
cervical artery dissection. Neuroradiology 55:595–602 carotid artery dissection with tandem internal carotid
68. Leclerc X, Godefroy O, Salhi A, Lucas C, Leys D, and middle cerebral artery occlusion. Stroke
Pruvo JP (1996) Helical ct for the diagnosis of extra- 38:2270–2274
cranial internal carotid artery dissection. Stroke 73. Pham MH, Rahme RJ, Arnaout O, Hurley MC, Bern-
27:461–466 stein RA, Batjer HH, Bendok BR (2011) Endovascular
69. Vertinsky AT, Schwartz NE, Fischbein NJ, stenting of extracranial carotid and vertebral artery
Rosenberg J, Albers GW, Zaharchuk G (2008) Com- dissections: a systematic review of the literature. Neu-
parison of multidetector ct angiography and mr rosurgery 68:856–866, discussion 866
Nonatherosclerotic Nondissection
Diseases of Carotid Artery 9
Santosh Kumar Kannath and T. R. Kapilamoorthy

Contents Carotid Aneurysms and Carotid Blowout . . . . . . . . 154


Carotid Aneurysm . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140 Carotid Blowout Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Fibromuscular Dysplasia (FMD) . . . . . . . . . . . . . . . . . . 140 Miscellaneous . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156
Natural History and Clinical Features . . . . . . . . . . . . . . . 140 Carotidynia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156
Pathology and Imaging-Pathologic Correlation . . . . . 141 Eagle Syndrome or Stylohyoid Syndrome . . . . . . . . . . 157
Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158
Takayasu Arteritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
Giant Cell Arteritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158
Other Uncommon Vasculitis . . . . . . . . . . . . . . . . . . . . . . . 145
HIV Vasculopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
Connective Tissue Diseases . . . . . . . . . . . . . . . . . . . . . . . . 147
Arterial Tortuosity Syndrome . . . . . . . . . . . . . . . . . . . . . . . . 147
Ehler-Danlos Syndrome Type 4 (EDS4) . . . . . . . . . . . . 147
Loeys Dietz Syndrome (LDS) . . . . . . . . . . . . . . . . . . . . . . . 148
Pseudoxanthoma Elasticum (PXE) . . . . . . . . . . . . . . . . . . 150
Marfan Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
Other Systemic Disease with Carotid
Involvement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
Neurofibromatosis Type 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . 150
PHACE(S) Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151
Tuberous Sclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153
Behcet Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154

S.K. Kannath (*)


Neurointervention Center, Department of Imaging
Sciences and Interventional Radiology, Neurointervention
Center, Sree Chitra Institute of Medical Sciences and
Technology, Trivandrum, Kerala, India
e-mail: santhoshkannath@sctimst.ac.in;
santhoshkannath@rediffmail.com
T.R. Kapilamoorthy
Department of Imaging Sciences and Interventional
Radiology, Sree Chitra Institute of Medical Sciences and
Technology, Trivandrum, Kerala, India
e-mail: kapil@sctimst.ac.in

# Springer Science+Business Media New York 2016 139


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_38
140 S.K. Kannath and T.R. Kapilamoorthy

Abstract
Fibromuscular Dysplasia (FMD)
Nonatherosclerotic and nondissection diseases
Fibromuscular dysplasia is an idiopathic, segmen-
of carotid are often unrecognized clinical
tal, nonatherosclerotic, and noninflammatory dis-
entity, primarily due to nonspecific imaging
ease involving the small- and medium-sized
findings and unavailability of diagnostic tests.
arteries, commonly affecting renal and carotid
This entity comprises of wide spectrum of clin-
arteries. The exact prevalence of the disease is
ical conditions such as nonspecific arteritis,
unknown. The prevalence of the symptomatic
vasculitis, connective tissue diseases, infective
disease based on earlier angiographic studies is
processes, and some acquired conditions.
estimated to be between 0.3 and 3 %, though
Oftentimes, carotid involvement may be the
obviously this could not be extrapolated to general
first and only manifestation of these diseases.
population due to the evident bias and its actual
Ancillary clinical and examination findings
incidence may even be much lower [1–3].
lend clue to the underlying disease process
and help the clinician to further investigate
into the specific pathology. The list is exhaus-
Natural History and Clinical Features
tive; however, some of the important disease
entities and their clinical implications are
The etiology of FMD is not known, and various
reviewed in this chapter.
factors such as familial predisposition, hormonal
influence, smoking, and mechanical forces are
Keywords attributed to the causation of FMD. FMD com-
Carotid aneurysms • Carotid stenosis • monly affects the truncal portion of the internal
Takayasu arteritis • Fibromuscular dysplasia • carotid artery which is prone for mechanical
Giant cell arteritis • Vasculitis • Connective stretching and shearing over the cervical verte-
tissue disorders • Marfan syndrome • Ehler- brae. As this segment relatively lacks vasa
Danlos syndrome • Neurofibromatosis • vasorum, an ischemic cause has also been attrib-
Arterial tortuosity syndrome • Carotidynia • uted. A congenital theory presumes that the arte-
Eagle syndrome rial lesions are persistent embryonic intimal
cushions that normally appear in the main trunk
during fetal life. The fact that proximal cervical
Introduction ICA is formed from dorsal aortic arch lends cre-
dence to this theory [1, 4].
Though atherosclerotic manifestation of the carotid Cervicocranial FMD presents at an older age,
artery is a more common and important vascular though classical FMD is commonly discovered in
pathology in middle and elderly patients, nonather- younger and female population [4]. Carotid and
osclerotic vascular diseases assume prominence in vertebral arteries are nearly as frequently affected
younger population and remain an important cause as renal arteries. FMD may be detected inciden-
for stroke in this age group. Nonatherosclerotic tally or may have ischemic or hemorrhagic pre-
diseases may remain asymptomatic and are inci- sentation. Stroke or transient ischemic attacks
dentally detected or may present with catastrophic (TIA) can occur due to thromboembolic phenom-
events. Due to its variable age of presentation, it ena, dissection, or hemodynamic compromise.
may be confused with atherosclerotic pathologies Spontaneous carotid artery dissections are a
if the latter presents prematurely. Thus understand- known complication, and FMD changes may be
ing of this group of pathologies is vital to design identified in up to 15 % of spontaneous dissec-
appropriate investigations for optimal therapeutic tions [5]. Recent registry data showed incidence
management. In this chapter, some of the important of carotid dissections and aneurysms to be 75 %
nonatherosclerotic and nondissection diseases of and 21 %, respectively. Besides, vertebral
carotid artery are reviewed. artery dissections are also not uncommon [6].
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 141

The incidence of cerebral aneurysms is reported to the distal carotid artery sparing the carotid
be 20–30 % in FMD, higher than the general bulb. These lesions are stable and do not pro-
population. Subarachnoid hemorrhage is a gress beyond 40 years.
dreaded complication of aneurismal rupture. 3. Perimedial fibroplasias: Occurs in 10 %.
Besides, rarer manifestations such as intracerebral Lesions have a collagen collar of variable
hemorrhage, carotidocavernous fistula or thickness in the outer half or the media or at
vertebrovenous fistulas are also described. Rarely, media-adventitia junction. It can be localized
FMD can involve intracranial arteries such as or diffused. Producing a corrugated appear-
middle cerebral artery, basilar artery, and distal ance, the caliber of outpouching does not
carotid artery in isolation or as extension of cervi- exceed the diameter of the normal proximal
cal FMD. Intracranial FMDs are observed in pedi- artery.
atric population and usually have fatal outcome 4. Medial hyperplasia : Occurs in 1–2 % . There is
[1]. In some follow-up studies, the risk of recur- smooth muscle hyperplasia without fibrosis.
rent TIAs or stroke is reported up to 5 % per year, Beads are less numerous than other subtypes.
and hence it is prudent to keep such patients on 5. Periarterial fibroplasia: Fibrous thickening of
long strict clinical surveillance. Natural history of adventitia with cicatrix formation causes local/
incidental intracranial aneurysms in FMD is diffuse area of narrowing in the artery . This is a
unknown [1, 7]. very rare type occurring in <1 % of cases.

Traditionally, angiography has been used to


Pathology and Imaging-Pathologic diagnose FMD. The frequently affected segments
Correlation include mid and distal parts of ICA and vertebral
artery at C1 and C2 levels. Bilateral involvement
Three types of FMDs are identified histologically is common, and carotid and vertebral artery
depending on the arterial layer involvement. The pathologies may coexist. The most common
angiographic appearance can be correlated with angiographic abnormality is the appearance of a
histological features; however, categorization string of beads which is a characteristic of FMD
into different subtypes is successful only in few and usually suggests subtype medial FMD
entities. The various pathological subtypes (Fig. 1). Fine bristling of lumen may be noted
and its angiographic appearances are described indicating the presence of microaneurysms.
below. Standing columns of spastic contractions are the
differential diagnosis. Tubular stenosis accounts
1. Intimal fibroplasias: Occurs in less than 10 %. for 20 % of angiographic observations which may
There is circumferential thickening of intima be focal or diffuse. It is not related to specific
by collagenous tissue. It is usually seen in subtypes and can be confused with other vascular
children and young adults with no gender pre- diseases such as atherosclerosis or vasculitis.
dilection. The lesions are localized or a long Rarely, a weblike defect at the origin of internal
segment; however, this feature is not specific to carotid artery is also described. Other rare atypical
this entity. manifestations include beading of external carotid
2. Medial hyperplasia: Occurs in 60–70 %. There branches, diverticular outpouching of carotid
are areas of thickened fibrotic ridges alternate artery [1, 2, 4, 8].
with thinned media producing a beaded Intracranial FMD is described in conjunction
appearance due to alternate hyperplasic change with carotid involvement or as isolated findings.
and local aneurysm formation. The beads are Angiography may show a typical string of bead
aneurismal outpouchings that exceed the nor- appearance or multiple aneurysms and arterial
mal proximal caliber of the artery, and constric- tortuosities. Intima is as commonly affected as
tions represent fibrotic ridges. The appearance media, and most cases are reported in pediatric
is a diagnostic of FMD and usually occurs in population [2].
142 S.K. Kannath and T.R. Kapilamoorthy

Fig. 1 Diagnostic angiogram of bilateral common carotid artery (a, b). MRI revealed bilateral parieto-occipital
artery in a 3-year-old boy reveals tapering and occlusion of region acute infarcts (not shown). Volumetric reconstruc-
the right internal carotid artery and typical arterial beading tion shows moderate focal stenosis of midsegment of the
and moderate stenosis of proximal left internal carotid main renal artery (c)

Though typical angiographic appearance is and symptoms can significantly hamper the daily
confirmatory of FMD, the test is invasive, and activities in more than two thirds of TA
similar findings can be observed by other nonin- patients [9].
vasive studies such as US, CTA, or MRA. Distal The cerebral manifestations of TA include
carotid, vertebral artery, and intracranial circula- headache, dizziness, visual disturbances, stroke,
tion are better evaluated with CT or MRI, and in TIA, or rarely subarachnoid hemorrhage and
the current scenario, angiography is only intracerebral bleeds. The reported incidence of
performed if revascularization is planned. Besides these symptoms is quite variable with minor
diagnosis, imaging also helps in the surveillance symptoms noted in more than 50 % of patients
of intracranial aneurysms. and dreaded events such as TIA or stroke
observed in 10–35 % of patients [10, 11]. Ischemic
manifestations may be due to hemodynamic
Vasculitis insufficiency or thromboembolic phenomenon.
Intracranial stenosis is a rare important cause for
Takayasu Arteritis stroke in TA [12]. Subarachnoid hemorrhage sec-
ondary to aneurismal rupture is also rarely
Takayasu arteritis (TA) is a chronic inflammatory reported in TA. The aneurysms are common in
disease of large arteries affecting predominantly the posterior circulation, and persistent hyperten-
aorta, subclavian, carotid, renal, and pulmonary sion and altered hemodynamic factors are thought
arteries. Though reported from all over the world, to play a role in its genesis. Necrosis and destruc-
the disease is more prevalent in Asian population. tion of wall and elastic lamina are also reported;
The affected individuals are usually young and however, primary arteritis seems unlikely [13].
female, and since the initial symptoms are Vasa vasorum (VV) is assumed to play an
nonspecific, the time to diagnosis is often delayed important role in the pathogenesis of
by months to years. Initial inflammatory symp- TA. Inflammatory cells infiltrate the VV and incite
toms include anorexia, weight loss, malaise, fever, the chronic and granulomatous inflammatory
myalgia, arthralgia, or carotidynia. The disease reaction that spreads across to involve the media
shows progressive or relapsing remitting course and adventitia layer of the artery. The adventitia
and finally burns out. The clinical presentation is and media are thickened, and intimal layer shows
often related to the extent of compensatory collat- hyperplasia. The smooth muscles and elastin of
eral development in the affected vascular territory, media are destroyed, and the panarterial layer is
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 143

gradually replaced by diffuse or nodular fibrotic imaging modalities such as CT/MRI and
lesions resulting in the development of stenosis or US. The angiography is performed as a part of
eventual occlusion of the artery [9, 14, 15]. Exten- interventional procedure or to assess the hemody-
sive destruction followed by inadequate fibrosis namic status of cerebral circulation in patients
response leads to the formation of arterial aneu- presenting with TIA/stroke or hemorrhage.
rysms. The prognosis of TA has improved over the Since carotid artery is superficial, US can
last decade primarily due to earlier diagnosis, uti- detect many of these changes involving common
lization of imaging to assess disease progression, carotid artery and internal carotid artery such as
and advances in medical therapy [16]. The major stenosis, occlusion, or aneurysm formation.
morbidity or mortality is related to the develop- Besides, it can demonstrate the extraluminal arte-
ment of cardiac failure or stroke. Premature rial wall thickening and helps in the diagnosis as
atherosclerosis further adds to the increased cere- well as monitoring of further progression. It also
bral risks. Diagnostic criteria introduced by provides information regarding the direction of
Ishikawa and the American College of Rheuma- flow in carotid and vertebral artery, which is use-
tology are purely clinical and demonstrated more ful while planning therapy. Limitations include
than 90 % sensitivity and specificity for TA. inability to visualize the arteries in entirety.
European consortium for diagnosis of childhood Cross-sectional imaging such as MRI and CT
TA includes imaging cognates as well and had can demonstrate mural thickening of the arterial
reported very high sensitivity and specificity wall, extent of thickening, intraluminal thrombo-
nearing 100 % [9]. sis, steno-occlusive lesions, collateral pathways,
The gold standard imaging study for the eval- and aneurysm formation. MRI has the advantage
uation of TA has been a conventional angiogram, of superior tissue characterization and helps to
though recently, CT and MRI have been increas- delineate inflamed/fibrosed media and adventitia-
ingly employed to study the luminal and periadventitial regions and edematous intima [16].
extraluminal features with high degree of accu- Disease activity monitoring: Activity is best
racy. Typical changes described in cerebral angi- followed up by hematological parameters includ-
ography include: ing erythrocyte sedimentation rate and C-reactive
protein assessment. Recent reports suggest good
1. Irregularity of contour of artery: This is the correlation between the disease activity and the
earliest change seen and is mainly due to inti- delayed gadolinium enhancement of the wall of
mal fibrous thickening. the artery especially aorta. Thus MRI could
2. Stenosis caused by circumferential thickening become an important modality in assessing the
indicating advanced disease. disease progression of the patient. Caution needs
3. Complete occlusion due to thrombosis in a to be exercised; however, while interpretation as
stenosed segment or due to the disease process MRI lacks specificity, wall edema and enhance-
itself. Stenosis or occlusion often occurs at or ment may be observed in clinically dormant phase
close to the origin of the branches of aorta. as well [17, 18].
4. Diffuse dilatation or ectasias due to destruction The imaging studies show stenosis and/or
of media leading to irregular uniform widening occlusion of common carotid arteries, aneurysms
of carotid. of unilateral or bilateral carotid arteries,
5. Aneurysm saccular or fusiform : As there is a intraluminal thrombosis, and rarely dissection of
thick adventitial covering rupture of blood ves- the carotid artery. Extensive collateralization of
sel which is uncommon. external carotid artery through ascending cervical
6. Combination of lesions: Stenosis and and thyrocervical arteries may be visualized
aneurysms. reforming the internal carotid artery. Tortuous
vascular channels may be visualized within the
The angiograms are infrequently performed arterial wall which is in fact the vasa vasorum that
and are increasingly replaced by noninvasive develops secondary to ischemia of the arterial wall
144 S.K. Kannath and T.R. Kapilamoorthy

Fig. 2 This young lady presented with multiple events of b). Thin tortuous channels are seen along the course of the
acute cerebral ischemia. Aortogram demonstrates tapering right common carotid artery that reconstitutes antegrade
of brachiocephalic artery, occlusion of bilateral subclavian flow in the right internal carotid artery (c, d). MRI shows
artery, and moderate intermediate length smooth stenosis old infarct in the right corona radiate (e)
of proximal left common carotid artery (white arrows in a,

or cerebral parenchyma. Intracranial arteries may or may develop later [22, 23]. Ischemic cranial
show occlusion or arterial wall thickening symptoms include headache, scalp tenderness, jaw
(Fig. 2). Rarely nonaneurysmal SAH has been claudication, amaurosis fugax (AF), and partial or
reported [19–21]. complete visual loss. AF is an important premoni-
tory symptom portending worse visual outcomes if
not recognized and intervened at earliest. The
Giant Cell Arteritis visual loss occurs due to narrowing or occlusion
of posterior ciliary arteries and rarely due to central
Giant cell arteritis (GCA), also known as temporal retinal artery occlusion, posterior ischemic optic
arteritis or Horton disease, is the most common neuritis, or retrobulbar neuritis [24]. Cerebral
primary systemic vasculitis affecting people older ischemic events are rare and are reported to occur
than 50 years. The disease incidence increases in 2–4 % of GCA patients [22, 25]. Ischemic events
with age and shows mild female predominance. are common in posterior circulation and may pre-
Vasculitic involvement involves large- and sent as TIAs or stroke. Atypical presentation
medium-sized arteries and frequently affects includes the presence of nonspecific constitutional
cervicocranial arteries such as superficial tempo- symptoms, unexplained fever, claudication pain of
ral artery, posterior ciliary artery, and vertebral extremities due to large vessel GCA involvement,
artery. Aorta, proximal large aortic arteries, and carotid tenderness, or development of aortic aneu-
upper limb arteries are also involved in disease rysm, which in the absence of systemic biomarkers
process; however, intracranial arteries are not typ- can delay timely diagnosis and institution of appro-
ically affected by this disease [22]. priate management.
Clinical symptoms are variable and are related Pathological studies demonstrated infiltration
to developing ischemia or nonspecific inflamma- of all the three layers of arterial wall by chronic
tory processes. Polymyalgic symptoms may be inflammatory cells such as lymphocytes, macro-
seen in 30–50 % of GCA at the time of diagnosis phages, and giant cells. The internal elastic lamina
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 145

shows fragmentation, and smooth muscles are of aortic or large arterial wall and has been found
destroyed. Typical histological findings such as to be useful in the diagnosis of GCA in its early
the presence of giant cells at the transition of phase or when biopsy is negative. Also this
intima and media are found only in 50 % of the increased uptake in acute phase of GCA has
cases. The hyperplasic changes of intima narrow been correlated with the development of aortic
or obliterate the vascular lumen and contribute to dilatation at late follow-up [30].
the ischemic pathogenesis. However, these
changes are neither specific nor sensitive, and
often a false negative result may occur due to Other Uncommon Vasculitis
segmental nature of the disease [22].
Diagnosis of GCA is made as per the guide- HIV Vasculopathy
lines of the American College of Rheumatology
1990 classification criteria. Ultrasonography and Owing to improved survival rates among HIV
Doppler studies are valuable in the diagnosis of patients, the primary vascular pathology due to
GCA, though they are not commonly employed. HIV infection is increasingly being documented
Ultrasonography may show a dark hypoechoic in clinical practice. Young males are predomi-
circumferential arterial wall thickening termed as nantly affected, and often the patients who are
“halo sign” that disappears 2–3 weeks after the immunocompetent may manifest the disease. Vas-
initiation of steroid therapy. A recent meta- cular pathology may be aneurismal or occlusive in
analysis revealed a sensitivity of 69 % and spec- nature and involve extracranial carotid as well as
ificity of 91 % for unilateral halo sign. For bilat- intracranial arteries. Carotid involvement is noted
eral halo sign, the specificity approaches 100 %, in 25 % of all HIV-related aneurysms, and majority
and some investigators initiate therapy without of these aneurysms present with clinical features
confirmatory evidence from temporal artery such as painful enlarging mass lesions, hoarseness
biopsy. Besides halo sign, stenosis or occlusion of voice, dysphagia, or stridor [31, 32]. Intracranial
of temporal artery could be identified; however, aneurysms may be multiple large or giant fusiform
these findings are neither specific nor sensitive. aneurysms, reported usually in children or isolated
Ultrasonography is operator dependent, and exper- saccular aneurysms in adults [33]. Among the
tise, knowledge of temporal artery anatomy, and patients presenting with stroke, HIV vasculopathy
availability of high-resolution probes can affect the has been found to be the cause in 20 % of the
diagnostic accuracy of GCA [22, 26, 27]. A recent patients [34].
small study reported improved visualization of Exact pathogenesis in unclear and probably
carotid wall, and its vascularization and luminal involves multiple factors. Pathological studies
border by contrast enhanced ultrasonography had demonstrated infiltration of vasa vasorum by
[28]. High-resolution MRI and MRA with 1.5T neutrophils, macrophages, and plasma cells. The
and 3T may show inflammation of temporal artery resultant vasa vasoritis leads to ischemia and
visualized as rim of thickening and enhancement destruction of arterial wall and incites the devel-
of the arterial wall with central dark lumen due to opment of arterial aneurysms. There is fragmen-
spin flow (Fig. 3). The performance of MRI/MRA tation and loss of internal elastic lamina, thinning
was found to be comparable to USG and of outer arterial wall, and intimal fibrosis. Bland
Doppler [29]. thrombus occluding the lumen may also be dem-
Angiography is rarely performed these days onstrated. Opportunistic bacterial infection of
and is largely superseded by noninvasive modal- arterial wall is a distant possibility and should be
ities. MRA and CTA are useful in detecting extra- evaluated for if tissue specimen is available for
cranial vascular abnormalities of aorta or other analysis [32, 34–36].
large arteries (Fig. 4). FDG-PET study has the Vascular abnormalities are detected during rou-
potential to detect early features of inflammation tine surveillance or while evaluating specific
146 S.K. Kannath and T.R. Kapilamoorthy

Fig. 3 Biopsy has proven giant cell arteritis. Extensive thrombus is visualized in the proximal left common carotid
aortic and arch vessel wall thickening and severe luminal artery (arrow in panel c)
narrowing and irregularity are noted (a, b, d). Intraluminal

clinical manifestations. Ultrasound is widely better delineate these pathologies and are useful
available and can easily demonstrate pseudo- in detecting intracranial vascular abnormalities
aneurysms, stenosis, or occlusion of extracranial as well. Intracranial aneurysms are common in
carotid artery. Pseudoaneurysms are commonly the circle of Willis, first- or second-order
localized to distal common carotid artery or branches of MCA, ACA, or PCA. Besides diag-
carotid bifurcation, and it is visualized as blow- nosis, imaging is also useful in monitoring the
out defect in the arterial wall with turbulent color growth of the aneurysm and in assessing the
flow. The adjacent arterial wall shows thickening response to retroviral therapy as some of the
and hyperechoic spotting [37]. CT and MRI can lesions resolve with treatment [31, 34, 38].
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 147

Fig. 4 Giant cell arteritis. Fat saturated T1W contrast arterial wall is subtly appreciable in source images of TOF
enhanced MRI (a, b) shows soft tissue enhancement in MRA (c, d), highlighted in panel e (arrow). The thickened
the right temporal fossa (arrow in a) which is readily arterial wall shows contrast enhancement (f)
apparent when compared to opposite side. Thickening of

features include dysmorphic facies, skin laxity,


Connective Tissue Diseases hyperextensible joints, arachnodactyly, high
arched palate, and bilateral inguinal hernia
Arterial Tortuosity Syndrome which may mimic other CTDs such as Ehler-
Danlos or Marfan syndrome [39–41].
Arterial tortuosity syndrome is a rare autosomal
recessive connective tissue disorder of large- and
medium-sized arteries due to mutations of
SL2A10 gene encoding the glucose transporter Ehler-Danlos Syndrome Type 4 (EDS4)
receptor GLUT10. Impaired GLUT10 activity
leads to upregulation of TGF beta activity in the Also known as vascular EDS, EDS4 is an autoso-
arterial wall and results in poor elastogenesis and mal dominant CTD caused by heterogeneous
extracellular matrix formation. The hallmark mutation of COL3A1 gene located in the chromo-
findings include arterial tortuosity and elonga- some 2 and encodes type 3 collagen that predom-
tion, aneurysms, and dissections of large- and inates the walls of arteries and abdominal hollow
middle-sized arteries, focal coarctation of aorta, visceral organs. This entity is rare and constitutes
stenosis of great vessels of the arch, and pulmo- <10 % of all EDS patients. The arterials walls are
nary artery (Fig. 5). Associated extravascular fragile and thin and are easily susceptible to shear
148 S.K. Kannath and T.R. Kapilamoorthy

Fig. 5 Arterial tortuosity syndrome in a 3-month-old child. Arch, brachiocephalic artery, left common carotid artery, and
descending aorta are tortuous and elongated (a–c). The left brachial artery is small in caliber and appears quite tortuous (d)

stress injuries especially in large- and medium- vessel wall, the arteries are easily damaged while
sized arteries [42–45]. The four cardinal features obtaining arterial access or during the cerebral
that help in the diagnosis of EDS4 include dys- angiography. Catastrophic and life-threatening
morphic facial features, thin skin with visible complications are reported during the procedure,
subcutaneous veins, bruises, echymosis or hema- and hence invasive diagnostic tests are best
tomas, and features of fragility or rupture of artery, avoided unless performed as a part of interven-
digestive tract, or uterus. The typical features of tional procedure [48–50]. Imaging surveillance by
EDS such as skin and joint laxity are however CT/MRI or US is advocated to detect or monitor
uncommon in this entity. The diagnosis is con- the progress of vascular disease.
firmed by demonstration of deficient synthesis of
type 3 procollagen by cultured skin fibroblasts or
detection of mutation of COL3A1 gene. Loeys Dietz Syndrome (LDS)
Major catastrophic events such as visceral or
arterial rupture are uncommon in childhood; how- LDS is an autosomal dominant connective tissue
ever, nearly 80 % suffer from at least one such disease caused by the heterozygous mutation of
episode by 40 years. The average life expectancy genes encoding the receptors such as TGFBR1,
is around 50 years, and cause of death is due to TGFBR2, SMAD3, and TGFB2 that are involved
vascular complications in more than 90 % of the in the transformation growth factor signaling
patients [45, 46]. The carotid and intracranial pathway. Phenotypic features include aortic aneu-
events are reported to occur in about 15 % of rysms, arterial tortuosity, hypertelorism, bifid
patients. The typical complications include arte- uvula, or cleft palate. The aortic aneurysms are
rial dissection, carotidocavernous fistula, carotid frequently observed in LDS patients, and it
or intracranial aneurysms, and arterial rupture remains the major cause of morbidity and mortal-
(Figs. 6 and 7). The EDS4 remains an important ity [51, 52]. Besides, generalized arterial tortuos-
differential diagnosis for ischemic stroke in youn- ity, tortuosity of head and neck arteries,
ger patients [44, 47]. Due to inherent weakness of dissections, and intracranial aneurysms are also
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 149

Fig. 6 A middle-aged lady presenting with left carotico- proximal cavernous internal carotid artery (arrow in a).
cavernous fistula with clinical features of EDS. Dilatation Panel b shows direct carotico-cavernous fistula with sig-
of distal cervical internal carotid artery and old dissection nificant cortical venous reflux. Also note fusiform dilata-
sequelae are noted (Arrow in a, c). Note small aneurysm of tion of V4 segment of the left vertebral artery (arrow in d)

Fig. 7 Another patient


with EDS. Extensive
tortuosity and mild ectasias
of bilateral cervical
vertebral arteries are evident
150 S.K. Kannath and T.R. Kapilamoorthy

reported. The other neurological features include long-term structural stability, transfer and reorient
Chiari 1 malformation, craniosynostosis, and the stress load, stabilize the TGF beta binding
hydrocephalus. The aneurysms are large and proteins, and regulate its signaling. Fibrillin 1
symptomatic, reported to occur in 10 % of the mutation is not identified in one third of the
patients causing a mortality rate due to cerebral patients with marfanoid features, and instead,
hemorrhage of 7 % [51, 53]. Among the several mutations of TGFBR1 and TGFBR2 may be
factors that are taken into consideration in the detected. These patients are grouped as MS type
management of these aneurysms, the presence of 2, and ocular features in this subset are uncommon
arterial tortuosity cannot be overlooked as it sig- [58–60]. Revised Ghent criterion enables the
nificantly hampers the distal access of interven- establishment of MS based on cardinal features,
tional devices. Both neurosurgical clipping and family history, genetic screening tests, and
endovascular coiling have been reported for the weighted systemic scores of ancillary clinical
management of cerebral aneurysms [51, 53, 54]. findings [61].
Typical pathological features of large caliber
arteries include cystic medial necrosis, elastin
Pseudoxanthoma Elasticum (PXE) fragmentation, smooth muscle necrosis, and apo-
ptosis. Cystic medial necrosis is uncommon in
PXE is an autosomal recessive disease due to the intracranial arteries. A few pathological studies
mutation of ABCC6 gene on chromosome 16, the reported intimal hyperplasia, medial degenera-
defect of which results in the accumulation of tion, and internal elastic lamina fragmentation of
mineralized and fragmented elastic fibers in the intracranial arteries – findings that may predispose
skin, eyes, and arterial wall. The intima of the to the formation of aneurysms [62, 63]. The vas-
artery is thickened, calcified, and fragmented. cular findings of carotid arteries include arterial
The typical manifestations of the disease include tortuosity (similar to LDS), intracranial and extra-
skin laxity, angioid streaks in retina, and cardio- cranial aneurysms, and dissection. The isolated
vascular changes. The disease is rare and cerebral involvement of the neck and intracranial arteries
complications are reported in 10 % of the PXE is extremely uncommon, limited to small series or
patients. The neurological manifestations include case reports. Dissecting aneurysms secondary to
cerebral ischemia due to carotid or vertebral artery extension of aortic dissection may be more fre-
occlusion, carotid artery calcifications, and cervi- quent observation and may present with neurolog-
cal or intracranial aneurysms. Dissections and ical deficits. The saccular aneurysms are observed
association of rete mirabile of carotid artery are in intracranial circulation, while fusiform type is
also rarely reported [43, 55–57]. common in extracranial carotid arteries. The
patients may be asymptomatic or may present
with pulsatile swelling in the neck, TIA or strokes,
Marfan Syndrome or rarely subarachnoid hemorrhage [62, 64–66].

Marfan syndrome (MS) is a relatively common


connective tissue disease characterized by typical Other Systemic Disease with Carotid
cardiovascular, musculoskeletal, spinal menin- Involvement
geal, and ocular features. The disease is autosomal
dominant inherited; however, in one third of Neurofibromatosis Type 1
patients, new mutations are observed. The defect
lies in the synthesis of fibrillin 1, an extracellular Neurofibromatosis type 1 is a rare autosomal
matrix protein that forms one of the important dominant disease caused by the mutation of
components of elastic microfibril. The microfi- tumor suppressor gene located in the chromosome
brils support elastic membrane and provide 17 that encodes the protein neurofibromin.
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 151

Neurofibromin regulates cellular mitogenesis by pediatric population was found to be 2.5 % in a large
inhibiting the reticular activating system (ras) institutional-based study. Besides moyamoya pat-
pathways, and hence a loss function leads to tern, isolated stenosis, occlusions, or ectasias of
overexpression of mitogenic signals within the intracranial arteries are observed (Figs. 8 and
cell leading to cellular proliferation or differenti- 9) [75].
ation [67]. The clinical hallmarks of the disease Vertebral arteriovenous fistula is a well recog-
are café-au-lait spots, cutaneous neurofibromas, nized complication of neurofibromatosis with
bone changes, and iris hamartomas. The vascular fewer than 50 cases reported in the literature
involvement is uncommon and is reported to (Fig. 10). The etiology of arteriovenous fistula is
occur in 0.4–6.4 % of patients with NF1 not clearly understood. Spontaneous rupture
[68]. NF1 vasculopathy affects large-, medium-, predisposed by arterial wall thinning and fragility
and small-sized arteries. The arteries may be sec- to adjacent venous plexus is thought to be one of
ondary involved when its wall is weakened by the mechanisms. A congenital theory assumes
extrinsic compression of extravascular neurofi- preexisting abnormal congenital vascular commu-
bromas or due to the proliferation of Schwann nications within the arterial wall which develops
cell within the arterial layers. Recent studies dem- into the fistula. Clinical features of this condition
onstrated a marked intimal and smooth muscle include pulsatile mass lesion, radiculo-
cell proliferation, and this observation has been myelopathy, cranial neuropathy, or tinnitus. The
related to decreased expression of neurofibromin fistula may be found involving the upper or lower
in the vascular wall. It is unsure whether the cervical vertebral arteries or may involve one of
vascular changes are congenital or acquired as a the branches of the vertebral artery. The epidural
pathological response to injury [68–71]. The path- venous sacs are often large and demonstrate rapid
ophysiology of vascular abnormality is related to shunting. Flow reversal and arterial steal may
the amount of intimal proliferation, spindle cell often be identified, and sometimes, small commu-
proliferation in the media, and the arterial diame- nication channels open up when the main fistula is
ter. Hence, small-sized arteries are often occluded, occluded, and hence long-term surveillance is
whereas larger arteries show aneurismal changes advocated. Recent reports suggest good clinical
due to weakening of arterial wall [69–72]. outcome with various endovascular strategies
The NF1 vasculopathy is the second leading [76–78].
cause of death among the NF1 patients younger
than 40 years. The carotid, vertebral, or intracra-
nial arteries are affected in one fifth of the patients, PHACE(S) Syndrome
and vascular abnormality may be in the form of
arterial stenosis, aneurismal changes, or arteriove- PHACE(S) is an acronym for a neurocutaneous
nous malformations. The aneurismal changes are syndrome with salient features of Posterior fossa
the commonest pathology and show female pre- malformation, Facial hemangiomas, Arterial
ponderance and predilection to rupture spontane- anomalies, Coarctation of aorta and Cardiac
ously during pregnancy. Extracranial aneurysms defects, Eye anomalies, and ventral developmen-
are large and fusosaccular and may present as pul- tal anomalies such as Sternal clefts or
satile or enlarging neck mass or acute rupture. Intra- Supraumbilical raphe. The vascular lesions are
cranial aneurysms are relatively uncommon and common in PHACES, seen in up to 78 % of
may affect intradural ICA, middle cerebral, anterior patients. The abnormalities are either congenital
cerebral, or basilar arteries [68, 69, 71, 73, 74]. or progressive and are related to the development
The aneurysms are incidentally detected or present or phase of growth of cutaneous hemangioma.
with subarachnoid hemorrhage. Rarely coexistent The arterial anomalies are detected in the location
intracranial and extracranial aneurysms also may be of hemangioma, and this association is hypothe-
found. The incidence of cerebral vasculopathy in sized due to vascular expression factors released
152 S.K. Kannath and T.R. Kapilamoorthy

Fig. 8 A young patient with neurofibromatosis. MRI (a) supraclinoid internal carotid artery (d) and excessive tor-
shows gliosis and atrophy of the right frontal operculum. tuosity of the left superior and inferior divisions of middle
MR angiogram (b, c) shows occlusion of the right internal cerebral artery (e). Cervical and intracranial vertebral arter-
carotid artery and small caliber of the right middle cerebral ies are also tortuous (f)
artery. Diagnostic angiogram reveals occlusion of the right

by the hemangioma either during embryogenesis carotid-vertebral communications [79–81]. Pro-


or proliferative growth. The hemangiomas of face, gressive vasculopathy such as stenosis,
orbit, or scalp are associated with vascular moyamoya pattern, or occlusions is observed in
malformations of intracranial or carotid arteries, 40 % of the patients. These vascular findings are
whereas mandibular, neck, or chest lesions are observed in more than 90 % of the patients
related to cardiac, aortic, or supraaortic abnormal- presenting with clinical features of acute stroke
ities [79, 80]. [82]. Internal carotid arteries are commonly
The congenital vascular anomalies observed affected followed by middle cerebral, anterior
include arterial dysplasias, aberrant course or ori- cerebral, and vertebrobasilar arteries. Associated
gin, hypoplasia, or agenesis of different intracra- brain structural anomalies are evident in the brain
nial arteries, dolichoectasia, saccular intracranial in 41 % of the patients, more commonly in poste-
aneurysms, and presence of persistent embryonic rior fossa [81].
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 153

Fig. 9 Young girl with neurofibromatosis 1, presenting middle cerebral artery (a, c) with striate collaterals (b)
with acute stroke. Diagnostic angiogram of left internal suggestive of Moyamoya disease
carotid artery reveals occlusion of proximal segment of

Fig. 10 A middle aged


female with pulsatile mass
lesion in the neck. MRI (a)
show tortuous and dilated
left cervical vertebral artery
emptying into a large
pouch. This is confirmed in
the diagnostic angiogram
(b) She was diagnosed to
have neurofibromatosis 1

Tuberous Sclerosis mapped on chromosome 9 and chromosome


16, respectively. Besides typical hamartomatous
Tuberous sclerosis (TS) is an autosomal dominant lesions discovered in different organs, vascular
multisystem disorder caused by the mutation of involvement is extremely rare and is more com-
tumor suppressor genes, TSC1 and TSC2, monly observed in the aorta. To date, only three
154 S.K. Kannath and T.R. Kapilamoorthy

extracranial carotid aneurysms and 19 intracranial mass or neurological symptoms such as TIA or
aneurysms are reported in the literature. Intracra- stroke. Indeed, luminal thrombus is noted in one
nial aneurysms are more often reported with the fifth of such lesions. Detailed discussion of this
most commonly affected being the internal carotid entity is dealt with elsewhere. Other causes of true
artery. The aneurysms are giant and fusiform and aneurysms are rare and are described in the pre-
may affect multiple arteries. Vascular wall disin- ceding sections. Pseudoaneurysms are relatively
tegration due to accumulation of mucopolysac- uncommon and accounts for 14 % of all extracra-
charides is posited in its genesis [83–85]. nial carotid aneurysms. The most common cause
of such aneurysm includes blunt or penetrating
trauma and previous carotid surgeries. Direct pen-
Behcet Disease etrating injury by sharp objects, shrapnels, or iat-
rogenic carotid puncture either intentionally or
Behcet disease (BD) is a multisystem inflamma- while central venous access can injure the vessel
tory disease of unknown etiology that presents wall and produce localized or expanding hema-
with typical clinical triad of uveitis and oral and toma and pseudoaneurysm. Blunt injuries damage
genital ulcers. Vascular involvement is observed the arterial wall, when the shear or rotary forces
in 7–29 % of patients, and venous systems are compress and stretch the arterial wall against the
predominantly affected. Arterial involvement is cervical vertebra or skull base and promote aneu-
noted in 3–5 % of the patients, and aneurismal rysm formation. Also the dissecting process of
rupture remains to be the most common cause of carotid artery splits the internal elastic lamina
mortality in patients with BD. Pathomechanism of and muscular media and weakens the wall which
aneurysm formation is thought to be due to vasa may eventually turn aneurismal in one third of
vasoritis and vasculitis of the arterial wall. Carotid dissection [92, 93]. The isolated aneurysms of
artery involvement is very rare and may affect the external carotid artery are very rare [94].
common carotid or internal carotid artery. The Untreated extracranial aneurysms carry poor
patient may present with enlarging pulsatile prognosis with high mortality [93, 95]. Besides
lump in the neck or subarachnoid hemorrhage central neurological features, aneurysms may
[86–88]. Intradural aneurysms are rarer and bulge into throat or compress adjacent structures
show no segmental predilection. Multiple aneu- such as the esophagus and lower cranial nerves.
rysms may be found, and these patients show The most feared complication is aneurysm expan-
tendency to develop aneurysms at different loca- sion and rupture resulting in a massive exsangui-
tions on follow-up [89, 90]. nation and fatal outcome. Initial diagnostic
evaluation is done with ultrasonography and
Doppler which confirm the clinical diagnosis,
Carotid Aneurysms and Carotid delineate the aneurysm boundary and extent, and
Blowout demonstrate intraluminal thrombosis. Larger
aneurysms, aneurysms close to skull base and
Carotid Aneurysm presence of calcifications hinder detailed analysis.
However, it is a useful tool in following the
Aneurysms of extracranial carotid artery are growth of the aneurysm in susceptible individuals
uncommonly observed and constitute about 4 % or in patients on conservative therapy. CT angiog-
of all peripheral aneurysms and less than 5 % of all raphy is currently the investigation of choice for
carotid surgeries [91]. Carotid aneurysms are cat- the evaluation of cervical aneurysms. CTA delin-
egorized into true aneurysms or pseudoaneurysms eates the extent and boundary of the aneurysm, its
depending on whether the layers of arterial wall relation to other vascular and airway structures,
are intact or not. Atherosclerosis remains the most presence of thrombosis or soft tissue hematoma,
common cause of true aneurysm, seen in elderly and status of intracranial vasculature. Besides
hypertensives, and presents with pulsatile neck diagnosis, it provides wealth of information
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 155

Fig. 11 Carotid aneurysms in two patients (a, b, and c, respectively) with no identifiable etiology. Luminal irregularity
and thrombosis with calcifications are present (not shown)

influencing the choice of treatment as well as over of characteristic signal abnormalities of hematoma
anticipated difficulties. Access vessel tortuosities, helps to clinch the diagnosis. Due to rarity of the
presence of kinks, thrombosis, and disrupted cir- disease, the role of CT and MRI as a comprehen-
cle of Willis are some of the factors considered sive imaging study is not explored, and the current
while planning the therapeutic approach [96, 97]. practice is dependent on local institutional protocol
Rare involvement of external carotid artery or and availability of resources. Conventional angiog-
concomitant intra-extracranial disease can also raphy is an invasive modality, usually performed as
be detected by CT angiography. Moreover, a part of interventional procedure (Fig. 11) [100].
craniocervical CTA as part of whole-body CT
protocol in trauma patients enables early recogni-
tion of vascular abnormalities and allows institu- Carotid Blowout Syndrome
tion of appropriate therapy at the earliest [98, 99].
Disadvantages such as contrast overload and radi- The carotid blowout syndrome (CBS) is a life-
ation concerns are largely superseded by the over- threatening emergency caused by the rupture of
all benefits. Potential pitfalls include overlooking the carotid artery or its branches due to the weak-
subtle imaging features such as vessel wall irreg- ening of adventitial layer. The important factors
ularity, spasm, or small peripheral arterial aneu- predisposing CBS include trauma, prior neck irra-
rysms especially when images are degraded by diation for head and neck malignancies, or radical
swallowing artifacts. neck surgeries. Radiation increases the risk by
MRI and MRA are complementary to CT in the 7.6-fold and can occur anytime between 2 and
diagnosis of these aneurysms. MRI is useful in the 20 years after the therapy. Radiation induces
diagnosis and characterization of luminal thrombus inflammatory changes, fibrosis, and ischemia of
and delineation of extent of the aneurysm and adventitia and results in the weakening of arterial
status of adjacent soft tissue structures. Besides wall. The patient may be clinically silent or may
distinct advantage of absent ionizing radiation, present with acute massive exsanguination. The
CT scores over MRI in the imaging workup of latter often carries high morbidity and mortality.
carotid aneurysms due to its easy availability and Despite successful treatment by endovascular par-
reduced scanning time except in situations of clin- ent artery occlusion or stent graft placement,
ical dilemma where CT is inconclusive and strong recurrent CBS and procedural or postprocedural
suspicion of pseudoaneurysm exists. Identification complications are reported to occur in more than
156 S.K. Kannath and T.R. Kapilamoorthy

one fourth of the patients, and overall prognosis is [106]. In the appropriate clinical setting, this
poor due to natural progression of the disease or imaging appearance should raise the suspicion
treatment-related issues [101, 102]. of CBS.
Clinically, CBS is categorized into three Angiogram is performed as a diagnostic pro-
groups – acute, impending, and threatened. cedure prior to intervention or when patients pre-
Acute CBS presents with massive hemorrhage sent with acute CBS. Angiography may
that is not self-limiting and cannot be controlled demonstrate acute contrast extravasation,
by packing or pressure. The artery ruptures freely pseudoaneurysm formation, focal luminal
into the communicating wound or into the airway. narrowing, or luminal irregularity. Findings may
Without immediate resuscitation and treatment, involve common carotid, internal carotid, or
the patient succumbs to rapid exsanguination. external carotid artery branches and may involve
The impending CBS bleeds intermittently through more than one site too. In threatened CBS, the
the oral cavity or surgical wound indicating a angiographic findings are often normal [101, 103,
weakened or ruptured artery with pseudo- 105]. Diffuse blush in the soft tissue suggests
aneurysm formation. Without further treatment, inflammatory or telangiectatic changes secondary
full-blown arterial rupture may ensue anytime. to radiation and merits treatment if patient is
Exposed artery as a result of wound infection, symptomatic.
flap necrosis, or tissue breakdown suggests inev-
itable rupture without any treatment, and it is
termed as threatened CBS [103]. Miscellaneous
Recent study has shown that CTA is very use-
ful in identifying the patients presenting with the Carotidynia
clinical features of impending CBS [104] The
CTA findings include extravasation of contrast, The idiopathic carotidynia (CD) is characterized by
pseudoaneurysms, recurrence of tumor, necrosis, unilateral neck pain and tenderness over the carotid
and exposed main or branches of carotid arteries. artery bifurcation. Due to the diagnostic confusion
The pseudoaneurysm and extravasation may be with other diseases that have similar clinical symp-
observed in the common carotid artery, internal toms, carotidynia was removed by the International
carotid artery, or external carotid artery branches. Headache Society from the 2004 classification of
Petrous ICA is a common location of aneurysm in headache disorders [107, 108]. However, there are
patients who have received radiotherapy for naso- many isolated reports that describe radiological and
pharyngeal carcinoma. The artery is defined as structural changes of the carotid wall, thus empha-
exposed if necrosis contacts more than half of its sizing its existence as a unique entity possibly
circumference. These findings showed good cor- induced by inflammation. The symptoms of
relation with angiographic features and have carotidynia include unilateral neck pain, radiating
value in prognosticating the outcome following to face and ear, aggravated by lateral head move-
intervention. The patients with normal CTA find- ment, swallowing, or chewing. The disease is self-
ings and impending CBS had better clinical out- limited and may be preceded by flu-like symptoms.
come as compared to the group of patients who CD commonly affects young and middle-aged
demonstrated these findings. Disadvantages of individuals and women. Imaging shows the
CTA include its inapplicability in acute CBS, absence of structural vascular and nonvascular
decreased sensitivity in detecting small causes, though many reports suggest specific imag-
pseudoaneurysms, or contrast extravasations ing appearances in carotidynia [109–115]. Acute
[101, 104, 105]. Imaging appearance of CBS in phase reactants such as C-reactive protein (CRP)
MRI is also reported. Pseudoaneurysm was visu- and serum amyloid antigen (SAA) are reported to
alized as enhancing lesion close to the carotid correlate with disease activity [115, 116]. A gamut
artery that appeared isointense on T1-WI of differential diagnosis need to be ruled out before
sequences and hyperintense on T2-WI sequences confirming the diagnosis of CD.
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 157

Pathological study was reported only in one bone, forms stylohyoid complex (SHC), and a
report where a patient with classical carotidynia reduced distensibility of this complex with sub-
was found to have a thickened carotid adventitia sequent irritation of neurovascular structures is
intraoperatively and histological analysis implicated in the pathogenesis of Eagle syn-
revealed adventitial infiltration of lymphocytes, drome (ES) [118]. The embryological develop-
vascular and fibroblasts proliferation, and early ment of SHC is controversial and is thought to
fibrosis – picture suggesting chronic inflamma- differentiate from Reichert cartilage which is the
tory process [112]. Noninvasive imaging modal- cartilage of the second branchial arch. One the-
ities detect structural abnormalities in the ory focuses on the disintegration of this cartilage
periarterial carotid space, though these changes into five components such as tympanohyal,
are not correlated with histological analysis. stylohyal, ceratohyal, hypohyal, and basohyal.
Ultrasonography and Doppler studies showed Tympanohyal and stylohyal contribute to the
eccentric hypoechoic wall thickening of carotid base and styloid process, respectively; hypohyal
bulb and distal common carotid artery which may and basohyal form the hyoid bone; and
or may not be associated with mild non-flow- ceratohyal in humans is thought to degenerate
limiting luminal narrowing. The hypoechoic and forms the stylohyoid ligament. The second
soft tissue may extend outwardly and corroborate theory assumes the division of the cartilage: the
with the location of tenderness or nodule proximal and larger part contributes to the for-
[109–111]. CT is not sensitive to detect subtle mation of styloid process, distal segment into
soft tissue changes due to lower resolution. PET hyoid bone, and the intervening mesenchyme
CT has shown increased [18 F] fluorodeox- develops into stylohyoid ligament [119–122].
yglucose (18 F-FDG) uptake in the region of The styloid process is related to important
enhancing the soft tissue around the carotid neuro and vascular structures, and thus a deviant
[117]. MRI study may reveal intermediate signal and angled styloid process may impinge upon
intensity tissue around the distal common carotid these structures and cause clinical symptoms.
and carotid bulb limited to carotid sheath on The styloid process is related to the 9th to 12th
T1-WI sequence. These abnormal tissues show cranial nerves, cervical sympathetic chain,
a marked homogenous enhancement, more con- branches of external carotid artery, internal carotid
spicuous on fat-saturated T1-WI sequence. The artery, and internal jugular vein. The syndrome is
luminal caliber of the artery is almost always commonly observed in women in the fourth or
normal. Clearly, MRI delineates the abnormal fifth decades. Typical symptoms include unilat-
tissue as well as the extent of involvement and eral sharp or dull aching pain in the lateral neck,
also helps to rule out other causes that may mimic angle of mandible, oral cavity, or base of the
CD [109, 111, 113–115]. tongue which are precipitated or aggravated by
neck movement, swallowing, or yawning. Com-
pression of carotid artery may cause dull aching
Eagle Syndrome or Stylohyoid pain in the parietal region, TIA, stroke, and
Syndrome Horner syndrome exacerbated by rotary move-
ments of head to contralateral side or it may
The long styloid process defined as more than even cause sudden death [119, 120, 123]. Direct
3 cm in length is seen in 4 % of population, mechanical impingement on the cervical carotid
though the typical symptoms attributed to it are artery also has been implicated as an important
seen in just about 4 % of these patients. The risk factor for carotid artery dissection or aneu-
styloid process is a long slender bone attached rysm [124, 125].
to the base of the temporal bone and is related A gamut of possibilities needs to be excluded
closely to many important neurovascular struc- before one considers the diagnosis of ES. Oral
tures. The styloid process, together with examination of tonsillar fossa may reveal unduly
stylohyoid ligament and lesser cornua of hyoid elongated styloid process. Palpation of tonsillar
158 S.K. Kannath and T.R. Kapilamoorthy

fossa may provoke the pain which is abolished by Gornik L (2012) The United States registry
instillation of local anesthetic along the styloid for fibromuscular dysplasia: results in the first
447 patients. Circulation 125:3182–3190
process. Radiographic studies such as submento- 7. Wells RP, Smith RR (1982) Fibromuscular dysplasia
vertical view or orthopantomogram are useful to of the internal carotid artery: a long term follow-up.
reveal enlarged styloid process. The styloid pro- Neurosurgery 10:39–43
cess is considered enlarged if its length exceeds 8. Mettinger KL, Ericson K (1982) Fibromuscular dys-
plasia and the brain observations on angiographic,
one third of the length of the ramus of the mandi- clinical and genetic characteristics. Stroke 13:46–52
ble or if the length is >3 cm. CT and CTA studies 9. De Souza AWS, de Carvalho JF (2014) Diagnostic
can accurately depict the length, orientation, and and classification criteria of Takayasu arteritis.
relation of styloid process to major vascular struc- J Autoimmun 48–49:79–83
10. Ishikawa K (1978) Natural history and classification
tures [126]. Though invasive, 3D rotational angi- of occlusive thromboaortopathy. (Takayasu’s dis-
ography can directly visualize the mechanical ease). Circulation 57:27–35
impression on carotid or other vessels by elon- 11. Kim JH, Suh DC, Kim JK, Kim SJ et al (2005) Cor-
gated styloid process, it is a useful tool to confirm relation of neurological manifestations of Takayasu’s
arteritis with cerebral angiographic findings. J Clin
and localize disease in doubtful and confusing Imaging 29:79–85
situations [127]. 12. Ringleb PA, Strittmatter EI, Loewer M, Hartmann M
et al (2005) Cerebrovascular manifestations of
Takayasu arteritis in Europe. Rheumatology
44:1012–1015
Summary 13. Kim DS, Kim JK, Yoo DS, Huh PW, Cho KS, Kang
JK (2002) Takayasu arteritis presenting with sub-
The clinical disease entities encompassing arachnoid hemorrhage. Report of two cases. J Korean
nonatherosclerotic and nondissection diseases of Med Sci 17:695–698
14. Terao C, Yoshifuji H, Mimori T (2014) Recent
carotid artery are exhaustive and have often advances in Takayasu arteritis. Int J Rheum Dis
nonspecific clinical and imaging findings. The 17:238–247
specific clinical inputs and typical imaging fea- 15. Hotchi M (1992) Pathological studies on Takayasu’s
tures are important clues to suspect these vascular arteritis. Heart Vessels Suppl 7:11–17
16. Hartlage GR, Palios J, Barron BJ, Stillman AE,
diseases and guide in further evaluation and man- Bossone E et al (2014) Multimodality imaging of
agement. Genetic and molecular analysis is aortitis. JACC Cardiovasc Imaging 7:605–619
confirmative in hereditary and connective tissue 17. Ohigashi H, Haraguchi G, Konishi M, Tezuka D,
disorders. Kamiishi T, Ishihara T et al (2012) Improved progno-
sis of Takayasu arteritis over the past decade – com-
prehensive analysis of 106 patients. Circ J
76:1004e11
References 18. Alibaz-Oner F, Aydin SZ, Direskeneli H (2013)
Advances in the diagnosis, assessment and outcome
1. Touze E, Oppenheim C, Tyrstram D et al (2010) of Takayasu’s arteritis. Clin Rheumatol 32:541–546
Fibromuscular dysplasia of cervical and intracranial 19. Caballero PE (2011) Common carotid artery aneu-
arteries. Int J Stroke 5:296–305 rysm revealing Takayasu’s arteritis. J Stroke
2. Plouin PF, Perdu J, Batide-Alanore AL et al (2007) Cerebrovasc Dis 20(6):556–558
Fibromuscular dysplasia. Orphanet J Rare Dis 2:28 20. Santhosh K, Jaydadevan ER, Kapilamoorthy TR,
3. Schievink WI, Bjornsson J (1996) Fibromuscular Sylaja PN (2013) Temporal evolution of
dysplasia of the internal carotid artery: a clinicopath- hypertrophied vasa vasorum of common carotid
ological study. Clin Neuropathol 15:2–6 artery triggered by cerebral ischemia: a serial angio-
4. Mettinger KL (1982) Fibromuscular dysplasia and the graphic investigation. Neurol India 61(3):312–314
brain. II. Current concept of the disease. Stroke 21. Shuaib UA, Kate M, Homik J, Jerrakathil T (2013)
13:53–58 Recurrent non-aneurysmal subarachnoid
5. Schievink WI (2001) Spontaneous dissection of the haemorrhage in Takayasu arteritis: is the cause immu-
carotid and vertebral arteries. N Engl J Med nological or mechanical? BMJ Case Rep
344:898–906 22. Borchers AT, Gershwin MG (2012) Giant cell arteri-
6. Olin JW, Froehlich J, Gu X, Bacharach JM, Eagle K, tis: a review of classification, pathophysiology,
Gray BH, Jaff MR, Kim ESH, Mace P, Matsumoto geoepidemiology and treatment. Autoimmun Rev
AH, McBane RD, Kline-Rogers E, White CJ, 11:A544–A554
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 159

23. Smeeth L, Cook C, Hall AJ (2006) Incidence of HIV-related vascular aneurysms. Br J Radiol
diagnosed polymyalgia rheumatica and temporal 75:884–888
arteritis in the United Kingdom, 1990–2001. Ann 38. Mazzoni P, Chiriboga CA, Millar WS, Rogers A
Rheum Dis 65:1093–1098 (2000) Immunodeficiency virus infection: case report
24. Hayreh SS, Podhajsky PA, Zimmerman B (1998) and literature review. Pediatr Neurol 23:252–255
Ocular manifestations of giant cell arteritis. Am J 39. Ritelli M, Drera B, Vicchio M, Puppini G, Biban P,
Ophthalmol 125:509–520 Pilati M, Prioli MA, Barlat S, Colombi M (2009)
25. Gonzalez-Gay MA, Vazquez-Rodriguez TR, Gomez- Arterial tortuosity syndrome in two Italian paediatric
Acebo I, Pego-Reigosa R, Lopez-Diaz MJ, Vazquez- patients. Orphanet J Rare Dis 4:20
Triñanes MC et al (2009) Strokes at time of disease 40. Callewaert BL, Willaert A, Kerstjens-Frederikse WS,
diagnosis in a series of 287 patients with biopsy- De Backer J, Devriendt K, Albrecht B et al (2008)
proven giant cell arteritis. Medicine (Baltimore) Arterial tortuosity syndrome: clinical and molecular
88:227–235 findings in 12 newly identified families. Hum Mutat
26. Nesher G (2014) The diagnosis and classification of 29:150–158
giant cell arteritis. J Autoimmun 48–49:73–75 41. Nauheim MR, Walcott BP, Nahed BV, Macrae CA,
27. Arida A, Kryprianou M, Kanakis M, Sfikakis PP Levinson JR, Oglivy CS (2011) Arterial tortuosity
(2010) The diagnostic value of ultrasonography- syndrome with multiple intracranial aneurysms.
derived edema of the temporal artery wall in giant Arch Neurol 68(3):369–371
cell arteritis: a second meta-analysis. BMC 42. Germain DP (2007) Ehlers- Danlos syndrome type
Musculoskelet Disord 11:44 4. Orphanet J Rare Dis 2:32
28. Schinkel AF, van den Oord SC, van der Steen AF, van 43. Gdynia HJ, Kuhnlein P, Ludolph AC, Huber R (2008)
Laar JA, Sijbrands EJ (2014) Utility of contrast- Connective tissue disorders in dissections of the
enhanced ultrasound for the assessment of the carotid carotid or vertebral arteries. J Clin Neurosci
artery wall in patients with Takayasu or giant cell 15:489–494
arteritis. Eur Heart J Cardiovasc Imaging 15:541–546 44. Boutouyrie P, Germanin DP, Fiessinger JN, Laloux P,
29. Bley TA, Reinhard M, Hauenstein C, Markl M, Perdu J, Laurent S (2004) Increased carotid wall stress
Warnatz K, Hetzel A, Uhl M, Vaith P, Langer M in vascular Ehlers-Danlos syndrome. Circulation
(2008) Comparison of duplex sonography and high- 109:1530–1535
resolution magnetic resonance imaging in the diagno- 45. Pepin M, Schwarze U, Superti-Furga A, Byers PH
sis of giant cell (temporal) arteritis. Arthritis Rheum (2000) Clinical and genetic features of Ehlers–Danlos
52:2470–2477 syndrome type IV, the vascular type. N Engl J Med
30. Blockmans D, Coudyzer W, Vanderschueren S, 342:673–680
Stroobants S, Loeckx D, Heye S, De Ceuninck L, 46. Oderich GS, Panneton JM, Bower TC, Lindor NM,
Marchal G, Bobbaers H (2008) Relationship between Cherry KJ, Noel AA, Kalra M, Sullivan T, Gloviczi P
fluorodeoxyglucose uptake in the large vessels and (2005) The spectrum, management and clinical out-
late aortic diameter in giant cell arteritis. Rheumatol- come of Ehlers-Danlos syndrome type IV: a 30-year
ogy 47:1179–1184 experience. J Vasc Surg 42:98–106
31. Robbs JV, Paruk N (2010) Management of HIV 47. Schievink WI (2004) Cerebrovascular involvement in
vasculopathy – a South African experience. Eur J Ehlers-Danlos Syndrome. Curr Treat Options
Vasc Endovasc Surg 39:S25–S31 Cardiovasc Med 6:231–236
32. Padayachy V, Robbs JV (2012) Carotid artery aneu- 48. Dohle C, Baehring JM (2012) Multiple strokes and
rysms in patients with human immunodeficiency bilateral carotid dissections: a fulminant case of newly
virus. J Vasc Surg 55:331–337 diagnosed Ehlers–Danlos syndrome type IV. J Neurol
33. Hamilton DK, Kassel NF, Jensen ME, Dumont AS Sci 318:168–170
(2007) Subarachnoid hemorrhage and diffuse 49. Horowitz MB, Purdy PD, Valentine RJ et al (2000)
vasculopathy in an adult infected with HIV. Remote vascular catastrophes after neurovascular
J Neurosurg 106:478–480 interventional therapy for type 4 Ehlers-Danlos syn-
34. Tipping B, de Villiers L, Wainwright H, Candy S, drome. AJNR Am J Neuroradiol 21:974–976
Bryer A (2007) Stroke in patients with human immu- 50. Usinskiene J, Mazighi M, Bisdorff A et al (2006)
nodeficiency virus infection. J Neurol Neurosurg Psy- Fatal peritoneal bleeding following embolization of
chiatry 78:1320–1324 a carotid-cavernous fistula in Ehlers-Danlos syn-
35. Maniker AH, Hunt D (1996) Cerebral aneurysm in the drome type IV. Cardiovasc Intervent Radiol
HIV patient: a report of six cases. Surg Neurol 29:1104–1106
46:49–54 51. Loeys BL, Schwarze U, Holm T et al (2006) Aneu-
36. Chetty R, Batitang S, Nair R (2000) Large artery rysm syndromes caused by mutations in the TGF-beta
vasculopathy in HIV-positive patients: another receptor. N Engl J Med 355(8):788–798
vasculitic enigma. Hum Pathol 31:374–379 52. Maccarrick G, Black JH III, Bowdin S, Hamamsy IE,
37. Woolgar JD, Ray R, Maharaj K, Robbs JV (2002) Frischmeier-Guerrerio PA, Guerrerio AL, Sponseller
Colour Doppler and grey scale ultrasound features of PD, Loeys B, Dietz HC (2014) Loeys–Dietz
160 S.K. Kannath and T.R. Kapilamoorthy

syndrome: a primer for diagnosis and management. 69. Okendi E, Moghaddam MB, Oderich GS (2010)
Genet Med 16:576–587 Internal carotid artery aneurysms in a patient with
53. Rahme RJ, Adel JG, Bendok BR, Bebawy JF, Gupta neurofibromatosis type 1. Vasc Endovascular Surg
DK, Batjer HH (2011) Association of intracranial 44:511–514
aneurysm and Loeys-Dietz Syndrome: case illustra- 70. Xu J, Ismat FA, Wang T et al (2007) NF1 regulates a
tion, management, and literature review. Neurosur- Ras-dependent vascular smooth muscle proliferative
gery 69:E488–E493 injury response. Circulation 116:2148–2156
54. Levitt MR, Morton RP, Mai JC, Ghodke B, Hallam 71. Ku YK, Chen HW, Chen HW, Fu CJ, Chin SC, Liu
DK (2012) Endovascular treatment of intracranial YC (2008) Giant extracranial aneurysms of both
aneurysms in Loeys-Dietz syndrome. J Neurointerv internal carotid arteries with aberrant jugular veins
Surg 4:e37 in a patient with neurofibromatosis type 1. Am J
55. van den Berg JSP, Hennekam RCM, Cruysberg JRM, Neuroradiol AJNR 29:1750–1752
Steijlen PM, Swart J, Tijmes N et al (2000) Prevalence 72. Reubi F (1945) Neurofibromatose et le´sions
of symptomatic intracranial aneurysm and ischaemic vasculaires. Schweiz Med Wochenschr 75:463–465
stroke in pseudoxanthoma elasticum. Cerebrovasc 73. Schievink WI, Riedinger M, Maya MM (2005) Fre-
Dis 10:315–319 quency of incidental intracranial aneurysms in neuro-
56. Schievink WI, Michels VV, Piepgras DG (1994) fibromatosis type 1. Am J Med Genet A 134:45–48
Neurovascular manifestations of heritable connective 74. You MW, Kim EJ, Choi WS (2011) Intracranial and
disorders: a review. Stroke 25:889–903 extracranial fusiform aneurysms in a patient with
57. Vasseur M, Carsin-Nichol B, Ebran JM, Willoteaux S, neurofibromatosis type 1: a case report.
Martin L, Leftheriotis G (2011) Carotid rete mirabile Neurointervention 6:34–37
and pseudoxanthoma elasticum: an accidental associ- 75. Rosser TL, Vezina G, Packer RJ (2005) Cerebrovas-
ation? Eur J Vasc Endovasc Surg 42:292–294 cular abnormalities in a population of children with
58. Dean JCS (2007) Marfan syndrome: clinical diagno- neurofibromatosis type 1. Neurology 64:553–555
sis and management. Eur J Hum Genet 15:724–733 76. Patro SN, Gupta AK, Aravinda HR, Jolapara MB,
59. Eberth JF, Taucer AI, Wilson E, Humphrey JD (2009) Saini J (2009) Combined transarterial and percutane-
Mechanics of carotid arteries in a mouse model of ous coiling of a spontaneous vertebrovertebral fistula
Marfan syndrome. Ann Biomed Eng 37:1093–1104 associated with neurofibromatosis Type 1.
60. Ferruzzi J, Collins MJ, Yeh AT, Humphrey JD (2011) J Neurosurg 111:37–40
Mechanical assessment of elastin integrity in fibrillin- 77. Deans WR, Bloch S, Leibrock L, Berman BM,
1-deficient carotid arteries: implications for Marfan Skultety FM (1982) Arteriovenous fistula in patients
syndrome. Cardiovasc Res 92:287–295 with neurofibromatosis. Radiology 144:103–107
61. Loeys BL, Dietz HC, Braverman AC et al (2010) 78. Gao P, Chen Y, Zhang H, Ling F (2013) Vertebral
Towards a revised Ghent nosology for the Marfan arteriovenous fistulae (AVF) in Neurofibromatosis
syndrome. J Med Genet 47:476–485 type 1: a report of two cases. Turk Neurosurg
62. Schievink WI, Parisi JE, Piepgras DG (1997) Intra- 23:289–293
cranial aneurysms in Marfan’s syndrome: an autopsy 79. Baccin CE, Krings T, Alvarez H, Ozanne A,
study. Neurosurgery 41:866–871 Lasjaunias PL (2007) A report of two cases with
63. Yuan SM, Jing H (2011) Cystic medial necrosis: dolichosegmental intracranial arteries as a new fea-
pathological findings and clinical implications. Rev ture of PHACES syndrome. Childs Nerv Syst
Bras Cir Cardiovasc 26:107–115 23:559–567
64. Bouziane Z, Bensaid B, Idrissi RE, Mesnaoui AE, 80. Heyer GL, Dowling MM, Licht DJ, Tay SKH,
Bensaid Y, Ratbi I (2013) Marfan syndrome and cer- Morel K, Garzon MC, Meyers P (2008) The cerebral
vical internal carotid artery aneurysm. Vasa vasculopathy of PHACES syndrome. Stroke
42:457–460 39:308–316
65. Sztajzel R, Hefft S, Girardet C (2001) Marfan’s syn- 81. Hess CP, Fullerton HJ, Metry DW, Drolet BA, Siegel
drome and multiple extracranial aneurysms. DH, Auguste KI, Gupta N, Haggstrom AN, Dowd CF,
Cerebrovasc Dis 11:346–349 Frieden IJ, Barkovich AJ (2010) Cervical and intra-
66. Matsuda M, Matsuda I, Handa H, Okamoto K (1979) cranial arterial anomalies in 70 patients with PHACE
Intracavernous giant aneurysm associated with syndrome. AJNR Am J Neuroradiol 31:1980–1986
Marfan’s syndrome. Surg Neurol 12:119–121 82. Siegel DH, Tefft KA, Kelly T, Johnson C, Mery D,
67. Riccardi VM (2000) The vasculopathy of NF1 and Burrows P, Pope E, Cordisco M, Holland KE,
histogenesis control genes. Clin Genet 58:345–347 Maheswari M, Keith P, Garzon M, Hess C, Frieden
68. Oderich GS, Sullivan TM, Bower TC, Glovickzi P, IJ, Fullerton HJ, Drolet BA (2012) Stroke in children
Miller DV, Babovic-Vuksanovic D, Macedo TA, with posterior fossa brain malformations, hemangi-
Stanson A (2007) Vascular abnormalities in patients omas, arterial anomalies, coarctation of the aorta and
with neurofibromatosis syndrome type I: clinical cardiac defects, and eye abnormalities (PHACE) syn-
spectrum, management, and results. J Vasc Surg drome: a systematic review of the literature. Stroke
46:475–484 43:1672–1674
9 Nonatherosclerotic Nondissection Diseases of Carotid Artery 161

83. Kirkwood ML, Chung J, Timaran CH, Valentine RJ 98. Langner S, Fleck S, Kirsch M, Petrik M, Hosten N
(2013) Extracranial carotid artery aneurysms in two of (2008) Whole-body ct trauma imaging with adapted
three monozygotic triplets with tuberous sclerosis and optimized CT angiography of the craniocervical
complex. J Vasc Surg 57:1120–1122 vessels: do we need an extra screening examination?
84. Jurkiewicz E, Jozwiak S (2006) Giant intracranial AJNR Am J Neuroradiol 29:1902–1907
aneurysm in a 9-year-old boy with tuberous sclerosis. 99. Fleck SK, Langner S, Baldauf J, Kirsch M,
Pediatr Radiol 36:463 Rosenstengel C, Schroeder HW (2010) Blunt
85. Hung PO, Wang HS, Chou ML, Wong AMC (2008) craniocervical artery injury in cervical spine lesions:
Tuberous sclerosis complex with multiple intracranial the value of CT angiography. Acta Neurochir
aneurysms in an infant. Pediatr Neurol 39:365–367 152:1679–1686
86. Sayed A, Elwan H, Fouad F, Taha A, Elhindawi K, 100. Ouffiah C, Hall E (2012) Imaging assessment of pen-
Khairy H, Gad A, Eldin HK (2010) Behcet extracra- etrating injury of the neck and face. Insights Imaging
nial carotid aneurysms: is there still a role for ligation. 3:419–431
Eur J Vasc Endovasc Surg 39:17–22 101. Chang FC, Lirng JF, Luo CB, Wang SJ, Wu HM, Guo
87. Albeyoglu S, Cinar B, Eren T, Filizcan U, WY, Teng MMH, Chnag CY (2008) Patients with
Bayserke O, Aslan C (2010) Extracranial carotid head and neck cancers and associated postirradiated
artery aneurysm due to Behcet’s disease. Asian carotid blowout syndrome: endovascular therapeutic
Cardiovasc Thorac Ann 18:574–576 methods and outcomes. J Vasc Surg 47:936–945
88. Bouarhroum A, Sedki N, Bouziane Z, Mahi OE, 102. Chang FC, Luo CB, Lirng JF, Lin CJ, Wu HM, Hung
Idrissi RE, Lahlou Z, Lekehel B, Sefiani Y, Mesnaoui SC, Guo WY, Teng MMH, Chang CY (2013) Evalu-
AE, Benjelloun A, Ammar F, Bensaïd Y (2006) ation of the outcomes of endovascular management
Extracranial carotid aneurysm in Behçet disease: for patients with head and neck cancers and associated
report of two new cases. J Vasc Surg 43:627–630 carotid blowout syndrome of the external carotid
89. Ozveren MF, Matsumoto Y, Kondo R, Takahashi A artery. Clin Radiol 68:e561–e569
(2009) Coil embolization of an unruptured intracra- 103. Chaloupka JC, Putman CM, Citardi MJ et al (1996)
nial aneurysm associated with Behcet’s disease. Endovascular therapy for the carotid blowout syn-
Neurol Med Chir (Tokyo) 49:471–473 drome in head and neck surgical patients: diagnostic
90. Kaku M, Hamada JI, Kuroda JI, Kai Y, Morioka M, and managerial considerations. AJNR Am J
Kuratsu JI (2007) Multiple peripheral middle cerebral Neuroradiol 17:843–852
aneurysms associated with Behcet’s disease. Acta 104. Lee CW, Yang CY, Chen YF, Huang A, Wang YH,
Neurochir (Wien) 149:823–827 Liu HM (2014) CT angiography findings in carotid
91. Garg K, Rockman CB, Lee V, Maldonaldo TS, blowout syndrome and its role as a predictor of 1-year
Jacobowitz GR, Adelman MA, Mussa FF (2012) survival. AJNR Am J Neuroradiol 35:562–567
Presentation and management of carotid artery aneu- 105. Luo CB, Teng MMH, Chang FC, Chang CY, Guo
rysms and pseudoaneurysms. J Vasc Surg WY (2008) Radiation carotid blowout syndrome in
55:1618–1622 nasopharyngeal carcinoma: angiographic features and
92. Longo GM, Kibbe MR (2005) Aneurysms of the endovascular management. Otolaryngol Head Neck
carotid artery. Semin Vasc Surg 18:178–183 Surg 138:86–91
93. Attigah N, Ku¨lkens S, Zausig N, Hansmann J, 106. Yanik B, Keyik B, Conkbayir I, Teber MA (2011)
Ringleb P, Hakimi M, Eckstein HH, Allenberg JR, Carotid blowout syndrome with oronasal hemor-
Bo¨ckler D (2009) Surgical therapy of extracranial rhage: magnetic resonance imaging findings. Jpn J
carotid artery aneurysms: long-term results over a Radiol 29:72–75
24-year period. Eur J Vasc Endovasc Surg 107. Headache Classification Committee of the Interna-
37:127–133 tional Headache Society (1988) Classification and
94. Nadig S, Barnwell S, Wax MK (2009) diagnostic criteria for headache disorders, cranial neu-
Pseudoaneurysm of the external carotid artery – ralgias and facial pain. Cephalalgia 8(Suppl 7):1–96
review of literature. Head Neck 31:136–139 108. Headache Classification Subcommittee of the Inter-
95. Cox MW, Whittaker DR, Martinez C, Fox CJ, national Headache Society (2004) The International
Feuerstein IM, Gillespie DL (2007) Traumatic Classification of Headache Disorders: 2nd edn.
pseudoaneurysms of the head and neck: early Cephalalgia 24(Suppl 1):9–160
endovascular intervention. J Vasc Surg 46:1227–1233 109. Kosaka N, Sagoh T, Uematsu H, Kimura H,
96. Maras D, Lioupis C, Magoufis G, Tsamopoulos N, Miyayama S, Noguchi M, Itoh H (2007) Imaging by
Moulakakis K, Andrikopoulos V (2006) Covered multiple modalities of patients with a carotidynia
stent-graft treatment of traumatic internal carotid syndrome. Eur Radiol 17:2430–2433
artery pseudoaneurysms: a review. Cardiovasc 110. Barker R, Massouh H (2009) Carotidynia – a valid
Intervent Radiol 29:958–968 cause of unilateral neck pain. Eur J Radiol Extra 71:
97. Welleweerd JC, Moll FL, de Borst GJ (2012) Techni- e97–e99
cal options for the treatment of extracranial carotid 111. Schaumberg J, Eckert B, Michels P (2011)
aneurysms. Expert Rev Cardiovasc Ther 10:925–931 Carotidynia. Magnetic resonance imaging and
162 S.K. Kannath and T.R. Kapilamoorthy

ultrasonographic imaging of a self-limiting disease. 120. Piagkou M, Anagnostopoulou S, Kouladouros K,


Clin Neuroradiol 21:91–94 Piagkos G (2009) Eagle’s syndrome: a review of the
112. Upton PD, Smith JRG, Charnock DR (2003) Histo- literature. Clin Anat 22:545–558
logic confirmation of carotidynia. Otolaryngol Head 121. Rodriguez-Vazquez JF, Merida-Velasco JR, Verdugo-
Neck Surg 129:443–444 Lopez S, Sanchez-Montesinos I, Merida-Velasco JA
113. Burton BS, Syms MJ, Petermann GW, Burgess LPA (2006) Morphogenesis of the second pharyngeal arch
(2000) MR imaging of patients with carotidynia. cartilage (Reichert’s cartilage) in human embryos.
AJNR Am J Neuroradiol 21:766–769 J Anat 208:179–189
114. Woo JKH, Jhamb A, Heran MKS, Hurley M, Graeb D 122. Lorman JG, Biggs JR (1983) The Eagle syndrome.
(2008) Resolution of existing intimal plaque in a patient AJR Am J Roentgenol 140:881–882
with carotidynia. AJNR Am J Neuroradiol 29:732–733 123. Ghosh LM, Dubey SP (1999) The syndrome of elon-
115. Commacho F, Bottin R, Brescia G, Tsilikas K, Volo T, gated styloid process. Auris Nasus Larynx
Tregnaghi A, Martini A (2012) Carotidynia: new 26:169–175
aspects of a controversial entity. Acta otorhino- 124. Dao A, Karnezis S, Lane JS III, Fujitani RM, Saremi
laryngol Ital 32:266–269 F (2011) Eagle syndrome presenting with external
116. Taniguchi Y, Horino T, Hashimoto K (2008) Is carotid artery pseudoaneurysm. Emerg Radiol
carotidynia syndrome a subset of vasculitis? 18:263–265
J Rheumatol 35:1901–1902 125. Muthusami P, Kesavadas C, Sylaja PN, Thomas B,
117. Amaravadi RR, Behr SC, Kousoubris PD, Raja S Harsha KJ, Kapilamoorthy TR (2013) Implicating the
(2008) [18F] Fluorodeoxyglucose positron-emission long styloid process in cervical carotid artery dissec-
tomography-CT imaging of carotidynia. AJNR Am J tion. Neuroradiology 55(7):861–867
Neuroradiol 29:1197–1199 126. Murtagh RD, Caracciolo RD, Fernandez G (2001) CT
118. Colby CC, Del Gaudio JM (2011) Stylohyoid com- findings associated with Eagle syndrome. AJNR Am J
plex syndrome-a new diagnostic classification. Arch Neuroradiol 22:1401–1402
Otolaryngol Head Neck Surg 137:248–252 127. Nakagawa D, Ota T, Iijima A, Saito N (2011) Diag-
119. Fusco DJ, Asteraki S, Spetzler RF (2012) Eagle’s nosis of eagle syndrome with 3-dimensional angiog-
syndrome: embryology, anatomy, and clinical man- raphy and near-infrared spectroscopy: case report.
agement. Acta Neurochir 154:1119–1126 Neurosurgery 68:E847–E849
Imaging of the Pathology of the
Vertebral Arteries 10
David Chiao and Max Wintermark

Contents Subclavian Steal Syndrome . . . . . . . . . . . . . . . . . . . . . . . . 180


Epidemiology and Clinical Findings . . . . . . . . . . . . . . . . 180
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164 Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182
Embryology and Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . 164 Imaging Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182
Embryology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164 Work-Up and Management . . . . . . . . . . . . . . . . . . . . . . . . . . 183
Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166 Bow Hunter’s Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . 184
Vascular Territories . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 Epidemiology and Clinical Findings . . . . . . . . . . . . . . . . 184
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170 Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
Atherosclerotic Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172 Imaging Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186
Epidemiology and Clinical Findings . . . . . . . . . . . . . . . . 172 Work-Up and Management . . . . . . . . . . . . . . . . . . . . . . . . . . 186
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172 Miscellaneous Conditions . . . . . . . . . . . . . . . . . . . . . . . . . . 188
Imaging Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172 Fibromuscular Dysplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
Work-Up and Management . . . . . . . . . . . . . . . . . . . . . . . . . . 174 Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
Vertebral Artery Dissection . . . . . . . . . . . . . . . . . . . . . . . . 175 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
Epidemiology and Clinical Findings . . . . . . . . . . . . . . . . 175
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
Imaging Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
Work-Up and Management . . . . . . . . . . . . . . . . . . . . . . . . . . 178

D. Chiao • M. Wintermark (*)


Department of Radiology, School of Medicine, University
of Virginia, Charlottesville, VA, USA
e-mail: dc.harmonic@gmail.com;
max.wintermark@gmail.com

# Springer Science+Business Media New York 2016 163


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_17
164 D. Chiao and M. Wintermark

encountered vertebral artery diseases, vertebral


Abstract
artery atherosclerosis and vertebral artery dissec-
The vertebral arteries are among the most clin-
tion. This is then followed by discussion of two
ically important arteries in the body, providing
functional disorders, subclavian steal syndrome
blood flow to the posterior circulation of the
and bow hunter’s syndrome. Finally, a brief over-
brain. Both radiologists and clinicians need to
view of miscellaneous conditions, such as
understand the basic anatomy and the pathol-
fibromuscular dysplasia and vasculitis, is covered
ogy that can affect the vertebral arteries in the
in the last section.
acute and non-acute settings. Radiologists, in
particular, need to be cognizant of the imaging
findings of vertebral artery pathology as
misdiagnosis of certain common conditions
Embryology and Anatomy
could be potentially disastrous. In this chapter,
the embryology and anatomy of the vertebral
Embryology
arteries is reviewed, with a focus on normal
variants and vascular territories. Vertebral
Vertebral artery embryology forms a foundation
artery diseases are then discussed, including
for understanding normal and variant anatomy,
atherosclerotic disease, vertebral artery dissec-
which is useful in discriminating normal anatomic
tion, subclavian steal syndrome, and a variety
variation from pathologic processes. Recognition
of other conditions. Each section is organized
of normal variants is also important prior to sur-
by epidemiology and clinical findings, patho-
gical or endovascular intervention.
physiology, imaging findings, and work-up
The vertebral arteries develop from the dorsal
and management.
longitudinal neural arteries, which arise from lon-
gitudinal plexiform anastomoses of the first six
Keywords
cervical intersegmental arteries (Fig. 1) [1]. The
Vertebral artery • Carotid-vertebrobasilar vari-
dorsal longitudinal arteries run parallel to the
ants • Vertebral artery fenestration • Basilar
primitive internal carotid arteries and appear by
artery fenestration • Posterior circulation • Ath-
day 30 of embryonic life. The cranial portions of
erosclerosis • Stroke • Vertebral artery stenosis
the dorsal longitudinal neural arteries gradually
• Vertebral artery dissection • Subclavian steal
appose each other, fusing in the midline around
syndrome • Bow hunter’s syndrome
day 35 to form the basilar artery [1]. As the basilar
artery matures, it begins supplying the posterior
circulation, which is previously supplied by the
Introduction primitive internal carotid arteries via the fetal pos-
terior communicating arteries.
Radiologists are frequently asked to evaluate the Prior to the development of the mature
course, caliber, and integrity of the vertebral arter- vertebrobasilar system, temporary anastomoses
ies. Successful evaluation of the vertebral arteries between the cranial portions of the dorsal longitu-
requires a thorough knowledge of vertebral artery dinal neural arteries and the primitive internal
anatomy and pathology. carotid arteries exist [1]. These carotid-
This chapter begins by reviewing the embryol- vertebrobasilar anastomoses, as well as the cervi-
ogy and anatomy of the normal vertebral arteries. cal intersegmental arteries, eventually regress
Normal variants are discussed in this section, as (with the exception of the seventh cervical
they are commonly seen in clinical practice. This intersegmental arteries which develop into the
is followed by a general discussion of the most subclavian arteries). If these anastomoses fail to
frequently employed imaging modalities used in regress, communication between the
evaluating the vertebral arteries. This is followed vertebrobasilar system and the internal carotid
by discussion of the two most commonly artery persists into adulthood. Four major
10 Imaging of the Pathology of the Vertebral Arteries 165

Fig. 1 Embryology of the Right internal carotid artery


vertebral arteries. The
vertebral arteries develop Right vertebral artery Right external carotid artery
from longitudinal plexiform
anastomoses of the first six Aorta
Right thyrocervical artery
cervical intersegmental
arteries. The seventh
cervical intersegmental
arteries develop into the
subclavian arteries

Cervical intersegmental arteries

1
2
3
4
5
6
7

carotid-vertebrobasilar variants have been neuralgia if there is significant mass effect on the
described in the literature and are named trigeminal nerve.
according to the cranial nerves that they parallel The persistent hypoglossal artery is the second
(Fig. 2): the persistent trigeminal artery, the per- most common carotid-vertebrobasilar variant,
sistent otic artery, the persistent hypoglossal present in 0.1–0.25 % of cerebral angiograms
artery, and the proatlantal intersegmental artery and autopsies [1, 3]. It arises from the distal cer-
[1–3]. In general, these variants are considered vical internal carotid artery (usually between the
incidental findings; however, an association with C1 and C3 vertebral bodies), passes through an
an increased incidence of aneurysms has been enlarged hypoglossal canal, and joins the proxi-
described [3]. mal basilar artery (Figs. 7 and 8). Patients are
The persistent trigeminal artery is the most typically asymptomatic but can uncommonly pre-
common carotid-vertebrobasilar variant, present sent with glossopharyngeal neuralgia and hypo-
in 0.1–1.0 % of cerebral angiograms and autopsies glossal paralysis if there is significant mass effect
[1, 3]. It arises from the cavernous internal carotid on the hypoglossal nerve.
artery and runs across the dorsum sellae to con- The persistent otic artery is exceedingly rare,
nect with the distal basilar artery (Fig. 3); the having only been described in a handful of case
basilar artery proximal to the aberrant connection reports [1, 3]. It arises from the petrous internal
is often hypoplastic. On sagittal imaging, the per- carotid artery, exits via the internal acoustic mea-
sistent trigeminal artery forms a characteristic tus, and joins the proximal basilar artery.
“tau” or “trident” sign (Fig. 4) [5]. The persistent A fourth variant, the proatlantal intersegmental
trigeminal artery can occasionally be mistaken for artery, is similar to the persistent hypoglossal
an aneurysm on cross-sectional imaging; 3D CTA artery in that it arises from the distal cervical
or DSA can be helpful in these difficult cases internal carotid artery and joins the proximal bas-
(Figs. 5 and 6). Patients are typically asymptom- ilar artery; however, instead of traveling through
atic but can uncommonly present with trigeminal the hypoglossal canal, the proatlantal
166 D. Chiao and M. Wintermark

Trigeminal artery

Otic artery
Hypoglossal
artery

Proatlantal
intersegmental
artery

Vertebral
artery
Carotid
artery Fig. 3 MRA in a 45-year-old female demonstrating a
persistent trigeminal artery (arrow) which connects the
cavernous internal carotid artery with the distal basilar
artery
Fig. 2 Carotid-vertebrobasilar variants. The persistent tri-
geminal artery is the most common variant, connecting the
cavernous internal carotid artery with the distal basilar common than vertebral artery fenestration, occur-
artery. The persistent otic artery connects the petrous inter-
nal carotid artery with the proximal basilar artery via the
ring in up to 5 % of cerebral angiograms and
internal acoustic meatus. The persistent hypoglossal artery autopsies, and also predisposes to aneurysms
connects the distal cervical internal carotid artery with the (Fig. 10) [1].
proximal basilar artery via the hypoglossal canal. The Duplication of the vertebral artery is very rare.
proatlantal intersegmental artery connects the distal cervi-
cal internal carotid artery with the proximal basilar artery
It is thought to be due to failure of fusion of the
via the foramen magnum cervical intersegmental arteries during
development [1].

intersegmental artery travels through the foramen


magnum [1, 5]. Anatomy
Vertebral artery fenestrations occur in 0.2–2.0 %
of cerebral angiograms and autopsies [1]. Extracra- The vertebral arteries usually arise from the prox-
nial fenestrations are thought to arise due to the imal subclavian arteries. However, in approxi-
persistence of the cervical intersegmental arteries mately 5 % of patients, the left vertebral artery
while intracranial fenestrations are thought to arise arises directly from the aortic arch (Fig. 11)
due to the persistence of basivertebral anastomoses. [5]. The vertebral arteries are typically asymmet-
On imaging, vertebral artery fenestrations are char- ric in size, most commonly with the left vertebral
acterized by a “gap” in the middle of a vertebral artery being dominant. When the vertebral arteries
artery; this can be confusing on axial imaging, with are approximately equal in size, they are consid-
visualization of two apparent lumens, but is nicely ered to be codominant. The normal luminal diam-
demonstrated on reformats (Fig. 9). Vertebral artery eter of the extracranial vertebral artery is
fenestrations are associated with other vascular approximately 3–5 mm.
anomalies and predispose to vertebral artery aneu- After arising from the subclavian arteries, the
rysms [1–3]. Basilar artery fenestration is more vertebral arteries ascend in the neck and are
10 Imaging of the Pathology of the Vertebral Arteries 167

Fig. 4 “Tau” sign (arrow)


on MRA sagittal reformat in
a patient with a persistent
trigeminal artery. The
persistent trigeminal artery
is the most common
carotid-vertebrobasilar
variant

foramen, and then exits superiorly through the


C2 transverse foramen to become the V3 segment.
The V2 segment resides inside the vertebral col-
umn (Fig. 13) and is vulnerable to vertebral frac-
tures, especially fractures which violate the
transverse foramina.

V3 Segment
The V3 segment courses posteriorly and medially
around the atlantooccipital joint (forming a
groove along the posterior ring of the atlas) and
then sharply turns anteriorly and superiorly to
pierce the dura to become the V4 segment. The
V3 segment resides outside the vertebral column
and is vulnerable to rotational forces which can
injure the artery at its dural insertion.
Fig. 5 3D carotid angiography nicely demonstrates a per-
sistent trigeminal artery (arrow) which was mistaken for an V4 Segment
aneurysm on prior MRA The V4 segment courses superiorly, anteriorly,
and medially through the foramen magnum and
categorized into four segments, abbreviated V1, then superomedially behind the clivus. The V4
V2, V3, and V4 (Fig. 12). segments unite to form the basilar artery near the
pontomedullary junction.
V1 Segment Along its course, the vertebral artery gives rise
The V1 segment is the first segment of the verte- to numerous arterial branches: cervical branches,
bral artery. The V1 segment typically arises from meningeal branches, and intracranial branches.
the subclavian artery, courses posteriorly and These arteries are described below.
superiorly in the superior mediastinum, and then
enters the C6 transverse foramen to become the Cervical Branches
V2 segment. The cervical branches originate from the extracra-
nial vertebral artery as it courses up the neck. The
V2 Segment cervical branches are categorized into the spinal
The V2 segment courses superiorly through the branches (which supply the spinal cord and verte-
C3–C6 transverse foramina, turns superolaterally bral bodies) and the muscular branches (which
through the “inverted L-shaped” C2 transverse supply the deep cervical musculature). The
168 D. Chiao and M. Wintermark

Fig. 6 Right carotid DSA


in the same patient as in
Fig. 5 with frontal (a) and
lateral (b) projections, again
demonstrating a persistent
trigeminal artery (arrow)

Intracranial Branches
The intracranial branches originate from the intra-
cranial vertebral artery (i.e., the V4 segment). The
intracranial branches are categorized into the ante-
rior spinal arteries, the posterior spinal arteries,
the posterior inferior cerebellar arteries, and the
perforating arteries. The anterior spinal arteries
unite in 50 % of cases and course inferiorly in
the anteromedian sulcus of the spinal cord to
supply the anterior spinal cord. The posterior
spinal arteries course inferiorly along the
posterolateral spinal cord to supply the posterior
spinal cord. The posterior inferior cerebellar
arteries course around the medullary tonsils to
supply portions of the medulla, inferior
cerebellum, and choroid plexus of the fourth ven-
tricle. The perforating arteries arise off the intra-
cranial vertebral artery to supply portions of the
medulla, inferior cerebellar peduncles, and
Fig. 7 CTA demonstrating a persistent hypoglossal artery olivary bodies.
(arrow) traversing through the hypoglossal canal
Variant anatomy of the posterior inferior cere-
muscular branches provide an important collateral bellar artery is common. Anomalous origin of the
pathway in the vertebrobasilar circulation, com- posterior inferior cerebellar arteries from V3
municating with the occipital branch of the exter- occurs in up to 10 % of cases. Duplication of the
nal carotid artery. posterior inferior cerebellar arteries is less com-
mon, occurring in approximately 2 % of cases
Meningeal Branches [4]. Occasionally, the posterior inferior cerebellar
The meningeal branches originate from the distal arteries arise from a single common trunk. Occa-
extracranial vertebral artery. The meningeal sionally, the vertebral artery terminates in the pos-
branches are categorized into the anterior menin- terior inferior cerebellar artery; in this situation,
geal artery and the posterior meningeal artery. The the contralateral vertebral artery supplies the
anterior meningeal artery arises from V2 and sup- majority of the posterior circulation blood flow
plies the dura around the foramen magnum. The [4]. Familiarity with these common variants helps
posterior meningeal artery arises from V3 and avoid confusion when interpreting the intracranial
supplies the falx cerebellum. branches of the vertebral arteries.
10 Imaging of the Pathology of the Vertebral Arteries 169

Fig. 8 Left carotid DSA in


the same patient as in Fig. 7
with frontal (a) and lateral
(b) projections
demonstrating a persistent
hypoglossal artery (arrow)

Fig. 9 (a) Axial time-of-


flight MRA demonstrates
two apparent lumens
(arrow) of the right
vertebral artery. (b) Coronal
time-of-flight MRA
maximum-intensity
projection reveals that this
represents a focal
fenestration (arrow) of the
V2 segment

Fig. 10 Right vertebral artery DSA (a) and 3D angiogram artery DSA after coil embolization demonstrates no signif-
(b) demonstrating a fenestrated proximal basilar artery icant filling of the aneurysm sac
with an associated saccular aneurysm. (c) Right vertebral

Vascular Territories insufficiency results in neurologic deficits that are


significantly different than those incurred by
The vertebrobasilar system supplies the posterior carotid artery insufficiency. Typical symptoms of
circulation of the brain. As such, vertebrobasilar vertebrobasilar insufficiency include bilateral
170 D. Chiao and M. Wintermark

motor and sensory deficits, nausea, vomiting, arteries, the posterior inferior cerebellar arteries,
vertigo, ataxia, diplopia, dysarthria, and dysmetria and the anterior inferior cerebellar arteries. The
[4, 5]. The precise deficits can be predicted based superior cerebellar arteries supply the superior
on the vascular territory of the arterial insult. surface of the cerebellum and the upper vermis.
The posterior inferior cerebellar arteries supply
Cerebellum the posterior and inferior surfaces of the cerebel-
The cerebellum is predominantly supplied by lum and inferior vermis. The anterior inferior cer-
three paired arteries: the superior cerebellar ebellar arteries supply the petrosal surface of the
cerebellum. The central portions of the vermis and
cerebellar hemispheres represent a watershed area
with variable contributions from all three vessels.

Brainstem
The brainstem is predominantly supplied by the
basilar artery and its perforating branches. How-
ever, the superior cerebellar arteries supply por-
tions of the superior pons while the distal vertebral
arteries and the posterior inferior cerebellar arter-
ies supply portions of the medulla. Occlusion of
one of the posterior inferior cerebellar arteries can
cause infarction of the lateral medulla, resulting in
Wallenberg syndrome. This syndrome is clini-
cally characterized by contralateral sensory defi-
cits affecting the body and extremities, as well as
ipsilateral sensory deficits affecting the face.

Imaging

Fig. 11 Oblique sagittal reformat NECT demonstrating The vertebral arteries can be evaluated with a
the left vertebral artery arising directly from the aorta variety of imaging modalities, including ultra-
(arrow) sound, MRA, CTA, and angiography. Ultrasound

Fig. 12 3D vessel analysis


in a normal patient with
frontal (a) and oblique (b)
projections demonstrating
the four standard vertebral
artery segments, V1–V4
10 Imaging of the Pathology of the Vertebral Arteries 171

overcome many of these limitations but is still


prone to poor spatial resolution, patient motion,
and metal artifacts.
An alternative to MRA is CTA, which provides
excellent visualization of the vertebral arteries
with superior spatial resolution and shorter imag-
ing times. In addition, CTA is generally offered
day and night at most facilities. The scanning
range typically includes the aortic arch to the
circle of Willis in order to visualize the whole
length of the vertebral arteries. Automatic trigger-
ing or test bolus techniques can be used to time the
contrast injection. An adjunct non-contrast and
post-contrast head CT is often performed with
the CTA. One weakness of CTA is that it tends
to overestimate arterial stenosis in the setting of
heavy vessel calcification. In addition, the direc-
tion of arterial flow cannot be determined with
conventional CT imaging.
Angiography has historically been considered
the “gold standard” in vertebral artery imaging but
is now rarely performed as the initial imaging
examination. In the modern era, angiography is
Fig. 13 3D vessel analysis in a normal patient demon- typically reserved for resolving diagnostic
strating the relationship of V2 with the bony cervical
vertebral column
uncertainties, planning interventions, or
performing therapeutic procedures. Vertebral
artery angiography is typically performed with
with pulsed Doppler using a linear 5.0–7.5 MHz rapid sequences during subclavian artery injec-
probe is a relatively inexpensive examination but tions. Selective imaging can also be obtained,
is significantly limited by osseous structures, cal- usually in the AP, lateral views, and oblique
cified atherosclerosis, patient body habitus, and views. Angiography carries a small inherent risk
operator experience. Thus, cross-sectional imag- of vascular injury or distal embolization. The risk
ing is typically performed for full evaluation of of TIA and stroke is approximately 3 % and <1 %
the vertebral arteries. respectively with a cerebral angiogram
Two-dimensional time-of-flight MRA typi- examination [4].
cally provides satisfactory visualization of the The ideal imaging modality for the evaluation
vertebral arteries. A superior saturation band is of the vertebral arteries depends on the clinical
commonly applied to eliminate jugular venous context. In cases of suspected vertebral artery
flow. However, it should be noted that a superior occlusion or dissection, CTA of the head and
saturation band can potentially hide reversed ver- neck is a reasonable initial examination, providing
tebral artery flow. Thus, in cases of suspected rapid visualization of the vertebral arteries with
subclavian steal syndrome or bow hunter’s syn- excellent spatial resolution, thus potentially min-
drome, a phase contrast sequence can be imizing the time to revascularization. However, in
performed to unmask reversal of flow. The general cases of subclavian steal syndrome or bow
limitations of time-of-flight MRA include hunter’s syndrome, dynamic imaging with
overestimation of stenosis in the setting of vessel contrast-enhanced MRA may be preferred.
calcification, vessel tortuosity, or slow flow. Finally, in patients with tenuous renal function,
Dynamic contrast-enhanced MRA is able to time-of-flight MRA may be selected in order to
172 D. Chiao and M. Wintermark

avoid contrast agents and the risk of nephrogenic Pathophysiology


systemic fibrosis.
Atherosclerosis is a complex pathologic process
with endothelial injury playing a central role in its
Atherosclerotic Disease pathogenesis [8, 9]. Endothelial injury results in
increased permeability of the arterial wall to
Epidemiology and Clinical Findings lipids, which deposit in the intima resulting in
the fibrofatty plaque. Microscopically, the
The most common nontraumatic condition affect- fibrofatty plaque is composed of lipids, foam
ing the extracranial vertebral arteries is atheroscle- cells, and connective tissue. A fibrotic cap, com-
rotic disease [6]. Atherosclerosis is a systemic posed of smooth muscle and collagen, forms over
arterial disease, prevalent in industrialized the surface of the fibrofatty plaque. Disruption of
nations. It is responsible for the majority of ische- the fibrotic cap results in exposure of the
mic strokes, resulting in healthcare-associated thrombogenic fibrofatty plaque to the blood-
costs over $30 billion each year [7]. There are stream, which can result in acute thrombosis
multiple well-known risk factors for atherosclero- and/or thromboembolus [9]. In addition, as the
sis, both modifiable and nonmodifiable. Common fibrofatty plaque enlarges, it progressively nar-
nonmodifiable risk factors include family history, rows the arterial lumen. In general, a 50 %
age, and male gender. Common modifiable risk decrease in luminal diameter is required before
factors include age, hypertension, hyperlipidemia, hemodynamic consequences occur, as compensa-
diabetes, smoking, and renal failure (Table 1) [8]. tory physiologic vasodilation mitigates the effects
There is a strong association between vertebral of mild atherosclerotic disease [9]. It should be
artery atherosclerosis and carotid artery athero- noted that tandem stenoses can be additive in their
sclerosis; however, the vertebral arteries tend to hemodynamic effects, much like serial resistances
be less severely and less diffusely affected than in a circuit [8]. Ultimately, clinical symptoms
the carotid arteries [6]. occur when the arterial demand outstrips the arte-
Most patients with vertebral artery atheroscle- rial supply, resulting in a demand-supply
rosis are asymptomatic; however, if the degree of mismatch and end-organ ischemia. If the
vertebral artery stenosis is significant, patients can mismatch is transient and infarction has not
present with vertebrobasilar insufficiency which occurred, then the clinical manifestations are tran-
can manifest as vertigo, ataxia, imbalance, syn- sient and the episode is referred to as a transient
cope, and cranial nerve deficits [8]. Patients can ischemic attack. However, if the mismatch is
also present with a myriad of non-neurologic prolonged and infarction has occurred, then the
symptoms related to systemic atherosclerosis, clinical manifestations are prolonged and the epi-
including angina from coronary artery disease sode is referred to as a cerebrovascular accident or
and claudication from peripheral vascular disease. stroke. Patients with prior transient ischemic
attacks have a ten times increased risk of
stroke [9].
Table 1 Risk factors for atherosclerotic disease
Nonmodifiable Family history
Imaging Findings
Age
Male gender
Vertebral artery stenosis severity is categorized
Modifiable Hypertension
per the North American Symptomatic Carotid
Hyperlipidemia
Diabetes
Endarterectomy Trial (NASCET) method
Smoking [10]. The NASCET method defines the percent
Renal failure stenosis as the ratio of the difference between the
normal and diseased luminal diameter to the
10 Imaging of the Pathology of the Vertebral Arteries 173

Table 2 Grading of atherosclerotic disease


Mild <50 %
Moderate 50–69 %
Severe 70 %

normal luminal diameter (i.e., percent stenosis =


(normal lumen minimal diseased lumen) / (nor-
mal lumen)  100%). Flow-limiting atheroscle-
rotic disease is graded by the percent stenosis,
with mild narrowing <50 %, moderate narrowing
50–70 %, and severe narrowing >70 % (Table 2).
Irregular atheromatous plaques are at an increased
risk of thromboembolism and are considered an
independent risk factor for stroke [4].
Ultrasound with pulsed Doppler has a very
limited role in evaluating vertebral artery athero-
Fig. 14 CTA demonstrating moderate stenosis of the left
sclerosis, largely due to its inability to visualize
vertebral artery origin (arrow) due to ostial calcified ath-
the vertebral artery in its entirety. As with other erosclerotic disease
blood vessels, arterial stenosis is characterized on
ultrasound by increased velocities, turbulence,
high-resistance waveforms, and elevated resistive
indices [6, 8, 11]. The post-stenotic segment is
characterized by decreased velocities, loss of
pulsatility, “parvus-tardus” waveforms, and
decreased resistive indices. Severe stenosis in
one vertebral artery can result in overestimation
in stenosis of the contralateral vertebral artery due
to compensatory increased blood flow [9].
CTA or MRA is often the initial diagnostic
examination for vertebral artery atherosclerosis,
with >90 % sensitivity and >90 % specificity
[5, 8, 9]. Both imaging modalities offer complete
visualization of the vertebral arteries and allow for
routine NASCET characterization of atheroscle-
rotic disease. In general, atherosclerosis is charac-
terized by smooth or irregular plaques resulting in
luminal narrowing, often with concomitant arte-
rial wall calcification [8]. Atherosclerotic disease
is typically most severe at the ostia of the vertebral
arteries (Fig. 14). Multiplanar reformats and/or Fig. 15 CTA demonstrating severe stenosis of the left
extracranial vertebral artery (arrow) due to atherosclerotic
3D vessel analysis can be useful in accurately disease
quantifying the degree of arterial stenosis, espe-
cially in vessel segments which do not run orthog- artery (Figs. 15 and 16 respectively). However,
onal to the axial plane. if the clinical suspicion for significant atheroscle-
Advanced atherosclerotic disease can manifest rosis is low (e.g., young patient without athero-
as near-occlusion or occlusion of the vertebral sclerotic risk factors), other causes of vertebral
174 D. Chiao and M. Wintermark

artery stenosis should be considered, such as ver- generally reserved for planning for endovascular
tebral artery dissection, radiation arteritis, interventions (Fig. 18).
fibromuscular dysplasia, or vasculitis [8]. In
these cases, MRA may provide additional infor-
mation by directly visualizing fibrofatty athero- Work-Up and Management
sclerotic plaque and excluding other causes of
luminal narrowing (Fig. 17). Lastly, the role of The management of vertebral artery atheroscle-
DSA in evaluating atherosclerotic disease is rotic disease has historically been with medical
therapy targeting atherosclerotic risk factors such
as hypertension or hypercholesterolemia. Sur-
gery, while frequently performed for carotid
artery atherosclerosis, is rarely performed for
vertebral artery atherosclerosis [12]. With the
advent of minimally invasive endovascular tech-
niques, angioplasty and stenting now have a role
in patients who fail maximum medical therapy.
Endovascular treatment involves aggressive pro-
cedural anticoagulation and deployment of a self-
expanding stent across the stenotic lesion. The
angiographic success rate of endovascular tech-
niques has been reported to be up to 95 %, with a
stroke rate of <2 % [9]. However, endovascular
stenting suffers from restenosis and neointimal
hyperplasia which often occurs at the margin of
previously placed stents (Fig. 19) [5]. Drug-
Fig. 16 CTA demonstrating complete occlusion of the eluting stents and other methods of reducing
right vertebral artery (arrow)

Fig. 17 Axial (a) and


coronal (b) MRA of the
neck demonstrating a
fibrofatty atherosclerotic
plaque (arrows) of the left
vertebral artery
10 Imaging of the Pathology of the Vertebral Arteries 175

Fig. 18 (a) Right vertebral


artery DSA demonstrates a
high-grade stenosis of V4.
(b) Right vertebral artery
DSA after angioplasty and
placement of a 2  8 mm
chromium cobalt stent
demonstrates an excellent
angiographic result

Fig. 19 (a) Right vertebral


artery DSA 2 years later in
the same patient as in
Fig. 18 demonstrates a >90
% stenosis at the distal
aspect of the previously
placed stent. (b) Right
vertebral artery DSA after
angioplasty and placement
of a second 2  8 mm
chromium cobalt stent
demonstrates an excellent
angiographic result

restenosis and neointimal hyperplasia are cur- fossa ischemic strokes [4, 13]. The mean age at
rently being developed and studied. presentation is approximately 45 years; there is a
slight male predominance [4, 14, 15]. Patients can
present with dizziness/vertigo, headache, neck
Vertebral Artery Dissection pain, ataxia, visual symptoms, nausea/vomiting,
nystagmus, Horner syndrome, sensory deficits,
Epidemiology and Clinical Findings cranial nerve palsies, dysphagia, and tinnitus
(Table 3) [13, 16]. In a meta-analysis of nearly
Vertebral artery dissection has an annual inci- 2,000 patients, Gottesman et al. found that the
dence of 1 per 100,000 patients and is one of the most common symptom was dizziness/vertigo
most common causes of stroke in young adult (58 %), followed by headache (51 %) and neck
patients [4]. Vertebral artery dissection constitutes pain (48 %) [16]. However, it should be noted that
approximately 27 % of all craniocervical arterial nearly one in four patients with proven vertebral
dissections and causes up to 40 % of all posterior artery dissection have no headache or neck pain at
176 D. Chiao and M. Wintermark

Table 3 Signs and symptoms of vertebral artery dissec- Table 5 Risk factors for vertebral artery dissection [16]
tion, sorted by frequency [16]
Trauma
Dizziness/vertigo 58 % Hypertension
Headache 51 % Connective tissue disorders
Neck pain 46 % Fibromuscular dysplasia
Ataxia 38 % Vasculitis
Visual symptoms 36 % Autosomal dominant polycystic kidney disease
Nausea/vomiting 35 % Hyperhomocysteinemia
Nystagmus 29 % Osteogenesis imperfecta
Horner syndrome 22 %
Sensory deficits 21 %
Cranial nerve palsies 21 %
Major trauma (e.g., motor vehicle accident or
Dysphagia 13 %
Tinnitus 7%
penetrating injury) is the single most important
risk factor for vertebral artery dissection. How-
ever, even minor trauma (e.g., abrupt head turn-
ing, sports injury, and chiropractic injury) can
Table 4 Complications of vertebral artery dissection, result in vertebral artery dissection, being reported
sorted by location [16] in 15–70 % of cases [16]. Other risk factors
Stroke Any 60 % include hypertension, connective tissue disorders,
Extracranial 68 % fibromuscular dysplasia, and vasculitis (Table 5)
Intracranial 32 % [17]. When dissection occurs in the absence of
TIA Any 14 % trauma, it is referred to as spontaneous vertebral
Extracranial 21 % artery dissection. While rare, spontaneous verte-
Intracranial 11 %
bral artery dissection is a potentially devastating
SAH Any 35 %
event because delayed diagnosis can result in poor
Extracranial 0%
outcomes.
Intracranial 57 %

Pathophysiology

the time of diagnosis; thus, the absence of these Vertebral artery dissection results from a primary
symptoms do not exclude the diagnosis. tear of the intima, which allows blood to travel
The major complication of vertebral artery dis- from the lumen to the tunica media. The dissec-
section is cerebral infarction due to vertebral tion flap extends cranially (i.e., with the direction
artery occlusion; this occurs in approximately of blood flow) and forms a false lumen which can
60 % of patients [16]. The risk of stroke is highest compress and obstruct the true lumen resulting in
in the first few weeks after dissection but can cerebrovascular ischemia. The dissection flap can
occur up to a month after dissection [16]. Ischemic also extend into the adventitia resulting in
events are particularly common in extracranial pseudoaneurysm formation. In addition, vertebral
vertebral artery dissection with stroke occurring artery dissection can result in vascular stasis
in 68 % of patients with extracranial vertebral and become a nidus for distal thromboembolism
artery dissection compared to 32 % in intracranial [4, 18].
vertebral artery dissection [16]. In contrast, sub- The extracranial vertebral segments are more
arachnoid hemorrhage occurs almost exclusively prone to dissection than the intracranial segments
in intracranial vertebral artery dissection with an due to their greater mobility and proximity to
incidence of approximately 60 % in patients with adjacent bony structures (Table 6) [16, 19]. How-
intracranial vertebral artery dissection ever, the intracranial segments are more prone to
(Table 4) [16]. rupture due to their thinner adventitia [4].
10 Imaging of the Pathology of the Vertebral Arteries 177

Table 6 Vertebral artery dissection by vertebral artery Table 8 Outcomes after VAD by MRS [16]
segment [16]
MRS 0–1 67 %
V1 28 % MRS 2–4 18 %
V2 34 % MRS 5–6 10 %
V3 36 % MRS modified Rankin scale, MRS 0 no symptoms, MRS 1
V4 34 % no significant disability, MRS 2 slight disability, MRS 3
moderate disability, MRS 4 moderately severe disability,
MRS 5 severe disability, MRS 6 death

Table 7 Direct and indirect signs of vertebral artery dis-


section [19] Two patterns of vertebral artery dissection
Direct Double lumen 22 % have been described in the literature: the steno-
Intimal flap 21 % occlusive pattern and the dissecting aneurysmal
Indirect Arterial stenosis 51 % pattern [4, 20]. The steno-occlusive pattern is due
String of pearls 48 % to subintimal dissection resulting in narrowing of
Arterial dilatation 37 % the true lumen and is characterized by a tapered
Arterial occlusion 36 % stenosis or occlusion; long-segment stenosis has
Pseudoaneurysm 22 % been referred to as the “string” sign. This pattern is
typically seen in extracranial vertebral artery dis-
section and is associated with thromboembolic
phenomenon (Fig. 20) [20]. The dissecting aneu-
Most cases of vertebral artery dissection heal rysmal pattern is due to subadventitial extension
spontaneously with 90 % of patients having sig- of the dissection flap and is characterized by focal
nificant improvement in the first 2–3 months fusiform aneurysmal dilatation with or without
[4]. Vertebral artery dissection has an overall stenosis; alternating stenosis and dilatation has
recurrence rate of 15 %, with 50 % of cases been referred to as the “string-of-pearls” sign.
recurring in the first month [4]. This pattern is typically seen in intracranial verte-
bral artery dissection and is associated with sub-
arachnoid hemorrhage (Fig. 21) [4]. A very specific
Imaging Findings appearance of vertebral artery dissection is a nar-
row, eccentric lumen surrounded by crescent-
Given the variable and often nonspecific clinical shaped mural thickening [22, 23, 24, 27]. In rare
presentation of vertebral artery dissection, imag- cases, the lumen is preserved but is surrounded by
ing plays a primary role in diagnosis. Imaging hyperdense intramural hematoma, referred to as the
findings of vertebral artery dissection can be clas- “rind” sign [4, 21].
sified as direct or indirect (Table 7). Direct find- Conventional angiography has historically
ings include visualization of a double lumen or an been considered the “gold standard” in the diag-
intimal flap. Indirect findings include arterial ste- nosis of vertebral artery dissection, though recent
nosis, dilatation, alternating stenosis and dilata- studies have demonstrated that CTA has a near
tion (“string of pearls”), occlusion, or 100 % sensitivity and specificity when compared
pseudoaneurysm formation. Overall, the most to either conventional angiography or with the
common finding in vertebral artery dissection is final clinical diagnosis [21]. Today, CTA is typi-
vertebral artery stenosis, which is seen in approx- cally the initial diagnostic examination for the
imately half of patients [19]. While direct signs of evaluation of acute vertebral artery dissection.
vertebral artery dissection are more intuitive than MRA is comparable to CTA but takes longer to
indirect signs, they are seen in less than one in four perform and is generally indicated for the
patients with proven vertebral artery dissection non-emergent evaluation of vertebral artery dis-
[19] (Table 8). section [19]. One advantage of MRA over CTA is
178 D. Chiao and M. Wintermark

Fig. 20 Forty-three-year-old female with vertebral artery projections demonstrating abrupt narrowing and irregular-
dissection status post-motor vehicle collision. CTA of the ity of the right extracranial vertebral artery (arrows). Note
neck with axial (a) and coronal (b) maximum-intensity the adjacent transverse foramina fractures (open arrows)

Fig. 21 Forty-eight-year-
old female with
spontaneous intracranial
vertebral artery dissection
after awakening with severe
headache. (a) NECT
demonstrating diffuse
subarachnoid hemorrhage
and intraventricular blood.
(b) CTA demonstrating
occlusion of the right V4
segment (arrow); the left
vertebral artery is patent
(open arrow)

its superior ability of visualizing an intramural occlusion is seen in 20 % of cases, and an isolated
hematoma, which is characterized as an eccentric aneurysm is seen in 7 % of cases [4, 27].
region of high T1 signal intensity, due to methe-
moglobin (Fig. 22) [20]. Another advantage of
MRA is its greater sensitivity for acute infarcts, Work-Up and Management
the major complication of vertebral artery dissec-
tion (Fig. 23). Because patients with vertebral artery dissection
The imaging findings of vertebral artery dis- can present with nonspecific symptoms and often
section on angiography are similar to CTA/MRA. do not have significant risk factors (e.g., major
The characteristic finding is an abrupt caliber trauma or connective tissue disorders), a high
change (Fig. 24): an isolated stenosis is seen in level of clinical suspicion is needed for diagnosis.
46 % of cases, a stenosis with a concomitant One possible algorithm for the work-up of vertebral
aneurysm is seen in 27 % of cases, an isolated artery dissection is given below (Fig. 25) [19].
10 Imaging of the Pathology of the Vertebral Arteries 179

Fig. 22 Forty-two-year-
old male with spontaneous
vertebral artery dissection
after “thunderclap”
headache. (a) T1-WI with
fat saturation demonstrating
hyperintense intramural
hematoma (arrow). (b) 3D
contrast-enhanced MRA
demonstrating long-
segment stenosis (“string
sign”) of the left vertebral
artery

Fig. 23 MRI/MRA of the


head in the same patient. (a)
Time-of-flight MRA
maximum-intensity
projection demonstrating an
abrupt cutoff of the left
P1-P2 junction (arrow).
DWI and ADC mapping (b,
c) demonstrating a left
posterior cerebral artery
territory infarct. Vertebral
artery dissection serves as a
nidus for thromboembolism

In this algorithm, patients with normal renal func- therapeutic intervention in extracranial dissec-
tion and no contrast allergy are evaluated with a tions [18]. In intracranial dissections, the risks of
CTA; patients with abnormal renal function or a anticoagulation are higher because of the
documented contrast allergy are evaluated with a increased incidence of subarachnoid hemorrhage.
time-of-flight MRA. Angiography is reserved for Endovascular or surgical intervention can be con-
troubleshooting in equivocal cases. sidered in selected patients.
Treatment of vertebral artery dissection is con- Follow-up imaging is often performed to eval-
troversial because randomized controlled studies uate for stability and/or resolution of vertebral
are lacking. Anticoagulation is the most frequent artery dissection (Fig. 26). On occasion, vertebral
180 D. Chiao and M. Wintermark

Fig. 24 Angiogram pre- (a) and post- (b) coil emboliza- representing thrombus formation. After coil embolization
tion of a right intracranial vertebral artery dissection (open of the right vertebral artery, the posterior circulation is
arrow) which involves the posterior inferior cerebellar supplied entirely by the left vertebral artery
artery. Note the intraluminal filling defect (arrow),

mainly in arteries not previously involved by the


dissection [18].
The clinical outcome of vertebral artery dissec-
tion is a function of the degree of cerebral ische-
mia caused by the dissection. Given that the
morbidity and mortality associated with vertebral
artery dissection is typically due to ischemic
infarcts, the modified Rankin scale (MRS) has
been used to categorize patient outcomes
(Table 6). Overall, patients have relatively good
outcomes after vertebral artery dissection, with
two-thirds of patients without significant perma-
nent disability [16].

Subclavian Steal Syndrome

Epidemiology and Clinical Findings


Fig. 25 An imaging work-up algorithm for vertebral
artery dissection The subclavian steal syndrome refers to the devel-
opment of neurologic symptoms secondary to
artery dissection can progress and/or convert into retrograde flow in a vertebral artery; it was first
pseudoaneurysms, necessitating endovascular or recognized by Contorni in 1960 and Reivich in
surgical intervention. In addition, follow-up imag- 1961 [29]. Retrograde flow without neurologic
ing is important for evaluating for recurrent ver- symptoms has been termed subclavian steal phe-
tebral artery dissection or delayed infarcts. Most nomenon. The Joint Study of Extracranial Arterial
recurrences occur within the first month and Occlusion reported that the incidence of subcla-
involve multiple cervical arteries [28]. After the vian steal phenomenon is approximately 2.5 %,
first month, the risk of recurrence is only about with only 5 % of patients with subclavian steal
1 % per year, and recurrent dissection occurs phenomenon presenting with neurologic
10 Imaging of the Pathology of the Vertebral Arteries 181

Fig. 26 CTA of the neck at


initial presentation (a) and
at 5-month follow-up (b)
after conservative
management of extracranial
vertebral artery dissection.
There is interval resolution
of the vertebral artery
stenosis (arrows)

Percentage of patients with neurologic symptoms

Fig. 27 Prevalence of 40%


neurologic symptoms in
patients with subclavian 35%
steal syndrome by brachial
blood pressure differential 30%

25%

20%

15%

10%

5%

0%
20-30 30-40 40-50 >50
Brachial blood pressure differential (mmHg)

symptoms [30]. The average age at presentation is cerebellum, and posterior cerebral hemispheres.
60 with a slight male predominance [4]. Because there is usually a proximal subclavian
Vertebrobasilar symptoms arise when there is artery stenosis resulting in the subclavian steal
inadequate blood flow to the posterior circulation phenomenon, patients can also present with ipsilat-
due to significant retrograde flow. Patients may eral ischemic arm symptoms such as arm claudica-
experience vertigo, ataxia, imbalance, and syn- tion, weakness, and paresthesias [4]. In fact, an
cope; in addition, cranial nerve deficits can occur important clinical marker for subclavian steal syn-
resulting in dysarthria, dysphagia, tinnitus, and drome is asymmetric brachial pressures [33, 34]. A
visual deficits [31–33]. Subclavian steal syndrome brachial blood pressure differential of greater than
rarely results in acute infarction because of the 20 mmHg is considered clinically significant, with
collateral blood supply from the contralateral ver- 55–85 % of cases demonstrating subclavian steal
tebral artery [4]. However, if the contralateral ver- phenomenon [33]. Increasing arm blood pressure
tebral artery is significantly diseased, patients are at differentials correlates with increasing incidence of
increased risk of infarction of the brainstem, subclavian steal syndrome (Fig. 27) [33].
182 D. Chiao and M. Wintermark

Pathophysiology Table 9 Grading of subclavian steal syndrome [33]


Grade I Reduced antegrade flow
As discussed above, the subclavian steal phenom- Grade II Alternating antegrade and retrograde flow
enon is typically due to a proximal, high-grade Grade III Permanent retrograde flow
subclavian artery stenosis or occlusion, resulting
in decreased distal subclavian artery pressures.
Vigorous exercise of the arm accentuates this Provocative maneuvers, such as ipsilateral arm
pressure gradient and provokes retrograde flow exercise or the hyperemia-ischemia cuff test,
from the ipsilateral vertebral artery, effectively may be employed to demonstrate blood flow
“stealing” blood flow from the posterior reversal.
circulation [4]. On Doppler ultrasound, the normal vertebral
The etiology of the inciting subclavian artery arteries demonstrate a low-resistance pattern with
stenosis is most commonly atherosclerosis cephalad flow throughout the cardiac cycle. In
[30]. In a study of over 7,000 neck ultrasounds, patients with subclavian steal syndrome, there is
Labropoulos et al. demonstrated that more than blunted antegrade or frank retrograde flow on the
80 % of cases of subclavian steal phenomenon affected side, often with compensatory increased
were due to atherosclerosis of the left subclavian flow (peak systolic velocity >60 cm/s) on the
artery [35]. This is thought to be due to the acute contralateral side [4, 36, 38, 39–42]. A grading
angle that the left subclavian artery takes as it system based on waveform characteristics has
arises off the aortic arch, predisposing to flow been developed, with grade I (mild steal) charac-
turbulence and atherosclerotic disease. Rarely, terized by reduced antegrade flow, grade II (mod-
subclavian steal syndrome can result from vascu- erate steal) characterized by alternating antegrade
litis, radiation angiopathy, aortic dissection, and retrograde flow, and grade III (severe steal)
extrinsic compression, or congenital stenosis [4]. characterized by permanent retrograde flow
A unique scenario can occur in patients with a (Table 9). The hyperemia-ischemia cuff test is
history of CABG with LIMA coronary bypass. In often used to provoke subclavian steal. The
these patients, the proximal subclavian artery ste- hyperemia-ischemia cuff test involves inflating a
nosis results in vascular steal not only from the blood pressure cuff at least 20 mmHg above the
ipsilateral vertebral artery but also from the LIMA systolic blood pressure for several minutes and
coronary bypass which can result in coronary then rapidly deflating the cuff with real-time
ischemia [36]. These patients may present with Doppler ultrasound of the ipsilateral vertebral
angina, especially after vigorous exercise of the artery. The rapid cuff deflation leads to transient
affected arm. increased blood flow to the arm, which can pro-
voke occult subclavian steal [36]. Infrequently,
the proximal subclavian artery stenosis can be
Imaging Findings visualized on Doppler ultrasound, characterized
by a high-resistance waveform, increased veloc-
Subclavian steal syndrome can be assessed with a ity, and turbulent flow.
variety of imaging modalities including ultra- While Doppler ultrasound is excellent at dem-
sound, CTA, MRA, and DSA. At some institu- onstrating the directional flow in the vertebral
tions, Doppler ultrasound is used as an initial arteries, it usually cannot visualize the proximal
screening test due to its availability and low cost subclavian artery stenosis. In these cases, cross-
[37]. CTA, MRA, and/or DSA are then performed sectional imaging with MRA or CTA can be
as confirmatory studies. The characteristic finding performed. The characteristic finding on cross-
is reversed flow in a vertebral artery with a prox- sectional imaging is a high-grade stenosis or
imal subclavian artery stenosis or occlusion. occlusion in the subclavian artery proximal to
10 Imaging of the Pathology of the Vertebral Arteries 183

Fig. 28 Forty-eight-year-
old female with history of
radiation therapy for
non-small cell lung cancer
who presents with
paroxysms of vertigo. (a)
CTA of the head and neck
demonstrating complete
occlusion of the proximal
subclavian artery (arrow).
(b) DSA with injection of
the left subclavian artery
showing the same finding
(arrow)

Fig. 29 Same patient as in


Fig. 28. (a) DSA with
injection of the right
vertebral artery. (b) Delayed
imaging demonstrates
retrograde filling of the left
vertebral artery (arrow)
with opacification of the left
subclavian artery (open
arrow), compatible with
subclavian steal
phenomenon

the takeoff of the vertebral artery (Figs. 28 and 29) “to-and-fro” flow in the affected vertebral artery,
[4]. Fibrofatty or calcified atherosclerotic plaque usually with a high-grade proximal subclavian
is a typical finding and may appear irregular artery stenosis (Fig. 32) [4].
and/or ulcerated. MRA is generally favored over
CTA because of its ability to visualize proximal
subclavian artery stenosis as well as its ability to Work-Up and Management
determine the direction of blood flow in the ver-
tebral arteries, especially with a phase contrast Only a small minority (approximately 18 %) of
sequence (Figs. 30 and 31). patients with subclavian steal phenomenon
As with most vascular pathology, angiography require therapeutic intervention [35]. Patients
is usually reserved for equivocal cases or prior to with mild symptoms are generally treated conser-
endovascular treatment. The characteristic finding vatively with medical treatment of the underlying
in subclavian steal syndrome is reversal of flow or atherosclerotic risk factors (such as
184 D. Chiao and M. Wintermark

Fig. 30 Fifty-five-year-old female with weakness and Fig. 31 Eighty-three-year-old male with lower extremity
paresthesias in her left hand after prolonged use. ataxia. 3D contrast-enhanced MRA demonstrates left sub-
Contrast-enhanced MRA maximum-intensity projection clavian artery occlusion (arrow)
demonstrates proximal left subclavian artery occlusion
(arrow)

Bow Hunter’s Syndrome


hyperlipidemia, hypertension, diabetes, or
smoking) [43]. Patients with severe symptoms Epidemiology and Clinical Findings
may require surgical or endovascular treatment
with carotid-subclavian bypass grafting or angio- Bow hunter’s syndrome, also known as positional
plasty/stenting. Surgical bypass has enjoyed wide or rotational occlusion of the vertebral artery, is a
success, with resolution of symptoms in the vast rare cause of vertebrobasilar insufficiency. The
majority of patients and patency rates of up to syndrome was originally described by Dr. Bruce
95 % at 10 years [44]. More recently, Sorensen in a case report in Neurosurgery in 1978
endovascular techniques have also been [48]. Sorensen described a 39-year-old man who
employed successfully, with patency rates of developed persistent right-sided weakness and
80 % at 5 years (Fig. 33) [45, 46]. Some authors left-sided numbness during archery practice.
argue that endovascular techniques are preferred Based on clinical findings, the patient was thought
over surgical techniques due to their similar effec- to have right medullary infarction resulting in
tiveness, lower complication rates, and shorter modified Wallenberg syndrome. However, the
hospitalizations; however, long-term patency is patient had no significant atherosclerotic risk fac-
less durable with endovascular therapy, with a tors. On angiography, the patient was found to
10-year target vessel revascularization rate of have significant right vertebral artery spasm.
47 % compared to 6 % for bypass [47]. Sorensen reasoned that the patient’s condition
10 Imaging of the Pathology of the Vertebral Arteries 185

Fig. 32 Sixty-year-old male with intermittent episodes of vertebral artery (arrow) with delayed opacification of the
diplopia and dizziness. (a) DSA with injection of the aortic right subclavian artery (open arrow). Right-sided subcla-
arch. The right subclavian artery is not seen. (b) Subse- vian steal syndrome is less common than left-sided subcla-
quent imaging demonstrates retrograde filling of the right vian steal syndrome

with their heads turned to the left (if right-


handed), the phenomenon of positional vertebral
artery occlusion is accentuated and could poten-
tially result in ischemia or infarction [48].
Since Sorensen’s original case report, at least
100 cases of bow hunter’s syndrome have been
reported in the literature [49]. The age at presen-
tation ranges widely, with most patients in their
40–60s [49, 50]. Patients typically present with
signs and symptoms of vertebrobasilar insuffi-
ciency, such as weakness, ataxia, numbness, tin-
gling, dysphagia, dysarthria, tinnitus, diplopia,
and nystagmus [4, 51]. Patients can also present
with nonspecific symptoms such as headache,
dizziness, nausea, and syncope [4, 52]. The key
finding is that clinical symptoms occur after a
sharp turn of the head.

Pathophysiology
Fig. 33 Same patient as in Fig. 32. DSA with injection of
the innominate artery after endovascular stent placement
(arrow) demonstrating antegrade flow in the right vertebral Bow hunter’s syndrome is thought to arise due to
(open arrow) and subclavian arteries positional occlusion of the contralateral vertebral
artery after sharply turning the head. In general,
stemmed from positional occlusion of the verte- transient positional vertebral artery occlusion is
bral artery during archery practice (i.e., turning considered physiologic and is asymptomatic due
the head sharply to the left will occlude the right to sufficient contralateral vertebral artery flow or
vertebral artery and vice versa). In archers and collateral flow via the posterior communicating
bow hunters, who stand perpendicular to a target arteries [50]. However, if the contralateral
186 D. Chiao and M. Wintermark

Fig. 34 DWI (a) and ADC


mapping (b) demonstrating
an area of restricted
diffusion in the right
thalamus, concerning for an
acute infarct

vertebral artery is hypoplastic or significantly dis- CTA can visualize areas of stenosis or occlusion.
eased, bow hunter’s syndrome is more likely to In addition, cross-sectional imaging has the
occur [50, 51]. Bow hunter’s syndrome is also advantage of evaluating extravascular pathology
more likely if the affected vertebral artery origi- which may explain or contribute to the patient’s
nates the dominant posterior inferior cerebellar symptoms. Complications of bow hunter’s syn-
artery [52]. drome, such as acute stroke, can also be visualized
Most cases of bow hunter’s syndrome occur at (Figs. 34 and 35).
the atlantoaxial level, which is especially prone to The “gold standard” for diagnosis remains
dynamic injury [4]. During neck rotation, the con- dynamic angiography with head turning
tralateral atlantoaxial joint rotates asymmetrically [52]. Angiography has the advantage of real-
forward and downward, potentially stretching the time visualization of stenosis or occlusion and
vertebral artery between the C1 and C2 foramina can demonstrate direction of flow in collateral
or under the atlantooccipital ligament [50]. Cervi- vessels (Fig. 36). Angiography is also frequently
cal spondylosis, herniated disks, and atlantoaxial performed to demonstrate angiographic improve-
instability can predispose to vertebral artery com- ment after an intervention is performed [53].
pression [50, 52]. In addition, cases of vertebral
artery compression by the longus colli and scalene
muscles have been reported [49]. Work-Up and Management

While bow hunter’s syndrome is rare, it can result


Imaging Findings in repeated vertebral artery ischemia and/or per-
manent neurologic deficits if not diagnosed and
Evaluation of bow hunter’s syndrome relies on treated appropriately. Work-up for bow hunter’s
dynamic imaging. Imaging is typically performed syndrome begins with a complete history and
in the neutral, left, and right positions. The key physical examination. In particular, a thorough
imaging finding is occlusion or stenosis of the description of the patient’s symptoms and exacer-
vertebral artery with the head turned to the left bating factors should be elicited, especially in
or right. Particular attention should be made to the patients with evidence of vertebrobasilar insuffi-
V3 segment of the vertebral artery (i.e., at the ciency. In particular, reproducibility of symptoms
atlantoaxial level), which is the most commonly with head turning is highly suggestive of bow
affected segment. hunter’s syndrome [49]. Occasionally, brainstem
Imaging is typically with MRA, CTA, or angi- auditory-evoked response testing is performed to
ography. Cross-sectional imaging with MRA or demonstrate increased latencies [51]. Dynamic
10 Imaging of the Pathology of the Vertebral Arteries 187

Fig. 35 CTA in the same


patient as in Fig. 34 with the
head turned to the left (a)
and with the head turned to
the right (b) demonstrating
positional narrowing of the
right vertebral artery
(arrow). Cervical
spondylosis, herniated
disks, and atlantoaxial
instability can predispose to
vertebral artery
compression

Fig. 36 DSA of the right


vertebral artery in the same
patient as in Fig. 34 in the
neutral position (a) and with
head turned to the left (b)
demonstrating positional
occlusion of the vertebral
artery (arrow). The V3
segment is the most
commonly affected
segment in bow hunter’s
syndrome

imaging studies can then be obtained to confirm patients, and surgical management, while effec-
the diagnosis. tive, can result in permanent restriction of neck
Patients with bow hunter’s syndrome are ini- rotation and can be complicated by a number of
tially managed with a cervical brace to restrict operative risks including infection, paralysis, and
neck rotation. Historically, medical treatment death [49]. Recently, endovascular therapy with
with anticoagulation was performed in mild self-expanding stents has been found to be an
cases and surgical decompression and/or cervical effective, minimally invasive treatment modality
spinal fusion was performed in severe cases. [52]. The downside to endovascular stenting is the
However, conservative management is effective potential for restenosis; data on long-term patency
in preventing stroke in only half of affected is unfortunately lacking. Nevertheless,
188 D. Chiao and M. Wintermark

endovascular therapy remains a treatment option Summary


and may supplant surgical management in
selected patients. The vertebral arteries are clinically important
blood vessels. Given the relatively high incidence
of vertebral artery pathology, thorough interroga-
tion of the vertebral arteries with diagnostic imag-
Miscellaneous Conditions ing is essential, with accurate interpretation
guiding management decisions. Collaboration
Fibromuscular Dysplasia among the radiology, neurology, and neurosur-
gery teams is of utmost importance. Clinical con-
Fibromuscular dysplasia is a noninflammatory text greatly aids in the selection of the most
condition, typically affecting middle-aged appropriate imaging modality. New and improved
women. It most commonly affects the renal, anatomic imaging is resulting in improved sensi-
carotid, or iliac arteries but can also affect the tivity and specificity in diagnosing vertebral artery
vertebral arteries in 10–40 % of cervical vascular diseases. In addition, dynamic imaging with or
cases. It is bilateral in two-thirds of patients. without provocative maneuvers is able to demon-
While there are several types of fibromuscular strate abnormal blood flow in functional vertebral
dysplasia, the most common type is artery disorders. Finally, interventional neuroradi-
medial fibroplasia characterized by a “string-of- ology is becoming increasingly prevalent in ther-
pearls” appearance. Fibromuscular dysplasia apeutic management, with a variety of minimally
predisposes to ischemia and infarct, likely due to invasive endovascular techniques being used in
the increased incidence of thromboembolism in the treatment of a number of vertebral artery
the affected arteries. It also predisposes to diseases.
spontaneous vertebral artery dissection. The
most sensitive examination for fibromuscular dys-
plasia is angiography, which is able to detect References
subtle cases. In symptomatic cases, treatment
with balloon angioplasty with or without stenting 1. Okahara M, Kiyosue H, Mori H, Tanoue S, Sainou M,
is warranted. Only rarely is surgical management Nagatomi H (2002) Anatomic variations of the cerebral
arteries and their embryology: a pictorial review. Eur
needed.
Radiol 12(10):2548–2561. doi:10.1007/s00330-001-
1286-x
2. Dimmick SJ, Faulder KC (2009) Normal variants of
Vasculitis the cerebral circulation at multidetector CT angiogra-
phy. Radiographics 29(4):1027–1043. doi:10.1148/
rg.294085730
Idiopathic vasculitis, including Takayasu’s arteri- 3. Luh GY, Dean BL, Tomsick TA, Wallace RC (1999)
tis and giant cell arteritis, can occasionally affect The persistent fetal carotid-vertebrobasilar anastomo-
the vertebral arteries. Vasculitis is characterized ses. AJR Am J Roentgenol 172(5):1427–1432.
doi:10.2214/ajr.172.5.10227532
by thickening of the arterial wall with long,
4. Osborn AG, Salzman KL, Barkovich AJ (2009) Diag-
smooth stenosis and/or “sausage-link” lesions. nostic imaging. Brain, 2nd edn. Amirsys, Salt Lake
Treatment is with steroids to reduce inflammatory City
injury. 5. Osborn AG, Jacobs JM, Osborn AG, Ralph Erskine
Conrad Memorial Fund (1999) Diagnostic cerebral
Radiation vasculitis can also affect the verte-
angiography, 2nd edn. Lippincott Willims & Wilkins,
bral arteries and can occur months to years Philadelphia, p 462
after radiation therapy. It is characterized by irreg- 6. Tay KY, U-King-Im JM, Trivedi RA et al (2005) Imag-
ular, calcified arteriopathy confined to the radia- ing the vertebral artery. Eur Radiol 15(7):1329–1343.
doi:10.1007/s00330-005-2679-z
tion portal. Treatment is challenging, as irradiated
7. Go AS, Mozaffarian D, Roger VL et al (2013) Execu-
vessels do not respond well to angioplasty or tive summary: heart disease and stroke statistics – 2013
surgery. update: a report from the american heart association.
10 Imaging of the Pathology of the Vertebral Arteries 189

Circulation 127(1):143–152. doi:10.1161/CIR. 24. Leclerc X, Godefroy O, Salhi A, Lucas C, Leys D,


0b013e318282ab8f Pruvo JP (1996) Helical CT for the diagnosis of extra-
8. Osborn AG (1994) Diagnostic neuroradiology. Mosby, cranial internal carotid artery dissection. Stroke
St. Louis, p 936 27(3):461–466
9. Kaufman JA, Lee MJ (2004) Vascular and interven- 25. Levy C, Laissy JP, Raveau V et al (1994) Carotid
tional radiology: the requisites. Mosby, Philadelphia, and vertebral artery dissections: three-dimensional
p 705 time-of-flight MR angiography and MR imaging ver-
10. Ferguson GG, Eliasziw M, Barr HW et al (1999) The sus conventional angiography. Radiology 190(1):
north american symptomatic carotid endarterectomy 97–103
trial: surgical results in 1415 patients. Stroke 26. Mascalchi M, Bianchi MC, Mangiafico S et al (1997)
30(9):1751–1758 MRI and MR angiography of vertebral artery dissec-
11. Vilela P, Goulao A (2005) Ischemic stroke: carotid and tion. Neuroradiology 39(5):329–340
vertebral artery disease. Eur Radiol 15(3):427–433. 27. Houser OW, Mokri B, Sundt TM Jr, Baker HL Jr,
doi:10.1007/s00330-004-2632-6 Reese DF (1984) Spontaneous cervical cephalic arte-
12. Stayman A, Nogueira RG, Gupta R (2013) Diagnosis rial dissection and its residuum: angiographic spec-
and management of vertebrobasilar insufficiency. Curr trum. AJNR Am J Neuroradiol 5(1):27–34
Treat Options Cardiovasc Med 15(2):240–251. 28. Dittrich R, Nassenstein I, Bachmann R et al (2007)
doi:10.1007/s11936-013-0228-7 Polyarterial clustered recurrence of cervical artery dis-
13. Arnold M, Bousser MG, Fahrni G et al (2006) Verte- section seems to be the rule. Neurology
bral artery dissection: presenting findings and predic- 69(2):180–186. doi:10.1212/01.wnl.0000265595.
tors of outcome. Stroke 37(10):2499–2503. 50915.1e
doi:10.1161/01.STR.0000240493.88473.39 29. Reivich M, Holling HE, Roberts B, Toole JF (1961)
14. Schievink WI, Roiter V (2005) Epidemiology of cer- Reversal of blood flow through the vertebral
vical artery dissection. Front Neurol Neurosci artery and its effect on cerebral circulation. N Engl J
20:12–15. doi:10.1159/000088125 Med 265:878–885. doi:10.1056/NEJM1961110226
15. Kristensen B, Malm J, Carlberg B et al (1997) Epide- 51804
miology and etiology of ischemic stroke in young 30. Fields WS, Lemak NA (1972) Joint study of extracra-
adults aged 18 to 44 years in northern sweden. Stroke nial arterial occlusion. VII. subclavian steal – a review
28(9):1702–1709 of 168 cases. JAMA 222(9):1139–1143
16. Gottesman RF, Sharma P, Robinson KA et al (2012) 31. Gosselin C, Walker PM (1996) Subclavian steal syn-
Clinical characteristics of symptomatic vertebral artery drome: existence, clinical features, diagnosis and man-
dissection: a systematic review. Neurologist agement. Semin Vasc Surg 9(2):93–97
18(5):245–254. doi:10.1097/NRL.0b013e31826754e1 32. Herring M (1977) The subclavian steal syndrome: a
17. Schievink WI (2001) Spontaneous dissection of the review. Am Surg 43(4):220–228
carotid and vertebral arteries. N Engl J Med 344 33. Osiro S, Zurada A, Gielecki J, Shoja MM, Tubbs RS,
(12):898–906. doi:10.1056/NEJM200103223441206 Loukas M (2012) A review of subclavian steal syn-
18. Rodallec MH, Marteau V, Gerber S, Desmottes L, Zins drome with clinical correlation. Med Sci Monit 18(5):
M (2008) Craniocervical arterial dissection: spectrum RA57–RA63
of imaging findings and differential diagnosis. Radio- 34. Tan TY, Schminke U, Lien LM, Tegeler CH (2002)
graphics 28(6):1711–1728. doi:10.1148/rg.286085512 Subclavian steal syndrome: can the blood pressure
19. Gottesman RF, Sharma P, Robinson KA et al (2012) difference between arms predict the severity of steal?
Imaging characteristics of symptomatic vertebral artery J Neuroimaging 12(2):131–135
dissection: a systematic review. Neurologist 35. Labropoulos N, Nandivada P, Bekelis K (2010) Prev-
18(5):255–260. doi:10.1097/NRL.0b013e3182675511 alence and impact of the subclavian steal syndrome.
20. Shin JH, Suh DC, Choi CG, Leei HK (2000) Vertebral Ann Surg 252(1):166–170. doi:10.1097/SLA.
artery dissection: spectrum of imaging findings with 0b013e3181e3375a
emphasis on angiography and correlation with clinical 36. Kliewer MA, Hertzberg BS, Kim DH, Bowie JD,
presentation. Radiographics 20(6):1687–1696 Courneya DL, Carroll BA (2000) Vertebral artery
21. Chen CJ, Tseng YC, Lee TH, Hsu HL, See LC (2004) doppler waveform changes indicating subclavian steal
Multisection CT angiography compared with catheter physiology. AJR Am J Roentgenol 174(3):815–819.
angiography in diagnosing vertebral artery dissection. doi:10.2214/ajr.174.3.1740815
AJNR Am J Neuroradiol 25(5):769–774 37. Vecera J, Vojtisek P, Varvarovsky I, Lojik M,
22. Bartels E, Flugel KA (1996) Evaluation of extracranial Masova K, Kvasnicka J (2010) Non-invasive diagnosis
vertebral artery dissection with duplex color-flow of coronary-subclavian steal: role of the doppler ultra-
imaging. Stroke 27(2):290–295 sound. Eur J Echocardiogr 11(9):E34. doi:10.1093/
23. Sturzenegger M, Mattle HP, Rivoir A, Baumgartner ejechocard/jeq068
RW (1995) Ultrasound findings in carotid artery dis- 38. Buckenham TM, Wright IA (2004) Ultrasound of the
section: analysis of 43 patients. Neurology extracranial vertebral artery. Br J Radiol 77(913):
45(4):691–698 15–20
190 D. Chiao and M. Wintermark

39. Flynn PD, Delany DJ, Gray HH (1993) Magnetic res- 47. Song L, Zhang J, Li J et al (2012) Endovascular
onance angiography in subclavian steal syndrome. stenting vs. extrathoracic surgical bypass for symptom-
Br Heart J 70(2):193–194 atic subclavian steal syndrome. J Endovasc Ther
40. Van Grimberge F, Dymarkowski S, Budts W, Bogaert J 19(1):44–51. doi:10.1583/11-3692.1
(2000) Role of magnetic resonance in the diagnosis of 48. Sorensen BF (1978) Bow hunter’s stroke. Neurosur-
subclavian steal syndrome. J Magn Reson Imaging gery 2(3):259–261
12(2):339–342 49. Kuether TA, Nesbit GM, Clark WM, Barnwell SL
41. Virmani R, Carroll TJ, Hung J, Hopkins J, Diniz L, (1997) Rotational vertebral artery occlusion: a mecha-
Carr J (2008) Diagnosis of subclavian steal syndrome nism of vertebrobasilar insufficiency. Neurosurgery
using dynamic time-resolved magnetic resonance angi- 41(2):427–432, discussion 432-3
ography: a technical note. Magn Reson Imaging 50. Netuka D, Benes V, Mikulik R, Kuba R (2005) Symp-
26(2):287–292. doi:10.1016/j.mri.2007.05.005 tomatic rotational occlusion of the vertebral artery –
42. Akin K, Kosehan D, Kirbas I, Yildirim M, Koktener A case report and review of the literature. Zentralbl
(2011) Diagnosis and percutaneous treatment of partial Neurochir 66(4):217–222. doi:10.1055/s-2005-
subclavian steal: doppler ultrasonography and phase 836600
contrast magnetic resonance angiography findings 51. Horowitz M, Jovin T, Balzar J, Welch W, Kassam A
and a brief review of the literature. Jpn J Radiol (2002) Bow hunter’s syndrome in the setting of
29(3):207–211. doi:10.1007/s11604-010-0521-2 contralateral vertebral artery stenosis: evaluation
43. Olsen KG, Lund C (2006) Subclavian steal syndrome. and treatment options. Spine (Phila Pa 1976)
Tidsskr Nor Laegeforen 126(24):3259–3262 27(23):E495–E498. doi:10.1097/01.BRS.0000035308.
44. Qi L, Gu Y, Zhang J et al (2010) Surgical treatment of 10464.37
subclavian artery occlusion. Zhongguo Xiu Fu Chong 52. Darkhabani MZ, Thompson MC, Lazzaro MA,
Jian Wai Ke Za Zhi 24(9):1030–1032 Taqi MA, Zaidat OO (2012) Vertebral artery
45. De Vries JP, Jager LC, Van den Berg JC et al (2005) stenting for the treatment of bow hunter’s syndrome:
Durability of percutaneous transluminal angioplasty report of 4 cases. J Stroke Cerebrovasc Dis 21(8):908.
for obstructive lesions of proximal subclavian artery: e1-908.e5. doi: 10.1016/j.jstrokecerebrovasdis.2011.
long-term results. J Vasc Surg 41(1):19–23. 09.006
doi:10.1016/j.jvs.2004.09.030 53. Velat GJ, Reavey-Cantwell JF, Ulm AJ, Lewis SB
46. Palchik E, Bakken AM, Wolford HY, Saad WE, Davies (2006) Intraoperative dynamic angiography to detect
MG (2008) Subclavian artery revascularization: an resolution of bow hunter’s syndrome: technical case
outcome analysis based on mode of therapy and report. Surg Neurol 66(4):420–423; discussion 423.
presenting symptoms. Ann Vasc Surg 22(1):70–78. doi: 10.1016/j.surneu.2006.03.040
doi:10.1016/j.avsg.2007.07.020
Carotid Artery Surgery
11
Roberto Montisci and Luca Saba

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191 Carotid endarterectomy is at present the most
frequent intervention on arterial vessels
Preoperative Study . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
performed in vascular surgery units. Currently
Surgical Technique of Endarterectomy . . . . . . . . . . . 196 the surgical technique is significantly
Carotid Body Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198 improved compared to 10 years ago with a
consequent reduction in terms of mortality
Aneurysms of the Extracranial Carotid
Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199 and morbidity. This chapter will cover the sur-
gical approaches as well as the most important
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 200
indications.
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 200
Keywords
Carotid Endarterectomy • Carotid Artery •
Carotid Artery Surgery

Introduction

Carotid endarterectomy is at present the most


frequent intervention on arterial vessels
performed in vascular surgery units. Current
wide development of this technique of reconstruc-
tion for occlusive carotid disease is a consequence
of a long arduous surgical course that had a deci-
sive impulse in the early 1950s with first success-
ful operation performed by DeBakey in 1953 on a
R. Montisci (*) 53-year-old patient with “intermittent episodes of
Department of Vascular Surgery, AOU of Cagliari, weakness of the right arm and leg, hesitancy and
Monserrato, Cagliari, Italy
difficulty in speaking, and difficulty in writing
e-mail: Roberto.montisci@iol.it
clearly” [1]. In the same period, other important
L. Saba
steps in carotid revascularization were climbed
Department of Radiology, AOU of Cagliari, Monserrato,
Cagliari, Italy through different methods like resection of the
e-mail: lucasaba@tiscali.it; lucasabamd@gmail.com segment of the internal carotid artery narrowed
# Springer Science+Business Media New York 2016 191
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_51
192 R. Montisci and L. Saba

and restoration of carotid circulation by direct introduction of embolic protection devices,


end-to-end anastomosis between the common improved the outcome of carotid stenting. In
carotid artery and the distal internal carotid artery 2010, the CREST trial assessed that “among
or using a graft after resection of a segment of the patients with symptomatic or asymptomatic
internal carotid artery (Eastcott 1954) [2]. In 1956 carotid stenosis, the risk of the composite primary
Lyons performed a subclavian to common carotid outcome of stroke, myocardial infarction, or death
graft bypass using a nylon prosthesis for a proxi- did not differ significantly in the group undergo-
mal common carotid occlusion [3]. In the same ing carotid-artery stenting and the group undergo-
year Cooley reported the first implant of a tempo- ing carotid endarterectomy” [13]. On this point,
rary shunt during carotid endarterectomy [4]. Suc- however, the debate is still open, and many cen-
cessively, the selective use of a temporary shunt ters continue to reserve CAS to selected patients
was recommended by Moore in 1969 on the basis with high risk for open surgery (hostile neck, post-
of the measurement of the stump pressure in the surgical restenosis, active coronary artery disease,
internal carotid artery after proximal cross- or congestive heart failure).
clamping and on the basis of EEG monitoring by
Collow in 1980 [5, 6].
Also anesthesia techniques shown a specific Preoperative Study
evolution from the simple general anesthesia
toward the current spectrum of possibilities that Vascular surgeons require from the vascular labo-
include local anesthesia via cervical plexus block ratory a large series of answers. In particular, a
or various intermediate techniques like conscious preoperative study of the patient with symptoms
sedation during regional anesthesia [7] or pre- of cerebral ischemia or suspected cerebral vascular
served consciousness in general anesthesia [8]. disease should be focused on identification of the
In the late 1990s, the results of some large carotid plaque and its extension on the vessel,
prospective randomized trials heavily increased evaluation of the myointimal thickness, the per-
the leaning to endarterectomy for carotid artery centage of stenosis, the classification of the
occlusive disease [9]. The North American Symp- plaque’s morphology, the localization of the carotid
tomatic Carotid Endarterectomy Trial (NASCET) artery and its bifurcation and its relationship with
[10] and the European Carotid Surgery Trial other neck anatomical structures, the possible pres-
(ECST) [11] demonstrated significative outcome ence of multiple stenotic lesions (tandem stenosis)
improving in symptomatic patients with high- both in extracranial and intracranial tracts, the pos-
grade (70–99 %) stenosis of the internal carotid sible presence of fresh intraluminal thrombus or
artery. A third large trial, the Asymptomatic plaque ulceration, the status of vertebral arteries
Carotid Atherosclerosis Study (ACAS) in 1995, and of the circle of Willis, and the presence of
showed a more modest benefit of carotid surgical cerebral ischemic lesions. These information can
treatment also in asymptomatic patients [12]. bring to modify the surgical or anesthesiologic
In 1977 Mathias reported the first percutaneous strategy or also induce to avoid surgery.
transluminal angioplasty of the internal carotid Most of these information can be acquired with
artery (Mathias 1977). In 1994 Marks reported duplex scan. According to the Society of Radiol-
the treatment of spontaneous dissection of the ogists in Ultrasound Consensus statement, Dopp-
internal carotid artery using the Palmaz stent. In ler sonography is increasingly becoming the sole
the first period of use of carotid artery stenting imaging technique used for the evaluation for
(CAS), the rate of neurologic intra- or post- carotid stenosis before surgery [14]. Ultrasound
procedural neurologic complications was very evaluation localizes the common, internal, and
high so that the first randomized trial of carotid external carotid artery and identifies the level of
artery stenting vs carotid endarterectomy was the bifurcation and the myointimal thickness. In
stopped (Naylor 1998). Subsequently, technical most of the cases, the presence of an atheroscle-
and material evolution, especially with the rotic plaque can be detected and the percentage of
11 Carotid Artery Surgery 193

Table 1 Angiographic criteria


NASCET (A–B)
—————————  100 % stenosis
A
ECST (C–B)
—————————  100 % stenosis
C

Table 2 Hemodynamic criteria


NASCET > = 70 % stenosis PSV > 220 cm/s
EDV > 80 cm s
ECST > = 70 % stenosis PSV > 190 cm/s
EDV > 65 cm s

The cutoff for the hemodynamically significative


stenosis is expressed in Table 2. There are some
differences between various authors in the defini-
tion of the cutoff threshold, ranging between
220 and 283 for NASCET criteria [14, 15],
Heijenbrok-Kal (2005), [16].
It’s evident that, in a minor number of patients,
Fig. 1 Schematic drawing that shows the measurements
for NASCET and ECST criteria the alternative use of NASCET or ECST criteria
can produce a different surgical decision. NASCET
criteria are just based on the narrowing of the
stenosis calculated. The main limit of this tech- hematic column in the carotid axis, and a stenosis
nique is represented by calcification of the arterial is detected when there is a reduction of this column
wall that hinders the ultrasound passage produc- diameter compared to the distal internal carotid
ing an “umbra” image. artery. ECST criteria instead compare the maxi-
One of the most important topics in Duplex mum narrowing with the entire internal carotid
scan examination is the calculation of the percent- diameter at the same level. Paradoxically, consid-
age of stenosis. This feature is extremely impor- ering the normal anatomical dilatation of the origin
tant because it is crucial for the surgical decision. of the internal carotid artery (carotid bulb), a plaque
However, there are various methods used for this that intersects just the dilated part of the bulb with-
calculation. The two most used methods derive out reducing the lumen with respect to the distal
from the NASCET and ECST trials (Fig. 1) and internal carotid artery determines a 0 % stenosis
are deeply different (Table 1). The NASCET and using NASCET criteria, while using ECST criteria
ECST criteria were originally established using the stenosis is detected (Fig. 2). If we consider that
contrast angiography. Afterward the same dimen- the risk of cerebral ischemia is consequent at the
sional criteria can be applied using ultrasound embolic potential of the plaque, in some cases
assessment. NASCET criteria could underestimate the real sit-
It is widely accepted that average Doppler uation. Due to the different systems of calculating,
velocity rises in direct proportion to the degree the percentage of stenosis results generally higher
of stenosis, Therefore, the internal carotid artery using ECST method.
peak systolic velocity (PSV) and the end-diastolic In the last years the role of plaque’s morphol-
velocity (EDV) can be used to identify hemody- ogy is emerging in the evaluation of embolic risk
namically significant carotid stenosis [15]. Maxi- (Fig. 3). The assessment of a stenosis is just one of
mal PSV and EDV are taken within the narrowing. the elements useful in the prediction of the clinical
194 R. Montisci and L. Saba

Fig. 2 Different criteria


can produce different
surgical decision

by a higher tendency to rupture, potentially


resulting in embolization and consequent cerebral
ischemia [19].
Therefore, characterization of the plaque mor-
phology can be important for the surgical decision,
mostly in patients with borderline level of stenosis.
Ultrasound morphologic evaluation of the
carotid plaque identifies the various plaque compo-
nents. In particular, anechogenic plaques have a
higher component of soft tissue, such as lipid or
intra-plaque hemorrhage, while echogenic plaques
are composed mainly of fibrous tissue. The Gray-
Weale’s scale modified by Geroulakos can identify
Fig. 3 Macroscopic view of the carotid specimen showing five types of carotid plaques: type 1 (anechogenic
a typical feature of vulnerable plaque: the intra-plaque with echogenic fibrous cap), type 2 (predominantly
hemorrhage (IPH)
anechogenic but with echogenic areas representing
less than 25 % of the plaque), type 3 (predominantly
outcome. Ouhlous [17] and Nandalur [18] in 2005 hyperechogenic but with anechogenic areas
pointed out the relationship between the compo- representing less than 25 % of the plaque), type
sition of the carotid plaque and the risk of cere- 4 (echogenic and homogeneous plaque), and type
brovascular ischemic events. Ohara in 2008 5 (unclassified plaques reflecting calcified plaques
demonstrated that eccentric stenosis was associ- with areas of acoustic shadowing which hide the
ated with a significant increase of ipsilateral ische- deeper part of the arterial layers).
mic events compared with concentric stenosis. Another aspect of plaque morphology is
The “vulnerable” carotid plaque is characterized represented by the analysis of the plaque surface.
11 Carotid Artery Surgery 195

Fig. 4 Longitudinal US view of an ulcerated (white


arrow) carotid artery plaque at the level of carotid
bifurcation

A smooth surface indicates a plain interface Fig. 5 MDCTA axial view of an ulceration involving the
ICA of the left side
between the plaque and the lumen. The presence
of plaque surface irregularities is related with an
increased risk of ischemic events [20, 21]. But the
most relevant surface alteration is the ulceration, characteristics. Extending the study to the
defined as an intimal defect larger than 1 mm that encephalic district, it’s possible to evaluate the
expose the necrotic core of the atheromatous plaque status of the circle of Willis. The presence of two
[22]. The presence of plaque irregularities or ulcer- or more interruption in the circle of Willis appears
ation (Fig. 4) exposes to major risk of embolization to be associated to high risk of intolerance to
due to possible platelet aggregation and thrombosis cross-clamping [24] (Fig. 6).
or embolic release of the lipidic plaque core. There- CT and MR are also useful in the evaluation of
fore, the presence of a “vulnerable plaque” should cerebral parenchyma. Sometimes also in clinically
be carefully evaluated for surgical treatment also in asymptomatic patients, these examination can
patients with noncritical stenosis. reveal areas of previous ischemic cerebral lesions.
Multi-detector computed tomography angiog- CT and MR provide the surgeon very important
raphy (MDCTA) is a reliable tool for the evalua- information about overall anatomy of the area.
tion of carotid atherosclerosis (Fig. 5). It’s Carotid endarterectomy can be quite simple or
generally used after ultrasound analysis. extremely difficult due to the wide variability of
In addition to the calculation of the stenosis anatomical disposition. In particular, a “high”
percentage, it’s useful in the detection of plaque bifurcation with a longitudinal extended vulnera-
irregularities and especially of ulceration with ble plaque in a deep internal carotid artery can
diagnostic accuracy higher than US-ECD [23]. represent a very challenging struggle. Consider-
Moreover, measuring Hounsfield unit (HU) atten- ing that carotid endarterectomy is a preventive
uation in the carotid plaque is useful to categorize intervention, often performed in asymptomatic
the plaque, also in case of calcification. patients, the availability of the maximum level
In alternative to CT, MR, with different acqui- of information is extremely useful in order to
sition techniques, is useful to obtain information minimize the risk of intraprocedural
about the percentage of stenosis and plaque complications.
196 R. Montisci and L. Saba

Fig. 6 MRA TOF (time-of-flight sequence) in the axial arrowhead) and both the PcoA (yellow open arrowheads).
and coronal views that show the circle of Willis configu- Moreover, there is no flow in the left PCA. This patient
ration (BA basilar artery, MCA middle cerebral artery, ACA showed cross-clamping intolerance during the carotid end-
anterior cerebral artery, PCA posterior cerebral artery). In arterectomy procedure requiring the shunt positioning dur-
this patient there is no A1 segment of the left ACA (yellow ing the surgery

with a vascular loop. Sometimes the superior thy-


Surgical Technique of Endarterectomy roid artery – normally the first branch of the exter-
nal carotid artery – originates directly from the
The intervention begins with an incision of common carotid artery at the bifurcation level and
10–12 cm along the anterior border of the needs to be isolated and passed with a loop. With
sternocleidomastoid muscle, centered at the level gentle traction on the external carotid artery, the
of carotid bifurcation. Cutting the platysma, it internal carotid is exposed and can be freed along
exposes the sternocleidomastoid muscle whose the adventitial plan, avoiding damage to the ner-
anterior margin is freed and retracted posteriorly. vous structures (Fig. 7) that lie near this vessel
The vascular phase begins; hence, it is useful to (in particular the hypoglossal nerve with its
magnify in order to identify the thin nerve struc- descending branch and the vagus nerve). The
tures that with great variability cross and surround internal carotid artery needs to be prepared exten-
the vascular structures and that must be mobilized sively until the end of the carotid plaque, with at
and resected. The internal jugular vein is found least 1 cm of healthy distal carotid for a safe
and the common facial vein is isolated, ligated, clamping.
and divided. At this point the common carotid In most of the cases, the end of the plaque can
artery is located by palpation. After cautious inci- be assessed both with visualization and gentle
sion of the vascular sheath, the adventitial plan is palpation. Preoperative imaging can be very help-
joined, and the common carotid artery is freed and ful on this point, in order to avoid hazardous
passed with a vascular loop. The opening of the handling of the carotid plaque. Also the internal
vascular sheath is progressively extended toward carotid artery is passed with a vascular loop. In
the bifurcation that is widely exposed; then the case of “high” bifurcation, with a plaque very
external carotid artery is isolated and encircled longitudinally extended, the exposure of the
11 Carotid Artery Surgery 197

Fig. 7 Surgical exposition


of the carotid bifurcation
during the carotid
endarterectomy procedure.
The CCA, ICA, and ECA
are visible together with the
hypoglossal nerve (white
arrowheads), the ansa
cervicalis (white open
arrow), and the transverse
cervical nerve (white
arrow)

internal carotid artery can require complex vascular flow avoiding cerebral damages. In that
maneuvers like mobilization of the hypoglossal case, after a wide arteriotomy that extended from
nerve, division of the digastric and stylohyoid the distal common carotid artery up to the end of
muscle, retraction of the parotid gland, subluxa- the plaque in the internal carotid artery, the shunt
tion of the mandibular condyle, and others skill (usually the Pruitt-Inahara shunt or the Javid
that can require a multidisciplinary approach. For- shunt) is inserted – previous rapid removing of
tunately, these cases are very rare. The exposition the clamp – before in the internal carotid artery
concluded that the patient is heparinized. and then in the common carotid artery. Both ends
At this point, the internal, external, and com- of the shunt are stabilized (the Pruitt-Inahara shunt
mon carotid arteries are sequentially clamped. inflating the balloons near the extremities, the
After cross-clamping, it’s mandatory to acquire Javid shunt with tourniquets). In most of the
information about tolerance to carotid cross- cases, the shunt positioning is followed by rapid
clamping. In fact, about 10–20 % of the patients restoring of the cerebral functions, easily recog-
don’t tolerate the interruption of the flow and nizable with clinical (local anesthesia) or instru-
show symptoms of cerebral ischemia. In the mental (general anesthesia) signs.
patient operated under local anesthesia, symptoms After cross-clamping, a longitudinal
are quickly detected: in most of the cases there is arteriotomy is performed on the posterolateral
loss of consciousness, mental confusion, and distal common carotid artery and extended toward
inability to squeeze a rattle in his or her contralat- the internal carotid artery. For direct endarterec-
eral hand. In the patient operated under general tomy the extension of the arteriotomy regards the
anesthesia, information about cross-clamping entire length of the plaque. With the use of a
intolerance is obtained with continuous dissector, the surgeon identifies the plan between
intraoperative electroencephalographic monitor- the atherosclerotic plaque and the residual arterial
ing or with somatosensory-evoked potential mon- wall. This plan can be found between the media
itoring. Information about intracranial circulation and the external elastic lamina or between the
can also be obtained both intraoperatively via intima and the internal elastic lamina. In my expe-
transcranial Doppler [25] and with near-infrared rience, the media is often involved, especially in
spectroscopy [26]. calcific plaques. In most of the cases, the plan of
The onset of intolerance of carotid cross- dissection of the plaque is easily identifiable. The
clamping requires immediate positioning of a plaque is circumferentially detached before in the
temporary shunt in order to ensure an adequate common carotid artery’s side and, then,
198 R. Montisci and L. Saba

continuing the dissection toward the external Jackson-Pratt drain is placed near the arterial
carotid artery and lastly toward the internal carotid plan and brought out with a little incision. The
artery. Distally, the plaque usually separates from sternocleidomastoid muscle is accosted to the
the normal intima but in some cases can be nec- median cervical fascia and the platysma is
essary to incise the distal intima. However, once reconstructed. The cutaneous plan is sutured
the plaque has been removed, an accurate check of with staples. Generally, the drain is removed in
the end point, i.e., the transition line between the the first post-op day.
endarterectomized area and the distal normal As alternative to direct endarterectomy, the
intima is mandatory. In fact, if the distal intima eversion endarterectomy requires the transection
is partially separated from the underlying plan, of the internal carotid artery at its origin. Then the
restoring the blood flow can totally dissect the plaque is grasped and the vessel wall is progres-
distal intima at the downstream edge, causing sively peeled back and everted. The plaque is
acute occlusion of the internal carotid artery. In completely removed and after meticulous
case of moderate instability of the distal intima, checking of the “end point,” the internal carotid
this can be stabilized with some stitches (Kunlin artery is reimplanted at the bifurcation with a
stitches), ensuring firm adhesion to the medio- circular anastomosis. This technique is used
adventitial plan. The internal surface of the inter- often in case of stenosis associated to kinking or
nal wall is then irrigated with heparinized saline coiling of the internal carotid artery: this tech-
solution, and any debris or microflap floating is nique allows a simple elimination of the redun-
removed. All these phases of the endarterectomy dancy of the vessel that is shortened before the
can be performed also when a shunt has been reimplantation.
inserted, although this result can be quite
cumbersome.
The suture begins at the internal carotid artery Carotid Body Tumors
and runs down toward the center of the
arteriotomy. A second suture begins at the com- Carotid body tumors are rare, generally benign,
mon carotid artery toward the first suture. When and occasionally malignant (5–7 %) tumors
the two sutures meet, before tying, a backflow (paraganglioma) of the carotid body, a little
from the internal carotid artery is flushed out via organ highly vascularized and placed in the saddle
arteriotomy in order to purge air bubbles and between external and internal carotid arteries. Its
microdebris from the lumen. After closure, we frequency is about 1 in 30,000 [27, 28]. The tumor
prefer to re-clamp the internal carotid artery at its presents as painless neck mass under the mandib-
origin and declamp at the same time the common ular angle. Color Doppler sonography can reveal
and the external carotid artery for about 10 s, the origin of the mass from the carotid body tumor
sending any possible microdebris toward the due to the splaying of the bifurcation and
external carotid artery’s district. Then the internal hypervascularity. MDCTA shows the relationship
carotid clamp is removed. Just when the circum- between the mass and the surrounding tissues and
ference of the internal carotid artery is small, the consents to classify the tumor according to the
arteriotomy can be closed using a patch (autoge- classical Shamblin’s classification (Fig. 8):
nous vein or polyester or polyurethane or bovine type I, tumor that grows splaying the two arteries
pericardium patch) in order to avoid a further (“lyre sign”) (Fig. 9) but without close contact
reduction of the lumen. Generally it is not with them; type II, tumor that begins to incorpo-
necessary. rate the two vessels but not completely (Fig. 10);
The regular flow can be assessed via and type III, tumor that has incorporated one or
intraoperative ultrasonography. both the vessels, sometimes together with nerve
Restoring the flow, careful hemostasis is structures.
achieved. If necessary, heparinization can be This classification is extremely important in
reversed with protamine infusion. A 4  10 the evaluation of the surgical risk. The contrast
11 Carotid Artery Surgery 199

Fig. 8 Schematic drawing ICA


of the Shamblin ECA
N,X
classification. See the other sup, laryn. n.
details in the text

N,XII
N,XII

sup.
laryngeal
nerve

N,x -

TYPE I TYPE II TYPE III

tumor presents a very rich vascularization, and


until the main peduncle is ligated, the risk of
blood loss is high. OctreoScan scintigraphy is
useful to assess the neuroendocrine character of
the mass and to localize any multicenter or meta-
static lesion. Selective angiography is possible to
treat preoperatively the mass with ultraselective
embolization of the feeding vessels.
Surgical excision is the treatment of choice.
Radiation therapy or embolization is considered
for patients that presents a high risk for surgery
both for local or general conditions [29].

Aneurysms of the Extracranial


Carotid Artery
Fig. 9 DSA view of a case with Shamblin type 1 tumor
(“lyre sign”): the tumor grows splaying the ICA and ECA
They are rare and more often of atherosclerotic
enhancement is useful in differentiation from origin than traumatic (also iatrogenic). Other very
other mass and, in the very early phase, can help rare causes are infections and fibromuscular dys-
to identify the main vascular peduncle. This infor- plasia [30, 31]. The clinical main sign for common
mation can simplify the surgical dissection. The carotid artery aneurysm is a laterocervical
200 R. Montisci and L. Saba

conquering an increasing popularity [34, 35, 36].


However, though an anticoagulation therapy,
about 6 % of thrombosis and occlusion of the
stent occur.

Conclusion

Nowadays the carotid endartectomy can be con-


sidered as an excellent modality to eliminate the
carotid artery plaque and to treat other pathologi-
cal conditions involving the carotid arteries.

References
1. DeBakey ME (1975) Successful carotid endarterec-
tomy for cerebrovascular insufficiency. JAMA
Fig. 10 CTA shows a Shamblin type 2 tumor (white 233:1083.rG
arrow); the tumor shows partial encasement of the ICA 2. Lin P, Javid H, Doyle E (1956) Partial internal carotid
and ECA (white arrowhead and white arrow, respectively) artery occlusion treated by primary resection and vein
graft. J Neurosurg 13:650
pulsatile mass, while internal carotid artery aneu- 3. Lyons C, Galbraith G (1957) Surgical treatment of
atherosclerotic occlusion of the internal carotid artery.
rysms often present in the pharynx. Nerve com- Ann Surg 146:487
pression of branches of cranial nerves can cause 4. Cooley D, Al-Naaman Y, Carton C (1956) Surgical
pain (V, VII) or paralysis (V, VI, VII, IX, X, XI, treatment of arteriosclerotic occlusion of common
XII). Thromboembolism from the aneurysm can carotid artery. J Neurosurg 13:500
5. Moore O, Karlan M, Sigler L (1969) Factors
cause transient ischemic attacks or stroke. Rup- influencing the safety of carotid ligation. Am J Surg
ture is a rare complication [32]. 118:666
In case of very high localization, Duplex scan 6. Callow A, David M (1980) Hume memorial lecture, an
can fail to identify the aneurysm. MDCTA is the overview of the stroke problem in carotid territory. Am
J Surg 140:181
gold standard, identifying the aneurysm, its exten- 7. Marrocco-Trischitta MM, Bandiera G, Camilli S,
sion and relation with the surrounding structures, Sillo F, Cirielli C, Guerrini P (2001) Remifentanil
the presence of intraluminal thrombus or dissec- conscious sedation during regional anaesthesia for
tion, and all information very useful for surgical carotid endarterectomy: rationale and safety. Eur J
Vasc Endovasc Surg 22:405
planning. 8. Marcucci G, Siani A, Accrocca F, Gabrielli R,
Whereas at the end of the twentieth century the Giordano A, Antonelli R et al (2011) Preserved con-
aneurysms involving the internal carotid artery sciousness in general anesthesia during carotid endar-
above the line between the angle of the mandible terectomy: a six-year experience. Interact Cardiovasc
Thorac Surg 13:601
and the tip of the mastoid process (Blaisdell line) 9. Robicsek F, Roush TS, Cook JW, Reames MK (2004)
were considered inaccessible [32], at present the From Hippocrates to Palmaz Schatz, the history of
development of reconstructive vascular surgery carotid surgery. Eur J Vasc Endovasc Surg 27:389
with multidisciplinary approach allows surgical 10. The North American Symptomatic Carotid Endarter-
ectomy Trial Collaborators (1991) Beneficial effect of
treatment of these lesions, also adjoining the base carotid endarterectomy in symptomatic patients with
of the skull [33]. Considering the complexity of the high grade stenosis. N Engl J Med 325:445
open surgical treatment of such cases that require a 11. European Carotid Surgery Trialists’s Collaborative
very extensive and invasive exposure of the base of Group (1998) Randomised trial of endarterectomy for
recently symptomatic carotid stenosis: final result of
the skull, with risks for nerve integrity, the the MRC European Carotid Surgery Trial (ECST).
endovascular approach with covered stenting is Lancet 351:1379
11 Carotid Artery Surgery 201

12. Executive Committee for the Asymptomatic Carotid multi-detector –row CT angiography. AJNR Am J
Atherosclerosis Study (1995) Endarterectomy for Neuroradiol 28:1061
asymptomatic carotid artery stenosis. JAMA 273:1421 24. Montisci R, Sanfilippo R, Bura R, Branca C, Piga M,
13. Brott TG, Hobson RW 2nd, Howard G, Roubin GS, Saba L (2013) Status of the circle of Willis and intol-
Clark WM, Brooks W, Mackey A, Hill MD, erance to carotid cross-clamping during carotid endar-
Leimgruber PP, Sheffet AJ, Howard VJ, Moore WS, terectomy. Eur J Vasc Endovasc Surg 45:107
Voeks JH, Hopkins LN, Cutlip DE, Cohen DJ, Popma 25. Stejskal L, Kramar F, Ostry S, Benes V, Mohapl M,
JJ, Ferguson RD, Cohen SN, Blackshear JL, Silver Limberk B (2007) Experience of 500 cases of neuro-
FL, Mohr JP, Lal BK, Meschia JF, CREST Investiga- physiological monitoring in carotid endarterectomy.
tors (2010) Stenting versus endarterectomy for treat- Acta Neurochir 149:681
ment of carotid-artery stenosis. N Engl J Med 26. Pennekamp CWA, Immink RV, den Ruijter HM,
363(1):11–23 Kappelle LJ, Bots ML, Buhre WF, Moll FL, de Borst
14. Grant EG, Benson CB, Moneta GL et al (2003) Carotid GJ (2013) Near-infrared spectroscopy to indicate selec-
artery stenosis: gray-scale and Doppler US diagnosis – tive shunt use during carotid endarterectomy. Eur J
society of radiologists in ultrasound consensus confer- Vasc Endovasc Surg 46:397
ence. Radiology 229:340 27. Sajid MS, Hamilton G, Baker DM (2007) Joint vascu-
15. Staikov IN, Nedeltchev K, Arnold M, Remonda L, lar research group. A multicenter review of carotid
Schroth G, Sturzenegger M, Herrmann C, Rivoir A, body tumour management. Eur J Vasc Endovasc Surg
Mattle HP (2002) Duplex sonographic criteria for mea- 34(2):127–30
suring carotid stenoses. J Clin Ultrasound 30:275 28. Ma D, Liu M, Yang H, Ma X, Zhang C (2010) Diag-
16. Saba L, Sanfilippo M, Montisci R, Mallarini G (2010) nosis and surgical treatment of carotid body tumor: a
Correlation between US-PSV and MDCTA in the report of 18 cases. J Cardiovasc Dis Res 1:122
quantification of carotid artery stenosis. Eur J Radiol 29. Davidovic LB, Djukic VB, Vasic DM, Sindjelic RP,
74:99 Duvnjak SN (2005) Diagnosis and treatment of carotid
17. Ouhlous M, Flach HZ, de Weert TT et al (2005) Carotid body paraganglioma: 21 years of experience at a clin-
plaque composition and cerebral infarction: MR imag- ical center of Serbia. World J Surg Oncol 3:10
ing study. AJNR Am J Neuroradiol 26:1044 30. Schechter DC (1979) Cervical carotid aneurysm. N Y
18. Nandalur KR, Baslurt E, Hagspiel K, Douglas State J Med 79:892
Philips C, Kramer CM (2005) Calcified carotid athero- 31. McCann RL (1990) Basic data related to peripheral
sclerotic plaque is associated less with ischemic symp- artery aneurysms. Ann Vasc Surg 4:411
toms than is non calcified plaque on MDTC. Am J 32. Stoney RJ, Qvarfordt PG (1987) Accessible and inac-
Roentgenol 184:295 cessible aneurysm of the extracranial carotid artery. In:
19. Saba L, Sanfilippo R, Montisci R, Atzeni M, Moore WS (ed) Surgery for cerebrovascular disease.
Ribuffo D, Mallarini G (2011) Vulnerable plaque: Churchill Livingstone, New York
detection of agreement between multidetector-row CT 33. Rosset E, Albertini JN, Magnan PE, Ede B, Thomassin
angiography and US-ECD. Eur J Radiol 77:509 JM, Branchereau A (2000) Surgical treatment of extra-
20. Kitamura A, Iso H, Imano H et al (2004) Carotid cranial internal carotid artery aneurysms. J Vasc Surg
intima-media thickness and plaque characteristics as a 31:713
risk factor for stroke in Japanese elderly men. Stroke 34. Li Z, Chang G, Yao C, Guo L, Liu Y, Wang M, Liu D,
35:2788 Wang S (2011) Endovascular stenting of extracranial
21. Prabhakaran S, Rundek T, Ramas R et al (2006) carotid artery aneurysm: a systematic review. Eur J
Carotid plaque surface irregularity predicts ischemic Vasc Endovasc Surg 42(4):419–26
stroke: the northern Manhattan study. Stroke 37:2696 35. Pickering GW, Rob CG (1954) Reconstruction of inter-
22. Sitzer M, Muller W, Siebler M et al (1995) Plaque nal carotid artery in a patient with intermittent attacks
ulceration and lumen thrombus are the main sources of hemiplegia. Lancet 264:994
of cerebral microemboli in high-grade internal carotid 36. Ohara T, Toyoda K, Otsubo R et al (2008) Eccentric
artery stenosis. Stroke 26:1231 stenosis of the carotid artery associated with ipsilateral
23. Saba L, Caddeo G, Sanfilippo R, Montisci R, Mallarini cerebrovascular events. AJNR Am J Neuroradiol
G (2007) CT and US in the study of ulcerated carotid 29:1200
plaque compared with surgical results. Advantages of
Part III
The Basics of Intracranial Arterial
Circulation
Intracranial Atherosclerosis
12
Xinyi Leng and David S. Liebeskind

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206 Atherosclerotic and arteriolosclerotic diseases
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206 of the cervicocerebral arteries are prevalent
Pathology and Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . 206 across the world, respectively, affecting arter-
Recurrent Risk in Symptomatic ICAS and Related ies from large to small and distal arterioles.
Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
Research Gaps . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207 This chapter is focused on atherosclerotic dis-
ease that affects large intracranial arteries, an
Neuroimaging Methods for Diagnosing
and Evaluating Symptomatic ICAS . . . . . . . . . . . . . . . 208
important cause of ischemic stroke and tran-
Introduction on Routine and Novel Imaging sient ischemic attack worldwide that requires
Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208 further understanding. An introduction covers
Routine Neuroimaging Methods to Evaluate the epidemiology and pathology of intracranial
ICAS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 208
Novel Neuroimaging Methods to Evaluate
atherosclerosis (ICAS), including both asymp-
ICAS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218 tomatic and symptomatic manifestations, and
the recurrent risk of symptomatic lesions. This
Treatment Strategies for Patients with ICAS . . . . 227
is followed by a detailed discussion on routine
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 229 and novel imaging methods to evaluate the
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 230 different aspects of symptomatic ICAS and
their roles in stratifying affected patients,
followed by a brief summary of recommenda-
tions and evidence for treatment of such
lesions based on current guidelines.
Imaging methods for diagnosing and eval-
uating symptomatic ICAS reviewed in this
chapter include noninvasive and invasive
methods to gauge the severity of luminal ste-
X. Leng nosis and direct and indirect methods to define
Department of Medicine and Therapeutics, The Chinese the extent and function of collateral circulation.
University of Hong Kong, Prince of Wales Hospital, We also reviewed routine and novel methods to
Hong Kong, SAR, China
reveal the status of the overall and territorial
e-mail: lengxinyi@gmail.com
tissue perfusion and to detect the salvageable
D.S. Liebeskind (*)
penumbra. Moreover, there is a discussion on
UCLA Department of Neurology, UCLA, Los Angeles,
CA, USA the emerging method of plaque imaging to
e-mail: davidliebeskind@yahoo.com delineate plaque morphology and constituents
# Springer Science+Business Media New York 2016 205
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_39
206 X. Leng and D.S. Liebeskind

and rising methods to directly and indirectly As summarized in a previous review article,
quantify the hemodynamic effects of such risk factors associated with ICAS can be catego-
lesions. For the particularly high risk of recur- rized as modifiable, nonmodifiable, and less well-
rent stroke in symptomatic ICAS, not only the documented risk factors [1]. Among all the
severity of luminal stenosis has been identified nonmodifiable risk factors, age, sex, and race are
as an independent predictor, other characteris- probably the most widely established. Based on
tics of the lesion revealed by imaging methods previous studies, there are several well-
as discussed in this chapter may also affect the documented modifiable risk factors for ICAS,
recurrent risk. Appropriate application and including smoking, hypertension, dyslipidemia,
interpretation of these imaging methods in diabetes, and metabolic syndrome, which are
cases suspected of symptomatic ICAS may among the main target risk factors to be controlled
facilitate reasonable clinical decision making in secondary prevention of stroke by current
for acute treatment and secondary prevention guidelines [2, 3]. There are also some less well-
of such lesions. documented factors that might be correlated to
ICAS, such as atherosclerosis of aortic, coronary
Keywords and carotid arteries, head and neck radiotherapy,
Intracranial atherosclerosis • Intracranial arte- and Alzheimer disease, which mandate further
rial stenosis • Symptomatic • Asymptomatic • investigation [1].
Cerebrovascular disease • Neuroimaging •
Ischemic stroke • Transient ischemic attack •
Recurrence • Risk stratification • Collateral Pathology and Pathogenesis
circulation • Perfusion imaging • Plaque imag-
ing • Fractional flow • Cerebral hemodynamics Atherosclerosis is a systemic disorder, involving
the coronary, carotid, intracranial, renal, and
peripheral arteries. Atherosclerosis progresses
Introduction with increasing age, which is a complex interaction
affected by variable risk factors, mediated by cel-
Epidemiology lular, molecular, and hemodynamic factors [4, 5].
In systemic atherosclerosis, atherogenesis of intra-
Intracranial atherosclerosis (ICAS) is of high prev- cranial arteries may be different between
alence throughout the world, particularly in Asian populations of different racial origins. Previous
populations. Symptomatic ICAS has been identi- studies carried out in Asian populations have
fied as the leading cause for ischemic stroke and found that carotid atherosclerosis might be more
transient ischemic attack (TIA) in Asian resembling to that of coronary atherosclerosis but
populations, which accounts for around 30–50 % not ICAS. The correlation between ICAS and
of ischemic stroke and around 50 % of TIA in carotid atherosclerosis in Caucasians may be stron-
Asians. ICAS is also predominant in stroke patients ger than that in Asians. Intracranial atherosclerosis
with Hispanic and African origins. Although it is probably occurs earlier than carotid atherosclerosis
relatively uncommon in Caucasians, ICAS has in Asians, while on the contrary, ICAS may be a
been found responsible for around 10 % of ische- more severe process than carotid atherosclerosis in
mic stroke and TIA in them. As for asymptomatic Caucasians [6].
ICAS, data are relatively scarce. Based on limited Pathology reveals that fatty streaks are the
data, asymptomatic ICAS affects about 7–13 % of earliest visible atherosclerotic lesions, which
community-dwelling Chinese aged over 40 years later progress to fibrous plaques and then
and those without histories of stroke or TIA, while advanced or complicated plaques [7]. Progression
it has been detected in 13 % of predominantly of atherosclerotic plaques could lead to the
Caucasian patients referred for carotid Doppler in narrowing of the vessel lumen, which may cause
a clinic-based [1]. turbulence or slow flow, in turn further promoting
12 Intracranial Atherosclerosis 207

atherosclerosis by activating platelets and other As for modifiable factors, elevated blood pressure
pathways. Complicated plaque constituents and and cholesterol levels, and the presence of metabolic
focal hemodynamic and cellular factors may also syndrome and diabetes, have been found to increase
alter the vulnerability of plaques [5]. Although the risk of stroke recurrence and other major vascu-
relevant data and studies are limited due to the lar events in patients with symptomatic ICAS [2].
difficulty in performing biopsy and autopsy stud- Furthermore, the severity of presenting stroke or
ies of intracranial arteries, investigating patholog- ischemia, number of stenotic cervicocerebral arter-
ical characteristics of ICAS lesions will facilitate ies, and number of ischemic lesions at baseline may
understanding of related mechanisms of stroke, also be associated with recurrent stroke in this
such as acute thrombosis, hypoperfusion, and patient subset.
artery-to-artery embolization, which needs to be Moreover, characteristics of the ICAS lesion
addressed in future studies [8]. as revealed by different imaging modalities and
methods could also affect subsequent risk of
stroke recurrence, which yield potential indica-
Recurrent Risk in Symptomatic ICAS tors for risk stratification of symptomatic ICAS.
and Related Risk Factors First of all, the severity of luminal stenosis or the
percent stenosis in ICAS has been established as
Among all principal causes of ischemic stroke or an independent predictor for stroke recurrence in
TIA, including intracranial and extracranial large- ischemic stroke or TIA due to ICAS, which has
artery disease, cardioembolism, and small vessel been widely used to define a “severe” or “mod-
disease, those due to ICAS are at particularly high erate” ICAS lesion from both the clinical and
risk of recurrence, despite medical and/or inter- imaging points of view [2, 14]. However, during
ventional treatment and risk factor modification the past few years, increasing evidence has
[9–11]. Until now, there have been several large emerged, challenging the role of percent luminal
clinical trials focusing on ischemic stroke or TIA stenosis in predicting the risk of stroke recur-
patients with symptomatic ICAS, such as the rence in symptomatic ICAS. For instance, factors
Warfarin-Aspirin Symptomatic Intracranial Dis- including but not limited to collateral status,
ease (WASID) trial and the Stenting and Aggres- plaque morphology and components, perfusion
sive Medical Management for Preventing status, and hemodynamic characteristics may
Recurrent Stroke in Intracranial Stenosis also affect the recurrent risk of symptomatic
(SAMMPRIS) trial that were performed predom- ICAS [15]. Considering the high risk of recur-
inantly in Caucasians and the Trial of cilOstazol in rence, appropriate evaluation and stratification of
Symptomatic intracranial arterial Stenosis symptomatic ICAS is required for accurate clin-
(TOSS) and TOSS 2 performed in Asians ical decision making. Routine and novel imaging
[9–12]. According to the results from these clini- methods for the evaluation of such lesions are
cal trials, the risk of stroke recurrence in ischemic discussed in details hereinafter.
stroke or TIA patients with a symptomatic ICAS
of 50–99 % luminal stenosis could be as high as
15–20 % at 1 year after ictus, with more than half Research Gaps
occurring in the territory of the index stenotic
artery [9, 11, 12]. As an important cause of ischemic stroke or TIA,
The recurrent stroke risk of symptomatic ICAS is with high risk of stroke recurrence worldwide,
related to several factors, including nonmodifiable symptomatic ICAS has not been adequately
and modifiable demographic and clinical factors, as investigated to date. Concerning the imaging eval-
well as imaging manifestations directly and indi- uation of symptomatic ICAS, the percentage of
rectly related to the ICAS lesion. Age has been luminal stenosis has been designated as almost the
established as a predictor for stroke recurrent only indicator for scaling the severity of such
in this patient subset in several studies [13]. lesions for years. Increasing evidence supports
208 X. Leng and D.S. Liebeskind

the potential clinical relevance of collateral circu- rising imaging methods to assess ICAS in clinical
lation, perfusion status, plaque characteristics, scenarios or the research area, is reviewed below
and other features of the lesion in stratifying this in this chapter.
patient subset and guiding clinical decisions.
However, most of these findings have not been
well established, partly due to the lack of a more Routine Neuroimaging Methods
wide-scale attention on this particular disorder, to Evaluate ICAS
the misleading focus on the severity of luminal
stenosis for diagnosis of this disorder, and proba- Currently, several neuroimaging methods, includ-
bly the heterogeneity of imaging methods to eval- ing routine and novel methods, are used in clinical
uate particular aspects of such lesions. Therefore, practice and the research area to evaluate ICAS
broad consensus is in great demand regarding a lesions. Common applications of routine imaging
practical paradigm for the assessment of selected methods in the evaluation of ICAS lesions are as
aspects of symptomatic ICAS and comprehensive follows: transcranial Doppler imaging (TCDI) for
interpretation of relevant imaging findings. Only diagnosing the severity of stenosis mainly based
in this way the ultimate values of different aspects on blood flow velocity, determining flow direc-
of such lesions in stratifying affected patients and tion, evaluating cerebral autoregulation, detecting
guiding clinical decisions could be systemically microemboli, and monitoring recanalization dur-
investigated in future studies. ing reperfusion therapy, of which the diagnostic
ability has been enhanced by the application of
transcranial color-coded duplex (TCCD) in recent
Neuroimaging Methods years [16]; different techniques of magnetic reso-
for Diagnosing and Evaluating nance angiography (MRA) for diagnosing the
Symptomatic ICAS degree of stenosis, assessing the collaterals, and
quantifying flow velocities and flow volumes
Introduction on Routine and Novel [17]; computed tomography angiography (CTA)
Imaging Methods for diagnosing the percentage of luminal stenosis,
detecting primary and secondary collaterals, and
Considering the high risk of stroke recurrence in assessing the thrombus burden [17, 18]; digital
symptomatic ICAS, accurate and timely risk strat- subtraction angiography (DSA), commonly
ification of the lesions is crucial for the sake of regarded as the gold standard for quantifying the
reducing the recurrent risk and consequent dis- luminal stenosis and the collateral status [14, 18];
ability and mortality, which requires a reasonable and CT perfusion (CTP) and dynamic susceptibil-
application of imaging methods to diagnose and ity contrast perfusion-weighted MR imaging
evaluate ICAS and comprehensive interpretation (DSC-PWI) for evaluating perfusion and collat-
of the imaging results. Currently, there are several eral status and for determining salvageable tissues
routine imaging methods to evaluate the severity for reperfusion therapies in acute cases. Each of
of luminal stenosis, cerebral blood flow, and col- the routine imaging methods has its unique advan-
lateral and perfusion status in the case of ICAS. tages and limitations, which are discussed in
There are also some novel methods to more accu- details below.
rately or less invasively delineate different aspects
of ICAS, including the above aspects that could be TCDI/TCCD
explored by routine modalities as well, and also TCDI, as a safe and noninvasive method to diag-
other applications that could not be achieved by nose ICAS, has become widely available through-
using routine methods only such as plaque imag- out the world during the past three decades
ing and selective perfusion territory visualization. [16]. Although the accuracy of TCDI to diagnose
Routine imaging methods most commonly used luminal stenosis in ICAS was reported to be infe-
in clinical examination of ICAS, and novel or rior to that of MRA or CTA, its unique advantage
12 Intracranial Atherosclerosis 209

lies in its ability to provide real-time flow infor- In recent years, TCCD (Fig. 2) has become a
mation, reveal blood flow velocities at different standard diagnostic technique to evaluate intracra-
vessel segments, evaluate cerebral autoregulation, nial arteries in ischemic stroke patients at some
detect microembolic signals (MES), and provide centers and has been increasingly used in guiding
evidence for flow direction, collateral compensa- thrombolysis of acute stroke [20]. The combina-
tion, and steal phenomenon. And by combining tion of flow and anatomic information of intracra-
with carotid duplex ultrasound, it could more nial arteries and adjacent structures enhanced the
accurately depict the cervicocerebral vasculature diagnostic accuracy of the severity of luminal
and reveal the hemodynamic impact of associated stenosis in ICAS, as compared with routine
arterial lesions [14, 16]. TCDI. Moreover, the use of contrast in TCCD
According to the Stroke Outcomes and Neuro- yields higher diagnostic value than TCDI in
imaging of Intracranial Atherosclerosis (SONIA) patients with insufficient temporal acoustic
trial, the largest prospective study so far to system- window [20].
ically examine the diagnostic accuracies of nonin- Real-time flow monitoring by TCDI or TCCD
vasive imaging methods TCDI, MRA, and CTA to yields an economic and effective method for
scale the severity of luminal stenosis in ICAS as detecting the recanalization of occluded arteries
compared with DSA, TCDI could reliably rule out in acute ischemic stroke (Fig. 3) and is possibly
50 % luminal stenosis, with negative predictive related to stroke severity and mortality [16]. In the
values (NPV) of higher than 80 % for both the past few years, TCDI and TCCD have also been
prospectively tested cut points and the cut points used to enhance the thrombolytic activity of intra-
modified to maximize the positive predictive value venous tissue plasminogen activator (tPA), which
(PPV). However, abnormal findings in intracranial was found to be efficient and safe. As compared
arteries suggested by TCDI with regard to luminal with tPA alone, tPA plus high-frequency TCDI/
stenosis (Fig. 1) may require further confirmation TCCD may correlate with higher rates of com-
by other imaging methods, since the PPV of TCDI plete recanalization, without increasing the risk of
was found to be lower than 50 % [14]. symptomatic intracranial hemorrhage [21].
Besides the most common application of TCDI TCDI, as a noninvasive and relatively inexpen-
in grading luminal stenosis of ICAS, it could also sive imaging method, is widely available through-
be used in the following manners. Quantified out the world. With its unique advantages as
vasomotor reactivity by TCDI could represent mentioned above, TCDI could be a useful tool to
the status of cerebral autoregulation, which prob- evaluate hemodynamic characteristics of symp-
ably is globally impaired in patients with symp- tomatic ICAS, and some of the characteristics
tomatic ICAS, and also a risk factor for stroke could be potential predictors for stroke severity
[16]. MES revealed by TCDI supports the theory and the risk of recurrence in patients with symp-
that artery-to-artery embolization may be a poten- tomatic ICAS. However, TCDI is a highly
tial mechanism of ICAS-related ischemic stroke, operator-dependent examination; inconsistency
and it has been found to be related to the worsen- of experience and skills of different operators
ing of neurological deficits in acute ischemic may impact on its diagnostic accuracy. Also,
stroke and also independently correlated to recur- insufficiency of temporal windows, especially in
rent events in ischemic stroke due to ICAS [16]. In elder females, leads to underestimation of dis-
addition, recent studies suggested that the steal eased intracranial arteries [16].
phenomenon termed “reversed Robin Hood syn-
drome” revealed by TCDI, referring to arterial MRA
blood flow stolen from ischemic to non-affected Several different techniques or sequences of
brain tissues, could be a possible mechanism for MRA, a noninvasive imaging modality, have
an early deterioration of acute ischemic stroke and been applied in clinical practice to evaluate the
correlated with the risk of stroke recurrence inde- cervicocerebral vasculature during the past
pendent of stroke subtypes [19]. decades [22]. Among all these techniques,
210 X. Leng and D.S. Liebeskind

a 30 30
40 40
50 50
mm

mm
60 60
70 70
80 80

100 140
80 120
60 100
cm/s

cm/s
40 80
20 60
0 40
–20 20

b 60 60

70 70

80 80
mm

mm
90 90

100 100

110 110

–160 –220
–140 –200
–120 –180
–100 –160
–80 –140

cm/s
cm/s

–60 –120
–40 –100
–20 –80
0 –60
20 –40
40 –20
60 0

c 60 60

70 70

80 80
mm
mm

90 90

100 100

110 110

–100
–80
–90
–70
–80
–60
–70
–50
cm/s

–60
–40
cm/s

–50
–30
–40
–20
–30
–10
–20
0
–10
10
0
20
10

Fig. 1 Intracranial atherosclerotic stenoses diagnosed by DSA (center), 84 % proximal BA stenosis; TCDI, delayed
transcranial Doppler imaging (TCDI) and digital subtrac- systolic upstroke in both proximal and mid-BA with MFVs
tion angiography (DSA). (a) DSA (center), 58 % stenosis of 20 and 23 cm/s, indicating a most severe or elongated
of the M1 segment of the middle cerebral artery (MCA); stenosis (Adapted from Zhao L, Barlinn K, Sharma VK,
TCDI, normal mean flow velocity (MFV) in the MCA at a Tsivgoulis G, Cava LF, Vasdekis SN, Teoh HL,
depth of 60 mm (insert on the left) and high MFV in the Triantafyllou N, Chan BP, Sharma A, Voumvourakis K,
mid-MCA at a depth of 49 mm (insert on the right), Stamboulis E, Saqqur M, Harrigan MR, Albright KC,
indicating a 50 % MCA stenosis. (SPR in the figure Alexandrov AV (2011) Velocity criteria for intracranial
indicates stenotic-to-prestenotic ratio). (b) DSA (center), stenosis revisited: an international multicenter study of
71 % mid-basilar artery (BA) stenosis; TCDI, normal MFV transcranial Doppler and digital subtraction angiography.
in the BA at a depth of 85 mm (insert on the left) and high Stroke 42: 3429–3434. doi: 10.1161/strokeaha.111.
MFV in the BA at a depth of 94 mm (insert on the right), 621235; with permission)
indicating a high-grade (70 %) mid-BA stenosis. (c)

time-of-flight MRA (TOF-MRA) is the most com- (QMRA) could also be used to reveal anatomic
monly used MRA technique in clinical investiga- severity of luminal stenosis as well as flow infor-
tion of intracranial arteries. In addition, phase- mation in the case of ICAS. Besides depicting
contrast MRA (PC-MRA), contrast-enhanced vessel structures, most of these MRA techniques
MRA (CE-MRA), and quantitative MRA bear flow information as well, which is a
12 Intracranial Atherosclerosis 211

MCA stenosis -210

-160

180

30 30

40 40
mm

50 50

60 60

70 70

80 80

Fig. 2 Appearance of a critical right middle cerebral (right). There is significantly increased mean flow velocity
artery (MCA) stem stenosis (white star) on transcranial (MFV, 220 cm/s) at the stenosis region and decreased MFV
color-coded duplex (TCCD) (upper left) and power (40 cm/s) and delayed upstroke (0.25 s) at the post-stenotic
motion-mode (PMD) transcranial Doppler imaging region (Adapted from Topcuoglu MA (2012) Transcranial
(TCDI) (lower left). Flow disturbance is easily discernible Doppler ultrasound in neurovascular diseases: diagnostic
on both TCCD and PMD. TCDI waveforms of the diseased and therapeutic aspects. J Neurochem 123 Suppl 2: 39–51.
right MCA and other cerebral arteries are displayed with a doi: 10.1111/j.1471-4159.2012.07942.x; with permission)
corresponding magnetic resonance angiography image

particular advantage of this imaging modality the signal contrast mechanism of TOF-MRA,
over others. However, in the case of symptomatic flow-related enhancement, which may lead to an
ICAS, MRA techniques may not be available in overestimation of the severity of luminal stenosis
acute settings, at least at primary or secondary in lesions with slow or minimal flow. Although
medical centers, which limit its values in acute the flow-dependent nature of TOF-MRA may
evaluation of such lesions. hinder its ability to scale the luminal stenosis in
TOF-MRA (Fig. 4) is an angiographic method the case of ICAS, it could to a certain extent reveal
without using any contrast agents, which could be the hemodynamic impact of an ICAS lesion. So
widely used in patients with contraindications to the inherent flow information yields a unique
contrast agents, such as renal failure and relevant advantage of this imaging method over some of
allergic reactions. According to the SONIA trial, the others, which is further discussed below [15].
the positive and negative predictive values for Like TOF-MRA, PC-MRA does not need con-
TOF-MRA to diagnose a 50–99 % intracranial trast agents either, though the latter technique is
stenosis were 59 % (95 % CI: 54–65 %) and not as commonly used as the former one in clin-
91 % (95 % CI: 89–93 %), with DSA being the ical practice to evaluate intracranial atheroscle-
reference standard [14]. Thus, TOF-MRA could rotic disease. By using PC-MRA, not only the
be a reliable tool to rule out an arterial stenosis in arterial structure could be revealed, flow velocities
ICAS, but positive diagnosis of luminal stenosis and cerebral blood flow could also be quantified,
by TOF-MRA needs to be confirmed by other based on phase shifts of flowing protons
imaging methods, which is similar to that of within the vessel lumen. By using TOF-MRA to
TCDI as mentioned above. This could be due to facilitate three-dimensional vessel localization,
212

Fig. 3 Description and illustration of the thrombolysis in brain ischemia (TIBI) (Adapted from Demchuk AM, Burgin WS, Christou I, Felberg RA, Barber PA, Hill
transcranial Doppler imaging (TCDI) flow grades (Grades 0–5) to classify residual MD, Alexandrov AV (2001) Thrombolysis in Brain Ischemia (TIBI) transcranial
flow through an occluded intracranial artery. Emergent TCDI TIBI classification was Doppler flow grades predict clinical severity, early recovery, and mortality in patients
found to be correlated with initial stroke severity, clinical recovery, and mortality in treated with intravenous tissue plasminogen activator. Stroke 32: 89–93. doi: 10.1161/
X. Leng and D.S. Liebeskind

stroke patients treated with intravenously administered tissue plasminogen activator 01.STR.32.1.89; with permission)
12 Intracranial Atherosclerosis 213

Fig. 4 Intracranial atherosclerotic lesions at the left middle cerebral artery (a, arrow), right middle cerebral artery (b,
arrow), and basilar artery (c, arrow), as revealed by time-of-flight magnetic resonance angiography

PC-MRA-based QMRA could significantly probably has higher diagnostic accuracy than
improve the accuracy of PC-MRA to quantify MRA to define an ICAS lesion of 50–99 % lumi-
cerebral blood flow, which is detailed below [23]. nal stenosis. However, based on results from the
In addition to the contrast agent-independent SONIA trial, the positive predictive value of CTA
MRA techniques as above, CE-MRA is also fre- to define a 50–99 % intracranial stenosis was low,
quently used in clinical practice to simulta- almost comparable to that of MRA and TCDI
neously visualize the entire supra-aortic [24]. The SONIA trial suggested that, like MRA
vasculature, including extracranial and intracra- or TCDI, CTA was also a useful tool to rule out the
nial arteries. With contrast injection, CE-MRA presence of arterial stenosis due to ICAS but not
could better delineate vessel structures as com- as much reliable in scaling the severity of luminal
pared with TOF-MRA, but the diagnostic accu- stenosis, with DSA as the reference standard,
racy of arterial stenoses by CE-MRA was lower while the results may be biased by the fact that
for intracranial arteries than that for extracranial only a small part of subjects analyzed in the
arteries. Further, CE-MRA does not bear flow SONIA trial underwent CTA examinations.
information in the evaluation of ICAS, unlike Thus, further prospective large studies should be
TOF- and PC-MRA. performed to validate the diagnostic accuracy of
CTA for detecting arterial stenosis by ICAS.
CTA Additionally, CTA could also be used to eval-
Routine single-phase CTA has been increasingly uate the thrombus burden in intracranial athero-
used in the clinical evaluation of ICAS during the sclerotic disease, for instance, the clot burden
past few years (Fig. 5). As a minimally invasive score (CBS), which has been found to be able to
imaging modality, routine CTA could yield a use- predict functional outcome, final infarct size, and
ful tool for scaling the severity of luminal stenosis the parenchymal hematoma risk [25]. Further,
in ICAS. By using intravenous contrast, CTA CTA source images (CTA-SI) are very useful in
could better depict the vessel geometry as com- the assessment of tissue status in acute ischemic
pared with MRA, but delineation of some seg- stroke. Ischemic brain tissues with low cerebral
ments of the intracranial vasculature, such as the blood flow are revealed as regions without the
petrous and cavernous portions of internal carotid enhancement on CTA-SI, which could be differ-
artery, and the distal vertebral artery, may be entiated from brain tissues with normal cerebral
affected by surrounding bones, though the associ- blood flow. By using the Alberta Stroke Program
ated artifacts could be improved by proper post- Early CT Score (ASPECTS) on CTA-SI, acute
processing techniques. ischemic stroke within the middle cerebral arterial
Regarding the diagnostic ability of CTA for territory could be more reliably identified, and the
defining the severity of luminal stenosis in final infarct volume and clinical outcomes could
ICAS, there have been no large prospective stud- be more accurately predicted, as compared with
ies. According to some small studies, CTA non-contrast CT [26].
214 X. Leng and D.S. Liebeskind

Fig. 5 Intracranial atherosclerosis on computed tomogra- parietal infarct. The CTA demonstrates a tight stenosis at
phy angiography (CTA). This patient presented with mul- the left M1 segment of the middle cerebral artery. The 3D
tiple acute ischemic lesions in the left hemisphere as shown reconstruction better delineates the finding and the vessel
in the diffusion-weighted imaging (DWI) and apparent morphology
diffusion coefficient (ADC) maps. Notice the old right
12 Intracranial Atherosclerosis 215

Fig. 6 Axial (a) and coronal


(b) maximum intensity
projections of computed
tomography angiography
demonstrating
leptomeningeal collateral
blood flow from the posterior
cerebral artery (arrows) to
distal segments of an
occluded middle cerebral
artery (From Liebeskind DS
(2003) Collateral circulation.
Stroke 34: 2279–2284.
doi: 10.1161/01.STR.
0000086465.41263.06)

Moreover, CTA could be used to evaluate pri- DSA


mary and secondary collaterals (Fig. 6). For DSA (Fig. 7), with its superb spatial resolution, is
instance, there have been several methods to eval- regarded as the gold standard to diagnose the
uate leptomeningeal collaterals by CTA-SI or severity of luminal stenosis, flow direction, and
reconstructed CTA, the interobserver agreement collaterals in ICAS. It has been used in enormous
of which methods ranged from moderate to excel- studies as the reference standard to validate the
lent [18]. Multiple studies have demonstrated the diagnostic accuracies of other imaging modalities
protective role of good collaterals as revealed by to assess the intracranial vasculature, for instance,
CTA in clinical outcomes of stroke patients in the SONIA trial [14]. It has also been used in
[26]. However, till now, there have been no com- large clinical trials to determine the severity of
monly accepted methods using CTA to evaluate luminal stenosis by ICAS, for the sake of patient
collateral circulation in the case of symptomatic selection for the investigation of certain treatment
ICAS that has been validated in large prospective strategies, for example, the WASID and the
studies. The inconsistency of these methods used SAMMPRIS trials [9, 10]. The WASID criteria
for evaluating collaterals may hinder the general- for measuring the percentage of luminal stenosis
ized use of this minimally invasive imaging in ICAS are now widely used in clinical practice
modality in multicenter studies to fully appreciate and relevant research areas. Further, although
the impact of collateral flow in the case of symp- there are still controversies in applying interven-
tomatic ICAS. tional therapy in symptomatic ICAS, when
Although CTA has multiple applications in employed, the severity of luminal stenosis defined
the evaluation of ICAS, it tends to eliminate by DSA is usually used to guide clinical decisions
any temporal flow information through the or to guide patient selection in the setting of clin-
lesions, which by itself is not a good tool for ical trials [27].
evaluating hemodynamics of ICAS lesions. But Moreover, DSA may be used to evaluate and
it may be a good source for the reconstruction of grade collateral status in symptomatic ICAS.
blood flow models of ICAS by using the compu- There have been multiple methods using DSA to
tational fluid dynamics (CFD) technique, which evaluate the collateral circulation, including col-
is discussed below [15]. Another limitation of laterals of the Circle of Willis and leptomeningeal
this imaging method is that it is contrast agent collaterals. For instance, the American Society of
dependent, which is contraindicated in patients Interventional and Therapeutic Neuroradiology/
with renal failure or allergic reactions to the Society of Interventional Radiology (ASITN/
contrast agent. SIR) Collateral Flow Grading System (Table 1)
216 X. Leng and D.S. Liebeskind

Fig. 7 Serial angiographic images from a selective left internal carotid artery injection in a patient who has middle
cerebral artery occlusion, illustrating retrograde collateral flow. (# David S. Liebeskind, MD.)

Table 1 The ASITN/SIR collateral flow grading system Collateral status graded by DSA has been found to
Grade 0 No collaterals visible to the ischemic site be able to alter the risk of stroke recurrence in
Grade 1 Slow collaterals to the periphery of the ischemic stroke or TIA due to ICAS, in the
ischemic site with persistence of some of the WASID cohort and other studies as well
defect [15]. With the collateral status identified as an
Grade 2 Rapid collaterals to the periphery of ischemic independent predictor for recurrent stroke, proba-
site with persistence of some of the defect and
to only a portion of the ischemic territory bly surpassing the role of percent stenosis, in
Grade 3 Collaterals with slow but complete patients with symptomatic ICAS [28], there is a
angiographic blood flow of the ischemic bed great need for the establishment and validation of
by the late venous phase generalizable grading methods, so that the role of
Grade 4 Complete and rapid collateral blood flow to collateral circulation could be further explored in
the vascular bed in the entire ischemic
larger, multicenter clinical studies.
territory by retrograde perfusion
DSA may differentiate antegrade and retro-
Adapted from Higashida RT, Furlan AJ, for the Technol-
ogy Assessment Committees of the American Society of grade flow by directly revealing the flow direc-
Interventional and Therapeutic Neuroradiology and the tions. However, with its invasive nature, DSA
Society of Interventional Radiology (2003) Trial design could result in periprocedural complications,
and reporting standards for intra-arterial cerebral throm- including transient or even permanent neurologi-
bolysis for acute ischemic stroke. Stroke 34: E109-E137.
doi: 10.1161/01.str.0000082721.62796.09; with permis- cal deficits in rare cases, risk of which may be
sion. ASITN/SIR indicates American Society of Interven- operator dependent. Also, the DSA procedure is
tional and Therapeutic Neuroradiology/Society of relatively more expensive than other imaging
Interventional Radiology methods. Thus, DSA could not be widely used
in clinical scenarios, especially in primary and
is one of the most commonly used grading secondary medical centers [29]. Moreover, the
methods to gauge the function of collateral circu- superb resolution of DSA to depict intracranial
lation. However, there were few studies investi- arteries may be affected by slow flow in severely
gating the interobserver reproducibility of stenosed arteries that prevent or slow the contrast
methods utilizing DSA to grade collateral circula- agents to fill in the vessel, which sometimes may
tion in the case of ICAS. According to the limited lead to the overestimation of the severity of lumi-
existing data, several of these methods were of nal stenosis or the hemodynamic impact of an
good and very good interobserver agreement [18]. ICAS lesion.
12 Intracranial Atherosclerosis 217

Fig. 8 MR perfusion/diffusion mismatch in a patient with positive mismatch with a large penumbra (Adapted from
left middle cerebral artery occlusion. Note the small area of Kloska SP, Wintermark M, Engelhorn T, Fiebach JB (2010)
infarct core (arrow) in the diffusion-weighted image (DWI; Acute stroke magnetic resonance imaging: current status and
a) in comparison with the large area of perfusion abnormal- future perspective. Neuroradiology 52: 189–201. doi:
ity (arrows) in mean transit time (MTT; b) resulting in 10.1007/s00234-009-0637-1; with permission)

Routine CT and MR Perfusion Imaging mismatch between the hypoperfused area as


In the presence of an occluded intracranial artery, revealed by CT and MR perfusion imaging and
downstream cerebral blood flow may fall below the acute infarct core serves as a reliable tool for
the threshold of 20 mL/100 g tissue/min, under the identification of the penumbra [27]. The
which the function of neurons is impaired. If the mismatch between hypoperfused area on
cerebral blood flow falls below 12 mL/100 g tis- DSC-PWI and the ischemic area on diffusion-
sue/min, irreversible damages could happen to weighted imaging (DWI) (Fig. 8), and the mean
neuronal cells [30]. Thus, hypoperfusion has transit time-cerebral blood volume mismatched by
been identified as an important mechanism for CTP (Fig. 9), has been tested in several large clin-
ischemia in patients with symptomatic ICAS, ical trials to guide selection of patients indicated for
with affected brain tissues becoming the infarct thrombolysis beyond the 3-h time window.
core or ischemic penumbra, depending on the Although definite clinical benefits of imaging-
extent of hypoperfusion in different ischemic guided thrombolysis in acute ischemic stroke
areas. The proportion of salvageable tissues, or have not yet been demonstrated in these trials,
the ischemic penumbra, and the infarct core, in the there was a trend that imaging-detected mismatch
case of acute ischemic stroke, to a great extent might be correlated to increased reperfusion
depends on the balance between the residual [27]. According to the most up-to-date guideline,
antegrade flow across the occlusive artery and CTP or DSC-PWI may be considered for the selec-
retrograde flow via collaterals. Therefore, routine tion of patients for acute reperfusion therapy
perfusion imaging methods CTP and DSC-PWI, beyond the time windows for intravenous fibrino-
respectively, based on the imaging modalities of lysis [27]. However, substantial heterogeneities in
CT and MRI, is valuable in evaluating the perfu- the acquisition and post-processing protocols for
sion status of focal and downstream brain tissues, CT and MR perfusion imaging in exiting studies
and hence reflecting the collateral status, in may limit comparisons between studies and gener-
patients with acute symptomatic ICAS [17]. alization of the study results. Thus, further studies
The potentially salvageable ischemic penum- are warranted and needed to validate the role of
bra is an ideal target for reperfusion therapies these two perfusion imaging methods in defining
which require proper patient selection, while the the penumbra for thrombolytic therapy.
218 X. Leng and D.S. Liebeskind

Fig. 9 Mean transit time (MTT, e)-cerebral blood volume (d), and MTT maps (e). doi:10.1371/journal.
(CBV, d) mismatch in a patient with acute ischemic stroke, pone.0075615.g001 (Adapted from van Seeters T, Biessels
approximately 1 h after symptom onset. The upper row and GJ, Niesten JM, van der Schaaf IC, Dankbaar JW, Horsch
lower rows, respectively, show the ischemic signs in the AD, Mali WPTM, Kappelle LJ, van der Graaf Y, Velthuis
left middle cerebral territory at the basal ganglia level and BK, on behalf of the Dust Investigators (2013) Reliability
the supraganglionic level. (a–e), respectively, represent of visual assessment of non-contrast CT, CT angiography
non-contrast CT (a), source images of CT angiography source images and CT perfusion in patients with suspected
(b), penumbra (green) and infarct (red) maps by CT perfu- ischemic stroke. PLoS ONE 8: e75615. doi: 10.1371/jour-
sion superimposed on CTA source image (c), CBV maps nal.pone.0075615. Copyright: # 2013 van Seeters et al.)

In addition to its application in detecting MR imaging (ASL-PWI) has been used to mea-
mismatch, perfusion imaging adds additional sure cerebral blood flow and reveal perfusion
information to angiographic investigations in the status of individual cerebral arteries [22]. In
evaluation of collateral circulation in symptom- some small studies, the ability of ASL-PWI has
atic ICAS, due to the reason that downstream been tested to detect the ischemic penumbra so as
perfusion status in the case of intracranial arterial to guide patient selection for intravenous throm-
occlusion could be partly determined by the col- bolytic therapy, which may be an alternative for
lateral compensating ability. Till now, there have patients with contraindications to the use of con-
been several methods using perfusion imaging trast agents. The application of ASL-PWI in
alone, or by combining with angiographic imag- ischemic stroke due to ICAS is further discussed
ing, to evaluate the extent of collateralization, below.
some of which suggested beneficial effects of
good collateral flow in acute ischemic stroke [18].
As compared with MR perfusion, CTP is more Novel Neuroimaging Methods
accessible and less time-consuming, which may to Evaluate ICAS
be more feasible to be applied in the hyperacute
phase of a symptomatic ICAS in clinical practice. Besides of the routine methods as mentioned
However, the need for contrast agents may limit above, there are several novel imaging methods
the use of routine CT and MR perfusion imaging increasingly applied in the assessment of ICAS,
in patients with contraindications and may also which may facilitate more comprehensive assess-
lead to contrast-induced complications. More ment and more efficient risk stratification of such
recently, arterial spin-labeling perfusion-weighted lesions. For instance, high-resolution magnetic
12 Intracranial Atherosclerosis 219

resonance imaging (HR-MRI) is increasingly validated with histological findings in both animal
used to evaluate constituents and morphology of models and humans, which reinforced the role of
intracranial plaques and the vessel wall, which is this technique to be the leading noninvasive
promising in detecting vulnerable plaques and in vivo imaging modality to characterize athero-
revealing mechanisms for ICAS-related stroke sclerotic plaques in these arteries [36]. Only in
[31]; QMRA to quantify cerebral blood flow, recent years has HR-MRI been applied in the
which could simultaneously visualize vascular characterization of intracranial plaques and the
anatomy and evaluate the hemodynamic impact adjacent vessel wall (Fig. 10). In recent relevant
of ICAS lesions [32]; ASL-PWI, with no need to studies, HR-MRI were usually performed under
employ any contrast agents, to delineate perfusion the field strength of 1.5 or 3.0 T, with higher field
status and detect mismatch for reperfusion thera- strength providing images of increased signal-to-
pies in acute stroke cases, to reveal perfusion noise ratio and higher quality, which could
territory and quantify cerebral blood flow of indi- improve delineation of complex intracranial
vidual intracranial arteries, and to reveal and plaques [31].
quantify collateral functions [22]; and four- In several studies using HR-MRI to depict
dimensional CTA (4D CTA) or time-resolved coronary and carotid plaques, it has been demon-
CTA or multiphase CTA to provide angiographic strated feasible and reliable to quantify the plaque
and perfusion information based on CTP without size, to characterize the vessel wall, and to define
adding additional single-phase CTA scan, which the plaque vulnerability in these arteries, while its
could improve the imaging quality as compared application in intracranial plaques has not been
with routine single-phase CTA and could reveal fully appreciated [31, 36]. Based on limited data,
flow directions and collateral status [33, 34]. intracranial arterial stenosis due to atherosclerosis
Moreover, there are another two novel methods could be differentiated from that of other causes
to evaluate the hemodynamic impact of symptom- by using HR-MRI, and intraplaque hemorrhage
atic ICAS, paralleling to the evaluation of frac- (Fig. 11), lipid core, and fibrous cap in ICAS
tional flow in coronary artery disease, respectively could also be detected. But components of intra-
based on two routine imaging methods cranial plaques as defined by HR-MRI have not
TOF-MRA and CTA [35]. The MRA-based been widely validated with histopathological find-
method is a newly developed index termed signal ings [31]. According to a small postmortem,
intensity ratio (SIR), representing the relative in vitro study, HR-MRI at a high field strength of
change of signal intensities (SI) across a symp- 7 T had substantial accuracy in detecting intracra-
tomatic ICAS lesion on TOF-MRA images; while nial plaque and its components [37].
the CTA-based method utilizes the CFD tech- In symptomatic carotid atherosclerosis,
nique to simulate blood flow, so as to depict the intraplaque hemorrhage revealed by HR-MRI
hemodynamic characteristics of symptomatic has been correlated with increased plaque vulner-
ICAS lesions [35]. All of these novel methods ability and the risk of stroke recurrence, while
are of potential values in facilitating more com- such characteristic of intracranial plaques has not
prehensive assessment and more accurate risk been well studied. According to the existing evi-
stratification of ICAS lesions, discussed in dence, intraplaque hemorrhage may be of higher
detailed below, which need further investigation prevalence in symptomatic ICAS lesions than
and validation in the near future. asymptomatic lesions, but the correlation between
intraplaque hemorrhage in symptomatic ICAS
HR-MRI and the risk of stroke recurrence has not been
HR-MRI has been used to delineate coronary and demonstrated. As for other constituents of athero-
carotid plaques in enormous studies, both athero- sclerotic plaques, the presence and extent of lipid
sclerotic and non-atherosclerotic. Morphology core and the thickness of fibrous cap have also
and components of atherosclerotic plaques in been well correlated with the stroke risk in other
these arteries revealed by HR-MRI have been arterial beds, while the prognostic values of these
220 X. Leng and D.S. Liebeskind

Fig. 10 High-resolution magnetic resonance imaging segment. The reference segment (a relatively normal seg-
(HR-MRI) images (3.0 T) of a symptomatic ICAS lesion ment) is also shown (e) (Adapted from Xu WH, Li ML,
at the left middle cerebral artery (MCA). A 74-year-old Gao S, Ni J, Zhou LX, Yao M, Peng B, Feng F, Jin ZY, Cui
male had multiple acute infarcts (arrow, a) in the distribu- LY (2010) In vivo high-resolution MR imaging of symp-
tion of stenotic left MCA (arrow, b). On consecutive tomatic and asymptomatic middle cerebral artery athero-
sagittal T2-weighted image slices, a plaque with a band sclerotic stenosis. Atherosclerosis 212: 507–511. doi:
of high signal (long arrows, c and d) is seen in the stenotic 10.1016/j.atherosclerosis.2010.06.035; with permission)

characteristics in intracranial plaques need to be QMRA


further investigated [31]. QMRA, based on PC-MRA, is a noninvasive tool
Although intracranial arteries differ with to measure blood flow in intracranial arteries,
carotid and coronary arteries in the structure of which has been used to quantify cerebral blood
vessel walls, findings of the carotid and coronary flow. By combining with TOF-MRA to facilitate
plaque and vessel wall imaging by HR-MRI shed three-dimensional vessel localization, QMRA sig-
light on corresponding studies in intracranial ath- nificantly improves the accuracy of PC-MRA to
erosclerotic plaques. Investigation of plaque mor- quantify cerebral blood flow [23]. Till now,
phology and components in ICAS by HR-MRI QMRA has been applied in the quantification of
will help reveal underlying mechanisms of related total cerebral blood flow and regional blood flow
ischemic stroke or TIA, identify lesions prone to supplied by specific intracranial arteries in relevant
rupture, and stratify patients with higher risk of studies. QMRA can quantify blood flow through
recurrence. Moreover, this technique may also be major intracranial arteries including the anterior,
useful in investigating the response of intracranial middle, and posterior cerebral arteries, and the bas-
plaques to medical therapies, such as antiplatelets ilar artery, as well as collaterals within the Circle of
and statins, as well as interventional therapies, Willis, so that it could provide information of terri-
which may guide individual treatment of patients torial blood flow and the hemodynamic impact of
with such lesions [31]. individual atherosclerotic lesions [38]. QMRA has
12 Intracranial Atherosclerosis 221

Fig. 11 In a patient with


(a) multiple acute ischemic
lesions on diffusion-
weighted images located in
the left temporal lobe, a
magnetic resonance
angiography shows (b) a
high-grade atherosclerotic
stenosis of the left middle
cerebral artery. On high-
resolution magnetic
resonance imaging (3.0 T),
an eccentric plaque is
identified on T2-weighted
imaging (arrow in c), and
T1-weighted fat-suppressed
imaging shows intraplaque
hemorrhage (arrow in d)
(Adapted from Xu W-H, Li
M-L, Gao S, Ni J, Yao M,
Zhou L-X, Peng B, Feng F,
Jin Z-Y, Cui L-Y (2012)
Middle cerebral artery
intraplaque hemorrhage:
Prevalence and Clinical
Relevance. Ann Neurol 71:
195–198. doi: 10.1002/
ana.22626; with
permission)

also been found to be able to depict arterial wave- and anterior circulations [18, 39]. Thus, QMRA is a
forms during systolic and diastolic phases of the promising tool to noninvasively and quantitatively
cardiac cycle, which provided evidence of the evaluate the collateral circulation in patients with
dampening of the arterial waveforms distal to symptomatic ICAS.
symptomatic ICAS lesions. As compared with In addition, QMRA can visualize the anatomic
TCDI examinations for arterial waveforms, and hemodynamic effects of interventional therapy
QMRA is not dependent on operators or limited of affected arteries in the case of symptomatic
by the absence of temporal window, and it could ICAS. As previously reported, QMRA could
simultaneously provide anatomic and hemody- reveal restoration of distal arterial waveforms and
namic information of intracranial arteries affected enhancement of distal cerebral blood flow in symp-
by ICAS lesions [38]. tomatic cervical arterial stenosis treated by stenting
QMRA may not only directly reveal the direc- therapy. Though there were few data concerning of
tion of blood flow via the collaterals within the such application of QMRA in symptomatic ICAS
Circle of Willis and quantify the compensating lesions, there were small studies indicating that
blood flow volume, but could also indirectly assess QMRA could help assess intracranial in-stent ste-
the presence and function of leptomeningeal col- nosis (Fig. 12). Therefore, this technique may facil-
laterals [18, 38, 39]. According to a preliminary itate noninvasive investigation and surveillance of
study, significantly increased flow rates were different responses of patients with symptomatic
observed in ipsilateral posterior cerebral artery in ICAS lesions to the stenting therapy and the related
some cases with a flow-limiting ICAS lesion in the factors, so that it may help identify those who will
middle cerebral artery or internal carotid artery, benefit from this therapy.
which indirectly reflected collateral flow via Furthermore, low flow distal to symptomatic
leptomeningeal anastomoses between the posterior ICAS lesions of the vertebrobasilar arteries as
222 X. Leng and D.S. Liebeskind

Fig. 12 Detection of an in-stent stenosis at the flow improved to 367 mL/min. (d) DSA demonstrating
supraclinoid right internal carotid artery (ICA) by quanti- in-stent restenosis with 83 % recurrent stenosis (arrow) at
tative magnetic resonance angiography (QMRA). (a) Con- 4 months of follow-up. QMRA flow decreased to
ventional digital subtraction angiography (DSA) 94 mL/min. Two weeks later, the patient had a right hemi-
demonstrating an ICAS lesion of 76 % luminal stenosis spheric stroke. (e) DSA demonstrating progressive in-stent
at the supraclinoid right ICA before treatment. (b) Three- stenosis of >90 % after the stroke, with further decrease in
dimensional surface rendering of the vasculature by the QMRA flow to 33 mL/min (Adapted from Amin-Hanjani-
Noninvasive Optimal Vessel Analysis (NOVA) system S, Alaraj A, Calderon-Arnulphi M, Aletich VA, Thulborn
(VasSol Inc., Chicago, Ill) demonstrating the scan line on KR, Charbel FT (2010) Detection of intracranial in-stent
the ICA proximal to the region of disease (arrow). QMRA restenosis using quantitative magnetic resonance angiog-
measured a flow of 139 mL/min before treatment. (c) DSA raphy. Stroke 41: 2534–2538. doi: 10.1161/
demonstrating mild residual stenosis (arrow) immediately strokeaha.110.594739; with permission)
after angioplasty and wingspan stent placement. QMRA

quantified by QMRA was found to be related to a secondary collaterals, and evaluate the structural
higher risk of recurrence, which needs to be and hemodynamic effects of stenting therapy of
further validated in prospective studies affected arteries, and it may also play a role in
[40]. Therefore, the noninvasive imaging method risk stratification of patients with symptomatic
of QMRA, by combining TOF- and PC-MRA ICAS. But these applications of QMRA in symp-
techniques, could simultaneously visualize vas- tomatic ICAS were almost all based on small
cular anatomy and measure blood flow in the studies, which need to be further verified in
presence of symptomatic ICAS lesions, assess larger studies, by validating against better-
the presence and functions of primary and established imaging modalities.
12 Intracranial Atherosclerosis 223

ASL-PWI arteries, so that it could reveal perfusion territories


As mentioned above, hypoperfusion is probably of individual cerebral arteries, commonly the left
an important mechanism in ischemic stroke due to and right internal carotid arteries and the basilar
ICAS, and the evaluation of perfusion status in artery (Fig. 13). This could facilitate more accu-
areas surrounding the infarct in acute ischemic rate identification of perfusion territories of differ-
stroke is useful in identifying salvageable brain ent cerebral arteries in patients with symptomatic
tissues for thrombolytic therapy. Unlike routine ICAS lesions, especially in those with anatomic
perfusion imaging based on CTP and DSC-PWI, variation of intracranial arteries [42]. Further, this
ASL-PWI relying on magnetic labeling of arterial technique also enables noninvasive quantification
blood does not require the use of contrast agents, of regional cerebral blood flow of individual feed-
which makes it a potential alternative for the eval- ing cerebral arteries, which leads to the identifica-
uation of perfusion status in stroke patients with tion of hemodynamically significant ICAS lesions
contraindications to contrast agents. Also, this [42]. Also, selective ASL was found to be able to
contrast agent-free imaging method could speed quantitatively assess impaired cerebrovascular
up screening and evaluation of all acute stroke reactivity at the brain-tissue level, in patients
patients indicated for perfusion status examina- with symptomatic internal carotid artery occlusive
tion, by eliminating the need for performing disease versus healthy controls [43].
renal function tests [17, 22]. Moreover, both nonselective and selective
During the past few years, ASL-PWI has ASLs could be used to evaluate the collateral
become a popular imaging method to evaluate circulation in the presence of symptomatic ICAS
the cerebral hemodynamics in ischemic cerebro- [18, 42]. Similar to DSC-PWI, nonselective ASL
vascular disease. The accuracy of ASL-PWI for could depict the collateral circulation in symptom-
evaluating cerebral perfusion status in ischemic atic ICAS by revealing the brain perfusion status,
stroke has been validated with DSC-PWI in sev- only that the latter does not require application of
eral small studies [41]. Overall, the results contrast agents. Compared with nonselective
suggested that ASL- and DSC-PWI provided ASL, selective ASL could provide more informa-
basically consistent perfusion map in acute ische- tion regarding the collateral status, including the
mic stroke, but ASL may overestimate the presence, the origin, and the function of collateral
hypoperfused volume as compared with flow. Collateral flow defined by selective ASL
DSC-PWI in some cases. As for the ability of was found to be substantially consistent with that
ASL-PWI in detecting the ischemic penumbra, confirmed by DSA in small studies [22]. More-
referring to the mismatch between hypoperfused over, selective ASL could reveal direct correla-
region on ASL and acute ischemic lesion on DWI, tions between collateral flow and the
a small retrospective study reported the moderate corresponding compensated perfusion territories,
inter-modality agreement of ASL- and DSC-PWI which could not be achieved by DSA examina-
to categorize mismatch status, which also found tion. Therefore, selective ASL is a promising tool
an overestimation of PWI lesion size by ASL in to noninvasively and quantitatively depict territo-
some cases. Therefore, as a promising alternative rial perfusion status and collateral circulation in
for contrast agent-dependent perfusion imaging, clinical scenarios of symptomatic ICAS.
practical application of ASL in assessing the
ischemic penumbra and guiding clinical decisions Four-Dimensional CTA
in acute ischemic stroke due to ICAS could prob- As mentioned above, routine CTA, or single-
ably be pursued by further refinement of the phase CTA, is used to evaluate the severity of
detecting threshold for hypoperfusion and luminal stenosis, the thrombus burden, and the
improvement of the technique [41]. collateral status in the case of symptomatic
As an advantage over routine perfusion imag- ICAS, and CTP is used to assess the perfusion
ing methods, the recently emerging selective ASL status, to detect the salvageable penumbra, and to
could encode blood flow of individual feeding indirectly reveal the collateral circulation in the
224 X. Leng and D.S. Liebeskind

Fig. 13 Illustration for selective arterial spin labeling right posterior cerebral artery (a, star). The perfusion terri-
(ASL). Transverse (a), sagittal (b), and coronal (c) maxi- tory images of the separate labeling cycles are
mum intensity projections of the circle of Willis show the superimposed and color coded according to the labeling
planning of oblique labeling planes for territorial ASL planes above (Adapted from Hartkamp NS, Petersen ET,
imaging of the right internal carotid artery (ICA) (red), De Vis JB, Bokkers RPH, Hendrikse J (2013) Mapping of
left ICA (green), and the posterior circulation (blue). Ter- cerebral perfusion territories using territorial arterial spin
ritorial ASL images (d–f) from caudal (d) to cranial (f) of labeling: techniques and clinical application. NMR
one patient with an ischemic lesion in the posterior circu- Biomed 26: 901–912. doi: 10.1002/nbm.2836; with
lation (e, star) due to a flow-limited arterial stenosis at the permission)

presence of such lesions. According to a study that contains hemodynamic information and delin-
systematically compared the diagnostic accura- eates the vascular geometry based on sequential
cies of CTP and single-phase CTA in the assess- images acquired at multiple phases [33].
ment of the site of arterial occlusion, infarct core, Four-dimensional CTA has recently been used
salvageable brain tissue, and collateral circulation in the evaluation of intracranial vasculature. The-
in patients with or without symptomatic ICAS oretically, 4D CTA images, based on perfusion
lesions, the combination of these two techniques CT images acquired over time and covering the
was the most accurate assessment method whole period of contrast inflow into the brain,
[44]. With the recent advances in CT scanning could improve the imaging quality as compared
technique, high-quality CTA data could be with routine CTA, in which depiction of intracra-
derived from whole-brain CTP source images, nial arteries could be suboptimal due to the differ-
which was referred to as 4D CTA, or time- ences in the contrast material arrival in different
resolved CTA, or multiphase CTA, as compared areas of the brain. According to available data
to the routine single-phase CTA [33, 34]. Unlike from clinical studies, 4D CTA based on CTP did
routine CTA images, which are reconstructed only have superior or at least comparable image quality
based on a short interval of the arterial phase after to detect intracranial arterial stenosis, as compared
contrast injection, 4D CTA derived from CTP with that of routine CTA, while the application of
12 Intracranial Atherosclerosis 225

Fig. 14 Antegrade flow across incomplete vessel occlu- (c) Serial right internal carotid artery injection DSA dem-
sion of the right middle cerebral artery (MCA) revealed by onstrates slow antegrade opacification distal to the occlu-
four-dimensional computed tomographic angiography sion (arrows), confirming an incomplete vessel occlusion
(CTA), as confirmed by digital subtraction angiography (Adapted from Frolich AMJ, Psychogios MN, Klotz E,
(DSA). (a) Coronal maximum intensity projection from Schramm R, Knauth M, Schramm P (2012) Antegrade
single-phase CTA in a patient with left-side hemiplegia flow across incomplete vessel occlusions can be distin-
demonstrates short-segment occlusion of the right MCA. guished from retrograde collateral flow using
(b) Early to late four-dimensional CTA (left to right) dem- 4-dimensional computed tomographic angiography. Stroke
onstrates early opacification distal to the occlusion, 43: 2974–2979. doi: 10.1161/strokeaha.112.668889; with
extending more distally in an antegrade fashion (arrows). permission)

the former technique could reduce the total radia- small study, it was suggested of high sensitivity
tion dose and the amount of contrast agents, by and specificity, as well as substantial interobserver
performing a CTP examination only without agreement, to differentiate antegrade residual flow
adding an additional single-phase CTA scan, in through incompletely occlusive vessels from ret-
the circumstances when both angiographic and rograde collateral flow in the case of symptomatic
perfusion information are required [33]. ICAS (Fig. 14), in which residual antegrade flow
4D CTA could be used to assess an important as revealed by 4D CTA was associated with an
determinator of the prognosis of stroke patients increased chance of early vessel recanalization in
with symptomatic ICAS lesions as mentioned those treated with thrombolytic therapy [34].
above, which is the collateral circulation. Though
relevant data were limited, 4D CTA could reflect A Novel Method to Gauge Hemodynamic
the extent and function of collateral flow in the Impact of ICAS Based on TOF-MRA
presence of such lesions, with better collateral As mentioned above, TOF-MRA is a noninvasive
compensation correlated with smaller acute ische- imaging method widely used in clinical practice to
mic lesions on DWI images [45]. Moreover, in a depict the intracranial vasculature. Although it is
226 X. Leng and D.S. Liebeskind

Fig. 15 The method for measurement of signal intensity the mean background signal intensity (401.1; mean of
ratio (SIR) of an intracranial atherosclerotic lesion on a 409.5 and 392.6), which is (1039.6 401.1)/
magnetic resonance angiography maximum intensity pro- (1340.3 401.1) = 0.68 (From Leng X, Wong KS,
jection. SIR of the lesion at the right middle cerebral artery Liebeskind DS (2014) Evaluating intracranial atheroscle-
is calculated as the ratio of mean signal intensities distal rosis rather than intracranial stenosis. Stroke 45: 645–651.
(1039.6) and proximal (1340.3) to the lesion, adjusted by doi: 10.1161/STROKEAHA.113.002491)

probably of relatively low positive predictive reduction or complete signal loss in situ and distal
value in quantifying the severity of luminal steno- to the stenosis. Therefore, changes of SIs across
sis of cerebral arteries [14], TOF-MRA yields a an ICAS on MIPs of TOF-MRA could to a certain
potential imaging modality to noninvasively extent represent the hemodynamic significance of
gauge the hemodynamic impact of ICAS, based the lesion. Thus, a novel term SIR as
on its unique contrast mechanism of inflow- abovementioned has been developed to quantify
enhancement effect, also known as flow-related the hemodynamic impact of symptomatic ICAS,
enhancement. TOF-MRA using gradient-echo which represented the ratio of post- and
sequences allows stationary tissues to become pre-lesional signal intensities on MRA MIP
saturated and produce minimal background sig- adjusted by the background SI (Fig. 15) [15, 35].
nals, whereas flowing blood to produce higher The existing evidence suggested that the index
SIs. The degree of enhancement of flowing of SIR was feasible and easy-to-perform and had
blood, referred to as SI on TOF-MRA images, substantial intra- and interobserver reproducibility
nonlinearly increases with increasing absolute as well [15, 46]. Moreover, there were also studies
flow velocity, with other parameters being con- suggesting the clinical relevance of this index to
stant. Laminar flow in normal and non-tortuous define the hemodynamic impact of symptomatic
arteries produces relatively uniform signal inten- ICAS lesions. For instance, significant linear cor-
sities along the longitudinal axis of a specific relation was identified between the SIR values of
vessel segment and appears bright on maximum symptomatic ICAS lesions and the infarct vol-
intensity projections (MIP) of TOF-MRA. While umes on DWI in the acute phase of ICAS-related
in the presence of flow-limited ICAS lesions, the ischemic stroke, which indicated that SIR of an
turbulence or slow flow might cause signal ICAS as evaluated on TOF-MRA might reflect its
12 Intracranial Atherosclerosis 227

functional and hemodynamic significance flow simulation could eliminate the invasive pro-
[46]. Further, dichotomized SIR values by the cedure of DSA and the consequent low but unde-
median have been found to be independently cor- niable risk of periprocedural complications.
related to recurrent ischemic stroke in the WASID Further, since the simulation of blood flow by
cohort [15]. Therefore, SIR of symptomatic ICAS the CFD technique is based on the reconstructed
could be a potential indicator to predict the recur- vessel geometry and that CTA could better depict
rent risk of affected patients, which may yield a the anatomy of intracranial arteries as compared
novel noninvasive and easy-to-perform method to with MRA, CTA may also be superior to MRA as
stratify symptomatic ICAS lesions. However, all the source for CFD modeling of ICAS. CFD
of the existing studies were retrospective, and modeling of symptomatic ICAS lesions based on
some were of small sample sizes. Also, till now, routinely obtained CTA source images has been
its ability to gauge the hemodynamic impact of demonstrated feasible, according to the limited
ICAS has not been validated. Therefore, future number of relevant studies [15]. According to
prospective studies are needed to validate this the limited data, CFD models could directly reveal
index against other imaging modalities capable the hemodynamic characteristics of such lesions,
of evaluating the hemodynamic impact of ICAS such as pressure gradient across the lesion, and
lesions, as well as to investigate its ultimate value turbulent flow and increased shear stress at the site
in risk stratification of affected patients. of arterial stenosis and downstream (Fig. 16).
CFD models could also directly depict the hemo-
CFD Modeling of ICAS to Reveal dynamic impact of interventional therapy of the
the Hemodynamic Characteristics ICAS lesions [48, 49]. However, till now, the few
In the past decade, the CFD technique has been studies utilizing the CFD technique to investigate
used in coronary and carotid artery diseases to intracranial atherosclerotic disease were all of
reveal hemodynamic characteristics of atheroscle- extremely small sample sizes, and there is still a
rotic stenoses [35]. As for coronary artery disease, long way to go before an extensive application of
the CFD technique has been applied in the simu- this technique in clinical evaluation of ICAS. But
lation of blood flow through the lesion based on encouraging findings regarding this technique in
coronary CTA images. The noninvasive fractional the field of cardiovascular diseases imply poten-
flow reserve derived from CTA (FFRCT) assessed tial values of its use in cerebrovascular diseases.
by using this technique has been found of sub- Future studies exploring the clinical relevance and
stantial diagnostic accuracy of the hemodynamic possible prognostic values of hemodynamic char-
significance of coronary artery disease, with inva- acteristics of symptomatic ICAS lesions as delin-
sive fractional flow reserve (FFR) obtained during eated by CFD models could shed new light on the
the performance of percutaneous coronary angi- assessment and diagnosis of such lesions.
ography as the reference standard [47]. Although
cerebral and coronary arteries differ in anatomic,
physiological, and other aspects, advanced use of Treatment Strategies for Patients
the CFD modeling technique in the cardiology with ICAS
field could inspirit similar application of this tech-
nique in ICAS, to facilitate direct evaluation of the Primary prevention of intracranial atherosclerotic
hemodynamic characteristics of such lesions. disease lies in the control of associated modifiable
CFD modeling of ICAS lesions could be risk factors, such as cigarette smoking, hyperten-
achieved based on different imaging modalities sion, dyslipidemia, diabetes, and metabolic syn-
providing angiographic information, such as drome, while the application of aspirin for
DSA, CTA, and MRA, while CTA may be a better primary prevention of ICAS in general
source for this purpose. Although it has been populations has not been well established
demonstrated feasible to construct CFD models [50]. Regarding the acute treatment and secondary
based on biplane DSA images, CTA-based blood prevention of patients with symptomatic ICAS, so
228 X. Leng and D.S. Liebeskind

Fig. 16 Computational a
fluid dynamics model Pressure
reconstructed based on
1.196e+002
computed tomography
angiography source images
showing the hemodynamic 9.660e+001
characteristics of a
symptomatic ICAS lesion at
the left middle cerebral 7.363e+001
artery: decreased pressure
(a, arrows), and increased
flow velocity (b, arrows) 5.066e+001
and wall shear stress (c,
arrows) at the site of the 2.769e+001
lesion and downstream [mm Hg]

b
Velocity
4.268e+000

3.202e+000

2.135e+000

1.069e+000

2.004e-003
[m s^-1]

c
Wall Shear
1.396e+002

1.047e+002

6.978e+001

3.489e+001

0.000e+000
[Pa]

far there have been no specific guidelines, which therapy, if within the 3-h time window and no
were usually incorporated in guidelines on ische- contraindications exist. For those who can be
mic stroke or TIA due to all causes [2, 3, 27]. treated within 3–4.5 h after ictus, intravenous
According to the current guidelines in the fibrinolytic therapy is probably reasonable for
United States and Europe, patients with acute more selective patients, with additional exclusion
ischemic symptoms due to ICAS identified by criteria. Multimodal imaging investigations pro-
imaging examinations that have not resolved are viding information regarding the infarct core and
recommended to receive intravenous fibrinolytic the penumbra may be considered to select eligible
12 Intracranial Atherosclerosis 229

patients for this therapy who are beyond the time identified as of higher risk than those with a
windows. For patients with acute symptomatic symptomatic ICAS of 50–69 % luminal stenosis
ICAS who are ineligible for or irresponsive to [10, 11]. As compared with medical treatment,
intravenous thrombolysis, other treatment strate- extracranial-intracranial bypass surgery has been
gies may be considered for carefully selected indi- demonstrated to be associated with worse out-
viduals, such as intra-arterial thrombolysis and comes for patients with symptomatic ICAS
mechanical thrombectomy, which procedures are lesions based on existing evidence, which is not
not routinely applied in clinical practice and need recommended for such patients [2].
to be further validated in randomized clinical tri- Despite of all these treatment strategies, the
als, while clinical application of emergent intra- risk of stroke recurrence remains particularly
cranial angioplasty and/or stenting for the high in patients with symptomatic ICAS, which
responsible artery has not been well established is up to 15–20 % at 1 year after ictus in lesions
for acute symptomatic ICAS [3, 27]. resulting in 50–99 % luminal stenosis
For the secondary prevention of patients with [9–11]. Therefore, accurate and timely risk strati-
ICAS-related ischemic stroke or TIA, the two fication of these patients based on their demo-
large, prospective, randomized clinical trials, graphic and clinical features, and especially
WASID and SAMMPRIS, provided the evidence findings from appropriate imaging investigations
for clinical decision making concerning for different aspects of ICAS lesions as discussed
antiplatelet, anticoagulant, or interventional ther- above, could help identify patients truly at high
apies for affected patients [2, 3]. According to the risk and facilitate clinical decision making.
WASID trial, aspirin was superior to warfarin in
secondary prevention of ischemic stroke or TIA
due to an ICAS of 50–99 % luminal stenosis, in Summary
the way that aspirin was as effective as warfarin in
reducing the risks of stroke recurrence and other Intracranial atherosclerosis is a disease of consid-
vascular events while at the mean time safer than erable prevalence throughout the world, espe-
warfarin by significantly decreasing the risks of cially in Asia. Among the ischemic stroke of
adverse events including death and major hemor- different causes, those due to ICAS are at partic-
rhage [9]. However, although several large ran- ularly high risk of recurrence. Currently, the eval-
domized clinical trials have been investigating the uation of symptomatic ICAS is usually based on
efficacy and safety of dual versus mono- the severity of luminal stenosis, which has been
antiplatelet therapies in the secondary prevention found to be an independent predictor for the recur-
of ischemic stroke and TIA patients, few data rent risk. However, with the emerging evidence on
existed specifically on that of ICAS-related the role of collateral circulation in risk stratifica-
stroke. Till now, there has been no agreement on tion of symptomatic ICAS, increasing attention
the application of dual antiplatelet therapy in the has been attracted to imaging methods capable
secondary prevention of symptomatic ICAS. of revealing the collateral status. In addition,
Besides the antiplatelet therapy, a risk factor con- plaque imaging and novel perfusion and angio-
trol should also be included in the medical treat- graphic imaging methods have been rising in the
ment of ICAS-related ischemic stroke or TIA, field of ICAS during the past few years to delin-
such as management of hypertension, diabetes, eate different features of ICAS lesions. Moreover,
and dyslipidemia [2, 3]. inspired by the advances in studies of the hemo-
Concerning the intracranial angioplasty and dynamic impact of coronary artery disease, there
stenting therapy, the lately released final results have been preliminary studies to investigate that
of the SAMMPRIS trial indicated early and last- of symptomatic ICAS respectively based on
ing benefits of aggressive medical treatment over TOF-MRA and CTA. In the near future, ICAS
stenting for those with a symptomatic ICAS of should be paid more attention in clinical practice
70–99 % luminal stenosis, who were traditionally and relevant studies. By the reasonable
230 X. Leng and D.S. Liebeskind

application of imaging methods and comprehen- Kasner SE, Benesch CG, Sila CA, Jovin TG, Romano
sive interpretation of the imaging results, affected JG (2005) Comparison of warfarin and aspirin for
symptomatic intracranial arterial stenosis. N Engl J
patients could be timely and accurately stratified Med 352:1305–1316. doi:10.1056/NEJMoa043033
by the risk of recurrence, and treatment strategies 10. Chimowitz MI, Lynn MJ, Derdeyn CP, Turan TN,
could be properly determined. This in the long run Fiorella D, Lane BF, Janis LS, Lutsep HL, Barnwell
may reduce the recurrent risk of such lesions. SL, Waters MF, Hoh BL, Hourihane JM, Levy EI,
Alexandrov AV, Harrigan MR, Chiu D, Klucznik RP,
Clark JM, McDougall CG, Johnson MD, Pride GL Jr,
Torbey MT, Zaidat OO, Rumboldt Z, Cloft HJ (2011)
References Stenting versus aggressive medical therapy for intra-
cranial arterial stenosis. N Engl J Med 365:993–1003.
1. Qureshi AI, Feldmann E, Gomez CR, Johnston SC, doi:10.1056/NEJMoa1105335
Kasner SE, Quick DC, Rasmussen PA, Suri MF, Taylor 11. Derdeyn CP, Chimowitz MI, Lynn MJ, Fiorella D,
RA, Zaidat OO (2009) Intracranial atherosclerotic dis- Turan TN, Janis LS, Montgomery J, Nizam A, Lane
ease: an update. Ann Neurol 66:730–738. doi:10.1002/ BF, Lutsep HL, Barnwell SL, Waters MF, Hoh BL,
ana.21768 Hourihane JM, Levy EI, Alexandrov AV, Harrigan
2. Furie KL, Kasner SE, Adams RJ, Albers GW, Bush MR, Chiu D, Klucznik RP, Clark JM, McDougall
RL, Fagan SC, Halperin JL, Johnston SC, Katzan I, CG, Johnson MD, Pride GL Jr, Lynch JR, Zaidat OO,
Kernan WN, Mitchell PH, Ovbiagele B, Palesch YY, Rumboldt Z, Cloft HJ (2014) Aggressive medical treat-
Sacco RL, Schwamm LH, Wassertheil-Smoller S, ment with or without stenting in high-risk patients with
Turan TN, Wentworth D, American Heart Association intracranial artery stenosis (SAMMPRIS): the final
Stroke Council, Council on Cardiovascular Nursing, results of a randomised trial. Lancet 383:333–341.
Council on Clinical Cardiology, Interdisciplinary doi:10.1016/s0140-6736(13)62038-3
Council on Quality of Care and Outcomes Research 12. Kwon SU, Hong KS, Kang DW, Park JM, Lee JH, Cho
(2011) Guidelines for the prevention of stroke in YJ, Yu KH, Koo JS, Wong KSL, Lee SH, Lee KB, Kim
patients with stroke or transient ischemic attack: a DE, Jeong SW, Bae HJ, Lee BC, Han MK, Rha JH, Kim
guideline for healthcare professionals from the Amer- HY, Mok VC, Lee YS, Kim GM, Suwanwela NC, Yun
ican Heart Association/American Stroke Association. SC, Nah HW, Kim JS (2011) Efficacy and safety of
Stroke 42:227–276. doi:10.1161/STR.0b013e3 combination antiplatelet therapies in patients with symp-
181f7d043 tomatic intracranial atherosclerotic stenosis. Stroke
3. The European Stroke Organisation Executive Commit- 42:2883–2890. doi:10.1161/strokeaha.110.609370
tee and the ESO Writing Committee (2008) Guidelines 13. Jung JM, Kang DW, Yu KH, Koo JS, Lee JH, Park JM,
for management of ischaemic stroke and transient Hong KS, Cho YJ, Kim JS, Kwon SU, Toss-2 Investiga-
ischaemic attack 2008. Cerebrovasc Dis 25:457–507. tors (2012) Predictors of recurrent stroke in patients with
doi:10.1159/000131083 symptomatic intracranial arterial stenosis. Stroke
4. Moore KJ, Tabas I (2011) Macrophages in the patho- 43:2785–2787. doi:10.1161/STROKEAHA.112.659185
genesis of atherosclerosis. Cell 145:341–355. 14. Feldmann E, Wilterdink JL, Kosinski A, Lynn M,
doi:10.1016/j.cell.2011.04.005 Chimowitz MI, Sarafin J, Smith HH, Nichols F,
5. Wentzel JJ, Chatzizisis YS, Gijsen FJH, Giannoglou Rogg J, Cloft HJ, Wechsler L, Saver J, Levine SR,
GD, Feldman CL, Stone PH (2012) Endothelial shear Tegeler C, Adams R, Sloan M, The SONIATrial Inves-
stress in the evolution of coronary atherosclerotic tigators (2007) The Stroke Outcomes and Neuroimag-
plaque and vascular remodelling: current understand- ing of Intracranial Atherosclerosis (SONIA) trial.
ing and remaining questions. Cardiovasc Res Neurology 68:2099–2106. doi:10.1212/01.
96:234–243. doi:10.1093/cvr/cvs217 wnl.0000261488.05906.c1
6. Leng XY, Chen XY, Chook P, Xiong L, Lin WH, Liu 15. Leng X, Wong KS, Liebeskind DS (2014) Evaluating
JY, Tomlinson B, Thomas GN, Lam TH, Lam KSL, intracranial atherosclerosis rather than intracranial
Cheung BMY, Wong KS (2013) Correlation of large stenosis. Stroke 45:645–651. doi:10.1161/
artery intracranial occlusive disease with carotid STROKEAHA. 113.002491
intima-media thickness and presence of carotid plaque. 16. Alexandrov AV, Sloan MA, Tegeler CH, Newell DN,
Stroke 44:68–72. doi:10.1161/strokeaha.112.675652 Lumsden A, Garami Z, Levy CR, Wong LK,
7. Consigny PM (1995) Pathogenesis of atherosclerosis. Douville C, Kaps M, Tsivgoulis G (2012) Practice
Am J Roentgenol 164:553–558. doi:10.2214/ standards for transcranial Doppler (TCD) ultrasound.
ajr.164.3.7863871 Part II. Clinical indications and expected outcomes. J
8. Gao S, Wang YJ, Xu AD, Li YS, Wang DZ (2011) Neuroimaging 22:215–224. doi:10.1111/j.1552-
Chinese ischemic stroke subclassification. Front 6569.2010.00523.x
Neurol 2:6. doi:10.3389/fneur.2011.00006 17. Khan R, Nael K, Erly W (2013) Acute stroke imaging:
9. Chimowitz MI, Lynn MJ, Howlett-Smith H, Stern BJ, what clinicians need to know. Am J Med 126:379–386.
Hertzberg VS, Frankel MR, Levine SR, Chaturvedi S, doi:10.1016/j.amjmed.2012.11.014
12 Intracranial Atherosclerosis 231

18. McVerry F, Liebeskind DS, Muir KW (2012) System- 28. Liebeskind DS, Cotsonis GA, Saver JL, Lynn MJ,
atic review of methods for assessing leptomeningeal Turan TN, Cloft HJ, Chimowitz MI (2011) Collaterals
collateral flow. AJNR Am J Neuroradiol 33:576–582. dramatically alter stroke risk in intracranial atheroscle-
doi:10.3174/ajnr.A2794 rosis. Ann Neurol 69:963–974. doi:10.1002/ana.22354
19. Palazzo P, Balucani C, Barlinn K, Tsivgoulis G, 29. Willinsky RA, Taylor SM, terBrugge K, Farb RI,
Zhang Y, Zhao L, Dewolfe J, Toaldo B, Tomlinson G, Montanera W (2003) Neurologic com-
Stamboulis E, Vernieri F, Rossini PM, Alexandrov plications of cerebral angiography: prospective analy-
AV (2010) Association of reversed Robin Hood syn- sis of 2,899 procedures and review of the literature.
drome with risk of stroke recurrence. Neurology Radiology 227:522–528. doi:10.1148/radiol.
75:2003–2008. doi:10.1212/WNL.0b013e3181ffe4e4 2272012071
20. Nedelmann M, Stolz E, Gerriets T, Baumgartner RW, 30. Kloska SP, Wintermark M, Engelhorn T, Fiebach JB
Malferrari G, Seidel G, Kaps M (2009) Consensus (2010) Acute stroke magnetic resonance imaging: cur-
recommendations for transcranial color-coded duplex rent status and future perspective. Neuroradiology
sonography for the assessment of intracranial arteries 52:189–201. doi:10.1007/s00234-009-0637-1
in clinical trials on acute stroke. Stroke 40:3238–3244. 31. Bodle JD, Feldmann E, Swartz RH, Rumboldt Z,
doi:10.1161/strokeaha.109.555169 Brown T, Turan TN (2013) High-resolution magnetic
21. Tsivgoulis G, Eggers J, Ribo M, Perren F, Saqqur M, resonance imaging: an emerging tool for evaluating
Rubiera M, Sergentanis TN, Vadikolias K, Larrue V, intracranial arterial disease. Stroke 44:287–292.
Molina CA, Alexandrov AV (2010) Safety and efficacy doi:10.1161/STROKEAHA.112.664680
of ultrasound-enhanced thrombolysis: a comprehen- 32. Prabhakaran S, Warrior L, Wells KR, Jhaveri MD,
sive review and meta-analysis of randomized and Chen M, Lopes DK (2009) The utility of quantitative
nonrandomized studies. Stroke 41:280–287. magnetic resonance angiography in the assessment of
doi:10.1161/strokeaha. 109.563304 intracranial in-stent stenosis. Stroke 40:991–993.
22. Donahue MJ, Strother MK, Hendrikse J (2012) Novel doi:10.1161/strokeaha.108. 522391
MRI approaches for assessing cerebral hemodynamics 33. Smit EJ, Vonken EJ, van der Schaaf IC, Mendrik AM,
in ischemic cerebrovascular disease. Stroke Dankbaar JW, Horsch AD, van Seeters T, van
43:903–915. doi:10.1161/STROKEAHA.111.635995 Ginneken B, Prokop M (2012) Timing-invariant recon-
23. Zhao M, Charbel FT, Alperin N, Loth F, Clark ME (2000) struction for deriving high-quality CT angiographic
Improved phase-contrast flow quantification by three- data from cerebral CT perfusion data. Radiology
dimensional vessel localization. Magn Reson Imaging 263:216–225. doi:10.1148/radiol.11111068
18:697–706. doi:10.1016/S0730-725X(00)00157-0 34. Frolich AMJ, Psychogios MN, Klotz E, Schramm R,
24. Liebeskind DS, Kosinski AS, Saver JL, Feldmann E, Knauth M, Schramm P (2012) Antegrade flow across
SONIA Investigators (2007) CT angiography in the incomplete vessel occlusions can be distinguished
Stroke Outcomes and Neuroimaging of Intracranial from retrograde collateral flow using 4-dimensional
Atherosclerosis (SONIA) study. Stroke 32:477 computed tomographic angiography. Stroke
25. Puetz V, Dzialowski I, Hill MD, Subramaniam S, 43:2974–2979. doi:10.1161/strokeaha.112.668889
Sylaja PN, Krol A, O’Reilly C, Hudon ME, Hu WY, 35. Liebeskind DS, Feldmann E (2013) Fractional flow in
Coutts SB, Barber PA, Watson T, Roy J, Demchuk AM, cerebrovascular disorders. Interv Neurol 1:87–99.
Calgary CTASG (2008) Intracranial thrombus extent doi:10.1159/000346803
predicts clinical outcome, final infarct size and hemor- 36. Corti R, Fuster V (2011) Imaging of atherosclerosis:
rhagic transformation in ischemic stroke: the clot bur- magnetic resonance imaging. Eur Heart J
den score. Int J Stroke 3:230–236. doi:10.1111/j.1747- 32:1709–1719b. doi:10.1093/eurheartj/ehr068
4949.2008.00221.x 37. Majidi S, Sein J, Watanabe M, Hassan AE, Van de
26. Mortimer AM, Simpson E, Bradley MD, Renowden Moortele PF, Suri MF, Clark HB, Qureshi AI (2013)
SA (2013) Computed tomography angiography in Intracranial-derived atherosclerosis assessment: an
hyperacute ischemic stroke: prognostic implications in vitro comparison between virtual histology by intra-
and role in decision-making. Stroke 44:1480–1488. vascular ultrasonography, 7 T MRI, and histopatho-
doi:10.1161/strokeaha.111.679522 logic findings. AJNR Am J Neuroradiol
27. Jauch EC, Saver JL, Adams HP, Bruno A, Connors JJ, 34:2259–2264. doi:10.3174/ajnr.A3631
Demaerschalk BM, Khatri P, McMullan PW, Qureshi 38. Brisman JL, Pile-Spellman J, Konstas AA (2012) Clin-
AI, Rosenfield K, Scott PA, Summers DR, Wang DZ, ical utility of quantitative magnetic resonance angiog-
Wintermark M, Yonas H, American Heart Association raphy in the assessment of the underlying
Stroke, Council CoCN, Council on Peripheral Vascular pathophysiology in a variety of cerebrovascular disor-
Disease, Council on Clinical Cardiology (2013) Guide- ders. Eur J Radiol 81:298–302. doi:10.1016/j.
lines for the early management of patients with acute ejrad.2010.12.079
ischemic stroke: a guideline for healthcare profes- 39. Ruland S, Ahmed A, Thomas K, Zhao MD, Amin-
sionals from the American Heart Association/Ameri- Hanjani S, Du XJ, Charbel FT (2009) Leptomeningeal
can Stroke Association. Stroke 44:870–947. collateral volume flow assessed by quantitative mag-
doi:10.1161/STR. 0b013e318284056a netic resonance angiography in large-vessel
232 X. Leng and D.S. Liebeskind

cerebrovascular disease. J Neuroimaging 19:27–30. weighted MRI in MCA occlusion. J Neuroimaging


doi:10.1111/j.1552-6569.2008.00249.x 21:225–228. doi:10.1111/j.1552-6569.2010.00496.x
40. Amin-Hanjani S, Du XJ, Zhao MD, Walsh K, Malisch 46. Leng X, Wong KS, Soo Y, Leung T, Zou X, Wang Y,
TW, Charbel FT (2005) Use of quantitative magnetic Feldmann E, Liu L, Liebeskind DS (2013) Magnetic
resonance angiography to stratify stroke risk in symp- resonance angiography signal intensity as a marker of
tomatic vertebrobasilar disease. Stroke 36:1140–1145. hemodynamic impairment in intracranial arterial stenosis.
doi:10.1161/01.STR.0000166195. 63276.7c PLoS One 8:e80124. doi:10.1371/journal.pone.0080124
41. Nael K, Meshksar A, Liebeskind DS, Coull BM, 47. Taylor CA, Fonte TA, Min JK (2013) Computational
Krupinski EA, Villablanca JP (2013) Quantitative anal- fluid dynamics applied to cardiac computed tomogra-
ysis of hypoperfusion in acute stroke: arterial spin phy for noninvasive quantification of fractional flow
labeling versus dynamic susceptibility contrast. Stroke reserve: scientific basis. J Am Coll Cardiol
44:3090–3096. doi:10.1161/strokeaha. 113.002377 61:2233–2241. doi:10.1016/j.jacc. 2012.11.083
42. van Laar PJ, van der Grond J, Hendrikse J (2008) Brain 48. Schirmer CM, Malek AM (2007) Prediction of complex
perfusion territory imaging: methods and clinical appli- flow patterns in intracranial atherosclerotic disease using
cations of selective arterial spin-labeling MR imaging. computational fluid dynamics. Neurosurgery
Radiology 246:354–364. doi:10.1148/radio1. 61:842–852. doi:10.1227/01.neu.0000298914.32248.dc
2462061775 49. Suh DC, Ko YB, Park ST, Yoon K, Lim OK, Oh JS,
43. Bokkers RP, van Osch MJ, Klijn CJ, Kappelle LJ, Jeong YG, Kim JS (2011) Computational flow dynam-
Hendrikse J (2011) Cerebrovascular reactivity within ics of the severe M1 stenosis before and after stenting.
perfusion territories in patients with an internal carotid Neurointervention 6:13–16. doi:10.5469/neuroint.
artery occlusion. J Neurol Neurosurg Psychiatry 2011.6.1.13
82:1011–1016. doi:10.1136/jnnp.2010.233338 50. Goldstein LB, Bushnell CD, Adams RJ, Appel LJ,
44. Tan JC, Dillon WP, Liu SL, Adler F, Smith WS, Braun LT, Chaturvedi S, Creager MA, Culebras A,
Wintermark M (2007) Systematic comparison of Eckel RH, Hart RG, Hinchey JA, Howard VJ, Jauch
perfusion-CT and CT-angiography in acute stroke EC, Levine SR, Meschia JF, Moore WS, Nixon JV,
patients. Ann Neurol 61:533–543. doi:10.1002/ Pearson TA (2011) Guidelines for the primary preven-
ana.21130 tion of stroke: a guideline for healthcare professionals
45. Jung SL, Lee YJ, Ahn KJ, Kim YI, Lee KS, Shin YS, from the American Heart Association/American Stroke
Lee KS, Kim BS (2011) Assessment of collateral flow Association. Stroke 42:517–584. doi:10.1161/STR.
with multi-phasic CT: correlation with diffusion 0b013e3181fcb238
Imaging of Spontaneous
Nontraumatic Intracerebral 13
Hemorrhage

Stephen Quinet and Patrick Turski

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234 The focus of this chapter is to provide an over-
view of the most commonly encountered etiol-
Hypertensive Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . 234
ogies of spontaneous intracerebral hemorrhage
Evolution of Hematoma on CT and MRI . . . . . . . . . 239 (SICH). Specifically, the goal is to review the
Coagulopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 241 differential diagnosis of nontraumatic hemor-
rhage based on clinical presentation, imaging
Cerebral Amyloid Angiopathy . . . . . . . . . . . . . . . . . . . . 244
characteristics, and location of hemorrhage. In
T2* Gradient Echo Imaging . . . . . . . . . . . . . . . . . . . . . . . 248 addition to reviewing the various potential
Susceptibility-Weighted Imaging (SWI) . . . . . . . . . . 248 causes of nontraumatic intracranial hemor-
Nontraumatic Subarachnoid Hemorrhage . . . . . . . 251 rhage, this chapter will provide a review of
the variable appearance of hemorrhage on CT
Vascular Malformations . . . . . . . . . . . . . . . . . . . . . . . . . . . 262
and MR. An emphasis will be placed on iden-
Hemorrhagic Transformation of Infarct . . . . . . . . . 270 tification and anticipation of the potential com-
Venous Thrombosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 274 plications of acute intracranial hemorrhage.
Hemorrhagic Neoplasm . . . . . . . . . . . . . . . . . . . . . . . . . . . . 276
Keywords
Radiation-Induced Vascular Disease Spontaneous intracranial hemorrhage •
and Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 283
Nontraumatic intracranial hemorrhage •
Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285 Hypertensive hemorrhage • Coagulopathy •
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290 Cerebral amyloid angiopathy • Cerebral
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290 microbleeds • Cerebral microhemorrhages •
Nontraumatic subarachnoid hemorrhage •
Intracranial aneurysm • Arteriovenous malfor-
mation • Dural arteriovenous fistula • Cavern-
ous malformation • Hemorrhagic
transformation of infarct • Venous thrombosis
• Hemorrhagic neoplasm • Radiation
vasculopathy • Radiation microhemorrhage •
S. Quinet (*) • P. Turski Radiation microangiopathy • Central nervous
Department of Radiology, School of Medicine and Public
system vasculitis
Health, Clinical Science Center, University of Wisconsin,
Madison, WI, USA
e-mail: squine@uw.edu; pturski@uwhealth.org

# Springer Science+Business Media New York 2016 233


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_47
234 S. Quinet and P. Turski

Introduction with perfusion, is often warranted if the etiology


is not immediately obvious. The choice of modal-
This chapter provides a review of nine etiologies ity will depend on patient issues (e.g., rapid clin-
of nontraumatic intracranial hemorrhage. Three of ical deterioration, MR-incompatible medical
these entities comprise the majority of devices, uncooperative patient) and the most
nontraumatic intracranial hemorrhage: hyperten- highly suspected underlying pathology (e.g., vas-
sion (36 %), aneurysm (36 %), and arteriovenous cular lesion versus a hemorrhagic mass). In some
malformation (11 %). Other etiologies account for cases, the patient will be best assessed and poten-
17 % of nontraumatic hemorrhage, the more com- tially treated by proceeding directly to conven-
mon of which are discussed in detail later in the tional catheter-directed angiography. Given the
chapter (Table 1). appearance and distribution of the hemorrhage
Hemorrhagic strokes represent 10–15 % of all and the clinical presentation, a reasonable short
strokes and are characterized by the sudden onset differential diagnosis can be developed which will
of a focal neurologic deficit due to intracranial guide selection of the appropriate follow-up imag-
hemorrhage, which can be intraparenchymal ing modality.
(IPH), subarachnoid (SAH), intraventricular
(IVH), or a combination of locations. IPH is
more likely to mimic acute ischemic stroke, Hypertensive Hemorrhage
while subarachnoid hemorrhage classically pre-
sents with “thunder clap” headache. IPH will Arteriopathy related to long-standing hyperten-
more commonly manifest as seizures and nau- sion is the most common cause of spontaneous
sea/vomiting. However, there is considerable nontraumatic intracranial hemorrhage in adults
overlap in the presenting symptoms for both IPH [3]. Chronic systemic hypertension produces
and SAH. degeneration of the deep penetrating arteriolar
Non-contrast-enhanced head CT is often the branches of the major intracranial arteries on the
imaging modality selected for the initial evalua- basis of intimal hyperplasia and medial degenera-
tion of suspected intracranial pathology. The wide tion. These affected arterioles are the very small
availability in various clinical settings, the ability vessels at termination points of the perforators
to scan without antecedent safety screenings (as in arising as end artery branches from the anterior
MR), and extremely rapid image acquisition and and posterior circulation. Over time, sustained
reconstruction have made CT the imaging modal- hypertension leads to deposition of hyaline, fibrin,
ity generally used in the setting of a neurologic and collagen in the arteriolar walls, leading to
emergency. If there is high clinical suspicion for degeneration. Arteriolar sclerosis leads to
vascular pathology, starting with advanced imag- narrowing of the vessel lumen and ischemia,
ing techniques, most often CTA or MRI/MRA can with the ultimate end result being lacunar infarcts.
be performed [1, 2]. Once intracranial hemorrhage Additionally, as arteriolar walls degenerate, they
is detected, follow-up imaging with contrast- become weakened and begin to dilate partially on
enhanced CT and/or MR imaging, with dedicated the basis of lipid and collagen replacing arteriolar
angiographic images (CTA/MRA) without or smooth muscle cells, leading to formation of
so-called Charcot–Bouchard microaneurysms
(Fig. 1). These are not true aneurysms but rather
Table 1 Etiologies of spontaneous intracerebral hemor-
rhage (SICH) are more accurately classified as degenerative
fusiform dilations of chronically diseased vascu-
Location Frequency (%)
lature. These weakened, dilated walls are thought
Hypertension 36
Aneurysm 36
to be more prone to rupture.
Arteriovenous malformation (AVM) 11
Hypertensive hemorrhages occur in predict-
Others 17 able locations, a characteristic that is useful in
identifying chronic hypertension as the most
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 235

Fig. 1 Acute hypertensive hemorrhage. (a) Axial CTA microaneurysm (arrow). (b) Axial source images from
maximum intensity projection image demonstrates the CTA again show this enhancing punctate structure
intraparenchymal hemorrhage centered within the right (arrow). The coronal MIP reformat CTA image (c) shows
caudate nucleus, with extension into the ventricular sys- that the microaneurysm is at the termination of a
tem. A punctate enhancing structure in the right basal lenticulostriate artery
ganglia represents an unruptured Charcot–Bouchard

likely etiology for intracranial hemorrhage. About Table 2 Location of hypertensive hemorrhage
66 % occur in basal ganglia, particularly in the Location Frequency (%)
putamen and external capsule, 20 % within the Putamen/external capsule 60
thalamus, 10 % within the pons, 5 % within the Thalamus 20
cerebellum, and 5 % within the subcortical white Pons 10
matter (Table 2). It is important to note that while Cerebellum 5
the basal ganglia, thalamus, and brainstem are the Subcortical white matter 5
most common locations for hypertensive hemor-
rhage, this etiology must be considered in the
differential diagnosis of spontaneous lobar hem- locations, as detailed above. However, given the
orrhage, especially in the appropriate clinical set- volume of brain MR imaging done for myriad
ting (Fig. 2). potentially unrelated indications, sequelae of
In the acutely symptomatic patient, acute hem- chronic hypertensive arteriopathy are often inci-
orrhage will be identified in characteristic dentally detected. Specifically, T2* GRE and SWI
236 S. Quinet and P. Turski

Fig. 2 CT images in four patients demonstrate the classic Note that the cerebellar hemorrhage has extended in to the
locations of acute hypertensive hemorrhages, including the fourth ventricle. Dilation of the temporal horns of the
(a) putamen, (b) thalamus, (c) pons, and (d) cerebellum. lateral ventricles indicates obstructive hydrocephalus

sequences will reveal loss of signal and “bloom- include loss of brain volume, a background of
ing,” often multifocal, in the distributions of the chronic ischemic microvascular disease in the
deep perforators. Multiple hypertensive white matter, and old small lacunar infarcts. It is
microhemorrhages (Fig. 3) may mimic the presen- important to consider the possibility of illicit drug
tation of cerebral amyloid angiopathy (CAA). This use, especially cocaine and other sympathomimetic
topic is discussed in detail later in the chapter. drugs, as potential etiologies of acute intracranial
Some other potential associated findings seen in hemorrhage in typical locations of hypertensive
the setting of acute hypertensive hemorrhage hemorrhage in otherwise healthy young patients.
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 237

Fig. 3 Series of unenhanced head CT images obtained follow-up approximately 3 weeks after presentation. (d)
from a 30-year-old male presenting with right-sided weak- Multiple microhemorrhages are identified within the bilat-
ness. Multiple hypodense foci were identified in the deep eral basal ganglia and thalami. There has been evolution of
gray matter and white matter, including within the white the dominant left putaminal hematoma. (b, c) Additional
matter of the right frontal lobe (a) and right caudate (b, c). microhemorrhages are identified in the central (e) pons and
A large acute hemorrhage was identified centered within (f) cerebellum
the left putamen. Series of T2* MR images obtained in

The size of a hematoma on initial imaging and presentation of acute hypertensive hemorrhage,
its subsequent expansion portend a worse progno- dynamic combined CT/CTA imaging can be help-
sis in this patient population. Expansion of hema- ful in assessing the severity of the hemorrhage and
toma is identified in approximately one-third of the relative urgency of surgical decompression
cases of acute hypertensive hemorrhage and is versus close clinical and imaging surveillance.
most likely to occur within several hours of symp- Specifically, delayed post-contrast CT imaging
tom onset. In the setting of active extravasation, performed shortly after the CTA acquisition is
the “swirl sign” may be seen on unenhanced head useful to assess for active blood–brain barrier
CT, characterized by hypodense acute liquid breakdown. Contrast pooling within the hema-
blood products surrounding a hyperdense orga- toma seen on the delayed post-contrast imaging
nizing clot. Although not routinely performed in is indicative of active arterial extravasation. Addi-
all institutions if there is a classic imaging tionally, the so-called spot sign, a focus or foci of
238 S. Quinet and P. Turski

Fig. 4 (a) Unenhanced CT shows large acute hemorrhage subsequently obtained CTA demonstrate the “spot sign”
centered in the left basal ganglia with associated rightward (arrow) – a focal blush of contrast extravasation centered
mass effect, subfalcine herniation, and intraventricular within the basal ganglia hemorrhage
extension of hemorrhage. (b) MIP images from

arterial phase enhancement within the center of the including acute hydrocephalus, herniation syn-
hematoma, is visualized in approximately dromes, and infarction. Many supratentorial
one-quarter of cases (Fig. 4) [4]. It has been spec- hematomas can be managed medically; however,
ulated that this “spot sign” may actually represent hematomas in the posterior fossa require close
visualization of the culprit microaneursym within monitoring, often necessitating timely neurosur-
the area of hemorrhage. The spot sign is present in gical decompression, given the potential for rapid
20–30 % of acute IPH patients presenting within expansion of the hematoma and surrounding
3–10 h of symptom onset. The spot sign provides edema within the compact confines of the poste-
prognostic information, as up to 75 % of these rior fossa. There is limited ability of the posterior
patients will go on to have extension of their hema- fossa structures to enlarge without clinically sig-
tomas. Additionally, the presence of active contrast nificant, often catastrophic, complications.
extravasation on CTA (Fig. 5) is a predictor of poor Supratentorial herniation, cerebellar tonsillar her-
clinical outcome and increased in hospital mortal- niation, and obliteration of the fourth ventricle
ity (OR >10). It should be noted that the sensitivity with resultant obstructive hydrocephalus can
and specificity of the spot sign appear to be depen- develop in an impressively rapid fashion.
dent upon the timing of CTA and whether delayed Hypertensive hemorrhages tend to dissect
post-contrast imaging is performed. along the white matter tracts, so that the damage
Factors associated with poor prognosis in to the brain tissue and associated neurologic dam-
patients with acute intracranial hemorrhage age incurred by hypertensive hemorrhages are
include age greater than 80 years, posterior fossa often much less than the injury related to trau-
location of hemorrhage, volume of hemorrhage matic hemorrhagic contusions. These patients
exceeding 30 mL, and intraventricular extension will have a relatively good prognosis if they
of hemorrhage [5]. It is imperative to be aware of, have not experienced any of the above-described
and actively search for, the potential secondary severe secondary effects associated with the
effects of acute hypertensive hemorrhage hematoma.
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 239

Fig. 5 Series of images obtained from a 41-year-old susceptibility in the right pons and left caudate and puta-
woman presenting with acute hypertensive hemorrhage men consistent with additional microhemorrhages that
(SBP >220 mmHg). (a) Unenhanced CT shows large were occult on initial unenhanced CT. Blood products
acute hemorrhage centered in the right basal ganglia with line the site of the evacuated hematoma, in the region of
associated mass effect, including rightward midline shift the external capsule and putamen. (f) Unenhanced head CT
and subfalcine herniation (Source images from CTA) (b), performed 12 months after craniectomy shows expected
and delayed post-contrast CT (c) demonstrate contrast evolution of the surgical bed, with volume loss and
pooling within the area of hemorrhage (arrows) consistent hypodensity in the right frontal lobe, subinsular region, as
with active extravasation. (d) and (e): T2* GRE images well as sites of initial hemorrhage in the putamen and
status post-craniectomy demonstrate punctate foci of external capsule

platelets) [6]. The attenuation of the hematoma


Evolution of Hematoma on CT and MRI may increase in the first few days after presenta-
tion (up to 80–100 HU) depending on the degree
On unenhanced CT, the increased attenuation on of organization of the clot. After the first few days,
CT (HU 50–60) within an acute hematoma is not an acute hemorrhage evolves in a relatively pre-
due to iron content, but rather it is due to clot dictable fashion on unenhanced CT related to its
retraction and the presence of dense protein con- hydration. As the hematoma evolves, it begins to
tent within the hematoma (e.g., fibrin, cells, draw in water, resulting in the margins of the
240 S. Quinet and P. Turski

Fig. 6 Series of unenhanced head CT examinations dem- attenuation within the hematoma on sequential images
onstrating evolution of an intraparenchymal hematoma. (a) related to increased water content. Pre- and post-contrast
Initial head CT demonstrates large acute hematoma cen- head CT (e) and (f) performed 84 days after the initial
tered in the right temporoparietal region. Subsequent head hemorrhage demonstrate a small amount of residual
CT examinations performed (b) 11 days, (c) 23 days, and enhancement at the margins of the hematoma
(d) 29 days later demonstrate expected reduction in

hematoma beginning to decrease in attenuation. element of inflammation is present at the margins


As water is further drawn into the hematoma, of the hematoma as it begins to resorb, leading to
there is continued expected reduced attenuation breakdown of the blood–brain barrier. As such, a
within the hematoma itself on the basis of peripheral rim of contrast enhancement can be
increased water content. On average, one can seen starting approximately 1 week after the initial
expect the attenuation within the hematoma to hemorrhage, which can persist for up to 8–12
decrease by approximately 1.5 HU per day. An weeks (Fig. 6).
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 241

Table 3 Evolution of intracerebral hematoma on MR


Predominant blood Mass
Stage product Time course T1 signal T2 signal effect
Hyperacute Oxyhemoglobin <6 h Isointense Mildly +++
hyperintense
Acute Deoxyhemoglobin 6–72 h Iso- to Hypointense +++
hypointense
Early Intracellular <3 days to 1 week Hyperintense Hypointense +++/++
subacute methemoglobin
Late Extracellular 1–2 weeks to Hyperintense Hyperintense +/
subacute methemoglobin months
Chronic Hemosiderin 2 weeks to years Hypointense Hypointense

MR imaging is even better suited to demon- of evolution of blood products detailed above is
strate the evolution of hematoma based on the most reliable for intraparenchymal hemorrhage.
signal characteristics of various stages of break- Extra-axial hemorrhage tends to progress in a
down blood products and the anticipated time similar order, but in a more delayed fashion, likely
points at which they appear and transform on the basis of lower oxygen tension. Addition-
(Table 3, Fig. 7) [7, 8]. Hyperacute hematoma ally, subarachnoid and intraventricular hemor-
(<6 h) is predominantly composed of oxyhemo- rhage signal will be affected by mixing with
globin, demonstrating T1 isointense and T2 CSF. Finally, it is possible that on occasion an
mildly hyperintense signal. Within an acute hema- acute hematoma may be confused with infarction.
toma in slightly later evolution (7 h–3 days), the The core of an acute hematoma may be
blood products become deoxygenated and there is hyperintense on DWI (representing “T2 shine
conversion of oxyhemoglobin to through”) with decreased ADC values
deoxyhemoglobin, resulting in hematoma demon- (representing “T2 dark through”), mimicking
strating T1 isointense-to-hypointense signal and arterial stroke [9]. However, correlation with
T2 markedly hypointense signal. In the early sub- other sequences and/or CT should be helpful to
acute stage, as the blood products continue to avoid this potential pitfall.
degrade from deoxyhemoglobin to methemoglo-
bin and the red blood cell remains intact, the
hematoma predominantly demonstrates character- Coagulopathy
istic signal of intracellular methemoglobin, with
T1 marked hypointensity and T2 marked Spontaneous intracranial hemorrhage related to
hyperintensity. In the late subacute stage, after coagulopathy, whether on the basis of hemato-
the red blood cells have lysed, characteristic sig- logic disease, metabolic disease, or therapeutic
nal of extracellular methemoglobin is seen, with anticoagulation, is an entity that must be
T1 and T2 marked hyperintensity. In the chronic suspected in the appropriate clinical context and
state, there is further degradation of blood prod- is one that may demonstrate potential clues to its
ucts and subsequent uptake by macrophages diagnosis on imaging. Similar to patients with
resulting in deposition of hemosiderin and ferritin low hematocrit, blood products within a hema-
within the hematoma cavity, with marked T1 and toma in the setting of coagulopathy may be
T2 hypointensity. isodense to brain parenchyma on CT imaging.
Hematomas often contain blood products at Identification of a fluid–fluid level within the
different stages of evolution, resulting in a mix- hematoma on CT is a characteristic finding of
ture of these imaging features, complicating intracranial hematomas in this patient population
image interpretation, and sometimes establishing (Figs. 8 and 9). These fluid–fluid levels can per-
chronicity of hemorrhage is difficult. The timing sist for several hours after the hemorrhage.
242 S. Quinet and P. Turski

Fig. 7 (continued)
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 243

Fig. 7 Evolution of hematoma on MRI. Multiplanar, expected edema surrounding the hematoma in the region of
multisequence MR imaging demonstrates a typical left the left basal ganglia. Bottom row: Demonstration of late
basal ganglia hypertensive hemorrhage. Top row: Demon- subacute stage hematoma (2–4 weeks after hemorrhage).
stration of late acute stage hematoma (1–2 days after hem- (f) Sagittal T1, (g) axial T2, (h) coronal T2 FLAIR, (i) axial
orrhage). (a) Sagittal T1, (b) axial T2, (c) coronal T2 T2* GRE, and (j) axial DWI sequences. As the hematoma
FLAIR, (d) axial T2* GRE, (e) and axial DWI sequences. continues to evolve over the course of several weeks, the
Several days out, expected signal changes associated with contents of the hematoma are mostly extracellular methe-
deoxyhemoglobin are seen, with mildly T1 hypointense moglobin, demonstrating hyperintense signal on both T1-
and marked T2 hypointense signal; the T2-WI also shows and T2-WI images

Similarly, on MR imaging, heterogeneous signal nonspecific finding, but could identify potential
within the hematoma cavity with identifiable brain tissue that is at risk due to compression,
fluid–fluid levels can suggest underlying increased permeability, and inflammation related
coagulopathy. Peri-hematoma edema is a to the hematoma (Fig. 10).
244 S. Quinet and P. Turski

Fig. 8 Coagulopathic hemorrhage. Series of unenhanced fluid–fluid levels (arrows), characteristic of intracranial
head CT images from inferior to superior (a–c) demon- hemorrhage in the setting of coagulopathy
strate a large right basal ganglia hemorrhage. Note

Fig. 9 Coagulopathic hemorrhage. Two unenhanced head heterogeneous attenuation and fluid levels (arrows)
CT images demonstrate a large amount of intraventricular involving the right lateral, third, and fourth ventricles
hemorrhage and resulting obstructive hydrocephalus, with

Cerebral Amyloid Angiopathy cerebral amyloid angiopathy (CAA) [10]. Beta


amyloid deposition at these sites increases with
Beta amyloid deposition within the small- and age, accounting for the estimated 20 % of sponta-
small-to-medium-sized arteries of the cortex and neous intracranial hemorrhage in the elderly. This
leptomeninges is the pathologic hallmark of entity is rare before age 60, but is a common cause
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 245

Fig. 10 Coagulopathic hemorrhage. (a) Unenhanced of hemorrhage. Sagittal T1 (f) and axial T2 (g) images
head CT demonstrates large right basal ganglia hemor- demonstrate heterogeneous hyperintense signal within the
rhage with fluid–fluid levels, characteristic of hemorrhage hemorrhage. (h) T2 FLAIR imaging best demonstrates
in the setting of coagulopathy. (b) Axial maximum inten- degree of vasogenic edema surrounding the hematoma.
sity projection image from the concurrently performed DWI (i) and ADC (j) demonstrate a large component of
CTA demonstrates displaced MCA branches. (c–e) Cere- “T2 shine-through” in the core of the hematoma, but also
bral blood flow maps performed part of the CTA exam true diffusion restriction at the anterior and medial portion
demonstrate reduced CBF within and surrounding the area of the hematoma
246 S. Quinet and P. Turski

Fig. 11 Lobar hemorrhage and microhemorrhages in subcortical left temporal lobe, left parahippocampal
CAA. (a) Sagittal T1 image demonstrates right temporal region, and left occipital lobe. (d) Axial T2* image at a
lobar hemorrhage with peripheral hyperintensity related to more superior location demonstrates additional
methemoglobin formation. (b) Coronal T2 FLAIR image microbleeds in the left temporal and occipital cortices and
demonstrates peripheral hyperintense rim of the lobar hem- subcortical white matter. (e) Axial T2 image demonstrates
orrhage and minimal surrounding edema. Periventricular peripheral hyperintense rim of the lobar hemorrhage. (f)
and subcortical white matter signal abnormalities are also Axial DWI demonstrates hyperintense signal throughout
seen. (c) Axial T2* image demonstrates hypointense signal the hematoma. (g) ADC map demonstrates hypointense
in the lobar hemorrhage but also reveals punctate signal throughout the hematoma on the basis of “T2 dark
hypointense foci representing microbleeds in the through,” mimicking an acute infarct

of spontaneous intracranial hemorrhage in the microhemorrhages are most conspicuous as foci


elderly population, second only to hypertension. of low signal on T2* GRE and SWI sequences
CAA can present as lobar macrohemorrhage or as [12]. Leptomeningeal amyloid angiopathy can
multiple microhemorrhages, often recurrent initially present as acute blood layering within
(Fig. 11). The hemorrhage associated with CAA the subarachnoid space. Superficial siderosis,
is often lobar or cortical or combined cortical/ characterized by diffuse low signal and blooming
subcortical in location [11]. Pathologically, beta on susceptibility-weighted images along the pial
amyloid deposition is identified within the brain, surfaces of the brain and within the basilar cis-
perivascular spaces, and outer walls of arterioles terns, can be seen on follow-up imaging, thought
and leptomeningeal vessels leading to fibrinoid to be related to recurrent subarachnoid hemor-
degeneration and necrosis leading to vascular fra- rhage (Fig. 12) [13].
gility, accounting for the association with hemor- The Boston criteria were established in 1990 in
rhages. Microaneurysms have also been described order to standardize the diagnosis of CAA, using
as resulting from this degenerative process, likely clinical, imaging, and/or pathologic parameters
also contributing to hemorrhage. Finally, vessel (Table 4). Definite cerebral amyloid angiopathy
lumen obliteration may result, leading to micro- can be diagnosed only postmortem, while proba-
vascular ischemia. Characteristically, these ble CAA is defined in a patient older than
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 247

Fig. 12 Superficial siderosis due to recurrent subarach- of the left frontal and right parietal lobes (arrows)
noid hemorrhage in a patient with leptomeningeal amyloid representing additional subarachnoid hemorrhage. (d)
angiopathy (top row, imaging at presentation; bottom row, Axial T2* GRE image obtained 7 months later demon-
follow-up imaging 7 years later). (a) Unenhanced head CT strates diffuse hemosiderin staining of the pial surface of
at presentation demonstrates subarachnoid hemorrhage the brain bilaterally, representing superficial siderosis. (e)
layering within the sulci of the left frontal lobe. (b) Axial Axial SWI image obtained at the same time of
T2* GRE image at the level of the midbrain demonstrates d demonstrates the increased sensitivity of this sequence
punctate foci of susceptibility along the pial surface of the for hemorrhage. (f) Axial SWI image more cephalad than
right temporal and occipital lobes (arrows) representing e again shows superficial siderosis to better advantage
subarachnoid hemorrhage. (c) Axial T2* GRE image compared to routine T2* GRE imaging
shows punctate foci of susceptibility along the pial surface

55 years, in the setting of appropriate clinical hypertensive arteriopathy can similarly present as
history, with MRI findings of multiple multiple small areas of hemorrhage, so there can be
microhemorrhages and lobar hemorrhage or con- substantial overlap between the imaging appear-
vexity subarachnoid hemorrhage without any ance of CAA and advanced hypertensive changes
other explanation. [14]. These two entities account for the majority of
CAA coexists with Alzheimer dementia and cerebral microhemorrhages detected at imaging.
other dementias in 20–30 % of patients (Fig. 13). However, the differential diagnosis of multiple
Symptoms of dementia may be due to leukoaraiosis microhemorrhages includes hemorrhagic metastatic
resulting from progressive microvascular ischemia. disease, vasculitis, diffuse axonal injury (DAI),
Unfortunately, as discussed above, advanced cerebral embolism (including fat emboli), multiple
248 S. Quinet and P. Turski

Table 4 Boston criteria for diagnosis of cerebral amyloid spin echo T1- and T2-WI sequences.
angiopathy (CAA) Susceptibility-weighted T2* images can be of
Diagnostic category Criteria benefit in further characterizing intracranial hem-
Definite CAA Postmortem examination: orrhage/hematoma. Gradient echo T2* acquisi-
Lobar, cortical, or cortical/ tions do not have a refocusing radiofrequency
subcortical hemorrhage
Evidence of severe CAA pulse and are thus very susceptible to static vari-
vasculopathy ations in the magnetic field, such as that produced
Probable CAA with Supportive clinical history by the paramagnetic properties of hemosiderin
pathologic evidence Pathologic specimen: lobar, [15]. This local field inhomogeneity presents as
cortical, or cortical/ loss of signal on T2* images. While T2* images
subcortical hemorrhage
do not have a high contrast resolution and image
Probable CAA Supportive clinical history
Age 55 clarity of other pulse sequences, they are highly
MRI evidence of multiple sensitive to blood products (Fig. 17).
lobar, cortical, or cortical/ One pitfall in using MR to evaluate hematomas
subcortical hemorrhages
is related to the signal intensity seen on diffusion-
without other explanation
Possible CAA Supportive clinical history
weighted images. The core of an acute hematoma
Age 55 can be hyperintense on DWI with decreased ADC
MRI evidence of a single values, simulating an area of infarction.
lobar, cortical, or cortical/
subcortical hemorrhage
without other explanation
Susceptibility-Weighted Imaging (SWI)

A more advanced imaging method, which enables


cavernomas, radiation-induced vasculopathy, increased sensitivity to the detection of
cerebral autosomal dominant arteriopathy with microhemorrhages superior to even that of T2*
subcortical infarcts and leukoencephalopathy GRE (Fig. 18), is susceptibility-weighted
(CADASIL), and Parry–Romberg syndrome imaging (SWI) and susceptibility-weighted angi-
(Figs. 14 and 15). ography (SWAN) [16]. The basis of SWI is a
Inflammatory CAA is a condition which is high-resolution, flow-compensated, three-
thought to represent an autoimmune response to dimensional gradient echo sequence
the deposited beta amyloid. The imaging manifes- [17–19]. SWI combines magnitude images and
tation of inflammatory CAA is distinct from those phase shift information related to T2* dephasing
of multiple microhemorrhages classically associ- to exploit field inhomogeneity to increase the
ated with CAA. Inflammatory CAA may manifest ability to detect foci of susceptibility (e.g., cortical
as extensive white matter edema, perivascular veins, calcification, hemosiderin). Variations in
inflammation, and vasculitis (Fig. 16). The imag- the local magnetic field result in dephasing of
ing appearance of inflammatory CAA may very signal and reduction of T2*. Nonheme-related
closely mimic acute hypertensive encephalopa- iron can be detected and may be useful for the
thy/posterior reversible encephalopathy syn- detection and surveillance of diseases that result in
drome (PRES). iron deposition in the brain, such as Parkinson,
Huntington, and Alzheimer dementias. SWI per-
formance and sensitivity are positively related to
T2* Gradient Echo Imaging field strength.
Using postprocessing techniques, following
GRE T2* images are significantly more sensitive application of a high-pass filter to reduce aliasing,
to both hyperacute hemorrhage and a phase mask is applied to the magnitude images
microhemorrhages compared to conventional to increase the conspicuity of foci of
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 249

Fig. 13 A 77-year-old male with rapidly progressive dementia. Multiple axial T2* GRE images (e–g) demon-
dementia, diagnosed with Alzheimer dementia 2 years strate innumerable foci of signal loss consistent with
prior to this imaging. Unenhanced head CT (a) and T1 microhemorrhages, most pronounced in the left parietal
(b), T2 (c), and T2 FLAIR (d) MR images demonstrate the lobe, in a cortical–subcortical distribution typical of
classic appearance of dilated temporal horns of the lateral CAA. Hippocampal and temporal lobe volume loss, seen
ventricles due to hippocampal and medial temporal lobe to better advantage on the previous figure, again is com-
atrophy characteristically seen in Alzheimer-type patible with Alzheimer dementia
250 S. Quinet and P. Turski

Fig. 14 Differential diagnosis of multiple bilateral radiation-induced telangiectasias and a cavernoma, respec-
microhemorrhages. (a) Axial T2* GRE image demon- tively. (c) Axial T2* GRE image demonstrates
strates punctate microhemorrhages in bilateral basal microhemorrhages in the subcortical white matter of the
ganglia in a patient with hypertensive arteriopathy. (b) right frontal and temporal lobes as well as the left thalamus
Axial T2* GRE image demonstrates multiple punctate in a patient with metastatic melanoma. (d) Axial post-
foci of susceptibility bilaterally at cortical and cortical–- contrast T1-WI (same patient as c) demonstrates enhance-
subcortical junction locations as well as a larger focus in ment of the right temporal (white arrow) and left thalamic
the right temporal lobe. Findings favored to represent (black arrow) lesions
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 251

Fig. 15 Patient with cerebral autosomal dominant higher location again shows the thalamic
arteriopathy with subcortical infarcts and leukoence- microhemorrhages. (c) Axial T2-WI in the same patient
phalopathy (CADASIL). (a) Axial T2* GRE image dem- shows multiple hyperintense lesions in the periventricular
onstrates microhemorrhages in bilateral thalami in a patient white matter bilaterally superimposed on diffuse confluent
with CADASIL. (b) Axial T2* GRE image at a slightly deep white matter signal abnormality

susceptibility. For paramagnetic substances, the


increase in magnetic field leads to a negative Nontraumatic Subarachnoid
phase (right-handed system) relative to the sur- Hemorrhage
rounding parenchyma and CSF. At the worksta-
tion, the separately acquired phase shift Nontraumatic subarachnoid hemorrhage (SAH) is
information and the magnitude images combined commonly due to three etiologies: arterial aneu-
into a single image can be displayed using mini- rysm rupture, extension of intraparenchymal hem-
mum intensity projections (MinIP) for ease of orrhage into the subarachnoid space, or
interpretation. The MinIPs are useful in perimesencephalic SAH (likely venous). Aneurys-
confirming the vascular nature of tortuous foci of mal SAH represents the vast majority (>85 %) of
susceptibility by demonstrating contiguity with nontraumatic SAH. Intracranial aneurysms occur
other vascular structures. SWI has been reported in approximately 2 % of the asymptomatic popu-
to provide twofold to fivefold increased sensitiv- lation. In order of descending frequency, the most
ity for the detection of microbleeds versus routine common sites of intracranial aneurysms are the
MPGR T2* sequences. anterior communicating artery (AComm) (30 %),
An important clinical use of SWI is the posterior communicating artery (PComm)
distinguishing diamagnetic materials (e.g., cal- (25 %), the middle cerebral artery (MCA) bifurca-
cium) from paramagnetic materials (e.g., hemo- tion/trifurcation (20 %), the internal carotid artery
siderin) [20]. Calcium and hemosiderin may both (ICA) bifurcation/terminus (7.5 %), basilar tip
present as increased attenuation on unenhanced (7 %), pericallosal arteries (4 %), and the posterior
CT images and as foci of signal hypointensity and inferior cerebellar artery (3 %). Additional miscel-
blooming on conventional gradient echo T2*WI laneous aneurysm locations account for approxi-
images. Diamagnetic materials such as calcium mately 3.5 % of aneurysms (Table 5). While this
appear as high signal on phase images, allowing chapter aims specifically to review the etiologies of
distinction from paramagnetic hemosiderin. nontraumatic SAH, both the radiologist and the
252 S. Quinet and P. Turski

Fig. 16 Inflammatory cerebral amyloid angiopathy. (a) same level of b demonstrates non-enhancing hypointense
Axial T2 FLAIR image demonstrates hyperintense signal signal corresponding to the T2 FLAIR abnormality in the
in the subcortical white matter of the left frontal lobe. (b) left temporal and occipital lobes. Abnormal
Axial T2 FLAIR image a more inferior level in the same leptomeningeal enhancement is present surrounding the
patient demonstrates subcortical white matter signal abnor- signal abnormality. (e) T2* GRE image at a similar level
mality in the right temporo-occipital region. (c) Axial post- to a and c demonstrates punctate foci in the cortex and
contrast T1-WI at the same level of a demonstrates juxtacortical locations in the left frontal lobe representing
non-enhancing hypointense signal corresponding to the microhemorrhages. (f) T2* GRE image at a similar level to
FLAIR abnormality in the right frontal lobe. Abnormal b and d demonstrates punctate foci in the cortex and
leptomeningeal enhancement is present surrounding the juxtacortical locations in the right temporal and parietal
signal abnormality. (d) Axial post-contrast T1-WI at the lobes representing additional microhemorrhages

clinical service should be alert to the clinical sce- evaluate for SAH. On non-contrast-enhanced head
nario of ruptured aneurysm and its concomitant CT, the hallmark of SAH is the identification of
subsequent neurologic deficit leading to the initial hyperdense hemorrhage within the basal cisterns,
traumatic event. Thus, if the source of trauma is not interstices of the sulci, and the interhemispheric
clear, further evaluation with dedicated vascular fissure. Of note, aneurysmal rupture may also result
imaging should be considered, as detailed below. in intraparenchymal hemorrhage in up to one-third
For reasons described in the introduction, of cases. Rare intraparenchymal hemorrhage with-
unenhanced CT is often the modality selected to out associated SAH has also been described in
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 253

Fig. 17 Intraparenchymal hemorrhage of unknown etiol- (b) T2* MR image demonstrates susceptibility-related sig-
ogy in a 24-year-old woman. (a) Initial unenhanced head nal loss within the hematoma. (c–d) Subtracted three-
CT demonstrates large parenchymal hemorrhage centered dimensional phase-contrast MR angiography demonstrates
in the right parietal lobe. Subarachnoid hemorrhage layers excellent subtraction of the hyperintense hematoma pre-
along the sulci of the right parietal and left frontal lobes. sent on the MR images

relation to aneurysm rupture. The aneurysm itself detect with CT. Additionally, the sensitivity of
may occasionally be visible on non-contrast- unenhanced CT for SAH decreases as time since
enhanced CT as a hyperdense round or lobular symptom onset increases, related to both evolu-
structure, a filling defect surrounded by SAH in a tion of blood products and dilution of blood
basilar cistern or fissure, and/or by mural calcifica- within the CSF. Using MR imaging, fluid attenu-
tions, if the aneurysm is long standing. Classifica- ation inversion recovery (T2 FLAIR) sequences
tion systems (Fig. 19, Tables 6 and 7) can be used to are a very sensitive method for detecting SAH
describe both the imaging findings (Fisher grading [21]. However, while FLAIR imaging can be uti-
scale) and the clinical presentation (Hunt and Hess). lized to detect very small amounts of blood in the
Although CT is very sensitive for the detection subarachnoid space in both the acute and subacute
of SAH, if only a small amount of blood is present setting, it is not very specific. Potential etiologies
in the subarachnoid space, it may be difficult to for hyperintense signal in the subarachnoid space
254 S. Quinet and P. Turski

Fig. 18 Comparison of T2* and SWI. (a) Axial T2* GRE image in the same patient shows the increased conspicuity
image demonstrates extensive multiple small hemorrhages of this sequence for microbleeds
related to disseminated aspergillosis. (b) Axial SWAN

Table 5 Frequency of intracranial aneurysm by location Furthermore, if using two-dimensional image


Location Frequency (%) acquisition, inflow enhancement of the CSF
AComm 30 within the subarachnoid space can also have a
PComm 25 similar appearance; this effect is eliminated with
MCA bifurcation/trifurcation 20 use of a 3D acquisition technique. Finally, incom-
ICA bifurcation/terminus 7.5 plete nulling of CSF signal may give false-
Basilar artery tip 7 positive findings in the subarachnoid space, par-
Pericallosal arteries 4 ticularly a problem in the basilar cisterns.
Posterior inferior cerebellar artery 3 The pattern of SAH may suggest the location
Others 3.5 of a culprit aneurysm. For example, SAH concen-
trated in the anterior interhemispheric fissure sug-
gests an aneurysm arising from the AComm, and
on FLAIR imaging other than SAH include SAH asymmetrically involving one of the Sylvian
increased protein content within the subarachnoid fissures implicates the ipsilateral MCA. When
space (e.g., meningitis), retained circulating detected, the patient usually proceeds to CTA
gadolinium-based contrast material from a prior expeditiously to identify a potential culprit aneu-
contrast-enhanced scan in a patient with renal rysm for treatment planning. CTA is currently the
failure, high oxygen tension (e.g., patients on or imaging modality of choice in working up acute
near 100 % supplemental oxygen), and intubated nontraumatic SAH [22, 23]. Regarding detection
or sedated patients receiving propofol. Metal arti- of SAH and aneurysms >3 mm, CT and CTA
fact (e.g., braces, ventriculostomy shunt catheter demonstrate a sensitivity of 98–100 %, specificity
hardware) can interfere with the inversion pulse of 100 %, positive predictive value of 98–100 %,
applied during the FLAIR sequence, resulting in negative predictive value of 96–100 %, and accu-
incomplete nulling of the cerebrospinal fluid. racy of 99 %. Smaller aneurysms <=3 mm may
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 255

Fig. 19 Series of
unenhanced head CT
examinations with
subarachnoid hemorrhage
of varying degrees of
severity demonstrating
Fisher grades 2–4. (a–b)
Fisher grade 1. Thin
subarachnoid hemorrhage
layers within the suprasellar
cistern, anterior
interhemispheric fissure,
and the Sylvian fissures.
(c–d) In a different patient,
a similar pattern of
subarachnoid hemorrhage
in the basilar cisterns is
seen, but the presence of a
focal clot in the
interhemispheric fissure
upgrades this case to Fisher
grade 2. (e–f) The presence
of intraventricular
hemorrhage (causing
obstructive hydrocephalus,
in this case) results in this
case being classified as
Fisher grade 4. Note the
presence of right posterior
approach ventriculostomy
catheter placed to
decompress the dilated
lateral ventricles
256 S. Quinet and P. Turski

Table 6 Fisher grading scale for subarachnoid hemor- arising from the parent vessel wall. It is imperative
rhage (SAH) to evaluate both source images, MIPs, and recon-
Grade CT findings structions as important morphologic characteris-
1 No subarachnoid hemorrhage tics of the aneurysm may be best illustrated on
2 Diffuse blood only one these displays (Fig. 20).
Clot <3 mm Aneurysms can be subdivided by morphology,
Thin layer <1 mm
including saccular (most commonly responsible
3 Clot >3 mm
Thick layer >1 mm for aneurysmal SAH), mycotic or oncotic,
4 Intraventricular hemorrhage dissecting, fusiform, and giant (>20 mm). The
morphology of the neck of the aneurysm (e.g.,
broad vs. narrow) determines whether the aneu-
rysm would be more amenable to coiling versus
Table 7 Hunt and Hess grading scale for subarachnoid clipping. Likewise, the fundus-to-neck ratio may
hemorrhage (SAH)
be of benefit for pretreatment planning. Further-
Grade Clinical findings more, the presence of calcifications at the aneurysm
1 Asymptomatic or minimal headache and slight neck and the presence of vessels arising from the
nuchal rigidity
dome of aneurysm also impact treatment. Addi-
2 Moderate-to-severe headache, nuchal rigidity,
and no neurologic deficit other than cranial tional descriptors include pear-shaped, spherical,
nerve palsy beehive, blister-like, and lobulated. Occasionally,
3 Drowsiness, confusion, of mild focal deficit an aneurysm be partially or completely thrombosed
4 Stupor, moderate-to-severe hemiparesis, and, on initial or follow-up imaging. Giant aneurysms,
possibly, early decerebrate rigidity and defined as aneurysms measuring greater than
vegetative disturbances
20 mm, are more likely to present on the basis of
5 Deep coma, decerebrate rigidity, moribund
appearance symptoms due to mass effect, rather than due to
SAH/hemorrhagic rupture.
The radiologist must be aware of potential emer-
gent complications associated with acute SAH.
be difficult to detect using CTA due to limits of Acute hydrocephalus can develop very rapidly in
spatial resolution of the modality. A thorough these patients. Particular attention should be paid
search for additional asymptomatic aneurysms to the temporal horns of the lateral ventricles,
once the symptomatic lesion is identified is cru- which should be only a few millimeters in diameter
cial, as intracranial aneurysms are multiple in younger patients. The temporal horns can
approximately 20 % of the time. dilate very quickly, along with the third ventricle
Several different display techniques are used in the setting of acute SAH, and thus should be
during the interpretation of CTA images for SAH scrutinized closely in follow-up examinations, as
and/or aneurysm work-up. Maximum intensity these may be the earliest indicators of developing
projection (MIP) images are generated from the hydrocephalus.
axial source images. The source images also can The annual rate of aneurysmal rupture for a
be reconstructed into three-dimensional single aneurysm is approximately 0.7–1 %.
(3D) models, providing the ability to show the Thus, aneurysm risk stratification is an important
aneurysm in more detail. Specifically, the 3D part of initial and follow-up imaging of patients
reconstructions can be rotated to best demonstrate with intracranial aneurysms. Findings associated
the size and morphology of the aneurysm with higher risk of aneurysmal rupture include
[24]. Furthermore, these reconstructions offer the growth, PComm or posterior circulation location,
ability to delineate the relevant intracerebral vas- tobacco smoking, initial size >5 mm at presenta-
cular anatomy important for presurgical/ tion, and the presence of a daughter aneurysm or a
preprocedural planning. On CTA, aneurysms are multi-lobulated appearance (Figs. 21 and 22)
identified as contrast-opacified outpouchings [25–29]. Thus, despite a widely used threshold
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 257

Fig. 20 (continued)
258 S. Quinet and P. Turski

Fig. 21 Ruptured bilobed AComm aneurysm. Initial intensity reconstructed CTA image (d) shows that the
unenhanced head CT images (a, b) demonstrate extensive aneurysm extends from the cephalad aspect of the
subarachnoid hemorrhage predominantly in the basilar AComm. Rotated three-dimensional reconstructed image
cisterns, anterior interhemispheric fissure, and Sylvian fis- from the CTA source data (e) shows to better advantage the
sure. A small amount of hemorrhage layers dependently bilobed morphology of the aneurysm. Lateral projection
within the frontal horn of the left lateral ventricle. Axial digital subtraction angiography via a right ICA injection (f)
source images from subsequently performed CTA (c) dem- again shows the aneurysm
onstrate a bilobed AComm aneurysm. Sagittal maximum

of 7 mm for treatment of intracranial aneurysms, Advanced vascular imaging techniques are


continued imaging surveillance of patients with able to determine and quantify flow within aneu-
aneurysms <7 mm is warranted. Clinically, rysms and arteriovenous malformations (AVMs).
hypertension and tobacco use are associated with Research is ongoing regarding the stratification of
increased risk of aneurysm rupture. patients into high- and low-risk categories for

Fig. 20 A 53-year-old woman presenting with the worst Sequential axial source images from CTA demonstrate a
headache of her life. (a–c) Series of unenhanced axial CT multi-lobulated right superior hypophyseal artery aneu-
images from inferior to superior demonstrate hyperdense rysm (arrows), extending posteriorly and inferiorly into
subarachnoid hemorrhage within the basilar cisterns, the parasellar region. (f) Three-dimensional reformatted
including the suprasellar cistern, interpeduncular fossa, image reconstructed from CTA source image best demon-
and perimesencephalic cistern. Blood is also present within strates a bleb/daughter aneurysm arising from the posterior
bilateral, left-greater-than-right, Sylvian fissures. The tem- aspect of the aneurysm (arrow). The bleb/daughter aneu-
poral horns of the lateral ventricles are dilated representing rysm area is at high risk for rupture
acute communicating obstructive hydrocephalus. (d–e)
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 259

Fig. 22 Risk factors for


aneurysm rupture and
hemorrhage. (a) Time-of-
flight MRA image
demonstrates an inflow jet
within an AComm
aneurysm. (b) Axial source
images from CTA
demonstrate a lobular
AComm aneurysm with a
small bleb at its anterior
margin. (c) MIP images
from the same study
demonstrate the bleb to
better advantage (arrow).
(d) Three-dimensional
reconstruction from CTA
demonstrates the
morphology of the
aneurysm with superb detail

spontaneous aneurysmal rupture. Phase-contrast falsely negative on the basis of only a small
MRA (4DFlow) is emerging as a method to char- amount of aneurysmal hemorrhage, thrombus
acterize the flow features of intracranial aneu- within the aneurysm, and/or vasospasm occurring
rysms. It has been proposed that regions of low during CTA and/or conventional angiography.
wall shear stress (slow flow along the wall) lead to The diagnosis can only be made after repeat diag-
aneurysm wall degeneration and inflammation nostic imaging, CTA and/or DSA, after the acute
contributing to aneurysm growth. Complex episode resolves. Alternatively, the patient may
flow patterns with multiple internal vortices and present with repeat symptomatic SAH, obviously
discrete inflow jets likely promote aneurysm necessitating earlier repeat imaging work-up.
rupture. Therefore, lumbar puncture remains the best
Another clinical scenario is the patient method to exclude CT-negative SAH in patients
presenting with a very small amount of SAH that in whom this entity is strongly suspected
cannot be detected by CT, the so-called “sentinel” clinically.
headache or hemorrhage (Fig. 23). Occasionally, Mycotic aneurysms represent an infectious
initial multimodality work-up of SAH may be process that involves the arterial wall, usually a
260 S. Quinet and P. Turski

Fig. 23 Sentinel hemorrhage. (a) Unenhanced CT dem- sentinel hemorrhage. (e) Initial pre-contrast imaging dem-
onstrates hyperdense subarachnoid blood layering along onstrates accumulation of subarachnoid blood predomi-
the sulci of the right temporal lobe. (b) Unenhanced head nantly within the right suprasellar cistern and interposed
CT slightly more cephalad than a demonstrates blood between the medial temporal lobe and the brainstem. (f)
within the suprasellar cistern. (c) T2 FLAIR MR image Axial source images from CTA demonstrate a saccular
demonstrates hyperintense subarachnoid hemorrhage aneurysm from the right PComm. (g) Maximum intensity
along the sulci of the left temporal lobe and within the projection (MIP) images show the right PComm aneurysm
suprasellar cistern, as seen on CT. (d) T2 FLAIR MR to better advantage. (h) Conventional catheter digital sub-
image at a more cephalad location also reveals subarach- traction angiography in the AP projection demonstrates
noid blood along the sulci of the right parietal lobe more vasospasm at the time of diagnosis indicating prior sentinel
posteriorly. Follow-up CTA examination in patient with hemorrhage
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 261

Fig. 24 Perimesencephalic subarachnoid hemorrhage. around the perimesencephalic cistern bilaterally. The
(a–c) Series of unenhanced head CT images from inferior patient subsequently underwent two conventional catheter
to superior demonstrate characteristic location of hemor- angiographic examinations which were normal
rhage within the interpeduncular fossa and wrapping

distal branch. Mycotic aneurysms may present as essentially a diagnosis of exclusion, arrived at
acute intracranial hemorrhages in locations atypi- after excluding an arterial source of the bleed. An
cal for aneurysmal hemorrhage and may raise initial CTA is usually the modality of choice to
suspicion for an underlying AVM, other vascular search for culprit lesions. Whether to follow a
malformation, or hemorrhagic mass. negative CTA with conventional angiography
Nontraumatic SAH may be non-aneurysmal in shortly after the initial CTA versus only follow-up
etiology, with perimesencephalic SAH an impor- with another CTA is controversial and varies based
tant differential diagnostic consideration, albeit on personal, institutional, and/or regional prefer-
one which is a diagnosis of exclusion. This entity ence. However, at a minimum, at least one follow-
has been reportedly responsible for up to up CTA examination is routinely performed sev-
two-thirds of angiographically negative SAH. eral weeks out, after the SAH has cleared, to search
Perimesencephalic SAH often presents as a for underlying aneurysm or vascular malformation
small amount SAH within the prepontine cistern that was potentially occult or obscured on initial
and extending along the perimesencephalic cistern imaging.
(Fig. 24). Occasionally, SAH is seen in basal por- Pseudosubarachnoid hemorrhage is an imag-
tions of the Sylvian fissures. This appearance may ing phenomenon related to marked, severe cere-
mimic the rupture of a basilar tip aneurysm. bral edema and swelling, seen in the setting of
Patients presenting with perimesencephalic SAH global anoxic injury. Imaging features of cerebral
characteristically follow a more benign clinical edema are seen, with loss of gray–white
course compared to those presenting with SAH differentiation, compression of sulci, and
due to other causes. Perimesencephalic SAH rarely hypoattenuating cerebral cortex (Fig. 25). As the
produces vasospasm and is felt likely to be venous brain swells, there is hyperemia and congestion of
in origin. Anomalies of the basal vein of Rosenthal the meninges at the base of the brain. The
have been found to be associated with perimesen- congested meninges appear to demonstrate
cephalic SAH. Perimesencephalic SAH is increased attenuation relative to brain tissue.
262 S. Quinet and P. Turski

Fig. 25 Pseudosubarachnoid hemorrhage in a patient and effacement of sulci and gyri, indicating diffuse cere-
with anoxic injury. (a–f) Series of unenhanced CT images bral edema. The congested meninges of the basal cisterns
in a patient presenting after an anoxic event. Note the appear thickened and hyperdense juxtaposed against the
diffuse hypoattenuation throughout the cortex and subcor- edematous brain parenchyma
tical white matter, with loss of gray–white differentiation,

However, the meninges are relatively normal in AVMs and dural arteriovenous fistulas (AVFs). Cav-
attenuation, but appear prominent and engorged ernous malformations (cavernomas), developmental
on the basis of congestion. The appearance of venous anomalies (DVAs), and capillary telangiec-
congested basilar meninges juxtaposed to brain tasias are other vascular malformations that are also
tissue markedly reduced in attenuation accentuates common causes of nontraumatic intracranial hem-
the attenuation difference between these structures, orrhage in the young adult population and will be
falsely giving the appearance that there is diffuse also covered in this section. Though capillary tel-
hyperattenuation in the subarachnoid space. angiectasias may arise on a congenital or sponta-
neous basis, given the increased incidence of these
lesions in the postradiation treatment setting, the
Vascular Malformations imaging appearance of capillary telangiectasias is
covered in the section of radiation-induced vascu-
Arteriovenous malformations (AVMs) and other lar malformations.
vascular malformations are the most common Pial AVMs are developmental lesions that tend
causes of spontaneous intracranial hemorrhage in to present in younger adults, aged 30–60 years.
young adults. Subtypes of AVM include pial Fifty percent of patients with pial AVMs present
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 263

Fig. 26 Pial arteriovenous malformation in a 32-year-old expansion of the right frontal lobar hemorrhage. Note is
man presenting with headache and nausea/vomiting. (a) made of a tangle of abnormal vascular structures at the
Axial unenhanced head CT demonstrates intrapar- superolateral aspect of the hematoma. (c) Lateral projec-
enchymal hemorrhage centered in the right frontal lobe, tion from catheter angiography via right internal carotid
with extension into the right lateral ventricle. (b) Axial injection demonstrates arterial vessels supplying a nidus
source images from subsequent CTA demonstrate along with early opacification of the draining veins

Fig. 27 A 65-year-old male presents with newly diag- level demonstrates hypertrophied feeding arterial vascular
nosed pial AVM presenting as spontaneous intracranial structures arising from the left MCA distribution, a large
hemorrhage. Presenting symptoms included severe head- partially calcified nidus, and a dominant draining vein
ache, nausea/vomiting, and lethargy upon awakening. (a) emptying into the left cerebral vein. (c) Lateral projection
Axial image from initial unenhanced head CT demon- from catheter angiography via left ICA injection demon-
strates intraventricular hemorrhage, obstructive hydro- strates en passant angular artery supplying the AVM nidus,
cephalus, and a partially calcified lesion near the but also continuing on to supply normal brain tissue. A few
ependymal surface of the left lateral ventricle. (b) Axial Rolandic branches are seen at the cephalad aspect, feeding
source image from subsequently obtained CTA at a similar the AVM nidus

with intraparenchymal hemorrhage. Pial AVMS prominent nidus which can be partially calcified,
are defined by an anomalous connection between and large draining veins (Figs. 26 and 27). On
pial arteries and veins in the absence of an inter- MR, flow voids can be seen within the larger,
vening capillary bed [30]. The classic imaging more high-flow vascular components of the
appearance of AVM includes the presence of mul- AVM. Arterial feeders to pial AVMs may arise
tiple hypertrophied arterial feeding vessels, a from either branches of the ICA or the
264 S. Quinet and P. Turski

vertebrobasilar system. Aneurysms may be asso- infratentorial location, as well as the spinal canal.
ciated with the arteries supplying the nidus. The Cavernous–orbital syndrome (e.g., exophthalmos)
intraparenchymal nidus is often surrounded by may result from dAVF involving the cavernous
gliotic brain tissue. sinus, while pulsatile tinnitus and/or a clinical
While spontaneous intracranial hemorrhage is sign of a bruit may be a clinical manifestation of
the most typical presentation of patients with pial dAVFs involving the transverse and/or sigmoid
AVMs, these lesions may less commonly be sinuses.
detected in patients presenting with seizures or Dural AVFs can be idiopathic, congenital, or
undergoing work-up for other clinical indications. acquired lesions. In the acquired setting, these
It is important to recognize that CTA, MRA, and lesions often arise due to prior occlusion of a
even conventional angiography may be falsely dural venous sinus. Venous hypertension, prior
negative in the setting of acute hematoma related craniotomy, and trauma have also been found to
to a pial AVM due to mass effect and compression be associated with the development of dAVF.
of the hematoma on the supplying arteries, the While CTA and MRA are helpful in the evaluation
nidus, and/or the draining vein. Thus, repeat angio- and diagnosis of dAVF, conventional angiography
graphic imaging should be performed after resolu- remains the gold standard diagnostic modality, as
tion of the hematoma if there is strong clinical the classification and the risk of subsequent hem-
suspicion for this entity. The Spetzler–Martin grad- orrhage are predicated on the pattern of venous
ing scheme attempts to stratify AVMs based on drainage of these lesions (Fig. 28). Additionally,
surgical risk and takes into account the size of the angiography-directed catheter-based therapy is
AVM, whether the lesion involves eloquent cortex, often the treatment modality of choice for uncom-
and the pattern of venous drainage (e.g., superficial plicated dAVFs. Importantly, the presence of
and/or deep). The risk of hemorrhage of an transosseous vascular channels should alert one
unruptured AVM is about 2 % per year, while the to the potential presence of a dural AVF.
chance of recurrent hemorrhage related to a previ- The Borden and Cognard classification schema
ously hemorrhagic AVM has been reported as up to have been developed to stratify dAVFs into
18 % in the first year after the initial hemorrhage. “benign” and “aggressive” subtypes, based on
Additional risk factors for hemorrhage include the site of venous drainage (specifically, the pres-
older age at presentation, exclusively deep venous ence or absence of cortical venous drainage), the
drainage, and deep location of the AVM. direction of flow within the involved or adjacent
Dural AVFs account for approximately 10–15 % dural sinus, and venous outflow architecture
of intracranial AVMs (accounting for 6 % of (Tables 8 and 9). These subtypes are intended to
supratentorial and 35 % of infratentorial vascular predict the likelihood of hemorrhage as well as
malformations) and are characterized by anomalous nonhemorrhagic neurologic deficits. In consider-
arteriovenous shunting predominantly between ing these subtypes, it is important to note that
supplying meningeal arteries and draining dural there is rare progression of “benign” subtypes to
venous sinuses or meningeal or cortical veins “aggressive” ones (approximately 2 %). Similarly,
[31]. In contradistinction to pial AVMs, no intrapar- spontaneous thrombosis and regression of dAVFs
enchymal nidus is present in dAVFs. In patients have also been described. As such, close clinical
with dAVF, venous ectasia is the finding most and imaging follow-up is necessary to exclude
highly associated with risk of hemorrhage using progression of existing “benign” lesions, espe-
the Cognard classification scheme (60 %). Patients cially in the setting of new symptoms referable
with dAVFs not drained by cortical veins, and there- to these lesions, which may herald a change in the
fore at lower risk of hemorrhagic complications, pattern of venous drainage.
may initially present clinically with symptoms Carotid-cavernous fistulas (CCFs) represent
related to dural venous hypertension. Involvement abnormal communication between the carotid arte-
of the cavernous sinus and posterior fossa is most rial system and the cavernous sinus. CCFs are
common, though dAVFs may involve any supra- or subdivided into direct, high-flow CCF (type A)
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 265

Fig. 28 (continued)
266 S. Quinet and P. Turski

Fig. 28 Dural arteriovenous fistula. (a) Sagittal occipital artery with direct communication to the trans-
reformatted non-contrast-enhanced head CT demonstrates verse sinus. Note is made of early venous drainage into
hemorrhage within the cerebellum and along the floor of cortical veins, some of which are ectatic (black arrows).
the posterior fossa extending into the foramen magnum. AP (e) and sagittal (f) projections from subsequent DSA
(b) Sagittal reformatted maximum intensity projection performed from a left external carotid injection demon-
image from CTA shows multiple dilated contrast-opacified strate multiple small arterial feeders (open white arrows)
vascular structures within the posterior fossa cerebellar arising from the left occipital artery with direct communi-
interhemispheric region just inferior to the torcular cation to the transverse sinus. Note is made of early venous
Herophili without obvious nidus visualized. A large varix drainage into cortical veins, some of which are ectatic
is identified posteriorly near the midline. AP (c) and lateral (open black arrows). Additional AP (g) and lateral (h)
(d) projections from subsequent DSA performed from a DSA images from selective right occipital artery injection
right external carotid injection demonstrate multiple small and AP (i) and lateral (j) DSA images from selective left
arterial feeders (white arrows) arising from the right occipital artery branch injection confirm the above findings

and low-flow, dural CCFs (types B–D). Direct While digital subtraction angiography remains
CCFs are high-flow lesions representing direct the gold standard for diagnosis of CCF, a number
communication between the cavernous ICA and of findings on CTA/MRA can suggest this diag-
the cavernous sinus, most commonly arising in nosis, including asymmetric enlargement of the
the setting of prior trauma or rupture of an ICA cavernous sinus ipsilateral to the CCF, enlarge-
aneurysm [32]. Rarely, an underlying connective ment of dural venous sinuses due to increased
tissue disease such as Ehlers–Danlos or venous pressure, and early enhancement of the
fibromuscular dysplasia may be the cause. Patients cavernous sinus in the arterial phase. Addition-
with direct CCFs typically present acutely, ally, orbital abnormalities on the basis of venous
with rapidly developing symptoms. Dural CCFs engorgement and increased venous outflow from
represent abnormal communication between the the cavernous sinus are also characteristic. Typical
cavernous sinus and multiple branches of the ICA findings include enlargement of the superior oph-
(type B), ECA (type C), or both (type D). thalmic vein, proptosis, enlargement of the
The clinical presentation of dural CCFs is charac- extraocular muscles, and edema within the orbital
terized by the insidious onset of symptoms. Dural fat. Subarachnoid and/or intraparenchymal hem-
CCFs often occur in the setting of chronic hyper- orrhage may occur due to rupture of a cortical vein
tension. Dural CCFs may represent sequelae of (Fig. 29).
cavernous sinus thrombosis and subsequent Cavernous malformations (cavernomas) are
revascularization. low-flow vascular malformations. Cavernomas
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 267

Table 8 Cognard classification of dural arteriovenous dark rim. This hypointense rim is especially nota-
fistulas (dAVFs) ble on GRE and SWI sequences, particularly at
Other Risk of higher field strengths, with small lesions occa-
Type Drainage characteristics hemorrhage sionally detectable only on these sequences [34].
I Dural venous Anterograde Low Cavernomas lack both a nidus and enlarged sup-
sinus flow in sinus
plying arteries, distinguishing these lesions from
IIa Dural venous Retrograde Low
sinus flow in sinus pial AVMs.
IIb Dural venous Anterograde Moderate Cavernomas can present as single or multiple
sinus flow in sinus + lesions and can occur sporadically or as an auto-
cortical vein somal dominant condition with variable penetra-
reflux
tion (Fig. 30). Clinically, these patients can
IIa + b Dural venous Retrograde Moderate
sinus flow in sinus +
present with headaches and/or seizures with spon-
cortical vein taneous hemorrhage being a less common presen-
reflux tation. However, cavernomas are often detected
III Cortical vein Cortical vein High incidentally. It is estimated that 10–33 % of
drainage only cavernomas are associated with developmental
IV Cortical vein Cortical vein High
venous anomalies (DVAs) at the time of diagnosis
drainage only +
venous ectasia (Fig. 31). This fact can be exploited using post-
V Cortical vein Associated High contrast imaging; if a DVA is identified adjacent
+ spinal with to an intraparenchymal hematoma, a ruptured
perimedullary progressive cavernoma compressed by the hematoma is
veins myelopathy
strongly suspected, given the extremely low like-
lihood that the DVA itself is responsible for the
Table 9 Borden classification of dural arteriovenous fis-
hemorrhage [35]. It is important to note that a
tulas (dAVFs) partially calcified lesion on unenhanced CT
Other Risk of
carries a differential diagnosis that includes
Type Drainage characteristics hemorrhage AVM and a calcified neoplasm, so further imaging
I Dural No cortical vein Low evaluation, ideally with contrast-enhanced MR
venous reflux and MRA, is recommended. In contradistinction
sinus to other intracranial vascular malformations,
II Dural Cortical vein Moderate cavernomas are most often angiographically
venous reflux
sinus occult, and as such, catheter angiography is not
III Cortical Cortical vein High routinely part of the work-up of these lesions.
vein drainage only DVAs are common and are seen routinely as
incidental lesions on post-contrast CT and MR
imaging. It is important to recognize that DVAs
pathologically are hamartomatous lesions com- represent the sole venous drainage of the affected
posed of single-layered endothelial-lined vessels brain, and thrombosis or resection of these lesions
containing blood and, occasionally, calcification will result in infarction of the drained territory. A
or mineralization, without intervening normal “caput medusa” appearance is common, with mul-
brain tissue [33]. As such, cavernomas may tiple small tributaries converging upon a single
appear as areas of indistinct or amorphous subtle large draining vein, extending to the ependymal
hyperdensity on CT. Cavernomas can also dem- surface of the lateral ventricle or to a cortical vein.
onstrate subtle hypodensity on unenhanced DVAs rarely hemorrhage unless associated with a
CT. On MR, these lesions are classically cavernoma or thrombosis of the prominent anom-
described as heterogeneous in signal intensity, alous draining vein, leading to venous infarct.
with areas of “popcorn-like” bright signal cen- Advanced perfusion imaging techniques, either
trally on both T1- and T2-WI sequences with a CTP or MRP, may demonstrate prolonged mean
268 S. Quinet and P. Turski

transit times and/or increased relative cerebral in the subcortical white matter surrounding an
blood volume, which may indicate venous con- unusually large DVA, which may represent
gestion within these engorged anomalous venous edema and/or gliosis related to the venous con-
structures. Occasionally, non-enhancing gestion and relative ischemia (Fig. 32). Further-
T2/FLAIR signal hyperintensity may be identified more, a recent publication has described the

Fig. 29 (continued)
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 269

ä
Fig. 29 (continued)
270 S. Quinet and P. Turski

presence of hypointense foci associated with and type 2 (minimal mass effect). On imaging,
DVAs in almost two-thirds of patients when petechial hemorrhage is punctate or confluent in
imaged with susceptibility-weighted sequences morphology with little or no associated mass
at 3-T [36]. It is thought that this hypointense effect, with a predilection for involvement of
signal represents microhemorrhage and/or small deep gray matter. The other type, which is more
cavernomas, and the increased sensitivity of SWI clinically significant, is parenchymal hematoma.
suggests that these entities are more commonly Parenchymal hematoma is divided into type
associated with DVAs than previously thought. 1 (hematoma involving <= 30 % of the infarct
Capillary telangiectasias represent dilated cap- area) and type 2 (involvement of >30 % of the
illaries with intervening normal brain tissue. Tel- infarct area). If the hematoma is relatively small,
angiectasias are most often clinically silent and these are usually not of major clinical conse-
the importance of their identification is quence. Type 2 parenchymal hematomas are asso-
distinguishing them from other lesions. Typical ciated with clinical deterioration and poor
locations include the brainstem (especially the outcome. Hemorrhagic transformation can be
pons), cerebellum, and spinal cord. On imaging, seen most commonly a few days after the infarct;
they classically present as focal subcentimeter, however, earlier transformation can be seen in the
non-mass-like areas of hypointense signal on setting of pharmacologic or mechanical
T2* and SWI sequences with associated thrombolysis.
ill-defined, “brushlike” enhancement (Fig. 33). The presence of preexisting microhemorrhages
Telangiectasias are often occult on other conven- correlates with a worse prognosis after post-
tional MR sequences. They are most often solitary thrombolysis reperfusion, presumably due to
in the sporadic form, while multiple lesions are microvasculature structures being diseased and
more commonly seen in the setting of prior radi- damaged (Fig. 35). These patients are also more
ation. Radiation-induced telangiectasias will be likely to have parenchymal hemorrhages. Resto-
covered later in the chapter. ration of blood flow to severely ischemic tissue
has been shown to increase the likelihood of hem-
orrhagic transformation, specifically the more
Hemorrhagic Transformation of Infarct clinically significant entity of parenchymal hema-
toma. Two of the most accepted and long-standing
There are two types of hemorrhage that can occur predictors of hemorrhagic transformation on
following reperfusion of an infarct (Fig. 34, unenhanced head CT are the extent of confluent
Table 10) [37]. The first is a petechial type of hypoattenuation in the cortex and subcortical
hemorrhage, representing a relatively normal evo- white matter (e.g., >1/3 MCA territory) and the
lution of infarct. It is an expected finding after presence of hyperattenuating thrombus within the
reperfusion in the area of infarct. Petechial hem- MCA (“dense MCA sign”). The use of CT perfu-
orrhage is subdivided into type 1 (no mass effect) sion (CTP)-derived blood–brain barrier

Fig. 29 Acquired traumatic carotid–cavernous fistula in a parietal cortical veins. The right frontal lobar hemorrhage
60-year-old woman with history of recurrent falls resulting is again seen. Frontal (e) and lateral (f) digital subtraction
in subdural hematomas. (a) Initial unenhanced head CT angiography images performed from a right ICA injection
demonstrates an acute intraparenchymal hematoma cen- in the early arterial phase demonstrate early filling of the
tered within the right frontal lobe. (b) Axial CTA source cavernous sinus. Lateral arterial phase images from the
image demonstrates early contrast enhancement and same injection in the frontal (g) and lateral (h) projections
enlargement of the right cavernous sinus (arrow) and the show filling of the contralateral cavernous sinus and its
right superior ophthalmic vein. Right proptosis is also drainage pathways including dilated right superior oph-
evident. (c) A more cephalad image from the same CTA thalmic veins. The dilated right cerebral veins seen on
again shows the dilated, early draining right superior oph- prior CTA are visualized. Post-coiling frontal (i) and lateral
thalmic vein. (d) An additional more cephalad image from (j) DSA projections show exclusion of the
the same study shows engorgement of right frontal and carotid–cavernous fistula
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 271

Fig. 30 Series of images in


a patient with familial
multiple cavernomas
(autosomal dominant). (a)
Axial unenhanced head CT
image at the level of the
brachium pontis
demonstrates a well-defined
round heterogeneous
attenuation lesion, with
both hyperdense and
hypodense components. (b)
Axial unenhanced head CT
image at a slightly more
inferior slice further
demonstrates the extent of
the lesion. (c) Sagittal
T1-WI without contrast
demonstrates
heterogeneous signal within
the lesion, with
predominantly T1
hyperintense rim and some
central T1 hyperintensity.
(d) Axial T2-WI without
contrast demonstrates the
classic “popcorn”-type
appearance of the lesion,
with mixed central T2
signal and a hypointense
rim, representing the
methemoglobin and
hemosiderin components of
the lesion, respectively. (e)
Axial T2-WI demonstrates
a smaller but similar
appearing lesion at the
cortical–subcortical
junction in the right
temporal lobe. (f) Axial
T2-WI at more cephalad
location demonstrates
another cavernoma in the
right frontal parasagittal
region

permeability measurements has been shown to be (rCBF), reduced relative cerebral blood volume
helpful in predicting hemorrhagic transformation (rCBV), and delayed transit times [38, 39]. In
in the setting of acute ischemic stroke, including particular, severe hypoperfusion, as demonstrated
findings of reduced relative cerebral blood flow by Tmax >14 s, has been shown to be predictive
272 S. Quinet and P. Turski

Fig. 31 Developmental venous anomaly and cavernoma, demonstrates increased signal in the right frontal lobe
sporadic pattern. Unenhanced axial head CT (a) demon- corresponding to the hyperattenuation on CT. Final multi-
strates faint hyperattenuation in the right frontal lobe plied and processed SWI image (d) demonstrates
(arrow). Contrast-enhanced T1 MR image (b) demon- hypointensity at this location. This appearance on phase
strates partial visualization of a right frontal developmental imaging and SWI is due to the susceptibility of effects of
venous anomaly (open arrow). Filtered phase image (c) diamagnetic calcium within the cavernoma

of the development of parenchymal hematoma. areas of very low CBV, large DWI lesion volume,
CTP-derived blood–brain permeability (BBBP) a very low ADC, and Tmax >14 s. Additional
and PWI-based assessments of BBB permeability measures of permeability on MR imaging have
measurements have also shown promise in also been suggested as predictive of post-
predicting hemorrhagic transformation. Using thrombolysis hemorrhage, including late-phase
MR, severely ischemic tissue is characterized by T2* signal loss in perfusion sequences or
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 273

Fig. 32 Series of images in a patient with a right frontal location shows similar signal but decreased conspicuity
developmental venous anomaly. (a) Axial post-contrast of the lesion, relative to its appearance on SWI. (d) Axial
T1-WI demonstrates abnormal enhancing tubular struc- T2/FLAIR image shows confluent signal hyperintensity in
tures in the right frontal lobe converging upon and draining the white matter in the region of the DVA, likely
into an ependymal vein in the right parasagittal region. (b) representing gliosis/leukoaraiosis in the territory drained
Axial SWI image at a similar location demonstrates by the DVA. MR perfusion imaging demonstrates elevated
hypointense signal within these enhancing structures, on cerebral blood volume (e) and prolonged first moment
the basis of deoxyhemoglobin in this slow-flow vascular transit times (f) to the territory of this lesion, probably on
malformation. (c) Axial T2* GRE image at a similar the basis of venous congestion

post-contrast FLAIR enhancement. An additional cerebrospinal fluid on post-contrast T2/FLAIR


potential sign thought to be predictive of hemor- images, may be a predictor of hemorrhagic
rhagic transformation includes early development transformation [40].
of T2/FLAIR signal hyperintensity within the area The European Cooperative Acute Stroke Study
of DWI in an acute infarct, indicating a more (ECASS) III randomized control trial demon-
severe ischemic injury. Furthermore, the strated the increased risk of symptomatic intracra-
hyperintense acute reperfusion marker (HARM) nial hemorrhage in the setting of patients with
sign, which has been shown to represent leakage acute ischemic stroke treated with intravenous
of gadolinium-based contrast agents across a alteplase thrombolysis compared to placebo
damaged blood–brain barrier into the [41]. In the ECASS III protocol, symptomatic
274 S. Quinet and P. Turski

Fig. 33 Series of MR images in a patient with presumed related signal loss on T2*, with increased conspicuity on
brainstem capillary telangiectasia. Axial unenhanced T1 SWI. The lesion is not apparent on T1 and T2 FSE
(a), post-contrast T1 (b), T2 (c), T2* (d), and SW (e) sequences. The lesion did not produce symptoms referable
images demonstrate a presumed capillary telangiectasia to its location and was stable on serial examinations
in the right paramedian pons (arrows). The lesion demon- performed for other reasons
strates faint, brushlike enhancement and susceptibility-

intracranial hemorrhage was defined as “any symptomatic ICH. Though less intensively stud-
apparently extravascular blood in the brain or ied, CTP-derived permeability measurements
within the cranium that was associated with clin- have shown promise in predicting development
ical deterioration, as defined by an increase of four of malignant edema following thrombolysis.
points or more in the score on the NIHSS, or that Malignant edema was defined in the ECASS III
led to death and that was identified as the predom- trial as brain edema with mass effect as the pre-
inant cause of the neurologic deterioration.” dominant cause of clinical deterioration (NIHSS
Importantly, despite the increased incidence of score increase >4 or death).
symptomatic ICH in the thrombolysis group,
there was no difference in mortality. The results
from an earlier arm of the study (ECASS II) are Venous Thrombosis
the basis for the well-established association
between the extent of parenchymal Thrombosis of a cortical vein and/or dural sinus
hypoattenuation on initial unenhanced head CT with subsequent hemorrhage is an important con-
and risk for symptomatic ICH. Hypoattenuation sideration in evaluating for potential etiologies of
involving more than one-third of the MCA terri- spontaneous intracranial hemorrhages in unusual
tory was associated with increased risk of locations (e.g., not within classic arterial
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 275

Fig. 34 Examples of hemorrhagic transformation. (a) patient demonstrates a hyperdense hemorrhage centered
Axial unenhanced head CT demonstrates hyperdense pete- in the right caudate and the periventricular white matter.
chial hemorrhage centered in the right putamen with sur- Note hypodensity throughout the cortex and subcortical
rounding edema. Given the minimal mass effect, this white matter in the distribution of the right MCA. The
would be classified as a type 2 petechial hemorrhage. (b) extent of hematoma is <30 % and can be classified as a
Axial unenhanced head CT in the same patient demon- type 2 hematoma. (d) Axial unenhanced head CT image at
strates and additionally shows hemorrhage within the right a slightly more cephalad location as C shows to better
caudate. (c) Axial unenhanced head CT in a different advantage the intraventricular extension of hemorrhage

distributions) or seemingly without an associated patient populations. The clinical presentation of


underlying mass or vascular anomaly. Particular patients with venous thrombosis is often not
attention should be paid to this entity in the pedi- straightforward, with severe, persistent headache
atric, young adult female, and hypercoagulable a common reason for referral to imaging.
276 S. Quinet and P. Turski

Table 10 Classification of hemorrhagic transformation unenhanced sagittal T1-WI (Figs. 37 and 38).
(HT) in ischemic strokea The presence of expansion of the sinus or vein
Type I Type II can be a helpful ancillary finding supporting the
Petechial No mass effect Minimal mass presence of thrombosis, particularly if the throm-
hemorrhage effect bus is not obviously T1 hyperintense. Likewise,
Parenchymal <30 % infarct >30 % infarct routine unenhanced spin echo images should be
hematoma territory territory
a
scrutinized for preserved flow voids in venous
Based on European Cooperative Acute Stroke Study
(ECASS) I and II definitions sinuses and cortical veins. However, because
lack of a flow void may be due to either thrombo-
sis or slow flow, additional sequences can be
In the pediatric setting, patients may present with helpful to distinguish these two entities (e.g.,
nausea, vomiting, and irritability. If there is exten- post-contrast imaging, TOF, posterior cerebral
sion of thrombus to involve the cortical veins with artery (PCA), contrast-enhanced time-resolved
subsequent venous infarction, the patient may 4D MRA – 4DCEMRA). TOF or PCA imaging
present with seizures. The sequence of events without and/or with contrast can be performed if
leading intracranial hemorrhage is thought to be there is high clinical suspicion prospectively
due to prolonged venous stasis leading to throm- (Figs. 37 and 38). TOF MRV images demonstrat-
bosis with subsequent venous hypertension, ing lack of flow-related enhancement can be sug-
ischemia, and infarction. gestive of the diagnosis; however, correlation
Given the vague presentation, unenhanced with other sequences is mandatory, as inflow
CT may be the first imaging obtained, depending dephasing may mimic thrombosis and T1
on the level of suspicion for this entity. hyperintense thrombus may mimic flow-related
Hypoattenuation and/or hemorrhage in non-arterial enhancement. Unenhanced TOF and PCA tech-
distributions can be a clue to venous infarction. niques are particularly valuable in the patient pop-
Assessment of venous vascular territories in addi- ulation unable to receive intravenous contrast
tion to those of arterial vascular territories is impor- agents. If post-contrast imaging or 4D CE MRA
tant, as certain patterns of signal abnormality is used, filling defects within the thrombosed
and/or hemorrhage can signal the presence of venous structures can be identified, analogous to
venous thrombosis and suggest the site of throm- post-contrast CT imaging (Fig. 39) [42]. Post-
bus. Examples include the posterior temporal lobe contrast imaging can also be helpful in
(vein of Labbe +/ transverse sinus), parasagittal distinguishing thrombosis versus a hypoplastic
frontoparietal lobes (superior sagittal sinus and/or sinus.
cortical veins), and bilateral thalami (internal cere- Venous infarcts less often demonstrate large
bral veins) (Figs. 36, 37, 38, and 39). areas of restricted diffusion compared to arterial
If infarction has not yet occurred, hyperdense infarcts, and the absence of restricted diffusion
thrombus within venous sinuses or cortical veins does not exclude venous ischemia or infarction
(“cord sign”) may be the only finding visible early (Fig. 36). As discussed above, DWI and ADC
in the process, though the clot tends to become maps should be interpreted with caution given
less dense after a week and may thus be incon- the possibility of T2 shine-through and dark-
spicuous on non-contrast imaging (Fig. 36). through effects complicating interpretation.
Contrast-enhanced CT, CTA, and optimally CTV
classically demonstrate normally enhancing dura
surrounding non-enhancing thrombus with a dural Hemorrhagic Neoplasm
sinus (the “empty delta” sign) (Fig. 37).
On MR imaging, a corollary to the cord sign is Hemorrhage into underlying malignant intracra-
presence of T1 hyperintense methemoglobin nial neoplasms, either primary or metastatic,
within the thrombus. This can be particularly con- accounts for approximately 6–10 % of spontane-
spicuous within the superior sagittal sinus on ous intracranial hematomas. Among primary CNS
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 277

Fig. 35 Risk factors for hemorrhagic transformation. (a) advantage (arrow). (c) Perfusion imaging demonstrates
Axial T2/FLAIR image demonstrates abnormal prolonged mean transit times through the majority of the
hyperintense signal within the cortex and subcortical right MCA distribution. (d) Very low relative cerebral
white matter within the infarcted territory. Note is made blood volume is also noted corresponding to the distribu-
of hypointense microhemorrhage in the left frontal lobe tion of the prolonged transit times. The presence of
(arrow). (b) Axial T2* GRE image at a similar level microhemorrhage increases the risk of hemorrhagic con-
demonstrates the left frontal hemorrhage to better version after thrombolysis

neoplasms, grade IV astrocytoma/glioblastoma and ependymomas. Metastatic lesions related to


multiforme is most likely to present as hemor- melanoma (Fig. 43), choriocarcinoma, renal cell
rhagic masses (Figs. 40, 41, and 42). Additional carcinoma, and thyroid carcinoma are most likely
malignant primary CNS neoplasms with a propen- to hemorrhage. Of note, despite the increased pro-
sity for hemorrhage include oligodendrogliomas pensity of these tumor types to hemorrhage, a
278 S. Quinet and P. Turski

Fig. 36 Series of CT and MR images in a 20-year-old hypointense area in the left thalamus becoming more con-
female patient with Crohn disease presenting with bilateral spicuous on this sequence. (e) Axial T2* GRE image
internal cerebral vein thrombosis. (a) Axial unenhanced demonstrates “blooming” of hypointense areas within the
head CT image demonstrates confluent hypodensity within left thalamus with a hyperintense core and petechial hem-
bilateral thalami, left greater than right. Note hyperdense orrhage in the right thalamus. (f) Axial DWI demonstrates
thrombus in the expected location of the internal cerebral hyperintensity within the left thalamus. (g) Corresponding
veins (arrow). (b) Axial unenhanced head CT image at a axial ADC map image demonstrates hypointensity in the
slightly more inferior location shows the same findings. (c) region of hyperintense signal on DWI, confirming apparent
Axial unenhanced T2-WI at a similar level as the previous diffusion restriction. (h) Sagittal TOF MIP image demon-
image demonstrates predominantly hyperintense signal in strates lack of inflow enhancement of the straight sinus and
the thalami corresponding to hypodensity on the CT bilateral internal cerebral veins. (i) Sagittal MIP flow image
images. Scattered foci of hypointensity are also present from the PC MRA examination demonstrates excellent
within these lesions. (d) Axial T2 FLAIR image at a similar subtraction of stationary tissue and absence of flow signal
location confirms the findings seen on T2-WI, with a in the straight sinus and internal cerebral veins
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 279

Fig. 37 (continued)
280 S. Quinet and P. Turski

Fig. 37 Series of CT and MR images in a 51-year-old frontal cortex and subcortical white matter. There is corti-
patient presenting with seizures and fever in the setting of cal enlargement and loss of gray–white distinction. Note
illicit drug use. (a) Axial image from unenhanced head CT linear hypointense signal at the lateral margin of the right
demonstrates subarachnoid hemorrhage layering along the frontal lobe (arrow) representing a thrombosed cortical
sulci of the right frontal lobe. Hypodensity is also noted vein. (h) Axial T2* GRE image demonstrates blooming
within the cortex and subcortical white matter of the right of hypointense signal within the thrombosed cortical vein
frontal lobe with blurring of gray–white distinction. (b) (arrow). (i) Axial T2* GRE image at a slightly higher level
Enhanced head CT at a slightly more cephalad location shows the further extent of the thrombosed cortical vein
again shows cortical enhancement. (c) Axial image from (arrows). (j) Axial T2/FLAIR image shows abnormal
subsequent CTA in the same patient demonstrates filling hyperintense signal within the cortex and subcortical
defect within the superior sagittal sinus (arrow) indicating white matter of the right frontal lobe. Abnormal
thrombosis at this site. (d) Axial CTA image at a higher hyperintense signal also interdigitates between the sulci
location again shows superior sagittal sinus thrombosis of the right frontal lobe within the subarachnoid space.
(arrow). (e) Sagittal reformatted CTA again shows the (k) Axial post-contrast T1-WI shows abnormal
sagittal sinus thrombosis (arrow). (f) Sagittal unenhanced leptomeningeal enhancement. (l) Axial T2-WI demon-
T1-WI image demonstrates hyperintense thrombus within strates linear hypodense thrombosis within a cortical vein
the superior sagittal sinus (arrow). (g) Coronal T2 FLAIR (arrow). (m) Axial DWI demonstrates hyperintensity
image demonstrates hyperintense signal within the right within the right frontal cortex

hemorrhagic metastatic lesion discovered on imag- Potential mechanisms responsible for


ing is most likely to be secondary to a lung or breast intratumoral hemorrhage include neovascularity/
primary malignancy, given the much higher inci- angiogenesis, rapid tumor growth and necrosis,
dence and prevalence of these tumors. and vascular invasion. Several factors have been
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 281

Fig. 38 Series of MR images in a patient with right frontal demonstrates the hematoma to be predominantly
intraparenchymal hemorrhage related to superior sagittal hyperintense with internal areas of hypointensity, indicat-
sinus thrombosis. (a) Sagittal unenhanced T1-WI near the ing different stages of hematoma evolution. Correlating
midline demonstrates hyperintense methemoglobin throm- with the T1 signal, the hematoma is likely in the acute
bus within the superior sagittal sinus. (b) Sagittal T1-WI stage. (e) Axial T2* GRE image demonstrates blooming of
right parasagittal near the midline demonstrates slightly the hypointense components of the hematoma due to sus-
hyperintense hemorrhage in the right frontal lobe. (c) Sag- ceptibility effects. (f) Axial DWI image demonstrates
ittal PC MRV demonstrates lack of flow signal within the hyperintense signal at the periphery and hypointense signal
majority of the superior sagittal sinus (arrows) with incom- in the center of the hematoma
plete thrombosis of the posterior aspect. (d) Axial T2-WI

suggested which help to favor a malignant etiol- favor hemorrhagic malignant neoplasm over a
ogy for intracranial hemorrhage rather than a benign entity.
benign cause, including locations atypical for Metastatic melanoma can present a diagnostic
other common benign etiologies of intracranial dilemma. Melanin can cause intrinsic T1
hemorrhage (e.g., hypertensive hemorrhage), hyperintensity, as does methemoglobin. Meta-
multiplicity, and early lesion enhancement. Addi- static melanoma can present as multiple areas
tionally, delay and/or distortion of the expected of hemorrhage within the metastasis and/or
orderly evolution of hematoma evolution on as recurrent hemorrhage from metastatic mela-
MR can be suggestive of underlying malignant noma leading to superficial siderosis (hemosid-
neoplasm, potentially on the basis of intratumoral erin staining of the subarachnoid space and
hypoxia [43]. Prominent perilesional edema pial surface of the brain). As such, this entity
which persists on follow-up imaging and lack may mimic severe hypertensive arteriopathy
of development of a complete hemosiderin rim and CAA.
282 S. Quinet and P. Turski

Fig. 39 Transverse sinus and vein of Labbe thrombosis. (a) (arrow). (d) Axial T2* GRE image at a slightly more
Axial T2-WI demonstrates hypointense lesion with sur- cephalad location shows similar findings as C. Vein of
rounding hyperintense signal involving the cortex and sub- Labbe thrombosis is again seen (arrow). (e) Axial post-
cortical white matter of the posterior left temporal lobe. (b) contrast T1-WI demonstrates a filling defect within the left
Axial post-contrast T1-WI demonstrates heterogeneous but transverse sinus (arrow). (f) Axial T2* GRE image demon-
predominantly hypointense signal within the affected left strates blooming of thrombus within the left transverse sinus
temporal lobe associated with abnormal leptomeningeal (arrow). (g) Coronal MRA image shows lack of enhance-
enhancement overlying the involved cortex. (c) Axial T2* ment of the left transverse sinus. (h) Coronal MRA image a
GRE image shows “blooming” of hypointense blood prod- slightly more posterior location demonstrates enhancement
ucts within the hematoma. Note the linear hypointense of the dura of the left transverse sinus surrounding a
signal and blooming at the lateral margin of the left temporal non-enhancing filling defect representing the thrombus
lobe representing thrombosis within the vein of Labbe (arrow), the so-called empty delta sign
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 283

Fig. 40 Grade IVastrocytoma. Axial post-contrast T1 (a), right temporal lobe. Extensive enhancing leptomeningeal
T2* (b), and SW (c) images demonstrate a metastatic disease is present within the Sylvian fissure and
microhemorrhage in the subcortical right frontal lobe studding the pial surfaces of the right frontal and temporal
(white arrow) and macrohemorrhage (black arrow) in the lobes

delayed radiation injury and are thought to repre-


Radiation-Induced Vascular Disease sent a proliferative vasculopathy involving small
and Hemorrhage vessels, arising on the basis of radiation injury to
the microvasculature [45]. These lesions are
Telangiectasias and cavernomas have been thought to arise along a different pathophysio-
described with increased frequency in patients logic pathway than large-vessel radiation-induced
with history of prior CNS irradiation, particularly vasculopathy, which is characterized by acceler-
in childhood, relative to the general population. ated atherosclerosis on the basis of hyalinization
Radiation-induced telangiectasias have been and fibrinoid necrosis of the vessel wall, intimal
shown to occur in at least 20 % of children receiv- proliferation, and thrombosis. Telangiectasias
ing childhood radiation, with a suggestion of have a much shorter latency period, on average
apparent increased frequency of telangiectasias arising under a year after radiation. Conversely,
occurring in patients receiving higher radiation cavernomas may arise as late as 26 years follow-
doses [44]. The increased frequency in the setting ing radiation. It has been speculated that given this
of childhood CNS radiation is thought to reflect discrepancy in latency periods, cavernomas may
the established increased radiation sensitivity of progress from telangiectasias. A shorter latency
the pediatric brain, with possibly a contribution period for development of cavernomas has been
from increased expression of angiogenic factors described with childhood radiation doses in
(e.g., vascular endothelial growth factor) in the excess of 30 Gy.
pediatric population. Pathologically, telangiecta- On MR imaging, radiation-induced telangiec-
sias represent dilated capillaries with thin walls tasias most often appear as homogeneous small
and intervening normal brain parenchyma. (often <5–10 mm) areas of T2 hypointensity with
Attempted collateralization in the setting of “blooming” on T2* GRE and SWI sequences
small venous thrombosis has been suggested as a (Fig. 44). Occasionally, telangiectasias can be het-
potential etiology of radiation-induced telangiec- erogeneous in signal with at T2 hypointense rim.
tasia development. Both telangiectasias and These lesions may demonstrate stippled or punc-
cavernomas can be categorized as a subtype of tate enhancement. The appearance of multiple
284 S. Quinet and P. Turski

Fig. 41 Grade IV astrocytoma. Initial unenhanced head soft tissue mass. (d) SWI MinIP image demonstrates
CT (a) demonstrates a large acute hemorrhage involving susceptibility-related signal loss throughout the hemor-
the right cerebral hemisphere. Post-contrast T1 image (b) rhagic mass. Vasogenic edema surrounds the mass. Diffu-
shows heterogeneous enhancement of an underlying mass. sion tensor imaging/tractography (e) and functional MRI
(c) T2 image demonstrates blood products in varying states (f) were subsequently performed for pretreatment planning
of degradation within the hematoma associated with the
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 285

Fig. 42 Grade IV astrocytoma. Unenhanced head CT (a) complex, with hemorrhagic and necrotic components with
demonstrates a large intraparenchymal hemorrhage cen- enhancement of the solid components. Contralateral exten-
tered within the right frontal lobe and surrounding sion via the corpus callosum is best appreciated on the
vasogenic edema causing marked mass effect. Follow-up coronal image. Increased cerebral blood volume within
MR imaging with T2 (b), T2* (c), perfusion (d), and the solid components of the mass is identified on perfusion
coronal reformatted post-contrast T1 sequences show the imaging
underlying mass in better detail. The mass is shown to be

radiation-induced telangiectasias can mimic imaging appearance as well as a typical relative


CAA. The lesions are most often asymptomatic, lack of surrounding edema.
unless associated with hemorrhage, which usually
occur in the setting of a cavernoma.
The appearance of radiation-induced Vasculitis
cavernomas is no different than those arising on
a congenital or genetic basis and is described Central nervous system vasculitis is a rare
earlier in this chapter [46]. The appearance of a cause of spontaneous intracranial hemorrhage,
cavernoma can often be distinguished from neo- which can be intraparenchymal or subarachnoid
plastic entities based on their characteristic in location [47]. In the absence of history or
286 S. Quinet and P. Turski

Fig. 43 Metastatic melanoma. Axial T1 images (a–b), and/or methemoglobin. T2* images demonstrate blooming
post-contrast T1 (c–d), and axial T2* (e–f) MR images of hypointense signal confirming the presence of methe-
demonstrate multifocal subcortical metastatic lesions in a moglobin. The presence of enhancement confirms the met-
patient with melanoma. Several of the lesions demonstrate astatic nature of these microhemorrhages
T1 hyperintense signal which may represent melanin

strong clinical suspicion for the diagnosis, (e.g., granulomatosis with polyangiitis), connec-
vasculitis can be an extremely difficult diagnosis tive tissue diseases (e.g., systemic lupus
to make prospectively in a patient presenting erythematosus), angioinvasive infection, and illicit
with intracranial hemorrhage. Vasculitis is drugs (most commonly cocaine, amphetamines,
defined as inflammation of the vessel wall. and other sympathomimetics). On imaging, CTA
Inflammatory changes can lead to irregularity or MRA may show nonspecific irregular beading
and compromise of the vessel lumen, which of the arterial structures (Figs. 45 and 46). Impor-
may ultimately manifest as ischemia, infarct, tantly, small vessel disease is below current imag-
and/or hemorrhage. ing resolution of routine MRI or MRA imaging,
CNS vasculitis is considered primary if there is and a negative examination does not exclude the
no underlying cause identified (i.e., primary diagnosis. DSA remains the most sensitive imag-
angiitis of the CNS). Secondary CNS vasculitis ing modality to suggest the diagnosis, but is simi-
is more common and due to systemic vasculitides larly nonspecific. DSA classically demonstrates
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 287

Fig. 44 Radiation-induced microangiopathy and telangi- susceptibility-related signal loss representing multiple pre-
ectasias and/or cavernomas. Axial unenhanced head CT at sumed capillary telangiectasias and/or cavernomas within
the time of presentation (a) shows a large partially calcified the radiation treatment bed. Axial high B-value DWI (e) and
mass centered in the left hemisphere with associated marked ADC map (f) images demonstrate small foci of restricted
mass effect, resected shortly thereafter and shown to be diffusion within the cortex of the left frontal lobe.
anaplastic ependymoma. (b–f) Series of MRI images Reconstructed maximum intensity projection image from
obtained approximately 2 years after resection and focused three-dimensional phase-contrast MRA (g) demonstrates
radiation in the setting of new right-hand weakness and attenuation of flow signal throughout the M1 segment and
dysarthria. Axial T2/FLAIR image (b) demonstrates distal branches of the left MCA. Digital subtraction angiog-
hyperintense signal in the subcortical white matter of the raphy images obtained shortly after this MR in coronal
left frontal lobe and periventricular region, compatible with projection of right (h) and left (i) common carotid injections
postradiation change. Axial T2 (c) and SWI (d) images confirm the findings on the PCA images. The right intracra-
demonstrate multiple foci of hypointensity and nial vasculature is normal and shown for comparison
288 S. Quinet and P. Turski

Fig. 45 Series of MR images in a patient with CNS imaging demonstrates prolonged transit times to the area
vasculitis. (a) Coronal T2/FLAIR image demonstrates of infarct (black solid arrow). Series of MRA images in a
hyperintense signal along the sulci of the right frontal patient with CNS vasculitis. (d–f) Coronal MIP images
lobe (white solid arrow) consistent with subarachnoid from TOF MRA demonstrate diffuse thinning and irregu-
hemorrhage. (b) DWI demonstrates an area of diffusion larity of the proximal arterial vasculature, most notably
restriction within the right parietal lobe (white open within the M1 segments of the MCS
arrow), representing infarction. (c) Perfusion-weighted

multifocal areas of small- and middle-sized arterial Other less common vascular disorders that
stenosis and dilations. Severe, diffuse atheroscle- uncommonly present with intracranial hemor-
rotic vascular disease can mimic vasculitis on CTA, rhage include moyamoya disease and syndrome,
MRA, and DSA imaging. Leptomeningeal biopsy acute hypertensive encephalopathy/posterior
may ultimately be required to arrive at the diagno- reversible encephalopathy syndrome (PRES),
sis in the absence of an established diagnosis of a and reversible cerebral vasoconstriction syn-
systemic disorder or suggestive clinical history. drome (RCVS).
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 289

Fig. 46 Series of CT and MR images in a patient with right thalamic hematoma on the basis of blood products of
vasculitis secondary to systemic lupus erythematosus differing ages. (e) Axial post-contrast T1-WI demonstrates
(SLE). (a) Axial unenhanced CT image demonstrates peripheral enhancement of the right thalamic hematoma.
right frontal subarachnoid hemorrhage (arrow). (b) Axial Care must be taken to distinguish this lesion from a mass
unenhanced CT image at a more inferior location demon- lesion or abscess. (f–h) Series of CTA images from the
strates right thalamic hematoma and intraventricular exten- same patient demonstrates diffuse irregularity of the intra-
sion of hemorrhage (open arrow) and a left parietal infarct cerebral arterial vasculature. As in the previous case, find-
(arrow). (c) Axial T2* GRE image demonstrates blooming ings are particularly evident in the M1 segments of the
of hypointense right thalamic hemorrhage. (d) T2-WI at a bilateral MCA
similar location demonstrates mixed intensity within the
290 S. Quinet and P. Turski

Summary 7. Gomori JM, Grossman RI (1988) Mechanisms respon-


sible for the MR appearance and evolution of intracra-
nial hemorrhage. Radiographics 8(3):427–440.
Unenhanced CT and dynamic CTA and post- doi:10.1148/radiographics.8.3.3380989
contrast CT are the primary tools for evaluating 8. Barkovich AJ, Atlas SW (1988) Magnetic resonance
intracranial hemorrhage. It is important to evaluate imaging of intracranial hemorrhage. Radiol Clin North
the hemorrhage based on location (e.g., cortical, Am 26(4):801–820
9. Silvera S, Oppenheim C, Touzé E, Ducreux D, Page P,
basal ganglia, SAH, IVH). Additionally, evidence Domigo V, Meder J-F (2005) Spontaneous intracere-
of active bleeding (e.g., spot sign), an underlying bral hematoma on diffusion-weighted images: influ-
lesion (e.g., vascular malformation, neoplasm), the ence of T2-shine-through and T2-blackout effects.
size of the hematoma, and associated mass effect Am J Neuroradiol 26(2):236–241
10. Viswanathan A, Greenberg SM (2011) Cerebral amy-
(e.g., subfalcine or transtentorial herniation, acute loid angiopathy in the elderly. Ann Neurol 70
hydrocephalus, infarct) require close attention. MR (6):871–880. doi:10.1002/ana.22516
imaging is well suited to age the hemorrhage, 11. Chao CP, Kotsenas AL, Broderick DF (2006) Cerebral
though evaluation can be complicated when asso- amyloid angiopathy: CT and MR imaging findings.
Radiographics 26(5):1517–1531. doi:10.1148/
ciated with an underlying lesion and/or recurrent rg.265055090
hemorrhage occurs. Advanced vascular imaging 12. Haacke EM, DelProposto ZS, Chaturvedi S, Sehgal V,
techniques, with both CT and MRI, are helpful in Tenzer M, Neelavalli J, Kido D (2007) Imaging cere-
prognostication and risk stratification of subse- bral amyloid angiopathy with susceptibility-weighted
imaging. Am J Neuroradiol 28(2):316–317
quent hemorrhage in a subset of entities causing 13. Wang J, Gong X (2011) Superficial siderosis of
nontraumatic intracerebral hemorrhage. the central nervous system: MR findings with
susceptibility-weighted imaging. Clin Imaging 35
(3):217–221. doi:10.1016/j.clinimag.2010.06.003
14. Schrag M, McAuley G, Pomakian J, Jiffry A, Tung S,
References Mueller C, Kirsch WM (2010) Correlation of
hypointensities in susceptibility-weighted images to
1. Khosravani H, Mayer SA, Demchuk A, Jahromi BS, tissue histology in dementia patients with cerebral
Gladstone DJ, Flaherty M, Aviv RI (2013) Emergency amyloid angiopathy: a postmortem MRI study. Acta
noninvasive angiography for acute intracerebral hem- Neuropathol 119(3):291–302. doi:10.1007/s00401-
orrhage. Am J Neuroradiol 34(8):1481–1487. 009-0615-z
doi:10.3174/ajnr.A3296 15. Chavhan GB, Babyn PS, Thomas B, Shroff MM,
2. Broderick JP, Adams HP, Barsan W, Feinberg W, Haacke EM (2009) Principles, techniques, and appli-
Feldmann E, Grotta J, Zuccarello M (1999) Guidelines cations of T2*-based MR imaging and its special appli-
for the management of spontaneous intracerebral hem- cations. Radiographics 29(5):1433–1449. doi:10.1148/
orrhage: a statement for healthcare professionals from a rg.295095034
special writing group of the Stroke Council, American 16. Goos JDC, van der Flier WM, Knol DL, Pouwels PJW,
Heart Association. Stroke 30(4):905–915. Scheltens P, Barkhof F, Wattjes MP (2011) Clinical
doi:10.1161/01.STR.30.4.905 relevance of improved microbleed detection by
3. Fischbein NJ, Wijman CAC (2010) Nontraumatic susceptibility-weighted magnetic resonance imaging.
intracranial hemorrhage. Neuroimaging Clin N Am Stroke 42(7):1894–1900. doi:10.1161/
20(4):469–492. doi:10.1016/j.nic.2010.07.003 STROKEAHA.110.599837
4. Wada R, Aviv RI, Fox AJ, Sahlas DJ, Gladstone DJ, 17. Haacke EM, Mittal S, Wu Z, Neelavalli J, Cheng Y-CN
Tomlinson G, Symons SP (2007) CT angiography (2008) Susceptibility-weighted imaging: technical
“spot sign” predicts hematoma expansion in acute aspects and clinical applications, part 1. Am J
intracerebral hemorrhage. Stroke 38(4):1257–1262. Neuroradiol 30(1):19–30. doi:10.3174/ajnr.A1400
doi:10.1161/01.STR.0000259633.59404.f3 18. Mittal S, Wu Z, Neelavalli J, Haacke EM (2008)
5. Rønning P, Sorteberg W, Nakstad P, Russell D, Helseth Susceptibility-weighted imaging: technical aspects
E (2008) Aspects of intracerebral hematomas – an and clinical applications, part 2. Am J Neuroradiol 30
update. Acta Neurol Scand 118(6):347–361. (2):232–252. doi:10.3174/ajnr.A1461
doi:10.1111/j.1600-0404.2008.01023.x 19. Barnes SRS, Haacke EM (2009) Susceptibility-
6. Huisman TAGM (2005) Intracranial hemorrhage: weighted imaging: clinical angiographic applications.
ultrasound, CT and MRI findings. Eur Radiol 15 Magn Reson Imaging Clin N Am 17(1):47–61.
(3):434–440. doi:10.1007/s00330-004-2615-7 doi:10.1016/j.mric.2008.12.002
13 Imaging of Spontaneous Nontraumatic Intracerebral Hemorrhage 291

20. Schweser F, Deistung A, Lehr BW, Reichenbach JR Multidisciplinary consensus on assessment of


(2010) Differentiation between diamagnetic and para- unruptured intracranial aneurysms: proposal of an inter-
magnetic cerebral lesions based on magnetic suscepti- national research group. Stroke 45(5):1523–1530.
bility mapping. Med Phys 37(10):5165. doi:10.1118/ doi:10.1161/STROKEAHA.114.004519
1.3481505 30. Mullan S, Mojtahedi S, Johnson DL, Macdonald RL
21. Noguchi K, Seto H, Kamisaki Y, Tomizawa G, (1996) Embryological basis of some aspects of cerebral
Toyoshima S, Watanabe N (2000) Comparison of vascular fistulas and malformations. J Neurosurg
fluid-attenuated inversion-recovery MR imaging with 85(1):1–8
CT in a simulated model of acute subarachnoid hem- 31. Gandhi D, Chen J, Pearl M, Huang J, Gemmete JJ,
orrhage. Am J Neuroradiol 21(5):923–927 Kathuria S (2012) Intracranial dural arteriovenous fis-
22. Papke K, Kuhl CK, Fruth M, Haupt C, Schlunz- tulas: classification, imaging findings, and treatment.
Hendann M, Sauner D, Brassel F (2007) Intracranial Am J Neuroradiol 33(6):1007–1013. doi:10.3174/ajnr.
aneurysms: role of multidetector CT angiography in A2798
diagnosis and endovascular therapy planning. 32. Sliker CW (2008) Blunt cerebrovascular injuries:
Radiology 244(2):532–540. doi:10.1148/radiol. imaging with multidetector CT angiography.
2442060394 Radiographics 28(6):1689–1708. doi:10.1148/
23. McKinney AM, Palmer CS, Truwit CL, Karagulle A, rg.286085521
Teksam M (2008) Detection of aneurysms by 33. Wilson CB (1992) Cryptic vascular malformations.
64-section multidetector CT angiography in patients Clin Neurosurg 38:49–84
acutely suspected of having an intracranial aneurysm 34. De Souza JM, Domingues RC, Cruz LCH, Domingues
and comparison with digital subtraction and 3D rota- FS, Iasbeck T, Gasparetto EL (2008) Susceptibility-
tional angiography. Am J Neuroradiol 29(3):594–602. weighted imaging for the evaluation of patients with
doi:10.3174/ajnr.A0848 familial cerebral cavernous malformations: a compari-
24. Yoon DY, Lim KJ, Choi CS, Cho BM, Oh SM, Chang son with T2-weighted fast spin-echo and gradient-echo
SK (2007) Detection and characterization of intracra- sequences. Am J Neuroradiol 29(1):154–158.
nial aneurysms with 16-channel multidetector row CT doi:10.3174/ajnr.A0748
angiography: a prospective comparison of volume- 35. Perrini P, Lanzino G (2006) The association of
rendered images and digital subtraction angiography. venous developmental anomalies and cavernous
Am J Neuroradiol 28(1):60–67 malformations: pathophysiological, diagnostic,
25. Villablanca JP, Duckwiler GR, Jahan R, Tateshima S, and surgical considerations. Neurosurg Focus 21(1):e5
Martin NA, Frazee J, Vinuela FV (2013) Natural history 36. Takasugi M, Fujii S, Shinohara Y, Kaminou T,
of asymptomatic unruptured cerebral aneurysms evalu- Watanabe T, Ogawa T (2013) Parenchymal
ated at CT angiography: growth and rupture incidence hypointense foci associated with developmental
and correlation with epidemiologic risk factors. Radiol- venous anomalies: evaluation by phase-sensitive MR
ogy 269(1):258–265. doi:10.1148/radiol.13121188 imaging at 3T. Am J Neuroradiol 34(10):1940–1944.
26. Wermer MJH, van der Schaaf IC, Algra A, Rinkel GJE doi:10.3174/ajnr.A3495
(2007) Risk of rupture of unruptured intracranial 37. Smith SD, Eskey CJ (2011) Hemorrhagic stroke.
aneurysms in relation to patient and aneurysm charac- Radiol Clin North Am 49(1):27–45. doi:10.1016/j.
teristics: an updated meta-analysis. Stroke 38 rcl.2010.07.011
(4):1404–1410. doi:10.1161/01.STR.0000260955. 38. Hom J, Dankbaar JW, Soares BP, Schneider T, Cheng
51401.cd S-C, Bredno J, Wintermark M (2010) Blood–brain
27. Wiebers DO, Whisnant JP, Huston J 3rd, Meissner I, barrier permeability assessed by perfusion CT predicts
Brown RD Jr, Piepgras DG, International Study of symptomatic hemorrhagic transformation and malig-
Unruptured Intracranial Aneurysms Investigators nant edema in acute ischemic stroke. Am J
(2003) Unruptured intracranial aneurysms: Neuroradiol. doi:10.3174/ajnr.A2244
natural history, clinical outcome, and risks of surgical 39. Jain AR, Jain M, Kanthala AR, Damania D, Stead LG,
and endovascular treatment. Lancet 362 Wang HZ, Jahromi BS (2013) Association of CT per-
(9378):103–110 fusion parameters with hemorrhagic transformation in
28. Wiebers DO, Piepgras DG, Meyer FB, Kallmes DF, acute ischemic stroke. Am J Neuroradiol 34
Meissner I, Atkinson JL, Brown RD Jr (2004) Patho- (10):1895–1900. doi:10.3174/ajnr.A3502
genesis, natural history, and treatment of unruptured 40. Warach S (2004) Evidence of reperfusion injury,
intracranial aneurysms. In: Mayo clinic proceedings, exacerbated by thrombolytic therapy, in human focal
vol 79. Elsevier, pp 1572–1583. Retrieved from http:// brain ischemia using a novel imaging marker of early
www.sciencedirect.com/science/article/pii/S00256196 blood–brain barrier disruption. Stroke 35(11_Suppl_1):
11618685 2659–2661. doi:10.1161/01.STR.0000144051.32131.09
29. Etminan N, Beseoglu K, Barrow DL, Bederson J, 41. Hacke W, Kaste M, Bluhmki E, Brozman M,
Brown RD, Connolly ES, Macdonald RL (2014) Dávalos A, Guidetti D, Machnig T (2008)
292 S. Quinet and P. Turski

Thrombolysis with alteplase 3 to 4.5 hours after acute and dose relation. Radiology 230(1):93–99.
ischemic stroke. N Engl J Med 359(13):1317–1329 doi:10.1148/radiol.2301021143
42. Ozsvath RR, Casey SO, Lustrin ES, Alberico RA, 45. Rabin BM, Meyer JR, Berlin JW, Marymount MH,
Hassankhani A, Patel M (1997) Cerebral venography: Palka PS, Russell EJ (1996) Radiation-induced
comparison of CT and MR projection venography. AJR changes in the central nervous system and head and
Am J Roentgenol 169(6):1699–1707 neck. Radiographics 16(5):1055–1072. doi:10.1148/
43. Atlas SW, Grossman RI, Gomori JM, Hackney DB, radiographics.16.5.8888390
Goldberg HI, Zimmerman RA, Bilaniuk LT (1987) 46. Jain R, Robertson PL, Gandhi D, Gujar SK, Muraszko
Hemorrhagic intracranial malignant neoplasms: spin- KM, Gebarski S (2005) Radiation-induced cavernomas
echo MR imaging. Radiology 164(1):71–77. of the brain. Am J Neuroradiol 26(5):1158–1162
doi:10.1148/radiology.164.1.3588929 47. Blitstein MK, Tung GA (2007) MRI of cerebral
44. Koike S, Aida N, Hata M, Fujita K, Ozawa Y, Inoue T microhemorrhages. Am J Roentgenol 189
(2004) Asymptomatic radiation-induced telangiectasia (3):720–725. doi:10.2214/AJR.07.2249
in children after cranial irradiation: frequency, latency,
ICA–ECA Collaterals
14
Maxim Mokin and Adnan H. Siddiqui

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294 Knowledge and identification of multiple anas-
tomotic connections between the external
Ophthalmic Artery Anastomoses . . . . . . . . . . . . . . . . . . 294
carotid artery and the internal carotid artery
Internal Carotid Artery Anastomoses . . . . . . . . . . . . 295 branches are paramount for the diagnosis and
Posterior Circulation Anastomoses . . . . . . . . . . . . . . . 298 treatment of vascular diseases of the brain,
head, neck, and spine. In this chapter, common
Noninvasive Imaging Modalities . . . . . . . . . . . . . . . . . . 299
extracranial–intracranial anastomoses of the
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301 ophthalmic artery, internal carotid artery, and
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301 of the posterior circulation that are critical for
the proceduralist to recognize in order to avoid
complications during embolization procedures
or open surgery are reviewed. Finally, the role
M. Mokin of noninvasive imaging in the evaluation of the
Department of Neurosurgery, School of Medicine and
collateral circulation and cerebrovascular
Biomedical Sciences, University at Buffalo, State
University of New York, Buffalo, NY, USA reserve in clinical conditions associated with
arterial occlusive disease is discussed.
Department of Neurosurgery, Gates Vascular Institute/
Kaleida Health, Buffalo, NY, USA
e-mail: maximmokin@gmail.com Keywords
A.H. Siddiqui (*) External carotid artery • Internal carotid artery
Department of Neurosurgery, School of Medicine and • Anastomosis • Stroke • Collaterals •
Biomedical Sciences, University at Buffalo, State Angiography
University of New York, Buffalo, NY, USA
Department of Neurosurgery, Gates Vascular Institute/ List of Abbreviations
Kaleida Health, Buffalo, NY, USA
AP Anteroposterior
Department of Radiology, School of Medicine and CRA Central retinal artery
Biomedical Sciences, University at Buffalo, State
ECA External carotid artery
University of New York, Buffalo, NY, USA
ICA Internal carotid artery
Toshiba Stroke and Vascular Research Center, University
MMA Middle meningeal artery
at Buffalo, State University of New York, Buffalo, NY,
USA OA Ophthalmic artery
Jacobs Institute, Buffalo, NY, USA
e-mail: asiddiqui@ubns.com; adnan.h.siddiqui@gmail.
com

# Springer Science+Business Media New York 2016 293


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_1
294 M. Mokin and A.H. Siddiqui

Introduction critical to avoid occlusion of the CRA, which


will result in monocular blindness. The CRA is
Knowledge and identification of multiple anasto- the first branch of the OA, when the OA crosses
motic connections between the external carotid above the optic nerve. When the OA crosses
artery (ECA) and the internal carotid artery below the optic nerve, the lateral posterior ciliary
(ICA) branches are paramount for the diagnosis artery originates from the OA first and the CRA
and treatment of vascular diseases of the brain, second. In cases in which the CRA cannot be
head, neck, and spine. The expanding role of reliably identified on diagnostic angiography
minimally invasive approaches for the treatment and embolization is planned, provocative testing
of cerebrovascular diseases, including arteriove- with superselective injection of sodium amobar-
nous malformations, fistulas, moyamoya disease, bital and lidocaine should be considered to iden-
and other conditions, such as epistaxis and head tify whether potential anastomoses to the eye
and neck tumors, requires careful exploration of exist at or distal to the site of microcatheter
the arterial supply to these structures to ensure placement.
success from the intervention. In this chapter, Visualization of a choroidal blush (best appre-
common extracranial–intracranial anastomoses ciated during angiography on lateral views as a
that are critical for the proceduralist to recognize thin crescent of contrast material) on ECA injec-
in order to avoid complications during emboliza- tions should raise suspicion for potential anasto-
tion procedures or open surgery are reviewed. The moses with the OA [6]. However, it should be
role of noninvasive imaging in the evaluation of noted that absence of the choroidal blush does
the collateral circulation and cerebrovascular not exclude the existence of such anastomoses.
reserve in clinical conditions associated with arte- For example, when embolization is executed in
rial occlusive disease is discussed. the territory known to have potential connections
The development of the cranial vascular sup- with the OA, repeat diagnostic angiographic runs
ply during embryonic stages and a comprehensive should be performed, as dangerous anastomoses
review of all potential extracranial–intracranial might become visible only following occlusion of
anastomoses are beyond the scope of this chapter. ECA branches with embolic agents. Identification
Readers are referred to the classic work by of OA–MMA anastomoses can also help avoid
Lasjaunias et al. [1] for a more detailed descrip- visual complications in cranial base surgery, such
tion of vascular anatomy on this subject. as during resection of meningiomas [7].
An isolated OA origin of the MMA is a rare
anatomic variant, especially when present bilater-
Ophthalmic Artery Anastomoses ally [8, 9]. In such cases, the entire MMA or some
of its branches are not visualized on a selective
The ophthalmic artery (OA) most commonly ECA injection. An ICA injection will opacify the
arises from the paraclinoid segment of the ICA MMA in a slightly delayed fashion, via the prox-
in adults. Variations in the OA origin can be seen imal OA through the superior orbital fissure
on angiography, such as from the cavernous por- (Fig. 1). Embolization of a lesion supplied by
tion of the ICA (the so-called “dorsal OA”), from such anomalous meningeal vessels is challenging
the middle meningeal artery (MMA), or from the because of the high risk for emboli into OA
middle cerebral artery (MCA) [2–4]. The order branches, causing retinal ischemia [10].
and appearance of the OA branches depend on Multiple anastomoses between branches of the
the course of this artery in relationship to the ECA and OA exist, and only the most clinically
optic nerve [5]. The OA branches are commonly relevant variants will be described here. For a
divided into three groups: ocular, orbital, and more detailed review, the articles by Hayreh [11]
extraorbital. The retina and choroid are supplied and Perrini et al. [5] are recommended. The recur-
by the central retinal artery (CRA) and the ciliary rent meningeal artery is a critical anastomosis
arteries. Their visualization on angiography is when attempting embolization of convexity
14 ICA–ECA Collaterals 295

Fig. 1 Ophthalmic origin of the middle meningeal artery. arrow) through the recurrent meningeal artery (black
Cerebral angiogram, lateral projection, early (a) and late arrowhead). (c) Notice that the middle meningeal artery
arterial phase (b) of selective left internal carotid artery does not opacify on the selective external carotid artery
injection demonstrate reconstitution of the middle menin- injection. sta superficial temporal artery, ima internal max-
geal artery (white arrows) via the ophthalmic artery (black illary artery

meningiomas. It typically projects posteriorly to anastomoses have been described between the
the OA through the superior orbital fissure. MMA, superficial temporal artery, occipital artery,
Another important group of anastomoses is and the cortical leptomeningeal arteries [13]
between the anterior and posterior ethmoidal (Fig. 4).
branches of the OA and the distal branches of
the internal maxillary artery, namely, the
sphenopalatine and greater palatine arteries Internal Carotid Artery Anastomoses
(Fig. 2). The presence of such anastomoses should
be explored on cerebral angiography when plan- Important anastomoses within the petrocavernous
ning embolization for epistaxis [12]. segment of the ICA are described below. They can
Additional examples of potential anastomoses often be identified on cerebral angiograms in
involving the OA include the superficial temporal cases of cervical ICA occlusion and are a critical
artery branches such as the zygomatico-orbital component of dural arteriovenous malformations
artery, terminal nasal branches of the facial artery, in this region.
and the infraorbital branch of the internal The inferolateral trunk is a remnant of the
maxillary artery (Fig. 3). Extracranial–intracranial dorsal OA and consists of several branches that
296 M. Mokin and A.H. Siddiqui

Fig. 2 Ophthalmic artery–internal maxillary artery anas- branches (black arrows point to the nasal blush at the site
tomoses. Cerebral angiogram in a patient with chronic of anastomoses within the nasal cavity). White arrowhead
right internal carotid artery occlusion (asterisk on – infraorbital artery of the internal maxillary artery. Black
anteroposterior [AP] view), AP (a) and lateral (b) views, arrowhead – cavernous internal carotid artery; ima internal
shows reconstitution of the ophthalmic artery (white maxillary artery
arrows) through the anterior and posterior ethmoidal

Fig. 3 Superficial temporal artery anastomoses to the (black arrowheads) via the zygomatico-orbital branch
ophthalmic artery. Cerebral angiogram, AP (a) and lateral (white arrow) of the superficial temporal artery. Another
(b) views, shows opacification of the ophthalmic artery anastomosis involves the infraorbital branch (white arrow-
(black arrows) and the supraclinoid internal carotid artery heads) of the internal maxillary artery (ima)
14 ICA–ECA Collaterals 297

Fig. 4 Cortical
leptomeningeal
anastomoses. Cerebral
angiogram, early arterial
intracranial AP (a) and
lateral (b) views and late
arterial intracranial AP (c)
and lateral (d) views, shows
cortical leptomeningeal
anastomoses between the
middle meningeal artery
(black arrows) and the
middle cerebral artery
(white arrows)

provide supply to the cranial nerves and the occlusion of the cervical ICA [14] (Fig. 7). Exten-
gasserian ganglion. Anteroinferiorly, a character- sive proliferation of the vasa vasorum can lead to
istic serpentine-like anastomosis with the maxil- anterograde recanalization of the ICA distally, such
lary artery through the artery of the foramen as at the level of the petrocavernous segment, and
rotundum can be observed (Fig. 5). Other anasto- can be confused with anastomoses originating from
moses include the recurrent artery of foramen the ascending pharyngeal artery. Visualization of
lacerum (which connects with the ascending pha- tortuous vascular channels that project over the
ryngeal artery), branches of the MMA (seen lat- expected course of the cervical ICA rather than
erally through the foramen spinosum), branches the ascending pharyngeal artery helps in differen-
of the accessory meningeal artery (seen posteri- tiating the two conditions. The ascending pharyn-
orly), and branches of the ascending pharyngeal geal artery typically originates from the posterior
artery (carotid branch of the superior pharyngeal wall of the ECA trunk, although rarely its origin
branch through the foramen lacerum) (Fig. 6). can be seen from the proximal cervical ICA.
A rare finding on cerebral angiography is The meningohypophyseal trunk of the ICA
hypertrophy of the vasa vasorum after complete supplies the pituitary gland, the clival region,
298 M. Mokin and A.H. Siddiqui

and the meninges of the tentorium. Through its


tentorial branches, which are seen more anteriorly,
it can form anastomoses with petrous branches of
the MMA. Another common anastomosis of the
meningohypophyseal trunk involves its lateral
clival artery and neuromeningeal branches of the
ascending pharyngeal artery.

Posterior Circulation Anastomoses

Early during embryologic development of the


central nervous system, the posterior circulation
forms primitive anastomoses with the ICAs (the
trigeminal, otic, hypoglossal, and proatlantal con-
nections) that eventually disappear, except for the
posterior communicating arteries, which remain
in adults. The most common anastomosis
involves the occipital artery and the vertebral
Fig. 5 Artery of foramen rotundum. Cerebral angiogram, artery connecting at the C1 or C2 segmental levels
AP view, demonstrates a characteristic serpentine-like artery
(Fig. 8). Other examples of anastomotic connec-
of foramen rotundum (white arrows) directed at the base of
the sella, which is an anastomosis between the internal tions with the vertebral artery include the ascend-
maxillary artery (ima) and the cavernous segment of the ing pharyngeal artery and the ascending cervical
internal carotid artery (black arrow). oa ophthalmic artery and deep cervical arteries.

Fig. 6 Ascending
pharyngeal artery
anastomosis. Cerebral
angiogram, AP (a) and
lateral (b) views, shows a
typical appearance of the
ascending pharyngeal
artery. The superior
pharyngeal branch of the
pharyngeal trunk (white
arrows) gives off the carotid
branch, which creates an
anastomosis with the
inferolateral trunk (not
shown). cca common
carotid artery, fa facial
artery, ima internal
maxillary artery, oa
occipital artery
14 ICA–ECA Collaterals 299

Fig. 7 Vasa vasorum of the


internal carotid artery.
Cerebral angiogram,
cervical AP (a) and lateral
(b) views and cranial AP (c)
and lateral (d) views in a
case of chronic cervical left
internal carotid artery
occlusion. The petrous
segment of the internal
carotid artery (black
arrows) reconstitutes via a
prominent network of vasa
vasorum, which appear as
tortuous channels following
the expected course of the
cervical internal carotid
artery (white arrows)

to estimate the ability of such connections to pro-


Noninvasive Imaging Modalities vide adequate cerebral blood flow in conditions
such as occlusive disease of the ICA and the
Catheter angiography remains the gold standard vertebrobasilar system and moyamoya disease.
for the identification of specific anastomotic con- Measurement of cerebrovascular reserve can
nections between the ECA and ICA branches help predict stroke risk in patients with carotid
because of its excellent spatial resolution and the stenosis or occlusion, based on a meta-analysis
proceduralist’s ability to perform superselective of studies evaluating cerebrovascular reserve
catheterization of individual vessels. Noninvasive impairment [15].
imaging can be used in clinical practice as a way
300 M. Mokin and A.H. Siddiqui

The techniques used most commonly to eval-


uate cerebrovascular reserve include computed
tomographic and magnetic resonance perfusion
imaging, single-photon emission computed
tomography, and positron emission tomography
[16–20] (Fig. 9). Systemic administration of cere-
bral vasodilator agents that can cross the
blood–brain barrier, such as acetazolamide,
allows the calculation of changes in cerebral
blood flow and visualization of reactivity in spe-
cific vascular territories in which vasoocclusive
disease is present [21] (Fig. 10).
Arterial spin-labeling magnetic resonance
imaging is another technique that can assess tem-
poral dynamics of arterial blood inflow and iden-
tify brain regions with impaired hemodynamics
[18, 22, 23]. It can be used to visualize the perfu-
Fig. 8 Occipital artery–vertebral artery anastomosis.
sion territory of the ECA in order to depict the
Cerebral angiogram, lateral view, selective occipital artery
(oa) injection, demonstrates opacification of the status of the collateral circulation and quantita-
vertebrobasilar system (white arrows) via the C1 segmen- tively compare flow pre- and postoperatively [23].
tal artery (black arrow)

Fig. 9 Watershed
perfusion deficit. In a
patient with chronic
occlusive disease of the left
internal carotid artery, there
is a perfusion deficit in the
anterior cerebral artery
(ACA)–middle cerebral
artery watershed territory
(indicated by dotted lines)
14 ICA–ECA Collaterals 301

Fig. 10 Acetazolamide challenge. (a) In a patient with (b) A robust increase in cerebral blood flow within the
chronic right internal carotid artery occlusion, there is right hemisphere is seen after administration of acetazol-
decreased cerebral blood flow in the right internal carotid amide, indicating good cerebrovascular reserve
artery territory, in comparison to the contralateral side.

3. Watanabe A, Hirano K, Ishii R (1996) Dural carotico-


Summary cavernous fistula with both ophthalmic arteries arising
from middle meningeal arteries. Neuroradiology
To ensure success and safety when treating cere- 38:806–808
4. Uchino A, Saito N, Takahashi M, Kozawa E,
brovascular diseases of the head and neck, Mizukoshi W, Nakajima R, Okano N (2013) Persistent
proceduralists should be aware of dangerous dorsal ophthalmic artery and ophthalmic artery arising
extracranial–intracranial carotid arterial anasto- from the middle meningeal artery diagnosed by MR
moses, which are highly variable in their appear- angiography at 3 T. Surg Radiol Anat 35:775–782
5. Perrini P, Cardia A, Fraser K, Lanzino G (2007) A
ance and location. Catheter angiography remains microsurgical study of the anatomy and course of the
the gold standard for the identification of such ophthalmic artery and its possibly dangerous anasto-
anastomotic routes and should be carefully moses. J Neurosurg 106:142–150
reviewed before performing embolization of arte- 6. Schurr PH (1951) Angiography of the normal ophthal-
mic artery and choroidal plexus of the eye. Br J
riovenous malformations, tumors, or epistaxis. Ophthalmol 35:473–478
Noninvasive imaging can estimate the ability of 7. Hayashi N, Kubo M, Tsuboi Y, Nishimura S,
such connections to provide adequate cerebral Nishijima M, Ahmed Abdel-Aal M, Endo S (2007)
blood flow in conditions such as occlusive disease Impact of anomalous origin of the ophthalmic artery
from the middle meningeal artery on selection of sur-
of the ICA and the vertebrobasilar system. gical approach to skull base meningioma. Surg Neurol
68:568–572
8. Plas B, Bonneville F, Dupuy M, Sol JC, Chaynes P
References (2013) Bilateral ophthalmic origin of the middle men-
ingeal artery. Neurochirurgie 59:183–186
9. Royle G, Motson R (1973) An anomalous origin of the
1. Lasjaunias P, Berenstein A, ter Brugge K (2001) Sur- middle meningeal artery. J Neurol Neurosurg Psychia-
gical neuroangiography: 1 clinical vascular anatomy try 36:874–876
and variations. Springer, Berlin 10. Trivelatto F, Nakiri GS, Manisor M, Riva R,
2. Sade B, Tampieri D, Mohr G (2004) Ophthalmic artery Al-Khawaldeh M, Kessler I, Mounayer C (2011) Pre-
originating from basilar artery: a rare variant. AJNR operative onyx embolization of meningiomas fed by
Am J Neuroradiol 25:1730–1731
302 M. Mokin and A.H. Siddiqui

the ophthalmic artery: a case series. AJNR Am J 18. Donahue J, Sumer S, Wintermark M (2014) Assess-
Neuroradiol 32:1762–1766 ment of collateral flow in patients with cerebrovascular
11. Hayreh SS (2006) Orbital vascular anatomy. Eye disorders. J Neuroradiol 41:234–242
(Lond) 20:1130–1144 19. van Laar PJ, van der Grond J, Bremmer JP, Klijn CJ,
12. Willems PW, Farb RI, Agid R (2009) Endovascular Hendrikse J (2008) Assessment of the contribution of
treatment of epistaxis. AJNR Am J Neuroradiol the external carotid artery to brain perfusion in patients
30:1637–1645 with internal carotid artery occlusion. Stroke
13. Handa J, Waga S, Handa H (1971) Dural-cortical arte- 39:3003–3008
rial anastomosis as a collateral channel in carotid occlu- 20. Ozgur HT, Kent Walsh T, Masaryk A, Seeger JF,
sive disease. Clin Radiol 22:302–307 Williams W, Krupinski E, Melgar M, Labadie E
14. Meguro T, Muraoka K, Terada K, Hirotsune N, (2001) Correlation of cerebrovascular reserve as mea-
Nishino S (2011) Recanalisation of the internal carotid sured by acetazolamide-challenged SPECT with angio-
artery via the vasa vasorum after coil occlusion. Br J graphic flow patterns and intra- or extracranial arterial
Radiol 84:e23–e26 stenosis. AJNR Am J Neuroradiol 22:928–936
15. Gupta A, Chazen JL, Hartman M, Delgado D, 21. Vagal AS, Leach JL, Fernandez-Ulloa M, Zuccarello
Anumula N, Shao H, Mazumdar M, Segal AZ, M (2009) The acetazolamide challenge: techniques and
Kamel H, Leifer D, Sanelli PC (2012) Cerebrovascular applications in the evaluation of chronic cerebral ische-
reserve and stroke risk in patients with carotid stenosis mia. AJNR Am J Neuroradiol 30:876–884
or occlusion: a systematic review and meta-analysis. 22. Bokkers RP, van Laar PJ, van de Ven KC, Kapelle LJ,
Stroke 43:2884–2891 Klijn CJ, Hendrikse J (2008) Arterial spin-labeling MR
16. Konstas AA, Goldmakher GV, Lee TY, Lev MH (2009) imaging measurements of timing parameters in patients
Theoretic basis and technical implementations of CT with a carotid artery occlusion. AJNR Am J
perfusion in acute ischemic stroke, part 1: theoretic Neuroradiol 29:1698–1703
basis. AJNR Am J Neuroradiol 30:662–668 23. Dang Y, Wu B, Sun Y, Mo D, Wang X, Zhang J, Fang J
17. van Laar PJ, van der Grond J, Mali WP, Hendrikse J (2012) Quantitative assessment of external carotid
(2006) Magnetic resonance evaluation of the cerebral artery territory supply with modified vessel-encoded
circulation in obstructive arterial disease. Cerebrovasc arterial spin-labeling. AJNR Am J Neuroradiol
Dis 21:297–306 33:1380–1386
Imaging Biomarkers of Angiogenesis
and the Microvascular Environment 15
in Cerebral Tumors

Alan Jackson, Ibrahim Djoukhadar, and David J. Coope

Contents Clinical Applications of Microvascular


Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304 Diagnosing Glioma Mimics . . . . . . . . . . . . . . . . . . . . . . . . . 321
Abnormalities of Tumor Blood Vessels . . . . . . . . . . . 304 Grading Glioma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322
Identifying Transformation in Low-Grade
Antiangiogenic Therapies . . . . . . . . . . . . . . . . . . . . . . . . . . 307 Glioma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322
Distinguishing Recurrent Tumor from Treatment
Imaging the Tumor Vascular
Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322
Microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
Predicting Treatment Response . . . . . . . . . . . . . . . . . . . . . . 323
Imaging Biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323
Biomarkers of Perfusion and the Vascular
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 324
Microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
Dynamic Susceptibility Contrast-Enhanced
MRI (DSCE-MRI) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
Dynamic Relaxivity Contrast-Enhanced
MRI (DRCE-MRI) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 310
Dynamic Contrast-Enhanced Computed
Tomography (DCE-CT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311
Imaging Microvessel Morphology and Hypoxia
with MRI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 312
Vessel Size Imaging (VSI) and Vessel
Architectural Imaging (VAI) . . . . . . . . . . . . . . . . . . . . . . . . . 312
Capillary Heterogeneity Imaging . . . . . . . . . . . . . . . . . . . . 314
Arteriovenous Overlap Imaging (AVOL) . . . . . . . . . . . 315
Imaging Hypoxia Using MRI . . . . . . . . . . . . . . . . . . . . . . . 316
Molecular Imaging of the Tumor Vascular
Microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316
Imaging Hypoxia Using PET . . . . . . . . . . . . . . . . . . . . . . . . 319
Imaging Integrin Expression . . . . . . . . . . . . . . . . . . . . . . . . . 319

A. Jackson (*) • I. Djoukhadar • D.J. Coope


Wolfson Molecular Imaging Centre, University of
Manchester, Withington, Manchester, UK
e-mail: Alan.Jackson@manchester.ac.uk; Ibrahim.
Djoukhadar@Manchester.ac.uk; david.
coope@manchester.ac.uk

# Springer Science+Business Media New York 2016 303


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_18
304 A. Jackson et al.

macromolecules, which can in turn directly affect


Abstract
the behavior of tumor cells. The development of
The structure and organization of blood vessels
therapeutic approaches, which target the angio-
within tumor tissue is very different from that
genic process or the neovasculature, has led to
seen in normal tissues. Tumor blood vessels
enormous research activity in this area over the
show abnormalities in microstructure and hier-
past 20 years. Imaging techniques have been
archical organization, which result from multi-
widely applied to study the process of angiogen-
ple factors including local tumor
esis and the characteristics of the tumor vascular
characteristics, angiogenic drive, and the abil-
microenvironment (TVM). Imaging has unique
ity of the angiogenic process to keep pace with
strengths allowing repeated, noninvasive assess-
tumor growth. Tumor microvasculature is inef-
ment of the whole tumor or multiple tumors in the
ficient compared to that seen in normal tissues,
same patient. In addition, imaging provides spa-
and, particularly in rapidly growing tumors,
tial data allowing the study of regional variations
blood flow is often inadequate to meet the
in the microenvironment within a single tumor.
demands for oxygen and nutrient delivery and
Imaging also allows the development of quantita-
clearance of waste material. Understanding the
tive metrics, which reflect the physiological, phar-
microvascular environment and its variation
macological, or pathological processes occurring
between and within tumors is critical for an
within the tumor. Such quantitative metrics are
understanding of tumor behavior and therapeu-
referred to as imaging biomarkers (IB), and con-
tic response. A wide range of quantitative
siderable research activity has been focused on the
imaging techniques have been developed in
discovery, technical, and biological validation of
an attempt to provide noninvasive, repeatable
IB that allow characterization of the TVM. In this
assays of microvascular characteristics which
chapter, the characteristics of the TVM and the IB
can then be studied in terms of their spatial
available for its characterization and their current
variability and change over time. This chapter
applications are reviewed.
reviews the currently available imaging bio-
markers and their current clinical application.
Abnormalities of Tumor Blood Vessels
Keywords
Angiogenesis • Blood vessels • Permeability • In normal tissues a single layer of endothelial cells
Flow • Dynamic contrast-enhanced imaging • (EC) lines blood vessels forming a barrier limiting
Positron emission tomography • Hypoxia • the passage of macromolecules. EC are
Integrins surrounded by tightly connected pericytes and
both of these enveloped in a basement membrane.
In normal tissues pericytes are heavily involved in
Introduction the suppression of EC proliferation and EC are
quiescent, showing no evidence of division or
The development of new blood vessels (angiogen- sprouting of new vessels [2]. Pericytes produce a
esis) is an essential component of tumor growth number of signaling molecules, which include
and is one of the classic hallmarks of the cancer vascular endothelial growth factor (VEGF),
cell [1]. The development of new blood vessels, angiopoietin-1 (Ang-1), fibroblast growth factors
their maturation, and organization give rise to a (FGFs), NOTCH, and chemokines, which support
distinctive vascular microenvironment that has normal endothelial cell repair. Endothelial cells
been shown to vary significantly between tumor can also respond directly to dissolved oxygen
types and even within individual tumors. The concentration and have receptors for hypoxia-
characteristics of the vascular microenvironment inducible factors such as prolyl-hydroxylase
will place constraints on the delivery of nutrients, domain 2 (PHD2) and hypoxia-inducible factor-
oxygen, and drugs as well as the distribution of 2α (HIF-2α) which support mechanisms to enable
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 305

remodeling of vessels to optimize oxygen delivery. capillary network, which drains into venules and
Within normal tissue blood vessels are arranged in larger veins. These vascular networks vary from
a clear hierarchy, with inflow of blood managed by tissue to tissue but are, in general, highly structured
arterial vessels and branching arterioles feeding a and internally consistent.

Loosening junctions
a Selection of tip cell (VE-Cadherin)
Matrix remodelling (MMPs)
Tip-cell formation
(VEGFR-2, DLL4, JAGGED1
NRP1, integrins, HIF-1α
MT1-MMP, PGC-1α)
Angiogenic factors
(VEGF, VEGF-C, FGFs,
ANG-2, chemokines)
Pericyte detachment
(ANG-2)
Quiescent vessel Flow
Permeability, vasodilation
and extravasation (VEGF)
b Stalk elongation and tip guidance
Lumen formation
(VE-cadherin, CD34, sialomucins, VEGF)

Pericyte recruitment (PDGF-B, ANG-1,


NOTCH, ephrin-B2, FGF)
Tip-cell guidance and adhesion
(semaphorins, ephrins, integrins)

Liberation of angiogenic
factors from ECM
(VEGF, FGFs)

ECM
Stalk elongation Myeloid cell
Flow recruitment (ANG-2, Adjacent vessel
(VEGFR-1, NOTCH, WNT,
SDF-1α, PIGF) sprout
NRARP, PIGF, FGFs, EGFL7)

c Quiescent phalanx resolution


Phalanx cell
Transendothelial lipid transport (VEGF-B)
(PHD2, HIF-2α
Vascular maintenance VE-cadherin, TIE-2)
(VEGF, ANG-1, FGFs, NOTCH)
Barrier formation
(VE-cadherin, ANG-1)

Pericyte maturation
Basement membrane (PDGF-B, PDGFR-β
Flow deposition (TIMPs, PAI-1) ephrin-B2, ANG-1,
NOTCH, TGF-β1)

Fig. 1 (continued)
306 A. Jackson et al.

Tumor vasculature demonstrates a wide range patent lumen in response to a range of signaling
of abnormalities dependent on the tumor type. As molecules. These new vessels fuse with existing
tumors develop, they depend on the development ones to initiate blood flow and, in time, develop a
of new blood vessels to meet increasing demand pericyte coverage. As they mature they may
for nutrient delivery and waste removal. This pro- reform EC tight junctions and basement
cess of neovascularization is referred to as angio- membranes.
genesis [2]. Angiogenesis is controlled by a In general terms, rapidly growing aggressive
number of signaling mechanisms mediated by tumors provide greater angiogenic drive and will
small molecules known as cytokines which may result in increasingly disordered microvascular
be released from tumor cells, EC, hypoxic normal environments. In some, slowly growing, tumors
cells, or inflammatory tissue [3]. In normal tissue such as low-grade meningiomas, the microvas-
the angiogenic process forms a normal part of cular architecture can be remarkably structured
development [2]. In tumors, the presence of hyp- with well-developed hexagonal arteriolar and
oxia and hypoglycemia promotes the release of capillary territories clearly seen. In more aggres-
pro-angiogenic signaling molecules including sive tumors such as glioma, tumor vessels are
VEGF, Ang-2, FGFs, and chemokines into the disorganized, irregular, and tortuous with bizarre
local microenvironment. In normal vessels branching patterns. They do not maintain the
exposed to these pro-angiogenic stimuli, pericytes normal arterial/capillaries/venous hierarchy and
detach from the basement membrane under the all components of the tumor vessel wall are
action of matrix metalloproteinases (MMP) and abnormal. EC tight junctions are absent, render-
the tight junctions between endothelial cells ing tumor vessels leaky to macromolecules and
loosen, increasing the permeability of the endo- often with a propensity to microhemorrhage.
thelial membrane (Fig. 1). These neovascular structures are dynamic, show-
Plasma proteins extravasate into the extravas- ing constant changes in blood vessel develop-
cular space and form an early extracellular matrix, ment and variations in hemodynamics so that
while integrin signaling stimulates migration of blood flow in individual regions may change in
EC onto the matrix. New vessels grow in an volume and direction even over short time
ordered process that involves migration of a single periods. Primary malignant brain neoplasms
EC, known as the tip cell along the new matrix. exhibit the greatest degree of vascular prolifera-
Nearby EC, known as stalk cells, divide to form tion of any tumor type, and angiogenesis is a key
an elongated vascular core that then develops a step in their progression [4].

Fig. 1 The consecutive steps of blood vessel branching are adhere to the extracellular matrix (mediated by integrins) to
shown, with the key molecular players involved denoted in migrate. Stalk cells behind the tip cell proliferate, elongate,
parentheses. (a) After stimulation with angiogenic factors, and form a lumen, and sprouts fuse to establish a perfused
the quiescent vessel dilates and an endothelial tip cell is neovessel. Proliferating stalk cells attract pericytes and
selected (DLL4 and JAGGED1) to ensure branch formation. deposit basement membranes to become stabilized.
Tip-cell formation requires degradation of the basement Recruited myeloid cells such as tumor-associated macro-
membrane, pericyte detachment, and loosening of endothe- phages (TAMs) and TIE-2-expressing monocytes (TEMs)
lial cell junctions. Increased permeability permits extrava- can produce pro-angiogenic factors or proteolytically liber-
sation of plasma proteins (such as fibrinogen and ate angiogenic growth factors from the ECM. (c) After
fibronectin) to deposit a provisional matrix layer, and pro- fusion of neighboring branches, lumen formation allows
teases remodel preexisting interstitial matrix, all enabling perfusion of the neovessel, which resumes quiescence by
cell migration. For simplicity, only the basement membrane promoting a phalanx phenotype, reestablishment of junc-
between endothelial cells and pericytes is depicted, but in tions, deposition of basement membrane, maturation of
reality, both pericytes and endothelial cells are embedded in pericytes, and production of vascular maintenance signals.
this basement membrane. (b) Tip cells navigate in response Other factors promote transendothelial lipid transport (From
to guidance signals (such as semaphorins and ephrins) and Ref. [3] with permission)
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 307

Most tumors have far less blood flow than antiangiogenic agents offer significant survival
would be expected from their vascularity, benefit in combination therapies and research
reflecting the inefficiency of the tumor vascula- into this mechanism of drug action continues,
ture. In addition, leakage of large molecules and albeit with new expectations. The main questions
the lack of normal lymphatic drainage can result which now face imaging researchers in this field
in high interstitial pressures, which further impair are more demanding and include: Can IB predict
regional perfusion. Consequently, regional hyp- therapeutic response to specific agents or combi-
oxemia and necrosis are common, particularly in nation therapies? Can IB guide selection of opti-
aggressive tumors. mal therapeutic combinations? Can IB be used to
optimize the timings of combination therapies?
Can IB identify and/or predict specific mecha-
Antiangiogenic Therapies nisms of resistance and appropriate subsequent
interventions?
The ubiquitous nature of the angiogenic process
led to great excitement in the 1990s with the hope
that therapeutic approaches, which targeted path- Imaging the Tumor Vascular
ological angiogenesis, would provide a “magic Microenvironment
bullet” that could be used to deprive a large
range of tumors of the blood vessels necessary It is clear that the TVM represents one of the most
for development and expansion. The development profoundly important factors both in tumor behav-
of antiangiogenic drugs and of antivascular ior and therapeutic response. It controls the ability
agents, which target abnormal, developing ves- of individual regions of proliferating tumor cells to
sels, led to one of the most extensive areas of obtain sufficient nutrients and oxygen. Failure of
anticancer research in recent decades. Despite vascular development, adequate to the require-
this, antiangiogenics, while showing efficiency ments of the developing tumor, results in regional
as single or combination agents in a number of hypoxia with the consequent effects on local tumor
tumor types, have failed to provide the prolonged and blood vessel growth and a drive toward geno-
tumor suppression of tumor activity commonly typic instability and tumor cell mutation. It can also
observed in animal models. Many mechanisms result in regional necrosis and tumor cell death.
have been identified which imbue tumor cells, Features of the TVM also control the passage of
EC, or other components of the tumor stroma macromolecules and inflammatory cells into the
with resistance to angiogenic inhibition, and tissue and the generation of hematogenous meta-
some agents have proven toxic when combined static deposits. The ability to study and measure
with chemotherapy [5]. Compensatory increase in these biological processes is essential, and many
other pro-angiogenic signaling pathways can sup- can be characterized and quantified in exquisite
port tumor angiogenesis independently from detail on the basis of tissue biopsy or even by
VEGF, and some evidence is beginning to suggest examination of circulating tumor cells and soluble
that antiangiogenic therapy may cause cancer biomarkers. Despite this, imaging techniques
cells to become increasingly malignant in behav- remain the only available approach to studying
ior. Secondary hypoxia can induce mutations the TVM in situ without the need for biopsy or
resulting in hypoxic-resistant tumor cells and surgical resection. Furthermore, imaging tech-
alternate pro-angiogenic “escape” mechanisms. niques support studies of the entire tumor, can
In glioma there is also evidence that EC can be provide information on the heterogeneity of indi-
formed from primary tumor stem cells via a pro- vidual biological features, and can, in many cases,
cess that is relatively insensitive to blockade of be optimized to provide biological data of direct
VEGF. While the lack of curative single-agent relevance to tumor biology. These features,
efficiency is disappointing, it is clear that together with the minimally invasive nature of
308 A. Jackson et al.

most imaging techniques and the ability to repeat used primarily in the intraoperative setting or in
studies whenever necessary, give imaging signifi- animal models of disease, and will not be
cant advantages over other methodological discussed further.
approaches. Most early imaging studies focused on the char-
In recent years, the concept of the imaging acterization of aspects of the TVM involved in the
biomarker (IB) has become the basis for the devel- angiogenic process, specifically the amount of
opment of novel imaging methods. A biomarker is blood vessels and the increases in capillary endo-
defined as “a quantitative parameter which directly thelial permeability induced by pro-angiogenic
reflects the physiology, anatomy or pathological cytokines. Both of these features can be estimated
process under investigation.” The use of soluble using dynamic contrast-enhanced imaging tech-
and tissue biomarkers, like prostate-specific anti- niques, which are now widely employed both in
gen or CA 125, has led to the development of clinical practice and in trials of antiangiogenic and
“biomarker roadmaps” designed to standardize vascular targeting agents. However, more recently
the discovery, technical, and biological validation there has been significant interest in developing
of individual biomarkers to ensure that they are fit biomarkers that target other aspects of the TVM,
for purpose. The concept of an IB is identical and leading to the description of an increasingly wide
requires identification of a potentially valuable, range of IB and, in particular, of PET-based molec-
measurable imaging feature, proof that it is techni- ular imaging agents.
cally measurable in single or multi-institutional
settings, characterization of its repeatability and
reproducibility, and evidence to support its biolog- Biomarkers of Perfusion
ical validity. For imaging techniques the technical and the Vascular Microenvironment
validation steps are often more extensive than those
required for soluble and tissue biomarkers and can The early development of IB for tumor microvas-
be technically challenging, particularly where cular structure focused on attempts to measure the
imaging techniques must be standardized across known histological features of angiogenic tissue.
different manufacturers, imaging platforms, and Classically, histochemical staining studies had
institutions. Despite this, the development of identified the importance of the density of new
novel IB now forms one of the main areas of vessels (microvascular density; MVD) measured
imaging research both in cancer and other diseases. on stained tissue. MVD has been shown to relate
In the remainder of this chapter, the IB avail- to malignancy and outcome in a number of tumors
able for quantified imaging of the TVM is and is routinely used in histological studies [6]. It
reviewed, the evidence for their technical and was quickly realized that a theoretical surrogate of
biological validity is discussed, and their applica- MVD could be extracted from dynamic contrast-
tions to the present time are reviewed. enhanced MRI.

Imaging Biomarkers Dynamic Susceptibility Contrast-


Enhanced MRI (DSCE-MRI)
The development of IB to characterize the tumor
microenvironment and the angiogenic process has Early studies concentrated on the use of dynamic
been and continues to be a major area of imaging T2*WI acquisitions, often referred to as Dynamic
research. All major modalities have been utilized, Susceptibility Contrast-Enhanced MRI (DSCE-
including CT, MR, single-photon emission com- MRI), in neoplastic and cerebral vascular disease.
puted tomography (SPECT), positron emission The initial focus on DSCE-MRI arose from the
tomography (PET), ultrasound, and optical imag- fact that system architecture at that time made
ing. The last two are largely experimental, being rapid dynamic acquisitions with T2* star
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 309

weighting more straightforward than dynamic unpopular since, although these do abrogate the
T1-WI imaging. More importantly, the signal effect of the so-called T1 shine through signal,
change in T2*WI images arises from the effects they also result in significant decreases in signal-
of the locally altered magnetic field which extends to-noise ratio with consequent reductions in the
some distance from the contrast molecule. In iso- accuracy of subsequent curve fitting analyses.
lated capillary beds this means that signal is gener- The growing application of DSCE-MRI in
ated from protons within the extravascular neuro-oncology led to a number of additional
extracellular space (EES), even if contrast mole- analytical approaches designed to provide addi-
cules remain entirely intravascular. This suscepti- tional data about the TVM. It was rapidly realized
bility effect gives rise to slightly higher that in tumor vasculature, the normal shape of the
proportional signal changes in the normal gray CC-TCC was distorted by retention of contrast
and white matter which is a significant advantage agent following the first passage of the contrast
in normal cerebral capillary beds. The analysis of bolus. This elevation of contrast concentration
the dynamic signal intensity time course curves was first described as the relative recirculation
(SI-TCC) from DSCE-MRI is also relatively (rR) parameter and has also been widely referred
straightforward. Dynamic time course signal to as the percentage recovery (describing the per-
changes can be transformed into estimates of con- centage of signal recovery to the baseline). This
trast concentration time course curves (CAA-TCC) has been variously attributed to contrast linkage
from which estimates of proportional cerebral and to retention of contrast agent within the abnor-
blood volume can be derived as the area under mal microvasculature due to vessel tortuosity and
the contrast agent concentration curve during its regional reductions in perfusion pressure. These
first passage. This area correlates with the propor- metrics allow identification of areas of tumor
tion of the volume of the voxel filled by blood where capillary flow is present, but delayed,
vessels often referred to as the relative cerebral corresponding to regions with reduced perfusion
blood volume (rCBV). By scaling to normal pressure and reduced flow and has been used to
white matter or pure vascular tissues (large veins), differentiate GBM from metastasis [9].
rCBV can be normalized to produce absolute The development of more complex image anal-
values (CBV), which have shown close agreement ysis techniques led to the development of pharma-
with histological assessment of MVD in a number cokinetic models designed with the intention of
of cancer types and show similar correlations with measuring regional blood flow and/or capillary
tumor grade, aggressiveness, and survival data [7]. endothelial permeability. These approaches require
The basic analysis approaches for the DSCE- correction of the tissue residue function from the
MRI assumed that contrast agent remains intra- individual tumor voxels to correct for variations in
vascular in the presence of an intact blood–brain the width of the injected arterial bolus, known as the
barrier. In tumor tissue this causes very significant arterial input function (AIF). Correcting for the
errors since contrast agent leakage into the EES variation between patients resulting from individual
produces contradictory signal changes, due to T1 variations in AIF allows estimations of regional
effects. These give rise to artificially low estimates capillary blood flow. Unfortunately, the accuracy
of CBV (so-called T1 shine through). These leak- of these estimates is greatly affected by delays in
age errors can be reduced by preloading the tissue the arrival of the contrast bolus within the tissue and
with a dose of contrast agent prior to the dynamic by variations in dispersion of the contrast bolus,
contrast study, by the use of low flip angle which result from a combination of flow delays
sequences with low T1 sensitivity, or by signal and irregular vessel flow patterns. The result of
post-processing [8]. Increasingly, contrast agent these effects is that regional capillary flow estimates
preloading, signal processing, or a combination of can only be acquired reliably where there can be an
the two is routinely used. The use of low flip angle assumption of normal arterial/capillary/venous hier-
acquisition sequences has been relatively archy, which is not the case in tumor tissue [10].
310 A. Jackson et al.

Most early antiangiogenic agents targeted approaches and the lack of available standardized
VEGF or VEGF signaling. Since VEGF produces software for clinical application (Fig. 2) [11].
rapid and significant increases in the permeability Despite this a desire for better standardization
of the endothelial membrane, considerable work across imaging systems and for greater physiolog-
was directed toward the quantification of perme- ical specificity led to the development and wide-
ability by measurement of MR contrast agent spread application of pharmacokinetic analytical
leakage. Analysis of DSCE-MRI using a approaches [12].
two-compartment leakage model was described Pharmacokinetic analysis of DRCE-MRI data
in an attempt to estimate permeability surface requires transformation of signal change measure-
area product of the local tumor capillary bed. ments into estimates of contrast agent concentra-
These pharmacokinetic analysis techniques tion to allow the measurement of CC-TCC (Fig. 3).
allow estimation of the contrast transfer coeffi- This can be performed assuming that the base-
cient (Ktrans) between the plasma and extravascu- line T1 values of the tissue are measured prior to
lar extracellular space (EES). However, modeling contrast agent administration. While accurate mea-
of leakage effects using T2* dynamic images is surement of T1 values can be time-consuming, the
challenging, and attention rapidly shifted to the use of multiple flip angle gradient echo sequences
use of T1-WI dynamic acquisitions. produces adequate accuracy in acceptable acquisi-
tion times and has become routine. Analysis of the
CC-TCC can then be undertaken using a variety of
Dynamic Relaxivity Contrast-Enhanced pharmacokinetic distribution models. All of these
MRI (DRCE-MRI) models require provision of an explicit input func-
tion representing the arterial input to the tissue
On T1-WI images, signal change observed in (AIF). The accuracy of measurement of AIF has a
response to intravascular and extravascular contrast considerable effect on the accuracy of derived
agent is additive, simplifying attempts to estimate values of parameters from the analysis, and many
the magnitude of contrast agent leakage in compar- authors have studied the optimal sampling and
ison to DSCE-MRI. Furthermore, the majority of modeling of the AIF. Where an AIF cannot be
clinical MR scanning systems are capable of acquir- measured with sufficient accuracy, then a
ing DRCE-MRI data. DRCE-MRI data can be ana- population-based AIF may be applied.
lyzed using simple semiquantitative metrics, The most common pharmacokinetic analysis
although in research applications these have com- approach is a modified version of the original
monly been avoided since intensity changes can Toft’s pharmacokinetic model that produces esti-
vary between systems due to differences in acqui- mates of vascular fraction (vp), extravascular
sition sequence design, receiver gain settings, and extracellular space fraction (ve), and the transfer
other technical variables. Early studies attempted to contrast coefficient (Ktrans) [14]. The Ktrans param-
minimize these sources of variation by the use of eter is widely used in DRCE-MRI studies, often as
simple, normalized, metrics such as the maximal a potential surrogate of capillary endothelial per-
rate of change of signal intensity or the ratio of pre- meability. However, although Ktrans will be
and postcontrast signal intensity. Subsequent affected by the permeability, it is a bulk transfer
improvements in hardware and particularly in gra- coefficient, so that it is also affected by the surface
dient coil design have minimized variations in area of the capillary endothelial bed and by rate of
observed signal change, and these together with delivery. This means that in very low flow states,
the use of normalized metrics have led to routine particularly where leakage is high, the transfer coef-
clinical use of semiquantitative measurements in ficient will depend almost entirely on flow, whereas
prostate, breast, pancreatic, cervical, colonic, and in high flow states with low permeability, it will
rectal cancers, bone sarcoma, as well as brain depend almost entirely on permeability. Nonethe-
tumors. The clinical preference for semiquantitative less it rapidly became evident that the VEGF inhi-
metrics reflects the complexity of pharmacokinetic bition was indeed associated with rapid and
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 311

High
Day0

Day0

Day2

Day90

low
R10 Ktrans vp ve SE1min SErel ∑SE ∑SErel Rse1/se2

Fig. 2 Axial view of central slices of 3D longitudinally uses a baseline value for normalization in order to reduce
co-registered kinetic and semiquantitative parametric maps the dependence on biological and imaging system
obtained in a 26-year-old woman, who has Type II neurofi- variables, (3) the sum of SE over a fixed post-injection
bromatosis with a progressive VS (arrow heads) and an duration (SE), (4) the sum of SErel over a fixed post-injection
occipital located meningioma (arrow) undergoing treatment duration (SErel), and (5) signal enhancement ratio,
with bevacizumab. Images show comparisons of pharmaco- commonly defined as the ratio of early  to late contrast

kinetic leak derived parameters Ktrans, vp, and ve with enhancement ratio, e.g., Rse1=se2 ¼ SI1min post  SIpre =
 
semiquantitative parameters: (1) absolute signal enhance- SI5min post  SIpre (From Ref. [11] with permission)
ment (SE1min), (2) relative signal enhancement (SErel) which

profound reductions in Ktrans [13]. Although the data with high signal-to-noise characteristics. The
modified Toft’s model has been widely used, a need to “square the circle” by producing very high
variety of alternate pharmacokinetic models exist, spatial resolution data with high temporal resolu-
ranging from simple two-compartment models, tion measurement of the AIF and high signal-to-
such as the original Toft’s model, to more complex noise characteristics has led to extensive research
models, such as the adiabatic tissue homogeneity into MR image acquisition leading to a number of
model, where more specific physiological informa- novel fast image acquisition approaches for
tion is sought by definition of additional biological DCE-MRI and the development of dual injection
variables (Table 1). methods where separate dynamic acquisitions are
The choice of appropriate pharmacokinetic acquired to provide matched high spatial and high
models depends on a wide range of factors. In temporal resolution data.
some instances the demands of an individual
study may require absolute estimation of physio-
logical variables such as regional capillary blood Dynamic Contrast-Enhanced
flow (F) which can be provided only by more Computed Tomography (DCE-CT)
complex models. In contrast, the use of increas-
ingly complex models introduces uncertainty into With the development of rapid multi-slice CT
parameter estimates due to the need to optimize to acquisitions, it was natural that the analytical
an increasing number of variables. This in turn approaches taken with DCE-MRI would be
places significant demands on the image acquisi- applied to CT data. Dynamic CT (DCT) has a
tion strategy requiring high temporal resolution number of potential advantages over MRI. The
312 A. Jackson et al.

R
Fig. 3 DCE-MRI data acquisition and analysis. (a) Multi- Abbreviations: [CA]dt area under the contrast agent–time
ple images (typically 25–100) are acquired as a bolus of curve, AIF arterial input function, Cp contrast agent concen-
contrast agent passes through tissue capillaries; (b) the tration in plasma, CA contrast agent, DCE-MRI dynamic
region of interest for a tumor and the feeding vessel arterial contrast-enhanced MRI, EF enhancing fraction, ETV
input function are defined; and (c) signal intensity values for enhancing tumor volume, IAUC60 initial area under con-
each voxel are converted into contrast agent concentration centration agent–time curve at 60 s, Ktrans volume transfer
using a map of T1 values. These steps allow calculation of constant between plasma and the extracellular extravascular
(d) WTV and T1 values. Next, voxels are classified as leakage space, ROI region of interest, S signal, T1 longitu-
enhancing or not after which parameters based on the dinal relaxation time, ve volume of extracellular extravas-
amount and proportion of enhancement can be defined, cular leakage space, vp blood plasma volume, WTV whole
along with the IAUC60. Finally, a pharmacokinetic model tumor volume (From Ref. [13] with permission)
may be applied to derive parameters such as Ktrans.

main one of these is that the concentration of


contrast agent is directly linearly related to the Imaging Microvessel Morphology
measured attenuation value; this makes measure- and Hypoxia with MRI
ments of semiquantitative parameters and the
application pharmacokinetic models more simple Vessel Size Imaging (VSI) and Vessel
and alleviates many of the problems associated Architectural Imaging (VAI)
with multicentered studies in MRI [16]. The main
problem with DCT is of course the radiation dose, As described above, a great deal of the develop-
which is significant and limits the application of ment of dynamic contrast-enhanced techniques
the technique into clinical trials requiring repeated arose from a desire to study changes in regional
imaging measurements. blood volume or capillary endothelial permeabil-
ity. An important feature of antiangiogenic
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 313

Table 1 Parameters derived in DCE-MRI


Parameter Definition Unit Notes
Primary end pointsa
Ktrans Volume transfer constant between min1 Composite measure of permeability, capillary
plasma and the EES surface are and flow
IAUC60 Initial area under concentration agent- mmol min Similar measure to Ktrans, but also influences
time curve at 60 s by Ve
Alternative functional biomarkersb
kep Rate constant between EES and plasma min1 NA
Ve Volume of EES per unit volume of tissue NA NA
Vp Blood plasma volume NA Relatively poor reproducibility
F Blood flow ml/ Temporal resolution achieved in most studies s
g min1 too slow
PS Permeability surface area product per ml/ Temporal resolution achieved in most studies is
unit mass of tissue g min1 too slow
Alternative biophysical measurementsc
WTV Whole tumor volume mm3 Easy to measure
ETV Enhancing tumor volume mm3 Easy to measure
EF Enhancing fraction (ETV:WTV ratio) none Easy to measure
T1 Longitudinal relaxation time ms Easy to measure
DCE-MRI dynamic contrast-enhanced MRI, EES extracellular extravascular leakage space, NA not applicable
a
As recommended by Leach et al. [15]
b
Require pharmacokinetic modeling
c
Do not require pharmacokinetic modeling

therapy includes reduction in the abnormal has been used, in drug trials, to demonstrate vessel
tumoral vasculature to produce the so-called “nor- normalization in response to antiangiogenic ther-
malization” of vascular structures. This normali- apies (Fig. 4) [20].
zation process is characterized by a reduced More recently this approach has been extended
number of vessels of increasingly normal caliber following the realization that the difference in
and reestablishment of the arterial/capillaries/ contrast mechanisms between T1 and T2*
venous hierarchy [17]. As discussed above the dynamic acquisitions produces not only the dif-
signal changes resulting from T1 and T2* relaxa- ference in signal intensity but also an apparent
tion measurements differ significantly. One prin- temporal variation between the maximal effect of
cipal difference is that the relaxivity effect, which T1- and T2*-based signal changes [21]. Modeling
produces the reduction in T1, is limited to the of the temporal discrepancies of these signal
immediate spatial environment of the proton, changes allows estimation of the homogeneity
while the variation in local field intensity which and direction of flow. Furthermore the signals
results in T2* contrast extends a significant dis- are differentially affected by oxygen tension so
tance beyond the proton, affecting not only the that an estimate of regional oxygen extraction can
vessels containing contrast but also the surround- also be calculated. This technique, known as ves-
ing tissue. As a consequence of this difference in sel architectural imaging (VAI), has also been
the contrast mechanisms, it is possible to model applied in studies of antiangiogenic therapies
the effect of vessel size and vessel distribution showing different patterns of change in the early
and, using paired T1 and T2*WI acquisitions, to phases of treatment in patients who respond to
derive a calculated parameter which reflects aver- therapy. VAI has also been used to study patients
age vessel size (Rv) [18]. This technique is with glioblastoma treated with cediranib and
referred to as vessel size imaging (VSI). VSI is chemoradiation, demonstrating that increases in
increasingly employed in research settings and perfusion and oxygenation, seen in approximately
314 A. Jackson et al.

Fig. 4 Reversibility of Drug Holiday


normalization: (a) Vascular
and volume changes as a Post-contrast T1-weighted MRI
function of time in a patient
who did not take drug from
days 43 to 56 and was
imaged on day 55 (shown as
drug holiday). T1-WI
anatomic images after
intravenous administration
of gadolinium-DTPA,
similar to Fig. 2. Note that at
day 55, there is a rebound in
tumor enhancement, which
decreases again after
restarting the drug as seen
on follow-up imaging on
day 110. In this patient,
maps of relative vessel size
(similar to Fig. 1c) also Relative Vessel Size
show fluctuation with the
drug holiday and −1 1 28 55 111
resumption of AZD2171 Study Days
treatment. (b)
Measurements of imaging Drug Holiday
parameters confirm the 120 Day 43 to 55
Value relative to pretreatment (%)

reversibility of vascular
normalization by drug
interruption followed by VS
renormalization after
80
AZD2171 is resumed
(From Ref. [19] with
P
permission)

40
FLAIR

CE-T1
0
−1 1 28 55 111
Study Days

50 % of patients, are associated with improved (PS), transfer constant (Ktrans), washout rate
survival. In comparison an increase was seen in (kep), interstitial volume (Ve), blood volume
only one of 14 patients with GBM treated with (Vb), capillary transit time (tc), and capillary het-
chemotherapy and radiation alone [21]. erogeneity (a1) [22]. This has been termed the
gamma capillary transit time (GCTT) model, and
the introduction of the capillary transit time and
Capillary Heterogeneity Imaging heterogeneity terms represents novel biomarkers
that could provide new evidence about microvas-
Extensions of current pharmacokinetic modeling cular structure. Jensen et al. [23] applied the
approaches have also been developed for DRCE- model in a group of patients with glioblastoma
MRI. St Lawrence et al. extended their original and found close correlation between the capillary
adiabatic tissue heterogeneity model to provide transit time, HIF-1, and VEGF expression in
spatial maps of tumor blood flow (F), extraction active tumor. Furthermore, patient survival corre-
fraction (E), permeability–surface area product lated with only two DCE parameters: capillary
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 315

Fig. 5 Example of Avol in a representative case of overlaid. The overlap of the two is indicated in yellow.
untreated GBM. (Top) Row 1 shows a series of T1-WI (Bottom) Mean MR signal within voxels defined by ICA
postcontrast images, and row 2 shows the same series with components and the overlap (From Ref. [24] with
the arterial (red) and venous (blue) ICA components permission)

heterogeneity in active tumor and interstitial vol- They used this arteriovenous overlap (AVOL)
ume in areas of peri-tumoral edema. as an index of abnormal tumor microvasculature,
showing that median survival was greater in
patients where the Avol decreased in response to
Arteriovenous Overlap Imaging (AVOL) bevacizumab treatment. They propose the Avol as
an independent biomarker of abnormal microvas-
La Violette et al. [24] studied a group of 43 patients cular hemodynamics. In a subsequent study the
with high-grade glioma using independent compo- same authors demonstrated significant sensitivity
nent analysis to separate the vasculature into arte- of the Avol measurement to contrast leakage and
rial and venous components. They observed that modify their recommendations to suggest that the
the abnormal vascular flow patterns in tumors gave measurement should be performed on a second
rise to a number of voxels that were classified both dose of contrast following a preloading dose to
as arterial and venous (Fig. 5). saturate the T1 effects of contrast leakage [25].
316 A. Jackson et al.

Imaging Hypoxia Using MRI However, although these signal changes may
have some diagnostic value, they have not been
Hypoxia plays a central role in tumor development, biologically qualified as an IB of tissue hypoxia,
angiogenesis, tumor growth, and resistance to treat- and it is clear that the signal would also be affected
ment. Hypoxia is intimately related to the structure by other causes of susceptibility contrast such as
and function of the vascular microenvironment. calcification or microhemorrhage.
Inadequate supply of oxygen and other nutrients Attempts to qualify the presence of hypoxia
to tumor cells and to tumor support and inflamma- using T1-WI oxygen-enhanced magnetic reso-
tory cells results in local hypoxia and which forms nance imaging (OE-MRI) measurements have
one of the main pro-angiogenic stimuli. Likewise, recently been described [27]. These and previous
failure of angiogenesis to keep pace with the rate of works have demonstrated that increasing the con-
tumor proliferation or metabolism will give rise to centration of inspired oxygen produces a signal
areas of hypoxic tissue within the tumor. The pres- detectable in blood and tissues on T1-WI imaging.
ence of hypoxia has a profound effect on tumor Animal models of the area under the curve
biology and has been shown to correlate with the resulting from oxygen enhancement showed pos-
probability of metastatic spread, tumor recurrence, itive correlation with histologically demonstrated
resistance to chemotherapy and radiotherapy, inva- hypoxic tissue and a negative correlation with
sive phenotype, and decreased patient survival. It is vessel density (Fig. 7).
clear that prolonged or severe hypoxia induces These relationships were not seen with param-
phenotypic changes such as loss of genotype sta- eters derived from DCE-MRI measurements.
bility, loss of apoptotic potential, and oncogene Clinical data confirm that the same changes were
activation. Imaging methods for the identification seen in patients with glioblastoma apparently
of hypoxic tissue have classically relied on the use identifying central areas of hypoxic tissue. The
of positron emission tomography (vide infra). use of T1-WI dynamic imaging together with
However, there is increasing interest in the use of oxygen enhancement appears highly promising
MR-based techniques to identify areas of hypoxic although further biological and technical valida-
tissue. As described in the previous section, VAI tion is still required .
has been shown to allow measurement of regional
oxygen consumption although the biomarker has
not yet been fully biologically qualified. Similarly, Molecular Imaging of the Tumor
capillary heterogeneity imaging demonstrates Vascular Microenvironment
close correlation with the expression of HIF-1.
Over the past several years, susceptibility- Understanding of the tumor vascular microenvi-
weighted imaging (SWI) has become part of rou- ronment and of the angiogenic process continues
tine neuroimaging. SWI is used to detect magnetic to increase at an almost exponential rate. Enor-
susceptibility of different tissues and is now clin- mous investment leads to the ongoing identifica-
ically used to assess neoplastic microvascular, tion of numerous novel therapeutic targets and
microbleeds and necrotic areas. One potential candidate compounds which will pass into pre-
cause of increased signal on SWI is the presence clinical and, eventually, for the more successful
of increasing concentrations of deoxyhemoglobin agents, into clinical trial. Although dynamic
providing another potential IB for hypoxic tissue. contrast-enhanced imaging methods have pro-
In a recent study, 64 patients with brain tumors vided, and continue to provide, the mainstay of
underwent SWI demonstrating that R2* values imaging biomarker support for such studies, they
were significantly different between low-grade describe only selected elements of the tumor
and high-grade glioma. The authors suggest that microvascular environment. There is a growing
the R2* value may reflect alterations in deoxygen- demand for biomarkers that can quantify specific
ated hemoglobin which are higher in high-grade molecular processes to provide a more compre-
than low-grade glioma [26] (Fig. 6). hensive toolkit for the investigation of the TVM
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 317

a
a b c

b 40.00 a

62

30.00
R2* values

20.00

10.00

0.00

low-grade high-grade meningiomas brain


gliomas gliomas metastasis
brain tumors

6.00 b

5.00

4.00
23
CBF

3.00
38
2.00
13
1.00

0.00
Low grade High grade Meningiomas Metastasis
gliomas gliomas
Brain tumors

Fig. 6 (a) A 46-year-old man with a histologically proven and rCBF values of four types of brain tumors. (a) The
glioblastoma multiforme (grade IV) located in the right decreasing order of R2* values is high-grade gliomas,
frontal lobe. (a) Axial T1-WI postcontrast image demon- brain metastasis, meningiomas, and low-grade gliomas. (b)
strates nodular enhancement in the tumor. (b) The R2* map The sequence rCBF values are meningiomas, high-grade
shows an area of high R2* values (31.22) in the tumor. gliomas, brain metastasis, and low-grade gliomas (From
There is hemorrhage at the bottom of the tumor. (c) The Ref. [26] with permission)
ASL CBF map demonstrates nodular hyperperfusion. b R2*
318 A. Jackson et al.

Fig. 7 (a) Tumors from five patients with GBM. Top row: scale with all images windowed to maximize visible
area under the DR1 curve for OE-MRI, AUCOE displayed dynamic range. Numerals indicate patient tumor index.
using a hot color scale for positive values and negative (b) Correlation between negative AUCOE areas following
values using a cool color scale. All images windowed to oxygen inhalation with vessel density (circles) and per-
maximize visible dynamic range symmetrically around centage hypoxic area (crosses) in all U87MG tumors
zero AUCOE change. Bottom row: area under the DR1 (From Ref. [27] with permission)
curve for DCE-MRI, IAUCGd displayed using a hot color

and of angiogenesis, antiangiogenic, and Many candidate tracers are currently in develop-
antivascular therapies. This has led to the devel- ment and have been used for preclinical studies.
opment of a number of novel radiotracers, princi- For a review of the current state-of-the-art in this
pally designed for use with positron emission field, the reader is directed toward a number of
tomography (PET), which target molecules major review articles. In many cases the focus of
expressed by vascular and vascular support cells. these agents is on systemic tumors, and there has
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 319

been little application in brain tumors in humans. ratio of the hypoxic volume to the MRI
The main area of interest with regard to human T2-defined tumor volume) and the net rate of
glioma has been around imaging of hypoxia and cell proliferation, as well as between the biologi-
integrin expression. cal aggressiveness ratio (defined as the ratio
between the net rate of cell proliferation and the
net rate of invasion) and relative hypoxia, scaled
Imaging Hypoxia Using PET to the blood activity of the tracer.
18F-FMISO and 15O-H2O PET have also
PET imaging is an ideal modality for evaluating been used in brain tumors to provide matched
hypoxia. The PET imaging agent fluoromiso- measures of tumor hypoxia and perfusion.
nidazole (18F-FMISO) is an 18F fluorinated Increased 18F-FMISO brain tumor retention was
derivative of misonidazole, an azomycin hypoxic observed in GBM (seven out of seven) and was
cell sensitizer. It binds covalently to intracellular associated with an increased 18F-FMISO tumor
molecules at a rate inversely proportional to intra- distribution volume, which was also used as a
cellular O2 concentration. 18F-FMISO diffuses quantitative criterion for hypoxia. 18F-FMISO
freely across the blood–brain barrier and is accumulated in both hypo- and hyperperfused
reduced at any hypoxic site where it is not tumor regions, suggesting that hypoxia in glio-
excreted or highly metabolized and its metabolites blastoma may develop irrespective of the magni-
have a rapid plasma clearance, so normalization tude of perfusion [28]. Other studies have
for delivery is not required. Quantitatively, the demonstrated correlations between 18F-FMISO
degree of hypoxia can be defined by hypoxic uptake and expression of vascular endothelial
fraction or volume or can be expressed by its growth factor (VEGF)-R1 and Ki67 expression
severity, defined as the region with the lowest in glioblastomas and a relationship between the
oxygen concentration, and its relative level. 18F- uptake volume and intensity and both tumor pro-
FMISO imaging studies cannot provide values of gression and overall survival (Fig. 8).
regional oxygen concentration, but the maximum Relatively poor tumor-to-background ratios
tumor/blood ratio is a convenient surrogate for the combined with slow clearance from normal tissue
worst level of hypoxia in the image. 18F-FMISO have led to the ongoing search for improved hyp-
is currently the most widely used positron emis- oxia imaging agents. For a fuller review of the
sion tomography (PET) imaging agent for hyp- potential advantages and disadvantages of these
oxia. The imaging procedure is a well-tolerated alternate imaging agents, the reader is directed to a
procedure by patients and imaging takes around number of comprehensive review articles.
20–30 min, starting from 75 to 150 min after
injection.
18F-FMISO has been validated in several ani- Imaging Integrin Expression
mal models and human disease conditions, and its
signal is independent of other factors associated Integrins are membrane receptors comprised of an
with hypoxia, such as regional glucose concentra- α and a β subunit that mediate interactions
tion, glutathione levels, other transporters, between cells. To date, 18 different α and 8 differ-
and pH. ent β subunits have been identified, forming
In 3 studies of 34 patients with glioma, only 24 different integrin receptors [29]. A common
glioblastoma demonstrated constant uptake of property of many integrins is their interaction with
18F-FMISO, whereas neither low-grade glioma the arginine-glycine-aspartic acid (RGD)
nor anaplastic astrocytoma showed this effect. A sequence found in extracellular matrix proteins
recent study in GBM patients, imaged preopera- like vitronectin, fibrinogen, thrombospondin, and
tively using 18F-FMISO, showed positive corre- fibronectin. The most extensively studied of these
lations between relative hypoxia (defined as the in the angiogenic process is the integrin αvβ3
320 A. Jackson et al.

Fig. 8 (a–c) 15O-H2O PET perfusion images in three inverse pattern compared with hypoxia, and in high perfu-
patients with glioblastoma multiforme. (d–f) sion. PET images are normalized to their own maximum
Corresponding late 18F-FMISO PET images show tumor (From Ref. [28] with permission)
hypoxia in low perfusion, in intermediate perfusion with an

which is highly expressed on the surface of acti- with immunohistochemical assays of αvβ3
vated EC. However, integrins are commonly integrin expression [32] (Fig. 9).
expressed on the surface of tumor cells, and the A number of modifications designed to
interpretation of the signal obtained from integrin improve the pharmacokinetic properties of RGD
imaging can therefore be complex [30]. The first peptides have been described, including the
integrin-specific PET tracer used in humans was development of multimeric tracers designed to
[18F]Galacto-RGD. The initial study of mixed more effectively identify tumors in areas of high
tumor types demonstrated wide variability of physiologic integrin expression. Direct compari-
tumor uptake with high background in the kid- son of monomeric, dimeric, and tetrameric cyclic
neys, liver, spleen, and bowel. Immunohis- RGD tracers conjugated with DOTA and
tological examination of resected tissue radiolabeled with 111In in mice with SK-RC-52
demonstrated that the number of αvβ3-positive renal cell carcinoma xenografts demonstrated
vessels per field of view correlated highly with tumor uptake of the tetramer which exceeded
tumor uptake of [18F]Galacto-RGD [31]. A sim- that of both the dimer and the monomer [34],
ilar correlation between uptake and immunohisto- and a 64Cu-DOTA-RGD octamer demonstrated
chemical staining was also demonstrated in a later, higher uptake than tetramer versions in human
larger study. In glial cell tumors there was moder- glioblastoma tumor grafts [35].
ate but variable uptake, most marked in areas of One significant alternate integrin-specific
high proliferation or tumor infiltration. Immuno- tracer is [18F]-fluciclatide, which has been devel-
histochemical staining was positive both in EC oped by GE Healthcare and is becoming widely
and in tumor cells, and tracer uptake correlated available. This commercially available tracer
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 321

Fig. 9 Examples of
patients with glioblastoma
multiforme of the left
frontal lobe (a, b) and right
parieto-occipital lobe (c, d).
Note the intense peripheral
enhancement in the
gadolinium-DTPA-
enhanced MRI scans in both
tumors (a, c). However, the
tumor in (b) shows only
very faint tracer uptake in
the [18F]Galacto-RGD PET
(maximum standardized
uptake value [SUVmax],
1.2), whereas the tumor in
(d) demonstrates
substantially more intense
[18F]Galacto-RGD uptake
(SUVmax, 2.8) (From Ref.
[33] with permission)

shows different binding affinities with the highest


affinity for αvβ5 (IC50 0.1 nM) followed by Clinical Applications of Microvascular
integrin αvβ3 (IC50 11.1 nM). A study in human Imaging
glioma heterografts treated with sunitinib demon-
strated a therapy-induced reduction in tumor The use of IB microvascular structure and func-
uptake over a 2-week period. This correlated tion has been the subject of extensive clinical
with a reduction in tumor MVD suggesting that research, and some better-characterized IB are
this agent is capable of demonstrating vascular now passing into routine clinical use. Since
responses to transgenic inhibition prior to the much of the development of DCE-MRI tech-
occurrence of significant volumetric changes [36]. niques occurred in the brain, a number of these
The potential role of integrin imaging in the have now been shown to have actual or potential
clinical environment remains uncertain. Observa- neuro-oncological application.
tions of associations between integrin expression
and tumor progression in melanoma and tumor
grade in sarcoma have suggested potential clinical Diagnosing Glioma Mimics
utility. Similarly, preclinical studies have demon-
strated that integrin αvβ3 has an important role in In a large series of 5,000 stereotactic biopsies,
promoting aggression and metastatic potential in only 64.4 % were glioma, and it remains true,
breast cancer and glioma. These observations sug- despite improvements in routine clinical imaging,
gest specific potential clinical utility for integrin that these lesions cannot always be confidently
imaging, particularly in the selection of patients diagnosed. DCE-MRI techniques have been
for antiangiogenic therapies. shown to allow improved differentiation between
322 A. Jackson et al.

glial cell tumors and common diagnostic mimics. that differentiation between grade III and grade IV
Distinguishing solitary metastases from high- glioma is unreliable with either technique.
grade glioma can be difficult; however, several Identification of areas of dedifferentiation in
studies have shown that rCBV values are higher low-grade tumors and of aggressive areas in
in GBM particularly in the region surrounding the HGG, particularly grade III, avoids erroneous
malignant tumor [9]. This has been attributed to undergrading of the tumor due to failure to sample
the presence of mixed vasogenic and cytotoxic the most aggressive component. In many clinical
edema due to tumor cell invasion around the mar- practices this has led to routine use of rCBV maps
gins of GBM. The diagnosis of cerebral lym- to identify optimal sites for biopsy.
phoma is also greatly strengthened by the low
rCBV values that these tumors exhibit, clearly
distinguishing them from GBM which shows Identifying Transformation in
high rCBV despite very similar enhancement pat- Low-Grade Glioma
terns on contrast-enhanced static images
[37]. Another major differential diagnosis of Low-grade glioma has a propensity to undergo
GBM is cerebral abscess. Abscess may have malignant transformation, but the time scale is
very similar classical imaging appearances but is highly variable between individuals. Monitoring
of course an important urgent medical diagnosis. of patients with low-grade disease is aimed at
Once again rCBV in the rim of an enhancing identifying transformation at an early stage and
access is found to be significantly lower than has classically relied on the development of con-
that seen in HGG [38]. Although there is signifi- trast enhancement within the tumor. Contrast
cant overlap between these groups, diagnostic enhancement can result from contrast leakage
sensitivity and specificity become very high due to increased endothelial permeability or
when combined with measurements of apparent from increased intravascular contrast due to
diffusion coefficient from diffusion-weighted increased proportional blood volume. Measure-
imaging, which demonstrates restricted diffusion ment of increased rCBV using the DSCE-MRI
in the glial tumors [38]. has been shown to allow early detection of dedif-
ferentiation by 6–12 months compared with con-
ventional imaging and is now in common clinical
Grading Glioma use [42].

Many studies have described variations in DCE-


MRI-derived biomarkers between different grades Distinguishing Recurrent Tumor from
of glioma [39]. To a large extent this is an inter- Treatment Effects
esting biological validation of the imaging bio-
marker but represents little or no clinical benefit in Following chemoradiotherapy there is a signifi-
the management of the patient. In all cases tissue cant incidence of apparent progression of the
will be obtained and tissue diagnosis will be tumor on conventional radiological criteria that
made, which remains, at the present time, the later resolves. This is referred to as pseudo-
gold standard diagnostic criteria [40]. Typically progression and can cause significant problems
studies show increases in angiogenic activity, pro- with management and treatment planning. The
portionate to increasing aggressiveness and grade pseudo-progression syndrome appears to result
of the tumor. Multiple authors have reported from leakiness of endothelial membranes due to
increases in rCBV related to grade, capable of vascular damage following chemoradiotherapy.
differentiating low-grade from high-grade glioma The incidence of pseudo-progression has been
[41]. Some authors have also demonstrated simi- reported as 9 % with radiation alone and 21–31 %
lar correlations between grade and vascular frac- with concomitant temozolomide. Routine MRI
tion from DRCE-MRI although it is noteworthy sequences are unable to distinguish between
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 323

true progression and early or late related treatment high-CBV tumor volume at week 3 vs. week
effects. Pseudo-progression is characterized on 1 [46]. These findings have been confirmed by a
DSCE-MRI by lower rCBV, lower relative peak number of other similar studies. Furthermore,
height of the enhancement curve, and higher rCBV following resection has also been shown
percentage signal recovery compared to recurrent to be predictive of overall survival in patients with
tumor [43]. However, although some investigators cerebral metastases.
have showed extremely high discriminative
power of rCBValone, others have failed to achieve
this despite the use of multimodality IB including Summary
MR spectroscopy and ADC measurements [43].
In the late stages following chemoradio- Despite an explosion of research concerning the
therapy, radiation necrosis may occur between biology of the TVM, knowledge of the subject
4 and 12 months after radiation. It is related to continues to increase exponentially as the drive
the areas of highest radiation dose and also causes for new anticancer therapeutics continues. The
significant problems with treatment planning. In availability of minimally invasive, accurate,
one study, 51 % of patients with radiological reproducible IB forms a major component of
evidence of glioma progression following that research drive. The past 20 years has seen
temozolomide were found to have radiation necro- the development and evolution of sophisticated
sis following surgical resection with no evidence of image acquisition and analysis techniques aimed
recurrent tumor. Microvascular IB, particularly at developing IB which address the most rele-
CBV, show differences between radiation necrosis vant components of disease physiology and
and recurrent tumor, being higher in tumor, but which are truly able to provide important infor-
these are not sufficiently great to allow confident mation of direct relevance. A vast amount of
diagnosis [44]. In one study sensitivity and speci- development continues to occur around dynamic
ficity were 83.3 % and 100 %, respectively. More contrast-enhanced methodologies, which have
recently Larsen et al. reported sensitivities and now become ubiquitous in drug discovery and
specificities of 100 % in a small study of 14 patients drug trials for a large range of antiangiogenic
[45]; however, these levels of accuracy have not and vascular targeting agents. Increasingly
been repeated in larger studies. Although DSCE- we are seeing these biomarkers being used
MRI may help make the differential diagnosis, it is in clinic although there remains a very signifi-
clear that the microvascular imaging biomarkers cant delay between the initial biological valida-
are not yet sufficiently discriminative to be used tions of novel IB in their eventual clinical
alone. qualification.
Despite major strides there are enormous chal-
lenges ahead. Although the development of IB has
Predicting Treatment Response been almost exponential in nature, there are sig-
nificant and growing anxieties about the complex-
Many groups have demonstrated a relationship ity of utilizing IB across multiple centers and in
between progression-free survival and/or overall large-scale studies. A number of international ini-
survival and IB describing the TVM. As early as tiatives are currently under way to try and stan-
2006 it was shown that glioma patients with dardize acquisition and analysis techniques and to
higher proportions of high rCBV had reduced improve both technical and biological validation
prognosis. Furthermore these workers showed and qualification strategies. This process of con-
that changes in tumor CBV during the early treat- solidation will continue in parallel with innova-
ment course also predicted for survival. Better tion because, although IB are expensive and
survival was predicted by a decrease in the frac- challenging to utilize, they offer many unique
tional low-CBV tumor volume at week 1 of RT properties and unparalleled advantages for the
vs. before RT and by a decrease in the fractional study of tumor biology.
324 A. Jackson et al.

References 15. Leach MO, Brindle KM, Evelhoch JL, Griffiths JR,
Horsman MR, Jackson A et al (2005) The assessment
1. Hanahan D, Weinberg RA (2000) The hallmarks of of antiangiogenic and antivascular therapies in early-
cancer. Cell 100(1):57–70 stage clinical trials using magnetic resonance imaging:
2. Patan S (2004) Vasculogenesis and angiogenesis. issues and recommendations. Br J Cancer
Cancer Treat Res 117:3–32 92(9):1599–1610
3. Carmeliet P, Jain RK (2011) Molecular mechanisms 16. Naish JH, McGrath DM, Bains LJ, Passera K,
and clinical applications of angiogenesis. Nature Roberts C, Watson Y et al (2011) Comparison of
473(7347):298–307 dynamic contrast-enhanced MRI and dynamic
4. Cheng SY, Huang HJ, Nagane M, Ji XD, Wang D, Shih contrast-enhanced CT biomarkers in bladder cancer.
CC et al (1996) Suppression of glioblastoma Magn Reson Med 66(1):219–226
angiogenicity and tumorigenicity by inhibition of 17. Jain RK (2001) Normalizing tumor vasculature with
endogenous expression of vascular endothelial growth anti-angiogenic therapy: a new paradigm for combina-
factor. Proc Natl Acad Sci U S A 93(16):8502–8507 tion therapy. Nat Med 7(9):987–989
5. Chen HX, Cleck JN (2009) Adverse effects of antican- 18. Schmainda KM, Rand SD, Joseph AM, Lund R, Ward
cer agents that target the VEGF pathway. Nat Rev Clin BD, Pathak AP et al (2004) Characterization of a first-
Oncol 6(8):465–477 pass gradient-echo spin-echo method to predict brain
6. Nico B, Benagiano V, Mangieri D, Maruotti N, tumor grade and angiogenesis. AJNR Am J
Vacca A, Ribatti D (2008) Evaluation of microvascular Neuroradiol 25(9):1524–1532
density in tumors: pro and contra. Histol Histopathol 19. Batchelor TT, Sorensen AG, di Tomaso E, Zhang WT,
23(5):601–607 Duda DG, Cohen KS et al (2007) AZD2171, a
7. Yao WW, Zhang H, Ding B, Fu T, Jia H, Pang L pan-VEGF receptor tyrosine kinase inhibitor, normal-
et al (2011) Rectal cancer: 3D dynamic contrast- izes tumor vasculature and alleviates edema in glio-
enhanced MRI; correlation with microvascular density blastoma patients. Cancer Cell 11(1):83–95
and clinicopathological features. Radiol Med 20. Sorensen AG, Batchelor TT, Zhang WT, Chen PJ,
116(3):366–374 Yeo P, Wang M et al (2009) A “vascular normalization
8. Jackson A, Kassner A, Annesley-Williams D, Reid H, index” as potential mechanistic biomarker to predict
Zhu XP, Li KL (2002) Abnormalities in the survival after a single dose of cediranib in recurrent
recirculation phase of contrast agent bolus passage in glioblastoma patients. Cancer Res 69(13):5296–5300
cerebral gliomas: comparison with relative blood vol- 21. Emblem KE, Mouridsen K, Bjornerud A, Farrar CT,
ume and tumor grade. AJNR Am J Neuroradiol Jennings D, Borra RJ et al (2013) Vessel architectural
23(1):7–14 imaging identifies cancer patient responders to anti-
9. Cha S, Lupo JM, Chen MH, Lamborn KR, McDermott angiogenic therapy. Nat Med 19(9):1178–1183
MW, Berger MS et al (2007) Differentiation of glio- 22. St Lawrence K, Verdecchia K, Elliott J, Tichauer K,
blastoma multiforme and single brain metastasis by Diop M, Hoffman L et al (2013) Kinetic model opti-
peak height and percentage of signal intensity recovery mization for characterizing tumour physiology by
derived from dynamic susceptibility-weighted con- dynamic contrast-enhanced near-infrared spectros-
trast-enhanced perfusion MR imaging. AJNR Am J copy. Phys Med Biol 58(5):1591–1604
Neuroradiol 28(6):1078–1084 23. Jensen RL, Mumert ML, Gillespie DL, Kinney AY,
10. Thacker NA, Scott ML, Jackson A (2003) Can Schabel MC, Salzman KL (2014) Preoperative dynamic
dynamic susceptibility contrast magnetic resonance contrast-enhanced MRI correlates with molecular
imaging perfusion data be analyzed using a model markers of hypoxia and vascularity in specific areas of
based on directional flow? J Magn Reson Imaging intratumoral microenvironment and is predictive of
17(2):241–255 patient outcome. Neuro Oncol 16(2):280–291
11. Jackson A, Li KL, Zhu X (2014) Semi-quantitative 24. LaViolette PS, Cohen AD, Prah MA, Rand SD,
parameter analysis of DCE-MRI revisited: monte- Connelly J, Malkin MG et al (2013) Vascular change
carlo simulation, clinical comparisons, and clinical val- measured with independent component analysis of
idation of measurement errors in patients with type dynamic susceptibility contrast MRI predicts
2 neurofibromatosis. PLoS One 9(3):e90300 bevacizumab response in high-grade glioma. Neuro
12. Jackson A (2004) Analysis of dynamic contrast Oncol 15(4):442–450
enhanced MRI. Br J Radiol 77(Spec No 2):S154–S166 25. LaViolette PS, Daun MK, Paulson ES, Schmainda KM
13. O’Connor JP, Jackson A, Parker GJ, Roberts C, Jayson (2014) Effect of contrast leakage on the detection of
GC (2012) Dynamic contrast-enhanced MRI in clinical abnormal brain tumor vasculature in high-grade gli-
trials of antivascular therapies. Nat Rev Clin Oncol oma. J Neurooncol 116(3):543–549
9(3):167–177 26. Liu Z, Liao H, Yin J, Li Y (2014) Using R2* values to
14. Tofts PS, Kermode AG (1991) Measurement of the evaluate brain tumours on magnetic resonance imag-
blood-brain barrier permeability and leakage space ing: preliminary results. Eur Radiol 24(3):693–702
using dynamic MR imaging. 1. Fundamental concepts. 27. Linnik IV, Scott ML, Holliday KF, Woodhouse N,
Magn Reson Med 17(2):357–367 Waterton JC, O’Connor JP et al (2013) Noninvasive
15 Imaging Biomarkers of Angiogenesis and the Microvascular Environment in Cerebral Tumors 325

tumor hypoxia measurement using magnetic resonance 38. Erdogan C, Hakyemez B, Yildirim N, Parlak M (2005)
imaging in murine U87 glioma xenografts and in Brain abscess and cystic brain tumor: discrimination
patients with glioblastoma. Magn Reson Med with dynamic susceptibility contrast perfusion-
28. Bruehlmeier M, Roelcke U, Schubiger PA, Ametamey weighted MRI. J Comput Assist Tomogr
SM (2004) Assessment of hypoxia and perfusion in 29(5):663–667
human brain tumors using PET with 39. Patankar TF, Haroon HA, Mills SJ, Baleriaux D, Buck-
18F-fluoromisonidazole and 15O-H2O. J Nucl Med ley DL, Parker GJ et al (2005) Is volume transfer
45(11):1851–1859 coefficient (K(trans)) related to histologic grade in
29. Takada Y, Ye X, Simon S (2007) The integrins. human gliomas? AJNR Am J Neuroradiol
Genome Biol 8(5):215 26(10):2455–2465
30. Desgrosellier JS, Cheresh DA (2010) Integrins in can- 40. Thompson G, Mills SJ, Stivaros SM, Jackson A (2010)
cer: biological implications and therapeutic opportuni- Imaging of brain tumors: perfusion/permeability. Neu-
ties. Nat Rev Cancer 10(1):9–22 roimaging Clin N Am 20(3):337–353
31. Haubner R, Kuhnast B, Mang C, Weber WA, Kessler H, 41. Yoon JH, Kim JH, Kang WJ, Sohn CH, Choi SH,
Wester HJ et al (2004) [18F]Galacto-RGD: synthesis, Yun TJ et al (2014) Grading of cerebral glioma
radiolabeling, metabolic stability, and radiation dose with multiparametric MR imaging and 18F-FDG-
estimates. Bioconjug Chem 15(1):61–69 PET: concordance and accuracy. Eur Radiol
32. Schittenhelm J, Schwab EI, Sperveslage J, Tatagiba M, 24(2):380–389
Meyermann R, Fend F et al (2013) Longitudinal 42. Law M, Young RJ, Babb JS, Peccerelli N, Chheang S,
expression analysis of alpha v integrins in human gli- Gruber ML et al (2008) Gliomas: predicting time to
omas reveals upregulation of integrin alpha v beta 3 as progression or survival with cerebral blood volume
a negative prognostic factor. J Neuropathol Exp Neurol measurements at dynamic susceptibility-weighted con-
72(3):194–210 trast-enhanced perfusion MR imaging. Radiology
33. Schnell O, Krebs B, Carlsen J, Miederer I, Goetz C, 247(2):490–498
Goldbrunner RH et al (2009) Imaging of integrin alpha 43. Di Costanzo A, Scarabino T, Trojsi F, Popolizio T,
(v)beta(3) expression in patients with malignant glioma Bonavita S, de Cristofaro M (2014) Recurrent glioblas-
by [18F] Galacto-RGD positron emission tomography. toma multiforme versus radiation injury: a
Neuro Oncol 11(6):861–870 multiparametric 3-T MR approach. Radiol Med
34. Liu S, Hsieh WY, Jiang Y, Kim YS, Sreerama SG, 119:616–624
Chen X et al (2007) Evaluation of a (99m)Tc-labeled 44. Sugahara T, Korogi Y, Tomiguchi S, Shigematsu Y,
cyclic RGD tetramer for noninvasive imaging integrin Ikushima I, Kira T et al (2000) Posttherapeutic
alpha(v)beta3-positive breast cancer. Bioconjug Chem intraaxial brain tumor: the value of perfusion-sensitive
18(2):438–446 contrast-enhanced MR imaging for differentiating
35. Li ZB, Cai W, Cao Q, Chen K, Wu Z, He L et al (2007) tumor recurrence from nonneoplastic contrast-
(64)Cu-labeled tetrameric and octameric RGD pep- enhancing tissue. AJNR Am J Neuroradiol
tides for small-animal PET of tumor alpha(v)beta 21(5):901–909
(3) integrin expression. J Nucl Med 48(7):1162–1171 45. Larsen VA, Simonsen HJ, Law I, Larsson HB, Hansen
36. Battle MR, Goggi JL, Allen L, Barnett J, Morrison MS AE (2013) Evaluation of dynamic contrast-enhanced
(2011) Monitoring tumor response to antiangiogenic T1-weighted perfusion MRI in the differentiation of
sunitinib therapy with 18F-fluciclatide, an 18F-labeled tumor recurrence from radiation necrosis. Neuroradi-
alphaVbeta3-integrin and alphaV beta5-integrin imag- ology 55(3):361–369
ing agent. J Nucl Med 52(3):424–430 46. Cao Y, Tsien CI, Nagesh V, Junck L, Ten Haken R,
37. Calli C, Kitis O, Yunten N, Yurtseven T, Islekel S, Ross BD et al (2006) Survival prediction in high-grade
Akalin T (2006) Perfusion and diffusion MR imaging gliomas by MRI perfusion before and during early
in enhancing malignant cerebral tumors. Eur J Radiol stage of RT [corrected]. Int J Radiat Oncol Biol Phys
58(3):394–403 64(3):876–885
Part IV
Stroke Imaging
Patterns of Ischemic Stroke: From
Lacunar to Territorial to Multiple 16
Embolic to Watershed Hypotensive

Jessica Rotman and Robert Zimmerman

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 329 Neuroimaging is integral to stroke diagnosis
and management. Ischemic stroke prognosis,
Etiologies of Ischemic Strokes . . . . . . . . . . . . . . . . . . . . . 330
risk of recurrence, and management decisions
Imaging Findings Associated with Ischemic are influenced by stroke subtype, which can be
Strokes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 332 categorized by anatomic distribution and caus-
CT Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 332
MRI Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 337 ative mechanism. Computed tomography
(CT) and multimodal magnetic resonance
Vascular Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341
imaging (MRI) facilitate the identification of
MR Perfusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341 the extent, etiology, and time of onset of ische-
Localization of Ischemic Strokes . . . . . . . . . . . . . . . . . . 344 mic strokes. Understanding the underlying
Anterior Territory Infarcts . . . . . . . . . . . . . . . . . . . . . . . . . . . 344 vascular anatomy and patterns of ischemic
Posterior Territory Infarcts . . . . . . . . . . . . . . . . . . . . . . . . 347 stroke enables physicians to localize diseased
Branch Occlusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 347 blood vessels and better personalize patient
Basilar Occlusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351 management.
Watershed Infarcts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353
Lacunar Infarcts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
Keywords
Ischemia • Lacunar infarct • Watershed infarct •
Anoxia and Hypoxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356
Anoxia • Hypoxia • Emboli • CT • MRI
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 359
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 360
Introduction

Stroke is a leading cause of mortality and morbid-


ity in the developed world. The goals of imaging
J. Rotman (*)
PGY-3 Diagnostic Radiology Resident, New York in the setting of acute stroke are to establish a
Presbyterian Hospital, Weill Cornell Medical Center, diagnosis as early as possible and to deduce accu-
New York, NY, USA rate information regarding the intracranial vascu-
e-mail: jessicarotman@gmail.com; jar9120@nyp.org
lature and brain perfusion to guide in the selection
R. Zimmerman of appropriate therapy.
Diagnostic Radiology Department, New York Presbyterian
The ability to determine the age of an
Hospital, Weill Cornell Medical Center, New York, NY,
USA ischemic stroke provides useful clinical
e-mail: rdzimmer@med.cornell.edu

# Springer Science+Business Media New York 2016 329


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_27
330 J. Rotman and R. Zimmerman

information for the patient, his or her family, and Table 1 Topographic clinical pattern of brain infarction
the medical team. The time window to administer according to modified Oxfordshire method [3]
intravenous tissue plasminogen activator is Type of infarct Clinical description
currently 4.5–6 h from the time when the patient Lacunar Acute stroke that includes one of the
was last seen to be normal [1, 2]. Different major recognized lacunar
syndromes: (1) pure motor, (2) pure
imaging characteristics associated with different sensory, (3) sensorimotor, (4) ataxic
stages of infarction significantly affect the deci- hemiparesis, (5) dysarthria (clumsy
sion of whether or not to administer thrombolytic hand syndrome)
therapy. Total anterior Ischemia in both deep and superficial
The various MRI patterns of acute ischemic circulation territories of the middle cerebral
artery characterized by higher
stroke, including topography, size, and multiplic- cerebral dysfunctions such as
ity, are essential factors that can reflect the most dysphasia, dyscalculia, visuospatial
likely mechanisms of origin. Leptomeningeal disorder, homonymous visual field
arteries comprise the terminal branches of the defects, ipsilateral motor, and/or
sensory deficit of at least two areas of
cerebral and cerebellar arteries, which penetrate the face, arm, and leg
the cortex and subjacent white matter. Infarcts Partial anterior Clinical syndrome includes only two
involving the leptomeningeal artery system are circulation of the three aforementioned
often described as territorial infarcts. Ischemic components, with higher cerebral
dysfunction alone or with more
infarcts secondary to hypoperfusion that occur at
restricted sensorimotor deficits than
the border zones between major arterial territories those classified as lacunar infarcts
are known as watershed infarcts, while deep cere- Posterior Ipsilateral cranial nerve palsy with
bral infarcts typically caused by hypertension are circulation contralateral motor and/or sensory
referred to as lacunar infarcts. In 1991, the deficit, bilateral motor and/or
sensory deficit, disorder of conjugate
Oxfordshire Community Stroke Project (OCSP) eye movement, cerebellar
proposed four subgroups of cerebral infarctions dysfunction without ipsilateral long-
based on presenting signs and symptoms tract deficit, or isolated
(Table 1). Patterns of brain lesions on MRI homonymous visual defect
according to a modified Oxfordshire method are Adapted with permission from Lippincott Williams and
Wilkins/Wolters Kluwer Health: Stroke, Adams HP Jr,
classified based on the anatomic distribution of
Bendixen BH, Kappelle LJ, Biller J, Love BB, Gordon
infarcts: (1) total anterior circulation infarcts, DL, Marsh EE 3rd, Classification of subtype of acute
(2) partial anterior circulation infarcts, (3) poste- ischemic stroke. Definitions for use in a multicenter clinical
rior circulation infarcts, (4) watershed infarcts, trial. TOAST. Trial of Org 10172 in Acute Stroke Treat-
ment [4], 1993
(5) centrum ovale infarcts, and (6) lacunar
infarcts [3].
In 1993, a multicenter clinical trial known as
TOAST, or Trial of ORG 10172 in Acute Stroke Etiologies of Ischemic Strokes
Treatment, classified ischemic strokes based on
their respective causative mechanisms: (1) large- Large-vessel disease is the culprit in approxi-
vessel atherosclerotic disease, (2) small-vessel mately 13 % of all patients who present with
atherosclerotic disease, (3) cardioembolism, first-time strokes [6]. Caused by atherosclerotic
(4) other determined etiologies, and debris and ulceration, large-vessel disease is
(5) undetermined of multiple possible etiologies defined as occlusion or stenosis of greater than
(Table 2) [4, 5]. 50 % of a major brain artery or branch cortical
In this chapter, the etiologies of ischemic artery. Large-vessel atherosclerosis leads to
stroke, characteristic imaging patterns associated infarction when there is hemodynamically signif-
with the evolution of ischemic strokes, and stroke icant narrowing, which is defined as a decrease in
categories based on causative mechanisms will be vessel lumen diameter by 50–70 % [7]. In such
reviewed. cases, however, it is also possible for blood flow to
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 331

Table 2 Stroke categories according to TOAST method Table 3 Sources of cardioembolism stratified by risk
[4, 5] [7, 9–12, 67]
1 Large-vessel disease High risk Mechanical prosthetic valve
2 Small-vessel disease Mitral stenosis with atrial fibrillation
3 Cardioembolism Atrial fibrillation
4 Other etiology Left atrial/atrial appendage thrombus
5 Undetermined or multiple possible etiologies Sick sinus syndrome
TOAST trial of ORG 10172 in acute stroke treatment Recent myocardial infarction (<4 weeks)
Left ventricular thrombus
Dilated cardiomyopathy
be preserved in the setting of collateral circulation
Akinetic left ventricular segment
involving the circle of Willis and leptomeningeal
Atrial myxoma
vessels. Conversely, complete internal carotid
Infective endocarditis
artery (ICA) occlusion in the neck may still Medium Mitral valve prolapse
cause cerebral infarctions in the setting of emboli, risk Mitral annulus calcification
which may be multiple and simultaneous or Mitral stenosis without atrial fibrillation
focally broken up to produce multiple infarctions. Left atrial turbulence
While 35 % of patients over the age of 50 have Atrial septal aneurysm
atherosclerotic changes in their cervical arteries, Patent foramen ovale
only one third will have symptomatic vascular Atrial flutter
disease [7]. Dissection of the carotid or vertebral Lone atrial fibrillation
artery should always be considered as a cause of Bioprosthetic cardiac valve
stroke in young otherwise healthy patients. Ische- Nonbacterial thrombotic endocarditis
mic infarction typically occurs as a result of distal Congestive heart failure
embolization from a cervical dissection rather Hypokinetic left ventricular segment
than occlusion and typically manifests on imaging Myocardial infarction (>4 weeks, <6
months)
several hours to days after the onset of the dissec-
Rovira [7] with kind permission from Springer Science and
tion, although may also be delayed for several Business Media
weeks.
Cardioembolism is the principal cause of
ischemic cerebrovascular disease. Cardiogenic probably secondary to atrial appendage embolism
embolism is responsible for approximately [11, 12]. On brain imaging, large cortical territo-
15–27 % of first-time strokes [6]. High and rial infarction is suggestive of a cardioembolic
medium risk sources of cardioembolism are sum- mechanism, especially if the patient does not
marized in Table 3. In those with large infarcts have ICA occlusion. Although simultaneous
without ICA occlusion, the frequency of acute infarctions are generally attributed to
cardioembolic disease is higher than in large cardioembolism, the majority of cases are gener-
infarcts with ICA occlusion or limited infarcts ally not caused by a proximal embolus from the
without ICA occlusion [8]. The incidence of heart or aortic arch, but rather by artery-to-artery
cardioembolism is greater in patients below the embolism or fragmentation of a proximal artery
age of 45, presumably due to a lower rate of embolus [7]. The median volume of infarcts sec-
atherosclerotic disease in this younger age ondary to cardiogenic embolism is greater than
group. Cardioembolic ischemic stroke is the twice that of infarcts caused by artery-to-artery
most common subtype associated with recurrent embolism [13].
strokes and is associated with the highest 1-month Although uncommon, acute stroke caused by
mortality and in-hospital mortality in comparison fat emboli is potentially life threatening. Acute
to other subtypes [9, 10]. ischemia in the presence of long bone or pelvic
Approximately 16 % of ischemic strokes are fractures, cardiac surgery, or joint replacement
associated with atrial fibrillation and 10 % are therapy should raise concern for fat embolism.
332 J. Rotman and R. Zimmerman

Fat emboli from epicardial or bone marrow fat can Imaging Findings Associated
pass through pulmonary capillaries without with Ischemic Strokes
shunting lesions and result in systemic emboliza-
tion, commonly to the brain. Patients typically Brain imaging can accurately localize an ischemic
present with the clinical triad of hypoxia, altered stroke lesion to a specific vascular distribution
mental status, and petechiae. Other signs that may [16, 17]. Identification of occlusion or stenosis
be present include tachycardia, fever, anemia, and of extracranial and/or intracranial arteries can
thrombocytopenia. delineate the ischemic nature of a lesion and
Small-vessel occlusion is characterized by one help determine its etiology – for example, whether
of the traditional lacunar syndromes without evi- it occurs as a consequence of slow flow, proximal
dence of cerebral or cortical dysfunction and is the embolic occlusion, or small-vessel disease.
cause of 25 % of all first-ever strokes [6]. Lacunes CT and MRI findings change rapidly through-
are thought to occur most often in patients with out the initial week after an infarct due to the
hypertension or diabetes [14]. In addition to athero- underlying pathophysiologic changes taking
sclerosis and lipohyalinosis, other potential causes place. Although CT is extremely useful for
of small-vessel occlusion include vasculopathy, detecting large ischemic infarcts of more than
artery-to-artery or cardiogenic embolism, arterial 6–8 h in duration, it does not reliably
dissection, and hypercoagulability-associated reveal infarcts of less than 4 h in duration or
thrombosis [15]. In patients with their first lacunar optimally characterize the extent of such
stroke, vascular imaging of the cervical and intra- infarctions. The goals of CT in the acute setting
cranial arteries for a cardiac source of embolism is include exclusion of intracranial hemorrhage or
indicated. involvement of greater than 33 % of the distribu-
Only 2 % of strokes are classified in the “other tion of the middle cerebral artery (MCA) territory,
etiology” category [6]. Strokes of other deter- which would preclude thrombolytic therapy, iden-
mined causes include non-atherosclerotic tification of any early features of infarction, and
vasculopathies, hypercoagulable states, and exclusion of other intracranial pathologies that
hematologic disorders, whereas strokes with neg- may mimic a stroke, such as a tumor.
ative or incomplete evaluations, or as a result of
two or more causes, are labeled strokes of
undetermined cause. CT Findings
Anoxic and hypoxic brain injuries occur when
there is decreased oxygen content in the blood. Within the first few hours of an ischemic stroke,
Anoxia results from near-complete absence of imaging features on CT include loss of
oxygen in the blood for more than 5 min, while gray–white matter differentiation and loss of the
hypoxia occurs when there is partial but more ability to differentiate the basal ganglia or thala-
prolonged hypoxemia [7]. Anoxia can occur mus from the internal capsule. Mass effect is
after cardiac arrest, prolonged seizures, strangu- typically absent. Areas of cortex with poor collat-
lation/hanging, near drowning, and smoke/car- eral blood supply, such as the insular ribbon
bon monoxide inhalation. Carbon monoxide (Fig. 1) are more vulnerable to ischemia [18] and
poisoning produces anoxic injury by preventing should be carefully inspected to detect early loss
oxygen from binding to hemoglobin. General of gray–white matter differentiation. The basal
sites of the brain susceptible to ischemia are ganglia should also be scrutinized for findings
those with increased metabolic rate and a paucity associated with hyperacute stroke; relative
of redundant blood supply, including the hippo- hypodensity in the lentiform nucleus (with loss
campus, globus pallidus (in particular in carbon of contrast difference to the adjacent white matter
monoxide poisoning), amygdala, cerebellum, in the internal capsule) can be seen in 75 % of
and occipital lobes, in decreasing order of patients at 3 h [18] (Fig. 2). The dense vessel sign
frequency. reflective of acute embolic occlusion can
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 333

phenomenon.” Administration of contrast will


demonstrate gyriform enhancement of the
“fogged” infarct area and can thus be performed
to prevent misinterpretation [19] (Fig. 5). Over the
weeks and months that follow, infarction gliosis
sets in, which appears as a region of low density
with focal volume loss (atrophy), such as in the
case of ex vacuo dilatation of the ventricles and
associated encephalomalacia (Fig. 6). Cortical
mineralization or laminar necrosis may also
occur, appearing as curvilinear hyperdensities fol-
lowing gyral anatomy.
Perfusion CT is superior in detecting the pres-
ence and extent of possible acute strokes that are
inconspicuous on unenhanced CT. It has emerged
as a critical tool for the selection of patients opti-
mal for reperfusion therapy, as it can identify the
penumbra surrounding the core of an infarct,
which is the region that is ischemic but has not
Fig. 1 Hyperacute M2/M3 MCA distribution infarct; yet undergone permanent infarction and may thus
insular ribbon sign. Axial CT of the brain demonstrates be possibly salvaged. The parameters associated
marked hyperdensity in the frontal and temporal lobes
with cerebral hemodynamics include cerebral
along the insular ribbon and cortex, consistent with acute
MCA distribution infarction blood flow (CBF), which represents instantaneous
capillary flow in tissue; cerebral blood volume
(CBV), which describes the blood volume of the
sometimes be seen on unenhanced CT and is cerebral capillaries and venules per cerebral tissue
helpful in the setting of normal-appearing acutely volume; the mean transit time (MTT), which mea-
infarcted parenchyma. Evaluation of CT angio- sures the length of time a certain volume of blood
gram source images may make acute infarction spends in the cerebral capillary circulation; and
more apparent because of absent or decreased the time to peak (TTP), which is inversely related
enhancement of hypoperfused gray matter to CBF in that reduction of blood flow results in an
(Fig. 3). increase in the time needed for contrast to reach its
Over the first week, hypoattenuation and peak in the perfused volume of brain tissue. These
edema associated with ischemia progressively parameters are linked by the central volume prin-
increase. In the case of a large infarct, substantial ciple which state that CBF = CBV/MTT. Specif-
mass effect is typical, often causing secondary ically, areas which demonstrate matched defects
damage to other areas of the brain, such as in in CBVor CBF and MTT represent unsalvageable
cases of herniation (Fig. 4). Contrast-enhanced infarct core, whereas those with prolonged MTT
scans may demonstrate parenchymal enhance- but preserved CBV are considered to be the ische-
ment, which is typically gyriform in cases of mic penumbra [20]. Recently the value of perfu-
cortical infarction. Approximately one week sion imaging in determining success of stroke
after the onset of cerebral infarction, the swelling intervention has come into question [21], leading
starts to subside and there are new cortical pete- some investigators to state that, given the impor-
chial hemorrhages and/or hyperemia, which result tance of time to treatment, a non-contrast head CT
in elevation of cortical attenuation (pseudonorma- is all that is needed to make a therapeutic decision
lization). In such cases, imaging of a stroke at this [22] (Fig. 2).
time can be misleading as the affected cortex will CT angiography (CTA) is utilized to
appear normal, known as the “CT fogging identify possible thrombus within an intracranial
334 J. Rotman and R. Zimmerman

Fig. 2 Hyperacute MCA infarct. (a) Axial CT of the brain artery and M1 segment of middle cerebral artery due to
demonstrates a dense distal right internal carotid artery and acute thrombosis. Note the normally enhancing patent left
M1 portion of the MCA, a sign of acute thrombus. Note the MCA and normal enhancement of right M2 branches is
normal less dense left MCA. (b) A more superior image consistent with collateral supply. (d) CT perfusion demon-
demonstrates loss of normal gray matter density in the strates decreased CBF and CBV and increased MTT
entire right MCA distribution. Note the relative corresponding to the subtle hypoattenuation seen on
hypodensity of the left and right sides of the ganglia and non-contrast CT. There is no mismatch
cortex. (c) The corresponding axial CTA of the brain dem-
onstrates a filling defect in the distal right internal carotid

vessel and guide intra-arterial thrombolysis or or carotid dissection, and identify any
clot retrieval. CTA can also be applied to the possible endovascular treatment access limita-
carotid and vertebral arteries in the neck to tions, such as arterial tortuosity or stenosis
establish stroke etiology, such as atherosclerosis (Figs. 2 and 3).
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 335

Fig. 3 MCA/ACA
distribution hyperacute
infarction. (a) Unenhanced
CT in a patient with atrial
fibrillation demonstrates
subtle loss of normal gray
matter density involving
lateral (MCA) and medial
(ACA) frontal cortex. (b)
On source images loss of
normal cortical gray density
is more apparent, reflecting
decreased blood volume in
the region of infarction.
Axial (c) and coronal (d)
MIP CTA images
demonstrate lack of
enhancement of the A1 and
M1 segments of the right
MCA, indicative of acute
thrombus and loss of
normal cortical density in
the MCA and ACA
distributions. Note the
presence of filling of some
right M2 and A2 branches
due to collateral supply
from the posterior
circulation

Fig. 4 Subacute “malignant” MCA infarct. (a) Axial CT lateral ventricle demonstrates marked hypodensity with
of the brain 3 days after an acute MCA territory stroke transfalcine herniation. Punctate foci of hyperdensity and
demonstrates marked hypodensity and mass effect with mild gyriform hyperdensity within the infarct are consis-
uncal herniation, as a result of increased vasogenic edema tent with a combination of reperfusion hemorrhage and
and intracranial pressure. (b) Image at the level of the hyperemia
336 J. Rotman and R. Zimmerman

Fig. 5 Subacute PCA P3


distribution infarct. (a)
Axial unenhanced CT of the
brain in a patient with a
subacute PCA territory
occipital lobe stroke, which
appears isodense to normal
gray matter due to
hyperemia and/or laminar
necrosis. (b) Contrast-
enhanced axial CT of the
brain demonstrated
gyriform enhancement that
confirms that non-enhanced
CT of the brain
demonstrated the “fogging
phenomenon”

Fig. 6 Chronic M2/M3


distribution infarct with
cystic encephalomalacia.
(a) Axial CT scan
demonstrates marked
hypodensity in the left
MCA distribution with ex
vacuo dilatation of the left
lateral ventricle. (b)
T1-weighted and (c) FLAIR
images demonstrate that
this region is fluid-filled
consistent with cystic
encephalomalacia. (d)
T2-weighted image at the
level of the mesencephalon
demonstrates marked
atrophy of the cerebral
peduncle consistent with
Wallerian degeneration
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 337

Table 4 Stages of ischemic stroke and their respective appearances on MRI


Stage Timing T1WI T2WI/FLAIR DWI ADC
Hyperacute 0–6 h Isointense, possible loss of Isointense to Hyperintense Hypointense
sulci mildly
hyperintense
Acute 6 h–4 Hypointense, mass effect Hyperintense Hyperintense Hypointense
days
Subacute 4–14 Hypointense Hyperintense Mildly hyperintense Isointense
days from T2 shine through
Chronic >14 Smaller area of Hyperintense Isointense Hyperintense
days hypointensity,
encephalomalacia
MRI magnetic resonance imaging, DWI diffusion-weighted imaging, ADC apparent diffusion coefficient

MRI Findings triphosphate, which in turn leads to breakdown of


normal cell membrane integrity. As a conse-
The acuity or chronicity of a stroke is better elu- quence, there is intracellular swelling and damage
cidated on MRI than CT due to different time- to intracellular proteins causing increased intra-
sensitive signal characteristics. The typical MRI cellular viscosity. These processes are indicators
findings that may help determine the relative ages of cell death. After 4–6 h T2 increases within the
of cerebral infarcts are displayed in Table 4. MRI area of infarction due to vasogenic edema.
is a much more sensitive modality than CT in the Restricted diffusion can be assessed independent
identification of hyperacute infarction (<6 h) of T2 by evaluating apparent diffusion coefficient
[23]. In the first 3 h after infarction, CT does not (ADC) maps. Restricted diffusion usually peaks at
typically demonstrate any parenchymal abnor- around 3 days and disappears by 5–10 days
malities [23, 24]. T2-weighted fluid attenuation (pseudonormalization) [27, 28]. Thereafter diffu-
inversion recovery (FLAIR) sequences are also sion increases in the area of infarction due to
relatively insensitive during this hyperacute removal of dead cells and development of gliosis.
period (and are less sensitive than CT in the Diffusion restriction and increased T2 combine to
3–6 h range) but the overall sensitivity of MR is produce hyperintensity on DWI images that lasts
82 % in the first 24 h, whereas detection rates for for up to 2 weeks after infarction. In circum-
acute infarction on CT are 58 % in the first 24 h stances where diffusion is equal to normal brain
[25]. Even more impressive is the 99 % sensitivity but T2 is increased, such as in the case of a
rate of diffusion-weighted imaging (DWI), asso- subacute infarction, the tissue will appear bright
ciated with detection of an ischemic stroke within on DWI and isointense on apparent diffusion
12 h of its onset (including the first 1 h), in coefficient (ADC) maps, a phenomenon known
contradistinction to the 39 % sensitivity rate asso- as T2 shine through (Fig. 9).
ciated with CT [26]. In addition, causes of stroke DWI nearly doubles the likelihood of detecting
such as venous thrombosis, vascular acute ischemic strokes compared to FLAIR
malformations, infections, and tumors are better [29]. Although T2-weighted imaging and FLAIR
identified and characterized with greater accuracy are less sensitive in detecting parenchymal change
on MRI than is possible with CT. in the first few hours, loss of normal flow voids in
Described as the “light bulb sign,” large arteries may be visible immediately
hyperintensity associated with acute infarction [30]. The MRI correlate of the early CT dense
usually occurs within 3 h and is easily detected vessel sign in acute embolic occlusion is
on DWI [7] (Figs. 7 and 8). This marked intraluminal hypointensity without associated
hyperintensity is initially due to diffusion restric- flow-related hyperintensity on gradient echo or
tion in the area of cerebral ischemia. Cytotoxic susceptibility-weighted images. Normally, large
edema occurs as a result of depletion of adenosine vessels are centrally hypointense with associated
338 J. Rotman and R. Zimmerman

Fig. 7 Hyperacute PCA P2 distribution infarct on MR FLAIR mismatch has been proposed as an indicator of
(<4 h); FLAIR/DWI mismatch. (a) Hyperintensity on hyperacute infarction when the clinical timing of infarction
DWI and (b) relative isointensity on FLAIR in the occipital is uncertain (e.g., “wake-up strokes”). In many of these
lobe and hippocampus. The presence of the diffusion- cases, CT will be positive before FLAIR

Fig. 8 Acute infarct internal capsule – anterior choroidal diffusion. (d) FLAIR reveals subtle hyperintensity indica-
distribution. (a) CT reveals ill-defined hypodensity in left tive of acute infarct. (e) DWI at the level of the cerebral
posterior limb of internal capsule consistent with infarct of peduncle reveals hyperintensity indicative of acute
indeterminate age. (b) DWI reveals marked hyperintensity. Wallerian degeneration
(c) ADC map reveals hypointensity indicative restricted
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 339

Fig. 9 Subacute thalamic infarct – T2 shine through. (a) hypointensity indicative of T2 shine through and not
Axial DWI MRI of the brain demonstrates hyperintensity restricted diffusion. (c) FLAIR MRI demonstrates marked
in the medial thalamus imparting mild mass on the third hyperintensity corresponding to region of hyperintensity
ventricle. (b) ADC map demonstrates absence of on DWI sequence

hyperintensity adjacent to the vessel due to signal of infarction, the extent of the DWI abnormality
from spatially displaced blood secondary to flow remains relatively unchanged (unless there has
effects. In the presence of acute clot, however, been interval progression of the infarct)
there is marked hypointensity that often extends (Fig. 10). In 50 % of cases, contrast enhancement
beyond the lumen of the vessel, referred to as can be seen [2]. Enhancement can be intra-arterial
“blooming,” and there is no corresponding flow- due to slow flow, in which case it can be seen at
related hyperintensity [7] (Fig. 10). the time of vascular occlusion. Subsequently tran-
Hyperacute infarction is T1 isointense and T2 sient leptomeningeal enhancement may occur and
isointense to mildly hypertense, which is best usually resolves by the end of the first week.
appreciated on FLAIR and is usually confined to Parenchymal enhancement due to blood–brain
the gray matter in cases of thromboembolic infarc- barrier damage and ingrowth of new immature
tion. It typically takes 4–5 h for hyperintensity to vessels may occur by the end of the first week
develop on FLAIR, which is actually less sensi- and last for several weeks [2].
tive than CT in the 3–6 h period [7]. The presence Blood flow to the affected portion of the brain
of restricted diffusion and normal signal on is typically reestablished 24–72 h after infarction,
FLAIR (diffusion-FLAIR mismatch) may poten- during the early subacute phase. By day 3 or
tially be used to determine the onset of infarction 4, new ingrowth of immature vessels with more
in patients in whom the time on onset is unknown permeable blood–brain barriers result in increased
(referred to as “wake-up” stroke), potentially vasogenic edema, which is extracellular swelling
allowing for therapeutic intervention in these indi- caused by leakage of fluid out of capillaries. In
viduals [31] (Fig. 7). large infarcts, mass effect typically peaks at
After 6–12 h, infarcted tissue becomes high around 5 days and can lead to transfalcine or
signal on T2-weighted images, while low inten- transtentorial herniation. Infarcted brain is mildly
sity on T1-weighted images typically mirrors this T1 hypointense and markedly T2 hyperintense,
high T2/FLAIR signal. In thromboembolic involving both gray and white matters with
infarcts, T2 hyperintensity is confined to the ill-defined margins. Intensity on DWI and ADC
gray matter. Although the extent and degree of maps is variable at this stage, reflecting the degree
T2 hyperintensity increase during the acute phase of cytotoxic edema (decreased ADC) and
340 J. Rotman and R. Zimmerman

Fig. 10 Subacute deep and superficial MCA infarct sec- diffusion. (e) FLAIR reveals hyperintensity and mass
ondary to carotid occlusion. (a) SWI image demonstrates effect in the entire right MCA distribution consistent with
hypointensity in the M1 segment of the right MCA. Note infarct of 2–3 days duration. (f) T1 reveals mild cortical
the normal hyperintense flow effects within the left MCA. hypointensity and mass effect with gyral swelling and
(b) MRA reveals absence of flow in the distal right internal sulcal obliteration. These findings are typical for an infarct
carotid artery and MCA. (c) DWI reveals marked after 3–5 days
hyperintensity. (d) ADC reveals marked restricted

vasogenic edema (increased ADC) (Figs. 10 and 11). and T2 hyperintensity persist into the late sub-
If the infarct involves the corticospinal tract, acute and chronic healing phases. Cortical laminar
Wallerian degeneration may occur and manifest as necrosis may be visualized as a ribbon of intrinsic
mild T2 hyperintensity and restricted diffusion T1 hyperintensity and is usually apparent after
acutely (Fig. 8). In such cases, signal abnormality 2 weeks [2] (Fig. 12). Cystic encephalomalacia
will be present in the ipsilateral cerebral peduncle and and lacunar infarcts appear as central regions of
pons and should not be mistaken for an additional T1 hypointensity, T2 hyperintensity, and FLAIR
area of infarction. In chronic infarcts Wallerian hypointensity (similar to spinal fluid intensity)
degeneration is manifested by volume loss in the surrounded by T2 hyperintensity and are best
ipsilateral cerebral peduncle without signal abnor- seen on FLAIR imaging (Fig. 6). On DWI,
mality (Fig. 6d). chronic infarction is isointense to mildly
Within 5–14 days, DWI demonstrates hypointense, while the infarct appears
isointensity to mild hyperintensity, while ADC hyperintense on ADC maps due to increased dif-
maps display hyperintensity reflective of fusion in the hypocellular infarcted brain
increased diffusion [7] (Fig. 9). T1 hypointensity (Fig. 12). If contrast is administered, parenchymal
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 341

Fig. 11 Malignant MCA infarct MR. (a–c) FLAIR marked mass effect with early medial displacement of the
images reveal an infarct involving the majority of the uncus (a) and mass effect upon and displacement of the
deep and superficial distributions of the MCA. Note the lateral ventricle (b and c)

enhancement usually begins near the end of the segments). Dissections of the extracranial internal
first week and lasts less than 12 weeks – if it lasts carotid artery produce four patterns of luminal
longer, the presence of an underlying lesion abnormality: smooth tapering to a pointed occlu-
should be considered [2]. sion, long segment asymmetric narrowing, double
lumen with an intimal flap, and pseudoaneurysm
formation [7]. CTA allows for the detection of
Vascular Imaging intimal flaps and double lumens and MRA is
excellent at identifying carotid dissection. MRI
Arterial and venous occlusion or slow flow can be imaging of the neck in particular with
visualized on FLAIR and enhanced T1-weighted fat-suppressed T1-weighted imaging can identify
images. Chronic vessel occlusion or extremely the intramural hematoma that is the direct result of
slow flow in large vessels manifests with the dissection (Fig. 13).
isointensity to hyperintensity on T1-weighted
images (Fig. 13) and hyperintensity on FLAIR
because the intrinsic hyperintensity of blood can MR Perfusion
be “captured” due to the absence of flow effects. If
contrast is administered, intraluminal Perfusion imaging can be performed with MR as
hyperintensity is more extensive distal to an well as CT. As with CT the brain is imaged as a
occlusion than in normal circulation because the bolus of contrast is being injected. The contrast
intrinsic intensity of the enhanced blood is detect- agent is paramagnetic so it causes intensity in the
able secondary to absent flow effects. brain to decrease proportionate to the amount of
Direct visualization of vascular abnormalities contrast in the vascular system. Whole brain cov-
(stenosis occlusion and dissection) can be erage is routinely obtained (as opposed to CT
obtained with CTA, magnetic resonance angiog- perfusion). The perfusion parameters are similar
raphy (MRA), and catheter angiography. The lat- to those obtained from CT, but it is not possible
ter is usually reserved for cases where intervention (or at least not easy) to obtain MTT measurements
is contemplated or when diagnosis is in doubt. and therefore quantification is difficult. However,
Atherosclerotic narrowing and occlusion occur since the perfusion diffusion mismatch is qualita-
in typical locations (e.g., carotid bifurcations in tively visible, there is no need for quantification to
the neck, M1 segments of middle cerebral delineate the core infarct and the penumbra
arteries, vertebral artery origins, and intracranial (Fig. 13).
342 J. Rotman and R. Zimmerman

Fig. 12 Chronic infarcts. (a) T1-weighted image demon- hyperintensity is due to pseudolaminar necrosis. (e) In an
strates cortical hyperintensity and atrophy in the M2 dis- example of a left chronic lacunar infarction, FLAIR images
tribution of the MCA. (b) FLAIR demonstrates cortical demonstrate hyperintensity. T1-weighted images would
and subcortical T2 hyperintensity and volume loss. (c) demonstrate corresponding minimal hypointensity. (f)
Gradient echo scan demonstrates no evidence of suscepti- Lacunar infarction is isointense on DWI and (g)
bility hyperintensity to suggest chronic hemorrhage. (d) T1 hyperintense on ADC map
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 343

Fig. 13 Carotid dissection with slow flow and large per- hyperintense clot in the vessel wall. (d) DWI image dem-
fusion diffusion mismatch. (a) T1- and (b) T2-weighted onstrates minimal restricted diffusion in the left corona
images demonstrate the presence of peripheral radiata (apparent hyperintensity overlying right hemi-
hyperintensity surrounding central hypointensity in the sphere is artifactual). (e) Contrast-enhanced T1-weighted
left internal carotid artery at the skull base (“target sign”) image demonstrates enhancement of MCA branches due to
representing intramural clot surrounding the narrow but slow flow. (f) MR perfusion demonstrates marked prolon-
patent artery. (c) MRA demonstrates the hyperintense gation of mean transit time and (g) moderate decrease in
flow with the lumen surrounded by more amorphous cerebral blood flow
344 J. Rotman and R. Zimmerman

Localization of Ischemic Strokes artery and its supply to the central artery of the
retina can result in blindness.
The intracranial circulation involves paired inter- Acute infarct involving the anterior choroidal
nal carotid arteries and vertebral arteries that arteries, which arise from the ICA, may involve
merge to form the basilar artery. An anastomotic the medial temporal cortex, posterior limb of the
network connects the carotid and vertebrobasilar internal capsule, posterior and medial aspect of
systems at the circle of Willis provides defense the lentiform nucleus, body of the caudate
against major-vessel occlusive disease. However, nucleus, and periventricular white matter
collateral circulation is more sparse distal to the [27–30] (Figs. 8 and 14). Patients with anterior
circle of Willis, which makes these areas more choroidal artery occlusion most commonly pre-
susceptible to infarction. It is thus important to sent with contralateral hemiplegia [29, 31]. Left
understand the vascular anatomy and recognize lesions may be the source of aphasia, constructive
territorial arterial supply to localize blood vessels apraxia, and amnesia [32, 33], while right lesions
with atherosclerotic disease/thrombosis. could be responsible for spatial neglect
[34–36]. Other possible manifestations include
impaired impulses, prosopagnosia, blindness
Anterior Territory Infarcts (optic tract and radiations), and paralysis (globus
pallidus and posterior limb of the internal cap-
The internal carotid artery (ICA) has cervical, sule). Large AchA territory ischemic lesions are
petrous, lacerum (or pre-cavernous), and cavern- more associated with embolic pathologies and
ous portions prior to piercing the dura to enter the portend worse prognoses than small AChA
intracranial space. The remainder of the internal strokes [37]. MRI findings include FLAIR and
carotid is defined by the clinoid, ophthalmic, and T2-weighted hyperintensity and acute restricted
supraclinoid segments. Branches of the ophthal- diffusion. Localization of small choroidal terri-
mic artery anastomose with those from the facial tories is easily missed on CT and in such cases
and internal maxillary arteries to provide a rich MRI with DWI proves very useful (Fig. 8).
network of collateral supply should the proximal In the setting of multiple small or subcortical
carotid artery become occluded. The ophthalmic infarcts within the same arterial territory, fragmen-
artery supplies the orbit, globe, frontal scalp tation of an embolus near its origin or within a
region, frontal and ethmoidal sinuses, and upper major proximal intracranial artery is implicated.
part of the nose. Occlusion of the ophthalmic The main cause of acute multiple brain infarctions

Fig. 14 Anterior choroidal distribution infarction. (a–c) Axial FLAIR images demonstrate acute infarct involving the
uncus, hippocampus, posterior thalamus, and posterior limb of internal capsule
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 345

in one cerebral hemisphere is typically MCA


and/or ICA disease (75 %) [6]. Nearly half of all
patients with ICA occlusion have territorial
infarction, although approximately 20 % of high-
grade symptomatic ICA territorial strokes are
associated with cardioembolic or lacunar
etiologies [6].
The ICA terminates at the bifurcation of the
anterior and middle cerebral arteries. Isolated
anterior cerebral artery (ACA) infarcts are rare
causes of ischemic stroke because extracranial
embolic material preferentially enters the middle
cerebral artery and the anterior communicating
artery protects the ACA distribution in the case
of cervical carotid occlusion. Isolated ACA distri-
bution infarction typically occurs as a result of
severe subfalcine herniation or intrinsic arterial
disease associated with occlusion, such as in
patients with diabetes, hypertension, vasospasm,
or vasculitis.
Fig. 15 ACA distribution infarction. FLAIR image dem-
onstrates an acute infarct in the right medial frontal lobe.
ACA Territory Note the hyperintensity in the ACA branches within the
The first portion of the ACA is termed the A1 infarction indicative of slow flow
segment, which turns along the genu of the corpus
callosum before forming the A2 segment. The function and sensation (medial precentral and
medial lenticulostriate arteries arise from the A1 postcentral gyri, respectively), memory, and emo-
segment to supply the anterior inferior basal tion (cingulate gyrus) [7].
ganglia and anterior limb of the internal capsule; On CT, ischemia or infarction in part or all of
specifically, a named branch called the recurrent the ACA vascular territory manifests with loss of
artery of Heubner supplies the head of the caudate gray–white matter differentiation (Fig. 3). Perfu-
nucleus, putamen, and anterior limb of the internal sion CT demonstrates decreased CBF/CBV ratio
capsule. A2 segment branches, such as the and increased MTT/TTP in the medial cerebral
pericallosal and callosomarginal arteries, primar- hemisphere. T2/FLAIR MRI exhibits medial
ily supply the corpus callosum and terminate as hemispheric hyperintensity, while DWI demon-
the splenial and cuneal arteries, respectively strates acute diffusion restriction and
(Fig. 15). corresponding hypointensity is present on the
The anterior communicating artery (ACoA) ADC map. Artery of Huebner infarcts produce
connects the ipsilateral A1 and contralateral A1 changes in the caudate head, anteriomedial puta-
segments of the ACA and should also always be men/globus pallidus, and anterior limb of the
evaluated for aneurysm. The ACA supplies the internal capsule. A2 distribution infarcts produce
medial anteroinferior frontal lobe, the anterior two changes in the parasagittal frontal cortex. There
thirds of the medial hemisphere surface, and a may be posterior extension into the parietal
variable amount of the cortical convexities. parasagittal region as well depending on the sup-
Since distal branches of the ACA supply the ply to this region by the posterior cerebral artery.
olfactory bulbs and tracts, corpus callosum, and
medial aspects of the frontal and parietal lobes, MCA Territory
infarctions affect olfaction, thought processes Infarcts involving the anterior territory are mostly
(medial inferior frontal lobe), lower limb motor related to the middle cerebral artery (MCA). MCA
346 J. Rotman and R. Zimmerman

occlusion on CT manifests as hypodensity involv- Patients with large MCA infarcts demonstrate
ing the frontal, parietal, and temporal lobes and/or higher frequencies of clinical deterioration and
basal ganglia and can occasionally be identified poor prognoses, frequently with high mortality
by the hyperdense vessel sign, reflective of inter- rates. Large MCA infarctions are associated with
nal thrombus (Figs. 2 and 10). The MCA cortical cardioembolism, ICA occlusion, and ICA
hemispheric supply includes the anterior temporal dissection.
lobe, insular, perisylvian, pre- and post-Rolandic Acute extracranial carotid occlusion may pro-
cortex. duce large areas of infarction involving the deep
The horizontal portion of the MCA is the M1 basal ganglia and cortical MCA distribution; these
segment, which gives off lateral lenticulostriate infarcts are typically the result of large distal
perforators to supply the putamen, globus emboli associated with proximal occlusion
pallidus, superior internal capsule and the adja- (Fig. 10). In such cases, the ACA territory is
cent corona radiata, and body and posterior head typically spared because of collateral supply
of the caudate. In cases of lateral lenticulostriate from the contralateral ACA with the ACoA of
infarctions, damage of the internal capsule typi- the circle of Willis. The ACA distribution may
cally results in hemiparesis and contralateral sen- be involved if the contralateral A1 segment of
sory deficits. Speech can be affected when the left ACA is absent or hypoplastic.
frontal and temporal lobes are involved, resulting Malignant MCA infarction is a life-threatening
in global aphasia due to cut off blood supply to complete or nearly complete MCA infarction
Broca (frontal) and Wernicke (superior temporal) associated with an 80 % mortality rate [6]
speech areas. Vision may be compromised by (Figs. 4 and 11). This type of infarction occurs in
contralateral hemianopsia/upper quandran- up to 10 % of all stroke patients and is uniformly
tanopsia if the optic radiations that sweep from characterized by clinical deterioration within the
the lateral geniculate nucleus to Meyer’s loops in first 2–3 days after stroke [6]. In the early phase of
the temporal lobes are involved. large territory infarction, DWI can accurately pre-
The M2 segment refers to the sylvian segment dict transformation into malignant MCA infarc-
of the MCA distal to its branching off into the tion (MCA volume >145 cm3) with 100 %
anterior division, posterior division, and anterior sensitivity and 94 % specificity [32] in patients
temporal artery. The distal branches of the MCA with persistent arterial occlusion and signs of total
course lateral to the insula and loop around the anterior circulation infarction. The main cause of
frontal operculum to form the “candelabra” effect death is severe post-ischemic brain edema, which
of the sylvian triangle and then become the corti- leads to increased intracranial pressure; hence,
cal M3 segment. The last branch of the MCA early detection is important since treatment with
sylvian vessels is the angular artery, which sup- hypothermia and/or hemicraniectomy can signifi-
plies the angular gyrus just beyond the sylvian cantly reduce mortality [33].
fissure (Figs. 1 and 12). Limited infarcts only involve one of the three
Infarction of the MCA may affect motor and MCA territories and are rarely associated with clin-
sensory functions of the face, arm, and trunk ical deterioration. These infarcts are caused by
(lateral precentral and postcentral gyri, respec- large-artery atherosclerosis and cardioembolism
tively), speech (inferior lateral frontotemporal in equal numbers. Since cardiac thrombi are
gyri), thought processes (anteroinferolateral fron- generally larger than thrombi of the large vessels,
tal lobes), hearing (superior temporal gyri), mem- there is a lower incidence of cardioembolism in
ory and naming of objects (temporal lobe), and limited MCA infarcts in contrast to large
taste (insular cortex) [7]. infarcts [34].
Large infarcts (Figs. 2, 4, 10, and 11) are
defined as those covering at least two of the Combined ACA/MCA
three MCA territories, including the deep, super- Involvement of both the ACA and MCA terri-
ficial anterior, and superficial posterior regions. tories is rare and typically fatal. These infarcts
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 347

are typically cardioembolic and occur in the set- As the V3 segment travels from the atlas to pierce
ting of atrial fibrillation. Large clots typically fill the dura to enter the intracranial compartment via
the distal internal carotid artery and A1 and M1 the foramen magnum, posterior meningeal
segments of anterior and middle cerebral arteries, branches are given off. In the intracranial com-
respectively. Combined ACA/MCA infarcts may partment where it is known as the V4 segment, the
also occur in the context of cervical carotid occlu- first branch is the PICA (although the PICA may
sion in the absence of the A1 segment of the ACA arise from the V3 extracranial segment in a sig-
contralateral to the carotid occlusion, thereby nificant number of patients).
eliminating protective effects of the Circle of The PICA supplies the posterolateral medulla,
Willis (Fig. 3). inferior cerebellar vermis, and inferior aspect of
the cerebellum. The inferior vermian artery and
tonsilohemispheric arteries are the terminating
Posterior Territory Infarcts branches of the PICA. Distal vertebral artery
and/or PICA occlusion can result in lateral med-
Proximal disease of the vertebral artery is the most ullary (Wallenberg) syndrome (Fig. 16), which
common finding in large-artery disease involving causes loss of pain and temperature sensation on
the posterior circulation. Vessels that are most the contralateral side of the body (lateral
frequently thrombosed by intra-arterial embolism spinothalamic tract) and face on the ipsilateral
are the intracranial vertebral artery and distal bas- side (descending trigeminothalamic tract), ataxia
ilar artery, leading to infarctions of the posterior (inferior cerebellar peduncle), ipsilateral
inferior cerebellar (PICA), superior cerebellar swallowing and taste disorders (CN IX), hoarse-
(SCA), and posterior cerebral arteries (PCA), ness and ipsilateral paralysis of the vocal cord
respectively. Posterior circulation infarcts are (CN X), vertigo and nystagmus (CN VIII), and
associated with severe brain atrophy, compatible ipsilateral Horner’s syndrome (hypothala-
with longstanding hypoperfusion. mospinal tract) [36–41]. As the name implies,
Posterior circulation infarcts can involve indi- the lateral medullary syndrome infarcts involve
vidual vertebrobasilar branches, perforating the lateral medulla and inferior cerebellum. The
branches or large portions of the posterior extent of infarction associated with a PICA occlu-
circulation. sion is quite variable given the reciprocal size of
PICA and AICA. It is common to have a dominant
PICA or AICA (with the other vessel being small
Branch Occlusions or absent). As a result, the vascular territories of
these vessels demonstrate considerable overlap,
The vertebral arteries arise from the subclavian and occlusion of either vessel can lead to infarc-
arteries and course superiorly before entering the tion in the same portions of the brain stem and
vertebral canal at the C6 transverse foramen (prior cerebellum [42] (Fig. 17). Given the variability of
to which they are known as V1 segments). The PICA and AICA, identification of an occluded
arteries then continue upward in the vertebral vessel may be difficult. This may be particularly
foramina (where they are referred to as V2 seg- challenging when there is a nondominant verte-
ments) before exiting at the C1–C2 level. In 75 % bral artery that does not appear to connect with the
of cases, the left vertebral artery is larger than the basilar artery. It is common for a nondominant
right and is known as the “dominant” artery [35]; vertebral artery to end in PICA.
thus, in cases of posterior fossa aneurysm or bas- Non-visualization of the V4 segment of the verte-
ilar artery stenosis, the left side should be care- bral artery in the absence of an ipsilateral PICA
fully evaluated. Branches of the vertebral artery should raise suspicion for a V4 and PICA
include muscular arteries to the neck, occipital occlusion.
artery, segmental spinal arteries, and anterior spi- AICA infarcts are less common than PICA
nal artery, which supplies most of the spinal cord. infarcts. The AICA arises laterally from the
348 J. Rotman and R. Zimmerman

Fig. 16 Lateral medullary infarct in PICA distribution. (a) distribution of the posterior inferior cerebellar artery. (c)
DWI and (b) FLAIR images demonstrate an acute infarct MRA demonstrates occlusion of the V4 segment of the
in the posterior lateral medulla and posterior medial infe- vertebral artery
rior cerebellum (lateral medullary syndrome) in the

Fig. 17 PICA/AICA distribution infarct. (a) DWI and (b) brain stem. (c) Follow-up CT scan 24 h later reveals devel-
T2-weighted images reveal a large acute infarct involving opment of hydrocephalus due to compression of the fourth
the right inferior cerebellum. Note the compression of the ventricle

basilar artery to supply the inferolateral pons and (CN VII), ipsilateral hearing loss and vertigo and
upper medulla via perforators as well as a small nystagmus (CN VIII), Horner’s syndrome
region of the petrous surface of the cerebellum, (hypothalamospinal tract), and ataxia (middle
the brachium pontis, and the fourth ventricle fora- and inferior cerebellar peduncles) [36,
men of Luschka and choroid plexus. After the 42–44]. With acute AICA strokes, MRI demon-
AICA loops in the auditory canal, it gives rise to strates diffusion restriction and hyperintensity
a labyrinthine branch, which supplies the inner ear involving the inferolateral cerebellum and pons.
cranial nerves VII and VIII then continues on to MRA or CTA may be used to evaluate the vessels.
supply the anteroinferior aspect of the cerebellum. While isolated unilateral AICA infarcts are likely
Patients with AICA infarcts may present with due to small artery atherosclerotic disease in dia-
lateral inferior pontine syndrome, involving con- betic patients, more widespread infarctions that
tralateral loss of pain and temperature include AICA territory are typically due to basilar
(spinothalamic tract), ipsilateral facial paralysis artery occlusive disease.
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 349

Fig. 18 Pontine perforator


infarct. (a) FLAIR imaging
reveals an infarct in the left
pons. Note that the infarct
stops abruptly in the
midline reflecting the
vascular supply to the pons.
(b) MRA reveals
atherosclerotic irregularity
of the mid-basilar artery

Isolated superior cerebellar artery infarcts are occipital or inferior temporal lobes including the
rare. The vessels are relatively large and often hippocampi, as well as regions supplied by the
duplicated. When they occur there is involvement small premammillary, posterior thalamoperforate,
of the cerebellum hemisphere above the great thalamogeniculate, and peduncular perforating
horizontal fissure and the superior vermis. The arteries. These branches arise from the P2 seg-
SCA is the last infratentorial branch off the basilar ment to supply the thalamus, hypothalamus,
artery and supplies the upper aspect of the cere- geniculate bodies, posterior limb of the internal
bellum prior to terminating as the superior capsule, upper midbrain, and choroid plexus. The
vermian artery. Fine SCA branches help supply PCA splenial branch supplies the splenium and
the pons, superior cerebellar peduncle, and infe- dorsal corpus callosum. There is variable supply
rior colliculus. The superior vermian vessel anas- to the parietal parasagittal cortex. The extent of
tomoses with the inferior vermian artery of the infarction depends upon the site of obstruction.
PICA. Predominantly caused by embolism, With complete occlusion of the PCA, there may
infarcts in the territory of the SCA are uncommon be involvement of the entire PCA distribution,
[45]. SCA occlusion can cause ipsilateral jaw including the upper brain stem, thalamus, hippo-
weakness and facial numbness (spinothalamic campi, posterior inferior temporal-occipital lobes,
tract and CN V), with the jaw deviating toward paramidline occipital lobes (including the
the lesion due to unopposed action of the contra- calcarine cortex), and parietal lobes (Fig. 19).
lateral medial pterygoid muscle. More distal occlusions will produce cortical
Infarcts commonly arise from pontine perforat- occipital and parietal infarcts but will spare the
ing branches of the basilar artery. The pontine brain stem and thalamus (Figs. 5 and 7).
perforators are paired paramidline structures that Although the posterior thalamoperforate arter-
extend posteriorly into the brain stem. Pontine ies are usually paired, the artery of Percheron
perforator infarcts therefore present as unilateral (a variant where there is bilateral supply to the
paramidline lesions that extend posteriorly along posterior superior brain stem and medial thala-
the course of the vessel into the posterior pons mus) and the thalamogeniculate branches arise
(Fig. 18). These are essentially lacunar infarcts from the proximal PCA (P2 segment), the poste-
(see below). rior communicating artery (PCoA) may contribute
The basilar artery terminates in two PCAs as minimal supply to these areas as well (Fig. 20).
well as a few small perforating vessels arising Occlusion of these vessels may affect memory
from its vertical dome. The PCA distribution and emotion (anterior thalamus), endocrine func-
includes the medial posterior third of the cerebral tion (hypothalamus), language (pulvinar), pain
hemispheres and infarcts commonly involve the sensation (thalami), sight (lateral geniculate),
350 J. Rotman and R. Zimmerman

Fig. 19 Basilar tip


occlusion. (a–c) FLAIR
image reveals an infarct
involving the superior brain
stem, right hippocampus,
and bilateral thalami. (d)
MRA reveals occlusion of
the distal basilar artery.
There is normal flow in the
P2 segment of the right
posterior cerebral artery due
to collateral supply from the
posterior communicating
artery

Fig. 20 Artery of
Percheron infarct. (a) DWI
and (b) FLAIR images
reveal bilateral medial
thalamic infarcts in the
distribution of the artery of
Percheron

and motor control (subthalamic nuclei). The P1 mammillary bodies. It is important to note
segment of the PCA joins the anterior circulation that PCA ischemia may arise from anterior circu-
via the PCoA. The PCoA thus connects the ante- lation emboli if PCA of fetal origin is present and
rior carotid circulation with the posterior that the PCoA allows emboli from both anterior
vertebrobasilar circulation, supplying parts of the and posterior circulations to affect PCA
thalamus, hypothalamus, optic chiasm, and distribution.
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 351

The PCA continues in its P3 and P4 distal Basilar Occlusion


segments, giving rise to anterior and posterior
temporal arteries and a posterior pericallosal Occlusion of the basilar artery produces bilateral
artery prior to terminating as parieto-occipital infarction in all vascular territories superior to the
branches and calcarine arteries to supply the distribution of the PICA. Depending on the com-
occipital lobe. PCA infarction most commonly petency of the posterior communicating arteries,
affects vision (occipital lobes) but can also affect there may be partial or complete unilateral or
memory (posteroinferior temporal lobe), smell bilateral sparing of the posterior cerebral artery
(hippocampal region), and emotion (posterior distribution. The distal basilar artery provides per-
fornix). forators of rostral pontine, median, paramedian, and
Occlusion of the PCA can result in medial lateral pontine branches and generates the SCA
midbrain (Weber) syndrome, characterized by prior to terminating as PCA branches. Pontine
contralateral spastic paresis (corticospinal artery occlusion may result in medial inferior
and corticobulbar tracts), ipsilateral CN III (Millard–Gubler) pontine syndrome, which is char-
palsy, and contralateral homonymous hemianopsia acterized by contralateral motor and sensory deficits
with macular sparing. Damage to the visual cortex (corticospinal tracts and medial lemniscus, respec-
can result in alexia without agraphia, as the right tively), internuclear ophthalmoplegia (medial lon-
visual cortex is unable to communicate with Broca gitudinal fasciculus), ipsilateral gaze paralysis
and Wernicke areas. Large- and small-vessel dis- (paramedian pontine reticular formation), ipsilateral
eases are the usual culprits, whereas cardiogenic lateral rectus paralysis (CN VI), and ipsilateral facial
embolism is rare [46]. PCA occlusion on CT dem- weakness and partial loss of taste (facial colliculus)
onstrates low attenuation or loss of gray–white [44, 47–50] (Figs. 19, 21, and 22).
margins in PCA territory, while MRI depicts dif- Acute occlusion of the basilar artery is a true
fusion restriction and FLAIR and T2-weighted neurointerventional emergency. Patients with
hyperintensity (Fig. 7). MRI is best for localizing acute occlusion of the basilar artery present with
small infarcts and subtle midbrain and/or temporal sudden and dramatic neurological impairment
lobe findings. and, without early treatment, rapid deterioration.

Fig. 21 Acute basilar


artery occlusion. (a and b)
CT scans reveal
hypodensity throughout the
left cerebellum, brain stem,
and bilateral occipital lobes
with a dense basilar artery.
Note the marked
compression of the
quadrigeminal plate cistern
and hydrocephalus
352 J. Rotman and R. Zimmerman

Fig. 22 Acute basilar


artery occlusion. (a–c) DWI
images reveal restricted
diffusion in the cerebellum,
right hippocampus, right
occipital lobe, and bilateral
thalami. (d) T2-weighted
image reveals the absence
of a flow void in the distal
basilar artery. (e) MRA
reveals complete absence of
flow in the basilar artery and
its branches

Complete bilateral lesions to the pons will affect Diagnosis of hyperacute basilar thrombosis on
the reticular activating substance, which affects CT is challenging because calcifications within
coordination and motor control and manifests as the basilar artery are common (much more so
locked-in syndrome with complete paralysis than within M1 segment of MCA) and density
[51]. Acute basilar artery occlusion can be due to changes within the brain are difficult to detect
either thromboembolism or thrombosis from ath- because of beam-hardening artifact. MR is supe-
erosclerosis or propagation of an intracranial rior to CT allowing for detection of parenchymal
dissection [52]. changes (hyperintensity on DWI and/or FLAIR)
The earliest sign on CT of acute occlusion is a and visualization of intraluminal clot (GrE and/or
hyperdense basilar artery (Fig. 21) analogous to SWI) (Fig. 22).
the dense MCA sign followed by the development Large-vessel atherosclerosis of the
of hypodensity and loss of gray–white differenti- vertebrobasilar arteries has been demonstrated in
ation in the bilateral temporal lobes, occipital more than two thirds of patients with cerebellar
lobes, midbrain, thalami, and peduncles. infarction [6], which only accounts for
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 353

approximately 1.5–2.3 % of all cerebral infarc- Watershed infarcts (WIs) that occur at the junction
tions [53, 54]. The cerebellum swells with an between two or three arterial territories account
infarction involving more than one third of its for approximately 10 % of ischemic strokes [6]
volume, a basilar artery with poor collateral sup- (Fig. 23).
ply, an embolus with reperfusion, and/or a mas- In patients with ICA occlusion, hypotensive
sive SCA infarction [7]. However, cerebellar cerebral infarcts account for 72 % of delayed
infarcts are often difficult to detect on CT due to strokes, although they are rarely the initial mani-
beam-hardening artifact or partial volume averag- festation of ICA occlusion [56]. Conversely, 50 %
ing, which can mask subtle regions of low density. of patients with high-grade ICA stenosis suffer
Attention to subtle asymmetry involving the watershed infarcts [6]. Cerebrovascular investiga-
fourth ventricle and quadrigeminal plate cistern tion of patients with MCA watershed infarcts pre-
may aid in the identification of cerebellar edema. sumably due to atherosclerotic disease should
Small cerebellar infarcts less than 2 cm are best include evaluation of the large extra- and intracra-
identified on MRI. Obstructive hydrocephalus nial vessels.
may occur, which manifests early as dilatation of Bilateral hypotensive cerebral infarcts are
the temporal horns (Fig. 17). Minimal mass effect mainly attributed to global reduction in perfusion
and slight ventriculomegaly can rapidly evolve to pressure in the setting of significant hypoxia/
large-volume strokes with compression of the hypovolemia or hemodynamic impairment in
brainstem and cerebellar herniation; the superior patients with diffuse cerebral vascular diseases,
vermis can herniate superiorly through the such as sickle cell anemia (Fig. 24). Unilateral
tentorium, while the tonsils and inferior vermis presentations after global hypoxic-ischemic
may herniate downward through the foramen events are possible in cases of focal underlying
magnum. vascular stenosis and relative hypoperfusion.
Decreased perfusion can also alter blood flow
and thus allow for microemboli to reach recipient
Watershed Infarcts blood vessels; therefore, even in the absence of
severe stenosis, reduction of flow can result in
Watershed, or border zone, infarctions occur at the ischemia due to decreased circulation of distal
most distal areas between arterial territories and emboli, which can enable regional ischemia to
are thought to be secondary to severe develop.
hypoperfusion, such as in cases of severe stenosis On CT, major arterial border zone infarcts pre-
or occlusion of the carotid artery and prolonged sent with hypodensity at gray–white matter junc-
hypotension [55]. Also called hypotensive cere- tions between vascular territories. In severe cases,
bral infarctions, they are typically seen at there is effacement of nearly all supratentorial
gray–white matter junctions as well as between gray–white matter junctions. The basal ganglia
perforating arteries in deep white matter. Reduc- and thalami may also be affected. Deep white
tion of flow primarily affects these zones to the matter hypotensive cerebral infarcts typically
greatest extent due to insufficient CBF to meet manifest with more than three white matter lesions
metabolic demands. within the centrum semiovale, similar in location
Two vascular border zone areas are present to areas affected by prolonged hypoxia, although
within the cerebral hemispheres, the cortical and changes appear multifocal rather than diffuse. A
the internal. Anterior cortical border zone areas “string of pearls” appearance is used to describe
include the cortical blood supply of the ACA and the linear orientation of multiple watershed
MCA, while the posterior cortical border zone infarcts that occur parallel to the lateral ventricles
areas are located between the MCA and PCA. (Fig. 23). CTA can be performed to determine the
Commonly associated with severe ICA or MCA degree of severity in cases of ICA occlusion.
occlusion/stenosis, internal border zone areas are There is decreased CBF to affected areas on CT
located between the ACA, MCA, and PCA. perfusion studies.
354 J. Rotman and R. Zimmerman

Fig. 23 Watershed (border


zone) infarct. (a–c) DWI
reveals multiple foci of
restricted diffusion (“string
of pearls sign”) at the
borders between the
anterior and middle cerebral
artery territories. (d) MRA
reveals focal stenosis of the
supraclinoid internal carotid
artery with decreased flow
in the proximal internal
carotid artery

Fig. 24 Chronic lacunar infarct. (a) FLAIR image dem- differentiation from a dilated perivascular space. (b) On
onstrates a right periventricular infarct which has central DWI the infarct is hypointense, while on (c) ADC it is
fluid intensity and peripheral T2 hyperintensity. The pres- hyperintense
ence of peripheral hyperintensity on FLAIR allows for
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 355

The best imaging tool for detecting hypoten- supplied by the perforating arteries arising from
sive cerebral infarction is MRI, including SWI, the MCA or ACA.
MRA, and DWI. Acute infarcts demonstrate Patients with lacunar infarcts may have normal
hypointensity and edematous gyri on CT examinations. MR is more sensitive and allows
T1-weighted images, while subacute infarcts for identification of most small lesions regardless
may display gyriform cortical hyperintensity, of their age (Figs. 8, 9, and 12). Most lacunar
termed pseudolaminar necrosis (Fig. 12), and infarcts are clinically silent, and subtle neurological
enhance after contrast administration. deficits may go unnoticed by patients and/or phy-
T2-weighted images show hyperintensity in sicians. Clinical presentation depends on the size,
involved areas as well as cisternal/sulcal compres- location, and number of lacunar infarcts. Pure
sion in severe cases. FLAIR images can demon- motor hemiparesis is the most common lacunar
strate intraluminal arterial hyperintensity in cases syndrome (55 %) [9, 58]. Patients who present
of proximal vessel thrombosis or severe stenosis. with classic lacunar syndromes have high proba-
bilities of recovery and are typically treated with
antiplatelet therapy for secondary prevention [5].
Lacunar Infarcts As aforementioned, acute lacunar infarcts are
often undetected and are typically impossible to
The word “lacune,” translated as “hole” in Latin, distinguish from chronic lacunar infarcts on CT,
is used to describe small fluid-filled cavities although the presence of distinct convex margins
mostly present in the basal ganglia and thalami. is suggestive of acute disease. Because of their
Lacunar infarcts (LIs) are deep cerebral infarcts small size, many lacunar infarcts are not visual-
less than 1.5 cm in diameter, although this size ized on CT scans. When they are found on CT,
criteria does not apply in the acute phase due to lacunar infarcts appear as small discrete round/
the presence of cellular swelling and extracellular ovoid foci of hypodensity that may enhance in
edema. LIs usually represent the healed stage of the late acute or early subacute stage. The Massa-
small deep infarcts, which likely occurred second- chusetts General Hospital group recommends
ary to occlusion of a single penetrator artery aris- narrowing CT window widths and levels to better
ing from the large arteries of the circle of Willis or identify subtle hypoattenuating lesions that por-
from the basilar artery. The most commonly tend infarct, although since the advent of
involved arteries involved include the low-dose algorithms to limit radiation exposure,
lenticulostriate, thalamoperforate, and anterior the increased noise inherent to such scans makes
choroidal artery perforators. the detection of subtle infarcts more difficult [6].
LIs are found in approximately 11 % of On MRI, acute lacunar infarcts present as foci
patients admitted with stroke [9]. LIs can involve of T1 iso- to hypointensity and T2 hyperintensity.
the upper two thirds of the basal ganglia, the Similar to the case of CT, they remain difficult to
internal capsule, the thalamus, or the paramedian distinguish from chronic lacunar infarcts on these
and lateral regions of the brainstem (Figs. 8, 9, and sequences, particularly in patients with chronic
12). Brainstem LIs are mainly found in the microvascular ischemia depicted as multiple foci
paramidline region of the pons, which is supplied of T2 hyperintensity in the white matter. DWI
from midline and anteromedial perforators arising makes detection of acute lacunar infarcts easy
from the basilar artery (Fig. 18). LIs are less since these lesions are hyperintense, while chronic
commonly located in the medulla and midbrain, white matter lacunar infarctions and ischemic
as these areas are supplied by short arterioles and changes are DWI isointense (Fig. 12). Lacunar
are thus less vulnerable to aging and hypertension. infarcts often demonstrate peripheral
While symptomatic initial LIs are usually located hyperintensity surrounding a fluid intensity core
in the area supplied by anterior choroidal artery on FLAIR thus allowing for differentiation from
[57] (Figs. 8 and 14), multiple asymptomatic LIs large VR spaces, which lack the peripheral
are commonly located within the territory hyperintensity of lacunar infarcts (Fig. 24).
356 J. Rotman and R. Zimmerman

Patients with thalamic lesions (Figs. 9, 11, Anoxia and Hypoxia


and 24) may present with sensorimotor and
behavioral symptoms due to interruption of recip- In anoxic injuries, the metabolically active areas of
rocal cortical-thalamic connections. There are the brain such as the basal ganglia, dentate nucleus,
four major thalamic vascular territories, each and hippocampus are the most commonly affected.
with a predilection for supplying particular groups Corresponding CT abnormalities may take as long
of nuclei: (1) tuberothalamic, (2) inferolateral, as 24 h to become visible. The earliest CT finding is
(3) paramedian, and (4) posterior choroidal ves- subtle loss of normal gray matter hyperdensity,
sels. Patients with tuberothalamic territory strokes leading to the inability to discern the basal ganglia
may display impaired arousal, orientation, learn- and thalami from the internal capsules (Fig. 25).
ing, memory, and executive function as well as Over time, as in the case with localized strokes,
superimposition of temporally unrelated informa- hypodensity and mass effect become apparent. The
tion and emotional facial paresis [59, 60]. delayed CT appearance of anoxic insult may result
Paramedian thalamic infarcts also cause from maintenance of the CBV (which in contrast to
decreased arousal (especially if bilateral) and hyperacute infarction, for example, is diminished);
impaired learning and memory [59]. Confusion, therefore, CT changes only become visible once
agitation, aggression, and apathy may be persis- vasogenic edema has set in. In contradistinction,
tent features [61–63]. Left paramedian and left anoxic brain injuries can be visualized within as
tuberothalamic lesions that include the ventrolat- early as 3 h on MRI due to cytotoxic edema and cell
eral nucleus produce language deficits, whereas death, which result in DWI hyperintensity and
right thalamic lesions in these territories result in restricted diffusion on ADC maps (Fig. 26).
visual-spatial deficits, including hemispatial Changes that result from carbon monoxide toxicity
neglect [59]. Inferolateral territory strokes cause are similar to those seen in anoxia but are most
contralateral hemisensory loss, hemiparesis pronounced in the bilateral globus pallidi (Fig. 27).
and hemiataxia, and pain syndromes that more If delayed encephalopathy occurs 2–3 weeks after
commonly occur with right thalamic lesions recovery, additional findings include T2-weighted
[59, 64, 65]. hyperintensity involving the corpus callosum, sub-
Arising from the PCA, the medial and lateral cortical U fibers, and internal and external capsules
posterior choroidal arteries supply the trigone and hypointensity in the thalamus and putamen
region of the lateral ventricle. The medial poste- [7]. In patients who survive, chronic anoxic injury
rior choroidal arteries pass around the midbrain results in basal ganglia and hippocampal atrophy
and course medially toward the pineal gland. with secondary dilation of the frontal and temporal
They supply the tectum, choroid plexus of the horns of the lateral ventricles.
third ventricle, and thalami. The lateral posterior Prolonged hypoxia leads to hypoperfusion and
choroidal arteries run behind the pulvinar into the the portions of the brain furthest from the circle of
choroidal fissure to supply the choroid of the Willis are preferentially affected. Thus, the
lateral and third ventricles, posterior thalamus, lesions begin in the border zone lesions and at
and fornix. The anterior choroidal artery territory least initially spare the more proximal but
anastomoses with posterior medial and lateral more metabolically active deep portions of the
choroidal branches. Patients with posterior cho- brain that are preferentially affected by
roidal lesions present with visual field deficits, anoxia. On CT there may be subtle loss of
variable sensory loss, weakness, dystonia, bilateral superficial gray–white differentiation
tremors, and occasional amnesia and language followed by diffuse brain swelling with sulcal
impairment [59]. MRI findings are similar to and ventricular compression. On MR there is dif-
those with anterior choroidal artery infarcts, with fuse symmetric cortical/subcortical hyperintensity
FLAIR and T2-weighted hyperintensity and acute on FLAIR and DWI may mimic a normal exam
restricted diffusion. (Fig. 28).
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 357

Fig. 25 Anoxia progressing to brain death. (a) Initial CT preserved density in the cortical gray matter. (c) Scan at
scan performed 6 h after an episode of near drowning 48 h reveals complete loss of normal gray matter density in
demonstrates normal gray matter density in the deep gray the basal ganglia and thalamus as well as the cortex. Sulci
matter and cortex. (b) Follow-up scan at 18 h reveals loss have been obliterated and the ventricles are small, indica-
of normal density in the basal ganglia and thalami with tive of brain death

Fig. 26 Anoxia. (a) CT


demonstrates diffuse
hypodensity in the bilateral
basal ganglia with mass
effect on the lateral
ventricles. (b) FLAIR
image demonstrates diffuse
T2 hyperintensity in the
same regions

In severe cases of anoxia or hypoxia, CT find- to complete cessation of CBF and resultant brain
ings progress to diffuse cerebral edema with sulcal death (Fig. 25).
and cisternal obliterations. The brain will appear Infarcts related to fat emboli commonly affect
diffusely hypodense without any gray–white mat- both gray and white matter and mimic thrombo-
ter distinction and will be nearly completely fea- embolic strokes. Diffuse white matter ischemia
tureless with the exception of visualization of can result directly from a massive fat embolism
major arterial structures near the circle of Willis or in combination with hypoxia from pulmonary
and the dura. These findings may mimic subarach- complications. A “hypodense MCA” sign may be
noid hemorrhage. Increased intracranial pressure the only sign present on initial CT, related to the
produces central and tonsillar herniations, leading presence of fat within the MCA. DWI will be
358 J. Rotman and R. Zimmerman

Fig. 27 Carbon monoxide


poisoning. (a and b) FLAIR
imaging demonstrates
hyperintensity in the
hippocampi and globus
palladi

Fig. 28 Severe prolonged


hypoxia. (a and b) Axial
DWI and (c and d) FLAIR
images demonstrate diffuse
gyral hyperintensity. Note
the sparing of the basal
ganglia. The global
abnormality creates a
“pseudonormal”
appearance
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 359

Fig. 29 Fat emboli. (a) Axial FLAIR and (b) DWI at the susceptibility hypointensity indicative of
level of the lateral ventricles demonstrate multiple foci of microhemorrhage from microemboli, referred to as the
T2 hyperintensity. (c) SWI demonstrates numerous foci of “starfield” pattern, consistent with fat embolization

positive in acute cases due to extensive small-


vessel ischemia and hypoxia; foci of cytotoxic Summary
edema from multiple microemboli manifest as
bright spots on a dark background, which is Stroke is a leading cause of mortality and morbid-
referred to as the “starfield” pattern [66] ity in the developed world. The goals of imaging
(Fig. 29). T1-weighted images are usually in the setting of acute stroke are to establish a
unremarkable, while T2 and FLAIR images may diagnosis as early as possible and to deduce accu-
demonstrate small, scattered foci of rate information regarding the intracranial vascu-
hyperintensity, mimicking the pattern of water- lature and brain perfusion to guide in the selection
shed infarcts. of appropriate therapy. While CT has the
360 J. Rotman and R. Zimmerman

advantage of detecting acute hemorrhage, MRI multicenter clinical trial. TOAST. Trial of Org 10172
offers more accurate pathophysiological informa- in Acute Stroke Treatment. Stroke 24(1):35–41
5. Kolominsky-Rabas PL et al (2001) Epidemiology of
tion. Different imaging characteristics associated ischemic stroke subtypes according to TOAST criteria:
with different stages of infarction significantly incidence, recurrence, and long-term survival in ische-
affect the decision of whether or not to administer mic stroke subtypes: a population-based study. Stroke
thrombolytic therapy. For example, 32(12):2735–2740
6. Rovira A, Grive E, Alvarez-Sabin J (2005) Distribution
hyperintensity on MRI diffusion-weighted imag- territories and causative mechanisms of ischemic
ing may be the only sequence to identify stroke. Eur Radiol 15(3):416–426
hyperacute infarct and thus indicate candidacy 7. Zimmerman RD (2010) Vascular diseases of the brain.
for acute stroke management. In: Yousem DM, Grossman RI (eds) Neuroradiology:
the requisites. Mosby, Philadelphia, pp 104–169
The most common causes of infarction include 8. Jung JM et al (2010) Difference in infarct volume and
large-artery atherosclerosis, cardioembolism, and patterns between cardioembolism and internal carotid
lacunes, and outcomes tend to differ depending on artery disease: focus on the degree of cardioembolic
subtype. For instance, large-artery lesions have risk and carotid stenosis. Cerebrovasc Dis 29
(5):490–496
higher associated mortality rates than lacunar 9. Arboix A, Alio J (2012) Acute cardioembolic cerebral
infarcts, while cardioembolic strokes are associ- infarction: answers to clinical questions. Curr Cardiol
ated with more frequent recurrence and higher Rev 8(1):54–67
1-month mortality rates. Since characteristic 10. Ferro JM (2003) Cardioembolic stroke: an update.
Lancet Neurol 2(3):177–188
MRI imaging patterns can discern specific ische- 11. Hart RG, Halperin JL (2001) Atrial fibrillation and
mic stroke etiologies, certain findings are able to stroke: concepts and controversies. Stroke 32
guide more personalized patient management. For (3):803–808
example, whereas large cortical territorial infarcts 12. Yesilot Barlas N et al (2013) Etiology of first-ever
ischaemic stroke in European young adults: the 15 cit-
are typically associated with cardioembolism and ies young stroke study. Eur J Neurol 20(11):1431–1439
warrant cardiac work-up, lacunar infarcts are typ- 13. Timsit SG et al (1993) Brain infarction severity differs
ically treated with anticoagulation. Moreover, according to cardiac or arterial embolic source. Neu-
understanding that intracranial emboli most com- rology 43(4):728–733
14. Horowitz DR et al (1992) Mechanisms in lacunar
monly produce MCA territory infarcts cues the infarction. Stroke 23(3):325–327
physician to investigate the internal carotid artery, 15. Lodder J et al (1990) Are hypertension or cardiac
since emboli that enter the carotid artery preferen- embolism likely causes of lacunar infarction? Stroke
tially lodge within MCA branches. Appreciating 21(3):375–381
16. Tatu L et al (1996) Arterial territories of human brain:
this pattern also affects patient treatment, since brainstem and cerebellum. Neurology 47
rather than medical management, carotid endar- (5):1125–1135
terectomy and stenting is indicated for large- 17. Tatu L et al (1998) Arterial territories of the human
vessel disease. brain: cerebral hemispheres. Neurology 50
(6):1699–1708
18. Nakano S et al (2001) Correlation of early CT signs in
the deep middle cerebral artery territories with angio-
graphically confirmed site of arterial occlusion. AJNR
References Am J Neuroradiol 22(4):654–659
19. Becker H et al (1979) CT fogging effect with ischemic
1. Picanco MR et al (2014) Reperfusion after 4.5 hours cerebral infarcts. Neuroradiology 18(4):185–192
reduces infarct growth and improves clinical outcomes. 20. Hopyan J et al (2010) Certainty of stroke diagnosis:
Int J Stroke 9(3):266–269 incremental benefit with CT perfusion over
2. Allen LM et al (2012) Sequence-specific MR imaging noncontrast CT and CT angiography. Radiology 255
findings that are useful in dating ischemic stroke. (1):142–153
Radiographics 32(5):1285–1297; discussion 1297–9 21. Wechsler LR (2011) Imaging evaluation of acute ische-
3. Bamford J et al (1991) Classification and natural his- mic stroke. Stroke 42(Suppl 1):S12–S15
tory of clinically identifiable subtypes of cerebral 22. Meretoja A et al (2014) Stroke thrombolysis: save a
infarction. Lancet 337(8756):1521–1526 minute, save a day. Stroke 45(4):1053–1058
4. Adams HP Jr et al (1993) Classification of subtype of 23. Gonzalez RG et al (1999) Diffusion-weighted MR
acute ischemic stroke. Definitions for use in a imaging: diagnostic accuracy in patients imaged within
16 Patterns of Ischemic Stroke: From Lacunar to Territorial to Multiple Embolic to Watershed. . . 361

6 hours of stroke symptom onset. Radiology 210 40. Norrving B, Cronqvist S (1991) Lateral medullary
(1):155–162 infarction: prognosis in an unselected series. Neurol-
24. Mitomi M et al (2014) Comparison of CT and DWI ogy 41(2 (Pt 1)):244–248
findings in ischemic stroke patients within 3 hours of 41. Kim JS (2003) Pure lateral medullary infarction:
onset. J Stroke Cerebrovasc Dis 23(1):37–42 clinical-radiological correlation of 130 acute, consecu-
25. Bryan RN et al (1991) Diagnosis of acute cerebral tive patients. Brain 126(Pt 8):1864–1872
infarction: comparison of CT and MR imaging. AJNR 42. Amarenco P et al (1993) Anterior inferior cerebellar
Am J Neuroradiol 12(4):611–620 artery territory infarcts. Mechanisms and clinical fea-
26. Brazzelli M (2009) Magnetic resonance imaging ver- tures. Arch Neurol 50(2):154–161
sus computed tomography for detection of acute vas- 43. Oas JG, Baloh RW (1992) Vertigo and the anterior
cular lesions in patients presenting with stroke inferior cerebellar artery syndrome. Neurology 42
symptoms. Cochrane Database Syst Rev (4):Cd007424 (12):2274–2279
27. Huang IJ et al (2001) Time course of cerebral infarction 44. Kim JS, Choi-Kwon S (1999) Sensory sequelae of
in the middle cerebral arterial territory: deep watershed medullary infarction: differences between lateral and
versus territorial subtypes on diffusion-weighted MR medial medullary syndrome. Stroke 30
images. Radiology 221(1):35–42 (12):2697–2703
28. Warach S et al (1995) Acute human stroke studied by 45. Kumral E, Kisabay A, Atac C (2005) Lesion
whole brain echo planar diffusion-weighted magnetic patterns and etiology of ischemia in superior cerebellar
resonance imaging. Ann Neurol 37(2):231–241 artery territory infarcts. Cerebrovasc Dis 19
29. Perkins CJ et al (2001) Fluid-attenuated inversion (5):283–290
recovery and diffusion- and perfusion-weighted MRI 46. Kim JS, Kim J (2005) Pure midbrain infarction: clini-
abnormalities in 117 consecutive patients with stroke cal, radiologic, and pathophysiologic findings. Neurol-
symptoms. Stroke 32(12):2774–2781 ogy 64(7):1227–1232
30. Toyoda K, Ida M, Fukuda K (2001) Fluid-attenuated 47. Onbas O et al (2005) Millard-Gubler syndrome: MR
inversion recovery intraarterial signal: an early sign of findings. Neuroradiology 47(1):35–37
hyperacute cerebral ischemia. AJNR Am J Neuroradiol 48. Ahdab R et al (2013) Pure ipsilateral central facial
22(6):1021–1029 palsy and contralateral hemiparesis secondary to
31. Emeriau S et al (2013) Can diffusion-weighted ventro-medial medullary stroke. J Neurol Sci 332
imaging-fluid-attenuated inversion recovery mismatch (1–2):154–155
(positive diffusion-weighted imaging/negative fluid- 49. Toyoda K et al (1996) Medial medullary infarction:
attenuated inversion recovery) at 3 Tesla identify analyses of eleven patients. Neurology 47
patients with stroke at <4.5 hours? Stroke 44 (5):1141–1147
(6):1647–1651 50. Kim JS, Kim HG, Chung CS (1995) Medial medullary
32. Oppenheim C et al (2000) Prediction of malignant syndrome. Report of 18 new patients and a review of
middle cerebral artery infarction by diffusion-weighted the literature. Stroke 26(9):1548–1552
imaging. Stroke 31(9):2175–2181 51. Caplan LR (1980) “Top of the basilar” syndrome.
33. Georgiadis D et al (2002) Hemicraniectomy and mod- Neurology 30(1):72–79
erate hypothermia in patients with severe ischemic 52. Verdelho A, Pereira JG, Ferro JM (1999) Multiple
stroke. Stroke 33(6):1584–1588 vertebro-basilar infarcts and cardio-embolism. Rev
34. Tei H et al (2000) Deteriorating ischemic stroke in Neurol 28(11):1027–1030
4 clinical categories classified by the Oxfordshire 53. Tohgi H et al (1993) Cerebellar infarction. Clinical and
Community Stroke Project. Stroke 31(9):2049–2054 neuroimaging analysis in 293 patients. The Tohoku
35. Yousem DM, Grossman RI (2010) Cranial anatomy. Cerebellar Infarction Study Group. Stroke 24
In: Yousem DM, Grossman RI (eds) Neuroradiology: (11):1697–1701
the requisites. Mosby, Philadelphia, pp 23–57 54. Macdonell RA, Kalnins RM, Donnan GA (1987) Cer-
36. Ross MA et al (1986) Magnetic resonance imaging in ebellar infarction: natural history, prognosis, and
Wallenberg’s lateral medullary syndrome. Stroke 17 pathology. Stroke 18(5):849–855
(3):542–545 55. Derdeyn CP et al (2001) Severe hemodynamic impair-
37. Ogawa K et al (2013) Clinical study of medial area ment and border zone–region infarction. Radiology
infarction in the region of posterior inferior cerebellar 220(1):195–201
artery. J Stroke Cerebrovasc Dis 22(4):508–513 56. Bogousslavsky J, Regli F (1986) Borderzone infarc-
38. Peterman AF, Sieker TR (1960) The lateral medullary tions distal to internal carotid artery occlusion: prog-
(Wallenberg) syndrome: clinical features and progno- nostic implications. Ann Neurol 20(3):346–350
sis. Med Clin North Am 44:887–896 57. Boiten J, Lodder J, Kessels F (1993) Two clinically
39. Grant G (1966) Infarction localization in a case of distinct lacunar infarct entities? A hypothesis. Stroke
Wallenberg’s syndrome. A neuroanatomical investiga- 24(5):652–656
tion with comments on structures responsible for nys- 58. De Reuck J, De Groote L, Van Maele G (2008) The
tagmus, impairment of taste and deglutition. classic lacunar syndromes: clinical and neuroimaging
J Hirnforsch 8(5):419–430 correlates. Eur J Neurol 15(7):681–684
362 J. Rotman and R. Zimmerman

59. Schmahmann JD (2003) Vascular syndromes of the groups defined by computerized tomography. Brain
thalamus. Stroke 34(9):2264–2278 108(Pt 2):485–516
60. Lisovoski F et al (1993) Left tuberothalamic artery 64. Caplan LR et al (1988) Lateral thalamic infarcts. Arch
territory infarction: neuropsychological and MRI fea- Neurol 45(9):959–964
tures. Eur Neurol 33(2):181–184 65. Nasreddine ZS, Saver JL (1997) Pain after
61. Castaigne P et al (1981) Paramedian thalamic and thalamic stroke: right diencephalic predominance and
midbrain infarct: clinical and neuropathological study. clinical features in 180 patients. Neurology 48
Ann Neurol 10(2):127–148 (5):1196–1199
62. Graff-Radford NR et al (1984) Nonhemorrhagic infarc- 66. Parizel PM et al (2001) Early diagnosis of cerebral fat
tion of the thalamus: behavioral, anatomic, and physi- embolism syndrome by diffusion-weighted MRI
ologic correlates. Neurology 34(1):14–23 (starfield pattern). Stroke 32(12):2942–2944
63. Graff-Radford NR et al (1985) Nonhaemorrhagic tha- 67. Han SW et al (2007) Frequency and significance of
lamic infarction. Clinical, neuropsychological and cardiac sources of embolism in the TOAST classifica-
electrophysiological findings in four anatomical tion. Cerebrovasc Dis 24(5):463–468
Territorial Strokes as a Tool to Learn
Vascular Territories 17
Behroze Adi Vachha and Pamela Whitney Schaefer

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364 Stroke is the fourth leading cause of death in
the United States and a leading cause of seri-
General Overview of Intracranial Arteries . . . . . . . 364
ous, long-term adult disability. One of the key
Anterior Circulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365 tasks in establishing the diagnosis of acute
Intradural Internal Carotid Artery . . . . . . . . . . . . . . . . . . . 365 ischemic stroke and determining its appropri-
Anterior Cerebral Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368
Middle Cerebral Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 370 ate treatment is establishing the arterial terri-
tory affected. Knowledge of cerebral vascular
Posterior Circulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 372
Intradural Vertebral Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . 372
territories helps identify abnormal vessels on
Basilar Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 374 CT and conventional angiograms, confirms
Posterior Cerebral Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . 376 that a DWI hyperintense lesion represents an
Border Zones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378 acute arterial stroke, and guides further inves-
tigations and treatment. Factors contributing to
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 379
understanding vascular territories include the
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 379 anatomy of the intracranial circulation and its
normal variants and the intrinsic variability in
the extent of brain supplied by the main
branches of the intracranial circulation. This
chapter reviews the normal anatomy of intra-
cranial arteries and describes the vascular dis-
tribution as seen on CT and MRI using major
territorial strokes as a learning tool.

Keywords
Stroke • Infarct • Vascular territories

B.A. Vachha • P.W. Schaefer (*)


Neuroradiology, Massachusetts General Hospital, Boston,
MA, USA
e-mail: bvachha@mgh.harvard.edu; bvachha@partners.
org; pschaefer@mgh.harvard.edu; pschaefer@partners.org

# Springer Science+Business Media New York 2016 363


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_10
364 B.A. Vachha and P.W. Schaefer

Introduction This chapter reviews the normal anatomy of


intracranial arteries and describes the vascular
Stroke is the fourth leading cause of death in the distribution as seen on CT and MRI using major
United States and a leading cause of serious, territorial strokes as a learning tool.
long-term adult disability [1]. Arterial
ischemic strokes account for 87 % of all
cases; the remaining strokes are mostly General Overview of Intracranial
hemorrhagic [2]. Arteries
Readily available computed tomography
(CT) and magnetic resonance imaging (MRI) in The intracranial circulation can be conveniently
emergency settings have allowed clinicians to divided into the anterior and posterior circulation.
improve correlations between clinical history The anterior circulation consists of the intradural
and precise anatomic lesion location in stroke internal carotid artery (ICA) and its branches, as
patients. One of the key tasks in establishing the well as the two terminal branches of the ICA – the
diagnosis of acute ischemic stroke and determin- anterior cerebral artery (ACA) and the middle
ing its appropriate treatment is establishing the cerebral artery (MCA). The anterior communicat-
arterial territory affected. Knowledge of cerebral ing artery (AComm) connecting the two ACAs
vascular territories helps identify abnormal ves- and the left and right posterior communicating
sels on CT and conventional angiograms, con- arteries (PComm) connecting the ipsilateral ICA
firms that a DWI hyperintense lesion represents to the ipsilateral posterior communicating artery
an acute arterial stroke, and guides further inves- (PCA) are also considered part of the anterior
tigations and treatment. Factors contributing to circulation [3]. The posterior circulation includes
understanding vascular territories include the the vertebral arteries, the basilar artery and its
anatomy of the intracranial circulation and its branches, and the terminal bifurcation of the bas-
normal variants and the intrinsic variability in ilar artery into the right and left PCAs [3]. Approx-
the extent of brain that the main branches of the imate vascular territories of the major intracranial
intracranial circulation supply. arteries are depicted in Fig. 1.

Fig. 1 Approximate vascular territories of the cerebral arteries and their branches as well as the posterior fossa vessels.
There is considerable variability in these territories particularly in the basal ganglia, brainstem, and posterior fossa
17 Territorial Strokes as a Tool to Learn Vascular Territories 365

Fig. 2 Anatomic diagram


of the normal circle of
Willis

At the base of the brain, branches of the inter-


nal carotid arteries and the vertebrobasilar Anterior Circulation
trunk anastomose to form an arterial polygon
known as the circle of Willis [3]. The circle of Intradural Internal Carotid Artery
Willis is formed by two terminal ICAs, two
proximal or horizontal (A1) segments of the The intracranial ICA is divided into six segments:
ACA, the AComm, two PComms, two proximal petrous (C2) segment, lacerum (C3) segment,
or horizontal (P1) segments of the PCAs, and the cavernous (C4), clinoid (C5) segment, ophthalmic
top of the basilar artery (BA). The MCAs are not (C6) segment, and communicating (C7) segment
considered part of the circle of Willis. The normal [4]. The ophthalmic (C6) segment is the first ICA
circle of Willis is depicted diagrammatically in segment that lies within the subarachnoid space
Fig. 2. and gives off two important branches: the oph-
The following sections discuss the thalmic artery which supplies the retina and the
normal anatomy and vascular distributions of the optic nerve [4] and the superior hypophyseal
anterior and posterior circulations as seen on CT artery which supplies the anterior pituitary lobe,
and MRI. the infundibular stalk, and the optic chiasm [5].
366 B.A. Vachha and P.W. Schaefer

The communicating (C7) segment extends Patients with infarctions confined to the ventral
from the PComm origin to the ICA terminus. anterior nucleus, anterior nuclei, paramedian
This segment has two important branches: the nuclei, mammillothalamic tract, and internal med-
PComm and the anterior choroidal artery. ullary lamina manifest differently from those with
the previously described larger tuberothalamic
Posterior Communicating Artery infarctions. In these patients, there is severe
The PComm joins the anterior and posterior cir- impairment of memory and new learning with
culation. It extends from the ICA to the junction of relative sparing of language [6]. Palipsychism is
the P1 and P2 segments of the PCA. Perforating a phenomenon associated with anterior thalamic
arteries arise from the posterior PComm and sup- infarcts in which patients demonstrate persevera-
ply the thalamus and hypothalamus [4]. tion which leads to overlap of sequential cognitive
The tuberothalamic artery or polar artery processes [9].
arises from the caudal part of the PComm, which
is close to the PCA, or from the border between Anterior Choroidal Artery
the caudal and middle third of the PComm [6, The anterior choroidal artery (AChA) arises from
7]. It is absent in approximately one-third of the the posterior wall of the ICA distal to the origin of
normal population and is considered to have an the PComm (although it occasionally arises from
anatomically complementary relationship with the MCA) and courses laterally in the suprasellar
the paramedian artery originating from the P1 cistern to enter the choroidal fissure of the tempo-
segment [7]. The tuberothalamic artery supplies ral horn of the lateral ventricle [3, 4].
the ventral section of the thalamus including the Although the vascular territory of the AChA
anterior thalamic nuclei (i.e., anteromedial, shows large variations, the most consistently
anteroventral, anterodorsal nuclei), ventral ante- documented area involves the amygdala, the lat-
rior nucleus, reticular nucleus, rostral portion of eral geniculate body, posterior two-thirds of the
the ventrolateral nucleus, ventral pole of the posterior limb of the internal capsule, most of the
medial dorsal nucleus, mammillothalamic tract, globus pallidus, the origins of the optic radiations,
ventral amygdalofugal pathway, and ventral por- the middle one-third of the cerebral peduncles,
tion of the internal medullary lamina [6]. and the lateral thalamic border [3, 4, 10, 11]. The
Infarction in the territory of the tuberothalamic AChA territory is reciprocal with those of the
artery results in severe neuropsychological defi- posterolateral and posteromedial choroidal arter-
cits [7]. A characteristic feature of tuberothalamic ies that arise from the PCA [4]. Figure 3 depicts an
artery infarctions is the impairment of recent infarct in the vascular territory supplied by the
memory, temporal disorientation, and new learn- AChA.
ing, which is more prominent with left-sided Interruption of blood flow from the AChA can
lesions, while right-sided lesions result in visual result in complete or partial manifestation of the
memory deficits, hemispatial neglect, and visual AChA syndrome which includes the triad of
spatial processing deficits [7, 6]. Language disor- hemiplegia (due to involvement of pyramidal
ders characterized by anomia, impaired fluency, tracts in the posterior limb of the internal capsule),
semantic and phonemic paraphasic errors, and hemisensory loss (due to involvement of the pos-
impaired comprehension are seen in left-sided terolateral nucleus of the thalamus), and homon-
lesions. The “amnestic syndrome” is associated ymous hemianopia (due to involvement of the
with a disconnection between the anterior tha- lateral geniculate body) [10, 12, 13]. Many
lamic nuclei and hippocampal formation, caused AChA territory infarcts present as lacunar
by the disruption of the mammillothalamic tract, syndromes [13].
as well as a disconnection between the amygdala
and anterior nuclei by disruption of the Normal Variants of the ICA
amygdalothalamic projections passing through Fetal origin of the posterior cerebral artery occurs
the internal medullary lamina [8]. when the caliber of the PComm may be the same
17 Territorial Strokes as a Tool to Learn Vascular Territories 367

Fig. 3 Thirty-six-year-old
female with right
hemiparesis and word-
finding difficulties.
Diffusion-weighted MRI
(DWI) obtained 2 h after
onset of symptoms
demonstrated restricted
diffusion in the left mesial
temporal lobe (a) and in the
left posterior limb of the
internal capsule (b)
consistent with acute
infarction in the left anterior
choroidal artery distribution

as or larger than that of the ipsilateral P1 segment trans-hypophyseal course and a lateral petrosal
of the posterior cerebral artery [14]. This variant variation in which the artery courses with the
has clinical significance since simultaneous sensory roots of the trigeminal nerve and exits
infarcts in the vascular territories of both the ante- the Meckel cave below the petroclinoid ligament
rior and posterior circulation can occur from ICA [18]. Saltzman classifies the PTA into three types:
emboli in the presence of fetal circulation [15]. Type I is defined when the PTA supplies the distal
A “hyperplastic” AChA is defined as a normal basilar artery, the proximal basilar artery is typi-
cerebrovascular variant in which the temporal- cally hypoplastic, and the ipsilateral PComm is
occipital branches of the posterior cerebral artery absent, Type II is defined when the PTA fills the
arise from the AChA [14]. The clinical signifi- anterior superior cerebellar arteries only and the
cance of this variant is the increased occurrence posterior cerebral arteries are supplied by the
of intracranial aneurysm formation [16]. PComms, and Type III demonstrates a PTA that
In a 4-mm embryo, there are sites of anasto- unites with a remnant of the primitive paired lon-
mosis between the paired dorsal aortic arches that gitudinal neural artery and supplies an ipsilateral
later form the internal carotid arteries and the cerebellar artery (usually the anterior-inferior cer-
paired longitudinal neural arteries that later form ebellar artery) [19]. Clinically, these anomalies
the vertebrobasilar system [17]. With the excep- may be responsible for ischemia and trigeminal
tion of the PComm, all other primitive arterial neuralgia [20].
connections regress when the definite circulation The persistent hypoglossal artery originates
develops. Failure of these vessels to regress from the ICA at the levels of the C1 through C3
results in persistent carotid-vertebrobasilar anas- vertebral bodies and courses through the hypo-
tomoses. From cephalad to caudal, these are the glossal canal to anastomose with the basilar artery
persistent trigeminal artery, persistent otic artery, [3]. This variant is the second most common
persistent hypoglossal artery, and proatlantal carotid-vertebrobasilar anastomosis and has been
intersegmental artery [14, 17]. reported to result in glossopharyngeal neuralgia
The persistent trigeminal artery (PTA) is seen and hypoglossal nerve paralysis [3, 14].
in 0.1–0.2 % of patients and is the most common The proatlantal intersegmental artery arises
of the carotid-vertebrobasilar anastomoses from the internal carotid artery (Type I) or the
[3]. Salas et al. classify the PTA into a medial external carotid artery (Type II) at the levels of
sphenoid variation which has an intrasellar or the C2 through C4 vertebral bodies and joins the
368 B.A. Vachha and P.W. Schaefer

suboccipital vertebral artery before coursing gives rise to the medial lenticulostriate arteries
through the foramen magnum [3, 4]. which supply the medial basal ganglia and the
The persistent otic artery is the least common anterior limb of the internal capsule [3, 4]
of the carotid-vertebrobasilar anastomoses. The (Fig. 1). The AComm has perforating arteries
actual existence of this variant is controversial, that supply the anterior hypothalamus, optic chi-
and it may represent overlapping vascular terri- asm, genu of the corpus callosum, cingulate
tories rather than persistence of an embryonic gyrus, and pillars of the fornix.
vessel [4, 14].
Occlusions of the ICA may have clinical man- Vertical (A2) Segment
ifestations related to embolism or low flow The vertical/A2 segment courses in the
(discussed in border zone infarctions) [15]. Epi- interhemispheric fissure and gives rise to the
sodes of transient monocular blindness due to orbitofrontal and frontopolar arteries that sup-
embolization to the retinal circulation are typical. ply the undersurface and the inferomedial aspect
An ICA occlusion may be asymptomatic in the of the frontal lobe [3, 4].
presence of a competent circle of Willis. When a
thrombus propagates up to the top of ICA and Callosal (A3) Segment
occludes both the MCA and the ACA, the occlu- The callosal/A3 segment curves around the cor-
sion is often called a “T” occlusion [15]. It carries pus callosal genu and divides into the pericallosal
a very poor prognosis unless complete recanaliza- and callosomarginal arteries. The pericallosal
tion is achieved very early [21–23]. artery courses over the superior surface of the
corpus callosum in the pericallosal cistern and
gives off many small branches to the corpus
Anterior Cerebral Artery callosum [3]. The callosomarginal artery
courses within the cingulate sulcus over the cin-
The ACA is the smaller terminal branch of the gulate gyrus [3]. The cortical branches of the
supraclinoid ICA. The ACA supplies the anterior pericallosal and callosal marginal arteries supply
two-thirds of the medial surface of the cerebral the medial portions of the frontal lobes, superior
hemisphere extending from the frontal pole to the medial portions of the parietal lobes, and the ante-
parieto-occipital sulcus; this region includes the rior corpus callosum [24]. Infarctions of these
motor and sensory cortical areas for the pelvis and vessels result in hemiparesis and hemianesthesia
the lower extremities. The ACA vascular territory of the contralateral leg due to involvement of the
includes the anterior four-fifths of the corpus medial precentral and postcentral gyri,
callosum, the anterior limb of the internal capsule, respectively.
the anterior-inferior head of the caudate nucleus,
and approximately 1 in. of the frontal and parietal Recurrent Artery of Heubner
cortex of the cerebral convexity adjacent to the The recurrent artery of Heubner arises from the
interhemispheric fissure [3, 4]. Figure 4 demon- proximal A2 or the distal A1 segment [25, 26] and
strates an acute infarct with hemorrhagic conver- supplies the anterior part of the caudate nucleus,
sion in the expected vascular distribution of the the anterior third of the putamen, the tip of the
ACA. The ACA has three segments: horizontal outer segment of the globus pallidus, and the
(A1), vertical (A2), and callosal (A3) segments. inferior anterior limb of the internal capsule [25,
26]. Hemiparesis with faciobrachial predomi-
Horizontal (A1) Segment nance has been attributed to occlusions within
The horizontal/A1 segment extends from its ori- this artery [26]. Figure 5 depicts an acute infarct
gin to the midline where it communicates with the in the vascular territory of the recurrent artery of
contralateral ACA by the AComm. The ACA Heubner.
17 Territorial Strokes as a Tool to Learn Vascular Territories 369

Fig. 4 Sixty-year-old male


with weakness of the right
shoulder, leg, and foot.
Non-contrast-enhanced CT
of the brain (a) obtained 6 h
after onset of symptoms
demonstrates
hypoattenuation with
superimposed hyperdensity
consistent with
hemorrhagic conversion of
acute infarct in the left
anterior cerebral artery
distribution. Diffusion-
weighted (b) and FLAIR (c)
images obtained 1 h later in
the same patient show the
full extent of acute left
anterior cerebral artery
infarct

Normal Variants of the Anterior Cerebral ACA (Type III anomaly) where there is an addi-
Arteries tional vessel arising from the anterior communi-
The azygos anterior cerebral artery is a rare vari- cating artery accompanied by two hypoplastic A2
ant of the anterior cerebral artery involving a segments [27]. The most common anomaly noted
common trunk in the A2 segment (above the by Baptista was the Type III anomaly, while the
anterior communicating artery) with a prevalence Type I anomaly was observed in only 1 of the
of 0.2–4.0 % [14, 27]. Baptista distinguished three 381 brain specimens examined by him [27]. An
types of this anatomical variant: (1) “unpaired” azygos anterior cerebral artery is associated with a
ACA or true azygos artery (Type I anomaly) in large number of cerebral anomalies including
which a single unpaired ACA provides branches agenesis of corpus callosum, porencephalic
to both cerebral hemispheres; (2) “bihemispheric” cysts, hydranencephaly, lobular
ACA (Type II anomaly) in which the A2 segment holoprosencephaly, septo-optic dysplasia, and
of one ACA sends branches across the midline, saccular aneurysms. Being aware of the presence
while the contralateral A2 segment either is hypo- of an azygos ACA is important, as an occlusion of
plastic or terminates early in its course toward the this vessel can result in infarcts involving the
genu of the corpus callosum; and (3) “accessory” bilateral medial cerebral hemispheres [14].
370 B.A. Vachha and P.W. Schaefer

transcortical aphasia, apraxia, apathy, abulia,


incontinence, and disinhibition [26]. Bilateral
ACA territory infarction causes a neurologic syn-
drome characterized by profound akinetic mut-
ism, paraparesis, and poor recovery [26].

Middle Cerebral Artery

The MCA has the largest vascular territory of all


of the major intracranial arteries and is the vessel
most commonly affected by cerebrovascular acci-
dents [30]. Cortical branches of the MCA supply
two-thirds of the lateral surface of the cerebral
hemisphere with the exception of a thin 1-in.
strip near the vertex that is supplied by the ACA
and the occipital and posteroinferior parietal lobes
that are supplied by the PCA (Fig. 1). Infarcts that
occur in different segments of the MCA lead to
diverse neurologic outcomes. The following sub-
Fig. 5 Fifty-five-year-old male with history of atrial fibril- sections detail the specific MCA territories sup-
lation presented with new onset seizures. Diffusion-
weighted MRI obtained 3 h after onset of symptoms dem- plied by the various segments of the MCA and
onstrates restricted diffusion in the caudate nucleus, ante- their correlation to specific neurologic deficits.
rior limb of the internal capsule, and anterior aspect of the
putamen and globus pallidus consistent with acute infarct Horizontal (M1) Segment
in the expected vascular distribution of the recurrent artery
of Heubner. There is an additional small acute infarct in the The horizontal/M1 segment of the MCA arises
right periatrial white matter from the carotid terminus and extends laterally
where it bifurcates or trifurcates just before it
enters the Sylvian fissure. The anterior temporal
Trifurcation of the anterior cerebral artery, artery arises from the M1 segment and supplies
defined as the presence of three A2 segments of the tip of the temporal lobe [4]. Lateral
the ACA, is seen in 2–13 % cases and most likely lenticulostriate arteries are deep penetrating
represents persistence of the median callosal branches of the M1 segment that supply the supe-
artery [14]. In the presence of thromboembolic rior part of the head and body of the caudate
disease, hypoplasia or absence of the A1 segment nucleus, the putamen, and the external capsule
of the ACA typically results in a decreased collat- [4, 31].
eral supply and consequently an increased risk of Complete proximal M1 occlusions (Fig. 6)
infarction [14]. result in sensory, motor, language, and executive
Clinical manifestations of unilateral ACA ter- function deficits due to disruption of both MCA
ritory infarction are spastic hemiparesis of the cortical representations and basal ganglia circuit
contralateral lower limb and to a lesser degree structures, while distal MCA occlusions spare the
paresis of the arm, with the face and tongue lateral lenticulostriate vascular territory but
largely spared [26, 28, 29]. Sensory dysfunction, involve the distributions of both the superior and
particularly proprioceptive and discriminative inferior division branches (see below).
impairment of the contralateral lower limb, may
be present but is usually mild. Other manifesta- Insular (M2) Segments
tions of unilateral ACA territory infarction In the majority of cases, the MCA divides into two
include speech disturbance with initial mutism, trunks – superior and inferior – although
17 Territorial Strokes as a Tool to Learn Vascular Territories 371

Fig. 6 Sixty-seven-year-old female with left hemiplegia


last seen well 9 h prior to presentation in the emergency
room. Diffusion-weighted MRI (DWI) demonstrated
restricted diffusion within the right frontal and temporal Fig. 7 Sixty-six-year-old male with right facial droop and
lobe consistent with acute infarct in the vascular distribu- expressive aphasia. Diffusion-weighted MRI (DWI) of the
tion of the superior and inferior divisions of the right brain obtained 4 h after symptom onset shows an acute
middle cerebral artery infarct involving the left frontal operculum and left anterior
insula in the expected vascular distribution of the superior
division of the left middle cerebral artery

occasionally it trifurcates into superior, middle,


and inferior divisions [3, 4, 30, 31]. The superior Normal Variants of the Middle Cerebral
division supplies the frontal and peri-Rolandic Arteries
regions including the anterior parietal lobe An early bifurcation or trifurcation of the M1
[30]. The inferior division supplies the lateral segment close to its origin at the ICA is a common
temporal and inferior parietal lobes [30]. Figures 7 finding and is of no definite clinical significance
and 8 demonstrate acute infarcts in the vascular [14]. A middle cerebral artery branch arising from
territories of the superior and inferior divisions of the distal internal carotid artery is called a “dupli-
the MCA respectively. cated” MCA, while a vessel arising from the ACA
and coursing parallel to the M1 segment of the
Opercular (M3) Segments MCA is called an “accessory MCA” [14]. The
The portions of the superior and inferior divisions frequency of accessory MCA is 0.3–0.4 %,
that course laterally under the frontal, parietal, and while that of MCA duplication is 0.2–2.9 %
temporal opercula are termed the opercular/M3 [14]. The accessory MCA supplies the territory
segments [30, 31]. of the orbitofrontal, prefrontal, precentral, and/or
central arteries [32]. The duplicated MCA sup-
Cortical (M4) Segments plies the territory of the lateral part of the orbital
The cortical/M4 segments are the portions of the surface of the frontal lobe and the anterior and/or
superior and inferior MCA divisions that exit the middle temporal territories. In the setting of MCA
Sylvian fissure and branch over the lateral surface infarcts, the accessory MCA can provide collat-
of the hemisphere [3, 4]. eral flow to the anterior frontal lobe, but it cannot
372 B.A. Vachha and P.W. Schaefer

More distal superior division branch occlu-


sions may produce a clinical syndrome of contra-
lateral hemiparesis and hemianesthesia
affecting the lower face and upper extremity
more than the leg and contralateral visual
field deficits predominantly affecting the lower
fields [30]. For superior division infarcts
involving the dominant hemisphere, global apha-
sia with mutism (aphemia) is seen initially with
eventual progression to nonfluent (expressive)
aphasia [30], while nondominant hemisphere
infarcts are associated with an acute confusional
state and a variable degree of hemispatial
neglect [34].
Inferior division infarcts result in visual field
defects (contralateral homonymous
hemianopia/quadrantanopia) with relative
sparing of sensorimotor involvement [30].
Inferior division branch occlusion of the
MCA supplying the dominant superior
Fig. 8 Eighty-nine-year-old female with new onset recep-
tive aphasia. Diffusion-weighted MRI (DWI) of the brain temporal lobe leads to fluent (receptive) aphasia
obtained 12 h after symptom onset shows an acute infarct without weakness, while infarcts of the
involving the left temporal lobe in the expected vascular nondominant hemisphere superior temporal lobe
distribution of the inferior division of the left middle cere- lead to behavioral disturbances and impairment
bral artery
of visuospatial skills [30]. Sensory aprosody,
probably as a result of damage to the right
posterosuperior temporal lobe, is a common find-
supply enough flow to the main MCA territory ing in nondominant inferior division territory
[32]. Similarly, the duplicated MCA can provide infarcts [35].
collateral flow to the anterior temporal lobe, but it
cannot supply enough flow to the main MCA
territory [32].
Clinical patterns of MCA territory infarcts Posterior Circulation
depend on the location, size, and side of the infarct
[30]. If the entire MCA is occluded at its origin, Intradural Vertebral Artery
clinical findings are contralateral hemiplegia,
hemianesthesia, homonymous hemianopia, and The intradural segment of the vertebral artery/V4
conjugate deviation of the eyes toward the side segment gives rise to the anterior and posterior
of the infarct in the acute phase [30, 33]. When the spinal arteries, the medullary perforating
speech/language-dominant hemisphere is branches, and the posterior inferior cerebellar
affected, severe global aphasia is present; when artery (PICA). Occlusions of the anterior spinal
the nondominant hemisphere is affected, the artery or vertebral (V4) artery result in infarcts of
patient presents with anosognosia, constructional the medial aspect of the medulla (Fig. 10). The
apraxia, and hemineglect [30]. A variant of MCA classic triad of the medial medullary syndrome
stem infarction is malignant infarction, a term (Déjerine-Roussy syndrome) includes contralat-
used to describe those cases with subsequent eral hemiplegia sparing the face, contralateral
extensive brain swelling and herniation loss of posterior column sensation, and ipsilateral
(Fig. 9) [22]. tongue paralysis due to involvement of the
17 Territorial Strokes as a Tool to Learn Vascular Territories 373

Fig. 9 Seventy-one-year-
old man with right
hemiplegia and receptive
aphasia. (a) Diffusion-
weighted MRI (DWI)
obtained 3 h after onset of
symptoms demonstrates an
acute infarct in the territory
of the superior and inferior
divisions of the left middle
cerebral artery.
Non-contrast axial (b) and
coronal (c) head CT
obtained 4 days later for
progressive worsening
mental status in the same
patient demonstrate right
midline shift with
subfalcine and uncal
herniation consistent with
malignant edema

Fig. 10 Fifty-five-year-old
male with acute onset of
nystagmus, right tongue
deviation, left-sided ataxia,
and sensory impairment.
Diffusion-weighted MRI
(DWI) obtained 6 h after
symptom onset
demonstrates restricted
diffusion in the right medial
medulla consistent with an
acute infarct in the expected
vascular distribution of the
intradural vertebral artery

corticospinal tract at the medullary pyramid, the Posterior Inferior Cerebellar Artery
medial lemniscus, and the hypoglossal nucleus or The PICA has a variable vascular territory
nerve [36, 37]. Frequently, however, the clinical depending on the size of the anterior-inferior cer-
syndrome is not confined to the triad and is more ebellar artery (AICA). PICA territory infarcts
heterogeneous [38]. involve the lateral medulla (Fig. 11a) as well as
374 B.A. Vachha and P.W. Schaefer

Fig. 11 (a) Thirty-seven-


year-old female with
Wallenberg syndrome.
Diffusion-weighted MRI
(DWI) obtained 3 h after
onset of symptoms
demonstrates restricted
diffusion in the left lateral
medulla consistent with
acute infarct. (b) Thirty-
three-year-old female with
acute onset of vomiting and
gait unsteadiness. DWI
demonstrates restricted
diffusion consistent with
acute infarct in the inferior
left cerebellar hemisphere.
Both case (a) and case (b)
represent acute infarcts in
the vascular territories of
the posterior inferior
cerebellar artery

the cerebellar tonsil and inferior cerebellar hemi- arteries, and an aberrant course of the distal ver-
sphere (Fig. 11b). Lateral medullary infarcts due tebral artery [41]. Knowledge of these variants is
to occlusion of the PICA result in the lateral important for both endovascular and surgical
medullary (Wallenberg) syndrome character- planning.
ized by ipsilateral limb ataxia, ipsilateral loss of
pain and temperature sensation in the face, loss of
contralateral pain and temperature sensation in the Basilar Artery
body, vomiting, vertigo, nystagmus away from
the lesion side, ipsilateral Horner syndrome, dys- The basilar artery is formed from the union of the
phagia, and hiccups [38, 39]. two vertebral arteries near the pontomedullary
Occlusions of the PICA result in infarcts of the junction. The basilar artery courses within the
inferior cerebellar hemisphere characterized by prepontine cistern and bifurcates within the
vertigo, gait ataxia, limb dysmetria, dysarthria, interpeduncular fossa into the right and left pos-
nausea, and vomiting [40]. If the infarct is large, terior cerebral arteries.
there may be mass effect on the brainstem and The basilar artery gives rise to several cerebel-
hydrocephalus. lar branches; occlusions in these vessels result in
territorial infarctions that are discussed below.
Normal Variants of the Vertebral Artery
Normal variations in the distal vertebral artery Superior Cerebellar Artery
have been described including C1 and C2 origins The superior cerebellar arteries (SCA) arise from
of the PCIA, duplication of the distal vertebral the distal basilar artery and supply the superior
17 Territorial Strokes as a Tool to Learn Vascular Territories 375

Fig. 12 Fifty-seven-year-old male with endocarditis infarct (a) and more inferiorly in the anterior right cerebel-
presenting with right-sided ataxia, dysarthria, nystagmus, lar hemisphere posterior to the temporal bone consistent
nausea, and vomiting. Diffusion-weighted MRI (DWI) with acute right anterior-inferior cerebellar artery infarct
obtained 3 h after onset of symptoms demonstrates (b). Foci of restricted diffusion are also noted within the
restricted diffusion in the right superior cerebellar hemi- right brainstem and right occipital lobe (a) and in the left
sphere consistent with an acute superior cerebellar artery cerebellum (b)

surface of the cerebellar hemispheres, the superior Infarctions of the basilar artery territory result
vermis, and the dentate nuclei (Figs. 1 and 12a). in a wide range of neurologic deficits. Complete
SCA occlusion causes ipsilateral ataxia, dysar- basilar artery occlusion leads to infarction of the
thria, contralateral pain and temperature sensory pons, midbrain, and thalamus. The medial por-
impairment, nystagmus, nausea, and vomiting tions of the temporal, inferior parietal, and occip-
[40]. Horner syndrome may be caused by involve- ital lobes also undergo infarction if the PCAs do
ment of the oculosympathetic fibers [40]. With not receive sufficient collateralization from the
large infarcts, mass effect may lead to compres- posterior communicating arteries. A combination
sion of the midbrain or cause hydrocephalus. of dysarthria, pupillary disorders, lower cranial
Sleep disorders due to lesions involving the nerve involvement, or reduced consciousness at
locus ceruleus have been documented [40]. initial admission appears to be strongly associated
with severe disability or death in the absence of
Basilar Perforating Arteries reperfusion [42].
Basilar perforating arteries arise from the dorsal A constellation of clinical syndromes caused
surface of the midbrain to supply the pons and the by involvement of brainstem nuclei and ascending
midbrain. or descending long tracts can be observed with
incomplete basilar artery occlusion and more
Anterior-Inferior Cerebellar Artery focal infarction. Several named syndromes are
The AICA supplies CN VII and CN VIII as well commonly associated with basilar artery occlu-
as a narrow strip of the cerebellar hemisphere that sions. Occlusions of the proximal and middle
lies directly behind the petrous temporal bone segments of the basilar artery result in infarction
(Figs. 1 and 12b). of the basis pontis with sparing of the tegmen of
the pons (Fig. 13), manifesting as the locked-in
Normal Variants of the Basilar Artery syndrome (quadriplegia with spared level of con-
Basilar artery fenestration is noted in 0.6 % of sciousness, preserved eye movements, and
cases and is most commonly located close to the blinking) [43]. Top of the basilar syndrome
vertebrobasilar junction [14]. (characterized by visual, oculomotor, and
376 B.A. Vachha and P.W. Schaefer

midbrain, subthalamic nucleus, and thalamus.


The vascular territory of the PCA includes the
posterior two-thirds of the medial temporal lobe,
nearly the entire occipital lobe and the medial
inferior parietal lobes, as well as the posterior
one-fifth of the corpus callosum (Fig. 1). The
PComm, arising from the ICA, extends posteri-
orly to join the PCA at the junction of the P1 and
P2 segments, thereby completing the circle of
Willis and providing an important source of col-
lateral circulation for the MCA territory. Infarcts
in the region of the PCA result in homonymous
hemianopia of the contralateral visual field with
macular sparing. Proximal PCA strokes result in
Fig. 13 Seventy-five-year-old male with acute onset of infarcts in the thalamus and/or midbrain as well as
right arm weakness and dysarthria. Diffusion-weighted in the cortex, while distal PCA strokes involve
MRI (DWI) obtained 3 h after onset of symptoms demon-
strates a wedge-shaped acute infarct involving the left
only cortical structures. The following subsec-
paramedian basis pontis with sparing of the tegmen in the tions describe four distinct PCA segments and
expected vascular distribution of the branches of the basilar their branches.
artery
Precommunicating (P1) Segment
behavioral abnormalities, often without signifi- The precommunicating/P1 segment of the PCA
cant motor dysfunction) occurs when there is extends laterally from the basilar artery to the
thromboembolic occlusion of the distal basilar junction with the PComm. Branches arising from
artery with resultant infarction of the rostral the P1 segment of the PCA include thalamoper-
brainstem and/or the posterior cerebral artery ter- forating or paramedian arteries to the thalamus
ritory [44]. Infarctions of the dorsolateral pons and brainstem branches [3, 4].
usually result from occlusion of the AICA (caudal The paramedian arteries have great variability
pons) or SCA (rostral pons) and cause the lateral in terms of the territorial contribution to the thal-
pontine syndrome (ipsilateral limb ataxia, ipsi- amus but principally supply the dorsomedial
lateral loss of pain and temperature sensation of nucleus and internal medullary lamina and
the face, contralateral loss of pain and temperature intralaminar nuclei (central lateral, centromedian,
sensation of the body, ipsilateral Horner syn- and parafascicular nuclei) [6]. When the
drome, and ipsilateral facial paralysis) [45]. tuberothalamic artery is absent, the paramedian
The medial pontine syndrome (contralateral arteries can supply the anterior thalamic areas as
spastic hemiparesis of the body, contralateral well [6, 7]. The artery of Percheron is an
loss of position and vibration of the body, and anatomic variant of the paramedian artery in
medial strabismus) results from occlusion of the which a single dominant paramedian artery sup-
paramedian perforating branches with infarction plies the bilateral medial thalami with variable
of the medial pons [45]. contribution to the midbrain [4]. Specific ischemic
patterns and syndromes that result from occlusion
of the artery of Percheron are discussed in detail in
Posterior Cerebral Artery a later section.
Unilateral thalamic infarction in the territory
The PCAs are paired vessels arising from the of the paramedian artery is characterized by
basilar artery. The PCAs supply parts of the deficits in arousal and memory. In the early
17 Territorial Strokes as a Tool to Learn Vascular Territories 377

stages, patients demonstrate decreased and fluctu-


ating levels of consciousness, while impaired
arousal with confusion, agitation, and apathy are
seen in later stages of the infarct [7]. Left-sided
lesions result in language and speech impairments
characterized by hypophonia and dysprosody
with reduced verbal fluency and perseveration.
Syntactic structure is preserved with occasional
paraphasic errors and normal repetition [6]. Right-
sided infarcts result in visuospatial deficits.
Bilateral paramedian thalamic strokes are typ-
ically characterized by altered mental status,
memory impairment, and vertical gaze palsy
[6, 46]. Altered mental status may present on a
spectrum ranging from disorientation and
hypersomnolence to deep coma and akinetic mut-
ism [6]. Anterograde and retrograde memory Fig. 14 Fifty-year-old male presenting with
impairment with confabulation tends to resolve prosopagnosia. Diffusion-weighted MRI (DWI) obtained
with time but may be severe and persistent. In 15 h after symptom onset demonstrates restricted diffusion
in the right occipital and medial temporal lobes consistent
the late stages, the patient may demonstrate emo- with acute right posterior cerebral artery territory infarct
tional blunting, loss of initiative, and inappropri-
ate social behaviors.
Occlusion of the P1 segment of the PCA that The medial branch supplies the external half of
supplies the midbrain can result in a CN III palsy the medial geniculate nucleus. The principal
or contralateral ataxia or hemiplegia [47]. inferolateral arteries supply the major part of the
ventral posterior nuclei (lateral, medial, and infe-
Ambient (P2) Segment rior nuclei) as well as the ventral and lateral parts
The medial and lateral posterior choroidal of the ventrolateral nucleus. The inferolateral
arteries arise from the P2 segment. The lateral pulvinar arteries supply the rostral and lateral
posterior choroidal artery shares a reciprocal rela- parts of the pulvinar and lateral dorsal nucleus [6].
tionship with the anterior choroidal artery (which The Déjerine-Roussy syndrome or thalamic
arises from the ICA) [4]. pain syndrome occurs following infarctions
The anterior and posterior temporal arter- related to the inferolateral arteries and is charac-
ies supply the inferior surface of the temporal terized by contralateral sensory loss followed by
lobe, while the thalamogeniculate and pedun- agonizing burning pain [6, 47, 48]. This syndrome
cular perforating arteries supply the midbrain is usually associated with infarcts of the right
[4]. Occlusion of the P2 segment causes visual thalamus.
field defects and more complex visual changes
such as prosopagnosia secondary to infarction of Quadrigeminal (P3) Segment
the medial temporal and occipital lobes The quadrigeminal/P3 segment is a short segment
(Fig. 14) [47]. without any significant named branches [4].
The inferolateral arteries comprise five to ten
arteries that arise from the P2 segment of the PCA Calcarine (P4) Segment
and supply the thalamus [6]. There are three main The calcarine/P4 segment divides into the medial
groups: the medial geniculate, principal and lateral trunks within the calcarine fissure.
inferolateral, and inferolateral pulvinar arteries. The medial trunk gives off medial occipital,
378 B.A. Vachha and P.W. Schaefer

Fig. 15 Sixty-nine-year-
old male with waxing and
waning mental status
progressively worsening
over the course of 3 days.
Diffusion-weighted MRI
demonstrates foci of
restricted diffusion within
the medial aspects of the
right and left thalami (a)
and medial midbrain (b)
consistent with an acute
artery of Percheron infarct

parieto-occipital, calcarine, and posterior the PCA has been noted but is a very rare
splenial arteries which supply the regions of the occurrence [14].
brain represented in their names [4]. The lateral
trunk gives rise to the lateral occipital artery [4].
Border Zones
Artery of Percheron
The central artery of Percheron is a rare but clin- Border zones occur at the junction between two
ically important PCA variant. This occurs when vascular territories. Two distinct border zone or
the bilateral, medial thalamic/rostral midbrain per- watershed territories are defined: (1) External
forators arise from a single trunk from one P1 (cortical) border zone territories include the
segment [4]. Occlusion may result in four distinct frontal and parietal border zone territory between
patterns of infarction in descending order of fre- the ACA and MCA territories and the posterior
quency: bilateral, paramedian, thalamic, and ros- parieto-occipital border zone territory between the
tral midbrain infarction (Fig. 15); bilateral, MCA and PCA territories. (2) Internal (subcor-
paramedian, and thalamic without midbrain tical) border zone territories include the border
involvement; bilateral, paramedian, and anterior zones in the corona radiata and centrum semiovale
thalamic with midbrain involvement; and bilat- between the penetrating branches (lenticulostriate
eral, paramedian, and anterior thalamic without and medullary perforating and anterior choroidal
midbrain involvement [46]. In addition to these arteries) and the main cerebral arteries (ACA,
four distinct patterns, Lazzaro et al. described a MCA, and PCA) [4]. Figure 16 is an example of
V-shaped hyperintense signal abnormality on diffusion-weighted MRI sequences demonstrat-
axial FLAIR images along the pial surface of the ing restricted diffusion consistent with border
midbrain in the interpeduncular fossa in their zone infarcts.
series of 37 patients with occlusions of the artery Clinical manifestations of patients with border
of Percheron [46]. Infarctions associated with zone infarcts depend upon the location of the
artery of Percheron occlusions produce long- lesion. Patients with unilateral precentral cortico-
lasting memory dysfunction, deficits in executive subcortical infarcts present with somnolence, bra-
function, and mood changes [49]. chial monoparesis, disproportionate motor
hemiparesis with face mostly spared, transcortical
Normal Variants of the Posterior Cerebral motor aphasia, and focal myoclonic jerks
Arteries [50]. Unilateral, postcentral cortico-subcortical
Fetal origin of the posterior cerebral artery has infarcts result in sensorimotor hemiparesis with
been described under normal variants of the stereoagnosia, cortical hemihypesthesia, visual
ICA. Fenestration of the P1 and P2 segments of and tactile hemispatial neglect, anosognosia,
17 Territorial Strokes as a Tool to Learn Vascular Territories 379

Summary

Neuroimaging plays a key role in the initial


workup of stroke patients. Knowledge of arterial
territories and their associated stroke syndromes is
needed to achieve accurate localization of ische-
mic lesions to guide further management.

References
1. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry
JD, Blaha MJ, Dai S, Ford ES, Fox CS, Franco S,
Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, How-
ard VJ, Huffman MD, Judd SE, Kissela BM, Kittner SJ,
Lackland DT, Lichtman JH, Lisabeth LD, Mackey RH,
Magid DJ, Marcus GM, Marelli A, Matchar DB,
McGuire DK, Mohler ER 3rd, Moy CS, Mussolino
ME, Neumar RW, Nichol G, Pandey DK, Paynter NP,
Reeves MJ, Sorlie PD, Stein J, Towfighi A, Turan TN,
Virani SS, Wong ND, Woo D, Turner MB (2014) Heart
disease and stroke statistics – 2014 update: a report from
Fig. 16 Eighty-seven-year-old man with atrial fibrillation the american heart association. Circulation 129(3):
with new onset right hemiplegia. Diffusion-weighted MRI e28–e292. doi:10.1161/01.cir.0000441139.02102.80,
(DWI) obtained 4 h after symptom onset demonstrates 01.cir.0000441139.02102.80 [pii]
bilateral border zone infarcts between the anterior cerebral 2. Grysiewicz RA, Thomas K, Pandey DK (2008) Epide-
artery and middle cerebral artery, presumed to be miology of ischemic and hemorrhagic stroke: inci-
cardioembolic in origin dence, prevalence, mortality, and risk factors. Neurol
Clin 26(4):871–895. doi:10.1016/j.ncl.2008.07.003,
S0733-8619(08)00104-7 [pii]
3. Osborn A (1999) Diagnostic cerebral angiography.
hemianopia particularly involving the lower Lippincott Williams & Wilkins, Philadelphia
quadrant, Wernicke-like aphasia, and focal myo- 4. Osborn A (2013) Osborn’s brain: imaging, pathology,
and anatomy. Amirsys, Manitoba
clonic jerks [50]. Patients with deep border zone 5. Joo W, Funaki T, Yoshioka F, Rhoton AL Jr (2012)
infarcts in the dominant hemisphere develop Microsurgical anatomy of the carotid cave. Neurosur-
global aphasia, nonfluent aphasia with or without gery 70(2 Suppl Operative):300–311. doi:10.1227/
comprehension and/or repetition deficits, ideomo- NEU.0b013e3182431767; discussion 311–302
6. Schmahmann JD (2003) Vascular syndromes of the
tor apraxia, and acalculia. Patients with internal thalamus. Stroke 34(9):2264–2278. doi:10.1161/01.
border zone infarctions in the nondominant hemi- STR.0000087786.38997.9E, 01.STR.0000087786.
sphere demonstrate anosognosia, neglect, or 38997.9E [pii]
dysprosody depending on the extent of the lesion. 7. Bogousslavsky J, Regli F, Assal G (1986) The syn-
drome of unilateral tuberothalamic artery territory
Bilateral infarcts in the border zone regions infarction. Stroke 17(3):434–441
between the MCA and PCA can result in 8. von Cramon DY, Hebel N, Schuri U (1985) A contri-
asimultagnosia (difficulty recognizing several bution to the anatomical basis of thalamic amnesia.
objects at the same time), optical ataxia (difficulty Brain 108(Pt 4):993–1008
9. Ghika-Schmid F, Bogousslavsky J (2000) The acute
coordinating hand and eye movements), and behavioral syndrome of anterior thalamic infarction: a
apraxia of gaze [50]. Bilateral infarcts in the bor- prospective study of 12 cases. Ann Neurol 48(2):
der zone regions between the ACA and MCA can 220–227
result in selective weakness of the upper and 10. Hupperts RM, Lodder J, Heuts-van Raak EP, Kessels F
(1994) Infarcts in the anterior choroidal artery territory.
lower extremities with sparing of the face, Anatomical distribution, clinical syndromes, presumed
hands, and feet referred to as “a man in a barrel” pathogenesis and early outcome. Brain 117(Pt 4):
distribution [50]. 825–834
380 B.A. Vachha and P.W. Schaefer

11. Ois A, Cuadrado-Godia E, Solano A, Perich-Alsina X, of distal medial striate artery (recurrent artery of
Roquer J (2009) Acute ischemic stroke in anterior Heubner). Surg Radiol Anat 34(1):15–20.
choroidal artery territory. J Neurol Sci 281 doi:10.1007/s00276-011-0888-5
(1–2):80–84. doi:10.1016/j.jns.2009.02.323, S0022- 26. Brust J (2012) Anterior cerebral artery. In: Caplan L,
510X(09)00055-0 [pii] Van Gijn J (eds) Stroke syndromes, 3rd edn. Cam-
12. Foix C, Chavany J, Hillemand P, Schiff-Wertheimer S bridge University Press, Cambridge, UK, pp 364–374
(1925) Obliteration de l’arte’re choroidienne 27. Baptista AG (1963) Studies on the arteries of the brain.
ante’rieure: ramollissement de son territoire ce’re’bral: Ii. The anterior cerebral artery: some anatomic features
he’miple’gie, he’mianesthe’sie, he’mianopsie. Bull and their clinical implications. Neurology 13:825–835
Soc Ophtalmol Paris 37:221–223 28. Bogousslavsky J, Martin R, Moulin T (1992)
13. Helgason C, Caplan L (2012) Anterior choroidal artery Homolateral ataxia and crural paresis: a syndrome of
territory strokes. In: Caplan L, Van Gijn J (eds) Stroke anterior cerebral artery territory infarction. J Neurol
syndromes, 3rd edn. Cambridge University Press, Neurosurg Psychiatry 55(12):1146–1149
Cambridge, UK, pp 375–386 29. Hiraga A, Uzawa A, Tanaka S, Ogawara K,
14. Dimmick SJ, Faulder KC (2009) Normal variants of Kamitsukasa I (2009) Pure monoparesis of the leg
the cerebral circulation at multidetector CT angiogra- due to cerebral infarctions: a diffusion-weighted imag-
phy. Radiographics 29(4):1027–1043. doi:10.1148/ ing study. J Clin Neurosci 16(11):1414–1416.
rg.294085730, 29/4/1027 [pii] doi:10.1016/j.jocn.2009.01.015, S0967-5868(09)
15. Schwarz S, Schwab S, Hacke W (2002) Large and 00156-8 [pii]
panhemispheric infarcts. In: Bogousslavsky J, Caplan 30. Mohr J, Kejda-Scharler J (2012) Middle cerebral artery
L (eds) Stroke syndromes, 2nd edn. Cambridge Uni- territory syndromes. In: Caplan L, Van Gijn J (eds)
versity Press, Cambridge, UK, pp 490–498 Stroke syndromes, 3rd edn. Cambridge University
16. Hyun DK, Shim YS, Park HS, Oh SY (2014) Throm- Press, Cambridge, UK, pp 344–363
boembolic complication following neurointervention 31. Gibo H, Carver CC, Rhoton AL Jr, Lenkey C, Mitchell
in ruptured anomalous hyperplastic anterior choroidal RJ (1981) Microsurgical anatomy of the middle cere-
artery aneurysm. Neuroradiol J 27(1):103–107, doi:IN. bral artery. J Neurosurg 54(2):151–169. doi:10.3171/
v27.i1.p103 [pii] jns.1981.54.2.0151
17. Caldemeyer KS, Carrico JB, Mathews VP (1998) The 32. Komiyama M, Nakajima H, Nishikawa M, Yasui T
radiology and embryology of anomalous arteries of the (1998) Middle cerebral artery variations: duplicated
head and neck. AJR Am J Roentgenol 170(1): and accessory arteries. AJNR Am J Neuroradiol
197–203. doi:10.2214/ajr.170.1.9423632 19(1):45–49
18. Salas E, Ziyal IM, Sekhar LN, Wright DC (1998) 33. Mohr JP, Foulkes MA, Polis AT, Hier DB, Kase CS,
Persistent trigeminal artery: an anatomic study. Neuro- Price TR, Tatemichi TK, Wolf PA (1993) Infarct topog-
surgery 43(3):557–561; discussion 561–552 raphy and hemiparesis profiles with cerebral convexity
19. Saltzman GF (1959) Patent primitive trigeminal artery infarction: the Stroke Data Bank. J Neurol Neurosurg
studied by cerebral angiography. Acta Radiol 51(5): Psychiatry 56(4):344–351
329–336 34. Mori E, Yamadori A (1987) Acute confusional state
20. Morita A, Fukushima T, Miyazaki S, Shimizu T, and acute agitated delirium. Occurrence after infarction
Atsuchi M (1989) Tic douloureux caused by primitive in the right middle cerebral artery territory. Arch
trigeminal artery or its variant. J Neurosurg 70(3): Neurol 44(11):1139–1143
415–419. doi:10.3171/jns.1989.70.3.0415 35. Darby DG (1993) Sensory aprosodia: a clinical clue to
21. Wijdicks EF, Diringer MN (1998) Middle cerebral lesions of the inferior division of the right middle
artery territory infarction and early brain swelling: pro- cerebral artery? Neurology 43(3 Pt 1):567–572
gression and effect of age on outcome. Mayo Clin Proc 36. Kim JS, Han YS (2009) Medial medullary
73(9):829–836. doi:10.4065/73.9.829, S0025-6196 infarction: clinical, imaging, and outcome study in 86 con-
(11)63788-9 [pii] secutive patients. Stroke 40(10):3221–3225. doi:10.
22. Hacke W, Schwab S, Horn M, Spranger M, De 1161/STROKEAHA.109.559864, STROKEAHA.109.
Georgia M, von Kummer R (1996) ‘Malignant’ middle 559864 [pii]
cerebral artery territory infarction: clinical course and 37. Dejerine J (1914) Semiologie des affections du
prognostic signs. Arch Neurol 53(4):309–315 systeme nerveux. Masson, Paris
23. Krieger DW, Demchuk AM, Kasner SE, Jauss M, 38. Kim J (2012) Medullary infarcts and hemorrhages. In:
Hantson L (1999) Early clinical and radiological pre- Caplan L, Van Gijn J (eds) Stroke syndromes, 3rd edn.
dictors of fatal brain swelling in ischemic stroke. Cambridge University Press, Cambridge, UK, pp
Stroke 30(2):287–292 461–468
24. Bogousslavsky J, Caplan L (eds) (2002) Stroke syn- 39. Kim JS (2003) Pure lateral medullary infarction:
dromes. Cambridge University Press, New York clinical-radiological correlation of 130 acute, consecu-
25. Zunon-Kipre Y, Peltier J, Haidara A, Havet E, tive patients. Brain 126(Pt 8):1864–1872. doi:10.1093/
Kakou M, Le Gars D (2012) Microsurgical anatomy brain/awg169, awg169 [pii]
17 Territorial Strokes as a Tool to Learn Vascular Territories 381

40. Mazighi M, Amarenco P (2012) Cerebellar infarcts. In: 46. Lazzaro NA, Wright B, Castillo M, Fischbein NJ,
Caplan L, Van Gijn J (eds) Stroke syndromes, 3rd edn. Glastonbury CM, Hildenbrand PG, Wiggins RH,
Cambridge University Press, Cambridge, UK, pp Quigley EP, Osborn AG (2010) Artery of percheron
469–479 infarction: imaging patterns and clinical spectrum.
41. Siclari F, Burger IM, Fasel JH, Gailloud P (2007) Devel- AJNR Am J Neuroradiol 31(7):1283–1289.
opmental anatomy of the distal vertebral artery in rela- doi:10.3174/ajnr.A2044, ajnr.A2044 [pii]
tionship to variants of the posterior and lateral spinal 47. Chaves C, Caplan L (2012) Posterior cerebral artery.
arterial systems. AJNR Am J Neuroradiol 28 In: Caplan L, Van Gijn J (eds) Stroke syndromes, 3rd
(6):1185–1190. doi:10.3174/ajnr.A0498, 28/6/1185 [pii] edn. Cambridge University Press, Cambridge, UK, pp
42. Baird TA, Muir KW, Bone I (2004) Basilar artery 405–418
occlusion. Neurocrit Care 1(3):319–329. doi:10.1385/ 48. Dejerine J, Roussy G (1906) Le syndrome thalamique.
NCC:1:3:319, NCC:1:3:319 [pii] Rev Neurol 14:521–532
43. Smith E, Delargy M (2005) Locked-in syndrome. BMJ 49. Jodar M, Martos P, Fernandez S, Canovas D, Rovira A
330(7488):406–409. doi:10.1136/bmj.330.7488.406 (2011) Neuropsychological profile of bilateral paramedian
330/7488/406, 330/7488/406 [pii] infarctions: three cases. Neurocase 17(4):345–352.
44. Caplan LR (1980) “Top of the basilar” syndrome. doi:10.1080/13554794.2010.509324, 931811693 [pii]
Neurology 30(1):72–79 50. Ringelstein EB, Dittrich R, Stogbauer F (2012)
45. Kumral E, Bayulkem G, Evyapan D (2002) Clinical Borderzone infarcts. In: Caplan L, van Gijn J (eds)
spectrum of pontine infarction. Clinical-MRI correla- Stroke syndromes, 3rd edn. Cambridge University
tions. J Neurol 249(12):1659–1670. doi:10.1007/ Press, Cambridge, UK, pp 480–500
s00415-002-0879-x
Hemorrhagic Stroke
18
Julius Griauzde, Elliot Dickerson, and Joseph J. Gemmete

Contents Magnetic Resonance Angiography


of Intracranial Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . 391
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 384
Time of Flight . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 392
Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 385
Time of Flight: Pitfalls in Evaluating Causes
Non-contrast Computed Tomography in of Intracranial Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . 393
Intracranial Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . 385
Contrast-Enhanced MRA . . . . . . . . . . . . . . . . . . . . . . . . . . 393
NCCT Technical Considerations and Pitfalls . . . . 386
Contrast-Enhanced MRA: Pitfalls in
Magnetic Resonance Imaging in Intracranial Evaluating Causes of Intracranial
Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 387 Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394
Magnetic Susceptibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . 387 MRA: Aneurysms, AVMs, DAVF,
and Neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394
MRI of Intracranial Hemorrhage:
Biologic Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 388 Imaging Patterns of Intraparenchymal
Hemoglobin Structure, Iron, and Oxygenation . . . . . . 388 Hemorrhage (ICH) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394
Plasma, Red Blood Cell Membranes, and Blood Hemorrhage Etiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394
Breakdown Products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 389 Hypertensive Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . 395
Illicit Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 395
MRI of Intracranial Hemorrhage: Technical Cerebral Amyloid Angiopathy . . . . . . . . . . . . . . . . . . . . . . 396
Considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 390 Vascular Malformations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 397
Arteriovenous Malformations (AVMs) . . . . . . . . . . . . . . 397
CT Angiography of Intracranial Hemorrhage . . . 391
Cavernous Malformations (CMs) . . . . . . . . . . . . . . . . . . . 397
CTA: Pitfalls in Evaluating Causes Dural Arteriovenous Malformations (DAVFs) . . . . . . 397
of Intracranial Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . 391 Aneurysm . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 397
Hemorrhagic Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 398
Hemorrhagic Transformation of Ischemic Stroke . . . 398
Venous Thrombosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 399
Trauma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 399
Additional Clinical Topics in ICH . . . . . . . . . . . . . . . . . 400
J. Griauzde (*) • E. Dickerson Active Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 400
Department of Radiology, Radiology Resident, University Intraventricular Extension, Associated
of Michigan, Ann Arbor, MI, USA Subarachnoid Hemorrhage, and Hydrocephalus . . . . 400
e-mail: jgriauz@med.umich.edu; Hemorrhage Location . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 402
dickerse@med.umich.edu Predicting Clinical Outcome . . . . . . . . . . . . . . . . . . . . . . . . . 402
Evolution of the Intraparenchymal Hematoma . . . . . . 403
J.J. Gemmete
Department of Radiology, Neurosurgery, and
Otolaryngology, University of Michigan, Ann Arbor,
MI, USA
e-mail: gemmete@med.umich.edu

# Springer Science+Business Media New York 2016 383


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_46
384 J. Griauzde et al.

Extra-axial Central Nervous System Abstract


Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 404 Neuroimaging is integral in the diagnostic
Subarachnoid Hemorrhage (SAH) . . . . . . . . . . . . . . . . . . . 404
Nonaneurysmal Perimesencephalic
algorithm and follow-up of patients with hem-
SAH (NPSAH) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 405 orrhagic stroke. The role of the neuroimager is
Subdural and Epidural Hematomas . . . . . . . . . . . . . . . . . . 405 to recognize and localize the hemorrhage and
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 407 attempt to determine its etiology. Non-contrast
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 407 computed tomography has long been the initial
imaging tool in the acute neurologic patient. As
MRI technology and angiographic imaging has
evolved, they too have proven to be beneficial
in narrowing the differential diagnosis and
triaging patient care. Several biological and
physical characteristics contribute significantly
to the appearance of blood products on neuro-
imaging. To adequately interpret images in the
patient with hemorrhagic stroke, the evaluator
must have a knowledge of the interplay
between imaging modalities and intracranial
blood products. Additionally, an understand-
ing of technical parameters as well as the lim-
itations of imaging modalities can be helpful in
avoiding pitfalls. Recognition of typical imag-
ing patterns and clinical presentations can fur-
ther aid the evaluator in rapid diagnosis and
directed care.

Keywords
Hemorrhagic stroke • Hemoglobin • Methemo-
globin • CT angiography • MR angiography •
Time of flight • Hypertension • Amyloid
angiopathy • Venous thrombosis • Aneurysm
• Arteriovenous malformation • Dural arterio-
venous fistula • Subarachnoid hemorrhage •
Subdural • Epidural

Introduction

Neuroimaging is undergoing a second revolution


in terms of understanding disease mechanisms
and therapeutics, fueled by rapid advancements
in advanced computed tomography (CT) and
magnetic resonance imaging (MRI). The accurate
identification and characterization of intracranial
hemorrhage is one of the most essential functions
of a neuroradiologist, because immediate
18 Hemorrhagic Stroke 385

recognition is important for further diagnostic Non-contrast Computed Tomography


work-up, clinical management, and ultimately in Intracranial Hemorrhage
patient outcome. CT and MRI are important for
the identification of intracranial hemorrhage, and Since its inception as a diagnostic tool,
MRI can further characterize the age based on non-contrast computed tomography (NCCT) has
specific signal characteristics. In this chapter, clin- been a highly sensitive first-line screening exami-
ical presentation will be discussed first, followed nation in the diagnosis of intracranial hemorrhage
by the physics and technical limitations of CT, [3, 4]. The inherent contrast between the CT atten-
MRI, computed tomography angiography (CTA), uation of acute blood products and brain paren-
and magnetic resonance angiography (MRA) in chyma allows for a straightforward diagnosis for
detecting intracranial hemorrhage. Then, specific the trained evaluator. Similarly, the speed, rela-
clinical patterns of intracranial hemorrhage will be tively low cost, and widespread availability of CT
presented, so the neuroradiologist can make an have preserved its place in the diagnostic algorithm
accurate diagnosis of the probable cause based on of the acute neurologic patient. Because no contrast
clinical history and imaging findings. Finally, the is needed to identify intracranial hemorrhage, there
CT and MRI findings of extra-axial central nervous is virtually no risk to the patient, although the small
system hemorrhage will be discussed, including associated radiation dose should be a cause of
subarachnoid hemorrhage (SAH) and subdural additional consideration in pediatric patients.
and epidural hematomas. The appearance of blood (or any other sub-
stance) on CT relates to its ability to attenuate
x-rays. This property is defined as substances’
Clinical Presentation attenuation coefficient. In clinical practice, the
CT attenuation of a substance is described by its
Rapid identification of intracranial hemorrhage Hounsfield density. Hounsfield density values
(ICH) is essential for the clinical management of (HU) range from 1,000 (air) to +1,000 (bone)
ischemic and hemorrhagic stroke, since this will with a center at zero (water) [5]. The attenuation
affect treatment algorithms. Both ischemic and of white matter ranges between approximately
hemorrhagic stroke are characterized by a sudden 25 and 34 HU and that of gray matter from 30 to
onset of symptoms and neurological deficits. The 40 HU [6]. Gray matter values can normally
severity and type of symptoms depend on the extend as high as 45 HU. The CT attenuation of
lesion type, location, and size. intracranial blood varies linearly with the hemat-
Patients with subarachnoid hemorrhage (SAH) ocrit value, and hemoglobin concentration is the
will typically describe “the worst headache of my most significant contributor to the attenuation of
life” prior to presenting with hemorrhage. One in an intracranial hematoma [7, 8]. Intracranial blood
five may report a less severe sentinel headache in in a patient with normal hematocrit can be
hours or days leading up to the ictus. This also expected to range between 30 and 60 HU imme-
may be accompanied by nausea/vomiting, sei- diately after the hemorrhagic event (Fig. 1). As
zure, loss of consciousness, and focal neurologi- time progresses, the attenuation of intracranial
cal signs. In contrast, ICH is classically described blood products will evolve in a somewhat predict-
as a gradual process with worsening symptoms able manner. In the case of intraparenchymal
over minutes to hours. Headache and nausea/ hematoma, the Hounsfield attenuation of the col-
vomiting are more frequent in ICH than ischemic lection will increase (particularly centrally) as clot
stroke. Given the overlap in the clinical presenta- forms and serum is extruded [9]. This process will
tion between ischemic and hemorrhagic stroke, occur in the first hours and days after the hemor-
imaging is vital for determining the etiology and rhagic event with HU values ranging from 60 to
clinical management of patients, particularly in 80. Higher values extending up to 90–100 HU can
the early presentation [1, 2]. be expected in the center of the hematoma with
386 J. Griauzde et al.

associated mass effect influences immediate treat-


ment planning, and the volume of the hematoma
has been identified as an important predictor of a
30-day patient outcome [14].

NCCT Technical Considerations


and Pitfalls

A simplified description of technical imaging con-


siderations is included to highlight solutions for
some common pitfalls that can be encountered.
Axial images using 5 mm reconstructions are gen-
erally used in NCCT of intracranial hemorrhage, as
this thickness has an adequate signal-to-noise ratio
to allow for evaluation of the brain parenchyma,
Fig. 1 Hounsfield densities. Cerebrospinal fluid has an identification of intracranial hemorrhage, and
HU of 0 (white arrow). White matter has an HU of 25–34
assessment of intracranial mass effect. Unfortu-
(open white arrow). Gray matter has an HU of 30–40
(white arrowhead). Acute blood has an HU of 60–80 nately, in some cases intracranial hematoma can
(black arrowhead). Bone has an HU near 1000 (open be obscured by nearby skeletal structures or be
black arrow) mistaken for dural prominence or calcifications
[15]. Reviewing a wider “blood” window can
low attenuation surrounding the hematoma improve the detection of intracranial hemorrhage
related to extruded serum and edema. which layers along the skull (Fig. 2a, b). Review of
At the midpoint of the first week after the hem- coronal and sagittal reformatted images is also par-
orrhagic event, the blood collection will begin to amount in all cases but particularly when findings
decrease in attenuation converging from the periph- of hemorrhage are equivocal. Reformatted images
ery centrally. The density is estimated to decrease by allow for visualization parallel and/or perpendicu-
approximately 1–2 HU per day [10]. This decrease lar to the long axis of the hematoma allowing for
in density is attributable to the breakdown of red improved diagnostic accuracy [16] (Fig. 3a, b).
blood cells and subsequent removal of the compo- Similarly, thin-cut images may help resolve any
nents of the hematoma, most importantly hemoglo- questionable areas (Fig. 2c). In the interest of
bin, by phagocytic macrophages [10]. This process time, thin section cuts should only be created and
leads to an inhomogeneous appearance which is reviewed when there is a specific area of concern.
mildly hyperdense to isodense to brain parenchyma Review of bone algorithm reconstructions is also
(HU approximately 30–50) in the subacute period suggested (particularly in cases of trauma) for iden-
(up to about 30 days) [11, 12]. After 30 days, the tification of skull fractures which will heighten the
collection is expected to be hypodense to brain readers’ suspicion of underlying intracranial hem-
parenchyma (<30 HU) [10, 11]. orrhage and help to exclude mimickers of hemor-
In addition to the initial identification of intra- rhage like small calcifications (Fig. 3c).
cranial hemorrhage, NCCT helps triage the care of It is important to note that in the first minutes
the patient presenting with intracranial hemor- after hemorrhage, there is a partial overlap
rhage. Identification of the location of the hema- between the HU of blood products and that of
toma, when taken together with the clinical brain parenchyma, creating a potential pitfall in
picture, aids in narrowing the differential diagno- diagnosis. Similarly, intracranial hemorrhage can
sis and deciding on additional imaging [13]. Eval- remain occult in patients with low hematocrit
uating the extent of the hematoma and the because of isodensity to brain parenchyma [17].
18 Hemorrhagic Stroke 387

Fig. 2 (a) Focus of extra-axial hemorrhage layering along “blood” window (black arrow). (c) Focus of hemorrhage
the left frontal region is poorly seen with standard is made even more conspicuous using thin-cut slices (black
windowing secondary to adjacent skull (black arrow). (b) arrow). Note the increased noise associated with the
Focus of hemorrhage is more conspicuous with wider thinner cut

Fig. 3 (a) Subtle focus of hemorrhage along the posterior (white arrow). (c) Bone algorithm window shows a skull
skull is very difficult to identify (black arrow). (b) Sagittal fracture, heightening the reader’s attention for subtle hem-
image in wider blood window allows visualization parallel orrhage (black arrow)
to the long axis of the hemorrhage increasing conspicuity

In such cases, and in those where there is a high understanding of magnetic properties as well as
index of suspicion for intracranial hemorrhage the biologic and technical factors which affect the
without definite CT abnormality, MRI should be appearance of intracranial blood products aids in
recommended for further evaluation. overcoming confusion and avoiding potential pit-
falls. What follows is a description of the key
factors involved and their effects on the appear-
Magnetic Resonance Imaging ance on intracranial blood products.
in Intracranial Hemorrhage

The evaluation of blood products on magnetic Magnetic Susceptibility


resonance imaging (MRI) is much more involved
than on CT. The appearance depends on a myriad Magnetic susceptibility refers to the measure of
of biologic factors which evolve with time and substances’ interaction and response to an applied
vary based on operator-dependent factors. A basic magnetic field [18, 19]. In simplified terms, the
388 J. Griauzde et al.

magnetic susceptibility of a substance can be Table 1 Purely diamagnetic and paramagnetic substances
attributed to the pairing of its valence electrons Purely diamagnetic Paramagnetic
[19]. Those substances which have no unpaired Water Deoxyhemoglobin
valence electrons are termed diamagnetic. Paired Oxyhemoglobin Methemoglobin
electrons in an orbital have opposing spins which Most soft tissue Hemosiderin/ferritin
cancel each other, resulting in no magnetic dipole Bone Gadolinium
when placed in a magnetic field [19]. When a
diamagnetic substance is placed in an applied
magnetic field, its valence electrons do experience is a brief description of these processes and the
minimal alterations in their orbital motion, how- resultant effects they have on the MRI signal
ever [19]. According to Lenz’s law, this results in produced.
a weak induced magnetic field which opposes the
applied field [20].
In contrast to diamagnetic substances, para- Hemoglobin Structure, Iron,
magnetic substances have unpaired valence elec- and Oxygenation
trons. When placed in a magnetic field, the spins
of the valence electrons align with the magnetic Hemoglobin is a metalloprotein composed of four
field creating a magnetic dipole which augments protein moieties, each of which contains a heme
the applied field [19, 20]. In human tissues, the subunit. Each heme subunit is composed of a
magnetic dipole of paramagnetic substances porphyrin ring with a central iron atom which
allows them to undergo proton-electron dipole can reversibly bind oxygen [25] (Fig. 4). In oxy-
interactions with local protons (i.e., those in hemoglobin, an iron atom is reversibly bound to
water molecules) [21]. These interactions have a oxygen in the ferrous state (+2). Oxyhemoglobin
profound effect on the magnetic microenviron- has no unpaired outer electrons and is therefore
ments of human tissues, most significantly affect- diamagnetic [26].
ing the longitudinal relaxation of nearby protons, In oxygen-poor environments (i.e., the intra-
resulting in T1 shortening [20, 22]. For this dis- cranial hematoma), hemoglobin releases its bound
cussion, it is important to note that the strength of oxygen leaving deoxyhemoglobin with four
dipole interactions varies inversely with the sixth unpaired electrons and the paramagnetic proper-
power of the distance between the dipoles [23]. ties that come with them [26]. From the discussion
Also, as magnetic field strength increases, above, this would lead the reader to believe that
paramagnetic properties become more pro- the presence of deoxyhemoglobin in human tis-
nounced because paramagnetic substances aug- sues would result in dipole interactions with con-
ment the applied field more so than diamagnetic sequential T1 shortening. The magnetic properties
substances oppose it [24]. Relevant compounds of deoxyhemoglobin are more complex, however.
are classified based on their magnetic susceptibil- For dipole interactions to occur between the
ity in Table 1. valence electrons of iron and the protons of
water molecules, they must be able to approach
within 3 Å of each other [23]. The loss of oxygen
MRI of Intracranial Hemorrhage: from hemoglobin causes a shift in its three-
Biologic Factors dimensional configuration, displacing the iron
atom of the heme subunit out of the plane of the
Several biologic factors contribute to the charac- porphyrin ring [25, 27]. This shift effectively
teristic appearance of intracranial blood products increases the dipolar distance between iron and
on MRI. As time passes, intracranial blood prod- nearby water proton dipoles, preventing any sig-
ucts are exposed to metabolic processes and var- nificant dipolar interactions [21, 27] (Fig. 5). The
iations in their local environment which alter their result is a lack of marked T1 shortening in the
chemical and magnetic properties. What follows presence of deoxyhemoglobin.
18 Hemorrhagic Stroke 389

With further exposure to an oxygen-poor present, there is a significant T1 shortening in the


environment, deoxyhemoglobin is oxidized into intracranial hematoma.
methemoglobin whose iron atom is in the Ferric As time progresses, methemoglobin is further
state (+3) and unable to bind oxygen [25]. The oxidized, metabolized, and phagocytosed yield-
associated conformational change decreases the ing residual iron atoms which are scavenged and
dipolar distance between the unpaired valence stored as hemosiderin and ferritin [29]. The scav-
electrons of iron and nearby water protons, enged iron is paramagnetic, but because of the
allowing for proton-electron dipole interactions structures of hemosiderin and ferritin, water can-
[22, 26, 28]. Therefore, when methemoglobin is not closely approach it and proton-electron dipo-
lar interactions are not possible [20, 30–32].

Plasma, Red Blood Cell Membranes,


and Blood Breakdown Products

Freshly extruded plasma (prior to clot formation)


has high water content, admixed serum proteins,
and intact red blood cells. The high water content
of extruded plasma causes T2 prolongation com-
pared to brain parenchyma [33]. As time pro-
gresses, clot begins to form, water is resorbed,
and serum proteins are concentrated with subse-
quent clot retraction. These processes all have
minor contributions to T2 shortening of the evolv-
Fig. 4 The heme subunit of hemoglobin ing hematoma [34]. Red blood cell (RBC)

Fig. 5 (a) In plane view of the oxyhemoglobin molecule. created using graphics reproduced under the GNU Free Doc-
The oxygen-iron bond is indicated by an arrow. (b) When umentation License (Citation: http://commons.wikimedia.
oxygen is released by hemoglobin, the iron atom shifts out of org/wiki/File:Heme_deoxy_vs_oxygenated.jpg created by
plane from the heme subunit. This shift prevents water mol- user Mrbean427 and http://commons.wikimedia.org/wiki/
ecules from approaching within 3 Å of the valence electrons File:Dipole_H2O.svg user NEUROtiker)
of iron, preventing dipolar interactions. This figure was
390 J. Griauzde et al.

Table 2 Biologic factors affecting T1 and T2


Field strength dependent How does it work on How does it work on
Biologic factors T1 T2 (T2) T1? T2?
Fresh plasma / " No High protein content High water content
#
Deoxyhemoglobin  # Yes N/A Susceptibility
Methemoglobin " # Yes Dipole interactions Susceptibility
Hemosiderin/  # Yes N/A Susceptibility
ferritin
RBC membrane  # Yes N/A Susceptibility

membranes remain intact early on after extrusion, compounds in relation to diamagnetic compounds
sequestering hemoglobin. In the oxygen-poor at higher field strengths [24]. This exaggerated
environment of the hematoma, oxyhemoglobin difference in magnetization results in greater
is converted to deoxyhemoglobin and then later magnetic field inhomogeneity between paramag-
to methemoglobin. In this way, the RBC effec- netic and diamagnetic microenvironments
tively creates a paramagnetic microenvironment causing more pronounced T2 shortening
in contrast to the diamagnetic microenvironment (T2 darkness).
external to it. Diffusing water molecules experi- Gradient recall echo (GRE) sequences are
ence the gradient created by this magnetic field highly sensitive to susceptibility effects caused
inhomogeneity, which dephases their protons and by magnetic field in homogeneities
results in T2 shortening [24, 35, 36]. When the [38–40]. GRE sequences are, in turn, highly sen-
RBCs lyse, the deoxy- and methemoglobin are no sitive for the detection of intracranial hemorrhage
longer sequestered, and the field inhomogeneity is and highly accurate at predicting its extent
lost. Finally, after progressive metabolism, para- [38, 41]. This makes GRE particularly valuable
magnetic iron atoms are taken up and sequestered in the hyperacute and chronic periods, when blood
by hemosiderin and ferritin. This again results in products might otherwise remain inconspicuous
magnetic field inhomogeneity which dephases [38, 39]. As a result, GRE sequences have become
diffusing water protons with resultant T2 shorten- widely incorporated in standard MRI protocols
ing [30]. The effects of biological materials on T1 for the evaluation of intracranial hemorrhage.
and T2 in intracranial hemorrhage are listed in Fast spin echo (FSE) sequences have become a
Table 2. mainstay of standard MRI protocols, widely
replacing conventional spin echo. By using
shorter interecho times and closely spaced pulses,
MRI of Intracranial Hemorrhage: FSE allows for significant reductions in scan times
Technical Considerations [42, 43]. These shorter echo times have been
shown to cause increased T2 prolongation by a
The appearance of intracranial blood products can variety of mechanisms [42]. An untoward result of
be significantly altered depending on the this T2 prolongation is a decreased sensitivity
magnetic field strength and the pulse sequence to T2 susceptibility effects, although the clinical
used. Magnetic field strength imparts its influence significance of this has been questioned [43].
by altering susceptibility-induced T2 effects. Nevertheless, it is recommended that GRE
The inverse of the T2 relaxation rate varies qua- sequences be included in standard MRI protocols
dratically with the field strength [24, 37, 38]. to ensure that high sensitivity for small amounts
This relationship is a product of the relatively of blood products is maintained despite the usage
increased magnetization of paramagnetic of FSE.
18 Hemorrhagic Stroke 391

CT Angiography of Intracranial CTA: Pitfalls in Evaluating Causes


Hemorrhage of Intracranial Hemorrhage

CT angiography (CTA) is a useful adjunct to both Although CTA plays an important role in the
NCCT and MRI in the evaluation of patients with evaluation of patients with intracranial hemor-
acute intracranial hemorrhage. CTA images can rhage, it has several notable limitations.
be rapidly acquired and reconstructed to allow Reconstructed images (i.e., volume-rendered and
intricate visualization of the intracranial vessels maximum-intensity-projection images) can
in multiple planes and projections. It has high enhance artifacts and are susceptible to data loss,
sensitivity, specificity, and diagnostic accuracy in potentially confusing the diagnosis of vascular
identifying vascular causes of intraparenchymal pathology [54]. Pitfalls such as this can be
hemorrhage and can be beneficial in evaluating avoided by reviewing the source images in all
the vascularity of hemorrhagic neoplasms cases and using multiplanar reformatted images
[44–48]. (which contain all source data), rather than relying
In addition to its role in diagnosis, CTA can on reconstructed images [54]. Acute extraluminal
provide valuable prognostic information in the blood products can also obscure evaluation of the
patient with intracranial hemorrhage. The CTA arteries, although this can be minimized with
“spot sign” is a high-attenuation focus within an proper arterial enhancement, as the Hounsfield
intracerebral hematoma which represents a site of density of intravenous contrast is significantly
extravasation [49] (Fig. 6). In patients with intra- higher than that of acute extraluminal blood.
cerebral hematoma, the “spot sign” has been iden- Unfortunately, other artifacts are not as easily
tified as a predictor of hematoma progression, avoided as these.
longer mean hospital stay, in-hospital mortality, Evaluation of patients with previous place-
and poor overall outcomes in survivors [50–53]. ment of embolic materials, endovascular coils,
and surgical aneurysm clips is often
suboptimal secondary to marked streak artifact.
Also, CTA is limited in evaluating small arter-
ies as well as arteries near bony structures,
secondary to limited spatial resolution, poor
contrast between arteries and adjacent bones,
and beam-hardening artifact [55, 56]. Because
of these limitations, catheter-based digital sub-
traction angiography (DSA) remains the gold
standard in the evaluation of intracranial hem-
orrhage [48, 57, 58].

Magnetic Resonance Angiography


of Intracranial Hemorrhage

Magnetic resonance angiography (MRA) is an


extensive topic whose breadth is beyond the
scope of this chapter. There are, however, impor-
tant concepts which will allow for a more thor-
ough understanding of its value and shortcomings
Fig. 6 The CTA “spot sign”: a high-attenuation focus
along the medial aspect of an intracranial hematoma in the diagnostic algorithm of intracranial hemor-
(arrow) rhage. The three main types of magnetic
392 J. Griauzde et al.

resonance angiography are time-of-flight MRA thickness and the velocity of the blood within
(TOFMRA), phase-contrast MRA (PCMRA), the vessel. Maximal signal is obtained when all
and contrast-enhanced MRA (CEMRA). saturated blood is replaced with unsaturated
PCMRA allows for evaluation of hemodynamic blood. This occurs when the velocity of flow is
flow, but its use in the anatomic evaluation of greater than or equal to the slice thickness
intracranial vasculature is at present dwarfed by divided by the repetition time, represented by
TOFMRA. Therefore, this discussion will focus the equation v  z/TR, where v is the velocity
on the important strengths and weaknesses of of flow, z is the slice thickness, and TR is the
non-contrast TOFMRA and CEMRA in the eval- pulse repetition time [60, 61]. Additional factors
uation of intracranial hemorrhage. that affect the signal of flowing blood include the
flip angle of the pulse, the T1 of the tissue in the
slice, and the orientation of the vessel to the
Time of Flight imaging slice [59, 61]. In the case of the intracra-
nial circulation, flow signals are optimized by
TOF imaging, in its simplest form, takes advan- selecting axial slice acquisition, which is perpen-
tage of the flow of blood. In TOF, radiofrequency dicular to the average overall intracranial arterial
(RF) pulses are applied at short TRs to a given flow. Similarly, optimal TR and slice thickness
slice thickness resulting in the saturation of all can be selected to ensure that the signal of the
the magnetic spins within the slice and suppres- static tissue is suppressed and that the signal in
sion of their longitudinal magnetization the vessels is maximally enhanced.
[59]. Spins that are moving perpendicularly into TOF imaging identifies any flow within an
and out of the slice are not saturated, however. In image slice; therefore, both arterial and venous
a given length of time, the blood that has been structures will produce signal. To selectively
saturated by the RF pulses will move out of the highlight the vascular structure of interest (artery
slice and be replaced by “new” unsaturated or vein) and to minimize contamination of the
blood. When exposed to an excitatory pulse, other, saturation bands are employed. These
this “new” blood is magnetized and can undergo pre-saturation bands are positioned a certain dis-
longitudinal relaxation with resultant bright sig- tance from the imaging slice to suppress the
nal (Fig. 7). The relative signal of the flowing undesired flow signal before it arrives within the
blood will be directly related to the slice imaging slice [62] (Fig. 8).

Fig. 7 New blood flows


into the imaging slice
displacing saturated blood. Saturating RF pulses
When the “new” blood
arrives, it can be excited and
undergoes longitudinal
Saturated Blood
relaxation with resultant
bright signal
Stationary Tissue in Slice

When V > z /TR

Flow
18 Hemorrhagic Stroke 393

Fig. 8 Blood within a


vessel is pre-saturated
upstream to the imaging
slice. When the blood
arrives in the imaging slice
(asterisk), it has no signal
because of pre-saturation
Saturated Imaging Slice

Saturating Pulses

Flow
Pre-Saturated Band

complex flow patterns which vary based on their


Time of Flight: Pitfalls in Evaluating size and morphology. Turbulent and slow flow
Causes of Intracranial Hemorrhage within the aneurysm results in loss of signal on
TOF, leading to underestimation of the aneurysm
Because of its dependence on flow and spin satu- volume and neck size [67]. Also, the presence of
ration, TOF is susceptible to many artifacts. When methemoglobin in thrombosed aneurysms can
flow within a vessel is slow or turbulent, or the lead to false-positive results [66]. Similarly,
orientation of flow is parallel (or near parallel) to AVMs and DAVFs pose a formidable diagnostic
the imaging slice, the blood spins have a longer challenge on TOFMRA because of their vessel
“dwell time” within the image slice [63]. Increased tortuosity, complex angioarchitecture, and slow-
“dwell time” exposes the spins to more saturating flow draining veins. These properties cause
pulses, leading to suppressed signal in these por- TOFMRA to overestimate the size of small AVMs
tions of the vessel, which can mimic vessel and underestimate the size of large AVMs [68].
narrowing or occlusion [63]. Additional sources Additionally, TOF is limited in correctly
of artifact include bright T1 substances, tissue inter- interpreting small feeding arteries and in identifying
faces, pulsation, and foreign bodies. Methemoglo- draining veins of AVMs and DAVFs, which can
bin (T1 bright) presents a particular challenge in have significant treatment implications [68, 69].
TOF because it is often present in hemorrhage The use of higher field strength has been shown to
and/or vessel thrombus. The T1 signal from met- improve characterization, but many shortcomings
hemoglobin can be difficult to suppress, and it remain [68].
recovers its T1 signal rapidly, decreasing the con-
spicuity of vessels from background tissue and
even mimicking flow where there is none
[58, 64]. Metallic foreign bodies (surgical clips) Contrast-Enhanced MRA
and tissue interfaces can dephase spins in nearby
vessels causing alteration in their apparent config- CEMRA employs fast spoiled gradient-recalled
uration and diameter, although these are a less echo-based sequences (FSPGR) and the paramag-
significant source of artifact [65, 66]. netic properties of gadolinium to intensify the
The artifacts associated with TOF can make signal within vessels. FSPGR is used to spoil the
proper evaluation of aneurysms, arteriovenous transverse magnetization of background tissues
malformations (AVM), and dural venous fistulas which accentuates the marked T1 shortening of
(DAVF) very challenging. Aneurysms can contain intravascular gadolinium [70]. FSPGR also
394 J. Griauzde et al.

employs short RF pulses allowing for fast Table 3 MRA and CTA: sources of artifact and limitation
sequence times [70]. Administration of gadolin- in intracranial hemorrhage
ium allows for improved signal-to-noise ratio and Source CTA TOFMRA CEMRA
minimizes flow-related artifacts [70]. Acute blood products +  
Methemoglobin  + +
Tissue interfaces  + 
Contrast-Enhanced MRA: Pitfalls Skeletal structures +  
in Evaluating Causes of Intracranial Metal/embolic ++ + +
materials
Hemorrhage Tortuous vessels  ++ 
Turbulent/slow flow  ++ 
The saturation and spin dephasing effects that Complex  ++ +
occur with TOF are of no consequence in angioarchitecture
CEMRA. It does, however, have its own limita- Venous enhancement + + +
tions. Correct bolus timing is paramount to creat- Small arteries + + +
ing a diagnostic study but unfortunately can be ++ significant effect, + some effect,  no significant effect
very challenging to obtain [70]. When bolus
timing is poor, significant artifact can limit evalu-
ation of vascular pathology [71]. Artifacts includ- combination of TOFMRA and CEMRA can reli-
ing distortion of the size of the vessel, “ringing” ably diagnose moderate- and high-flow DAVF, and
within the vessel, and nontarget vessel enhance- CEMRA has the added ability to identify the pres-
ment can obscure pathology [70, 72]. In addition ence of cortical venous drainage [69]. MRA plays a
to bolus timing, the marked T1 shortening of limited role in the evaluation of intracranial neo-
methemoglobin can obscure contrast between plasms. It can identify encasement and mass effect
vessels and background tissue, as it does in TOF of large arteries but does not reliably identify small
images. Finally, limited spatial resolution in arterial feeders [77].
CEMRA makes it insensitive in the evaluation of
small aneurysms (<3 mm) and in the evaluation
of complex AVMs [73]. Artifacts and limitations Imaging Patterns of Intraparenchymal
of CTA and MRA are summarized in Table 3. Hemorrhage (ICH)

Hemorrhage Etiology
MRA: Aneurysms, AVMs, DAVF,
and Neoplasms There are numerous etiologies of ICH including
both traumatic and nontraumatic. Among
Despite the limitations noted above, TOFMRA and nontraumatic etiologies, about 80–90 % are “pri-
CEMRA play a role in the diagnostic work-up of mary” ICH which is related to hypertension
patients with intracranial hemorrhage. MRA has and/or cerebral amyloid angiopathy and the asso-
been shown to adequately depict aneurysms greater ciated changes of vessel walls which leads to
than 3 mm, which encompasses most clinically weakening and rupture [78–80]. Less common
significant aneurysms [66, 74, 75]. Additionally, but still clinically relevant “secondary” causes of
MRA has been shown to be sensitive in the detec- ICH include ruptured vascular malformations,
tion of AVMs greater than 3 cm [48]. Furthermore, hemorrhagic transformation of ischemia, tumor,
the addition of contrast allows for improved delin- vasculitis, anticoagulation, and cerebral venous
eation of aneurysm anatomy and AVM nidus as thrombosis. Determining an exact etiology for an
well as improved assessment of AVM drainage ICH can be challenging because multiple
pattern and small arterial feeders [71, 73, 76]. The dynamic factors may be present in the acute
18 Hemorrhagic Stroke 395

neurologic patient; for instance, ICH can arise


from hemorrhagic transformation of infarcted
tissue, but a large ICH may also cause mass effect
on surrounding tissue that restricts the perfusion
gradient of the tissue and leads to surrounding
infarction. Familiarity with characteristic imag-
ing factors can improve the interpreter’s ability to
troubleshoot in challenging cases.

Hypertensive Hemorrhage

Hypertension induces changes of intracranial arte-


rial vessel walls, weakening them and allowing
them to rupture leading to hypertensive ICH.
These changes include smooth muscle cell hyper-
plasia, smooth muscle cell death, collagen
replacement of the vascular walls, and arteriolar
ectasia [78, 79]. The most classic locations of
hypertensive ICH are the basal ganglia, thalami, Fig. 9 Hypertensive hemorrhage into the thalamus. A
pons, and cerebellum; the cerebral lobes are some- 62-year-old female presenting with altered mental status
related to hypertensive ICH. The hematoma is centered in
what less common (Fig. 9) [80]. A relatively
the left thalamus, a classic location for hypertensive ICH
homogeneous single blood collection in these (solid arrow). Note that blood has dissected into the lateral
locations, without imaging evidence of an under- ventricle (open arrow). Also, there is a mild edema around
lying etiology such as an aneurysm, is a the hematoma and lateral ventricle
classical appearance of these hemorrhages.
Patient clinical factors include hypertension as
well as other cardiovascular risk factors such as pressure after an ICH is of critical importance as
old age, smoking, and excessive alcohol use discussed in the American Stroke Association’s
[79]. Imaging markers of diffuse microvascular most recent guidelines on intracranial hemorrhage
disease can suggest long-standing vascular (Class 1, Level A recommendation) [82].
changes which predispose to hypertensive ICH.
These include periventricular low attenuation on
CT or FLAIR hyperintensity on MRI and numer- Illicit Drugs
ous foci of susceptibility artifact on T2* MR
sequences in characteristic locations which repre- Although far less common than hypertensive
sent evidence of prior clinically silent ICH, illicit drug-induced ICH is presented here
“microbleeds” [81]. because its imaging findings are similar to hyper-
In the absence of other recognized etiologies, tensive hemorrhage in terms of bleed location.
ICH is typically assumed to be related to hyper- Commonly implicated drugs include those caus-
tension. It is possible that some of the ICHs attrib- ing hypertension, vasospasm, or even cerebral
uted to hypertension are in fact occult vasculitis such as cocaine, amphetamines, and
presentations of other etiologies. Nevertheless, MDMA (“ecstasy”). Differentiation for hyperten-
the pathologic effects of hypertension on other sive ICH is typically based on clinical factors
risk factors for ICH (i.e., vascular malformations) including a history of illicit drug use, younger
cannot be dismissed, and control of blood age, and absence of classical cardiovascular risk
396 J. Griauzde et al.

Fig. 10 A 66-year-old female with mental status changes subcortical location (solid arrows) and scattered spots of
from hemorrhages related to cerebral amyloid angiopathy. susceptibility artifact from remote microhemorrhage
T2* GRE images show multifocal hemorrhages based in a

factors. For young adults, drug use may surpass angiopathy may play an important role in several
other biologic risk factors in causing ICH, dementias, and most patients presenting with ICH
although these patients often have a more favor- related to amyloid are elderly (usually >70 years).
able clinical course compared to other patients The imaging hallmark of cerebral amyloid
with ICH [83]. angiopathy is hemosiderin staining secondary to
multiple prior microbleeds, particularly in the cere-
bral lobes (Fig. 10). This is in contrast to the tha-
Cerebral Amyloid Angiopathy lamic and basal ganglia microbleeds seen in
hypertensive angiopathy. These microbleeds are
Amyloid angiopathy is a common etiology of most readily detected as scattered areas of suscepti-
nontraumatic ICH, perhaps second only to hyper- bility artifact on T2* images [85, 86]. The charac-
tension [84]. Amyloid angiopathy is characterized teristic finding of multiple lobar microhemorrhages
by the deposition of β-amyloid protein into small- suggests the diagnosis of cerebral amyloid
and medium-sized cerebral arteries [79]. The vas- angiopathy even for patients who have not
cular insults related to cerebral amyloid presented with a major ICH [85, 86].
18 Hemorrhagic Stroke 397

Vascular Malformations significant risk of future hemorrhage (approxi-


mately 2 % per year) [89]. Findings which predict
Vascular malformations are the most common that an AVM will lead to future bleeding include
cause of ICH which are secondary to specific presenting with hemorrhage, deeper location, an
anatomic abnormalities and are a leading cause associated aneurysm (reflecting multifocal weak-
of ICH in young adults. Vascular malformations ening of vessel walls), and drainage exclusively
often include vessel wall components which are into deep cerebral veins (veins which drain into
not capable of withstanding the high pressure the vein of Galen and inferior sagittal sinus as
exerted within the vascular malformation, leading opposed to superficial veins) [90, 91].
to ICH [87]. By far the most clinically relevant
vascular malformations are arteriovenous
malformations (AVMs), which lead to about Cavernous Malformations (CMs)
80 % of ICH due to vascular malformations
[88]. These are followed in importance by cavern- Cavernous malformations are benign low-flow
ous malformations (CMs) and dural arteriovenous malformations comprised of endothelial-lined
fistulas (DAVFs). Developmental venous anoma- blood vessels not separated by brain parenchyma.
lies (DVAs) and capillary telangiectasias usually CMs are characterized by a heterogeneous central
do not lead to ICH unless they occur in association appearance including regions of high T1 and T2
with other vascular anomalies. Identification of signal with a surrounding area of hemosiderin
the causative vascular malformation is rarely pos- staining most obvious as signal loss on T2*
sible using non-contrast examination, and CTA, images. CMs can be found in association with
contrast-enhanced MRI/MRA, and conventional DVAs, and several hereditary syndromes of mul-
angiography are much more sensitive. tiple CMs have been described [92].

Dural Arteriovenous Malformations


Arteriovenous Malformations (AVMs) (DAVFs)
Arteriovenous malformations (AVMs) are develop- DAVFs are fistulas from meningeal arteries to a
mental lesions in which pial arteries connect directly meningeal vein or dural venous sinus. DAVFs can
into pial veins without an intervening capillary bed, be either sequela of prior trauma such as a fracture
allowing for rapid shunting of blood through the or craniotomy or be developmental. Just as with
malformation and elevating pressures on the venous developmental AVMs in the brain, DAVFs are
side. AVMs often have a characteristic “bag of characterized by large feeding and draining ves-
worms” appearance both on imaging and gross sels, which are best demonstrated by angiographic
examination due to the high flow of blood through imaging. In addition to ICH, symptoms can
the feeding and draining vessels (Fig. 11) [87]. A include neurologic deficits and tinnitus related to
central nidus at the actual site of connection venous hypertension. Treatment often centers
between the arterial and venous vessels is a charac- around curative endovascular techniques without
teristic feature although the size can vary from the need for later surgery [89].
microscopic (nearly impossible to detect on most
imaging modalities) to several centimeters in size
(potentially detectable on even non-contrast CT). Aneurysm
The gold standard for the treatment of AVMs
remains surgical excision, but therapeutic guide- Aneurysmal hemorrhage most typically causes
lines endorse a role for endovascular and radiation subarachnoid hemorrhage (discussed below), but
treatments prior to surgery for some patients. intraparenchymal hemorrhage can also occur if
When AVMs are detected incidentally, the care the aneurysm is enclosed in brain parenchyma
team will usually opt for treatment, as they have a [50, 79, 93].
398 J. Griauzde et al.

Fig. 11 AVM with hemorrhage. CT of a 43-year-old appearance on T2 image (b, open arrow). The arteriove-
woman presenting with headache and mental status nous malformation which led to the hemorrhage is best
changes with a large hemorrhage in the left cerebral lobe shown on DSA injection of the left internal carotid artery
(a); areas of irregular high attenuation are presented ante- (ICA) (c–e) including early filling of draining veins
rior to hematoma (solid arrow). Further evaluation with (arrowheads)
MRI reveals large flow voids with a “bag of worms”

Hemorrhagic Tumors because they are more common tumors overall.


Among primary brain tumors, glioblastoma
Both primary and metastatic CNS tumors can be multiforme, astrocytoma, oligodendroglioma, and
complicated by clinically significant hemorrhage. mixed astrocytoma/oligodendroglioma all bleed
Identification of contrast enhancement can help with significant frequency, but glioblastoma
delineate the underlying lesion from surrounding multiforme is the most common etiology due to
blood products, and the presence of multiple sim- its high overall prevalence [94, 95].
ilar appearing lesions is highly suggestive of met-
astatic disease [94]. About 5 % of CNS
malignancies will be complicated by macroscopic Hemorrhagic Transformation
bleeding although the risk varies dramatically by of Ischemic Stroke
the type of tumor. For instance, among metastatic
masses, about a third of melanoma metastasis will After an ischemic stroke, bleeding can occur
be complicated by hemorrhage (Fig. 12). Meta- within the area of infarcted tissue. Hemorrhagic
static adenocarcinomas and squamous cell carci- transformation of ischemic stroke which is symp-
nomas have a lower risk of bleeding on a per lesion tomatic occurs in about 6 % of patients treated
basis but are likely etiologies for hemorrhage with thrombolytic therapy and about 1 % of
18 Hemorrhagic Stroke 399

Fig. 12 Hemorrhagic melanoma metastasis. A 59-year- collection demonstrates intrinsic high T1 (a) and high T2
old man with history of melanoma and headache. Note the signal (c) compatible with a late subacute hemorrhage.
enhancement of the mass between pre-contrast (a) and Also note vasogenic edema surrounding the hematoma
post-contrast (b) axial T1 images as well as the fluid (c, open arrow)
collection just lateral to the mass (solid arrow). This fluid

patients treated without thrombolysis. It often seizure [99]. The impaired venous drainage
carries a dismal clinical prognosis with estimates reduces the perfusion pressure of the brain paren-
of severe morbidity and/or mortality exceeding chyma which in turn can lead to ischemia and
90 % [96]. subsequent hemorrhagic transformation. ICH
Imaging findings which predict an increased complicates about a third of cerebral venous
risk of hemorrhagic transformation include the thrombosis (Fig. 14). The most characteristic find-
CT findings of early stroke (low attenuation in ing of ICH related to venous thrombosis is irreg-
the infarcted area, loss of gray-white differentia- ular or flame-shaped hemorrhage that occurs in the
tion, and parenchymal swelling), perhaps because region of parenchyma drained by the thrombosed
areas of ischemia visible on CT tend to be further cerebral vein. These findings are almost always
progressed than those visible only with MRI accompanied by other parenchymal abnormalities
(Fig. 13). The size of these abnormalities is also caused by the thrombosis such as edema and per-
important; several authors have found that CT fusion/diffusion restriction [100].
changes suggesting stroke over a region larger
than a third of the middle cerebral artery (MCA)
distribution are more likely to hemorrhage Trauma
[97]. Studies with MRI have also found that
greater volumes of restricted diffusion also predict When the head is exposed to external trauma,
a greater risk of hemorrhage [97, 98]. common sites of injury are at the site of external
force (coup) and at the area of the brain opposite to
the external force (contrecoup). Intraparenchymal
Venous Thrombosis hemorrhage has a propensity to occur at the
contrecoup site due to anchoring structures
Cerebral venous thrombosis can present in a num- such as falx cerebri and bridging cortical
ber of prothrombotic clinical scenarios including veins pulling on the parenchyma as the brain is
pregnancy, recent surgery, oral contraceptives, accelerated. Other areas which are at particular
coagulation disorders, dehydration, and infection risk of ICH following trauma are the areas of the
(both systemic and local) and present as either brain adjacent to the jagged portions of the skull
acute or chronic symptoms including headache, base such as the petrous ridges and the ethmoid
neurologic deficits, mental status changes, and plate.
400 J. Griauzde et al.

Fig. 13 Hemorrhagic
transformation of ischemia.
A 69-year-old male with
stroke demonstrated on
diffusion-weighted images
(a) and non-contrast CT (b)
(solid arrows). Repeat
non-contrast CT at 1 day (c)
and 1 month (d)
demonstrates hemorrhagic
transformation of the
infarction (open arrows)

Small, punctate regions of parenchymal hem- around one another to produce a “swirl sign”
orrhage can also be seen at sites of other traumatic which has also been associated with hemorrhage
injuries such as shearing and diffuse axonal inju- growth and poor outcomes [103]. If the patient
ries, although low attenuation from edema may receives a CTA study, active extravasation can
predominate at these sites [101, 102]. present as a tiny spot on arterial phase images
(the “spot sign”) which can rapidly expand on
more delayed phase images (Fig. 15) [50]. Even
Additional Clinical Topics in ICH when a spot of precise extravasation is not identi-
fied on arterial phase images, more delayed
Active Hemorrhage images can directly demonstrate extravasated con-
trast indicating ongoing hemorrhage [104–106].
Observing active extravasation of blood into the
area of hemorrhage is an ominous radiologic find-
ing that suggests both ongoing expansion of the Intraventricular Extension, Associated
hematoma and a poor clinical outcome [103]. On Subarachnoid Hemorrhage,
non-contrast examinations, freshly extravasated and Hydrocephalus
blood will have attenuation characteristics differ-
ent from the blood which has been present in the Although intraparenchymal hemorrhage begins
hematoma for a longer period, and these hetero- within the brain parenchyma, blood can dissect
geneous groups of blood products can circle through the relatively pliant tissues of the central
18 Hemorrhagic Stroke 401

Fig. 14 Venous sinus


thrombosis with
hemorrhage. A 13-year-old
female that recently
initiated oral contraceptive
pills presenting with
headache. MR venogram
(a) demonstrates occlusion
of the straight sinus and left
transverse sinus
(arrowheads). Diffusion
restriction (b) in the right
thalamus related to the
thrombosis (closed arrow).
Rim of hemorrhage in the
area of the prior diffusion
restriction is demonstrated
on non-contrast CT (c, open
arrow). Follow-up MRI
3 months later (d) after
resolution of thrombosis
shows (not shown)
susceptibility artifact from
hemosiderin staining at the
site of prior hemorrhage
(arrowhead)

Fig. 15 A 51-year-old male with a hypertensive hemor- aneurysm or vascular malformation reveals a tiny high-
rhage in the left basal ganglia seen on non-contrast CT (a, attenuation spot compatible with active extravasation
solid arrow). CT angiography performed to screen for (b and c, open arrows)

nervous system and breech the ependymal linings that it will evolve to include an intraventricular
of the cerebral spinal fluid spaces to present as component also increases. This volume also
intraventricular and subarachnoid hemorrhage. appears to depend upon the location of the hem-
As the size of the ICH increases, the probability orrhage as certain regions of the brain may permit
402 J. Griauzde et al.

Means of
describing ICH
location

Infratentorial Supratentorial

Brainstem Cerebellar Deep Lobar

Pons Medulla Basal ganglia Thalami

Fig. 16 Classification of areas of hemorrhage within the brain

more hemorrhage volume before forcing blood to hemorrhages account for about a third of
decompress into the ventricular system; for infratentorial ICH with the remainder occurring
instance, many more lobar ICHs achieve a volume in the cerebellum [109] (Fig. 16). Multifocal ICH
over 50 ml without breeching the ventricles than is well described but uncommon (about 1 % of
thalamic ICHs [107]. ICHs) [109, 110].
Just as in primary subarachnoid hemorrhage, Studies of the prognostic implications of
hydrocephalus is a common complication of ICH location as an independent risk factor
intraparenchymal hemorrhage (about half of have shown inconsistent results, and
patients with ICH). Hydrocephalus is also an hematoma volume and growth have been identi-
independent predictor of mortality [108]. fied as more significant prognostic factors [84,
109–111].

Hemorrhage Location
Predicting Clinical Outcome
ICH may occur almost anywhere within the intra-
cranial parenchyma. The most basic distinction in As alluded to above, a variety of radiologic and
hemorrhage location is above or below the clinical factors can assist in prognosticating for
tentorium cerebelli (supratentorial versus patients with ICH. Among the most widely used is
infratentorial). Supratentorial hemorrhagic is the validated “ICH Score,” described by Hemphill
most common and is estimated to occur in et al. in 2001 [84, 112]. Radiologic predictors of
approximately 90 % of cases [109]. About half poor clinical outcome in the ICH Score include an
to two thirds of the supratentorial ICHs are infratentorial origin of the ICH, an ICH volume
based in the deep cerebral structures including above 30 ml, and an intraventricular extension of
the basal ganglia and thalami with the the hemorrhage, while clinical predictors include
remaining supratentorial ICHs based in the the Glasgow Coma Scale and age at presentation.
cerebral lobes [109, 110]. Infratentorial hemor- For instance, approximately 50 % of patients scor-
rhage is less common, accounting for approxi- ing as the lowest risk category on the ICH Score
mately 10 % of ICH [109]. Brainstem will have favorable clinical outcomes at 12 months
18 Hemorrhagic Stroke 403

(total or near total recovery, “patient is able to 60–80 HU which gradually fades to approach
carry out all usual duties and activities”), while water attenuation over the coming months of
those in the highest categories will have virtually evolution. The MRI appearance is more
no chance of recovering to ambulatory status complex: the central portion of the hematoma
(moderate to severe disability or death) containing residual oxyhemoglobin will be T2
[112]. Other radiologic factors discussed above hyperintense and T1 hypointense, and the periph-
such as active extravasation or growth of ICH ery containing deoxyhemoglobin will be T2
provide additional important prognostic informa- hypointense and T1 isointense. As the proportion
tion; for example, active extravasation on a of oxyhemoglobin drops and deoxyhemoglobin
contrast-enhanced CT such as a CTA raises the prevails, the hematoma will become more uni-
risk of a 30-day mortality by an odds ratio of formly T2 hypointense and T1 isointense to
4.7 [103]. hyperintense, especially as protein content pre-
dominates in the extracellular space favoring T1
signal gain and further T2 signal loss. Because of
Evolution of the Intraparenchymal mass effect, T2 hyperintense surrounding
Hematoma vasogenic edema is usually observed until the
brain parenchyma has remodeled around the
A more detailed description of the biophysical mass effect [114].
factors involved in the CT and MRI appearance As the clot continues to contract and the hema-
of intracranial blood products has been provided toma progresses into the “early subacute” phase
above; this section aims to provide a clinically (typically several days), deoxyhemoglobin is oxi-
oriented guide to aid in understanding the evolv- dized into methemoglobin within the still-intact
ing appearance of intracranial hemorrhage over red blood cells; the presence of methemoglobin
time. Recognizing different phases of hemorrhage dramatically increases the T1 signal of the clot,
can be useful for identifying ongoing hemorrhage but because the methemoglobin is sequestered in
requiring further management and correlating the intact red blood cells, the field inhomogeneity
onset of neurologic deficits and occasionally in between intracellular and extracellular spaces
medicolegal settings such as non-accidental leads to marked T2 hypointensity. In the “late
trauma in children. subacute” phase several days to a month follow-
In the minutes immediately following ing the initial bleed, the red blood cells finally
extravasation, the CT and MRI appearance of lyse, allowing the methemoglobin to distribute
hemorrhage is similar to intravascular evenly throughout the clot, and finally both T1
blood excepting flow-related artifacts that and T2 hyperintensity prevail.
can cause signal void in patent blood vessels In the months and years of the “chronic” phase,
on T2 images. On CT, this is usually manifest macrophages will gradually digest the clot, reduc-
as fluid collections of 40–60 HU which can be ing its volume and transforming extracellular met-
difficult to differentiate from the adjacent hemoglobin into hemosiderin; in this phase, the
brain, although early clot products including clumps clot is hypointense on T1 and markedly
of fibrin, platelets, and white blood cells will intro- hypointense on T2 due to susceptibility effects
duce heterogeneous attenuation [113]. of the hemosiderin (Fig. 17).
The hours and days following extravasation, As evidenced above, the appearance of the
the “hyperacute” and “acute” phases, are charac- ICH at different periods of time depends consid-
terized by clot formation where the water compo- erably upon a number of factors. For instance, in
nent is squeezed out leaving behind a matrix of early phases, the hematocrit and protein levels of
red and white blood cells, platelets, and clotting the hematoma will dramatically alter the CT atten-
proteins. The attenuation of these protein- uation in the hematoma. In later phases, factors
rich components is higher than water, leading to such as oxygen tension at the hematoma will
a hyperdense collection on CT reaching up to determine how quickly deoxyhemoglobin
404 J. Griauzde et al.

Late subacute (days-month)


hypointense ← T2 signal → hyperintense

Extracellular met-Hb
Lysed RBC’s

Hyperacute (<12 h)
oxy-Hb
Intact RBC’s

Acute (hours-days)
deoxy-Hb
Intact RBC’s
Early subacute (days)
Chronic (month-years) Intracellular met-Hb
Hemosiderin and ferritin Intact RBC’s
Macrophage digested clot

hypointense ← T1 signal → hyperintense


Fig. 17 Evolution of blood products on MRI (Derived from a figure created by Dr. Frank Gaillard as presented on http://
radiopaedia.org/articles/ageing-blood-on-mri, with permission)

transitions into methemoglobin and how quickly tumor, dural AVM, intracranial dissection, vascu-
red blood cells finally lyse and decrease the field litis, mycotic aneurysm, blood dyscrasias, amy-
inhomogeneity effects of sequestered methemo- loid angiopathy, moyamoya disease,
globin. The integrity of the blood-brain barrier complications of pregnancy, and spinal cord
also helps to determine the degree to which AVM [115, 116]. Fifteen percent of patients with
hemosiderin-laden macrophages remain trapped SAH have no aneurysm found and no other cause
in the parenchyma causing hemosiderin staining noted after four vessel angiography [117].
long after the vast majority of the hematoma mass SAH is best identified on non-enhanced CT of
has been resorbed [113]. the brain. This shows hyperdensity in the CSF
spaces surrounding the brain parenchyma and
may often be seen layering in the ventricles
Extra-axial Central Nervous System when there is intraventricular extension
Hemorrhage (Fig. 18). High-density blood products in acute
SAH are identified within cisterns and subarach-
Subarachnoid Hemorrhage (SAH) noid spaces in 90–97 % of patients within the first
24 h following SAH [118, 119]. The sensitivity
The most common cause of atraumatic SAH in the for SAH decreases to 80 % within 3 days, 70 %
adult population is a ruptured intracranial aneu- within 5 days, 50 % within 1 week, and 30 %
rysm. Other causes include secondary leakage of within 2 weeks. CT is useful in directing which
blood from a primary intraparenchymal hemor- blood vessel should be injected first for the
rhage, trauma, intracranial AVM, hemorrhagic angiogram.
18 Hemorrhagic Stroke 405

one does not detect subarachnoid methemoglobin


if the patient is imaged a few days after an ictus.
MRI and MRA can be helpful in a patient with a
SAH with multiple aneurysms, since the aneu-
rysm that bled may have high intensity of methe-
moglobin in the adjacent sulci or aneurysm wall
[125–127].
The rate of rehemorrhage from a ruptured
aneurysm has been reported to be between 4 %
and 20 % within the first 24 h [128, 129]. The rate
of rupture then decreases to between 1 % and 2 %
per day for the next 2 weeks. Vasospasm occurs at
3–15 days after SAH and is the chief cause of
significant morbidity and mortality in SAH.
Symptomatic vasospasm can be treated with
hypervolemia, hypertension, hemodilution,
nimodipine, intra-arterial nicardipine, and balloon
angioplasty [130, 131]. Cerebral aneurysms can
be treated by surgical clip placement of
endovascular coil embolization [132].
Fig. 18 A 45-year-old female who presents with worst
headache of her life. Axial NCCT of the brain shows
diffuse high-attenuation material with the subarachnoid
space surrounding the midbrain, within the sylvian fis-
Nonaneurysmal Perimesencephalic
sures, and interhemispheric fissure SAH (NPSAH)

Patients with NPSAH have a similar presentation to


The diagnosis of SAH on MRI may be first traditional SAH patients with sudden onset of the
identified using fluid-attenuated inversion recov- “worst headache of their life.” The radiologic
ery (FLAIR) or susceptibility-weighted (SW) criteria to diagnose NPSAH consist of (1) hemor-
imaging [120, 121]. On FLAIR images, SAH rhage centered immediately anterior to the mid-
can be seen as high signal intensity in the sulci brain with or without extension of blood into the
due to blood protein shortening the T2 of the CSF anterior part of the ambient cistern or to the basal
[122, 123]. On conventional T1-WI and T2-WI, part of the sylvian fissure, (2) no blood filling the
acute SAH is not visible. The oxygen pressure of anterior interhemispheric fissure and no extension
CSF is approximately 43 mmHg. Given the sig- to the lateral sylvian fissure (except for minute
moid shape of the oxyhemoglobin dissociation amounts of blood), and (3) the absence of intraven-
curve, this gives a saturated hemoglobin of tricular hemorrhage [133]. These cases have an
approximately 72 %. Therefore, only 28 % of uncomplicated clinical course [134]. The cause of
the hemoglobin is in the deoxygenated form. NPSAH is not known although the current belief is
The hematocrit of a SAH is usually about 5 %, that it may result from a capillary or venous
and thus the low hematocrit together with the rupture [135].
small amount of deoxyhemoglobin makes acute
hemorrhage difficult to detect on MR [124]. Oxy-
hemoglobin has no unpaired electrons and there- Subdural and Epidural Hematomas
fore behaves like other nonparamagnetic
substances, being isointense to the brain on Subdural and epidural hematomas can be readily
T1-WI and T2-WI. A majority of the time, identified by T1-WI and T2-WI MR images,
RBCs are cleared from the CSF so that normally because the contrast between the calvarium and
406 J. Griauzde et al.

Fig. 19 Axial NCCT shows a (a) crescentic extra-axial hemisphere consistent, subacute subdural hematoma; and
high-attenuation area along the right cerebral hemisphere, (c) crescentic extra-axial low-attenuation material (HU =
acute subdural hematoma; (b) crescentic extra-axial inter- 2) along the right cerebral hemisphere, chronic subdural
mediate attenuation area along the right cerebral hematoma

Fig. 20 Axial T1-WI (a)


and T2-WI (b) MR images
show crescentic extra-axial
material predominantly
high signal on T1, with
areas of low and high signal
of T2 over the left cerebral
hemisphere, consistent with
predominantly subacute
subdural hematoma

blood is high [136]. Precise compartmentalization subdural hematomas evolve, there is breakdown of
of these extra-axial collections on MR images can the methoglobin with a gradual decrease in the
be determined by traditional criteria that are based signal intensity on T1-WI images. Chronic sub-
on CT (Fig. 19). Isodense subdural hematomas on durals can be only minimally hyperintense to CSF
CT can be easily deciphered on MR because these on T1-WI MR, reflecting either a very low concen-
subacute hematomas typically contain methemo- tration of paramagnetic methemoglobin or a high
globin which is hyperintense on MR (Fig. 20). The concentration of nonparamagnetic protein.
signal intensity patterns of evolving subdural Rebleeding into a subdural collection should be
hematomas on conventional MR have been suspected if irregular hypointense membranes and
reviewed in an earlier portion of this chapter and loculating areas of different signal are found or if
appear similar to intraparenchymal hematomas in there are debris-fluid levels seen within the collec-
acute and subacute stages [137, 138]. Chronic sub- tion. In cases of suspected child abuse, subdural
dural hematomas are markedly hyperintense on hematomas typically have a crescentic appearance
MR due to the methemoglobin content. As chronic and cross suture lines extending across a greater
18 Hemorrhagic Stroke 407

Fig. 21 (a) Axial non-contrast head CT shows a convex intermediate attenuation area within the right posterior
high-attenuation area within the right posterior temporal temporal lobe, subacute epidural hematoma
lobe, acute epidural hematoma, and (b) convex

Table 4 Causes of subdural and epidural hematomas is important in determining the age and etiology of
Trauma the hemorrhage. In the future, MR will play a
Dural-based metastases larger role in the evaluation of acute hemorrhagic
Meningioma stroke.
Dural intracranial (or intraspinal) vascular malformation
Superficial intraparenchymal hemorrhage from any cause
Bleeding diathesis due to disease
References
Iatrogenic coagulopathy
1. Broderick J, Connolly S, Feldmann E et al (2007)
Peripheral aneurysm Guidelines for the management of spontaneous intra-
cerebral hemorrhage in adults: 2007 update: a guide-
line from the American Heart Association/American
Stroke Association Stroke Council, High Blood Pres-
area of the brain surface. In contrary, epidural sure Research Council, and the Quality of Care and
hematomas have a convex appearance and do not Outcomes in Research Interdisciplinary Working
cross suture lines involving a smaller portion of the Group. Stroke 38(6):2001–2023
brain surface (Fig. 21). MR plays an important role 2. Adams HP Jr, del Zoppo G, Alberts MJ et al (2007)
Guidelines for the early management of adults with
in detecting multiple sites of intracranial hemor- ischemic stroke: a guideline from the American Heart
rhage at different stages of evolution. The clinical Association/American Stroke Association Stroke
etiologies of subdural and epidural hematomas are Council, Clinical Cardiology Council, Cardiovascu-
shown in Table 4. lar Radiology and Intervention Council, and the Ath-
erosclerotic Peripheral Vascular Disease and Quality
of Care Outcomes in Research Interdisciplinary
Working Groups: the American Academy of Neurol-
Summary ogy affirms the value of this guideline as an educa-
tional tool for neurologists. Stroke 38(5):1655–1711
3. Scott WR, New PF, Davis KR, Schnur JA (1974)
The role of the neuroimager in evaluating CT and Computerized axial tomography of intracerebral and
MR studies in suspected cases of hemorrhagic intraventricular hemorrhage. Radiology 112(1):73–80
stroke is to recognize and localize the hemorrhage 4. Greenberg JO, Skubick DL (1977) Unexpected brain
and attempt to determine its etiology. Understand- hemorrhages and the value of computerized tomogra-
phy. Comput Tomogr 1(4):349–357
ing the limitations of CT in the evaluation of 5. Hounsfield GN (1973) Computerized transverse axial
hemorrhagic stroke and how signal intensity scanning (tomography). 1. Description of system. Br J
varies on MRI based on the stage of hemorrhage Radiol 46(552):1016–1022
408 J. Griauzde et al.

6. Weinstein MA, Duchesneau PM, MacIntyre WJ 22. Koenig SH, Brown RD 3rd, Lindstrom TR (1981)
(1977) White and gray matter of the brain differenti- Interactions of solvent with the heme region of met-
ated by computed tomography. Radiology 122(3): hemoglobin and fluoro-methemoglobin. Biophys J
699–702 34(3):397–408
7. New PFJ, Aronow S (1976) Attenuation measure- 23. Gomori JM, Grossman RI (1988) Mechanisms
ments of whole-blood and blood fractions in com- responsible for the MR appearance and evolution of
puted tomography. Radiology 121(3):635–640 intracranial hemorrhage. Radiographics 8(3):
8. Norman D, Price D, Boyd D, Fishman R, Newton TH 427–440
(1977) Quantitative aspects of computed tomography 24. Thulborn KR, Waterton JC, Matthews PM, Radda GK
of blood and cerebrospinal-fluid. Radiology (1982) Oxygenation dependence of the transverse
123(2):335–338 relaxation time of water protons in whole blood at
9. Bergstrom M, Ericson K, Levander B, Svendsen P, high field. Biochim Biophys Acta 714(2):265–270
Larsson S (1977) Variation with time of the attenua- 25. Ingram VM (1961) Hemoglobin and its abnormali-
tion values of intracranial hematomas. J Comput ties. Thomas, Springfield, Ill
Assist Tomogr 1(1):57–63 26. Pauling L, Coryell CD (1936) The magnetic proper-
10. Dolinskas CA, Bilaniuk LT, Zimmerman RA, Kuhl ties and structure of hemoglobin, oxyhemoglobin and
DE (1977) Computed tomography of intracerebral carbonmonoxyhemoglobin. Proc Natl Acad Sci
hematomas. I. Transmission CT observations on U S A 22(4):210–216
hematoma resolution. AJR Am J Roentgenol 129(4): 27. Dickerson RE, Geis I (1983) Hemoglobin: structure,
681–688 function, evolution, and pathology. Benjamin/Cum-
11. Tans JT (1977) Computed tomography of intracere- mings Publication, Menlo Park
bral hematoma. Clin Neurol Neurosurg 79(4): 28. Di Chiro G, Brooks RA, Girton ME et al (1986)
285–295 Sequential MR studies of intracerebral hematomas
12. Messina AV, Chernik NL (1976) Computed tomogra- in monkeys. AJNR Am J Neuroradiol 7(2):193–199
phy – resolving intracerebral hemorrhage. Radiology 29. Matz P, Turner C, Weinstein PR, Massa SM, Panter
118(3):609–613 SS, Sharp FR (1996) Heme-oxygenase-1 induction
13. Zhu XL, Chan MS, Poon WS (1997) Spontaneous in glia throughout rat brain following experimental
intracranial hemorrhage: which patients need diag- subarachnoid hemorrhage. Brain Res 713(1–2):
nostic cerebral angiography? A prospective study of 211–222
206 cases and review of the literature. Stroke 30. Thulborn KR, Sorensen AG, Kowall NW et al (1990)
28(7):1406–1409 The role of ferritin and hemosiderin in the MR appear-
14. Broderick JP, Brott TG, Duldner JE, Tomsick T, ance of cerebral hemorrhage: a histopathologic bio-
Huster G (1993) Volume of intracerebral hemorrhage. chemical study in rats. AJNR Am J Neuroradiol
A powerful and easy-to-use predictor of 30-day mor- 11(2):291–297
tality. Stroke 24(7):987–993 31. Bizzi A, Brooks RA, Brunetti A et al (1990) Role of
15. Gado M, Phelps M (1975) The peripheral zone of iron and ferritin in MR imaging of the brain: a study in
increase density in cranial computed tomography. primates at different field strengths. Radiology
Radiology 117(1):71–74 177(1):59–65
16. Zacharia TT, Nguyen DT (2010) Subtle pathology 32. Gillis P, Koenig SH (1987) Transverse relaxation of
detection with multidetector row coronal and sagittal solvent protons induced by magnetized spheres:
CT reformations in acute head trauma. Emerg Radiol application to ferritin, erythrocytes, and magnetite.
17(2):97–102 Magn Reson Med 5(4):323–345
17. Smith WP Jr, Batnitzky S, Rengachary SS (1981) 33. Zimmerman RD, Heier LA, Snow RB, Liu DP, Kelly
Acute isodense subdural hematomas: a problem in AB, Deck MD (1988) Acute intracranial hemorrhage:
anemic patients. AJR Am J Roentgenol 136(3): intensity changes on sequential MR scans at 0.5 T.
543–546 AJR Am J Roentgenol 150(3):651–661
18. Schenck JF (1996) The role of magnetic susceptibility 34. Clark RA, Watanabe AT, Bradley WG Jr, Roberts JD
in magnetic resonance imaging: MRI magnetic com- (1990) Acute hematomas: effects of deoxygenation,
patibility of the first and second kinds. Med Phys hematocrit, and fibrin-clot formation and retraction on
23(6):815–850 T2 shortening. Radiology 175(1):201–206
19. Saini S, Frankel RB, Stark DD, Ferrucci JT Jr (1988) 35. Atlas SW, DuBois P, Singer MB, Lu D (2000) Diffu-
Magnetism: a primer and review. AJR Am J sion measurements in intracranial hematomas: impli-
Roentgenol 150(4):735–743 cations for MR imaging of acute stroke. AJNR Am J
20. Thulborn KR, Brady TJ (1989) Iron in magnetic Neuroradiol 21(7):1190–1194
resonance imaging of cerebral hemorrhage. Magn 36. Gomori JM, Grossman RI, Yu-Ip C, Asakura T (1987)
Reson Q 5(1):23–38 NMR relaxation times of blood: dependence on field
21. Bloembergen N (1948) Nuclear magnetic relaxation. strength, oxidation state, and cell integrity. J Comput
M. Nijhoff, The Hague Assist Tomogr 11(4):684–690
18 Hemorrhagic Stroke 409

37. Gomori JM, Grossman RI, Goldberg HI, Zimmerman 51. Delgado Almandoz JE, Yoo AJ, Stone MJ et al (2010)
RA, Bilaniuk LT (1985) Intracranial hematomas: The spot sign score in primary intracerebral hemor-
imaging by high-field MR. Radiology 157(1):87–93 rhage identifies patients at highest risk of in-hospital
38. Weingarten K, Zimmerman RD, Deo-Narine V, mortality and poor outcome among survivors. Stroke
Markisz J, Cahill PT, Deck MD (1991) MR imaging 41(1):54–60
of acute intracranial hemorrhage: findings on sequen- 52. Demchuk AM, Dowlatshahi D, Rodriguez-Luna D
tial spin-echo and gradient-echo images in a dog et al (2012) Prediction of haematoma growth and
model. AJNR Am J Neuroradiol 12(3):457–467 outcome in patients with intracerebral haemorrhage
39. Edelman RR, Johnson K, Buxton R et al (1986) MR using the CT-angiography spot sign (PREDICT): a
of hemorrhage: a new approach. AJNR Am J prospective observational study. Lancet Neurol 11(4):
Neuroradiol 7(5):751–756 307–314
40. Atlas SW, Mark AS, Grossman RI, Gomori JM 53. Delgado Almandoz JE, Kelly HR, Schaefer PW
(1988) Intracranial hemorrhage: gradient-echo MR et al (2012) CT angiography spot sign predicts
imaging at 1.5 T. Comparison with spin-echo imaging in-hospital mortality in patients with secondary intra-
and clinical applications. Radiology 168(3):803–807 cerebral hemorrhage. J Neurointerv Surg 4(6):442–447
41. Patel MR, Edelman RR, Warach S (1996) Detection 54. Tomandl BF, Kostner NC, Schempershofe M
of hyperacute primary intraparenchymal hemorrhage et al (2004) CT angiography of intracranial aneu-
by magnetic resonance imaging. Stroke 27(12): rysms: a focus on postprocessing. Radiographics
2321–2324 24(3):637–655
42. Melki PS, Mulkern RV, Panych LP, Jolesz FA (1991) 55. Ng SH, Wong HF, Ko SF et al (1997) CT angiography
Comparing the FAISE method with conventional of intracranial aneurysms: advantages and pitfalls.
dual-echo sequences. J Magn Reson Imaging 1(3): Eur J Radiol 25(1):14–19
319–326 56. Tanabe S, Uede T, Nonaka T, Ohtaki M, Hashi K
43. Jones KM, Mulkern RV, Mantello MT et al (1992) (1998) Diagnosis of cerebral arteriovenous
Brain hemorrhage: evaluation with fast spin-echo and malformations with three-dimensional CT angiogra-
conventional dual spin-echo images. Radiology phy. J Clin Neurosci 5(Suppl):33–38
182(1):53–58 57. Chappell ET, Moure FC, Good MC (2003) Compar-
44. Delgado Almandoz JE, Schaefer PW, Forero NP, ison of computed tomographic angiography with dig-
Falla JR, Gonzalez RG, Romero JM (2009) Diagnos- ital subtraction angiography in the diagnosis of
tic accuracy and yield of multidetector CT angiogra- cerebral aneurysms: a meta-analysis. Neurosurgery
phy in the evaluation of spontaneous 52(3):624–631; discussion 630–621
intraparenchymal cerebral hemorrhage. AJNR Am J 58. Tanaka H, Numaguchi Y, Konno S, Shrier DA,
Neuroradiol 30(6):1213–1221 Shibata DK, Patel U (1997) Initial experience with
45. Wong GK, Siu DY, Abrigo JM et al (2011) Computed helical CT and 3D reconstruction in therapeutic plan-
tomographic angiography and venography for young ning of cerebral AVMs: comparison with 3D time-of-
or nonhypertensive patients with acute spontaneous flight MRA and digital subtraction angiography.
intracerebral hemorrhage. Stroke 42(1):211–213 J Comput Assist Tomogr 21(5):811–817
46. Tsuchiya K, Hachiya J, Mizutani Y, Yoshino A (1996) 59. Gullberg GT, Wehrli FW, Shimakawa A, Simons MA
Three-dimensional helical CT angiography of skull (1987) MR vascular imaging with a fast gradient
base meningiomas. AJNR Am J Neuroradiol 17(5): refocusing pulse sequence and reformatted images
933–936 from transaxial sections. Radiology 165(1):241–246
47. Hirai T, Korogi Y, Ono K, Uemura S, Yamashita Y 60. Bradley WG Jr, Waluch V (1985) Blood flow: mag-
(2004) Preoperative embolization for meningeal netic resonance imaging. Radiology 154(2):443–450
tumors: evaluation of vascular supply with angio- 61. Keller PJ, Drayer BP, Fram EK, Williams KD,
CT. AJNR Am J Neuroradiol 25(1):74–76 Dumoulin CL, Souza SP (1989) MR angiography
48. Gross BA, Frerichs KU, Du R (2012) Sensitivity of with two-dimensional acquisition and three-
CT angiography, T2-weighted MRI, and magnetic dimensional display. Work in progress. Radiology
resonance angiography in detecting cerebral arterio- 173(2):527–532
venous malformations and associated aneurysms. 62. Felmlee JP, Ehman RL (1987) Spatial presaturation: a
J Clin Neurosci 19(8):1093–1095 method for suppressing flow artifacts and improving
49. Dowlatshahi D, Wasserman JK, Momoli F depiction of vascular anatomy in MR imaging. Radi-
et al (2014) Evolution of computed tomography angi- ology 164(2):559–564
ography spot sign is consistent with a site of active 63. van Tyen R, Saloner D, Jou LD, Berger S (1994) MR
hemorrhage in acute intracerebral hemorrhage. Stroke imaging of flow through tortuous vessels: a
45(1):277–280 numerical simulation. Magn Reson Med 31(2):
50. Wada R, Aviv RI, Fox AJ et al (2007) CT angiography 184–195
"spot sign" predicts hematoma expansion in acute 64. Huston J 3rd, Rufenacht DA, Ehman RL, Wiebers DO
intracerebral hemorrhage. Stroke 38(4):1257–1262 (1991) Intracranial aneurysms and vascular
410 J. Griauzde et al.

malformations: comparison of time-of-flight and tumours: comparison with selective digital subtrac-
phase-contrast MR angiography. Radiology 181(3): tion angiography. Neuroradiology 37(1):42–47
721–730 78. Sessa M (2008) Intracerebral hemorrhage and hyper-
65. Siegelman ES, Charafeddine R, Stolpen AH, Axel L tension. Neurol Sci 29(Suppl 2):S258–S259
(2000) Suppression of intravascular signal on 79. Qureshi AI, Tuhrim S, Broderick JP, Batjer HH,
fat-saturated contrast-enhanced thoracic MR arterio- Hondo H, Hanley DF (2001) Spontaneous intracere-
grams. Radiology 217(1):115–118 bral hemorrhage. N Engl J Med 344(19):1450–1460
66. Deutschmann HA, Augustin M, Simbrunner J 80. Mutlu N, Berry RG, Alpers BJ (1963) Massive cere-
et al (2007) Diagnostic accuracy of 3D time-of-flight bral hemorrhage. Clinical and pathological correla-
MR angiography compared with digital subtraction tions. Arch Neurol 8:644–661
angiography for follow-up of coiled intracranial aneu- 81. Nighoghossian N, Hermier M, Adeleine P et al (2002)
rysms: influence of aneurysm size. AJNR Am J Old microbleeds are a potential risk factor for cerebral
Neuroradiol 28(4):628–634 bleeding after ischemic stroke: a gradient-echo T2*-
67. Bosmans H, Wilms G, Marchal G, Demaerel P, Baert weighted brain MRI study. Stroke 33(3):735–742
AL (1995) Characterisation of intracranial aneurysms 82. Morgenstern LB, Hemphill JC 3rd, Anderson C
with MR angiography. Neuroradiology 37(4): et al (2010) Guidelines for the management of spon-
262–266 taneous intracerebral hemorrhage: a guideline for
68. Heidenreich JO, Schilling AM, Unterharnscheidt F healthcare professionals from the American Heart
et al (2007) Assessment of 3D-TOF-MRA at 3.0 Association/American Stroke Association. Stroke
Tesla in the characterization of the angioarchitecture 41(9):2108–2129
of cerebral arteriovenous malformations: a prelimi- 83. McEvoy AW, Kitchen ND, Thomas DG (2000) Intra-
nary study. Acta Radiol 48(6):678–686 cerebral haemorrhage and drug abuse in young adults.
69. Noguchi K, Melhem ER, Kanazawa T, Kubo M, Br J Neurosurg 14(5):449–454
Kuwayama N, Seto H (2004) Intracranial dural arte- 84. Hemphill JC 3rd, Bonovich DC, Besmertis L, Manley
riovenous fistulas: evaluation with combined 3D GT, Johnston SC (2001) The ICH score: a simple,
time-of-flight MR angiography and MR digital sub- reliable grading scale for intracerebral hemorrhage.
traction angiography. AJR Am J Roentgenol 182(1): Stroke 32(4):891–897
183–190 85. Vernooij MW, van der Lugt A, Ikram MA et al (2008)
70. Zhang H, Maki JH, Prince MR (2007) 3D contrast- Prevalence and risk factors of cerebral microbleeds:
enhanced MR angiography. J Magn Reson Imaging the Rotterdam Scan Study. Neurology 70(14):
25(1):13–25 1208–1214
71. Isoda H, Takehara Y, Isogai S et al (2000) MRA of 86. Greenberg SM, Vernooij MW, Cordonnier C
intracranial aneurysm models: a comparison of et al (2009) Cerebral microbleeds: a guide to detection
contrast-enhanced three-dimensional MRA with and interpretation. Lancet Neurol 8(2):165–174
time-of-flight MRA. J Comput Assist Tomogr 87. McCormick WF (1966) The pathology of vascular
24(2):308–315 (“arteriovenous”) malformations. J Neurosurg
72. Maki JH, Prince MR, Londy FJ, Chenevert TL (1996) 24(4):807–816
The effects of time varying intravascular signal inten- 88. Al-Shahi R, Bhattacharya JJ, Currie DG et al (2003)
sity and k-space acquisition order on three- Prospective, population-based detection of intracra-
dimensional MR angiography image quality. J Magn nial vascular malformations in adults: the Scottish
Reson Imaging 6(4):642–651 Intracranial Vascular Malformation Study (SIVMS).
73. Unlu E, Temizoz O, Albayram S et al (2006) Stroke 34(5):1163–1169
Contrast-enhanced MR 3D angiography in the assess- 89. Meyers PM, Schumacher HC, Higashida RT
ment of brain AVMs. Eur J Radiol 60(3):367–378 et al (2010) Indications for the performance of intra-
74. White PM, Wardlaw JM, Easton V (2000) Can non- cranial endovascular neurointerventional procedures.
invasive imaging accurately depict intracranial aneu- A scientific statement from the American Heart Asso-
rysms? A systematic review. Radiology 217(2): ciation Council on cardiovascular radiology and inter-
361–370 vention, Stroke council, council on cardiovascular
75. Locksley HB (1966) Natural history of subarachnoid surgery and anesthesia, interdisciplinary council on
hemorrhage, intracranial aneurysms and arteriove- peripheral vascular disease, and interdisciplinary
nous malformations. Based on 6368 cases in the council on quality of care and outcomes research.
cooperative study. J Neurosurg 25(2):219–239 J Neurointerv Surg 2(2):177–188
76. Suzuki M, Matsui O, Kobayashi K et al (2003) 90. Stapf C, Mast H, Sciacca RR et al (2006) Predictors of
Contrast-enhanced MRA for investigation of cerebral hemorrhage in patients with untreated brain arterio-
arteriovenous malformations. Neuroradiology 45(4): venous malformation. Neurology 66(9):1350–1355
231–235 91. Kidwell CS, Jahan R, Gornbein J et al (2013) A trial
77. Wilms G, Bosmans H, Marchal G et al (1995) Mag- of imaging selection and endovascular treatment for
netic resonance angiography of supratentorial ischemic stroke. N Engl J Med 368(10):914–923
18 Hemorrhagic Stroke 411

92. Zabramski JM, Wascher TM, Spetzler RF et al (1994) 107. Hallevi H, Albright KC, Aronowski J et al (2008)
The natural history of familial cavernous Intraventricular hemorrhage: anatomic relationships
malformations: results of an ongoing study. and clinical implications. Neurology 70(11):848–852
J Neurosurg 80(3):422–432 108. Diringer MN, Edwards DF, Zazulia AR (1998)
93. Huisman TA (2005) Intracranial hemorrhage: ultra- Hydrocephalus: a previously unrecognized predictor
sound, CT and MRI findings. Eur Radiol 15(3): of poor outcome from supratentorial intracerebral
434–440 hemorrhage. Stroke 29(7):1352–1357
94. Salmaggi A, Erbetta A, Silvani A, Maderna E, Pollo B 109. Sacco S, Marini C, Toni D, Olivieri L, Carolei A
(2008) Intracerebral haemorrhage in primary and met- (2009) Incidence and 10-year survival of intracerebral
astatic brain tumours. Neurol Sci 29(Suppl 2): hemorrhage in a population-based registry. Stroke
S264–S265 40(2):394–399
95. Kondziolka D, Bernstein M, Resch L et al (1987) 110. Zahuranec DB, Gonzales NR, Brown DL et al (2006)
Significance of hemorrhage into brain tumors: Presentation of intracerebral haemorrhage in a com-
clinicopathological study. J Neurosurg 67(6): munity. J Neurol Neurosurg Psychiatry 77(3):
852–857 340–344
96. Lansberg MG, Albers GW, Wijman CA (2007) 111. Davis SM, Broderick J, Hennerici M et al (2006)
Symptomatic intracerebral hemorrhage following Hematoma growth is a determinant of mortality and
thrombolytic therapy for acute ischemic stroke: a poor outcome after intracerebral hemorrhage. Neurol-
review of the risk factors. Cerebrovasc Dis 24(1): ogy 66(8):1175–1181
1–10 112. Hemphill JC 3rd, Farrant M, Neill TA Jr (2009) Pro-
97. Lansberg MG, Thijs VN, Bammer R et al (2007) Risk spective validation of the ICH Score for 12-month
factors of symptomatic intracerebral hemorrhage functional outcome. Neurology 73(14):1088–1094
after tPA therapy for acute stroke. Stroke 38(8): 113. Parizel PM, Makkat S, Van Miert E, Van Goethem
2275–2278 JW, van den Hauwe L, De Schepper AM (2001)
98. Singer OC, Humpich MC, Fiehler J et al (2008) Risk Intracranial hemorrhage: principles of CT and MRI
for symptomatic intracerebral hemorrhage after interpretation. Eur Radiol 11(9):1770–1783
thrombolysis assessed by diffusion-weighted mag- 114. Linfante I, Llinas RH, Caplan LR, Warach S (1999)
netic resonance imaging. Ann Neurol 63(1):52–60 MRI features of intracerebral hemorrhage within
99. Bousser MG (2000) Cerebral venous thrombosis: 2 hours from symptom onset. Stroke 30(11):
diagnosis and management. J Neurol 247(4):252–258 2263–2267
100. Leach JL, Fortuna RB, Jones BV, Gaskill-Shipley MF 115. Kim YW, Lawson MF, Hoh BL (2012)
(2006) Imaging of cerebral venous thrombosis: cur- Nonaneurysmal subarachnoid hemorrhage: an
rent techniques, spectrum of findings, and diagnostic update. Curr Atheroscler Rep 14(4):328–334
pitfalls. Radiographics 26(Suppl 1):S19–S41; discus- 116. Carvi y Nievas MN, Archavlis E (2009) Atypical
sion S42-13 causes of nontraumatic intracranial subarachnoid
101. Cianfoni A, Colosimo C (2011) Problem solving in hemorrhage. Clin Neurol Neurosurg 111(4):354–358
neuroradiology: expert consult, 1st edn. Elsevier, 117. Delgado Almandoz JE, Jagadeesan BD, Refai D
Philadelphia, pp 427–472 et al (2012) Diagnostic yield of repeat catheter angi-
102. Tong KA, Ashwal S, Holshouser BA et al (2004) ography in patients with catheter and computed
Diffuse axonal injury in children: clinical correlation tomography angiography negative subarachnoid
with hemorrhagic lesions. Ann Neurol 56(1):36–50 hemorrhage. Neurosurgery 70(5):1135–1142
103. Kim J, Smith A, Hemphill JC 3rd et al (2008) Contrast 118. Gee C, Dawson M, Bledsoe J et al (2012) Sensitivity
extravasation on CT predicts mortality in primary of newer-generation computed tomography scanners
intracerebral hemorrhage. AJNR Am J Neuroradiol for subarachnoid hemorrhage: a Bayesian analysis.
29(3):520–525 J Emerg Med 43(1):13–18
104. Goldstein JN, Fazen LE, Snider R et al (2007) Con- 119. Mark DG, Hung YY, Offerman SR et al (2013)
trast extravasation on CT angiography predicts hema- Nontraumatic subarachnoid hemorrhage in the setting
toma expansion in intracerebral hemorrhage. of negative cranial computed tomography results:
Neurology 68(12):889–894 external validation of a clinical and imaging predic-
105. Ederies A, Demchuk A, Chia T et al (2009) tion rule. Ann Emerg Med 62(1):1–10 e11
Postcontrast CT extravasation is associated with 120. Verma RK, Kottke R, Andereggen L et al (2013)
hematoma expansion in CTA spot negative patients. Detecting subarachnoid hemorrhage: comparison of
Stroke 40(5):1672–1676 combined FLAIR/SWI versus CT. Eur J Radiol
106. Becker KJ, Baxter AB, Bybee HM, Tirschwell DL, 82(9):1539–1545
Abouelsaad T, Cohen WA (1999) Extravasation of 121. Wu Z, Li S, Lei J, An D, Haacke EM (2010) Evalu-
radiographic contrast is an independent predictor of ation of traumatic subarachnoid hemorrhage using
death in primary intracerebral hemorrhage. Stroke susceptibility-weighted imaging. AJNR Am J
30(10):2025–2032 Neuroradiol 31(7):1302–1310
412 J. Griauzde et al.

122. Spickler E, Lufkin R, Teresi L et al (1990) MR imag- 131. Kimball MM, Velat GJ, Hoh BL (2011) Participants
ing of acute subarachnoid hemorrhage. Comput Med in the international multi-disciplinary consensus con-
Imaging Graph 14(1):67–77 ference on the critical care management of subarach-
123. Ibrahim GM, Weidauer S, Macdonald RL (2011) noid H. Critical care guidelines on the endovascular
Interobserver variability in the interpretation of management of cerebral vasospasm. Neurocrit Care
computed tomography following aneurysmal sub- 15(2):336–341
arachnoid hemorrhage. J Neurosurg 115(6): 132. Bakker NA, Metzemaekers JD, Groen RJ, Mooij JJ,
1191–1196 Van Dijk JM (2010) International subarachnoid aneu-
124. Atlas SW, Thulborn KR (1998) MR detection of rysm trial 2009: endovascular coiling of ruptured
hyperacute parenchymal hemorrhage of the brain. intracranial aneurysms has no significant advantage
AJNR Am J Neuroradiol 19(8):1471–1477 over neurosurgical clipping. Neurosurgery 66(5):
125. Sailer AM, Wagemans BA, Nelemans PJ, de Graaf R, 961–962
van Zwam WH (2014) Diagnosing intracranial aneu- 133. Brinjikji W, Kallmes DF, White JB, Lanzino G, Mor-
rysms with MR angiography: systematic review and ris JM, Cloft HJ (2010) Inter- and intraobserver agree-
meta-analysis. Stroke 45(1):119–126 ment in CT characterization of nonaneurysmal
126. Cirillo M, Scomazzoni F, Cirillo L et al (2013) Com- perimesencephalic subarachnoid hemorrhage. AJNR
parison of 3D TOF-MRA and 3D CE-MRA at 3T for Am J Neuroradiol 31(6):1103–1105
imaging of intracranial aneurysms. Eur J Radiol 134. Kong Y, Zhang JH, Qin X (2011) Perimesencephalic
82(12):e853–e859 subarachnoid hemorrhage: risk factors, clinical pre-
127. Pierot L, Portefaix C, Rodriguez-Regent C, Gallas S, sentations, and outcome. Acta Neurochir Suppl
Meder JF, Oppenheim C (2013) Role of MRA in the 110(Pt 1):197–201
detection of intracranial aneurysm in the acute phase 135. Buyukkaya R, Yildirim N, Cebeci H et al (2014) The
of subarachnoid hemorrhage. J Neuroradiol 40(3): relationship between perimesencephalic subarach-
204–210 noid hemorrhage and deep venous system drainage
128. Roos YB, Beenen LF, Groen RJ, Albrecht KW, pattern and calibrations. Clin Imaging 38(3):226–230
Vermeulen M (1997) Timing of surgery in patients 136. Gentry LR, Godersky JC, Thompson B, Dunn VD
with aneurysmal subarachnoid haemorrhage: (1988) Prospective comparative study of intermediate-
rebleeding is still the major cause of poor outcome field MR and CT in the evaluation of closed head
in neurosurgical units that aim at early surgery. trauma. AJR Am J Roentgenol 150(3):673–682
J Neurol Neurosurg Psychiatry 63(4):490–493 137. Ganz JC, Thuomas KA, Vlajkovic S et al (1993)
129. Nieuwkamp DJ, de Gans K, Algra A et al (2005) Changes in intracranial morphology, regional cerebral
Timing of aneurysm surgery in subarachnoid water content and vital physiological variables during
haemorrhage–an observational study in The Nether- epidural bleeding. An experimental MR study in
lands. Acta Neurochir (Wien) 147(8):815–821 dogs. Acta Radiol 34(3):279–288
130. Velat GJ, Kimball MM, Mocco JD, Hoh BL (2011) 138. Han JS, Kaufman B, Alfidi RJ et al (1984) Head
Vasospasm after aneurysmal subarachnoid hemor- trauma evaluated by magnetic resonance and com-
rhage: review of randomized controlled trials and puted tomography: a comparison. Radiology 150(1):
meta-analyses in the literature. World Neurosurg 71–77
76(5):446–454
Medical Therapy of Acute Stroke
19
Keith G. DeSousa and Albert S. Favate

Contents Management of Body Temperature . . . . . . . . . . . . . . . 420


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 414 Neuroprotection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 420
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 414 Deep Venous Thrombosis (DVT) Prophylaxis . . . 420
Initial Assessment of the Stroke Patient . . . . . . . . . . 415 Acute Stroke Units . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 416
Acute Therapy: Hemorrhagic Stroke . . . . . . . . . . . . . 421
Acute Therapy: Ischemic Stroke . . . . . . . . . . . . . . . . . . 416
Management of Intracranial Pressure . . . . . . . . . . . . 421
Intravenous Thrombolysis . . . . . . . . . . . . . . . . . . . . . . . . . 416
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 422
Antiplatelet Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 422
Anticoagulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418
Statin Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419
Blood Pressure Management
in Ischemic Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419
Fluid Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419
Management of Hyperglycemia . . . . . . . . . . . . . . . . . . . 420

K.G. DeSousa (*)


Department of Neurology, NYU Langone Medical Center,
New York, NY, USA
Vascular Neurology, University of Miami, Miami,
FL, USA
e-mail: Keith.DeSousa@nyumc.org;
drkeith5@gmail.com; k.desousa@med.miami.edu
A.S. Favate
NYU Langone Medical Center, New York, NY, USA
e-mail: Albert.Favate@nyumc.org

# Springer Science+Business Media New York 2016 413


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_40
414 K.G. DeSousa and A.S. Favate

heart, and vascular structures. Every stroke patient


Abstract
is an individual, and treatment must be guided
Approximately 800,000 strokes occur in the
with an understanding of the basic mechanism
United States each year. However, first and
that leads to the incident.
recurrent strokes are preventable. Stroke is
the fourth leading cause of death and a major
cause of disability in the United States (Go AS
Pathophysiology
et al, Heart disease and stroke statistics – 2014
update: a report from the American Heart
Ischemic stroke results when there is a decrease in
Association. Circulation 129(3):e28–e292,
blood flow to the brain resulting in tissue damage.
2014). Recent advances in medical therapy of
Criteria for classifying strokes by their underlying
stroke have resulted in greatly improved sur-
etiology are available. The stroke subtype classi-
vival rates from stroke. Early treatments as
fication includes large artery atherosclerosis,
well as preventative therapies can help mitigate
cardioembolism, small vessel occlusion, other
the brain damage that could be caused by
unusual but determined etiology, and
stroke.
undetermined etiology or cryptogenic [1]. The
The treatment of stroke varies by the type of
TOAST (Trial of Org 10172 in Acute Stroke
stroke, i.e., ischemic or hemorrhagic. The time
Treatment) classification system is still used in
since symptom onset is also critical to stroke
clinical practice as well as for studies.
management. Risk factor management and
At a molecular level, a process called
reduction also plays a key factor in stroke
“excitotoxicity” is responsible for cellular death
prevention. This chapter aims to discuss cur-
in ischemic tissue. Glutamate, which is normally
rent medical therapy of acute stroke.
present in synaptic terminals, is cleared from the
extracellular space in an energy-dependent pro-
Keywords
cess. Lack of blood flow results in decreased
Stroke/diagnosis • Stroke/therapy tissue plas-
levels of ATP (adenosine triphosphate) available
minogen activator • Telemedicine • Evidence-
for clearing glutamate. The high levels of gluta-
based medicine • Practice guidelines as topic •
mate cause calcium channels to remain open and
Quality assurance health care/standards
continuous membrane depolarization. There is an
influx of calcium, sodium, and chloride ions as
well as an efflux of potassium [2]. The elevated
Introduction calcium causes activation of destructive enzymes.
The osmotic gradient leads to cellular swelling.
Stroke is a term given to any brain damage caused All these events lead to a loss of cellular integrity.
by dysfunction in blood supply to the brain. The This correlates with MRI (magnetic resonance
term applies generally when symptoms begin imaging) diffusion-weighted imaging (DWI) as
abruptly. Stroke refers to an extremely broad this sequence helps demonstrate increased intra-
range of cerebral pathology that vary consider- cellular water. As a result of the aforementioned
ably. Rupture of a blood vessel and hemorrhage cellular cascade of events, the DWI image shows
into the cranial vault and occlusion of a miniscule high signal within minutes of onset of an acute
blood vessel with a small amount of tissue damage ischemic stroke (Fig. 1).
in an important area are both classified as strokes The brain does not, for the most part, have its
but the treatment varies considerably. C. Miller own energy stores and relies on blood flow for its
Fisher, one of the pioneers of vascular neurology, survival. Thus, it is susceptible to damage with
said “neurology is learned stroke by stroke.” To even short periods of lack of blood flow. Normal
treat a stroke patient, the clinician must be inti- cerebral blood flow (CBF) is around 50–60
mately familiar with the anatomy, physiology, and ml/100 g/min. The brain may respond to changes
pathology of not only the brain but the spinal cord, in blood flow by local vasodilatation, opening
19 Medical Therapy of Acute Stroke 415

collaterals and/or increasing the oxygen and glu- The majority of strokes are due to a small area
cose extraction from blood. Neuronal dysfunction of tissue that is deprived of blood flow. Often, a
occurs when the CBF decreases to around single blood vessel becomes blocked. The imme-
20 ml/100 g/min. Permanent damage occurs diate region surrounding the blood vessel is most
when the CBF goes below 10 ml/100 g/min [3]. affected – this is called the ischemic core where
cells are damaged and may die. Further away from
the core, the cells may still be receiving a small
amount of oxygen and glucose through diffusion
from collateral vessels. The CBF in these regions
is around 25–50 % of normal. The cellular integ-
rity may be preserved in these regions. These cells
can recover if blood flow is restored to them
quickly. This region is called the penumbra
[3]. Saving this tissue, the penumbra, is the goal
of reperfusion therapy. Perfusion imaging can
help visualize and separate tissue which is poten-
tially salvageable from that which has already
been infarcted (Fig. 2).

Initial Assessment of the Stroke


Patient

The initial assessment of a stroke patient requires


a broad approach as there are many other diagno-
ses which could mimic a stroke as well as stroke
patients generally have multiple comorbidities.
Fig. 1 Area of high signal intensity seen on diffusion- The basics should not be overlooked and the
weighted imaging suggestive of acute ischemic stroke patient should be assessed for airway, breathing,

Fig. 2 MRI perfusion imaging. (a) CBV (cerebral blood contrast in the left hemisphere). The regions outside of
volume reduced in an area which likely corresponds with the infarcted area with slow arrival of contrast represent
infarcted tissue), (b) CBF (cerebral blood flow which is potentially salvageable tissue amenable to reperfusion
reduced in an area larger than ischemic core), and (c) MTT strategies
(mean transit time which demonstrates slow arrival of
416 K.G. DeSousa and A.S. Favate

and circulation. Attention should be paid to any • Further investigations should be performed for
other condition which could easily be reversible – evaluation of stroke etiology.
such as hypoglycemia. A quick, concise history
should be obtained including last known time
normal as this will guide further therapy. A rapid Acute Therapy: Ischemic Stroke
assessment to determine if a patient is a candidate
for thrombolytic therapy should be completed The phrase “time is brain” was intended to empha-
including National Institutes of Health Stroke size that brain tissue is rapidly lost during a stroke
Scale (NIHSS) and evaluation of inclusion and and rapid evaluation and treatment is necessitated.
exclusion criteria for tPA (tissue plasminogen During each minute in which a large vessel ische-
activator/brand name: Alteplase) as described mic stroke goes untreated, the patient can lose 1.9
later in this chapter. Point-of-care blood glucose million neurons, 13.8 billion synapses, and 12 km
should be obtained. The NIHSS, which quantifies (7 miles) of axonal fibers [5].
the neurological exam, should be performed in the Candidates for acute therapy should be identi-
first 10 min from arrival prior to imaging if possi- fied as rapidly as possible. The goal of treatment
ble. Two IV lines (preferably 18 g) should be during an ischemic stroke should be the restora-
placed in case thrombolytic therapy or if tion of blood flow as quickly and safely as possi-
contrast-enhanced imaging is necessary. Comput- ble. The approach to acute ischemic stroke
erized tomography (CT) should be done within treatment can be divided into vascular and neuro-
minutes of arrival and interpreted shortly thereaf- nal strategies. Noninterventional vascular strate-
ter. Labs including CBC (complete blood count), gies focus on recanalization, increasing blood
CMP (comprehensive metabolic panel), PT (pro- flow to the penumbra, and avoidance of clot prop-
thrombin time), PTT (partial thromboplastin agation. Neuronal strategies focus on novel treat-
time), INR (international normalized ratio), and ments such as neuroprotection.
cardiac enzymes (if indicated) should be drawn.
Lytic therapy may be initiated in the imaging suite
to avoid delays. It may be started without waiting Intravenous Thrombolysis
for laboratory test results unless the patient is on
anticoagulation or has history of known bleeding The first NINDS (National Institute of Neurolog-
diathesis. An electrocardiogram (EKG) should be ical Disorders and Stroke) trial had shown that IV
completed as well. Next, investigations should be tPA had improved patient functional outcome at
tailored to determine the cause of the stroke in 3 months if the patient was treated within 3 h of
each individual patient [4]. symptom onset [6]. Later trials including ECASS
3 (European Cooperative Acute Stroke Study)
showed that IV tPA not only was safe but patients
Summary benefitted from receiving the medication up to
4.5 h after stroke onset [7]. However, although
• Obtain a quick, concise history including last the use of IV tPA within the 3–4.5 h window is
known time normal, past medical history, risk generally accepted, it is currently not FDA (Food
factors for stroke, and medications, and per- and Drug Administration) approved.
form evaluation of inclusion and exclusion Multiple prior analyses of data of patients who
criteria for tPA. received tPA have shown that each 15 min reduc-
• NIHSS should be performed. tion in time to ignition of tPA treatment was asso-
• IV lines placed. ciated with an increase in the odds of walking
• Non-contrast head CT should be done and rap- independently at discharge and being discharged
idly interpreted. to home rather than an institution. The same
• Labs should be drawn including CBC, CMP, 15 min reduction in time to tPA treatment was
PT, PTT, and INR and EKG completed. associated with a decrease in the odds of death
19 Medical Therapy of Acute Stroke 417

Table 1 Guidelines for the use of IV tissue plasminogen activator (tPA)


Warningsa (relative
Indications Contraindications contraindications)
>18 years of age Hemorrhage on imaging Abnormal blood glucose (<50
mg/dL or >400 mg/dL)
Measurable deficit Subarachnoid hemorrhage on imaging Severe neurologic deficit
NIHSS (NIHSS > 22)
Symptom onset within Serious head or spinal trauma within the last 3 months Major and early infarct signs on
3h CT
No hemorrhage on CT History of intracranial hemorrhage Minor neurologic deficit
No contraindications Uncontrolled hypertension SBP > 185 or diastolic >110 Rapidly improving symptoms
(see next column) mmHg
Seizures at onset of stroke
Active internal bleeding
Intracranial neoplasm, AVM, or aneurysm
Known bleeding diathesis (INR > 1.7, PT > 15, heparin
within the past 48 h w/elevated aPTT
Platelet count <100,000
a
In these certain situations, benefits should be weighed against risks

before discharge and symptomatic hemorrhagic taken into consideration when offering tPA to
transformation of infarction. Given these findings, patients. The results of ECASS 3 showed that
one should strive to give tPA as rapidly as possible significantly more patients had a better outcome
after identification of a potential candidate rather with IV tPA than placebo. There was no difference
than later [8]. in mortality between IV tPA and placebo. Symp-
The initial NINDS stroke trial included tomatic intracranial hemorrhage was significantly
patients with ischemic stroke who could be treated more likely to occur with IV tPA administration
with IV tPA within 3 h of symptom onset. Six than with placebo [7].
hundred twenty-four patients were randomized IV tPA should be offered to patients presenting
to treatment with placebo or IV tPA (0.9 mg/kg within the 4.5 h (3–4.5 h use is recommended by
up to 90 mg, 10 % as a bolus followed by the rest American Heart Association/American Stroke
over 60 min). At 3 months a complete or near Association (AHA/ASA) but not FDA approved
recovery occurred in a statistically greater number and therefore off-label) window who meet the
of patients assigned to IV tPA than placebo (38 % inclusion and exclusion criteria. Selection of can-
vs. 21 %). Three-month mortality was not signif- didates for intravenous thrombolysis requires a
icantly different between the two groups. Severe neurologic evaluation, neuroimaging, determina-
systemic hemorrhage occurred in less than 1 % of tion of eligibility in accordance with the inclusion,
patients [6]. and exclusion criteria (Table 1).
ECASS 3 trial assigned 821 patients with acute Today’s guidelines for tPA administration
ischemic stroke to receive IV tPA or placebo. come from the original trials designed to show
Additional exclusion criteria were included in the efficacy of tPA in acute stroke patients.
this study. Patients greater than 80 years old, Noncompliance with the guidelines has been
NIHSS >25, those with a combination of previ- associated with an increase in the risk of hemor-
ous stroke and diabetes, and those on anticoagu- rhage [9]. However, strict compliance with these
lants regardless of INR were excluded from criteria has excluded patients who may have
participation in the study. This was done to satisfy benefitted from receiving tPA. Therefore, in
safety concerns from the European regulatory extreme emergent situations, a rational individu-
agency. Depending on local stroke center guide- alized decision may be necessary for a certain
lines, the exclusion criteria from ECASS 3 may be patient.
418 K.G. DeSousa and A.S. Favate

Before starting IV tPA, the treating physician be initiated to evaluate for the source of bleeding
should make sure that treatment is within 4.5 h of and tailored to the individual circumstance [9].
onset of symptoms, there is a measurable
neurologic deficit, inclusion and exclusion criteria
are met, non-contrast head CT or MRI is done Antiplatelet Agents
and does not show hemorrhage or large infarct,
blood pressure parameters are met, IV lines are in Timely restoration of blood flow in acute ischemic
place, and accurate body weight has been stroke is the most effective way for saving brain
determined. tissue that has not yet been infarcted. Again, IV
Serum glucose should be checked to ensure thrombolytic therapy should always be consid-
that hypoglycemia is not a cause of the precipitat- ered in patients who meet the eligibility criteria.
ing neurologic findings. Patients with low platelet Early administration of aspirin has been shown
counts or elevated prothrombin times should be to be beneficial in large randomized controlled
excluded. Uncontrollable hypertension is also trials [10]. Recently, dual antiplatelet therapy
exclusionary. with aspirin and clopidogrel has been shown to
The tPA dose is calculated at 0.9 mg/kg of have better outcomes in a Chinese population with
actual body weight with a maximum dose of TIA or minor stroke [11].
90 mg. Ten percent of the dose is given as a The International Stroke Trial (IST) showed
bolus over one minute. The rest is infused over that among 19,435 patients who received
an hour. Following administration of tPA, close 300 mg aspirin, there was a significant reduction
monitoring of the patient is required with frequent in the number of patients who had recurrent stroke
assessment of vital signs and neurologic status as at 14 days (2.8 % vs. 3.9 %). There was also a
per local institution guidelines. A follow-up CT reduction in the combined outcome of nonfatal
should be done 24 h post-tPA administration or stroke or death (11.3 % vs. 12.4 %) [10].
sooner if there is any deterioration in The CHANCE (Clopidogrel in High-risk
clinical exam. Patients with Acute Non-disabling Cerebrovascu-
Tight blood pressure control is essential during lar Events) trial randomized Chinese patients to
the administration of thrombolytic therapy. The receive either dual antiplatelet therapy with aspi-
blood pressure must be less than or equal to rin and clopidogrel or placebo plus aspirin within
185/110 mmHg before starting the medication. 24 h after the onset of TIA or minor stroke. The
Aggressive blood pressure lowering can be study showed that after 90 days there was a sig-
achieved using a variety of antihypertensives. nificant reduction in stroke for the aspirin and
Labetalol or IV nicardipine are routinely used. If clopidogrel group vs. the aspirin plus placebo
blood pressure cannot be reliably controlled, group (8.2 % vs. 11.7 %). The absolute risk reduc-
thrombolytic therapy should not be administered. tion was 3.5 % (hazard ratio 0.68, 95 % CI
Following administration of tPA, the blood pres- 0.57–0.81). Hemorrhagic stroke occurrence was
sure needs to be kept below 180/105 for at least similarly low in both groups (0.3 %) [11].
24 h.
Symptomatic intracranial hemorrhage is one of
the most feared complications of thrombolytic Anticoagulation
therapy. If such an event is suspected, discontinu-
ation of tPA is warranted. A stat non-contrast head Anticoagulation has consistently not been shown
CT or MRI should be completed for evaluation. to be beneficial in many situations of acute ische-
Administration of medication to reverse the mic stroke and in some studies have been shown
effects of the thrombolytic therapy should be con- to have worse outcomes. Guidelines by the Amer-
sidered including cryoprecipitate and/or platelets. ican Heart Association issued in 2013 have not
If systemic bleeding occurs, tPA administration recommended anticoagulation to be routinely
should also be halted, and further workup should used for treatment of acute ischemic stroke [9].
19 Medical Therapy of Acute Stroke 419

However, certain circumstances may warrant hypertension or other stroke-mediated causes.


anticoagulation such as intracardiac thrombus, The elevated blood pressure may be required and
mechanical heart valves, TIA (transient ischemic a natural protective response to help under-
attack) with a high chance of recurrence or some perfused tissues [15]. In this case further
arterial dissections, etc. In these situations, ther- neurovascular imaging is required to help guide
apy should be tailored to the individual patient. In treatment. It has been seen in multiple studies that
patients with large territorial infarctions on imag- aggressive acute blood pressure lowering in acute
ing, anticoagulation should be avoided if possible. stroke has been associated with increased neuro-
Also, if anticoagulation is required, therapy logic deterioration. Sometimes patients may ben-
should be started without a bolus as there may efit from artificially raising blood pressure if it is
be an increased risk of hemorrhagic transforma- too low. Caution must be taken as patients may
tion of the stroke. develop hypertensive encephalopathy from blood
pressures that are too high.
As described earlier, patients who are receiving
Statin Therapy thrombolytic therapy should have their blood
pressure treated so that it is less than or equal to
Lipid-lowering therapy with a statin has been 185/110 mmHg. It should be maintained at or
shown to reduce the risk of recurrent stroke and below 180/105 mmHg for at least 24 h after tPA
other cardiovascular events [12]. An observa- administration.
tional study of patients who were already on statin For patients presenting with ischemic stroke
therapy during a stroke and continued statin ther- who are not candidates for reperfusion therapy,
apy while inpatient had improved outcomes at treatment of blood pressure is not recommended
discharge as well as increased survival [13]. Initi- during the first 24 h unless the pressure elevation
ation of statin therapy early during hospitalization is >220 mmHg or there is evidence of other
has been shown to increase survival [14]. end-organ damage such as resulting renal failure,
aortic dissection, hypertensive encephalopathy,
eclampsia/preeclampsia, etc. In most situations,
Blood Pressure Management antihypertensive medication can slowly be
in Ischemic Stroke restarted after the first 24 h following a stroke
unless there is specific contraindication. Chroni-
Cerebral blood flow can be controlled by chang- cally hypertensive patients would require a more
ing the diameter of vessels which alters their gradual lowering of blood pressure because of
resistance. Larger diameters result in increased improper cerebral blood flow autoregulation. If
blood volume and more cerebral blood flow. treatment is required, the generally accepted
Smaller diameters have the opposite effect. blood pressure lowering is 15 % in the first
Changes in the cerebral perfusion pressure will 24 h [4].
also affect the blood flow. Cerebral blood flow is
under autonomic control and can self-adjust as a
response to changes in many different parameters. Fluid Management
Autoregulation can maintain blood flow within a
mean arterial pressure (MAP) of 60–150 mmHg. Most patients with acute ischemic stroke are
When the MAP is outside this range, cerebral euvolemic or hypovolemic. The goal should be
autoregulation fails as the brain is unable to com- to maintain the patient at a euvolemic state.
pensate. Therefore, there is a risk of ischemia at Hypovolemia may result in hypoperfusion of
low and high pressures [15]. ischemic tissue and thus worsen the infarct.
During an acute ischemic stroke, blood pres- Hypervolemia can worsen cerebral edema. The
sures may be elevated by an acute sympathetic hypovolemic patient should be rapidly fluid resus-
response, and the patient has underlying chronic citated if possible (care must be taken in heart
420 K.G. DeSousa and A.S. Favate

failure patients) followed by maintenance fluids. outcomes. Around one-third of patients with
Euvolemic patients can be maintained based on stroke may present with concomitant temperature
body weight and estimated fluid losses. Normal >37.6  C [16]. The source of the fever should be
saline is recommended because it is distributed sought by the physician. Urinary tract infections,
evenly into the extracellular spaces. Half-normal pneumonia, and sepsis are all sources of fever
saline can also worsen cerebral edema and should which could be treated. Acetaminophen or aspirin
be avoided if possible [4]. has some effect at bringing down body tempera-
ture. Cooling blankets or cool saline infusions
may also be used [4].
Management of Hyperglycemia There is some possible evidence that hypother-
mia may be beneficial especially in the setting of
Patients with diabetes have approximately twice anoxia or cardiac arrest. However, the use of
the risk of stroke. Diabetes affects approximately hypothermia as a method of neuroprotection is
8 % of the population of the United States. The currently being studied and results are not
prevalence of diabetes in stroke patients is available.
approximately 15–33 %. Hyperglycemia is com-
mon during acute stroke, and multiple trials have
shown that more than 40 % of stroke patients have
Neuroprotection
hyperglycemia on admission.
Normal fasting glucose is defined as <100
There have been multiple neuroprotective agents
mg/dL. Impaired fasting glucose is 100–125
that have shown benefit in animal models. How-
mg/dL. A fasting plasma glucose level >126
ever, none has been shown to have any benefit in
mg/dL, hemoglobin A1C >6.5 %, and casual
humans. There are multiple clinical trials that are
plasma glucose >200 mg/dL with symptoms
ongoing currently. One is currently looking at the
meet diagnostic criteria for diabetes [4].
use of high-dose lovastatin given quickly after the
Multiple studies have found worse outcomes
onset of stroke. Currently this study is still enroll-
associated with hyperglycemia. In patients who
ing patients and no data is available yet.
received IV tPA with hyperglycemia, it has been
shown that there are higher rates of intracerebral
hemorrhage and worse outcomes.
Currently, there are no large-scale trials Deep Venous Thrombosis (DVT)
looking at aggressive glucose control and stroke Prophylaxis
outcomes. Currently, the guidelines state that glu-
cose should be maintained between 140 and DVT or pulmonary embolism (PE) is a feared
180 mg/dL in all hospitalized patients as per complication of stroke. A nonfunctional,
American Diabetes Association guidelines. nonmobile limb is a setup to form a clot. All
There are multiple protocols for glucose control patients with acute stroke should be started on
and can vary from hospital to hospital. Care must some sort of DVT prophylaxis and it can be
be taken while managing blood glucose as the started soon after a stroke if there is no evidence
main risk from aggressive blood glucose control of intracranial hemorrhage. DVT prophylaxis
result is hypoglycemia. should not be started until after 24 h and negative
imaging for hemorrhage after receiving IV tPA.
Low molecular weight heparin has been shown to
Management of Body Temperature be slightly more effective at lowering the risk of
DVT/PE than unfractionated heparin in ischemic
If possible stroke patients should be maintained as stroke patients. IV heparin may need to be started
close to normothermic as possible. Hyperthermia in the event of an acute PE or DVT followed by
has been associated with poor neurological long-term anticoagulation [4]. Starting DVT
19 Medical Therapy of Acute Stroke 421

treatments should be tailored to the patient based sulfate should be considered in patients on IV
on comorbidities. heparin therapy. Platelet transfusion is question-
able in patients with intracranial hemorrhage on
antiplatelet therapy. Platelet transfusion may be
Acute Stroke Units necessary in patients with thrombocytopenia. Cer-
tain factor replacement may be necessary in
New evidence suggests that patients who have patients with severe coagulation factor deficiency.
been hospitalized in a unit specializing in stroke Hypertension is often a cause of hemorrhagic
care consistently have better outcomes regardless stroke and therefore blood pressure control plays a
of the type of stroke [17]. These specialized units key role in management. High blood pressure may
have dedicated physicians trained in stroke care as cause continued bleeding; however, too low blood
well; dedicated support staff including nurses and pressure may inhibit cerebral perfusion. The
physical and occupational therapists; and also INTERACT 2 (Intensive Blood Pressure Reduc-
quick access to multimodal imaging including tion in Acute Cerebral Hemorrhage Trial 2) trial
MRI, CT, ultrasound, and diagnostic angiography randomized patients to either aggressive blood
as well as trained neurointerventionalists. As per pressure lowering with a target systolic blood
the current guidelines, patients should be hospi- pressure (SBP) <140 mmHg or traditional man-
talized in stroke units whenever possible. Emer- agement with a target of SBP <180 mmHg.
gency medical services should also attempt to Adverse events were similar in the two groups,
bring patients to dedicated stroke centers [4]. and there was reduced disability in patients receiv-
The NIH has set recommended response times ing aggressive blood pressure control
for acute ischemic stroke. This is known as the [18]. Smaller studies have shown that there is
“golden hour” of evaluation and treatment. Within reduced hematoma expansion in patients who
10 min the patient should be seen by a physician underwent aggressive blood pressure manage-
and history, lab work, and NIHSS completed. The ment in the setting of intracerebral hemorrhage.
stroke team should be notified within 10 min. CT
scanning should be done within 25 min and labs
and CT should be interpreted within 45 min. IV Management of Intracranial Pressure
thrombolytic therapy should be initiated in less
than 60 min from arrival [4]. The Monroe-Kellie hypothesis recognizes that the
cranial vault is a fixed structure. The volume
inside the cranium has limited compliance. There-
Acute Therapy: Hemorrhagic Stroke fore, the volume of blood, cerebrospinal fluid
(CSF), and brain tissue should be in a state of
Although ischemic stroke is the more common equilibrium. If there is an increase in one of the
form of stroke, hemorrhagic stroke is an important components, there must be a decrease in another.
cause of morbidity and mortality. Initial treatment Increased intracranial pressure (ICP) can result
includes both medical and surgical treatment. The from the hematoma itself but also from the
most important goal should be the prevention of resulting edema (this can also result from edema
hemorrhage extension and management of intra- caused by ischemic stroke). As a result, the dam-
cranial pressure. aged brain may crush or shift normal tissue and
For all patients with intracerebral hemorrhage cause hydrocephalus or, worse, brain herniation.
on anticoagulation or antiplatelet agents, these Therefore, raised ICP should be managed aggres-
agents should be discontinued immediately and sively. Steps to reduce ICP can vary from simple
reversal of their effects should be initiated. measures to more invasive and radical
Vitamin K, fresh frozen plasma (FFP), or pro- approaches.
thrombin complex concentrate (PCC) should be Elevation of the head of bed to increase venous
given to patients on warfarin therapy. Protamine drainage from the head can lower ICP.
422 K.G. DeSousa and A.S. Favate

Hyperventilation to lower the PaCO2 to 25–30 can • In settings of intracerebral hemorrhage, steps
produce a large lowering of ICP; however, this is should be taken to reverse coagulopathy if
only a temporizing measure and is only effective indicated. Aggressive blood pressure control
for a few hours. Intravenous mannitol can be used appears to be safe in these patients.
as well. It is given as a bolus of 1 g/kg followed by • ICP management is critical management of
0.25–0.5 g/kg every 6 h. Hypertonic saline to acute stroke patients. There are various
achieve a serum sodium level of around methods that can be used to reduce ICP and
150 mEq/L may also be used. Further manage- should be tailored to the individual patient.
ment including surgical decompression or
ventriculostomy placement may be necessary to
reduce ICP [19]. References
1. Adams HP Jr et al (1993) Classification of subtype of
acute ischemic stroke. Definitions for use in a multi-
Summary center clinical trial. TOAST. Trial of Org 10172 in
acute stroke treatment. Stroke 24(1):35–41
• Early treatment of stroke starts with emergency 2. Rossi DJ, Oshima T, Attwell D (2000) Glutamate
medical services (EMS). Prompt recognition, release in severe brain ischaemia is mainly by reversed
uptake. Nature 403(6767):316–321
assessment, and transport to a qualified stroke
3. Markus HS (2004) Cerebral perfusion and stroke.
center are recommended. J Neurol Neurosurg Psychiatry 75(3):353–361
• An organized protocol of triage and evaluation 4. Jauch EC et al (2013) Guidelines for the early manage-
should be in place at the stroke center. Rapid ment of patients with acute ischemic stroke: a guideline
for healthcare professionals from the American Heart
evaluation using imaging, blood work, and a
Association/American Stroke Association. Stroke
stroke rating scale such as the NIHSS should 44(3):870–947
be completed. 5. Saver JL (2006) Time is brain–quantified. Stroke
• All acute ischemic stroke patients should be 37(1):263–266
6. (1995) Tissue plasminogen activator for acute ischemic
evaluated for possible thrombolytic therapy.
stroke. The National Institute of Neurological Disor-
• Blood pressure should be controlled as per ders and Stroke rt-PA Stroke Study Group. N Engl J
guidelines if the patient is a candidate for Med 333(24):1581–1587
thrombolytic therapy. Otherwise they should 7. Hacke W et al (2008) Thrombolysis with alteplase 3 to
4.5 hours after acute ischemic stroke. N Engl J Med
have their blood pressure cautiously lowered.
359(13):1317–1329
• Limited evidence is available for the use of 8. Hacke W et al (2004) Association of outcome with
anticoagulation in acute ischemic stroke and early stroke treatment: pooled analysis of ATLANTIS,
should be avoided except for certain ECASS, and NINDS rt-PA stroke trials. Lancet
363(9411):768–774
circumstances.
9. Graham GD (2003) Tissue plasminogen activator for
• Oral administration of antiplatelet agents such acute ischemic stroke in clinical practice: a meta-
as aspirin is recommended for patients with analysis of safety data. Stroke 34(12):2847–2850
acute ischemic stroke within 24 h of onset of 10. (1997) The International Stroke Trial (IST): a
randomised trial of aspirin, subcutaneous heparin,
stroke symptoms. Antiplatelet agents should
both, or neither among 19435 patients with acute
not be started until at least 24 h after receiving ischaemic stroke. International Stroke Trial Collabora-
thrombolytic therapy and follow-up imaging tive Group. Lancet 349(9065):1569–1581
has not revealed any major hemorrhage. 11. Wang Y et al (2013) Clopidogrel with aspirin in acute
minor stroke or transient ischemic attack. N Engl J Med
• Statin therapy is generally safe to be continued
369(1):11–19
in patients with acute ischemic stroke or may 12. Amarenco P et al (2006) High-dose atorvastatin after
be administered for secondary stroke stroke or transient ischemic attack. N Engl J Med
prevention. 355(6):549–559
13. Blanco M et al (2007) Statin treatment withdrawal in
• Blood sugar should be managed according to
ischemic stroke: a controlled randomized study. Neu-
standard guidelines. rology 69(9):904–910
19 Medical Therapy of Acute Stroke 423

14. Ni Chroinin D et al (2011) Association between acute 17. Candelise L et al (2007) Stroke-unit care for acute
statin therapy, survival, and improved functional out- stroke patients: an observational follow-up study. Lan-
come after ischemic stroke: the North Dublin popula- cet 369(9558):299–305
tion stroke study. Stroke 42(4):1021–1029 18. Anderson CS et al (2013) Rapid blood-pressure lower-
15. Qureshi AI (2008) Acute hypertensive response in ing in patients with acute intracerebral hemorrhage.
patients with stroke: pathophysiology and manage- N Engl J Med 368(25):2355–2365
ment. Circulation 118(2):176–187 19. Dunn LT (2002) Raised intracranial pressure. J Neurol
16. Azzimondi G et al (1995) Fever in acute stroke worsens Neurosurg Psychiatry 73(Suppl 1):i23–i27
prognosis. A prospective study. Stroke 26(11):2040–2043
Endovascular Treatment of Stroke
20
Paolo Machi and Paolo Garofalo

Contents Keywords
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 425
Ischemic stroke • Thrombectomy •
Synthesis Trial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 425 Endovascular treatment • Stent retrievers •
Interventional Management of Stroke III . . . . . . . . . . . . 426 Clot retrievers • Tandem occlusions • Intrave-
MR Rescue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 426 nous fibrinolysis • Embolectomy • Intracranial
Stroke Trials of the “New Era” . . . . . . . . . . . . . . . . . . . . . . 427
MR CLEAN Trial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 427
revascularization
ESCAPE Trial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 427
EXTEND IA Trial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 428
Intra-arterial Recanalization Techniques . . . . . . . . . 428 Introduction
Clot (Stent) Retrievers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 428
Procedure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 429 In 2013 results of three trials comparing the effi-
Selection of Patients for Endovascular
cacy of intra-arterial approach (IAA) versus intra-
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 430
Tandem Occlusions: Mechanical Recanalization venous fibrinolysis (IVF) for the treatment of the
Techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 430 acute ischemic stroke were published in the New
Rationale for Endovascular Treatment of Tandem England Journal of Medicine. IVF was at that
Occlusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 431
time the treatment of reference for acute ischemic
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 437 stroke, even if several retrospective clinical series
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 437 found that anatomical and clinical outcomes were
better when such treatment was associated with an
endovascular approach. These trials and their neg-
ative results have not influenced the
neurovascular community, above all, because of
several study biases.

Synthesis Trial
P. Machi (*)
Department of Neuroradiology Unit, CHU Gui de This trial [1] compared the 90-day clinical out-
Chauliac, Montpellier University Hospital, Montpellier, come of two groups of patients presenting within
France
e-mail: paolo.machi@gmail.com 4.5 h from the onset of the acute ischemic stroke
treated with IVF or IAA. One hundred and eighty-
P. Garofalo
Medicine School, University of Parma, Parma, Italy one patients assigned to the standard care arm
e-mail: paolo.garofalo89@gmail.com were administered rt -PA 2.75 h (median) after
# Springer Science+Business Media New York 2016 425
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_41
426 P. Machi and P. Garofalo

the stroke onset. One hundred and eighty-one 434 patients of this cohort received IVF, while
patients assigned to the intervention arm received, 222 IVF plus endovascular approach. The 90-day
by a median of 3.75 h after the stroke onset, intra- outcome, assessed by using the mRS, did not sig-
arterial (“in situ”) fibrinolysis or mechanical clot nificantly differed through the two groups. 40.8 %
fragmentation or removal or a combination of of patients of the intervention group presented with
these therapies. The 90-day evaluation showed a mRS score 2 similarly to the 38.7 % of the
better clinical evolution for patients assigned to standard care group. The study showed no signif-
the control group. Sixty three out of 181 patients icant difference in terms of clinical outcome com-
(34.8 %) of the control group presented without paring the interventional and the standard care
disability with a modified Rankin scale (mRS) groups. Again, imaging criteria selection of this
score 2, compared to 55 out of 181 patients trial (as for the Synthesis trial) represented an inclu-
(30.4 %) of the intervention group. The study sion bias because, at least in the early period, no
showed that mechanical treatment was not supe- brain vessel investigation was performed before
rior to standard care in the treatment of acute randomization. Furthermore, only 4 out of
ischemic stroke. One of the biases of this trial 434 patients of this cohort received endovascular
was that patients randomized to the intervention treatment performed with a dedicated stent
arm received different approaches, considered retriever. In addition, subjects of the interventional
nowadays not effective, giving to the study a group had to be treated within 7 h from the stroke
very low level of reproducibility. In 109 patients onset. In this group late endovascular treatment led,
of the intervention arm, the treatment was in some instances, to useless recanalization.
performed without the use of a “real” Authors offered as explanation of the neutral
thrombectomy device. This subgroup of subjects results of their study the fact that patients of the
received “in situ” intra-arterial administration of intervention group were treated with “old-gener-
rt-PA; in some instance, manual clot fragmenta- ation” thrombectomy devices and this was related
tion was performed by using a micro-wire. Only to low rate of vessel recanalization.
56 patients of the intervention group were treated
with a “real” clot removal device, and only
23 patients out of 163 of the intervention group MR Rescue
were treated with a stent retriever. Moreover,
patients of the intervention group did not receive This third trial [3] was designed, as well, to eval-
rt-PA while awaiting for the mechanical treatment. uate the 90-day outcome of stroke patients treated
Another bias of this study was the imaging used with IVF or IVF associated with IAA until 8 h
for patient selection. The only radiological inves- after stroke. In this trial CT and MRI scan were
tigation performed before randomization was a used to stratify randomization in favorable and
brain non-contrast-enhanced CT scan. This exam- non- favorable imaging patterns. Among the
ination presents limitations for the assessment of 127 patients randomized, 70 undergo
the infarct extension and vessel occlusion thrombectomy, while 57 received IVF. Only
identification. 58 % of the treated subjects presented a favorable
penumbral pattern. In 67 % of patients of the
intervention group, an occluded arterial recanali-
Interventional Management zation was achieved. At the 3-month follow-up,
of Stroke III the mean mRS score did not differ between
patients of the two groups (3.9 vs. 3.9). Interven-
This was a randomized trial [2] comparing out- tional treatment was not superior to IVF either in
come of stroke patients treated within 3 h from subjects with or without a favorable penumbral
stroke onset by stand-alone IVF or by IVF and pattern. In the analysis of the 90-day outcome, no
IAA. This trial was stopped prematurely because interaction was found between pretreatment imag-
of futility after randomization of 656 subjects. ing pattern and treatment assignment.
20 Endovascular Treatment of Stroke 427

The study showed that a favorable penumbral conducted by randomizing 500 eligible patients
pattern on neuroimaging did not identify patients presenting with acute ischemic stroke caused by
who would benefit from IAA. Moreover, an intracranial occlusion of the distal intracranial
thrombectomy was not superior to standard IFV carotid artery, middle cerebral artery (M1/M2), or
for the treatment of acute ischemic stroke. Never- anterior cerebral artery (A1/A2) and NIHSS score
theless, authors suggested that probably “second-  of 2. Patients were selected with baseline CT
generation” stent retrievers, due to their high effec- scan to evaluate the ischemic core by using the
tiveness in arterial reopening, would have a treat- ASPECT score. CT angiography, MR angiography,
ment benefit in patients with a ischemic penumbra. or DSA were used to evaluate the baseline vessel
imaging. Two hundred and thirty-three patients
(46.6 %) were assigned to the intervention group
Stroke Trials of the “New Era” and 267 patients (53.4 %) were assigned to the
control group. Mechanical treatment was
The trial reported above failed in demonstrating the performed in 195 out of 233 patients (83.7 %)
superiority of the IAA over standard IVF for the and clot retrievers were used in 190 out of
treatment of acute ischemic stroke. However, these 195 (97 %) patients. A good brain vessel reperfu-
trials presented several biases; subjects randomized sion was obtained in 115 out of 196 (58.7 %)
to the intervention group did not receive the same patients in the intervention group. This was related
treatment and only a small number among them to an increase in functional independence and
were treated with “second-generation” clot reduced mortality at 90-day follow-up. The study
retrievers. In addition, imaging selection criteria showed, for the first time, that the mechanical
were not the same in all participating centers. More- thrombectomy was effective and safe when admin-
over, given that clinical evolution is related to the istered within 6 h after stroke onset to patients with
delay of the artery reopening, the delay of treatment acute ischemic stroke caused by a proximal intra-
of patients randomized to the intervention group cranial occlusion of the anterior circulation.
was in some instances too long (up to 8 h). Based
on results of these trials, other trials have been
designed in order to better evaluate comparison ESCAPE Trial
between standard fibrinolysis and interventional
treatment. In these subsequent trials patients ESCAPE [5] was a multicenter, prospective ran-
assigned to the intervention arm were treated within domized, open-label controlled trial with blinded
4.5 h after the stroke onset by experimented opera- outcome evaluation, conceived to evaluate the
tors with dedicated clot removal devices following benefits obtained by rapid endovascular treatment
radiological study showing the infarct core along for acute ischemic stroke patients, who were
with the ischemic penumbra zone and the occluded selected on the basis of results of CT and CT
brain vessel, eventually. angiography. Recruitment could occur up to 12 h
after the stroke onset. CT and CT angiography
were performed to identify subjects with a small
MR CLEAN Trial infarct core, defined as an ASPECT score of 6–10,
and an occluded large vessel in the anterior circu-
MR CLEAN [4] was a phase III, multicenter clin- lation with moderate or good collateral circula-
ical trial with randomized treatment group assign- tion, defined as the filling of 50 % or more of the
ment, open-label treatment, and blinded end-point middle cerebral artery territory. A total of
evaluation. This study, together with the other two 316 patients underwent randomization and
reported below, compensating for many aspects the divided into two groups. The intervention group
imbalances that were in the previous trials, con- (165) underwent to rapid endovascular treatment
firmed the superiority of the interventional treat- using clot retrievers. Among them, 151 (91.5 %)
ment over the standard IVF. This study was underwent to endovascular treatment, and
428 P. Machi and P. Garofalo

120 (72.7 %) received intravenous rt-PA. Clot patients of the intervention group showed greater
retrievers were used in 130 out of 151 patients early neurologic recovery at 72 h in comparison to
(86.1 %) who underwent to an endovascular pro- the control group. Some essential differences
cedure. 150 patients were assigned to the control between EXTEND IA and the previous trials have
arm and received standard of care. Both groups been mentioned. They include: the use of CT per-
received intravenous rt-PA within 4.5 h after fusion imaging to select patients with the greatest
stroke symptom onset. Successful reperfusion potential to benefit from endovascular therapy,
with TICI score of 2b was observed in 113 out excluding patients with large ischemic cores and
of 156 enrolled patients (72.4 %) in the interven- without evidence of clinically significant recover-
tion group. Results of this trial showed close cor- able ischemic brain; shorter time from stroke onset
relation with results of MR CLEAN trial that to treatment, 30 min compared with 100 min in the
showed benefits and low complication rates with MR CLEAN trial, which may also have contributed
endovascular treatment performed predominantly to the higher proportion of patients with indepen-
with retrievable stents. Nevertheless, there are dent functional outcome observed; and improved
main factors that distinguish ESCAPE from MR rates of angiographic revascularization. Under the
CLEAN and the other prior trials of endovascular light of the results obtained in the trials reported
treatment for stroke: first of all the use of imaging above, it must be undisputed that early administra-
to exclude subjects with a large infarct core and tion of endovascular thrombectomy without delay
poor collateral circulation and then a shorter inter- after the initiation of intravenous rt-PA, among
val from symptom onset to treatment initiation. patients with ischemic stroke with a proximal cere-
bral arterial occlusion and salvageable tissue, had
led to a better outcome in comparison to standard
EXTEND IA Trial care treatment.

The EXTEND IA [6] was a multicenter, prospec-


tive, randomized, open-label, blinded end-point Intra-arterial Recanalization
study designed to evaluate whether patients with Techniques
anterior circulation stroke, with evidence of sal-
vageable tissue on perfusion imaging within 4.5 h Clot (Stent) Retrievers
after the onset of stroke, will have improved reper-
fusion and early neurologic improvement when Clot retrievers are stent-like that are navigated via
treated by endovascular approach, within 6 h after a micro-catheter through the occluded artery and
stroke onset, employing the Solitaire FR device placed within the thrombus. Such devices present
after intravenous administration of rt-PA, compared a double functionality; just after deployment they
with rt-PA alone. This study enrolled 70 eligible act as temporary bypass, restoring the flow
patients who had an ischemic stroke due to occlu- through the thrombus, and in addition, they
sion of the internal carotid artery or of the first or allow clot removal by engaging the thrombus
second segment of the middle cerebral artery eval- during the retrieving.
uated by CT angiography. Thirty five patients Different types of clot retriever tools have been
underwent endovascular therapy while the same launched into the market so far. They can be
number of subjects received standard care. distinguished, depending on which part of the
Endovascular therapy resulted in increased reperfu- thrombus they apply force, in proximal and distal
sion rate at 24 h (P < 0.001), and the rate of intra- clot removers [7–9]. In addition, in some
cranial recanalization after mechanical approach instances other intracranial devices not specific
was nearly 90 % versus 34 % of the control group for clot removal like coronary stents or angio-
without symptomatic intracerebral hemorrhage, as plasty balloons have been “off-label” used with
compared with the control group. Furthermore, this purpose [9, 10].
20 Endovascular Treatment of Stroke 429

In the Multi MERCI trial, a study on MERCI due to the fact that the TIMI score, used to eval-
retriever, the first tool approved by the FDA as a uate the degree of reopening of the target artery,
clot removal tool, a recanalization rate of 69.5 % does not take into consideration the degree of
was reported when mechanical treatment was parenchymal reperfusion. Hence, a high degree
associated with intra-arterial injection of rt-PA of the TIMI score could not be related to good
[9]. Hemorrhagic complication rate of patients clinical recovery. Nowadays, also Penumbra Inc.
who received thrombectomy was 9.8 % (16 out produces a stent-based dedicated clot retriever.
of 131 patients) while perioperative adverse Intracranial self-expanding stents (not dedi-
events rate was 5.5 % (9 out of 131 patients). cated stent retrievers) have been, in the past,
The mean number of passes of the device used as temporary or permanent endovascular
performed per procedure was 2.9. Comparison bypasses for the treatment of ischemic stroke
of results obtained in studies in which the tool [10–13]. In this technique, which could be con-
used for thrombectomy was a dedicated, second- sidered the background for the use of the modern
generation clot retriever (i.e., Solitaire FR) with stent retrievers, a self-expanding stent was placed
results obtained in the Multi MERCI trial shows through the occluding thrombus, displacing it and
higher target vessel recanalization rate recorded allowing for blood flow restoring. This mecha-
with second-generation devices, up to 89.2 % nism facilitates removal of pro-thrombotic fac-
versus 65.5 %. Furthermore, a lower number of tors. Moreover, it allows interactions between
device passes performed per intervention (2.9 fibrinolytic drugs and the clot [14].
vs. 2) along with lower rates of perioperative Since 2010, results of several clinical series
complications has been reported for tools of sec- documenting results of Solitaire FR device used
ond generation. Even if MERCI ® retriever was for the endovascular treatment of acute stroke
the first clot retriever device with FDA approval, patients have been reported in literature
its relatively low efficacy has not encouraged [17]. Results of overall studies are concordant in
worldwide use of such tool. A second tool confirming the efficacy of such device, in terms of
launched into the market in 2007, with the vessel recanalization rate and 3-month clinical
approval of the FDA, was the Penumbra system. outcome. Nowadays, even under the light of the
The first version of this device was a kit made of a results of the trial mentioned above, stent
pump, a micro-catheter, and a mechanical retrievers are considered as the device of reference
thrombectomy tool shaped like a little brush. for mechanical endovascular thrombectomy.
The micro-catheter had to be navigated up to the From a technical point of view, stent-based clot
clot and then the clot had to be fragmented with retrievers are easily advanced to the target vessel
the “brush” tool while clot fragments were aspi- via a micro-catheter and placed within the clot.
rated by a micro-catheter connected to a pump. Hence the stent, fully expanded, is retrieved
Clinical evaluation of this device was done with through the brain vasculature and removed. The
the Penumbra Pivotal Stroke Trial [10]. This was a efficacy of this technique is less operator depen-
multicenter registry on results of 125 subjects dent in comparison with other endovascular pro-
treated with the Penumbra System. 81.6 % of cedure (i.e., the treatment of a cerebral aneurysm).
patients of this cohort had target vessels reopening Furthermore, this technique enables reproducible
with a TIMI score of 2–3. Eighteen perioperative results throughout an entire interventionist team.
adverse events were recorded in 16 patients (12.8
%). At 3 months of follow-up, 25 % of patients of
this series presented a mRS score of  2. The low Procedure
rate of patients with good clinical outcome at the
90-day follow-up seems not related with the Mechanical thrombectomies are performed via a
extremely high incidence of target vessel recana- common femoral artery approach with the patient
lization of this series (81.6 %). This was probably under conscious sedation or general anesthesia.
430 P. Machi and P. Garofalo

A guide catheter is placed in the concerned carotid an ASPECT score of >5 are also good candidates
or vertebral artery. Hence, a micro-catheter is nav- for the combined approach: intravenous fibrinoly-
igated distal to the “occlusion point” through the sis and mechanical recanalization. This dual
clot over a micro-wire, and the stent is then approach, in case of tandem occlusion or terminal
deployed within the thrombus. Subsequently, an “T” carotid occlusion, leads to a higher rate of
angiographic run is carried out in order to verify vessel recanalization in comparison with stand-
its placement. Stent retrievers are usually alone fibrinolysis.
maintained in place before the retrieving for few
(4–5) minutes in order to obtain its best expan-
sion. Thereafter, the stent is gently pulled back to Tandem Occlusions: Mechanical
the guiding catheter. During the retrieving, aspi- Recanalization Techniques
ration through the guiding catheter can be
performed both manually and by using a pump Intravenous fibrinolysis showed a very low rate of
aspirating system. In the cases of vessel tortuosity, target vessel recanalization in cases of carotid
a smaller guiding catheter (a coaxial catheter) can bifurcation (carotid “T”) or extra- and intracranial
be navigated within the first one in order to get tandem occlusions [17, 27]. Consequently, more
more stability and to reach distal intracranial ves- “invasive” approaches, such as intra-arterial fibri-
sel. A coaxial catheter is usually used also in cases nolysis or mechanical thrombectomy, have been
of tandem occlusions; in such instance the first associated with standard intravenous treatment.
guide catheter is placed in the common carotid The efficacy and the safety of these endovascular
artery while the second one, with a tapered profile, approaches to tandem occlusions stroke have been
is navigated through the proximal occlusion. confirmed by several clinical series.
When faced with extra- and intracranial tan-
dem occlusions, operators have to evaluate if the
Selection of Patients for Endovascular first target of the mechanical treatment should be
Treatment the extracranial or the intracranial occlusion.
Authors of the present chapter described a tech-
Patients who could benefit of an endovascular nique in which the intracranial thrombectomy was
treatment has to present an NIHSS >8. Patient performed as first stage, and then the proximal
clinical status on admission, after the treatment, cervical occlusion treatment was tapered
and at discharge must be assessed using the same depending on the type of underlying etiology
scale. The 90-day clinical outcome has to be eval- (Fig. 1). In this setting the proximal cervical
uated by using the modified Rankin scale (mRS). carotid thrombosis is firstly partially treated by
A score of 2 is considered as a good clinical angioplasty and manual aspiration in order to
outcome. Radiological imaging used for patient allow the advancement of the guide catheter, or
selection, MRI or CT scan, has to evaluate the of the coaxial guiding catheter, beyond the prox-
presence of a large vessel occlusion, the extension imal occlusion point. After the positioning of the
of the ischemic core, and the presence of the guide catheter, the intracranial thrombus is
penumbra zone along with the collateral supply removed by clot retrieving. In cases in which the
to the ischemic zone. The ASPECT score, evalu- proximal occlusion is due to a dissection, the
ated with DWI or CT perfusion, is currently used guide catheter is advanced over a micro-catheter
to determine the extension of the ischemic core, in through the true lumen of the dissection. Thereaf-
cases of MCA stroke. A patient presenting with an ter, the guide catheter is navigated above the dis-
ASPECT score of >5 is a good candidate for sected segment and the intracranial clot is
mechanical revascularization. Furthermore, removed as previously described. The theoretical
patients with tandem occlusions, involving an purpose of this approach is the immediate revas-
intracranial occlusion associated with a cervical cularization of the intracranial vessel in order to
common carotid occlusion, when presenting with obtain immediate reperfusion of the ischemic
20 Endovascular Treatment of Stroke 431

Fig. 1 (a–f) DWI shows the extension of the ischemic core

brain zone. Thereafter, the treatment of the extra- Rationale for Endovascular Treatment
cranial occlusion and the underlying stenosis is of Tandem Occlusions
performed, eventually. The stenting of the cervical
carotid is performed only in cases of tight stenosis Tandem occlusions have been previously treated
or unstable ulcerated plaque. For cases in which by stand-alone intravenous fibrinolysis. Neverthe-
stand-alone angioplasty is sufficient to restore less, efficacy of such approach in comparison with
normal flow within the artery, the treatment of cases of isolated M1 occlusions seems to be
the cervical stenosis, endovascular or surgical, is reduced [18, 19]. Tandem occlusion is an inde-
scheduled after the acute phase. Such “staged” pendent risk factor for poor clinical outcome for
procedure reduces the need for antiplatelet medi- patients treated exclusively with intravenous fibri-
cations and the risk of bleeding of the ischemic nolysis [20]. Furthermore, intravenous fibrinoly-
core in the immediate post-acute phase. In cases of sis presents reduced efficacy also in cases of
acute carotid stenting, patients are given intrave- carotid bifurcation “T” occlusions [21]. In cases
nous aspirin (250–500 mg) intraoperatively. In of tandem occlusion, prompt intracranial recana-
cases of carotid dissection, the stenting of the lization is related to improved clinical outcome
dissected segment is performed when collateral regardless of persistence of the proximal cervical
supply to the dissected artery, via the circle of occlusion [22].
Willis, is not present or adequate (Figs. 2, 3, 4, Ozdemir et al. [23] published results of a study
5, and 6). involving eight patients presenting with tandem
432 P. Machi and P. Garofalo

Fig. 2 (a, b) MR
angiography shows the
presence of a proximal
carotid occlusion associated
with M1 intracranial
occlusion (tandem
occlusion)

occlusion strokes. In this series the intracranial treatment seems to be more effective than the
clot was treated by local injection of rt-PA standard intravenous treatment. They did not
released through a micro-catheter navigated via specify whether this approach would be effective
the communicating artery system to the intracra- also in cases of tandem occlusions due to cervical
nial thrombus. The proximal cervical occlusion of carotid atherosclerotic plaques.
patients of this series was not treated and the Malik et al. [25] reported a series of 27 patients
vessel remained occluded. This technique led to presenting with tandem occlusions treated by
intracranial recanalization only in two out of eight intra-arterial administration of rt-PA performed
patients, in spite of low rate of cerebral revascu- prior to stenting of the cervical carotid. In this
larization; all patients with a functioning commu- series clot removal/disruption was performed
nicating artery system had favorable outcome at with different techniques and tools: Merci clot
3 months. Nevertheless, the persistence of the retrieval device, Penumbra system, or by angio-
cervical thrombotic occlusion could be a source plasty and/or stent stenting. Patients of this series
of potential new emboli; this is an independent were treated under systemic heparinization. More-
risk factor for early re-occlusion after rt-PA over, patients received a bolus of intravenous
[21]. Lavalée et al. [24] reported on a series of eptifibatide upon stent deployment and, after the
ten patients presenting with tandem occlusion procedure, double antiplatelet regimen with
strokes due to cervical carotid dissection. clopidogrel and aspirin. Complete proximal and
Among them, four out of ten patients received distal recanalization was obtained in 75.3 % of
intravenous rt-PA, while six patients underwent patients; hemorrhagic complications were
mechanical clot fragmentation. In all patients recorded in 10.4 % of the subjects, while the
treated by mechanical approach, a complete mortality rate of this series was 24.6 %. A favor-
recanalization of the involved vessel was able clinical outcome was recorded in 41.6 % of
achieved. Authors stated that endovascular patients. Authors stated that the reopening of the
20 Endovascular Treatment of Stroke 433

Fig. 3 (a, b) DSA shows the occlusion of the cervical was performed with a coaxial catheter navigated through
carotid artery due to an atherosclerotic plaque. (c, d) DSA the proximal stenosis
shows the intracranial M1 occlusion; the angiographic run

cervical carotid allows immediate intracranial jails the thrombus, reducing the risk of distal
revascularization and a better positioning of the embolization. By the way the authors state that
guiding catheter. At the same time, the stent this approach could require prolonged time for
deployed at the level of the proximal stenosis stent placement and that the dual antiplatelet
434 P. Machi and P. Garofalo

Fig. 4 (a, b) DSA run performed with a micro-catheter DSA run performed after retrieving of the stent retriever:
navigated within the M1 occlusion. (c, d) DSA run complete reopening of M1, presence of an embolic occlu-
performed after deployment of the stent retriever. (e, f) sion of the anterior cerebral artery

therapy could result in hemorrhagic transforma- atherosclerotic plaque. In the other two cases the
tion especially in patients with a large stroke proximal occlusion was due to a dissection and to
extension or ineffective intracranial clot removal. the consequences of an endoartherectomy. Six
The authors suggest that a tapered stand-alone patients presented a concomitant M1 occlusion.
cervical stenosis angioplasty could reduce the Complete recanalization was obtained in all cases
risk of hemorrhagic complications. of M1 occlusion by “in situ” rt-PA injection
Srinivasan et al. [26] reported a series of seven performed through a micro-catheter navigated
patients presenting with tandem occlusion within the cervical occlusion. Management of
strokes. In five out of seven cases, the proximal the proximal occlusion varied depending on the
occlusion was due to thrombosis developed on an type of underlying cause. Patients with cervical
20 Endovascular Treatment of Stroke 435

Fig. 5 (a, b) Stand-alone angioplasty of the proximal cervical carotid stenosis. (c, d) Final DSA control shows reopening
of the right anterior cerebral artery
436 P. Machi and P. Garofalo

Fig. 6 (a–d) Stenting of the cervical stenosis performed 15 days after the acute phase initial treatment

atherosclerotic plaque received angioplasty and first target to be treated should be the intracranial
stenting. In the case of cervical dissection, the occlusion because it is the cause of the neurolog-
artery was treated by deploying two covered ical symptoms. They agree with such aptitude
stents along the dissected segment. Four out of because in their opinion, the first purpose of the
six patients of this cohort showed a good clinical endovascular approach to tandem occlusions
outcome at 1-month follow-up examination. The should be the prompt and complete recanalization
authors claim that in tandem occlusion stroke, the of the intracranial vessel [27–30]. The first
20 Endovascular Treatment of Stroke 437

approach to the cervical carotid is performed in References


order to navigate the guiding catheter above the
proximal occlusion. Thereafter, the intracranial 1. Ciccone A, Valvassori L, Nichelatti M et al (2013)
Endovascular treatment for acute ischemic stroke. N
occlusion is treated by using clot retriever device.
Engl J Med 368:904–913
Finally, the carotid cervical occlusion/stenosis is 2. Broderick J, Palesch Y, Demchuk A et al (2013)
treated by medical management, stand-alone Endovascular therapy after intravenous t-PA versus
angioplasty, or angioplasty and stenting. Acute t-PA alone for stroke. N Engl J Med 368:893–903
3. Kidwell C, Jahan R, Gornbein J et al (2013) A trial of
stenting is performed in cases of dissection, with-
imaging selection and endovascular treatment for
out collateral supply, or unstable, occlusive ath- ischemic stroke. N Engl J Med 368:914–923
erosclerotic plaque. In such instance, patient is 4. Berkhemer OA, Fransen PSS, Beumer D (2015) A
given intravenous aspirin (250–500 mg) just randomized trial of intraarterial treatment for acute
ischemic stroke (MR CLEAN). N Engl J Med
upon stent placement. Stand-alone angioplasty
372:11–20
seems to be the best approach in cases of 5. Goyal M, Demchuk AM, Menon M (2015) Random-
nonocclusive thrombus. The stenting of the cervi- ized assessment of rapid endovascular treatment of
cal carotid or the surgical treatment is in this case ischemic stroke. N Engl J Med
6. Campbell BCV, Mitchell TJ, Kleining HM (2015)
performed a few days (10–15) after the acute
Endovascular therapy for ischemic stroke with perfu-
phase. This staged approach may reduce the risk sion imaging selection. N Engl J Med
of hemorrhagic transformation and leads to a bet- 7. Higashida RT, Furlan AJ, Roberts H et al (2003) Trial
ter antiplatelet preparation of the patient. design and reporting standards for intra-arterial cere-
bral thrombolysis for acute ischemic stroke. Stroke 34:
e109–e137
8. The IMS II Trial Investigators (2007) The Interven-
Summary tional Management of Stroke (IMS) II study. Stroke
38:2127–2135
In patients presenting with acute ischemic stroke 9. Smith WS, Sung G, Saver J et al (2008) Mechanical
due to a large vessel occlusion, an early recanaliza- thrombectomy for acute ischemic stroke: final results
of the Multi MERCI trial. Stroke 39:1205–1212
tion of the occluded artery is related to better clin-
10. Penumbra PivotalStroke Trial Investigators (2009) The
ical outcome. From the beginning of 2015 several Penumbra Pivotal Stroke trial: safety and effectiveness
trials demonstrated improvements, in terms of ana- of a new generation of mechanical devices for clot
tomical and clinical results, when stroke patients removal in intracranial large vessel occlusive disease.
Stroke 40:2761–2768
were treated by standard intravenous therapy asso-
11. Mazighi M, Serfaty JM, Labreuche J et al (2009) Com-
ciated with intra-arterial mechanical therapy. parison of intravenous alteplase with a combined intra-
Before then, recommended therapy for acute ische- venous –endovascular approach in patients with stroke
mic stroke was intravenous administration of rt-PA and confirmed arterial occlusion (RECANALISE
study): a prospective cohort study. Lancet Neurol
within 4.5 h from symptom onset. Results of the
8:802–809
early randomized, controlled trials appeared in lit- 12. Gralla J, Schroth G, Remonda L et al (2006) Mechan-
erature since 2013, comparing mechanical therapy ical thrombectomy for acute ischemic stroke:
associated with intravenous rt-PA (standard care) thrombus-device interaction, efficiency, and complica-
tions in vivo. Stroke 37:3019–3024
versus stand-alone intravenous rt-PA, failed to
13. Hussain MS, Kelly ME, Moskowitz SI et al (2009)
demonstrate the superiority of the mechanical Mechanical thrombectomy for acute stroke with the
approach over standard care. alligator retrieval device. Stroke 40:3784–3788
In the present chapter they firstly report results 14. Levy EI, Ecker RD, Horowitz MB et al (2006) Stent-
assisted intracranial recanalization for acute stroke:
of randomized trials as unfavorable to mechanical
early results. Neurosurgery 58:458–463
approach and discuss their limitation and biases. 15. Kelly ME, Furlan AJ, Fiorella D et al (2008) Recana-
Then they report about recent trials appeared in lization of an acute middle cerebral artery occlusion
2015 demonstrating the superiority of this using a self-expanding, reconstrainable, intracranial
microstent as a temporary endovascular bypass. Stroke
approach. Moreover, they describe clot retrieval
39:1770–1773
techniques and the endovascular treatment of tan- 16. Machi P, Costalat V, Lobotesis K et al (2012) Solitaire
dem occlusions. FR thrombectomysystem. immediateresults in
438 P. Machi and P. Garofalo

56 consecutive acute ischemic stroke patients. J tandem cervical carotid artery/ middle cerebral artery
Neurointerv Surg 4:62–66 occlusion. AJNR Am J Neuroradiol 29:1596–1600
17. Costalat V, Machi P, Lobotesis K et al (2011) Rescue, 24. Lavallée PC, Mickaël M, Saint-Maurice JP et al (2007)
combined, and stand-alone thrombectomy in the man- Stent-assisted endovascular thrombolysis versus intra-
agement of large vessel occlusion stroke using the venous thrombolysis in internal carotid artery dissec-
Solitaire device: a prospective 50-patient single-center tion with tandem cerebral artery occlusion. Stroke
study: timing, safety, and efficacy. Stroke 38:2270–2274
42:1929–1935 25. Malik AM, Nirav AV, Ridwan L et al (2011)
18. Kim K, Garami Z, Mikulik R et al (2005) Early recan- Endovascular treatment of extracranial/intracranial
alization rates and clinical outcomes in patients with anterior circulation occlusion. Stroke 42:1653–1657
tandem internal carotid artery/middle cerebral artery 26. Srinivasan A, Goyal M, Stys P, Sharma M, Lum C
occlusion and isolated middle cerebral artery occlu- (2006) Microcatheter navigation and thrombolysis in
sion. Stroke 36:869–871 acute symptomatic cervical internal carotid occlusion.
19. Linfante I, Llinas RH, Selim M et al (2002) Clinical AJNR Am J Neuroradiol 27:774–779
and vascular outcome in internal carotid artery versus 27. Rubiera M, Ribo M, Delgado-Mederos R et al (2006)
middle cerebral artery occlusions after intravenous tis- Tandem internal carotid artery/middle cerebral
sue plasminogen activator. Stroke 33:2066–2071 artery occlusion: an independent predictor of poor out-
20. The National Institute of Neurological Disorders and come after systemic thrombolysis. Stroke
Stroke rtPA Stroke Study Group (1995) Tissue plas- 37:2301–2305
minogen activator for acute ischemic stroke. N Engl J 28. Zaidat OO, Suarez JI, Sunshine JL et al (2005) Throm-
Med 333:1581–1587 bolytic therapy of acute ischemic stroke: correlation of
21. Christou I, Feldeberg RA, Demchuk AM et al (2002) angiographic recanalization with clinical outcome.
Intravenous tissue plasminogen activator and improve- AJNR Am J Neuroradiol 26:880–884
ment in acute ischemic stroke patients with internal 29. Kimura K, Iguchi Y, Shibazaki K et al (2008) Large
carotid artery occlusion. J Neuroimaging 12:119–123 ischemic lesions on diffusion-weighted imaging done
22. Mourand I, Brunel H, Vendrell JF et al (2010) before intravenous tissue plasminogen activator throm-
Endovascular stent-assisted thrombolysis in acute bolysis predicts a poor outcome in patients with acute
occlusive carotid artery dissection. Neuroradiology stroke. Stroke 39:2388–2391
52:135–140 30. Malgorzata M, Michels P, Jensen AM et al (2008)
23. Ozdemir O, Bussiere M, Leung A et al (2008) Intra- Endovascular treatment in proximal and intracranial
arterial thrombolysis of occluded middle cerebral carotid occlusion 9 hours after symptom onset. Neuro-
artery by use of collateral pathways in patients with radiology 50:599–604
Sickle Cell Disease and Stroke
21
Akifumi Fujita, Chie Asai, Yu-Ming Chang, Nadja Kadom,
Martin H. Steinberg, Naoko Saito, and Osamu Sakai

Contents Blood Transfusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 444


Hematopoietic Cell Transplantation . . . . . . . . . . . . . . . . . 444
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 440
Etiology, Classification, and Pathophysiology . . . . . . 441 Neurovascular Complications Concept and
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 442 Imaging Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 444
Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 444
Clinical Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 442 Moyamoya Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449
Painful Episodes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 442 Extracranial Vasculopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . 451
Sickle Cell Acute Events . . . . . . . . . . . . . . . . . . . . . . . . . . . . 442
Infarcts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 443 Posterior Reversible Encephalopathy
Cognitive Deficits . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 443 Syndrome (PRES) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 454
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 443 Other Sickle Cell Disease Imaging
Hydroxyurea (HU) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 443 Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 454
Craniofacial Bone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 454
Temporal Bone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456

A. Fujita • C. Asai • Y.-M. Chang • N. Kadom Advanced Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 457


Departments of Radiology, Boston University, Boston Diffusion Tensor Imaging (DTI) . . . . . . . . . . . . . . . . . . . . . 457
Medical Center, Boston, MA, USA Perfusion Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
e-mail: akifuji@jichi.ac.jp; chie_asai1116@yahoo.co.jp; MR Spectroscopy (MRS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
ychangneuro@gmail.com; nadja.kadom@bmc.org Quantitative MRI (qMRI) . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
Nuclear Medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
M.H. Steinberg
Departments of Medicine, Pediatrics, and Pathology and Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
Laboratory Medicine, Boston University, Boston Medical Prevention of Primary Stroke . . . . . . . . . . . . . . . . . . . . . . . . 461
Center, Boston, MA, USA Prevention of Secondary Stroke . . . . . . . . . . . . . . . . . . . . . 462
e-mail: mhsteinb@bu.edu
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
N. Saito
Department of Diagnostic Radiology, Saitama Medical References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
University International Medical Center, Saitama, Japan
e-mail: naoko.saito.ns@gmail.com
O. Sakai (*)
Departments of Radiology, Boston University, Boston
Medical Center, Boston, MA, USA
Department of Otolaryngology – Head and Neck Surgery,
Boston University, Boston Medical Center, Boston, MA,
USA
Department of Radiation Oncology, Boston University,
Boston Medical Center, Boston, MA, USA
e-mail: osamu.sakai@bmc.org

# Springer Science+Business Media New York 2016 439


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_11
440 A. Fujita et al.

• Vasculopathy • Vaso-occlusion • Hemolytic


Abstract
anemia • Stroke • Ischemic stroke • Hemor-
Sickle cell disease (SCD) is a hereditary disor-
rhagic stroke • Silent cerebral infarct (SCI) •
der that is so named for the presence of char-
Cerebral fat embolism • Moyamoya syndrome
acteristic malformed sickle-shaped red blood
• Posterior reversible encephalopathy syn-
cells (RBCs). These abnormal RBCs are short-
drome (PRES) • Bone infarction • Osteomye-
lived and can also cause vaso-occlusion
litis • Labyrinthine hemorrhage (LH) •
throughout the body, which may lead to ische-
Labyrinthitis ossificans (LO) • Arterial spin
mia and infarcts in multiple organ systems.
labeling (ASL) • Positron emission tomogra-
Neurovascular complications of SCD include
phy (PET) • Single-photon emission computed
cerebral vasculopathy, such as stroke and
tomography (SPECT) • Transcranial Doppler
neurocognitive deficits, which are significant
(TCD)
causes of morbidity and mortality in both chil-
dren and adults. In current clinical practice,
various conventional imaging tools, such as
Introduction
computed tomography (CT), magnetic reso-
nance imaging (MRI), and catheter angiogram,
Sickle cell disease (SCD) is an inherited hemato-
still play an important role in diagnosis and
logic disorder characterized by the formation of
follow-up of neurovascular complications of
misshapen red blood cells (RBCs) when sickle
SCD. Ultrasounds, including transcranial
hemoglobin becomes deoxygenated and polymer-
Doppler (TCD) ultrasound, are useful in
izes. SCD causes a broad spectrum of clinical
screening for intra- and extracranial
manifestations in various organ systems and is
vasculopathy. Advanced imaging techniques,
associated with significant morbidity and mortal-
such as diffusion tensor imaging (DTI), perfu-
ity in children and adults. Imaging plays a crucial
sion imaging, MR spectroscopy, quantitative
role in the detection and description of the extent
MRI (qMRI), and nuclear medicine such as
of involvement of SCD in the brain, head and
single-photon emission computed tomography
neck, and spine.
(SPECT) and positron emission tomography
Neurovascular complications, such as cerebro-
(PET), are emerging as diagnostic tools in the
vascular ischemia/infarcts, hemorrhage, various
evaluation of SCD.
intra- and extracranial vasculopathy, and cogni-
In this chapter, a brief overview of the eti-
tive impairment, are among the most significant
ology, classification, pathophysiology, epide-
complications in SCD. SCD also affects the osse-
miology, clinical manifestation, and treatment
ous structures of the head and neck, including the
of SCD is provided. The complications of SCD
inner ears. Neuroimaging plays an important role
seen across the spectrum of conventional to
in diagnosing neurological complications of SCD
advanced imaging modalities are also illus-
as well as in monitoring the effects of medical and
trated and discussed. The focus is on
surgical treatments. Currently, neuroimaging of
neurovascular complications and its manage-
SCD largely relies on various conventional imag-
ment, but there will also be a discussion of
ing modalities such as computed tomography
posterior reversible encephalopathy syndrome,
(CT), magnetic resonance imaging (MRI), Dopp-
craniofacial osseous complications, and inner
ler ultrasound, and catheter angiogram. Recently,
ear complications, which may be encountered
quantitative MRI (qMRI) and functional MRI
in the daily neuroradiology practice.
techniques, such as diffusion tensor imaging
(DTI), perfusion imaging, and MR spectroscopy,
Keywords have been introduced to assess and monitor
Sickle cell disease (SCD) • Quantitative MRI patients with SCD. In this chapter, the pathophys-
(qMRI) • Diffusion tensor imaging (DTI) • iology, epidemiology, clinical features, and char-
Perfusion imaging • MR spectroscopy (MRS) acteristic neuroimaging manifestations of SCD
21 Sickle Cell Disease and Stroke 441

with conventional as well as advanced imaging shape after oxygen tension is restored. These sick-
techniques are reviewed. Current therapeutic led cells and other RBCs can be sequestered in the
approaches and management of SCD and the spleen and other reticuloendothelial cell-
potential role of imaging in management will containing organs and also lyse in the circulation,
also be discussed. leading to anemia.
Patients who carry one abnormal HBB allele
and a normal β-globin gene are said to have sickle
Etiology, Classification, cell trait (HbAS) and have 60 % HbA and 40 %
and Pathophysiology HbS. HbAS is considered a benign entity with
only rare complications. In carriers of HbAS,
The term SCD refers to a variety of β-globin because each RBC contains 40 % HbS, polymer-
(HBB) genotypes that all result in abnormally ization of HbS occurs only under special condi-
shaped RBCs. The mutation underlying SCD is tions, like in the hypoxic, acidotic, and
an A-to-T transversion in the codon for amino hyperosmolar conditions of the renal medulla
acid position 6 in the β-globin gene. Because of and in the spleen of some people who are hypoxic.
this mutation, a valine residue replaces the normal One prospective study in patients with HbAS
glutamic acid residue (glu6val) and HbS β-globin showed that even otherwise asymptomatic
chains are substituted for normal β-globin chains patients had a high percentage of vasculopathy,
[1, 2]. Each hemoglobin molecule contains two such as arterial tortuosity, but the pathophysio-
β-globin and two α-globin subunits. Hemoglobin logic consequences remain unclear [3]. Patients
with two normal β-globin chains is referred to as with HbAS may develop hyposthenuria and occa-
hemoglobin A (HbA) and is the major hemoglo- sional hematuria and have a higher incidence of
bin of adults. Fetal hemoglobin (HbF), a molecule thromboembolic disease.
of two γ-globin and two α-globin chains, is most The two major pathophysiological processes in
prevalent in the fetus and newborns up to about SCD are vaso-occlusion and hemolytic anemia
three months of age. Other variant hemoglobin that is both intra- and extravascular [1, 2, 4].
that can be present as compound heterozygotes 3Vaso-occlusion can occur at both the micro-
with HbS are hemoglobin C (HbC), hemoglobin E and macrovascular levels. At the microvascular
(HbE), and hemoglobin D (HbD) [1, 2]. SCD is a level, the abnormal adherence of sickled RBCs
recessive trait, which means that patients must to the capillary and venule endothelium can lead
carry two abnormal alleles of HBB in order for to intravascular stasis of blood flow. Stasis attracts
the disease to fully manifest. Homozygosity for leukocytes and fosters platelet adherence and
the HbS gene or sickle cell anemia is the most degranulation, as well as fibrin deposition. This
common genotype of SCD [1, 2]. HbSC disease is microvascular occlusion can lead to tissue ische-
the second most common genotype. SCD can also mia and infarct. Inflammation can enhance the
be caused by compound heterozygosity for HbS expression of adhesion molecules, further increas-
and one of the many forms of β-thalassemia. ing the tendency of sickled erythrocytes to adhere
Deoxygenation causes HbS molecules to poly- to the vascular endothelium, worsening vaso-
merize, causing RBCs to assume various abnor- occlusion [1, 2, 4]. At the macrovascular level,
mal shapes, some of which resemble a sickle. The abnormal adhesion of sickled RBCs and white
change in shape causes RBC membrane damage blood cells (WBCs) to vessel sites of high flow
and is associated with a decrease in RBC elastic- turbulence may damage the vascular wall,
ity, rendering them unable to pass through small resulting in intimal hyperplasia of large vessels,
capillaries. Hemolytic anemia or a shortened stenosis, and progressive occlusion of the large
lifespan of the RBC is also a feature of this cellular cerebral arteries [5]. Chronic hemolytic anemia
damage [1, 2]. Sickled cells can become irrevers- from the accelerated breakdown of sickled RBCs
ibly fixed in the “sickle” shape because of perma- may also result in macrovascular occlusion from
nent membrane damage and fail to resume normal intimal hyperplasia secondary to an inflammatory
442 A. Fujita et al.

cascade triggered by increased free hemoglobin older ages and SCD is considered a chronic con-
levels [1, 2, 4]. In addition, sickled RBCs may dition requiring comprehensive, lifelong manage-
adhere to damaged vessel walls, which can result ment [4]. However, in low-resource countries,
in secondary thrombosis and distal branch emboli more than 50 % of children younger than five
[5–7]. In the past, small vessel occlusion by intra- years of age die due to complication of SCD [4].
vascular sickling and slugging of RBCs was con-
sidered to be the primary cause of strokes in SCD.
It is now understood, however, that occlusion of Clinical Manifestations
small vessels accounts for about 25 % of cerebral
infarct in SCD while macrovascular complica- Painful Episodes
tions account for 75 % [8].
Pain, both acute and chronic, is a hallmark of
SCD. Acute painful episodes are the most com-
Epidemiology mon vaso-occlusive events with inflammatory
and ischemic consequences in patients with SCD
SCD is most common in people from sub-Saharan of all ages, although they are more common in
Africa, South and Central America, the Caribbean teenagers and young adults [1, 2, 4]. In young
islands, the Mediterranean countries, India, Saudi children, one of the first manifestations of SCD
Arabia, and their descendants worldwide [1, 2]. It is the so-called hand-foot syndrome (i.e., sickle
is now estimated that 200,000 children affected cell dactylitis), a painful swelling of the hands and
with SCD are born every year, approximately feet due to inflammation of the metacarpal and
2,000 children in the USA. The estimated US metatarsal periosteum. Painful episodes may
prevalence ranges from 70,000 to 140,000 involve any bones, muscles, mesentery, and
[4]. About 8 % of African Americans have other organs. Although acute vaso-occlusive
HbAS. In some regions of Africa, more than a pain is generally self-limiting and does not
quarter of the populations are carriers; very high result in permanent organ damage, increased fre-
gene frequencies are also found in some parts of quency of pain is associated with early death in
Saudi Arabia and Greece, in tribal populations of patients with SCD who are older than 20 years
India, and in Brazil [1, 2, 4]. HbAS confers some [12]. The cause of chronic pain is poorly under-
protection from Plasmodium falciparum malaria; stood, but it can be debilitating and may result
the underlying protective mechanism is still sub- from leg ulcers, avascular necrosis of bone, and
ject of scientific investigation [9]. neuropathic complications [4]. An important con-
Mortality in patients with SCD has signifi- sideration for the neuroradiologist is that the skull
cantly improved in high-resource countries. The and maxillofacial bones may be affected by acute
universal newborn screening for hemoglobinopa- pain. Typical symptoms are headaches or local-
thies has become standard practice in such coun- ized pain and swelling. Frequently, these are
tries, enabling early diagnosis and patient caused by bone infarcts with subperiosteal
management [1, 4]. In 2000, the universal admin- hemorrhage [13].
istration of the pneumococcal vaccine has further
decreased the mortality and morbidity of child-
hood invasive pneumococcal disease, including in Sickle Cell Acute Events
children with SCD. As a result of these programs,
SCD-related death fell by 42 % from 1999 Besides the painful episodes described above,
through 2002 [10]. It is estimated that 94 % of patients with SCD may also experience splenic
children with SCD now live to adulthood, and and hepatic sequestration crisis, aplastic crisis,
more than 98 % of children with SCD in high- acute chest syndrome (ACS), and stroke. In addi-
resource countries are living into adulthood tion, SCD patients are more susceptible to infec-
[11]. This has caused a mortality shift toward tion and its complications, such as sepsis,
21 Sickle Cell Disease and Stroke 443

pulmonary embolism, pulmonary hypertension, without corresponding neurological deficits last-


cardiomyopathy, and hepatic disease [1, 2, 4]. ing longer than 24 h. SCIs are clinically signifi-
Infection with B19 parvovirus can suppress the cant because they indicate a higher likelihood of
production of RBCs in patients with SCD for 2 to subsequent overt stroke [18]. Although SCD is
3 days, causing an acute, life-threatening aplastic recognized as a hypercoagulable state, the inci-
anemia with pallor, tachycardia, and fatigue. dence of venous infarction due to thrombosis is
Severe anemia can also result acutely from unknown [19].
sequestration of blood in the spleen or liver
[2]. ACS is the second most common cause of
hospitalization among patients with SCD and a Cognitive Deficits
leading cause of mortality [14]. ACS is an acute
lung injury that causes development of a new As more SCD patients survive to adulthood, the
alveolar pulmonary infiltrate involving at least neurocognitive dysfunction caused by SCD is
one lung segment. ACS is caused by a combina- becoming an emerging problem in young adults
tion of pulmonary infection, fat embolism, and [20]. Studies correlating MRI evidence of brain
intravascular pulmonary sequestration of sickled damage have come to controversial conclusions,
erythrocytes, resulting in lung injury and infarct. with some stating that neurocognitive deficits in
Recurrent ACS can lead to chronic respiratory SCD patients are associated with focal brain
insufficiency. Occasionally, ACS culminates injury and silent strokes in children while others
with multi-organ failure [2]. state that these deficits are associated more with
anemia and age. A recent study suggests that
volume reduction of the basal ganglia and thala-
Infarcts mus may correlate with cognitive deficits in adult
with SCD [21]. These reports suggest that the
Infarcts are the consequences of vaso-occlusive cause of the neurocognitive impairment is multi-
crises. Infarcts may involve the brain, liver, factorial and that further correlating studies using
spleen, kidney, and bones. Infections often pre- functional imaging for cerebral blood flow and
cede vaso-occlusive episodes in children, hypoxia may be needed [20].
suggesting that fever, dehydration, and acidosis
may be contributing factors [6]. Bone infarction
can occur anywhere in the skeleton. It results
directly from the sickling of RBCs in the bone Treatment
marrow, which causes stasis of blood and seques-
tration of cells [13, 15]. Hydroxyurea (HU)
Stroke is one of the most severe complications
of SCD because it may result in permanent neu- Hydroxyurea (HU) is a drug that promotes the
rological deficits or death. The incidence of stroke production of HbF, the normal hemoglobin
varies depending on the SCD genotype. The prev- which is more prevalent in newborns. HU is cur-
alence of cerebral vascular accident (CVA) was rently the only established preventive pharmaco-
11 % in patients under 20 years of age with homo- logic treatment for both children and adults with
zygous HBSS [16]. Ischemic strokes are more SCD. The presence of HbF inhibits HbS polymer-
common in the children, adolescent, and old ization and higher levels of HbF are associated
adult SCD patients, while the incidence of hem- with reduced mortality in SCD patients [2]. In a
orrhagic strokes peaks between the ages of 20–29 randomized controlled trial, HU was shown to
years [16]. Silent cerebral infarcts (SCIs) occur decrease the frequency of painful episodes and
more commonly than overt strokes, with a preva- acute chest syndrome and the need for blood
lence of 21.8 % in children with HbSS ages 6–19 transfusion and hospital admissions in SCD
years [17]. SCIs are defined as a lesion on MRI, patients [22].
444 A. Fujita et al.

Blood Transfusion hemorrhagic, or transient ischemic attack


[16]. The special forms of ischemic stroke in
Chronic blood transfusions have been demon- SCD patients are the so-called silent cerebral
strated to reduce the risk of both primary and infarcts (SCIs) and stroke caused by cerebral fat
secondary strokes and SCIs [23] and prevent embolism [27].
repeated ACS [2, 4]. There is currently no specific Imaging approaches for stroke in SCD patients
treatment for acute stroke in SCD. However, are the same as for non-SCD patients. The pur-
blood transfusion within 48 hours after acute pose of acute stroke imaging in SCD is to deter-
stroke may decrease the incidence of subsequent mine the extent of brain damage, to differentiate
strokes [24]. The aim of transfusion therapy is to ischemic stroke from hemorrhagic stroke, and to
reduce HbS to below 30 %, which effectively help identify and delineate underlying
prevents overt stroke and SCIs [25]. However, it vasculopathy.
can result in morbidity related to iron overload CT of the brain without contrast is usually the
and iron deposition in multiple organs, including first imaging modality used in the acute setting,
the liver, heart, and endocrine systems, and often which allows for quick assessment of hemor-
necessitates iron chelation therapy [2, 4]. rhagic versus ischemic stroke, for detection of
other causes to explain new neurological deficits,
and for evaluation of the extent of damage and
Hematopoietic Cell Transplantation complications. Other benefits of CT include its
easier accessibility and 24/7 availability at most
Hematopoietic cell transplantation is the only institutions, as well as short exam times [28]. Dis-
curative treatment for SCD thus far. However, advantages of CT include ionizing radiation expo-
there is a lack of suitable allogeneic marrow sure, low sensitivity for detection of small
donors (usually HLA-matched siblings) for infarcts, and low sensitivity for stroke detection
about 90 % of eligible patients [2]. There is a in the first several hours [29].
5–10 % mortality rate associated with this treat- MRI with diffusion-weighted imaging (DWI)
ment, mostly due to graft failure, lack of response is very useful in detecting acute infarcts as well as
to first-line immunosuppressive therapy, graft- determining the exact location and extent of ische-
versus-host disease (GVHD), relapse after the mic lesions [30, 31]. Apparent diffusion coeffi-
initial successful treatment, and secondary clonal cient (ADC) maps in conjunction with DWI help
and malignant disease [26]. There are especially to pinpoint regions of early ischemia and infarct
poor outcomes in patients over the age of 16 years [32]. T2-weighted (T2-WI) imaging and fluid-
who are subjected to myeloablative conditioning attenuated inversion recovery (FLAIR) imaging
regimens, making the use of non-myeloablative are used for detecting T2 high signal intensity
regimens standard in older patients [2]. lesions [33], such as acute stroke and silent
infarcts. Gradient echo T2*-weighted (T2*WI)
images and susceptibility-weighted images
Neurovascular Complications Concept (SWI) are used to detect hemorrhagic lesions
and Imaging Features [34]. While MRI offers the benefits of increased
sensitivity for pathology compared to CT and
Stroke avoidance of radiation exposure, it requires con-
siderable patient cooperation and in many pediat-
Stroke or CVA is defined as a sudden-onset neu- ric patients, may require sedation. The other
rological deficit that persists for more than disadvantages of MRI are limited access for
24 hours. SCD is one of the most common causes acutely ill patients at many institutions and rela-
of stroke in children and young adults. The Coop- tively long duration of imaging studies.
erative Study of Sickle Cell Disease (CSSCD) Vascular imaging studies in the head and neck
classified stroke in SCD patients as ischemic, may be considered in all patients with SCD,
21 Sickle Cell Disease and Stroke 445

Fig. 1 A 38-year-old woman with SCD, presenting with map (c) shows multiple foci of restricted diffusion in the
altered mental status. (a) Axial FLAIR image shows mul- left periventricular white matter consistent with acute
tiple high signal intensity lesions of the bilateral infarcts (arrows). Right periventricular lesions suggest
periventricular white matter (arrows). (b, c) chronic ischemia or prior infarcts
Corresponding axial DWI (b = 1,000) (b) with ADC

irrespective of whether they had previous epi- high intensity on T2-WI images. With FLAIR
sodes of stroke [30, 35]. CTA and MRA can be imaging, abnormal T2 high signal may be more
used to detect intracranial arterial occlusion or easily detected than conventional T2-WI imaging,
stenosis and aneurysms. CTA exposes patients to particularly in the periventricular and subcortical
high levels of ionizing radiations and contrast regions (Figs.1, 2, 3, 4, and 5).
media; therefore, MRA is preferred in children Arterial stenosis or occlusion is often seen in
[30]. MR venography can be useful when venous conjunction with infarcts of the cerebral gray mat-
thrombosis is suspected. Conventional catheter ter (the cortex, basal ganglia, and thalamus)
angiogram is an invasive procedure but yields (Fig. 2). However, white matter infarcts can be
higher anatomical detail of vascular anatomy and seen in the setting of arterial stenosis or occlusion
pathology than other imaging modalities, includ- as well, particularly in border (watershed) zones
ing information on hemodynamic function. Cath- [8] (Fig. 3). The anterior circulation is more often
eter angiogram is usually indicated prior to involved than the posterior circulation. The radi-
surgery or endovascular interventions [31]. A ologist should screen for loss of vascular flow
careful risk–benefit evaluation should be voids on T2-WI images, which may indicate
performed prior to subjecting an SCD patient to vasculopathy (Fig. 2e). FLAIR imaging can
catheter angiogram. If anesthesia is necessary, show high signal in cortical vessels in the setting
special care should be taken in the perioperative of altered flow velocities, such as slow distal flow
care of patients with SCD [36]. of a stenotic arterial branch or leptomeningeal
collaterals [37] (Fig. 6). In these cases, the radiol-
Ischemic Stroke ogist may decide to add brain MRA or recom-
The majority of stroke in SCD is caused by large mend other angiographic imaging such as CTA or
vessel disease, which may involve the proximal catheter angiogram for further evaluations regard-
cerebral arteries and the neck arteries [8, 27]. ing diagnosis, treatment, and management.
Macrovascular stroke in SCD has imaging
features of cortical involvement and is more com- Hemorrhagic Stroke
monly unilateral [8]. DWI with ADC map is Hemorrhagic stroke in SCD is relatively more
essential for diagnosing acute ischemic infarct common in adults than in children. Although it
and for differentiating it from chronic ischemic is rare, primary hemorrhagic stroke, such as sub-
changes. Both acute and chronic changes can have arachnoid and parenchymal hemorrhage, is
446 A. Fujita et al.

Fig. 2 A 6-year-old girl presenting with right-sided weak- border zone larger than that shown on FLAIR images con-
ness. (a, b) Axial FLAIR images show multiple high-signal- sistent with acute infarct (arrows). (e) Axial T2-WI image
intensity lesions at the left frontal border zone (watershed) shows loss of flow voids in the left proximal MCA (arrow).
areas (arrows). (c, d) Corresponding axial DWI (b = 1,000) (f) MRA shows obvious stenosis of left MCA (arrow) and
shows restricted diffusion in the left MCA territory and decrease of flow-related signal distally

Fig. 3 A 16-year-old woman presenting with headache zone (watershed) distribution indicating old ischemic
and syncope. (a) Axial FLAIR image demonstrates changes. (b) MRA shows decreased caliber and flow-
encephalomalacia (asterisks) with surrounding gliosis related signal with wall irregularity of the visualized right
(arrows) in the right frontoparietal ACA-MCA border extracranial ICA (arrows)
21 Sickle Cell Disease and Stroke 447

Fig. 4 A 22-year-old woman presenting with seizure.


Axial T2-WI image shows bilateral frontal high signal Fig. 5 A 14-year-old girl presenting with past episode of
intensity, indicating chronic ischemic changes and right hemiplegia. Axial FLAIR image shows high signal
encephalomalacia (asterisks), predominantly in (arrows) and prominent sulci (asterisks), indicating vol-
ACA-MCA border zone (watershed) distribution ume loss and gliosis with cortical involvement in the left
cerebral hemisphere

frequently fatal in SCD patients. Aneurysms are


the most common identified cause of subarach- acute parenchymal hemorrhage is similar to CT,
noid hemorrhage in adult patients with SCD [27], especially when gradient echo sequences, such as
although their exact prevalence is unknown. T2*-WI imaging and SWI, are used [39]. Catheter
Compared with patients without SCD, aneurysms angiogram using three-dimensional (3D) digital
in SCD patients are often multiple, have an subtraction angiography is still the gold standard
increased propensity for the posterior cerebral for the diagnosis and preoperative delineation of
circulation, and may be prone to rupture at smaller intracranial aneurysms. Noninvasive techniques
sizes [38]. Moyamoya syndrome is a common such as MRA and CTA are more commonly
cause of parenchymal hemorrhage in SCD in chil- used, and recently their accuracy is considered
dren [8], which will be discussed later in more satisfactory [40, 41]. CTA is often the first-line
detail. modality to search for underlying aneurysms in
Non-contrast CT is the first imaging modality the setting of acute subarachnoid hemorrhage,
of choice in the diagnosis of acute hemorrhagic while MRA is commonly used in the non-acute
stroke (Figs. 7 and 8). In the current clinical prac- setting (Figs. 9 and 10).
tice, MRI may occasionally be obtained prior to
CT when there is high suspicion for acute stroke Silent Cerebral Infarct (SCI)
based on clinical symptoms. Radiologists should SCI is the most common neurovascular complica-
be familiar with the MR imaging features of acute tion in both pediatric and adult patients with SCD.
hemorrhage that are the same as non-SCD SCIs have been seen in children under the age of
patients, to avoid delayed diagnosis of hemor- 6 years [42]. They may represent a different path-
rhagic strokes. The accuracy of MRI in detecting ogenesis from either ischemic or hemorrhagic
448 A. Fujita et al.

Fig. 6 A 10-year-old girl


presenting with left facial
droop, drooling, and hand
tingling. (a) (b) Axial
FLAIR images show
multiple high signal
intensity vessels in the
Sylvian fissure and
frontoparietal sulci
(arrows), indicating slow
flow or thrombosis. (c)
MRA shows loss of flow-
related signal in the right
MCA distribution
(asterisks) and bilateral
proximal ACAs (arrows)

strokes as they involve small vessels in watershed be progressive [50]. Small foci of acute SCI may be
distributions instead of the larger cerebral vessels found in asymptomatic children with SCD [42].
[43]. Measurement of arterial velocities with SCIs may continue to progress in spite of initiation
transcranial Doppler (TCD) ultrasound are not of chronic transfusion therapy after a stroke [51];
predictive of SCI risk, unlike with overt ischemic however, a recent study showed that in patients
stroke, which supports a different pathophysiol- without a stroke and not at high risk of a stroke,
ogy between overt stroke and SCI [44]. Several chronic transfusions reduced the occurrence of new
studies showed that SCIs are more frequent in SCIs by 56 % over a median observation period of
patients with arterial stenosis and intracranial arte- 3 years [23].
rial tortuosity, suggesting that large vessel disease SCIs are defined as T2 high signal intensity
may play a role in the pathophysiology of these lesions of at least 3 mm in the greatest linear
lesions [42, 45, 46]. Children with arterial stenosis dimension in children and 5 mm in adults, visible
are at higher risk of brain parenchymal injury as on at least two planes of T2-WI images [20, 52]
they have more SCIs [47] (Fig. 10). (Figs. 11 and 12). Because of their small size,
The presence of SCI is considered a predictor of detection of SCI depends largely on the MR imag-
future strokes [17, 48, 49]. SCIs have been shown ing technique, such as the use of FLAIR, slice
to progress in both number and size over time [49]. thickness, multiplanar acquisitions, and the mag-
Associated neurocognitive abnormalities also may netic field strength [53]. With further advances in
21 Sickle Cell Disease and Stroke 449

Fig. 7 A 20-year-old man presenting with altered mental


status. Axial non-contrast CT shows a high density area in
the right frontal lobe white matter with surrounding low Fig. 8 A 14-year-old boy presenting with severe head-
density indicating edema (arrow), compatible with acute ache. Axial non-contrast CT shows high density in the right
hemorrhage Sylvian fissure consistent with acute subarachnoid hemor-
rhage (arrows)

imaging and more utilization of 3.0-T or higher


magnet field units, it is likely that more cases of the cerebral white matter, basal ganglia, thalamus,
SCI will be detected in individuals with SCD [49]. brain stem, and cerebellum. This MR imaging
pattern, sometimes called a “starfield” pattern, is
Cerebral Fat Embolism not very specific and can also be seen in cardio-
Fat embolism syndrome is a very rare but poten- genic or septic emboli, vasculitis, and tiny hem-
tially lethal complication of SCD that is not orrhagic metastases [54]. T2*-WI images and
widely recognized [54]. In non-SCD patients, fat SWI frequently demonstrate microhemorrhages
embolism syndrome is usually a complication of in cerebral fat embolism [55].
long bone fractures. The pathogenesis of fat
embolism syndrome in SCD remains controver-
sial. It is thought to be caused by bone marrow Moyamoya Syndrome
infarcts and necrosis, with subsequent emboliza-
tion through osseous venous channels into the Vascular pathology in SCD includes arterial tor-
venous circulation. Neurologic dysfunction in tuosity, stenosis, occlusion, and aneurysm forma-
the setting of fat embolism syndrome may result tion. A special form of cerebrovascular
directly from vessel occlusion by fat emboli, from manifestation in SCD patients is “moyamoya syn-
disruption of the blood–brain barrier by toxic free drome,” a progressive stenosis of major intracra-
fatty acids, or both [54, 55]. nial arteries with formation of numerous collateral
Typical MR imaging findings of cerebral fat circulations, typically via lenticulostriate and
embolism include tiny foci of restricted diffusion thalamoperforating arteries (Fig. 13). Moyamoya
and corresponding T2 high signal intensities syndrome typically involves the distal ICA (inter-
representing microinfarcts. Lesions can involve nal carotid artery) and proximal ACA (anterior
450 A. Fujita et al.

Fig. 9 A 34-year-old man


presenting with headache.
(a) MRA shows an ovoid
focus of flow-related signal
arising from the junction of
the cavernous–clinoid
segments of the left ICA
extending inferiorly,
consistent with an
aneurysm (arrow). (b)
Catheter angiogram of the
left ICA lateral view
confirmed the aneurysm
(arrow)

Fig. 10 A 22-year-old man presenting with headache and cerebral infarcts. (b) MRA shows a 3 mm aneurysm at the
vertigo. (a) Axial FLAIR image shows multiple high signal medial portion of the left IC-ophthalmic segment (arrow).
foci (arrows) in the left frontal white matter without abnor- Tortuous extracranial right internal carotid artery and bilat-
mal diffusion restriction (not shown) consistent with silent eral vertebral arteries are also noted (open arrows)

cerebral artery) and MCA, with relative sparing of moyamoya syndrome in SCD patients has been
the posterior circulation. The name moyamoya reported for 35 % of conventional catheter angio-
syndrome is derived from moyamoya disease, a gram studies [7] and in 20–40 % of MRA studies
rare, presumed genetic, progressive cerebrovascu- [8, 58]. It was shown that 75 % of patients with
lar disorder. The term “moyamoya” means “puff moyamoya syndrome demonstrated radiographic
of smoke” in Japanese and refers to the appear- progression of arteriopathy and 65 % of patients
ance of collateral vessels on cerebral angiogram showed clinical progression during follow-up
[56]. The pathophysiology of moyamoya syn- [59] (Fig. 14).
drome in SCD is poorly understood. It is hypoth- Moyamoya syndrome can be detected on
esized that sickle-shaped RBCs occlude the vasa MRA as progressive large vessel occlusions and
vasorum of the carotid arteries [31], leading to major collateral flow patterns, the so-called
intimal hyperplasia [57]. The prevalence of moyamoya vessels. On T2-WI images,
21 Sickle Cell Disease and Stroke 451

moyamoya syndrome presents with loss of flow neurosurgery and allows for timely revasculariza-
voids in the circle of Willis (Fig. 15) and on time- tion therapy [59]. Patients with moyamoya syn-
of-flight MRA as a loss of flow-related signal drome may have a particularly poor prognosis and
(Fig. 16). Collaterals typically present as multiple may benefit from revascularization of ischemic
flow voids on T2-WI and T1-WI images in the brain parenchyma by establishing collaterals
thalamoperforate and lenticulostriate territories. from the external carotid artery (ECA) branches
High signal intensity vessels within the cortical to the ICA territories [60]. Superficial temporal
sulci on FLAIR images can indicate a decreased artery (STA) to middle cerebral artery (MCA)
flow of cortical arteries [37] (Fig. 6). bypass is a direct revascularization procedure,
Incidentally discovered asymptomatic which is technically difficult. Indirect revascular-
moyamoya syndrome in children has the potential ization procedures such as encephaloduroarterio-
to progress, both radiographically and clinically. synangiosis (EDAS) and pial synangiosis are
Monitoring and screening with imaging in high- becoming popular for the surgical treatment of
risk patients facilitates early referral to moyamoya syndrome [61]. Pial synangiosis is a
technique in which the dura and the arachnoid are
opened and the STA adventitia is sutured directly
to the pia [62], and it has a relatively low risk of
complications [63]. Pial synangiosis typically
results in an increase in collaterals from the STA
or middle meningeal artery (MMA) to the brain
[62]. MRA or CTA can be used to monitor the
patency of synangiosis between STA branches
and pial arterial branches after cerebral revascu-
larization (Fig. 14).

Extracranial Vasculopathy

Extracranial ICA stenosis is rare, but it has been


reported that there is an association between the
Fig. 11 A 6-year-old girl presenting with SCD for screen-
ing MRI. Coronal FLAIR image shows high signal foci in
presence of extracranial internal carotid
the right frontal subcortical white matter (arrow), compat- arteriopathy and stroke risk in SCD
ible with silent cerebral infarct [64–66]. Doppler ultrasound screening studies

Fig. 12 A 42-year-old
woman presenting with left
facial numbness. (a) Axial
FLAIR image shows
multiple high signal
intensity lesions in the right
frontoparietal white matter
(arrows). (b) DWI
(b = 1,000) shows
corresponding restricted
diffusion in the postcentral
gyrus consistent with acute
infarct (arrow)
452 A. Fujita et al.

Fig. 13 A 24-year-old man presenting with multiple epi- ICA shows occlusion of the supraclinoid ICA (arrow).
sodes of ischemic infarct. (a) Axial FLAIR image shows Posterior communicating artery (PComm) is markedly
bilateral periventricular high signal intensity with cystic prominent (asterisk) and diffuse moyamoya-like collateral
foci compatible with old ischemic change (arrows). (b) vessels are noted (open arrows). (d) Subsequent image of
MRA shows occlusion of the right ICA terminus (asterisk) the ICA catheter angiogram shows more apparent
and severe stenosis of the left ACA (arrow) and distal “moyamoya” (puff-of-smoke) appearance of the collateral
MCA (open arrow). (c) Catheter angiogram of the right vessels (open arrows)

found stenosis or occlusion of the cervical ICA in detected with submandibular Doppler ultrasound
3–6 % of the children, and the majority of these is highly predictive of intracranial arterial stenosis
also had strokes [65]. One MRA study reported seen on MRA [67]. The differential diagnosis of
that patients with an acute neurologic event, TCD extracranial ICA stenosis and/or occlusion
abnormalities, and previous history of stroke also includes atherosclerosis, arteritis, and
showed cervical ICA stenosis and/or occlusion. fibromuscular dysplasia. Age, location, and lack
The most commonly involved artery is the prox- of systemic inflammation or hypercoagulability
imal ICA, within 1 cm from its origin from the help differentiate SCD-related extracranial inter-
CCA [66] (Fig. 17). In addition, a recent study nal carotid arteriopathy from other
showed that the high velocity of extracranial ICA conditions [68].
21 Sickle Cell Disease and Stroke 453

Fig. 14 A 15-year-old man


presenting with recurrent
headache. (a, b) In 2005,
Axial FLAIR image (a)
shows normal brain and
MRA (b) shows possible
stenosis of the right
proximal ACA and bilateral
proximal MCA (asterisk).
(c, d) In 2006, axial FLAIR
image (c) still shows normal
brain, but MRA (d) shows
obvious occlusion of the
right terminal ICA
(asterisk). The patient
received bilateral pial
synangiosis for cerebral
revascularization in 2007.
(e, f) In 2013, axial FLAIR
image (e) shows bilateral
frontal old infarcts with
encephalomalacia and
gliosis (arrows). MRA (f)
shows bilateral occlusion of
the terminal ICAs
(asterisks) and bilateral
prominent superficial
temporal arteries
(arrowheads) after
revascularization. Serial
FLAIR images and MRA
images over 8 years
demonstrate progression of
arterial stenosis, bilateral
frontal lobe infarcts, and
atrophy in spite of
revascularization

Arterial tortuosity is considered to be an tortuosity may serve as a predictor of chronic


adaptive response to chronic anemia in patients brain hypoxia [53]. On imaging, tortuosity can
with SCD. Chronic anemia is associated with be diagnosed when the following features are
increased cardiac output and high blood flow present: dilatation or ectasia of an artery segment,
velocity due to the increased need for blood sup- abnormal increase in length of an arterial segment,
ply, which can lead to arterial tortuosity and can and obvious bowing of an artery [53, 69]
occur in all cervical arteries [68]. Cerebrovascular (Figs. 17, 18, and 19).
454 A. Fujita et al.

Posterior Reversible Encephalopathy


Syndrome (PRES)

There have been several case reports of posterior


reversible encephalopathy syndrome (PRES), also
known as reversible posterior leukoence-
phalopathy syndrome (RPLS), in patients with
SCD [70–74]. The vast majority of the patients
are in the pediatric population [71]. It is believed
that there are two major pathomechanisms in
PRES: a cytotoxic event after an increase in blood
pressure that causes vasoconstriction and cerebral
ischemia or a vasogenic etiology where hyperten-
sion in combination with endothelial dysfunction
and failure of cerebral autoregulation causes vaso-
dilation [70]. Studies have identified PRES as a
common neurological complication of SCD, and
this entity needs to be considered when evaluating
a child with acute neurological deterioration, espe-
Fig. 15 A 22-year-old woman presenting with seizure. cially if the patient is hypertensive, has history of
Axial T2-WI image shows loss of normal flow voids of ACS, and has received systemic steroid therapy or
the bilateral MCA and ACA (arrows). In contrast, promi- blood transfusion [72–74]. A diagnosis of PRES
nent posterior circulation flow voids are noted
(arrowheads)
may predict a high risk for morbidity and mortality
[73]. On MR imaging, PRES usually presents as
symmetrical areas of high signal intensities on
T2-WI and FLAIR images without associated
restricted diffusion, suggesting vasogenic edema,
which is the same as non-SCD patients [73]
(Fig. 20).

Other Sickle Cell Disease Imaging


Manifestations

There are several osseous complications related to


SCD. These include chronic bone marrow
changes in response to long-standing anemia and
acute bone infarcts as well as osteomyelitis. Oste-
oporosis and iron deposition in the bone marrow
due to repeated transfusions can also be seen [13,
15, 68, 75, 76].

Craniofacial Bone
Fig. 16 A 38-year-old woman with SCD presenting with
altered mental status. MRA shows multiple stenoses and Bone Marrow Hyperplasia
occlusions of the right ICA terminus, bilateral ACAs,
bilateral PCAs (posterior cerebral artery), and left MCA As a result of chronic anemia in SCD patients, the
(arrows) red marrow in bony erythrocyte production never
21 Sickle Cell Disease and Stroke 455

Fig. 17 A 30-year-old woman with SCD, evaluation for left CCA (lateral view) shows stenosis of the ICA at the
revascularization. (a) Catheter angiogram of the right CCA ophthalmic segment (arrow). (c) Catheter angiogram of the
(lateral view) shows complete occlusion of the extracranial left subclavian artery shows tortuosity of the left VA
ICA near its origin (arrow). (b) Catheter angiogram of the (arrows)

bones. The non-conversion of the bone marrow is


demonstrated by diffuse low signal intensity on
T1-WI images relative to the signal in the
intervertebral disks [15] (Figs. 21 and 22).

Bone Infarct
Bone infarcts are more common than osteomyeli-
tis among children with SCD. Ischemia causes
pain, even before infarct occurs. For SCD patients
presenting with headache or localizing facial pain,
acute craniofacial bone infarct should be a major
diagnostic consideration [13, 15]. The orbital
walls, mandible, cranial vault, and skull base are
Fig. 18 A 34-year-old woman with SCD, presenting for commonly affected by bone infarcts in SCD
annual MRI follow-up. MRA shows no significant stenosis
patients [13, 15] (Fig. 23). In the spine, it is
or occlusion of the intracranial arteries; however, dilatation
and elongation of the extracranial left ICA are seen hypothesized that bone infarcts contribute to the
(arrows) characteristic “letter H” deformity of vertebral
bodies that is typical of patients with SCD [77]
undergoes the conversion to fatty yellow marrow (Fig. 22).
[68, 75]. Furthermore, as the anemia becomes MR imaging is more sensitive than CT in
more severe and long-standing, the bone marrow detecting bone infarcts. MR imaging findings of
spaces expand secondary to increased RBC pro- acute bone infarcts include increased signal on
duction [68, 75]. This marrow expansion is most T2-WI images, particularly with fat-suppression
frequently noted clinically in the skull and facial techniques such as STIR, and associated high
456 A. Fujita et al.

Fig. 20 A 5-year-old girl with SCD, presenting with


headache and hypertensive status. Axial FLAIR image
shows high signal intensity lesions in the bilateral occipital
lobes (arrows). Given the distribution, clinical course, and
Fig. 19 A 20-year-old man with SCD. MRA shows tor- documented hypertension, the findings are compatible
tuous left extracranial ICA (arrows) and aneurysmal dila- with PRES
tation of the extracranial segment of the left VA (open
arrow)
infarcts. In the presence of fluid collections, MR
signal intensity on DWI with corresponding low susceptibility artifact is suggestive of blood prod-
signal intensity on ADC map (Fig. 24). There may ucts from subperiosteal hematoma, which is more
also be subperiosteal hemorrhage. The high signal commonly seen with bone infarcts. Infectious
intensity of the affected bones on fat-suppressed fluid collections may exhibit restricted
T1-WI images, which may represent the seques- diffusion [13].
tered RBCs in the infarcted marrow [13, 78], can
be very helpful in the differentiation of bone
infarcts from osteomyelitis (Fig. 25). Temporal Bone

Osteomyelitis Sensorineural hearing loss (SNHL) is a well-


Osteomyelitis is less common than bone infarcts recognized inner ear complication of SCD [68,
and typically affects long bones. However, osteo- 79]. The most accepted pathogenesis of SNHL is
myelitis may also occur in craniofacial bones, the recurrent vaso-occlusion of the labyrinthine
particularly the mandible, because of its relatively blood vessels, either in the distribution of the
poor blood supply [13, 68] (Fig. 26). Differentia- anteroinferior cerebellar artery or a branch of the
tion between infarct and osteomyelitis in early basilar artery, which can result in labyrinthine
presentation with imaging alone can be quite chal- hemorrhage (LH) and labyrinthitis ossificans
lenging. The typical MR imaging findings, (LO) [79]. In the past, LH was considered rela-
including bone marrow edema, heterogeneous tively rare in patients of SCD with sudden-onset
bone marrow enhancement, and soft tissue swell- SNHL and vertigo, but it was recently shown that
ing, can be seen in osteomyelitis as well as bone abnormalities can be detected by MR imaging in
21 Sickle Cell Disease and Stroke 457

Fig. 21 An 18-year-old man with SCD. Sagittal T1-WI


image shows the diffuse expansion of the marrow space of
the skull (arrows)

Fig. 23 A 6-year-old boy presenting with temporoman-


dibular joint pain and headache. Axial fat-suppressed
T2-WI image demonstrates high signal intensity in the
left mandibular ramus (arrow), compatible with bone
infarct

tumors such as schwannomas and hemangiomas


that would show intense and localized enhance-
ment, and fat-suppressed MR imaging or high-
resolution temporal bone CT may be used to
exclude lipoma and fat-containing tumors (Fig. 27).
The characteristic imaging finding of LO is the
high density of the membranous labyrinth on
CT. On high-resolution T2-WI images, the normal
T2 high signal from the fluid in the membranous
labyrinth is lost. LO is the end stage of
labyrinthitis, characterized pathologically by pro-
liferation of fibroblasts and finally osteoblasts. In
Fig. 22 A 10-year-old boy with SCD, presenting with patients with SCD presenting with inner ear com-
back pain. Sagittal T1-WI image of the lumbar spine plaints, dedicated temporal bone imaging should
shows low signal in the vertebral bodies (asterisks) relative be performed, preferably by MR imaging
to the intervertebral disk, compatible with an activated (Fig. 28).
bone marrow triggered by chronic anemia. Note also the
“H”-shaped deformity of vertebral bodies, compatible with
end-plate infarcts and subsequent height loss
Advanced Imaging
approximately one-third of SCD patients with
inner ear symptoms, with males preferentially Diffusion Tensor Imaging (DTI)
affected [79].
High signal intensity on non-contrast high-reso- Diffusion tensor imaging (DTI), which measures
lution T1-WI images is a characteristic finding for the directional tendencies of Brownian water mol-
LH. Contrast-enhanced T1-WI images can exclude ecule motion in biologic tissues, has a potential to
458 A. Fujita et al.

Fig. 24 A 15-year-old boy with SCD, presenting with subgaleal fluid collection consistent with hemorrhage
headache. (a) Coronal T2-WI image shows high signal (open arrow). (b) Axial DWI (b = 1,000) shows an area
intensity in the skull consistent with bone marrow edema of increased signal in the skull, compatible with bone
(arrows), as well as mixed abnormal signal intensity in the infarct (arrows)

Fig. 26 A 25-year-old man presenting with left jaw swell-


Fig. 25 A 17-year-old man presenting with headache. ing and pain. Coronal contrast-enhanced CT shows periph-
Sagittal T1-WI image shows high signal intensity of the erally enhancing low density fluid collections medial and
bone marrow in the parietal bone consistent with bone lateral to the left mandibular ramus (arrows), consistent
infarct with hemorrhage (arrow) (Adapted with permission with subperiosteal abscess formation
from Lippincott Williams and Wilkins/Wolters Kluwer
Health: Journal of Computer Assisted Tomography [13],
copyright (2013))
fractional anisotropy (FA), a measure of the pre-
ponderant directionality of water diffusion, are
two of the most frequently used DTI metrics for
detect and quantify microstructural brain changes measuring microstructural tissue damage in
earlier than conventional MRI [80]. This patients with brain disease [80]. DTI directional-
advanced MRI technique can show the orientation ity information is used for fiber tractography,
and integrity of white matter fibers in vivo. The which is the only way to obtain 3D fiber architec-
apparent diffusion coefficient (ADC), which is ture of white matter tracts in vivo [80].
comparable to mean diffusivity as a measure of Using region of interest (ROI) based analysis
the degree of restriction to water diffusion, and in a study comparing SCD patients with controls,
21 Sickle Cell Disease and Stroke 459

FA values in the prefrontal segment of the corpus corpus callosum and sensory segment of the cor-
callosum (which covers the first sixth of the cor- pus callosum (which covers the posterior
pus callosum), upper frontal white matter area, one-third but not including the posterior
centrum semiovale, anterior periventricular one-fourth), caudate nucleus, thalamus, and pons
white matter area, posterior periventricular white [81]. The increase in ADC along the fibers may
matter area, and brain stem were significantly indicate extracellular water content increase sec-
lower in SCD patients than controls. ADC values ondary to axonal loss and fiber density reduction,
were significantly higher in SCD patients com- both of which could be attributed to chronic ische-
pared to controls in the prefrontal segment of the mia. One study found a significant reduction in
DTI fiber counts in the corpus callosum of SCD
patients compared to controls. A significant
reduction in DTI fiber counts was also observed
in the corticospinal tracts bilaterally. These results
can be explained by axonal damage due to
vasculopathy [81].

Perfusion Imaging

Perfusion imaging can be achieved with contrast-


enhanced CT, contrast-enhanced MRI,
non-contrast MRI with arterial spin labeling
(ASL) technique, and positron emission tomogra-
phy (PET) imaging [82]. Perfusion imaging does
not affect treatment decisions in SCD patients.
However, there is value in studying brain perfu-
sion in SCD to better understand the pathophysi-
ology of the disease. Perfusion imaging allows the
Fig. 27 A 14-year-old man presenting with sudden onset
calculation of relative cerebral blood flow (CBF)
of right-sided hearing loss. Axial T1-WI image shows high
signal intensity in the right cochlea (arrow) compared to and blood volume in the brain. Because of the
left, suggesting labyrinthine hemorrhage very high radiation dose compared to

Fig. 28 A 12-year-old boy presenting with hearing loss. CT shows ossification of the cochlea corresponding to
(a) Axial high-resolution T2-WI image (DRIVE sequence) signal loss on MRI (a), consistent with labyrinthitis
shows signal loss of the right cochlea (asterisk). (b) Axial ossificans (arrow)
460 A. Fujita et al.

Fig. 29 A 10-year-old boy with SCD presenting with lobes (arrows). (c) Axial DWI (b = 1,000) 2 days after
acute visual symptoms. (a) Axial DWI (b = 1,000) the initial MRI shows discrete areas of diffusion restriction
shows mildly increased signal in the bilateral parietal in the bilateral parietal lobes (arrows) and in the left frontal
lobes. (b) Arterial spin labeling perfusion image shows periventricular watershed distribution (open arrows)
decreased blood flow in the bilateral parieto-occipital

conventional head CT, CT perfusion and PET MR Spectroscopy (MRS)


imaging should be cautiously used in children
[82]. Perfusion MRI techniques commonly used MR spectroscopy (MRS) is an advanced MR
include dynamic susceptibility contrast (DSC) imaging method for quantitative and qualitative
MRI and ASL. Compared with ASL, DSC MRI assessment of the metabolic state of brain tissue.
perfusion provides higher spatial resolution, Reports of MRS in SCD are limited. In 1992, one
requires shorter scanning time, and can measure study showed that normally appearing brain tis-
other hemodynamic parameters such as cerebral sues on conventional MRI did not show abnormal
blood volume (CBV) and mean transit time MRS findings [88], meaning there was no addi-
(MTT) simultaneously [83]. ASL using either con- tional increase in sensitivity in disease detection
tinuous or pulsed ASL sequence is a noninvasive with MRS. The same group reported a reversible
MR perfusion imaging technique that uses arterial temporary decrease of N-acetylaspartate (NAA),
blood water protons as an endogenous tracer of which is considered a marker of neuronal tissue, at
perfusion [84–86] (Fig. 29). Perfusion imaging the time of a stroke episode [89]. Another study
studies can show changes of cerebral blood flow showed that NAA was increased in the basal
immediately after the event [87]. ASL studies of ganglia and generally in the brain of SCD patients
whole brain CBF in SCD patient compared to [90]. These results suggest that brain NAA
controls have been controversial [84–86]. ASL appears not to be a reliable marker of viable neu-
has been used to correlate CBF with both full- rons in SCD patients at this moment.
scale and performance IQ [85]. Using ASL, studies
have identified right-left CBF asymmetries that
could be early indicators of subclinical pathologi- Quantitative MRI (qMRI)
cal changes in microvasculature and/or hemody-
namics in SCD patients. The described perfusion Remarkable advances in MR imaging have led to
asymmetries, however, could not always be asso- the development of a variety of quantitative MR
ciated with the presence of ischemic lesions imaging (qMRI) techniques, including diffusion-
[84–86]. weighted imaging (DWI), diffusion tensor
21 Sickle Cell Disease and Stroke 461

imaging (DTI), relaxometry, magnetization trans- with normal brain MR images. One study found
fer imaging, perfusion imaging, MR spectros- that neurologically intact adult SCD patients
copy, and volumetry [91]. exhibit brain perfusion decreases in areas such as
qMRI studies of T1 spin–lattice relaxation the basal ganglia, thalamus, watershed areas of the
times have shown reduced gray matter relaxivity anterior circulation, and cerebellar and occipital
in pediatric patients with SCD compared to cortex [97]. These findings may contribute to the
healthy controls [92, 93], especially in the caudate understanding of the mechanisms underlying the
nucleus and in cortical gray matter. In addition, evolution of cerebral vasculopathy in SCD
volumetric deficits of gray matter in patients with patients. Calculation of cerebrovascular reserve
SCD were reported in the absence of volumetric with acetazolamide (Diamox)-induced cerebro-
deficits in the white matter [94]. These observa- vascular vasodilatation in conjunction with
tions support the idea that gray matter is selec- SPECT imaging has a role in identifying SCD
tively vulnerable to injury in pediatric SCD patients who are at a high risk for stroke [98].
patients. In the future, the use of qMRI tech- Since the late 1990s, perfusion and metabolic
niques, such as T1 values and volumetry, may imaging techniques, such as positron emission
serve as early markers of brain damage in young tomography (PET), have been increasingly used
SCD patients [92–94]. to investigate the microvascular blood flow in
qMRI studies examined with mixed TSE pulse patients with SCD. One study showed that PET
sequence showed T1 lengthening, T2 and secular- may detect abnormalities missed by structural
T2 shortening, and an increase in bone marrow MRI in patients with SCD and a history of stroke
volume in patients with SCD [75, 76]. The T1 [99]. Another study found that metabolic compro-
lengthening supports the fact that there is less mise in SCD patients exceeded the corresponding
yellow marrow volume and more red marrow anatomic abnormality demonstrated on
volume in patients with SCD, which suggests the MRI [100].
failure of red-to-yellow marrow conversion. T2
and secular-T2 shortening also supports the fail-
ure of red-to-yellow marrow conversion. Management
Increased iron deposition as a result of hemolysis
in subjects with severe disease and repeated blood Prevention of Primary Stroke
transfusions can account for the shortening of T2
and secular-T2 times. By using the specific Early screening of SCD patients with TCD ultra-
changes seen in T1, T2, and secular-T2 relaxation sound is the gold standard for assessing stroke risk
times, qMRI can be a useful tool in the assessment [25]. In the absence of early TCD screening,
and monitoring of disease severity in patients with stroke occurs in 7.4 % and 11 % of patients with
SCD [75, 76]. In addition, qMRI has been used to SCD by 14 and 20 years of age, respectively, [16,
evaluate the extracranial structures, such as sali- 101] and by age 30 and 45 years, 15 % and 24 %
vary glands [95] and lacrimal glands [96]. will experience overt stroke, respectively [16,
102]. In a newborn cohort, an early TCD screen-
ing and transfusion program employed in patients
Nuclear Medicine at risk decreased the overt stroke risk to 1.9 % by
18 years of age [103]. It has been recommended to
Brain perfusion single-photon emission computed obtain serial TCD and brain MRI/MRA studies to
tomography (SPECT) is a functional neuroimag- monitor the effectiveness of transfusion in chil-
ing technique that allows for noninvasive evalua- dren with abnormal TCD velocities [104]
tion of the physiological and pathophysiological (Fig. 30).
status of the brain. SPECT imaging may demon- The time-averaged mean of the maximal veloc-
strate cerebral perfusion deficits in SCD patients ity (TAMMV) in large intracranial arteries is a
462 A. Fujita et al.

Fig. 30 An 8-year-old boy


with SCD presenting with
annual follow-up with
transcranial Doppler
screening. Peak systolic
velocity (PSV) in the left
MCA was 258.7 cm/s
(<200, normal; 200–249,
conditional; 250
abnormal). Follow-up
MRA showed left MCA
stenosis (not shown)

powerful, noninvasive tool that can be utilized to primary initial measurement for stroke-risk
classify an individual patient’s risk of stroke stratification [107].
[25]. According to the TCD criteria for SCD,
TAMMV < 170 cm/s in any intracranial artery is
considered normal, and TAMMV of 170–199 Prevention of Secondary Stroke
cm/s is conditional. TAMMV > 200 cm/s is con-
sidered abnormal and indicates the need for trans- SCIs that may be found in approximately 20–37 %
fusion. TAMMV > 200 cm/s is associated with a of children with SCD are associated with increased
40 % risk of stroke within the next 3 years risk of subsequent overt stroke [42, 44, 53]. Trans-
[105]. The Stroke Prevention Trial in Sickle Cell fusion therapy has been shown to reduce overt
Anemia (STOP) documented that chronic transfu- strokes in patients with SCIs. Despite transfusion
sion therapy in patients with a mean blood flow treatment, there remains a 20 % secondary stroke
velocity of >200 cm/s with TCD prevented the rate [108] and a 25 % likelihood of additional
initial occurrence of stroke, decreasing the annual SCIs [51]. Hydroxyurea (HU) has also been
incidence of stroke from 10 % to <1 % [25]. TCD shown to reduce TCD velocities and has shown
is repeated every 12 months after a normal scan some benefit in reducing the risk of secondary
and every 3 months with a conditional scan. stroke [109]. A randomized clinical trial, Stroke
Although TAMMV is the gold standard TCD With Transfusions Changing to Hydroxyurea
measurement in the setting of SCD, in clinical (SWiTCH), evaluated HU with phlebotomy as an
practice, peak systolic velocity (PSV) measure- alternative to the ongoing transfusion therapy,
ments are more commonly used in vascular ultra- but the study was closed early after an interim
sound practice and have been shown to be highly analysis revealed equivalent liver iron between
correlated with TAMMV in the prediction of the two groups, failing to meet the primary end
future stroke [106] (Fig. 30). However, a recent point [110]. MRI can monitor the progression and
abstract has demonstrated that PSV alone may extent of the ischemic change, and MRA can mon-
overestimate the risk of stroke in children, dem- itor the stenosis and/or occlusion of the intra- and
onstrating the importance of TAMMV as the extracranial arteries (Fig. 14).
21 Sickle Cell Disease and Stroke 463

Summary 7. Moran CJ, Siegel MJ, DeBaun MR (1998) Sickle cell


disease: imaging of cerebrovascular complications.
Radiology 206:311–321
Sickle cell disease (SCD) is one of the most 8. Moritani T, Numaguchi Y, Lemer NB, Rozans MK,
important hereditary hematologic disorders Robinson AE, Hiwatashi A, Westesson PL (2004)
which may result in severe neurovascular compli- Sickle cell cerebrovascular disease: usual and unusual
cations due to vaso-occlusion. Radiological eval- findings on MR imaging and MR angiography. Clin
Imaging 28:173–186
uation is extremely important in daily practice, 9. Glushakova S, Balaban A, McQueen PG, Coutinho R,
such as TCD in stroke-risk screening with veloc- Miller JL, Nossal R, Fairhurst RM, Zimmerberg J
ity measurements of the cerebral arteries, CT in (2014) Hemoglobinopathic erythrocytes affect the
detecting hemorrhagic strokes, MRI in diagnosing intraerythrocytic multiplication of Plasmodium
falciparum in vitro. J Infect Dis 210:1100–1109
acute infarct, white matter changes and cerebral 10. Halasa NB, Shankar SM, Talbot TR, Arbogast PG,
vasculopathy, and catheter angiogram in evaluat- Mitchel EF, Wang WC, Schaffner W, Craig AS, Grif-
ing precise vascular features and in treatment fin MR (2007) Incidence of invasive pneumococcal
planning. Radiologic imaging features may also disease among individuals with sickle cell disease
before and after the introduction of the pneumococcal
support the current understanding of the patho- conjugate vaccine. Clin Infect Dis 44:1428–1433
physiology underlying neurovascular complica- 11. Quinn CT, Rogers ZR, McCavit TL, Buchanan GR
tions of SCD. Advanced MR imaging and (2010) Improved survival of children and adolescents
nuclear imaging have the potential to provide with sickle cell disease. Blood 115:3447–3452
12. Platt OS, Brambilla DJ, Rosse WF, Milner PF,
additional findings that may improve the manage- Castro O, Steinberg MH, Klug PP (1994) Mortality
ment of the neurovascular complications of SCD, in sickle cell disease. Life expectancy and risk factors
but further investigations are still necessary. for early death. N Engl J Med 330:1639–1644
Finally, radiologists and clinicians should be 13. Watanabe M, Saito N, Nadgir RN, Liao JH, Flower
EN, Steinberg MH, Sakai O (2013) Craniofacial bone
familiar with the pathophysiology and imaging infarcts in sickle cell disease: clinical and radiological
findings of the neurovascular complications of manifestations. J Comput Assist Tomogr 37:91–97
SCD as well as various complications in the cra- 14. Gladwin MT, Vichinsky E (2008) Pulmonary compli-
niofacial region and spine, which may be crucial cations of sickle cell disease. N Engl J Med
359:2254–2265
for timely diagnosis and appropriate treatment. 15. Ejindu VC, Hine AL, Mashayekhi M, Shorvon PJ,
Misra RR (2007) Musculoskeletal manifestations of
sickle cell disease. Radiographics 27:1005–1021
References 16. Ohene-Frempong K, Weiner SJ, Sleeper LA, Miller
ST, Embury S, Moohr JW, Wethers DL, Pegelow CH,
Gill FM (1998) Cerebrovascular accidents in sickle
1. Rees DC, Williams TN, Gladwin MT (2010) Sickle- cell disease: rates and risk factors. Blood 91:288–294
cell disease. Lancet 376:2018–2031 17. Pegelow CH, Macklin EA, Moser FG, Wang WC,
2. Steinberg MH (2008) Sickle cell anemia, the first Bello JA, Miller ST, Vichinsky EP, DeBaun MR,
molecular disease: overview of molecular etiology, Guarini L, Zimmerman RA, Younkin DP, Gallagher
pathophysiology, and therapeutic approaches. Sci DM, Kinney TR (2002) Longitudinal changes in
World J 8:1295–1324 brain magnetic resonance imaging findings in chil-
3. Steen RG, Hankins GM, Xiong X, Wang WC, Beil K, dren with sickle cell disease. Blood 99:3014–3018
Langston JW, Helton KJ (2003) Prospective brain 18. Miller ST, Macklin EA, Pegelow CH, Kinney TR,
imaging evaluation of children with sickle cell trait: Sleeper LA, Bello JA, DeWitt LD, Gallagher DM,
initial observations. Radiology 228:208–215 Guarini L, Moser FG, Ohene-Frempong K,
4. Kanter J, Kruse-Jarres R (2013) Management of Sanchez N, Vichinsky EP, Wang WC, Wethers DL,
sickle cell disease from childhood through adulthood. Younkin DP, Zimmerman RA, DeBaun MR, Cooper-
Blood Rev 27:279–287 ative Study of Sickle Cell D (2001) Silent infarction
5. Connes P, Verlhac S, Bernaudin F (2013) Advances in as a risk factor for overt stroke in children with sickle
understanding the pathogenesis of cerebrovascular cell anemia: a report from the Cooperative Study of
vasculopathy in sickle cell anaemia. Br J Haematol Sickle Cell Disease. J Pediatr 139:385–390
161:484–498 19. Sidani CA, Ballourah W, El Dassouki M,
6. Lonergan GJ, Cline DB, Abbondanzo SL (2001) Muwakkit S, Dabbous I, Dahoui H, Al-Kutoubi A,
Sickle cell anemia. Radiographics 21:971–994 Abboud MR (2008) Venous sinus thrombosis leading
464 A. Fujita et al.

to stroke in a patient with sickle cell disease on globulin for acquired severe aplastic anemia.
hydroxyurea and high hemoglobin levels: treatment Cochrane Database Syst Rev 7, CD006407
with thrombolysis. Am J Hematol 83:818–820 27. Webb J, Kwiatkowski JL (2013) Stroke in patients
20. Vichinsky EP, Neumayr LD, Gold JI, Weiner MW, with sickle cell disease. Expert Rev Hematol
Rule RR, Truran D, Kasten J, Eggleston B, Kesler K, 6:301–316
McMahon L, Orringer EP, Harrington T, Kalinyak K, 28. Arkuszewski M, Melhem ER, Krejza J (2010)
De Castro LM, Kutlar A, Rutherford CJ, Johnson C, Neuroimaging in assessment of risk of stroke in chil-
Bessman JD, Jordan LB, Armstrong FD, Neuropsy- dren with sickle cell disease. Adv Med Sci
chological Dysfunction, Neuroimaging Adult Sickle 55:115–129
Cell Anemia Study Group (2010) Neuropsychologi- 29. Smajlovic D, Sinanovic O (2004) Sensitivity of the
cal dysfunction and neuroimaging abnormalities in neuroimaging techniques in ischemic stroke. Med
neurologically intact adults with sickle cell anemia. Arh 58:282–284
JAMA 303:1823–1831 30. Paonessa A, Limbucci N, Tozzi E, Splendiani A,
21. Mackin RS, Insel P, Truran D, Vichinsky EP, Gallucci M (2010) Radiological strategy in acute
Neumayr LD, Armstrong FD, Gold JI, Kesler K, stroke in children. Eur J Radiol 74:77–85
Brewer J, Weiner MW, Neuropsychological Dysfunc- 31. Roach ES, Golomb MR, Adams R, Biller J, Daniels S,
tion, Neuroimaging Adult Sickle Cell Anemia Study Deveber G, Ferriero D, Jones BV, Kirkham FJ, Scott
Group (2014) Neuroimaging abnormalities in adults RM, Smith ER, American Heart Association Stroke
with sickle cell anemia: associations with cognition. Council, Council on Cardiovascular Disease in the
Neurology 82:835–841 Young (2008) Management of stroke in infants and
22. Charache S, Terrin ML, Moore RD, Dover GJ, Barton children: a scientific statement from a Special Writing
FB, Eckert SV, McMahon RP, Bonds DR (1995) Group of the American Heart Association Stroke
Effect of hydroxyurea on the frequency of painful Council and the Council on Cardiovascular Disease
crises in sickle cell anemia. Investigators of the Mul- in the Young. Stroke 39:2644–2691
ticenter Study of Hydroxyurea in Sickle Cell Anemia. 32. Gonzalez RG, Schaefer PW, Buonanno FS,
N Engl J Med 332:1317–1322 Schwamm LH, Budzik RF, Rordorf G, Wang B,
23. DeBaun MR, Gordon M, McKinstry RC, Noetzel MJ, Sorensen AG, Koroshetz WJ (1999) Diffusion-
White DA, Sarnaik SA, Meier ER, Howard TH, weighted MR imaging: diagnostic accuracy in
Majumdar S, Inusa BP, Telfer PT, Kirby-Allen M, patients imaged within 6 hours of stroke symptom
McCavit TL, Kamdem A, Airewele G, Woods GM, onset. Radiology 210:155–162
Berman B, Panepinto JA, Fuh BR, Kwiatkowski JL, 33. Rydberg JN, Hammond CA, Grimm RC, Erickson
King AA, Fixler JM, Rhodes MM, Thompson AA, BJ, Jack CR Jr, Huston J 3rd, Riederer SJ (1994)
Heiny ME, Redding-Lallinger RC, Kirkham FJ, Initial clinical experience in MR imaging of the
Dixon N, Gonzalez CE, Kalinyak KA, Quinn CT, brain with a fast fluid-attenuated inversion-recovery
Strouse JJ, Miller JP, Lehmann H, Kraut MA, Ball pulse sequence. Radiology 193:173–180
WS Jr, Hirtz D, Casella JF (2014) Controlled trial of 34. Cheng AL, Batool S, McCreary CR, Lauzon ML,
transfusions for silent cerebral infarcts in sickle cell Frayne R, Goyal M, Smith EE (2013) Susceptibility-
anemia. N Engl J Med 371:699–710 weighted imaging is more reliable than T2*-weighted
24. Hulbert ML, Scothorn DJ, Panepinto JA, Scott JP, gradient-recalled echo MRI for detecting
Buchanan GR, Sarnaik S, Fallon R, Chu JY, microbleeds. Stroke 44:2782–2786
Wang W, Casella JF, Resar L, Berman B, 35. Strouse JJ, Lanzkron S, Urrutia V (2011) The epide-
Adamkiewicz T, Hsu LL, Smith-Whitley K, miology, evaluation and treatment of stroke in adults
Mahoney D, Woods G, Watanabe M, DeBaun MR with sickle cell disease. Expert Rev Hematol
(2006) Exchange blood transfusion compared with 4:597–606
simple transfusion for first overt stroke is associated 36. Frietsch T, Ewen I, Waschke KF (2001) Anaesthetic
with a lower risk of subsequent stroke: a retrospective care for sickle cell disease. Eur J Anaesthesiol
cohort study of 137 children with sickle cell anemia. 18:137–150
J Pediatr 149:710–712 37. Maeda M, Tsuchida C (1999) “Ivy sign” on fluid-
25. Adams RJ, McKie VC, Hsu L, Files B, Vichinsky E, attenuated inversion-recovery images in childhood
Pegelow C, Abboud M, Gallagher D, Kutlar A, moyamoya disease. AJNR Am J Neuroradiol
Nichols FT, Bonds DR, Brambilla D (1998) Preven- 20:1836–1838
tion of a first stroke by transfusions in children with 38. Brandao RA, de Carvalho GT, Reis BL, Bahia E, de
sickle cell anemia and abnormal results on Souza AA (2009) Intracranial aneurysms in sickle cell
transcranial Doppler ultrasonography. N Engl J Med patients: report of 2 cases and review of the literature.
339:5–11 Surg Neurol 72:296–299 (discussion 299)
26. Peinemann F, Bartel C, Grouven U (2013) First-line 39. Santhosh K, Kesavadas C, Thomas B, Gupta AK,
allogeneic hematopoietic stem cell transplantation of Thamburaj K, Kapilamoorthy TR (2009) Susceptibil-
HLA-matched sibling donors compared with first-line ity weighted imaging: a new tool in magnetic reso-
ciclosporin and/or antithymocyte or antilymphocyte nance imaging of stroke. Clin Radiol 64:74–83
21 Sickle Cell Disease and Stroke 465

40. Lu L, Zhang LJ, Poon CS, Wu SY, Zhou CS, Luo S, Woods GM, Casella JF, Inusa B, Howard J,
Wang M, Lu GM (2012) Digital subtraction CT angi- Kirkham FJ, Anie KA, Mullin JE, Ichord R,
ography for detection of intracranial aneurysms: com- Noetzel M, Yan Y, Rodeghier M, Debaun MR
parison with three-dimensional digital subtraction (2011) Silent cerebral infarcts occur despite regular
angiography. Radiology 262:605–612 blood transfusion therapy after first strokes in children
41. Sailer AM, Wagemans BA, Nelemans PJ, de Graaf R, with sickle cell disease. Blood 117:772–779
van Zwam WH (2014) Diagnosing intracranial aneu- 52. Casella JF, King AA, Barton B, White DA, Noetzel
rysms with MR angiography. systematic review and MJ, Ichord RN, Terrill C, Hirtz D, McKinstry RC,
meta-analysis. Stroke 45:119–126 Strouse JJ, Howard TH, Coates TD, Minniti CP,
42. Kwiatkowski JL, Zimmerman RA, Pollock AN, Campbell AD, Vendt BA, Lehmann H, Debaun MR
Seto W, Smith-Whitley K, Shults J, Blackwood- (2010) Design of the silent cerebral infarct transfusion
Chirchir A, Ohene-Frempong K (2009) Silent infarcts (SIT) trial. Pediatr Hematol Oncol 27:69–89
in young children with sickle cell disease. Br J 53. Steen RG, Emudianughe T, Hankins GM, Wynn LW,
Haematol 146:300–305 Wang WC, Xiong X, Helton KJ (2003) Brain imaging
43. Pegelow CH, Wang W, Granger S, Hsu LL, findings in pediatric patients with sickle cell disease.
Vichinsky E, Moser FG, Bello J, Zimmerman RA, Radiology 228:216–225
Adams RJ, Brambilla D, Trial S (2001) Silent infarcts 54. Gibbs WN, Opatowsky MJ, Burton EC (2012) AIRP
in children with sickle cell anemia and abnormal best cases in radiologic-pathologic correlation: cere-
cerebral artery velocity. Arch Neurol 58:2017–2021 bral fat embolism syndrome in sickle cell beta-
44. Dowling MM, Quinn CT, Rogers ZR, Buchanan GR thalassemia. Radiographics 32:1301–1306
(2010) Acute silent cerebral infarction in children 55. Mossa-Basha M, Izbudak I, Gurda GT, Aygun N
with sickle cell anemia. Pediatr Blood Cancer (2012) Cerebral fat embolism syndrome in sickle
54:461–464 cell anaemia/beta-thalassemia: importance of
45. Kinney TR, Sleeper LA, Wang WC, Zimmerman RA, susceptibility-weighted MRI. Clin Radiol
Pegelow CH, Ohene-Frempong K, Wethers DL, Bello 67:1023–1026
JA, Vichinsky EP, Moser FG, Gallagher DM, DeBaun 56. Currie S, Raghavan A, Batty R, Connolly DJ,
MR, Platt OS, Miller ST (1999) Silent cerebral Griffiths PD (2011) Childhood moyamoya disease
infarcts in sickle cell anemia: a risk factor analysis. and moyamoya syndrome: a pictorial review. Pediatr
The Cooperative Study of Sickle Cell Disease. Pedi- Neurol 44:401–413
atrics 103:640–645 57. Amlie-Lefond C, Bernard TJ, Sebire G, Friedman
46. Wang WC, Gallagher DM, Pegelow CH, Wright EC, NR, Heyer GL, Lerner NB, DeVeber G, Fullerton
Vichinsky EP, Abboud MR, Moser FG, Adams RJ HJ, International Pediatric Stroke Study Group
(2000) Multicenter comparison of magnetic reso- (2009) Predictors of cerebral arteriopathy in children
nance imaging and transcranial Doppler ultrasonog- with arterial ischemic stroke: results of the Interna-
raphy in the evaluation of the central nervous system tional Pediatric Stroke Study. Circulation
in children with sickle cell disease. J Pediatr Hematol 119:1417–1423
Oncol 22:335–339 58. Dobson SR, Holden KR, Nietert PJ, Cure JK, Laver
47. Arkuszewski M, Krejza J, Chen R, Ichord R, JH, Disco D, Abboud MR (2002) Moyamoya syn-
Kwiatkowski JL, Bilello M, Zimmerman R, Ohene- drome in childhood sickle cell disease: a predictive
Frempong K, Melhem ER (2014) Sickle cell anemia: factor for recurrent cerebrovascular events. Blood
Intracranial stenosis and silent cerebral infarcts in 99:3144–3150
children with low risk of stroke. Adv Med Sci 59. Lin N, Baird L, Koss M, Kopecky KE, Gone E,
59:108–113 Ullrich NJ, Scott RM, Smith ER (2011) Discovery
48. Schatz J, White DA, Moinuddin A, Armstrong M, of asymptomatic moyamoya arteriopathy in pediatric
DeBaun MR (2002) Lesion burden and cognitive syndromic populations: radiographic and clinical pro-
morbidity in children with sickle cell disease. gression. Neurosurg Focus 31:E6
J Child Neurol 17:891–895 60. Arias EJ, Derdeyn CP, Dacey RG Jr, Zipfel GJ (2014)
49. DeBaun MR, Armstrong FD, McKinstry RC, Ware Advances and surgical considerations in the treatment
RE, Vichinsky E, Kirkham FJ (2012) Silent cerebral of moyamoya disease. Neurosurgery 74:S116–S125
infarcts: a review on a prevalent and progressive 61. Adelson PD, Scott RM (1995) Pial synangiosis for
cause of neurologic injury in sickle cell anemia. moyamoya syndrome in children. Pediatr Neurosurg
Blood 119:4587–4596 23:26–33
50. Steen RG, Miles MA, Helton KJ, Strawn S, Wang W, 62. Robertson RL, Burrows PE, Barnes PD, Robson CD,
Xiong X, Mulhern RK (2003) Cognitive impairment Poussaint TY, Scott RM (1997) Angiographic
in children with hemoglobin SS sickle cell disease: changes after pial synangiosis in childhood
relationship to MR imaging findings and hematocrit. moyamoya disease. AJNR Am J Neuroradiol
AJNR Am J Neuroradiol 24:382–389 18:837–845
51. Hulbert ML, McKinstry RC, Lacey JL, Moran CJ, 63. Kennedy BC, McDowell MM, Yang PH, Wilson CM,
Panepinto JA, Thompson AA, Sarnaik SA, Li S, Hankinson TC, Feldstein NA, Anderson RC
466 A. Fujita et al.

(2014) Pial synangiosis for moyamoya syndrome in relaxometry of mandibular bone marrow: a
children with sickle cell anemia: a comprehensive monomodal distribution in sickle cell disease.
review of reported cases. Neurosurg Focus 36:E12 J Magn Reson Imaging 37:1182–1188
64. Calviere L, Viguier A, Guidolin B, Tall P, Larrue V 77. Marlow TJ, Brunson CY, Jackson S, Schabel SI
(2007) Cervical artery stenoses in sickle cell disease. (1998) “Tower vertebra”: a new observation in sickle
Eur Neurol 58:120–121 cell disease. Skeletal Radiol 27:195–198
65. Deane CR, Goss D, Bartram J, Pohl KR, Height SE, 78. Jain R, Sawhney S, Rizvi SG (2008) Acute bone
Sibtain N, Jarosz J, Thein SL, Rees DC (2010) Extra- crises in sickle cell disease: the T1 fat-saturated
cranial internal carotid arterial disease in children sequence in differentiation of acute bone infarcts
with sickle cell anemia. Haematologica from acute osteomyelitis. Clin Radiol 63:59–70
95:1287–1292 79. Saito N, Watanabe M, Liao J, Flower EN, Nadgir RN,
66. Telfer PT, Evanson J, Butler P, Hemmaway C, Steinberg MH, Sakai O (2011) Clinical and radiologic
Abdulla C, Gadong N, Whitmarsh S, Kaya B, findings of inner ear involvement in sickle cell dis-
Kirkham FJ (2011) Cervical carotid artery disease in ease. AJNR AmJ Neuroradiol 32:2160–2164
sickle cell anemia: clinical and radiological features. 80. Le Bihan D, Mangin JF, Poupon C, Clark CA,
Blood 118:6192–6199 Pappata S, Molko N, Chabriat H (2001) Diffusion
67. Verlhac S, Balandra S, Cussenot I, Kasbi F, Vasile M, tensor imaging: concepts and applications. J Magn
Kheniche A, Elmaleh-Berges M, Ithier G, Reson Imaging 13:534–546
Benkerrou M, Bernaudin F, Sebag G (2014) Extracra- 81. Balci A, Karazincir S, Beyoglu Y, Cingiz C,
nial carotid arteriopathy in stroke-free children with Davran R, Gali E, Okuyucu E, Egilmez E (2012)
sickle cell anemia: detection by submandibular Dopp- Quantitative brain diffusion-tensor MRI findings in
ler sonography. Pediatr Radiol 44:587–596 patients with sickle cell disease. AJR Am J
68. Saito N, Nadgir RN, Flower EN, Sakai O (2010) Roentgenol 198:1167–1174
Clinical and radiologic manifestations of sickle cell 82. Behpour AM, Shah PS, Mikulis DJ, Kassner A (2013)
disease in the head and neck. Radiographics Cerebral blood flow abnormalities in children with
30:1021–1034 sickle cell disease: a systematic review. Pediatr
69. Steen RG, Reddick WE, Glass JO, Wang WC (1998) Neurol 48:188–199
Evidence of cranial artery ectasia in sickle cell disease 83. Kirkham FJ, Calamante F, Bynevelt M, Gadian DG,
patients with ectasia of the basilar artery. J Stroke Evans JP, Cox TC, Connelly A (2001) Perfusion
Cerebrovasc Dis 7:330–338 magnetic resonance abnormalities in patients with
70. Frye RE (2009) Reversible posterior leukoence- sickle cell disease. Ann Neurol 49:477–485
phalopathy syndrome in sickle-cell anemia. Pediatr 84. Oguz KK, Golay X, Pizzini FB, Freer CA, Winrow N,
Neurol 40:298–301 Ichord R, Casella JF, van Zijl PC, Melhem ER (2003)
71. Geevasinga N, Cole C, Herkes GK, Barnett Y, Lin J, Sickle cell disease: continuous arterial spin-labeling
Needham M (2014) Sickle cell disease and posterior perfusion MR imaging in children. Radiology
reversible leukoencephalopathy. J Clin Neurosci 227:567–574
21:1329–1332 85. Strouse JJ, Cox CS, Melhem ER, Lu H, Kraut MA,
72. Henderson JN, Noetzel MJ, McKinstry RC, White Razumovsky A, Yohay K, van Zijl PC, Casella JF
DA, Armstrong M, DeBaun MR (2003) Reversible (2006) Inverse correlation between cerebral blood
posterior leukoencephalopathy syndrome and silent flow measured by continuous arterial spin-labeling
cerebral infarcts are associated with severe acute (CASL) MRI and neurocognitive function in children
chest syndrome in children with sickle cell disease. with sickle cell anemia (SCA). Blood 108:379–381
Blood 101:415–419 86. van den Tweel XW, Nederveen AJ, Majoie CB, van
73. Khademian Z, Speller-Brown B, Nouraie SM, der Lee JH, Wagener-Schimmel L, van Walderveen
Minniti CP (2009) Reversible posterior leuko- MA, Poll The BT, Nederkoorn PJ, Heijboer H,
encephalopathy in children with sickle cell disease. Fijnvandraat K (2009) Cerebral blood flow measure-
Pediatr Blood Cancer 52:373–375 ment in children with sickle cell disease using contin-
74. Kolovou V, Zampakis P, Ginopoulou A, uous arterial spin labeling at 3.0-Tesla MRI. Stroke
Varvarigou A, Kaleyias J (2013) Reversible posterior 40:795–800
leukoencephalopathy syndrome after blood transfu- 87. Sanelli PC, Sykes JB, Ford AL, Lee JM, Vo KD,
sion in a pediatric patient with sickle cell disease. Hallam DK (2013) Imaging and treatment of patients
Pediatr Neurol 49:213–217 with acute stroke: an evidence-based review. AJNR
75. Elias EJ, Liao JH, Jara H, Watanabe M, Nadgir RN, Am J Neuroradiol 35:1045–1051
Sakai Y, Erbay K, Saito N, Ozonoff A, Steinberg MH, 88. Wang Z, Bogdan AR, Zimmerman RA, Gusnard DA,
Sakai O (2013) Quantitative MRI analysis of cranio- Leigh JS, Ohene-Frempong K (1992) Investigation of
facial bone marrow in patients with sickle cell disease. stroke in sickle cell disease by 1H nuclear magnetic
AJNR Am J Neuroradiol 34:622–627 resonance spectroscopy. Neuroradiology 35:57–65
76. Liao JH, Jara H, Nadgir R, Elias E, Nowrouzi N, 89. Wang Z, Zimmerman RA, Sauter R (1996) Proton
Saito N, Steinberg MH, Sakai O (2013) qMRI MR spectroscopy of the brain: clinically useful
21 Sickle Cell Disease and Stroke 467

information obtained in assessing CNS diseases in 101. Quinn CT, Rogers ZR, Buchanan GR (2004) Survival
children. AJR Am J Roentgenol 167:191–199 of children with sickle cell disease. Blood
90. Steen RG, Ogg RJ (2005) Abnormally high levels of 103:4023–4027
brain N-acetylaspartate in children with sickle cell 102. Switzer JA, Hess DC, Nichols FT, Adams RJ (2006)
disease. AJNR Am J Neuroradiol 26:463–468 Pathophysiology and treatment of stroke in sickle-cell
91. Watanabe M, Liao JH, Jara H, Sakai O (2013) Multi- disease: present and future. Lancet Neurol 5:501–512
spectral quantitative MR imaging of the human brain: 103. Bernaudin F, Verlhac S, Arnaud C, Kamdem A,
lifetime age-related effects. Radiographics Chevret S, Hau I, Coic L, Leveille E,
33:1305–1319 Lemarchand E, Lesprit E, Abadie I, Medejel N,
92. Steen RG, Langston JW, Ogg RJ, Xiong X, Ye Z, Madhi F, Lemerle S, Biscardi S, Bardakdjian J,
Wang WC (1999) Diffuse T1 reduction in gray matter Galacteros F, Torres M, Kuentz M, Ferry C,
of sickle cell disease patients: evidence of selective Socie G, Reinert P, Delacourt C (2011) Impact of
vulnerability to damage? Magn Reson Imaging early transcranial Doppler screening and intensive
17:503–515 therapy on cerebral vasculopathy outcome in a new-
93. Steen RG, Schroeder J (2003) Age-related changes in born sickle cell anemia cohort. Blood
the pediatric brain: proton T1 in healthy children and 117:1130–1140, quiz 436
in children with sickle cell disease. Magn Reson 104. Sheehan VA, Hansbury EN, Smeltzer MP, Fortner G,
Imaging 21:9–15 McCarville MB, Aygun B (2013) Transcranial Dopp-
94. Steen RG, Emudianughe T, Hunte M, Glass J, Wu S, ler velocity and brain MRI/MRA changes in children
Xiong X, Reddick WE (2005) Brain volume in pedi- with sickle cell anemia on chronic transfusions to
atric patients with sickle cell disease: evidence of prevent primary stroke. Pediatr Blood Cancer
volumetric growth delay? AJNR Am J Neuroradiol 60:1499–1502
26:455–462 105. Adams RJ, Nichols FT, Figueroa R, McKie V, Lott T
95. Liao J, Saito N, Ozonoff A, Jara H, Steinberg M, (1992) Transcranial Doppler correlation with cerebral
Sakai O (2012) Quantitative MRI analysis of salivary angiography in sickle cell disease. Stroke
glands in sickle cell disease. Dentomaxillofac Radiol 23:1073–1077
41:630–636 106. Jones A, Granger S, Brambilla D, Gallagher D,
96. Buch K, Watanabe M, Elias EJ, Liao JH, Jara H, Vichinsky E, Woods G, Berman B, Roach S,
Nadgir RN, Saito N, Steinberg MH, Sakai O (2014) Nichols F, Adams RJ (2005) Can peak systolic veloc-
Quantitative magnetic resonance imaging analysis of ities be used for prediction of stroke in sickle cell
the Lacrimal gland in sickle cell disease. J Comput anemia? Pediatr Radiol 35:66–72
Assist Tomogr 38:674–680 107. Ishola T, Quinn CT (2013) Transcranial Doppler peak
97. Deus-Silva L, Bonilha L, Damasceno BP, Costa AL, systolic velocities overestimate the risk of stroke in
Yasuda CL, Costa FF, Santos AO, Etchebehere EC, sickle cell anemia. Blood 122:2240
Oquendo-Nogueira R, Fockink R, de Freitas CF, 108. Scothorn DJ, Price C, Schwartz D, Terrill C,
Camargo EE, Li LM, Cendes F, Saad ST (2013) Brain Buchanan GR, Shurney W, Sarniak I, Fallon R, Chu
perfusion impairment in neurologically asymptomatic JY, Pegelow CH, Wang W, Casella JF, Resar LS,
adult patients with sickle-cell disease shown by Voxel- Berman B, Adamkiewicz T, Hsu LL, Ohene-
based analysis of SPECT images. Front Neurol 4:207 Frempong K, Smith-Whitley K, Mahoney D, Scott
98. Kedar A, Drane WE, Shaeffer D, Nicole M, Adams C JP, Woods GM, Watanabe M, Debaun MR (2002)
(2006) Measurement of cerebrovascular flow reserve Risk of recurrent stroke in children with sickle cell
in pediatric patients with sickle cell disease. Pediatr disease receiving blood transfusion therapy for at
Blood Cancer 46:234–238 least five years after initial stroke. J Pediatr
99. Powars DR, Conti PS, Wong WY, Groncy P, Hyman C, 140:348–354
Smith E, Ewing N, Keenan RN, Zee CS, Harold Y, Hiti 109. Madden NA, Jones GL, Kalpatthi R, Woods G (2014)
AL, Teng EL, Chan LS (1999) Cerebral vasculopathy Practice patterns of stroke screening and hydroxyurea
in sickle cell anemia: diagnostic contribution of posi- use in children with sickle cell disease: a survey of
tron emission tomography. Blood 93:71–79 health care providers. J Pediatr Hematol Oncol 36:
100. Reed W, Jagust W, Al-Mateen M, Vichinsky E (1999) e382–e386
Role of positron emission tomography in determining 110. Ware RE, Helms RW, Investigators SW (2012) Stroke
the extent of CNS ischemia in patients with sickle cell with transfusions changing to Hydroxyurea
disease. Am J Hematol 60:268–272 (SWiTCH). Blood 119:3925–3932
Hypoxic–Ischemic Encephalopathy
(Preterm, Term, and Adult) 22
Mauricio Castillo and Francisco Chiang

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 470 Hypoxic–ischemic injury (HII) to the brain is
usually a devastating event and an important
Physiopathology of HII and Factors
Influencing the Patterns of Injury . . . . . . . . . . . . . . . . . 470
cause of morbidity and mortality in the United
States and elsewhere in the world. Neuroimag-
HII in the Preterm Neonate . . . . . . . . . . . . . . . . . . . . . . . . 472 ing plays a pivotal role in diagnosis, treatment,
Severe Asphyxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 472
Mild-to-Moderate Asphyxia . . . . . . . . . . . . . . . . . . . . . . . . . 473 and long-term prognosis determination for
these patients. The correct diagnosis made on
HII in the Term Neonate . . . . . . . . . . . . . . . . . . . . . . . . . . . 478
Severe Asphyxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 478
the basis of different imaging modalities
Partial Asphyxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 482 requires knowledge of the different manifesta-
tions of this type of injury. Some of the factors
HII in Postnatal Infants and Young Children . . . . 483
Severe Asphyxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 483 that contribute to the different findings are
Mild-to-Moderate Asphyxia . . . . . . . . . . . . . . . . . . . . . . . . . 484 brain maturity, duration and severity of the
HII in Older Children and Adults . . . . . . . . . . . . . . . . 485 insult, underlying cause, and associated
disorders.
Delayed White Matter Injury . . . . . . . . . . . . . . . . . . . . . 486
Severe HII will result in preferentially deep
Role of Proton MR Spectroscopy in gray matter damage in preterm and term
the Evaluation of HII . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 488 infants, with peri-rolandic involvement more
Imaging Choices in HII . . . . . . . . . . . . . . . . . . . . . . . . . . . . 490 frequently observed in the latter age group. In
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 492 these patients, a less profound insult will result
in germinal matrix hemorrhages or
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 492
periventricular leukomalacia (PVL) in preterm
neonates and parasagittal watershed infarcts in
term neonates. In the postnatal period, severe
insults produce diffuse gray matter injury, with
relative sparing of the peri-rolandic cortex and
posterior circulation structures. In older chil-
dren and adults, profound insults produce
injury in the deep gray matter nuclei, cortices,
M. Castillo (*) • F. Chiang hippocampi, and cerebellum.
Division of Neuroradiology, Department of Radiology,
The use of advanced MRI techniques such
University of North Carolina, Chapel Hill, NC, USA
e-mail: mauricio_castillo@med.unc.edu; as DWI and MR spectroscopy is useful in
franciscochiang@gmail.com

# Springer Science+Business Media New York 2016 469


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_28
470 M. Castillo and F. Chiang

making the diagnosis especially in the acute It is necessary to emphasize, however, that
setting where conventional imaging might be findings in HII are variable. They depend on
less sensitive. many different factors such as age (brain matu-
rity), duration, severity, and exact type of insult
Keywords and also the modality and timing of the imaging
HII • Hypoxic–ischemic injury • Brain ische- studies. Because of this difficult imaging scenario
mia • Global brain hypoxia and the necessity of making a fast and accurate
diagnosis, the radiologist requires a thorough
knowledge of the imaging patterns of HII and be
Introduction able to identify subtle findings by looking for the
specific areas most likely to be injured when HII is
Hypoxic–ischemic injury (HII) to the brain is suspected clinically.
usually a devastating event, still considered the In this chapter, first the pathophysiologic fea-
third leading cause of death in the United States tures of HII and how they influence the imaging
with approximately half a million new victims per patterns are discussed. Then, the specific imaging
year. Death will occur in nearly one third of these manifestations in term (>36 weeks gestational
patients, while another third will end up suffering age) and preterm (<36 weeks gestational age)
severe neurologic deficits with important func- neonates, postnatal infants, children, and adults
tional impairment [1]. Even more, perinatal are reviewed.
asphyxia is considered an important risk factor
for cognitive and behavioral difficulties in surviv-
ing children [2]. Physiopathology of HII and Factors
This serious and life-threatening condition is Influencing the Patterns of Injury
most often caused by insults as cardiac arrest,
asphyxia, poisoning (drug overdose or carbon Earlier in this chapter, some of the most important
monoxide intoxication), and head trauma. Sup- causes of HII were mentioned. Independent from
portive care is the most used primary treatment the underlying causes, the main pathophysiologic
strategy, which in majority of cases fails to pre- processes that lead to HII are remarkably similar.
vent the brain injury that quickly follows the The main two processes that cause HII are ische-
primary insult. Other treatment techniques are mia and hypoxemia. Depending on the type of
being studied, including promising new targeted injury, the duration of injury, and the patient’s
neuroprotective approaches such as hypothermia age (brain maturity), these two processes will
and excitatory amino acid antagonists. However, play different roles influencing the pathologic
most of these new treatments have a limited win- and imaging findings.
dow of time to be effective even as little as 6 h post In general, infants and small children are more
insult [3]. For this reason, times spent in the trans- inclined to suffer asphyxia, due to perinatal
portation of patients to the hospital and early asphyxia or being traumatic in nature (pool acci-
detection of this injury are extremely important dents, etc.). In asphyxia, the chain of events that
to the final outcomes of these patients [4]. As lead to brain HII starts with hypoxemia. If the
treatment protocols seize being based on time hypoxemia is maintained long enough and if the
and become based on tissue status, neuroimaging redistribution of oxygenated blood fails to com-
plays an important role with ultrasonography pensate for it, cardiac hypoxia and decreased car-
(US), computed tomography (CT), and magnetic diac output occur. After this, brain ischemia
resonance (MR) imaging permitting early inter- ensues due to decreased blood flow. For this rea-
vention. Also, imaging in the subacute stages is son, the type of brain injury caused by asphyxia is
useful in predicting long-term outcomes of these ischemia superimposed on hypoxia. Even more,
patients by providing precise information about there is evidence that ischemia superimposed on
the severity and extent of injury [5–8]. hypoxia is required for the HII to occur as acute
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 471

hypoxia alone is unlikely to cause damage to the peroxynitrite. Also, damage to the mitochondria
neonatal brain unless it is prolonged. This reflects may diminish their capacity of handling multiple
the resistance of the immature brain to hypoxia oxidation reactions and results in further loss of
when compared to the adult brain as well as better ATP enabling the production of more free radicals
compensatory mechanisms in babies [9]. In con- and perpetuating the cycle of membrane depolar-
trast to this, in adults, direct blood flow reductions ization and NMDA receptor channel opening.
as in cardiac arrest or cerebrovascular disease, Finally, delayed depletion of energy and elevated
which lead to primary ischemia with secondary brain lactate levels are associated with neuronal
hypoxia, are common underlying mechanisms. death and resultant neurodegeneration after
Another important aspect of HII physiopathol- asphyxia.
ogy is that not all brain structures are affected to Finally, from all of these biochemical and path-
the same degree in a global hypoxic–ischemic ophysiological data mentioned before, there are
event. It is known that there are certain tissues some important conclusions to be kept in mind
more likely to be injured or injured earlier because when analyzing the imaging patterns of HII.
of their different sensitivity to hypoxia. This con- First, some areas of the brain are more susceptible
cept is known as selective vulnerability. HII to ischemic injury than others primarily
causes damage through a process called because of their concentrations of glutamate and
excitotoxicity [9]. This process refers to cellular other excitatory amino acid receptors (primarily
death due to the excessive stimuli of certain amino located in the gray matter), a concept known as
acid receptors that normally mediate physiologic selective vulnerability. Second, there are some
excitatory effects of dicarboxylic acid glutamate, areas of the brain with more energy demand
a ubiquitous neurotransmitter in the brain. This than others. In these, energy will be depleted
stimuli end up triggering a complex cascade of faster and therefore the injury will ensue earlier.
biochemical events that finally produces selective Third, due to delayed neuron death (apoptosis),
death of certain populations of neurons. This pro- not all of the injury is evident until days after the
cess appears to be even more important in devel- initial insult.
oping brains than in adults. It is important to realize that in any given
Brain ischemia, on the other hand, produces a patient, the sites of the brain that are most vulner-
change from oxidative phosphorylation to able and are first affected by HII will be deter-
anaerobic metabolism which is inefficient. Rapid mined by the degree of maturity of the brain,
depletion of adenosine triphosphate (ATP) occurs, which depends on the age of the patient. This is
and lactate accumulates in cells with loss of one of the reasons why HII manifestations in the
normal functions in their membrane. The failure perinatal period (up to 1 month of age) differ from
to produce ATP and the subsequent depolarization those seen in older infants and in adults. Even
of the neuronal cell membranes cause excessive between term and preterm infants, there are
release of glutamate from nerve terminals, which important differences in patterns of injury. It is
in combination with the reduced activity of glial relevant to be cognizant of the degree of brain
pumps that normally keep synaptic glutamate maturity to correctly interpret the studies for
levels low produces a rapid increase in glutamate suspected HII. Another important factor determin-
concentration. When this process occurs, ing the distribution of injuries is the severity and
neurons and other cells with the appropriate duration of the HII. Severe episodes of
receptors die due to a complex neurotoxic cas- hypoxia–ischemia produce different injury pat-
cade. In immature brains, glutamate and reduced terns when compared to lesser ones. In insults of
membrane potential contribute to the opening of short duration, usually there is no brain injury
N-methyl-D-aspartate (NMDA) receptors which especially in the immature brain due to its
cause a massive influx of calcium (Ca2+) into enhanced resistance to hypoxia. In the pediatric
neurons. Direct effects of Ca2+ include generation population, the time of hypoxia–ischemia
of oxygen free radicals such as nitric oxide and required to produce brain injury has been
472 M. Castillo and F. Chiang

estimated to be approximately 15 min and in Severe Asphyxia


adults 3–4 min [10]. With all these factors that
influence the patterns of injury in mind, we can The injury pattern of preterm babies following a
now review the different manifestations of HII in severe but brief hypoxic event resembles the pat-
imaging studies in preterm, term, and older tern seen in term infants (see below) but with
children and adults. some differences [12]. These patients often show
lesions in the basal ganglia, thalami, brainstem
structures, cerebellum, and corticospinal tracts, as
HII in the Preterm Neonate well as decreased cerebral hemispheric white mat-
ter. Although basal ganglia injury is frequent, it is
Preterm neonates are susceptible to suffer HII, and less severe than the involvement of the thalami in
this type of injury is more common in this group this age group and especially among those less than
than in term neonates. The consequences of HII 32 weeks of age. When involved, the basal ganglia
can be devastating, as at least 5 % of infants born tend to atrophy without scarring. Overall, the thal-
before 32 weeks gestational age and approxi- ami, anterior vermis, and dorsal brainstem are the
mately 19 % of preterm neonates less than most commonly involved structures when pro-
28 weeks develop cerebral palsy. If one takes all found asphyxia happens [3, 13]. The peri-rolandic
cases of cerebral palsy, approximately 50 % of cortex is usually spared in these patients. Germinal
them occur in preterm neonates [11]. This situa- matrix hemorrhages and periventricular white mat-
tion can be explained mainly by two reasons. First, ter injuries may also be seen in this group.
in this group, there is increased frequency in events The reason for the differences in the extent of
leading to HII, such as respiratory distress syn- injury between the putamen and thalamus that is
drome, pneumothorax, patent ductus arteriosus, seen in these patients is likely related to the degree
pregnancy infection, etc. Second, preterm infants of myelination of these structures. The thalamus,
have less autoregulation and compensatory capac- the pallidothalamic fibers of the posterior internal
ity when compared to term neonates. Also, in this capsule, and globus pallidus myelinate at about
age group, making a diagnosis is more difficult 24–25 weeks of gestation, whereas the corpus
because the clinical examination is limited, signs striatum (caudate nucleus and putamen) does not
of HII may be lacking, or if present, they can be myelinate until 35–36 weeks. Myelinated struc-
mistaken for developmental immaturity. Further- tures are more metabolically active, as shown in
more, preterm infants not uncommonly have tran- F-18 FDG positron emission tomography studies
sient neurological abnormalities and decreased [14], when compared to less myelinated areas, as
muscle tone as part of their immature central ner- the putamen and caudate, and therefore they are
vous system, and most of these patients do not more susceptible to hypoxic injury [15].
develop long-term deficits. For this is the reason, Usually, the first imaging study performed in
imaging is the key to arrive at the correct diagnosis. preterm neonates is a cranial transfontanelle
Imaging manifestations of HII in preterm US. As this is a noninvasive, bedside examina-
babies differ from those seen in term infants, tion, it can be easily used in the intensive care unit
mainly because of their degree of immaturity. In as a screening tool to determine significant brain
terms of the imaging findings, HII can be divided injury. The findings in US suggesting HII include
in two groups: those with severe and those with increased echogenicity in the thalami at 2–3 days
mild-to-moderate asphyxia. One must realize that following the insult; nevertheless, US may appear
this is an arbitrary and somewhat simplistic divi- normal, especially in the first 2 days. MRI is
sion because in any given patient, there is a con- generally the next study performed in preterm
tinuous range of insults that are variable in terms neonates with suspected HII and is complimen-
of duration and severity, and therefore it is not tary to US and necessary to accurately assess the
surprising to have different imaging features in the extent of the injury and predict outcomes espe-
same patient. cially cognitive delay [16].
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 473

Fig. 2 T1WI in a preterm neonate who suffered severe


asphyxia shows hyperintensity in the thalami and posterior
Fig. 1 Severe asphyxia in a preterm neonate. DWI image
lentiform nuclei. The high signal in T1 can persist into the
shows hyperintensity in the thalami
chronic stage

Conventional MRI sequences can be normal or


show only subtle abnormalities within the first day involute by the first half of the third trimester so
after injury. DWI is the most sensitive sequence in that by the 34th week, the germinal zones have
the first 24 h showing abnormalities in the thalami almost completely involute. For this reason, ger-
(Fig. 1). Then, after approximately 2 days post minal matrix hemorrhages are infrequent before
injury, T2 hyperintensity can be seen in the thal- this age [3]. A very important anatomic landmark
ami and basal ganglia. By the third day, T1 short- to recognize is the caudothalamic notch. The
ening ensues in the injured areas. As in term caudothalamic notch is a groove where the last
infants, the DWI abnormalities peak at about remnant of the germinal matrix to involute – the
3–5 days and then tend to pseudo-normalize ganglionic eminence – is located. The
[3, 10, 17]. Thereafter, T2 hyperintensity appears caudothalamic notch is a groove located between
at approximately 7 days and T1 shortening per- the caudate head and the thalamus and is where
sists into the chronic stage (Fig. 2). most of germinal matrix hemorrhages originate.
The pathogenesis of germinal matrix hemorrhage
is related to the relative higher vascularization of
Mild-to-Moderate Asphyxia this region and the properties of the vascular bed
in it. The capillaries in this region are fragile,
The most characteristic pattern of injury in mild- mainly because they are lined only by simple
to-severe asphyxia in preterm babies is deter- endothelium and lack the muscular or collagenous
mined by direct injury and hemorrhage of the layers that are present in the larger blood vessels.
germinal matrix. The germinal matrix is a highly Second, cells that compose their endothelium
cellular region that lines the walls of the lateral have high concentration of mitochondria
ventricles in fetal life and from where neurons and reflecting their high oxidative metabolic require-
glia arise and migrate. It is most active between ments. This makes them susceptible to hypoxic
the second half of the first trimester and the first conditions. It is believed that first, this fragile
half of the second trimester. Thereafter, it starts to endothelium suffers a loss of integrity due to
474 M. Castillo and F. Chiang

hypoxia and then with restoration of the normal parenchymal periventricular hemorrhagic
circulation by resuscitation, bleeding ensues. This infarcts, probably venous in origin, with extension
hemorrhage can be localized in the caudothalamic to the ventricular system. There is a correlation
notch or extend to the ventricles. The prevalence between higher hemorrhage grade and higher
of intraventricular hemorrhage in preterm neo- perinatal mortality rates as well as a higher prev-
nates weighting less than 2 Kg has been estimated alence of long-term neurological sequelae [10].
at approximately 25 % and most of them are Germinal matrix and intraventricular hemor-
related to hemorrhages of the germinal matrix. rhages can be adequately evaluated with cranial
Also, it is known that most hemorrhages happen US (Figs. 3, 4, and 5), keeping in mind that
within the first 24 h of life and that infants who are sometimes the findings can be subtle and difficult
very premature and with a very low birth weight to visualize. US of the posterior fossa by a poste-
are at higher risk for developing intraventricular rior fontanelle approach, as a complement to the
hemorrhage [18, 19]. classic anterior fontanelle examination, can help
Germinal matrix hemorrhages are divided in to better visualize the posterior supra- and
four grades reflecting their locations and degree of infratentorial structures. This can help to diagnose
dilatation of the ventricles (Table 1). subtle intraventricular hemorrhages when the ven-
Grades I–III are hemorrhages that arise from tricles are not dilated and cerebellar hemorrhages
the germinal matrix and have variable extension that are believed to be underdiagnosed. These last
to the lateral ventricles. Grade IV hemorrhages are types of hemorrhages are clinically silent but are
not germinal matrix hemorrhages but are not uncommon and are recognized in 10–20 % of
autopsies. In fact, cerebellar hemorrhages are
believed to be no different in origin than
Table 1 Germinal matrix hemorrhage (GMH) –
periventricular hemorrhage (IVH) grading caudothalamic notch hemorrhages also arising
from germinal matrix remnants within the exter-
Grade I Subependymal GMH (mostly in the
caudothalamic groove) nal granule cell layer of the cerebellum and in the
Grade II GMH and IVH with or without mild subependymal layer of the roof of the fourth ven-
ventriculomegaly tricle [3, 20, 21]. Cerebellar hemorrhages are seen
Grade III GMH and IVH with ventriculomegaly as lentiform or crescent-shaped hyperechoic
Grade IV Above + periventricular parenchymal lesions located posterior and peripheral in the
hemorrhagic infarction (not true GMH) cerebellar hemispheres. MRI is usually the next

Fig. 3 US image in a preterm patient. Coronal (a) and sagittal image confirms the location in the caudothalamic
sagittal (b) images demonstrate bilateral areas of groove. Choroid plexus is large and thought not to be
subependymal echogenicity, right greater than left. The related to the hemorrhage. GMH grade I
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 475

Fig. 4 US coronal (a) and sagittal (b) images in a preterm neonate demonstrate extension of the left side hemorrhage into
the lateral ventricles, right greater than left. The ventricles are not enlarged. GMH grade II

Fig. 5 US coronal (a) and sagittal (b) images in a preterm neonate demonstrate bilateral intraventricular hemorrhage with
enlargement of the lateral ventricles. GMH grade III

study used most of the times to detect concomitant anatomic and physiological immaturity of the
injuries such as white matter injury of prematurity blood vessels in preterm patients [22, 23]. These
or deep gray matter injury. oligodendrocyte precursors are late precursors
Another common manifestation that can be known as preoligoendrocytes, and the white mat-
seen in mild-to-moderate asphyxia in preterm ter in the period prior to myelination is populated
babies is periventricular leukomalacia (PVL), with them. This is the period of higher risk for
also known as white matter injury of prematurity. PVL, as these late precursor cells are believed to
This type of injury also appears to be inversely be even more susceptible to hypoxia than the
related to gestational age at birth. Its pathogenesis earlier precursor cells or the mature oligodendro-
is believed to be related to selective vulnerability cytes [24]. This explanation is supported by the
of oligodendrocyte precursors and to perturba- fact that the prevalence of PVL declines after
tions in cerebral blood flow in a context of 32 weeks, the same time that the population of
476 M. Castillo and F. Chiang

Fig. 6 US coronal (a) and sagittal (b) images in a preterm neonate demonstrate the third stage of PVL with the
development of bilateral periventricular cysts

these late precursors in the periventricular white collapse resulting in gliosis and loss of white
matter maturates into oligodendrocytes. Also, matter volume that is seen in imaging studies [3,
damage to a particular subpopulation of vulnera- 26].
ble neurons plays a role in the development of By US, there are four stages of PVL that some-
PVL. These are called subplate neurons and they what correlate with its histological characteristics.
contribute to cortical development and in particu- First, there is congestion in the periventricular
lar to the formation of thalamocortical connec- white matter, which in US is seen as increased
tions. They form a transient cell population that echogenicity that usually adopts an elongated and
peaks at approximately 24 weeks (the onset of the globular morphology, sometimes referred as
developmental window of vulnerability) and later “flares.” This increased echogenicity usually is
undergoes apoptosis [25]. The subplate which can seen in the first 48 h. In the second stage, there is
reach up to four times the width of the cortical a relative return to normal which occurs mostly by
plate has been shown by MRI to be affected by the 2–4 weeks. In the third stage, the development
hypoxic injury. of cysts is evident in US at approximately 3–6
PVL is most commonly seen in the peritrigonal weeks (Fig. 6). Finally, in the fourth and last stage,
region and adjacent to the foramina of Monro [33, there is resolution of the cysts, with evidence of
37]. It can have a cavitary or non-cavitary presen- volume loss with enlargement of the lateral ven-
tation, this last type being more frequent. The tricles. This last stage happens at approximately
most commonly encountered neurological 6 months of age [27].
sequelae are motor and visual impairments US is usually used as the first examination in
because of the direct injury to the corticospinal evaluating suspected HII cases. Nevertheless, it
tracts and geniculocalcarine tracts that pass lacks the sensitivity and positive predictive value
through affected regions in the periventricular and, as mentioned before, the study can be normal
white matter [3, 26]. Spastic diplegia is also a in patients that develop PVL. Conversely, in other
common motor sequelae of PVL, in which the cases, US shows increased echogenicity in the
degree of motor impairment is greater in the periventricular areas of normal neonates. The
lower extremities, and occurs more frequently in presence of prolonged hyperechogenicity of the
preterm infants with PVL than in term infants periventricular white matter has a fairly low sen-
[3, 11]. At a histological level, PVL evolves first sitivity and positive predictive value for the detec-
with necrosis and cavitation that thereafter pro- tion of PVL [28]. Serial US examinations improve
gress to porencephalic cysts. Later, these cysts substantially the detection of transient cystic
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 477

Fig. 7 (a, b) T2WI of a preterm neonate who suffered dark fluid levels can be seen inside the lateral ventricles
mild-to-moderate asphyxia shows T2 hyperintensity in the compatible with intraventricular hemorrhage. Small left
periventricular white matter in the setting of PVL. Also, side periventricular cyst is present

lesions and can be better than MRI studies for this


purpose. This is important because it has prognos-
tic value as most of patients with cystic changes
present neurological sequelae [29]. For these rea-
sons, the primary roles of US are to detect germi-
nal matrix hemorrhages in the immediate
postnatal period and detect cystic changes later
in perinatal life [3].
MRI allows better visualization of the
periventricular white matter lesions and is a useful
complement to cranial US especially among
patients without cystic lesions. It also allows
better depiction of hemorrhages and/or white
matter volume loss which also has prognostic
value [29]. In MR images, early injury to the
white matter appears as foci of T1 hyperintensity
in the larger areas of T2 hyperintensity. These T1 Fig. 8 T2WI of a preterm neonate who suffered moderate
asphyxia demonstrates characteristic hyperintensity in the
hyperintense foci must be distinguished periventricular white matter, more pronounced at the level
from hemorrhages, and they do not produce T2 of the peritrigonal regions compatible with PVL. Also,
shortening. These T1 abnormalities may represent note the “wavy” appearance of the ventricular walls
focal areas of mineralization, while in the white
matter reactive gliosis develops [30]. These CT is usually avoided in neonates because of
changes are usually evident at the third–fourth the exposure to ionizing radiation. It does not
days post injury, and then they give way to a mild provide much more information than the US and
T2 shortening of the white matter at days 6–7. The MRI, but it could be important and helpful in
high T2 signal is most evident in the peritrigonal confirming PVL end-stage injuries later in life.
regions (Figs. 7 and 8). In the last stage of PVL, MRI and CT show a
478 M. Castillo and F. Chiang

characteristic loss of volume of the periventricular Table 2 HII in preterm neonates


white matter and centra semiovale with secondary Severe Injury in the deep gray matter, mostly
enlargement of the lateral ventricles in particular the thalami but also the basal ganglia,
their trigones. MRI and CT also show the charac- dorsal brainstem, cerebellum, and
corticospinal tracts as well as a
teristic irregular outline and wavy appearance of diminished volume of the cerebral
the outer wall of the lateral ventricles. MRI better hemispheric white matter
shows the loss of volume in the corpus callosum, Mild to Germinal matrix hemorrhage
particularly in the posterior aspect of the body and moderate IVH
splenium [31]. PVL

HII in the Term Neonate


These patients, in the first hours after a severe
As mentioned before, HII is also considered an insult, may present with depressed consciousness,
important cause of death, neurodevelopmental dis- periodic breathing or apnea, or bradycardia. In
orders, and disability in term neonates, although its cases where severe injury to cortical regions has
incidence and prevalence has declined over the last ensued, hypotonia and seizures may occur. In
decade and is now estimated to be between 2 and patients that survive, severe HII typically develops
4 per 1,000 live term births [3, 32, 33]. Risk factors including quadriparesis, choreoathetosis, severe
for HII can be divided in antepartum factors and seizure, and/or mental retardation. In cases of mod-
intrapartum factors. Antepartum risk factors erate HII, spastic diplegia or quadriplegia almost
include maternal hypotension, infertility treatment, always develops and is usually referred to as cere-
multiple gestation, prenatal infection, gestation bral palsy. On the other hand, in mild cases of HII,
41 weeks, and thyroid disease. Among the most term infants may develop mild developmental
important intrapartum factors are forceps delivery, delay or recover completely.
breech extraction, umbilical cord prolapse, The imaging patterns can be subdivided
abruptio placentae, tight nuchal cord, maternal depending on the severity of the hypoxic injury
fever, prolonged membrane rupture, abnormal into severe and partial asphyxia (Table 2).
cardiotocography, shoulder dystocia, and thick In term neonates, transfontanelle US is the first
meconium. The most popular hypothesis is that imaging study to be obtained when HII is
most of the HII cases are attributable only to suspected. Although some abnormalities can be
antepartum risk factors; however, there are new detected by US, it has a low sensitivity and there-
reports that point to the intrapartum factors as nec- fore a negative study should not be used as a
essary to develop this condition. In approximately definite evidence of an absence of hypoxic injury.
10 % of HII cases, there are postnatal complica- If there is strong clinical suspicion of HII and US is
tions such as sepsis, shock, and/or severe respira- negative, MR imaging should be obtained to eval-
tory distress [33–35]. uate the presence and severity of the injury. It is
The clinical manifestations at birth of HII in important to remember that, as mentioned before,
term infants include nonspecific signs and symp- the biochemical and histological features of HII
toms that evolve over a period of days. Data that influence the imaging findings vary with time
suggest that the infants at risk for severe HII can so that a study performed only hours after the event
be reliably identified by a group of clinical man- will be different from one done several days later.
ifestations that include evidence of intrapartum
distress (e.g., fetal heart rate abnormality), severe
functional depression (low 5 min Apgar score), Severe Asphyxia
need for resuscitation in the delivery room, severe
fetal acidemia, abnormal early neurologic exami- In term neonates, severe asphyxia results mainly
nation, and abnormal electroencephalogram. in a central pattern of injury that usually involves
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 479

Fig. 9 (a, b) DWI in a term neonate with severe asphyxia demonstrates diffusion restriction in the ventrolateral thalami
and peri-rolandic cortex

the deep gray matter including the putamina, ven- hyperechogenicity generally suggests a severe
trolateral thalami, hippocampi, dorsal brainstem, insult and poor outcome [40]. At a later stage,
and lateral geniculate nuclei. Occasionally, the the imaging pattern reflects the loss of volume
peri-rolandic cortex is also involved. The expla- including prominence of the ventricles and extra
nation for this pattern of injury is, as we men- axial CSF-containing spaces, likely due to atro-
tioned before, the active state of myelination of phy. Doppler US during the initial US examina-
these areas and the high concentration of NMDA tion may be useful and improves sensitivity and
receptors which makes them more susceptible to specificity by showing diminished resistive
neonatal HII [6, 36]. The rest of the cortex is indexes (<60) in the anterior and middle cerebral
usually spared or shows mild abnormalities since arteries. These lower resistive indexes have been
it is generally less metabolically active. However, also associated with poorer clinical outcome, even
if the injury is prolonged, the remaining cortex in absence of other US abnormalities [41].
will be injured and portrays a worse MRI is probably the most accurate modality to
prognosis [10]. assess neonatal HII especially when performed
Transfontanelle US, although it is the most with diffusion-weighted imaging (DWI) in the
commonly used technique and usually the first first 24 h, when DWI is most sensitive to detect
one in cases of suspected HII, is less sensitive injuries which may still not be visible in conven-
(about 50 % in the first week of life) and specific tional T1- and T2-weighted images. DWI shows
compared with CT and MRI and carries less high signal (with corresponding low ADC values)
interobserver agreement [3, 37, 38]. Its sensitivity in the ventrolateral thalami and basal ganglia (par-
increases when it is performed after 7 days. Early ticularly the posterior putamina), peri-rolandic
US findings include a generalized increase in regions, and along the corticospinal tracts
cerebral echogenicity and diffuse cerebral edema (Fig. 9). It is important to highlight the fact that
with obliteration of the cerebrospinal fluid (CSF) even with the high sensitivity of this technique,
containing spaces. Subtle increased echogenicity the findings in DWI in the first 24 h usually
in the basal ganglia, thalami, and brainstem can be underestimate the ultimate extent of the injury,
seen in the first week but are more apparent after and although rare, some reports of normal find-
7 days [38, 39]. The presence of thalamic ings in the first 24 h have been reported [17, 42].
480 M. Castillo and F. Chiang

Fig. 10 (a, b) T1WI showing hyperintensity in the posterior lentiform nuclei, ventrolateral thalami, and pericentral
cortex in a preterm neonate who suffered profound asphyxia

It is believed that the reason for this delay in


showing the full extent of the injury may be
based in the important role of apoptosis in HII,
and as explained before, the time that takes for
ATP to be depleted which precedes the death of
neurons and the resultant neurodegeneration at a
macroscopic level. Abnormalities on DWI peak at
3–5 days. By the end of the first week, the
hyperintensity in injured areas in DWI tends to
decrease, phenomenon known as “pseudo-nor-
malization” [4, 6, 10, 20]. It is important to realize
that although the images seemingly improve, this
does not imply that there is a real reversal or
improvement of the underlying injury, just reso-
lution of signal abnormalities on DWI. Because of
the rare possibility of a false-negative DWI study
Fig. 11 T1WI. A different patient showing T1
when performed in the first days, it is
hyperintensity in the posterior lentiform nuclei and ventro-
recommended to repeat the examination at 2–4 lateral thalami
days when the signal abnormality is expected to
be greatest or perform an evaluation with proton
MR spectroscopy (MRS). nuclei and ventrolateral thalami (Figs. 10 and 11).
It is well known that conventional MRI Sometimes, signal intensity changes may also be
sequences with T1- and T2-weighted images seen in the dorsal brainstem and basal ganglia
obtained within the first day are frequently normal [36]. The T2 hyperintensity usually develops
and therefore are less useful than DWI to diagnose later than the T1 shortening and usually by the
acute HII. By the second day, conventional second week, affects the thalami and posterior
sequences, especially T1-weighted images, start putamina. As mentioned before, cortical abnor-
to show hyperintensity in the posterior lentiform malities can also occur.
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 481

Fig. 12 (a, b) T2WI in a term neonate with chronic changes from HII shows hyperintensity in the corticospinal tracts,
putamina, and ventrolateral thalami. Also, some loss of volume is noted

The cause of the abnormalities seen with the


MRI conventional sequences in the basal ganglia
and thalami remains incompletely understood,
with possible explanations including hemorrhage,
transient calcium deposition, lipid release from
myelin breakdown, free fatty acids, and even
paramagnetic effects from free oxygen radicals.
In infants, cortical abnormalities are likely due to
laminar necrosis [36, 43]. A possible explanation
for the delay in appearance of the T2
hyperintensity changes is the high water content
of the white matter, so subtle abnormalities are
obscured and difficult to identify at first. The T1
shortening in posterior putamina, thalami, and
peri-rolandic cortex can persist for several
months. Because of all these reasons, DWI is
very useful in the first days, especially in the first Fig. 13 NECT in acute severe HII shows hypodensity of
24 h when conventional MR images are likely to the basal ganglia. CT is not usually the imaging modality of
be normal. On the other hand, conventional MRI choice in these patients
sequences are useful at the end of the first week
when the DWI images pseudo-normalize. Later,
in the chronic phase, the imaging findings reflect ganglia lesions includes HII and inborn errors of
atrophy of injured structures and T2 metabolism. The latter ones are suspected if there
hyperintensity especially in the ventrolateral thal- is no history of HII and if other imaging features
ami, posterior putamina, and corticospinal tracts outside of the typical spectrum of HII, like local-
(Figs. 12 and 13) [36]. The major imaging differ- ized white matter and cortical abnormalities, atro-
ential diagnosis in newborns with bilateral basal phy, or heterotopias, are seen.
482 M. Castillo and F. Chiang

Fig. 14 (a, b) DWI in a term neonate who suffered partial asphyxia demonstrates hyperintensity in watershed areas with
the corresponding low signal in the ADC map, compatible with watershed infarcts

Partial Asphyxia modality of choice when studying term infants


with suspected partial asphyxia. Regarding the
When partial asphyxia occurs, the pattern of MRI sequences that are more useful, again DWI
injury changes and as mentioned before, the neo- are the most sensitive and the first to show abnor-
natal brain is more resistant to hypoxia than the malities in the first 24 h. These abnormalities
adult brain. For this reason, in mild or moderate include hyperintensity with corresponding low
insults of short duration, there may be little or no ADC values (diffusion restriction) in the water-
injury [10]. Compensatory mechanisms that take shed territories (Fig. 14). Interpretation of DWI is
place when hypoxia is established have been well sometimes difficult because of the high content of
studied in animal models. With prolonged fetal water of the brain at this age which makes the
hypoxia, blood shunting to vital brain structures hyperintensity of HII subtle. To facilitate the
occurs, including the brainstem, basal ganglia, correct diagnosis, it is important to interpret
hippocampi, and cerebellum. Thus, less metabol- DWI with the corresponding ADC map
ically active regions of the brain receive less blood which will show areas of low signal confirming
and are more susceptible to injury, specifically the true diffusion restriction [17]. T1 and T2 images
cortex and white matter. This is the reason why in may be normal in the first 24 h, but by the
mild-to-moderate HII, the brainstem, cerebellum, second day, they show T2 hyperintensity in the
and deep gray matter are generally spared. When cortex and subcortical areas related to
the autoregulatory mechanisms are exceeded, the cortical swelling and loss of differentiation
result is injury to the watershed zones which between the gray and white matter contrast.
become relatively hypoperfused. The clinical These findings are more evident in watershed
manifestations of this process generally are sei- zones but occasionally can be appreciated in the
zures, hypotension, and possibly proximal hemispheres [3]. Deep gray matter structures will
extremity weakness and/or spasticity [10]. be most likely spared in these patients. In the
US diagnosis of this type of injury is difficult as chronic stage, there are signs of atrophy with
this technique has low sensitivity for examination loss of volume of the white matter and cortical
of the cortical and subcortical areas that are close thinning predominantly in the parasagittal water-
to the calvarium. For this reason, MRI is the shed zones (Table 3).
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 483

Table 3 HII in term neonates


Severe Injury of the deep gray nuclei
(putamina, ventrolateral thalami),
hippocampi, dorsal brainstem,
and lateral geniculate nuclei
Occasionally, the peri-rolandic
cortex
Partial or less Cortical watershed zones
severe asphyxia

HII in Postnatal Infants and Young


Children

In this group of patients, the most common causes


of HII are accidents such as drowning or choking
and non-accidental trauma. There are differences
Fig. 15 NECT of severe diffuse anoxic injury shows the
between the pattern of injury seen in neonates and “reversal sign.” Note the subtle decreased density of the
that in infants and young children. These differ- cortical gray matter and basal ganglia relative to the denser
ences occur mostly because of disparities in the white matter
brain maturation that advances rapidly during the
perinatal period. It has been estimated that the infants may be due to redistribution of blood flow
myelination process is completed by about from the anterior circulation to the posterior cir-
2 years of age, and around this period, the pattern culation after asphyxia [36]. Also, some differ-
of injury starts to resemble the one encountered in ences may be due to physiologic and
adults. biochemical changes that occur with maturation;
this results in changes in regional energy require-
ments and thus also in changes in the regional
Severe Asphyxia susceptibility to hypoxia. By the time the anterior
fontanelle closes (at approximately 4 months), US
Severe episodes of asphyxia in infants between cannot be used anymore and CT becomes the
1 and 2 years of age result in injuries to the study of choice. CT examinations that are done
caudate nuclei, putamina, lateral geniculate too early, before 24 h, can show only subtle
nuclei, hippocampi, and cerebral cortex. The ante- hypodensity in the deep gray matter structures or
rior frontal and parieto-occipital cortex will be be negative. Later, CT demonstrates diffuse basal
most affected with relative sparing of the peri- ganglia abnormalities along with diffuse cortical
rolandic cortex and thalami [36]. hypoattenuation with loss of gray-white matter
In patients who experience asphyxia after the differentiation and sulcal and cisternal efface-
immediate perinatal period, but before 1 year of ment, all of which are consequences of cerebral
age, findings are often a mixture between those of edema. The peri-rolandic cortex may be relatively
neonatal asphyxia and later infantile asphyxia and spared [13, 44, 45]. At 4–6 days, hemorrhagic
result in the involvement of the basal ganglia in infarcts may be evident in the basal ganglia. In
particular the posterior putamina and lateral thal- some patients, the “reversal sign” can be seen
ami and also in involvement of the dorsal mid- (Fig. 15). The reversal sign refers to a reversal in
brain and cortex. The reasons for these differences the normal CT attenuation patterns between gray
between neonates and older children are not and white matter probably due to the congestion
entirely understood, but it has been suggested of deep medullary veins secondary to obstruction
that the relative sparing of the thalami in older of venous outflow by cerebral edema and
484 M. Castillo and F. Chiang

Fig. 17 FLAIR image demonstrates affectation of the


Fig. 16 NECT demonstrating the “white cerebellum” basal ganglia with sparing of the thalami. The occipital
sign in a child with severe HII. Note the diffuse cortex is also slightly hyperintense
hypodensity of the cerebral parenchyma that makes the
cerebellum look denser
Table 4 HII in postnatal infants and young children
Severe 1–2 years: injury to the caudate nuclei,
subsequent compression of them. The other sign putamina, lateral geniculate nuclei,
in CT studies is the “white cerebellum sign” hippocampi, and cerebral cortex
(especially anterior frontal and parieto-
(Fig. 16), in which the cerebral hemispheres are occipital)
hypodense due to diffuse edema, making the cer- <1 year: mixture of the features of
ebellum and brainstem appear relatively neonatal asphyxia and later infantile
hyperdense. This finding may be related to blood asphyxia
flow redistribution to the posterior circulation that Mild to Cortical watershed zones
moderate
occurs during anoxia [44]. Both the “reversal
sign” and “white cerebellum sign” are associated
with a worst outcome [44–46]. of bilateral basal ganglia abnormalities is wide
In MRI studies, the abnormalities on DWI are and includes inborn errors of metabolism, hypo-
evident in the first 12–24 h. First, images show glycemia, osmotic myelinolysis, hemolytic ure-
high intensity in the posterolateral lentiform mic syndrome, toxic exposure, and infectious
nuclei, and if the thalami are involved, usually encephalitis.
the ventrolateral nuclei will be most affected. In
the next 48 h, there is progression and involve-
ment of the rest of the basal ganglia and cortex. Mild-to-Moderate Asphyxia
Conventional T1- and T2-weighted images are
usually normal during the first day and may In mild hypoxic insults to older infants, watershed
remain normal for 48 h [43]. After this, zone abnormalities in the cortex and subcortical
T2-weighted images show diffuse basal ganglia white matter are seen. White matter lesions may
and cortical hyperintensities with a relative spar- also be seen but are more common in younger
ing of the thalami and peri-rolandic cortex children (under 1 year of age) [7]. Relative spar-
(Fig. 17). These T2 hyperintensities are believed ring of the periventricular white matter is common
to represent edema [3]. The differential diagnosis [31] (Table 4).
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 485

HII in Older Children and Adults they are not able to generate energy during an
anoxic event. The Purkinje cells die along with
In older children, just as in younger ones, the most cells in the granular layer [13].
common causes of asphyxia are drowning and In older patients, the first imaging study
choking accidents. In adults on the other hand, performed for a suspected brain anoxic injury is
the causes of HII are different. The leading causes commonly a CT. In CT, the most common find-
for hypoxic adult events are related to chronic ings are diffuse hypoattenuation of the cortex and
diseases as cerebrovascular disease and/or cardiac basal ganglia, consistent with edema, effacement
arrest with secondary hypoxemia. In this group of of the CSF-containing spaces, and loss of gray-
patients, mild-to-moderate injury will manifest as white matter differentiation (Fig. 18). In older
watershed infarcts. More severe insults have del- patients, as in children, the “reversal sign” and
eterious effects in the cortical gray matter and “white cerebellum” sign may be present and also
deep gray structures. The cortex is usually dif- indicate a poor prognosis (Fig. 19) [3].
fusely affected predominantly in the peri-rolandic As in other age groups, DWI is the earliest to
and visual areas, and the cerebellum and hippo- show abnormalities, usually starting after a few
campi may also be affected. The pathophysiology hours after the injury. During the first 24 h,
of this gray matter injury pattern is probably hyperintensity in DWI is evident in the basal
related to the fact that the gray matter contains ganglia, cerebellar hemispheres (Fig. 20), and cere-
most of the postsynaptic glutamate receptors and bral cortex (especially in peri-rolandic and occipital
therefore is most susceptible to the excitotoxic areas), and the thalami, brains tem and basal
effects of glutamate. Also, gray matter is more ganglia may also be involved (Fig. 21) [7, 47, 48].
metabolically active than white matter. Abnormalities on T1- and T2-weighted images
Cerebellar injury is common in older patients. may be delayed in comparison with DWI. After
The reason for this is not completely understood, the first 24 h, T2 images begin to show abnormal-
but it is believed that the relative immaturity of the ities that consist of hyperintensity and swelling of
Purkinje cells in neonates has a protective. These gray matter structures that may persist until the
cells are particularly prone to ischemia because end of the second week (Figs. 22, 23 and 24).

Fig. 18 NECT in severe HII injury after a cardiac arrest shows total loss of gray-white matter differentiation with
hypoattenuation of the cortex and basal ganglia. There is also effacement of the CSF-containing spaces
486 M. Castillo and F. Chiang

DWI images usually pseudo-normalize by the end


of the first week. In the chronic stage, T2-weighted
images demonstrate some residual hyperintensity
in basal ganglia, and the T1 may show
hyperintensity in the affected cortex, representing
cortical laminar necrosis in addition to diffuse atro-
phy (Figs. 25 and 26) [48] (Table 5).

Delayed White Matter Injury

Delayed white matter injury, also known as


postanoxic leukoencephalopathy, is a rare compli-
cation of global hypoxia and takes place weeks
after the injury. It is an uncommon syndrome
Fig. 19 NECT showing the “white cerebellum sign.” The which may be seen in approximately 3 % of
cerebellum appears “white” when compared with the carbon monoxide intoxications. Patients usually
abnormal hypodense brain present with a period of relative clinical stability

Fig. 20 (a–d) DWI in


severe HII shows diffuse
diffusion restriction of the
cerebellar parenchyma with
the corresponding ADC
map hypointensity
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 487

Fig. 21 (a–f) DWI showing diffuse diffusion restriction throughout the cortex, caudate nuclei, and left lentiform nucleus
in a severe HII

or improvement, after which they develop acute


neurologic deterioration 2–3 weeks after the ini-
tial hypoxic event. Symptoms include delirium,
personality changes, intellectual impairment,
movement disorders, and/or seizures [49, 50].
This condition has a relatively good prognosis
with approximately 75 % of patients showing
near-complete or complete recovery in 6–12
months. In the rest of patients, there maybe resid-
ual dementia or rarely the condition may progress
to paresis, coma, or even death [3]. It is important
to identify patients with this condition recogniz-
ing their clinical and imaging features to be able to
provide them time to recover without abrupt with-
drawal of care [51].
MR is the study of choice in suspected delayed
white matter injury. DWI can fail to demonstrate
abnormalities when performed immediately after
Fig. 22 T2WI shows hyperintensity and swelling of the the insult, but when performed during the period
parieto-occipital cortex in HII
488 M. Castillo and F. Chiang

Fig. 23 (a, b) DWI in HII shows diffusion restriction in the cortex and basal ganglia, with the corresponding ADC map
hypointensity

Although this syndrome is typically a delayed


process, it can present as a progressive leukoence-
phalopathy not separable from the primary injury
and without an intervening lucid period. In these
cases, the abnormalities seen in DWI appear as
early as 2 days. In other cases, clinical findings
may not be apparent and MRI may be the first to
detect white matter injury. Clinical improvement
appears to be associated with improvement of the
signal intensity abnormalities in the white matter;
however, some residual hyperintense areas may
persist beyond 18 months [3, 53].
The pathophysiology of this delayed injury is
not known. It is believed that the findings repre-
sent a process similar to Wallerian degeneration
although apoptotic cell death may also play a role
and help explain the delay in the presentation of
the disease [47].

Role of Proton MR Spectroscopy


in the Evaluation of HII
Fig. 24 T2WI of the same patient shows diffuse cortical
and basal ganglia hyperintensity and swelling
Magnetic resonance spectroscopy (MRS) is a
of delayed neurologic decline demonstrate diffuse technique that allows the identification and quan-
confluent areas of restricted diffusion in the white tification of some metabolites in tissues. It differs
matter [3, 47–49, 52]. T2-weighted images also from the conventional MR imaging in that it pro-
show corresponding hyperintensities in these areas. vides physiological and pathophysiological data
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 489

Fig. 25 (a, b) FLAIR images in HII demonstrate hyperintensity in the temporoparietal cortex

Fig. 26 (a–c) T1WI show areas of hyperintense cortex representing cortical laminar necrosis

instead of anatomic information. MRS has life [55]. In HII, MRS shows increased lactate
become a valuable tool in the identification of levels which appear as a doublet at 1.3 ppm at
HII, especially in the perinatal period. Together 1.5 T in the deep gray matter, parieto-occipital
with DWI, they are the most sensitive modalities regions, or watershed zones by 2–8 h [17, 54,
for detecting HII in the acute period [3, 17, 54]. 56]. A glutamine–glutamate peak may be detected
Furthermore, MRS is very useful in the first 24 h, at 2.3 ppm [54, 55] and could reflect the glutamate
being more sensitive than the conventional release that occurs in HII (Fig. 27). Evidence
MR sequences and DWI in this period. MRS suggests that the elevation of lactate takes place
also has value in predicting the severity of injury in two different phases. A first elevation occurs
[10, 17, 42]. very early in the acute stage of the injury and is
The main alteration seen in MRS when HII probably due to hypoxemia and the secondary
ensues is elevation of lactate. Lactate is not nor- anaerobic glycolysis that takes place. Next, it
mally present or seen only as a very small peak in returns to normal as perfusion is restored. Then,
normal newborns and only the first few hours of a second peak occurs over the following 24–48 h
490 M. Castillo and F. Chiang

Table 5 HII in older children and adults findings at MRS in HII are reduced NAA, ele-
Severe The cortex usually diffusely affected, vated lactate, and elevated glutamine–glutamate.
deep gray structures (basal ganglia and These alterations in the spectra correlate with a
thalami), hippocampi, and cerebellum bad prognosis including persistent vegetative
Mild to Cortical watershed zones state or death [27]. Although HII cannot be
moderate
completely ruled out with normal examinations
until day 3–4, a normal MRS study and no abnor-
which is believed to be caused by a process known mal findings in conventional MRI sequences cor-
as “secondary energy failure” in which surviving relate with a good outcome [6].
neurons suffer delayed energy depletion most
probably due to mitochondrial failure. Injury
caused by this mechanism results in lactate eleva- Imaging Choices in HII
tion after 24 h and is associated with poor prog-
nosis [5, 6]; this partially explains the As HII characteristics are variable, highly depen-
underestimation of injury seen in DWI during dent on the time since onset and patient’s age
the first hours. (CNS maturity), many factors must be taken into
To correctly interpret the MRS studies in pre- account when choosing the type of imaging study.
term and term infants, it is important to realize that Some of the most important considerations are the
the spectra of premature babies differ significantly patient’s condition, age, availability of imaging
from the one seen in term infants and adults. This modalities, and concerns about ionizing radiation
is the result of the differences in the concentration exposure.
of metabolites in the developing brain which does Many neonates with suspected HII are in inten-
not become similar to the adult brain until 2 years sive care units and are hemodynamically unstable,
of age [55]. In newborns, it is normal to have a making the transportation difficult. In this regard,
lower N-acetylaspartate (NAA) and higher myo- performing cranial US may be best, taking in
inositol (Myo) and choline (Cho) levels compared consideration that it is fast and easy to perform
with older children and adults. In preterm babies, in a bedside setting, does not involve ionizing
a small peak of lactate is present and makes the radiation, and may provide useful information.
interpretation of MRS in suspected HII more dif- The major limitations of US are its operator
ficult. Lactate diminishes and NAA increases as dependence and its low sensitivity (especially in
the brain matures, but trace amounts of lactate some areas such as the convexities). When US
may be present even in term infants demonstrates HII, it is very helpful but if it is
[3, 55–57]. Because of this, it is very useful to negative, another study is required.
take into consideration the gestational age of the In neonates, the next study should be MRI. CT
infant at birth when interpreting the MRS to avoid is not recommended in these patients because it
false-positive interpretations. involves the use of ionizing radiation and is no
One useful tool is using the lactate–NAA ratio more sensitive than US due to the high water
in preterm neonates. The normal ratio in the thal- content of the neonatal brain [3]. The MR protocol
ami for normal control neonates (term and pre- should include at least DWI, ADC map, and
term) is around 0.25 [56]. In preterm and term T1–T2-weighted images. MR spectroscopy can
neonates with HII, lactate–NAA ratios are greater be performed in the acute stage, especially when
than 0.4, while in more severe injury, ratios are DWI is negative and a clinical suspicion for HII is
greater than 0.5. When ratios are above the 95 % high. In these situations, long echo time
confidence limits, there is a clear association with (135 msec) and short echo time (30 msec) MRS
major impairments or death at 1 year [56]. NAA is with voxels positioned in the basal ganglia and
usually normal in acute HII. If it descends in the centra semiovale are most useful. If the study is
subacute phase, it is also associated with a poor negative in the first 24 h, it should be repeated
neurologic outcome [5, 58]. In older children, the within 2–4 days. If subtle abnormalities are
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 491

Fig. 27 (a, b) MR I : Integral


spectroscopy in a term
neonate with HII. Short Cho
(top) and long (bottom) Ins dd1 I : 93.9
echo times show lactate a I : 72.5
doublet centered at 1.3 ppm
20
in the short echo time and an NAA
inverted lactate doublet in I : 140
the long echo time. There is
also elevated Cr
glutamine–glutamate (red 15 I : 59.0
arrow) in the short echo Cr2
I : 74.2
time and reduced NAA in
the long echo time
10 Lac
I : 72.4

ppm
4 3 2 1

I : Integral
Cho
b
I : 49.2
6

Cr
I : 32.3 NAA
4
I : 29.4

Lac
I : 11.5

ppm
3 2 1

present on MRI in the acute setting, follow-up In adults and infants with closed anterior fon-
imaging at the end of the first week is tanelles, unenhanced head CT is the initial exam-
recommended to define progression and overall ination of choice. If CT is positive, performing an
extent of injury [6]. MRI to assess the extent of the injury can be
492 M. Castillo and F. Chiang

considered. Currently, there is no clear role for – MRS and DWI are the most sensitive modali-
MRI perfusion imaging in this clinical setting. ties for detecting HII in the acute period.

Summary References

HII is a highly variable entity that can pose sig- 1. Dugan LL, Choi DW (1999). Hypoxia-ischemia and
brain infarction. In: Siegel GJ, Agranoff BW, Albers
nificant difficulties to the diagnosis from a clinical
RW et al (eds) Basic neurochemistry: molecular, cellu-
and neuroimaging standpoint especially in lar and medical aspects, 6th edn. Lippincott-Raven,
newborns. Philadelphia. Available from: http://www.ncbi.nlm.
Different patterns are observed in these nih.gov/books/NBK28046/
2. Armstrong-Wells J, Bernard TJ, Boada R, Manco-
patients and depend mainly on brain maturity,
Johnson M (2010) Neurocognitive outcomes following
severity and length of the injury, and type and neonatal encephalopathy. NeuroRehabilitation
timing of the imaging studies. A clear understand- 26(1):27–33. doi:10.3233/NRE-2010-0533
ing of the pathophysiological events related to the 3. Huang BY, Castillo M (2008) Hypoxic-ischemic brain
injury: imaging findings from birth to adulthood.
global hypoxic–ischemic injury may help in
Radiographics 28(2):417–439. doi:10.1148/rg.2820
understanding its different imaging patterns. The 75066; quiz 617
concept of selective vulnerability allows imagers 4. Khurshid F, Lee KS, McNamara P, Whyte H, Mak W
to focus their attention in specific areas that are (2011) Lessons learned during implementation of ther-
apeutic hypothermia for neonatal hypoxic ischemic
most likely injured. The most sensitive imaging
encephalopathy in a regional transport program in
modalities in the detection of HII in the Ontario. Paediatr Child Health 16(3):153–156
hyperacute stage are DWI and MR spectroscopy. 5. Barkovich AJ, Baranski K, Vigneron D et al (1999)
DWI can underestimate the extent of injury in Proton MR spectroscopy for the evaluation of brain
injury in asphyxiated, term neonates. AJNR Am J
very young children, so performing delayed imag-
Neuroradiol 20:1399–1405
ing is recommended to assess the extent of injuries 6. Grant PE, Yu D (2006) Acute injury to the immature
and shed light on the long-term prognosis of these brain with hypoxia with or without hypoperfusion.
patients. Radiol Clin North Am 44:63–77, viii
7. Christophe C, Fonteyne C, Ziereisen F et al (2002)
Value of MR imaging of the brain in children with
Key Points hypoxic coma. AJNR Am J Neuroradiol 23:716–723
– Imaging findings in HII are highly variable and 8. Wijdicks EF, Campeau NG, Miller GM (2001) MR
depend mainly on brain maturity, severity and imaging in comatose survivors of cardiac resuscitation.
AJNR Am J Neuroradiol 22:1561–1565
duration of the injury, and timing and imaging
9. Johnston MV, Trescher WH, Ishida A, Nakajima W
modality used to study these patients. (2001) Neurobiology of hypoxic-ischemic injury in the
– Conventional T1- and T2-weighted images are developing brain. Pediatr Res 49(6):735–741
most useful at the end of the first week when 10. Barkovich AJ (2005) Brain and spine injuries in
infancy and childhood. In: Barkovich AJ (ed) Pediatric
DWI abnormalities have pseudo-normalized.
neuroimaging, 4th edn. Lippincott Williams &
– DWI is useful in the first week. When used for Wilkins, Philadelphia, pp 190–290
the evaluation of the neonatal brain, it should 11. O’Shea TM (2002) Cerebral palsy in very preterm
be reviewed carefully in conjunction with infants: new epidemiological insights. Ment Retard
Dev Disabil Res Rev 8:135–145
ADC maps because true restriction may be
12. Barkovich AJ, Sargent SK (1995) Profound asphyxia
masked by intrinsic T2 hyperintensity of the in the premature infant: imaging findings. AJNR Am J
unmyelinated brain at this age. Neuroradiol 16:1837–1846
– In preterm neonates, profound ischemia pre- 13. Castillo M (2007) Selective vulnerability and the cere-
bellum in neonates. AJNR Am J Neuroradiol 28:20–21
sents with damage to the deep gray matter
14. Chugani HT, Phelps ME, Mazziotta JC (1987) Positron
and brainstem. Moderate-to-mild insults pre- emission tomography study of human brain functional
sent as germinal matrix hemorrhage, intraven- development. Ann Neurol 22:487–497
tricular hemorrhage, and/or periventricular 15. Hasegawa M, Houdou S, Mito T, Takashima S,
Asanuma K, Ohno T (1992) Development of
white matter damage (PVL).
22 Hypoxic–Ischemic Encephalopathy (Preterm, Term, and Adult) 493

myelination in the human fetal and infant cerebrum: a histopathologic findings of the brain in extremely sick
myelin basic protein immunohistochemical study. preterm infants. AJNR Am J Neuroradiol
Brain Dev 14:16 20:1349–1357
16. Whyte HE, Blaser S (2013) Limitations of routine 31. Barkovich AJ, Truwit CL (1990) Brain damage from
neuroimaging in predicting outcomes of preterm perinatal asphyxia: correlation of MR findings with
infants. Neuroradiology 55(Suppl 2):3–11. gestational age. AJNR Am J Neuroradiol
doi:10.1007/s00234-013-1238-6 11:1087–1096
17. Barkovich AJ, Westmark K, Partridge C, Sola A, 32. Vannucci RC, Perlman JM (1997) Interventions for
Ferriero DM (1995) Perinatal asphyxia: MR findings perinatal hypoxic-ischemic encephalopathy. Pediatrics
in the first 10 days. AJNR Am J Neuroradiol 100:1004–1014
16:427–438 33. Martinez-Biarge M, Diez-Sebastian J, Wusthoff CJ,
18. Volpe JJ (1989) Intraventricular hemorrhage in the Mercuri E, Cowan FM (2013) Antepartum and
premature infant: current concepts – I. Ann Neurol intrapartum factors preceding neonatal hypoxic-
25:3–11 ischemic encephalopathy. Pediatrics 132(4):
19. Paneth N, Pinto-Martin J, Gardiner J et al (1993) Inci- e952–e959. doi:10.1542/peds.2013-0511
dence and timing of germinal matrix/intraventricular 34. Ferriero DM (2004) Neonatal brain injury. N Engl J
hemorrhage in low birth weight infants. Am J Med 351:1985–1995
Epidemiol 137:1167–1176 35. Nelson KB (2002) The epidemiology of cerebral palsy
20. Merrill JD, Piecuch RE, Fell SC, Barkovich AJ, Gold- in term infants. Ment Retard Dev Disabil Res Rev
stein RB (1998) A new pattern of cerebellar hemor- 8:146–150
rhages in preterm infants. Pediatrics 102:E62 36. Barkovich AJ (1992) MR and CT evaluation of pro-
21. Correa F, Enríquez G, Rossello J, Lucaya J et al (2004) found neonatal and infantile asphyxia. AJNR Am J
Posterior Fontanelle Sonography: An Acoustic Win- Neuroradiol 13:959–972
dow into the Neonatal Brain. AJNR Am J Neuroradiol 37. Blankenberg F, Loh N, Bracci P, D’Arceuil H
25:1274–1282 et al (2000) Sonography, CT, and MR imaging: a
22. Ballabh P, Braun A, Nedergaard M (2004) Anatomic prospective comparison of neonates with suspected
analysis of blood vessels in germinal matrix, cerebral intracranial ischemia and hemorrhage. AJNR Am J
cortex, and white matter in developing infants. Pediatr Neuroradiol 21:213–218
Res 56:117–124 38. Babcock DS, Ball W Jr (1983) Postasphyxial enceph-
23. Back SA (2006) Perinatal white matter injury: the alopathy in full-term infants: US diagnosis. Radiology
changing spectrum of pathology and emerging insights 148:417–423
into pathogenetic mechanisms. Ment Retard Dev 39. Hertzberg BS, Pasto ME, Needleman L, Kurtz AB,
Disabil Res Rev 12(2):129–140 Rifkin MD (1987) Postasphyxial encephalopathy in
24. Back SA, Luo NL, Borenstein NS, Levine JM, Volpe term infants: sonographic demonstration of increased
JJ, Kinney HC (2001) Late oligodendrocyte progeni- echogenicity of the thalamus and basal ganglia. J Ultra-
tors coincide with the developmental window of vul- sound Med 6:197–202
nerability for human perinatal white matter injury. 40. Connolly B, Kelehan P, O’Brien N et al (1994) The
J Neurosci 21:1302–1312 echogenic thalamus in hypoxic ischaemic encephalop-
25. Ferriero D, Miller S (2010) Imaging selective vulnera- athy. Pediatr Radiol 24:268–271
bility in the developing nervous system. J Anat 41. Stark JE, Seibert JJ (1994) Cerebral artery Doppler
217:429–435. doi:10.1111/j.1469-7580.2010.01226.x ultrasonography for prediction of outcome after peri-
26. Flodmark O, Lupton B, Li D et al (1989) MR imaging natal asphyxia. J Ultrasound Med 13:595–600
of periventricular leukomalacia in childhood. AJR Am 42. Robertson RL, Ben-Sira L, Barnes PD et al (1999) MR
J Roentgenol 152:583–590 line-scan diffusion-weighted imaging of term neonates
27. Dubowitz LM, Bydder GM, Mushin J (1985) Devel- with perinatal brain ischemia. AJNR Am J Neuroradiol
opmental sequence of periventricular leukomalacia: 20:1658–1660
correlation of US, clinical, and nuclear magnetic reso- 43. Dubowitz DJ, Bluml S, Arcinue E, Dietrich RB (1998)
nance functions. Arch Dis Child 60:349–355 MR of hypoxic encephalopathy in children after near
28. Inder TE, Anderson NJ, Spencer C, Wells S, Volpe JJ drowning: correlation with quantitative proton MR
(2003) White matter injury in the premature infant: a spectroscopy and clinical outcome. AJNR Am J
comparison between serial cranial sonographic and Neuroradiol 19:1617–1627
MR findings at term. AJNR Am J Neuroradiol 44. Harwood-Nash DC (1992) Abuse to the pediatric cen-
24:805–809 tral nervous system. AJNR Am J Neuroradiol
29. Murgo S, Avni EF, David P, Muller MF, Golzarian J, 13:569–575
Balériaux D, Struyven J (1999) Periventricular 45. Bird CR, Drayer BP, Gilles FH (1989) Pathophysiol-
leukomalacia in premature infants: prognostic role of ogy of “reverse” edema in global cerebral ischemia.
ultrasonography and MRI. J Radiol 80(7):715–720 AJNR Am J Neuroradiol 10:95–98
30. Felderhoff-Mueser U, Rutherford MA, Squier WV 46. Han BK, Towbin RB, De Courten-Myers G,
et al (1999) Relationship between MR imaging and McLaurin R, Ball WS Jr (1989) Reversal sign on CT:
494 M. Castillo and F. Chiang

effect of anoxic/ischemic cerebral injury in children. resonance images in patients with delayed encephalop-
AJNR Am J Neuroradiol 10:1191–1198 athy after acute carbon monoxide poisoning. Psychia-
47. Arbelaez A, Castillo M, Mukherji S (1999) Diffusion try Clin Neurosci 51:421–423
weighted MR imaging of global cerebral anoxia. AJNR 54. Hanrahan JD, Sargentoni J, Azzopardi D et al (1996)
Am J Neuroradiol 20:999–1007 Cerebral metabolism within 18 hours of birth asphyxia:
48. Takahashi S, Higano S, Ishii K et al (1993) Hypoxic a proton magnetic resonance spectroscopy study.
brain damage: cortical laminar necrosis and delayed Pediatr Res 39:584–590
changes in white matter at sequential MR imaging. 55. Bertholdo D, Watcharakorn A, Castillo M (2013) Brain
Radiology 189:449–456 proton magnetic resonance spectroscopy: introduction
49. Roychowdhury S, Maldjian JA, Galetta SL, Grossman and overview. Neuroimaging Clin N Am
RI (1998) Postanoxic encephalopathy: diffusion MR 23(3):359–380. doi:10.1016/j.nic.2012.10.002
findings. J Comput Assist Tomogr 22:992–994 56. Penrice J, Cady EB, Lorek A et al (1996) Proton
50. Bass E (1985) Cardiopulmonary arrest: pathophysiol- magnetic resonance spectroscopy of the brain in nor-
ogy and neurologic complications. Ann Intern Med mal preterm and term infants, and early changes after
103:920–927 perinatal hypoxia-ischemia. Pediatr Res 40:6–14
51. Salazar R, Dubow J (2012) Delayed posthypoxic 57. Leth H, Toft PB, Pryds O, Peitersen B, Lou HC,
leukoencephalopathy following a morphine overdose. Henriksen O (1995) Brain lactate in preterm
J Clin Neurosci 19(7):1060–1062. doi:10.1016/j. and growth retarded neonates. Acta Paediatr
jocn.2012.01.001 84:495–499
52. Kim JH, Chang KH, Song IC et al (2003) Delayed 58. Groenendaal F, Veenhoven RH, van der Grond J, Jan-
encephalopathy of acute carbon monoxide intoxica- sen GH, Witkamp TD, De Vries LS (1994) Cerebral
tion: diffusivity of cerebral white matter lesions. lactate and N-acetyl-aspartate/choline ratios in asphyx-
AJNR Am J Neuroradiol 24:1592–1597 iated full-term neonates demonstrated in vivo using
53. Inagaki T, Ishino H, Seno H, Umegae N, Aoyama T proton magnetic resonance spectroscopy. Pediatr Res
(1997) A long-term follow-up study of serial magnetic 35:148–151
Part V
Aneurysms
Aneurysmal Subarachnoid
Hemorrhage 23
Frédéric Clarençon, Nader-Antoine Sourour, Vincent Degos,
Aurélien Nouet, Federico Di Maria, Eimad Shotar,
Joseph Gabrieli, Lise Le Jean, and Jacques Chiras

Contents Surgical Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 506


Conservative Management . . . . . . . . . . . . . . . . . . . . . . . . . . 508
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 498
Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 508
Intracranial Aneurysms: Epidemiology . . . . . . . . . . 498 SAH-Related Complications . . . . . . . . . . . . . . . . . . . . . . . . 508
Clinical Symptoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499 Iatrogenic Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . 512

Diagnostic Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 501 Prognostic Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 513


Diagnosis of SAH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 501 Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 515
Imaging Modalities for the Depiction of IC
Aneurysms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 502 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
DSA-Negative SAH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 503 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 504
General Considerations on Ruptured Intracranial
Aneurysms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 504
Endovascular Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 504

F. Clarençon (*) • N.-A. Sourour • F. Di Maria •


E. Shotar • J. Gabrieli • J. Chiras
Department of Interventional Neuroradiology,
Pitié-Salpêtrière Hospital, Paris VI University, Paris,
France
e-mail: fredclare5@msn.com; nsourour@gmail.com;
federico.dimaria@gmail.com; eimad.shotar@gmail.com;
wjosephw@libero.it; jacques.chiras@psl.aphp.fr
V. Degos • L. Le Jean
Department of Anesthesia and Critical Care, Neuro-
intensive Care Unit, Pitié- Salpêtrière Hospital, Paris VI
University, Paris, France
e-mail: degosv@gmail.com; lise.lejean@psl.aphp.fr
A. Nouet
Department of Neurosurgery, Pitié-Salpêtrière Hospital,
Paris VI University, Paris, France
e-mail: aurelien.nouet@psl.aphp.fr

# Springer Science+Business Media New York 2016 497


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_19
498 F. Clarençon et al.

Abstract
Intracranial Aneurysms: Epidemiology
Intracranial aneurysms are a non-exceptional
Intracranial (IC) aneurysms are non-exceptional
disease affecting 1–3 % of the general popula-
disease that, according to angiographic and
tion. The incidence of aneurysmal subarach-
autopsy studies, affects, in the Western countries,
noid hemorrhage (SAH) is about 1/10,000/
about 1–3 % of the population [1, 2]. A higher
year. Aneurysmal SAH is a devastating disease
prevalence of IC aneurysms has been reported in
leading to death approximately in 50 % of the
Norway and Japan [3, 4], suggesting genetic
cases and to neurological sequels in about 25 %
predisposing factors for this disease [5]. The IC
of the cases. Management of aneurysmal SAH
aneurysms’ rupture incidence has been evaluated
consists in early exclusion of the aneurysm’s
at 1/10 000/year [3, 6–9].
sac either by endovascular means or by micro-
The cumulative individual rupture risk per year
surgical clipping. Intensive care management
is difficult to calculate and has been estimated
is of tremendous importance to treat
around 0.5–1 %/year [4]. Most of the IC aneu-
SAH-related complications like hydrocephalus
rysms are located within the anterior circulation
and to prevent delayed complications such as
(85 %) [10]. Anterior communicating artery
vasospasm. Current perspectives for intracra-
(AComm) aneurysms account for 30 % of IC
nial aneurysms management consist in a better
aneurysms, posterior communicating artery
selection of the unruptured aneurysms to treat,
(PComm) aneurysms 25 %, middle cerebral artery
in order to propose a preventive treatment of
(MCA) aneurysms 20 %, internal carotid artery
exclusion before they bleed.
(ICA) termination 7.5 %, pericallosal artery
(PA) 4.5 %, basilar artery (BA) 7–10 %, and
Keywords
posterior inferior cerebellar artery 3–4 %
Subarachnoid hemorrhage • Intracranial aneu-
[11]. Saccular bifurcation aneurysms account for
rysm • Rupture • Coiling • Clipping • Hydro-
more than 90 % of IC aneurysms, the
cephalus • Arterial vasospasm
remaining being sidewall or dissecting aneu-
rysms [12]. Intracranial aneurysms have a female
predominance. Indeed, females have a relative
Introduction
risk of 1.24 for IC aneurysms, compared to
males [3]. Some genetic diseases may also be
Subarachnoid hemorrhage (SAH) consists in blood
associated with arterial wall abnormalities that
effusion in the subarachnoid space. The most fre-
may promote the formation of IC aneurysms.
quent cause of SAH is head trauma. In case of
Among them, the most frequently encountered is
spontaneous SAH, intracranial aneurysm is the
the polycystic kidney disease (PCKD) [13, 14].
leading cause and therefore should be carefully
In this specific population, the relative risk to
looked for with appropriate imaging modalities.
have intracranial aneurysm is 4.4-fold higher
Due to the high morbidity and mortality rate of
than in the general population [15]. Type IV
aneurysmal subarachnoid hemorrhage, early aneu-
Ehlers-Danlos disease (anomaly of type III colla-
rysm sac exclusion should be performed either by
gen) [16], Marfan disease (anomaly of fibrillin)
microsurgical technique or by endovascular means.
[17], and alpha-1 antitrypsin deficit [18] are other
Specific SAH-related complications that may occur
diseases of the elastic tissue that may be associ-
within the following days after the hemorrhage
ated with higher incidence of IC aneurysms.
require hospitalization in intensive care unit and
Familial forms of IC aneurysms are rare; com-
adapted management. We present an overview of
pared to incidental aneurysms, their size is fre-
the intracranial aneurysms’ epidemiology, of the
quently over 10 mm, and they are often multiple,
clinical and radiological signs suggesting an aneu-
located on the MCA, and prone to rupture earlier
rysmal SAH, and of the therapeutic management of
[19, 20].
this severe disease.
23 Aneurysmal Subarachnoid Hemorrhage 499

Clinical Symptoms presented an aneurysmal SAH will die from the


rupture or from the rupture-related complications
Most of the IC aneurysms are asymptomatic and [21]. Among the survivors, half will present
are discovered incidentally during imaging work- sequels from the SAH at long-term follow-up
up. The most severe and feared complication of IC [22]. Clinically, SAH is revealed by a typical
aneurysms is the rupture, leading to SAH thunderclap headache and is associated with clin-
(Fig. 1a). Indeed, about 50 % of the patients who ical signs of meningeal irritation (neck stiffness,

Fig. 1 Different patterns of intracranial hemorrhage fissure, close to the ruptured aneurysm (arrow). (b)
related to aneurysmal rupture. (a) Typical subarachnoid Intraparenchymal hemorrhage related to a right MCA
hemorrhage in a patient with an AComm aneurysm. Spon- aneurysm rupture. (c) Acute subdural hematoma related
taneous hyperdensities are seen within the cortical sulci. to a calcified left MCA aneurysm rupture
Note a thickener amount of blood in the interhemispheric
500 F. Clarençon et al.

photophobia, phonophobia). In case of massive sometimes be responsible for intraparenchymal


SAH, focal neurological deficit may be seen. hemorrhage [23] (Fig. 1c) or even subdural hema-
Finally, SAH may lead to comatose state (Tables 1 toma [24] (Fig. 1b). Rarely, the rupture may pre-
and 2). Although the great majority of IC aneu- sent as an acute and severe headache without
rysms’ rupture leads to SAH, this rupture may blood leakage within the subarachnoid space.
This entity corresponds to a “pre-rupture” status
and is described as a “cracking syndrome”
Table 1 World Federation of Neurological Surgeons
(WFNS) scale [25]. PComm aneurysms associated with sudden
headache and cranial nerve (CN) III palsy should
Grade Glasgow Coma Scale Motor deficit
be considered as ruptured aneurysms and treated
1 15 Absent
in emergency (Fig. 2).
2 13–14 Absent
3 13–14 Present
In case of large or giant aneurysms, especially
4 7–12 Present or absent on the carotid siphon, IC aneurysms may be
5 3–6 Present or absent revealed by progressive CN palsy (oculomotor
palsy or trigeminal neuralgia) [26]. These symp-
toms are related to the mass effect and the
edema that may compress/irritate the CNs of the
cavernous sinus (Fig. 3) [27]. ICA giant/large
Table 2 Hunt and Hess scale
aneurysms may also be revealed by epistaxis
I Asymptomatic or minimal headache and slight
when an erosion of the wall of the sphenoid
nuchal rigidity
II Moderate to severe headache, nuchal rigidity, no
sinus is present – the aneurysm presenting a risk
neurological deficit (except cranial nerve palsy) of rupture in the nasal cavities, leading to a poten-
III Drowsiness, confusion, or mild focal deficit tially cataclysmic and life-threatening
IV Stupor, moderate or severe hemiparesis, possible nosebleeding [28]. Finally, headache may reveal
early decerebrate rigidity, and vegetative intracranial aneurysms. However, the relationship
disturbance between the pain and the aneurysm is often diffi-
V Deep coma, decerebrate rigidity, moribund
cult to establish.

Fig. 2 A 59-year-old female presenting with severe head- after embolization showing a satisfactory occlusion of the
ache and acute CN III palsy. (a) Left ICA digital subtrac- aneurysm’s sac and the patency of the PComm. The CN III
tion angiography in lateral projection showing a 17 mm palsy completely recovered 5 weeks after the symptoms’
PComm aneurysm. (b) Control DSA in lateral projection onset
23 Aneurysmal Subarachnoid Hemorrhage 501

Fig. 3 Giant left cavernous ICA aneurysm revealed by a gaze and (b) left position gaze. Abduction deficit of the left
CN VI palsy in a 68-year-old female. (a) and (b) photo- eye is seen in left position gaze. (c) Left ICA DSA in AP
graphs showing the left CN VI palsy: (a) right position projection showing the giant cavernous ICA aneurysm

that of CT scan. The best sequence to depict


Diagnostic Imaging SAH is the T2*WI. SAH will appear as a
hypointense signal within the cortical sulci
Diagnosis of SAH (Fig. 4a). The interest of this sequence is also to
depict SAH at the late phase [31]. Fluid-
Unenhanced computed tomography (CT) is the attenuated inversion recovery (FLAIR) sequence
best imaging option at the acute phase of the is also helpful do depict SAH showing a
SAH. Indeed, the sensitivity of CT scan to detect hyperintense signal within the cortical sulci
SAH is over 98 % within the first 48 h following (Fig. 4b). However, even if FLAIR-WI may be
the hemorrhage [29]. It appears with a typical positive for SAH at the acute phase whereas CT
pattern of spontaneous hyperdensity in the sub- scan is negative, this sequence is not the panacea
arachnoid space that insinuates inside the cortical for SAH diagnosis. Indeed, the FLAIR-WI will be
sulci. In case of massive hemorrhage, intraven- positive in case of negative CT scan in only less
tricular inundation may also be seen. It is note- than 20 % of the cases [32]. A recent study has
worthy that the thickest amount of blood may nonetheless showed that an MRI protocol com-
orientate to the location of the aneurysm. Finally, bining FLAIR-WI and susceptibility-WI (SWI)
CT scan may also help to depict associated hydro- had a high sensitivity for the depiction of
cephalus. However, this sensitivity of CT for SAH SAH [33].
progressively decreases with time (58 % at 5 days, Despite the high sensitivity of these various
50 % at 1 week) [30]. Brain magnetic resonance imaging techniques, the gold standard exam for
imaging (MRI) may increase the sensitivity of CT the positive diagnosis of SAH remains the lumbar
scan for the detection of SAH, especially few days puncture (LP) [34, 35].
after the rupture. Indeed, LP will find incoagulable blood in the
MRI is rarely performed in suspected SAH. CSF and products of hemoglobin breakdown,
Indeed, the accessibility of MRI is far lower than confirming the diagnosis of SAH [36].
502 F. Clarençon et al.

Fig. 4 Brain MRI in a 69-year-old female presenting with the right lateral ventricle (white arrow head). (b) Axial
an aneurysmal subarachnoid hemorrhage. (a) Axial fluid-attenuated inversion recovery-WI showing
T2*WI showing hypointense signal within the subarach- hyperintense signal in the subarachnoid space of both
noid space (black arrows) corresponding to subarachnoid sylvian fissures (black arrows). (c) MR angiography; 3D
hemorrhage and hypointense signal in the occipital horn of TOF showing an AComm aneurysm (arrow)

Finally, even if aneurysmal rupture is the most Imaging Modalities for the Depiction
frequent cause of spontaneous SAH, one should of IC Aneurysms
keep in mind that the most frequent cause of SAH
is trauma. Other causes of SAH should not be The aim of vascular diagnostic imaging is to
missed when the presence of an aneurysm has depict the IC aneurysm responsible for the SAH.
been ruled out, like cerebral vein thrombosis, The presence of other IC aneurysms on the circle
amyloid angiopathy, or rupture of an intracranial of Willis should always be looked for, since IC
vascular malformation [37]. It is noteworthy that aneurysms are multiple in more than 25 % of the
in case of brain arteriovenous malformation cases. Additionally, cerebral vasospasm should be
(bAVM) rupture, the SAH is often associated assessed on vascular imaging, especially for
with an intraparenchymal hemorrhage [38]. patients admitted few days after the SAH. Finally,
23 Aneurysmal Subarachnoid Hemorrhage 503

Fig. 5 Comparison of 3D time-of-flight (TOF) MR angi- bifurcation MCA aneurysm. Note that on DSA, the small
ography (a) and digital subtraction angiography (DSA) (b) branches close to the aneurysm are more precisely seen
in left oblique anterior projection for visualization of a left than on 3D TOF MRA

differential diagnosis, like brain AVM or cerebral angiography developed in the early 2000s, by
vein thrombosis, will be searched on vascular providing a manipulable volume, increased dra-
imaging if no IC aneurysm is found. matically the understanding of IC aneurysms’
CT angiography (CTA) is a valuable tool for the anatomy and is very helpful to tailor their treat-
detection of IC aneurysms [39–41]. This exam can ment [45]. Thus, DSA is the best imaging modal-
be performed just after the CT scan that authenti- ity to depict very small aneurysms and to see
cated the SAH. It requires an iodinated contrast accurately and precisely small branches originat-
media injection via a venous access (most of time ing from the neck or from the parent artery close to
antebrachial). Its sensitivity for the detection of IC the aneurysm (Fig. 5). Except in case of extreme
aneurysms is about 95 % [39]. However, very emergency (e.g., ruptured MCA aneurysm with
small aneurysms (<3 mm) and aneurysms close clinically non-tolerated hematoma requiring sur-
to the skull base may be missed by CTA [42]. It is gical evacuation), DSA should always be
noteworthy that the continuous improvement of performed before treatment (either endovascular
CT technologies increases the sensitivity of CTA or surgical) of an IC aneurysm. Indeed, DSA helps
for the depiction of IC aneurysms [43]. The other to clearly demonstrate the shape, size, neck, and
limitations for CTA are due to the fact that it relationships of the aneurysm with the parent
delivers X-rays, which limits its use for pregnant artery. DSA should always be performed via the
women or children, and due to the potential risk of four supra-aortic arterial vessels (i.e., both ICAs
allergy to the iodinated contrast media. and both vertebral arteries) in order not to miss
Three-dimensional (3D) time-of-flight (TOF) multiple aneurysms on the circle of Willis.
magnetic resonance angiography (MRA) has a
lower spatial resolution than CTA. However, due to
the fact that it does not require contrast media injec- DSA-Negative SAH
tion, this sequence may be interesting for patients
with renal insufficiency or in pregnant patients [44]. DSA-negative SAHs are defined as proven SAHs
Finally, digital subtraction angiography (DSA) with negative initial DSA for the diagnosis of IC
is the gold standard exam for the visualization of aneurysm or other IC vascular malformations [46].
IC aneurysms. Indeed, its spatial resolution is very The frequency of such SAH is as high as about
high: 200 μm. Moreover, 3D rotational 20 % of the cases [47]. This subgroup of SAH is
504 F. Clarençon et al.

divided into perimesencephalic and Selective Coiling


non-perimesencephalic (i.e., diffuse, cortical, or Since they have been developed by Guglielmi
xanthochromia only) SAHs. In perimesencephalic et al. in the early 1990s [51], detachable coils
SAH overall diagnostic yield of repeat angiogra- have revolutionized the treatment of IC aneu-
phy is 0 %, while it reaches 12.5 % in rysms. Most of these coils are made of platinum,
non-perimesencephalic ones [48]. Even if and all of them are detachable. They are deployed
DSA-negative SAHs have a better prognosis than inside the aneurysm via a microcatheter posi-
aSAH, and despite the very low rate of rebleeding, tioned in the sac and detached from a stainless-
in such SAH delayed complications may be steel microguidewire by an electrical current
observed like hydrocephalus or arterial (Fig. 6). The objective is to fill the aneurysm by
vasospasm [47]. means of decreasing size coils until total angio-
graphic occlusion, while preserving the patency of
the parent artery and other adjacent branches.
Treatment Even if safe and efficient for the treatment of
most IC aneurysms, large-necked aneurysms
General Considerations on Ruptured may be difficult to treat by regular coiling due to
Intracranial Aneurysms the risk of coil protrusion in the parent artery that
may be responsible for thromboembolic
The first-line treatment for ruptured IC aneu- complication.
rysms, since the publication of the ISAT study
[49], became the endovascular one. Indeed, “Remodeling” Technique
when eligible for both treatments, a patient with The remodeling technique has been developed in
ruptured aneurysm is less likely to have poor the mid-1990s to overcome the limitation of the
clinical outcome (death or severe disability) at presence of a large neck for the endovascular
1-year follow-up when treated by endovascular treatment of IC aneurysms [52]. The principle of
means than by surgical clipping (23.7 % vs. this technique consists in inflating temporarily a
30.6 %). However, the treatment option is largely balloon in front of the aneurysm’s neck during the
dependent on the experience of the operator of coil deployment. This balloon helps to constrain
each specialty in a given center. Moreover, neuro- the positioning of the coil’s loops in the aneu-
surgical clipping has still some consensual rysm’s sac during its delivery (Fig. 7). With this
indications like very large-necked aneurysms technique, almost all IC aneurysms are now eligi-
or aneurysms associated with clinically ble for endovascular treatment. Despite some con-
non-tolerated parenchymal hematoma, for which troversies [53], there are no evidences that the
hematoma evacuation and aneurysm clipping will balloon remodeling technique increases the
be performed during the same treatment. thromboembolic complication rate [54, 55].

Parent Artery Occlusion


Endovascular Treatment Parent artery occlusion is a potential therapeutic
option for ruptured aneurysms, but can only be
The aim of the endovascular treatment is to performed in case of sufficient collaterals,
“secure” the aneurysm sac by avoiding especially for large parent artery [26]. Moreover,
blood circulation in the sac. The treatment the potential risks of delayed vasospasm that
should be performed in emergency due to the can lead to ischemic complications in patients
potential risk of acute rebleeding of the ruptured with ruptured IC aneurysms may prevent from
IC aneurysm within the first days following the occluding a large vessel to preserve the circle of
SAH [50]. Willis.
23 Aneurysmal Subarachnoid Hemorrhage 505

Fig. 6 Regular coiling in an AComm ruptured aneurysm. aneurysm. (c) Unsubtracted snapshot during the aneurysm
(a) and (b) right ICA DSA: (a) 3D rotational angiography, sac coiling. (d) Post-coiling DSA in working projection
(b) working projection showing the saccular AComm showing the complete occlusion of the aneurysm

Treatment with Biological Glue artery harboring the dissecting aneurysm


Biological glue (n-butyl-cyanoacrylate [NBCA]) [56]. The other main indication for the use
may be used in ruptured IC aneurysms for of glue in aSAH is the presence of
parent artery occlusion in dissecting aneurysms infectious IC aneurysms (aka mycotic aneu-
(e.g., posterior inferior cerebellar artery [PICA] rysms). As for dissecting IC aneurysms, the
dissecting aneurysm). The aim of this aim of the treatment is to obtain a segmental
treatment is to perform a segmental occlusion by occlusion of the vessel harboring the
means of glue of the portion of the parent aneurysm [57].
506 F. Clarençon et al.

Fig. 7 Ruptured basilar tip aneurysm treated by during the coiling under balloon protection (remodeling
remodeling technique in a 49-year-old female. (a) Left technique). Note the positioning of the balloon in front of
vertebral artery DSA showing the basilar tip aneurysm the aneurysm’s neck to constrain the coils in the aneu-
with a relatively large neck (arrow). (b) 3D rotational rysm’s sac (arrow). (d) Final DSA control showing the
angiography reconstruction showing more precisely the total angiographic exclusion of the aneurysm
anatomy of the aneurysm. (c) Snapshot of the road map

Intracranial Stenting blister-like aneurysms, the use of flow diverter


The use of intracranial stents is not recommended stents may be a valuable option, either at the
for the treatment of ruptured IC aneurysms at the acute phase or few days after the SAH
acute phase. Indeed, these devices require a dual (Fig. 8) [59].
anti-aggregation platelet therapy that may be
responsible for hemorrhagic complications. How-
ever, in some selected cases, IC stents may be
used for the ruptured IC aneurysms. Indeed, in Surgical Treatment
case of coil protrusion in the parent artery, the
deployment of a stent in front of the aneurysm’s The aim of the surgical treatment is the same as
neck may be on option to avoid thromboembolic that for the endovascular one: to avoid aneurysm
complication. This technique is called the “bailout sac circulation. The main technique used is the
technique” [58]. In case of uncoilable blood surgical clipping.
23 Aneurysmal Subarachnoid Hemorrhage 507

Fig. 8 Aneurysmal subarachnoid hemorrhage in a blister-like” aneurysm of the P1 segment of the left PCA
39-year-old female. (a) Unenhanced CT scan demonstrat- (arrow). (d) Flat-panel CT-like acquisition after the stent
ing a prepontine spontaneous hyperdensity (arrow). (b) deployment showing its satisfactory apposition on the par-
DSA in AP projection showing a “blood blister-like” aneu- ent artery wall. (e) DSA performed at 6 months showing
rysm located on the left P1 segment (arrow). (c) 3D rota- the complete occlusion of the aneurysm covered by the
tional angiography, confirming the presence of a “blood flow diverter stent
508 F. Clarençon et al.

Walter Dandy first reported the surgical clip- Complications


ping of an aneurysm in 1937.
After having performed a craniotomy and a Complications that may be observed in aneurys-
microsurgical dissection around the aneurysm’s mal SAH are divided into those related to the SAH
sac and the parent artery, this technique and those related to the treatment.
consists in excluding the aneurysm from the intra-
cranial circulation by positioning a surgical clip at
the neck of the aneurysm (Fig. 9). In case of SAH-Related Complications
complex shape aneurysms, few clips may be nec-
essary to exclude the aneurysm and reconstruct SAH-related complications may be severe and life
the parent artery. Most of IC aneurysms can be threatening. Their consequences should thus not
treated by surgical means. However, IC aneu- be underestimated. Among these complications,
rysms located in the posterior circulation are hydrocephalus, arterial vasospasm, and neurolog-
more difficult to treat, leading to a higher compli- ical/neuropsychological deficits may be seen at
cation rate [60]. the acute phase of the SAH or with a delay
In complex aneurysms, for which a clipping [67]. Their management requires a close coordi-
seems too risky, or for blood blister-like aneu- nation between neurosurgical, interventional neu-
rysms, the “wrapping technique” may be an roradiology, and intensive care teams.
option to prevent rebleeding. This technique con-
sists in wrapping the aneurysm and the parent Spontaneous Rebleeding
artery by surgical gauze or plastic sheeting in Spontaneous rebleeding is the most severe com-
order to prevent further rebleeding [61]. Finally, plication in aSAH. Indeed, the clinical conse-
bypass associated with parent artery sacrifice may quences of the rebleeding are frequently worse
be an option for the treatment of some very com- than the ones from the first bleeding [68]. The
plex IC aneurysms, especially for giant/large rebleeding is related to the lysis of the clot that
aneurysms of the MCA [62]. stopped the first hemorrhage. The early rebleeding
has been reported with a rate around 14 % [69]
and is higher within the first 6 h [68]. This high
Conservative Management rate of spontaneous rebleeding and the poor clin-
ical consequences of such rebleeding are the ratio-
Conservative management and imaging follow- nale for an early treatment of ruptured IC
up may be a valuable option for unruptured intra- aneurysms.
cranial aneurysms, especially for small ones
[63]. In ruptured IC aneurysms, conservative Hydrocephalus
management should not be proposed, since Hydrocephalus may be observed at the acute
rebleeding rate is high. However, for untreatable phase of the hemorrhage or can be delayed. The
aneurysms in patients in poor clinical condition, a mechanisms that may lead to hydrocephalus are
“wait and see” strategy could be proposed, the acute obstruction of the cerebrospinal fluid path-
treatment being to be discussed few weeks later, way by a clot and abnormal blood resorption due
if the patient recovers from the aSAH and its to the SAH. Hydrocephalus may occur in the
complications. It is noteworthy that, in aSAH, following weeks after the SAH. It is related to a
the spontaneous rebleeding rate within 2 weeks fibrosis of the subarachnoid spaces and an inflam-
ranges from 15 % to 25 % and is about 50 % matory proliferation of arachnoid villosities’ cells.
within 6 months. Six months after the SAH, the Such delayed hydrocephalus is more frequent in
cumulative annual risk for rebleeding is 2–3 % per patients in poor initial grade (WFNS). Clinically,
year [11, 64–66]. consciousness disorders and psychiatric disorders
23 Aneurysmal Subarachnoid Hemorrhage 509

Fig. 9 A 60-year-old female with a ruptured left MCA the positioning of the clip at the aneurysm’s neck. (d) DSA
bifurcation treated by surgical clipping. (a) Left ICA DSA in working projection after the clipping showing a satis-
in working projection showing a small, large-necked, factory exclusion of the aneurysm (arrow). (e) On
MCA bifurcation aneurysm (arrow). (b) Photographs dur- non-subtracted image, the clip can be seen at the aneu-
ing the surgical clipping. Exposition of the aneurysm rysm’s neck (arrows)
(arrow) by microsurgical dissection. (c) Photograph after
510 F. Clarençon et al.

Table 3 Fisher grading scale [99] evaluating the appear- Daily low doses of statins have proven their effec-
ance of SAH on CT scan tiveness in preventing cerebral vasospasm
1 No hemorrhage evident [73]. The diagnosis of vasospasm should be
2 Subarachnoid hemorrhage less than 1 mm thick suspected in case of fever, disorders of conscious-
3 Subarachnoid hemorrhage more than 1 mm thick ness, and motor or sensitive deficit or on ultraso-
4 Subarachnoid hemorrhage of any thickness with nographic findings (accelerated mean velocities
intraventricular hemorrhage (IVH) or parenchymal
extension
on arterial vessels). Such signs should incite to
perform brain CT scan to rule out ischemic infarct,
hydrocephalus, parenchymal hematoma, or
rebleeding and a vascular imaging (CTA or
may be observed. Communicant hydrocephalus DSA). On vascular imaging, arterial vasospasm
may require a ventriculoatrial or ventriculo- typically appears as narrowing of the ICA termi-
peritoneal shunting and can be temporarily treated nation, A1 and M1 segments (Fig. 10). Cortical
by serial lumbar punctures [70]. defect may also be seen on capillary phase on
DSA.
Arterial Vasospasm In case of symptomatic vasospasm, aggressive
Arterial vasospasm may occur few days after the treatment should be performed in emergency.
hemorrhage (> day 3) until day 21. Its frequency Intra-arterial chemodilatation with various drugs
and severity increase with the volume of blood in (nimodipine [74], nicardipine [75], milrinone
the subarachnoid space (see Fisher score; [76]) has proven its effectiveness. However, the
Table 3). Arterial vasospasm is seen on angio- effect of such intra-arterial chemodilatation is
graphic exams in about 70 % of the cases in only temporary (<24 h). The balloon angioplasty
aSAH. In 30 % of the cases, arterial vasospasm is another option for the treatment of proximal
may lead to ischemic lesions [71]. Thus, vaso- vasospasm; it consists in the inflation of a
spasm should be prevented by aggressive thera- nondetachable balloon at the level of the arterial
pies and treated, when present, to prevent narrowing. This treatment is more effective and
potential severe complications. The clear patho- definitive, but presents some risks of severe com-
physiology of arterial vasospasm related to aSAH plications (dissection, artery rupture, etc.) [77].
remains unclear, some data suggesting an inflam-
matory vasculitis rather than a true Epilepsy
vasospasm [72]. Seizures may occur at the acute phase or lately.
The “H therapy” is one of the first-line thera- Seizures are present at the acute phase from 6 % to
pies in the armamentarium of the treatment of 25 % [78] and may be related to hypoperfusion of
aSAH-related arterial vasospasm. This therapy the brain parenchyma during the bleeding; they
associates hypertension while keeping are associated with a poor prognosis. Late seizures
normovolemia and normalized cardiac flow. may be related to rebleeding. Persistent seizures
Vasoconstrictor therapy with noradrenalin is after discharge are observed in about 5 % of the
administered in an intensive care unit, with con- cases [79]; independent predicting factors for such
tinuous monitoring comprising, at least, an inva- seizures are the presence of a subdural hematoma
sive measurement of the blood pressure. The and ischemic infarct [79]. Finally, it is noteworthy
“triple-H therapy” that associates hemodilution, that the incidence of post-SAH epilepsy is higher
hypervolemia, and arterial hypertension is no lon- in patients treated by clipping than for those
ger used to prevent arterial vasospasm because it treated by endovascular means [79].
did not prove its efficacy for the treatment of
cerebral vasospasm. The use of a vasodilator Dysnatremia
with cerebral tropism like nimodipine is system- The incidence of hyponatremia ranges from 30 %
atically performed during the first 3 weeks after to 40 %. Most of the time, this hyponatremia is
the hemorrhage either per os or intravenously. related to the so-called cerebral salt-wasting
23 Aneurysmal Subarachnoid Hemorrhage 511

Fig. 10 A 53-year-old male who presented an AComm capillary phase, extensive parenchymal defect is seen in the
aneurysm rupture. Increasing of blood velocities on Dopp- left ACA and MCA territories, corresponding to a severe
ler sonography at day 5. Left ICA DSA in anteroposterior distal vasospasm (b, arrow heads). (c) Unenhanced CT
projection: arterial phase (a) and capillary phase (b). scan performed the same day, showing multiple
Narrowing of A1 and M1 segments is seen (a, arrows) vasospasm-related ischemic lesions in both hemispheres,
consistent with a proximal arterial vasospasm. Note the predominating in the left side
delayed opacification of the left ACA (a, arrow head). On

syndrome. This syndrome, first described in the natriuretic peptide, analog of the atrial natriuretic
1950s, is characterized by an important loss of peptide. The cerebral salt-wasting syndrome
water and sodium in the urine. This syndrome is associates a depletive hyponatremia and a
the consequence of a central hypersecretion of hypovolemia. These conditions may increase the
512 F. Clarençon et al.

vulnerability of the brain for vasospasm. The


“syndrome of inappropriate antidiuretic hormone
secretion” (SIADH) is less frequent in SAH. This
syndrome associates hyponatremia and
hypervolemia. Hypernatremia, independently
from its mechanism, may worsen cerebral edema.
Finally, hypernatremia may be related to dys-
function of the hypothalamic-pituitary axis. Dia-
betes insipidus is characterized by a hypo-osmolar
polyuria (urine density close to the one of water),
hypernatremia, and hyponatriuresis, responsible
for intracellular dehydration and hypovolemic sta-
tus. Diabetes insipidus is treated by compensation
of hydric loss using hypotonic solute and by
antidiuretic hormone.

Cardiac Failure
The stimulation of the sympathetic nervous sys-
tem during the aneurysm’s rupture is responsible
for an “adrenergic storm” responsible for cardiac Fig. 11 Brain CT scan in a 46-year-old female who expe-
disease [80]. Arrhythmia may be observed as well rienced acute severe aneurysmal subarachnoid hemor-
rhage. Note the hyperdensity (arrow) in the posterior
as myocardial infarction and congestive heart fail- compartment of the eye, corresponding to blood as part
ure. These cardiac complications may be life of the so-called Terson syndrome
threatening.
A massive release of catecholamines in the
myocardial wall, which may occur during the Terson Syndrome
aSAH, will result in myocardial stunning poten- The so-called Terson syndrome corresponds to
tially responsible for cardiac failure. This syn- vitreous hemorrhage secondary to acute SAH
drome leading to a left ventricle insufficiency is (Fig. 11) [82]. This complication may lead to
known as the “adrenergic myocarditis.” Clini- severe decrease of visual acuity. The mechanism
cally, this syndrome presents as cardiogenic leading to this complication is still unclear. Sud-
shock. The duration of this cardiac failure is var- den increase of intracranial pressure may hamper
iable: from few hours to 5 days. Its evolution is the venous drainage leading to a rupture of the
most of the time favorable under tonicardiac treat- hematoretinal barrier. In bilateral non-remitting
ment, with ad integrum recovery of the cardiac cases, vitrectomy can be proposed [83].
function.

Neurological/Neuropsychological Iatrogenic Complications


Deficits
Neurological sequels may be seen secondary to Perforation/Periprocedural Rebleeding
aSAH; they may be related to the hemorrhage Perforation during the coiling is the most feared
itself, to iatrogenic complications, or to ischemic complication during the treatment of ruptured IC
vasospasm. Neuropsychological deficits, like aneurysms by endovascular means. Indeed, this
memory loss or frontal syndrome, may be seen, hemorrhagic complication may lead to death
especially in ruptured aneurysms of the AComm [84]. Rupture during endovascular treatment is
[81]. These neuropsychological disorders may be reported from 2 to 5 % [85–89]. The rupture
responsible for severe handicap and may limit the may be related to perforation with the
daily living and the return to work. microguidewire or the microcatheter during the
23 Aneurysmal Subarachnoid Hemorrhage 513

microcatheterization of the aneurysms’ sac or dur- these complications are easily managed during
ing the deployment of the coils’ loops (usually the the procedure by dropping the blood pressure or
first one) (Fig. 12). This complication, if not ade- by IVaspirin injection (Fig. 13). Sometimes, more
quately and quickly managed, may be fatal, even aggressive managements are required like IA
in the angiographic suite [90]. If a perforation injection of GPIIb/IIIa inhibitors [95], mechanical
occurs during endovascular treatment, the coiling thrombectomy [96], or even “bailout” stenting
must be performed or pursued in emergency to [58]. Rarely, delayed (within the few hours after
stop the bleeding. It is noteworthy that this com- the coiling) thromboembolic complications may
plication is more frequent in the endovascular occur. Such complication may be observed when
coiling of ruptured IC aneurysms than in the contact surface between the coils’ cast and the
unruptured ones. The mortality rate related to parent artery is large.
aneurysm perforation is relatively high Ischemic complications during the surgical
(33 %) [91]. clipping may be related to the inopportune clip-
During surgical clipping, rebleeding may also ping of a normal branch close to the aneurysm’s
occur, either during the microsurgical dissection neck, to hypoperfusion in the territory of an arte-
or during the positioning of the clip. The fre- rial branch narrowed by a clip not satisfactorily
quency of such rebleeding during surgery is esti- positioned, or to compression of surgical retrac-
mated for 10–20 %. Ruptured aneurysms are more tors on the brain parenchyma. Most of ischemic
likely to rupture during the surgical clipping than complications are silent (i.e., non-symptomatic):
unruptured ones [92]. Even if the clinical conse- 8 %. Symptomatic ischemic complications related
quences of intraoperative rupture in surgical clip- to surgical clipping only account for about
ping are less pejorative than in endovascular 2 % [97].
coiling, the clinical outcome of patients who expe-
rienced a rupture during the clipping is worse than
the other ones [86]. PCA, AComm and PComm Prognostic Factors
aneurysms were more liable to rupture
intraoperatively. The occurrence of intraoperative Some factors may predict the clinical outcome of
rupture for early surgery is not significantly higher patients who had aSAH or the occurrence of
than for surgery performed more than 3 days after delayed complications. Indeed, the World Feder-
subarachnoid hemorrhage [92]. To avoid such ation of Neurological Surgeons (WFNS) scale
complication, a temporary occlusion of the parent (Table 1) and the Hunt and Hess score (Table 2)
vessel may be used [92]. are predictive, when high (4–5) for poor clinical
outcome [98].
Thromboembolic/Ischemic The Fisher score is based on the evaluation of
Complications the SAH-related blood burden on initial CT scan
Symptomatic thromboembolic complication may (Table 3). When the Fisher score is high, the risk
occur in 5–10 % of the cases in embolization of IC and potential severity of delayed cerebral vaso-
aneurysms [93]. Even if small ischemic infarcts spasm increase [99]. Age is a controversial prog-
are relatively frequently seen on diffusion- nostic factor for clinical outcome. Indeed, even if
weighted images after endovascular treatment elderly patients (70 years) present comorbidities
(as frequently as in half of the cases [94]), most that may hamper the clinical outcome, 37 % of
of them remain asymptomatic, and among the them will have a good clinical outcome at long-
symptomatic ones, only a few will be responsible term follow-up after endovascular coiling of a
for permanent deficit. Technical difficulties that ruptured aneurysm, independently from the initial
are associated with a higher risk of thromboem- WFNS score at admission [100]. However, hydro-
bolic complications are microcatheter cephalus in elderly patients may be associated
repositioning, coil removal and repositioning, with a worse clinical outcome [101]. Finally,
and size of the aneurysmal neck [55]. Most of some scores combining clinical and biomarker
514 F. Clarençon et al.

Fig. 12 Example of perforation during the coiling in a kept inflated in front of the aneurysm. (c) DSA performed
ruptured left PComm aneurysm. (a) Left ICA DSA in after the coiling showing the complete occlusion of the
working projection showing a 4 mm saccular PComm PComm aneurysm and the stop of the bleeding. (d)
aneurysm (arrow). Also note the presence of a second Unenhanced CT scan performed after the procedure show-
small anterior choroidal artery aneurysm (arrow head). ing a diffuse SAH and contrast media extravasation. (e)
(b). DSA after the deployment of the first coil loop show- Unenhanced CT scan performed at day 10 showing a total
ing an acute rebleeding (arrows). The heparin was quickly regression of the SAH. The patient eventually discharged
reversed and the coiling pursued while the balloon was 3 weeks after the hemorrhage without any deficit
23 Aneurysmal Subarachnoid Hemorrhage 515

data, like the ABC score [102], have been pro- Perspectives
posed to predict the 1-year clinical outcome in
patients with coiled ruptured aneurysms. Using Even if the management of ruptured intracranial
the ABC score, high GCS and elevated serum aneurysms has dramatically improved during the
levels of S100β protein (a marker of glial destruc- past decades, the major challenge for the future in
tion) and troponin I were predictive of 1-year the treatment of IC aneurysms will be to treat them
mortality in this cohort. before they bleed and thus to accurately select

Fig. 13 (continued)
516 F. Clarençon et al.

Fig. 13 Example of thromboembolic complication during arrow) under balloon inflation (white arrow). (e) DSA in
the coiling of a ruptured basilar tip aneurysm. (a) DSA in working projection at the end of the coiling showing clot
working projection showing the basilar tip aneurysm with formation at the origin of the P1 segment of the left PCA
a large neck. (b) 3D rotational angiography showing more (white arrow) and of the left superior cerebellar artery
precisely the anatomy of the aneurysm. (c) Snapshot of the (black arrow). (f) After increasing of the blood pressure
road map in anteroposterior projection during the treatment and IV aspirin injection, the control DSA showed a nearly
of the aneurysm. The microcatheter tip is positioned in the complete disappearance of the clots. (g) Unenhanced CT
aneurysm’s sac (black arrow) and the remodeling balloon scan performed at D7 showing no infarct in the left PCA
in front of the neck (white arrow). (d) Snapshot of the road territory
map in lateral projection during the coil deployment (black

those which are prone to rupture. At this Although some rupture risk factors have been
moment, trials designed to compare the recognized and are used in daily practice to dis-
endovascular treatment vs. conservative manage- cuss the indications for the treatment of
ment for unruptured aneurysms have failed, unruptured IC aneurysms [104], no absolute
mostly due to the difficulty to include patients in markers for further rupture of IC aneurysms
these studies [103]. have been identified. Some tools are currently
23 Aneurysmal Subarachnoid Hemorrhage 517

under development to individualize, among all the treatment in experienced hands in the period prior to
IC aneurysms, those that are prone to rupture in the advent of endovascular coiling. J Neurol
Neurosurg Psychiatry 74:1680–1684
their natural history. The most promising of these 7. Fraser JF, Riina H, Mitra N et al (2006) Treatment of
tools is the computational fluid dynamics (CFD) ruptured intracranial aneurysms: looking to the past to
technologies [105]. CFD is a computational sim- register the future. Neurosurgery 59:1157–1166, dis-
ulation of the flow in the parent artery and the cussion 1166–1157
8. Wermer MJ, van der Schaaf IC, Algra A et al (2007)
aneurysm sac. Many parameters can be obtained Risk of rupture of unruptured intracranial aneurysms
from this simulation like the wall shear stress, in relation to patient and aneurysm characteristics: an
blood velocity, and impingement patterns, some updated meta-analysis. Stroke J Cereb Circ
of them being possibly predictive for aneurysm 38:1404–1410
9. (1998) Unruptured intracranial aneurysms–risk of
rupture [106]. rupture and risks of surgical intervention. Interna-
tional study of unruptured intracranial aneurysms
investigators. N Engl J Med 339:1725–1733
Summary 10. Bonneville F, Sourour N, Biondi A (2006) Intracra-
nial aneurysms: an overview. Neuroimaging Clin N
Am 16:371–382
Despite the advances in the management of aneu- 11. Brisman JL, Song JK, Newell DW (2006) Cerebral
rysmal subarachnoid hemorrhage, this disease aneurysms. N Engl J Med 355:928–939
remains associated with a poor prognosis. The 12. Mizutani T, Miki Y, Kojima H et al (1999) Proposed
classification of nonatherosclerotic cerebral fusiform
management of ruptured IC aneurysms can only and dissecting aneurysms. Neurosurgery 45:253–259,
be multidisciplinary, with trained diagnostic neu- discussion 259–260
roradiologists, experienced interventional neuro- 13. Niemczyk M, Gradzik M, Niemczyk S et al (2013)
radiologists and neurosurgeons, and intensivists Intracranial aneurysms in autosomal dominant poly-
cystic kidney disease. AJNR Am J Neuroradiol
specialized in neurointensive care. 34:1556–1559
14. Rozenfeld MN, Ansari SA, Shaibani A et al (2014)
Should patients with autosomal dominant polycystic
References kidney disease be screened for cerebral aneurysms?
AJNR Am J Neuroradiol 35:3–9
15. Rinkel GJ, Djibuti M, Algra A et al (1998) Prevalence
1. Johnston SC, Higashida RT, Barrow DL et al (2002) and risk of rupture of intracranial aneurysms: a sys-
Recommendations for the endovascular treatment of tematic review. Stroke J Cereb Circ 29:251–256
intracranial aneurysms: a statement for healthcare 16. Schievink WI (2004) Cerebrovascular involvement in
professionals from the Committee on Cerebrovascu- Ehlers-Danlos syndrome. Curr Treat Options
lar Imaging of the American Heart Association Coun- Cardiovasc Med 6:231–236
cil on Cardiovascular Radiology. Stroke J Cereb Circ 17. Schievink WI (1999) Marfan syndrome and intracra-
33:2536–2544 nial aneurysms. Stroke J Cereb Circ 30:2767–2768
2. Bardach NS, Zhao S, Gress DR et al (2002) Associ- 18. Schievink WI, Katzmann JA, Piepgras DG
ation between subarachnoid hemorrhage outcomes et al (1996) Alpha-1-antitrypsin phenotypes among
and number of cases treated at California hospitals. patients with intracranial aneurysms. J Neurosurg
Stroke J Cereb Circ 33:1851–1856 84:781–784
3. de Rooij NK, Linn FH, van der Plas JA et al (2007) 19. Ruigrok YM, Rinkel GJ (2008) Genetics of intracra-
Incidence of subarachnoid haemorrhage: a systematic nial aneurysms. Stroke J Cereb Circ 39:1049–1055
review with emphasis on region, age, gender and time 20. Ruigrok YM, Rinkel GJ, Algra A et al (2004) Char-
trends. J Neurol Neurosurg Psychiatry 78:1365–1372 acteristics of intracranial aneurysms in patients with
4. Muller TB, Sandvei MS, Kvistad KA et al (2013) familial subarachnoid hemorrhage. Neurology
Unruptured intracranial aneurysms in the Norwegian 62:891–894
Nord-Trondelag Health Study (HUNT): risk of rup- 21. Wiebers DO, Whisnant JP, Huston J 3rd et al (2003)
ture calculated from data in a population-based cohort Unruptured intracranial aneurysms: natural history,
study. Neurosurgery 73:256–261, discussion 260; clinical outcome, and risks of surgical and
quiz 261 endovascular treatment. Lancet 362:103–110
5. Caranci F, Briganti F, Cirillo L et al (2013) Epidemi- 22. Nieuwkamp DJ, Setz LE, Algra A et al (2009)
ology and genetics of intracranial aneurysms. Eur J Changes in case fatality of aneurysmal subarachnoid
Radiol 82:1598–1605 haemorrhage over time, according to age, sex, and
6. Lafuente J, Maurice-Williams RS (2003) Ruptured region: a meta-analysis. Lancet Neurol 8:635–642
intracranial aneurysms: the outcome of surgical
518 F. Clarençon et al.

23. Stapleton CJ, Walcott BP, Fusco MR, Butler WE, 37. Marder CP, Narla V, Fink JR et al (2014) Subarach-
Thomas AJ, Ogilvy CS (2015) Surgical management noid hemorrhage: beyond aneurysms. AJR Am J
of ruptured middle cerebral artery aneurysms with Roentgenol 202:25–37
large intraparenchymal or sylvian fissure hematomas. 38. Geibprasert S, Pongpech S, Jiarakongmun P
Neurosurgery. 76(3):258–64 et al (2010) Radiologic assessment of brain arteriove-
24. Boujemaa H, Gongora-Rivera F, Barragan-Campos H nous malformations: what clinicians need to know.
et al (2006) Bilateral acute subdural hematoma Radiogr Rev Publ Radiol Soc North Am Inc
from ruptured posterior communicating artery aneu- 30:483–501
rysm. A case report. Interv Neuroradiol J 39. Li Q, Lv F, Yao G et al (2014) 64-section
Peritherapeutic Neuroradiol Surg Proc Relat Neurosci multidetector CT angiography for evaluation of intra-
12:37–40 cranial aneurysms: comparison with 3D rotational
25. Feely M, Kapoor S (1987) Third nerve palsy due to angiography. Acta Radiol 55(7):840–6
posterior communicating artery aneurysm: the impor- 40. Taschner CA, Thines L, Lernout M et al (2007) Treat-
tance of early surgery. J Neurol Neurosurg Psychiatry ment decision in ruptured intracranial aneurysms:
50:1051–1052 comparison between multi-detector row CT angiog-
26. Clarencon F, Bonneville F, Boch AL et al (2011) Par- raphy and digital subtraction angiography.
ent artery occlusion is not obsolete in giant aneurysms J Neuroradiol 34:243–249
of the ICA. Experience with very-long-term follow- 41. Teksam M, McKinney A, Casey S et al (2004) Multi-
up. Neuroradiology 53:973–982 section CT angiography for detection of cerebral
27. Roccatagliata L, Guedin P, Condette-Auliac S aneurysms. AJNR Am J Neuroradiol 25:1485–1492
et al (2010) Partially thrombosed intracranial aneu- 42. Zhang H, Hou C, Zhou Z et al (2012) Evaluating of
rysms: symptoms, evolution, and therapeutic man- small intracranial aneurysms by 64-detector CT angi-
agement. Acta Neurochir 152:2133–2142 ography: a comparison with 3-dimensional rotation
28. Davies A, Dale O, Renowden S (2011) Spontaneous DSA or surgical findings. J Neuroimag Off J Am Soc
rupture of an intra-cavernous internal carotid artery Neuroimag
aneurysm presenting with massive epistaxis. 43. Luo Z, Wang D, Sun X et al (2012) Comparison of the
J Laryngol Otol 125:1070–1072 accuracy of subtraction CT angiography performed
29. Cortnum S, Sorensen P, Jorgensen J (2010) Determin- on 320-detector row volume CT with conventional
ing the sensitivity of computed tomography scanning CT angiography for diagnosis of intracranial aneu-
in early detection of subarachnoid hemorrhage. Neu- rysms. Eur J Radiol 81:118–122
rosurgery 66:900–902, discussion 903 44. Yanamadala V, Sheth SA, Walcott BP et al (2013)
30. Latchaw RE, Silva P, Falcone SF (1997) The role of Non-contrast 3D time-of-flight magnetic resonance
CT following aneurysmal rupture. Neuroimaging angiography for visualization of intracranial aneu-
Clin N Am 7:693–708 rysms in patients with absolute contraindications to
31. Sohn CH, Baik SK, Lee HJ et al (2005) MR imaging CT or MRI contrast. J Clin Neurosci 20:1122–1126
of hyperacute subarachnoid and intraventricular hem- 45. Missler U, Hundt C, Wiesmann M et al (2000) Three-
orrhage at 3T: a preliminary report of gradient echo dimensional reconstructed rotational digital subtrac-
T2*-weighted sequences. AJNR Am J Neuroradiol tion angiography in planning treatment of intracranial
26:662–665 aneurysms. Eur Radiol 10:564–568
32. Mohamed M, Heasly DC, Yagmurlu B et al (2004) 46. Hui FK, Tumialan LM, Tanaka T et al (2009) Clinical
Fluid-attenuated inversion recovery MR imaging and differences between angiographically negative, dif-
subarachnoid hemorrhage: not a panacea. AJNR Am J fuse subarachnoid hemorrhage and perimesen-
Neuroradiol 25:545–550 cephalic subarachnoid hemorrhage. Neurocrit Care
33. Verma RK, Kottke R, Andereggen L et al (2013) 11:64–70
Detecting subarachnoid hemorrhage: comparison of 47. Lin N, Zenonos G, Kim AH et al (2012) Angiogram-
combined FLAIR/SWI versus CT. Eur J Radiol negative subarachnoid hemorrhage: relationship
82:1539–1545 between bleeding pattern and clinical outcome.
34. Sidman R, Connolly E, Lemke T (1996) Subarach- Neurocrit Care 16:389–398
noid hemorrhage diagnosis: lumbar puncture is still 48. Dalyai R, Chalouhi N, Theofanis T et al (2013) Sub-
needed when the computed tomography scan is nor- arachnoid hemorrhage with negative initial catheter
mal. Acad Emerg Med 3:827–831 angiography: a review of 254 cases evaluating patient
35. Davis SM, Donnan GA (2008) Thunderclap head- clinical outcome and efficacy of short- and long-term
ache: CT and lumbar puncture but occasionally repeat angiography. Neurosurgery 72:646–652, dis-
more! Stroke J Cereb Circ 39:1396 cussion 651–642
36. Cruickshank A, Auld P, Beetham R et al (2008) 49. Molyneux A, Kerr R, Stratton I et al (2002) Interna-
Revised national guidelines for analysis of tional Subarachnoid Aneurysm Trial (ISAT) of neu-
cerebrospinal fluid for bilirubin in suspected sub- rosurgical clipping versus endovascular coiling in
arachnoid haemorrhage. Ann Clin Biochem 2143 patients with ruptured intracranial aneurysms:
45:238–244 a randomised trial. Lancet 360:1267–1274
23 Aneurysmal Subarachnoid Hemorrhage 519

50. Wong GK, Boet R, Ng SC et al (2012) Ultra-early 65. Rosengart AJ, Schultheiss KE, Tolentino J
(within 24 hours) aneurysm treatment after subarach- et al (2007) Prognostic factors for outcome in patients
noid hemorrhage. World Neurosurg 77:311–315 with aneurysmal subarachnoid hemorrhage. Stroke J
51. Guglielmi G, Vinuela F, Dion J et al (1991) Cereb Circ 38:2315–2321
Electrothrombosis of saccular aneurysms via 66. Feigin VL, Findlay M (2006) Advances in
endovascular approach. Part 2: preliminary clinical subarachnoid hemorrhage. Stroke J Cereb Circ
experience. J Neurosurg 75:8–14 37:305–308
52. Moret J, Cognard C, Weill A et al (1997) Reconstruc- 67. Koenig MA (2012) Management of delayed cerebral
tion technic in the treatment of wide-neck intracranial ischemia after subarachnoid hemorrhage. Continuum
aneurysms. Long-term angiographic and clinical 18:579–597
results. Apropos of 56 cases. J Neuroradiol 24:30–44 68. Inagawa T, Kamiya K, Ogasawara H et al (1987)
53. van Rooij WJ, Sluzewski M, Beute GN et al (2006) Rebleeding of ruptured intracranial aneurysms in the
Procedural complications of coiling of ruptured intra- acute stage. Surg Neurol 28:93–99
cranial aneurysms: incidence and risk factors in a 69. Ohkuma H, Tsurutani H, Suzuki S (2001) Incidence
consecutive series of 681 patients. AJNR Am J and significance of early aneurysmal rebleeding
Neuroradiol 27:1498–1501 before neurosurgical or neurological management.
54. Shapiro M, Babb J, Becske T et al (2008) Safety and Stroke J Cereb Circ 32:1176–1180
efficacy of adjunctive balloon remodeling during 70. Hasan D, Lindsay KW, Vermeulen M (1991) Treat-
endovascular treatment of intracranial aneurysms: a ment of acute hydrocephalus after subarachnoid hem-
literature review. AJNR Am J Neuroradiol orrhage with serial lumbar puncture. Stroke J Cereb
29:1777–1781 Circ 22:190–194
55. Albayram S, Selcuk H, Kara B et al (2004) Throm- 71. Dorsch NW (1995) Cerebral arterial spasm–a clinical
boembolic events associated with balloon-assisted review. Br J Neurosurg 9:403–412
coil embolization: evaluation with diffusion-weighted 72. Dumont AS, Dumont RJ, Chow MM et al (2003)
MR imaging. AJNR Am J Neuroradiol 25:1768–1777 Cerebral vasospasm after subarachnoid hemorrhage:
56. Crowley RW, Albuquerque FC, Ducruet AF putative role of inflammation. Neurosurgery
et al (2012) Technical considerations in the 53:123–133, discussion 133–125
endovascular management of aneurysms of the pos- 73. Sanchez-Pena P, Nouet A, Clarencon F et al (2012)
terior inferior cerebellar artery. Neurosurgery Atorvastatin decreases computed tomography and
71:ons204–ons217, discussion ons217–ons208 S100-assessed brain ischemia after subarachnoid
57. Chapot R, Houdart E, Saint-Maurice JP et al (2002) aneurysmal hemorrhage: a comparative study. Crit
Endovascular treatment of cerebral mycotic aneu- Care Med 40:594–602
rysms. Radiology 222:389–396 74. Biondi A, Ricciardi GK, Puybasset L et al (2004)
58. Yoo E, Kim DJ, Kim DI et al (2009) Bailout stent Intra-arterial nimodipine for the treatment of symp-
deployment during coil embolization of tomatic cerebral vasospasm after aneurysmal sub-
intracranial aneurysms. AJNR Am J Neuroradiol arachnoid hemorrhage: preliminary results. AJNR
30:1028–1034 Am J Neuroradiol 25:1067–1076
59. Gonzalez AM, Narata AP, Yilmaz H et al (2014) 75. Linfante I, Delgado-Mederos R, Andreone V
Blood blister-like aneurysms: single center experi- et al (2008) Angiographic and hemodynamic effect
ence and systematic literature review. Eur J Radiol of high concentration of intra-arterial nicardipine in
83:197–205 cerebral vasospasm. Neurosurgery 63:1080–1086,
60. Spetzler RF, McDougall CG, Albuquerque FC discussion 1086–1087
et al (2013) The barrow ruptured aneurysm trial: 76. Shankar JJ, dos Santos MP, Deus-Silva L et al (2011)
3-year results. J Neurosurg 119:146–157 Angiographic evaluation of the effect of intra-arterial
61. Cudlip SA, Kitchen ND, McKhahn GM et al (1998) milrinone therapy in patients with vasospasm from
Wrapping of solitary ruptured intracranial aneurysms, aneurysmal subarachnoid hemorrhage. Neuroradiol-
outcome at five years. Acta Neurochir ogy 53:123–128
140:1167–1170, discussion 1170–1161 77. Dorsch NW (2002) Therapeutic approaches to vaso-
62. Kivipelto L, Niemela M, Meling T et al (2014) Bypass spasm in subarachnoid hemorrhage. Curr Opin Crit
surgery for complex middle cerebral artery aneu- Care 8:128–133
rysms: impact of the exact location in the MCA tree. 78. Hart Y, Sneade M, Birks J et al (2011) Epilepsy after
J Neurosurg 120:398–408 subarachnoid hemorrhage: the frequency of seizures
63. Valenca MM (2013) “Sit back, observe, and wait.” Or after clip occlusion or coil embolization of a ruptured
is there a pharmacologic preventive treatment for cerebral aneurysm: results from the International Sub-
cerebral aneurysms? Neurosurg Rev 36:1–9, discus- arachnoid Aneurysm Trial. J Neurosurg
sion 9–10 115:1159–1168
64. Edlow JA, Malek AM, Ogilvy CS (2008) Aneurysmal 79. Claassen J, Peery S, Kreiter KT et al (2003) Predictors
subarachnoid hemorrhage: update for emergency and clinical impact of epilepsy after subarachnoid
physicians. J Emerg Med 34:237–251 hemorrhage. Neurology 60:208–214
520 F. Clarençon et al.

80. Ahmadian A, Mizzi A, Banasiak M et al (2013) Car- 94. Altay T, Kang HI, Woo HH et al (2011) Thromboem-
diac manifestations of subarachnoid hemorrhage. bolic events associated with endovascular treatment
Heart Lung Vessels 5:168–178 of cerebral aneurysms. J Neurointerv Surg 3:147–150
81. Proust F, Martinaud O, Gerardin E et al (2009) Qual- 95. Aggour M, Pierot L, Kadziolka K et al (2010)
ity of life and brain damage after microsurgical clip Abciximab treatment modalities for thromboembolic
occlusion or endovascular coil embolization for rup- events related to aneurysm coiling. Neurosurgery
tured anterior communicating artery aneurysms: neu- 67:503–508
ropsychological assessment. J Neurosurg 110:19–29 96. Fourie P, Duncan IC (2003) Microsnare-assisted
82. Terson A (1900) De l’hémorrhagie dans le corps vitre mechanical removal of intraprocedural distal middle
au cours de l’hémorrhagie cerebrale. Clin Ophthalmol cerebral arterial thromboembolism. AJNR Am J
6:309–312 Neuroradiol 24:630–632
83. Stienen MN, Lucke S, Gautschi OP et al (2012) 97. Krayenbuhl N, Erdem E, Oinas M et al (2009) Symp-
Terson haemorrhage in patients suffering aneurysmal tomatic and silent ischemia associated with microsur-
subarachnoid haemorrhage: a prospective analysis of gical clipping of intracranial aneurysms: evaluation
60 consecutive patients. Clin Neurol Neurosurg with diffusion-weighted MRI. Stroke J Cereb Circ
114:535–538 40:129–133
84. Chen M (2013) A checklist for cerebral aneurysm 98. Wang T, Zhang JH, Qin X (2011) Analysis on death-
embolization complications. J Neurointerv Surg associated factors of patients with subarachnoid hem-
5:20–27 orrhage during hospitalization. Acta Neurochir Suppl
85. Ricolfi F, Le Guerinel C, Blustajn J et al (1998) Rup- 110:219–223
ture during treatment of recently ruptured aneurysms 99. Fisher CM, Kistler JP, Davis JM (1980) Relation of
with Guglielmi electrodetachable coils. AJNR Am J cerebral vasospasm to subarachnoid hemorrhage
Neuroradiol 19:1653–1658 visualized by computerized tomographic scanning.
86. Elijovich L, Higashida RT, Lawton MT et al (2008) Neurosurgery 6:1–9
Predictors and outcomes of intraprocedural rupture in 100. Iosif C, Di Maria F, Sourour N et al (2013) Is a high
patients treated for ruptured intracranial aneurysms: initial World Federation of Neurosurgery (WFNS)
the CARAT study. Stroke J Cereb Circ 39:1501–1506 grade really associated with a poor clinical outcome
87. Vinuela F, Duckwiler G, Mawad M (1997) Guglielmi in elderly patients with ruptured intracranial aneu-
detachable coil embolization of acute rysms treated with coiling? J Neurointerv Surg
intracranial aneurysm: perioperative anatomical and 101. Degos V, Gourraud PA, Tursis VT et al (2012) Elderly
clinical outcome in 403 patients. J Neurosurg age as a prognostic marker of 1-year poor outcome for
86:475–482 subarachnoid hemorrhage patients through its inter-
88. Cognard C, Weill A, Castaings L et al (1998) Intra- action with admission hydrocephalus. Anesthesiol-
cranial berry aneurysms: angiographic and clinical ogy 117:1289–1299
results after endovascular treatment. Radiology 102. Degos V, Apfel CC, Sanchez P et al (2012) An admis-
206:499–510 sion bioclinical score to predict 1-year outcomes in
89. Doerfler A, Wanke I, Egelhof T et al (2001) Aneurys- patients undergoing aneurysm coiling. Stroke J Cereb
mal rupture during embolization with Guglielmi Circ 43:1253–1259
detachable coils: causes, management, and outcome. 103. Raymond J, Molyneux AJ, Fox AJ et al (2008) The
AJNR Am J Neuroradiol 22:1825–1832 TEAM trial: safety and efficacy of endovascular treat-
90. Ryu CW, Lee CY, Koh JS et al (2011) Vascular ment of unruptured intracranial aneurysms in the pre-
perforation during coil embolization of an intracranial vention of aneurysmal hemorrhages: a randomized
aneurysm: the incidence, mechanism, and clinical comparison with indefinite deferral of treatment in
outcome. Neurointervention 6:17–22 2002 patients followed for 10 years. Trials 9:43
91. Cloft HJ, Kallmes DF (2002) Cerebral aneurysm per- 104. Lall RR, Eddleman CS, Bendok BR et al (2009)
forations complicating therapy with Guglielmi Unruptured intracranial aneurysms and the assess-
detachable coils: a meta-analysis. AJNR Am J ment of rupture risk based on anatomical and mor-
Neuroradiol 23:1706–1709 phological factors: sifting through the sands of data.
92. Leipzig TJ, Morgan J, Horner TG et al (2005) Anal- Neurosurg Focus 26:E2
ysis of intraoperative rupture in the surgical treatment 105. Cebral JR, Mut F, Weir J et al (2011) Quantitative
of 1694 saccular aneurysms. Neurosurgery characterization of the hemodynamic environment in
56:455–468, discussion 455–468 ruptured and unruptured brain aneurysms. AJNR Am
93. Brooks NP, Turk AS, Niemann DB et al (2008) Fre- J Neuroradiol 32:145–151
quency of thromboembolic events associated with 106. Cebral JR, Sheridan M, Putman CM (2010) Hemody-
endovascular aneurysm treatment: retrospective case namics and bleb formation in intracranial aneurysms.
series. J Neurosurg 108:1095–1100 AJNR Am J Neuroradiol 31:304–310
Blister Aneurysms
24
James V. Byrne and Svein Harald Mørkve

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 522 Blister aneurysms comprise about 1 % of rup-
tured intracranial aneurysms. They are associ-
Etiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 522
ated with higher rates of rebleeding and, as a
Demography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 522 result, with greater mortality and morbidity
Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 522 than typical aneurysms. Because of rarity, the
etiology and pathology are poorly understood.
Presentation and Incidence . . . . . . . . . . . . . . . . . . . . . . . . 523
Diagnosis relies on a stereotypical appearance
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 523 and location at non-branch points on the inter-
Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 525 nal carotid artery at angiography, together with
Surgical Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 526 inspection at surgery. They are increasingly
seen involving other intracranial arteries and
Endovascular Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . 528
generally considered to be caused by athero-
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 531 sclerotic mural disease.
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 532 Surgical clipping is difficult because of their
small size, sessile shape, and thin sac walls. It
is frequently complicated by operative rupture,
and alternative wrapping and trapping are often
required. For these reasons, endovascular treat-
ments are increasingly used as an alternative.
The recent introduction of high mesh density
stents, designed to redirect blood flow away
from aneurysms and reinforce the parent artery,
has stimulated the development of new pro-
cedures to manage these lesions. Initial expe-
riences with flow-diverting stents have shown
J.V. Byrne (*) a substantial improvement in outcomes, and
Nuffield Department of Surgical Sciences, Oxford stent procedures are rapidly being accepted as
University, John Radcliffe Hospital, Oxford, UK
the intervention of choice.
e-mail: james.byrne@nds.ox.ac.uk
S.H. Mørkve
Department of Neurosurgery, Haukeland University
Hospital, Bergen, Norway
e-mail: snhm@helse-bergen.no; smorkve@gmail.com

# Springer Science+Business Media New York 2016 521


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_20
522 J.V. Byrne and S.H. Mørkve

Etiology
Keywords
Aneurysm • Intracranial aneurysm • Blister The primary cause is probably atherosclerosis of
aneurysm • Blood blister aneurysm • Blister- the arterial wall combined with particular hemo-
like aneurysms • Subarachnoid hemorrhage • dynamic forces generated by blood flow in the
Stent • Flow diverter • Wrapping • Clip wrap- supraclinoid internal carotid artery [1, 8]. The lat-
ping • Microsurgical clipping • Endovascular ter is most likely related to the shape of the
therapy supraclinoid section and forces generated at
curves of the arterial wall [4]. Other mechanisms
have been proposed, including local dissection
Introduction [9], although no classic flap has been demon-
strated, or the presence of occult small branch
These are a well-recognized but poorly defined arteries. Abe et al. postulated that mural hemor-
subgroup of intracranial aneurysms. They were rhages at sites of atheroma were important etiol-
first described in the neurosurgical literature ogy factors and that subsequent growth into a
30 years ago [1, 2] as small focal arterial wall saccular shape was caused by organization of the
dilations occurring at non-branching points of the hematoma overlying a wall defect [7]. Such a
internal carotid artery. About 5 % of intracranial mechanism would explain differences in histol-
aneurysms are found at non-branch points, of ogy and natural history reported for this type of
which a minority look like a blister of the arterial aneurysm.
wall on inspection at surgery [3, 4]. This type of
aneurysm has been reported to arise on the
anteromedial, dorsal, anterior, and superior internal
carotid artery wall [5]. The term blister aneurysm Demography
has therefore been applied to describe a small
aneurysm with a thin wall found on the wall of They are more commonly found in women than
the supraclinoid internal carotid artery. Aneurysms men [9–11], and patients are generally younger in
with the same appearance, on the basis of inspec- age than those with typical intracranial aneurysms
tion at surgery or on angiography, have also been [12]. They are reported to occur more frequently
found arising from other basal cerebral arteries. on the right side [13]. Systemic hypertension, [9,
Another characteristic feature of blister aneu- 12] and atherosclerosis [7, 9] are associated risk
rysms is their behavior. This is a potential source factors.
of confusion since relying on small size and stereo-
typical angiographic appearance to distinguish this
type of aneurysm is unreliable because serial angio- Pathology
grams have shown that they may enlarge within
days of presentation. This behavior means that There have been very few reports of histological
their appearance on angiograms may vary from a examinations of blister aneurysms. Ishikawa
small bulge of the arterial wall to sessile or saccular et al. [5] described the autopsy findings of a man
sac shapes [6]. Changing size and shape together after fatal bleeding from an aneurysm of the supe-
with frequent rebleeding are considered their hall- rior surface of the internal carotid artery. This
mark behavior and the best criteria for report described the histological examination as
distinguishing them from other types of intracranial follows: “The internal elastic lamina and media
aneurysms. Because of the thin wall evident at sur- had disappeared at the border between the eccen-
gery, the terms blister, blister-like [3, 4], or blood trically sclerotic and normal carotid wall. The gap
blister aneurysm [7] have been used synonymously in the internal elastica was covered with normal
by different authors. In this chapter, the simple term adventitia and fibrinous tissue. This portion was
blister aneurysm will be used throughout. not composed of collagenous tissue as ordinarily
24 Blister Aneurysms 523

seen in aneurysm walls. Neither infiltration of of large series of ruptured intracranial aneurysms,
inflammatory cells nor dissection of the artery their frequency is around 1 %. Yasargil reported
were observed.” Thus, ruling out dissection, they 3 (0.9 %) in a series of 319 [2], Nakagawa et al.
concluded that the aneurysm was the result of 8 (1.7 %) in a series of 460 [4], and Meling et al.’s
degeneration of the internal elastic lamina. In 14 (1.5 %) in a series of 912 [13] ruptured aneu-
another case report by Kim et al. [6], the histology rysms. In Meling et al.’s series, all blister aneu-
of a resected blister aneurysm showed smooth rysms occurred at non-branching points of the
muscle in the wall, and the authors considered it internal carotid artery, and these constituted
to be the same as a “true aneurysm.” In this 6.6 % of ruptured internal carotid artery
patient, clipping was performed 12 days after aneurysms [13].
SAH and after a second angiogram had shown
the aneurysm to have grown from sessile to a
saccular shape. Imaging
These are the only two reports quoted in the
translated English literature though there have Because of their small size, blister aneurysms can
been sufficient reports of in vivo observations at be difficult to identify on CT or magnetic reso-
surgery to conclude that the blister aneurysm has a nance imaging (MRI) scans, and catheter angio-
distinct pathology and no clear evidence that they gram (DSA) is the best imaging modality to
are the result of focal dissection. The observation identify these lesions. On DSA, a focal arterial
that a blister aneurysm can enlarge within days of dilation is seen with a variety of possible shape
SAH suggests a dynamic process, which may be configurations, including fusiform, sessile, or sac-
associated with the proximity and organization of cular dilations of the artery at non-branch points.
an acute blood clot [7]. Local exposure to endog- These changes are often difficult to distinguish
enous lytic agents could be a factor in the process. from mural dissections (see Fig. 1). The location
on the internal carotid artery helps to suggest the
diagnosis, but as discussed above, the classic
Presentation and Incidence in vivo description of this lesion was made by
direct inspection at surgery, and this remains the
Most patients present with acute subarachnoid best method of confirming the angiogram diagno-
hemorrhage (80–90 %), and the rest are diagnosed sis. However, small focal dilations demonstrated
coincidental with other aneurysms. In the reports in similar circumstances on arteries other than the

Fig. 1 Axial CT (a) showing acute subarachnoid hemor- shaped” dilation of the internal carotid artery on the
rhage caused by rupture of a blister aneurysm. The aneu- anterior-superior surface between the posterior communi-
rysm was not diagnosed on preliminary CTA, but cating and anterior choroidal artery origins (arrows)
subsequent DSA (b and c) showed a typical “thorn-
524 J.V. Byrne and S.H. Mørkve

Fig. 2 Axial CT (a) and lateral DSA (b) at presentation Control angiograms after deployment of the stent (c) and
5 days after spontaneous subarachnoid hemorrhage caused after insertion of coils (d) show how the aneurysm enlarged
by a blister aneurysm of the right internal carotid artery. in the interval between angiograms and developed a more
The aneurysm was treated with a combination of an saccular shape. Small markers can be seen at the two ends
endovascular stent and endosaccular coils after the patient of the stent (arrows)
suffered repeat subarachnoid hemorrhage 12 days later.

internal carotid artery may also be blister aneu- confirmation of the diagnosis by surgical inspec-
rysms, and proposing this diagnosis after angiog- tion is not available (see Fig. 3).
raphy alone seems appropriate. For the time being, it can be argued that the
Sequential angiograms often show the aneu- blister aneurysm is a precursor of a saccular aneu-
rysm to enlarge within days of the presenting rysm, which spontaneous subarachnoid hemor-
subarachnoid hemorrhage, which helps to distin- rhage brings to medical attention and after
guish them from very small non-blister aneurysms rupture is, for a short period of time, in an unstable
[6, 9] (see Fig. 2). This transformation into a larger state. Whether such aneurysms left untreated
saccular shape on serial DSA is usually inevitably evolve to become saccular aneurysms
interpreted to indicate dissection or mural hemor- is unknown, but it has been reported that sponta-
rhage as the initiating event for aneurysms at sites neous complete regression can occur [14]. In the
other than the internal carotid artery. It creates a future, higher resolution imaging than available
potential difficulty in classification for future on current magnetic resonance imagers may be
authors, since blister aneurysms are increasingly able to better detect mural hemorrhage and pro-
managed by endovascular treatment and vide noninvasive in vivo confirmation of the
24 Blister Aneurysms 525

Fig. 3 Unruptured tandem small aneurysms (large surface of the artery. Their location is typical of blister
arrows) of the right internal carotid artery. DSA performed aneurysms, but inspection at surgery was not available to
before (a) and after (b) placing a stent to cover the necks of confirm the diagnosis. The small arrows show the markers
the aneurysms, which arise from the superior and lateral at the ends of the stent (Courtesy of Dr. M. Cellerini)

postulated etiology and diagnosis at other arterial high risks, which significantly increases the total
locations. risk of iatrogenic morbidity [15]. Adding to the
total risk is the possibility that these small lesions
are frequently not diagnosed on primary CT angi-
Management ography or initial catheter angiograms and are
only evident on repeat angiograms after growth
Blister aneurysms are notoriously difficult to treat of the aneurysmal sac. This delay prolongs the
regardless of the approach employed, and inter- period that patients are at risk of repeat bleeding
ventions are associated with overall rates of mor- and additional morbidity if rebleeding
tality and morbidity that are significantly higher occurs [15].
than those for typical ruptured intracranial aneu- The best treatment of blister aneurysms has,
rysms [9, 15, 16]. The thin, fragile wall of this since their first recognition, been controversial,
type of aneurysm, in combination with their small and various strategies have over time been advo-
sizes and broad necks, are some of the features cated. Most recently, this has centered on the
that render them particularly prone to relative strength and weaknesses of open micro-
intraprocedural rupture when treated using either surgery compared to endovascular treatments.
surgical or endovascular techniques [12, 17]. Parent artery sacrifice (i.e., a deconstructive tech-
Blister aneurysms are thus considered particularly nique) was for a long time considered the best
dangerous lesions and carry an overall primary treatment for these lesions and is still by
re-rupture risk that is greater than that of most many used as a rescue therapy if other techniques
saccular aneurysms [12, 17]. Another feature of fail. Advances in both microsurgical and
these lesions that adds to the high mortality and endovascular techniques, however, have allowed
morbidity rates reported after spontaneous an increasing proportion of blister aneurysms to
subarachnoid hemorrhage is a frequently be treated preserving the parent artery (i.e., recon-
observed rapid regrowth of the aneurysmal sac structive techniques) or preserving the blood flow
within weeks of apparently initial successful treat- carried through the parent artery to its distal terri-
ment [15]. Blister aneurysms consequently may tory through various bypass procedures. These
require multiple treatments, all carrying similar techniques generally appear to offer better
526 J.V. Byrne and S.H. Mørkve

anatomical results and patient outcomes than par- uncommon occurrence during attempted direct
ent artery sacrifice [13, 18]. clipping of these lesions [9, 13]. Intraoperative
Other measures in addition to direct interven- rupture may create a situation where sacrifice of
tions to occlude the aneurysm are important adju- the parent vessel (ICA) to control bleeding is the
vant treatments, which should be considered in only possible way to regain control. However,
order to prevent aneurysm re-rupture. These parent artery sacrifice has been shown by Meling
include tight control of arterial blood pressure, et al. to be associated with poor outcomes if
careful drainage of cerebrospinal fluid, and the performed in SAH patients within 48 h of their
use of antifibrinolytic drugs such as aminocaproic bleeding even if collaterals appeared to be ade-
acid [18]. quate on preoperative angiography [13]. These
Most treatment techniques proposed and authors suggested that this was due to vasospasm
described in the literature have been directed secondary to subarachnoid hemorrhage reducing
toward the classical blister aneurysms arising the available blood flow support to the distal vas-
from the internal carotid artery. Similar lesions cular territory via collateral arteries and that parent
can, however, as already mentioned sometimes artery occlusion in the acute phase after hemor-
be identified on angiography at other locations. rhage should be avoided if at all possible [13].
The typical “thorn-shaped” arterial wall dilation Due to the high risk of intraoperative rupture of
(Fig. 1) has for example been demonstrated in blister aneurysms during attempted clipping,
aneurysms of the basilar artery, and because of alternative methods for surgical repair have been
this appearance, these aneurysms have also been proposed. Several authors have advocated treat-
termed blister aneurysms [19]. However, since ment with various wrapping or combined clipping
endovascular therapists do not have the possibility and wrapping techniques [7, 21]. The vessel-
of direct visual inspection, it can only be assumed encircling clip graft, or Sundt clip graft, was intro-
that aneurysms with these appearances at other duced over 40 years ago, and recently, Park and
locations have the same characteristics as those Meyer described the application of Sundt clip
of the ICA. grafts as a rescue technique when vessel dehis-
cence occurred perioperatively. They achieved
good outcomes in 76 % of patients, most of
Surgical Treatment whom were treated for blister aneurysms
[22]. Sundt clip grafts have also been successfully
The thin, frail walls of blister aneurysms easily applied as a salvage operation after failed
rupture during surgical dissection or during endoluminal stenting [23], and their application
attempted aneurysm repair [9, 12, 13, 20]. Appli- remains an important tool in the arsenal of surgical
cation of clips directly onto the aneurysm is treatment techniques for selected patients. How-
treacherous, and gentle dissection needs to be ever, the encircling clip grafts have important
performed with careful planning of aneurysm limitations when it comes to the anatomy of the
clip placement. Ideally, the clip should be applied vessel segment around which they are to be
completely parallel to the parent vessel, which applied. The clip grafts cannot be applied to blister
may be facilitated by clipping under systemic aneurysms arising close to branch arteries without
hypotension [9, 12]. Parallel clip placement occluding them and thereby causing ischemic
ensures an even distribution of traction from the infarctions and potentially permanent neurologi-
aneurysm clip on the vessel wall, minimizing cal deficits.
distortion of the wall and allowing a more secure Although generally less effective than clipping
and stable hold of the clip on the wall surrounding in preventing re-rupture of saccular aneurysms,
the thin-walled and fragile sac of the blister aneu- different wrapping techniques have been
rysm [4, 7, 10]. Despite surgical manipulation employed successfully by several authors to treat
being performed with the utmost care, avulsion unclippable blister aneurysms [24–26]. Various
and rupture of the aneurysm wall is not an materials have been used for wrapping including
24 Blister Aneurysms 527

muscle [27], muslin gauze followed by a layer of A further consideration in treatment planning
temporal fibrous fascia [28], and cotton [26]. Blis- is the possible late development of chronic gran-
ter aneurysms have also been coated with various ulation tissue after wrapping. Granulomas or
plastic adhesives [29] including formulated cya- so-called gauzomas have been found to some-
noacrylates, such as Biobond or Aron Alpha [30, times form around wrapped aneurysms [26].
31], and methyl methacrylate [29]. Wrapping These may cause cranial neuropathies, which is
using muscle and fascia seems to be of limited the most frequent complication of aneurysm
value when it comes to preventing re-rupture, wrapping apart from re-rupture [26]. To minimize
most likely because of the gradual necrosis and the risk of this happening, the wrapping material
disappearance of the tissue over time [27, 31, 32]. should therefore never be placed in direct contact
Wrapping with cotton or gauze, however, either with cranial nerves [21].
alone or combined with coating the wrapped Wrapping of blister aneurysms is now usually
aneurysm with plastic adhesives, offers better pro- performed in combination with some form of clip
tection over time [32]. A potential disadvantage of placement. Aneurysmal clips can be placed across
using plastic adhesives relates to their potential to the lesion after it has been wrapped with gauze or
cause chronic inflammatory changes with necro- cotton (e.g., Surgicel) in a technique called “wrap-
sis and fibrosis of the media layer of the vessel holding clipping.” Here the wrapping material both
wall. Used alone, cotton and muslin gauze cause reinforces the arterial wall and prevents slippage
fibrosis in the adventitia but no changes in the and displacement of the clip which may cause
arterial wall media [33]. This selective reaction aneurysm re-rupture. The wrapping material can
of the outer layer of the vessel wall is thought to also be used to encase the arterial segment
better reinforce the aneurysm sac [27]. Several containing the aneurysm, and an aneurysm clip
authors therefore consider cotton used without can then be placed over the wrapping material to
additional adhesive coating the best material for hold it in place without incorporating the aneurysm
wrapping ruptured aneurysms [21, 26, 33]. or vessel wall in the clip. This technique is known
In one study, re-rupture rates for aneurysms as “wrap clipping” [35]. Both techniques have been
wrapped between 1965 and 1975 using muslin reported to achieve better results for difficult blister
gauze with or without additional muscle or adhe- aneurysms than clipping or wrapping alone
sives were found to be 8.6 % for early re-rupture, [9]. Wrapping materials commonly in use are cot-
i.e., within the first 6 months, and 1.5 % per ton (e.g., Bemsheet or Surgicel) [35], rayon [21],
annum for late re-ruptures up to 10 years postop- Gore-Tex [18, 26], or Dacron [36]. The latter two
eratively [24]. This does not compare favorably to fabrics are mostly used for wrap clipping.
an annual re-rupture risk of around 0.3–0.4 % per Delayed aneurysm growth and branch occlu-
year for aneurysms that were clipped in the same sions have been reported after wrap clipping, and
period [24]. However, in the cooperative study frequent follow-up imaging is therefore
performed prior to the widespread availability of recommended in particular for patients treated
CT scanning, Nishioka reported a confirmed fatal with this technique [35]. Another way to combine
rebleeding rate of 25 % in untreated patients with wrapping and clipping techniques is to put the
all types of intracranial aneurysms [34], and it is wrapping material around a primary clipped
thus clear that wrapping of aneurysms offers a lesion. Similar to wrap-holding clipping, this is
significant degree of protection compared to the also performed in order to prevent slippage of the
re-rupture risk of untreated aneurysms. The use- clip, to strengthen the arterial wall, and to provoke
fulness of this technique has also been confirmed formation of scar tissue over time [18]. This tech-
in the era of microsurgical neurosurgery, and one nique has been used as a salvage operation
more recent study reported no rebleeding during a together with arterial suturing or tying the blades
follow-up interval of 5 years for patients with of a clip to the carotid trunk with silk sutures
solitary aneurysms wrapped using muslin following perioperative rupture of blister aneu-
gauze [25]. rysms during primary clip placement [20].
528 J.V. Byrne and S.H. Mørkve

Over recent years, it has become increasingly with this kind of bypass surgery, however, espe-
clear that blister aneurysms of the supraclinoid cially in the setting of acute subarachnoid hemor-
internal carotid artery may involve a large portion rhage, is significant, and 25 % of patients in the
of the vessel circumference. Often nearly half same review had greater neurological deficits after
(i.e., 180 ) of the vessel’s circumference is surgery than before, mostly due to thromboem-
directly involved in the aneurysm with its charac- bolic events [39]. Because of this high complica-
teristic diseased thin and fragile vessel wall. This tion rate, Spetzler and colleagues have recently
leaves a very limited amount of normal vessel stated that they consider the EC-IC high-flow
wall onto which it is possible to place aneurysm bypass and parent artery occlusion as the last
clips and can make clip reconstruction, with pres- option for treating blister aneurysms to be used
ervation of a vessel diameter sufficient to sustain only after failure of other techniques, i.e., primary
adequate perfusion in the territory supplied by the clipping or wrap clipping [18]. They point out that
artery, impossible [12]. Extracranial to intracra- although good results are obtainable using bypass
nial (EC-IC) bypass followed by parent artery surgery, the complication rates of this kind of
occlusion or trapping has therefore been increas- surgery immediately after subarachnoid hemor-
ingly advocated by several authors as a means of rhage remain high in clinical practice [18].
treating blister aneurysms [13, 18, 37, 38]. Various
bypass techniques are used depending on the
magnitude of the blood flow the bypass needs to Endovascular Treatment
sustain and the inherent availability of collateral
blood flow. A low-flow bypass from the superfi- The standard endovascular treatment for ruptured
cial temporal artery (STA) to the middle cerebral intracranial aneurysm is performed by packing the
artery (MCA), a so-called STA-MCA bypass, can aneurysm sac with platinum coils (i.e.,
only provide limited supportive blood flow. A endosaccular coiling). This approach, however,
high-flow bypass between the external carotid like standard microsurgical clipping is not optimal
artery (ECA) and the MCA with an interposed for treating blister aneurysms. Attempted
arterial or venous graft (e.g., radial artery endosaccular coiling of these lesions has at best
(RA) or saphenous vein (SV)) can provide much yielded mixed results and is often not possible
more substantial blood flow. When treating aneu- because of their broad-based shapes and small
rysms of the supraclinoid internal carotid artery sizes [13, 17]. Both these features make it difficult
(ICA), a high-flow bypass such as the ECA-RA- for coils to be retained within the aneurysm sac,
MCA bypass is usually required [37, 38]. and treatment of small-sized aneurysms is gener-
In one small study, 4 out of 5 patients with ally associated with higher rates of periprocedural
ruptured internal carotid artery blister aneurysms rupture. Thus endosaccular coil embolization
had good outcomes after treatment with ECA- carries a high risk of coil herniation into the parent
RA-MCA bypass surgery and parent artery occlu- artery as well as intraoperative aneurysm rupture
sion [38]. A temporary low-flow STA-MCA [16]. The wide dome and inherent recurrence ten-
bypass was used during the procedure to provide dency of blister aneurysms also prevent balloon
adequate perfusion to the MCA territory while the remodeling from achieving more permanent aneu-
permanent anastomosis between the ICA and rysm occlusion [17, 40]. Consequently, most blis-
MCA using a RA graft was established ter aneurysms are unsuitable for endosaccular
(so-called double-insurance bypass) [38]. This is coiling, and parent artery occlusion has histori-
a complex procedure, but the authors’ results were cally been considered the more practical and safer
in line with the results from a large systematic endovascular treatment option [17].
review of different aneurysms in the anterior cir- However, for the reasons already mentioned,
culation treated with EC-IC bypass surgery, in outcomes after parent artery sacrifice are often
which good outcomes (mRS 0–1) were reported poor in the acute period after subarachnoid hem-
in 81 % of the patients [39]. The complication rate orrhage unless supported by bypass surgery
24 Blister Aneurysms 529

[13, 15], even if the native collateral blood flow is to induce progressive aneurysm thrombosis,
capacity appeared sufficient when assessed before which usually occurs within weeks of treatment,
treatment [13]. Parent artery occlusion also carries and neointimal growth covering the stent and
the inherent risk of occluding important side closing the aneurysm neck over the following
branch arteries of the internal carotid artery [15], months [43, 44].
and therefore in many centers microsurgical repair The intra-aneurysmal reduction in blood flow
remained the primary treatment approach for blis- induced by flow-diverting stents is closely related
ter aneurysms until self-expanding endovascular to the porosity and pore density of the device
stents for intracranial use became available on the [44]. Porosity is a measure of the proportion of
commercial market. open area to the total area of the stent wall, and for
Combining endosaccular coil embolization flow-diverting stents, this is typically in the range
with stenting has several potential advantages. of 65–70 % (i.e., a metal coverage of 30–35 %). In
The most important in the context of blister aneu- comparison, conventional stents have higher
rysms is that stents deployed in the parent artery porosities (>90 %) and therefore considerably
act to retain coils in the aneurysm sac. Coil and less metal coverage [43, 44]. Flow-diverting
stent treatments are performed either by first plac- stents appear to offer a better endovascular treat-
ing the stent and then introducing coils through ment alternative for blister aneurysms in part
the struts of the stent or by catheterizing the aneu- because they can be used without placement of
rysm sac and then placing the stent over the cath- any adjunct endosaccular coils. This theoretically
eter before introducing endosaccular coils into the reduces the risk of periprocedural aneurysm rup-
aneurysm (so-called “jailing” technique) (see ture compared to the stent and coil technique but
Fig. 2). Stent-assisted coiling has allowed a sig- needs to be proven in practice [45, 46].
nificantly larger proportion of blister aneurysms to There are currently four commercially avail-
be treated successfully with an endovascular able flow-diverting stents that are in common use.
approach, and good clinical outcomes have been These are the Silk/Leo flow diverter (Balt Extru-
reported in 62–89 % of patients [16, 40–42]. A sion, Montmorency, France), the Pipeline Embo-
majority of treated patients, however, still had to lization Device (PED - Covidien, Dublin,
have more than one procedure, either for place- Ireland), the Flow Redirection Endoluminal
ment of a second stent within the first or place- Device (FRED – Microvention, Tustin, CA,
ment of additional coils after aneurysm regrowth USA), and the Surpass flow diverter (Surpass,
becomes evident on follow-up imaging [40, 41]. Stryker Neurovascular, Fremont, CA, USA).
Early postoperative or perioperative hemorrhage, Flow-diverting stents have been used to treat blis-
most often fatal, has also been reported to occur in ter aneurysms with encouraging results [45, 47].
13–28 % of blister aneurysm patients treated Good outcomes (mRS 0–2 or unchanged/
using stent and coil techniques [16, 41, 42]. The improved neurological status) and combined mor-
results of initial attempts to utilize stents to aug- bidity and mortality rates of 0–10 % were reported
ment endovascular treatment by endosaccular in 86–100 % of patients treated with flow-
coiling were mixed. diverting stents for blister aneurysms not
So-called flow-diverting stents were intro- presenting acutely after subarachnoid hemorrhage
duced in 2007 and provide an alternative [46, 48]. Recently, good outcomes (mRS 0–2)
endovascular treatment option for aneurysms were also reported in 92 % of patients treated for
with a difficult anatomy such as the blister aneu- ruptured blister aneurysms using Silk stents with a
rysm. These stents work by reconstructing lami- combined morbidity and mortality rate of
nar flow in the parent vessel and redirecting blood 18 % [19]. Similar results have been obtained by
flow away from the aneurysm sac [43, 44]. Inflow others treating ruptured blister aneurysms with
jets of blood into the aneurysm are reduced, and both Silk stents and PEDs with good clinical out-
the shear stress on the aneurysm wall is eliminated comes (mRS 0–2 or GOS 4–5) in 66–94 % of
immediately after deployment [43, 44]. The effect patients [49–51]. Procedure-related morbidity
530 J.V. Byrne and S.H. Mørkve

and mortality rates, however, remain as high as some time with clopidogrel [54]. This precaution
15–25 %, and thromboembolic or hemorrhagic has been recommended in order to reduce the
complications in the postoperative period have incidence of hemorrhagic complications that
been reported in 5–33 % [49–51]. Aneurysms have been shown to occur preferentially in
treated with flow-diverting stents tend to patients overtreated with clopidogrel [54]. Treat-
gradually occlude as endosaccular thrombosis ment with two antiplatelet drugs (e.g., aspirin and
occurs over a period of time after treatment, clopidogrel) is typically continued for a minimum
and complete occlusion has been reported in of 3–6 months after the procedure, and then aspi-
75–100 % at 3–6 months and in 94–100 % at rin is usually continued for at least an additional
12 months [19, 49, 51, 52]. 6 months or in some instances for the remainder of
Deployment of any stent, and especially flow- the patient’s life [54, 55].
diverting stents with their larger metal content, The most widespread method of performing
requires the use of drugs to prevent spontaneous platelet function tests in endovascular therapy
in-stent thrombosis. Patients are therefore obliged units is the point-of-care system VerifyNow
to receive antiplatelet drugs with attendant risks of (Accumetrics, San Diego, CA). This system can
spontaneous hemorrhage and more severe bleed- measure a patient’s response to all three main
ing should the aneurysm rupture or re-rupture. In classes of antiplatelet agents in current use.
elective treatments of unruptured aneurysms, These are blocking the formation of thromboxane
most centers today start treatment with two A2 in platelets (aspirin), platelet receptor P2Y12
antiplatelet drugs, typically aspirin and inhibitors (thienopyridines such as clopidogrel),
clopidogrel, 5–17 days before treatment and platelet glycoprotein IIb/IIIa receptor inhibi-
[53–55]. Such dual antiplatelet therapy is tors (e.g., abciximab). The system measures plate-
employed in part to overcome potential resistance let function qualitatively by exposing patient
to one or the other drug, which occurs in about whole-blood samples to a mixture of agonists
20 % of people. The heterogeneity of individuals’ causing platelet activation and fibrinogen-coated
responses to antiplatelet drugs is idiosyncratic for microparticles. Platelet function is measured
aspirin, but for clopidogrel, it is attributable to this based upon the activated platelets’ ability to bind
being a prodrug. Clopidogrel is administered to the microparticles causing them to aggregate.
orally and though usually well absorbed requires Aspirin inhibition is reported as aspirin reaction
activation by cytochrome P450 enzymes in the units (ARU), whereas clopidogrel inhibition (and
liver. Its effectiveness is thus liable to be affected other thienopyridines) is reported as P2Y12 reac-
by interactions with other commonly used drugs tion units (PRU). Importantly, when it comes to
such as atorvastatin and verapamil [54]. Whereas neuroendovascular treatments using stents, PRU
15–20 % of the population are hyperresponders to values <60 or >240 have been shown to be a
clopidogrel, as many as 25–32 % are strong predictor for thromboembolic and hemor-
hyporesponders to the same drug [53–55]. Platelet rhagic complications, respectively, up to 6 months
function tests are therefore usually recommended postoperatively [53, 54].
at least once before surgery to make sure patients In the setting of acute subarachnoid hemor-
are not under- or overtreated and to make any rhage and the use of stents to treat ruptured aneu-
necessary adjustments in dose or switch from rysms, administration of antiplatelet drugs
clopidogrel to an alternative antiplatelet drug requires careful consideration. Though various
such as ticagrelor or prasugrel before the proce- therapy regimens have been used for prophylaxis
dure [54]. To further complicate matters, up to against stent thrombosis in this situation, there is
75 % of patients will also show an increasing still no consensus on the optimal treatment. In
response to clopidogrel during long-term treat- most centers, however, loading doses of
ment and may thus become overtreated. It has clopidogrel and aspirin or ticagrelor are used
therefore been suggested to perform follow-up [16, 54, 55]. Typically, emergency loading doses
platelet function tests in all patients treated over in the range of 325–500 mg aspirin and
24 Blister Aneurysms 531

300–600 mg clopidogrel are administered at the ruptured blister aneurysms. A majority of the
time of, or up to 8 h prior to, endovascular treat- patients were treated surgically, but 28 % required
ment. Both drugs are then continued in lower more than one therapeutic approach. Both the
maintenance doses from the day after treatment overall morbidity (mRS >2 or new neurological
as for elective cases [49, 55]. Platelet function deficit following treatment) and mortality were
tests are under these circumstances not commonly higher in patients who received surgical treat-
recommended prior to treatment, as they will not ment, 18 % and 14 % respectively, as compared
affect the planned treatment strategy, but they can to 3.4 % and 11.5 % respectively in patients
be of value at a later stage to ensure adequate post- receiving endovascular treatment [15]. Thus
procedural antiplatelet therapy. despite the need for antiplatelet drugs being a
It has been suggested that the risk of intracere- major constraint for the endovascular treatment
bral hemorrhage in stented patients on antiplatelet of ruptured blister aneurysms with flow-diverting
drugs is independent of a preexisting subarach- stents, this technique appears the most promising
noid hemorrhage [47, 56], and similar delayed prospect for improving treatment in these difficult
hemorrhage rates have been reported among aneurysms.
patients treated for ruptured or unruptured aneu-
rysms using flow-diverting stents [45–52]. How-
ever, dual antiplatelet therapy in the acute period Summary
after subarachnoid hemorrhage is a delicate bal-
ance and complicates, in particular, the manage- Blister aneurysms are rare and comprise less than
ment of hydrocephalus. Operative intracranial 1 % of ruptured intracranial aneurysms. Various
procedures, such as insertion of an external ven- terms, such as blood blister and blister-like, have
tricular drain (EVD) or placement of a ventricu- been used for this type of aneurysm. A common
loperitoneal shunt (VP shunt), are associated with feature is a higher frequency of rebleeding than
significantly increased risks of causing intracra- occurs in the usual type of ruptured intracranial
nial hemorrhages in this setting [15, 57, 58], and aneurysms and, as a result, greater mortality and
particular attention to surgical hemostasis is there- morbidity. The pathological cause for this differ-
fore warranted during such procedures. ent behavior is poorly understood because of their
Gonzales et al. recently published their experi- rarity. In vivo diagnosis relies on a stereotypical
ence with ruptured blister aneurysms together appearance on angiography or after inspection at
with a systematic review of the existing literature surgery and a typical location at non-branch
on treatment of these aneurysms [15]. The authors points of the internal carotid artery. However, the
reported good outcomes (mRS 0–2) in 91 % of same appearances and natural histories are
their own patients treated from 2003 to 2013. increasingly reported for aneurysms involving
Endovascular therapy was their primary approach other intracranial arteries. They are generally con-
and was initially performed using conventional sidered to be caused by atherosclerotic mural
stents combined with endosaccular coiling but disease.
was later changed to flow-diverting stents (single Surgical clipping is difficult because of their
or multiple stents telescoped within each other) small size, sessile shape, and thin sac walls. It is
without adjunctive endosaccular coils. If uncer- frequently complicated by operative rupture, and
tainty as to the likely best approach for the patient alternative but less secure treatments by wrapping
existed after imaging by CT angiography, catheter or trapping are often considered a safer option. For
angiography was performed in a hybrid theater these reasons, endovascular treatments have been
where endovascular or surgical treatment could proposed and used as an alternative. The recent
proceed directly after assessment of the aneurysm introduction of high mesh density endovascular
anatomy by DSA performed in the operating stents, which are designed to redirect blood flow
room. In their review of the literature, they found away from the aneurysm lumen and reinforce the
63 original papers including 322 patients with parent artery wall, has stimulated the development
532 J.V. Byrne and S.H. Mørkve

of new procedures to manage these lesions. Initial subarachnoid haemorrhage: treatment and outcome.
experiences of treatment with endovascular stents J Neurosurg 108:662–671
14. Ueta T, Ichi S, Ochi T, Suzuki I (2004) Spontaneous
have shown a substantial improvement in out- regression of an aneurysm at a nonbranching site of the
comes, and stent procedures are rapidly being supraclinoid internal carotid artery. Case report.
accepted as the intervention of choice. J Neurosurg 101:1070–1072
15. Gonzalez AM, Narata AP, Yilmaz H, Bijlenga P,
Radovanic I, Schaller K, Lovblad KO, Pereira VM
(2014) Blood blister-like aneurysms: single center
References experience and systematic literature review. Eur J
Radiol 83:197–205
1. Ohara H, Sakamoto T, Suzuki J (1979) Sclerotic brain 16. Meckel S, Singh TP, Undrén P, Ramgren B, Nilsson
aneurysms. In: Suzuki J (ed) Cerebral aneurysms. Neu- OG, Phatouros C, McAulife W, Cronqvist M (2011)
ron, Tokyo, pp 673–682 Endovascular treatment using predominantly stent-
2. Yasargil MG (1984) Clinical considerations, surgery of assisted coil embolization and antiplatelet and
the intracranial aneurysm and results. Georg Thieme anticoagulation management of ruptured blood
Verlag, Stuttgart/New York blister-like aneurysms. AJNR Am J Neuroradiol
3. Yoshimoto Y, Ochiai C, Nagai M (1996) Cerebral 32:764–771
aneurysms unrelated to arterial bifurcations. Acta 17. Park JH, Park IS, Han DH, Kim SH, Oh CW, Kim JE,
Neurochir (Wien) 138:958–963 Kim HJ, Han MH, Kwon OK (2007) Endovascular
4. Nakagawa F, Kobayashi S, Takemae T, Sugita K treatment of blood blister-like aneurysms of the inter-
(1986) Aneurysms protruding from the dorsal nal carotid artery. J Neurosurg 106:812–819
wall of the internal carotid artery. J Neurosurg 18. Garrett M, Spetzler RF (2012) Surgical treatment of
65:303–308 blister-like aneurysms. World Neurosurg 77:76–77
5. Ishikawa T, Nakamura N, Houkin K, Nomura N (1997) 19. Aydin K, Arat A, Sencer S, Hakyemez B,
Pathological consideration of a “blister-like” aneurysm Barburoglu M, Sencer A, Izgi N (2014) Treatment of
at the superior wall of the internal carotid artery: case ruptured blood blister-like aneurysms with flow
report. Neurosurgery 40:403–405 diverter Silk stents. J Neurointerv Surg. doi:10.1136/
6. Kim JH, Kwon TH, Kim JH, Park YK, Chung HS neurintsurg-2013-011090
(2006) Internal carotid artery dorsal wall aneurysm 20. Joo SP, Kim TS, Moon KS, Kwak HJ, Lee JK, Kim JH,
with configurational change: are they all false aneu- Kim SH (2006) Arterial suturing followed by clip
rysms? Surg Neurol 66:441–443 reinforcement with circumferential wrapping for
7. Abe M, Tabuchi K, Yokoyama H, Uchino A (1998) blister-like aneurysms of the internal carotid artery.
Blood blisterlike aneurysms of the internal carotid Surg Neurol 66:424–428
artery. J Neurosurg 89:419–424 21. Figueiredo EG, Foroni L, Monaco BA, Gomes MQ,
8. Sano K (1979) Internal carotid posterior communicat- Sterman Neto H, Teixeira MJ (2010) The clip-wrap
ing anterior choroidal region aneurysms. Microsurgical technique in the treatment of unclippable aneurysms.
treatment. In: Pia HW, Langmaid C, Zierski J (eds) Arq Neuropsiquiatr 68:115–118
Cerebral aneurysms. Advances in diagnosis and ther- 22. Park PJ, Meyer FB (2010) The Sundt clip graft. Neu-
apy. Springer, Berlin/Heidelberg/New York, p 252 rosurgery 66:300–305
9. Ogawa A, Suzuki M, Ogasawara K (2000) Aneurysms 23. Cho TG, Hwang SN, Nam TK, Park SW (2012) Sal-
at nonbranching sites in the supraclinoid portion of the vage surgical treatment for failed endovascular proce-
internal carotid artery: internal carotid artery trunk dure of a blood blister-like aneurysm. J Cerebrovasc
aneurysms. Neurosurgery 47:578–583 Endovasc Neurosurg 22:295–303
10. Shigeta H, Kyoshima K, Nakagawa F, Kobayashi S 24. Todd NV, Tocher JL, Jones PA, Miller JD (1989) Out-
(1992) Dorsal internal carotid artery aneurysms with come following aneurysm wrapping: a 10-year follow-
special reference to angiographic presentation and sur- up review of clipped and wrapped aneurysms.
gical management. Acta Neurochir (Wien) 119:42–48 J Neurosurg 70:841–846
11. Kinouchi H, Mizoi K, Nagamine Y, Yanagida N, 25. Cudlip SA, Kitchen ND, McKahn GM, Bell BA (1998)
Mikawa S, Suzuki A, Sasajima T, Yoshimoto T Wrapping of solitary intracranial aneurysms, outcome
(2002) Anterior paraclinoid aneurysms. J Neurosurg at five years. Acta Neurochir (Wien) 140:1167–1171
96:1000–1005 26. Desmukh VR, Kakarla UK, Udaya K, Figueiredo EG,
12. Sim SY, Shin YS, Cho KG, Kim SY, Kim SH, Ahn YH, Zabramski M, Spetzler R (2005) Long-term clinical
Yoon SH, Cho KH (2006) Blood blister-like aneu- and angiographic follow-up of unclippable wrapped
rysms at nonbranching sites of the internal carotid intracranial aneurysms. Neurosurgery 58:434–442
artery. J Neurosurg 105:400–405 27. Sachs E Jr (1972) The fate of muscle and cotton
13. Meling TR, Sorteberg A, Bakke SJ, Slettebø H, wrapped about intracranial carotid artery aneurysms.
Hernesniemi J, Sorteberg W (2008) Blood blister-like A laboratory and clinico-pathological study. Acta
aneurysms of the internal carotid artery causing Neurochir (Wien) 26:121–137
24 Blister Aneurysms 533

28. Gillingham FJ (1958) The management of ruptured assisted coil embolization. Clin Neurol Neurosurg
intracranial aneurysms. Hunterian lecture. R Coll 115:920–925
Surg Engl 23:89–117 42. Ihn YK, Kim SH, Sung JH, Kim TG (2012) The
29. Dutton JEM (1956) Intracranial aneurysms. A new efficacy of endovascular treatment of ruptured blood
method of surgical treatment. Br J Med 2:585–586 blister-like aneurysms using stent-assisted coil embo-
30. Cartoon CA, Kennedy JC, Heifetz MD et al (1965) The lization. Interv Neuroradiol 18:432–441
use of a plastic adhesive (methyl-2-cyanoacrylate 43. Sadasivan C, Cesar L, Seong J, Rakian A, Hao Q, Tio
monomer) in the management of intracranial aneu- FO, Wakhloo AK, Lieber BB (2009) An original flow
rysms and leaking cerebral vessels. A report of diversion device for the treatment of intracranial aneu-
15 cases. In: Sahs AL, Field WS (eds) Intracranial rysms: evaluation in the rabbit elastase-induced model.
aneurysms and subarachnoid haemorrhage. Charles Stroke 40:952–958
C. Thomas, Springfield 44. Zanaty M, Chaloui N, Tjoumakaris SI, Rosenwasser
31. Minakawa T, Koike T, Fujii Y, Ishii R, Tanaka R, Arai RH, Gonzalez LF, Jabbour P (2014) Flow-diversion
H (1987) Long term results of ruptured aneurysms panacea or poison? Front Neurol. doi: 10.3389/
treated by coating. Neurosurgery 21:660–663 fneur.2014.00021
32. Mount LA, Antunes JL (1975) Results of treatment of 45. Martin AR, Cruz JP, Matouk CC, Marotta TR (2012)
intracranial aneurysms by wrapping and coating. The pipeline flow-diverting stent for exclusion of rup-
J Neurosurg 42:189–193 tured intracranial aneurysms with difficult morphol-
33. Herrera O, Kawamura S, Yasui N, Yoshida Y (2005) ogies. Neurosurgery 70:21–28
Histological changes in the rat common carotid artery 46. Chalouhi N, Zanati M, Tjoumakaris S, Gonzalez LF,
induced by aneurysmal wrapping and coating mate- Hasan D, Kung D, Rosenwasser RH, Jabbour P (2014)
rials. Neurol Med Chir (Tokyo) 39:134–139 Treatment of blister-like aneurysms with the
34. Nishioka H (1966) Report on the cooperative study of pipeline embolization device. Neurosurgery
intracranial aneurysms and subarachnoid 74:527–532
haemorrhage. Section VII. I. Evaluation of the conser- 47. Rasskazoff S, Silvaggio J, Brouwer PA, Kaufmann A,
vative management of ruptured intracranial aneurysms. Nistor A, Iancu D (2010) Endovascular treatment of a
J Neurosurg 25:574–592 ruptured blood blister-like aneurysm with a flow-
35. Kim YB, Hong CK, Chung J, Joo JY, Huh SK (2014) diverting stent. Interv Neuroradiol 16:255–258
Long-term clinical and angiographic outcomes of 48. Wagner A, Cortsen M, Hauerberg J, Romner B, Wag-
wrap- clipping strategies for unclippable cerebral aneu- ner MP (2012) Treatment of intracranial aneurysms.
rysms. Yonsei Med J 55:401–409 Reconstruction of the parent artery with flow-diverting
36. Nakano S, Iseda T, Yoneyama T, Goya T, Wakisaka S SILK-stent. Neuroradiology 54:709–718
(2000) A combination of wrapping and clipping using 49. Cruz JP, O’Kelly C, Kelly M, Wong JH, Alshaya W,
a collagen-impregnated dacron fabric (Hemashield). Martin A, Spears J, Marotta TR (2013) Pipeline embo-
Surg Neurol 53:330–333 lization device in aneurysmal subarachnoid
37. Kazumata K, Nakayama N, Nakamura T, haemorrhage. AJNR Am J Neuroradiol 34:271–276
Kamiyama H, Terasaka S, Houkin K (2014) Changing 50. Causin F, Pascarella R, Pavesi G, Marasconi R,
treatment strategy from clipping to radial artery graft Zambon G, Battaglia R, Munari M (2011) Acute
bypass and parent artery sacrifice in patients with rup- endovascular treatment (<48 hours) of uncoilable rup-
tured blister-like internal carotid artery aneurysms. tured aneurysms at non-branching sites using Silk
Neurosurgery 10(Suppl 1):66–73 flow-diverting devices. Interv Neuroradiol 17:357–364
38. Murai Y, Mizunari T, Umeoka K, Tateyama K, 51. Nerva JD, Morton RP, Levitt MR, Osbun JW, Ferreira
Kobayashi S, Teramoto A (2012) Ischemic complica- MJ, Ghodke BV, Kim LJ (2014) Pipeline embolization
tions after radial artery grafting and aneurysmal trap- device as primary treatment for blister aneurysms and
ping for ruptured internal carotid artery anterior wall iatrogenic pseudoaneurysms of the internal carotid
aneurysms. World Neurosurg 77:166–171 artery. J Neurointerv Surg. doi:10.1136/neurintsurg-
39. Schaller B (2008) Extracranial-intracranial bypass 2013-011047
to reduce the risk of ischemic stroke in intracranial 52. Brinjikji W, Murad M, Lanzino G, Cloft HJ, Kallmes
aneurysms of the anterior cerebral circulation: DF (2013) Endovascular treatment of intracranial
a systematic review. J Stroke Cerebrovasc Dis aneurysms with flow diverters. A metaanalysis. Stroke
17:287–298 44:442–447
40. Lee BH, Kim BM, Park MS, Park SI, Cheung EC, Suh 53. Delgado Almandoz JE, Crandall BM, Scholz JM,
SH, Choi CS, Won YS, Yu IK (2009) Reconstructive Fease JL, Anderson RE, Kadkhodayan Y, Tubman
endovascular treatment of ruptured blood blister-like DE (2013) Pre-procedure P2Y12 reaction units value
aneurysms of the internal carotid artery. J Neurosurg predicts perioperative thromboembolic and hemor-
110:431–436 rhagic complications in patients with cerebral aneu-
41. Fang YB, Li Q, Yang PF, Huang QH, Zhao WY, Xu Y, rysms treated with the Pipeline embolization device.
Hong B, Liu JM (2013) Treatment of blood blister-like J Neurointerv Surg 5. doi:10.1136/neurintsurg-2012-
aneurysms of the internal carotid artery with stent- 010582
534 J.V. Byrne and S.H. Mørkve

54. Delgado Almandoz JE, Crandall BM, Scholz JM, 56. Pearl M, Gregg L, Gailloud P (2010) Endovascular
Fease JL, Anderson RE, Kadkhodayan Y, Tubman treatment of aneurysmal subarachnoid haemorrhage.
DE (2014) Last-recorded P2Y12 reaction units value Neurosurg Clin N Am 21:271–280
is strongly correlated with thromboembolic and hem- 57. Mahaney KB, Chalouhi N, Viljoen S, Smietana J,
orrhagic complications occurring up to 6 months after Kung DK, Jabbour P, Bulsara KR, Howard M, Hasan
treatment in patients with cerebral aneurysms treated DM (2013) Risk of hemorrhagic complication associ-
with the Pipeline embolization device. AJNR Am J ated with ventriculoperitoneal shunt placement in
Neuroradiol 35:128–135 aneurysmal subarachnoid hemorrhage patients on
55. Faught RW, Satti SR, Hurst RW, Pukenas BA, Smith dual antiplatelet therapy. J Neurosurg 119:937–942
MJ (2014) Heterogeneous practice pattern regarding 58. Kung DK, Policeni BA, Capuano AW, Rossen JD,
antiplatelet medications for neuroendovascular Jabbour PM, Torner JC, Howard MA, Hasan D
stenting in the USA: a multicenter survey. (2011) Risk of ventriculostomy-related hemorrhage in
J Neurointerv Surg. doi:10.1136/neurintsurg-2013- patients with acutely ruptured aneurysms treated using
010954 stent-assisted coiling. J Neurosurg 114:1021–1027
Surgical Treatment of Aneurysms
25
Saul F. Morales-Valero, Shanna Fang, and Giuseppe Lanzino

Contents Abstract
Introduction and Historical Overview . . . . . . . . . . . . 536 Although an increasing number of aneurysms
are treated with endovascular therapy, tradi-
Indications for Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . 537
tional surgical treatment continues to be an
Choice of Treatment Modality . . . . . . . . . . . . . . . . . . . . . 537 important therapeutic option for both ruptured
Preoperative Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 538 and unruptured aneurysms. The goal of aneu-
rysm treatment is to isolate the aneurysm from
Surgical Principles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 538
Anesthetic Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 540 the circulation while ensuring patency of par-
Critical Steps in Aneurysm Surgery (Table 1) . . . . . . 540 ent artery and/or perforator branches. With
Intraoperative Assessment of Aneurysm advanced neurosurgical approaches focused
Occlusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 543 on minimizing brain manipulation, careful
Intraoperative Angiography . . . . . . . . . . . . . . . . . . . . . . . . . 543 arachnoid dissection, adjunctive intraoperative
Microvascular Doppler Ultrasonography . . . . . . . . . . . . 543 monitoring, and imaging techniques, aneu-
Indocyanine Green Fluorescent
Videoangiography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 543 rysm surgery can reliably achieve definitive
occlusion with low complication rates. In the
Surgical Approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 543
Pterional Craniotomy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 544
last decade, the widespread utilization of non-
Other Surgical Approaches for the Treatment invasive vascular imaging techniques has led
of Aneurysms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 545 to an increased number of unruptured aneu-
Postoperative Care . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 545 rysms that are detected and treated. Noninva-
sive imaging studies such as MRA and CTA
Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 546
play an ever-increasing role not only for the
Follow-Up of Surgically Treated Aneurysms . . . . . 546 preoperative assessment but also postoperative
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 547 monitoring of patients with intracranial
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 547
aneurysms.

Keywords
S.F. Morales-Valero • G. Lanzino (*) Intracranial aneurysms • Cerebral aneurysms •
Department of Neurologic Surgery, Mayo Clinic, Vascular disorder • Surgical clipping • Surgical
Rochester, MN, USA
treatment • Carotid balloon occlusion • Digital
e-mail: saulfmv@gmail.com;
lanzino.giuseppe@mayo.edu subtraction angiography • Magnetic resonance
angiography • Computed tomography angiog-
S. Fang
Mayo Medical School, Mayo Clinic, Rochester, MN, USA raphy • Indocyanine green angiography •
e-mail: fang.shanna@mayo.edu Intraoperative angiography • Ruptured
# Springer Science+Business Media New York 2016 535
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_5
536 S.F. Morales-Valero et al.

aneurysm • Unruptured aneurysm • Aneurys- of preoperative temporary carotid occlusion test-


mal subarachnoid hemorrhage ing, development of adjustable clamps for gradual
carotid occlusion, and introduction of adjuvant
extracranial-to-intracranial arterial bypass [8, 9,
Introduction and Historical Overview 11, 12]. Another well-accepted method of carotid
occlusion for aneurysm treatment at the time was
Aneurysm rupture and subarachnoid hemorrhage parent artery sacrifice with permanent balloon
are associated with significant morbidity and mor- occlusion, although the success of treatment was
tality. Patients who suffer aneurysmal subarach- similarly dependent on the degree of collateral
noid hemorrhage carry an average case fatality circulation available to prevent ischemia
rate as high as 52 %, while as many as 46 % of [13–16]. Long-term complications of carotid and
those surviving the subarachnoid hemorrhage parent artery occlusion include the risk of imme-
have significant long-term cognitive impairment, diate or late cerebral ischemia, as well as devel-
and a third of survivors are left dependent on opment of flow-associated aneurysms [11,
lifelong care [1–4]. For these patients, the immi- 17–20]. In a review of patients treated with ther-
nent danger is rebleeding from the ruptured aneu- apeutic carotid occlusion, 4.3 % of patients were
rysm, so the therapeutic goal is to isolate and found to have de novo aneurysm formation on
secure the aneurysm to prevent this catastrophic average 9.1 years after treatment, thus suggesting
complication. The mortality associated with rup- the need for extensive follow-up [21]. Nowadays,
tured aneurysms has decreased in the last three carotid sacrifice is used only in selected cases of
decades with early surgical intervention, which large and giant aneurysms that are not amenable to
has been shown to reduce the risk of rebleeding surgical clipping or endovascular treatment and
and improve the chance for good recovery in has shown good outcomes and low mortality rates
patients with subarachnoid hemorrhage [5–7]. In [20, 22–24].
recent years, advances and widespread utilization In the late 1960s and early 1970s with the
of noninvasive imaging have led to a large number introduction of the surgical microscope and
of unruptured and often incidental aneurysms to improvements in surgical clip technology, direct
be diagnosed, and this has been associated with an clipping with the use of microsurgical techniques
increase in the number of invasive procedures to became the modus operandi and led to the devel-
treat intact aneurysms as well. opment of modern aneurysm surgery as we know
Surgical clipping has long been considered the it today. The next important step in the treatment
gold standard for the treatment of intracranial of intracranial aneurysms was the introduction of
aneurysms. Direct surgical clipping excludes the the Guglielmi Detachable Coil in 1992, which
aneurysm from the circulation and removes the represented a dramatic paradigm shift with the
risk of growth and rupture. In recent years, paral- eventual establishment of endovascular therapy
lel to the development of endovascular tech- as an acceptable alternative, and even first-line
niques, there has been a progressive reduction in option, to traditional clip ligation at many centers
the number of surgically treated aneurysms. Nev- [25]. However, microsurgery remains the funda-
ertheless, surgery continues to be an important mental gold standard for certain aneurysms with
therapeutic strategy in the treatment of these complex morphological features such as small
lesions. size, broad-based neck, those located in small
Prior to 1970, the method of treating aneu- diameter parent arteries. Surgical treatment is
rysms was carotid ligation, which was shown to also preferred for certain patients such as those
decrease the risk of rebleeding compared to con- of younger age and good functional status, as it
servative treatment [8–10]. The high rates of treat- provides immediate and long-term aneurysmal
ment failure and ischemic complications obliteration. Given the variety of considerations
associated with this procedure were greatly that factor into decision making for the best ther-
reduced with modifications that included the use apeutic approach for each individual aneurysm,
25 Surgical Treatment of Aneurysms 537

a multidisciplinary cerebrovascular team is neces- each patient and aneurysm should be considered
sary to work together to consider both surgical and individually, a consensus group of international
endovascular options in every case [26–28]. This experts has recently come up with a scoring sys-
chapter provides an overview of the principles of tem that can be used in deciding whether or not an
modern intracranial aneurysm surgery. unruptured aneurysm should be considered for
invasive treatment [30].

Indications for Treatment


Choice of Treatment Modality
Invasive treatment of an intracranial aneurysm is
dependent on many different conditions, the most Surgical clipping of intracranial aneurysms has
important one being the rupture status. A patient long been considered the modality of choice and
who presents with intracranial hemorrhage from a the gold standard for the treatment of intracranial
ruptured aneurysm has a greater than 60 % risk of aneurysms. However, since the introduction of the
re-rupture over the next 6 months, with the risk Guglielmi detachable coil in clinical practice in
being much higher during the first 2–3 weeks after 1992, endovascular treatment has emerged as a
the original bleed [29]. Therefore, treatment of a valid alternative to direct surgical clipping. Due to
ruptured aneurysm is indicated in the vast major- technological advances, an increasing number of
ity of cases except in the very elderly who present aneurysms are treated nowadays by endovascular
with impaired neurological condition and those means. Three prospective studies have been
patients in deep coma who fail to improve after conducted in patients with ruptured aneurysms
maximal neurological and systemic resuscitation. comparing surgical clipping to endovascular
While the decision to treat is fairly clear in a coiling [31–33]. A meta-analysis of these studies
patient with a ruptured aneurysm, the decision- showed similar results in the three studies, dem-
making process is much more difficult in the onstrating unequivocally that the percentage of
patient who is found to harbor an unruptured patients with unfavorable outcome (modified
intracranial aneurysm. Detailed analyses of the Rankin score of 4–6) is significantly lower in
factors that play a role in the decision to treat patients treated endovascularly compared to
unruptured aneurysms are beyond the scope of those treated with surgical clipping. This conclu-
this chapter. sion indicates that for patients with ruptured intra-
In general, treatment of an unruptured aneu- cranial aneurysms amenable to either treatment,
rysm is recommended for patients harboring endovascular coiling is associated with a better
aneurysms that are symptomatic, those functional outcome and should be the preferred
documented to have increased in size on serial treatment modality [34]. However, concerns exist
imaging, those larger than 6 mm, and aneurysms about the long-term durability of endovascular
in certain locations that seem to harbor a worse coiling with the risk of recurrence, need for
natural history (e.g., posterior communicating retreatment, and underlying risk of long-term
artery (PComm), the posterior circulation, and risk of re-rupture of the aneurysm. Because of
the anterior communicating artery complex). these concerns, surgical clipping might be pre-
Patient age, presence of risk factors such as ferred for very young patients with ruptured
smoking and hypertension, as well as family his- aneurysms [35].
tory of a ruptured intracranial aneurysm also play There are no available randomized studies
a pivotal role in the decision to treat an unruptured comparing endovascular to surgical treatment in
aneurysm. The patient’s overall attitude about the patients with unruptured intracranial aneurysms.
knowledge of having an aneurysm and whether or Overall, given the perceived less invasiveness of
not this has impacted negatively their quality of endovascular treatment, an increasing number of
life is yet another factor to take into account. unruptured aneurysms are treated by
Although generalizations cannot be made and endovascular means rather than surgical clipping.
538 S.F. Morales-Valero et al.

More recent developments in endovascular tech- provides high-resolution images and allows for
niques, such as stent assisted coiling [36] and flow accurate characterization of the aneurysm anat-
diversion [37], have further expanded the indica- omy (Fig. 1). Moreover, it allows immediate treat-
tions for endovascular treatment of unruptured ment if endovascular therapy is considered
aneurysms. In general, endovascular treatment is suitable. Despite its invasive nature, the compli-
preferred for unruptured aneurysms that are ame- cation rate in hands of experienced neuroradiolo-
nable to safe treatment using this modality. How- gists is very low [40]. Classically, a
ever, surgical clipping continues to have an two-dimensional digital subtraction angiography
important role when treating certain aneurysms with three different projections is obtained to pro-
of very small size or those with a geometry and vide appropriate detail. However, three-
location (particularly MCA (middle cerebral dimensional reconstruction (Fig. 1b) is increas-
artery) bifurcation aneurysms) that is unfavorable ingly being used to obtain a better morphological
for endovascular treatment. visual depiction with the advantage of manipula-
tion and free rotation of the vascular tree, which is
very useful for treatment planning. This technique
Preoperative Imaging has also been shown to improve the detection of
additional small aneurysms when compared to the
The diagnosis of a ruptured intracranial aneurysm classic two-dimensional angiography [41].
is often suggested by the presence of subarach- CTA (Fig. 2) is a fast and widely available
noid hemorrhage on non-contrast head computed modality that can be performed quickly in the
tomography (CT) in patients with the typical clin- acute setting after subarachnoid hemorrhage.
ical presentations [38, 39]. However, many aneu- The sensitivity and specificity of this modality
rysms are detected incidentally during brain are reported as 98 % and 100 %, respectively
imaging studies performed in the workup of neu- [42]. It is possible to derive three-dimensional
rological complaints. In order to confirm the diag- renderings of the aneurysm, and this is particu-
nosis and accurately characterize these lesions, larly useful in depicting the relationship of the
proper vascular imaging techniques are necessary aneurysm to its surrounding bony structures
to accurately guide treatment decisions. From the [38]. The use of CTA has greatly increased during
surgical point of view, it is of particular interest to the last years [43], and some authors support its
assess the anatomy of the aneurysm particularly role as the sole preoperative study for intracranial
with attention to the orientation, diameter, neck aneurysms [42]. Technical advances including
width, and presence of perforator branches increased field strength (from 1.5 to 3-T) and
derived from the sac or neck of aneurysms. It is refinement of techniques such as time of flight
also useful to determine the relationship of the (TOF) have improved the diagnostic value of
aneurysm to the parent vessel and surrounding MRA over time [44]. Although this modality is
neural or osseous structures. Other features such not appropriate for the acute setting, it is widely
as the presence of calcifications at the neck level utilized in the diagnosis and characterization of
are also important, since this can imply a more unruptured aneurysms and is the method of choice
challenging treatment. in patients with absolute contraindications (preg-
Many different imaging techniques are avail- nancy, anaphylactic reactions, etc.) to contrast
able to evaluate intracranial aneurysms. Digital administration.
subtraction angiography (DSA), computed
tomography angiography (CTA), and magnetic
resonance angiography (MRA) have been Surgical Principles
implemented in the evaluation of these vascular
lesions [38]. Unquestionably, catheter angiogra- The goal of surgical treatment of intracranial
phy remains the gold standard for the diagnosis of aneurysms is complete obliteration with minimal
intracranial aneurysms [39]. This modality brain manipulation, while preserving flow in
25 Surgical Treatment of Aneurysms 539

Fig. 1 (a) Selective right internal carotid artery (ICA) aneurysm remnant after clipping (arrows). (d) Selective
digital subtraction angiography injection, oblique projec- right ICA injection shows the postoperative remnant. At
tion, showing a complex aneurysm arising from the takeoff surgery, it was found that the origin of the anterior choroi-
of the anterior choroidal artery (arrows). (b) 3-dimensional dal artery was duplicated (arrows), and therefore a small
reconstruction of the digital subtraction angiogram show- remnant was intentionally left to protect the more distal
ing the irregular shape of the aneurysm. (c) Unsubtracted duplicated anterior choroidal artery
right ICA angiogram, oblique projection, showing an

Fig. 2 (a) 3D reconstruction of computed tomography manipulation shows a different projection of the aneurysm.
angiogram in a patient with a small, 2.6 mm, posterior Here, the relationship of the neck of the aneurysm and the
inferior cerebellar artery aneurysm (arrows), showing a origin of the posterior inferior cerebellar artery is well
detailed profile of the aneurysm. (b) Rotational identified (arrows)
540 S.F. Morales-Valero et al.

parent artery and associated perforator vessels. tight and edematous brain. Additional measures
The success of the surgical procedure depends have to be considered when a prolonged
on a variety of factors, including appropriate temporary clipping is necessary, such as pharma-
anesthetic management and an organized cologic metabolic suppression. The role of
surgical technique. All of the procedures, moderate hypothermia to extend the duration of
regardless of the location, involve certain critical tolerable occlusion was felt to be protective as it
steps that allow adequate exposure and theoretically reduces the release of excitotoxic
obliteration of the aneurysm. Further principles amino acids. However, a randomized trial
have to be taken into account depending on the failed to show an effect of mild intraoperative
particular anatomic location. Once the hypothermia on neurologic outcome in patients
aneurysm has been surgically treated, there are with favorable clinical grade subarachnoid
tools available in the intraoperative setting to con- hemorrhage [45].
firm proper obliteration of the aneurysm and
ensure patency of the parent artery and perforator
branches. Critical Steps in Aneurysm Surgery
(Table 1)

Anesthetic Management
Table 1 Critical steps for the surgical treatment of intra-
Appropriate anesthetic management plays a very cranial aneurysms
important role in the surgical treatment of intra-
1. Subarachnoid Opening of cisterns and arachnoid
cranial aneurysms. The baseline clinical condition dissection planes during initial exposure can
of the patient and the rupture status of the aneu- help drain CSF
rysm often dictate the anesthetic plan. While early Dissection of arachnoid fissures
surgical treatment is the best measure to reduce exposes proximal parent artery
and allows proximal vascular
the risk of rebleeding in patients with aneurysmal control
subarachnoid hemorrhage, tight control of hemo- Sharp dissection along artery frees
dynamics and certain precautions must be aneurysm neck with focus on
implemented to minimize the risk of avoiding damage to arterial
periprocedural rupture. The transmural pressure perforators
gradient should be minimized while an adequate Trans-sylvian veins should be
preserved in order to limit venous
cerebral perfusion pressure should be maintained. congestion and edema
Appropriate prophylactic measures should be 2. Minimize CSF drainage can help brain
adopted to prevent excessive hypertension that brain retraction relaxation via opening of basal
can be present during procedures such as tracheal cisterns, fenestration of lamina
terminalis, or placement of external
intubation or surgical stimuli such as skin inci-
ventricular drain in patients with
sions or pinning for the craniotomy. During the acute hemorrhage from aneurysm
surgical procedure, pharmacologic measures can rupture
be established in order to increase brain Osmotic diuretics and steroid
relaxation, minimize brain retraction, and facili- administration minimizes cerebral
edema
tate dissection. Osmotic diuresis with mannitol
Suction device should be used for
reduces the brain tissue volume, while changes applying gentle retraction and
in the ventilatory parameters such as controlled countertraction during aneurysm
hyperventilation can achieve mild hypocapnia dissection
and reduced cerebral blood volume to provide Avoid excessive parenchymal
retraction to decrease risk of
additional brain relaxation. These latter maneu-
ischemic injury or avulsion of an
vers are especially important in patients with adherent aneurysm
acutely ruptured aneurysms who often have a (continued)
25 Surgical Treatment of Aneurysms 541

Table 1 (continued) structures. The suction device can be used to


3. Bony exposure Advanced skull base approaches apply gentle retraction as the dissection pro-
and modification of traditional gresses as well as to provide countertraction
approaches remove bony structures when cutting tissue (Fig. 3a, c). If fixed retrac-
(e.g., sphenoid ridge, anterior and
posterior clinoid, etc.) to extend the tors are necessary, these should be placed only
available operative corridor and after thorough dissection to maximize brain
minimize brain retraction that is relaxation and minimize retraction pressure.
required Drainage of cerebrospinal fluid by opening
4. Vascular Avoidance of dome manipulation the basal cisterns, fenestrating the lamina
control and careful exposure of afferent and
efferent arteries are important to terminalis, lowering an existing external ven-
establish proximal and distal tricular drain (EVD), or placement of a new
control for temporary clipping EVD intraoperatively (in ruptured cases) are
5. Temporary Temporary clipping can soften the all acceptable maneuvers. In addition, osmotic
clipping aneurysm and reduce risk of rupture
diuretics, as previously described, can be given
during aneurysm manipulation
6. Permanent Clip selection depends on size and
to decrease the brain tissue volume, and ste-
clipping direction of the aneurysm dome and roids can be administered to minimize edema
neck from retraction.
Select proper blade length to avoid 3. Maximizing exposure through bone resec-
poor visibility and inadvertent tion: Bone removal might be necessary to
occlusion of perforating vessels or
nearby cranial nerves maximize the exposure and decrease brain
Booster clips can be utilized in manipulation, for example, the drilling of the
atherosclerotic calcified aneurysms sphenoid ridge for pterional approaches. A
7. Clip inspection Clip blades should be placed variety of advanced skull base approaches
completely across neck and parallel have been used in the treatment of intracranial
to branch vessel or bifurcations to
aneurysms. While these are not necessary in
avoid small remnants
the majority of cases, they continue to have a
role in selected patients with aneurysms at
challenging locations such as the mid-basilar
1. Subarachnoid dissection (Fig. 3a–c): Intra- trunk.
cranial aneurysms are commonly located 4. Vascular control (Fig. 3d–f): The afferent
along the circle of Willis within the subarach- and efferent arteries that supply anterograde
noid space, so the surgical treatment is entirely or retrograde blood flow to an aneurysm
extraparenchymal. By opening the arachnoidal should be exposed in case temporary
planes and respecting the pia-arachnoid cover- clipping is necessary. Proximal control
ing the brain, there is no violation of the cortex. should be established as early as possible, and
Progressive opening of the subarachnoid cis- the most appropriate point is defined according
terns en route to the aneurysm allows CSF to the location of the aneurysm; the most
drainage, and a wide opening of the surgical complete and preferable is that located adja-
corridor allows mobilization of the brain with- cent to the aneurysm. Avoidance of the
out transmitting traction or undue forces on the aneurysm dome while establishing proximal
surrounding structures. The dissection usually and distal control is critical in order to
follows the path of a major artery with special prevent intraoperative rupture. If vascular con-
caution to avoid damage of small perforators. trol cannot be established, a careful dissection
2. Brain retraction: Proper view of the surgical focused on the aneurysm neck should be made,
corridor occasionally requires some degree of and the dome should be avoided to prevent
brain retraction. However, this should be rupture.
applied in a delicate fashion to avoid local 5. Temporary clipping (Fig. 3d, e): Temporary
ischemia or damage to neurovascular clips are often placed to facilitate aneurysm
542 S.F. Morales-Valero et al.

manipulation and to optimize visualization of


its anatomy, including small perforating arter-
ies and efferent vessels. In the case of complex
aneurysms, temporary clipping is especially
helpful for softening of the neck to allow for
more aggressive permanent clipping. In a
minority of cases, such as thrombosed or pre-
viously coiled aneurysms, the use of both prox-
imal and distal temporary clipping allows for
complete trapping of the segment harboring
the aneurysm.
6. Permanent clipping (Fig. 3e, f): Permanent
clipping is certainly the most important and
critical part of the surgical treatment of intra-
cranial aneurysms. The goal is to achieve com-
plete occlusion of the aneurysm while
preserving adequate flow through the parent
vessel and small perforating branches. There-
fore, complete visualization of the aneurysm
and its surrounding structures is essential
before proceeding with the permanent clip
application (Fig. 3c). Small aneurysms with
narrow necks are often treated with a single
clip. However, large aneurysms with broad
necks and complex anatomy often require mul-
tiple clips in order to achieve complete
obliteration.
7. Clip inspection: Once the permanent clips are
in place, the aneurysm can be mobilized in order
to confirm the patency of the parent artery as
well as the efferent branches and perforating
vessels. In addition, complete aneurysm obliter-
ation can be confirmed with visual inspection
and by palpating the aneurysm sac looking for
the absence of pulsatile flow. Puncture of the
dome is often done at this stage to confirm
complete exclusion by the clip.
Fig. 3 Artist depiction of the basic steps of microsurgery
for intracranial aneurysms. (a) The arachnoid surrounding
the aneurysm and its parent artery is widely opened. (b)
The proximal portion of the feeding vessel is exposed in
order to obtain proximal control. (c) The aneurysm neck is ä
circumferentially separated from surrounding tissues and
carefully inspected prior to clipping. (d) Temporary clip- Fig. 3 (continued) ideal placement of the clip blades will
ping can be used to “soften” the aneurysm and facilitate completely cross the neck of the aneurysm while preserv-
definitive clipping. In addition, presence of the temporary ing patency of the parent artery. (f) The temporary clip is
clip protects from massive bleeding in the case of removed and the blades of the permanent clip are inspected
intraoperative aneurysm rupture. When applying the tem- to ensure that clip blades completely cross the aneurysm
porary clip, the utmost care is exercised to avoid any neck without compromising the parent artery. It is impor-
compromise of small perforating vessels. (e) The perma- tant to check for any small perforators that may have
nent clip is applied while the temporary clip is in place; the inadvertently been included inside the clip blades
25 Surgical Treatment of Aneurysms 543

Intraoperative Assessment method is inexpensive, easily performed, does not


of Aneurysm Occlusion significantly prolong the operative time, and can
be used repeatedly if clip adjustments are
Visual inspection alone is often insufficient to required. Although partial aneurysm occlusion
confirm proper clip placement. Therefore, various can be easily detected due to persistent flow in
techniques have been developed to assess aneu- the aneurysmal dome, the residual neck of a par-
rysm occlusion as well as parent artery and perfo- tially obliterated or thrombosed aneurysm cannot
rating branch patency. In the intraoperative be detected with this modality because of the lack
setting, these techniques can be used alone or in of flow in the dome.
combination to further adjust surgical clips.
Adjustments are undertaken to avoid vessel ste-
nosis, occlusion, and subsequent cerebral ische- Indocyanine Green Fluorescent
mia or aneurysm remnant, which may predispose Videoangiography
to aneurysm recurrence and rupture.
Near-infrared indocyanine green (ICG) fluores-
cent videoangiography is a relatively new method
Intraoperative Angiography for intraoperative assessment of blood flow during
aneurysm surgery. ICG is a near-infrared fluores-
Intraoperative digital subtraction angiography cent compound that is injected intravenously and
(DSA) is considered by many as the gold standard binds to plasma proteins to reach the cerebral
for assessment of aneurysm occlusion and vessel vasculature within 30 s. A near-infrared light
patency. The rate of clip readjustment after source and a near-infrared-sensitive video camera,
intraoperative angiography is highly variable and which are included in most modern surgical
ranges between 8 % and 34 %. Images in different microscopes, are necessary to visualize ICG in
projections can be obtained for comparison with the vasculature. This technique allows for real-
the preoperative studies and to ensure proper clip time visualization of aneurysm filling and patency
placement. Despite its invasive nature, the com- of efferent vessels or perforating branches within
plication rate is minimal in experienced hands, the surgical field (Fig. 4). This method of
and many authors advocate the routine use of intraoperative assessment is especially useful for
intraoperative angiography in all cases of aneu- evaluating patency of very small perforating
rysm clipping. However, it is not widely available, branches (Fig. 4c–e), as many of these are
requires the presence of trained personnel during beyond the resolution of standard DSA. Recent
the surgical procedure, and is time-consuming. studies have shown similar rates of clip
Moreover, some small perforators are beyond its readjustment when comparing intraoperative
resolution and cannot be visualized. With the ICG videoangiography to intraoperative DSA,
availability of newer techniques of intraoperative supporting the routine use of ICG
imaging to verify proper clip placement, this tech- videoangiography while DSA is utilized only in
nique is now used in selected cases, particularly selected cases.
when treating large and complex aneurysms.

Surgical Approaches
Microvascular Doppler
Ultrasonography The goal of aneurysm surgery is to attain occlu-
sion of the aneurysm while preserving flow in the
Micro-Doppler ultrasonography is a noninvasive associated vasculature. The majority of aneu-
method often used during intracranial aneurysm rysms involve proximal arterial branches at or
surgery for the assessment of parent vessel close to the base of the brain. A handful of surgical
patency and complete aneurysm occlusion. This approaches along with some of their variations are
544 S.F. Morales-Valero et al.

Fig. 4 (a) Intraoperative high-magnification view of an small perforating vessel (arrows) that is incorporated in
irregular anterior communicating artery aneurysm after the distal neck of the aneurysm. (d) Intraoperative view of
surgical exposure, with areas of lobulations (arrows) indi- aneurysm after multiple-clip reconstruction of this com-
cating possible areas of weakness in the aneurysm wall. (b) plex aneurysm, with sparing of the associated small perfo-
ICG fluorescent videoangiography view showing the aneu- rating vessel (arrows). (e) Post-clipping ICG fluorescent
rysm prior to clipping. This image is obtained to establish videoangiography shows no evidence of the previously
the baseline against which post-clipping ICG fluorescent visualized aneurysm, while patent flow is preserved
videoangiography can be compared. (c) Baseline ICG through the small perforating vessel (arrows)
fluorescent videoangiography showing presence of a

utilized in the treatment of these aneurysms: the versatility and access to most anterior circulation
pterional (and its numerous variations), the aneurysms and many posterior circulation aneu-
subtemporal, the interhemispheric, and the rysms. Not only is it the preferred exposure for
retrosigmoid/far lateral. A detailed description of most aneurysms, but it is also widely utilized to
these approaches and their pros and cons is treat other vascular and nonvascular lesions of the
beyond the scope of this chapter. sellar/parasellar region, the orbital roof and fis-
sure, the cavernous sinus, as well as the frontal
and anterior temporal regions [46]. The patient is
Pterional Craniotomy positioned supine with the head fixed in three-
point fixation, slightly rotated to the contralateral
The pterional craniotomy, also known as the side, and extended so that the ipsilateral zygoma
frontotemporal craniotomy, is the most commonly becomes the highest point of the patient’s head.
used approach in aneurysm surgery because of its This “classic” position maximizes the effects of
25 Surgical Treatment of Aneurysms 545

gravity in allowing the brain to “fall down” after Table 2 Craniotomy selection for intracranial aneurysms
bony opening, thus minimizing the need for by location
mechanical retraction. The skin incision is curvi- Surgical approach Aneurysm location
linear, starting 1 cm anterior to the tragus begin- Pterional approach and ICA, anterior
ning at the level of the zygomatic arch, and arcs its modifications communicating artery
(AComm), middle cerebral
forward behind the hairline. artery, proximal anterior
Over the years, numerous modifications of the cerebral artery, posterior
“classic” pterional approach have been described cerebral artery, basilar
and applied to the treatment of intracranial aneu- bifurcation, superior
cerebellar artery
rysms. With advances in skull base techniques,
Interhemispheric Distal anterior cerebral
some skull base-specific approaches have been approach artery, pericallosal artery,
proposed to treat intracranial aneurysms. Of callosal marginal artery
these, the orbitozygomatic craniotomy has gained Far lateral approach/ Vertebral artery,
traction in the past as a variation of the pterional retrosigmoid and vertebrobasilar junction,
suboccipital approach mid-basilar artery, anterior
approach. With this approach, complete or
inferior cerebellar artery,
partial removal of the frontal or temporal posterior inferior cerebellar
portion of the zygomatic arch provides the artery
required exposure to minimize the need for brain Petrosal approach Mid-basilar artery, anterior
retraction. On the other hand, the impetus for less inferior cerebellar artery
invasive techniques in recent years has led to a
progressive reduction of the size of the craniot-
omy for the treatment of most anterior circulation
aneurysms. These include development of the Postoperative Care
“key-hole” approach and the lateral supraorbital
approach. There are a variety of different Following craniotomy and clipping of aneurysms,
approaches for the treatment of most anterior cir- patients are typically admitted to the neurological
culation aneurysms from which to choose from, intensive care unit. In the absence of complica-
and the selection of one modification of the tions, patients are awakened and extubated in the
pterional approach over another is often related operating room. In the presence of immediate
to surgeon preference, degree of comfort with a postoperative deficits, an immediate CT scan
specific approach, and aneurysm characteristics or should be performed. If imaging studies show
position. evidence of vessel compromise responsible for
these deficits, return to the operating room may
be considered. However, this is exceedingly rare
Other Surgical Approaches with the available tools utilized intraoperatively
for the Treatment of Aneurysms nowadays.
Patients are usually observed in the neurolog-
Approaches other than the pterional and its mod- ical intensive care unit overnight, though the
ifications are utilized for aneurysms in specific patients with ruptured aneurysms are observed
locations [47, 48]. These approaches and their for a longer period of time because of the risk of
respective indications include the subtemporal SAH-related complications such as vasospasm
approach for some aneurysms of the upper basilar and hydrocephalus. With modern neurosurgical
artery and the proximal posterior cerebral artery, techniques and high-power microscopes, surgery
the interhemispheric approach for aneurysms of for intracranial aneurysms is usually done within
the pericallosal artery, and the retrosigmoid and the subarachnoid space, which minimizes the
the far lateral craniotomy for aneurysms of the amount of brain manipulation that is required.
lower basilar trunk and the origin of the posterior With careful preservation of venous structures,
inferior cerebellar artery (Table 2). postoperative seizures are uncommon. Therefore,
546 S.F. Morales-Valero et al.

the routine use of anticonvulsant medications is discontinued after 6 months to 1 year. An aneu-
not recommended. Anticonvulsant medications rysm remnant can occasionally be seen after clip-
delay recovery due to the high incidence of side ping of an aneurysm. Sometimes, the remnant is
effects such as fatigue and drowsiness seen with left intentionally in order to decrease the risk of
even the last-generation agents. If there are no compromise to the parent artery (Fig. 1d). At other
postoperative complications, intravenous fluids times, the remnant can be an unexpected finding
are discontinued the day after the operation, and on postoperative imaging studies. In general,
the diet is advanced as tolerated. Similarly, Foley small remnants after clipping of an aneurysm
catheter and arterial lines are disconnected early have a benign natural history and can be followed
and the patient is rapidly mobilized. over time, especially if their correction is associ-
ated with a high risk of potential complications.

Complications
Follow-Up of Surgically Treated
Complications related to surgical treatment of Aneurysms
intracranial aneurysms are uncommon due to the
use of advanced microsurgical techniques in com- We usually obtain an immediate follow-up vascu-
bination with intraoperative tools such as electro- lar study to document completeness of aneurysm
physiologic monitoring, microvascular Doppler exclusion and to rule out complications. Although
ultrasound, ICG videoangiography, and this was traditionally done using catheter angiog-
intraoperative catheter angiography in select raphy, CTA with 3D reconstruction has been
cases. However, symptomatic epidural and sub- increasingly relied upon in recent years for post-
dural hematomas can be seen postoperatively in operative imaging. With modern software, arti-
less than 1 % of patients, and ischemic complica- facts related to the clips can be minimized, and
tions are still a risk despite use of these advanced good correlation with postoperative angiography
intraoperative tools and microsurgical techniques. has been shown. Long-term follow-up of surgi-
Ischemic complications are often related to cally treated aneurysm has established surgery as
manipulation of very small perforating vessels a durable treatment modality with an exceedingly
during the dissection of the aneurysm or their low incidence of recurrence after adequate clip-
compromise during clip application. This occurs ping. Patients who presented with subarachnoid
more frequently in areas rich in small perforating hemorrhage have a higher cumulative incidence
vessels, where the aneurysm sac is often adherent of recurrent hemorrhage when treated with
to or in close relationship with vessels like the coiling as compared to clipped aneurysms
basilar bifurcation or the anterior communicating (2.6 % vs. 0.4 %). However, recurrent
artery complex (Fig. 4d). hemorrhage can be also related to de novo aneu-
With advanced microsurgical techniques, post- rysms [1, 49, 50]. Follow-up imaging every
operative seizures are an uncommon occurrence, 5 years can be considered in patients who
but when encountered, an urgent CT scan should presented with SAH in order to rule out de novo
be obtained to rule out the possibility of an intra- aneurysm formation, although no study has defin-
cranial hematoma. They can also be the conse- itively shown the validity of this approach in
quence of a subpial dissection and compromise of preventing future SAH. In the presence of a
the cortical mantle. If the patient suffers a postop- known remnant, follow-up imaging study with
erative seizure, anticonvulsant medications are CTA should be performed approximately 1 year
started. In the absence of repeated seizures and after the original surgery. In the presence of a
after an EEG rules out any evidence of an active stable remnant, further follow-up can be consid-
seizure focus, the anticonvulsants are ered every 2–3 years thereafter.
25 Surgical Treatment of Aneurysms 547

Summary 8. Nishioka H (1966) Results of the treatment of intracra-


nial aneurysms by occlusion of the carotid artery in the
neck. J Neurosurg 25:660–704
In conclusion, despite rapid advances in 9. Poppen JL, Fager CA (1960) Intracranial aneurysms.
endovascular techniques, traditional surgical clip Results of surgical treatment. J Neurosurg 17:283–296
ligation continues to be an important therapeutic 10. Jha AN, Butler P, Lye RH, Fawcitt RA (1986) Carotid
option for patients with ruptured and unruptured ligation: what happens in the long term? J Neurol
Neurosurg Psychiatry 49:893–898
aneurysms. Surgical clipping provides higher 11. Drake CG, Peerless SJ, Ferguson GG (1994) Hunterian
degree of complete aneurysm exclusion when proximal arterial occlusion for giant aneurysms of the
compared to endovascular techniques and carries carotid circulation. J Neurosurg 81:656–665
a lower risk of aneurysm recurrence over time. 12. Hopkins LN, Grand W (1979) Extracranial-intracranial
arterial bypass in the treatment of aneurysms of the
Strict adherence to microsurgical principles focus- carotid and middle cerebral arteries. Neurosurgery
ing on minimizing brain manipulation, extensive 5:21–31
arachnoid dissection, and utilization of 13. Fox AJ, Vinuela F, Pelz DM, Peerless SJ, Ferguson
intraoperative adjuncts are the key to low compli- GG, Drake CG et al (1987) Use of detachable
balloons for proximal artery occlusion in the treatment
cation rates in aneurysm surgery. Noninvasive of unclippable cerebral aneurysms. J Neurosurg
vascular imaging studies such as MRA and CTA 66:40–46
play an ever-increasing role in the preoperative 14. Higashida RT, Halbach VV, Dowd C, Barnwell SL,
and postoperative assessment of patients with Dormandy B, Bell J et al (1990) Endovascular detach-
able balloon embolization therapy of cavernous carotid
intracranial aneurysms undergoing surgical artery aneurysms: results in 87 cases. J Neurosurg
treatment. 72:857–863
15. Elhammady MS, Wolfe SQ, Farhat H, Ali Aziz-Sultan-
M, Heros RC (2010) Carotid artery sacrifice for
unclippable and uncoilable aneurysms: endovascular
occlusion vs common carotid artery ligation. Neuro-
References surgery 67:1431–1436, discussion 1437
16. Serbinenko FA (1974) Balloon catheterization and
1. Rinkel GJ, Algra A (2011) Long-term outcomes of occlusion of major cerebral vessels. J Neurosurg
patients with aneurysmal subarachnoid haemorrhage. 41:125–145
Lancet Neurol 10:349–356 17. Roski RA, Spetzler RF, Nulsen FE (1981) Late com-
2. Hackett ML, Anderson CS (2000) Health outcomes plications of carotid ligation in the treatment of intra-
1 year after subarachnoid hemorrhage: an international cranial aneurysms. J Neurosurg 54:583–587
population-based study. The Australian Cooperative 18. Fujiwara S, Fujii K, Fukui M (1993) De novo aneu-
Research on Subarachnoid Hemorrhage Study Group. rysm formation and aneurysm growth following thera-
Neurology 55:658–662 peutic carotid occlusion for intracranial internal carotid
3. Hop JW, Rinkel GJ, Algra A, van Gijn J (1997) Case- artery (ica) aneurysms. Acta Neurochir (Wien)
fatality rates and functional outcome after subarach- 120:20–25
noid hemorrhage: a systematic review. Stroke 19. Larson JJ, Tew JM Jr, Tomsick TA, van Loveren HR
28:660–664 (1995) Treatment of aneurysms of the internal carotid
4. Suarez JI, Tarr RW, Selman WR (2006) Aneurysmal artery by intravascular balloon occlusion: long-term
subarachnoid hemorrhage. N Engl J Med 354:387–396 follow-up of 58 patients. Neurosurgery 36:26–30,
5. Haley EC Jr, Kassell NF, Torner JC (1992) The inter- discussion 30
national cooperative study on the timing of aneurysm 20. Matouk CC, Kaderali Z, terBrugge KG, Willinsky RA
surgery. The North American experience. Stroke (2012) Long-term clinical and imaging follow-up of
23:205–214 complex intracranial aneurysms treated by
6. Lovelock CE, Rinkel GJ, Rothwell PM (2010) Time endovascular parent vessel occlusion. AJNR Am J
trends in outcome of subarachnoid hemorrhage: Neuroradiol 33:1991–1997
population-based study and systematic review. Neurol- 21. Arambepola PK, McEvoy SD, Bulsara KR (2010) De
ogy 74:1494–1501 novo aneurysm formation after carotid artery occlusion
7. Nieuwkamp DJ, Setz LE, Algra A, Linn FH, de Rooij for cerebral aneurysms. Skull Base 20:405–408
NK, Rinkel GJ (2009) Changes in case fatality of 22. Javedan SP, Deshmukh VR, Spetzler RF, Zabramski
aneurysmal subarachnoid haemorrhage over time, JM (2001) The role of cerebral revascularization in
according to age, sex, and region: a meta-analysis. patients with intracranial aneurysms. Neurosurg Clin
Lancet Neurol 8:635–642 N Am 12:541–555, viii
548 S.F. Morales-Valero et al.

23. Sughrue ME, Saloner D, Rayz VL, Lawton MT (2011) seen in the international subarachnoid aneurysm trial? J
Giant intracranial aneurysms: evolution of manage- Neurosurg 108:437–442
ment in a contemporary surgical series. Neurosurgery 36. Lanzino G, Kanaan Y, Perrini P, Dayoub H, Fraser K
69:1261–1270, discussion 1270–1261 (2005) Emerging concepts in the treatment of intracra-
24. Lawton MT, Spetzler RF (1999) Surgical strategies for nial aneurysms: stents, coated coils, and liquid embolic
giant intracranial aneurysms. Acta Neurochir Suppl agents. Neurosurgery 57:449–459, discussion
72:141–156 449–459
25. Leake CB, Brinjikji W, Kallmes DF, Cloft HJ (2011) 37. D’Urso PI, Lanzino G, Cloft HJ, Kallmes DF (2011)
Increasing treatment of ruptured cerebral aneurysms at Flow diversion for intracranial aneurysms: a review.
high-volume centers in the united states. J Neurosurg Stroke J Cereb Circ 42:2363–2368
115:1179–1183 38. Hacein-Bey L, Provenzale JM (2011) Current imaging
26. Barker FG 2nd, Amin-Hanjani S, Butler WE, Ogilvy assessment and treatment of intracranial aneurysms.
CS, Carter BS (2003) In-hospital mortality and mor- AJR Am J Roentgenol 196:32–44
bidity after surgical treatment of unruptured intracra- 39. Connolly ES Jr, Rabinstein AA, Carhuapoma JR,
nial aneurysms in the united states, 1996–2000: the Derdeyn CP, Dion J, Higashida RT et al (2012)
effect of hospital and surgeon volume. Neurosurgery Guidelines for the management of aneurysmal sub-
52:995–1007, discussion 1007–1009 arachnoid hemorrhage: a guideline for healthcare pro-
27. Rabinstein AA, Lanzino G, Wijdicks EF (2010) fessionals from the American Heart Association/
Multidisciplinary management and emerging therapeu- American Stroke Association. Stroke J Cereb Circ
tic strategies in aneurysmal subarachnoid 43:1711–1737
haemorrhage. Lancet Neurol 9:504–519 40. Fifi JT, Meyers PM, Lavine SD, Cox V, Silverberg L,
28. Bederson JB, Connolly ES Jr, Batjer HH, Dacey RG, Mangla S et al (2009) Complications of modern
Dion JE, Diringer MN et al (2009) Guidelines for the diagnostic cerebral angiography in an
management of aneurysmal subarachnoid hemorrhage: academic medical center. J Vasc Interv Radiol: JVIR
a statement for healthcare professionals from a special 20:442–447
writing group of the stroke council, american heart 41. van Rooij WJ, Sprengers ME, de Gast AN, Peluso JP,
association. Stroke 40:994–1025 Sluzewski M (2008) 3d rotational angiography: the
29. Jane JA, Kassell NF, Torner JC, Winn HR (1985) The new gold standard in the detection of additional intra-
natural history of aneurysms and arteriovenous cranial aneurysms. AJNR Am J Neuroradiol
malformations. J Neurosurg 62:321–323 29:976–979
30. Etminan N, Beseoglu K, Barrow DL, Bederson J, 42. Westerlaan HE, van Dijk JM, Jansen-van der Weide
Brown RD Jr, Connolly ES Jr et al (2014) Multidis- MC, de Groot JC, Groen RJ, Mooij JJ et al (2011)
ciplinary consensus on assessment of unruptured intra- Intracranial aneurysms in patients with subarachnoid
cranial aneurysms: proposal of an international hemorrhage: Ct angiography as a primary examination
research group. Stroke J Cereb Circ 45:1523–1530 tool for diagnosis–systematic review and meta-
31. Koivisto T, Vanninen R, Hurskainen H, Saari T, analysis. Radiology 258:134–145
Hernesniemi J, Vapalahti M (2000) Outcomes of 43. McDonald JS, Kallmes DF, Lanzino G, Cloft HJ
early endovascular versus surgical treatment of rup- (2013) Use of ct angiography and digital
tured cerebral aneurysms. A prospective randomized subtraction angiography in patients with
study. Stroke J Cereb Circ 31:2369–2377 ruptured cerebral aneurysm: evaluation of a large
32. McDougall CG, Spetzler RF, Zabramski JM, Partovi S, multihospital data base. AJNR Am J Neuroradiol
Hills NK, Nakaji P et al (2012) The barrow ruptured 34:1774–1777
aneurysm trial. J Neurosurg 116:135–144 44. Sailer AM, Wagemans BA, Nelemans PJ, de Graaf R,
33. Molyneux A, Kerr R, Stratton I, Sandercock P, van Zwam WH (2014) Diagnosing intracranial aneu-
Clarke M, Shrimpton J et al (2002) International sub- rysms with mr angiography: systematic review and
arachnoid aneurysm trial (isat) of neurosurgical clip- meta-analysis. Stroke J Cereb Circ 45:119–126
ping versus endovascular coiling in 2143 patients with 45. Todd MM, Hindman BJ, Clarke WR, Torner JC (2005)
ruptured intracranial aneurysms: a randomised trial. Mild intraoperative hypothermia during surgery for
Lancet 360:1267–1274 intracranial aneurysm. N Engl J Med 352:135–145
34. Lanzino G, Murad MH, d’Urso PI, Rabinstein AA 46. Yasargil MG, Fox JL (1975) The microsurgical
(2013) Coil embolization versus clipping for ruptured approach to intracranial aneurysms. Surg Neurol
intracranial aneurysms: a meta-analysis of prospective 3:7–14
controlled published studies. AJNR Am J Neuroradiol 47. Davies JM, Lawton MT (2014) Advances in open
34:1764–1768 microsurgery for cerebral aneurysms. Neurosurgery
35. Mitchell P, Kerr R, Mendelow AD, Molyneux A 74(Suppl 1):S7–S16
(2008) Could late rebleeding overturn the superiority 48. Park J, Hamm IS (2004) Anterior interhemispheric
of cranial aneurysm coil embolization over clip ligation approach for distal anterior cerebral artery aneurysm
25 Surgical Treatment of Aneurysms 549

surgery: preoperative analysis of the venous anatomy aneurysm trial (isat): long-term follow-up. Lancet
can help to avoid venous infarction. Acta Neurochir Neurol 8:427–433
146:973–977, discussion 977 50. Schaafsma JD, Sprengers ME, van Rooij WJ,
49. Molyneux AJ, Kerr RS, Birks J, Ramzi N, Yarnold J, Sluzewski M, Majoie CB, Wermer MJ et al (2009)
Sneade M et al (2009) Risk of recurrent subarachnoid Long-term recurrent subarachnoid hemorrhage
haemorrhage, death, or dependence and standardised after adequate coiling versus clipping of ruptured
mortality ratios after clipping or coiling of an intracra- intracranial aneurysms. Stroke J Cereb Circ
nial aneurysm in the international subarachnoid 40:1758–1763
Endovascular Treatment of Brain
Aneurysms 26
Luca Quilici and Edoardo Boccardi

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 552 Brain aneurysms are the most frequent cause of
Myths, Faith, and a Little Bit of Science . . . . . . . . . . . . 552 intracranial hemorrhage in young people. In
Stone Age (Balloon and Parent Artery
the last 40 years the treatment of brain aneu-
Occlusion) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 553 rysm has been revolutionized by the advent
Procedure (Tips and Tricks) . . . . . . . . . . . . . . . . . . . . . . . . . 553 and the growth of endovascular techniques.
Iron Age (Coiling and Balloon Remodeling) . . . . . 554 The introduction of detachable balloons by
Procedure (Tips and Tricks) . . . . . . . . . . . . . . . . . . . . . . . . . 557 Serbinenko in the mid-1970s to occlude rup-
Renaissance (Stent-Assisted Coiling) . . . . . . . . . . . . . . 563
tured or symptomatic aneurysms was the first
Procedure (Tips and Tricks) . . . . . . . . . . . . . . . . . . . . . . . . . 568 step of the neurovascular progress that goes on
today. In the early 1990s detachable coils with
Modern Times (Flow Diverters) . . . . . . . . . . . . . . . . . . . 571
Procedure Tips and Tricks . . . . . . . . . . . . . . . . . . . . . . . . . . . 577 the aid of the balloon remodeling technique
before and of self-expanding stents later were
The Promises . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
the following steps of this path. Nowadays
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579 these techniques are well established, safe,
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579 effective, and usually used all over the world,
helping to secure ruptured aneurysms and to
prevent bleeding by unruptured lesions. The
recent advent of flow-diverter stents represents
the latest improvement, pretending to obtain in
most of the cases not only the exclusion of the
aneurysm from the flowing blood but also its
complete disappearance.

Keywords
Aneurysm • Blister-like aneurysm • Brain hem-
orrhage • Fusiform • Partially thrombosed
aneurysm • Clipping • Detachable balloon •
Coil • GDC • Balloon remodeling • Stent •
Flow diverter • Flow disruptors • Resorbable
L. Quilici (*) • E. Boccardi
stent
Ospedale Niguarda – Ca’ Granda, Milan, Italy
e-mail: Luca.quilici@ospedaleniguarda.it; Edoardo.
boccardi@ospedaleniguarda.it

# Springer Science+Business Media New York 2016 551


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_6
552 L. Quilici and E. Boccardi

Introduction one person every 12–15 is the bearer of a brain


aneurysm. This observation would lead to an
Myths, Faith, and a Little Bit of Science obvious consequence, to estimate that 6–8 % of
the world’s population, over the course of their
What is an aneurysm? Providing an accurate and lives, would be affected by a brain hemorrhage
complete definition of an aneurysm is not easy. sooner or later. However, the scientific biblio-
The official definition of an aneurysm is a focal graphic data show that the rate of brain hemor-
dilation of the wall of an artery [1]. However, this rhage from a ruptured aneurysm is constant: 5–10/
is a very simplistic and unsatisfactory definition. 100,000 individuals per year in Western countries
What should the cutoff size of the dilation of the (in Japan and Finland, 20/100,000) [2–6]. It is
wall be, to define it an aneurysm? And how should obvious that only a very small percentage of the
we differentiate an aneurysm, for example, from a aneurysms that are diagnosed each day will be
normal variation in caliber of a section of an responsible for a brain hemorrhage. So the syllo-
artery? Moreover, this definition of “aneurysm” gism, unruptured aneurysm = time bomb, is most
would exclude some vascular lesions, like blister likely a myth of modern medicine, with no scien-
aneurysms, which are a particular type of dis- tific basis. Hundreds of millions of people cur-
sected lesion often invisible on angiographic rently live their lives as bearers of a brain
images. aneurysm, but only a few thousand of them will
Even if a perfect definition and classification of be affected by a brain hemorrhage from that diag-
aneurysms does not exist, sharing a common lan- nosed aneurysm during their lifetimes.
guage and universal definitions is fundamental to The purpose of the treatment of an aneurysm is
communicate thoughts, ideas, and activities. the prevention of bleeding. In the most common
Cerebral aneurysms are certainly where the cases the presentation is already a rupture with
field of neurovascular disease focuses its greatest bleeding, so the goal is to prevent further bleed-
attention and interest, likely due to the fact that ings. Historically neurosurgery has found a very
they are pretty common and probably the most good solution to exclude aneurysms from the cir-
common cause of brain hemorrhages in younger culation, by using clips which would compress the
people – frequently a dramatic, often fatal, event. aneurysmal sac and remodel the vessel to a normal
This is probably the reason why – over the past anatomy.
few decades and especially since the concept of Nowadays, endovascular treatment is in many
preventive medicine began to assert itself – the places the therapeutic option of choice in most
medical community has created the aura of the cases of brain aneurysms. The evolution of the
imminent threat of a ticking time bomb around the endovascular techniques, since its birth 40 years
idea of a brain aneurysm, even when unruptured ago, has been overwhelming. History, more than
and asymptomatic. For the majority of doctors, science, has shown that the endovascular
specialists or not, every aneurysm is a possible approach, compared to the neurosurgical
source of hemorrhage, but the fears of the medical approach, is almost always safer, simpler, and
community should come to terms with reality. cheaper [7]. Probably the intrinsic characteristics
Every day in the world, tens of thousands of of a more simple, physiologically respectful, min-
neuroradiological exams are performed with imally invasive approach have led to the gradual
various indications and – as a result of these expansion and success of endovascular therapy
exams – thousands of new, totally asymptomatic, over the past four decades. The evolution of mate-
aneurysms are incidentally identified. The more rials and techniques has provided a further boost
diagnostic techniques progress, the more in-depth to the remarkable progress of the method. Even if
and detailed tests are carried out, with new, in the late 1970s it was only possible to occlude
smaller and smaller, aneurysms being diagnosed. the parent vessel of an aneurysm with homemade
Recent estimates suggest that, globally, roughly balloons, nowadays, with the flow diverters, it is
26 Endovascular Treatment of Brain Aneurysms 553

possible to occlude the aneurysm, and obtain the The idea and the technique of detachable bal-
definitive disappearance of the lesion. loons became widespread and had a significant
impact on intervention strategy for direct arterio-
venous fistulas and brain aneurysms. Starting
Stone Age (Balloon and Parent Artery from there, therapeutic endovascular artery occlu-
Occlusion) sion has become routine all over the world, in all
cases of inoperable aneurysms [12, 13]. Even
The beginning of the “endovascular age” is dat- today, the choice of occluding the parent vessel
able to the mid-1970s [8, 9]. Until that time, remains a valuable and safe option in cases of
before the discovery of CT and MRI, catheter extremely complex lesions, such as giant aneu-
angiography was only one of the few diagnostic rysms of the internal carotid or dissected aneu-
tools useful in identifying vascular lesions or rysms (Fig. 2).
other space-occupying lesions that conditioned
displacement of the brain vessels. At that time
the treatment of a brain aneurysm was exclusively Procedure (Tips and Tricks)
a surgeon affair. The surgeon, in the case of a
ruptured aneurysm or a symptomatic one, decided The procedure of vessel occlusion has to be pre-
whether, how, and when to place one or more viously confirmed by an occlusion test. The test
clips, on the basis of angiographic images and allows the interventionist to know whether it is
the direct exploration of the lesion. In the most possible to occlude the target vessel. A rough test
complicated cases, when clipping was impossible could be done with a manual compression at the
for the site, or for the size of the sac, the aneurysm neck of the vessel one plans to occlude while
would be wrapped, a technique with limited effi- carrying out angiographic runs of the other ves-
cacy. Then it came the time of detachable bal- sels. To obtain more complete information on
loons. These devices allowed the exclusion of collaterals and safety, it is necessary to perform a
arteriovenous fistulas, especially the carotid- balloon test occlusion (BTO).
cavernous type, the occlusion of vessels, and in The BTO is done with the patient awake and
some cases the filling of aneurysms. The idea of under systemic anticoagulation with heparin
the detachable balloon catheter was conceived by (80 IU/kg). Two femoral sheaths must be placed:
a Russian neurosurgeon, Fedor Serbinenko. He one for the guiding catheter and the other one for
developed the idea and produced the balloons in angiographic control from the other vessels dur-
his small workshop. The balloons were made in ing the occlusion test. A complete angiographic
latex, of various sizes, and were mounted on cath- study of the cerebral circulation must always be
eters days prior to the procedure. His idea came obtained before starting the procedure. The
from observing a child playing with a balloon angiographer has to carefully evaluate the func-
inflated with helium during the May Day parade tion of the communicating arteries and the pres-
on Red Square in 1959. In 1969, Serbinenko ence of anastomosis, particularly among the
successfully performed the first endovascular branches of the external carotid artery and internal
treatment of a carotid-cavernous fistula. Over the carotid or vertebral artery. The second step is to
following decade, he executed 162 endovascular perform BTO. The BTO is realized by placing an
treatments of arterial occlusion therapy, mostly in inflatable balloon into the target vessel under fluo-
inoperable brain aneurysms. He published his roscopic guidance at the point where one wants to
experience in a Russian scientific journal in 1971 occlude the artery. The point of occlusion should
[10]. Notoriety for him and his invention arrived be chosen very carefully, in light of the previous
in 1974 with the publication of his work in a angiographic study, to avoid occluding the artery
Western scientific journal (Journal of Neurosur- in the wrong location, i.e., a location where the
gery) [11] (Fig. 1). occlusion may leave a persistent filling of the
554 L. Quilici and E. Boccardi

Fig. 1 Fedor Serbinenko, the Russian neurosurgeon that conceived the idea of detachable balloons (a) and developed the
technique of their use. Historical picture of latex detachable balloons (b). The mechanism of detachment of balloons (c)

aneurysm sac through anastomosis. The balloon is finally occlude the artery, by detaching the bal-
inflated up to the point of occluding the lumen of loon (in case of a detachable balloon) or by
the vessel; the occlusion is maintained for at least substituting the balloon with coils or a different
15–30 min during which time, through angio- occlusion device.
graphic runs; the intra- and extracranial collater- Arterial occlusion is simple and very effective,
alization is evaluated. Neuropsychological tests but it is certainly not riskless [14, 15]. Detachable
are performed to assess the possible occurrence balloons are not available anymore, mostly due to
of a neurological deficit. It is always possible to commercial reasons. For this reason in recent
perform stress tests, provoking a drug-controlled years, therapeutic occlusion is almost always
hypotension, in such a way as to highlight sub- performed using coils (Figs. 3, 4, and 5).
liminal neurological deficit.
The balloon test occlusion has two results: an
angiographic result and a clinical one. The angio- Iron Age (Coiling and Balloon
graphic test is passed only when the vein Remodeling)
opacification of the territory downstream from
the occluded artery is contemporary to the rest of In the early 1990s a new device changed forever
the brain up to, but not more than, two seconds the treatment of cerebral aneurysms: the electrical
later. The clinical test is passed if the occlusion detachable coil invented by Guido Guglielmi
does not provoke any neurological deficits. If the (Guglielmi detachable coil). Coils (metallic fila-
occlusion test is not tolerated, the balloon is ments that would curl up on themselves) were
deflated, the procedure is discontinued, and one already being used for filling aneurysms, but the
may consider the realization of a surgical bypass. problem was that by pushing them through a
If the test occlusion is instead tolerated, one can microcatheter it was almost impossible to control
26 Endovascular Treatment of Brain Aneurysms 555

Fig. 2 Preparing the detachable balloon (Goldbal2; Balt) and ½ contrast medium (b). Testing the continence of the
(a–e). A GBV2 mounted on the needle of a 2 cc syringe GBV (c). The GBV is inflated and mounted on the tip of the
and a Magic 1.2 Fr catheter (Balt) (a). Inflation test of the Magic catheter with the help of a Stylet wire (d, e)
balloon. The syringe is prepared with a solution of ½ saline

their correct final position, with the risk of losing that it was necessary to fill the sac with filaments
them in distal arteries (Fig. 6). rather than causing thrombosis [16, 17].
Guglielmi achieved the goal of controlling the The first GDCs made their appearance on the
detachment of the coils from the pusher wire. One world stage in 1991 and very quickly demon-
could finally detach the coil only once there was strated their effectiveness. The first coils had a
the evidence of a correct positioning. The detach- filament diameter of 0.010 in., and subsequently
ment is realized with an electrolytic mechanism. coils of 0.015 in. were produced, making
The aneurysm is then filled through the release of more rapid filling of large aneurysms
several consecutive coils, obtaining a small ball of possible; the 0.015 in. versions were later
thread. Initially the idea had been to cause the replaced by those measuring 0.018 in. Platinum
thrombosis of the aneurysm sac by coagulating was chosen because it is a metal that does not
the blood through an electric current necessary to undergo electrolysis and because it is spontane-
detach the coil inside the aneurysm. It was soon ously radiopaque, as well as being thrombogenic
evident that that approach would not work and and inert.
556 L. Quilici and E. Boccardi

Fig. 3 Images obtained in a 50-year-old with a giant showing the collateralizations through communicating
cavernous aneurysm symptomatic for headache. arteries and through pial collaterals (c, d). T2-weighted
Endovascular treatment by vessel occlusion. Frontal view brain MR images of the aneurysm before (e) and after
of the left internal carotid injection (ICA) (a). The occlu- 2 years from the treatment (f). Note the complete shrinking
sion of left ICA by 2 gold valve balloon (Balt) (b). Frontal of the aneurysm
views of the right ICA and the left vertebral (VA) injections

The detachment of the coil takes place by the defined as the diameter of a single loop of the
closing of an electric circuit powered by a battery, coil and the length of the entire filament, was
in which the positive pole is coupled to the tail of very limited. Nevertheless, the idea had an imme-
the coil, placed outside the microcatheter, and the diate extraordinary success. Endovascular treat-
negative pole is coupled to a needle inserted under ment with coils showed clear advantages
the skin of the patient. The passage of a compared to neurosurgical clipping: in many “dif-
low-voltage current causes the electrolysis of the ficult” cases the treatment was relatively easy, fast
micro-welding causing the detachment of the plat- and, especially, less aggressive than a neurosurgi-
inum filament. The newest detachment systems cal intervention, at the same time ensuring a more-
do not need needle insertion anymore. than-satisfactory result in excluding the aneu-
GDCs were initially used for the treatment of rysm. At the beginning of the coiling experience,
ruptured aneurysms which neurosurgeons consid- there was no scientific proof of the truth of these
ered technically impossible – or at least very dif- observations, but the efficacy of the method was
ficult – to address. It could be because of their simply evident [19–22]. Over time different com-
location (e.g., the tip of the basilar) or of their panies provided a more comprehensive range of
characteristics (Fig. 7). The selected patients sizes of coils with different degrees of stiffness
were mostly elderly or in poor clinical condition (normal, soft, ultrasoft, etc.) and with different
[18]. The range of measurements and spatial con- spatial conformations of the loops (2D, 3D, com-
formations of the initially available GDCs, plex, etc.).
26 Endovascular Treatment of Brain Aneurysms 557

Fig. 4 Images obtained in a 68-year-old man suffering after the creation of a high-flow bypass between right
from right CN III palsy caused by a partially thrombosed CCA and ipsilateral MCA (b). Note the initial slow
cavernous aneurysm. A combined surgical and downing of the ICA. Occlusion of right ICA with balloons
endovascular treatment: bypass and artery occlusion. Fron- (*, c). Frontal view of bypass injection with excellent
tal view of right ICA injection before the treatments (a). revascularization of the right hemisphere and reflow in
Frontal view of right common carotid (CCA) injection the supraclinoid ICA and in the ophthalmic artery (d)

Procedure (Tips and Tricks) usually required, and some authors would
consider also antiplatelet therapy. A good
The procedure is performed under general practice in neurointerventions is that all catheters
anesthesia to ensure immobility of the – such as the guiding catheter and the
patient and to allow the catheterization to be as microcatheter – must be constantly perfused
safe as possible. Systemic anticoagulation is using heparinized saline.
558 L. Quilici and E. Boccardi

Fig. 5 Endovascular treatment by artery occlusion with branches rising from the dissected tract. Oblique views of
coils after repeated subarachnoid bleedings in a 63-year- left and right VAs showing the complete occlusion of the
old lady. Oblique view of the left VA injection (a) showing left artery by coils and good supply of the posterior circu-
an intracranial dissection of the VA. Note the absence of lation by the right one (b, c)

It is highly recommended to start the procedure complete and compact filling of the aneurysm.
by performing an accurate angiographic study The common practice is to start with a coil of a
with 3D reconstructions of the aneurysm, in diameter corresponding to the maximal diameter
order to define the working projections that should of the aneurysm and then fill the interstices with
be used during the intervention. The projections progressively smaller coils. It is advisable to use a
must ensure the best possible view of the aneu- Y connector with rotating hemostatic valve for
rysmal neck, namely, the plane passing between both the guiding catheter and the microcatheter
the aneurysm and parent vessel. The working to ensure maximum stability of the system. The
projection should also take account of the pres- microcatheter with the microwire is tracked inside
ence of other vessels, especially if they originate the guiding catheter and under fluoroscopic vision
in the vicinity of the sac. The accurate study of 3D pushed close to the aneurysmal neck.
and 2D images is crucial in allowing the operator The entrance of the wire and entrance of the
to build a mental image of the aneurysm and its catheter into the aneurysm are two key moments,
surroundings. In addition, the 3D helps in choos- especially in the case of ruptured aneurysms. The
ing both the measures of the first coil and the maneuver should therefore be performed with
curve that the tip of the microcatheter should great care, avoiding touching the walls of the
have. The tip of the microcatheter might come aneurysm if possible. Once the aneurysm is
pre-curved in the package or it might be prepared catheterized, the microwire should be withdrawn
using steam, depending on the characteristics of and replaced with the first coil. The release of the
each single aneurysm. It is also very important to coil must be very gentle, a smooth movement,
bend the tip of the microwire. continuous, progressive, and slow. In order to
The choice of the size and shape of the coils for allow the adaptation of the coil to the walls of
the treatment is entirely operator dependent: the the aneurysm, creating a so-called basket. The
goal being to place coils in order to obtain a release of the first coil is crucial: if well realized,
26 Endovascular Treatment of Brain Aneurysms 559

Fig. 6 Guido Guglielmi (a, b) and his revolutionary coils

it allows easier filling of the basket by the subse- blood clots, along catheters or on the coils placed
quent coils. The loop protrusion outside the aneu- at the level of the aneurysmal neck. The risk of
rysmal neck in the parent artery is an occurrence thromboembolic events is directly related to the
that should obviously be avoided due to the length of the procedure. Simple and fast
chance that this will stimulate platelet aggregation procedures have major advantages in terms of
with subsequent embolism risk. After the release reducing complication rates. The ischemic
of the first coil, coils with a progressively smaller complications are best taken care of with a good
diameter are usually chosen. The filling of the sac technical approach, clean sheets and catheters,
should be continued until the coil release becomes and heparin injections. Only rarely one has to try
too difficult. to dissolve some major clot in cerebral
Along with the advantages of this technique, vessels. The use of fibrinolytics is forbidden,
its limitations have also quickly emerged. The because of high chances of having a rebleed
wider the aneurysmal neck is, the more difficult from the aneurysm. Glycoprotein IIb/IIIa inhibi-
the treatment of an aneurysm with coils is. The tors may be of help in some cases, but often the
most frequent issues with coiling have been ische- clot especially if limited in size will dissolve
mic complications determined by the formation of spontaneously.
560 L. Quilici and E. Boccardi

Fig. 7 Images from one of the oldest cases (first in Italy) Frontal views of left vertebral injection (a–c). Before the
of endovascular treatment with GDC coils in 1993. A treatment, after the detachment of the first coil, and in the
34-year-old lady with a basilar tip ruptured aneurysm. 5-year follow-up

Hemorrhages caused by the rupture of aneu- the world has highlighted another important lim-
rysms during catheterization of the sac or during itation of this type of treatment: the tendency of
the release of coils are less frequent. In this coiled aneurysms toward recanalization. Recana-
unlikely event, the only thing to do is to deploy lization is variable from aneurysm to aneurysm,
coils inside the aneurysm as quickly as possible partly depending on the coil density at the end of
until the bleeding stops (Figs. 8 and 9). treatment. Recanalization is more frequent in
Another complication of this type of treatment large and giant aneurysms and in wide-neck aneu-
is stretching of the coil: by pushing and pulling rysmal necks, but it often occurs even in those
repeatedly the platinum filament may stretch to aneurysms which appear to be perfectly filled at
the point of rupture. At that point placing the coil the end of treatment. The first cause of the phe-
into the aneurysm or withdrawing it could be nomenon has to be found in a spontaneous settling
impossible. Coil stretching was more frequent in and compaction of the coils toward the bottom of
the past. Now, with the introduction of the stretch- the sac, under the effects of blood flow and blood
resistant mechanism, constituted by a further fila- pressure. The degree of recanalization of the sac,
ment which links the first and the last loop of the as we have said, is variable and is usually
coil, stretching has become a more rare phenom- highlighted in angiographic controls already at
enon. Stretching a coil is still possible though, 6–12 months after treatment, and then, in most
particularly in cases of long and difficult position- cases, it stabilizes over a longer period of 2–5
ing with continuous advances and withdrawals of years. The main consequence of this observation
the coil. If the event occurs stretched coils may be has been that a high proportion of treated aneu-
retrieved using a retrieval system (goose neck or rysms have undergone a second, a third, or addi-
similar). If unsuccessful one can stretch even the tional endovascular treatments in pursuit of the
proximal part of the coil down into the aorta, so best possible compaction of the coils inside the
that the downward blood stream will keep it in sac, increasing, for each treatment, the overall risk
place. of clinical and technical complications [22].
Angiographic follow-up of the tens of thou- In 1994, a French neuroradiologist, Jacques
sands of aneurysms treated with coils all over Moret, devised and put into practice a strategy
26 Endovascular Treatment of Brain Aneurysms 561

Fig. 8 (continued)

for limiting the problem of wide necks. The idea positioned in the aneurysmal sac. The inflated
was to place a catheter with an undetachable com- balloon allows an easier deployment of coils
pliant balloon in the parent vessel in front of the preventing coil prolapse in the parent vessel.
aneurysmal neck and inflate it during the release Moreover, the presence of the inflated balloon
of the coils through a second catheter previously allows to place more coils in the sac, but most
562 L. Quilici and E. Boccardi

Fig. 8 Endovascular treatment of a ruptured aneurysm of consecutive steps of the procedure (b–e). The positioning
the AComm. Images obtained in a 52-year-old lady who of the microcatheter in the aneurysmal sac (b). Progressive
suffers of a subarachnoid hemorrhage. Preliminary 3D filling of the lesion with consecutive deployment and
rotational angiography (a) and frontal view of the left detachment of four coils (c, d). Final result after removal
internal carotid injection showing the work projection. of the catheter (e)
AP views subtracted and un-subtracted showing the

importantly, in cases of a rupture of the aneurysm, procedure is obviously best performed in a condi-
the balloon can immediately stop the bleeding. tion of systemic heparinization in the majority of
Moret named this advance in endovascular cases.
techniques the remodeling technique. This Endovascular treatment of aneurysms of the
improvement quickly proved its effectiveness bifurcation became possible thanks to the contem-
and allowed the treatment of aneurysms until porary positioning of two balloons: one balloon
then considered extremely difficult [23]. for each of the branches of the bifurcation, in
Nowadays single-lumen balloon and double- order to protect both of them.
lumen balloon catheters are available. The balloon Thanks to the remodeling technique the treat-
is inflated and deflated by a small (1–2 ml) syringe ment of aneurysms with coils had a further exten-
injection. The balloon is maintained inflated for sion of its indications and, during the 1990s,
the time necessary for the deployment of each became quikly the treatment of choice for all
coil, but usually for no longer than 5 min, in aneurysm [24, 25]. All over the world surgery
order to avoid ischemic complications. The was progressively replaced by the endovascular
26 Endovascular Treatment of Brain Aneurysms 563

Fig. 9 Intraprocedural rupture of an AComm aneurysm in the left ICA injection. Fast, consecutive deployment of
a 49-year-old lady. The tip of catheter punches the aneu- three coils with the arrest of bleeding (d, e) and good
rysm at the level of the neck (a, b). Immediate leakage of exclusion of the aneurysm
contrast medium (b, c) in the subarachnoid spaces during

approach, and in some countries neurovascular


surgery has progressively declined almost to the Renaissance (Stent-Assisted Coiling)
point of disappearance.
In 2002, The Lancet published the results of the Treatment with coils and the balloon remodeling
ISAT (International Subarachnoid Aneurysm technique aroused great enthusiasm and interest,
Trial) which compared the outcomes of the two and many interventionists tried new ways to
types of intervention in a population of over 2000 advance the techniques. From the mid-1990s
patients with subarachnoid hemorrhage from onward, some of them experimented sporadically
ruptured aneurysm suitable for both endovascular with intracranial stenting procedures – the idea
treatment or surgical clipping. ISAT compared being that a stent might produce the cure of an
the rate of mortality and the degree of aneurysm by changing the flow inside the sac and
disability at 2 months and at 1 year after by being a scaffold for endothelial regrowth. They
treatment. The final results showed a reduction used coronary balloon-mounted stents, but the
of 7.4 % in the absolute risk of death or depen- maneuvers were extremely difficult and cumber-
dency at 1 year from the endovascular treatment some due to the high rigidity of the systems
[26, 27]. The trial results certified what experience [28, 29]. The idea of an intracranial stent subse-
and history had already shown: endovascular quently considered the possibility to better fill the
treatment with coils is a safe and effective way difficult wide-neck aneurysms, being a support to
to treat aneurysms, and in most cases it is safer the coils, in an even more effective way than did
than neurosurgical clipping. For ruptured aneu- the remodeling technique. The need of a stent
rysms it is the treatment of choice, thanks to its dedicated to the specificity of intracranial vessels
simplicity and to its low degree of invasiveness became apparent: the new stent should be softer
(Figs. 10 and 11). and easily trackable along curves and tortuosities
564 L. Quilici and E. Boccardi

Fig. 10 The “balloon remodeling technique” of a rup- inflated (Scepter; MicroVention). Note the little adjustment
tured aneurysm in an 81-year-old lady. Frontal view of (tiny white stripes) of the loops caused by the inflation of
the right internal carotid injection before the treatment of the balloon (b). The cast of coils and the position of both
a wide-neck supraclinoid aneurysm (a). Road mapping catheter and balloon at the end of the filling (c). Final
image of the first coil deployment protected by the balloon angiogram of the right internal carotid (d)

and should be self-expandable, in order to avoid stent, preloaded in the microcatheter, was an
possible ruptures of vessels by balloon inflation open-cell stent, formed by radiolucent filaments
(Fig. 12). made of nitinol. The diameters of the devices
The first prototype of a self-expandable intra- ranged from 3 to 4.5 mm, while the lengths ran
cranial stent, the Smart, appeared in 2001: it was from 15 to 20 mm. At the two extremities of the
developed by Dr Peter Kim Nelson and his team at stent, four radiopaque markers ensured visibility
Smart Therapeutics (San Leandro, California), and controllability. The delivery catheter was 3 Fr
and renamed Neuroform in 2002 when the patent in outer diameter. The stabilizer was a second
was acquired by Boston Scientific and approved microcatheter of smaller diameter (0.25 in.) with
by the US FDA [30, 31]. The Smart system was a radiopaque marker at the distal end. The whole
composed of three parts, a self-expandable stent, a system was navigated by an exchange 0.01400
microcatheter, and a stabilizer over the wire. The wire, needed to reach the landing zone. Both the
26 Endovascular Treatment of Brain Aneurysms 565

Fig. 11 Images obtained in a 65-year-old lady with a vertebral injections (a–d). The arrowheads indicate the
ruptured basilar tip aneurysm. The endovascular treatment catheter (Echelon 10, Covidien-Ev3) and two balloons
with coils and double balloon. Frontal views of left (Transform, Striker), one in every PCA

catheter and the stabilizer were equipped with “Y” Upon reaching the area of release, the
valve connectors. The system was assembled on microcatheter was withdrawn, keeping the stent
the sterile surgical field by inserting the stabilizer in position thanks to the stabilizer; so doing, the
in the proximal end of the microcatheter stent was released at the target point, across the
containing a preloaded stent. At the time of the neck of the lesion. The Neuroform (Smart) was
release of the stent, the distal end of the stabilizer followed by other competing devices: Leo (Balt,
was placed next to the proximal end of the stent. Montmorency, France), Enterprise (Cordis,
566 L. Quilici and E. Boccardi

Fig. 12 Oblique view of left internal carotid angiograms showing the initial exclusion of the aneurysm (c). Note the
in a 53-year-old lady suffering from headache caused by a positions of the guiding catheter and the tip of the exchange
large supraclinoid aneurysm (a). An un-subtracted image wire, respectively, in the petrosal tract of ICA and in a
immediately after the deployment of a stent graft (arrow- distal branch of MCA to guarantee enough support. Final
head); the balloon is still inside (b). Left ICA angiogram left ICA angiogram (d)

Neurovascular J&J), Solitaire (Ev3, Covidien, open, and hence need an intrinsic radial force,
Paris, France), and LVIS (MicroVention, Tustin, produced in part by the material of which they
California, USA), characterized by different consist and in part by the geometry with which the
design philosophies [32–35] (Fig. 13). cells that constitute them are joined. The memory
Intracranial stents used for assisted coiling are metal alloys, at a predetermined temperature (usu-
tubular structures in cells, made of a metal alloy ally 37  C), tend to shape themselves, expanding
with shape memory, typically nitinol or cobalt- and bending in space, allowing the stents to
chromium. The stents are released into the parent achieve a preprogrammed diameter once
vessel of an aneurysm, across the aneurysm sac, unconstrained by the catheter. It is mainly the
usually before filling the aneurysm with coils. The different types of construction – open cell
self-expandable stents do not need a balloon to (Smart, Neuroform I, II, and III), braided stent
26 Endovascular Treatment of Brain Aneurysms 567

Fig. 13 Stent-assisted coiling of an aneurysm of the right (a). The presence of the stent (Neuroform, Boston S.)
superior cerebellar artery (SCA) (a–c). Images obtained in avoids the protrusion of loops in the basilar artery and in
a 36-year-old man suffering from headache. The right both PCAs. Coils have been placed away from the bottom
vertebral angiogram showing the origins of the right SCA of the aneurysm in order to keep regularly patent the SCA
and the ipsilateral PCA, respectively, from the bottom of (b, c)
the aneurysm and from the basilar artery, behind the sac

(Leo and LVIS), closed cell (Enterprise), or folded 1. The stent should be a stable support frame on
sheet (Solitaire) – which influence the character- which to create a compact cast of coil, resulting
istics of different stents: their radial force, their in a reduced incidence of prolapsing coil loops
capacity in adapting to vessel tortuosity, their in the parent vessel and of recanalization of the
opening and closing resistances, and their ability sac in the follow-up.
to retrieve the stent. Every stent has its unique 2. The stent should change the hemodynamic
combination of these characteristics. The Solitaire blood flow in the vessel in which it is released,
adds the specificity of being fully retrievable. especially at the level of the aneurysmal neck.
The rationale of using a stent in the treatment This change is related to the geometric modi-
of aneurysms is tied to three potential beneficial fication that the stent imposes on the artery -
effects: typically a distension and a straightening - as
568 L. Quilici and E. Boccardi

well as to the presence of the stent meshes placed, the filling of the aneurysm sac with coils is
which cut into and alter the flow along the usually easier and faster and the coil packing in
artery walls. the aneurysm can be denser. It might be tricky
3. The stent should become integrated into the though, because coil filaments may become
wall of the arterial vessel, covered by a layer stuck in the meshes, especially in long and diffi-
of fibroblasts and an endothelium, to further cult procedures.
help to achieve the complete healing of the The advent of stent-assisted coiling techniques
disease. has allowed a more effective treatment of wide-
neck aneurysms as well as of those lesions located
The disadvantages are mainly related to an at artery bifurcations: the basilar tip, the bifurca-
increase in the complexity and technical difficulties tion of the middle cerebral artery (MCA), the
of the procedures, resulting in longer durations of termination of the carotid artery, and the anterior
interventions. The increased complexity is due to communicating artery (AComm). As with the
the number of steps to be performed (first stenting, remodeling technique, and even more so, in
then coiling, or vice versa), as well as the need to these cases the positioning of a stent in the most
use microcatheters for releasing stents which have appropriate branch of the bifurcation determines
larger caliber than those used for coils. This the constitution of a new artificial aneurysmal
increases rigidity and decreases navigability of neck most favorable to the coiling. Thanks to the
the system. With the first generations of stents, experience gained with balloons and with the
precisely because of the reduced navigability of remodeling technique, very soon it was devised,
the proper microcatheters, it was necessary to per- in cases of bifurcation aneurysm, to put two stents,
form an exchange with a “normal” catheter and a crossed or side by side.
300 cm long 0.01400 wire at the beginning of the Operators came up with different spatial con-
procedure. Another disadvantage is that stent figurations of stents. The most commonly used is
implantation also requires the establishment of an the “Y stenting”, widely utilized for basilar tip
effective antiplatelet therapy to prevent in-stent aneurysms. A stent is placed from the posterior
platelet aggregation, responsible of increased risk cerebral artery (PCA) to the basilar artery; the
for ischemic embolic events or for the occlusion of second stent is placed symmetrically from the
the parent vessel. However, the antiplatelet therapy other PCA to the basilar artery. The second device
carries of course its own hemorrhagic risks. is released by passing through the meshes of the
first stent at the level of the aneurysmal neck. The
proximal ends of both stents are placed one inside
Procedure (Tips and Tricks) the other in the basilar trunk (Fig. 14).
One more possible configuration is the X
The introduction of self-expanding intracranial stenting: used for aneurysms of the AComm. A
stents has led operators to redesign the treatment stent is released from the A2 tract of the anterior
of aneurysms in two separate phases: the place- cerebral artery (ACA) to the A1 tract of the con-
ment of the stent and the filling of the sac with tralateral ACA. The second device is positioned
coils. Usually the positioning of the stent precedes symmetrically, opening it through the mesh of the
the release of coils inside the aneurysm. In this first stent. T, 1/2-T, and Z stenting and more are
case, the microcatheter is placed inside the aneu- very rarely used.
rysm after the deployment of the stent by passing The procedure of intracranial stenting is
the catheter through the meshes of the stent. In performed, as are all intracranial procedures,
other cases, the catheter can be placed into the under general anesthesia in order to minimize
aneurysm before the deployment of the stent the movements of the patient. The positioning of
(jailing technique). The technique is especially a stent requires a pharmacological preparation of
useful for very large wide-neck aneurysms, but it the patient with antiplatelet drugs. The prepara-
has been used also in simpler cases. Once a stent is tion typically requires at least 4–5 days of intake
26 Endovascular Treatment of Brain Aneurysms 569

Fig. 14 Oblique views of successive treatment steps in a deployment of a Neuroform stent (Boston S.) in the left
Y stenting and coiling of a basilar aneurysm in a 46-year- PCA (b), an Enterprise stent (Codman, J&J) in the right
old man (a–d). Un-subtracted images of progressive PCA (c), and coils (d)

to be effective. The drugs used most commonly in branches. The angiographic and 3D studies
many labs are clopidogrel (75 mg daily) and allow the choice of working projections for the
acetylsalicylic acid (ASA, 81–325 mg daily). placement of the stent and release of coils. The
The procedure, as already stated, should be projections for the two phases of treatment often
always be preceded by a careful angiographic do not coincide. For the choice of working pro-
study and a three-dimensional representation of jection for coiling, apply the suggestions given in
the aneurysm and of the parent vessel with its the previous section. The working projections for
570 L. Quilici and E. Boccardi

the release of the stent should depict in the best incidence and extent of recanalization of aneu-
possible way the vessel receiving the stent and rysms compared with the treatment of aneurysms
also its perforating branches with respect to the with bare coils. On the other hand, the new tech-
aneurysm. This will allow you to identify the best nique has brought with it an increase both in the
area of opening, landing, and deployment of the incidence of periprocedural ischemic complica-
stent. The preliminary angiographic and 3D tions and in their severity, because of the presence
recostructions will also be helpful in choosing of more catheters and material in cerebral arteries
the most appropriate sizes of stent and coil. The for a longer time. In addition, thrombotic occlu-
measurements (diameter and length) of the stent sions of the artery occurred more frequently, due
will have to take into account the diameters of the to ineffective antiplatelet therapy [36, 37].
receiving vessel, but one must also consider the However, the appearance of intracranial stents
changes in length that each stent undergoes has also represented a new opportunity for treat-
depending on its opening diameter. It is important ment of a particular group of aneurysms: those
to remember that for braided stents (Leo, LVIS, fusiform, partially thrombosed. This type of aneu-
Leo Baby), the smaller the diameter of the receiv- rysm is very often localized at the level of the
ing vessel is when compared with the diameter of vertebrobasilar circulation and involves long seg-
the unconstrained stent, the greater the elongation ments of these arteries. These lesions are rarely
of the stent within the vessel will be. Open- and the cause of bleeding in the brain. In most cases
closed-cell stents do not change in length, but we they behave like a neoplasm: they grow slowly
shall find out soon that this concept will be very over years to an advanced state, exerting a pro-
important for another type of device, the flow gressively worsening mass effect on the brainstem
diverters. and cranial nerves. Not infrequently, the diagnosis
The guiding catheter must have a diameter of at of these aneurysms is by the onset of an obstruc-
least 6 Fr. The delivering microcatheter inner tive hydrocephalus due to compression of the
diameter is 0.027" for an “old generation” stent, cerebral aqueduct. Before the introduction of
and can now be 0.021" or 0.017". in the cases of stents the only treatment option available, if pos-
the LVIS Jr. and Leo Baby. Both microcatheters sible, was vessel occlusion by balloon or coils.
can navigate with a 0.014 wire. As for all Some neurointerventionists started to try the use
neurovascular treatment, the guiding catheter of self-expandable stents in this kind of aneu-
and the microcatheter should be perfused with rysms. The rationale for the use of stents is not
heparinized saline (4000–5000 units per liter). It so much to help the optimal filling of the aneu-
is advisable to use the silicone on-off Y valves, to rysm sac by the coils, but rather to be a true
obtain an optimal management of the tensions and intravascular endoprosthesis. The stent should
pressures in the releasing phase of the stent. It is divert the flow in the lumen of the diseased vessel
better to push the delivering catheter into the and constitute the skeleton on which to favor the
target vessel distal to the initial point of planned creation of a new wall inside of the vessel itself, so
stent release. This will give more space to maneu- that over time the natural diseased wall of the
ver in the opening of the stent and in the apposing vessel is progressively excluded from circulation.
to the walls of the artery. In the majority of stent With this idea in mind, and having no other effec-
models, the release of the stent is achieved by tive treatment options, over the last decade there
withdrawing the microcatheter while maintaining have been many treatments of these aneurysms
the position of the stent pusher. using, in most cases, the Leo stent (Balt). The
The technique of stent-assisted coiling for the braided construction makes the Leo meshes very
treatment of cerebral aneurysms has definitely dense, so as to have some effect on blood flow. To
been another milestone along the way toward cover the segment of pathological arteries, often
affirmation of endovascular techniques. It has involving both vertebral and the basilar arteries,
made the treatment of very large wide-neck aneu- Leo stents are released one inside the other, par-
rysms possible and has enabled a reduction in the tially overlapping, to form long chains
26 Endovascular Treatment of Brain Aneurysms 571

(telescoping technique). Unfortunately, this tech- healing of the sac by changing the hemodynamic
nique has rarely been successful in these aneu- environment around it. This tool allows us to
rysms, and the results of arrested growth of these obtain the definitive cure of an aneurysm by its
aneurysms or even their healing are anecdotal disappearance, for the first time in the history of
[38, 39]. However, this technique has provided endovascular treatment of aneurysms. The devel-
the ideological and technical support on which opment of flow diverters has also changed the
to base the latest evolution of the technique of method of evaluating the success or failure of
endovascular treatment of aneurysms by flow treatment: with these devices we no longer talk
diversion (Fig. 15). merely about percentages of filling or non-filling
of the aneurysm; mostly we discuss whether or not
the aneurysm has disappeared.
Modern Times (Flow Diverters) As with the advent of coils in 1991, the first flow
diverters have been implanted for the treatment of
In 2007 the neurovascular world changed again, unruptured aneurysms previously considered tech-
as dramatically as it changed in 1991 with the nically impossible and extremely difficult or char-
advent of GDCs. In 2007, over a short period of acterized by a high probability of relapse-
time, two new intracranial devices became avail- recanalization with traditional treatments: internal
able, both the result of a new idea, a new theory in carotid aneurysms of the petrosal segment, cavern-
the treatment of aneurysms: the Silk (Balt, Mont- ous aneurysms, or para-ophthalmic aneurysms,
morency, France) and the Pipeline large and giant, aneurysms without neck or with a
(Ev3-Covidien, Paris, France). The first two flow very large neck, aneurysms from which cerebral
diverters (FDs), built by hand by weaving 48 thin arteries emerged, or recurrent aneurysms after pre-
strands of metal alloy – nitinol for the Silk and vious endovascular treatments (Fig. 16). In most
cobalt-chromium for the Pipeline – to form tubes cases it was sufficient to implant one FD for every
with very high density meshes and very low aneurysm, but in lesions without neck or with a
porosity. Released in front of the neck of the very large neck often more than one or two FDs
aneurysm, flow diverters redirect the flow in the have been implanted. They have been deployed in
parent vessel, away from the aneurysm, causing a partially overlapping fashion, one inside the other
its exclusion from circulation. Over the following to form a chain or a telescopic column.
weeks and months, this causes progressive and With a small percentage of technical failures in
spontaneous intra-aneurysmal thrombosis and the delivery of devices, and with a rate of intra-
then the formation of a clot that subsequently and periprocedural complications comparable to
retracts, leading to deflation of the sac until the those of the classical methods, the flow-diverter
aneurysm has totally shrunk. devices showed a high or very high (up to more
The impact of these devices on the than 90 %) rate of exclusion from circulation of
endovascular treatment of aneurysms has been these aneurysms in the medium term (6 months to
truly revolutionary. With the flow diverters, it is 1 year). Not only in the statistics that have taken
no longer necessary, in the great majority of cases, account of these issues over an identical follow-up
to care or worry about the aneurysm sac. The period, more than half (66 %) of these aneurysms
possibility of rupturing the aneurysm during treated with flow diverters have disappeared,
endovascular treatment no longer exists or nearly while another good proportion of these have
so. The possibility of coils dislodging during or decreased in size, mitigating or resolving the
after endovascular treatment no longer exists. mass effect on the brain and on cranial nerves
Aneurysm recanalization no longer exists or and allowing the bony structures eroded by the
nearly so. presence of the aneurysm to grow back – all these
It’s a totally different vision of endovascular by keeping the parent vessel and the vessels that
treatment of aneurysms – no more unsuccessful emerged from the sac properly open [40–42]
filling of a sac. We have finally realized the self- (Fig. 17).
572 L. Quilici and E. Boccardi

Fig. 15 A blister aneurysm treated with telescopic stents deployment of the stents one inside the other (c).
technique in a 43-year-old lady after two subarachnoid Six-month follow-up angiogram with complete exclusion
hemorrhages. The 3D rotational angiography and the left of the aneurysm (d). The un-subtracted image documents
ICA angiogram document the absence of collateral the straightening of the supraclinoid tract of ICA and of the
branches rising from the neck of the aneurysm (a). Final stents (e)
angiogram after the endovascular treatment (b). Note the
26 Endovascular Treatment of Brain Aneurysms 573

Fig. 16 Pretreatment (a, c) and follow-up (b, d) images obtained 6 months after endovascular treatment (b). Note
obtained in a 53-year-old lady with a large left VA aneu- the normal patency of PICA that rises directly from left
rysm symptomatic for CN VI palsy, treated using a flow- VA. Axial T2-weighted MR images showing pretreatment
diverter stent (PED, Covidien-Ev3). Left VA injection and aneurysm impression on the pons (c) and complete shrink-
the 3D rotational angiograms showing the aneurysm (a). ing and healing of the aneurysm in the follow-up images at
The posterior inferior cerebellar artery (PICA) arises from 1 year (d)
the lateral wall of the aneurysm. Left VA angiogram

Fig. 17 A giant cavernous right ICA aneurysm symptom- It is difficult to identify both in the angiogram and in the CT
atic for headache and CN III palsy treated with a Silk stent scan the previous presence of the aneurysm
(a, b, d). Images obtained in the 6-month follow-up (c, e).
574 L. Quilici and E. Boccardi

Fig. 18 Artery arrangement after endovascular treatment in the pretreatment, and the origin of ACA seems to be a
of a right MCA large aneurysm with a stent flow diverter. little detached from the ICA (ghost junction). After the
Pre-treatment (a) 6-month follow-up images (b) and an treatment most of perforators of MCA seem to rise from
enlarged image of follow-up (c). In the follow-up images the superior branch of MCA. All the artery arrangement
the superior branch of MCA and the ACA are thinner than occurred asymptomatic

In the light of these incredible results, soon The FD has even been used for the treatment of
flow-diverter implantation came to be performed giant, fusiform, partially thrombosed aneurysms
successfully even in “normal” aneurysms and in of the basilar artery, but again in this type of
“distal” aneurysms, placed on small arteries, the pathology, the successes are anecdotal [45, 46]
AComm, the pericallosal artery, branches of the (Fig. 19).
MCA (Fig. 18), or distal tracts of the PCA, As was the case for the Guglielmi detachable
confirming the excellent results already shown. coils, flow diverters have become, over the course
They proved their efficacy even in small aneu- of just a few years, an essential tool in the
rysms, while certainly becoming the definitive endovascular treatment of unruptured aneurysms
solution in cases of recanalization of those aneu- for nearly all the neuro-interventionists in the
rysms already treated with coils or with stent and world.
coils [43]. In the wake of this incredible success, compet-
Soon FDs were implanted in dissected aneu- itors have proposed flow-diverter devices with the
rysms, like blister lesions of the carotid siphon, same philosophy, but with different technical
responsible for recent subarachnoid hemorrhages. characteristics: Fred (MicroVention), Surpass
Blister aneurysms are lesions that are very diffi- (Stryker), P64 (Phenox), and Derivo (Acandis).
cult to treat by clipping or by coiling because their The power of the FD is very strong. When
walls are extremely fragile, like a tiny sheet of implanted, their hemodynamic influence is
paper. In these very rare cases, usually one FD is exerted not only on the aneurysm but all over the
able to prevent the rebleeding and help to create a surrounding arteries. The FDs may decrease the
new vessel wall and protect the patient [44]. flow in the arterial branches in front of which they
26 Endovascular Treatment of Brain Aneurysms 575

Fig. 19 Images obtained in a 16-year-old girl with acute angiograms before and immediately after the deployment
onset of headache and right ischemic hemiplegia caused by of a Pipeline stent (c, d). Note the dissected stenosis at the
a dissected aneurysm of MCA involving the M1 segment neck of the aneurysm and the absence of the lateral perfo-
and the perforators branches. Axial T2 FLAIR images rators of M1. Left ICA angiogram at 1-year follow-up (e)
showing the aneurysm (intramural hematoma) (a) and a showing the normalization of diameter and course of MCA
left lenticular ischemia (b). Frontal view of left ICA and its branches
576 L. Quilici and E. Boccardi

Fig. 20 Vascular plasticity after endovascular treatment artery before the treatment origins from the thinner fenes-
of a left vertebrobasilar junction aneurysm with a stent tration arm (a). The follow-up images show the progres-
FD. Oblique views of left VA angiograms obtained before sive cure of the aneurysm (star) and the changes of the
the stent deployment, at 6-month, at 1-year, and at 2-year course and diameter (b, c, d) of the AICA artery (arrow-
follow-up (a–d). The aneurysm is located on the larger arm head). All these modifications occurred totally
of a fenestration. The anteroinferior cerebellar (AICA) asymptomatic

are positioned, leading over time to a progressive intra-aneurysmal thrombosis of the aneurysm sac
reduction in the caliber of these arteries also. The excluded from circulation after stent placement
reduction in size of the vessel can be up to the can cause rupture of the sac itself. In cases of
point of angiographic disappearance, especially if aneurysms larger than 15 mm, it is possible to
the artery has a small caliber (perforator arteries) reduce the risks of this complication by adding
or has a small distribution area. The reduction of coiling of the sac, usually with a jailing technique.
size and flow occurs slowly and progressively; it The idea is that the coiling will make the throm-
takes weeks or months and the changes are almost bosis more gradual, slow, and fragmented by the
always asymptomatic. During this time the other presence of the filaments. The second expedient is
arteries, not subjected to the effect of the stent, also the administration of steroids for a few weeks
increase in caliber and flow and take charge of the of post-treatment in order to moderate the inflam-
vascular territories lost from the branches covered mation caused by the intra-aneurysmal
by the stent. In the end, the flow diverters reshape thrombosis [47].
the local anatomy, changing the territorial distri- Another new kind of complication appeared
bution of the different branches (Figs. 18 and 20). with the development of flow-diverter experience:
The dark side of FDs is mainly represented by intraparenchymal hemorrhage in locations far
the emergence of a new kind of complications, from the treated aneurysm. This is a less frequent,
never occurred before. One new kind of compli- but certainly more insidious, complication that
cation is the aneurysms rupture few weeks after usually develops within a few days after treat-
FD deployment. This type of hemorrhage is often ment, with moderate clinical-neurological conse-
fatal or severely disabling. This complication quences, rarely fatal, sometimes even
rarely happens and is more frequent with large or asymptomatic. The etiology is still unknown
giant aneurysms (in about 2–3 % of aneurysms although there has been a higher incidence in
larger than 15 mm) in which a quick and massive cases where more than one FD has been released.
26 Endovascular Treatment of Brain Aneurysms 577

At first it was thought that the bleeding had the measurements of the FD. The working projections
same pathogenesis as that which occurs with tra- should allow the best areas for opening and
ditional treatment (stents and coils), caused by the releasing the device along the parent vessel, just
perforations of arteries produced by wires during as in the procedures with conventional stents.
the treatments. Yet in the case of FD treatments, However, differently from these, they also allow
these hemorrhages often are far away from where us to perfectly locate the origin of the arterial
the microwires had been during the intervention. branches that it is possible to cover or not with
One possible explanation is that the FD itself is the the stent.
problem. Once it is released, it changes the cere- The 3D study is also useful for the choice of the
bral hemodynamic by diverting a large amount of size (diameter and length) of the FD, a choice that
blood flow to brain areas which were not “pre- is certainly crucial for the success of the procedure,
pared” to receive blood in this amount. This much more crucial than for procedures with con-
excessive perfusion caused by the FD would ventional stents. A device with an overestimated
lead to a kind of hemorrhagic infarction. Other diameter with respect to the receiving vessel will
interventionists have speculated that, during the not open properly. The cell sizes of an oversized
release and the opening of the FD, an emboliza- device will remain too large, limiting or canceling
tion occurs via a shower of angiographically their effect of flow diversion. Moreover, not
invisible air microbubbles which had been previ- reaching its nominal diameter, the device will be
ously trapped in the small meshes of the stent. much longer than expected in the vessel. Vice
This shower of microbubbles could cause the versa, an undersized FD with respect to the receiv-
occlusion of very small vessels and an ischemia ing vessel will tend to open up more than its nom-
with subsequent hemorrhagic infarction. Still inal diameter; the device will not appose correctly
much about this complication remains to the vessel walls, making the treatment ineffec-
unknown [48]. tive; and finally it might easily displace. This will
also result in a considerable shortening of the stent.
These features greatly affect the phases of opening,
Procedure Tips and Tricks release, and implantation of the stent, which are not
linear as with conventional stents.
Endovascular treatment with flow diverters is a The deployment should always be done in no
simple procedure and in most cases quicker than hurry, taking time, slowly and carefully. The
other procedures using conventional systems. All opening and the start of the release of the FD
needed is a coaxial system that consists of a 6 Fr should begin on a section of the artery down-
guiding catheter and a 0.02700 microcatheter with a stream of the desired point of landing of the
0.01400 microwire. The procedure requires phar- device. Once the distal end is opened and fixed
macological preparation with dual antiplatelet at the walls of the artery in the desired point, the
therapy. Specific technical maneuvers are rarely release of the remaining portion of the device
necessary mainy in cases where the access road to must be done in consecutive and alternating
the parent vessel of the aneurysm is complicated steps. The point is that the FD has to be pushed
or tortuous. In these instances, it is useful or and shortened in order to be well open. Only then
necessary to switch to a triaxial system interpos- the flow diversion effect will be maximal. There-
ing an distal access catheter between the guiding fore, it is necessary to carefully pull and push the
catheter and the microcatheter. The procedure, as device so that subsequent portions of it will
usual, must always be preceded by an accurate appose correctly the vessel walls. A good deploy-
angiographic study and a three-dimensional ment of an FD takes much longer than the deploy-
reconstruction of the aneurysm and the parent ment of a traditional stent: 5, 10, or even 15 min
vessel. Even more than for conventional stent versus 30 seconds. Notably, this phase, for an FD,
procedures, the 3D study is needed to choose the is much more critical than with a traditional stent.
working projections and especially the If a portion of the FD is not well deployed, or is
578

Fig. 21 Treatment of a basilar tip aneurysm with an endosaccular device WEB. Frontal and lateral views of left vertebral angiogram and 3D rotational angiography before the
treatment (a). Frontal and lateral views both un-subtracted (b) and subtracted (c) of left VA angiograms (b) after the deployment of device
L. Quilici and E. Boccardi
26 Endovascular Treatment of Brain Aneurysms 579

not completely opened, it is better to resheath it or detachable balloons by Serbinenko in the


retrieve it and restart the procedure. mid-1970s to occlude ruptured or symptomatic
As coils represent the best solution for the aneurysms was the first step of the neurovascular
treatment of ruptured aneurysms because of their progress that goes on today. In the early 1990s
simplicity, safety, and effectiveness in most cases, detachable coils with the aid of the balloon
the FD, in our opinion, equally represents the best remodeling technique before and of self-
solution for unruptured aneurysms. As previously expanding stents later were the following steps
stated, in most cases they represent a definitive of this path. Nowadays these techniques are well
cure of the aneurysm. established, safe, effective, and usually used all
over the world, helping to secure ruptured aneu-
rysms and to prevent bleeding by unruptured
The Promises lesions. The recent advent of flow diverter stents
represents the latest improvement, pretending to
Exploiting FD technology, a new type of device obtain in most of the cases not only the exclusion
has been presented in recent years: intra- of the aneurysm from the flowing blood but also
aneurysmal flow disruptors, such as the WEB its complete disappearance.
(Sequent Medical) and the Luna ev3. These
devices are small bags that behave like a plug,
which has to be positioned inside a saccular aneu-
rysm. Their structure, made by filaments like an References
FD, slows down the flowing of blood inside the
1. Osborn AG, Salzman KL, Barkowich AJ, Katzman GL
aneurysm and provokes the formation of a clot et al (2010) Diagnostic imaging brain. Amirsys, Salt
with subsequent thrombosis of the aneurysm. Lake City
Flow disruptors follow the old philosophy that 2. International Study of Unruptured Intracranial Aneu-
the target of the treatment is the aneurysmal sac rysms Investigators (1998) Unruptured intracranial
aneurysms – risk of rupture and risks of surgical inter-
and their goal is to thrombose the sac by staying vention. N Engl J Med 339:1725–1733
inside it. They are a promising technology, espe- 3. Menghini VV, Brown RD Jr, Sicks JD et al (1998)
cially for ruptured aneurysms, and in the near Incidence and prevalence of intracranial aneurysms
future they might be able to substitute coils in and hemorrhage in Olmsted County, Minnesota, 1965
to 1995. Neurology 51:405–411
emergency cases [49, 50]. 4. Ronkainen A, Hernesesiemi J, Ryynanen M (1993)
On the FD side hopefully the next generation Familial subarachnoid hemorrhage in east Finland
of flow diverters will not need any antiplatelet 1977–1990. Neurosurgery 33:787–797
therapy. Or perhaps we will come up with a 5. Juvela S, Porras M, Poussa K (2000) Natural history of
unruptured intracranial aneurysms: probability of and
resorbable flow diverter which could be easily risk factors for aneurysm rupture. J Neurosurg
positioned for any aneurysm, ruptured or 93:379–387
unruptured [51]. The dream of course would be 6. Winn HR, Jane JA Sr, Taylor J et al (2002) Prevalence
the invention of a pill, which you take in the of asymptomatic incidental aneurysms: review of 4568
arteriograms. J Neurosurg 96:43–49
morning and by night the aneurysm is gone 7. Lad SP, Babu R, Rhee MS, Franklin RL et al (2013)
(Fig. 21). Long-term economic impact of coiling vs clipping for
unruptured intracranial aneurysms. J Neurosurg
72(6):1000–1011
8. Djindjian R, Picard L, Manelfe C, Merland JJ, Theron J
Summary (1978) Development de la neuroradiologie
therapeutique. Neuroradiology 16:381–384
Brain aneurysms are the most frequent cause of 9. Wolpert SM (1974) In re: Serbinenko FA. Balloon
intracranial hemorrhage in young people. In the catheterization and occlusion of major cerebral vessels.
J Neurosurg 41:1974
last 40 years the treatment of brain aneurysm has 10. Serbinenko FA (1971) Balloon occlusion of cavernous
been revolutionized by the advent and the growth portion of the carotid artery as a method of treating
of endovascular techniques. The introduction of carotid-cavernous fistulae. Zh Vopr Neirokhir 6:3–9
580 L. Quilici and E. Boccardi

11. Serbinenko FA (1974) Balloon catheterization and aneurysms: a retrospective, multicenter study. Am J
occlusion of major cerebral vessels. J Neurosurg Neuroradiol 22:345–351
41(2):125–145 26. ISUIA (2003) International study of unruptured intra-
12. Debrun G, Lacour P, Caron J-P et al (1978) Detachable cranial aneurysms: natural history, clinical outcome,
balloon and calibrated-leak balloon techniques in the and risks of surgical and endovascular treatment.
treatment of cerebral vascular lesions. J Neurosurg Lancet 362(12):103–110
49:635–649 27. Molyneux AJ, Kerr RS, Clarke M et al (2002) Interna-
13. Fox AJ, Vinuela F, Pelz DM, Sydney I et al (1987) Use tional subarachnoid aneurysm trial (ISAT) of neurosur-
of detachable balloons for proximal artery occlusion in gical clipping versus endovascular coiling in 2143
the treatment of unclippable cerebral aneurysms. patients with ruptured intracranial aneurysms:
J Neurosurg 66:40–46 a randomised trial. Lancet 360:1267–1274
14. Langford KH, Vitek JJ, Zeiger E (1983) Migration of 28. Wakhloo AK, Lanzino G (1998) Stents for intracranial
detachable mini-balloon from the ICA causing occlu- aneurysms: the beginning of a new endovascular era?
sion of the MCA. J Neurosurg 58:430–434 Neurosurgery 43(2):377–379
15. Swann KW, Heros RC, Debrun G et al (1986) Inadver- 29. Lanzino G, Wakhloo AK (1999) Efficacy and current
tent middle cerebral artery embolism by a detachable limitations of intravascular stents for intracranial inter-
balloon: management by embolectomy. J Neurosurg nal carotid, vertebral, and basilar artery aneurysms.
64(2):309–312 J Neurosurg 91(4):538–546
16. Guglielmi G, Vinuela F, Sepetka I, Macellari V (1991) 30. Henkes H, Bose A, Felber S (2002) Endovascular coil
Electrothrombosis of saccular aneurysms via occlusion of intracranial aneurysms assisted by a novel
endovascular approach. Part 1: electrochemical basis, self-expandable nitinol microstent (neuroform). Interv
technique, and experimental results. J Neurosurg Neuroradiol 8:107–119
75:1–7 31. David F, Felipe A, Vivek D, Cameron GM (2005)
17. Guglielmi G, Vinuela F, Dion J, Duckwiller G (1991) Usefulness of the Neuroform stent for the treatment
Electrothrombosis of saccular aneurysms via of cerebral aneurysms: results at initial (3–6 mo)
endovascular approach. Part 2: preliminary clinical follow-up. Neurosurgery 56:1191–1202
experience. J Neurosurg 75:8–14 32. Mocco J, Snyder KV, Albuquerque FC et al (2009)
18. Guglielmi G, Vinuela F, Dion J, Duckwiller G, Lylyk P Treatment of intracranial aneurysms with the Enterprise
(1992) Endovascular treatment of posterior circulation stent: a multicenter registry. J Neurosurg 110:35–39
aneurysms by electrothrombosis using electrically 33. Pumar JM, Blanco M, Vasquez F (2005) Preliminary
detachable coils. J Neurosurg 77:515–524 experience with Leo self-expanding stent for the treat-
19. Moret J, Pierot L, Boulin A, Castaings L (1996) ment of intracranial aneurysms. Am J Neuroradiol
Endovascular treatment of anterior communicating 26:2573–2577
artery aneurysms using Guglielmi detachable coils. 34. Liebig T, Henkes H, Reinartz J (2006) A novel self-
Neuroradiology 38:800–805 expanding fully retrievable intracranial stent (SOLO):
20. Houdart E (1996) Traitement par spires (coils) à experience in nine procedures of stent-assisted aneu-
detechement controlé électrique de 315 aneurysmes rysm coil occlusion. Neuroradiology 48:471–478
intracraniens. Acad Natl Med 180:1173–1183 35. Turner RD, Turk A, Chaudry I (2013) Low-profile
21. Cognard C, Pierot L, Boulin A et al (1997) Intracranial visible intraluminal support device: immediate out-
aneurysms: endovascular treatment with mechanical come of the first three US cases. J Neurointerv Surg
detachable spirals in 60 aneurysms. J Radiol 5:157–160
202:783–792 36. Geyik S, Yavuz K, Yurttutan N (2013) Stent-assisted
22. Brilstra EH, Rinkel GJE et al (1999) Treatment of coiling in endovascular treatment of 500 consecutive
intracranial aneurysms by embolization with coils a cerebral aneurysms with long-term follow-up. Am J
systematic review. Stroke 30:470–476 Neuroradiol 34:2157–2162
23. Moret J, Cognard C, Weill A et al (1997) The 37. Nishido H, Piotin M, Bartolini B (2014) Analysis of
“remodeling technique” in the treatment of wide complications and recurrences of aneurysm coiling
neck intracranial aneurysms. Angiographic results with special emphasis on the stent-assisted technique.
and clinical follow up in 56 cases. Interv Neuroradiol Am J Neuroradiol 35(2):339–344
3:21–35 38. Pumar JP, Lete I, Pardo MI (2008) Leo stent
24. Aletich VA, Debrun GM, Mukesh M (2000) The monotherapy for the endovascular reconstruction of
remodeling technique of balloon-assisted Guglielmi fusiform aneurysms of the middle cerebral artery. Am
detachable coil placement in wide-necked aneurysms: J Neuroradiol 29:1775–1776
experience at the University of Illinois at Chicago. 39. Juszkat R, Nowak S, Mol S (2007) Leo stent for
J Neurosurg 93:388–396 endovascular treatment of broad-necked and fusiform
25. Cottier JP, Pasco A, Gallas S, Gabrillargues J intracranial aneurysms. Interv Neuroradiol 13:255–269
et al (2001) Utility of balloon-assisted Guglielmi 40. Saatci I, Yavuz K, Ozer C (2012) Treatment of intra-
detachable coiling in the treatment of 49 cerebral cranial aneurysms using the pipeline flow-diverter
26 Endovascular Treatment of Brain Aneurysms 581

embolization device: a single center experience with large or giant fusiform vertebrobasilar aneurysms.
long-term follow-up results. Am J Neuroradiol J Neurosurg 116:1258–1266
33:1436–1446 47. Siddiqui A, Kan P, Abla AA et al (2013) Complication
41. Piano M, Valvassori L, Quilici L (2013) Midterm and after treatment with pipeline embolization for giant
long-term follow-up of cerebral aneurysms treated with intracranial aneurysms with or without coil emboliza-
flow diverter devices: a single center experience. tion. Neurosurgery 71(2):509–513
J Neurosurg 118:408–416 48. Tomas C, Benaissa A, Herbreteau D et al (2014)
42. Lanzino G (2013) Flow diversion for intracranial aneu- Delayed ipsilateral parenchymal hemorrhage follow-
rysms. J Neurosurg 118:405–407 ing treatment of intracranial aneurysms with flow
43. Brinjikji W, Murad MH, Lanzino G (2013) diverter. Neuroradiology 56:155–161
Endovascular treatment of intracranial aneurysms with 49. Pierot L, Liebig T, Sychra V et al (2012) Intrasaccular
flow diverters: a meta-analysis. Stroke 44:442–447 flow-disruption treatment of intracranial aneurysms:
44. Çinar C, Oran I, Bozkaya H et al (2013) Endovascular preliminary results of a multi center clinical study.
treatment of ruptured blister-like aneurysms with spe- Am J Neuroradiol 33:1232–1238
cial reference to the flow-diverting strategy. Neurora- 50. Lubicz B, Klisch J, Gauvrit Y et al (2014)
diology 55:441–447 Web-endovascular treatment of wide-neck bifurcation
45. van Oel LI, van Rooij WJ, Sluzewski M et al (2013) aneurysms: short- and mid-term results in a European
Reconstructive endovascular treatment of fusiform and study. Am J Neuroradiol 35:432–438
dissecting basilar trunk aneurysms with flow diverters, 51. Wang K, Yuan S, Zhang X (2013) Biodegradable flow-
stents and coils. Am J Neuroradiol 34:589–595 diverting device for the treatment of intracranial
46. Siddiqui AH, Abla AA, Kan P et al (2012) Panacea or aneurysm: short term results of a rabbit experiment.
problem: flow diversion the treatment of symptomatic Neuroradiology 55:621–628
Postop Evaluation of Aneurysms
(Including Vasospasm) 27
Michael Mayich, Brian P. Walcott, Christopher J. Stapleton,
and Daniel Thomas Ginat

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 583 Both endovascular and microsurgical
approaches are used to treat cerebral aneu-
Overview of Treatment Options . . . . . . . . . . . . . . . . . . . 584
rysms. Endovascular techniques include coil
Imaging Modalities and Expected Posttreatment embolization, stenting, or a combination of
Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585 these. Microsurgical procedures include clip-
Vasospasm and Posttreatment Complications . . . 589 ping, without or with vessel bypass. Radiolog-
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 599 ical imaging, particularly with digital
subtraction angiography, CTA, and MRA,
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 599
plays a critical role in the posttreatment assess-
ment of patient with cerebral aneurysms. The
indications for various imaging modalities
after treatment of cerebral aneurysms, potential
artifacts related to imaging, and strategies to
mitigate these, expected imaging findings fol-
lowing treatment, as well as the imaging find-
ings for associated complications are discussed
and illustrated in this chapter.

Keywords
Endovascular • Microsurgical • Cerebral aneu-
rysms • Coil embolization • Stenting • Digital
M. Mayich
subtraction angiography • CTA • MRA • Arti-
Department of Radiology, University of Chicago, Chicago,
IL, USA facts • Complications
e-mail: Michael.Mayich@uchospitals.edu
B.P. Walcott • C.J. Stapleton
Department of Neurosurgery, Massachusetts General Introduction
Hospital, Boston, MA, USA
e-mail: Walcott.Brian@mgh.harvard.edu;
Cerebral aneurysms are rather common in the
Stapleton.Christopher@mgh.harvard.edu
general adult population, with a prevalence of
D.T. Ginat (*)
1–6 %, and aneurysm rupture accounts for the
Department of Radiology, University of Chicago, Chicago,
IL, USA majority of the estimated 30,000 cases of
e-mail: ginatd01@gmail.com nontraumatic subarachnoid hemorrhage per year
# Springer Science+Business Media New York 2016 583
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_44
584 M. Mayich et al.

in the United States [1]. The lesions can result in clipping and led to an increased acceptance of coil
considerable morbidity and mortality, particularly embolization clinically. There remains a well-
if there is rupture into the subarachnoid space. established role for both therapies; however, an
Consequently, large numbers of cerebral aneu- understanding of their features and potential com-
rysms undergo treatment on a yearly basis plications is essential for those involved in the
throughout the world. The two main treatment management of patients with intracranial aneu-
options include open surgical approaches, which rysm. Certainly, whether either type of interven-
involve clipping the base of the aneurysm with or tion is performed, medical therapy also plays an
without accompanying bypass, versus important role in the management of cerebral
endovascular techniques, which includes coil aneurysms and includes smoking cessation and
embolization, with or without accompanying blood pressure control.
stenting, and the use of more recent flow-diverting Microsurgical clipping is an open neurosurgical
devices. Diagnostic imaging plays a crucial role in procedure that excludes a saccular aneurysm from
the posttreatment evaluation of cerebral aneu- the parent circulation and offers definitive treat-
rysms. Various imaging modalities are available, ment. Aneurysm clips basically consist of blade
including computer tomography (CT)/computer and spring components and are almost universally
tomography angiography (CTA), magnetic reso- constructed of titanium in the modern era. Thus, the
nance imaging (MRI)/magnetic resonance angi- titanium aneurysm clips are MRI compatible. The
ography (MRA), and conventional catheter clips are placed under direct visualization with the
angiography. The treatment option, imaging aid of the operating microscope. A variety of sur-
modalities and associated artifact and technical gical approaches (craniotomy techniques) can be
advances, and expected and complicated utilized, which permit direct line of sight to the area
posttreatment findings related to aneurysms, with of interest. Examples include mini-pterional, lateral
an emphasis on vasospasm, are reviewed in the supraorbital, and orbital-pterional craniotomies.
following sections. Additional clinoidectomy, or removal of portions
of the clinoid process, may be necessary to more
readily access the ophthalmic segment ICA aneu-
Overview of Treatment Options rysms. In addition to bone removal, extensive
Sylvian fissure subarachnoid dissection is often
The optimal treatment of aneurysms has been a necessary to allow for the separation of the frontal
source of intense investigation which is evolving and temporal lobes in order to access circle of
as our understanding of the benefits and draw- Willis aneurysms. By creating a working space
backs of endovascular versus microsurgical ther- with exposure of the circle of Willis, the aneurysm
apeutic approaches continues to deepen. While clips can be delivered via delicate handheld appli-
endovascular approaches including coiling or cators to the neck of an aneurysm. The main goal of
flow diversion offer the possibility of avoiding a aneurysm clipping is to minimize the risk of rup-
craniotomy, there are a unique set of consider- ture. Another important goal of any surgery is to
ations to take into account when considering this preserve blood flow through the parent artery, since
approach. The International Subarachnoid Aneu- any stenosis or occlusion of the parent artery circu-
rysm Trial (ISAT) released in 2005 was a land- lation could result in cerebral ischemia. In order to
mark study which prospectively compared preserve adequate cerebral perfusion, direct extra-
endovascular coiling versus neurosurgical clip- cranial to intracranial arterial bypass is indicated
ping in aneurysms amenable to either therapy for the treatment of complex aneurysms and con-
[2]. The trial demonstrated a risk reduction of sists of anastomosing relatively large-caliber ves-
7.4 % for death or dependence at 1 year with an sels or grafts, most commonly the superficial
early survival advantage maintained for up to temporal artery (STA) to the middle cerebral artery
7 years in patients who underwent coiling versus (MCA) [3].
27 Postop Evaluation of Aneurysms (Including Vasospasm) 585

Endovascular therapy for cerebral aneurysms intervention in order to assess adequate collateral
is a minimally invasive alternative to microsurgi- circulation and risk of developing a stroke if the
cal clipping. Endovascular techniques include coil vessel is sacrificed. Vessel occlusion can be
embolization of the aneurysm sac for narrow-neck performed through the use of coils or clips.
saccular aneurysm and stent-assisted coil emboli- Ultimately, the overall success of aneurysm
zation for wide-neck saccular aneurysms or fusi- treatment can be gauged by the degree of aneu-
form aneurysms. The coils form a “mass” within rysm obliteration, patency of parent and perforat-
the lumen of the aneurysm. The formation of ing arterial vasculature, and the durability of the
thrombus associated with the coils is responsible aneurysm obliteration. Recanalization, recur-
for occluding the aneurysm. When stents are used, rence, and incomplete obliteration are complica-
these span the neck of the aneurysm in the parent tions known to occur infrequently following any
vessel in order to prevent prolapse of the coils and aneurysm treatment, and their incidence may be
maintain patency of the parent vessel. Nitinol, related to the treatment modality [5]. ISAT and
platinum, nickel, iridium, and tungsten have other studies have demonstrated the superiority of
been the primary metals used in construction of clipping compared with coiling providing long-
coils and stents, which are MRI-compatible. term protection from re-hemorrhage [2]. In ISAT,
Matrix coils have also been developed, which two patients that had documented complete angio-
consist of thin platinum coils covered with a graphic occlusion of their aneurysm following
bioabsorbable, polymeric material, such as coiling suffered from re-hemorrhage events. It
polyglycolic acid/lactide, and can accelerate aneu- should be noted that aneurysm growth after clip-
rysm fibrosis and neointima formation without ping has also been reported in several cases
parent artery stenosis. Occasionally, liquid embo- [6]. Regardless of whether an aneurysm is clipped
lization agents such as Onyx may be used in or coiled, follow-up imaging is mandated to
conjunction with coils for the treatment of large ensure the durability of treatment. The various
cerebral aneurysms. imaging modalities available for accomplishing
More recently, flow-diverting stents, such as this task and the potential complications that
the Pipeline Embolization Device and SILK may be encountered after aneurysm treatment
Flow Diverter, have been introduced for the treat- are reviewed in the subsequent sections.
ment of aneurysms without coil embolization and
are mainly indicated for large, wide-necked inter-
nal carotid artery (ICA) aneurysms. These devices Imaging Modalities and Expected
consist of a porous tubular tight mesh that can be Posttreatment Findings
positioned across the aneurysm neck while
maintaining patency of the patent vessel. The The main purpose of diagnostic imaging follow-
strategy behind the use of flow-diverting stents is ing aneurysm clipping and endovascular therapy
to channel blood flow away from the aneurysm, is to confirm adequate exclusion of the aneurysm.
which consequently thrombose. The devices are In general, CTA is superior to MRA for the eval-
composed of platinum and nickel-cobalt-chro- uation of aneurysms after surgical clipping, while
mium alloy and are thus MRI-compatible. These MRA is the preferred noninvasive imaging
flow-diverting devices provide successful and modality following coil embolization [10]. Diag-
durable occlusion of wide-neck intracranial aneu- nostic digital subtraction angiography is generally
rysms without severe complications [4]. reserved as second-line imaging modality for
As a last resort, parent vessels may be occluded addressing issues that cannot be adequately
via an open surgical or endovascular approach, answered on the noninvasive cross-sectional
particularly for posterior circulation fusiform imaging exams.
aneurysms or dissecting aneurysms. Typically a Axial source images, coronal and sagittal
balloon occlusion test is performed before such an multiplanar reformats, and interactive 3D volume
586 M. Mayich et al.

by using CTA post-processing bone subtraction


techniques. However, this approach requires an
additional scan and increased radiation dose.
The advent of dual-energy CT (DECT) imag-
ing in conjunction with Metal Artifact Reduction
Software (MARS) has been shown to improve
image quality of aneurysm, parent vessel, and
adjacent parenchyma when compared to single-
energy conventional CT examinations and offers a
potential future application for CT in surveillance
of treated lesions [8]. Additionally, DECT offers
the capability of constructing virtual unenhanced
images, offering the capability of monitoring rel-
ative contributions of hemorrhage versus contrast
to hyperattenuation in the subarachnoid space
related to pre- or post-procedure aneurysm rup-
ture, extravasated contrast from the procedure
itself, or both [9]. Furthermore, DECT can be
useful for bone subtraction without an additional
Fig. 1 Aneurysm clip. The CT image shows the spring
(arrow) and blade (arrowhead) components of a titanium
scan. Extracranial-intracranial bypass performed
aneurysm clip in conjunction with cerebral aneurysm clipping is
most commonly performed between the superfi-
cial temporal artery and MCA or between the
renderings are used to evaluate the treated aneu- occipital artery and posterior cerebral artery
rysm and the rest of the cerebral circulation. CTA (PCA). Patency of the anastomoses between
can often adequately depict small aneurysm rem- bypass vessels can be assessed on CTA and
nants, as well as demonstrate patency, stenosis, or MRA (Fig. 2). The STA often increases in caliber
vasospasm in the adjacent parent vessels. Cer- after successful revascularization.
tainly, metal artifacts can be a limiting factor for Following endovascular coil embolization,
the use of CTA following aneurysm clipping. metallic streak artifact related to platinum alloy
However, the presence of artifacts is much less coils can considerably obscure the aneurysm and
with titanium aneurysm clips compared with the parent vessel as well as the adjacent paren-
cobalt clips, and the components of the clips can chyma, limiting the usefulness of CTA. On the
even be delineated in detail (Fig. 1). In addition, other hand, several studies have demonstrated the
the orientation of the clips can affect artifacts in sensitivity of MRA for residual aneurysm after
the peri-clip region. For example, clips position- coil embolization or stent-assisted coil emboliza-
ing perpendicular relative to the parent vessel tion to be between 90 % and 100 % [9], while
results in fewer artifacts than those parallel to the others have determined that MRA may be at least
parent vessel. In addition, clips that lie oblique to as sensitive as DSA if performed carefully (Fig. 3)
the scanning plane produce fewer artifacts. [10]. TOF-MRA and contrast-enhanced MRA
Another challenge pertaining to postoperative have similar accuracy for the evaluation of coiled
via CTA is the proximity of osseous structures, aneurysms. Nevertheless, some institution proto-
such as the skull base or clinoid processes, to cols perform both techniques sequentially,
segments of the cerebral circulation. Indeed, employing CE-MRA data to supplement assess-
beam-hardening artifact from the dense bone can ment by TOF-MRA. However, with regard to
further obscure the treated aneurysm in addition to post-stent-assisted coiling, the excess susceptibil-
the effects of the clips. This issue can be mitigated ity artifacts related to the stent material on time-of-
27 Postop Evaluation of Aneurysms (Including Vasospasm) 587

Fig. 2 Aneurysm clipping with extracranial-intracranial corresponding time-of-flight MTA (c) show a patent right
bypass. Axial CT image (a) shows a right paraclinoid extracranial bypass graft and anastomosis with the right
aneurysm clip (arrow). 3D CTA image (b) and MCA (arrowhead)

Fig. 3 CT versus MRA.


Non-contrast CT (a) after
coiling demonstrates
extensive metallic streak
artifact that obscures
adjacent structures. The
corresponding time-of-
flight 3-T MRA (b)
demonstrates minimal
susceptibility artifact (*)
allowing visualization of
the patent parent vessel
(arrow) adjacent to the
coil mass
588 M. Mayich et al.

Fig. 4 The utility of


contrast MRA. (a) 3D
TOF-MRA post-stenting of
basilar fusiform aneurysm
at 3-T basilar artery
obscured by susceptibility
artifact (white arrow).
Contrast-enhanced MRA at
the same level (b)
demonstrates both
opacification of the vessel
lumen (arrow) and a small
focus of residual flow in the
aneurysm (arrowhead)

flight MRA can obscure the aneurysm neck and versus 1.5-T [12]. They also showed that the
parent vessel. Thus, in such cases, follow-up with volume of artifact depended on packing density
contrast-enhanced MRA may be more beneficial of the aneurysm, with a more densely packed
(Fig. 4). In the case of embolization devices, the aneurysm resulting in significant differences in
used CTA for follow-up is often preferable due to volume overestimation. They found that the use
even greater artifact of MRA. Indeed, the lumen of a short TE protocol improved visualization,
of the embolization devices and status post the especially on 3D TOF images, concluding that
treated aneurysms can be defined in detail on with all variables being equal, 1.5-T was superior
CTA (Fig. 5). to 3-T when 3D TOF images were used. However,
There are several variables which may be con- a prospective clinical study by Kaufmann
sidered when optimizing imaging parameters. For et al. showed no significant difference in sensitiv-
example, increasing receiver bandwidth can min- ity for detecting an aneurysm remnant between
imize susceptibility artifact related to metal 1.5-T and 3-T [14]. Ultimately, an accurate inter-
implants [11]. By minimizing the amount of time pretation of the posttreatment MRA requires a
during which echo sampling occurs (TE), familiarity with the pretreatment and immediate
susceptibility-related distortion of the image can post-embolization conventional angiograms. A
be decreased, although this occurs to the detriment review of the images from the initial treatment
of signal-to-noise ratio. While protocols may vary, provides the reader with a 2D projection of the
as a general principle, doubling the readout band- 3D anatomy, demonstrating the orientation of the
width yields good results. Shortening TE time coil mass and aneurysm neck with respect to
may also decrease susceptibility artifact related the parent vessel.
to the coil mass and/or associated hardware Digital subtraction angiography (DSA) is the
[12]. Gonner et al. found that decreasing TE modality of choice for treated aneurysm follow-up
from 6 to 2.4 ms significantly reduced the volume in cases when examination with MRA is
of susceptibility artifact associated with the coil contraindicated or noninvasive diagnostic imag-
mass and additionally allowed improved visuali- ing is inconclusive. DSA allows for exceptional
zation of the adjacent artery in 36 % of cases [13]. spatial resolution, multiplanar projection, and the
Although imaging at 3-T provides greater added dimension of dynamic temporal assessment
signal-to-noise ratio than at lower field strengths, of the contrast bolus, eliminating issues such as
the degree of susceptibility artifact associated venous contamination encountered in cross-
with the coil mass is also increased. An in vitro sectional techniques. The procedure is invasive,
study by Walker et al. showed a significant however, carrying an associated risk of neurologic
increase in the amount of susceptibility artifact complication in 2.6 %, stroke with permanent
and coil mass volume overestimation at 3-T disability in 0.14 %, and death in 0.06 %
27 Postop Evaluation of Aneurysms (Including Vasospasm) 589

Fig. 5 Pipeline device.


Pretreatment coronal CTA
(a) shows a large right
superior hypophyseal
aneurysm (*). Fluoroscopic
image (b) obtained shows
the Pipeline device
(arrowheads) in position
within the right ICA. Digital
subtraction angiography (c)
at the completion of
Pipeline Embolization
device insertion shows
persistent filling of the
aneurysm sac (*). Follow-
up CTA obtained 9 months
later shows near-complete
exclusion of the aneurysm
due to thrombosis of the sac
with minimal residual
filling of the neck (arrow)
and Pipeline device in
position (arrowheads).
Curved planar reformatted
CTA image (e) shows the
full length of the patent
flow-diverting stent (arrow)

[7, 15]. 2.6 % is a commonly quoted value,


although it is notable that this large series showed Vasospasm and Posttreatment
a decreasing rate of complications in examina- Complications
tions performed more recently in the series.
Meta-analyses also demonstrate that the risk of Aneurysmal subarachnoid hemorrhage is associ-
angiographic complication is significantly lower ated with a high mortality rate and is attributable
in patients with SAH, cerebral aneurysm, or AVM to the initial hemorrhage event or aneurysm
than in patients with TIA/stroke [16]. In general, re-hemorrhage prior to surgical or endovascular
CTA tended to overestimate aneurysm volume and obliteration. In survivors, the days that follow
MRA consistently underestimated volume, while aneurysm obliteration are a critical period where
DSA can both over- and underestimate aneurysm patients are monitored intensely for the develop-
volume, providing a more accurate value on aver- ment of cerebral vasospasm. It is this secondary
age than either of the other modalities [17]. insult of vasospasm that impairs cerebral blood
590 M. Mayich et al.

flow and is the major cause of delayed ischemia institution to institution, where various means of
and stroke [18]. Although the pathophysiology of radiographic and sonographic surveillance aid to
vasospasm is incompletely understood, it is gen- augment serial clinical examinations. In patients
erally accepted that the rapid institution of thera- with Glasgow Coma Scale scores less than
pies, including systemic hypertension and intra- 9 (comatose), these radiographic surveillance
arterial vasodilator therapy, can be used in symp- methods are the main method of vasospasm detec-
tomatic patients to prevent permanent deficits tion, as it is challenging to detect subtle differ-
[19]. Therefore, the period following aneurysmal ences in neurological exam in a poor neurological
subarachnoid, whether the aneurysm is treated with state.
clipping or coiling, represents a critical period Doppler ultrasound is a noninvasive modality
where clinical care is heavily influenced by the that can be used to measure cerebral blood flow
information provided by neuroimaging. The defi- [21]. The general concept of the modality is that
nition of cerebral vasospasm is new onset of intra- blood velocity increases as vessel diameter
cranial arterial narrowing and it typically described decreases, as is seen in the setting of vasospasm.
qualitatively (mild, moderate, or severe) as subjec- However, hyperemia can also result in elevated
tively interpreted on catheter or CT angiography velocity recordings, confounding interpretation.
[20]. Elevated transcranial Doppler velocities of A variety of measurement techniques have been
>200 cm/s not thought to be a result of hyperemia used to help differentiate between these two con-
are also classified as cases of vasospasm. Symp- ditions (vasospasm and hyperemia), such as the
tomatic vasospasm is a subgroup of vasospasm calculation of the Lindegaard ratio, which com-
where clinical neurological symptoms are referable pares the blood flow in the more proximal carotid
to a region of radiographic or sonographic vaso- artery with the MCA [22]. Identification of these
spasm in the absence of alternative explanations. differences in blood flow is potentially useful in
The purpose of any imaging surveillance for determining focal areas of vasospasm as com-
vasospasm is to identify changes in vessel caliber pared to global changes in blood flow that are
and/or blood flow prior to neurological deteriora- seen with hyperemic states or diffuse vasospasm.
tion. Not all patients with subarachnoid hemor- Drawbacks of transcranial Doppler ultrasound to
rhage from ruptured cerebral aneurysms go on to detect vasospasm are inherent in its limited sensi-
develop cerebral vasospasm. Radiographic grad- tivity and specificity. Some reports caution that
ing scales, such as the Fisher scale, have been there is a lack of evidence and lack of accuracy of
developed to describe a relationship between transcranial Doppler, criticizing its use in routine
quantity or pattern of blood and the risk of future clinical practice [23]. Nevertheless, because of its
vasospasm development. However, patients with noninvasive nature, lack of radiation exposure,
similar hemorrhage patterns and quantitative and ease of daily bedside acquisition, it is rou-
amount of subarachnoid blood may have a drasti- tinely used at several high volume neurovascular
cally different clinical course (with some patients institutions.
experiencing no vasospasm at all). This empha- CTA is also useful for the detection of cerebral
sizes that in addition to the subarachnoid hemor- vasospasm following subarachnoid hemorrhage
rhage itself, there are patient-specific factors that post-clipping, particularly when there is severe
predispose an individual to cerebral vasospasm. spasm located in proximal artery locations, such
Development of radiographic biomarkers and as the A1 segment of the anterior cerebral artery
studies assessing the efficacy of surveillance (ACA) or the M1 segment of the MCA (Fig. 6). In
imaging will be important to improve manage- addition, CT perfusion can be performed concur-
ment in these critical patients. However, there is rently to provide insight into the extent of cerebral
no evidence from randomized controlled trials ischemia resulting from vasospasm. However,
that guide the detection or management of vaso- CTA may be less accurate for detecting mild and
spasm following aneurysmal subarachnoid hem- moderate spasm in distal vessel locations
orrhage. Practice patterns vary widely from [24]. The use of CTA in patients with renal
27 Postop Evaluation of Aneurysms (Including Vasospasm) 591

Fig. 6 Cerebral vasospasm


depicted on CTA and CT
perfusion. Axial
non-contrast CT (a) shows
extensive subarachnoid
hemorrhage related to a
ruptured basilar artery
aneurysm. Axial CTA MIP
image (b) obtained several
days after aneurysm coiling
shows severe diffuse
narrowing of the proximal
circle of Willis branches,
consistent with vasospasm.
The corresponding CBV (c)
and MTT (d) maps show
perfusion deficits,
particularly severe in the
left parietal lobe (arrows)

impairment is limited secondary to the necessity that may be able to detect aberrations in cerebral
of iodinated contrast. CTA is also associated with perfusion irrespective of changes in large vessel
a radiation dose that can be significant in critical caliber. This is an important consideration
patients undergoing serial imaging in the vaso- because vasospasm of the microcirculation may
spasm “window.” Future study in the form of not routinely be detected on either CT angiogra-
randomized trials is needed to define the efficacy phy or digital subtraction angiography. The most
of either symptom-prompted CTA or routine sur- commonly used dynamic imaging technique for
veillance CTA following aneurysmal subarach- this purpose is CT perfusion, likely as a result of
noid hemorrhage and its effect on outcome. its widespread availability and ease of acquisition.
Ultimately, catheter-based angiography has Other dynamic imaging techniques, such as posi-
been considered to be the historical gold standard tron emission tomography, have also been used to
to diagnose vasospasm [25]. Advantages of identify regions of impaired regional blood flow
catheter-based angiography include its high spec- or regional cerebral metabolic rate of oxygen.
ificity and sensitivity. It also allows for the imme- Emphasis on imaging of brain perfusion may be
diate administration of intra-arterial therapy able to better evaluate for vasospasm, particularly
should severe vasospasm be detected (Fig. 7). since “large vessel” constriction may only occur
The main risks of the diagnostic procedure in a portion of affected cases.
include stroke (estimated at 1 % or less), local Periprocedural aneurysm rupture is a
access site complications, the need for iodinated documented complication of coil embolization
contrast, and radiation exposure [16]. occurring in 4.6 % of all cases and 20.4 % of
In addition to direct imaging of the cerebral balloon-assisted cases in one large series (Fig. 8)
vasculature, there are several imaging modalities [26]. While a distressing complication, the
592 M. Mayich et al.

Fig. 7 Digital subtraction angiography of cerebral vaso- stenosis of the MCA (arrow). The patient was treated with
spasm. Five days postoperatively, the patient was intra-arterial administration of milrinone and nicardipine,
suspected to have developed left MCA vasospasm based with nearly resolved spasm of the MCA (arrow), as dem-
on elevated transcranial Doppler velocities in that distribu- onstrated on follow-up lateral DSA projection (b)
tion. Initial lateral projection DSA image (a) shows severe

majority of patients demonstrated an absence of


clinical sequelae (74 %) related to the event. Early
rebleeding is also a well-documented complica-
tion following coil embolization [27]. The ISAT
study found rates of rebleeding among ruptured
aneurysms treated with coiling of 2.5 % in the first
year and 0.2 % in subsequent years. The CARAT
trial demonstrated rebleeding risk in the first
3 years to be 2.2 %, 0.2 %, and 0 %, respectively
[28]. Hemorrhage at any point following surgical
clipping is considered to be a rare event with
accumulative estimated risk of between 1 % and
2 % [3]. Unruptured aneurysms treated with coil
embolization demonstrate an exceptionally low
rate of subsequent bleeding, estimated at 0.2 %
annually in one meta-analysis [29].
Thromboembolic complications are a well-
described complication of coil embolization and
range from silent incidental findings on subse-
quent imaging to devastating findings. Silent
thromboembolic events are common, presenting
Fig. 8 Intraprocedural aneurysm rupture: DSA demon-
strates extravasation of contrast from a partially coiled as multifocal diffusion restriction on DWI
ACoA aneurysm (arrow) sequence images in the postoperative period.
27 Postop Evaluation of Aneurysms (Including Vasospasm) 593

Fig. 9 Silent
thromboembolic events. At
6 days post-coiling of a
ruptured 6 mm ACoA
aneurysm, there is
multifocal diffusion
restriction depicted on DWI
(a) and ADC (b)

Lesions are typically small, often multifocal, and compared to coil embolization alone. In addition
typically localize to the vascular territory of the to early thrombosis, stent-assisted coiling results
vessel being treated (Fig. 9). One of the earlier in the risk of both delayed in-stent stenosis and
descriptions by Rordorf et al. showed multifocal delayed thrombosis. A recent meta-analysis deter-
restricted diffusion in 61 % of patients who were mined a rate of delayed stenosis of 5.3 % (range of
imaged within 48 h of uncomplicated coiling 0–20.6 %), which correlates with a prospective
[30]. A recent larger series of 342 patients by database assessing the Enterprise stent that found
Kang et al. showed an incidence of rates of delayed stenosis and thrombosis of 3 %
DWI-positive lesions of 54.5 %, while symptoms each [34]. Rates of in-stent stenosis and thrombo-
were only detected in 4.1 % of patients, resolving sis compare favorably with rates documented fol-
in approximately half. The presence of 6 lesions lowing stenting of intracranial vessels for
was, however, correlated with the presence of atherosclerosis during the SAMMPRIS trial of
symptoms – 78.6 % of symptomatic patients had 28.3 % and 3.9 %, respectively [35].
6 lesions implying the burden of DWI could Clinically significant thromboembolism is a
potentially be employed as a surrogate marker much less common complication, although this
for symptomatic ischemia [31]. Occasionally, is the most common cause of periprocedural mor-
larger territorial infarcts result when a branch bidity associated with coil embolization of intra-
associated with an aneurysm is sacrificed or as a cranial aneurysms, reported to occur in 1–17 % of
result of parent vessel is purposely embolized cases, including 4.7 % of cases in one large retro-
(Fig. 10). Stent-assisted coiling is also associated spective series [36, 37]. During as many as 11 %
with silent thromboembolism, although limited of procedures, distal emboli or intraluminal
literature at present precludes assessment of thrombus related to the coil mass/aneurysm may
whether the risk is significantly elevated in this be demonstrated (Fig. 11) [38]. If encountered
population. Altay et al. found DWI lesions in during the coiling procedure, therapy including
30 % of 46 patients with unruptured aneurysms infusion of a glycoprotein IIB-IIIA inhibitor
treated with stent-assisted coiling [32]. Hahne- such as abciximab (ReoPro™) may be employed
mann et al. recently reported on a series of to acutely lyse thrombus, although thromboem-
75 patients treated with stent assistance in which bolic complications occurring after the comple-
48 (64 %) were found to develop ischemic lesions tion of the procedure pose a more difficult
[33]. Stent-assisted coiling techniques are associ- therapeutic conundrum, especially if the treated
ated with an increased risk of thrombosis aneurysm was ruptured at the time of presentation.
594 M. Mayich et al.

Fig. 10 Infarct due to


parent vessel sacrifice with
coil embolization.
Pretreatment 3D surface-
rendered CTA image (a)
shows a dissecting
aneurysm of the basilar
artery with proximal
stenosis. Post-embolization
3D surface-rendered CTA
image (b) shows coils
within the lesion and absent
opacification of the distal
basilar artery (arrowhead).
Axial DWI (c) and ADC
map (d) shows associated
restricted diffusion in the
pons, which correspond to
acute infarcts in the
perforator artery territory

Risk factors for symptomatic thromboembolism thromboembolic complications. The increased


in one series of 159 procedures included mass risk of thromboembolism with neck remodeling
effect by the treated lesion, residual flow within is at least partially attributable to selection bias
the coil mass at completion of the procedure, wherein larger, more complex aneurysms requir-
prolapsed coils, and mean aneurysm diameter ing longer procedure times may impart an inher-
larger than 12 mm [37]. Remodeling of the aneu- ently higher risk of thromboembolism.
rysm neck is also a risk factor for thromboembolic Distant coil migration is a rare intraprocedural
complication. Use of a stent construct to remodel or delayed complication of aneurysm emboliza-
the aneurysm neck resulted in acute or subacute tion (Fig. 12) [41]. This phenomenon can occur
in-stent thrombosis in 4.5 % of cases in a series of despite the use of balloon remodeling or stent-
161 patients, and 2.3 % experienced delayed ste- assisted techniques and can lead to territorial
nosis or occlusion [39]. These results were in infarction due to vessel occlusion. Vascular imag-
keeping with a recent meta-analysis which found ing can be used to localize the migrated coil, and
a 4.6 % risk of intraprocedural and 4.3 % post- MRI is useful for confirming and delineating the
procedural risk of stent-related thromboembolism extent of any associated cerebral infarction.
[40]. Balloon remodeling of the neck was also Aneurysm expansion or overpacking of aneu-
associated with an increased risk of rysms from liquid agent or coil embolization can
27 Postop Evaluation of Aneurysms (Including Vasospasm) 595

Fig. 11 Intraprocedural thrombus formation related to a


prolapsed coil loop. Immediate administration of IV hepa-
rin halted clot propagation and decreased clot burden.
Antiplatelet therapy was optimized postoperatively, and Fig. 13 Cranial nerve compression after embolization.
follow-up examination at 10 days failed to demonstrate The patient presented with diplopia following coil and
clot at the coil mass or occlusion of distal vessels. Onyx embolization of a giant left cavernous carotid aneu-
Intraprocedural thrombus formation may complicate as rysm. The coronal T2-WI image shows that the low signal
many as 11 % of procedures intensity embolic material exerts considerable mass effect
upon the cavernous sinus (arrow)

factor and affected cranial nerves (Fig. 13). Symp-


toms can persist or sometimes improve spontane-
ously over time.
A late complication of coil embolization is the
risk of aneurysm recurrence due to coil compac-
tion or aneurysm expansion, which manifests as a
discrete region of increased signal intensity on
MRA in contrast to the signal void from the coil
material (Fig. 14). Potential false positives include
hyperintense thrombus being mistaken for flow-
or contrast-related enhancement or enhancement
peripheral to the coil mass on CE-MRA which is a
normal finding most likely attributable to periph-
eral thrombus, inflammation, or healing. A recent
meta-analyses determined recurrence occurs in an
Fig. 12 Coil migration. Sagittal MIP CTA obtained after estimated 20.8 % of patients, necessitating
stent-assisted coiling of an ACOM aneurysm (arrowhead) re-treatment in 10 % of cases [43]. This rate stands
shows a coil (arrow) located in a more distal pericallosal in contrast to the exceptional stability of clipped
branch
aneurysms; one study demonstrated an annual
recurrence rate of only 0.5 % at 4.4 years
occasionally lead to new symptoms due to the [44]. Recurrence has been reported to be more
increased mass effect [42]. The most commonly common following treatment of ruptured (17 %)
involved structures are the optic and abducens versus unruptured (7 %) aneurysms [45]. Other
nerves. MRI is useful for evaluating the causative factors that have been suggested to correlate with
596 M. Mayich et al.

Fig. 14 Recurrent
aneurysm after coil
embolization. Initial post-
embolization lateral DSA
image (a) shows no filling
of a pericallosal artery
aneurysm. Follow-up
lateral DSA image (b)
shows new partial contrast
opacification (arrow) of the
aneurism sac associated
with coiled compaction
(arrowhead). Time-of-flight
MRA images obtained at
1.5-T with a TE of 2.9 ms
(c) and 3-T with a TE of
3.4 ms (d) show
corresponding flow-related
enhancement within a
portion of the aneurysm sac
(arrows) and coil
compaction (arrowheads).
There is similar image
quality with regard to the
coiled aneurysm at 1.5-T
and 3-T

higher rate of recurrence include large aneurysm incidence of aneurysm recurrence and exceed-
size, presence of intraluminal thrombus, low ingly low risk of subarachnoid hemorrhage in
packing density, initial incomplete occlusion, lon- patients whose treated aneurysms remain
ger follow-up, and wide neck, although specific occluded on examination at 6 months.
contributions by each or any of these risk factors Both the ISAC and CARAT studies suggest a
are debatable on the basis of differences in study low risk of late rebleeding, an observation corrob-
design [46]. The frequency at which recurrence orated by Sluzewskia and Van Rooij who found
occurs has led to the standard practice of follow- four instances in their series of 392 patients
up imaging, although there is an absence of con- followed for a mean of 51.5 months, with the
sensus guidelines regarding the specific fre- latest occurring at 40 months [50]. This compares
quency. One series of 126 patients found that favorably with series of clipping, where hemor-
100 % of recurrences occurred within 6 months rhage following treatment is considered to be a
[47], although the LOTUS I study showed 4 of rare event with an estimated risk of between 1 %
111 treated lesions reopened 5–11 years following and 2 % [3]. However, with regard to flow-
coiling, one of which necessitated re-treatment diverting devices, both early and delayed aneu-
[48]. A study assessing the incidence of subarach- rysm ruptures have been reported in the literature.
noid hemorrhage following successfully occluded The underlying mechanism for this phenomenon
lesions on a 6-month follow-up exam demon- may include changes in the wall, shear stress at the
strated a cumulative incidence of recurrent sub- aneurysm dome, unfavorable hemodynamic
arachnoid hemorrhage of 0.4 % within 8 years of changes, and autolysis of the aneurysm wall sec-
coiling in a series of 283 patients [49]. These ondary to red thrombus formation with accompa-
studies together demonstrate an extremely low nying inflammation. The hemorrhage associated
27 Postop Evaluation of Aneurysms (Including Vasospasm) 597

Fig. 16 Postoperative intraparenchymal hemorrhage and


edema. Coronal CT image shows acute hemorrhage and
vasogenic edema in the left temporal lobe (arrows) after
Fig. 15 Aneurysm recurrence after clipping. Lateral dig- recent left MCA and ICA aneurysm clipping, ruptured
ital subtraction angiogram shows aneurysm recurrence at intraoperatively
the origin of the posterior communicating artery (PComm),
adjacent to the clip (arrow)
and is important for excluding the presence of
with the presence of flow-diverting stents can be herniation and midline shift, which may require
distant from the treated aneurysm and can be surgical decompression (Fig. 16). Diagnostic
intraparenchymal. imaging can be useful to confirm suspected
Aneurysm recurrence after clipping is much unintended infarction. While hypoattenuation
less common than after coiling but can neverthe- and swelling can be identified on CT (Fig. 17),
less lead to devastating subarachnoid hemorrhage CT is relatively insensitive for the detection of
and warrants evaluation via CTA or DSA acute infarcts. Rather, MRI, particularly DWI
(Fig. 15). On the other hand, procedure-related sequences, can be useful for further investigation.
surgical complications are relatively common, Although superficial wound infections may be
with an overall incidence of approximately clinically apparent, diagnostic imaging can be
20 %, although this estimate can vary depending useful for evaluating the extent of the infection,
on aneurysm location and surgeon experience. particularly if intracranial involvement is
Aneurysms of the basilar artery and the anterior suspected. MRI with contrast is particularly useful
choroidal artery are more difficult to access via to rule out the presence of a pyogenic abscess,
craniotomy and thus have higher rates of compli- which appears as a rim enhancing fluid collection
cations than for clipping of MCA aneurysms, for with restricted diffusion. Analogous to coil
example. Procedure-related complications mainly masses, aneurysm clips can impinge upon the
consist retraction-related injury, unintended cere- cisternal segments of the cranial nerves, which
bral infarction, wound infections, CSF leakage, can be assessed using MRI cisternography-type
hydrocephalus, and cranial nerve injury. CT of sequences, such as CISS or FIESTA.
the brain is routinely performed in the early post- The major complications associated with
operative period to evaluate many of these com- extracranial-intracranial bypass performed with
plications, while MRI can be obtained to further aneurysm clipping include ischemic injury and
evaluate complications that are not explained by hyperperfusion syndrome [7]. Cerebral ischemia
CT. Indeed, CT can readily depict acute hemor- can result from thromboembolism, hemodynamic
rhage and edema associated with retraction injury ischemia, or watershed shift phenomenon, which
598 M. Mayich et al.

Fig. 17 Perioperative
infarct. Axial CT images (a,
b) obtained after left PCOM
aneurysm clipping show
hypoattenuation in the left
medial temporal lobe
(arrow). MRI was obtained
shortly afterward and the
DWI (c) and ADC map (d)
confirms the presence of an
acute infarct in the left
medial temporal lobe
(arrows)

is a dynamic change in cerebral hemodynamics Finally, while aneurysm recurrence is a pri-


caused by bypass flow. MRI findings can confirm mary indication for surveillance imaging, it is
acute cerebral infarction by showing restricted also important to consider the possibility of
diffusion. Vascular imaging may show a filling de novo aneurysms or enlargement of
defect in the graft, anastomosis, or supplied vas- concurrent aneurysms that were not treated
cular territory. Perfusion imaging can reveal def- on follow-up cerebrovascular imaging.
icits in cerebral blood flow. Alternatively, Tsutsumi et al. reported an annual risk of 0.89 %
extracranial-intracranial bypass can cause an of de novo aneurysm formation when they studied
increase in cerebral blood flow that can lead to a series of 112 patients treated for 140 clipped
hyperperfusion syndrome after direct aneurysms. This translates to a cumulative risk
extracranial-intracranial bypass can appear as of 10 % at 9 years, prompting the recommenda-
high signal intensity of the donor STA and dilated tion that follow-up is warranted at 9 years even in
branches of the MCA around the site of the anas- the context of completely clipped aneurysms
tomosis, which is most apparent with 3T versus (Fig. 18) [5]. Alternatively, the incidence of de
1.5T MRI and approximately 1 week after sur- novo aneurysm formation is estimated to be
gery. Perfusion imaging can help differentiate 1.5 %, and approximately 20 % of patients with
hyperperfusion from ischemia, which is important treated aneurysms have additional cerebral aneu-
because these conditions have contradictory rysms, of which a minority of these demonstrates
management. growth on 5-year follow-up imaging [51].
27 Postop Evaluation of Aneurysms (Including Vasospasm) 599

Fig. 18 Aneurysm follow-up. The patient has a history of stability of an additional left cavernous carotid artery aneu-
right paraclinoid aneurysm, status post right pterional cra- rysm (arrowheads) but increase in size of a partially
niotomy for clipping. Follow-up CTA images show thrombosed fusiform basilary artery aneurysm (arrows)

Nevertheless, these lesions infrequently require aneurysm coiling. Ultimately, conventional cathe-
treatment. These observations have led to the sug- ter angiography remains the gold standard imaging
gestion of a simplified algorithm for aneurysm modality if there are any questions that cannot be
follow-up that all patients should be followed with answered with the noninvasive imaging tech-
MRA at 6 months and if satisfactorily occluded are niques. It is important to recognize the expected
considered cured. Those not satisfactorily occluded and complicated posttreatment imaging findings.
could be re-treated at that point or undergo contin-
ued imaging follow-up. Continued follow-up may
be warranted in higher-risk patients, including References
those with giant aneurysms, multiple aneurysms,
positive family history of aneurysm, heritable dis- 1. Bederson JB, Connolly ES Jr, Batjer HH et al (2009)
American Heart Association. Guidelines for the man-
ease associated with aneurysm formation, or with
agement of aneurismal subarachnoid hemorrhage: a
proven growth of additional aneurysms to assess statement for healthcare professionals from a special
both the treated lesion and surveillance for devel- writing group of the Stroke Council, American Heart
opment of any additional lesions. Association. Stroke 40(3):994–1025
2. Molyneux AJ, Kerr RSC, Yu L-M, Clarke M,
Sneade M, Yarnold JA et al (2005) International sub-
arachnoid aneurysm trial (ISAT) of neurosurgical clip-
Summary ping versus endovascular coiling in 2143 patients with
ruptured intracranial aneurysms: a randomised com-
parison of effects on survival, dependency, seizures,
Imaging is indispensable for the evaluation of
rebleeding, subgroups, and aneurysm occlusion. Lan-
aneurysms after both endovascular and microsur- cet 366(9488):809–817
gical treatment. Multiple imaging modalities are 3. Ginat DT, Smith ER, Robertson RL, Scott RM,
available, each with advantages and disadvan- Schaefer PW (2013) Imaging after direct and indirect
extracranial bypass surgery. AJR Am J Roentgenol
tages. In general CT is routinely obtained after
201(1):W124–W132
aneurysm clipping to evaluate for major compli- 4. Deutschmann HA, Wehrschuetz M, Augustin M,
cations, such as hemorrhage, stroke, and hydro- Niederkorn K, Klein GE (2012) Long-term follow-up
cephalus, while MRI is performed if CT does not after treatment of intracranial aneurysms with the Pipe-
line embolization device: results from a single center.
reveal anything to explain the patient’s symptoms. AJNR Am J Neuroradiol 33(3):481–486. doi:10.3174/
Furthermore, CTA is preferable for imaging resid- ajnr.A2790
ual or recurrent aneurysms after aneurysm clip- 5. Tsutsumi K, Ueki K, Morita A, Usui M, Kirino T
ping, while MRA is suitable for imaging after (2001) Risk of aneurysm recurrence in patients with
600 M. Mayich et al.

clipped cerebral aneurysms: results of long-term 19. Dankbaar JW, Slooter A, Rinkel G, Schaaf IC (2010)
follow-up angiography. Stroke 32(5):1191–1194 Effect of different components of triple-H therapy on
6. David CA, Vishteh AG, Spetzler RF, Lemole M, Law- cerebral perfusion in patients with aneurysmal sub-
ton MT, Partovi S (1999) Late angiographic follow-up arachnoid hemorrhage: a systematic review. Crit Care
review of surgically treated aneurysms. J Neurosurg 14(1):R23
91(3):396–401 20. Wilson DA, Nakaji P, Abla AA, Uschold TD, Fusco
7. Wallace RC, Karis JP, Partovi S et al (2007) Noninva- DJ, Oppenlander ME et al (2012) A simple and quan-
sive imaging of treated cerebral aneurysms. Part I. MR titative method to predict symptomatic vasospasm after
angiographic follow-up of coiled aneurysms. AJNR subarachnoid hemorrhage based on computed tomog-
Am J Neuroradiol 28:1001–1008 raphy: beyond the Fisher scale. Neurosurgery
8. Shinohara Y, Sakamoto M, Iwata N, Kishimoto J, 71(4):869–875
Kuya K, Fujii S, Kaminou T, Watanabe T, Ogawa T 21. Aaslid R, Markwalder T-M, Nornes H (1982) Nonin-
(2014) Usefulness of monochromatic imaging with vasive transcranial Doppler ultrasound recording of
metal artifact reduction software for computed tomog- flow velocity in basal cerebral arteries. J Neurosurg
raphy angiography after intracranial aneurysm coil 57(6):769–774
embolization. Acta Radiol 55:1015–1023. 22. Lindegaard K-F, Nornes H, Bakke SJ, Sorteberg W,
doi:10.1177/028418511351049 Nakstad P (1988) Cerebral vasospasm after subarach-
9. Phan CM, Yoo AJ, Hirsch JA et al (2012) Differentia- noid hemorrhage investigated by means of transcranial
tion of hemorrhage from iodinated contrast in different Doppler ultrasound. In: Proceedings of the 8th
intracranial compartments using dual-energy head European Congress of Neurosurgery, 6–11 September
CT. AJNR Am J Neuroradiol 33:1088–1094 1987. Springer, Barcelona
10. Kaufmanna TJ, Huston J III, Cloft HJ, Mandrekar J, 23. Lysakowski C, Walder B, Costanza MC, Tramèr MR
Gray L, Bernstein MA, Atkinson JL, Kallmes DF (2001) Transcranial Doppler versus angiography in
(2010) A prospective trial of 3T and 1.5T time-of-flight patients with vasospasm due to a ruptured cerebral aneu-
and contrast-enhanced MR angiography in the rysm a systematic review. Stroke 32(10):2292–2298
follow-up of coiled intracranial aneurysms. AJNR 24. Anderson GB, Ashforth R, Steinke DE, Findlay JM
31:912–918 (2000) CT angiography for the detection of cerebral
11. Olsen RV, Munk PL, Lee MJ et al (2000) Metal artifact vasospasm in patients with acute subarachnoid hemor-
reduction sequence: early clinical applications. Radio- rhage. Am J Neuroradiol 21(6):1011–1015
graphics 20(3):699–712 25. Chappell ET, Moure FC, Good MC (2003) Compari-
12. Walker MT, Tsai J, Parish T et al (2005) MR angio- son of computed tomographic angiography with digital
graphic evaluation of platinum coil packs at 1.5T and subtraction angiography in the diagnosis of
3T: an in vitro assessment of artifact production: tech- cerebral aneurysms: a meta-analysis. Neurosurgery
nical note. Am J Neuroradiol 26:848–853 52(3):624–631
13. Gonner F, Heid O, Remonda L, et al. (1998) MR 26. van Rooij WJ, Sluzewski M, Beute GN, Nijssen PC
angiography with ultrashort echo time in cerebral aneu- (2006) Procedural complications of coiling of ruptured
rysms treated with Guglielmi detachable coils. AJNR intracranial aneurysms: incidence and risk factors in a
Am J Neuroradiol 19:1324–1328 consecutive series of 681 patients. Am J Neuroradiol
14. Kaufmann TJ, Huston J III, Cloft HJ et al (2010) A 27:1498–1501
prospective trial of 3T and 1.5T time-of-flight and 27. Sluzewski M, van Rooij WJ (2005) Early rebleeding
contrast-enhanced MR angiography in the follow-up after coiling of ruptured cerebral aneurysms: incidence,
of coiled intracranial aneurysms. Am J Neuroradiol morbidity, and risk factors. Am J Neuroradiol
31:912–918 26:1739–1743
15. Leffers AM, Wagner A (2000) Neurologic complica- 28. Johnston SC, Dowd CF, Higashida RT, Lawton MT,
tions of cerebral angiography. A retrospective study of Duckwiler GR, Gress DR, CARAT Investigators
complication rate and patient risk factors. Acta Radiol (2008) Predictors of rehemorrhage after treatment of
41:204–210 ruptured intracranial aneurysms: the Cerebral Aneu-
16. Cloft HJ, Joseph GJ, Dion JE (1999) Risk of cerebral rysm Rerupture After Treatment (CARAT) study.
angiography in patients with subarachnoid hemor- Stroke 39(1):120–125
rhage, cerebral aneurysm, and arteriovenous malfor- 29. Naggara ON, White PM, Guilbert F et al (2010)
mation. A meta-analysis. Stroke 30(2):317–320 Endovascular treatment of intracranial unruptured
17. Hanley M, Zenzen WJ, Brown MD, Gaughen JR, aneurysms: systematic review and meta-analysis of
EvansHanley AJ et al (2008) Comparing the accuracy the literature on safety and efficacy. Radiology
of digital subtraction angiography, CT angiography 256:887–897
and MR angiography at estimating the volume of cere- 30. Rordorf G, Bellon RJ, Budzik RE Jr et al (2001) Silent
bral aneurysms. Interv Neuroradiol 14(2):173–177 thromboembolic events associated with the treatment
18. Kassell N, Sasaki T, Colohan A, Nazar G (1985) Cere- of unruptured cerebral aneurysms by use of Guglielmi
bral vasospasm following aneurysmal subarachnoid detachable coils: prospective study applying diffusion-
hemorrhage. Stroke 16(4):562–572 weighted imaging. Am J Neuroradiol 22:5–10
27 Postop Evaluation of Aneurysms (Including Vasospasm) 601

31. Kang D-H et al (2013) MR-DWI–positive lesions and initially successful balloon-assisted aneurysm coiling:
symptomatic ischemic complications after coiling of technical case report. Neurosurgery 64(2):E391–E392;
unruptured intracranial aneurysms. Stroke 44:789–791 discussion E392
32. Altay T, Kang HI, Woo HH et al (2011) Thromboem- 42. Bhatti MT, Peters KR, Firment C, Mericle RA (2004)
bolic events associated with endovascular treatment of Delayed exacerbation of third nerve palsy due to aneu-
cerebral aneurysms. J Neurointerv Surg 3:147–150 rysmal regrowth after endovascular coil embolization.
33. Hahnemann ML, Ringelstein A, Sandalcioglu IE, J Neuroophthalmol 24(1):3–10
Goericke S, Moenninghoff C, Wanke I, Forsting M, 43. Murayama Y, Nien YL, Duckwiler G, Gobin YP,
Sure U, Schlamann M (2014) Silent embolism after Jahan R, Frazee J et al (2003) Guglielmi detachable
stent-assisted coiling of cerebral aneurysms: diffusion- coil embolization of cerebral aneurysms: 11 years’
weighted MRI study of 75 cases. J Neurointerv Surg experience. J Neurosurg 98(5):959–966
6:461–465 44. Ferns SP, Sprengers ME, van Rooij WJ et al (2009)
34. Mocco J, Fargen KM, Albuquerque FC, Bendok BR Coiling of intracranial aneurysms: a systematic review
et al (2011) Delayed thrombosis or stenosis following on initial occlusion and reopening and retreatment
enterprise-assisted stent-coiling: is it safe? Midterm rates. Stroke 40:e523–e529
results of the interstate collaboration of enterprise 45. Cognard C, Weill A, Spelle L et al (1999) Long-term
stent coiling. Neurosurgery 69(4):908–914 angiographic follow-up of 169 intracranial berry aneu-
35. Albuquerque FC, Levy EI, Turk AS, Niemann DB, rysms occluded with detachable coils. Radiology
Aagaard-Kienitz B, Pride GL Jr et al (2008) Angio- 212:348–356
graphic patterns of Wingspan in-stent restenosis. Neu- 46. van Rooij WJ, Sluzewski M (2009) Opinion: imaging
rosurgery 63(1):23–27 follow-up after coiling of intracranial aneurysms. Am J
36. Song JK, Niimi Y, Fernandez PM, Brisman JL, Neuroradiol 30:1646–1648
Buciuc R, Kupersmith MJ, Berenstein A (2004) 47. Sluzewski M, van Rooij WJ, Rinkel GJ et al (2003)
Thrombus formation during intracranial aneurysm Endovascular treatment of ruptured intracranial aneu-
coil placement: treatment with intra-arterial abciximab. rysms with detachable coils: long-term clinical and
Am J Neuroradiol 25:1147–1153 serial angiographic results. Radiology 227:720–724
37. Derdeyn CP, Cross DT III, Moran CJ, Brown GW 48. Sprengers ME, Schaafsma J, van Rooij WJ et al (2008)
et al (2002) Postprocedure ischemic events after treat- Stability of intracranial aneurysms adequately
ment of intracranial aneurysms with Guglielmi detach- occluded 6 months after coiling: a 3T MR angiography
able coils. J Neurosurg 96(5):837–843 multicenter long-term follow-up study. Am J
38. Cognard C, Weill A, Castaings L et al (1998) Intracra- Neuroradiol 29(9):1768–1774
nial berry aneurysms: angiographic and clinical results 49. Schaafsma JD, Sprengers ME, van Rooij WJ
after endovascular treatment. Radiology et al (2009) Long-term recurrent subarachnoid hemor-
206:499–5108 rhage after adequate coiling versus clipping of ruptured
39. Kanaan H, Jankowitz B, Akeu A, Kostov D et al (2010) intracranial aneurysms. Stroke 40:1758–6
In-stent thrombosis and stenosis after neck-remodeling 50. McLaughlin N, Bojanowski MW (2004) Early
device-assisted coil embolization of intracranial aneu- surgery-related complications after aneurysm clip
rysms. Neurosurgery 67(6):1523–1533 placement: an analysis of causes and patient outcomes.
40. McLaughlin N, McArthur D, Martin D (2013) Use of J Neurosurg 101(4):600–606
stent-assisted coil embolization for the treatment of 51. Sprengers ME, van Rooij WJ, Sluzewski M, Rinkel GJ,
wide-necked aneurysms: a systematic review. Surg Velthuis BK, de Kort GA, Majoie CB (2009) MR
Neurol Int 4:43 angiography follow-up 5 years after coiling: frequency
41. Fiorella D, Kelly ME, Moskowitz S, Masaryk TJ of new aneurysms and enlargement of untreated aneu-
(2009) Delayed symptomatic coil migration after rysms. AJNR Am J Neuroradiol 30(2):303–307
Part VI
Other CNS Vascular Diseases
Brain Arteriovenous Malformations
28
Daniel Sahlein and Nathan Manning

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 605 Brain arteriovenous malformations (bAVMs)
are relatively rare complex vascular lesions
bAVM Natural History: The Etiology
of Controversy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 606
involving a shunt between the artery and vein
Estimating Hemorrhage Risk in an Individual within the subpial space. This physiology can
bAVM: The Need for An result in hemorrhage, which can cause severe
Individualized Approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 608 neurological morbidity and death. There is tre-
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 611 mendous controversy surrounding appropriate
The Brain Arteriovenous Malformation . . . . . . . . . . 612
treatment, particularly for unruptured bAVMs,
Embryology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 612 which stems at least in part from debate as to
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 614 the natural history of bAVMs as well as vari-
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622 ability from lesion to lesion. This chapter will
Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 631
review bAVM imaging and in doing so will
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 635 summarize the origins of the current debate
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 635 over bAVM treatment utilizing a detailed
review of the natural history literature. Further-
more, this chapter will discuss bAVM epide-
miology, embryology, and treatment.

Keywords
Brain arteriovenous malformation • Stroke •
Intracranial hemorrhage • Vascular disorders •
Embolization • Microsurgery • Gamma Knife
surgery • Angiography • CT angiography • MR
angiography

D. Sahlein (*)
Goodman Campbell Brain and Spine, Indianapolis, Introduction
IN, USA
e-mail: dsahlein@goodmancampbell.com
A brain arteriovenous malformation (bAVM) is
N. Manning
defined as a direct communication from an artery
Department of Neurological Surgery, Columbia University
Medical Centre, New York, NY, USA to a vein through an intervening “nidus” which is
e-mail: dr.nathan.manning@gmail.com located in the subpial meningeal space [1]. Brain
# Springer Science+Business Media New York 2016 605
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_21
606 D. Sahlein and N. Manning

arteriovenous malformations are relatively rare management with an intervention (surgery, embo-
vascular lesions but nevertheless represent as lization, and/or Gamma Knife) or medical man-
high as 33.5 % of intracerebral hemorrhage in agement alone. Randomization began in April of
patients under the age of 40 [2]. There remains 2007 and was stopped in April of 2013 when a
considerable controversy with regard to their data and safety monitoring board recommended
pathogenesis, natural history, and optimal man- halting randomization because of superiority of
agement. Controversies notwithstanding, it the medical management group. Specifically,
seems likely these lesions form during early after a mean follow-up of 33.3 months (with a
embryologic development. Arteriovenous standard deviation of almost 20 months), the pri-
malformations are nonneoplastic; however, they mary outcome of death or symptomatic stroke was
are highly dynamic as a result of molecular sig- reached by 10.1 % of patients in the medical
naling molecules intimately involved in management arm and 30.7 % in the intervention
vasculogenesis and angiogenesis. Brain arteriove- group. Without getting too enmeshed in the details
nous malformations have a complex natural his- of this study, or why the study may have been
tory, the predictors of which have not entirely biased toward the medical therapy arm, the very
been elucidated but are likely to be dependent existence of this study raises a broader question as
upon bAVM architecture which may be under- to why such a basic aspect of bAVM management
stood through neuroimaging, and may evolve is still open to debate after decades of rigorous
over time. The diagnostic radiologist can there- study. Why was an NIH-sponsored study enroll-
fore play a critical role in understanding bAVMs ing unruptured bAVMs in a randomized trial com-
and in contributing to an understanding of both paring intervention versus medical therapy as late
natural history and treatment-related risk. as 2007? Nowhere is the answer to this question
Treatment is not without risk and is a source of clearer than in the variability and lack of clarity
recent controversy, particularly with respect to the provided by the bAVM natural history literature.
management of unruptured bAVMs [3]. Neverthe- Determining the true natural history of bAVMs
less, treatment risk has been extensively studied, is a daunting task to be sure. First, bAVMs are
and a prospectively tested pretreatment risk strat- fairly rare lesions, occurring with an incidence of
ification scale is available for microsurgical resec- approximately 1 per 100,000, and, second, they
tion [4, 5] though not for endovascular account for only around 4 % of all intracranial
embolization [6] likely as a result of fundamental hemorrhage [7]. However, the seemingly precise
differences in these two treatment modalities. numbers quoted in the previous sentence belie the
This chapter will review the fundamentals of true controversy that exists in this field and mask
bAVM formation, natural history, and treatment the variability observed from paper to paper with
with an eye toward diagnostic neuroradiology. regard to disease incidence, prevalence, and rup-
ture risk (see below for a thorough discussion of
the epidemiology). Very quickly does a review of
bAVM Natural History: The Etiology the bAVM literature become more a study in epis-
of Controversy temology and less an exploration of a complex
vascular lesion. The lack of a consensus under-
Few subjects in neurovascular disease are as con- standing of bAVM natural history is the direct
troversial or as current as the debate over optimal result of the complexity of studying a rare lesion
bAVM management. Much of the debate sur- and the variability in the disease from patient to
rounding bAVMs is the result of the publication patient that manifests as inconsistency between
of A Randomized trial of Unruptured Brain AVMs papers. This lack of clarity convolutes counseling
(ARUBA) in the Lancet in 2014 [3]. The ARUBA of individual patients in ways that will be
trial was a multicenter, randomized, non-blinded discussed in detail below.
trial that enrolled patients with unruptured The number of citations that a paper receives
bAVMs into one of two arms: medical gives a general sense of its relative value to the
28 Brain Arteriovenous Malformations 607

biomedical community [8]. The five most cited [10] in another; and tended to be larger, deeper,
bAVM natural history papers in order of publica- and closer to eloquent cortex in a third study
tion year are Fults et al. [9] published in 1984, [12]. The Brown et al. paper states that patients
Crawford et al. [10] from 1986, Brown et al. [11] treated immediately were excluded from natural
from 1988, Ondra et al. [12] from 1990, and Mast history analysis but gives no data on the number
et al. [13] from 1997. While the Ondra study of patients or differences between the treated and
found a remarkably stable per annum risk of hem- followed cohorts. The Mast et al. paper included
orrhage independent of clinical presentation and all patients until they were treated, creating huge
time from presentation of 3.9–4.0 % [12], there is ranges in the duration of follow-up in the study of
a tremendous variability in hemorrhage rate per 0.1–96.2 months in the hemorrhage arm and
annum from study to study ranging from 0 % to 0.4–90.9 months in the non-hemorrhage arm. In
32.9 % [13] depending on the type of presentation other words, some patients were contributing
and time elapsed. Furthermore, the associated almost 1000 the patient-year data in the study
mortality resulting from a hemorrhage is remark- than others. Furthermore, there is no discussion of
ably variable ranging from 0 % [13] to 29 % [11] the differences in the lesions or patient demo-
per hemorrhage. The incidence of significant neu- graphics in the patients treated within days of
rological morbidity is similarly variable, ranging presentation and those who were followed for
from 0.4 % per annum in one subset of the Fults months or years, and this was not a randomized
cohort [9] to 3.5–4.7 % per annum in patients study. In summary, each paper for which data is
presenting with hemorrhage [9, 11]. This degree given excludes approximately 40 % of the cohort
of variability makes it difficult if not impossible to from the natural history analysis, and excluded
accurately determine the natural history risk in an bAVMs are significantly different from those
individual patient. Many practitioners take some that were included. It is from these data that prac-
amalgam of this data and suggest a 2–4 % per titioners are estimating risk and determining
annum risk of hemorrhage and discuss the poten- whether treatment is necessary, i.e., calculating a
tial associated morbidity and mortality at least risk/benefit ratio for intervention. Without even
partially based on bAVM location [9–13]. delving into a discussion of treatment risk, it is
However, a closer examination of these papers not surprising that well-informed clinicians come
as “natural history” studies would suggest that to very different conclusions about appropriate
attempts at creating an informal average or formal clinical management [14, 15].
meta-analysis would not necessarily result in a For the sake of general approximation, two
more accurate calculation of natural history recent papers would suggest that the average of a
despite a theoretical increase in precision, a subtle 2–4 % per annum risk of hemorrhage is within a
but critical distinction. In each of the above reasonable range. The prospective, randomized
referenced natural history papers, a cohort of ARUBA study [3] found a 2.2 % per annum risk
patients was treated immediately and therefore of spontaneous rupture, and a recent meta-
excluded from the natural history observation, analysis which harmonized data from multiple
leading to serious questions about the generaliz- large databases to include over 6000 patient-
ability of the findings. A not-insignificant 36.6 % years of follow-up found an annual 2.3 % risk of
[9], 36.7 % [10], and 36 % [12] of patients were hemorrhage with a 95 % confidence interval of
treated immediately and excluded from three of 2.0–2.7 % [16]. The latter study however elimi-
the five studies. Furthermore, those patients nated patients once treated using a similar meth-
treated were statistically significantly different odology to the Mast paper [13] and like Mast
from those who were included in the natural his- et al. gives no data about the differences between
tory analysis. Patients in the nonsurgical arms patients treated early and those followed.
tended to have larger and deep bAVMs [9] in However, even if the 2–4 % per annum risk of
one study; tended to be left-sided, cross the mid- hemorrhage is accurate over large numbers of
line, larger, deeper, and more posterior bAVMs patients, variability between studies regarding
608 D. Sahlein and N. Manning

both hemorrhage risk and morbidity/mortality retrospective single-center study, 121 patients
raises a critical question. Can these averages with 122 bAVMs at NYU Langone Medical Cen-
truly be applied to an individual patient in the ter from 1996 to 2006 were entered into the anal-
context of this variability? In other words, is this ysis. All patients received full diagnostic
“average” approach appropriate for making treat- angiography with superselective microcathe-
ment decisions in individual cases when disease terization [43]. Detailed angioarchitectural data
variability renders such averages likely to be included bAVM location, depth, nidus size (mea-
highly inaccurate for an individual patient? Mod- sured in three planes), bAVM flow physiology
ern prospective data would suggest significant (nidus predominant, fistula predominant, or
disability in approximately 33 % of patients fol- mixed – see below for further details), arterial
lowing an initial hemorrhage and in 45 % of supply (organized by major feeding territory and
patients following a second hemorrhage [17], categorized as cortical or perforator), associated
and older studies suggest mortality of up to 29 % aneurysms (number and location characterized as
[11] per hemorrhage, considerable numbers for a nidal, flow-related, proximal [circle of Willis],
disease that presents on average in patients in their venous), pial collaterals (described by whether
early 30s. the collaterals originated from the same arterial
distribution [i.e., middle cerebral artery (MCA) to
middle cerebral artery] or different [i.e., anterior
Estimating Hemorrhage Risk cerebral artery (ACA) to middle cerebral artery]),
in an Individual bAVM: The Need for An bAVM border morphology (compact versus
Individualized Approach diffuse – see below for a detailed explanation),
the presence of moyamoya-type changes, venous
Estimating hemorrhage risk in an individual characteristics (number of draining veins, superfi-
bAVM is particularly critical in calculating a cial versus deep drainage, the presence of venous
risk/benefit ratio for therapy. A significant litera- angiopathy), and bAVM eloquence as defined by
ture has attempted to address this issue [9–11, 13, Spetzler and Martin [5]. Indisputable intracranial
18–40], and two meta-analyses have studied the hemorrhage was confirmed by computed tomog-
aggregate data more recently [41, 42]. Many fea- raphy scan in all patients [43].
tures of bAVMs have been found to be statistically Interestingly, bAVMs with a single draining
significantly associated with hemorrhage presen- vein were more likely to present with hemorrhage
tation. However, there is no proffered hypothesis than those with two or more draining veins
for why many of these bAVM features might [43]. This is not the first study to find this rela-
increase rupture risk [43]. For example, bAVM tionship between hemorrhage risk and a single
deep location [10, 32, 35, 36, 41], bAVM nidal draining vein [19, 22, 34, 38, 39]. However, it is
size [10, 18, 21, 23, 26, 28, 35–39, 41, 42], deep/ the first to test the physiologic implications of this
only deep venous drainage [13, 18, 20–22, 27–30, finding by analyzing different venous outflow
36, 37, 40–42, 44], ventricular/periventricular conditions. The study hypothesized that bAVMs
location [19, 22], posterior fossa location [9, 28], with a single vein were more often associated with
and arterial nonborderzone location [26] have all hemorrhage because of restricted outflow imped-
been statistically significantly associated with ance. This hypothesis implies that bAVMs with
rupture risk without any compelling explanation multiple draining veins would be protected from
as to causality. In the absence of a clear rationale hemorrhage by lower outflow impedance. Vari-
as to why these factors are associated with hem- ability in the venous drainage of patients with
orrhage, clinicians cannot confidently use them to two or more draining veins provided an opportu-
predict hemorrhage prospectively [43]. nity to test this hypothesis. Specifically, the study
However, one study lays the foundation for a looked at bAVMs with two or more draining veins
model of bAVM rupture risk based on parameters with significant venous stenosis and those with-
with clear physiologic causality [43, 45]. In a out. Interestingly, those bAVMs with multiple
28 Brain Arteriovenous Malformations 609

draining veins and a stenosis had significant asso- two or more), the presence of stenosis, and the
ciation with hemorrhage with an odds ratio very presence of any aneurysm). Not surprisingly, all
similar to those with one draining vein, thereby were statistically significant in the multivariate
validating the hypothesis [43]. logistic regression analysis with adjusted odds
The study also found that small nidal size, deep ratios ranging from 2.4 to 6.6 (one draining vein,
location, exclusively deep venous drainage, and 6.6; venous stenosis, 2.6; the presence of aneu-
the absence of pial-to-pial collaterals were signif- rysm, 2.4). Refer to Figs. 1 and 2 as examples of
icantly associated with hemorrhage and venous bAVMs with one draining vein versus multiple
outflow stenosis and the presence of any aneu- draining veins and a stenosis.
rysm showed a trend toward significance in the Furthermore, those variables without clear
univariate analysis. Only those potentially physi- physiologic implication that were significantly
ologic parameters were added to the multivariate associated with hemorrhage in the univariate anal-
analysis (the number of draining veins (one versus ysis were assessed as confounders using

Fig. 1 A 29-year-old female presented to the ER with arrows) and superior parietal artery (yellow arrowhead)
severe headaches. She was found to have a small supplying a subcentimeter bAVM (black arrowhead).
paramedian right parietal intracerebral hemorrhage. Sagit- There is a single paramedian superficial parietal draining
tal CTA (a) shows an enlarged paramedian vein (white vein which drains into the superior sagittal sinus. (blue
arrowheads). One can almost make out a nidus (black arrowheads). The patient was treated with open surgical
arrowhead). AP (b) and lateral (c) angiography show an resection
enlarged anterior cerebral pericallosal artery (white
610 D. Sahlein and N. Manning

Fig. 2 A 46-year-old male was evaluated with arterial and mid arterial phases (d and e, respectively).
noncontrast head CT following a seizure. Transaxial The mid arterial phase depicts an enlarged angular branch
noncontrast head CT (a) shows a right angular cortical of the right MCA (black arrowhead) terminating within the
and subcortical hyperdensity (white arrowheads). Two nidus (white arrowheads). Venous drainage is into two
sagittal precontrast T1-WI images (b, c) show the lesion dominant veins: the veins of Labbe and Trolard. There is
spanning the cortices of the superior and middle temporal a venous ectasia on Labbe (white arrow) and stenosis
gyri (arrowhead) and involving the angular gyrus. Right (black arrow) associated with the vein of Trolard
ICA angiograms in the lateral projection in the early

agreement analysis. In each case, the less clearly implications likely serve as confounders in
physiologic parameter was highly associated with assessing hemorrhagic risk, thereby validating the
one of the more plausibly physiologic parameters, selection of parameters with potential physiologi-
confirming that the less physiologic parameters cal implications for the multivariate analysis [43].
were acting as confounders. Small nidal size, a Not only does this study explain many of the
characteristic without clear physiologic implica- literature findings of bAVM features associated
tion, for example, showed 80 % agreement with with hemorrhage without clear physiologic impli-
one draining vein. Likewise, the presence of only cation, it paves the way for a patient-specific
deep venous drainage showed an 84 % agreement analysis of bAVMs from the standpoint of natural
with one draining vein. The absence of pial-to-pial history risk assessment [45]. The diagnostic radi-
collaterals from different distributions had a 71 % ologist’s role in assessing for venous stenosis,
agreement with one draining vein. Deep location number of draining veins, and aneurysms also
had a 77 % agreement with one draining vein. becomes critical. Future studies will elaborate on
These results confirmed that various bAVM char- and refine this model. While this model uses ana-
acteristics with less plausible physiological tomic features as physiologic surrogates, with
28 Brain Arteriovenous Malformations 611

inherent limitations, new MRI modalities have of headache in the general population, suggesting
tremendous potential to directly interrogate that this is an unlikely attributable symptom of
bAVM physiology [46, 47]. bAVMs, a controversial conclusion.
The Scottish Intracranial Vascular Malforma-
tion Study (SIVMS) [50] was the first large
Epidemiology population-based, prospective study of bAVM
epidemiology. It used multiple sources of case
The nature of the bAVM disease process, physi- ascertainment over a prospective 24-month period
cian and institutional tendency, and difficulties in to identify “first-ever-in-life” diagnosis of any
case detection lead to significant bias in the avail- intracranial vascular malformation in Scottish
able epidemiological data and result in persistent adults. In this study, the incidence of bAVM was
uncertainty with regard to the epidemiology and 1.12/100,000 person-years with a hemorrhagic
natural history of bAVMs. The first population- presentation incidence of 0.89/100,000 person-
based study to address the incidence and preva- years and a nonhemorrhagic presentation inci-
lence of bAVMs (in addition to intracranial vas- dence of 0.23/100,000 person-years. Two addi-
cular malformations (IVMs)) as a whole was the tional population-based studies from North
Olmsted County study [48]. Medical records from America, the New York Islands AVM Study and
Olmstead County, Minnesota, were reviewed for data from the Northern Manhattan Stroke Study
all IVMs from 1965 to 1992. This study demon- (NOMASS), were published in 2003 and 2002,
strated an incidence of 2.05 per 100,000 person- respectively [44, 51]. The NOMASS found three
years. The vast majority (85 %) of detected patients with bAVMs out of 207 patients with
bAVMs were symptomatic, and the mean age at first-ever intracranial hemorrhage, suggesting
diagnosis was ~40 years. The authors noted an that bAVMs account for 1.4 % of first-ever intra-
increasing detection rate during the course of the cranial hemorrhage in an unselected population
study that was presumably the result of improved [44]. The estimated crude incidence for first-ever
understanding of bAVMs and technological bAVM hemorrhage was 0.55/100,000 person-
advances such as improved medical imaging. In years. However, two of the three discovered
a population-based study from the state of West- bAVMs were in the posterior fossa, disproportion-
ern Australia, a considerably lower incidence of ately high with respect to bAVM location, thereby
0.89/100,000 person-years was determined with a casting doubt as to how representative the data is
prevalence of only 5.47/100,000 [49]. In this pop- and, by extension, the generalizability of the
ulation, the mean age at diagnosis was ~34 years, study. In spite of these concerns, the New York
approximately 60 % of patients presented with Islands AVM Study produced similar results
hemorrhage and 30 % presented with seizures. [51]. The New York Islands AVM Study encoun-
Only 2.5 % of bAVMs in this study were inciden- tered 284 AVM patients from a total of
tal suggesting a degree of under-detection, a met- 21,216,467 person-years of observation for an
ric that has implications for our understanding of incidence of 1.34/100,000 person-years. The
natural history. The authors of the Western crude incidence for AVM-related hemorrhage of
Australia study noted that the incidence of 0.51/100,000 person-years was remarkably simi-
bAVMs rose during the study period peaking at lar to that from the NOMASS data set. The mean
1.46/100,000 person-years in 1989, exceeding the age of bAVM rupture was ~31 years.
population rise. However, the incidence decreased A Swedish population-based cohort study also
from this point in spite of increasing availability reported similar findings with a bAVM incidence
of CT and MRI. The authors conclude that this of 1.24/100,000 person-years [52]. Hemorrhage
trend argues for a low incidence of occult bAVM; was by far the most common presentation seen in
however, this remains speculative. Interestingly, ~70 % with a presentation incidence of 0.87/
the authors also noted that headache, as a 100,000 person-years. Like others before, these
presenting symptom, did not exceed the incidence authors concluded that incidental bAVMs are rare.
612 D. Sahlein and N. Manning

The issue of incidental bAVMs has been hemorrhage. This figure rises to as high as
addressed, at least obtusely, by several population 33.5 % of hemorrhages in young adults (< age
MRI studies. A German Air Force recruit study in 40). Contrary to later reports, Al-Shahi and
which 2,536 males aged 17–35 years old Warlow estimated that 15 % of bAVMs are
underwent brain MRI as part of medical screening asymptomatic at the time of diagnosis. However,
for flying service found bAVMs in a surprising in line with later studies, they estimated that
0.2 % [53]. While this is significantly higher than approximately 60–70 % present with hemorrhage
previous clinical studies, it is within the range and 20 % with seizures. A more recent study has
reported in autopsy series, as summarized in a provided updated data [59]. Review of a 10-year
review of the epidemiology of bAVMs by Berman period of Kaiser Permanente Medical Care Pro-
et al. [54]. However, several other MRI screening gram in northern California medical records, radi-
studies have failed to identify any patients with ology reports, and administrative databases
bAVM [55–57]. This may in part be due to the yielded an overall bAVM detection rate of 1.42/
younger age of participant screened in the German 100,000 person-years [59].
study [53] in that older individuals are less likely
to harbor an unruptured or otherwise asymptom-
atic bAVM [10]. Furthermore, the imaging param- The Brain Arteriovenous Malformation
eters appear to be more rigorous in the German
study. Embryology
There are considerable challenges associated
with estimating epidemiology in a rare and poten- It is likely that bAVMs develop in utero, during
tially occult disease. Appropriate studies require the vascular development of the central nervous
adequate radiological and/or pathological investi- system. An understanding of bAVM anatomy and
gation to reliably diagnose bAVMs as well as to physiology therefore requires a basic understand-
exclude other entities that may have a similar ing of the embryology of the neurovasculature.
presentation such as cavernous malformations or The neural tissue and vascular tissue develop
dural arteriovenous fistulas (dAVF) [7]. While this in concert in the early embryo. Two processes are
seems reasonably achievable, clinicians may responsible for the vascularization of the central
forgo aggressive investigation due to a myriad of nervous system and indeed the developing fetus:
reasons including clinical urgency or futility. vasculogenesis, the formation of the primitive
Pathological investigations may lack sufficient endothelial cells [60], and angiogenesis, which
zeal given the minor role bAVMs may play on a follows vasculogenesis and is responsible for
population basis in the theater of intracranial hem- remodeling and expansion of the network
orrhage. Representative cohorts should preferably [61]. While there remains some debate based on
include individuals diagnosed at large referral rare observations, bAVMs are generally consid-
centers as well as small peripheral hospitals in ered to be congenital lesions that likely form, at
addition to those who have died in the community. least as immature precursors, during angiogenesis
To this end, many of the larger, population-based of the central nervous system [62]. It is impossible
trials would be expected to be fairly accurate. to convey the current theories on bAVM patho-
While the challenge is significant, some comfort physiology without at least an introduction to the
can be taken by the relatively similar results concepts of vasculogenesis and angiogenesis as
reported by large, population-based studies they pertain to the central nervous system. The
[48, 58]. The epidemiology data prior to 2001 is following sections will attempt to cover these
well reviewed by Al-Shahi and Warlow [7]. The complicated subjects in an expedient fashion.
authors reported a bAVM incidence of 1/100,000 Numerous texts and reviews are available which
person-years and a prevalence of 18/100,000. cover these processes in greater detail [1, 63–65].
Rupture of a bAVM was estimated to account for Closure of the neural tube occurs at around 3–4
1–2 % of all strokes and 4 % of intracerebral weeks gestation and signals the beginning of the
28 Brain Arteriovenous Malformations 613

embryonic stage. In regard to the morphogenesis adhesion molecules resulting in leakage of plasma
of the cerebral circulation, the embryonic stage proteins. These plasma proteins facilitate the
can be further classified into the prechoroidal, breakdown of extracellular matrix components
choroidal, and brachial phases. During the and at the same time provide the necessary tem-
prechoroidal stage, blood vessels develop by porary scaffolding allowing migration of endothe-
way of vasculogenesis from the meninx primitiva lial cells [66]. Pseudopodia are formed with a lead
on the neural tube surface. The meninx primitiva cell that penetrates between glial elements
is the mesenchymal covering from which the [67]. These early vascular channels are orientated
dura, arachnoid, and pia mater are derived. Within at right angles to the pial surface with little
vasculogenesis, the critical interaction occurs branching.
between the principal signaling molecules, vascu- A similar pattern occurs from the ependymal
lar endothelial-derived growth factors (VEGF), surface of the primitive ventricles. For both the
and their receptors (VEGFR). This is a relatively more dominant ventriculopetal and the more mod-
large group of molecules (VEGF A-E), which est ventriculofugal cerebral vascular develop-
have recurring roles in vasculogenesis and angio- ment, VEGF is again a key factor driving the
genesis both in physiological and pathological development with high VEGF levels found in
states. The interaction between VEGF-A and the deep subventricular zone acting as a powerful
VEGFR-2 promotes angioblasts to form chemoattractant to vascularization [60]. The
angiocysts, which ultimately migrate to form the migrating and dividing endothelial cells initially
early vascular network of the meninx [60]. form solid cords of elongated endothelial cells
The neural tissue is supplied by simple diffu- directed toward high concentrations of VEGF,
sion from the primitive vascular plexus on its particularly in the subventricular zone. Following
surface during the prechoroidal phase. As the the formation of solid cords, there is canalization
cranial neural tube develops into the five vesicles with lumen formation. This type of angiogenesis
(telencephalon, diencephalon, mesencephalon, is referred to as “sprouting” and is the dominant
metencephalon, and myelencephalon), the neural form in the developing CNS [68]. The definitive
tissue becomes too abundant to be supported by intraparenchymal vascular pattern is not arrived at
simple diffusion from the peripheral vascular immediately. Immature vessels, which are little
plexus. Support is added when the choroid more than continuous vascular channels at this
develops as an invagination of the meninx from stage, without recognizable arterial, capillary, or
the roof of the neural tube into the central canal as venous components, undergo further selection
the choroid is quite metabolically active and with dominant vessels undergoing further
brings with it its vascular supply, thereby initiat- branching, while other channels regress. To this
ing the choroidal phase [63–65]. end, an interaction between the endothelial cells
The brachial phase sees the emergence of the and their supportive structures, namely, smooth
adult-type arterial pattern by a series of regres- muscle cells (SMCs) and extracellular matrix
sions and enlargements of preexisting transverse (ECM), is critical. Both the ECM and the SMCs
vessels. As the developing central nervous system provide a source of growth factors and supportive
exceeds the capacity of simple diffusion, vascular ligands, which lend permanency to the developing
channels form within the parenchyma to keep up vascular structures. During both vasculogenesis
with the metabolic demands of the rapidly devel- and angiogenesis within the developing CNS,
oping primitive brain. Initially there is vasodila- the VEGF family of signaling molecules plays
tion directly mediated by increases in nitric oxide the dominant role and in particular is critical
(NO). Following vasodilation, there are signifi- for endothelial cell maturation and replication
cant increases in vascular permeability again [60, 63–65].
mediated by nitric oxide, however, this time via In addition to these critical signaling pathways,
upregulation of VEGF family molecules. VEGF several other molecules have been identified
acts directly on endothelial cells to effect cell-cell which play a role not only in endothelial cell
614 D. Sahlein and N. Manning

signaling but also in directing the supporting from the venous outflow. The bAVMs that dem-
SMCs and interactions with the ECM. ANG-1 onstrate a more direct physiologic shunt, and
interacts with the TIE2 receptor to encourage therefore possess less of a vascular network
recruitment and interaction between the endothe- between the arterial and venous compartments,
lial cell and the supporting environment. Equally instead possess a shunt in which the artery and
important is the ANG-2 molecule, which has an vein are more directly connected [75, 76]. In the
antagonistic effect on the TIE2 receptor neces- end, the nidiform and fistulous designations exist
sary for appropriate vascular remodeling to form not as absolutes but along a continuum. The fistu-
the final organized functional arterial tree lous physiology is more rare than the nidiform
[60]. Further signaling molecules include type, representing only around 2 % of patients in
endoglin and the related Alk1 receptors which some series [6]. Interestingly, this morphological
play a role in endothelial cell to cell interactions type is typically reported on the surface of the
during capillary formation and act on supporting central nervous system [1].
structures, triggering mesenchymal cells to Brain arteriovenous malformations are found
differentiate into pericytes and SMCs [69, 70]. in the supratentorial compartment in the over-
There has been an increasing interest in these whelming majority (>90 %) and demonstrate a
molecules as their role in pathological processes predilection for the MCA territory [6]. Deep struc-
becomes more apparent, particularly given their tures are involved in approximately 15 % of cases
role in genetic forms of bAVM (Osler-Weber- [43]. In approximately 50 %, there is deep venous
Rendu (also called hereditary hemorrhagic telan- drainage [77].
giectasia)) [66, 71, 72]. There are many ways of categorizing and
understanding bAVMs. Some authors have
suggested that there is a degree of predictability
Pathophysiology in both the pattern of arterial supply and venous
drainage based on the location of the bAVM
On gross inspection, bAVMs are composed of one [1]. This is useful to keep in mind when
or more, often enlarged, afferent arterial feeders interpreting the imaging appearance of lesions,
with one or more, frequently dilated, efferent particularly in addressing arterial supply and
veins. The core pathophysiology in bAVMs is venous drainage. Cerebral bAVMs may be topo-
that of arteriovenous shunting. The degree of graphically divided into those that reach the cor-
shunting has been categorized as either tex, those restricted to a deep location, and those
“nidiform” or “fistulous,” though these two types involving the choroid [1]. Lesions involving the
can coexist within a single lesion (Fig. 3) cortex are further classified into four categories:
[1, 73, 74]. (It is important that the reader not
confuse this distinction with dural (Fig. 4) or pial 1. Cortical lesions are on or reach the cortex and
(Fig. 5) arteriovenous fistula, which in the former supplied exclusively by cortical arteries with
reflects an abnormal connection between the only superficial venous drainage. It should be
artery and vein within the dura and in the latter noted that the basal veins of Rosenthal are not,
refers to an abnormal single connection within the anatomically speaking, deep cerebral veins as
pia, without a subpial component). Rather, this is a they lie on the brain surface and may drain
description of the physiologic shunting between cortical bAVM (i.e., temporal lobe lesions).
the arterial and venous compartments of a bAVM. They are, however, considered deep veins
The nidiform type demonstrates a larger network from the neurosurgical point of view given
of abnormal vascular channels leading from the their relatively inaccessible location and,
arterial compartment to the venous compartment. when involved in the surgical treatment of
These channels comprise the “nidus” which gives bAVMs, have higher associated morbidity.
the bAVM its mass-like appearance and which Cortical lesions have also been referred to as
adds a conduit that separates the arterial feeders sulcal bAVMs.
28 Brain Arteriovenous Malformations 615

Fig. 3 A 58-year-old male with a left posterior temporal/ Galen (red arrowhead), and straight sinus (white arrows), a
temporo-occipital brain AVM discovered incidentally fol- unique “deep” venous drainage for an otherwise superficial
lowing a motor vehicle accident. T2-WI transaxial MRI (a) bAVM. Lateral angiogram (e) following a single pedicle
shows the classic appearance of the nidus within the cortex embolization shows both fistulous (white arrowhead) and
and subcortical white matter of the left temporo-occipital nidiform (black arrowhead) components of the bAVM.
region (white arrowheads). The decision was made to treat Note the en passage supply from the parieto-occipital
the patient with embolization and surgery. (b) Left internal branch which goes on to supply the normal brain (yellow
carotid angiography, lateral projection, in the early arterial arrowheads). Superselective microcatheter angiography
phase demonstrates two dominant arterial feeders, both prior to embolization demonstrates discrete outflow into
grossly enlarged: the posterior temporal (white arrows) separate veins from each microcatheter injection, a
and temporo-occipital (black arrows) divisions of the left multicompartmental physiology. (f) The catheter tip is
MCA supplying the nidus (white arrowheads). The capil- depicted by the black arrowhead. Venous drainage from
lary (c) and venous (d) phases depict the bAVM venous the bAVM (white arrowheads) is directed inferiorly into
outflow. Venous outflow is predominantly into the left the sigmoid sinus (white arrow). (g) This pedicle opacifies
sigmoid sinus (white arrows) as well as retrograde into a separate compartment which, in distinction, drains pos-
the vein of Labbe (black arrows) and out via Trolard teriorly along the occipital pole (white arrowheads). The
(yellow arrowheads). There is lesser outflow within corti- catheter tip is depicted by a black arrowhead. The shadow
cal occipital veins and into the posterior aspect of the of the glue cast can be seen (yellow arrowheads). (h) An
superior sagittal sinus (black arrowhead). (d) A superficial unsubtracted lateral x-ray shows the glue cast following
vein (blue arrowheads) courses along the inferior and three pedicle embolizations (yellow arrowheads). Lateral
posterior occipital lobe superficially and then heads ante- angiogram in the arterial phase (i) shows significant
riorly along the paramedian occipital lobe, draining into devascularization of the nidus (white arrowheads) follow-
the basal vein of Rosenthal (yellow arrowheads), vein of ing three pedicles of embolization
616 D. Sahlein and N. Manning

Fig. 4 A 53-year-old male presented to the ER for wors- venous outflow is into the petrosal vein (black arrowhead)
ening episodes of dizziness. CTA was performed (a) which into the basal vein of Rosenthal (white arrow) which drains
demonstrated increased vascularity abutting the right out via the vein of Galen (black arrow) and straight sinus
petrous ridge (white arrowhead) with a few enlarged (red arrow). Additionally, there is infratentorial drainage
enhancing curvilinear structures adjacent to the into a superficial cerebellar hemispheric vein (white arrow-
pontomedullary junction on the right (black arrowheads). heads). Reformatted CTA can assist with surgical planning
This could easily be confused for a bAVM given its almost- (c). The vascular fullness along the petrous ridge is appar-
nidiform appearance. (b) Catheter angiography (right ent (white arrowheads). The petrosal vein courses toward
external carotid anterior trunk in the arterial phase) shows the viewer in this projection (black arrowheads) into the
a dural arteriovenous fistula along the right petrous ridge basal vein of Rosenthal (at the top of the image, yellow
with dominant supply from the petrosquamosal (yellow arrows). The internal auditory canal (red arrow) is
arrowheads) and basal tentorial (blue arrowheads) included as a helpful osseous landmark for surgery
branches of the right middle meningeal artery. Dominant
28 Brain Arteriovenous Malformations 617

Fig. 5 A 6-year-old girl presented with subarachnoid arrowhead). The individual feeding artery was
hemorrhage and intraventricular hemorrhage. Transaxial catheterized and an arteriogram was performed (f, g).
time-of-flight MRA (a) demonstrates unusual ectasias or Note the absence of a nidus or subpial component. The
aneurysms in the region of the ACAs, one oriented towards artery drains directly into the ectatic vein (junction marked
the right and the other to the left (white arrows). Left with a white arrow). The microcatheter was advanced
internal carotid AP (b) and lateral (c) angiograms in the across the fistula and into the ectactic venous outflow. (h)
early arterial phase demonstrate a rapid early opacification depicts the microcatheter tip within the first venous ectasia
from a small frontal branch of the ACA, arising from the (black arrowhead). The fistula was then coiled. Follow-up
right ACA (white arrow shows the junction of the artery internal carotid angiogram in the lateral projection (i) dem-
and vein). Capillary phase AP (d) and lateral (e) angio- onstrates no residual shunting. The coil mass is positioned
grams demonstrate the early opacification of a paramedian within the ectactic primary draining vein, extending prox-
cortical vein with multiple ectasias (black arrow), draining imally into the arterial supply (white arrowhead)
into the anterior aspect of the superior sagittal sinus (white

2. Cortico-subcortical bAVMs, also referred to as the transcerebral venous pathways remain open
gyral bAVM, are supplied by cortical arteries (Figs. 6 and 7). Unusual cortical pathways into
and drained by cortical veins. However, they the medial temporal cortical venous drainage
may also drain into the deep venous system if can also lead to deep venous drainage.
618 D. Sahlein and N. Manning

Fig. 6 A cortico-subcortical bAVM in a 33-year-old the venous outflow into the superior sagittal sinus (skinny
patient found to have a bAVM during a work-up for head- black arrows)). More unusual is the transcerebral pathway
aches. (a) A sagittal T1-WI precontrast MRI depicts the that remains open in this case and leads to deep venous
cortical and subcortical bAVM (white arrow). Lateral angi- drainage despite the relatively superficial nidus (white
ography from a right ICA injection in the mid arterial (b) arrowheads). Finally, it drains into the vein of Galen (red
and late arterial (c) phases depicts a parietal cortical and arrowhead) and straight sinus (yellow arrowhead). These
subcortical bAVM (white arrow) with supply from the latter two veins barely opacify because of the relative
ACA parietal divisions (black arrowhead) and MCA pari- paucity of opacified blood coming through the
etal division (white arrowhead). The venous outflow is transcerebral venous conduit relative to the large amount
depicted in (c). There is superficial venous drainage into of unopacified blood coming through both cerebral hemi-
the superior sagittal sinus which might be expected in a spheres (it is still the late capillary phase on the right- the
parasagittal bAVM such as this one (white arrows depict side of the injection.) into the deep venous system

3. Corticoventricular bAVMs extend from subventricular zone. These lesions will be


the cortex to the ventricular surface (Figs. 7 supplied by both cortical and perforating arter-
and 8). These lesions often demonstrate the ies and have both superficial and deep
classic pyramidal morphology of bAVM with drainage.
a broad cortical surface and apex pointing to or 4. Corticocallosal bAVMs share many similari-
indeed reaching the ventricular margin or ties with corticoventricular lesions, having
28 Brain Arteriovenous Malformations 619

Fig. 7 An 11-year-old boy was sent for an MRI as part of (white arrowheads). Note the classic perforator supply
an evaluation for with headaches. MRI was performed from the lateral lenticulostriate arteries (white arrow).
which revealed a right parietal corticoventricular brain The late arterial phase clearly depicts both superficial
arteriovenous malformation with superficial and deep (black arrow) and deep (black arrowhead) drainage. In
venous drainage and measuring 5.1  2.0  2.0 cm in addition, there is an unusual transcerebral vein which
maximum transverse, craniocaudal, and AP dimensions, drains from the deeper aspect of the lesion into the super-
respectively. (a) Transaxial T2-WI images demonstrate the ficial venous system (yellow arrowhead). That
classic flow voids extending from the cortex to the transcerebral vein is visible on the MRI T2-WI sequence
ependymal surface (white arrowheads). The large flow (magnified view (d–j) going from the level of the ventricle
void within the ventricle represents a deep vein (black sequentially to the level of the vertex), a testimony to the
arrowhead). Right internal carotid angiography in the excellent soft tissue contrast that MRI affords. It would be
early arterial (b) and later arterial (c) phases shows the difficult, however, to see this finding prospectively in the
nidus extending from the cortex to the ependymal surface absence of the catheter angiogram (yellow arrowheads)

both deep and superficial draining veins. The prospectively identify as their extra-axial location
arterial supply to the corpus callosum is makes them both surgically and endovascularly
derived from cortical vessels; however, it may accessible. This extra-axial location can be chal-
appear to represent perforators, particularly lenging to identify on cross-sectional imaging in
anteriorly and at the splenium [1, 78]. extensive lesions or in those lesions that do not
respect anatomical boundaries. The key to appre-
Deep lesions can be found in any structure of ciating this topographical subtype of bAVM is in
either the supra- or infratentorial compartments identifying supply by choroidal and
(Fig. 9). They are exclusively supplied by perfo- subependymal arteries arising from the circle of
rating arteries and drained via the deep venous Willis including the basilar tip. Cortical arterial
system. These lesions obviously pose the greatest supply is not a feature of these lesions. Drainage is
challenge to open surgical treatment. Choroidal typically via ventricular veins but may also
bAVMs are of particular importance to involve transcerebral veins. While these features
620 D. Sahlein and N. Manning

increasingly frequent as one approaches the site


of shunt [62]. One of the hallmarks of these abnor-
mal vessels is the marked variability in the wall
thickness, particularly involving the media. There
is both thickening and focal thinning of the vas-
cular wall with medial fibrosis and duplication
and degradation of the elastic lamina [62]. The
media itself may be partially or completely absent
in sections or alternatively demonstrate marked
structural disarray with separation into an inner
and outer coat by an aberrant elastic lamina [61,
62, 79, 80]. Veins often have an “arterialized”
appearance with lack of elastin and hypertrophy
of the smooth muscle layer [69]. However, in
sections there may be near complete loss of nor-
mal venous mural architecture, being apparently
Fig. 8 A sagittal postcontrast image to the right of midline replaced by the fibrous tissue [62, 79].
demonstrating a classic pyramidal or wedge-shaped The vast majority of bAVMs are sporadic;
corticoventricular bAVM in a 33-year-old, discovered dur- however, bAVMs associated with genetic syn-
ing a work-up for headache. Note the excellent soft tissue
contrast on MRI and the precise anatomic localization
dromes have lead to insight regarding the roles
that various genes play in vascular development
and in the development of bAVMs [1, 61]. It
are largely reliable, it must be remembered that comes as little surprise that many genes with a
bAVMs are dynamic lesions capable of evolution role in either vasculogenesis or angiogenesis dem-
and adaptation. This is particularly true for venous onstrate altered levels of expression of functional
drainage which may shift significantly if previ- proteins in bAVMs.
ously established drainage undergoes thrombosis Studies into hereditary hemorrhagic telangiec-
or becomes stenotic [1]. tasia (HHT) (Rendu-Osler-Weber syndrome) 1 and
2 have provided some insights into the disrupted
Histology and Histopathology pathways that may be involved in bAVM forma-
Microscopic examination of bAVMs demon- tion. Hereditary hemorrhagic telangiectasia is an
strates abnormal vessels from both the arterial autosomal dominant condition of vascular dyspla-
and venous compartments, with still further ves- sia involving multiple organ systems. The clinical
sels that do not easily fit into either venous or symptoms in HHT are caused by direct arteriove-
arterial categories [62]. While it is commonly nous connections without an intervening capillary
stated that the bAVM nidus lacks brain paren- bed. This results in multiple mucocutaneous telan-
chyma, gliotic neural tissue is often seen at the giectasias particularly about the oral cavity, finger-
margin and even within the lesions [61, 79]. There tips, and within the nasal cavity. The latter leads to
is frequently evidence of microhemorrhage that recurrent epistaxis, which is a common presenting
does not have a clinical event correlation and thus symptom. In addition to these superficial telangi-
implies subclinical extravasation [79]. Likely ectasias, there are visceral AVMs involving, for
related to this phenomenon, evidence of calcifica- example, the lungs, liver, and gastrointestinal tract
tion can be seen within bAVM vessel walls and in around one third of patients. Cerebral AVMs are
has even been reported within the adjacent paren- seen in around 10 % of patients; however, they are
chyma [80]. This calcification is more easily iden- probably under-recognized [82]. A mutation of
tified histologically [80], although may endoglin, a component of the transforming growth
occasionally be identified on imaging [81]. Clus- factor (TGF) receptor complex, is responsible for
ters of abnormal, muscularized arteries become HHT1 [61, 82]. The gene responsible for HHT2
28 Brain Arteriovenous Malformations 621

Fig. 9 A 16-year-old with a history of a ruptured diffuse temporize the lesion for Gamma Knife therapy. The
thalamic bAVM. (a) depicts the intracerebral and intraven- microcatheter tip is depicted by the black arrow. A
tricular acute hematoma. Note the hydrocephalus. The pseudoaneurysm (white arrowhead) was successfully
patient was sent for catheter angiography and possible targeted. Two sequential transaxial slices from a
embolization. AP Towne projection from a left vertebral noncontrast head CT (d and e) acquired 4 years later
injection (b) shows a diffuse thalamic and ventricular nidus show the onyx (embolic) cast, the very hyperdense curvi-
(white arrows) with supply from right posterior cerebral linear material in the right thalamus and posterior body of
artery (PCA) perforators and the posteromedial choroidal the right lateral ventricle. The patient has been stable
artery. The patient was sent for embolization (c) to neurologically

has also been identified: ALK1, which belongs to While these genes play a known role in hered-
the TGF receptor superfamily [61, 82]. Transgenic itary bAVMs, they may very well play a role in
mice carrying homozygous defects in endoglin die sporadic bAVMs as well. In fact, a study from
in utero with defective vascular remodeling. Het- 2005 found an association between a common
erozygous mice demonstrate a phenotype consis- polymorphism of the ALK1 and sporadic
tent with HHT1 including the development of bAVMs [84], suggesting that these genes may
bAVMs [70]. Endoglin appears critical for normal play a larger role in sporadic bAVMs than previ-
capillary remodeling and extension, being highly ously believed. Gene expression studies have
expressed during capillary extension prior to the identified numerous genes with either increased
commencement of blood flow. It has been or decreased levels of expression in bAVMs com-
suggested that failure of this process leaves the pared with normal cerebral vasculature [85]. How-
newly formed capillaries vulnerable to opportunis- ever, the challenge remains to identify potential
tic micro-shunts at the time of the first blood flow, therapeutic targets. Interestingly, increased VEGF
which, over time, develop into bAVMs [83]. and VEGF receptor expression has been reported
622 D. Sahlein and N. Manning

Fig. 10 A 16-year-old male who presented with acute parenchymal, subarachnoid, and intraventricular hemor-
onset of severe headache found to have a 2 cm left insular rhage (e). Repeat angiogram (f) (left internal carotid injec-
bAVM. Lateral angiogram in the early arterial (a) and mid tion in the lateral projection in the early arterial phase)
arterial (b) phases from a left ICA injection shows a 2 cm demonstrated a 3.4 cm left insular bAVM. Venous drainage
left insular bAVM supplied by the middle cerebral artery (g) was via Labbe (white arrows) as well and numerous
(MCA) (white arrowheads). Venous drainage is into the smaller frontal cortical veins (black arrowheads) into the
vein of Labbe (white arrows). Repeat lateral angiogram superior sagittal sinus. This was an unusual case of a
following surgical resection of the lesion in the mid arterial bAVM that was thought to be cured following microsurgi-
phase (c) and late arterial phase (d) shows no nidal cal resection, but which recurred and ruptured 9 years later.
opacification or early venous opacification and the lesion Cases such as this one stress the need for interval imaging
was thought to be cured. At the age of 25, the patient follow-up in younger patients even in cases that are
sustained a sudden onset of severe headache and loss of thought to be cured
consciousness. Noncontrast head CT showed a large

following partial embolization of bAVMs Imaging


[86, 87]. Given VEGF’s role in angiogenesis,
this may be central to the dynamic and recurrent Whether a bAVM is identified incidentally or due
nature of bAVMs and is an attractive target for to clinical sequelae such as seizure or focal neu-
therapeutic intervention in combination with rological deficit resulting from hemorrhage or
endovascular treatments. Figure 10 depicts a venous congestion, imaging studies establish the
recurrent bAVM 9 years following what was diagnosis. Imaging studies in their totality are
thought to be curative microsurgical resection. critical to define simple bAVM characteristics
28 Brain Arteriovenous Malformations 623

such as location and size as well as establishing may give a clue to the presence of regional paren-
more complex characteristics such as drainage chymal volume loss. Hydrocephalus may also be
pattern, feeding arteries, and the presence of com- present due to mass effect from the nidus itself, or
plicating factors such as associated aneurysms. more commonly a prominent draining vein.
Therefore, an understanding of the concepts CT angiography (CTA) will often establish the
already presented and applied to various imaging diagnosis of bAVM by demonstrating the com-
studies allows the radiologist to provide critical plex mesh-like network of the nidus and, if flow is
information in regard to the characteristics of a sufficient, enlarged arterial feeders and abnormal
bAVM, thus providing support for clinical deci- draining veins. However, as traditional CTA lacks
sion making as well as therapeutic planning. temporal resolution, the key diagnostic feature of
Every opportunity should be taken to provide bAVMs, namely, arteriovenous shunting, cannot
more than a simple diagnosis of bAVM and help be identified. In the absence of temporal resolu-
better understand individual bAVM physiology tion, other intracranial vascular abnormalities
and procedural risk. must be necessarily considered in the differential
diagnosis, specifically developmental venous
Computed Tomography anomalies (DVAs) (Fig. 12) and capillary telangi-
Noncontrast computed tomography (CT) is not ectasia. DVAs and capillary telangiectasias have
well suited to assessing the detailed angioarchi- features that may be mistaken for a nidus, and
tectural components critical in making treatment DVAs demonstrate a prominent vein, which may
decisions for bAVMs, however, as it is often the lead to further confusion. Modern CT angiogra-
first neuroimaging patients have for practical rea- phy (CTA) provides excellent spatial resolution
sons, and therefore every radiologist should be allowing characterization of nidal characteristics
aware of the CT imaging features. In particular, including size and to a lesser extent intranidal
patients who initially present with hemorrhage aneurysms, though the spatial resolution remains
secondary to bAVM will most likely undergo CT a fraction of that of conventional angiography
as their first diagnostic procedure, and radiologists (Figs. 1 and 13) [89].
should always be cognizant to the possibility of With proper adherence to the technique, using
bAVM as the cause of hemorrhage. When intra- individually timed acquisitions to ensure that the
cerebral hemorrhage is encountered in young study is acquired in the early to mid arterial phase,
patients (<45 years old), has a lobar distribution, arteriovenous shunting may be inferred by the
or is associated with parenchymal calcifications or presence of contrast within venous structures.
obvious hyperdense serpiginous structures, More recently, dynamic 3D-CTA techniques
bAVM must be considered as a possible etiology have been developed which allow even greater
[88]. In the case of unruptured bAVMs, slightly diagnostic accuracy, albeit at higher radiation
hyperdense serpiginous structures may be seen doses [90]. As CTA techniques improve, greater
and generally represent draining veins which at angioarchitectural detail will be gleaned from
their largest are typically quite a bit larger than the these noninvasive studies, allowing for the iden-
largest feeding arteries. Parenchymal calcification tification of associated aneurysms as well as the
can be seen in up to 20 % of bAVM either relating to interrogation of the venous drainage including the
previous hemorrhage or venous thrombosis number and location of draining veins as well as
(Fig. 11) [80]. Gliosis as a consequence of vascular the possibility of associated angiopathy such as
shunting or recurrent microhemorrhage may be stenoses. In fact, CTA is currently able to identify
directly identifiable or alternatively may be demon- intranidal aneurysms with reliability approaching
strated by volume loss and asymmetry. The latter that of digital subtraction angiography (DSA), the
may be obvious or, alternatively, may come to the gold standard (Fig. 13) [91].
radiologist’s attention by appreciating asymmetry In the setting of rapid clinical decline, CTA
in the CSF spaces such as the sulci or Sylvian may provide time-efficient information to assist
fissure. Ex-vacuo dilatation of ventricular structures surgical planning in the emergent setting. The
624 D. Sahlein and N. Manning

Fig. 11 A 6-year-old girl had a noncontrast head CT for first mesh-like network of the nidus as well as associated features
seizure (a). Note the calcified, slightly hyperdense pyramidal such as abnormal, enlarged draining veins (c). Note the
shape structured within the anterior left frontal lobe cortex scalloped inner table abutting the venous varix owing to
and subcortical white matter (white arrowheads). Transaxial chronicity (black arrowheads with white center). Transaxial
CTA confirms the diagnosis (b), with a pyramidal complex T2-WI MRI (d, e) demonstrates the classic flow voids
28 Brain Arteriovenous Malformations 625

routine use of CTA in the investigation of all imaging with low flip angles. Recent evidence
intracerebral hemorrhage is being adopted not suggests that those patients with evidence of pre-
only to identify and characterize potential vascu- vious microhemorrhage are at greater risk for
lar causes but also to assist in prognostication and subsequent clinically significant hemorrhage
possibly to identify patients that may benefit from [95]. The gliotic parenchymal tissue is generally
new treatments [92]. best visualized on FLAIR imaging where it
appears hyperintense. Venous hypertension may
Magnetic Resonance Imaging lead to vasogenic edema and mass effect in the
Magnetic resonance imaging (MRI) offers several involved territory, again best demonstrated by the
theoretical advantages over CT imaging of FLAIR sequence.
bAVMs. In particular, MRI’s unparalleled soft While convenient because of the lack of intra-
tissue contrast allows for the assessment of more venous contrast administration, traditional time-
subtle clinically significant epiphenomena of of-flight (TOF) magnetic resonance angiography
bAVMs such as clinically silent chronic hemor- (MRA) has limited utility in bAVM imaging,
rhage, gliosis, and venous congestion. Further- being inferior to both CTA and standard MRI in
more, MRI’s anatomic detail is unmatched direct comparisons [91]. Technical limitations in
allowing precise localization of the nidus and anatomic coverage and artifacts related to slow
characterization of its relationship to adjacent ana- flow result in poor delineation of the nidus itself
tomical structures. Using modern tractography and failure to depict associated aneurysms and
techniques as well as functional imaging, MRI some venous drainage pathways. Modified MRA
provides detailed information with regard to func- techniques are under development that allow
tional anatomy that may assist in both open surgi- superior vascular imaging and possibly even pro-
cal and radiosurgery planning [93, 94]. vide time resolution, critical to competing with
The bAVM nidus appears, similarly to CT, as a DSA. Postcontrast, dynamic MR subtraction
mesh-like network. However, due to the flow angiography has shown promise in early work;
within the vascular structures on typical spin however, it is time demanding [96].
echo sequences, the nidal structures are depicted More recently, intracranial 4D flow MRI has
as black flow voids (Figs. 7, 11, 14). Typically this been used not only to depict time-resolved MRA
is best appreciated on T2-WI which has been but also to assess flow dynamics within vascular
reported to have a diagnostic sensitivity as high structures [47]. This may prove of particular inter-
as 97 % in the case of unruptured bAVM est to endovascular surgeons. Changes in lesion
[91]. Intralesional or perilesional hemorrhage hemodynamics following partial embolization
will be demonstrated dependent upon the age of may in part be responsible for post-embolization
the blood. Typically, chronic hemorrhage with hemorrhage [6]. Initial results demonstrate
deposition of hemosiderin will appear marked changes in flow velocities of
hypointense on all sequences with blooming on vascular structures both in the immediate vicinity
susceptibility-weighted imaging or gradient echo of the lesion and at a distance from the nidus as

Fig. 11 (continued) associated with the nidus. Note, how- second trunk. Drainage is entirely superficial, with the
ever, the high signal within the large venous varix (e) deep aspect of the AVM and draining superiorly into the
attributable to turbulent and slower flow (white arrow- superior sagittal sinus via cortical veins and the superficial
heads). MRI signal characteristics must be interpreted in aspect draining via cortical veins into the superficial
the context of the flow physiology. Left ICA LAO angio- Sylvian territory and out of via the vein of Labbe. There
grams in the early arterial and capillary blush phases (f and is a 1.5 cm venous varix (small white arrow) associated
g) clarify the detailed angioarchitecture of the bAVM. with the deep aspect of the AVM and a 2.5 cm venous varix
There is direct supply from a trunk of the prefrontal divi- (small black arrow) associated with the superficial aspect
sion of left MCA with additional en passant supply from a
626 D. Sahlein and N. Manning

Fig. 12 A 6-year-old boy was worked up for unexplained view (c) and magnified lateral oblique (d) in the mid
loss of consciousness. Noncontrast CT had revealed a venous and late venous phases (e) and (f), respectively.
calcified lesion in the left temporal lobe, and CTA had Note the appearance of the classic “caput medusae” (white
demonstrated unusual vascularity in the region. The patient arrows on c and d). Note the delayed washout as a result of
was referred for angiography for evaluation of bAVM a stenosis (black arrow on f). The reported calcification is
versus tumor. Left internal carotid artery (ICA) injection likely the result of chronic venous congestion with possible
in the LAO view (a) and magnified lateral oblique (b) in component of venous infarct. There is no current way to
the mid arterial phase demonstrating a normal study. LAO treat this abnormality
28 Brain Arteriovenous Malformations 627

Fig. 13 An 11-year-old presented with severe refractory pseudoaneurysmal. Repeat angiography 2 weeks later
headaches which began while he was playing soccer. (a) showed interval thrombosis of the small pseudoaneurysm
Noncontrast head CT demonstrates a left temporo-occipital which is not uncommon for pseudoaneurysms in the sub-
acute hematoma. (b) CTA was obtained and the axial MIP acute phase following rupture. Also notice the mass effect
demonstrates two potential foci of hemorrhage. A larger, with splaying of arterial branches (small black arrow-
medial focus (large white arrow) and a smaller, posterior heads) and central oligemia, the angiographic appearance
and slightly lateral focus (small white arrow). The patient of hematoma. Microcatheterization of pedicles 1 (e) and
was sent for catheter cerebral angiography to further elu- 2 (f) demonstrates a fairly diffuse border and a single
cidate. Left vertebral artery LAO oblique angiogram in the physiologic compartment, meaning that both pedicles
early arterial (c) and late arterial/capillary phases (d). (c) share the same draining vein. (Catheter tip is depicted by
Note the same two features on the angiogram. The large the black arrowhead.) The point at which each pedicle
white arrow points to the larger outpouching, while the injected enters a shared vein is depicted as a white arrow-
smaller points to the smaller. In the late capillary/early head. The bAVM was embolized to cure with nBCA
venous phase (d), the larger outpouching is clearly the embolic pushed into the venous outflow on the second
proximal draining vein (small white arrowheads) with a injection
small associated varix, whereas the smaller focus looks

well [47]. A greater understanding of the signifi- venous vascular compartments. Currently, DSA is
cance of these changes may lead to safer and more the most reliable modality for evaluation of
effective endovascular treatment. bAVMs as a result of its fine temporal resolution.
Furthermore, the extremely high spatial resolution
Digital Subtraction Angiography has the potential to depict the exquisite vascular
Digital subtraction angiography (DSA) remains the detail required for the analysis of angioarchitectural
gold standard for both the detection and character- features as well as related hemodynamics. For
ization of bAVMs. At the heart of bAVM pathology example, voxel size on the latest Siemens equip-
is the shunting of the blood between the arterial and ment is 0.1  0.1 mm (mm) for DSA versus an
628 D. Sahlein and N. Manning

Fig. 14 A 47-year-old female with seizures. (a) arrowhead) that drains into the superior sagittal sinus.
Transaxial fast spin echo T2-WI images near the vertex. The patient was sent for Gamma Knife treatment. (d)
Note the classic “mesh-like” appearance of black flow Depicts the angiography for Gamma Knife targeting pur-
voids within the nidus (white arrow). MRI yields excellent poses. The fiducial markers on the Leksell headframe are
soft tissue contrast and depicts precise anatomic localiza- centered within the images (“X” and “+”). At a 6-month
tion. The bAVM nidus is centered within the cortex and follow-up, there was a signal abnormality surrounding the
subcortical white matter of the right postcentral gyrus. AP lesion seen well on the transaxial FLAIR sequence (e) near
(b) and lateral (c) catheter angiography in the early arterial the vertex (white arrow). The FLAIR signal extends into
phase from a right ICA injection demonstrate the arterio- the precentral gyrus, likely the result of perilesional radia-
venous shunt associated with the paramedian right parietal tion exposure. The patient remained asymptomatic.
nidus (white arrows depict the nidus) supplied from the Follow-up catheter angiography at 3 years (f) demonstrates
precentral and superior parietal divisions of the ACA complete bAVM obliteration. Note that the ACA has
(skinny black arrows), with pial-to-pial collaterals second- diminished in caliber (skinny black arrow) owing to the
arily supplying from the right MCA (black arrowheads). resolution of arteriovenous shunting
Venous drainage is into a single superficial vein (white

average of 0.5  0.5  .5 mm for CTA and characteristics. The bAVM location and by asso-
1.0  1.0  0.9 mm for an average clinical MRA. ciation (as described earlier) arterial supply and
DSA allows a precise analysis of the origin and venous drainage are well demonstrated, although
course of arterial feeders as well as supply to the anatomic position on DSA is inferred via the
normal brain structures, if present. Aneurysms, proximity to known arterial inflow and venous
either found proximally about the circle of Willis, outflow, as well as by bony landmarks rather
arising from arterial feeders, or arising within the than directly visualized as it is with CT or MRI.
nidus, are most easily identified on DSA, particu- Finally, the all-important venous drainage can be
larly with superselective microcatheter angiogra- assessed. The temporal resolution of DSA allows
phy [6, 20]. The lesion itself can be classified as easy assessment of not only the bAVM draining
fistulous or nidiform type, or possessing both veins but also their relationship to venous
28 Brain Arteriovenous Malformations 629

structures subservient to brain parenchyma. This Table 1 A list of suggested characteristics of bAVMs
distinction is of significant practical importance to Angioarchitectural
the surgeon at resection. The use of superselective characteristics Notes
microcatheter injections allows a further level of AVM location Hemispheric lobe, basal ganglia,
angioarchitectural detail to be revealed. This is the thalamus, caudate, callosal,
intraventricular, cerebellar
most sensitive method for identifying nidal aneu- hemispheric, cerebellar vermis,
rysms [6, 20] as well as nidal flow characteristics subarachnoid, brain stem,
and compartmentalization, which are of critical perimesencephalic
importance in embolization procedures. AVM depth Categorized as hemispheric
supratentorial (with subcategories
cortical, cortical/subcortical,
Angioarchitecture periventricular, cortical/subcortical/
A separate discussion is warranted on angioarch- periventricular, subependymal),
itecture because of its paramount significance in callosal, deep nuclei, ventricular,
both risk estimation and treatment decisions. Fur- cerebellar, brain stem
AVM laterality Right-sided, left-sided, midline
thermore, while it is classically assessed via DSA,
Nidus size Measured in three planes or by
it forms the framework by which all radiologists cross section imaging or
should approach bAVM description irrespective angiography with fiducial
of what imaging modality has been performed. A markers. Additionally categorized
more thorough discussion of angioarchitectural as <3 cm, 3–6 cm, and >6 cm
features and their descriptions is available bAVM flow Nidus-predominant, large fistula-
physiology predominant, or mixed
[1]. We limit this discussion to an overview rele-
Arterial supply Organized by major feeding
vant to diagnostic imaging. Table 1 gives a sum- territory and categorized as
mary of relevant angioarchitectural features to cortical or perforator where
keep in mind during radiologic evaluation. appropriate
Associated The presence, number, and
aneurysms location (nidal, flow-related,
Arteries proximal (circle of Willis), venous)
Arterial feeders may be broadly classified by their Pial collaterals The presence of pial collaterals
relationship to the nidus as either direct feeders or and a description of whether the
en passage feeders (Fig. 3). The former supply the collaterals originated from the
nidus as an end artery, terminating at the nidus, same (i.e., MCA branch to
adjacent MCA branch) or different
while the later give branches to the nidus in pass- (i.e., ACA branch to MCA branch)
ing (en passage) and course distal to the bAVM to major arterial distributions
supply normal structures. This distinction has bAVM border Compact versus diffuse
obvious treatment implications. When en passage morphology
arterial feeders are identified, efforts should be Moyamoya-type The presence of moyamoya-type
changes arterial changes
made to identify downstream structures that may
Venous
be at risk if the artery is lost at surgery or during
characteristics
embolization. Either due to intrinsic changes to Number of Number of principal veins (1, 2,
the bAVM and its arterial feeders or due to partial draining veins 3, or more) directly draining the
or staged treatments, en passage feeders may nidus
evolve, with increasing contribution in bAVM Superficial versus Utilization of superficial, deep, or
supply. In such cases, changes in hemodynamics deep drainage both types of venous drainage
Venous angiopathy Outflow stenosis (defined
secondary to the bAVM’s “sump effect” will alter
as >70 % stenosis) or ectasia.
the location of preexisting watershed regions of Venous or dural sinus thrombosis
the brain. This may lead to vascular steal until a or occlusion
new perfusion equilibrium is achieved with adja- bAVM eloquence The presence and criteria of
cent vascular territories. These dynamic vascular eloquence as defined by Spetzler
and Martin2
changes, which effect normal brain regions, may
630 D. Sahlein and N. Manning

Fig. 15 AP (Towne projection) (a) and lateral (b) angi- calcarine and parieto-occipital divisions of the PCA as
ography from a left vertebral injection demonstrating a well as en passant supply from the posterior temporal
right occipital bAVM in an 18-year-old found incidentally division. There is dural supply from the posterior menin-
after a concussion. There is dominant supply from the geal artery (white arrowheads)

explain the development of seizure activity in 58 %, and is likely dependent upon high-quality
previously asymptomatic lesions. superselective angiography [6, 97]. Redekop and
Dynamic changes in arterial supply may result colleagues proposed a classification system for
when supply from an arterial feeder is reduced, bAVM-related aneurysms that remains useful
either from arterial stenosis due to “high-flow [98]. In the classification by Redekop et al.,
arteriopathy” or as the result of partial emboliza- aneurysms are divided into three broad catego-
tion and atypical and somewhat bizarre arterial ries: intranidal, flow-related, and unrelated (dys-
channels of supply may develop. These channels plastic). Intranidal aneurysms are found within
may cause some diagnostic confusion if the the boundaries of the nidus and demonstrate rel-
dynamic and evolving nature of these lesions is ative early filling during angiography, prior to
not understood. Arterial supply may develop from opacification of the venous outflow. This should
adjacent vascular territories or from vessels not distinguish intranidal aneurysms from venous
predicted to be involved (i.e., cortical bAVM with ectasia and aneurysms. Flow-related aneurysms
deep perforator supply). The nidus may even are those found along the course of arteries that
recruit dural supply in some cases (Fig. 15). This contribute supply to the bAVM. The authors fur-
process reflects non-sprouting angiogenesis prob- ther classified flow-related aneurysms into prox-
ably in the setting of relative nidal ischemia and imal if the aneurysms were found on or about the
the liberation of pro-angiogenic signaling mole- circle of Willis or vertebrobasilar trunk or distal
cules such as VEGF. If this is suspected on any if the aneurysm occurred beyond these locations.
imaging modality, it should be conveyed, as this Unrelated aneurysms were those that occurred on
may increase the risk of surgery or mandate an arteries with no supply to the bAVM
atypical endovascular approach. [98]. Unrelated aneurysms are seen in fewer
patients with bAVM (0.8 %) than the general
Arterial Aneurysms public, with a generally reported rate of 2–3 %
Arterial aneurysms are seen with increased fre- [98]. This is likely to reflect the younger age of
quency in bAVM patients. The precise incidence bAVM patients with respect to the typical aneu-
is unclear, being variously reported from 7 % to rysm patient. Both flow-related and intranidal
28 Brain Arteriovenous Malformations 631

aneurysms are seen at significantly higher rates which is unique to one or a few arterial feeders
suggesting a causative effect of the bAVM or with distinct draining veins filling from separate
possibly a shared etiology [24, 99]. It is likely pedicles. This classification is primarily of signif-
that a combination of hemodynamic factors icance to embolization.
including flow velocity and the hyperdynamic Of further significance to embolization is the
circulatory environment associated with classification of the abnormal arteriovenous com-
bAVMs are at least partly involved. Furthermore, munication as fistulous or nidiform type. In the
regression and even resolution of associated former, there is a more direct communication
aneurysms have been reported with definitive between the artery and vein. As stated previously,
treatment of the bAVMs [100]. The risk of hem- in practice both fistulous and nidiform types occur
orrhage due to the presence of an associated in the same lesion. However, points of shunting
aneurysm is also variably reported. There does must be identified and monitored to prevent pre-
appear to be an association with the presence of mature embolization of venous outflow. Both
aneurysms and initial hemorrhagic presentation; compartmental characteristics and shunt charac-
however, the effect an aneurysm has on the teristics can only accurately be assessed on cath-
rate of subsequent hemorrhage remains unclear eter angiography.
[16, 24, 33, 42, 98, 101].
Veins
The Nidus Analysis of venous drainage in the setting of
Perhaps the most easily discernible nidal charac- bAVM should already be part of routine imaging
teristic on noninvasive imaging is the distinction interpretation given its importance in surgical
between compact and diffuse nidi. Diffuse lesions morbidity and in endovascular embolization.
have intervening brain parenchyma, often demon- The presence of deep venous drainage should be
strate multiple, more normal-appearing arterial evaluated on all bAVMs for calculation of the
feeders, and numerous morphologically Spetzler-Martin grade [5]. In addition, the pres-
unremarkable draining veins [1]. Diffuse lesions ence of high-flow venopathy including venous
pose a significant management conundrum as they stenosis and ectasia (Fig. 2) should be
are often extensive, with intervening functional documented, as well as the presence of venous
parenchyma. aneurysm. The presence of venous thrombosis
Nidal size is of importance when considering should be included in any analysis of bAVMs as
treatment options and associated risk. Particularly, well (Fig. 16).
the maximum nidal dimension should be mea-
sured and categorized as 0–3 cm, >3–6 cm,
or >6 cm for the sake of Spetzler-Martin classifi- Management
cation [5] (see further discussion below). Nidal
size should not be confused with the compact and A discussion of the management of bAVMs
diffuse classification as undoubtedly compact should be divided into those patients presenting
bAVM may be larger than 3 cm. with or having a history of clinically significant
Using superselective angiography, the nidus hemorrhage and those patients presenting inci-
may be further classified as single compartment dentally or with nonhemorrhagic symptoms.
(Fig. 13) or multi-compartment bAVM (Fig. 3). Hemorrhage rates are significantly higher in
Those lesions that demonstrate a single draining patients with a history of hemorrhage estimated
vein or veins which opacify after every arterial at 4.5 % per year in meta-analyses [42]. The risk is
feeder are injected are considered single compart- greatest in the first year following hemorrhage
ment. That is to say all arterial feeders have a with rates estimated between 6 % and 32.9 %
common pathway to a shared venous outflow, [13, 42]. Therefore, patients presenting with
whether single or multiple. A multi-compartment bAVM hemorrhage are generally offered some
bAVM demonstrates distinct venous drainage, form of eradicative treatment in the hope that
632 D. Sahlein and N. Manning

Fig. 16 A 70-year-old female was found down, and outflow (black arrow). AP (d) and lateral projection (e) right
noncontrast head CT revealed a right brachium pontis and internal carotid injection in the venous phase demonstrate no
vermis/medial right cerebellar hemispheric intracerebral opacification of the deep venous system on the right, and
hemorrhage (a). CTA was performed and read as normal. extensive medullary venous congestion, with enlarged
The patient was sent for catheter angiography because of the transcerebral veins diverting the deep venous territory out-
unusual location of the hemorrhage. Lateral mid arterial (b) flow superficially which can be appreciated as increased
and late arterial (c) angiograms from a left vertebral artery vascularity within the brain parenchyma. Note the lack of
injection show a diffuse superior vermian bAVM (white the internal cerebral veins, vein of Galen, or straight sinus on
arrowheads), likely being compressed by the hematoma. the lateral projection (white arrows depict location where
Supply is from vermian branches of the superior cerebellar the deep venous system should opacify, but does not). AP (f)
artery (black arrowhead). The lesion drains into a very short and lateral projection left internal carotid injection in the
segment of the precentral cerebellar vein and into an venous phase demonstrate normal opacification of the left
excluded pouch within the vein of Galen (red arrowhead) deep venous system (white arrow) depicted en face. (g) Left
which cannot drain into the straight sinus and instead internal carotid injection in the venous phase demonstrates
refluxes into the right internal cerebral vein (yellow arrow- normal opacification of the left internal cerebral vein (white
head) and into the right basal vein of Rosenthal (light blue arrows), vein of Galen (black arrow), and straight sinus (red
arrowheads) which drains out via an infratemporal vein arrow) which proves that the vein of Galen and straight
(white arrow). In a slightly later phase (capillary phase, c), sinus remain patent and that the primary bAVM drainage
note the opening of falcine veins for internal cerebral venous into Galen occurs into an excluded pouch
28 Brain Arteriovenous Malformations 633

this risk can be eliminated or palliative treatment deficit (scored as 0 for no eloquence and 1 for
in the hope that the risk can be reduced. The eloquence). Eloquent regions typically
question of what management has to offer include the sensorimotor cortex, occipital (pri-
bAVM patients without a history of hemorrhage mary visual) cortex, speech areas, deep nuclei
is more complex. of the cerebellum and brain stem, thalamus and
The recently published ARUBA explored the hypothalamus, internal capsule, and cerebral
issue of management of unruptured bAVM, com- and cerebellar peduncles. This scale offers a
paring active treatment with conservative medical fairly simple and effective risk stratification
management [3]. The trial was stopped early when system for patients undergoing surgical resec-
a data and safety monitoring board recommended tion, and its effectiveness was prospectively
halting randomization because of the superiority validated [4]. However, it does presume nor-
of the medical management group. The trial itself mal topographical orientation of brain func-
has generated considerable controversy as a result tions which may not be the case in bAVM
of poor recruitment, a skewed patient population, patients. Where possible, fMRI will provide
heterogeneous treatment strategies, and relative additional insight. The Spetzler-Martin scale
short-term follow-up [102–105]. However, it has been accurate in stratifying risk, with
does generate a need to apply more stringent risk Grade Is and IIs experiencing less than 5 %
determination tools, like those discussed previ- major morbidity or mortality, Grade IIIs
ously, to offer patients the optimum care. experiencing approximately 5 %, and Grade
Treatment consists of one or a combination of IVs and Vs experiencing 7–12 % [5]. The
three approaches: microsurgical resection, scale is generalizable between surgeons in
endovascular embolization, and/or radiotherapy. large part because the procedural end point
for surgery is more or less the same – complete
Microsurgical Resection resection of the entire bAVM. The same cannot
The goal of open surgery is complete excision of be said about the procedural end point for
the bAVM and, with that in mind, achieves the embolization, a critical factor that will be
most robust results. The Spetzler-Martin grading discussed below.
scale was developed to predict surgical difficulty
and complications [5]. The scale ranges from 0 to Embolization
5 based on three factors: Unlike microsurgical resection, the role of emboli-
zation is quite variable from institution to institution
1. Assessment of the nidus size (graded as or even practitioner to practitioner [6]. Embolization
0 for <3 cm, 1 for 3–6 cm, and 2 for >6 cm), might be carried out for the following reasons:
oftentimes most easily measured on cross- (1) endovascular occlusion, (2) devascularization
sectional images. of as much of the bAVM as possible prior to or
2. Determination of the pattern of venous drain- following resection or radiosurgery, and
age, either superficial or deep (0 or 1, respec- (3) targeting of a focal angioarchitectural weakness,
tively). (Of note the term “deep venous either alone or in combination with surgery or
drainage” is a surgical one in this context rather Gamma Knife. Each of these strategies carries its
than an anatomical one. As stated earlier, while own unique risk profile and should be assessed
the basal veins of Rosenthal are not considered separately with respect to whether the procedural
deep venous structures anatomically, they are end point has been met [6]. In the absence of
considered deep venous structures at opera- documented procedural intent, procedural efficacy
tion. So too are other venous structures which cannot be calculated, nor can results be generalized
are challenging to access at craniotomy such as across medical centers or between practitioners.
the interhemispheric cortical veins.) This lack of procedural standardization and docu-
3. Involvement of eloquent areas or regions if mentation makes it impossible to develop a reliable
injured would cause immediate neurologic pre-embolization risk stratification scale [6].
634 D. Sahlein and N. Manning

The literature on bAVM embolization is limited Radiotherapy


by a lack of class 1 evidence and is characterized by The first Gamma Knife surgery (GKS) for bAVMs
small series, often without a control group or quan- was performed in 1970. Multiple research studies
titative outcome measure. Nevertheless, proposed subsequently demonstrated that GKS is safe and
benefits of embolization include better long-term effective in small to medium-sized bAVMs, even
clinical outcomes with embolization and surgery in deep and eloquent territories [120], with oblit-
than surgery alone [106]; the ability to surgically eration rates of approximately 60–90 %
resect otherwise unresectable large, complex [121–124], at least in part dependent on bAVM
bAVMs [107]; a reduction in morbidity following volume [125]. The major limitations of GKS are
surgery in embolized bAVMs in eloquent brain the average latency to bAVM cure (approximately
areas [108]; equal operating time and blood loss 2–3 years) and potential harm to perilesional brain
in larger, more complex embolized bAVMs as parenchyma. During the intervening time between
compared to non-embolized smaller AVMs [109]; treatment and cure, the patient remains at risk for
and diminished surgical difficulty [110]. hemorrhage or may even be at slightly greater risk
Traditionally, the principal role of embolization [126, 127].
in most practices is as adjunct treatment prior to While bAVM obliteration rates following GKS
surgical resection or radiotherapy (Fig. 3). A bal- are directly proportional to radiation dose, unfor-
ance must be struck between the reduction of arte- tunately morbidity associated with GKS is also
rial inflow and the preservation of balanced correlated with radiation dose (as well as the total
outflow. If the arterial inflow is reduced signifi- volume treated- larger volume bAVMs are associ-
cantly but not completely obliterated, venous out- ated with greater morbidity) [128]. In order, there-
flow may be reduced to the point of spontaneous fore, to minimize the risk of morbidity, doses at
thrombosis. Delayed hemorrhage can result [111]. the periphery of the lesion, abutting normal brain
It should be appreciated from the previous parenchyma, should be the lowest within the
discussion of the hemodynamically balanced targeted region, and minimum doses of approxi-
nature of these lesions that if any arterial inflow mately 17 Gy at the margin have been demon-
remains in the setting of occluded venous outflow, strated to lead to obliteration rates of
hemorrhage becomes increasingly likely. How- approximately 80 % and are an independent pre-
ever, in the setting of attempting cure via emboli- dictor of procedural efficacy (Fig. 14) [121].
zation alone, ultimately the venous outflow must The direct correlation between dose delivered
be embolized to prevent recurrence. This tech- and both obliteration rates and morbidity and the
nique is being increasingly used for curative direct relationship between total volume treated
embolization alone using either transarterial and morbidity have particular impact on the treat-
[112] or transvenous approaches [113]. ment of large bAVMs using GKS, where total
The liquid embolic agents, N-butyl cyanoacry- lesion dose required for obliteration can cause
late (NBCA) and onyx, a copolymer of ethylene unacceptable levels of morbidity. There are differ-
vinyl, are the primary agents used for the ent strategies to safely increase radiation delivery
endovascular treatment of cerebral AVMs. The to the bAVM while limiting perilesional injury,
two agents have very different radiographic mostly by fractionating radiation dose by time or
appearances as a result of the contrast agents by volume [129]. Multiple fractioned radiation
used in each (Ethiodol versus tantalum). delivery protocols have been utilized, with
The risk associated with embolization varies mixed results [130–135]. A more recent trial dem-
widely in the literature. The morbidity associated onstrated that bAVMs larger than 15 cm3 in vol-
with embolization in the large nBCA series ranges ume can be treated successfully using fractionated
from 0.8 % to 9.0 % per patient [6, 114] and dosing with a staged-volume protocol [136]. Typ-
mortality from 0.8 % to 3.7 % [6, 115] and onyx ically GKS is used alone; however, it has been
series morbidity from 4.6 % to 15.5 % [116, 117] used with prior embolization to reduce the volume
and mortality from 2.1 % to 4.3 % [118, 119]. of large bAVMs to that suitable for GKS with
28 Brain Arteriovenous Malformations 635

mixed results [135, 137]. Diminished efficacy of publication on the part of treating clinicians.
GKS following embolization is likely related to Given the uncertain natural history of individual
obscuration or incomplete opacification of the lesions, the significant risks and limitations of the
nidus following endovascular treatment and there- available management, and the complexity of the
fore incomplete nidal targeting [137]. imaging features and their interplay with risk-
Modern equipment allows excellent dose shap- benefit assessment, bAVM diagnosis and manage-
ing, achieving high nidal doses with minimal dose ment is best suited to collaborative multidis-
to surrounding tissues. Radiotherapy causes dam- ciplinary teams. This allows management
age to the endothelium and smooth muscle cells of combinations to be individually tailored to suit
the bAVM. This induces an inflammatory reaction both the characteristics of the lesion and the patient.
and scarring with vascular narrowing and even-
tual obliteration of the nidus. GKS leads to signif-
icant morbidity in approximately 4.8 % [126]. References
1. Lasjaunias PL, Berenstein A, Brugge KGT (2001)
Conclusion Surgical neuroangiography, 2nd edn. Springer,
Berlin/New York
2. Ruiz-Sandoval JL, Cantu C, Barinagarrementeria F
bAVMs are complex and relatively rare vascular (1999) Intracerebral hemorrhage in young people:
lesions involving a shunt between the artery and analysis of risk factors, location, causes, and progno-
vein within the subpial space. Despite decades of sis. Stroke 30(3):537–541
study, there is scant true natural history data even 3. Mohr JP et al (2014) Medical management with or
without interventional therapy for unruptured brain
among highly cited “natural history” papers, the arteriovenous malformations (ARUBA): a
source of tremendous controversy about the true multicentre, non-blinded, randomised trial. Lancet
natural history of the disease and secondarily the 383(9917):614–621
most appropriate management. Furthermore, var- 4. Hamilton MG, Spetzler RF (1994) The prospective
application of a grading system for arteriovenous
iability from lesion to lesion calls into question the malformations. Neurosurgery 34(1):2–6,
accuracy of applying “average” data to individual discussion 6–7
patients. Risk estimates using individual lesion 5. Spetzler RF, Martin NA (1986) A proposed grading
characteristics based on parameters with a clear system for arteriovenous malformations. J Neurosurg
65(4):476–483
potential physiologic impact should be utilized. 6. Sahlein DH et al (2012) Nidal embolization of brain
Current models use anatomic features as physio- arteriovenous malformations: rates of cure, partial
logic surrogates, though advances in MR technol- embolization, and clinical outcome. J Neurosurg
ogy might soon avail the neurovascular 117(1):65–77
7. Al-Shahi R, Warlow C (2001) A systematic review of
community of direct noninvasive physiologic the frequency and prognosis of arteriovenous
interrogation of bAVMs. With current technology, malformations of the brain in adults. Brain
bAVMs are best imaged using digital subtraction 124(Pt 10):1900–1926
catheter angiography, which has the best temporal 8. Garfield E (1972) Citation analysis as a tool in journal
evaluation. Science 178(4060):471–479
and spatial resolution, though modern dynamic 9. Fults D, Kelly DL Jr (1984) Natural history of arte-
CT angiography has good temporal resolution, riovenous malformations of the brain: a clinical study.
albeit with a higher radiation dose to the patient. Neurosurgery 15(5):658–662
Management of bAVMs typically comes down 10. Crawford PM et al (1986) Arteriovenous malforma-
tions of the brain: natural history in unoperated patients.
to microsurgical resection, endovascular emboli- J Neurol Neurosurg Psychiatry 49(1):1–10
zation, Gamma Knife surgery, or some combina- 11. Brown RD Jr et al (1988) The natural history of
tion thereof. The efficacy and risks of each of unruptured intracranial arteriovenous malformations.
these procedures have been well studied, though J Neurosurg 68(3):352–357
12. Ondra SL et al (1990) The natural history of symp-
new treatment strategies are constantly being tomatic arteriovenous malformations of the brain: a
developed, particularly using the latter two 24-year follow-up assessment. J Neurosurg
modalities, necessitating ongoing research and 73(3):387–391
636 D. Sahlein and N. Manning

13. Mast H et al (1997) Risk of spontaneous haemorrhage 29. Marks MP et al (1990) Hemorrhage in intracerebral
after diagnosis of cerebral arteriovenous malforma- arteriovenous malformations: angiographic determi-
tion. Lancet 350(9084):1065–1068 nants. Radiology 176(3):807–813
14. Cockroft KM (2007) Unruptured brain arteriovenous 30. Pollock BE et al (1996) Factors that predict the bleed-
malformations should be treated conservatively: ing risk of cerebral arteriovenous malformations.
no. Stroke 38(12):3310–3311 Stroke 27(1):1–6
15. Stapf C, Mohr JP (2007) Unruptured brain arteriove- 31. Todaka T et al (2003) Analysis of mean transit time of
nous malformations should be treated conservatively: contrast medium in ruptured and unruptured arterio-
yes. Stroke 38(12):3308–3309 venous malformations: a digital subtraction angio-
16. Kim H et al (2014) Untreated brain arteriovenous graphic study. Stroke 34(10):2410–2414
malformation: patient-level meta-analysis of hemor- 32. Yamada S et al (2007) Risk factors for subsequent
rhage predictors. Neurology 83(7):590–597 hemorrhage in patients with cerebral arteriovenous
17. Choi JH et al (2006) Clinical outcome after first and malformations. J Neurosurg 107(5):965–972
recurrent hemorrhage in patients with untreated brain 33. da Costa L et al (2009) The natural history and pre-
arteriovenous malformation. Stroke 37(5):1243–1247 dictive features of hemorrhage from brain arteriove-
18. Duong DH et al (1998) Feeding artery pressure and nous malformations. Stroke 40(1):100–105
venous drainage pattern are primary determinants of 34. Alexander MD et al (2015) Association between
hemorrhage from cerebral arteriovenous venous angioarchitectural features of sporadic brain
malformations. Stroke 29(6):1167–1176 arteriovenous malformations and intracranial hemor-
19. Nataf F et al (1997) Angioarchitecture associated with rhage. AJNR Am J Neuroradiol 36:949–952
haemorrhage in cerebral arteriovenous 35. Stefani MA et al (2002) Angioarchitectural factors
malformations: a prognostic statistical model. Neuro- present in brain arteriovenous malformations associ-
radiology 39(1):52–58 ated with hemorrhagic presentation. Stroke
20. Turjman F et al (1995) Correlation of the angioarch- 33(4):920–924
itectural features of cerebral arteriovenous 36. Hernesniemi JA et al (2008) Natural history of brain
malformations with clinical presentation of hemor- arteriovenous malformations: a long-term follow-up
rhage. Neurosurgery 37(5):856–860, discussion study of risk of hemorrhage in 238 patients. Neuro-
860–2 surgery 63(5):823–829, discussion 829–31
21. Kader A et al (1994) The influence of hemodynamic 37. Langer DJ et al (1998) Hypertension, small size, and
and anatomic factors on hemorrhage from cerebral deep venous drainage are associated with risk of
arteriovenous malformations. Neurosurgery hemorrhagic presentation of cerebral arteriovenous
34(5):801–807, discussion 807–8 malformations. Neurosurgery 42(3):481–486, discus-
22. Miyasaka Y et al (1992) An analysis of the sion 487–9
venous drainage system as a factor in hemorrhage 38. Shi X, Wang Z, Yu C (1993) An analysis of the
from arteriovenous malformations. J Neurosurg correlation between angiographic and clinical find-
76(2):239–243 ings in cerebral arteriovenous malformations. Chin
23. Mansmann U et al (2000) Factors associated with Med Sci J 8(1):35–37
intracranial hemorrhage in cases of cerebral arterio- 39. Albert P et al (1990) A study on the venous drainage
venous malformation. Neurosurgery 46(2):272–279, of 150 cerebral arteriovenous malformations as
discussion 279–81 related to haemorrhagic risks and size of the lesion.
24. Meisel HJ et al (2000) Cerebral arteriovenous Acta Neurochir (Wien) 103(1–2):30–34
malformations and associated aneurysms: analysis 40. Nataf F et al (1998) Therapeutic strategy for cerebral
of 305 cases from a series of 662 patients. Neurosur- arteriovenous malformations. Proposal for classifica-
gery 46(4):793–800, discussion 800–2 tion of individual hemorrhagic risk. Neurochirurgie
25. Halim AX et al (2004) Longitudinal risk of intracra- 44(2):83–93
nial hemorrhage in patients with arteriovenous mal- 41. Abecassis IJ et al (2014) Natural history of brain
formation of the brain within a defined population. arteriovenous malformations: a systematic review.
Stroke 35(7):1697–1702 Neurosurg Focus 37(3), E7
26. Stapf C et al (2000) Incident hemorrhage risk of brain 42. Gross BA, Du R (2013) Natural history of cerebral
arteriovenous malformations located in the arterial arteriovenous malformations: a meta-analysis.
borderzones. Stroke 31(10):2365–2368 J Neurosurg 118(2):437–443
27. Brown RD Jr, Wiebers DO, Forbes GS (1990) 43. Sahlein DH et al (2014) Features predictive of
Unruptured intracranial aneurysms and arteriovenous brain arteriovenous malformation hemorrhage:
malformations: frequency of intracranial hemorrhage extrapolation to a physiologic model. Stroke
and relationship of lesions. J Neurosurg 45(7):1964–1970
73(6):859–863 44. Stapf C et al (2002) Incidence of adult brain arterio-
28. Khaw AV et al (2004) Association of infratentorial venous malformation hemorrhage in a prospective
brain arteriovenous malformations with hemorrhage population-based stroke survey. Cerebrovasc Dis
at initial presentation. Stroke 35(3):660–663 13(1):43–46
28 Brain Arteriovenous Malformations 637

45. Zammar SG et al (2014) Precision medicine in brain 61. Gault J et al (2004) Pathobiology of human cerebro-
arteriovenous malformation management: arteries vascular malformations: basic mechanisms and clini-
steal the show but veins may hold the crystal ball. cal relevance. Neurosurgery 55(1):1–16, discussion
Neurosurgery 75(6):N13–N14 16–7
46. Wolf RL et al (2008) Arteriovenous shunt visualiza- 62. Meng JS, Okeda R (2001) Histopathological structure
tion in arteriovenous malformations with arterial spin- of the pial arteriovenous malformation in adults:
labeling MR imaging. AJNR Am J Neuroradiol observation by reconstruction of serial sections of
29(4):681–687 four surgical specimens. Acta Neuropathol
47. Ansari SA et al (2013) Intracranial 4D flow MRI: 102(1):63–68
toward individualized assessment of arteriovenous 63. Raybaud C (2010) Normal and abnormal embryology
malformation hemodynamics and treatment-induced and development of the intracranial vascular system.
changes. AJNR Am J Neuroradiol 34(10):1922–1928 Neurosurg Clin N Am 21(3):399–426
48. Brown RD Jr et al (1996) Incidence and prevalence of 64. Wittko-Schneider IM, Schnieder FT, Plate KH (2014)
intracranial vascular malformations in Olmsted Cerebral Angiogenesis During Development: Who Is
County, Minnesota, 1965 to 1992. Neurology Conducting the Orchestra. In: Milner R (ed) Cerebral
46(4):949–952 angiogenesis: methods and protocols. Methods in
49. ApSimon HT et al (2002) A population-based study molecular biology. Springer, New York, PP 3–15
of brain arteriovenous malformation: long-term treat- 65. Byrne JV (2012) Tutorials in endovascular neurosur-
ment outcomes. Stroke 33(12):2794–2800 gery and interventional neuroradiology. In: Byrne JV
50. Al-Shahi R et al (2003) Prospective, population-based (ed) Vascular malformations of the brain. Springer
detection of intracranial vascular malformations in Heidelberg Dordrecht , London, New York
adults: the Scottish Intracranial Vascular Malformation 66. Carmeliet P (2000) Mechanisms of angiogenesis and
Study (SIVMS). Stroke 34(5):1163–1169 arteriogenesis. Nat Med 6(4):389–395
51. Stapf C et al (2003) The New York Islands AVM 67. Mullan S et al (1996) Embryological basis of some
Study: design, study progress, and initial results. aspects of cerebral vascular fistulas and
Stroke 34(5):e29–e33 malformations. J Neurosurg 85(1):1–8
52. Hillman J (2001) Population-based analysis of arte- 68. Risau W (1997) Mechanisms of angiogenesis. Nature
riovenous malformation treatment. J Neurosurg 386(6626):671–674
95(4):633–637 69. Matsubara S et al (2000) Analysis of endoglin expres-
53. Weber F, Knopf H (2006) Incidental findings in mag- sion in normal brain tissue and in cerebral arteriove-
netic resonance imaging of the brains of healthy nous malformations. Stroke 31(11):2653–2660
young men. J Neurol Sci 240(1–2):81–84 70. Atri D et al (2013) Endothelial signaling and the
54. Berman MF et al (2000) The epidemiology of molecular basis of arteriovenous malformation. Cell
brain arteriovenous malformations. Neurosurgery Mol Life Sci 71(5):867–883
47(2):389–396, discussion 397 71. Carmeliet P, Jain RK (2011) Molecular mechanisms
55. Katzman GL, Dagher AP, Patronas NJ (1999) Inci- and clinical applications of angiogenesis. Nature
dental findings on brain magnetic resonance imaging 473(7347):298–307
from 1000 asymptomatic volunteers. JAMA 72. Risau W et al (1988) Vasculogenesis and angiogenesis
282(1):36–39 in embryonic-stem-cell-derived embryoid bodies.
56. Tsushima Y, Taketomi-Takahashi A, Endo K (2005) Development 102(3):471–478
Prevalence of abnormal findings on brain magnetic 73. Houdart E et al (1993) A proposed angiographic
resonance (MR) examinations in adult participants of classification of intracranial arteriovenous fistulae
brain docking. BMC Neurol 5:18 and malformations. Neuroradiology 35(5):381–385
57. Yue NC et al (1997) Clinically serious abnormalities 74. Niimi Y et al (1991) Pial arteriovenous fistula of the
found incidentally at MR imaging of the brain: data brain. Neuroradiology 33 (Suppl) (Proceedings of the
from the Cardiovascular Health Study. Radiology XIV Symposium Neuroradiologicum): 545–547
202(1):41–46 75. Forsting M, Wanke I (2008) Intracranial vascular
58. Jessurun GA et al (1993) Cerebral arteriovenous malformations and aneurysms: from diagnostic
malformations in The Netherlands Antilles. High work-up to endovascular therapy, 2nd rev. edn. Med-
prevalence of hereditary hemorrhagic telangiectasia- ical radiology – diagnostic imaging and radiation
related single and multiple cerebral arteriovenous oncology. Springer, Berlin, 291 p
malformations. Clin Neurol Neurosurg 76. Stapf C, Mohr JP (2000) New concepts in adult brain
95(3):193–198 arteriovenous malformations. Curr Opin Neurol
59. Gabriel RA et al (2010) Ten-year detection rate of 13(1):63–67
brain arteriovenous malformations in a large, multi- 77. Choi JH, Mohr JP (2005) Brain arteriovenous
ethnic, defined population. Stroke 41(1):21–26 malformations in adults. Lancet Neurol 4(5):299–308
60. Moftakhar P et al (2009) Cerebral arteriovenous 78. Valavanis A, Yasargil MG (1998) The endovascular
malformations. Part 1: cellular and molecular biology. treatment of brain arteriovenous malformations. In:
Neurosurg Focus 26(5):E10 Cohadon F et al (eds) Advances and technical
638 D. Sahlein and N. Manning

standards in neurosurgery, vol 34. Springer/Wien, the visual pathway. Neurosurgery 58(2):331–337,
New York discussion 331–7
79. Navarrete MG et al (2014) Brain vascular lesions: a 94. Latchaw RE et al (1995) Functional magnetic reso-
clinicopathologic, immunohistochemistry, and nance imaging as a management tool for cerebral
ultrastructural approach. Ann Diagn Pathol arteriovenous malformations. Neurosurgery
18(4):193–198 37(4):619–625, discussion 625–6
80. McCormick WF (1966) The pathology of vascular 95. Guo Y et al (2012) Silent intralesional
(“arteriovenous”) malformations. J Neurosurg 24 microhemorrhage as a risk factor for brain arteriove-
(4):807–816 nous malformation rupture. Stroke 43(5):1240–1246
81. George U et al (2010) Susceptibility-weighted imag- 96. Griffiths PD et al (2000) Brain arteriovenous
ing in the evaluation of brain arteriovenous malformations: assessment with dynamic MR digital
malformations. Neurol India 58(4):608–614 subtraction angiography. AJNR Am J Neuroradiol
82. Letteboer TG et al (2006) Genotype-phenotype rela- 21(10):1892–1899
tionship in hereditary haemorrhagic telangiectasia. 97. D’Aliberti G et al (2015) Arterial and venous aneu-
J Med Genet 43(4):371–377 rysms associated with arteriovenous malformations.
83. Mahmoud M et al (2010) Pathogenesis of arteriove- World Neurosurg 83(2):188–196
nous malformations in the absence of endoglin. Circ 98. Redekop G et al (1998) Arterial aneurysms associated
Res 106(8):1425–1433 with cerebral arteriovenous malformations: classifica-
84. Pawlikowska L et al (2005) Polymorphisms in tion, incidence, and risk of hemorrhage. J Neurosurg
transforming growth factor-beta-related genes ALK1 89(4):539–546
and ENG are associated with sporadic brain arterio- 99. Okamoto S, Handa H, Hashimoto N (1984) Location
venous malformations. Stroke 36(10):2278–2280 of intracranial aneurysms associated with cerebral
85. Shenkar R et al (2003) Differential gene expression in arteriovenous malformation: statistical analysis.
human cerebrovascular malformations. Neurosurgery Surg Neurol 22(4):335–340
52(2):465–477, discussion 477–8 100. Lasjaunias P et al (1988) Cerebral arteriovenous
86. Sure U et al (2004) Hypoxia-inducible factor and malformations (C. AVM) and associated arterial aneu-
vascular endothelial growth factor are expressed rysms (AA). Analysis of 101 C. AVM cases, with
more frequently in embolized than in nonembolized 37 AA in 23 patients. Acta Neurochir (Wien)
cerebral arteriovenous malformations. Neurosurgery 91(1–2):29–36
55(3):663–669, discussion 669–70 101. Rutledge WC et al (2014) Hemorrhage rates and risk
87. Sure U et al (2001) Endothelial proliferation, factors in the natural history course of brain
neoangiogenesis, and potential de novo generation arteriovenous malformations. Transl Stroke Res
of cerebrovascular malformations. J Neurosurg 5(5):538–542
94(6):972–977 102. Fiehler J, Stapf C (2008) ARUBA–beating natural
88. Delgado Almandoz JE et al (2010) Practical scoring history in unruptured brain AVMs by intervention.
system for the identification of patients with intrace- Neuroradiology 50(6):465–467
rebral hemorrhage at highest risk of harboring an 103. Cockroft KM et al (2012) A perfect storm: how a
underlying vascular etiology: the Secondary Intrace- randomized trial of unruptured brain arteriovenous
rebral Hemorrhage Score. AJNR Am J Neuroradiol malformations’ (ARUBA’s) trial design challenges
31(9):1653–1660 notions of external validity. Stroke 43(7):1979–1981
89. Sanelli PC, Mifsud MJ, Stieg PE (2004) Role of CT 104. Mocco J et al (2013) Randomized clinical trials: the
angiography in guiding management decisions of newly double edged sword. J Neurointerv Surg
diagnosed and residual arteriovenous malformations. 5(5):387–390
AJR Am J Roentgenol 183(4):1123–1126 105. Amin-Hanjani S (2014) ARUBA results are not appli-
90. Matsumoto M et al (2007) Dynamic 3D-CT angiog- cable to all patients with arteriovenous malformation.
raphy. AJNR Am J Neuroradiol 28(2):299–304 Stroke 45(5):1539–1540
91. Gross BA, Frerichs KU, Du R (2012) Sensitivity of 106. DeMeritt JS et al (1995) Outcome analysis of preop-
CT angiography, T2-weighted MRI, and magnetic erative embolization with N-butyl cyanoacrylate in
resonance angiography in detecting cerebral arterio- cerebral arteriovenous malformations. AJNR Am J
venous malformations and associated aneurysms. Neuroradiol 16(9):1801–1807
J Clin Neurosci 19(8):1093–1095 107. Spetzler RF et al (1987) Surgical management of large
92. Meretoja A et al (2014) The spot sign and tranexamic AVM’s by staged embolization and operative exci-
acid on preventing ICH growth–AUStralasia Trial sion. J Neurosurg 67(1):17–28
(STOP-AUST): protocol of a phase II randomized, 108. Pasqualin A et al (1991) Treatment of cerebral arte-
placebo-controlled, double-blind, multicenter trial. riovenous malformations with a combination of pre-
Int J Stroke 9(4):519–524 operative embolization and surgery. Neurosurgery
93. Kikuta K et al (2006) Early experience with 3-T 29(3):358–368
magnetic resonance tractography in the surgery of 109. Jafar JJ et al (1993) The effect of embolization with N-
cerebral arteriovenous malformations in and around butyl cyanoacrylate prior to surgical resection of
28 Brain Arteriovenous Malformations 639

cerebral arteriovenous malformations. J Neurosurg gamma knife radiosurgery. J Neurosurg 93(Suppl


78(1):60–69 3):96–101
110. Vinuela F et al (1991) Combined endovascular embo- 124. Ding D et al (2013) Radiosurgery for patients with
lization and surgery in the management of cerebral unruptured intracranial arteriovenous malformations.
arteriovenous malformations: experience with J Neurosurg 118(5):958–966
101 cases. J Neurosurg 75(6):856–864 125. Wegner RE et al (2011) A modified radiosurgery-
111. Ovalle F, Shay SD, Mericle RA (2012) Delayed intra- based arteriovenous malformation grading scale and
cerebral hemorrhage after uneventful embolization of its correlation with outcomes. Int J Radiat Oncol Biol
brain arteriovenous malformations is related to vol- Phys 79(4):1147–1150
ume of embolic agent administered: multivariate anal- 126. Kano H et al (2013) Stereotactic radiosurgery after
ysis of 13 predictive factors. Neurosurgery 70(2 Suppl embolization for arteriovenous malformations. Prog
Operative):313–320 Neurol Surg 27:89–96
112. van Rooij WJ et al (2012) Curative embolization of 127. Parkhutik Vet al (2012) Postradiosurgery hemorrhage
brain arteriovenous malformations with onyx: patient rates of arteriovenous malformations of the brain:
selection, embolization technique, and results. AJNR influencing factors and evolution with time. Stroke
Am J Neuroradiol 33(7):1299–1304 43(5):1247–1252
113. Iosif C et al (2015) Endovascular transvenous cure for 128. Barker FG 2nd et al (2003) Dose-volume prediction
ruptured brain arteriovenous malformations in com- of radiation-related complications after proton beam
plex cases with high Spetzler-Martin grades. radiosurgery for cerebral arteriovenous
J Neurosurg 122(5):1229–38 malformations. J Neurosurg 99(2):254–263
114. Taylor CL et al (2004) Complications of preoperative 129. Pollock BE et al (2000) The rationale and technique
embolization of cerebral arteriovenous of staged-volume arteriovenous malformation
malformations. J Neurosurg 100(5):810–812 radiosurgery. Int J Radiat Oncol Biol Phys
115. Debrun GM et al (1997) Embolization of the nidus of 48(3):817–824
brain arteriovenous malformations with n-butyl cya- 130. Firlik AD et al (1998) Staged volume radiosurgery
noacrylate. Neurosurgery 40(1):112–120, discussion followed by microsurgical resection: a novel treat-
120–1 ment for giant cerebral arteriovenous malformations:
116. van Rooij WJ, Sluzewski M, Beute GN (2007) Brain technical case report. Neurosurgery 43(5):1223–1228
AVM embolization with Onyx. AJNR Am J 131. Karlsson B et al (2007) Is repeated radiosurgery an
Neuroradiol 28(1):172–177, discussion 178 alternative to staged radiosurgery for very large brain
117. Perez-Higueras A, Lopez RR, Tapia DQ (2005) arteriovenous malformations? J Neurosurg
Endovascular treatment of cerebral AVM: our experi- 107(4):740–744
ence with onyx. Interv Neuroradiol 11(Suppl 132. Karlsson B et al (2005) Long-term results after frac-
1):141–157 tionated radiation therapy for large brain arteriove-
118. Pierot L et al (2005) Endovascular treatment of brain nous malformations. Neurosurgery 57(1):42–49,
arteriovenous malformations using onyx: preliminary discussion 42–9
results of a prospective multicenter study. Interv 133. Foote KD et al (2003) Salvage retreatment after fail-
Neuroradiol 11(Suppl 1):159–164 ure of radiosurgery in patients with arteriovenous
119. Pierot L et al (2013) Endovascular treatment of brain malformations. J Neurosurg 98(2):337–341
arteriovenous malformations using a liquid embolic 134. Lindvall P et al (2003) Hypofractionated conformal
agent: results of a prospective, multicentre study stereotactic radiotherapy for arteriovenous
(BRAVO). Eur Radiol 23(10):2838–2845 malformations. Neurosurgery 53(5):1036–1042,
120. Nagy G et al (2012) Stereotactic radiosurgery for discussion 1042–3
arteriovenous malformations located in deep critical 135. Veznedaroglu E et al (2004) Fractionated stereotactic
regions. Neurosurgery 70(6):1458–1469, discussion radiotherapy for the treatment of large arteriovenous
1469–71 malformations with or without previous partial embo-
121. Flickinger JC et al (1996) A dose–response analysis lization. Neurosurgery 55(3):519–530, discussion
of arteriovenous malformation obliteration after 530–1
radiosurgery. Int J Radiat Oncol Biol Phys 136. Huang PP et al (2012) Long-term outcomes after
36(4):873–879 staged-volume stereotactic radiosurgery for
122. Karlsson B, Lindquist C, Steiner L (1997) Prediction large arteriovenous malformations. Neurosurgery
of obliteration after gamma knife surgery for cerebral 71(3):632–643, discussion 643–4
arteriovenous malformations. Neurosurgery 40 137. Schwyzer L et al (2012) Long-term results of gamma
(3):425–430, discussion 430–1 knife surgery for partially embolized arteriovenous
123. Chang JH et al (2000) Factors related to complete malformations. Neurosurgery 71(6):1139–1147,
occlusion of arteriovenous malformations after discussion 1147–8
Dural AVF and Treatment
29
Pierre Guedin

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 642 Intracranial arteriovenous dural shunts
(ICADS) account for 10–15 % of all intracra-
Physiopathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 642
nial vascular malformations.
Classification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 643 The natural history of ICADS is based on
Clinical Symptoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 646 the venous drainage and the presence or
absence of leptomeningeal venous reflux.
Natural History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 646
Benign ICADS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 646 CT and MR can provide useful informations
Malignant ICADS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 647 for diagnosis, but DSA is mandatory to analyze
Imaging Strategy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 647 the lesion and to understand the venous drain-
Benign ICADS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 647 age of the shunt and of the brain.
Malignant ICADS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 648 ICADS are dynamic lesions, i.e., a benign
Digital Subtraction Angiography (DSA) . . . . . . . . . . . . 648 fistula can turn into a malignant ICADS with
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 648 time, but may also regress spontaneously.
Transvenous Embolization . . . . . . . . . . . . . . . . . . . . . . . . . . . 648 The spontaneous morbi-mortality of the dis-
Transarterial Embolization . . . . . . . . . . . . . . . . . . . . . . . . . . . 650
Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 650
ease dramatically increases in case of
Stereotactic Radiosurgery (SRS) . . . . . . . . . . . . . . . . . . . . 655 leptomeningeal venous reflux and require
thus a curative treatment.
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 655
Endovascular approach is nowadays the
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 655 first intention treatment, either transarterially
or transvenously. This should be proposed in
case of malignant ICADS and benign ICADS
with intolerable symptoms.

Keywords
Intracranial dural fistula • Cortical venous
drainage • Embolization • Glue • Onyx

P. Guedin (*)
Department of Diagnostic and Therapeutic
Neuroradiology, Hôpital Foch, Suresnes, France
e-mail: p.guedin@hopital-foch.org

# Springer Science+Business Media New York 2016 641


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_43
642 P. Guedin

Introduction Physiopathology

Intracranial arteriovenous dural shunts (ICADS) A wide range of pathologies or conditions (e.g.,
represent abnormal communications between traumatisms, surgery, infectious diseases,
arteries and veins that are located inside the dura coagulopathy, dehydration, hormonal deficits)
mater. They account for 10–15 % of all intracra- have been reported in the medical history of
nial vascular malformations [1]. patients presenting with ICADS. However, not
Arterial feeders are most frequently meningeal all of the patients having suffered from these
branches arising from the external carotid artery diseases develop ICADS, even after many years
(ECA), the internal carotid artery (ICA), or the of evolution. This points out the fact that an indi-
vertebral artery (VA), while the venous drainage vidual’s susceptibility influences the occurrence
can be taken in charge by either dural sinuses or of these acquired lesions.
veins or by cerebral veins. Venous hypertension has been supposed to be
The term “shunt” encompasses two architec- the main pathogenic factor in the creation of these
tural types: AV fistulas and niduses. Nidus-type lesions. It can occur associated with
shunts are characterized by a network of abnormal non-thrombotic diseases (like direct sinusal com-
vessels placed between feeding arteries and pression by a space occupying lesion, or surgical
draining veins (either sinuses or leptomeningeal sinusal sacrifice) but is mainly considered to be
veins), whereas fistulas are AV connections with related to thrombotic events.
arterial feeders ending directly into draining Three potential steps in the creation of ICADS
veins. In our daily practice, we regularly use the have thus been described. The first step is a sinusal
term “dural fistula” only in this latter thrombophlebitis and/or stenosis reducing the
angioarchitecture. venous outflow and inducing a venous hyperten-
The venous drainage of ICADS influences the sion. The second step consists in the opening of
natural history of these lesions [2–4]. If the microshunts inside the dura mater related to this
venous drainage is exclusively in dural sinuses venous hypertension and congestion, associated
or dural veins, the venous drainage of the brain with the release of angiogenic factors due to the
is not compromised; those lesions are thus con- thrombus itself (as vascular endothelial growth
sidered as “benign” because they do not carry a factor (VEGF), basic fibroblast growth factor
risk of hemorrhage or neurological symptoms. If (FGF)) and to the ischemic brain parenchyma.
however the shunt induces reflux into cortical The third step is the recanalization of the sinus.
leptomeningeal or deep veins, this correlates Incomplete recanalization associated to the shunts
with a high spontaneous risk of morbi-mortality and arterialization of the venous outflow can pro-
related to venous congestion and rupture giving, voke a reflux in leptomeningeal veins and give
respectively, rise to seizures, deficits, or hemor- rise to an aggressive dural shunt with neurological
rhage. Those lesions are thus considered as consequences [6].
“aggressive.” Even if conceptually seducing, this initial
Pediatric dural shunts are different from hypothesis has been counterbalanced by clinical
adult ones: not only do they vary according evidences. In fact very few patients suffering from
to the age groups that can be affected thrombophlebitis will develop an ICADS, and the
(dural sinus malformations in neonates or vast majority of patients with ICADS do not pre-
infants, juvenile dural shunts in infants, sent clinical and radiographical signs of present or
adult-ype shunts in older children), but they previous thrombophlebitis [1]. Other hypotheses
also have specific symptoms and natural have thus been imagined to understand the crea-
histories [5]. This group of lesions will not be tion of ICADSs: inflammation occurs at the pen-
addressed here. etrating point of the emissary vein on the dura due
29 Dural AVF and Treatment 643

to idiopathic or secondary causes. Local inflam- Table 1 Classification of ICADS


matory reactions induce vessel dilatation and ICADS Classification
neovascularization and subsequently create arte- Venous
riovenous connections on the arteriole level. Localization drainage Borden Lariboisière
Although emissary vein (EV) communicating Dural sinus Sinusal, 1 I
with dural arteries might play a role as draining anterograde
Dural sinus Sinusal, 1 IIa
routes at first, they start to degrade due to com-
anterograde/
pression of enlarged emissary arteries or to a retrograde
hemodynamic shift to the drainage pathway of Dural sinus Sinusal, 1 IIa
least resistance following, the occlusion of drain- (occluded) retrograde
age pathway through EV into the sinus or cortical Dural sinus Sinusal, 2 IIb
veins may form, resulting in clinically detectable anterograde,
CVR
ICADS. The shunt then expands to the surround-
Dural sinus Sinusal, 2 II a + b
ing dura associated with recruitment of feeders anterograde/
from distant sites induced by expression of angio- retrograde,
genic factors and a shift in the hemodynamic CVR
balance. In sinus-type ICADS, the sinus is pro- Cortical vein CVR 3 III
gressively compartmentalized and finally Dural sinus CVR 3 III
(tandem
occludes due to thrombogenesis with activated occlusion with
coagulopathy or to hemodynamic hypertrophy of CVR)
the sinus wall. This progression results in the Ectatic CVR 3 IV
mature, aggressive fistula with drainage impair- cortical vein
ments. Previous mechanistic hypotheses focusing Perimedullary CVR 3 V
vein
on sinus hypertension and sinus thromboses can-
not explain the pathogenesis of non-sinus type of
ICADS [7].

Classification

ICADSs are most frequently classified according


to either Lariboisière or Borden classification
(Table 1). Both are based on the analysis of the
angiographic venous drainage and correlate to the
natural history [8, 9]. The leptomeningeal or deep
venous reflux appears to be the best predictive
factor of the severity of the lesion. The “benign”
types are represented by Lariboisière I (Fig. 1) and
IIa and Borden 1, whereas “malignant” types
encompass Lariboisière IIb (Fig. 2), III (Fig. 3),
IV, and V and Borden 2 and 3 [3, 10, 11].
Recently Lasjaunias et al. proposed a revised
classification of cranial and spinal dural shunts
Fig. 1 Cerebral angiogram, lateral view. Selective
based on embryologic and anatomical specific- catheterism of the occipital artery: intracranial dural arte-
ities [12]. This classification considers epidural riovenous shunt of the lateral sinus with exclusive sinusal
compartments in three groups: the ventral drainage
644 P. Guedin

Fig. 2 Cerebral angiogram, lateral view, selective lateral sinus with reflux into the vein of Labbé.
catheterism of the maxillary (a) and occipital artery (b). Posttreatment lateral view of the cast of glue into the lateral
Intracranial dural arteriovenous shunt of a thrombosed sinus (“sinus suspendu”)

epidural, the dorsal epidural, and lateral epidural the bony structures, and they drain outside the
group. Three observations of the venous develop- bony limits, thus resulting in no subarachnoid
ment of the brain and spinal cord and adjacent (spinal or cortical) venous reflux. However,
bony structures contributed to the generation of some factors may precipitate development of
this classification. CVR in these regions such as thrombosis within
The venous system of the notochord and the epidural space surrounding, remote, or distal
corresponding sclerotome extends from the to the shunt. This will produce reflux in the lateral
basisphenoid (cavernous plexus) to the sacrum epidural space as seen in rare cases of ventral
and gives rise to the ventral epidural drainage epidural spinal shunts with perimedullary venous
group. It collects the blood from spongious bony reflux. CVR can also be encountered in high-flow
structures and has no primary role in the drainage shunts forcing the emissary-bridging vein open-
of the central nervous system. It includes the ing after reflux in the lateral epidural space. Ven-
basioccipital bone, the petrous pyramid, the tral epidural shunts are characterized by a strong
basisphenoid with its adjacent sphenoid wings, female predominance and absence of CVR unless
and their related dural structures. The venous extensive thrombosis of epidural drainage or
afferents of these regions are closely related to high-flow shunts.
29 Dural AVF and Treatment 645

mean age, and can be multiple. Usually no


leptomeningeal venous reflux is seen, and
craniofugal flow is normally observed.
Leptomeningeal reflux may occur following asso-
ciated venous outflow restriction or with high-
flow shunts. Variations of the venous opening
into this system or distal restriction of the sinus
opening will also be responsible for cortical
venous reflux.
The veins draining the central nervous system
(both spinal and cranial) are not related to
the peripheral nerves but use “emissary” or
“bridging veins.” These latter join the lateral
epidural venous spaces as connecting drainage
system. The leptomeningeal venous drainage
has no direct confluent communication with
the ventral and dorsal epidural venous
Fig. 3 Cerebral angiography, lateral view. Selective plexuses, which drain the skull and spine.
catheterism of the middle meningeal artery. Intracranial While the typical lateral epidural shunts corre-
dural arteriovenous shunt of the dural wall of the lateral
sinus with direct fistula into a temporal vein with secondary
spond to spinal dural fistulas that drain into emis-
opacification of the superior longitudinal sinus and lateral sary or radicular veins joining the perimedullary
sinus venous plexuses, intracranial pathological loca-
tions involve emissary-bridging veins of the
The dorsal epidural venous space includes the brainstem and their homologs draining deep
superior sagittal sinus (SSS), torcular, transverse cerebral structures such as the condyloid vein
sinus, medial occipital sinus, and posterior mar- at the foramen magnum, the superior petrosal
ginal sinus at the foramen magnum. It is poorly vein, the basal vein, the vein of Galen, the veins
developed at the spinal level, and the presence of of the anterior cranial fossa, and the orbit. Thus,
dorsally located dural sinuses intracranially is the following DAVS locations are encountered:
therefore the major difference between the venous marginal sinus, falco-tentorial angle, petrosal
systems of the brain and spine. Their formation is and basi-tentorial areas, Breschet sinus,
linked to the appearance, during evolution, of the paracavernous and intraorbital regions, and lam-
dural falces and tentorium and associated with the ina cribriformis. The drainage is always directed
development of the paleo- and neopallial struc- towards leptomeningeal veins, and this type of
tures. These sinuses result from the confluence in DAVS is therefore always considered to be
the epidural space of two different venous sys- aggressive. Lateral epidural shunts are character-
tems: the osseous system draining the cranial ized by a male predominance and present at
vault and the leptomeningeal system draining the later ages.
brain. The venous pressure within the dural The significant difference of the patient char-
sinuses is usually low; thus, antegrade and acteristics (sex and age) between the groups of the
craniofugal flow of the shunts is normally new classification suggests the existence of fun-
observed. CVR may occur after associated venous damental differences, either biological or hor-
outflow restriction or with high-flow shunts; in monal, within the epidural venous systems in
case of variations of the venous opening into this each region and may point towards different eti-
system such as presence of an accessory epidural ologies of these three different groups of arterio-
sinus, distal restriction of the sinus opening will venous shunts. Furthermore, this classification
rapidly produce CVR. Dorsal epidural shunts allows a better understanding of various physio-
have no sex predominance, present at lower pathological phenomena.
646 P. Guedin

Clinical Symptoms Table 2 Differential diagnosis of pulsatile tinnitus


Pulsatile tinnitus etiologies
Two modes of presentation can be distinguished: Arterial Venous Nonvascular
those due to hemorrhage and those due to venous Developmental Benign Myoclonia
congestion [2]. Hemorrhagic complications, i.e., Aberrant intracranial
internal carotid hypertension
parenchymal, subarachnoid, or subdural hemor-
artery
rhages, present acutely in a stroke-like manner, Persistent
whereas nonhemorrhagic neurological presenta- stapedial artery
tions have often a subacute or slowly progressive Neurovascular
conflict
onset. Cranial nerve palsy, ocular symptoms,
Arteriopathy Venous Semicircular
dementia, seizures, and encephalopathy are the
Atheroma dehiscent canal
usual symptoms associated to venous Fibromuscular jugular bulb dehiscence
hypertension. dysplasia thrombophlebitis
Those symptoms will vary accordingly to the Dissection
Aneurysm
localization and the type of the shunt.
Vascular tumors
Pulsatile tinnitus (PT) is due to arterialization Paraganglioma
and flow turbulences in the shunting zone and the Endolymphatic
surrounding venous structures, which can be trans- sac tumor
mitted to the petrous bone. They are presented in Meningioma
Hemangioma
60 % of patients, mainly those presenting with Hypervascular
direct sinusal drainages. The differential diagnoses metastasis
of PT are listed in Table 2 [1, 10]. Osseous
Cavernous sinus ICADS typically present with dysplasia
ophthalmoplegia, proptosis, chemosis, retro- Otospongiosis
Paget disease
orbital pain, or decreased visual acuity.
Vascular shunts
During the follow-up of non-treated patients Arteriovenous
with ICADS, any modification of the noise inten- malformation
sity (increase, decrease, or disappearance) should Dural fistulas
be explored by a MRI/MRA and/or DSA. Indeed,
changes in the architecture of the ICADS as
thrombosis of the sinus may reroute the venous 73 patients (62 %). Intolerable bruit or ophthal-
drainage through cortical veins and would put the mological sequelae were deemed indications for
patient in a risky situation because of the transfor- palliative embolization in 43 patients and surgical
mation of the lesion from a benign into an aggres- treatment in one patient. A median follow-up
sive one. period of 27.9 months (range 1 month – 17.5
years) was available in 112 patients (95.7 %),
among whom repeated angiography was
Natural History performed in 50. Overall, observational and palli-
ative management resulted in a benign and toler-
Benign ICADS able level of disease in 110 (98.2 %) of 112 cases.
In two cases managed conservatively CVR devel-
Satomi et al. reported a series of 117 patients oped with spontaneous progressive thrombosis of
harboring benign cranial DAVFs. At the initial venous outlets. The disease course of a cranial
assessment, four patients were asymptomatic. DAVF without CVD is indeed benign, obviating
Most patients presented with benign symptoms: the need for a cure of these lesions. However, a
chronic headache, bruit, or orbital phenomena. benign DAVF carries a 2 % risk of developing
Observational management was instituted in CVD, mandating close clinical follow-up and
29 Dural AVF and Treatment 647

renewed radiological evaluation in response to presenting event. Three patients bled after diag-
any deterioration in the patient’s condition [4]. nosis. One of these patients died. Apart from
deficits caused by hemorrhage, no patient
reported adverse neurological symptoms. In
Malignant ICADS patients presenting with an intracranial hemor-
rhage, the annual risk for hemorrhage is approx-
The natural history of aggressive (Borden 2 and 3) imately 7.4 %, and in those not presenting with a
cranial dural arteriovenous fistulas is not well hemorrhage, it is approximately 1.5 %. They
described. Reported annual mortality and hemor- conclude that the risk of intracranial hemorrhage
rhage rates vary widely and range up to 20 % per from a dural arteriovenous shunt with cortical
year. Van Dijk et al. reported a consecutive single- venous drainage is most likely smaller than pre-
center cohort of 236 cases that presented with a viously proposed, but presentation with hemor-
cranial DAVF between June 1984 and May 2001. rhage is a risk factor for hemorrhage [14].
A group of 118 cranial DAVFs was selected for the Duffau et al. reported a series of 20 patients
presence of CVR. All patients were offered treat- suffering from dural fistulas with cortical venous
ment aimed at the disconnection of the CVR. drainage revealed by intracranial hemorrhage.
Patients who declined or had partial treatment with The mean duration between the first hemorrhage
persistence of the CVR had long-term clinical and and treatment was 20 days. Seven patients (35 %)
angiographic follow-up to study the disease course presented with acute worsening during this delay
of this select group. Fourteen non-treated patients due to radiologically proven early rebleeding with
(11.9 %) and six partially treated patients were graver consequences than the first hemorrhage.
assessed clinically and angiographically over time. They therefore advocate complete and early treat-
The mean follow-up in this select group was 4.3 ment in all cases of AVF with cortical venous
years (86.9 patient-years). During follow-up, seven drainage revealed by an ICH [15].
patients suffered an intracranial hemorrhage (35 %).
The incidence of nonhemorrhagic neurological def-
icit was 30 %. Nine patients (45 %) died: six patients Imaging Strategy
expired after a hemorrhage, and three patients died
of progressive neurological deterioration. Two Benign ICADS
patients demonstrated a spontaneous closure of the
DAVF (10 %). Authors concluded that persistence For high-flow shunts, cervical Doppler may find
of the CVR in dural shunts yields an annual mor- an increase of the output in the primitive carotid
tality rate of 10.4 %. Excluding events at presenta- and a diastolic flow in the external carotid artery.
tion, in this series the annual risk for hemorrhage or Cerebral CT is usually normal.
nonhemorrhagic neurological deficit during follow- Contrast-enhanced CT may reveal a mottling
up was 8.1 % and 6.9 %, respectively, resulting in an of the skull base due to transosseous arterial
annual event rate of 15.0 % [13]. feeder arising from the occipital artery in lateral
In a retrospective study of 85 patients suffering sinus shunt (Fig. 4).
from an ICADS with CVR, Soderman CTA may be useful in static depiction of
et al. evaluated the records of 85 patients with angioarchitecture, showing enlarged arterial
dural arteriovenous shunts with cortical venous feeders and early opacification of the sinus and
drainage or reflux hospitalized in their institution the jugular vein.
from 1978 to 2007. MR parenchymal sequence is useful to rule out
Fifty-three patients did not have an intracra- the absence of parenchymal edema.
nial hemorrhage as the presenting event. One of 3D TOF may be positive in large shunt with
these patients bled after diagnosis. Thirty-two high flow but may also be negative for smaller or
patients had an intracranial hemorrhage as the slow-flow shunts.
648 P. Guedin

for gross depiction of angioarchitecture and


dynamics.
MRI/MRA is useful in post therapeutic follow-
up [16].

Digital Subtraction Angiography (DSA)

DSA is the gold standard modality to detect and


classify ICADS. It should be performed every
time an ICADS is suspected, in order to make
the diagnosis, to distinguish benign and aggres-
sive type of shunt and then to plan the treatment.
Selective serial should be performed in both
carotid and vertebral arteries, also in both external
carotid arteries. Long serial should be done in
order to understand the venous drainage of the
brain, the patency of the sinus, the flow direction
in each sinus, and the venous drainage of the
fistula itself. Selective serial should be done in
each arterial feeders of the region to depict spe-
Fig. 4 T1 post-contrast MR: transosseous arterial feeders cific angioarchitecture of the shunt.
arising from the right occipital artery

3DMRA is useful for gross depiction of Treatment


angioarchitecture and dynamics.
For cavernous shunts, the signs are specific, The therapeutic strategy will vary accordingly to
encompassing a dilated superior ophthalmic vein, the venous drainage pattern. Indications of treat-
exophthalmos, and a dilated cavernous sinus. ment of benign ICADS are rare. They are limited
to intolerable clinical symptoms, mostly double
vision and pulsatile tinnitus. The risk of the treat-
Malignant ICADS ment should be extremely low and less than the
natural history of the disease itself. Aggressive
Cerebral non-contrast CT can reveal subarachnoid ICADS require curative treatment in order to dis-
hemorrhage, hematoma, and parenchymal edema. connect the leptomeningeal reflux and to prevent
Brain parenchyma calcifications reflecting the future intracranial hemorrhage or neurological
chronicity of the venous hypertension could rarely deficit. Several therapeutic approaches can be pro-
be seen. In cerebral atrophy, hydrocephaly can be posed: surgery, radiosurgery, and embolization.
pointed out as consequences of the shunt on the Endovascular treatment, transarterially or
brain parenchyma. transvenously, is often considered as the first
Contrast-enhanced CT may show tortuous intention treatment.
feeding arteries and dilated and tortuous cortical
veins.
MR Flair sequences are useful to depict the Transvenous Embolization
edema due to venous hypertension and ischemia
(Fig. 5). In case of type V fistulas, hyperintensity Several modalities of transvenous embolizations
in the cervical medulla could be found. As for have been described: catheterization of the
benign ICADS 3D TOF and MRA could be useful affected sinus by venous route followed by
29 Dural AVF and Treatment 649

Fig. 5 MRI, T2 coronal sequence (a) and T1 post contrast anteroposterior view (c) and lateral view (d, e): dural
(b): right cerebellar edema in posterior fossa due to venous shunt with cortical reflux in cerebellar vein and venous
congestion of cerebellar vein. Cerebral angiography in congestion in the late venous phase
650 P. Guedin

deposition of coils and/or liquid embolic agent the shunt. Progression of the microcatheter is
adjacent to the shunt, transcranial approach and helped by the use of a microguidewire in order
direct puncture of the pathological sinus if selec- to allow distal catheterization as close as possible
tive endovascular navigation is considered impos- to the shunt. The ideal injections of glue are
sible for anatomical or architectural reasons, or performed under a wedge position [20]. When a
selective obliteration of the CVR after intravenous safe embolization position is achieved, selective
navigation without occlusion of the dural sinus injection of contrast is performed in order to
[17–19]. This latter technique remains excep- depict the focal angioarchitecture of the
tional because of the tortuosity and fragility of malformative compartment. The dilution of the
the leptomeningeal veins and the risk of rupture cyanoacrylate glue mixed with Lipiodol will
during selective catheterization. Transvenous vary accordingly to the position of the catheter,
management is reserved to cases in which the the size of the vessel, and the flow of the shunt. n-
sinus is already partially thrombosed (Fig. 6). A BCA is injected in liquid form and solidifies on
patent sinus draining ICADS should not be contact with ionic solutions as blood. The injec-
occluded transvenously in first intention: the nor- tion duration is relatively short. Glue presents a
mal venous drainage of the brain could be thrombogenic effect that can promote secondary
compromised and lead to delayed ischemic com- occlusion of residual shunt [21] (Figs. 7 and 8).
plications. Furthermore, secondary development Onyx is a copolymer of ethylene-vinyl alcohol
of de novo ICADS can occur, leading then to mixed with a solvent, DMSO and tantalum pow-
potential impairments in the intracranial venous der, that reacts as a gelling agent. Mixed with
drainage if one sinus is already suppressed. blood, the solvent is volatile and disappears in
the arterial flow. The deposit of the polymer is
slow. The polymer initially precipitates within the
Transarterial Embolization peripheral area of blood vessel with secondary
occlusion of the central vessel. Onyx injection
The concept of this approach is to superselectively can alternatively be stopped and started again,
catheterize the arterial feeders and occlude via this with the polymer going in another vessel. This
route the shunt itself. Nowadays, transarterial technique allows to maintain the dural sinus pat-
embolization can be achieved with liquid embolic ent with focusing on arterio-arterial reflux sur-
agent (glue and Onyx) and particles. The use of rounding the shunting zone [22].
particles as sole embolic agent is getting more and
more restricted because of their instability and
short-term effects. They have thus no interest in Surgery
ICADS with CVR, and when needed, they are
mostly used in particular lesions as an adjuvant When surgical management is decided, the oper-
embolus to reduce the flow in collateral vessels ative technique has to be adapted to the type of
and help to induce thrombosis. venous drainage of the lesion: in aggressive dural
All vessels potentially giving rise to arterial shunts with leptomeningeal venous drainage, its
feeders of the shunt are catheterized in order to aim is to disconnect the origin of the draining
build up the lesional anatomy and architecture of vein. First described by Grisoli in 1984, surgery
the lesion. Particular attention is paid to the is considered to be the efficacious way of curing
venous drainage of the brain as compared to the the ICADS. If the shunt drains first into the sinus
drainage of the lesion. and secondarily in cortical veins, by
With glue (n-BCA), when embolization is disconnecting the CVR, the surgical technique
decided, and according to the size and aspects of leaves the sinusal drainage intact, converts the
the vessels, a microcatheter is used to aggressive DAVF into a benign lesion, and
superselectively catheterize the main feeder of changes thus the natural history of the shunt [23].
29 Dural AVF and Treatment 651

Fig. 6 (continued)
652 P. Guedin

Fig. 6 Cerebral angiogram, selective catheterism of left occluded. Treatment: transvenous approach through the
maxillary (a, b) and occipital artery (c) revealing a left inferior petrosal sinus and selective coiling of the cavern-
dural cavernous shunts with reflux into superior and infe- ous sinus (e), in association with a superselective injection
rior ophthalmic vein and cortical venous reflux into the left of glue inside the sinus. (f) Posttreatment selective injec-
sylvian vein. Selective catheterism of petrosquamous tion of internal maxillary artery (g, h) revealed the occlu-
branch of the middle meningeal artery (d) showing more sion of the shunt. Appreciate the specific exophthalmos
precisely the venous congestion of ophthalmic and left with vascular congestion of the left eye (i) pre- and
hemispheric veins. The left inferior petrous sinus is posttreatment (j)
29 Dural AVF and Treatment 653

Fig. 7 Angiograms obtained in a 49-year-old man who arteriovenous shunt of fistulous type draining initially into a
presented with two recurrent hemorrhages in the posterior petrous vein and resulting in congestion of the posterior fossa
fossa. (a, b) Lateral views of the left internal maxillary (a) veins. Other feeding vessels arose from the left internal and
and occipital (b) arteries demonstrating a petrotentorial dural ascending pharyngeal arteries (not shown). (c): Left VA
654 P. Guedin

Fig. 8 Angiograms obtained in a 75-year-old man who (b) Image obtained after superselective catheterization of
presented with acute headaches due to a right frontal lobe the ethmoidal feeding vessel was performed via the OA
hemorrhage. (a) Lateral view of a right ICA injection and confirmed this architecture. (c) Image showing embo-
showing an ethmoidal shunt located at the level of the lization of the whole shunt performed in wedge position
lamina cribriformis. The fistula is vascularized by a poste- with NBCA. (d) Right ICA angiogram obtained immedi-
rior ethmoidal artery arising from the ipsilateral ophthal- ately after treatment confirming cure of the shunt with
mic artery (OA). It drains into a frontal vein harboring a respect to the OA and its branches. The patient remained
false aneurysm (indicating the rupture point of the lesion) asymptomatic. Follow-up MR imaging and MR angiogra-
and reaches secondarily the superior sagittal sinus. phy obtained 3 months later confirmed cure of the lesion

Fig. 7 (continued) angiogram in anteroposterior view thus occluding the ICDAVS. (e–g) Immediate postembo-
confirming poor filling of left hemispheric cerebellar lization images confirming cure of the fistula. Injection of
veins due to congestion of the veins because of the all the previously described arteries, internal maxillary
ICADS. All of the feeding vessels converged towards a artery (e), occipital artery (f), as well as the internal carotid
single shunting zone. (d) Image obtained at the end of the artery (ICA) and ascending pharyngeal artery (not shown),
angiographic session showing the cast of glue injected into does not opacify the shunt anymore but fills all the regional
the shunt. Distal catheterization of the basal tentorial arteries without opacification of the venous drainage. Late
branch of the middle meningeal artery, which extended phase of the postembolization left VA angiogram (g) shows
into the fistula, was performed with a Magic 1.2 normal filling of the hemispheric veins, confirming the
microcatheter and a 50 % mixture of NBCA, and Lipiodol suppression of the venous congestion. A 5-month control
was injected in a wedge position to cast the shunt and angiogram (not shown) confirmed total obliteration of the
penetrate into the proximal portion of the petrous vein, fistula. The patient returned to normal life
29 Dural AVF and Treatment 655

Stereotactic Radiosurgery (SRS) 3. Davies MA, TerBrugge K, Willinsky R, Coyne T,


Saleh J, Wallace MC (1996) The validity of classifica-
tion for the clinical presentation of intracranial dural
SRS provides effective long-term relief of symp- arteriovenous fistulas. J Neurosurg 85(5):830–837
toms. It cannot be applied however to every type of 4. Satomi J, van Dijk JMC, Terbrugge KG, Willinsky RA,
ICADS and should be reserved to benign fistulas Wallace MC (2002) Benign cranial dural arteriovenous
because of the delay needed before obtaining the fistulas: outcome of conservative management based
on the natural history of the lesion. J Neurosurg
classical radiation-induced obliterative arteritis that 97(4):767–770
leads to cure of the shunt. The poor natural history 5. Berenstein A, Lasjaunias P, TerBrugge K (1987) Dural
of ICADS with CVR requires indeed more emer- arteriovenous shunts. In: Surgical neuroangiography.
gent treatments and rapid results. Furthermore, the Springer, Berlin, pp 389–451
6. Kraus JA, St€ uper BK, Nahser HC, Klockgether T,
results obtained by radiosurgery seem Berlit P (2000) Significantly increased prevalence of
unpredictable, as 30 % of lesions are not cured by factor V Leiden in patients with dural arteriovenous
this technique. CVR, a risk factor for intracranial fistulas. J Neurol 247(7):521–523
hemorrhage, is considered thus as a relative contra- 7. Miyachi S, Izumi T, Matsubara N, Naito T,
Haraguchi K, Wakabayashi T (2011) Mechanism of
indication to radiosurgery; if needed, it should be the formation of dural arteriovenous fistula: the role
proposed as a complementary treatment for failures of the emissary vein. Interv Neuroradiol J
of surgical or endovascular treatments [24, 25]. Peritherapeutic Neuroradiol Surg Proced Relat
Neurosci 17(2):195–202
8. Borden JA, Wu JK, Shucart WA (1995) A proposed
classification for spinal and cranial dural arteriovenous
Summary fistulous malformations and implications for treatment.
J Neurosurg 82(2):166–179
The natural history of ICADS is based on the 9. Cognard C, Gobin YP, Pierot L, Bailly AL, Houdart E,
Casasco A et al (1995) Cerebral dural arteriovenous
venous drainage. ICADS are dynamic lesions, i. fistulas: clinical and angiographic correlation with a
e., a benign fistula can turn into a malignant revised classification of venous drainage. Radiology
ICADS with time, but may also regress spontane- 194(3):671–680
ously. The spontaneous morbi-mortality of the 10. Davies MA, Saleh J, Ter Brugge K, Willinsky R,
Wallace MC (1997) The natural history and manage-
disease dramatically increases in case of ment of intracranial dural arteriovenous fistulae.
leptomeningeal venous reflux and requires thus a Part 1: benign lesions. Interv Neuroradiol J
curative treatment. Peritherapeutic Neuroradiol Surg Proced Relat
CT and MR can provide useful informations Neurosci 3(4):295–302
11. Davies MA, Ter Brugge K, Willinsky R, Wallace MC
for diagnosis, but DSA is mandatory to perform (1997) The natural history and management of intra-
the analysis of the lesion and to understand the cranial dural arteriovenous fistulae. Part 2: aggressive
venous drainage of the shunt and of the brain. lesions. Interv Neuroradiol J Peritherapeutic
Endovascular approach is nowadays the first Neuroradiol Surg Proced Relat Neurosci 3(4):303–311
12. Geibprasert S, Pereira V, Krings T, Jiarakongmun P,
intention treatment, either transarterially or Toulgoat F, Pongpech S et al (2008) Dural arteriove-
transvenously. This should be proposed in case nous shunts: a new classification of craniospinal epi-
of malignant ICADS and benign ICADS with dural venous anatomical bases and clinical
intolerable symptoms. correlations. Stroke J Cereb Circ 39(10):2783–2794
13. Van Dijk JMC, terBrugge KG, Willinsky RA, Wallace
MC (2002) Clinical course of cranial dural arteriove-
nous fistulas with long-term persistent cortical venous
References reflux. Stroke J Cereb Circ 33(5):1233–1236
14. Söderman M, Pavic L, Edner G, Holmin S, Andersson
1. Berenstein A, Lasjaunias P, TerBrugge K (2004) Dural T (2008) Natural history of dural arteriovenous shunts.
arteriovenous shunts. Therapeutic aspects. In: Surgical Stroke J Cereb Circ 39(6):1735–1739
neuroangiography. Springer, Berlin, p 2.2:605 15. Duffau H, Lopes M, Janosevic V, Sichez JP,
2. Van Rooij WJ, Sluzewski M, Beute GN (2007) Dural Faillot T, Capelle L et al (1999) Early rebleeding
arteriovenous fistulas with cortical venous drainage: from intracranial dural arteriovenous fistulas: report
incidence, clinical presentation, and treatment. AJNR of 20 cases and review of the literature. J Neurosurg
Am J Neuroradiol 28(4):651–655 90(1):78–84
656 P. Guedin

16. Mossa-Basha M, Chen J, Gandhi D (2012) Imaging of management of intracranial dural arteriovenous shunts
cerebral arteriovenous malformations and dural arte- with leptomeningeal venous drainage: report of 53 con-
riovenous fistulas. Neurosurg Clin N Am 23(1):27–42 secutive patients with emphasis on transarterial embo-
17. Roy D, Raymond J (1997) The role of transvenous lization with acrylic glue. J Neurosurg 112(3):603–610
embolization in the treatment of intracranial dural arte- 22. Cognard C, Januel AC, Silva NA, Tall P (2008)
riovenous fistulas. Neurosurgery 40(6):1133–1141 Endovascular treatment of intracranial dural arteriove-
18. Luo C-B, Chang F-C, Wu H-M, Chung W-Y (2007) nous fistulas with cortical venous drainage: new man-
Transcranial embolization of a transverse-sigmoid agement using Onyx. AJNR Am J Neuroradiol
sinus dural arteriovenous fistula carried out through 29(2):235–241
a decompressive craniectomy. Acta Neurochir 23. Da Costa LB, Terbrugge K, Farb R, Wallace MC
(Wien) 149(2):197–200 (2007) Surgical disconnection of cortical venous
19. Mironov A (1998) Selective transvenous embolization reflux as a treatment for Borden type II dural arterio-
of dural fistulas without occlusion of the dural sinus. venous fistulae. Acta Neurochir (Wien)
AJNR Am J Neuroradiol 19(2):389–391 149(11):1103–1108
20. Nelson PK, Russell SM, Woo HH, Alastra AJG, 24. Koebbe CJ, Singhal D, Sheehan J, Flickinger JC,
Vidovich DV (2003) Use of a wedged microcatheter Horowitz M, Kondziolka D et al (2005) Radiosurgery
for curative transarterial embolization of complex for dural arteriovenous fistulas. Surg Neurol
intracranial dural arteriovenous fistulas: indications, 64(5):392–398 (discussion 398–399)
endovascular technique, and outcome in 21 patients. J 25. Söderman M, Edner G, Ericson K, Karlsson B, Rähn T,
Neurosurg 98(3):498–506 Ulfarsson E et al (2006) Gamma knife surgery for dural
21. Guedin P, Gaillard S, Boulin A, Condette-Auliac S, arteriovenous shunts: 25 years of experience.
Bourdain F, Guieu S et al (2010) Therapeutic J Neurosurg 104(6):867–875
Carotid Cavernous Fistula
30
Timothy R. Miller, Ravishankar Shivashankar, Gaurav Jindal,
and Dheeraj Gandhi

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 658 Carotid cavernous fistulas (CCF) are complex
lesions involving the abnormal shunting of
Anatomy of the Cavernous Sinus
and Cavernous Segment of the Internal Carotid
arterial blood into the cavernous sinus. There
Artery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 658 are two distinct types: one is a direct commu-
nication between the internal carotid artery and
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 659
the cavernous sinus, while the other is a dural
Classification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 660 shunt involving meningeal branches of the
Etiology and Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . 662 external and/or internal carotid arteries.
Clinical Presentations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 664 Resulting patient symptoms as well as poten-
tial complications is determined primarily by
Complications and High-Risk Features . . . . . . . . . . . 665
the degree of arteriovenous shunting as well as
Imaging Evaluation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 667 the direction of venous outflow from the cav-
Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 670 ernous sinus. Catheter angiography remains
Conservative Management . . . . . . . . . . . . . . . . . . . . . . . . . . 670 the gold standard imaging evaluation of
General Considerations of Endovascular CCFs, although both MR and CT angiography
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 671
Endovascular Treatment of Direct CCF . . . . . . . . . . . . . 673
can be used to screen patients in whom the
Endovascular Treatment of Indirect or diagnosis is suspected. Current treatment for
Dural CCF . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 676 these lesions consists primarily of conservative
Surgical Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 677 management and endovascular embolization.
Stereotactic Radiotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . 678
The latter can be performed via transarterial
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 679 and/or transvenous approaches, using various
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 680 materials such as detachable balloons, coils,
and liquid embolic agents. A multidisciplinary
approach, including neurointerventionalists,
neurosurgeons, and ophthalmologists, is often
required to achieve the best outcomes for
T.R. Miller (*) • R. Shivashankar • G. Jindal • D. Gandhi patients.
Department of Diagnostic Radiology, Neuroradiology,
University of Maryland Medical Center, Baltimore, MD, Keywords
USA
Carotid cavernous fistula • Endovascular
e-mail: Tmiller5@unm.edu; trmiller12@gmail.com;
Rshiva@umm.edu; Gjindal@umm.edu; embolization • Cortical venous reflux • Intra-
Dgandhi@umm.edu cranial hemorrhage
# Springer Science+Business Media New York 2016 657
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_7
658 T.R. Miller et al.

Introduction cavernous segment of the ICA and cranial nerves


III, IV, V1, V2, and VI [5–7]. Cranial nerves III,
A carotid cavernous fistula (CCF) is an abnormal IV, V1, and V2 run in the lateral wall of the sinus,
arteriovenous shunt involving the cavernous sinus while VI is located within the sinus itself. The
[1, 2]. The term actually applies to two distinct cavernous sinus normally receives venous blood
lesions, which vary in their etiology, potential from the ipsilateral orbit and brain, including the
clinical presentations, treatment, and outcomes. superior ophthalmic, inferior ophthalmic, superfi-
The first is a direct CCF, which consists of a tear cial middle cerebral, and deep middle cerebral
in the cavernous segment of the internal carotid veins, as well as the sphenoparietal sinus
artery (ICA) with direct shunting of arterial blood [4, 8]. Venous outflow from the cavernous sinus
into the sinus [2]. The second is an indirect CCF, is via superior and inferior petrosal sinuses to the
which represents a dural arteriovenous fistula transverse sinus and internal jugular vein respec-
involving the cavernous sinus wall, which is fed tively, as well as inferiorly via the pterygoid
by meningeal arterial branches that normally sup- venous plexus as well as emissary veins associ-
ply this region [1, 2]. Despite their differences, ated with the foramina ovale and rotundum
both lesions involve shunting of arterial blood into [4, 8]. Finally, the cavernous sinuses are
the normally low-pressure cavernous sinus, with interconnected via a venous network centered
resulting symptoms and complications deter- around the hypophysis, the so-called circular
mined primarily by the degree and route of venous sinus [2, 9]. Although the circular sinus generally
drainage from the diseased sinus [3, 4]. The fol- consists of a larger anterior and smaller posterior
lowing chapter explores the characteristics of both limbs, either may be absent or hypoplastic in a
types of CCF, highlighting their differences as particular patient.
well as similarities.
Contents of the Cavernous Sinus
Types of Carotid Cavernous Fistula • Cavernous segment of the internal
• Direct fistulas: A direct fistulous com- carotid artery
munication between the cavernous seg- • Cranial nerve VI
ment of the internal carotid artery and the • Cranial nerves III, IV, V1, and V2 in the
surrounding cavernous sinus lateral wall of the sinus
• Indirect fistulas: A dural arteriovenous
fistula involving meningeal branches of
the internal and external carotid arteries
that normally supply the region of the Normal Hemodynamics of the Cavernous
cavernous sinus Sinus
• Venous inflow
– Orbital veins: superior, inferior oph-
thalmic, and central retinal
– Superficial and deep middle cerebral
Anatomy of the Cavernous Sinus veins
and Cavernous Segment – Sphenoparietal sinus
of the Internal Carotid Artery • Venous outflow
– Superior petrosal sinus
Cavernous Sinus: The cavernous sinus is a con- Drains to transverse sinus
tiguous network of trabeculated venous sinusoids – Inferior petrosal sinus
surrounded by dura matter that is located centrally Drains to internal jugular vein
in the skull base adjacent to the pituitary sella and
sphenoid sinus [5–7]. It contains both the (continued)
30 Carotid Cavernous Fistula 659

most common branch to be involved by an indirect


– Pterygoid venous plexus
CCF [5, 6]. The ILT also has three main branches:
– Emissary veins around foramina
superior or tentorial, anterior, and posterior
ovale and rotundum
branches, which supply dura as well as cranial
• Intercavernous flow
nerves III, IV, V1, V2, and VI in and around the
– Circular sinus
cavernous sinus and orbit [5]. Finally, the cavern-
ous ICA may also give rise to McConnell’s capsu-
lar arteries, although these vessels are inconstantly
present [6].
Internal Carotid Artery: The ICA ascends ver-
tically in the skull base from the foramen lacerum
into the cavernous sinus, before turning anteriorly Branches of the Cavernous Internal Carotid
into a horizontal segment approximately 2 cm in Artery
length [6]. The artery then ascends superiorly in a • Meningohypophyseal trunk
second vertical segment, extending along the – Artery to the tentorium (Bernasconi-
medial aspect of the anterior clinoid process Cassonari)
before exiting the sinus. The cavernous segment – Dorsal meningeal artery
of the ICA can be divided into five segments, as – Inferior hypophyseal artery
detailed by Debrun et al. [10]. Going from caudal • Inferolateral trunk
to cranial, these include the posterior ascending – Superior or tentorial branch
segment, junction of posterior ascending segment – Anterior branch
and horizontal segment, horizontal segment, junc- – Posterior branch
tion of the horizontal segment and anterior • Capsular arteries
ascending segment, and ascending segment
[10]. Although any segment of the cavernous
ICA may be involved by a direct CCF, the hori-
zontal portion is the most common site of
fistulization [10]. Pathophysiology
The cavernous segment of the ICA has two
main branches that, along with distal branches of All CCFs demonstrate the same basic pathophys-
the internal maxillary artery from the external iologic mechanism of abnormal shunting of arte-
carotid, supply the dura surrounding the cavernous rial blood into the normally low-pressure venous
sinus as well as the nearby cranial nerves cavernous sinus. The resulting increase in pres-
[2, 5, 11]. Small anastomoses between these sure and volume results in reversal of blood flow
branches of the external and internal carotid arteries in venous structures that normally drain the cav-
are normally present in the surrounding dura and ernous sinus, including the superior and inferior
are responsible for the dural arteriovenous shunts ophthalmic veins as well as the superior and infe-
found in indirect CCF [1, 5, 11]. The first is the rior petrosal sinuses [4]. The resulting signs and
meningohypophyseal trunk, present in 100 % of symptoms associated with a CCF are determined
patients, which arises from the apex of the proximal both by the degree of arteriovenous shunting and
curve of the vessel in the cavernous sinus [6]. This the route of venous drainage from the cavernous
artery in turn has three main branches: the artery to sinus [3, 4]. The latter is determined by the loca-
the tentorium (Bernasconi-Cassonari), the dorsal tion of the arteriovenous shunt, the presence of
meningeal artery to the clivus, as well as the infe- thrombus in the involved sinus, as well as stenosis
rior hypophyseal artery [5]. The second branch of or thrombosis of potential venous drainage path-
the cavernous ICA is the inferolateral trunk (ILT), ways [10]. If the hemodynamic changes associ-
found in 84 % of patients, which originates from ated with a CCF persist over a long period of time,
the distal horizontal segment of the vessel and is the the involved veins may become arterialized with
660 T.R. Miller et al.

Fig. 1 Venous drainage pathways. (a) Frontal projection petrosal sinus (thick black arrows), right superior ophthal-
and (b) lateral projection of a right internal carotid artery mic vein (thin white arrows), right inferior ophthalmic vein
angiogram demonstrating a direct right-sided CCF with (thick white arrows), right superficial middle cerebral vein
bilateral venous drainage via the circular sinus. Multiple (thin blue arrows), right deep middle cerebral vein (thin
venous drainage pathways are illustrated, including the left red arrow), and pterygoid venous plexus (thick red arrow)
superior petrosal sinus (thin black arrows), left inferior

thickened, hyalinized walls [1]. Other chronic


changes include recruitment of capillary connec- Potential Routes of CCF Venous Drainage
tions in the conjunctiva of the eye, which are • Anterior drainage via the orbital veins
called specific limbal loops [1]. – Eventually draining into angular and
There are various patterns of venous drainage facial veins
from the cavernous sinus that can be encountered • Posterior drainage
with a CCF, which have important implications – Superior and inferior petrosal sinuses
for patient symptomatology as well as the risk of • Contralateral drainage
intracranial complications (Fig. 1) [4, 10, – Circular sinus
12]. Anterior drainage is commonly present, • Superior drainage
with reflux of blood into the ipsilateral orbit via – Superficial middle cerebral vein
the superior and inferior ophthalmic veins, with • Inferior drainage
subsequent drainage into angular and facial veins – Pterygoid venous plexus & emissary
[1, 12]. An additional anterior drainage route con- veins
sists of the sphenoparietal sinus running along the
anterior middle cranial fossa [2]. Posterior drain-
age from the cavernous sinus into the inferior and
superior petrosal sinuses is also often encoun-
tered, either alone or in combination with anterior Classification
as well as contralateral drainage via the circular
sinus [2, 4, 10, 12]. It has been speculated that the CCFs can be categorized using several criteria
majority, if not all, of low-flow indirect CCF orig- [5, 13]. These include classifications systems
inally drain posteriorly, with anterior drainage based on etiology (i.e., traumatic vs. spontaneous),
only commencing once the posterior pathway hemodynamics (high-flow vs. low-flow shunts), as
becomes obstructed, often due to thrombosis well as angiographic criteria [1, 5, 8, 13]. The latter
[12]. Finally, superior and inferior venous drain- was characterized by Barrow et al. [5] and is based
age from the cavernous sinus can proceed via the on both the hemodynamics of the shunt as well as
Sylvian vein and pterygoid venous plexus, the arterial supply to the fistula as demonstrated on
respectively [4]. angiography [5].
30 Carotid Cavernous Fistula 661

Fig. 2 Type A direct CCF.


(a) Frontal projection and
(b) lateral projection of a
left internal carotid artery
angiogram demonstrating a
high-flow direct shunt
between the left internal
carotid artery and the
ipsilateral cavernous sinus.
Venous drainage is
primarily via the circular
and contralateral cavernous
sinuses

Fig. 3 Indirect type B


CCF. (a) Frontal projection
and (b) lateral projection of
a left internal carotid artery
angiogram demonstrating
an indirect CCF supplied by
meningeal branches of the
ICA arising from the
meningohypophyseal trunk
(thin black arrow) and
inferolateral trunk (thick
black arrow)

Fig. 4 Indirect type C CCF.


(a) Frontal projection of a
right internal carotid artery
angiogram demonstrating no
evidence of arteriovenous
early cavernous shunting.
(b) Frontal projection right
external carotid artery
angiogram demonstrating
dural shunts between
external carotid meningeal
branches and the ipsilateral
cavernous sinus (white
arrows)

Type A CCFs are high-flow direct shunts branches and the cavernous sinus (Fig. 4). Finally,
between the ICA and the cavernous sinus type D CCFs are dural shunts with arterial supply
(Fig. 2). Type B CCFs are dural shunts between from meningeal branches of both the ICA and
meningeal branches of the cavernous ICA (arising ECA (Fig. 5) [5]. Rarely, a direct fistula between
from the meningohypophyseal or ILT trunks) and the ophthalmic artery and an ophthalmic vein can
the cavernous sinus (Fig. 3). Type C CCFs are present in a similar fashion to a CCF, a so-called
dural shunts between external carotid meningeal orbital shunt [1] (Fig. 6; Table 1).
662 T.R. Miller et al.

Fig. 5 Indirect type D


CCF. (a) Frontal projections
right internal carotid and (b)
right external carotid artery
angiograms demonstrating
an indirect CCF with
arterial supply from
meningeal branches arising
from both internal and
external carotid arteries
(white arrows)

Fig. 6 Cortical venous reflux. (a) Frontal projection right venous infarct (white arrows c, d). (e) Lateral right external
external carotid artery angiogram and (b) flat panel com- carotid artery angiogram and (f) axial FLAIR imaging
puted tomography demonstrates a type C indirect CCF following coil embolization demonstrates no residual arte-
with cortical venous reflux (black arrows a, white riovenous shunting (black arrows e) as well as resolution
arrows b). (c) Axial FLAIR and (d) susceptibility MR of ischemic changes in the brain stem (white arrows f)
imaging demonstrates a resulting hemorrhagic brainstem

most commonly found in young males, presum-


Etiology and Epidemiology ably due to the increased prevalence of head
trauma in this population [13–15]. Debrun
Direct Fistulas: Direct CCFs often result from et al. [10] found that the arterial tear in traumatic
blunt or penetrating trauma that results in a tear fistulas can vary in size anywhere from 1 to 5 mm,
in the cavernous segment of the ICA, with and rarely multiple rents may be present. Despite
resulting rapid arteriovenous shunting into the the high-flow arteriovenous shunting associated
cavernous sinus [8, 10, 14]. These lesions are with direct fistulas, patients often present in a
30 Carotid Cavernous Fistula 663

Table 1 Barrow angiographic classification of carotid


cavernous fistulas Etiologies of Direct Carotid Cavernous
Fistula Fistulas
type Arterial supply • Traumatic
Type A Direct Cavernous ICA – Blunt
Type B Indirect Meningeal ICA branches – Penetrating
Type C Indirect Meningeal ECA branches – Iatrogenic
Type D Indirect Meningeal ICA and ECA
• Spontaneous
branches
– ICA aneurysm rupture
– Collagen vascular disorders
Ehlers-Danlos type IV syndrome
somewhat delayed fashion, anywhere from days Pseudoxanthoma elasticum
to a few weeks following the inciting trauma Fibromuscular dysplasia
[16]. The mechanism of vessel injury in many of – Minor episode of trauma or valsalva
these cases is likely a sudden increase in Coughing and sneezing
intraluminal arterial pressure associated with the
traumatic event [17]. Penetrating trauma is
another possible cause, as well as rarely arterial
injury during skull base surgery [14–16, An additional cause of spontaneous direct
18]. Finally, traumatic direct CCFs have been CCFs includes various genetic syndromes that
associated with fractures involving the skull can weaken the arterial wall and predispose to
base, with Liang et al. [16] in a retrospective rupture after minor trauma or episodes of valsalva,
review reporting an 8.3 % incidence of direct including coughing or sneezing [1, 18, 19]. Spe-
fistulas in patients with fractures involving the cific genetic conditions that can affect vascular
middle cranial fossa, particularly those with a wall structural integrity and have been associated
transverse or oblique orientation. with CCF include fibromuscular dysplasia,
Direct CCF fistulas can also be encountered in Ehlers-Danlos type IV syndrome, and
patients without a history of preceding trauma pseudoxanthoma elasticum [23–25]. Finally, a
[8, 19]. These lesions are often encountered in direct CCF can be associated with a persistent
middle-age women, but are not exclusive to this trigeminal artery extending from the cavernous
age group [20]. These spontaneous direct fistulas ICA to the basilar artery [22]. These so-called
have several potential etiologies, the most com- trigemino-cavernous fistulas may arise either
mon of which is rupture of a cavernous segment from an inherent weakness in the aberrant vessel
ICA aneurysm [18, 19]. Cavernous ICA aneu- wall or due to aneurysm formation at the
rysms represent somewhere between 1.9 % and ICA-trigeminal artery branch point [22].
9 % of all intracranial aneurysms, are more com- Indirect or Dural Fistulas: Indirect CCFs rep-
monly associated with CCF development as resent a subset of dural arteriovenous fistulas
opposed to subarachnoid hemorrhage, and give involving the cavernous sinus, which receive
rise to roughly 20 % of direct CCFs [14, 19, 21]. blood flow from meningeal branches of the inter-
In a retrospective review of all cavernous nal and/or external carotid arteries that normally
ICA aneurysms presenting to a single medical supply the cavernous sinus dura [2, 26]. These
center, Kupersmith et al. [21] found that 13 of lesions most often occur spontaneously in post-
193 lesions were associated with a CCF (6.7 %). menopausal women [15, 26, 27]. However, as is
However, it is important to remember that a the case with direct CCFs, these lesions may pre-
predisposing cavernous aneurysm may not be sent in all age groups, including children and
identified following direct fistula formation due infants, and can develop following minor trauma
to the presence of high-flow arteriovenous or episodes of valsalva [12]. Altogether, dural
shunting [22]. arteriovenous fistulas represent approximately
664 T.R. Miller et al.

10–15 % of intracranial vascular malformations shunting results in orbito-ocular congestion and


[28]. Potential arterial feeders to indirect CCFs is responsible for the classic clinical triad asso-
include branches of the external carotid artery, ciated with CCFs, namely, pulsatile exophthal-
such as the internal maxillary, middle meningeal, mos, orbital bruit, and chemosis of the ipsilateral
accessory meningeal, and ascending pharyngeal globe [1, 3, 4, 14, 19]. The contralateral orbit
arteries, as well as internal carotid artery branches may also be affected due to reflux of blood across
[26]. The latter include meningohypophyseal, the circular sinus, occasionally in the absence of
capsular, inferolateral trunk arteries, as well as ipsilateral orbital symptoms depending on the
ethmoidal branches arising from the ophthalmic pattern of venous drainage [1, 9, 12]. Other
artery [26]. signs and symptoms of orbito-ocular congestion
As is the case with dural arteriovenous fistulas include medically refractory glaucoma,
found elsewhere in the intracranial compartment, ophthalmoplegia, retro-orbital pain, vision loss,
the etiology of indirect CCF remains uncertain as well as dilatation and arterialization of con-
[12, 15]. One theory speculates that these fistulas junctival and episcleral veins on ophthalmologic
may result from breakdown in small thin-walled exam [1, 12]. Although this latter finding may be
dural arteries that normally cross the cavernous seen in the setting of other diseases such as
sinus [12]. Alternatively, venous thrombosis conjunctivitis, a tortuous corkscrew appearance
and/or elevated venous pressure may result in the of these vessels is highly specific for the diagno-
opening of normally closed, small anastomotic sis of CCF [12]. General differential consider-
channels in the dura [8, 12]. Risk factors for the ations for physical exam findings suggestive of
development of these dural fistulas include athero- orbito-ocular congestion include vascular
sclerosis, hypertension, diabetes, sinusitis, preg- malformations involving the orbit or cavernous
nancy, and collagen vascular disease [8, 27, 29]. sinus, cavernous sinus thrombosis, as well as
inflammatory processes such as scleritis, with
vortex vein blockage [1].
Etiology of Indirect Carotid Cavernous
Some CCFs may present without classic symp-
Fistulas
toms, depending on the pattern of venous drainage
• Uncertain, two possible mechanisms
as well as the degree of arteriovenous shunting [3,
– Breakdown of small thin-walled dural
11, 15, 18, 28]. For example, low-flow indirect
arteries that transverse the cavernous
CCFs with exclusively posterior venous drainage
sinus
characteristically lack signs and symptoms of
– Opening of small anastomotic dural
orbito-ocular congestion, including pulsatile
channels
exophthalmos and orbital bruit [3, 4, 12, 29,
Venous sinus thrombosis
30]. Instead, these lesions often are either asymp-
Elevated venous pressure
tomatic or associated with nonspecific symptoms
• Risk factors
including headache, tinnitus, trigeminal neuropa-
– Atherosclerosis, diabetes, hyperten-
thy, facial nerve palsy, or isolated oculomotor palsy
sion, and pregnancy
due to the involvement of the corresponding cranial
nerves [11, 12, 30]. In these instances, patient
symptoms may be due, at least in part, to brainstem
congestion resulting from the posterior venous
drainage [12, 31]. The atypical presentation of
Clinical Presentations these posteriorly draining fistulas often leads to a
delay in their diagnosis, which is unfortunate
The majority of symptomatic CCFs have anterior given the association between drainage via the
drainage via the ophthalmic veins, with resulting superior petrosal sinus and cortical venous reflux,
reflux of high-pressure arterial blood into the a high-risk feature discussed subsequently in this
ipsilateral orbit [3, 12, 15]. This arteriovenous chapter [1, 4].
30 Carotid Cavernous Fistula 665

the choroid or iris, displacement of the iris-lens


Clinical Presentation of Carotid Cavernous diaphragm, or neovascularity secondary to
Fistulas chronic ischemia [12]. Finally, loss or deteriora-
• Fistulas with anterior drainage via orbital tion of vision in the involved eye can be secondary
veins to venous stasis retinopathy with resulting retinal
– Pulsatile exophthalmos ischemia, secondary glaucoma with optic nerve
– Chemosis damage, or spontaneous choroidal detachment [1,
– Glaucoma 11, 12]. Vision loss is more commonly seen with
– Vision loss direct CCF, although this finding may be encoun-
– Ophthalmoplegia tered in as many as 20–30 % of indirect fistulas,
• Fistulas with exclusively posterior drain- particularly chronic lesions [12].
age via petrosal sinuses
– Asymptomatic
– Headache Ophthalmologic Complications of Carotid
– Tinnitus Cavernous Fistulas
– Trigeminal neuropathy • Glaucoma
– Isolated oculomotor palsy – Elevated orbital venous pressure
– Choroid or iris congestion
– Iris-lens diaphragm displacement
– Neovascularity
Finally, symptom progression in CCFs is often • Ophthalmoplegia
determined by the degree of associated arteriove- – Extraocular muscle swelling
nous shunting, with high-flow lesions often – Cranial nerve palsy
presenting acutely with rapid deterioration, while • Vision loss
low-flow fistulas may demonstrate an insidious – Venous stasis retinopathy
onset and subsequent benign course – Glaucoma with optic nerve damage
[1, 15]. Low-flow shunts are most often encountered – Spontaneous choroidal detachment
in the setting of an indirect, dural CCF, although
direct lesions may also behave in a similar manner
if the tear in the cavernous ICA is relatively small or
if there is partial thrombosis of the involved cavern- Additional complications of CCFs include
ous sinus [1]. Recurrent fistulas following treatment intracranial hemorrhage, both intraparenchymal
may also present insidiously, again depending on the and subarachnoid, venous infarct, epistaxis
amount of residual arteriovenous shunting and avail- (which may be fatal), as well as increased intracra-
able venous drainage pathways [22]. nial pressure with venous hypertension [18, 26,
32]. Intraparenchymal hemorrhage is often a result
of reflux of high-pressure arterialized blood into
Complications and High-Risk Features cortical veins overlying adjacent brain parenchyma
[1, 15, 18]. Posterior venous drainage of a CCF via
Ophthalmologic complications of CCFs include the superior petrosal sinus has been associated with
vision loss, ophthalmoplegia, and medically the development of this high-risk feature and may
refractory glaucoma [1, 12]. Ophthalmoplegia be precipitated by the spontaneous thrombosis or
can result from either entrapment of the iatrogenic occlusion of alternative venous drainage
extraocular muscles due to swelling of these struc- pathways [1, 4, 15, 18]. Overall, cortical venous
tures or due to cranial nerve palsy secondary to reflux has been reported in anywhere from 10 % to
mechanical compression of the corresponding 55 % of CCFs, is often symptomatic due to
nerve(s) [1, 12]. Glaucoma in CCFs may develop increased intracranial pressure, and carries up to a
from high orbital venous pressure, congestion of 30–40 % chance of hemorrhagic stroke if untreated
666 T.R. Miller et al.

[15, 18, 26, 33]. Furthermore, the reported annual


mortality rate of intracranial dural arteriovenous High-Risk Features of Carotid Cavernous
fistulas with cortical venous reflux, regardless of Fistulas
location, is 10.4 % [33]. • Cortical venous reflux
Subarachnoid hemorrhage from a CCF has been – Between 10 % and 55 % of CCFs
associated with the development of a cavernous – Up to 30–40 % risk of hemorrhagic
sinus varix or pseudoaneurysm (Fig. 7) [18]. In a stroke untreated
retrospective review of the angiographic features of – Associated with superior petrosal
155 patients with CCFs, Halbach et al. [18] found sinus drainage
that cavernous sinus venous varix was present in – Can arise from blockage of venous
three of four patients who presented with subarach- drainage pathways
noid hemorrhage, all of which were fatal. Although • Subarachnoid hemorrhage
cavernous sinus varices were encountered in – Rare, but often fatal
patients without intracranial hemorrhage, the – Associated with cavernous sinus
authors argued the risk of fatal subarachnoid hem- varix
orrhage warranted emergent fistula treatment with • Drainage into sigmoid and transverse
this finding is present [18]. Finally, drainage into sinuses
the sigmoid and transverse sinuses has been asso- – Risk of elevated intracranial pressure
ciated with elevation of intracranial pressure from from resulting venous hypertension
resulting venous hypertension [18].

Fig. 7 Cavernous sinus


pseudoaneurysm. (a) Axial
and (b) coronal
reconstructed CT
angiography images show
an ectatic left cavernous
sinus (white arrow a) with
multiple lateral projecting
outpouchings (white arrows
b), which may represent
possible pseudoaneurysm
and/or draining venous
tributaries. (c) Frontal
projection and (d) lateral
projection of a left internal
carotid artery angiogram
demonstrate a direct CCF
with associated
pseudoaneurysm (black
arrow c)
30 Carotid Cavernous Fistula 667

Imaging Evaluation insensitive for lesions demonstrating less rapid


arteriovenous shunting [40].
Noninvasive Cross-Sectional Imaging: Although 3D TOF MRA can increase the sensitivity for
many patients with a CCF present with classic detection of CCF by depicting flow-related
signs and symptoms suggestive of the diagnosis, enhancement in the involved cavernous sinus as
others may demonstrate either atypical symptom- well as arterial feeders in the setting of an indirect
atology and/or an insidious, slowly progressive fistula [35, 37, 40]. It is important to note however
course [26]. In these instances, conventional that venous flow signal can be normally seen in
MR, time-of-flight (TOF) and contrast-enhanced the cavernous and inferior petrosal sinuses on 3D
MR angiography (MRA), Doppler ultrasound, as TOF MRA in the absence of a CCF [41]. In addi-
well as CT angiography (CTA), may be used to tion, 3D TOF MRA is limited by the lack of
screen patients for the presence of an arteriove- temporal resolution, which precludes full charac-
nous fistula involving the cavernous sinus terization of dynamic shunting lesions such as a
[34–36]. Conventional MR imaging finding sug- CCF [40]. However, time-resolved contrast-
gestive of a CCF includes prominent flow voids in enhanced MRA can help to overcome some of
the involved cavernous or inferior petrosal sinuses these challenges by imaging the suspected dis-
on spin-echo sequences, dilated intercavernous eased cavernous sinus during passage of a gado-
venous channels, as well as the sequelae of linium contrast bolus [40]. This technique has
orbito-ocular congestion including enlargement been shown to be both sensitive and specific for
of the superior ophthalmic vein and extraocular the diagnosis of CCF and is a promising tool both
muscles and proptosis [37–39]. However, these for screening patients as well as surveillance for
findings may only be present in high-flow fistulas, fistula recurrence following treatment
with conventional MR imaging being relatively (Fig. 8) [40].

Fig. 8 Conventional MR and MRA finding suggestive of (horizontal white line). (d) Post-contrast MRA demon-
a right CCF. (a, b) Prominent flow voids are noted on spin- strates enhancement of the involved cavernous sinus
echo sequences in the right cavernous and inferior petrosal (white arrows). (e, f) Time-resolved contrast-enhanced
sinuses (white thick and thin arrows respectively a) as well MRA demonstrates early opacification of right cavernous
as dilated intercavernous venous channels (thin black sinus (thin white arrow e) as well as the fistulous shunt and
arrows b). (c) Sequelae of orbito-ocular congestion are drainage via the superior ophthalmic vein (thick and thin
present in the ipsilateral orbit with enlargement of the white arrows, respectively f)
extraocular muscles (white thin arrows) and proptosis
668 T.R. Miller et al.

CT angiography has also been demonstrated to angiography remains the gold standard for the
be a promising technique for screening patients diagnosis and characterization of these lesions
suspected of having a CCF [42]. With the avail- due to its superior spatial and temporal resolution
ability of 256 and 320 slice CT scanners, excellent [26, 40]. The diagnosis of a CCF is readily made
quality dynamic studies can be performed with on catheter angiography by the demonstration of
high spatial and acceptable temporal resolution. abnormal arteriovenous shunting into the cavern-
CT and CT angiography findings of a CCF include ous sinus from either the ipsilateral cavernous
an enlarged cavernous sinus that demonstrates ICA or meningeal branches to the sinus wall in
early enhancement in the arterial phase, as well the case of an indirect fistula. Goals of catheter
as dilatation of draining venous tributaries (e.g., angiography when evaluating a CCF include the
superior ophthalmic vein) [42, 43]. Chen identification of the exact site of fistulization,
et al. [37] performed a retrospective study com- evaluation of the degree of arteriovenous
paring noninvasive 3D TOF MRA and CTA to the shunting, determination of arterial supply to the
gold standard of catheter angiography. They lesion, evaluation of the pattern of venous out-
found that CTA performed better at detecting flow, the presence of high-risk features including
CCF than TOF MRA, particularly for fistula cortical venous reflux and venous varix, potential
involving the more proximal aspect of the cavern- dangerous external or internal carotid artery anas-
ous ICA (Table 2). tomoses, as well as the presence of atherosclerotic
Catheter Angiography: Despite advances in disease if carotid compression is contemplated as
cross-sectional imaging of CCF, catheter a potential treatment [26].

Table 2 Noninvasive imaging modalities for evaluation Catheter Angiography Evaluation of Carotid
of CCFs Cavernous Fistulas
Potential • Determination of type of fistula
Modality Findings disadvantages • Arterial supply with indirect lesions
MRI Flow voids, Insensitive for • Localization of fistulous tear with direct
cavernous and moderate- to
petrosal sinuses low-flow fistulas lesions
Distension of the • Evaluation of degree of arteriovenous
cavernous sinus shunting
Dilated • Analysis of venous drainage pathways
intercavernous • Identification of high-risk features,
channels
including cortical venous reflux
Sequelae of orbito-
ocular congestion
TOF Flow-related Not 100 % specific
MRA enhancement in No temporal Huber’s Maneuver: In the setting of a high-
cavernous sinus resolution flow CCF, the tremendous arteriovenous shunting
CE Flow-related Improved temporal may obscure the underlying ICA tear, and the
MRA enhancement in resolution carotid artery more distally may not fill due to
cavernous sinus May not detect essentially complete diversion of blood flow into
high-risk features
the cavernous sinus [2, 10, 22]. In these instances,
CTA Distension of the Poor temporal
cavernous sinus resolution
injection of a vertebral artery during manual com-
Early sinus May not detect pression of the ipsilateral ICA may allow for
enhancement in high-risk features better characterization of both the location and
arterial phase the size of the fistulous communication (Fig. 9)
Dilatation of [10, 44]. This technique works by allowing a
draining venous
limited amount of contrast to reach the shunt via
tributaries
a posterior communicating artery with subsequent
30 Carotid Cavernous Fistula 669

Fig. 9 Huber maneuver.


(a) Frontal projection left
internal carotid artery
angiogram demonstrates a
direct CCF with the high-
flow arteriovenous shunting
obscuring the underlying
tear in the carotid artery. (b)
Injection of the contralateral
ICA or (c) vertebral artery
during manual or balloon
compression of the
ipsilateral ICA (Huber’s
maneuver) may allow for
better characterization of
both the location and the
size of the fistulous
communication. (d) C-arm
angiographic CT (flat panel
rotational computed
tomography) is also a
exceptional technique for
locating and studying the
anatomy of fistula as well

Fig. 10 Angiographic balloon occlusion test. (a) Frontal no significant filling of the left middle cerebral artery. (c)
projection left internal carotid artery angiogram demon- Frontal projection right internal carotid artery angiogram
strates high-flow arteriovenous shunting associated with a during balloon occlusion of the cervical left internal carotid
left direct CCF. There is relatively poor opacification of the artery (black arrow) demonstrates brisk collateral flow to
more distal left internal carotid artery due to vascular steal. the left middle cerebral artery territory (white arrows)
(b) Frontal projection right internal carotid artery angio-
gram opacifies the left anterior cerebral artery, but there is

retrograde flow down the ipsilateral supraclinoid Balloon Occlusion Test (BOT): As a significant
ICA [10]. Other maneuvers that can help delineate minority of direct CCFs may require ipsilateral
the fistula size and location include very high ICA sacrifice for successful closure, performing
frame rate imaging and 3D subtraction angiogra- a balloon occlusion test during the diagnostic
phy. Finally, C-arm angiographic CT (flat panel workup can provide invaluable information to
rotational computed tomography) is another the treating physician (Fig. 10). First, a guide
exceptional technique for locating and studying catheter is placed in the ipsilateral common
the anatomy of fistula. carotid artery. Next, following full heparinization,
670 T.R. Miller et al.

a soft, compliant balloon (typically a HyperForm disability on follow-up compared to those who
or HyperGlide Occlusion Balloon, ev3, Irvine, did (41.5 days compared to 140.8 days). Finally,
California) is navigated into the cervical ICA although complete fistula cure is often attempted,
proximal to the fistula. The balloon is then care- partial occlusion/embolization may be adequate if
fully inflated under fluoroscopic imaging, and a it addresses either intolerable symptomatology or
gentle injection is performed through the guide high-risk features such as cortical venous reflux.
catheter to confirm vessel occlusion. Continuous The following sections further explore the var-
neurologic monitoring, including testing of ious management options available to patients
patient speech and contralateral strength, is then with CCFs.
performed for a total of 30 min. The test is imme-
diately stopped if the patient develops symptoms
suggestive of ischemia involving the ipsilateral Conservative Management
cerebral hemisphere. The patient can be further
challenged during a balloon occlusion test by When discussing possible treatment of a CCF
purposefully dropping the systolic blood pressure with a patient, the potential risks and benefits of
by 30 %, although all operators do not routinely therapy should be weighed against the natural
perform this maneuver. Finally, collateral flow to history of the patient’s particular lesion [18, 26].
the affected vascular territory via the Circle of Spontaneous closure of indirect CCF has been
Willis (i.e., an angiographic balloon occlusion reported anywhere between 10 % and 73 % of
test) can be performed during balloon inflation cases, although spontaneous closure of direct fis-
by injection of the contralateral carotid and verte- tulas is thought to be considerably more rare
bral arteries using a second diagnostic catheter. [9, 14, 18, 19, 26]. Interestingly, spontaneous
fistula closure may be precipitated by catheter
angiography [18, 19, 26]. Accordingly, low-flow
Management fistulas without significant orbito-ocular conges-
tion, high-risk angiographic features, or other
The management of CCFs is often determined by intolerable symptoms can often be followed con-
multiple interdependent factors, including the rate servatively [1, 9, 15]. However, close clinical as
of arteriovenous shunting, the venous pathways well as periodic imaging follow-up is required
recruited by the fistula, the degree of associated though to exclude the development of aggressive
orbito-ocular congestion, as well as the presence fistula features [1]. Stiebel-Kalish et al. [9] found
of cortical venous reflux or venous varix in a retrospective review that clinical signs sug-
[15, 45]. Treatment options include conservative gestive of the presence of cortical venous reflux in
management with close imaging follow-up as indirect CCF include bilateral orbital symptoms as
well as endovascular or surgical repair. Indica- well as the presence of a postauricular bruit.
tions for aggressive fistula treatment include pro- Patients with these features should undergo cath-
gressive vision loss, cranial nerve palsies, rapidly eter angiography for further evaluation.
worsening proptosis with corneal exposure, corti- During this time, patients may help to promote
cal venous reflux, and intractable retro-orbital fistula closure by manually compressing the ipsi-
pain [14, 19]. It is important to note that these lateral cervical carotid artery and internal jugular
signs and symptoms may be reversible only when vein several times a day (Fig. 11)
treatment is initiated early in the course of the [47, 48]. Higashida et al. [48] found that this
disease, emphasizing the importance of prompt resulted in cure of indirect CCF in 30 % of
diagnosis and prompt treatment [1, 27, 46]. Liang patients anywhere from several minutes to
et al. [16] found in their retrospective review of 6 months. This maneuver can even result in lesion
post-traumatic direct CCFs that the time from first closure in a minority of direct CCF (17 %). Kai
symptom onset to treatment was significantly et al. [49] found that factors associated with a
shorter in patients who had no fistula-related higher likelihood of achieving fistula closure
30 Carotid Cavernous Fistula 671

Fig. 11 Manual compression for treatment of a low-flow arrows). Patient was managed conservatively and asked
indirect type D CCF. (a) Frontal and (b) lateral projections to perform intermittent manual carotid artery compres-
of a right internal carotid and as well as frontal (c, d) lateral sions. Follow-up right internal (e, f) and external carotid
projections of a right external carotid artery angiogram (g, h) artery angiogram performed 6 months later demon-
demonstrating arterial supply to an indirect CCF from strates resolution of the fistula
external and internal carotid meningeal branches (black

with manual compression for indirect lesions General Considerations


included a shorter time between symptom onset of Endovascular Treatment
and initiation of treatment, lower ocular pressure,
and venous drainage exclusively via the superior A large majority of both direct and indirect CCFs
ophthalmic vein. Patients should be instructed to can be successfully treated using modern
be sitting during the maneuver and to use the endovascular techniques with a low rate of
contralateral arm [49]. Otherwise, transient lack procedure-related complications [11, 50]. Emboli-
of blood flow to the ipsilateral cerebral hemi- zation materials currently used for the treatment of
sphere may lead to syncope as well as inadvertent CCFs include fiber and platinum microcoils,
prolonged compression if the ipsilateral arm is detachable latex balloons, as well as liquid
used (which would not be affected by the resulting embolic agents including n-butyl cyanoacrylate
ischemia). (nBCA) or ethylene vinyl alcohol (Onyx, ev3,
Irvine, California) [50–52]. Transarterial,
Carotid Manual Compression of Carotid transvenous, as well as combined approaches
Cavernous Fistulas may be utilized for fistula closure (Fig. 12) [11,
• Appropriate for low-flow lesions without 50, 53]. The endovascular method used to treat a
high-risk features or progressive given lesion often is determined by multiple fac-
symptoms. tors, including the type of shunt (direct
• Patient should be sitting in case of vs. indirect), the size of the ICA tear with direct
syncope. fistulas, the rate of arteriovenous shunting, as well
• Patients should perform maneuver sev- as the accessibility of venous pathways to the
eral times a day. involved cavernous sinus [15].
• Patient should be instructed to use con- General risks of endovascular treatment of
tralateral hand in case of ipsilateral cere- CCFs include stoke, hemorrhage, vessel injury,
bral ischemia. and parent artery occlusion. In addition, patients
672 T.R. Miller et al.

Fig. 12 Endovascular access routes. (a) Transarterial superior ophthalmic vein (black arrows). (d) Transvenous
microcatheter injection of a distal external carotid artery access of a CCF through the inferior petrosal sinus (black
branch supplying an indirect CCF. Black arrows demon- arrows) (e) Transvenous access of a CCF with a
strate the distal aspect of the microcatheter. (b) microcatheter extending through the intercavernous chan-
Transarterial access of a direct left CCF with a nels (white arrows). (f) Direct cavernous sinus access
microcatheter passing from the ipsilateral internal carotid through percutaneous of the inferior orbital fissure (black
artery (white arrows) and into the cavernous sinus (black arrows)
arrows). (c) Transvenous access of a CCF through the

should be counseled that their symptoms may patients should be advised that some symptoms,
temporarily, or rarely permanently, worsen fol- particularly cranial nerve palsies, may not
lowing therapy as the fistula thromboses, with improve despite successful fistula closure [46].
alterations of venous drainage [15]. Obtaining a
complete diagnostic catheter angiogram prior to
Endovascular Treatment of Carotid
embolization is essential to define important fis-
Cavernous Fistulas
tula characteristics such as arterial supply and
• Materials
venous drainage, as well as to evaluate for high-
– Detachable Balloons
risk features including cortical venous reflux
– Platinum or fiber coils
[45]. In addition, the interventionalist must pay
– Liquid embolic agents
close attention to alterations of venous drainage
n-Butyl cyanoacrylate (nBCA, glue)
from the cavernous sinus during embolization of a
Ethylene vinyl alcohol copolymer
CCF. Inadvertent diversion of blood into the
(Onyx)
sphenoparietal or superior petrosal sinuses after
– Covered Stents
partial fistula embolization may lead to cortical
• Endovascular Access
venous reflux and hemorrhagic stroke [15, 26, 45,
– Ipsilateral carotid artery for direct
54, 55]. Similarly, redirection of venous drainage
fistula
anteriorly into the ipsilateral orbit can lead to
– Venous routes
worsening of orbito-ocular congestion, with pos-
sible resulting loss of vision [28, 55]. Finally, (continued)
30 Carotid Cavernous Fistula 673

compression of the ICA and cranial nerves


Inferior petrosal sinus
[14, 56, 60]. In addition, some patients following
Facial and angular veins
detachable balloon treatment have developed
Pterygoid venous plexus
recanalization of a true cavernous ICA aneurysm,
Superior petrosal sinus
which was presumably responsible for develop-
Percutaneous puncture of superior
ment of the fistula [60]. Transarterial balloon
ophthalmic vein
embolization of direct CCFs has been abandoned
– Transarterial catheterization of men-
in the USA due to the lack of commercially avail-
ingeal feeders of indirect fistulas
able detachable balloons.
Coils and Liquid Embolic Agents: An alterna-
tive endovascular treatment of direct CCFs is the
placement of coils into the involved cavernous
Endovascular Treatment of Direct CCF sinus via a transarterial approach [11, 19, 55].
Once the microcatheter is in position in the
Detachable Balloons: The goal of endovascular sinus, multiple fiber or platinum microcoils can
treatment of direct CCF is complete fistula closure be carefully deployed in the venous
with preservation of the ICA [14, 56]. Early sinusoids closest to the point of arteriovenous
endovascular approaches to direct CCF consisted fistulization [55]. Similar, to detachable balloons,
of transarterially navigating a catheter with a challenges to coil embolization include difficul-
detachable balloon mounted at its tip past the tear ties navigating the microcatheter into the cavern-
in the cavernous ICA and into the cavernous sinus. ous sinus if the ICA tear is small, as well as
Once in position, one or more of these balloons inadvertent coil prolapse into the ICA with larger
were sequentially inflated and detached in the cav- rents [52, 55]. The latter may be particularly dif-
ernous sinus to achieve fistula closure [12, 22, ficult to visualize in the setting of a high-flow
33, 53]. Detachable balloons used for CCF treat- shunt and can result in parent vessel occlusion
ment are made of latex and inflated with contrast and/or stroke. The risk of coil prolapse into the
medium in position [19]. Serbinenko [57] was the ICA may be mitigated by placement of an
first to use such an approach for the treatment of endovascular stent and/or non-detachable balloon
CCFs, followed by Debrun et al. [58]. This method in the vessel prior to embolization [13, 52]. Finally,
is very successful at achieving fistula closure, with it is possible to occlude a large majority of the
high rates of ICA preservation and low procedure- diseased cavernous sinus with coils and still have
related morbidity and mortality [10, 11, 14, 59]. residual arteriovenous shunting if the portion of
Lewis et al. [14] demonstrated an 88 % rate of the sinus closest to the fistula remains patent [50].
complete occlusion of direct CCF via the Embolization of a direct CCF may be
transarterial balloon embolization method, with a supplemented or replaced by liquid embolic
75 % rate of ICA preservation. The permanent agents such as nBCA or ethylene vinyl alcohol
neurologic complication rate was only 4 %. (Onyx) [56, 61]. The non-adhesive nature of
Challenges to transarterial balloon emboliza- Onyx offers several advantages over nBCA,
tion of direct CCFs include difficulties navigating including the reduced risk of microcatheter reten-
the relatively stiff balloon catheters, inability to tion, which allows for longer injection times and
cannulate small ICA tears, inability to inflate a more thorough penetration of the liquid embolic
balloon in small-sized venous compartments agent into the lesion [50]. Potential risks of liquid
with the cavernous sinus, balloon compression embolic embolization of direct CCF include non-
and compromise of the cavernous ICA or cranial target embolization of the ipsilateral ICA and its
nerves, as well as subsequent balloon deflation or branches as well as cranial neuropathy. The risk of
migration [10, 14, 55, 56, 59, 60]. Balloon defla- reflux of liquid embolic agent may be mitigated by
tion or migration could result in fistula recurrence, inflation of a balloon into the ipsilateral cavernous
the development of an ICA pseudoaneurysm, or ICA during liquid embolic injection [62].
674 T.R. Miller et al.

Fig. 13 Carotid artery


sacrifice. (a) Frontal and (b)
lateral projections of a right
internal carotid artery
angiogram demonstrate a
post-traumatic right internal
carotid artery
pseudoaneurysm (white
arrows) and associated
direct CCF. (C) Frontal
projection of a right internal
carotid artery angiogram
following vessel sacrifice
by coil embolization
demonstrates no residual
filling of the fistula. Coil
mass is outlined by black
arrows. (d) Frontal
projection of a left internal
carotid artery angiogram
following right internal
carotid artery sacrifice
demonstrates cross-filling
of the contralateral right
anterior and middle cerebral
arteries

Alternative Routes to the Cavernous Sinus: If a approach via direct surgical cannulation/percuta-
transarterial approach is either not technically neous puncture of the ipsilateral superior ophthal-
possible or fails to adequately close a direct mic vein [27, 28]. This method has been shown to
CCF, transvenous treatment may be attempted be both safe and effective, with cannulation of the
[54]. A transvenous approach can be performed vein possible even if it appears thrombosed on
by placement of a guide catheter in the ipsilateral imaging [27]. Once microcatheter access to the
internal jugular vein followed by navigation of a cavernous sinus has been achieved, embolization
microcatheter into the cavernous sinus via the can then proceed with coils and/or liquid embolic
inferior petrosal sinus [45, 53, 54]. However, it agents [61]. Potential complications of direct
is not always possible to access the cavernous puncture of the superior ophthalmic vein include
sinus via this route due to either thrombosis or rupture of the vessel with retroocular hemorrhage
stenosis of the inferior petrosal sinus [15, 53, and rapid visual loss, infection, and damage to
63]. In these instances, alternative routes to the other orbital structures [28]. Finally, there are a
cavernous sinus include the ipsilateral facial and few report of direct percutaneous cannulation of
angular veins, the contralateral pterygoid venous the inferior ophthalmic vein performed for fistula
plexus, as well as the superior petrosal sinus via treatment [64].
the transverse sinus [15, 29, 53, 63]. Internal Carotid Artery Sacrifice: If other
On occasion, it may only be possible to reach endovascular methods of closing a direct CCF
the involved cavernous sinus from a transvenous fail, ICA sacrifice should be considered (Fig. 13)
30 Carotid Cavernous Fistula 675

Fig. 14 Covered stent treatment of a direct CCF. (a) pterygoid venous plexus (arrow head). (b) Lateral projec-
Lateral projection internal carotid artery angiogram dem- tion fluoroscopic native image demonstrates a covered
onstrates a high-flow direct CCF (black arrow) with exten- stent (black arrows) placed in the cavernous internal
sive shunting into the superior ophthalmic (white arrow) carotid artery spanning the defect
and inferior ophthalmic veins (red arrow) as well as the

[56]. The latter technique has proven to be highly polytetrafluoroethylene (PTFE)-covered stents,
successful in the treatment of direct CCFs, all of which had successful fistula closure and no
although there is a risk of ipsilateral cerebral immediate procedure-related morbidity or mortal-
hemispheric stroke. This risk may be mitigated, ity. However, one patient subsequently developed
in part, by performing a balloon occlusion test asymptomatic in-stent occlusion [51]. Tiewei
prior to artery closure. However, a successful et al. [65] reported similarly good results in a
balloon occlusion test does not guarantee a good group of eight patients with traumatic direct
outcome. High-flow direct CCFs that demonstrate CCF treated with covered coronary stents. Five
complete diversion of blood into the lesion with of the eight patients had occlusion of their fistula
non-opacification of the supraclinoid ICA repre- following stent placement, while six went on to
sent a special circumstance when carotid artery have resolution of their symptoms. However,
sacrifice should be considered [60]. If the patient once again, there was one case of subsequent
has not suffered from stroke or TIA-like symp- asymptomatic in-stent occlusion [65]. Another
toms, the hemodynamics of the fistula provide group, Wang et al. [66], reported successful clo-
strong evidence that carotid occlusion will be sure of 8 out of 10 direct CCF that failed more
tolerated. On the other hand, as many of these conventional endovascular therapy, with no
patients are relatively young, carotid artery sacri- in-stent stenosis or occlusion during the follow-
fice must be weighed against the risk of the patient up period of the study. These results suggest that
developing stenosis or occlusion of another major covered stents are a promising new endovascular
artery in the head and neck later in life. treatment of direct CCFs. However, more data is
Covered Stents: Covered stents have also been needed, particularly in regard to the long-term
used to treat direct CCFs, either alone or in com- patency of these devices. Finally, as is the case
bination with coils and liquid embolic agents with other endovascular stents, patients must be
(Fig. 14) [51, 65, 66]. Gomez et al. [51] treated maintained on dual antiplatelet therapy for at least
seven patients with post-traumatic CCFs with 3 months following treatment [51]. This is a
676 T.R. Miller et al.

Table 3 Endovascular treatment options for direct carotid significant drawback to treatment with covered
cavernous fistulas stents, particularly in patients who recently expe-
Treatment Potential Potential rienced significant head trauma (Table 3).
modality advantages disadvantages
Detachable Proven, safe Difficulty navigating
balloons and effective stiff delivery catheter
method to fistula Endovascular Treatment of Indirect or
Extensive Inability to inflate Dural CCF
operator balloon in small
experience venous Transvenous embolization of indirect, dural CCF
compartments is a safe and efficacious treatment for patients who
Potential for balloon
have progressive symptoms and high-risks fea-
deflation or
migration following tures on imaging or who fail conservative man-
deployment agement (Fig. 15) [15, 28, 45, 50, 63, 67]. Similar
Coils More controlled Potential for coil to all intracranial dural arteriovenous fistulas, the
deployment prolapse into goal of therapy is closure of the arteriovenous
compared to cavernous ICA,
liquid embolic especially in setting
shunts connecting meningeal arteries to feeding
agents of high-flow shunts veins [45]. For indirect CCF, this has been most
or large rents commonly accomplished by placement of multi-
Small, flexible Potential for residual ple coils in the diseased cavernous sinus [45,
microcatheters fistula following coil 50]. Kirsch et al. [46] reported either complete
may be used for embolization of
coil placement majority of lesion closure or minor residual shunt without
cavernous sinus cortical or ocular drainage in 94 % of 141 patients
Can be expensive if treated using this approach. Meyers et al. [26]
using platinum coils reported similar results with 90 % of patients
Liquid Excellent Less controlled cured following transvenous embolization of indi-
embolic penetration of embolization with
rect CCF in a retrospective review of 135 patients.
agents small venous potential for
compartments nontarget closure of However, transvenous coil embolization of
of the arterial supply to the indirect CCF may not always be successful due
cavernous sinus brain, eye, cranial to the trabeculated structure of the cavernous
nerves
sinus, which can preclude adequate coil place-
Carotid Effective Potential for
artery method ipsilateral ischemic
ment in the venous compartment(s) involved by
sacrifice Relatively safe stroke, even if patient the shunt [45, 50]. In these instances, emboliza-
if fistula results passes BOT tion may be successfully performed using liquid
in complete embolic agents, either alone or in combination
arterial steal or with coils (Fig. 16) [45, 67, 68]. Furthermore, if
patient passes
BOT a transvenous approach to an indirect CCF is not
Covered Shown to be Concern for long- possible due to either venous stenosis or occlu-
stents effective in term patency of sion, a transarterial approach with a liquid
small series stents embolic agent may also be utilized [50]. Onyx,
Option if more Need for antiplatelet with its ability to penetrate small arterial feeders
traditional therapy
endovascular
during prolonged injections, is uniquely well
methods fail suited to reach the small dural shunts from an
arterial pedicle [50]. Care must be taken however
30 Carotid Cavernous Fistula 677

Fig. 15 Coil embolization of an indirect CCF. Frontal Unsubtracted right internal carotid artery angiogram dem-
projections of a (a) right internal carotid artery, (b) left onstrating coil mass in the bilateral cavernous and circular
internal carotid artery, and (c) left external carotid artery sinuses (black arrows). Follow-up frontal projections of (e)
angiograms demonstrating meningeal supply (black right internal carotid artery and (f) left external carotid
arrows) to a type D indirect CCF. The patient subsequently artery angiograms demonstrate no residual arteriovenous
underwent transvenous coil embolization of the fistula. (d) shunting (white arrows)

as retrograde filling of non-catheterized arterial Surgical Treatment


pedicles is possible with Onyx, which may lead
to nontarget embolization of the arterial supply to Prior to the advent of modern endovascular tech-
the brain, eye, or other cranial nerves. Finally, niques, surgical closure of CCFs was the standard
other treatment options for indirect CCF of care [11]. Early surgical treatment for these
include transarterial embolization using a particu- lesions consisted primarily of carotid artery sacri-
late agent such as polyvinyl alcohol (PVA) fice by vessel trapping [10, 11, 70]. Parkinson [71]
(Fig. 17) [69]. A major limitation of this subsequently developed a direct surgical
latter approach however is a high rate of fistula approach to the cavernous sinus via a triangular
recurrence, which is less likely following emboli- space in the lateral sinus wall demarcated by the
zation using coils or liquid embolic agents [69] III and IV cranial nerves superiorly and the V and
(Table 4). VI cranial nerves inferiorly. With the aid of
678 T.R. Miller et al.

Fig. 16 Percutaneous onyx injection of an indirect CCF. map and (f) native fluoroscopy demonstrate a needle cours-
(a) Frontal and (b) lateral projections of a left internal ing through the inferior orbital fissure (white arrows c and
carotid artery angiogram demonstrate a type B CCF sup- d) and onyx cast within the cavernous sinus (black arrows
plied by left internal carotid artery meningeal branches. c, d). A balloon was inflated in the left internal carotid
Due to difficult access by transarterial or transvenous artery to prevent nontarget onyx embolization (red arrows
approaches, the lesion was accessed by direct transorbital c). Follow-up (e) frontal and (f) lateral left internal carotid
puncture of the left cavernous sinus following by instilla- artery angiograms demonstrates closure of the fistula
tion of onyx. Lateral projections of a (c) subtraction road

circulatory arrest and induced hypothermia, he associated morbidity and technical challenges of
went on to use this approach to treat both direct surgical repair of CCFs, as well as the success of
and indirect CCF, often preserving the carotid endovascular approaches, these procedures are
artery [72]. Modern surgical treatment of CCFs most often performed only after failure of
consists of a combined extradural-intradural endovascular treatment [64].
approach to the cavernous sinus, with disconnec-
tion of arterial feeders in the case of indirect
fistulas and clipping or suture repair of fistulous Stereotactic Radiotherapy
rents in the case of direct lesions [73, 74]. In
addition, both direct and indirect fistulas may Several small case series have demonstrated the
be indirectly occluded by packing the cavernous efficacy of stereotactic radiosurgery for the treat-
sinus with various materials, including muscle, ment of indirect, low-flow CCFs without high-
glue, thrombus, and wires [18, 73, 74]. Due to the risk features [75–77]. Successful closure of the
30 Carotid Cavernous Fistula 679

Fig. 17 Particulate embolization of a type C indirect CCF. two right external carotid artery pedicles (c, d) again dem-
(a) Frontal and (b) lateral projections of a right external onstrate supply to the fistula, which were subsequently
carotid artery angiogram demonstrating dural shunts embolized with polyvinyl alcohol particles (PVA). (e)
between external carotid meningeal branches and the cav- Frontal and (f) lateral projections of a right external carotid
ernous sinus (white arrows). Microcatheter injections of artery angiogram demonstrates occlusion of the fistula

arteriovenous shunt has been reported between However, stereotactic radiotherapy was
12 and 36 months following treatment [1]. supplemented by transarterial particulate emboli-
Onizuka et al. [76] reported their results of a zation in 13 of these patients [77].
small case series consisting of four elderly
women with symptomatic indirect CCF who
received stereotactic radiosurgery targeted to the
compartment of the involved cavernous sinus Conclusion
with a marginal dose of 13–15 Gy and a maximum
dose of 36–30 Gy. All four fistulas were success- Carotid cavernous fistulas are complex lesions
fully closed, and patients experienced symptom that can be challenging to diagnose and manage.
relief in 1–3 months. There were no instances of A multidisciplinary approach, including
lesion recurrence or adverse events from treat- neurointerventionalists, neurosurgeons, and oph-
ment during the follow-up period [76]. Pollock thalmologists, is often required to achieve the best
et al. [77] reported similarly high rates of symp- outcomes for patients. The benefits as well as the
tom improvement and fistula closure in 20 patients risks of treatment need to be carefully weighed
with indirect CCF (95 % and 93 %, respectively). against the natural history of these lesions,
680 T.R. Miller et al.

Table 4 Endovascular treatment options for indirect 3. Kurata A, Takano M, Tokiwa K et al (1993) Spontaneous
carotid cavernous fistulas carotid cavernous fistula presenting only with cranial
nerve palsies. AJNR Am J Neuroradiol 14:1097–1101
Treatment Potential Potential
4. Stiebel-Kalish H, Setton A, Nimii Y et al (2002) Cav-
modality advantages disadvantages
ernous sinus dural arteriovenous malformations: pat-
Transvenous Proven, safe and Trabeculated terns of venous drainage are related to clinical signs
coiling effective method cavernous sinus and symptoms. Ophthalmology 109:1685–1691
may preclude 5. Barrow DL, Spector RH, Braun IF et al (1985) Classi-
fistula closure fication and treatment of spontaneous carotid-
More controlled May not always cavernous sinus fistulas. J Neurosurg 62:248–256
deployment be possible due to 6. Harris FS, Rhoton AL (1976) Anatomy of the cavern-
compared to lack of venous ous sinus. A microsurgical study. J Neurosurg
liquid embolic access to 45:169–180
agents cavernous sinus 7. Hashimoto M, Yokota A, Yamada H et al (2000)
Transvenous Excellent Potential for Development of the cavernous sinus in the fetal period:
liquid penetration of nontarget closure a morphological study. Neurol Med Chir 40:140–150
embolic small venous of arterial supply 8. Ellis JA, Goldstein H, Connolly ES Jr et al (2012)
embolization compartments of to the brain, eye, Carotid-cavernous fistulas. Neurosurg Focus 32:E9
the cavernous cranial nerves 9. Stiebel-Kalish H, Setton A, Berenstein A et al (2002)
sinus Bilateral orbital signs predict cortical venous drainage
Can be used in May not always in cavernous sinus dural AVMs. Neurology
combination of be possible due to 58:1521–1524
coils lack of venous 10. Debrun G, Lacour P, Vinuela F et al (1981) Treatment
access to of 54 traumatic carotid-cavernous fistulas. J Neurosurg
cavernous sinus 55:678–692
11. Barry RC, Wilkinson M, Ahmed RM et al (2011) Inter-
Transarterial Excellent Potential for
ventional treatment of carotid cavernous fistula. J Clin
liquid penetration of nontarget closure
Neurosci Off J Neurosurg Soc Australas 18:1072–1079
embolic small venous of arterial supply
12. Miller NR (2007) Diagnosis and management of dural
embolization compartments of to the brain, eye,
carotid-cavernous sinus fistulas. Neurosurg Focus
the cavernous cranial nerves due
23:E13
sinus to either reflux of
13. Gemmete JJ, Ansari SA, Gandhi D (2009) Endovascular
Option when agent or
treatment of carotid cavernous fistulas. Neuroimaging
venous access to retrograde filling
Clin N Am 19:241–255, Table of Contents
cavernous sinus of
14. Lewis AI, Tomsick TA, Tew JM Jr (1995) Management
not possible non-catheterized
of 100 consecutive direct carotid-cavernous fistulas:
arterial pedicles
results of treatment with detachable balloons. Neuro-
Particulate Safe method High rate of surgery 36:239–244; discussion 244–235
embolization fistula recurrence 15. Annesley-Williams DJ, Goddard AJ, Brennan RP
et al (2001) Endovascular approach to treatment of
indirect carotico-cavernous fistulae. Br J Neurosurg
particularly low-flow, indirect fistulas. Finally, 15:228–233
16. Liang W, Xiaofeng Y, Weiguo L et al (2007) Traumatic
identification of high-risk features on catheter carotid cavernous fistula accompanying basilar skull
angiography, including cortical venous reflux fracture: a study on the incidence of traumatic carotid
and cavernous sinus venous varix, is essential to cavernous fistula in the patients with basilar skull fracture
appropriately triage these patients. and the prognostic analysis about traumatic carotid cav-
ernous fistula. J Trauma 63:1014–1020; discussion 1020
17. Helmke K, Kruger O, Laas R (1994) The direct carotid
cavernous fistula: a clinical, pathoanatomical, and
References physical study. Acta Neurochir 127:1–5
18. Halbach VV, Hieshima GB, Higashida RT et al (1987)
Carotid cavernous fistulae: indications for urgent treat-
1. De Keizer R (2003) Carotid-cavernous and orbital ment. AJR Am J Roentgenol 149:587–593
arteriovenous fistulas: ocular features, diagnostic and 19. Kobayashi N, Miyachi S, Negoro M et al (2003)
hemodynamic considerations in relation to visual Endovascular treatment strategy for direct carotid-
impairment and morbidity. Orbit 22:121–142 cavernous fistulas resulting from rupture of
2. Peeters FL, Kroger R (1979) Dural and direct cavern- intracavernous carotid aneurysms. AJNR Am J
ous sinus fistulas. AJR Am J Roentgenol 132:599–606 Neuroradiol 24:1789–1796
30 Carotid Cavernous Fistula 681

20. Gossman MD, Berlin AJ, Weinstein MA et al (1993) 34. Akiba H, Tamakawa M, Hyodoh H et al (2008) Assess-
Spontaneous direct carotid-cavernous fistula in child- ment of dural arteriovenous fistulas of the cavernous
hood. Ophthal Plast Reconstr Surg 9:62–65 sinuses on 3D dynamic MR angiography. AJNR Am J
21. Kupersmith MJ, Stiebel-Kalish H, Huna-Baron R Neuroradiol 29:1652–1657
et al (2002) Cavernous carotid aneurysms rarely 35. Chen JC, Tsuruda JS, Halbach VV (1992) Suspected
cause subarachnoid hemorrhage or major neurologic dural arteriovenous fistula: results with screening MR
morbidity. J Stroke Cerebrovasc Dis Off J Natl Stroke angiography in seven patients. Radiology
Assoc 11:9–14 183:265–271
22. Miller TR, Jindal G, Mohan S et al (2014) Diagnosis 36. Flaharty PM, Lieb WE, Sergott RC et al (1991) Color
and management of trigemino-cavernous fistulas: case Doppler imaging. A new noninvasive technique to
reports and review of the literature. J Neurointerv Surg diagnose and monitor carotid cavernous sinus fistulas.
45:674–687 Arch Ophthalmol 109:522–526
23. Hieshima GB, Cahan LD, Mehringer CM et al (1986) 37. Chen CC, Chang PC, Shy CG et al (2005) CT angiog-
Spontaneous arteriovenous fistulas of cerebral vessels raphy and MR angiography in the evaluation of carotid
in association with fibromuscular dysplasia. Neurosur- cavernous sinus fistula prior to embolization: a com-
gery 18:454–458 parison of techniques. AJNR Am J Neuroradiol
24. Rios-Montenegro EN, Behrens MM, Hoyt WF (1972) 26:2349–2356
Pseudoxanthoma elasticum. Association with bilateral 38. Elster AD, Chen MY, Richardson DN et al (1991)
carotid rete mirabile and unilateral carotid-cavernous Dilated intercavernous sinuses: an MR sign of
sinus fistula. Arch Neurol 26:151–155 carotid-cavernous and carotid-dural fistulas. AJNR
25. Schievink WI, Piepgras DG, Earnest FT et al (1991) Am J Neuroradiol 12:641–645
Spontaneous carotid-cavernous fistulae in Ehlers- 39. Hirabuki N, Miura T, Mitomo M et al (1988) MR
Danlos syndrome Type IV. Case report. J Neurosurg imaging of dural arteriovenous malformations with
74:991–998 ocular signs. Neuroradiology 30:390–394
26. Meyers PM, Halbach VV, Dowd CF et al (2002) Dural 40. Meckel S, Maier M, Ruiz DS et al (2007) MR angiog-
carotid cavernous fistula: definitive endovascular man- raphy of dural arteriovenous fistulas: diagnosis and
agement and long-term follow-up. Am J Ophthalmol follow-up after treatment using a time-resolved 3D
134:85–92 contrast-enhanced technique. AJNR Am J Neuroradiol
27. Goldberg RA, Goldey SH, Duckwiler G et al (1996) 28:877–884
Management of cavernous sinus-dural fistulas. Indica- 41. Ouanounou S, Tomsick TA, Heitsman C et al (1999)
tions and techniques for primary embolization via the Cavernous sinus and inferior petrosal sinus flow signal
superior ophthalmic vein. Arch Ophthalmol on three-dimensional time-of-flight MR angiography.
114:707–714 AJNR Am J Neuroradiol 20:1476–1481
28. Quinones D, Duckwiler G, Gobin PY et al (1997) 42. Coskun O, Hamon M, Catroux G et al (2000) Carotid-
Embolization of dural cavernous fistulas via superior cavernous fistulas: diagnosis with spiral CT angiogra-
ophthalmic vein approach. AJNR Am J Neuroradiol phy. AJNR Am J Neuroradiol 21:712–716
18:921–928 43. Ohtsuka K, Hashimoto M (1999) The results of serial
29. Klisch J, Huppertz HJ, Spetzger U et al (2003) dynamic enhanced computed tomography in patients
Transvenous treatment of carotid cavernous and dural with carotid-cavernous sinus fistulas. Jpn J Ophthalmol
arteriovenous fistulae: results for 31 patients and 43:559–564
review of the literature. Neurosurgery 53:836–856; 44. Huber P (1976) A technical contribution of the exact
discussion 856–837 angiographic localization of carotid cavernous fistulas.
30. Acierno MD, Trobe JD, Cornblath WT et al (1995) Neuroradiology 10:239–241
Painful oculomotor palsy caused by posterior-draining 45. Wakhloo AK, Perlow A, Linfante I et al (2005)
dural carotid cavernous fistulas. Arch Ophthalmol Transvenous n-butyl-cyanoacrylate infusion for com-
113:1045–1049 plex dural carotid cavernous fistulas: technical consid-
31. Bussiere M, Lownie SP, Pelz DM et al (2009) Direct erations and clinical outcome. AJNR Am J Neuroradiol
carotid-cavernous fistula causing brainstem venous 26:1888–1897
congestion. J Neuro-Ophthalmol Off J N Am Neuro- 46. Kirsch M, Henkes H, Liebig T et al (2006)
Ophthalmol Soc 29:21–25 Endovascular management of dural carotid-cavernous
32. Turner DM, Vangilder JC, Mojtahedi S et al (1983) sinus fistulas in 141 patients. Neuroradiology
Spontaneous intracerebral hematoma in carotid- 48:486–490
cavernous fistula. Report of three cases. J Neurosurg 47. Halbach VV, Higashida RT, Hieshima GB et al (1987)
59:680–686 Dural fistulas involving the cavernous sinus: results of
33. Cognard C, Januel AC, Silva NA Jr et al (2008) treatment in 30 patients. Radiology 163:437–442
Endovascular treatment of intracranial dural arteriove- 48. Higashida RT, Hieshima GB, Halbach VV et al (1986)
nous fistulas with cortical venous drainage: new man- Closure of carotid cavernous sinus fistulae by external
agement using Onyx. AJNR Am J Neuroradiol compression of the carotid artery and jugular vein. Acta
29:235–241 Radiol Suppl 369:580–583
682 T.R. Miller et al.

49. Kai Y, Hamada J, Morioka M et al (2007) Treatment of embolization of a dural carotid cavernous sinus fistula.
cavernous sinus dural arteriovenous fistulae by exter- AJNR Am J Neuroradiol 23:1153–1155
nal manual carotid compression. Neurosurgery 64. White JB, Layton KF, Evans AJ et al (2007)
60:253–257; discussion 257–258 Transorbital puncture for the treatment of cavernous
50. Gandhi D, Ansari SA, Cornblath WT (2009) Success- sinus dural arteriovenous fistulas. AJNR Am J
ful transarterial embolization of a Barrow type D dural Neuroradiol 28:1415–1417
carotid-cavernous fistula with ethylene vinyl alcohol 65. Tiewei Q, Ali A, Shaolei G et al (2010) Carotid cav-
copolymer (Onyx). J Neuro-Ophthalmol Off J N Am ernous fistulas treated by endovascular covered stent
Neuro-Ophthalmol Soc 29:9–12 grafts with follow-up results. Br J Neurosurg
51. Gomez F, Escobar W, Gomez AM et al (2007) Treat- 24:435–440
ment of carotid cavernous fistulas using covered stents: 66. Wang C, Xie X, You C et al (2009) Placement of
midterm results in seven patients. AJNR Am J covered stents for the treatment of direct carotid cav-
Neuroradiol 28:1762–1768 ernous fistulas. AJNR Am J Neuroradiol
52. Moron FE, Klucznik RP, Mawad ME et al (2005) 30:1342–1346
Endovascular treatment of high-flow carotid cavernous 67. Arat A, Cekirge S, Saatci I et al (2004) Transvenous
fistulas by stent-assisted coil placement. AJNR Am J injection of Onyx for casting of the cavernous sinus for
Neuroradiol 26:1399–1404 the treatment of a carotid-cavernous fistula. Neurora-
53. Biondi A, Milea D, Cognard C et al (2003) Cavernous diology 46:1012–1015
sinus dural fistulae treated by transvenous approach 68. Suzuki S, Lee DW, Jahan R et al (2006) Transvenous
through the facial vein: report of seven cases and treatment of spontaneous dural carotid-cavernous fis-
review of the literature. AJNR Am J Neuroradiol tulas using a combination of detachable coils and
24:1240–1246 Onyx. AJNR Am J Neuroradiol 27:1346–1349
54. Halbach VV, Higashida RT, Hieshima GB 69. Vinuela F, Fox AJ, Debrun GM et al (1984) Spontane-
et al (1988) Transvenous embolization of direct ous carotid-cavernous fistulas: clinical, radiological,
carotid cavernous fistulas. AJNR Am J Neuroradiol and therapeutic considerations. Experience with
9:741–747 20 cases. J Neurosurg 60:976–984
55. Siniluoto T, Seppanen S, Kuurne T et al (1997) 70. Vitek JJ, Smith MJ (2009) The myth of the Brooks
Transarterial embolization of a direct carotid cavernous method of embolization: a brief history of the
fistula with Guglielmi detachable coils. AJNR Am J endovascular treatment of carotid-cavernous sinus fis-
Neuroradiol 18:519–523 tula. J Neurointerv Surg 1:108–111
56. Luo CB, Teng MM, Chang FC et al (2006) 71. Parkinson D (1967) Transcavernous repair of carotid
Transarterial balloon-assisted n-butyl-2-cyanoacrylate cavernous fistula. Case report. J Neurosurg 26:420–424
embolization of direct carotid cavernous fistulas. 72. Parkinson D (1973) Carotid cavernous fistula: direct
AJNR Am J Neuroradiol 27:1535–1540 repair with preservation of the carotid artery. Technical
57. Serbinenko FA (1974) Balloon catheterization and note. J Neurosurg 38:99–106
occlusion of major cerebral vessels. J Neurosurg 73. Day JD, Fukushima T (1997) Direct microsurgery of
41:125–145 dural arteriovenous malformation type carotid-
58. Debrun G, Lacour P, Caron JP et al (1978) Detachable cavernous sinus fistulas: indications, technique, and
balloon and calibrated-leak balloon techniques in the results. Neurosurgery 41:1119–1124; discussion
treatment of cerebral vascular lesions. J Neurosurg 1124–1116
49:635–649 74. Tu YK, Liu HM, Hu SC (1997) Direct surgery of
59. Norman D, Newton TH, Edwards MS et al (1983) carotid cavernous fistulae and dural arteriovenous
Carotid-cavernous fistula: closure with detachable sil- malformations of the cavernous sinus. Neurosurgery
icone balloons. Radiology 149:149–157 41:798–805; discussion 805–796
60. Marques MC, Caldas JG, Nalli DR et al (2010) Follow- 75. Barcia-Salorio JL, Soler F, Barcia JA et al (1994) Ste-
up of endovascular treatment of direct carotid- reotactic radiosurgery for the treatment of low-flow
cavernous fistulas. Neuroradiology 52:1127–1133 carotid-cavernous fistulae: results in a series of
61. Goto K, Hieshima GB, Higashida RT et al (1986) 25 cases. Stereotact Funct Neurosurg 63:266–270
Treatment of direct carotid cavernous sinus fistulae. 76. Onizuka M, Mori K, Takahashi N et al (2003) Gamma
Various therapeutic approaches and results in knife surgery for the treatment of spontaneous dural
148 cases. Acta Radiol Suppl 369:576–579 carotid-cavernous fistulas. Neurol Med Chir
62. Zhao P, Liu L, Jiang C et al (2012) Coils and onyx 43:477–482; discussion 482–473
embolization of traumatic carotid-cavernous fistula 77. Pollock BE, Nichols DA, Garrity JA et al (1999) Ste-
caused by an intradural internal carotid artery reotactic radiosurgery and particulate embolization for
pseudoaneurysm. Neuroradiol J 25:231–236 cavernous sinus dural arteriovenous fistulae. Neurosur-
63. Mounayer C, Piotin M, Spelle L et al (2002) Superior gery 45:459–466; discussion 466–457
petrosal sinus catheterization for transvenous
Imaging of the Neurovasculitides
31
Adam J. Davis

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 684 Neurovasculitis is a term used to describe a
diverse spectrum of diseases characterized by
Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 685
Classification of the Vasculitides Affecting
inflammation of the blood vessels that may
the Central and Peripheral Nervous Systems . . . . . . . . 685 progress to ischemic injury of the central or
Diagnostic Approach to the Neurovasculitides . . . . . 687 peripheral nervous system. The overlapping
Laboratory and Histopathology . . . . . . . . . . . . . . . . . . . . . 690 clinical and radiographic features and the com-
Clinical Signs and Symptoms . . . . . . . . . . . . . . . . . . . . . . . 693
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 693
plex pathophysiology make it difficult to
quickly or clearly identify neurovasculitis as
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 694 the etiology of a patient’s symptoms. Nonethe-
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 694
Indirect Imaging Signs: Parenchymal . . . . . . . . . . . . . . . 694 less, simple yet comprehensive categorization
Indirect Imaging Signs: Vessel Lumen . . . . . . . . . . . . . . 700 schemes established by consensus meetings
Direct Imaging Signs: Vessel Wall . . . . . . . . . . . . . . . . . . 705 and based upon the anatomy, pathophysiology,
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 709 and imaging findings help to clarify the situa-
Mimics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 710 tion. For both the radiologist and the treating
Atherosclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 710 clinician, a systemic approach considering the
Tumor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 710
Reversible Cerebral Vasoconstrictive Syndrome . . . . 710
triad of organ systems involved, laboratory
results, and vessel size and type affected sim-
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 713
plifies the situation. Vessel size is particularly
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 715 important for the radiologist to consider as it
broadly categorizes the neurovasculitides
within the differential diagnosis. Imaging
plays an essential role in the diagnosis of
these diseases and may be divided into those
studies that investigate the vessel lumen, the
vessel wall, and the parenchyma. Demonstra-
tion of the vessel wall or perivascular inflam-
matory change is a specific and compelling
finding, which should be specifically pursued
by the radiologist. All imaging modalities con-
A.J. Davis (*)
tribute to the analysis; conventional catheter
Neuroradiology, Department of Radiology, NYU Langone
Medical Center, New York, NY, USA angiography is no longer the preeminent diag-
e-mail: adam.davis@nyumc.org nostic standard. Despite the uncommon
# Springer Science+Business Media New York 2016 683
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_22
684 A.J. Davis

occurrence of neurovasculitis, it is important to vessel stenosis or occlusion. Endothelial inflam-


be familiar with these diseases. They play a mation promotes intraluminal coagulation and
particularly important role in the differential thrombosis [1]. Perivascular inflammatory
diagnosis of stroke in young and middle-aged changes and edema contribute to the pathologic
adults and typically have devastating clinical picture. Arterial and venous components may be
consequences. If diagnosed early, they may be involved separately or together, and dural sinuses
effectively treated, relieving symptoms and may be affected. Generalized inflammatory pro-
avoiding permanent central and peripheral cesses may produce a secondary encephalitis or
neurologic damage. myelitis. Ischemia of the small vessel vasa
nervorum typically causes mononeuritis multi-
Keywords plex or polyneuropathy.
Neurovasculitides • Neurovasculitis • Cerebral Neurovasculitis has a wide clinical, radio-
vasculitis • Central nervous system vasculitis • graphic, and pathologic spectrum.
Stroke • Cerebral ischemia • Cerebral The overlapping clinical and radiographic fea-
infarction tures as well as the complex pathophysiology often
make it difficult to clearly identify neurovasculitis
List of Abbreviations as the etiology of the patient’s symptoms. The
ACA Anterior cerebral artery central and peripheral inflammatory and ischemic
CA Catheter angiography events associated with vasculitis produce symp-
CT Computed tomography toms that are similar to the more common
CTA Computed tomography angiography noninflammatory occlusive vascular diseases. The
18
FDG- F-fluorodeoxyglucose positron differential diagnosis of neurovasculitis may be
PET emission tomography overwhelming and confusing. The vasculitides
GCA Giant cell arteritis are sometimes confused with noninflammatory pri-
ICA Internal carotid artery mary vasculopathies both clinically and in the med-
MCA Middle cerebral artery ical literature. This includes noninflammatory
MRA Magnetic resonance angiography collagen vascular diseases and reversible vasocon-
MRI Magnetic resonance imaging strictive syndromes. Some authors will only con-
PACNS Primary angiitis of the central ner- sider the neurovasculitides within the context of
vous system autoimmune or autoinflammatory disease, exclud-
PCA Posterior cerebral artery ing vascular inflammation secondary to general-
RCVS Reversible cerebral vasoconstrictive ized disorders such as infection. Other authors
syndrome disregard the more generalized autoimmune and
SLE Systemic lupus erythematosus autoinflammatory diseases that are not primarily
TA Takayasu arteritis related to the vasculature but nonetheless directly
US Ultrasound affect the blood vessels, including systemic lupus
erythematosus (SLE). These disparities lead to
confusion and misunderstanding regarding the
Introduction nomenclature and classification of the neurovas-
culitides. This situation makes it difficult for phy-
Neurovasculitis is a term used to describe a sicians to have a common point of reference and
diverse spectrum of diseases characterized by interferes with the logical and efficient process of
inflammation of the blood vessels that may pro- diagnosis and treatment. Reiterating simple yet
gress to ischemic injury of the central or periph- comprehensive categorization, as outlined by con-
eral nervous system, resulting in a myriad of focal sensus meetings and based upon the anatomy, path-
and generalized neurologic symptoms. The injury ophysiology, and imaging findings helps to clear
is usually secondary to mural changes resulting in the conundrum.
31 Imaging of the Neurovasculitides 685

The primary vasculitides, inflammatory dis- inflammatory diseases that are systemic, affecting
ease affecting only the vasculature, are uncom- the vasculature throughout the entire body. Next
mon, even more so for clinicians interested in the are the generalized systemic autoimmune and
nervous system since only a fraction of these autoinflammatory diseases that produce vascular
patients have central or peripheral CNS involve- inflammation in addition to a generalized inflam-
ment. The estimated incidence of Churg–Strauss matory process affecting the body organs. These
syndrome is 20 per million, granulomatosis with include the connective tissue diseases. These cat-
polyangiitis (Wegener’s) 10 per million, and egories are distinct from those diseases that pro-
polyarteritis nodosa 0.9 per million people duce vasculitis secondarily by proximity
[2, 77]. Takayasu arteritis (TA) has a worldwide including infection, radiation, drugs, toxins, and
estimated incidence of 2.6 per million people [3] neoplasm. Table 1 organizes the more common
although this varies widely by geography; examples of these diseases.
autopsy studies in Japan report an incidence of The neurovasculitides are typically approached
1 per 3,000 population (333 per million) [4], as a long list of disease entities, which is inevitable
while studies in England report an incidence of as the etiologies of neurovasculitis are protean.
0.3 per million population [5]. It is apparent that Unfortunately, this leaves the clinician with no
the frequency of presentation for any given phy- practical coherent organizational system to refer
sician will vary by practice locale. Secondary to. A systemic approach to these diseases based
vasculitides, typically secondary to connective primarily upon the triad of:
tissue diseases including SLE, affect organ
parenchyma in addition to vasculature and are 1. Clinical presentation particularly the organ
more common. systems involved
Despite the uncommon occurrence of 2. Laboratory results
neurovasculitides, it is important to be familiar 3. Vessel size and type afflicted
with these diseases. They play a particularly
important role in the differential diagnosis of organizes and simplifies the subject for the radiol-
stroke in young and middle-aged adults, where ogist and clinician alike. Vessel size is particularly
the prevalence of atherosclerotic disease is small important for the radiologist to consider as it helps
[6, 7]. The neurovasculitides typically have dev- to broadly categorize the neurovasculitides within a
astating clinical consequences for the afflicted differential diagnosis (Fig. 1).
patient. If diagnosed early, they may be effectively Discussion of vessel size is problematic as there
treated with a myriad of immunosuppressive are many different classifications based on histo-
agents, relieving symptoms and avoiding perma- logic, pathologic, and clinical definitions, generally
nent damage to the brain, spinal cord, and periph- in reference to the entire human body and systemic
eral nerves. disease processes. The cerebrovascular system
skews this paradigm. Division into large elastic
arteries, medium highly muscular arteries, and
Overview small thin muscular arteries/arterioles as is utilized
by the prevailing systemic vasculitis classification
Classification of the Vasculitides systems [8, 9] fails to adequately describe the dis-
Affecting the Central and Peripheral tribution and pathology of neurovasculitides. The
Nervous Systems entire cerebrovascular system would coarsely be
divided into large elastic arteries from the aorta
The simplest classification systems divide the through the common carotid and vertebral arteries
neurovasculitides into primary vascular inflam- and then the muscular arteries, which correspond to
matory diseases affecting only the central and everything else distally to the capillary level. These
peripheral nervous systems from primary vascular classifications tend to disregard the unique
686 A.J. Davis

Table 1 Common etiologies of the neurovasculitides by which may effect the distribution of disease within
disease category the brain.
I. Nonsystemic primary vasculitis affecting only the In order to better organize the neurovas-
central or peripheral nervous system culitides, a more relevant classification is
Primary angiitis of the central nervous system (PACNS) required. Kuker [10, 25] proposed that the internal
Nonsystemic peripheral nervous system vasculitis
carotid arteries through their intracranial skull
(NSPNSV)
II. Systemic primary vasculitis
base segments including their primary divisions
Large vessel into the anterior cerebral artery (ACA) A1 seg-
Giant cell arteritis ments; the middle cerebral artery (MCA) M1 seg-
Takayasu arteritis ments; the vertebral artery, basilar artery, and
Medium vessel posterior cerebral artery (PCA) P1 segments; and
Polyarteritis nodosa their structural collaterals including the anterior
Kawasaki disease and posterior communicating arteries all be con-
Small-vessel vasculitis sidered to be of the large-vessel type. All cerebral
(More arteriolar) cortex, convexity, and branching arteries and their
Granulomatosis with polyangiitis (Wegener’s) penetrating segments are medium-vessel type,
Churg–Strauss syndrome discernable by catheter angiography (CA) and
Microscopic polyangiitis depending upon the technique by magnetic reso-
(More capillary venule) nance angiography (MRA) and computed tomo-
Henoch–Schönlein purpura graphic angiography (CTA) at the more proximal
Essential cryoglobulinemic vasculitis
second- and third-order branching segments.
Cutaneous leukocytoclastic angiitis
More distally, the small vessels are those not dis-
(Diverse involvement with emphasis on capillaries,
venules, and veins) cernible by imaging, corresponding to the smallest
Behçet’s disease muscular arteries and arterioles within the brain
III. Systemic autoimmune/autoinflammatory diseases parenchyma as well as the capillaries and proximal
affecting organ systems with a vasculitic component venules. Although Kuker did not discuss vascular
Systemic lupus erythematosus dimensions, these small vessels beneath the reso-
Rheumatoid lution of imaging correspond to 200 μm or less [11]
Scleroderma although even CA is severely limited below
Mixed connective tissue disease 500 μm [12]. Using this helpful and relevant clas-
Sjogren’s disease sification system, etiologies tend to cluster into the
IV. Secondary vasculitis following three groups:
Infection
Viral, bacterial, fungal, protozoan, spirochetes
1. Extracranial elastic, extracranial large vessel,
Neoplasm
and intracranial skull base large-vessel
Lymphoma
involvement
Lymphomatoid granulomatosis
2. Intracranial medium-vessel involvement
Lymphocytic vasculitis
Drugs
3. Intracranial small-vessel involvement
Amphetamines, hydralazine, levamisole, cocaine,
synthetic marijuana and other marijuana substitutes, Additionally, some etiologies tend to involve
immunosuppressive drugs for transplant patients the venous system with greater frequency and
(Tacrolimus), interferon sometimes even exclusively, though these small
Radiation venular diseases are difficult if not impossible to
detect with direct vascular imaging. One should
characteristics of brain arteries including a constantly bear in mind that these are categorical
decreased wall thickness to lumen ratio, lack of definitions to aid diagnosis; in reality, there is
external elastic lamina, diminished adventitia, and considerable overlap in vessel size involvement
perivascular subarachnoid fluid environment, all of among the neurovasculitides.
31 Imaging of the Neurovasculitides 687

Fig. 1 Giant cell arteritis. 78-year-old female with severe temporal arteries (white arrows). Maximum intensity pro-
worsening headache, diplopia, and jaw pain. MRA at pre- jection images of the ICAs (c) demonstrate intraluminal
sentation (a) demonstrates multifocal luminal narrowing linear defects (white arrows) consistent with dissection, a
and contour irregularity within the bilateral ICAs (white known complication of advanced GCA (Case courtesy of
arrows). CTA volume rendering (b) reveals multifocal Adam Davis, MD, New York University Langone Medical
short-segment luminal narrowing of the superficial Center, New York)

Table 2 Primary neurovasculitic diseases by distribution of vascular size and type


Aorta, large Medium Small arteries, Capillaries Venules Veins
arteries arteries arterioles
Takayasu arteritis ++ +
Giant cell arteritis ++ +
Kawasaki disease ++ +
PAN ++ +
ANCA + arteritides (CS, + ++ ++ +
MPA, GPA)
Henoch–Schonlein + ++ ++
Behçets + + ++ ++ ++
PACNS ++ ++ ++ ++ +
Based on reference [76]

A comprehensive clinical organization enables Diagnostic Approach to the


both the radiologist and clinician to arrive more Neurovasculitides
quickly and assuredly at the diagnosis, providing
a clear mandate for biopsy and/or treatment and The neurovasculitides tend to cluster into groups
resulting in improved outcome for the patient. An characterized by the size and type of vessel
organization of the most common neurovas- afflicted, an attribute that may be capitalized
culitides by vessel size is a helpful starting point upon for organizing these diseases (Table 2). Dis-
for practicing clinicians who may encounter these eases affecting the large extracranial and skull
patients, typically radiologists, internists, rheuma- base arteries may be differentiated from those
tologists, dermatologists, neurologists, and involving the intracranial medium and small arter-
neurosurgeons. ies and arterioles and, in a similar, fashion from
688 A.J. Davis

those preferentially affecting capillaries, venules, Table 3 Differential diagnosis of the common neurovas-
and veins. There is sometimes considerable ana- culitides and noninflammatory neurovasculopathies orga-
nized by vessel size and type
tomic overlap although clinical and laboratory
characterizations help to further define these I. Predominantly large artery extracranial disease
entities. Vasculitis
Systemic with CNS involvement
This approach is most helpful for the radiolo-
Takayasu/giant cell arteritis
gist who typically is able to separate the radio-
Atherosclerosis
graphic findings into broad vascular anatomic
Arterial dissection
categories, depending on the appearance of the
Fibromuscular dysplasia
vessels and the anatomic distribution of the imag- II. Predominantly medium and small artery intracranial
ing abnormalities. For example, an imaging eval- disease
uation, including an MRI of the brain Vasculitis
demonstrating multifocal small and punctate 1. Systemic with CNS Involvement – PAN and the
regions of T2 or FLAIR hyperintensity within ANCA-positive diseases: Churg–Strauss/GPA/
the cerebral white matter and irregularity of the microscopic polyangiitis
2. Autoimmune/autoinflammatory diseases –
most distal discernible intracranial vessels on
connective tissue diseases (SLE, rheumatoid, Sjögren’s)
CTA or CA, is more likely to correlate with a and Behçets
medium and/or small-vessel arteritis than an 3. Primary angiitis of the CNS
imaging demonstrating a large territory infarction 4. Vasculitis 2 , infection, radiation
and mural enhancement of the aorta or Atherosclerosis
brachiocephalic arteries, which likely is second- Diabetes
ary to a large-vessel arteritis. Moyamoya
The radiologic findings of the neurovas- Sickle cell
culitides may be similar to those of the more Neurofibromatosis
common noninflammatory vasculopathies Vasospasm – subarachnoid hemorrhage
including atherosclerosis. Noninflammatory Reversible cerebral vasoconstrictive syndrome
vasculopathies are associated with central III. Diseases involving the venules and veins
Vasculitis
and peripheral nervous system symptoms
Behçets, ANCA-positive diseases,
(Table 3), and separating inflammatory from Henoch–Schonlein, hypersensitivity vasculitis,
noninflammatory entities may be difficult, partic- lymphomatoid granulomatosis, lymphocytic vasculitis
ularly when imaging characteristics are nearly Hypercoagulable state, neurofibromatosis
indistinguishable. In these cases, clinical criteria IV. No vessel size predilection
typically help. For example, intracranial athero- Thromboembolic occlusion
sclerosis may be difficult to separate from a small Cardio-embolic including septic emboli, arterioarterial
artery-type vasculitis such as primary angiitis of 2 to atherosclerotic plaque, hypercoagulable states
the central nervous system (PACNS), SLE, or
Churg–Strauss syndrome. However, young age
or the absence of large-vessel, skull base, and As an example, vascular mural thickening with
calcified mural thickening should cast doubt on intramural or perivascular enhancement associ-
the diagnosis of atherosclerosis. Similarly, dis- ated with a segmental luminal narrowing at extra-
tinctive clinical features such as the presence of cranial sites would suggest TA or giant cell
a facial malar rash with elevated renal function arteritis (GCA). In this circumstance, the more
tests or serum eosinophilia with asthma would common noninflammatory vasculopathies includ-
steer the diagnosis toward SLE (Fig. 2) or ing atherosclerosis or spontaneous dissection sec-
Churg–Strauss disease (Fig. 3). Alternatively, in ondary to connective tissue disease would be
some cases, the imaging characteristics of unlikely.
neurovasculitis are distinctive and compelling, Peripheral neuropathy is an area of consider-
allowing for a primary radiographic diagnosis. able clinical overlap between the inflammatory
31 Imaging of the Neurovasculitides 689

Fig. 2 Systemic lupus erythematosus. 39-year-old female involvement of the parietal and occipital gray matter. Right
with known systemic lupus erythematosus and transient medial parietal cortical hyperintensity on diffusion-
worsening, bilateral lower extremity weakness, and numb- weighted imaging (a, middle, black arrow) with ADC
ness. MRI FLAIR sequence at presentation (a left) reveals low signal intensity (not shown) is consistent with acute
bilateral hemispheric white matter signal abnormality with ischemia in the right PCA parieto-occipital division.
690 A.J. Davis

and noninflammatory causes of vasculopathy. typically performed to evaluate for thrombotic


Many noninflammatory vasculopathies, including disorders that may be mistaken for vasculitis.
hypertension, atherosclerosis, and diabetes, may This typically includes prothrombin time, partial
cause extremity sensory neuropathies and cerebro- thromboplastin time, protein C, protein S, and
vascular abnormalities that can be difficult to dis- fibrinogen levels. More sophisticated clotting
tinguish from vasculitis secondary to polyarteritis tests are pursued if suspicion is high and a hema-
nodosa, granulomatosis with polyangiitis tology consultation is involved.
(Wegener’s), microscopic polyangiitis, Acute-phase reactants, usually erythrocyte
Churg–Strauss, SLE, or a myriad of other inflam- sedimentation rate and C-reactive protein, may
matory vascular diseases that have a similar pre- be elevated. Serologic/antibody testing typically
sentation. Again, the presence and pattern of includes rheumatoid factor, antinuclear antigen,
associated systemic organ involvement in conjunc- antibody to double-stranded DNA, anticardiolipin
tion with the laboratory results will help to steer the antibodies, and cryoglobulins. These tests are
differential diagnosis in the correct direction. helpful for evaluating the more common systemic
autoinflammatory and autoimmune diseases
including SLE and rheumatoid arthritis. Antinu-
Laboratory and Histopathology clear cytoplasmic antibodies (p-ANCA and
c-ANCA) are routinely obtained to evaluate for
Familiarity with the basics of the laboratory eval- primary systemic vasculitis including
uation for vasculitis is helpful to the radiologist. granulomatosis with polyangiitis (Wegener’s),
They are a constant and essential component in Churg–Strauss, and microscopic polyangiitis.
the diagnostic evaluation of these diseases. Perti- These tests are negative or weakly positive in
nent laboratory tests should be well regarded by polyarteritis nodosa, an important differentiating
the radiologist as they help to guide the imaging factor.
differential diagnosis toward the correct Infectious etiologies may be the primary or
conclusion. secondary causative factor in neurovasculitis.
A complete blood count is invariably Bacterial infection leading to leptomeningitis is a
performed. Anemia and leukocytosis are frequent particularly common etiology that may produce a
components of the systemic vasculitides. Eosino- secondary vascular inflammation resulting in
philia may be present, but when excessively ele- ischemia and infarction. Infectious agents may
vated, may indicate Churg–Strauss arteritis in the not only lead directly to inflammatory changes
right clinical setting. A coagulation panel is of the blood vessels but have also been postulated

Fig. 2 (continued) Gradient echo imaging (a, right, black- within the bilateral MCA M1 segments (black arrows).
outlined arrow) demonstrates focal susceptibility effect, Similar changes are present within the distal ICAs (not
indicating hemosiderin deposition from microhemorrhage. marked). The right PCA demonstrates multifocal luminal
The patient was treated for presumed hypercoagulable narrowing and abrupt occlusion of the parieto-occipital
thrombotic complications of SLE and discharged. The branch (black-outlined arrows). The extensive, large- and
patient presented 3 months later with profound left medium-vessel, multisegmental, tandem, luminal irregu-
hemiparesis and sensory deficits. Diffusion MRI of the larity and narrowing are more indicative of a vasculitic
brain (b) reveals restricted diffusion characterized by process than a thromboembolic process. CA (d) confirms
increased T2 diffusion and decreased ADC signal intensity the long-segment irregular luminal narrowing with inter-
within a large confluent region of the medial right parietal spersed segments of normal luminal caliber and contour
lobe and occipital lobes, extending to the splenium of the within all vascular territories. Immunosuppressive therapy
corpus callosum, consistent with the extension of acute was intensified for treatment of presumed lupus vasculitis.
infarction within the right PCA territory. MRA (c) demon- The patient’s clinical condition stabilized over the follow-
strates multifocal luminal narrowing, mostly short segment ing several months (Case courtesy of Adam Davis, MD,
and occasionally severe, as well as long-segment stenoses New York University Langone Medical Center, New York)
31 Imaging of the Neurovasculitides 691

Fig. 3 Churg–Strauss disease vs. eosinophilic vasculitis. performed for complaints of headache (b, FLAIR right,
45-year-old female with a history of rheumatoid arthritis, ADC left) reveals a punctate right basal ganglia chronic
asthma, and colon cancer in situ (PET confirmed not met- lacunar infarct (black arrows). CTA of the brain (c) dem-
astatic) presents with abdominal pain, nausea, vomiting, onstrates multifocal short- and long-segment luminal
lower extremity pain, and cyanosis. WBC 39,000, eosino- narrowing within the ACA (black arrows), MCA (white
phil count >15,000, C-reactive protein 110, rheumatoid arrows), and superficial temporal artery (black arrow-
factor elevated, and pANCA (positive). MRA of the heads). Intracranial vessels demonstrate marked luminal
extremities (a) demonstrates multifocal brachial, radial, contour irregularity, while the extracranial arteries demon-
posterior tibial, and dorsalis pedis artery stenoses and strate a more long-segment smooth and concentric
occlusions. Emergent attempt at thrombolysis was unsuc- narrowing, reflecting the variability of the radiographic
cessful and ischemia progressed to a gangrenous right foot findings of these diseases even in the same individual
requiring amputation. Initially assumed to be a multi- (Case courtesy of Adam Davis, MD, New York University
embolic event, no cardiac source was uncovered by Langone Medical Center, New York)
transesophageal echocardiography. MRI of the brain
692 A.J. Davis

Fig. 4 Lymphocytic vasculitis. 15-year-old male with enhancement within the deep cerebellum and brain stem
X-linked lymphoproliferative disease. Worsening confusion. were also present (not shown). The patient did not respond
MRI FLAIR axial imaging (a, left) demonstrates extensive to ganciclovir for suspected Epstein–Barr virus encephalitis;
confluent abnormal hyperintense signal throughout the bilat- a common complication of X-linked lymphoproliferative
eral hemispheric white matter as well as more patchy disease and concern for lymphocytic vasculitis or lymphoma
involvement of the deep nuclei and cortical gray matter. were raised. Brain biopsy was performed. Histopathology (b,
Diffusion imaging (not shown) failed to demonstrate an black arrow) confirms a perivascular polyclonal lymphocytic
evidence for acute infarction. Post-gadolinium T1-weighted infiltrate consistent with vasculitis. Treatment with cortico-
sagittal images (b, right) demonstrate striking radial linear steroids resulted in rapid clinical improvement (Case cour-
enhancement corresponding to the periventricular venous tesy of Yvonne Lui, MD, New York University Langone
system (white arrows). Additional regions of linear Medical Center, New York)

to be a predisposing factor in the development of encephalopathies. Regardless, it is important to


several vasculitic diseases including PACNS investigate CNS infection as well as exclude
[1, 13]. Consequently, CSF and serum investiga- demyelinating disease as a possible etiology.
tion for infection is often performed. Examination Biopsy is widely considered the gold standard
of the CSF including routine cell count, glucose, in the evaluation of neurovasculitis. When labora-
protein, Gram stain, and serologic testing for tory and imaging tests do not provide a clear diag-
syphilis, HIV, hepatitis B, Epstein–Barr virus, nosis or when the systemic evaluation is normal
varicella or other suspected infectious agents is and PACNS is suspected, brain biopsy is required.
routinely acquired. Granulomatous entities, par- Typically, sampling of the affected brain paren-
ticularly tuberculosis and sarcoidosis, may need to chyma and particularly the adjacent leptomeninges
be excluded as their imaging signs, including with a wedge biopsy guided by the imaging find-
multifocal regions of parenchymal signal abnor- ings [15, 16] provides the highest rate of success,
mality as well as leptomeningeal and parenchy- although nearly 15–38 % of biopsies are reportedly
mal enhancement, may overlap with vasculitic negative [14–18] in studies investigating all causes
processes including granulomatosis with of neurovasculitis. Children tend to have a higher
polyangiitis (Wegener’s) and PACNS. rate of positive brain biopsies than adults [19]. It is
Conversely, a negative laboratory evaluation is uncertain whether random sampling decreases the
a very important diagnostic component in the yield; it is generally not recommended but may be
evaluation of neurovasculitis. Lack of systemic necessary if no focal lesions are detected, if the
inflammatory markers is strongly supportive for lesions identified occupy eloquent regions of the
the diagnosis of PACNS, although even in these brain, or if the lesions are located deep within the
patients up to two-thirds may have nonspecific spinal cord parenchyma. It is important to consider
inflammatory serum markers including elevated that in approximately one-half of positive brain
erythrocyte sedimentation rate [14]. biopsies performed for the evaluation of PACNS,
CSF evaluation is usually nonspecific as an an alternative diagnosis is made, including neo-
elevated protein and leukocytosis may be present plasm (Fig. 4), demyelinating disease, and infec-
in many infectious and inflammatory tion [15, 18].
31 Imaging of the Neurovasculitides 693

The risk of biopsy is not negligible although it cortex. It may be acute and fulminant in presenta-
varies widely within the literature. A mortality tion or chronic and indolent.
rate of up to 1.5 %, a morbidity of up to 2 %, Systemic signs and symptoms frequently
and a complication rate of up to 20 % have been accompany the neurologic dysfunction and may
reported, although most complications are tran- at first be misleading. Fever, night sweats, weight
sient [20, 21]. Very few patients suffer permanent loss, weakness, rash, and arthropathy may initiate
neurologic deficit [15]. This relatively small risk an evaluation for infection or cancer. In fact, many
needs to be weighed against the risk of erroneous of the imaging studies that the radiologist encoun-
diagnosis and possible inappropriate or unneces- ters that ultimately result in a diagnosis of vascu-
sary treatment with potentially toxic immunosup- litis will initially be requested for other
pressive medications. Marsh et al. [2] warn that in indications. Even when signs and symptoms are
a series of five patients referred for a treatment of isolated to the neurologic system, demyelinating
primary CNS vasculitis, in which four of whom disease, infectious meningitis, acute disseminated
had already began immunosuppressive treatment, encephalomyelitis, atherosclerosis, and cardio-
none were ultimately found to have vasculitis. genic or thrombophilic embolic stroke are often
With this in mind, it is important to consider that the working diagnoses. When vasculitis is sys-
75 % of cases of neurovasculitis are diagnosed temic and not suspected, imaging of the brain is
and managed without biopsy [22]. Alternatively, often performed in symptomatic patients to eval-
when systemic organs are involved and a primary uate for hemorrhage or infarct. In these patients,
vasculitis or systemic autoimmune disease is con- other organ systems are frequently involved and
sidered, a biopsy of non-neurologic tissues may the neuroradiologist should take particular note
provide a less morbid means to confirm the diag- when imaging is performed for the indication of
nosis. This should be a strong inclination to per- neurovascular disease or encephalopathy and
form neck, chest, and abdomen imaging in there is a concurrent evaluation for suspected pul-
patients with suspected CNS vasculitis. monary, renal, liver, musculoskeletal, or dermato-
logic disease. The same is true when there is an
evaluation for elusive, cryptic, oncologic, or
Clinical Signs and Symptoms infectious disease in a patient with profound cen-
tral or peripheral CNS symptoms.
The signs and symptoms of neurovasculitis are
reflections of the many diseases that cause it [20,
23, 24]. It is roughly divided into symptoms Treatment
related to central nervous system and peripheral
nervous system involvement, the latter typically The treatment of the neurovasculitides is beyond
presenting as mononeuritis multiplex or sensory the scope of this chapter. Nonetheless, it is impor-
polyneuropathy. Cranial nerve involvement is a tant for the radiologist to consider that prompt and
prominent component of some of the neurovas- confident diagnosis of neurovasculitis and its
culitides, particularly granulomatosis with underlying etiology is critically important for
polyangiitis (Wegener’s), and is a distinguishing these patients. Many of these illnesses may be
feature. The involvement of the central nervous effectively treated with immunosuppressive
system results in a more diverse symptomology drugs [20], typically corticosteroids and cyclo-
and may be generalized or focal. Headache, phosphamide. Alternative agents including aza-
encephalopathy, seizure, stroke-like events, cog- thioprine, methotrexate, thalidomide, or
nitive impairment, and movement disorders par- interferon may be used. Plasmapheresis may be
ticularly chorea and myoclonus are described. helpful for certain diseases including
Behavioral abnormalities may occur, particularly cryoglobulinemia. Early treatment may induce
in children. Visual change may result from an long-term remission or cure, while delayed treat-
impairment of the optic nerve, tract, radiation, or ment may result in permanent vascular and
694 A.J. Davis

neurologic damage. As these drugs may have evaluating neurovasculitis. Gomes [26] conve-
serious and debilitating side effects, increased niently divides the imaging into three groups:
confidence in the diagnosis of vasculitis via imag- studies that investigate the vessel lumen, the ves-
ing provides a clear mandate to the referrer for sel wall, and the parenchyma. Parenchymal find-
their use. ings are the least specific but are usually necessary
to detect the presence of disease as well as follow
progression or remission. Vessel lumen abnormal-
Imaging ities are highly suggestive for vasculitis when
present but are not specific and the sensitivity is
Introduction low for the small-vessel vasculitides. Demonstra-
tion of vessel wall or perivascular inflammatory
Imaging plays an important if not essential role in changes are a more specific and compelling find-
the diagnosis of neurovasculitis. Nearly all ing, although it is only inconsistently found, likely
patients suspected of harboring an inflammatory due to the limitations of imaging technique. MRI
vasculopathy with neurologic signs and symp- and nuclear medicine imaging provide the most
toms undergo imaging of the brain and/or spinal sensitive evaluation for vessel wall inflammation
cord as well as dedicated imaging of the although extracranial large elastic vessel inflam-
cervicocerebral vasculature. Unfortunately, mation is seen particularly well with CT.
parenchymal imaging provides little, if any,
pathognomic findings of neurovasculitis. The
presence of ischemia, edema, intraparenchymal Indirect Imaging Signs: Parenchymal
and subarachnoid hemorrhage, and abnormal
parenchymal and leptomeningeal enhancement CT of the brain or spine is often employed as the
may be present in many different cerebral vaso- first imaging examination in patients presenting
occlusive, infectious, inflammatory, demyelinat- with acute or progressively worsening symptoms.
ing, and neoplastic conditions. Additionally, the It provides a rapid evaluation for the etiology of
radiographic findings and the specificity and sen- neurologic emergencies including hemorrhage,
sitivity of neuroimaging are variable and depen- infarction, or mass. Other than hemorrhage,
dent upon the specific disease entity causing the CT is both relatively nonspecific and insensitive
neurovasculitis. PACNS has a vastly different to the findings that are most indicative of
appearance than large-vessel primary vasculitis neurovasculitis.
such as GCA or a secondary vasculitis from infec- MRI of the brain or spinal cord is typically
tion. Nonetheless, the constellation of imaging employed as the next examination, although for
features may lead the radiologist to the correct patients with nonurgent, subacute, or chronic
diagnosis or at least a focused differential diagno- signs and symptoms, it is likely to be the first
sis within the given clinical context. diagnostic modality performed. MRI has become
Kuker [13, 25] divided the imaging findings of the standard of imaging for the initial evaluation
neurovasculitis into indirect and direct signs, the of neurovasculitis [13] since the sensitivity to
former related to ischemia, hemorrhage, and vas- parenchymal change is unequaled by other modal-
cular stenoses thought to be unlikely secondary to ities. Furthermore, MRI may still be positive in
hypertensive or atherosclerotic disease. Direct patients with reversible symptoms.
signs include findings related to vessel wall MRI findings are diverse [10, 13, 25, 27,
inflammatory changes including mural thickening 28]. The most common findings include
and enhancement. The radiologist should bear in T2/FLAIR hyperintense lesions secondary to
mind that regardless of the imaging modality ischemia distributed throughout the subcortical
employed (CT, CTA, MRI, MRA, CA, US, and deep white matter, the deep gray nuclei, and
nuclear medicine), the imaging findings will the cortex. The MCA territory is the most
have varying importance and specificity when involved vascular distribution [29, 30].
31 Imaging of the Neurovasculitides 695

Diffusion-weighted imaging helps to distinguish However, when the clinical information is consid-
acute, subacute, and chronic ischemia and is man- ered, including patient ethnicity along the geo-
datory. Lesions are frequently bilateral and of graphic “silk route” from the Middle East to
differing ages. Involvement of multiple vascular Japan, HLA-B51 variation, and stereotypical
territories or lesions within a frankly nonvascular physical signs such as oral and genital ulcers or
territorial distribution is a clue to the diagnosis uveitis, the imaging findings are virtually patho-
although it should not be confused with a gnomonic (Fig. 6a–c).
thrombophilic or cardiogenic multi-embolic pro- Intracranial hemorrhage, typically intrapar-
cess producing ischemia. Ischemic lesions are enchymal or subarachnoid, may be a presenting
reportedly present in nearly half of patients with or associated radiographic finding for nearly all of
PACNS [28] and are a constant feature in all the the neurovasculitides [33] although it occurs less
neurovasculitides (Fig. 5a). commonly than ischemia. Ischemia is present in
These nonspecific white matter changes may nearly 40 % of patients with PACNS, while hem-
be the only finding in symptomatic patients [21, orrhage is present in only 4–12 % [16, 34,
23]. In the younger patient population, these find- 35]. There is uncertainty regarding the signifi-
ings are unlikely to be attributed to atherosclerotic cance of micro-hemorrhage in these patients.
hypertensive disease as in older patients but may Prior literature mainly focused on frank hemor-
be difficult to distinguish from demyelinating rhagic lesions easily visualized with CT or con-
disease. ventional MRI sequences. Cortical petechial
Occasionally, diseases exhibit a more charac- hemorrhage, best visualized with T2* technique
teristic radiographic appearance. Behçet’s disease (gradient echo or susceptibility weighted imag-
is a notable example [31, 32]. Both ischemic and ing), has been suggested to be more commonly
nonischemic changes secondary to inflammation associated with vasculitis than the previously con-
or demyelination occur in either a mesodien- sidered [34] although in one series [36] of
cephalic junction pattern (46 % of patients) or a 25 patients with variable etiologies of intracranial
pontobulbar pattern (40 % of patients). It is typi- neurovasculitis, none demonstrated T2* changes
cally tegmental in distribution. More posterior to suggest acute or chronic blood products
mesodiencephalic lesions tend to extend caudal (Fig. 2a). This may be a helpful distinguishing
to the pontine tegmentum and superior cerebellar radiographic characteristic as diseases frequently
peduncles, while more anterior lesions tend to included in the differential diagnosis of
extend along the corticospinal tracts. The red neurovasculitis, particularly hypertension and
nucleus is spared. Upward extension typically amyloid angiopathy, are commonly associated
involves the posterior limbs of the internal cap- with cortical and white matter T2* susceptibility
sules and the contiguous globus pallidus and/or artifacts secondary to micro-hemorrhages.
putamen. There is no consensus as to the preva- Leptomeningeal enhancement by MRI is pre-
lence of basal ganglia involvement; it is reported sent in up to 9 % of patients with PACNS [27, 35]
as occurring in a majority of patients in one study and may be associated with granulomatous histol-
[32] and within a minority of patients in another ogy at biopsy (Fig. 5b). This finding is important
[31]. T2 hyperintensity at the onset eventually as this subset of patients may have negative MRA
disappears or becomes less prominent, leaving a and CA [35]. Leptomeningeal enhancement may
T1 iso- or hypointense lesion that is smaller in also be secondary to infection, itself a separate
size. Nodular enhancement in the acute phase is etiology for vasculitis, as well as carcinomatous
frequently present, while hemorrhage is rare. In meningitis and granulomatous inflammatory dis-
isolation from the clinical scenario, the radio- eases including neurosarcoidosis. This subset of
graphic appearance might be confusing. Mass- patients with PACNS typically present with an
like radiographic characteristics and/or nodular acute clinical onset and cognitive dysfunction,
enhancement may raise concern for a primary creating a clinical scenario that may be further
glial neoplasm, metastasis, or CNS lymphoma. confused with infection or neoplasm. Laboratory
696 A.J. Davis

Fig. 5 Primary angiitis of the central nervous system. white matter and left basal ganglia (black arrows) are
59-year-old male with remote history of cocaine and alco- sharply defined, without mass effect and likely reflect old
hol abuse. New onset lethargy and confusion progressing infarctions. Both the cortex and underlying white matter of
to delirium and agitation. Clinical and laboratory evalua- the right occipital lobe are involved, as is the right
tion for systemic disease, including toxicology, was nega- splenium of the corpus callosum (white arrows). In these
tive. Initial imaging evaluation reveals numerous infarcts locations, the margins are more ill defined and there is
of varying ages, situated within multiple vascular terri- subtle mass effect characterized by sulcal and ventricular
tories, with both small perforator and large artery etiology. effacement, suggesting acute ischemia in the right PCA
Non-contrast CT (a, top) demonstrates multifocal regions territory. MR FLAIR imaging (a, middle) demonstrates
of low attenuation. Those in the right frontal subcortical central low and peripheral high signal intensities within
31 Imaging of the Neurovasculitides 697

Fig. 6 Behçet’s disease. 38-year-year old Jordanian male respectively, white arrows) within the left paramedical
presenting with ataxia, dysphagia, and dysarthria. Patient’s caudal midbrain reflect acute or early subacute infarction.
brother carried a diagnosis of Behçet’s disease. Axial and Behçet’s disease often affects the venous system, account-
sagittal FLAIR imaging (a, left and right respectively) ing for the “non-territorial” vascular distribution. Hemor-
demonstrates a strikingly geographic abnormal rhage is unusual. Axial and coronal post-gadolinium
hyperintense signal intensity within the tegmentum of the T1-weighted images (c, left and right respectively) dem-
midbrain and pons. There is little mass effect. This brain onstrate peripheral, curvilinear, and punctate nodular
stem distribution is a common appearance of Behçet’s enhancement typically associated with Behçet’s disease.
disease and often extends cephalad to the basal ganglia, The appearance may sometimes be confusing – this patient
internal capsules, and diencephalon. Areas of diffusion was initially referred for evaluation of a brain stem tumor
restriction characterized byT2 diffusion hyperintense sig- (Case courtesy of Adam Davis, MD, New York University
nal and ADC hypointense signal (b left and right Langone Medical Center, New York)

Fig. 5 (continued) the frontal and periventricular white arrows) with intervening regions of normal-appearing vas-
matter lesions (black arrows) consistent with chronic culature. At the bottom of the image, vascular narrowing
encephalomalacia from old infarctions. The FLAIR within the PCA (not marked) is present. More than two
hyperintense signal within the right occipital lobe is more lesions within at least two territories is an important diag-
confluent and extends to the posterior temporal lobe and nostic criterion for MRA to avoid false-positive results. CA
splenium, involving both the cortex and white matter (d and e) reveals completely normal extracranial vascula-
(white arrows), and better delineates the extent of the ture, helping to narrow the differential diagnosis toward the
acute infarct. Diffusion imaging (c, bottom) demonstrates diagnosis of PACNS. The ACA (black arrowheads), MCA
restricted diffusion confirming the acuity of the occipital (black arrows), and PCA (black-outlined arrows) vascula-
ischemia and the chronicity of the other lesions. ture demonstrate short-segment mild-to-severe stenoses.
T1-weighted imaging pre- and post-gadolinium (b) dem- The larger vessels are not involved nor are the branch
onstrates extensive leptomeningeal enhancement along the points, a feature more commonly associated with athero-
cortical surface of the posterior temporal and occipital sclerotic disease further helping to arrive at the diagnosis of
lobes, a well described feature of granulomatous-type PACNS (Case courtesy of Adam Davis, MD, New York
PACNS. CTA (c) demonstrates multifocal vascular University Langone Medical Center, New York)
narrowing within several branches of the MCA (white
698 A.J. Davis

evaluation including CSF examination and resonance spectroscopy may reveal elevated glu-
leptomeningeal biopsy may be required to differ- tamate and glutamine and reduced N-acetyl aspar-
entiate these patients. tate [39, 41]. While there is no consensus on the
Unusual MRI findings associated with cerebral role of choline [39], absence of a choline elevation
vasculitis have been reported and may be mislead- helps to exclude a neoplastic etiology, although
ing [27]. For example, a tumor-like appearance the presence of a choline peak does not exclude
characterized by single or multiple discrete PACNS or inflammatory lupus vasculopathy
lesions with increased T2/FLAIR and decreased [41, 42].
T1 signal intensity, surrounding white matter MRI is the most sensitive imaging modality
edema pattern, enhancement, and mass effect available to the clinician for the evaluation of the
was seen in vasculitis patients, particularly those neurovasculitides. Its utility and reliability is
with Behçets, PACNS, and lymphocytic vasculitis nonetheless dependent on the clinical scenario.
[37–40] (Figs. 4, 5, and 6). Up to 4 % of patients For example, patients with focal sensorimotor
with PACNS present with this finding and may be deficits secondary to infarction will have 100 %
associated with amyloid angiopathy at sensitivity on MRI, while patients with more
histology [35]. nonspecific symptoms such as headache second-
The variability of imaging findings of cerebral ary to vascular or leptomeningeal inflammation
vasculitis is nowhere more clearly demonstrated will have a lower MRI sensitivity. The literature
than in a case report by Qu et al. [40] of a patient reports the sensitivity for MRI and parenchymal
with the ultimate diagnosis of PACNS who over a abnormalities for all types of cerebral vasculitis
7-month period of time was sequentially diag- ranges from 83 % to 100 %, mostly exceeding
nosed with idiopathic headache following a nor- 90 % [15, 28, 29, 36, 43–46]. Some studies report
mal head CT; multifocal basal ganglia infarcts a 100 % sensitivity of MRI for all types of auto-
secondary to tuberculous meningitis following immune neurovasculitis (though more accurately,
an abnormal CT with lesions in the basal ganglia; this sensitivity is based on cross-sectional imag-
viral meningoencephalitis following an abnormal ing since some of these studies were combined
CT and MRI with ischemic lesions in the basal with CT) and propose that normal MRI excludes
ganglia; malignant glioma following an MRI the diagnosis [30, 44–46]. Other studies conclude
demonstrating a parietal lobe mass lesion with that MRI is sensitive but not infallible, citing
midline shift; and, finally, cerebral vasculitis diag- series of patients with angiographic, clinical, or
nosed only when the MRA and conventional biopsy-proven cerebral vasculitis but negative
angiogram demonstrated diffuse vascular steno- MRI [29, 30, 36, 47]. However, many of these
ses. Brain biopsy confirmed the diagnosis. series are problematic in that FLAIR imaging,
Figure 7 demonstrates a case of HIV vasculitis commonly considered the most sensitive of the
presenting with ischemia which initially was diag- long TR sequences for detecting gray and white
nosed and followed as congenital fusiform aneu- matter and T2 signal intensity abnormalities, was
rysms and thromboembolic events. not included in the imaging evaluation.
Perfusion CT or MRI imaging may be helpful Despite the high sensitivity of MRI, specificity
in the evaluation of vasculitis since symptomatic has been reported to be as low as 19 % [15]. It
patients with normal MRI findings may have per- might be expected that CA, which provides the
fusion abnormalities [36]. These abnormalities greatest vascular luminal resolution (see “Indirect
include delayed perfusion and reduced relative Imaging Signs: Vessel Lumen” below), would
cerebral blood volume [27, 39]. Perfusion imag- significantly improve the diagnostic sensitivity
ing is suggested for all symptomatic patients with and specificity. Yet surprisingly, studies demon-
initial negative CT or MR imaging studies not strate that CA provides little additional benefit in
only in the evaluation of cryptic vasculitis but to specificity when differentiating vasculitis from
exclude the presence of ischemia secondary to the noninflammatory vasculopathies [15, 21, 36]. Fur-
more common vasculopathies. Magnetic thermore, CA does not improve sensitivity as
31 Imaging of the Neurovasculitides 699

Fig. 7 HIV vasculitis. 19-year-old female with congenital bilateral, long-segment, irregular, fusiform dilatation of the
HIV. History of cerebral infarction at the age of 10 years old. ICA and the most proximal MCA and ACA segments (white
Imaging evaluation at age 10 with MRA (not shown) arrows). The right ICA and MCA reveal areas of luminal
revealed intracranial bilateral ICA supraclinoid segment stenosis not appreciated on earlier examinations. CTA
fusiform dilatation with an irregular patulous contour reveals a more detailed and diagnostic view of the intracra-
extending into the proximal middle and anterior cerebral nial vasculature. The right MCA (c) demonstrates regions of
arteries. Findings were presumed to represent congenital short- and long-segment concentric vascular stenoses (black
aneurysmal malformations and the patient was followed arrows) with intervening luminal dilatation (black-outlined
periodically with MRA. MRI FLAIR imaging (a, right) arrows), a hallmark of vasculitis. Infectious thromboem-
demonstrates abnormal hyperintense signal and volume bolic disease may produce mycotic aneurysm that result in
loss within the cortex and underlying white matter of the vascular dilatation secondary to pseudoaneurysm formation
right parietal and far lateral occipital lobes consistent with but may be distinguished from vasculitis as it is typically
remote right MCA territory infarction. T1-weighted imaging eccentric, not fusiform as in this case, and often causes
(a, left) demonstrates gyriform increased signal intensity small-vessel occlusion. The ACA demonstrates short- and
which was stable for years likely secondary to dystrophic long-segment concentric stenoses (white arrows) and a
calcification or laminar necrosis. MRA (b)) demonstrates small pericallosal saccular aneurysm (white-outlined
700 A.J. Davis

compared with MRI. There are numerous case Spatial resolution and temporal resolution are
reports and series [15, 21, 30, 41, 44] of patients important performance benchmarks to consider
with clinical or biopsy-proven cerebral vasculitis when comparing the common angiographic tech-
with a positive MRI and a negative catheter angio- niques. Catheter angiography, a minimally inva-
gram, traditionally considered the imaging gold sive procedure, provides the highest spatial
standard. resolution (up to 0.2 mm) and fastest temporal
In summary, the literature confirms the impor- resolution (0.5–0.25 s for a typical study)
tant, if not essential, role of MRI in the diagnostic [11]. By comparison, the spatial resolution of
evaluation of the neurovasculitides. A negative modern multi-detector CTA, which is dependent
MRI may not definitively exclude cerebral vascu- on detector row thickness, is approximately
litis in a symptomatic patient, but it makes the 0.4–0.75 mm. CA provides detailed information
diagnosis extremely unlikely. Given a negative regarding hemodynamics that is generally absent
MRI, other etiologies for the patient’s symptoms from both CTA and MRA. However, the most
should be investigated. If strong clinical suspicion recent 320 row-detector CT technology does pro-
for vasculitis remains, dedicated vascular imaging vide limited hemodynamic information with a
including CTA, MRA, or CA should be temporal resolution of 1 s and a spatial resolution
performed. of 0.5 mm [48]. Indices for MRA spatial resolu-
tion are even less precise than these other
techniques.
Indirect Imaging Signs: Vessel Lumen
Catheter Angiography
Indirect imaging techniques in the investigation of CA is the oldest of the indirect imaging modalities
neurovasculitis characterize changes in the vessel currently utilized for the evaluation of the
lumen secondary to mural or perivascular inflam- neurovasculitides. It had been considered the
matory processes and include MRA, CTA, and gold standard for diagnosis but now plays a less
CA. While they do not provide direct evidence important role with the evolution of noninvasive
of inflammation, these studies convey salient vascular imaging techniques that provide similar
information about the pathologic process causing and, in some circumstances, equivalent
ischemia, infarction, and hemorrhage. The infor- information.
mation is crucial to the diagnosis, risk stratifica- The hallmark of vasculitis is multifocal lumi-
tion, and management of these patients. While nal narrowing, vascular contour irregularity, and
debate exists as to the sensitivity and specificity vascular dilatation occurring within multiple ves-
of each of these techniques, their utilization sels and multiple vascular territories. The classic
remains essential to the diagnosis of cerebral imaging finding is segmental luminal narrowing
vasculitis. followed by vessel dilatation, an appearance

Fig. 7 (continued) arrow). This constellation of radio- ICA, and cervical ICA segments (white arrows). The bilat-
graphic findings moved the diagnosis from congenital eral ACAs are occluded (open white arrows) and the left
aneurysm to HIV vasculitis. The patient presented one MCA is now markedly stenotic (white arrowhead). Find-
year later with right hemiparesis. MRI now demonstrates ings are consistent with disease progression and luminal
extensive abnormal FLAIR hyperintensity within the left thrombosis. CTA (g, volume-rendered imaging) demon-
basal ganglia and left frontal lobe cortex and underlying strates extensive collateral circulation to the ACA (open
white matter, with restricted diffusion (d left and right white arrows) from the right PCA via cortical
respectively). Findings are consistent with left MCA and leptomeningeal branches and posterior choroidal branches
ACA acute infarctions. CTA (e, maximum intensity pro- (white arrows) (Case courtesy of Adam Davis, MD,
jection, AP on the left and oblique on the right) now New York University Langone Medical Center, New York)
demonstrates occlusion of the skull base, left petrous
31 Imaging of the Neurovasculitides 701

Fig. 8 Primary angiitis of the central nervous system. PACNS, as opposed to the more irregular appearance asso-
74-year-old male with hypertension and no other signifi- ciated with atherosclerotic disease. Branch points are not
cant medical problems. Recurrent cerebral infarctions. involved. There is no abrupt distal vessel occlusion as
Systemic evaluation including CSF analysis was negative. might be expected with thromboembolic disease (Case
CA (a and b) reveals multifocal long- and short-segment courtesy of Adam Davis, MD, New York University
stenoses. Note the concentric appearance and relatively Langone Medical Center, New York)
smooth contour of the vessel lumen consistent with

referred to as “string of beads” (Figs. 3c and 7b, the ICA bulb, ICA petrocavernous junction, and
c, d). Unfortunately, vascular dilatation is an ICA cavernous segment. This helps to differenti-
inconstant finding. More typically, angiography ate vasculitis from atherosclerosis which com-
of vasculitis reveals multifocal stenoses with monly occurs at these locations. Multifocal
intervening vessel ectasia or normal luminal stenosis and luminal irregularity within the same
diameter (Figs. 5e, and 8a, b). Luminal contour vessel segment with intervening normal vessel
may be smooth (Figs. 5e, and 8a, b) or irregular contour favor vasculitis over atherosclerosis. Sim-
(Fig. 2c, d). Stenoses are classically short segment ilarly, isolated stenoses within separate vascular
and discrete, but occasionally may be more territories and otherwise normal vessel contours
elongated. (Figs. 5c, e, and 8a, b) are an unusual appearance
The size of the vessels affected and the distri- for atherosclerosis and favor vasculitis. It is
bution of lesions within each of the vessels vary important to remember that discrete lesions within
with the etiology of the neurovasculitis and help to multiple vascular territories may occur secondary
differentiate them. Large-vessel extracranial to multi-embolic disease, although emboli are
narrowing is typically long segment and undulat- typically more obstructive than stenotic and tend
ing when associated with TA (Fig. 9) and GCA not to occur in tandem within the same vessel as
(Fig. 1). Intracranial skull base large-vessel and does vasculitis.
cortical medium-vessel involvements are more There are some attributes which help to distin-
typical of the systemic primary vasculitides such guish medium-vessel disease secondary to sys-
as the ANCA positive diseases and the autoim- temic vasculitis and autoimmune disease from
mune diseases such as SLE and PACNS. Intracra- PACNS. Abrupt vascular truncation and occlu-
nial luminal manifestations tend to be more sion are typically seen with medium- and small-
variable than extracranial disease. vessel autoimmune vasculitides, but less so for
PACNS does not have a predilection for vessel PACNS. Microaneurysmal formations, likely
branch points in contradistinction to the autoim- pseudoaneurysms, are more commonly associated
mune inflammatory diseases [49] and atheroscle- with autoimmune inflammatory diseases, particu-
rosis. Luminal narrowing secondary to vasculitis larly polyarteritis nodosa (Fig. 10a–d) and in the
is typically not at regions of laminar flow disrup- author’s experience with drug-induced vasculitis,
tion or regions of high shear stress, which includes SLE and infectious vasculitis. Microaneurysmal
702 A.J. Davis

Fig. 9 Takayasu arteritis and collateral circulation. Case aortic arch. The brachiocephalic, subclavian, common
1: 25-year-old Indian female with long-standing TA and carotid, and vertebral arteries are among the most fre-
syncopal episodes. CA and CTA (a and b) demonstrate quently diseased in TA. The vertebral arteries are opacified
complete occlusion of the great vessels arising from the by collateral circulation (a, black arrows). Note the
31 Imaging of the Neurovasculitides 703

formation is uncommon with PACNS; none were [15]. This is an important benchmark since imag-
seen in an angiographic series reported by Cloft ing is essential to the diagnosis of PACNS because
et al. [50]. there are no systemic laboratory tests or clinical
Vessel dissection is a rare and serious event and signs or symptoms to otherwise confirm the diag-
may be associated with the transmural necrotizing nosis (Figs. 5, and 8).
vasculopathies including polyarteritis nodosa Additionally, PACNS and other autoimmune
(Fig. 10b, c, d). It is an unusual complication of vasculitides may present with a predominant
extracranial GCA (Fig. 1c). small-vessel involvement that is beneath the res-
When stenoses are severe or vascular occlusion olution of CA. Many of these patients are symp-
is present, typically within the large and medium tomatic, with a positive MRI but negative
vessels, collateral circulation should be investi- angiography. Multiple studies have confirmed
gated (Fig. 9a–f). Abnormal cerebral hemody- the poor correlation between findings on MRI
namics, typically characterized by slow and CA [15, 30, 50]. Pomper et al. noted that
antegrade flow, diminished distal luminal size only 65 % of lesions detected by MRI had a CA
(Fig. 9e, f), or prolonged circulation time, is com- correlate and only 44 % of lesions detected by
monly described. When present, it may be addi- angiography were evident on MRI. Similarly, in a
tionally quantified with CT or MR perfusion series of patients with PACNS, of 41 territories
imaging. involved by MRI, approximately 15 % were nor-
Despite the long experience with CA and the mal on angiography and conversely within 50 vas-
widespread notion that it represents the “gold cular territories involved by CA, 34 % were
standard” of diagnostic imaging, the utility of normal on MRI.
CA has been questioned. Numerous case reports The utility of CA is important to consider as
and studies describe patients with clinical, MRI, these studies carry risk, with reported permanent
or biopsy-proven cerebral vasculitis and negative and transient neurologic morbidities of 1 % and
angiography. The sensitivity of CA for PACNS 10 %, respectively, during the evaluation for vas-
has been reported to be from 40 % to 100 % culitis [51]. In the author’s experience, intrave-
[18, 21, 27, 28, 30, 44, 46, 49, 50] depending nous corticosteroids prior to the procedure may
upon whether clinical, radiographic, or histologic help reduce complications. The benefit of the
criteria are utilized to ultimately confirm the diag- study needs to be carefully considered with regard
nosis. For example, the specificity of CA for mak- to the potential harm. Nonetheless, when clinical
ing the diagnosis of PACNS as compared with suspicion remains high and noninvasive imaging
histology is reported to be no higher than 37 % is inconclusive, the high spatial resolution of CA

Fig. 9 (continued) residual stumps of the brachiocephalic, (white arrows) to the anterior spinal artery (white arrow-
common carotid, and left subclavian arteries (b, black heads). Case 2: 46-year-old Indian female with worsening
arrows). CA (c) demonstrates extensive collaterals to the dizziness. CTA demonstrates isolated involvement of the
cephalic circulation via the intercostal arteries (black left common carotid artery, beginning at the origin and
arrows) to the deep cervical and vertebral artery (black extending the length of the vessel. Note the severe contin-
arrowheads). The distal deep cervical branches (d, black uous long-segment luminal narrowing characteristic of the
arrows) opacify the occipital artery (black arrowhead) disease (g, left, maximum intensity projection; middle,
which via retrograde flow fills the ICA (black-outlined volume-rendered image, white-outlined arrows). The
arrows). CTA (e) documents the same collateral circulation internal and external carotid arteries (g, right, volume-
although flow dynamics are absent (deep cervical artery, rendered image, black-outlined arrows) are diminished in
white arrows; occipital artery, white arrowheads; ICA, caliber but with an otherwise smooth lumen and normal
white-outlined arrows). Even very small vasculature that morphology, likely resulting more from decreased flow
provides clinically significant collateral circulation can be than from direct disease involvement (Case courtesy of
demonstrated by CTA (f) including the ascending cervical Adam Davis, MD, New York University Langone Medical
artery to the vertebral artery to a spinal radicular artery Center, New York)
704 A.J. Davis

Fig. 10 Polyarteritis
nodosa. 43-year-old man
with known polyarteritis
nodosa presents with neck
swelling and bruit. CA of
the celiac axis performed
5 years prior to presentation
(a) demonstrates multifocal
luminal narrowing (black
arrows) and aneurysmal
dilatation (black
arrowhead). Both MRA
and CTA (b, c) demonstrate
a collection of prominent,
tortuous vasculature at the
posterior cervical space
enveloping the distal left
vertebral artery. Prominent
draining veins are noted
(white arrows). CA of the
left vertebral artery (d,
black arrow) reveals the site
of arteriovenous shunting
(black arrowhead) into a
dilated, tortuous draining
vein confirming the
diagnosis of an
arteriovenous fistula. This
was likely due to vascular
necrosis or aneurysmal
rupture. Both CTA and CA
(e, left and right
respectively) demonstrate a
discrete region of luminal
irregularity and narrowing
within the ICA proximal
cervical segment (black
arrows). In isolation, this
might be confused with
fibromuscular dysplasia
(Case courtesy of Adam
Davis, MD, New York
University Langone
Medical Center, New York)
31 Imaging of the Neurovasculitides 705

continues to make it an important adjunct in the studies does not require contrast administration.
assessment of the neurovasculitides. It possesses the lowest spatial resolution of the
common vascular imaging modalities and is prone
CT Angiography to overestimation of stenosis secondary to dimin-
The utility of CTA has been well established for ished signal intensity from vessel tortuosity or
the large-vessel vasculitides, particularly TA slow flow. MRA reveals vascular findings that
(Fig. 9) and GCA. Common conventional angio- are similar to the other luminal imaging modalities
graphic findings including stenosis, occlusion, including intracranial stenoses and occlusions
dilatation, and luminal irregularity are well dem- (Fig. 2c). More subtle findings including intracra-
onstrated by CTA as are the distribution and nial vessel irregularity are more difficult to assess
severity of involvement [52]. The sensitivity and due to lower spatial resolution. Compared with
specificity of CTA for TA are 93–95 % and CA in a series of patients with suspected vasculitis
98–100 % respectively [53]. Furthermore, CTA (not all of whom were ultimately diagnosed with
provides direct imaging evidence of inflammatory the disease), MRA revealed a sensitivity and spec-
vasculopathies by detecting mural changes that ificity of 79 % and 87 % for diagnosing vasculitis
are not evident on CA (Fig. 11). This is particu- while CA was 95 % and 97 % [54]. In a separate
larly important in the early stages of the disease study of patients with various etiologies for
when mural inflammatory changes may precede neurovasculitis, MRA provided a sensitivity of
luminal contour changes. The role of CTA in the 92 %. Given the criteria that more than two ste-
medium-vessel intracranial vasculitides is not as noses are identified within at least two vascular
well defined. CTA is routinely utilized to assess territories, the false-negative rate of MRA
the vessels at the circle of Willis and the 2nd- and improves markedly, approaching that of CA [54]
3rd-order branches of the anterior, middle, and eliminating the need for further indirect vascular
posterior cerebral arteries. These segments are luminal imaging. Unfortunately, if MRA is nega-
frequently involved by vasculitis (Fig. 3c). Steno- tive or if these stringent criteria are not met, CA or
sis, dilatation, and occlusion are well seen, while CTA is still required.
luminal irregularity is less consistent, given the
limits of the resolution of the modality. CTA is a
non-dynamic study, and collateral flow informa- Direct Imaging Signs: Vessel Wall
tion is not easily discernable as with CA but can
still be inferred by the angioarchitecture (Fig. 7g) The literature clearly supports the utility of direct
of the opacified vessels. Overall, CTA is noninva- vessel investigation in the diagnosis of
sive, with high spatial resolution, and may be neurovasculitis since demonstrating vessel wall
obtained quickly at any time of day, prompting and/or perivascular inflammatory changes pro-
its frequent use as the initial imaging study for vides direct evidence of the disease. The sensitiv-
suspected neurovasculitis. The radiation dose ity and specificity depend not only upon the
penalty, generally slightly greater than a routine modality utilized but the etiology as well. Large-
head CT, should always be considered. This may vessel extracranial vascular disease is typically
dissuade the clinician from utilizing this modality more obvious than intracranial vasculopathy
with children and young adults or as a screening where the smaller vessel size and close proximity
examination for adults. of arteries and veins make investigation more
difficult.
MRA
MRA is a frequently utilized modality in the CT and CTA
assessment of extracranial and intracranial vascu- TA and GCA, both large-vessel extracranial gran-
lar diseases, owing to its distinct advantages as ulomatous transmural arteritides, demonstrate
compared with CA and CTA: it is noninvasive, vessel wall abnormalities with all imaging modal-
lacks ionizing radiation, and for intracranial ities (Fig. 11). CT readily identifies abnormal
706 A.J. Davis

Fig. 11 Large-vessel inflammatory disease with features of localized narrowing within the external carotid artery
both Takayasu arteritis and giant cell arteritis. 51-year-old branches (arrowheads) typically found with TA. CTA vol-
male from Mali with fever, chills, night sweats, neck swell- ume rendering (c) demonstrates narrowing of the superficial
ing, and jaw pain. CTA (a) demonstrates marked mural temporal artery (black arrow) as would be expected in giant
thickening with heterogeneous enhancement within the bilat- cell arteritis. Biopsy demonstrated internal elastic lamina
eral cervical vasculature, (left to right) left subclavian artery, fragmentation and microaneurysm formation although with-
left CCA, and left ICA. The black arrows indicate the outer out granulomatous change as would be expected for either
wall contour, normally a barely imperceptible margin, while disease – failing to provide a definitive diagnosis. Following
the white arrows indicate the enhancing, narrowed vessel prompt and aggressive treatment with corticosteroids, the
lumen. The appearance is consistent with Takayasu arteritis. mural thickening within the left subclavian artery, CCA,
CTA volume rendering (b) demonstrates long-segment and ICA resolved (d) (Case courtesy of Adam Davis, MD,
smooth narrowing of the left ICA (black arrow) with more New York University Langone Medical Center, New York)
31 Imaging of the Neurovasculitides 707

mural thickening as defined by a thickness greater signal intensity changes are less sensitive than
than 1 mm in 93 % of patients with clinical evi- enhancement. Similar to CT, MRI provides
dence of TA [52]. Seventy-three percent of these radiopathologic correlation. Utilizing both mural
patients demonstrate changes within the morphology and signal intensity, these same
cervicocerebral vessels, most commonly the authors identified six distinct MRI patterns that
aorta, brachiocephalic artery, and common carotid likely correlate with histopathologic inflamma-
artery where wall thickening of up to 10 mm is tory changes, vascularization, and scarring/
observed. CA, once considered the gold standard fibrosis.
for detecting abnormal vasculature, may be nega- High-resolution MRI evaluation of extracra-
tive in the early stages of TA when mural abnor- nial vessel walls for inflammatory changes can
malities are present [52, 55]. This is an important play an important role in the diagnosis and
consideration as the disease is most effectively assessment of patients with suspected GCA.
treated with immunosuppressive therapy during Utilizing high-resolution, gadolinium-enhanced,
the earliest phase of the illness; a time when CA fat-saturation, T1 spin-echo technique, Bley
may be nondiagnostic. et al. [57] stratified MRI findings based on the
CTA is an excellent option in these circum- severity of wall thickening and prominence of
stances as it provides not only information regard- mural enhancement within the frontal and parietal
ing luminal abnormalities, including stenosis, superficial temporal arteries and occipital arteries.
occlusion, aneurysmal dilatation, and contour They demonstrated 81 % sensitivity and 97 %
irregularities similar to CA, but direct diagnostic specificity of MRI as compared with the final
findings including wall thickening, calcification, rheumatologists’ diagnosis based on clinical
and abnormal enhancement [52, 55]. While not exam, laboratory studies, and/or biopsy results.
specific to the cerebrovascular circulation, Some positive MRI results were biopsy negative,
Yamada et al. [53] demonstrated thoracic CTA to although the final diagnosis of GCA was con-
have a sensitivity and specificity of 95 % and firmed clinically. This may be attributed to the
100 %, respectively, for the diagnosis of TA. The intermittent vessel involvements (“skip lesions”)
utility of direct imaging is well documented by that occur with GCA (Fig. 1b), leading to false-
Park et al. [55] who correlate the vessel wall CT negative biopsy results when a normal segment of
abnormalities with the histopathologic findings. artery is surgically sampled. MRI is able to assess
The authors demonstrated that heterogeneous the entire length of the vessel, leading to a more
mural enhancement and an inner concentric low confident diagnosis.
attenuation ring which enhances at delayed imag- Obtaining clinical history regarding immuno-
ing correlate with the acute-phase histopathologic suppressive therapy is mandatory when imaging
findings of tunica media, vascularization, and inti- patients with suspected neurovasculitis. When
mal inflammatory swelling. patients are imaged following treatment with cor-
ticosteroids, false-negative MRI results may
MRI occur. Bley et al. [57] report that if only GCA
MRI provides similar and in some cases more patients who received treatment with steroids for
sensitive information than CT regarding vessel less than 10 days are considered, sensitivity
wall changes, particularly in those neurovas- increased from 81 % to 85 %. For those receiving
culitides affecting the extracranial vasculature. In steroids for more than 10 days, sensitivity falls to
patients with TA, mural thickening and enhance- 33 %. Therefore, radiologists should question
ment of the aorta, as compared with the myocar- patients regarding steroid therapy since patients
dium as a reference, correlate significantly with typically present for imaging days to weeks after
clinical disease activity as defined by erythrocyte seeing the referring physician. Vigilance for false-
sedimentation rate and C-reactive protein [56]. T2 negative tests is important, although it is
708 A.J. Davis

reassuring to know that MRI still provides sensi- 89 % for this disease [78]. Greater diagnostic
tive and specific information if imaging is sensitivity (80 %) and specificity (89 %) from
performed within days following the initiation of FDG-PET in cases of GCA have been obtained
immunosuppressive treatment. [59]. The specificity of FDG-PET is degraded by
MRI provides direct evidence of intracranial the presence of atherosclerosis as active inflam-
vasculitis [10, 36]. Findings include asymmetry matory plaques may produce false-positive find-
and thickening of the vessel wall, an eccentric or ings [60] and nuclear medicine studies should be
narrowed lumen, and mural enhancement. Thin- interpreted with caution in the elderly.
sliced images and flow saturation technique are The role of FDG-PET for monitoring disease
mandatory since images may be marred by an activity in patients with known vasculitis is still
artifact from laminar flow adjacent to the vessel not well defined. Some patients deemed inactive
wall or by periarterial venous enhancement. by clinical criteria have biopsy-proven active
Enhancement may be limited to the vessel wall inflammation [61]. Studies indicate that
or extend into the adjacent leptomeninges. Kuker FDG-PET may be a sensitive and helpful diag-
et al. [36] reported on 27 patients with nostic study for identifying these patients with
neurovasculitis secondary to varying etiologies subclinical active disease [66]. Eighty-three per-
in which 25 patients had mural thickening and cent of patients with biopsy-proven GCA had
23 demonstrated abnormal enhancement, con- positive FDG-PET studies [62]. The sensitivity
firmed in more than one plane of imaging within and specificity of FDG-PET to identify active
the same study. Approximately, the same number disease as compared with both clinical signs and
of patients demonstrated abnormal stenosis with laboratory criteria are 100 % and 89 % [63]. Inter-
MRA or abnormal parenchymal T2 signal inten- estingly, one of two patients considered false pos-
sity with MRI, although these findings were rela- itive due to lack of clinical and laboratory
tively nonspecific. Kuker [36] contends that for evidence went on to develop active disease within
intracranial large-vessel disease, the greater spec- several weeks, suggesting that subclinical disease
ificity of direct vascular wall inflammatory may be uncovered with nuclear medicine studies.
changes provides more convincing evidence than The issue is clouded by the fact that these patients
secondary imaging findings and perhaps exceeds are treated with immunosuppressive agents that
that of biopsy. may halt overt clinical manifestations and prompt
false-negative nuclear medicine studies, despite
Nuclear Medicine the presence of indolent subclinical disease activ-
Nuclear medicine evaluation of neurovasculitis ity. This is similar to the difficulties with MRI in
remains promising but problematic. Conceptually, detecting mural enhancement following immuno-
the ability to monitor metabolic activity within the suppressive therapy discussed earlier. Nonethe-
vessel wall should be a good indicator of inflam- less, nearly all practitioners recommend the
matory activity. 18F-fluorodeoxyglucose positron utilization of nuclear medicine studies to assess
emission tomography (FDG-PET) has been the disease progression, treatment efficacy, and dis-
most well-studied radionuclide for this indication, ease recurrence particularly when there is uncer-
particularly large-vessel diseases including TA tainty in the clinical assessment as defined by
and GCA. A meta-analysis of the literature [58] physical signs and symptoms or laboratory
demonstrated a wide variability of diagnostic sen- markers.
sitivities for TA ranging from 28 % to 100 %,
while the range of specificity was 50–100 %. Ultrasound
The pooled analysis nonetheless indicates modest Ultrasound (US), specifically color duplex imag-
results with a diagnostic sensitivity and specificity ing, may be a helpful modality in the diagnosis of
of 70 % and 77 % respectively. FDG-PET CT vasculitis by providing direct imaging evaluation
provides superior anatomic localization and of the vessel wall. It has been most extensively
improved sensitivity and specificity of 91 % and studied in the more common extracranial large
31 Imaging of the Neurovasculitides 709

artery vasculitides, TA and GCA. It is an appeal- disappeared at a mean of 16 days. Stenoses and
ing technique as it is noninvasive, without ioniz- occlusions are commonly seen in all symptomatic
ing radiation; can be easily administered; and may patients, and while significantly more common in
be repeated frequently to follow disease progres- patients with GCA (80 %) than those without, it is
sion or treatment response. Additionally, US pro- not as specific a finding as the halo. Similar find-
vides a high-resolution imaging evaluation as ings are present in the occipital arteries [69]
compared with MRI, detecting wall thickness dif- although the sensitivity is less when compared
ferences with a resolution of 0.1 mm [64]. with the superficial temporal arteries. Nonethe-
Concentric homogeneous mural thickening, less, it is a useful exam for symptomatic patients
stenosis, and occlusion of the aorta and presenting with nuchal pain, occipital headache,
brachiocephalic branches are typical US findings or occipital scalp tenderness when occipital artery
of TA [60, 65, 66] which may be differentiated involvement may be the only imaging manifesta-
from atherosclerotic disease by the absence of tion of the disease. Specificity is increased when
plaque formation, concentric morphology, long more than 3 arterial segments are involved. A
segment involvement, and location. US reveals meta-analysis of the literature [70] demonstrated
abnormalities more frequently in the common that if all US findings are considered – halo sign,
carotid artery than the internal carotid artery with stenosis, and occlusion – the sensitivity and spec-
vessel wall thickness greater than that of control ificity are 88 % and 78 %, respectively, as com-
subjects. In patients with TA, the common carotid pared with biopsy. The specificity increases to
artery wall thickness is greater than that of the 82 % when only the halo sign is considered.
carotid bulb, the reverse of normal control sub-
jects. US demonstrates subtle mural changes char-
acterized by a homogenous, circumferential, Conclusion
mid-echoic wall thickening within the subclavian
and carotid arteries in the early stages of the dis- The data for parenchymal, luminal, and vessel
ease – before abnormalities may be detected by wall imaging may at first glance suggest that the
CA [64]. Common carotid artery intima–media evaluation of vasculitis is a daunting task, partic-
thickness ratio is increased in patients with TA ularly for PACNS and the primary autoimmune
as compared with normal control patients and vasculitides, but this is true only if each imaging
the degree correlates with disease activity modality is considered in isolation. Diagnosis of
[67]. The sensitivity and specificity of this finding neurovasculitis should be considered on a
as compared with clinical criteria are 82 % and per-patient basis and not a per-territory or
70 %. per-modality basis. The largest single series of
Ultrasound has been well studied in patients PACNS [28] demonstrated that 77 % of CT,
with GCA. The wall diameter of the superficial 97 % of MRI, 59 % of MRA, and 90 % of
temporal artery (common, frontal, and parietal conventional angiographic studies were ulti-
divisions) is significantly greater in patients with mately positive. By comparison, only 62 % of
GCA than in symptomatic patients without the the brain biopsies were positive. Consequently,
disease or asymptomatic age-matched controls when all imaging modalities including those that
[68]. A hypoechoic halo surrounding a patent provide parenchymal, luminal, and mural evalua-
vessel lumen was found in 73 % of biopsy-proven tion are brought to bear on a given patient with
vasculitis patients but not in any symptomatic suspected vasculitis, the entire constellation of
patients without GCA or asymptomatic controls, findings typically brings clarity to the situation.
indicating that it may be a very specific imaging While not specific, few patients are missed by
finding. Interestingly, histopathologic findings imaging evaluation. When combined with the
characterized by mural cellular infiltrate did not clinical and laboratory results as well as biopsy,
correlate with the hypoechoic halo which has been the diagnosis becomes more assured even in the
attributed to edema. In all treated patients, the halo most difficult of clinical scenarios [15].
710 A.J. Davis

Mimics consistent with inflammatory vasculopathy. Mural


enhancement by CT or MRI may definitively dif-
There are numerous diseases that present with ferentiate vasculitis. A positive PET-CT will be
clinical manifestations and imaging findings that helpful but it should always be remembered that
are easily confused with and difficult to distin- atherosclerosis may be falsely positive.
guish from neurovasculitis. Any vasculopathy
resulting in extracranial or intracranial luminal
stenosis, occlusion, or irregularity and associated Tumor
with cerebral or spinal cord ischemia, hemor-
rhage, or inflammation may be suspect. Marsh Neoplastic involvement of the vasculature is an
et al. [2] report on 5 patients referred for treatment important entity to exclude. Tumor may directly
of PACNS who were ultimately found to have invade the vessel wall or propagate within the
intracranial atherosclerosis, B-cell lymphoma, lumen and may be difficult to differentiate from
cerebral amyloid angiopathy, and RCVS. Neel vasculitis.
et al. [71] report that RCVS, intracranial athero- Intravascular B-cell lymphoma is a rare type of
sclerosis, infection, malignancy, and embolism extranodal large B-cell lymphoma typically
may be confused with PACNS. More frequently, occurring in older patients which may present
a patient will present with ischemia secondary to a with CNS manifestations and constitutional
common noninflammatory vasculopathy (e.g., symptoms mimicking a systemic vasculitis
atherosclerosis) and suffer from an incidental sys- [72]. CNS symptoms are typically secondary to
temic inflammatory disease (e.g., rheumatoid ischemia. Morphologically, the tumor growth is
arthritis). The combination of cerebral ischemia, restricted to the lumen of small vessels and is
vasculopathy, and systemic inflammatory labora- often widely disseminated at presentation. Biopsy
tory abnormalities may mistakenly lead to a diag- is required for diagnosis. Secondary to the
nosis of vasculitis. These cases may be difficult to nonspecificity of the symptoms and the elusive
clarify. Careful search for direct vascular imaging intravascular location of the disease, the diagnosis
signs of mural inflammation or a characteristic is frequently made at autopsy. Radiologic findings
pattern of vasculitic involvement may help. in the brain (Fig. 12) are strikingly similar to
Sometimes biopsy is required to differentiate. vasculitis [72] and may be partially reversible
with treatment.
Lymphomatoid granulomatosis is a rare
Atherosclerosis Epstein–Barr virus associated lymphoproli-
ferative disease characterized by the presence of
Atherosclerosis particularly when it is predomi- T cells and EBV-infected B cells that produce an
nantly or exclusively intracranial is the most com- angiocentric and angiodestructive pattern. It typi-
mon and sometimes most difficult disease to cally affects the lungs but has been associated
differentiate from vasculitis. Likewise, hyperten- with the central nervous system, skin, and kid-
sive vasculopathy of the medium and small neys. Similar to intravascular B-cell lymphoma,
intracranial vessels may cause confusion. Indirect the appearance may mimic vasculitis.
luminal imaging, whether MRA, CTA, or CA,
provides subtle differentiating clues. Atheroscle-
rotic plaque tends to occur at branch points and Reversible Cerebral Vasoconstrictive
is typically better defined, shorter in length, and Syndrome
more eccentric than the smooth, concentric, tapered
morphology and distal location of primary and Reversible cerebral vasoconstrictive syndrome
secondary vasculitides. The presence of mural cal- (RCVS) [73–75] is a disorder characterized by
cification is helpful as it is more commonly asso- severe acute onset (“thunderclap”) headache,
ciated with atherosclerosis. Venous involvement is peaking within 1–2 min, with or without focal
31 Imaging of the Neurovasculitides 711

Fig. 12 Intravascular B-cell lymphoma. 49-year-old male likely as there is no predilection for a single vascular terri-
with new onset memory difficulties. Patient was referred for tory and the distribution is sporadic, affecting both cortical
evaluation of demyelinating disease. MRI FLAIR and T2 and deep perforating vascular territories. An alternative
diffusion imaging (top and bottom respectively) demon- diagnosis of infection was considered as well. Systemic
strate extensive abnormal hyperintense signal within the evaluation and CSF analysis were unrevealing and concern
bilateral hemispheric white matter, cortex, and splenium of for PACNS was raised. CTA (c) reveals multifocal, discrete,
the corpus callosum (black arrows) without mass effect. A eccentric luminal narrowing and irregularity affecting the
focal area of cortical/leptomeningeal high attenuation on CT MCA and ACA distal vasculatures (white arrows). Normal
with corresponding post-gadolinium enhancement on MRI intervening vascular segments are noted. CA (d) confirms
T1-weighted imaging was noted (b left and right respec- multifocal luminal narrowing within the distal vasculature.
tively) and retrospectively represented a region of lympho- The proximal vessels and branch points are normal. There is
matous involvement. Other areas of abnormal CT no luminal dilatation. Suspicion for vasculitis was height-
attenuation and abnormal MRI signal intensity were ened. Brain biopsy revealed intravascular B-cell lymphoma.
non-enhancing (black-outlined arrow). Findings were not Tumor invasion is rare but must always be included in the
felt to be consistent with demyelinating disease. Restricted differential diagnosis when the neurovasculitides are con-
diffusion raised concern for multifocal, acute, and chronic sidered (Case courtesy of Adam Davis, MD, New York
ischemia. Thromboembolic disease was considered most University Langone Medical Center, New York)
712 A.J. Davis

Fig. 13 Reversible vasoconstrictive syndrome. 31-year- consistent with a PCA parieto-occipital artery distribution.
old female with history of schizophrenia treated with MRA at admission (c) demonstrates short-segment
multiple psychotropic medications including a selective multifocal narrowing within the distal bilateral vertebral
serotonin reuptake inhibitor (SSRI). The patient presented arteries, the basilar artery, and the bilateral MCA and PCA
with acute-onset severe headache and right hemiparesis, vasculatures (white arrows). CTA demonstrates moderate
slowly improving. MRI FLAIR imaging on presentation narrowing within the right PCA P2 segment and a more
(a) demonstrates multifocal abnormal hyperintense signal severe narrowing distally within the P3 parieto-occipital
within the bilateral hemispheric white matter, more prom- segment (d, left, black arrows). Mild narrowing is present
inent in the parietal and occipital lobes where it extends to within the ACA A1 segment (d, right, black arrow). CA
the cortex. T2 diffusion imaging (b) demonstrates confirms multifocal narrowing within the bilateral PCAs
restricted diffusion consistent with acute ischemia. The (e, black arrows). The systemic evaluation including lum-
white matter distribution within the left hemisphere strad- bar puncture was normal. The patient demonstrated rapid
dles the ACA, MCA, and PCA vascular territories, a and near-complete clinical improvement with only limited
“watershed” distribution. The cortical ischemia is steroid treatment. Follow-up CTA (e, at presentation on the
31 Imaging of the Neurovasculitides 713

neurologic symptoms or seizures. Vascular imag- for several weeks following the onset of headache,
ing demonstrates abnormal multifocal segmental indicating a progressive worsening of stenosis
vascular constriction and dilatation that resolve even when headache resolves. This finding corre-
completely within 3 months (Fig. 13). It is asso- lates with the somewhat delayed appearance of
ciated with cortical subarachnoid hemorrhage and ischemia. Angiography demonstrates diffuse,
ischemia – including TIA and infarction, intrapar- multisegmental, arterial narrowing in all patients,
enchymal hemorrhage, and posterior reversible typically with intervening regions of dilatation
encephalopathy syndrome. The abrupt headache (“string of bead”) or normal luminal diameter. It
is an essential component of the diagnosis and may be difficult or impossible to distinguish this
helps to differentiate the disease from inflamma- appearance from vasculitis, particularly PACNS,
tory vasculopathies, which typically have a more although the inflammatory conditions may some-
subacute onset of headache. The patient median times appear more isolated, asymmetric, and
age is 45 years old and it occurs in females more eccentric with areas of vessel occlusion. The hall-
than males. Some cases are spontaneous. Many mark of the disease is the complete remission of
occur in the postpartum period or are secondary to vascular narrowing within 3 months, an arbitrary
vasoactive substances, the most common being milestone but consistent with the literature.
cannabis, serotonin reuptake inhibitors, and CT and MRI reveal cortical subarachnoid hem-
nasal decongestants. Numerous other etiologies orrhage most frequently (22 %) followed by
are cited. The presence of neurologic deficits intraparenchymal hemorrhage (6 %) and infarc-
varies widely by study, but when permanent, tion, often with a vascular watershed distribution
they are due to infarction or hemorrhage. Hemor- (4 %). Bilateral symmetric FLAIR signal
rhage and posterior reversible encephalopathy hyperintensity (10 %), usually in a posterior cir-
tend to occur early, within days of the illness culation distribution or involving the occipital/
onset, while ischemia may be more delayed, temporal–parietal/posterior frontal lobe distribu-
occurring a week or so after the onset of headache. tion, may correlate with the associated posterior
Pathologically, it is differentiated from reversible encephalopathy.
neurovasculitis as the abnormal vasoconstriction
is not inflammatory and is presumed to be second-
ary to a loss of control of vasomotor tone. Summary
In one study of 67 patients with RCVS [74],
MRA revealed multifocal segmental arterial Neurovasculitis is a term used to describe a
narrowing in 88 % of patients at a mean of diverse spectrum of diseases characterized by
8 days following the onset of headache. It is inflammation of the blood vessels that may pro-
important to note that a small percentage of gress to ischemic injury of the central or periph-
patients did not demonstrate vascular narrowing eral nervous system. The overlapping clinical and
until two weeks post the onset of symptoms. Mean radiographic features and the complex pathophys-
flow by transcranial Doppler typically increases iology make it difficult to quickly or clearly

Fig. 13 (continued) left, 5-month follow-up on the right) imaging is a common appearance. RVCS is frequently
reveals complete resolution of the original findings. Note secondary to medication or drug use and SSRI have been
the normal caliber and contour of the vertebral, basilar, implicated. The resolution of vascular imaging abnormal-
superior cerebellar, and posterior cerebral arteries on the ities within 3 months is an essential criterion of the diag-
follow-up exam (white arrows). Severe (thunderclap) nosis of RVCS; it would be a very rare outcome with
headache is the most common presentation of RVCS; PACNS, the leading alternative diagnosis (Case courtesy
focal neurologic signs and symptoms are common. Infarc- of Adam Davis, MD, New York University Langone Med-
tion is more unusual. A posterior frontal, parieto-occipital, ical Center, New York)
or temporo-occipital junctional zone distribution on
714 A.J. Davis

identify neurovasculitis as the etiology of a involved, as well as the patient demographics,


patient’s symptoms. Nonetheless, simple yet com- clinical presentation, and clinical course, helps to
prehensive categorization schemes established by differentiate inflammatory from noninflammatory
consensus meetings and based upon the anatomy, vasculopathies.
pathophysiology, and imaging findings help to Imaging plays an essential role in the diagnosis
clarify the situation. For both the radiologist and of these diseases. All modalities contribute to the
the treating clinician, a systemic approach consid- analysis of neurovasculitis; conventional catheter
ering the triad of (1) organ systems involved, angiography is no longer the preeminent diagnos-
(2) laboratory results, and (3) vessel size and tic standard. The radiologist should be familiar
type affected simplifies the situation. with the findings of a given disease process using
Vessel size is particularly important for the all the available imaging modalities as the infor-
radiologist to consider as it broadly categorizes mation is complimentary. When all imaging find-
the neurovasculitides within the differential diag- ings are taken together, they provide a more
nosis. Current systemic classification systems fail comprehensive analysis and a more certain diag-
to adequately describe the distribution and pathol- nosis. Imaging may be divided into those studies
ogy of the neurovasculitides as they tend to disre- that investigate the vessel lumen, the vessel wall,
gard the unique characteristics of brain and the parenchyma. Parenchymal findings are the
vasculature. A more specific cerebrovascular clas- least specific but are usually necessary to detect the
sification is more helpful, which roughly divides presence of disease as well as follow progression
the anatomic involvement of the neurovas- or remission. Vessel lumen abnormalities are
culitides into the following three areas: highly suggestive for vasculitis when present but
are not specific and the sensitivity is low for the
1. Extracranial elastic, extracranial large vessel, small-vessel vasculitides. Demonstration of vessel
and intracranial skull base large-vessel wall or perivascular inflammatory change is the
involvement most specific and compelling radiographic find-
2. Intracranial medium-vessel involvement ing, although it is only inconsistently found likely
3. Intracranial small-vessel involvement due to the limitations of routine imaging protocols.
Nonetheless, direct findings of mural inflamma-
This approach is most helpful for the radiolo- tory change should be aggressively sought by the
gist who typically is able to separate the radio- radiologist. MRI and nuclear medicine imaging
graphic findings into broad vascular anatomic provide the most sensitive evaluation for vessel
categories, depending on the appearance of the wall inflammation although extracranial large elas-
vessels and the anatomic distribution of the imag- tic vessel inflammation is seen particularly well
ing abnormalities. Even so, there is considerable with CT/CTA.
anatomic overlap and the radiologic appearance Biopsy is frequently required, not only to con-
of the neurovasculitides may be similar. In these firm the diagnosis but, in half the positive cases, to
cases, clinical criteria help and the radiologist provide an alternative diagnosis. Imaging should
should consider the patient’s laboratory values or be used to guide the site of surgical biopsy.
the other organ systems involved as a means to Despite the uncommon occurrence of
narrow the differential diagnosis. neurovasculitis, it is important to be familiar
Imaging findings of neurovasculitis may be with these diseases. They play a particularly
similar to the more common noninflammatory important role in the differential diagnosis of
vasculopathies including atherosclerosis or stroke in young and middle-aged adults and typ-
uncommon vasculopathies, including reversible ically have devastating clinical consequences. If
cerebral vasoconstrictive syndrome. In this cir- diagnosed early, they may be effectively treated,
cumstance, consideration of the anatomic distri- relieving symptoms and avoiding permanent cen-
bution, laboratory values, and other organs tral and peripheral neurologic damage.
31 Imaging of the Neurovasculitides 715

References 20. Joseph FG, Scolding NJ (2002) Cerebral vasculitis: a


practical approach. Pract Neurol 2:80–93
1. Giannini C, Salvarani C, Hunder G, Brown RD (2012) 21. Neel A, Pangnoux C (2009) PACNS. Clin Exp
Primary central nervous system vasculitis: pathology Rheumatol 27(Suppl 52):S95–S107
and mechanisms. Acta Neuropathol 123:759–772 22. Lie JT (1997) Biopsy diagnosis of systemic vasculitis.
2. Marsh EB, Zeiler SR, Levy M, Llinas RH, Urrutia VC Bailliere Clin Rheumatol 11:219–236
(2012) Diagnosing CNS Vasculitis: The Case Against 23. Rossi CM, Comite G (2009) The clinical spectrum of
Empiric Treatment. Neurologist 18:233–238 the neurological involvement in vasculitides. J Neurol
3. Weyand CM, Goronzy JJ (2003) Giant-Cell Arteritis Sci 285:13–21
and Polymyalgia Rheumatica. Ann Intern Med 24. Adams HP (2014) Cerebral vasculitis. In: Biller J,
139:505–515 Ferro JM (eds) Neurologic aspects of systemic disease
4. Hotchi M (1992) Pathological studies on Takayasu part I. Handbook of clinical neurology. vol 119 (3rd
Arteritis. Heart Vessels Suppl 7:11–17 series). Elsevier BV
5. Watts R, Al-Taiar A, Mooney J, Scott D, Macgregor A 25. Kuker W (2007) Imaging of cerebral vasculitis. Int J
(2009) The epidemiology of Takayasu Arteritis in the Stroke 2:184–190
UK. Rheumatology 48:1008–1011 26. Gomes LJ (2010) The role of imaging in the diagnosis
6. Adams HP, Kapelle J, Biller J et al (1995) Ischemic of central nervous system vasculitis. Curr Allergy
stroke in young adults. Iowa Registry of Stroke in Asthma Rep 10:163–170
Young Adults. Arch Neurol 52:491–495 27. Zuccoli G, Pipitone N, Haldipur A, Brown RD Jr,
7. Gemmete JJ, Davagnanam I, Toma AK, Brew S, Hunder G, Salvarani C (2007) Imaging findings in
Ganesan V (2013) Arterial ischemic stroke in children. primary central nervous system vasculitis. Clin Exp
Neuroimaging Clin of N Am 23:781–798 Rheumatol 29(Suppl 64):S104–S109
8. Hunder GG, Arend WP, Bloch DA, Calabrese LH, 28. Salvarani C, Brown RD Jr, Calamia KT et al (2007)
Fauci AS, Fries JF et al (1990) The American College Primary central nervous system vasculitis: analysis of
of Rheumatology 1990 criteria for the classification of 101 patients. Ann Neurol 62:442–451
vasculitis. Introduction. Arthritis Rheum 29. Wasserman BA, Stone JH, Hellman PMG (2001) Reli-
33:1065–1067 ability of normal findings on MR imaging for exclud-
9. Jennette JC, Falk RJ, Andrassy K, Bacon PA, Churg J, ing the diagnosis of vasculitis of the central nervous
Gross WL et al (1994) Nomenclature of systemic vas- system. AJR Am J Roentgenol 177:455–459
culitides. Proposal of an international consensus con- 30. Pomper M, Miller T, Stone J, Tidmore W, Hellman D
ference. Arthritis Rheum 37:187–192 (1999) CNS vasculitis in autoimmune disease: MR
10. Kuker W (2007) Cerebral vasculitis: imaging signs imaging findings and correlation with angiography.
revisited. Neuroradiology 49:471–479 AJNR Am J Neuroradiol 20:75–85
11. Kaufman TJ, Kallmes DF (2008) Diagnostic cerebral 31. Koçer N, Islak C, Siva A, Saip S, Akman C,
angiography: archaic and complication-prone or here Kantarci O, Hamuryudan V (1999) CNS involvement
to stay for another 80 years? AJR Am J Roentgenol in Neuro-Behçet syndrome: an MR study. AJNR Am J
190:1435–1437 Neuroradiol 20:1015–1024
12. Hajj-Ali RA, Calabrese LH (2013) PACNS. 32. Akman-Demir G, Serdaroglu P, Tasçi B (1999) Clinical
Autoimmun Rev 12:463–466 patterns of neurological involvement in Behçet’s dis-
13. Lie JT (1996) Vasculitis associated with infectious ease: evaluation of 200 patients. Brain 122:2171–2182
agents. Curr Opin Rheumat 8:26–29 33. Spitzer C, Mull M, Rohde V, Kosinski CM (2005)
14. Calabrese LH, Mallek JA (1988) PACNS. Report of Non-traumatic cortical subarachnoid haemorrhage:
8 new cases, review of the literature and proposal for diagnostic work-up and etiological background. Neu-
diagnostic criteria. Medicine 67:20–39 roradiology 47:525–531
15. Chu CT, Gray L, Goldstein LB, Hulette CM (1998) 34. Ay H, Sahin G, Saatci I, Soylemezoglu F, Saribas O
Diagnosis of intracranial vasculitis: a multi-disciplinary (2002) PACNS and silent cortical hemorrhages. AJNR
approach. J Neuropathol Exp Neurol 57:30–38 Am J Neuroradiol 23:1561–1563
16. Miller DV, Salvarani C, Hunder GG, Brown RD, Parisi 35. Salvarani C, Brown RD Jr, Gene G (2012) Adult pri-
JE, Christianson TJ, Giannini C (2009) Biopsy findings mary central nervous system vasculitis. Lancet
in PACNS. Am J Surg Pathol 33:35–43 380:767–777
17. Hankey G (1991) Isolated angiitis/angiopathy of the 36. K€uker W, Gaertner S, Nägele T, Dopfer C,
CNS. Prospective diagnostic and therapeutic experi- Schöninge M, Jens Fiehler J, Rothwell PM, Herrlinger
ence. Cerebrovasc Dis 1:2–15 U (2008) Vessel wall contrast enhancement: a diagnos-
18. Alrawi A, Trobe J, Blaivas M, Musch DC (1999) Brain tic sign of cerebral vasculitis. Cerebrovasc Dis
biopsy in PACNS. Neurology 53:858–860 26:23–29
19. Moharir M, Shroff M, Benseler S (2013) Childhood 37. Kumar RS, Singh A, Rathore C, Kesa-Vades C (2010)
central nervous system vasculitis. Neuroimaging Clin Primary angiitis of central nervous system: tumor-like
N Am 23:293–308 lesion. Neurol India 58:147–149
716 A.J. Davis

38. Panchal NJ, Niku S, Imbesi SG (2005) Lymphocytic 54. Demaerel P, De Ruyter N, Maes F, Velghe B, Wilms G
vasculitis mimicking aggressive multifocal cerebral (2004) Magnetic resonance angiography in suspected
neoplasm: MR imaging and MR spectroscopic appear- cerebral vasculitis. Eur Radiol 14:1005–1012
ance. AJNR Am J Neuroradiol 26:642–645 55. Park JH (1996) Conventional and CT angiographic
39. Muccio CF, DiBlasi A, Espositio G, Brunese L, diagnosis of Takayasu’s arteritis. IntJ Cardiol 54:
D’Arcos F, Caranc F (2013) Perfusion and spectros- SI65–SI71
copy magnetic resonance imaging in a case of lympho- 56. Choe YH, Han BK, Koh EM, Kim DK, Do YS, Lee
cytic vasculitis mimicking brain tumor. Pol J Radiol WR (2000) Takayasu’s arteritis: assessment of disease
78:66–69 activity with contrast-enhanced MR imaging. AJR Am
40. Qu SB, Khan SW, Hua L (2009) Primary central ner- J Roentgenol 175:505–551
vous system vasculitis mimicking brain tumour: case 57. Bley TA, Uhl M, Carew J, Markl M, Schmidt D, Peter
report and literature review. Rheumatol Int 30:127–134 HH, Lander M, Wieben M (2007) Diagnostic value of
41. Park MS, Marlin AE, Gaskill SJ (2013) Angiography- high-resolution MR imaging in giant cell arteritis.
negative PACNS in childhood. J Neurosurg Pediatr. AJNR Am J Neuroradiol 28:1722–1727
doi:10.3171/2013.9.PEDS13159. Published online 58. Cheng Y, Lv N, Wang Z, Chen B, Dang A (2013)
25 Oct 2013 18F-FDG-PET in assessing disease activity in
42. Sundgren PC, Jennings J, Attwood JT et al (2005) MRI Takayasu’s arteritis: a meta-analysis. Clin Exp
and 2D-CSI MR spectroscopy of the brain in the eval- Rheumatol 31:S22–S27
uation of patients with acute onset of neuropsychiatric 59. Besson FL, Arienti JJ, Bienvenu B et al (2011) Diag-
systemic lupus erythematosus. Neuroradiology nostic performance of F18 fluorodeoxyglucose posi-
47:76–585 tron emission tomography in giant cell arteritis: a
43. Woolfenden AR, Tong DC, Marks MP, Ali AO, Albers systematic review and meta-analysis. Eur J Nucl Med
GW (1998) Angiographically defined PACNS: is it Mol Imaging 38:1764–1772
really benign? Neurology 51:183–188 60. Amezcua-Guerra LM, Pineda C (2007) Imaging stud-
44. Harris KG, Tran DD, Sickels WJ, Cornell SH, Yuh WT ies in the diagnosis and management of vasculitis. Curr
(1994) Diagnosing intracranial vasculitis: the roles of Rheumatol Rep 9:320–327
MR and angiography. AJNR Am J Neuroradiol 61. Direskeneli H, Aydın SZ, Merkel PA (2011) Assess-
15:317–330 ment of disease activity and progression in Takayasu’s
45. Greenan TJ, Grossman RI, Goldberg HI (1992) Cere- arteritis. Clin Exp Rheumatol 29:S86–S91
bral vasculitis: MR imaging and angiographic correla- 62. Blockmans D, DeCeuninck L, Vander-Schueren S,
tion. Radiology 182:65–72 Knockaert D, Mortel-Mans L, Bobbaers H (2006)
46. Duna GF, Calabrese LH (1995) Limitations of invasive Repetitive 18F-fluorodeoxyglucose positron emission
modalities in the diagnosis of PACNS. J Rheumatol tomography in giant cell arteritis: a prospective study
22:662–667 of 35 patients. Arthritis Rheum 55:131–137
47. Imbesi SG (1999) Diffuse cerebral vasculitis with nor- 63. Karapolat I, Kalfa M, Keser G, Yalçin M, Inal V,
mal results on brain MR imaging. AJR Am J Kumanlioglu K, Pirildar T, Aksu K (2013) Comparison
Roentgenol 173:1494–1496 of 18F-FDG PET/CT findings with current clinical
48. Brouwer PA, Bosman T, van Walderveen MAA, disease status in patients with Takayasu’s arteritis.
Krings T, Leroux AA, Willems PWA (2010) Dynamic Clin Exp Rheumatol 31:S15–S21
320-section CTA in cranial arteriovenous shunting 64. Schmidt WA, Nerenheim A, Seipelt E, Poehls C,
lesions. AJNR Am J Neuroradiol 31:767–770 Gromnica-Ihle E (2002) Diagnosis of early Takayasu’s
49. Birnbaum J, Hellman DB (2009) PACNS. Arch Neurol arteritis with sonography. Rheumatology 41:496–502
66:704–709 65. Gotway M, Araoz PA, Macedo TA, Stanson AW
50. Cloft HJ, Phillips CD, Dix JE, McNulty BC, Zargado et al (2004) Imaging findings in Takayasu’s arteritis.
MT, Kallmes DF (1999) Correlation of angiography AJR Am J Roentgenol 184:1945–1950
and MR imaging in cerebral vasculitis. Acta Radiol 66. Kissin EY, Merkel PA (2004) Diagnostic imaging in
40:83–87 Takayasu’s arteritis. Curr Opin Rheumatol 16:31–37
51. Hellmann DB, Roubenoff R, Healy RA, Wang H 67. Seth S, Goyal NK, Jagia P, Gulati G, Karthikeyan G,
(1992) Central nervous system angiography: safety Sharma S, Talwar KK (2006) Carotid intima–medial
and predictors of a positive result in 125 consecutive thickness as a marker of disease activity in Takayasu’s
patients evaluated for possible vasculitis. J Rheumatol arteritis. Int J Cardiol 108:385–390
19:568–572 68. Schmidt WA, Kraft HE, Vorpahl K, Volker L, Erika J,
52. Khandelwal N, Kalra N, Garg MK, Kang M, Lal A, Gromnica-Ihle EJ (1997) Color duplex ultrasonogra-
Jain S, Suri S (2009) Multidetector CT angiography in phy in the diagnosis of temporal arteritis. New Engl J
Takayasu’s arteritis. Eur J Radiol 77:369–374 Med 337:1336–1342
53. Yamada I, Nakagawa T, Himeno Y, Numano F, 69. Pfadenhauer K, Weber H (2003) GCA of the occipital
Shibuya H (1998) Takayasu’s arteritis: evaluation of arteries: a prospective color coded duplex sonography
the thoracic aorta with CTA. Radiology 209:103–109 study in 78 patients. J Neurol 250:844–849
31 Imaging of the Neurovasculitides 717

70. Karassa FB, Matsagas MI, Schmidt WA, Ioannidis JP syndrome. A prospective series of 67 patients. Brain
(2005) Meta-analysis: test performance of ultrasonog- 130:3091–3101
raphy for giant cell arteritis. Ann Intern Med 75. Ducros A, Bousser MG (2009) Reversible cerebral
142:359–369 vasoconstriction syndrome. Pract Neurol 9:256–267
71. Néel A, Auffray-Calvier E, Guillon B, Fontenoy AM 76. Younger DS (2004) Vasculitis of the nervous system.
et al (2012) Challenging the diagnosis of PACNS: a Curr Opin Neurol 17:317–336. Lippincott Williams &
single-center retrospective study. J Rheumatol Wilkins
39:1026–1034 77. Mohammad AJ, Jacobsson LT, Westman KW
72. Orwat DE, Batalis NI (2012) Intravascular large B-cell et al (2009) Incidence and survival rates in Wegener’s
lymphoma. Arch Pathol Lab Med 136:333–338 granulomatosis, microscopic polyangiitis,
73. Calabrese LH, Dodick DW, Schwedt TJ, Singhal AB Churg–Strauss syndrome and polyarteritis nodosa.
(2007) Narrative review: reversible cerebral vasocon- Rheumatology 48:1560–1565
striction syndromes. Ann Intern Med 146:34–44 78. Kobayashi Y, Ishii K, Oda K et al (2005) Aortic wall
74. Ducros A, Boukobza M, Porcher R, Sarov M, inflammation due to TA imaged with 18F-FDG PET
Valade D, Bousser MG (2007) The clinical and radio- co-registered with enhanced CT. J Nucl Med
logical spectrum of reversible cerebral vasoconstriction 46:917–922
Miscellaneous Vascular Malformations
(Cavernous Malformations, 32
Developmental Venous Anomaly,
Capillary Telangiectasia, Sinus Pericranii)

Luiz Celso Hygino da Cruz and Cintia Elias Pires

Contents Capillary Telangiectasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . 741


Clinical Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 742
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 720 Histological Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 742
Cerebral Cavernous Malformations . . . . . . . . . . . . . . 720 Neuroimaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 742
Clinical Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723 Treatment, Differential Diagnosis, and Final
Histological Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 725 Considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 743
Neuroimaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 726 Sinus Pericranii . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 744
Differential Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 733 Clinical Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 744
Treatment and Final Considerations . . . . . . . . . . . . . . . . . 733 Neuroimaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 745
Developmental Venous Anomaly . . . . . . . . . . . . . . . . . . 733 Treatment, Differential Diagnosis, and Final
Clinical Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 736 Considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 745
Histological Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 739 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 747
Neuroimaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 739
Treatment and Final Considerations . . . . . . . . . . . . . . . . . 741 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 747

L.C.H. da Cruz (*)


Department of Radiology, Clinics CDPI/DASA and IRM,
Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
e-mail: celsohygino@hotmail.com
C.E. Pires
Department of Radiology, Clinics CDPI/DASA, National
Institute of Cardiology, Federal University of Rio de
Janeiro, Rio de Janeiro, Brazil
e-mail: cintiaepires@yahoo.com.br

# Springer Science+Business Media New York 2016 719


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_8
720 L.C.H. da Cruz and C.E. Pires

malformations without arteriovenous shunting


Abstract
[1, 2]. This latter group includes conditions of
Vascular malformations involving the brain are
miscellaneous etiologies such as venous anoma-
uncommon lesions that are seen increasingly
lies (cavernous hemangioma or cavernoma and
more often in clinical practice, primarily
developmental venous anomaly), capillary anom-
because of advances in cross-sectional brain
alies (capillary telangiectasia), and sinus
imaging. They are typically divided into two
pericranii, a special condition considered as a
main categories: arteriovenous malformations
venous malformation. Some can be angiographic-
and vascular malformations without arteriove-
ally occult, like cavernomas and capillary telangi-
nous shunting, including cavernous hemangi-
ectasias [3]. All these lesions must be
oma, developmental venous anomaly, capillary
distinguished from one another due to their differ-
telangiectasia, and sinus pericranii. This latter
ent natural histories and various treatment strate-
group is the topic of interest of this chapter.
gies. As the main focus of this chapter, these
Although all these lesions are clinically rele-
conditions are and will be discussed in detail. In
vant, their natural history and prognosis
this chapter, we attempt to provide a concise
depend on several factors, including lesion
review of the imaging methods utilized in the
type, location, size, and associated conditions.
clinical diagnosis and treatment of the cerebral
Understanding the associated imaging findings
malformations listed above.
and potential complications of these lesions
Combined with digital subtraction angiogra-
assists in determining the appropriate treatment
phy (DSA), cross-sectional imaging, including
options. In this chapter, we attempt to provide a
multiplanar computed tomography (CT) and mag-
concise review of the imaging methods used in
netic resonance imaging (MRI), plays an essential
the clinical diagnosis and treatment of the cere-
role not only in the differential diagnosis of brain
bral malformations listed above.
malformations but also in the long-term follow-up
monitoring of patients with lesions that do not
Keywords
require immediate intervention. It is well
Cerebral vascular malformations • Cavernous
established that MRI is the best noninvasive imag-
hemangioma • Cavernoma • Capillary telangi-
ing modality for the detection of a vascular mal-
ectasia • Developmental venous anomaly •
formation. In certain clinical settings and
Sinus pericranii • SWI
pathologies, it has become apparent that MRI
alone could be sufficient to evaluate some types
of vascular malformations and play an important
Introduction role in treatment management. In addition, it is
extremely helpful in presurgical planning to
Vascular malformations of the brain are uncom- assess the extent of the lesion, define borders,
mon lesions that manifest by imaging tests as and plan the surgical approach, based on the
abnormal vessels in the brain; they are clinically extent of the lesion and its relationship with sur-
relevant since clinical outcome may include hem- rounding areas and localization of eloquent corti-
orrhagic stroke, epilepsy, and focal neurologic cal areas.
deficits. Their pathogenesis involves abnormal
vasculogenesis and/or angiogenesis. The evalua-
tion of a vascular malformation of the brain may Cerebral Cavernous Malformations
be a diagnostic challenge to radiologists due to
their similar clinical manifestations and imaging Cerebral cavernous malformations (CCMs), also
features. known as cavernous angiomas, cavernous heman-
Vascular brain lesions are typically divided giomas, or simply cavernomas, are common cere-
into two main categories: brain arteriovenous bral vascular malformations consisting of clusters
malformations (AVMs) and vascular of deformed and dilated vessels, with little or no
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 721

Fig. 1 Right temporal lobe cavernous malformation. (a) FLAIR images (c). A subtle contrast enhancement can be
T1-weighted MR. (b) Postcontrast T1-weighted MR. (c) seen after intravenous gadolinium administration (b). GRE
FLAIR image. (d) Gradient-echo (GRE) T2*-weighted T2* weighted confirms the hemorrhagic component of the
MR. (e) Diffusion-weighted MR. The lesion presents the lesion. Subacute hemorrhagic foci within the lesion can
characteristic “popcorn” magnetic resonance appearance, demonstrate restricted diffusion, due to the presence of
secondary to blood components in different stages of deg- methemoglobin (e)
radation (a–c). A hypointense rim is well demonstrated on

intervening brain parenchyma, lined by endothe- familial form, usually characterized by multiple
lium and filled with thrombosed blood at various lesions, with an autosomal dominant pattern of
stages [4, 5]. These lesions usually have a charac- inheritance [7]. Typically, patients with a single
teristic appearance on MRI. Modern MR imaging lesion have a sporadic form of the disease, while
sequences are highly sensitive for detecting those with multiple lesions (10–30 % of all cases)
CCMs as well as associated hemorrhage at vari- often have an autosomal dominant form localiz-
ous stages of thrombosis and reorganization able to the CCM1/KRIT1, CCM2/MGC4607, or
(Fig. 1). CCM is combined with capillary telangi- CCM3/PDCD10 gene loci. Genetic linkage ana-
ectasia and thrombosed arteriovenous malforma- lyses mapped three CCM loci to chromosome 7q
tion, under the term occult cerebrovascular (CCM1), 7p (CCM2), and 3q (CCM3) [8]. The
malformation, since none of these lesions is dem- hallmark of familial CCM is the presence of
onstrated by conventional angiography [3]. multifocal lesions throughout the brain with the
CCMs are considered one of the four most appearance of new lesions over time (Fig. 2).
common types of vascular malformations, However, in about 20 % of individuals with mul-
accounting for approximately 8–16 % of all cere- tiple lesions, no mutation is observed [8]. The
brovascular malformations. Their estimated prev- sporadic form of CCM is often characterized by
alence on the basis of MR imaging findings is up a solitary lesion (or a cluster of lesions). There is a
to 0.4–0.5 % of the population [6]. Two forms high association between CCM and DVA (Fig. 3).
have been described: a sporadic form, in which This association with a DVA is more likely to
patients usually have an isolated lesion, and a define the nongenetic familial form of the disease.
722 L.C.H. da Cruz and C.E. Pires

Fig. 2 Multiple cavernous malformations in a patient with left frontal lobe. Note multiple small foci hypointense
familial form. Axial T2-weighted (a) and GRE T2*- lesions on GRE T2*-weighted images (b–d, arrows)
weighted (b) images demonstrate a CCM lesion in the consisting with small cavernous angiomas

Familial CCMs are unlikely to be associated with Although CCM can occur in virtually all parts
DVA, while sporadic forms have a high associa- of the central nervous system, they are much more
tion rate with this vascular malformation [9]. Even common in the cerebral hemispheres, followed by
though the multiplicity of lesions is characteristic the infratentorial compartment. A superficial cere-
of the familial form, it has been reported in 10 % bral hemisphere location with proximity to the
to up to 33 % of supposed sporadic cases. Addi- subarachnoid space (Fig. 5) and the ventricle is
tionally, they are commonly seen after therapeutic very common. Infratentorial involvement corre-
irradiation of the brain (Fig. 4), along with capil- sponds roughly to one fourth of the cases.
lary telangiectasias in the radiotherapy bed [10]. Although the brain stem and the cerebellum
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 723

Fig. 3 Cavernous malformation with associated develop- lesion, hypointense on GRE T2* weighted, without mass
mental venous anomaly. Axial postcontrast T1-weighted effect, in the right cerebellum peduncle, adjacent to the
(a) and FLAIR images (b) and coronal T2-weighted (c) fourth ventricle. There is a developmental venous anomaly
image demonstrate an heterogeneous signal intensity adjacent to the CCM (a)

hemispheres are equally affected, the pons is the T2-weighted sequences and intense homogenous
most common location of the brain stem (Fig. 6). enhancement. Dural-based cavernomas in associ-
CCM can also rarely occur in the spinal cord ation with dural sinuses have also been described
(Fig. 7), where it frequently coexists with multiple [13–15].
brain lesions (Fig. 8) [11, 12].
Extra-axial lesions are extremely atypical and
most lesions arise from the leptomeninges or the Clinical Manifestations
cranial nerves (Fig. 9). They have been described
in the cerebellopontine angle, intraventricularly, Most lesions are found incidentally in neuroimag-
in the pituitary fossa, optic chiasm, and cavernous ing studies and may remain asymptomatic
sinus. Extra-axial cavernous lesions usually dem- throughout life. They can be discovered by an
onstrate atypical findings on cross-sectional imag- episode of symptomatic hemorrhage or gradual
ing, including hyperintense signal intensity on and nonspecific symptoms can be seen. Usually,
724 L.C.H. da Cruz and C.E. Pires

Fig. 4 A 15-year-old male with germinoma was treated T1-weighted (c), axial FLAIR (d), SWI (e), and SWI phase
with surgery followed by radiation therapy and chemother- (f) images obtained 4 years after the radiation therapy show
apy. FLAIR (a) and SWI (b) obtained 2 years after the a round lesion of mixed signal intensity with peripheral
treatment show postsurgical changes in the right frontal hypointense rim in the left occipital lobe, suggestive of
lobe, and no lesion is seen in the left occipital lobe. Sagittal cavernous angioma

patients clinically present with CCMs between the inflammatory reaction, as well as to some degree
fourth and sixth decades of life, but symptoms can of cortex compression. A long-standing seizure
start early in life. Common clinical manifestations without any acute episode of hemorrhage can be
are seizures, focal neurological deficits, and hem- frequently observed.
orrhage (Fig. 10) [11, 12]. Symptoms related to Focal neurologic deficits can occur in 10–45 %
mass effect and headache can also occur. of the symptomatic patients. It is a rare condition,
When clinically apparent, seizures are reported mostly seen in infratentorial involvement.
as the most common symptom, occurring in Progressive deficits are more related to lesion
40–60 % of the symptomatic patients, and are growth and less commonly to an episode of hem-
the most frequent manifestation of the orrhage. The enlargement of the lesion is more
supratentorial lesions. Seizures are most often likely to be caused by chronic or recurrent extrav-
focal, can be refractory to treatment, and are asation of blood and thrombosis.
thought to be related to cortical involvement asso- It is well known that small and subclinical
ciated with irritative effect of hemorrhage and the hemorrhages are a common occurrence.
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 725

Fig. 5 Cavernous malformation with associated develop- demonstrates the heterogeneous signal characteristics of
mental venous anomaly located at the subcortical region of hemorrhage in various stages of resolution. A developmen-
the left frontal lobe. (a) Postcontrast T1-weighted MR. (b) tal venous anomaly is seen after the intravenous contrast
FLAIR MR. (c) GRE T2*-weighted MR. The lesion administration

Symptomatic hemorrhage is a rare condition. The structures. A rim of gliotic brain tissue usually
risk of a hemorrhage from a CCM is thought to delimits the periphery of the CCM with a variable
range from 0.1 % to 0.7 % per year and can reach degree of extralesional hemosiderin pigment from
up to 25 % in patients with a prior history of prior hemorrhages. There is no capsule involving
hemorrhage. The risk of bleeding is increased in the lesion [4, 5].
young and female patients with infratentorial Upon microscopic examination, CCM has a
lesions. Seizures and acute neurological deficits classic appearance of “popcorn” or a “mulberry-
are the most frequent symptoms reported. Exten- like” cluster of sinusoidal blood cavities
sion of hemorrhage to the subarachnoid space and surrounded by a single layer of endothelium,
to the ventricles is uncommon, as well as superfi- with little or no intervening neural tissue. The
cial siderosis, secondary to multiple hemorrhagic vascular structures are filled with blood, thrombi
episodes [11, 12, 16]. in different stages, and organized connective tis-
CCMs can exhibit a wide range of behaviors, sue. Adjacent brain parenchyma is usually gliotic.
including enlargement, regression, and de novo Almost all CCMs have peripheral deposition of
formation. These lesions have also been described hemosiderin, suggesting the occurrence of multi-
to be associated with prior brain radiotherapy. ple episodes of asymptomatic bleeding. Calcifica-
CCMs take about 1–26 years to develop after tions and cholesterol deposit are common findings
radiation [10]. The diagnosis of cavernoma within them. Arteriovenous shunting is absent,
should be considered when a new hemorrhagic and irrigation, as well as drainage, is performed
lesion appears in the radiation bed, particularly if by normal vasculature. Different from arteriove-
the radiation therapy was performed in childhood nous malformations, no muscular or elastic tissue
(Fig. 4). is present in vessel walls, no matter their thickness
[4, 5, 17].
Although the exact mechanism underlying the
Histological Findings pathogenesis of CCMs is unknown, the impor-
tance of the mechanism involved in the prolifera-
By gross pathology, CCM presents as a well- tion and differentiation of angiogenic precursors
circumscribed, red-colored spherical or lobulated and of members of the apoptotic mechanism has
mass lesion, with a grapelike configuration, been pointed out as a key regulator of the patho-
containing multiple thin-walled vascular genesis of CCM [6].
726 L.C.H. da Cruz and C.E. Pires

Fig. 6 Cavernous malformation with associated develop- MR (c, d) images, a developmental venous anomaly is
mental venous anomaly located in the left aspect of the identified with a drainage vein at the inferior aspect of the
pons. Axial FLAIR (a) and coronal T2-weighted MR (b) fourth ventricle, the left ambiens cistern, and the
images show a round heterogeneous lesion, with a quadrigeminal cistern
hypointense rim. On the axial postcontrast T1-weighted

Neuroimaging infratentorial compartments. Lesions vary in size


from few millimeters to several centimeters
Approximately 80 % of the CCM cases are across, with an average size of around 1.5
supratentorial intra-axial lesions. However, they cm. They usually lack mass effect, unless
can be found anywhere in the central nervous there have been recent episodes of bleeding. Typ-
system, including the brain stem and spinal cord. ical lesions show little or no enhancement [7, 11,
Extra-axial lesions have also been described. 12, 14].
They are usually solitary, although up to one Computed tomography (CT) can detect CCM
third of patients with sporadic lesions have more as a spontaneous high-attenuation lesion, with a
than one lesion. In the familial form, multiple variable presence of calcification. However, CT
lesions are usually seen in supratentorial and can be normal in around 30–50 % of the cases.
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 727

Fig. 7 Cavernous malformation in the cervical spine. A T1-weighted image (b) is seen. On T1-weighted MR after
heterogeneous cervical spine lesion with peripheral intravenous contrast administration (c), a venous structure
hypointense rim on the T2-weighted (a) and GRE T2*- is demonstrated inferiorly to the described cavernoma
weighted (d) images and spontaneous hyperintense on (arrow)

Although CCM may be diagnosed by using CT microhemorrhage, CCM contains large amounts
scans, MR imaging is much more sensitive for of deoxyhemoglobin or hemosiderin, which gen-
demonstrating these lesions. Owing to the erate the susceptibility effect, causing marked sig-
blood stagnation phenomenon, or chronic nal loss in specific MR sequences, including
728 L.C.H. da Cruz and C.E. Pires

Fig. 8 Multiple cavernous malformations. Axial FLAIR T2-weighted (d) and axial GRE T2*-weighted (e) images
image (a) demonstrates a subependymal CCM in the also demonstrate a cavernoma in the posterior and right
atrium of the right lateral ventricle. On susceptibility- aspect of the cervical spine, with an exophytic component
weighted images (SWI), multiple cavernomas can be (d, e)
detected as hypointensity lesions (b, c). Sagittal
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 729

Fig. 9 Cavernous malformation of the left trigeminal cranial nerve, invading the ipsilateral Meckel cave. On
nerve. An expansive, extra-axial lesion of mixed signal GRE T2*-weighted (d) image, the lesion presents areas
intensity on both T2-weighted (a, b) and T1-weighted (c) of low signal intensity, as well as a hypointense rim
images, involving the cisternal component of the left fifth

susceptibility-weighted imaging (SWI) and GRE other modality and has largely eliminated
sequences, particularly a T2*-weighted GRE misdiagnosis of CCM. MRI is also used in the
sequence. These sequences are highly sensitive follow-up monitoring of patients with known
for the susceptibility artifact, which make them CCM and for the assessment of family members
especially useful in the detection of smaller and in whom similar lesions are suspected. In addition,
concomitant cavernous lesions that may not be it is extremely helpful in presurgical planning to
detected with traditional sequences. assess the extent of the lesion, define borders, and
Thus, the sensitivity of MRI to flowing blood plan the surgical approach and exposure.
and blood products of varying ages, combined with Computed Tomography: CT scanning is not
the greater contrast resolution of MRIs, greatly sensitive for the diagnosis of CCM and can be
increases the specificity of MRI compared to any normal in 30–50 % of patients. When this lesion is
730 L.C.H. da Cruz and C.E. Pires

Fig. 10 Supratentorial cavernous malformation with a surrounded by moderate amount of edema, which also
recent hemorrhage. An 80-year-old man with mental con- involves the precentral gyrus. The postcontrast
fusion, seizures, and right hemiparesis. Axial T1-weighted T1-weighted image (b) shows mild contrast enhancement.
(a) and T2-weighted (c) images demonstrate an expansive, The susceptibility artifacts generated by the blood products
round, and hemorrhagic lesion in the left postcentral gyrus, can easily be seen on GRE T2*-weighted image (d)

detected on CTscans, it usually appears as a rounded not bring additional information, unless there are
slightly hyperdense lesion that may or may not have associated malformations, such as DVAs.
interspersed calcifications. If there has been a recent Magnetic Resonance Imaging: MRI is the
episode of hemorrhage, then it is more conspicuous method of choice, demonstrating a characteristic
and may be surrounded by a mantle of edema. “popcorn” or “mulberry” appearance with a rim of
Although most lesions do not enhance, faint signal loss due to hemosiderin, better depicted on
enhancement can occasionally be seen. CTA does susceptibility-weighted sequences [12, 14].
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 731

Fig. 11 Cavernous malformation associated to a develop- classic “popcorn” MRI appearance, surrounded by a
mental venous anomaly. (a) Axial T1-weighted MR. (b) hypointense rim. The lesion causes compression of the
Coronal T2-weighted MR. (c) Coronal postcontrast fourth ventricle. After the intravenous contrast administra-
T1-weighted MR. Axial GRE T2*-weighted MR. A left tion, a DVA is observed adjacent to the inferior aspect of
pontine and middle cerebellar peduncle CCM with the the CCM

Recently, more advanced imaging techniques subacute bleeding shows hyperintense signal.
such as high-field and susceptibility-weighted Hypointense areas correspond to hemosiderin
magnetic resonance imaging (SWI) have been deposition, related to chronic hemorrhage.
employed for the evaluation of CCMs. Further- – T2-weighted sequences reveal a heterogeneous
more, diffusion tensor imaging (DTI) and func- lesion surrounded by a marked hypointense
tional magnetic resonance imaging (fMRI) have rim, reflecting hemosiderin deposition.
been applied to the preoperative and – FLAIR sequences can demonstrate
intraoperative management of these lesions. perilesional edema in cases with recent epi-
sodes of bleeding.
– T1-weighted sequences can demonstrate a – Since cavernomas contain large amounts of
round heterogeneous lesion, with a “popcorn” deoxyhemoglobin (blood stagnation) or hemo-
or “mulberry” appearance, containing blood siderin (chronic microhemorrhage), sequences
components in different stages of degradation. that are highly sensitive for susceptibility artifact
Deoxyhemoglobin related to acute bleeding such as SWI and GRE T2*-weighted sequences
has intermediate signal intensity on T1W are extremely useful for detecting marked signal
images, whereas methemoglobin related to loss within the lesions (Fig. 11). Especially for
732 L.C.H. da Cruz and C.E. Pires

Fig. 12 A 74-year-old male patient with familial CCM. frontoparietal regions. SWI (c) shows a higher number of
Axial T2-weighted image (a) shows areas of white matter lesions and confirms the presence of larger lesions
high signal intensity and a small focus of low signal inten- (arrows), which are not seen on the T2-weighted GRE
sity on the left (arrow). T2-weighted GRE image (b) dem- image (Reprinted with permission from Ref. [18])
onstrates multiple foci of low signal intensity on the

multiple lesions, frequently associated with the not demonstrate arteriovenous shunting. When
familial form of the disease, these techniques are they are evident on angiograms, the findings are
of great use. SWI seems to be more sensitive nonspecific and include minimal vascular blush in
than T2* to detect cavernomas (Fig. 12) [18]. the late venous phase. When they occur in com-
The use of the blood pool agent ferumoxytol in bination with other complex vascular
susceptibility-weighted MRI has been recently malformations, angiography can be helpful in
discussed and seems to enhance the visibility of defining other lesions.
CCM, among other vascular malformations A classification system based on imaging and
[19]. Since ferumoxytol has a good biocompat- pathologic features has been reported to stratify
ibility profile, has no known long-term toxicity, these lesions [20]. Type I lesions are characterized
and, in contrast to the gadolinium-based contrast by hyperintensity on both T1WI and T2WI, which
agents, can be safely given to patients with is consistent with subacute hemorrhage. In Type II,
decreased renal function, it seems to be a prom- loculated regions of hemorrhage are surrounded by
ising tool for the diagnosis and follow-up of gliosis and hemosiderin-stained brain parenchyma.
cavernomas. However, further studies are neces- These CCMs exhibit a mixed signal intensity core
sary to support the practical use of this agent. on both T1WI and T2WI, with a well-
– Diffusion-weighted images (DWI) usually do circumscribed hypointense rim on T2WI, and are
not demonstrate restricted diffusion unless the classic CCM with a “popcorn” appearance.
there is subacute hemorrhage (Fig. 1). Type III lesions demonstrate a core that is iso- or
– Contrast-enhanced T1-weighted sequences hypointense on T1WI and hypointense on T2WI,
can demonstrate minimal or absent gadolinium surrounded by a hypointense rim on T2WI, consis-
enhancement. Associated DVAs can also be tent with chronic resolved hemorrhage or hemosid-
detected in T1-weighted postcontrast images. erin within and surrounding the lesion. Type IV
corresponds to tiny lesions often seen as
Digital Subtraction Angiography: On DSA, punctate hypointense foci on susceptibility
most CCMs are angiographically occult and do sequences [21].
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 733

Differential Diagnosis vascularized lesions. The association with DVA


should be considered before performing a surgical
Although cavernomas show a classic aspect on removal, due to the risk of causing a venous infarct
MRI, some conditions can be misdiagnosed as in the surrounding parenchyma. Multiple sequence
the multiple familial forms, giant cavernomas, MRIs play an important role in presurgical plan-
and CCM associated with brain parenchyma hem- ning to assess the extent of the lesion, define bor-
orrhage. Multiple foci lesions with hypointense ders, and plan the surgical approach and exposure.
lesions on susceptibility sequences can be a variety
of pathologic conditions that may mimic CCMs Cerebral Cavernous Malformation
and have to be considered in the differential diag-
General Imaging Neuroimaging
nosis. This variety of conditions include residual features modalities characteristics Hot spot
hyperintense hemorrhage; infections, specially Common MRI is the Characteristic Look for
treated toxoplasmosis, and fungus infection, like cerebral modality of “popcorn” or rounded
vascular choice “mulberry” areas of
aspergillosis and candidiasis; prior radiation ther- malformation appearance on marked
apy; disseminated intravascular coagulopathy; with classic MRI with signal loss in
amyloid angiopathy; diffuse axonal injury and appearance heterogeneous susceptibility-
on MRI signal intensity weighted MR
metastatic lesions secondary to melanoma and cho- within the sequences for
riocarcinoma; and metastasis from the lung, kid- lesion and a detection of
ney, and thyroid neoplasms (Fig. 13) [22]. rim of signal smaller and
loss due to concomitant
The distinction of CCM from hemorrhagic hemosiderin, cavernous
neoplasm may be difficult (Fig. 14). Some imag- better depicted lesions that
on may not be
ing features can be useful and can allow differen- susceptibility- detected with
tiation from a neoplasm. Some imaging signs are weighted traditional
more likely to be observed in CCMs, such as sequences sequences
(SWI and GRE
absence or little surrounding edema, absence of T2*)
identifiable nonhemorrhagic neoplasm, complete Isolated or CT scans can be Lack of mass
hemosiderin ring in the adjacent parenchyma, and multiple normal in effect, unless
lesions. In the 30–50 % of cases there has been
expected temporal evolution of the hematoma. latter type, recent bleeding
The presence of T1 hyperintensity in the they are often
familial, with
perilesional edema around a hemorrhagic lesion autosomal
can be able to differentiate CCM from other hem- dominant
orrhagic masses, like hemorrhagic tumors and transmission
Occur in all Angiographically Little or no
intracerebral hemorrhages (Fig. 15) [23].
parts of the occult enhancement
central
nervous
system but
Treatment and Final Considerations most
commonly in
Although most CCMs can simply be followed over the cerebral
hemispheres
time, surgical removal is an option in lesions caus-
ing significant morbidity related to mass effect,
epileptic activity, or repeated hemorrhage. Since
cavernomas are well circumscribed and Developmental Venous Anomaly
surrounded by a gliotic rim, surgical removal is
relatively simple. Control of hemorrhage is usually Developmental venous anomaly (DVA), also
easier because blood flow through the lesions is known as venous angioma or venous vascular
slower than that expected in more highly malformation, is the most frequently encountered
734 L.C.H. da Cruz and C.E. Pires

Fig. 13 A 77-year-old woman with lung cancer. Multiple enhancing small lesions in the cortical-subcortical junction
hemorrhagic metastatic lesions are better demonstrated on of the hemispheres and at the subependyma of the right
SWI (b) image than on T2-weighted image (a). lateral ventricle
Postcontrast axial T1-weighted image (c) shows multiple

cerebral vascular malformation. Usually asymp- common locations are the frontoparietal regions,
tomatic, it is frequently reported as a fortuitous accounting for 36–64 %, usually draining toward
finding in imaging studies. DVAs represent a the frontal horn of the lateral ventricle. The cere-
purely venous entity, with no arterial component. bellar hemispheres are also a very common loca-
They are mostly considered extreme anatomical tion, estimated to represent 14–27 % of the cases,
variations of the transmedullary veins that are draining toward the fourth ventricle [27]. They
necessary for the drainage of normal brain tissue. may be isolated, draining one territory, or may
Although DVA was thought to be rare before the compose a complex venous draining system of a
advent of CT and MR imaging, it is now consid- whole cerebral or cerebellar hemisphere.
ered the most common anomaly of the intracranial DVAs frequently coexist with other types of
vasculature, accounting for 63 % of vascular CNS vascular malformations such as arteriove-
malformations, with an overall incidence of nous malformation, capillary telangiectasia, and,
around 2.5 % [24]. especially, cavernous angiomas (15–30 % of
DVAs are believed to be adaptations to acci- patients) (Fig. 16) [28]. Indeed, the association
dents occurring during embryogenesis between the between DVAs and cavernous malformations is
fourth and seventh stages of embryologic develop- so common that the presence of a DVA on an
ment, resulting in occlusion or maldevelopment of imaging study should promptly demand a search
either the superficial or deep veins. Due to the for a cavernoma, which is more clinically impor-
plasticity of the vascular system at this stage, tant. They are also associated with head and neck
DVAs are formed as compensatory pathways, venous malformations (Fig. 17) and hemangi-
recruiting and dilating preexisting transmedullary omas and, more rarely, with cortical dysplasia,
veins [25, 26]. Since the involved vessels are not since both occur at the same developmental
abnormally formed, but apparently merely dilated stage. DVA is thought to be a manifestation of
without any proliferative potential, the term DVA the underlying abnormal neuronal migration and
seems to be more appropriate than “venous angio- not its cause [29]. In a prior publication, the fre-
mas” or “venous vascular malformations” [26]. quency of DVAs in patients with cervicofacial
Although DVAs can be seen anywhere in infra- venous malformations was described as around
or supratentorial compartments, draining either to 20 %. Most of these cervicofacial malformations
superficial or deep vein systems, the most were superficial and extensive [30].
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 735

Fig. 14 A solitary breast metastatic lesion on the right by edema. The lesion causes compression to the anterior
lateral portion of the pons in a 65-year-old woman can be and lateral portion of the fourth ventricle. Enhancement of
confused with CCM. Axial T1-weighted (a), T2-weighted the central portion of the lesion was seen after gadolinium
(c), and GRE T2*-weighted (d) images demonstrate an administration on T1-weighted image (b). The histological
expansive mixed signal intensity lesion, with hemorrhagic diagnosis was made after biopsy
component, associated with a hypointense rim, surrounded

Arterialized DVAs are an atypical form the supratentorial and cerebellar white matter, as
described when an early angiographic well as in the basal ganglia and the caudate
opacification occurs during the mid- or late arte- nucleus. This calcification may represent chronic
rial phases, typically found in large supratentorial hemorrhage, long-standing cerebral ischemia, or
DVAs. In some particular examples, these sub- venous hypertension [31]. White matter abnor-
types can be associated with an arteriovenous malities usually present as a slight hyperintense
malformation (AVM) [24]. area that characteristically respects the boundaries
DVAs can also be associated with regional of the DVA drainage territory (Fig. 18). They are
cerebral abnormalities other than CCMs within stable over time, do not enhance after contrast
the drainage territory in about 65 % of the cases. administration, and do not have mass effect. The
The most frequent abnormality is locoregional etiology of signal intensity changes is unclear but
cerebral atrophy, accounting for almost one third may be related to edema, gliosis, or leukoaraiosis
of the cases, followed by white matter lesions secondary to focal vascular narrowing within the
(ranging from 8 % to 30 %) and dystrophic calci- common venous stem that can alter the hemody-
fication (nearly 10 %), which occurs typically in namics of the drainage area [32].
736 L.C.H. da Cruz and C.E. Pires

Fig. 15 Axial FLAIR (a), coronal T2-weighted (b), and the centripetal pattern of the T1 hyperintense perilesional
SWI (c) images demonstrate a hemorrhagic mass with signal intensity sign, observed only at the deep area around
heterogeneous signal intensity, surrounded by edema in the hemorrhagic lesion, but not observed at the peripheral
the right frontal lobe. Sagittal T1-weighted image (d) dem- margin. The histological diagnosis of CCM was confirmed
onstrates hyperintensity within the vasogenic edema. Note after surgical excision

Clinical Manifestations less familiar with the field of intracranial vascular


malformations. Also, their management requires
DVAs represent a physiologically competent thorough understanding of the nature of DVAs,
venous drainage pathway of normally functioning including their frequent coexistence with other
brain tissue. Only rarely have neurologic symp- types of vascular malformations.
toms been attributed to them. Although DVAs are DVAs are typically benign, asymptomatic, and
usually found incidentally in neuroimaging stud- uncomplicated. However, they can rarely hemor-
ies performed for other reasons and are asymp- rhage and become symptomatic, most notably after
tomatic in most cases, their diagnosis often venous ischemia or infarctions from spontaneous
generates concern among physicians who are thrombosis of the venous collector. On the other
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 737

Fig. 16 Developmental venous anomaly with associated angioma associated to a hemorrhagic lesion, suggestive
cavernous malformation located at the left cerebellum of cavernous malformation. A drainage vein is better seen
hemisphere. Axial FLAIR (a), postcontrast T1-weighted at SWI (c, arrow)
MR (b), and SWI (c) images show a typical venous

Fig. 17 Postcontrast T1-weighted images demonstrate a right frontotemporal lymphangioma (arrows) associated with a
right cerebral hemisphere developmental venous anomaly, which has a deep drainage vein

hand, based on current literature, most instances of as seizures probably originated from the accompa-
hemorrhage with DVAs have been in patients with nying vascular malformation, rather than from the
combined vascular malformations, especially DVA alone. The association between DVAs and
CCM. It is controversial to attribute clinical symp- CCMs has been widely described. DVA is
toms to DVAs. In the vast majority of symptomatic associated with one or more cavernomas in about
cases, hemorrhage and clinical manifestations such 13–40 % of cases [28]. These CCMs are thought to
738 L.C.H. da Cruz and C.E. Pires

Fig. 18 Axial FLAIR (a) and SWI (b) images demonstrated a venous angioma with deep venous drainage, associated to
marked signal intensity abnormality, extending to the ventricular margin

Fig. 19 Axial postcontrast T1-weighted (a–c) images Axial FLAIR (d) and SWI (e, f) images demonstrate a left
demonstrate the characteristic enhancement and caput cerebellum hemisphere hematoma associated with devel-
medusae distribution of the developmental venous anom- opmental venous anomaly. No cavernoma was observed
aly, which drains anteriorly via an enlarged medullary vein. associated to the DVA

be responsible for the vast majority of symptomatic very rarely cause intracerebral hemorrhage when
cases, previously attributed to the DVAs [24]. It is they are not associated with CCM. The bleeding
interesting that CCMs associated to DVAs have a risk associated with DVAs is greater in the poste-
more aggressive clinical course than CCMs alone rior fossa (Fig. 19). However, it can very rarely
and are more likely to present with symptomatic occur in other regions of the brain parenchyma
hemorrhage [33]. Although DVAs have a very low [35]. Thrombosis of the collector vein of a DVA
risk of hemorrhage (0.22–0.68 %) [34], DVAs can is a rare complication that can lead to cerebral
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 739

venous infarction in about 53 % of the cases, which structure, and enhancement of the dilated
can be associated with hemorrhage (37 %), and, transmedullary veins with a radial distribution
less frequently, subarachnoid and intraventricular converging to the venous collector (caput medu-
hemorrhage (5 %) [36]. sae) can also be observed.
DVAs may also rarely cause symptoms by their Computed Tomography: DVAs are usually not
mechanical relationship with adjacent brain struc- identified by non-enhanced brain CT. However,
tures. Some reports have described clinical symp- they can rarely appear as focal hyper-attenuations,
toms related to DVAs, such as cranial nerve especially when associated with a cavernoma.
compression [37] and hydrocephalus, secondary After intravenous iodinated contrast administra-
to cerebral aqueduct obstruction by DVA [38]. tion, DVAs can be identified as radially distributed
vascular structures converging into a unique
venous collector, which in turn drains into a corti-
Histological Findings cal or deep vein, or even into a dural venous sinus.
When the anomaly is extremely small, a linear
A cluster of venous radicles that converge into a enhancing structure (venous collector) running in
collecting vein characterizes DVA, resulting in the the direction of the ventricular or pial surface may
typical appearance of caput medusae. The be the only finding suggestive of DVA.
collecting vein has a variable length and joins Although it is usually unnecessary, a more
the superficial or the deep venous system after detailed evaluation of a DVA’s venous drainage
crossing the brain parenchyma. In some particular can be obtained with angio-CT with 3D MIP
cases, the drainage pathway can be done by both reformation.
the superficial and deep venous systems, with one Magnetic Resonance:
of those as predominant [24]. – T1-weighted sequences: Although DVAs are
Macroscopically, DVAs are characterized by usually not depicted on non-enhanced T1WI,
multiple medullary or subcortical dilated veins, especially when they are small, large lesions
which are radially disposed with a centripetal can eventually demonstrate flow voids.
drainage system into one venous collector that is – T2-weighted sequences: With T2WI, flow
also dilated. This collector drains into either a voids possibly associated with degradation
superficial (subcortical) or a deep (pial) venous blood products can be seen.
system. The usual drainage vein of the specific – FLAIR: Hyperintense foci on FLAIR sequences
location is commonly absent. related to venous ischemia or hemorrhage in
Microscopically, DVAs appear as a conglom- different phases can also be demonstrated.
erate of dilated veins with minimal parietal thick- – Magnetic susceptibility sequences:
ening and hyalinization, with the absence of Susceptibility-weighted imaging (SWI) and
elastic lamina, and loosely arranged smooth mus- T2* gradient echo demonstrate that the DVA
cle layers, intermingled with normal brain paren- has a marked hypointensity secondary to the
chyma [24]. The intervening brain tissue paramagnetic deoxyhemoglobin with venous
associated with DVA is usually normal, with no blood (Fig. 20). These sequences can also dem-
evidence of hemorrhage or gliosis [22]. It can be onstrate hypointense foci if DVAs are associ-
extended, compromising a whole cerebral hemi- ated with old bleeding or a cavernoma (Fig. 21).
sphere, or limited to a small territory. – As described for cavernomas, the use of the
blood pool agent, ferumoxytol, seems to demon-
strate additional tributary veins and improve the
Neuroimaging detection of DVAs in susceptibility-weighted
MRI [19]. However, further studies are neces-
DVAs are best depicted in contrast-enhanced sary to support practical use of this agent.
imaging studies. The venous collector is usually – Contrast-enhanced T1WI sequences: A T1WI
identified as a linear or curvilinear enhancing sequence after gadolinium administration is
740 L.C.H. da Cruz and C.E. Pires

Fig. 20 Venous angioma with intravascular hypointensity T1-weighted MR (b) images. A marked hypointense signal
on susceptibility image. Right frontal venous angioma is intensity on SWI (c) is seen, probably secondary to
demonstrated on axial FLAIR (a) and postcontrast deoxyhemoglobin in venous blood

Fig. 21 Axial T1-weighted image after contrast adminis- hypointensity can also be observed and can be secondary
tration (a, b) and FLAIR images (c) demonstrate a DVA to deoxyhemoglobin in venous blood (d, arrow). Round
with deep venous drainage. Associated signal intensity hypointense areas within the lesion can be also demon-
abnormality surrounds the DVA, extending into the white strated (e, arrows), which may correspond to cavernomas
matter (c). On SWI images (d, e), linear areas of or even previous hemorrhage

the best sequence to identify a DVA, which medusae pattern, this sequence is usually not
appears as a tubular-enhanced structure directed necessary for the diagnosis of DVAs.
to a pial or ventricular surface. The small
transmedullary veins draining radially into the
collector are also seen. Although venous angio- Digital Subtraction Angiography: DSA is
MR can easily demonstrate the classic caput only rarely indicated if CT or MR interpretations
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 741

are doubtful. The classic angiographic feature is Developmental Venous Anomaly


the typical caput medusae appearance of the med-
General Imaging Neuroimaging
ullary veins seen only in the late venous phase of features modalities characteristics Hot spot
angiography. Larger lesions can demonstrate a Usually Best depicted Radially Always look
dense capillary stain; however, the lack of arterial incidental by contrast- distributed for other
finding in enhanced CT small vascular associated
enlargement suggests a DVA [39]. neuroimaging or MRI structures vascular
In rare cases, the transmedullary veins can studies (caput malformations,
be seen earlier in the late capillary phase performed for medusae especially
other reasons pattern) cavernomas,
due to dilatation of capillary spaces, which pro- converging in cases of
vides a more rapid transit time, a finding that into a unique symptomatic
should not be misinterpreted as an arteriovenous venous DVAs
collector,
shunt. which in turn
Angiography ideally details the venous drain- drains into a
cortical or
age to the cortical or ventricular surface or to the deep vein, or
dural sinus and is the gold standard method to even into a
evaluate permeability of the whole extension of venous dural
sinus
the collector vein. Asymptomatic Angiography:
in most cases rarely
indicated
No age or SWI:
Treatment and Final Considerations gender important for
predilection the
DVAs should be considered as simple varia- identification
of associated
tions of the brain’s venous system and usually vascular
do not require intervention. Since the vast malformations,
especially
majority of DVAs are clinically asymptomatic,
cavernomas
most experts question the benefit of surgical
treatment, since the resulting risk of an iatro-
genic venous infarct and hemorrhagic compli-
cations would far exceed the risk of irreversible Capillary Telangiectasia
damage from the DVA itself during a patient’s
lifetime. Additionally, DVAs that become Brain capillary telangiectasias (BCTs) are a dis-
symptomatic by bleeding frequently coexist tinct category of vascular malformations that are
with other types of vascular malformations, occult on angiography but are being increasingly
suggesting that the symptoms are actually discovered incidentally on MRIs when the brain is
caused by the associated vascular malformation imaged for other reasons [40]. They consist of
other than by the DVA itself. Therefore, any localized collections of multiple thin-walled vas-
signal intensity anomaly of the adjacent paren- cular channels interposed between normal brain
chyma should prompt the search for other vas- parenchyma.
cular lesions. Most capillary telangiectasias are considered
However, it is always important to mention benign entities, generally asymptomatic, and
the existence of a DVA in a medical report, without further implications for patient manage-
especially if it is an incidental finding in a ment. The true incidence of these malformations
neuroimaging test performed for other surgical is difficult to estimate because most are likely to
reasons, such as tumors. In this case, the neu- be clinically asymptomatic, but autopsy series
rosurgeon must be alerted to the fact that struc- suggest they are not uncommon, representing
tural damage of the venous anomaly could approximately 16–20 % of all CNS vascular
compromise the drainage of normal brain malformations [41, 42]. They are also considered
parenchyma. the second most common vascular anomaly after
742 L.C.H. da Cruz and C.E. Pires

developmental venous anomalies on imaging hemorrhages or small areas of altered coloration


studies [43]. of the brain tissue, far more common in the pons.
The area of involvement of the brain is typically Microscopically, it is composed of numerous
small, ranging from several millimeters to 2 cm in thin-walled vessels without smooth muscle or
size, and common sites of involvement include the elastic fibers that are separated by neural tissue.
pons, cerebral hemispheres, and spinal cord. The This is in contrast to cavernous malformations,
pons is the classic location of these lesions. BCT is which have no normal brain tissue within their
inconspicuous on conventional pre-contrast MR confines. There is little or no gliosis, calcification,
images and in most cases is detected due to faint or gross hemorrhage [40].
enhancement after intravenous contrast administra-
tion [41]. It can be difficult to differentiate CCMs
from BCTs when the lesion is in the pons. The Neuroimaging
association with hemorrhage implies the diagnosis
of CCM rather than BCT. Typically, BCT is occult Capillary telangiectasias are mostly located
on angiography studies. within the brain stem and pontine regions,
although they can be seen virtually anywhere in
the brain parenchyma or spinal cord [41]. They
Clinical Manifestations are usually seen as subtle lesions without mass
effect or circumjacent edema. As they are almost
The vast majority of capillary telangiectasias are always invisible both on CT and DSA, introduc-
incidental findings, as these malformations are tion of MRI was definitive for the wise recogni-
nearly always asymptomatic, without further tion of this entity by the radiology community.
implications for patient management Computed Tomography: Unenhanced CT can
[40]. Although headaches, confusion, weakness, almost never depict capillary malformations,
dizziness, visual changes, vertigo, tinnitus, and although punctate calcifications can be seen in
seizures can be rarely associated with capillary exceptional cases. Enhanced CT can rarely dem-
telangiectasias, confidently attributing such gen- onstrate capillary malformations as small areas of
eralized symptoms to these lesions is difficult. subtle enhancement in typical locations (pontine
Furthermore, the precise cause of these sup- region).
posed symptoms is usually not known because Magnetic Resonance Imaging: Contrast-
reports of apoplectic hemorrhage developing enhanced MRI is the best imaging modality for
from capillary telangiectasia are extremely rare the diagnosis of capillary telangiectasia.
[44]. In fact, similar to DVAs, the detection of
hemorrhage on imaging studies of patients with – T2 or FLAIR sequences can be normal or
capillary malformations should prompt consider- demonstrate a discreet area of slightly
ation of coexisting lesions, such as cavernomas, or increased signal intensity, without mass effect
may indicate a “mixed” form of lesion. or adjacent edema.
To avoid the diagnostic dilemma, a character- – Susceptibility-weighted magnetic resonance
istic signal intensity loss on susceptibility- sequences (SWI/T2*-weighted GRE sequence)
weighted sequences has been described, which, can demonstrate typical moderate hypointense
in conjunction with postcontrast enhancement, is signal in the whole malformation or just in the
virtually diagnostic of BCT [45]. center of the lesion and therefore play an impor-
tant role in the differential diagnosis with other
brain stem lesions, such as neoplastic or sub-
Histological Findings acute ischemic lesions. This hypointensity may
be explained by the low-velocity blood flow
Macroscopically, capillary telangiectasias can be in ectatic venous channels, resulting in a drop
undetectable or present merely as petechial of oxygen saturation and thus increasing
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 743

Fig. 22 Typical case of a central pontine capillary telan- signal intensity drop on SWI (c). Note that SWI (d) more
giectasia. The lesion is slightly hyperintense on clearly depicts a prominent vessel related to the BCT
T2-weighted image (a), faint homogeneous enhancement (arrow), which may correspond to a drainage vein or an
on postcontrast T1-weighted image (b), and considerable associated developmental venous anomaly

deoxyhemoglobin, causing susceptibility effect – Diffusion-weighted imaging is usually isointense


[40] or presumably representing residual sub- or hypointense, which is helpful in discerning
clinical bleeding. SWI is more sensitive to sus- BCTs from subacute ischemic disease [46].
ceptibility changes than T2* gradient echo,
being able to demonstrate small BCT lesions Digital Subtraction Angiography: Catheter
[43, 45]. angiograms are usually normal but can rarely
– On T1-weighted sequences, capillary demonstrate a faint blush.
malformations are typically iso- to hypointense
compared with the brain parenchyma. After IV
gadolinium administration, the lesion appears Treatment, Differential Diagnosis,
as an ill-defined focal enhancement, sometimes and Final Considerations
with a lineariform and mottled pattern. A prom-
inent draining vein can be depicted on both Since capillary telangiectasias are benign lesions
postcontrast T1WI and SWI (Fig. 22) [43]. that are almost always asymptomatic, have
744 L.C.H. da Cruz and C.E. Pires

interspersed normal brain parenchyma, and are Sinus Pericranii


most frequently located in the pons, surgical treat-
ment becomes impractical and unnecessary. No Sinus pericranii (SP) is an unusual venous mal-
follow-up is required if the imaging appearances formation characterized by anomalous commu-
are typical. nication between the intracranial dural sinuses
BCT is a benign entity and should be differ- and dilated epicranial venous blood vessels
entiated from some conditions to avoid unneces- through a bony defect. It consists of a collection
sary follow-up imaging evaluation, surgical of nonmuscular venous structures adhered
biopsy, or even instigation of treatment for pre- tightly to the outer surface of the skull, which
sumed pathology. Its MRI appearance should be communicates with an intracranial venous sinus
distinguished from primary neoplasms like astro- through diploic veins. The varicosities, inti-
cytomas, lymphomas, metastases, inflammatory mately associated with periosteum, are distensi-
processes, active demyelination lesions, and sub- ble and vary in size with changes in intracranial
acute ischemia. In the vast majority of cases, pressure [47].
demonstration of typical signal intensity loss on Sinus pericranii can appear at any age. The
susceptibility sequences (SWI, T2*GE) in an vast majority of cases are congenital in nature,
enhancing focal brain lesion, otherwise but they can also be acquired, traumatic, or spon-
unremarkable on conventional MR images, is taneous. In congenital cases, there is often associ-
highly specific for BCT, and no differential diag- ation with other venous anomalies and
nosis needs to be considered in this scenario syndromes, especially blue rubber bleb nevus
[45]. In cases of atypical features, the lack of syndrome.
mass effect or edema should favor the diagnosis Traumatic causes can be related to a skull frac-
of capillary malformations instead of glioma or ture, a tear in the dural sinuses or emissary veins, a
metastasis. Additionally, the lack of water depressed bone fracture leading to a dural sinus
restriction in DWI sequences is useful to differ- tear, or an epidural venous hematoma caused by a
entiate these lesions from subacute ischemic sinus tear. SP can be associated with other con-
disease [46]. genital vascular anomalies, such as an aneurysmal
internal cerebral vein, cavernous hemangioma,
General Imaging Neuroimaging
features modalities characteristics Hot spots
and systemic angioma [48].
Usually Best Poorly Demonstration Three different pathophysiological types of
incidental depicted demarcated of signal sinus pericranii have been described: circulation
finding in by area of slightly intensity loss
neuroimaging contrast- increased signal on SWI/T2* in
in a closed system in which the blood comes from
studies enhanced intensity on T2 an enhancing the sinus and also returns to it, circulation origi-
performed for MRI and FLAIR focal brain nating in the sinus with peripheral passage of the
other reasons sequences, with lesion,
a faint otherwise venous drainage, and a connection between an
ill-defined unremarkable angioma and the sinus [48].
enhancement on
on T1WI conventional
MR images, is
highly specific Clinical Manifestations
for BCT
Almost always Almost Moderately No mass effect,
asymptomatic always hypointense on edema, or In congenital cases, it usually manifests in the first
invisible SWI/T2* restricted months or years of life as a compressible
both on diffusion
CT and
nonpulsatile soft tissue scalp mass, most com-
DSA monly located in the frontal region (40 %) along
No age or No mass effect or close to the midline. Other possible locations
gender or edema
predilection
include the parietal (34 %), occipital (23 %), and,
more rarely, temporal regions (4 %) [49, 50].
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 745

Patients typically present with a nontender, angiography is useful for excluding arterial
nonpulsatile fluctuant soft tissue mass in the abnormalities and can demonstrate heteroge-
scalp. The varicosities typically reduce in upright neous enhancement of the extracranial venous
position and distend when prone or with the component because of the typical slow flow sta-
Valsalva maneuver, crying, coughing, and jugular tus of some of the connections. CT venography
vein compression. They are usually stable or show (CTV) or delayed imaging usually shows
discreet enlargement with time, although there homogenous enhancement throughout the vascu-
have been rare reports of spontaneous lar mass. In cases of thrombosis, CTV may
regression [51]. demonstrate filling defects within the
Since it is usually asymptomatic and painless, varicosities [47].
the primary concern is often cosmetic, but occa- Magnetic Resonance Imaging: MR images
sionally vague symptoms may occur, including demonstrate a paramedian extracranial mass
headache, vertigo, feelings of fullness, and local with a T2 hyperintense signal in most cases.
pain [52]. A sudden clinical alteration of the Signal intensity in T1WI sequences is variable.
lesion from soft and painless to firm and painful Flow voids may be present in a rapid-flowing
is a concern and should raise suspicion for varix or venous malformation. Contrast-
complications such as thrombosis or, more enhanced MRI demonstrates homogenous “vas-
rarely, hemorrhage, emboli, and infection. The cular” enhancement in cases of varix and hetero-
occurrence of partial thrombosis is not unex- geneous enhancement in cases of venous
pected because of the typical slow flow malformations. MR venography (MRV) delin-
of blood within these lesions. Other reported eates sharply demarcated vascular components
complications include hemorrhage and air of the lesion. In cases of thrombosis, a subacute
embolisms [53]. thrombus shows hyperintensity on T1-weighted
sequence, in correspondence to filling defects
(Fig. 23) [47].
Neuroimaging Doppler Sonography: Ultrasound imaging
demonstrates anechoic or hypoechoic extracra-
The diagnosis of SP is made from a combination nial lesions with lobulating and bulging con-
of clinical and radiologic features. It is based on tours. Doppler flowmetry shows intracranial
the demonstration of an extracranial venous com- and extracranial venous communication through
ponent communicating with a dural sinus through a calvarial defect. SP shows pulsatile Doppler
a bony defect. The most commonly involved sinus wave patterns and high-flow/low-flow vascular
is the sagittal superior, followed distantly by the shunting independent of changes in intracranial
transverse sinus. The extracranial venous compo- pressure [54].
nent may include a venous varix, venous malfor- Digital Subtraction Angiography: The arterial
mation, or multiple veins. phase of DSA usually shows no arterial abnormal-
Radiography: Plain skull radiography ities with exception for an eventual arterial blush.
shows bony defects, cortical thinning, or focal DSA during the venous phase confirms the
bony erosions, in close relation to a soft transosseous connection between the pericranial
tissue mass. varicosities and the dural sinus [50].
Computed Tomography: Unenhanced CT
scans reveal a paramedian extracranial soft tissue
mass overlying a calvarial defect, usually in close Treatment, Differential Diagnosis,
relation to the sagittal superior sinus. A and Final Considerations
transosseous defect within the skull and connec-
tions between the epicranial varicosities with an The differential diagnosis is extensive and
underlying dural sinus can be demonstrated. CT includes most of scalp lesions that appear as soft
746 L.C.H. da Cruz and C.E. Pires

Fig. 23 Sinus pericranii. Midsagittal postcontrast subcutaneous part of the venous anomaly through a small
T1-weighted MR images (a–c) and a tridimensional con- bone defect at the parietal region (c, d arrows) to the
structive interference in steady-state (3D CISS) MR posterior part of the superior sagittal sinus
sequence (d). An emissary vein extends from a

compressible masses such as meningocele, Although SP is usually asymptomatic, it may


encephalocele, skin or skull lesions, and other rarely cause fatal complications such as thrombo-
vascular abnormalities like hemangiomas, arterio- sis, traumatic air embolism, or hemorrhage. Thus,
venous malformations, lipoma, dermoid cysts, prompt treatment after diagnosis is required for
and epidermoid cysts [52]. In general, neuroim- prophylactic purposes to prevent complications,
aging modalities such as CT and MR are able to as well as for aesthetic purposes.
differentiate these conditions. In case of atretic Treatment options include surgery in most
cephalocele, absence of enhancement usually cases and endovascular therapy for smaller lesions
helps to discern this entity from sinus pericranii. [50]. In surgical treatment, the key point is to
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 747

resect the extracranial venous package and ligate arteriovenous malformations) from those that do
the emissary-communicating vein. not have shunting. The latter group is the main
topic of our chapter and includes miscellaneous
General Imaging Neuroimaging
features modalities characteristics Hot spots
etiology conditions, such as venous anomalies
Most cases Diagnosis is Extracranial Think of SP (cavernous hemangioma or cavernoma and devel-
are based on the paramedian in extracranial opmental venous anomalies), capillary anomalies
congenital demonstration soft tissue masses in
and become of an mass with close relation
(capillary telangiectasia), and sinus pericranii, a
clinically extracranial vascular to the sagittal special condition characterized by an abnormal
apparent in venous enhancement superior sinus communication between the dural venous sinus
the first years component in the
or months as communicating venography and extracranial venous system. All these lesions
a to dural sinus phase must be distinguished from one another by their
compressible through a bony overlying a different natural histories and the various treat-
nonpulsatile defect bony defect
soft tissue in close ment strategies. Cross-sectional imaging, includ-
scalp mass relation to a ing multiplanar computed tomography and
dural sinus
magnetic resonance imaging, is important not
Traumatic Best depicted Sagittal Think of
etiology on CT or MR superior sinus thrombosis in only in the differential diagnosis of brain
more venography much more painful and malformations but also in the long-term follow-
uncommon common noncompressible
lesions with
up monitoring of patients with lesions that do not
filling defects in require immediate intervention. It is also of great
delayed contrast- importance in presurgical planning to assess the
enhanced
imaging (CTV, extent of the lesion, define borders, and plan the
MRV, or DSA) surgical approach. In this chapter, we attempt to
Doppler provide a concise review of the imaging methods
ultrasound is
useful for utilized in the clinical diagnosis and management
neonates of the vascular lesions listed above.
DSA during the
venous phase
confirms the
diagnosis
References
1. Chaloupka JC, Huddle DC (1998) Classification of
vascular malformations of the central nervous system.
Summary Neuroimaging Clin N Am 8(2):295–321
2. MacDonald RL, Stoodley M, Weir B (2001) Vascular
malformations of the central nervous system.
Cerebral vascular malformations are uncommon Neurosurg Q 11(4):231–247
lesions of great clinical relevance, since clinical 3. Sigal R, Krief O, Houtteville J, Halimi P, Doyon D,
outcome may include serious neurological dis- Pariente D (1990) Occult cerebrovascular
malformations: follow-up with MR imaging. Radiol-
ability in a significant proportion of individuals
ogy 176:815–819
bearing them. These lesions are seen increasingly 4. Tagle P, Huete I, Méndez J, del Villar S (1986) Intra-
in clinical practice, primarily because of advances cranial cavernous angioma: presentation and manage-
in cross-sectional brain imaging, and their evalu- ment. J Neurosurg 64(5):720–723
5. Russel DS, Rubistein LJ (1989) Pathology of tumors of
ation provides a diagnostic challenge to radiolo-
the nervous system, 5th edn. Williams & Wilkins,
gists due to their similar clinical manifestations Baltimore
and imaging features. Different classification sys- 6. Cavalcanti DD, Kalani MY, Martirosyan NL, Eales J,
tems have been put forward. The most commonly Spetzler RF, Preul MC (2012) Cerebral cavernous
malformations: from genes to proteins to disease. J
used classification system separates vascular
Neurosurg 116:122–132
lesions that demonstrate shunting from arterial 7. Brunereau L, Labauge P, Tournier-Lasserve E
to venous systems malformations (brain et al (2000) Familial form of intracranial cavernous
748 L.C.H. da Cruz and C.E. Pires

angioma: MR imaging findings in 51 families. French 23. Yun TJ, Na DG, Kwon BJ et al (2008) A T1
Society of Neurosurgery. Radiology 214(1):209–216 hyperintense perilesional signal aids in the differentia-
8. Riant F, Bergametti F, Ayrignac X, Boulday G, tion of a cavernous angioma from other hemorrhagic
Tournier-Lasserve E (2010) Recent insights into cere- masses. AJNR Am J Neuroradiol 29(3):494–500
bral cavernous malformations: the molecular genetics 24. Ruiz DS, Yilmaz H, Gailloud P (2009) Cerebral devel-
of CCM. FEBS J 277(5):1070–1075 opmental venous anomalies: current concepts. Ann
9. Petersen TA, Morrison LA, Schrader RM, Hart BL Neurol 66:271–283
(2010) Familial versus sporadic cavernous 25. Truwit CL (1992) Venous angioma of the brain: his-
malformations: differences in developmental venous tory, significance, and imaging findings. AJR Am J
anomaly association and lesion phenotype. AJNR Am Roentgenol 159:1299–1307
J Neuroradiol 31:377–382 26. Cheong WY, Tan KP (1993) Cerebral venous
10. Jain R, Robertson PL, Gandhi D, Gujar SK, Muraszko angioma – a misnomer? Ann Acad Med Singapore 22
KM, Gebarski S (2005) Radiation-induced cavernomas (5):736–741
of the brain. AJNR Am J Neuroradiol 26:1158–1162 27. Lee C, Pennington MA, Kenney CM (1996) MR eval-
11. Robinson JR, Awad IA, Little JR (1991) Natural his- uation of developmental venous anomalies: medullary
tory of the cavernous angioma. J Neurosurg 75 venous anatomy of venous angiomas. AJNR Am J
(5):709–714 Neuroradiol 17(1):61–70
12. Batra S, Lin D, Recinos PF, Zhang J, Rigamonti D 28. Huber G, Henkes H, Hermes M et al (1996) Regional
(2009) Cavernous malformations: natural history, diag- association of developmental venous anomalies with
nosis and treatment. Nat Rev Neurol 5(12):659–670 angiographically occult vascular malformations. Eur
13. Sohn CH, Kim SP, Kim IM et al (2003) Characteristic Radiol 6:30–37
MR imaging findings of cavernous hemangiomas in 29. Desai K, Bhayani R, Nadkarni T et al (2002) Develop-
the cavernous sinus. AJNR Am J Neuroradiol mental deep venous system anomaly associated with
24:1148–1151 congenital malformation of the brain. Pediatr
14. Ide C, De Coene B, Baudrez V (2000) MR features of Neurosurg 36:37–39
cavernous angioma. JBR-BTR 83(6):320 30. Boukobza M, Enjolras O, Guichard JP et al (1996)
15. Meyer FB, Lombardi D, Scheithauer B, Nichols DA Cerebral developmental venous anomalies associated
(1990) Extra-axial cavernous hemangiomas involving with head and neck venous malformations. AJNR Am
the dural sinuses. J Neurosurg 73(2):187–192 J Neuroradiol 17(5):987–994
16. Kondziolka D, Monaco EA, Lunsford LD (2013) Cav- 31. San Millán Ruíz D, Delavelle J, Yilmaz H et al (2007)
ernous malformations and hemorrhage risk. Prog Parenchymal abnormalities associated with develop-
Neurol Surg 27:141–146 mental venous anomalies. Neuroradiology 49:987–995
17. Pinker K, Stavrou I, Knosp E et al (2006) Are cerebral 32. Santucci GM, Leach JL, Ying J, Leach SD, Tomsick
cavernomas truly nonenhancing lesions and thereby TA (2008) Brain parenchymal signal abnormalities
distinguishable from arteriovenous malformations? associated with developmental venous anomalies:
MRI findings and histopathological correlation. Magn detailed MR imaging assessment. AJNR Am J
Reson Imaging 24(5):631–637 Neuroradiol 29(7):1317–1323
18. de Souza JM, Domingues RC, Cruz LC Jr, Domingues 33. Wurm G, Schnizer M, Fellner FA (2005) Cerebral
FS, Iasbeck T, Gasparetto EL (2008) Susceptibility- cavernous malformations associated with venous
weighted imaging for the evaluation of patients with anomalies: surgical considerations. Neurosurgery 57
familial cerebral cavernous malformations: a compari- (1 Suppl):42–58; discussion 42–58
son with T2-weighted fast spin-echo and gradient-echo 34. McLaughlin MR, Kondziolka D, Flickinger JC
sequences. AJNR Am J Neuroradiol 29(1):154–158 et al (1998) The prospective natural history of cerebral
19. Dósa E, Tuladhar S, Muldoon LL, Hamilton BE, venous malformations. Neurosurgery 43:195–200; dis-
Rooney WD, Neuwelt EA (2011) MRI using cussion 200–191
ferumoxytol improves the visualization of central ner- 35. Ku MG, Rhee DY, Park HS, Kim DN (2009) Repeated
vous system vascular malformations. Stroke 42 intracerebral hemorrhage from developmental venous
(6):1581–1588 anomaly alone. J Korean Neurosurg Soc 45:46–49
20. Zabramski JM, Wascher TM, Spetzler RF et al (1994) 36. Gama RL, Nakayama M, Tavora DGF, Bomfim RC,
The natural history of familial cavernous Carneiro TC, Pimentel LHC (2008) Thrombosed
malformations: results of an ongoing study. J developmental venous anomaly associated with cere-
Neurosurg 80:422–432 bral venous infarct. Arq Neuropsiquiatr 66
21. Campbell PG, Jabbour P, Yadla S, Awad IA (2010) (3-A):560–562
Emerging clinical imaging techniques for cerebral cav- 37. Peterson AM, Williams RL, Fukui MB, Meltzer CC
ernous malformations: a systematic review. Neurosurg (2002) Venous angioma adjacent to the root entry zone
Focus 29(3):E6. 1–15 of the trigeminal nerve: implications for management
22. Atlas SW (ed) (2008) Magnetic resonance imaging of of trigeminal neuralgia. Neuroradiology 44:342–346
the brain and spine, 4th edn. Lippincott Williams & 38. Paulson D, Hwang SW, Whitehead WE, Curry DJ,
Wilkins, Philadelphia Luerssen TG, Jea A (2012) Aqueductal developmental
32 Miscellaneous Vascular Malformations (Cavernous Malformations, Developmental Venous. . . 749

venous anomaly as an unusual cause of congenital 46. Finkenzeller T, Fellner FA, Trenkler J et al (2010) Cap-
hydrocephalus: a case report and review of the litera- illary telangiectasias of the pons: does diffusion-
ture. J Med Case Rep 6:7 weighted MR increase diagnostic accuracy? Eur J
39. Lee M, Kim MS (2012) Image findings in brain devel- Radiol 74:e112–e116
opmental venous anomalies. J Cerebrovasc Endovasc 47. Carpenter JS, Rosen CL, Bailes JE, Gailloud P (2004)
Neurosurg 14(1):37–43 Sinus pericranii: clinical and imaging findings in two
40. Lee RR, Becher MW, Benson ML et al (1997) Brain cases of spontaneous partial thrombosis. AJNR Am J
capillary telangiectasia: MR imaging appearance and Neuroradiol 25:121–125
clinic histopathologic findings. Radiology 48. Wen C, Chang Y, Wang H, Kuo M, Tu Y (2005) Sinus
205:797–805 pericranii: from gross and neuroimaging findings to
41. Castillo M, Morrison T, Shaw J, Bouldin T (2001) MR different pathophysiological changes. Childs Nerv
imaging and histologic features of capillary telangiec- Syst 21:482–488
tasia of the basal ganglia. AJNR Am J Neuroradiol 49. Vaquero J, de Sola RG, Martinez R (1983) Lateral
22:1553–1555 sinus pericranii. Case report. J Neurosurg 58:139–140
42. Huddle DC, Chaloupka JC, Sebgal V (1999) Clinically 50. Lee CH, Lee YS, Lee JH, Lee HG et al (2009) Sinus
aggressive diffuse capillary telangiectasia of the brain pericranii: a case report and the literature review. Kor J
stem: a clinical radiologic-pathologic case study. Cerebrovasc Surg 11:174–178
AJNR Am J Neuroradiol 20:1674–1677 51. Hayakawa I, Fujiwara K, Sasaki A et al (1978) Spon-
43. El-Koussy M, Schroth G, Gralla J, Brenkenfeld C, taneous regression of sinus pericranii: report of a case
Andres RH, Jung S et al (2012) Susceptibility- (in Japanese). No Shinkei Geka 6:91–95
weighted MR imaging for diagnosis of capillary telan- 52. Sheu M, Fauteux G, Chang H et al (2002) Sinus
giectasia of the brain. AJNR Am J Neuroradiol pericranii: dermatologic considerations and literature
33:715–720 review. J Am Acad Dermatol 46:934–941
44. Bland LI, Lapham LW, Ketonen L, Okawara SH 53. Vinas FC, Valenzuela S, Zuleta A (1994) Literature
(1994) Acute cerebellar hemorrhage secondary to cap- review: sinus pericranii. Neurol Res 16:471–474
illary telangiectasia in an infant: a case report. Arch 54. Kim YJ, Kim IO, Cheon JE, Lim YJ, Kim WS, Yeon
Neurol 51:1151–1154 KM (2011) Sonographic features of sinus pericranii in
45. Barr RM, Dillon WP, Wilson CB (1996) Slow-flow 4 pediatric patients. J Ultrasound Med 30:411–417
vascular malformations of the pons: capillary telangi-
ectasias? AJNR Am J Neuroradiol 17:71–78
Miscellaneous Vascular Diseases
33
Matylda H. Machnowska and Richard I. Aviv

Contents Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 763


MRI Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 763
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 752
CARASIL . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 763
Fabry’s Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 752
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 752 Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 764
Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 752 PACNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 764
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753 Takayasu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
CT Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753 Giant Cell Arteritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
MRI Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753 Polyarteritis Nodosa . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
Kawasaki Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
PRES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 753 Churg-Strauss Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 754 Wegener’s . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 770
Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 754
Imaging Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 755 Systemic Lupus Erythematosus . . . . . . . . . . . . . . . . . . . 771
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 771
RCVS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 756 Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 756 Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
Clinical . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 756 Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
Moyamoya . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 758 Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 758 Special Procedures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 773
Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 759 Infectious Vasculitides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 773
Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 759 Varicella Zoster Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . 773
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 759 HIV Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 773
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760 Bacterial Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 774
Cerebral Autosomal Dominant Tuberculous Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 774
Arteriopathy with Subcortical Infarcts Fungal Vasculitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 774
and Leukoencephalopathy (CADASIL) . . . . . . . . . . 762 Neuro-Behcet’s Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 775
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 762 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 776
Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 762
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 777

M.H. Machnowska (*) • R.I. Aviv


Division of Neuroradiology, Department of Medical
Imaging, School of Medicine, Sunnybrook Health
Sciences Centre, University of Toronto, Toronto,
ON, Canada
e-mail: Matylda.machnowska@sunnybrook.ca;
Richard.aviv@sunnybrook.ca

# Springer Science+Business Media New York 2016 751


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_23
752 M.H. Machnowska and R.I. Aviv

Abstract
Fabry’s Disease
This chapter discusses a variety of inflamma-
Fabry’s disease is a multisystemic X-linked lyso-
tory, infectious, and not otherwise easily cate-
somal storage disorder which results from muta-
gorized diseases that can affect the intracranial
tions in the α-galactosidase A gene (GLA) leading
vessels. These include diseases with known
to deficiencies of α-galactosidase A.
genetic mutations such as Fabry’s disease,
cerebral autosomal-dominant arteriopathy
with subcortical infarcts and leukoence-
Epidemiology
phalopathy (CADASIL), and cerebral
autosomal recessive arteriopathy with
Estimates of the prevalence of Fabry’s disease in
subcortical infarcts and leukoencephalopathy
younger stroke patients (<55 years) range up to
(CARASIL); those arising as a result of genetic
4 %. A recent prospective study including the
and environmental factors such as moyamoya;
largest cohort of patients to date reported a lower
and those with yet unknown etiologies such as
prevalence of 0.5 % overall (0.8 % in females and
Takayasu’s, posterior reversible encephalopa-
0.4 % in males) [1]. A more conservative estimate
thy syndrome (PRES) and Behcet’s disease. In
of disease prevalence is also reflected in a recent
each case, the preferred imaging methods and
study of young patients with expected neurologic
characteristic imaging findings are discussed.
hallmarks of the disease, including not only stroke
We concentrate on the standard imaging
but also transient ischemic attack, intracranial
methods of MRI, cerebral angiography, and
hemorrhage, unexplained white matter lesions,
noninterventional angiography. Where appli-
and vertebrobasilar dolichoectasia [2].
cable, advanced imaging techniques including
Although an X-linked disorder, lyonization in
cerebrovascular reactivity, vessel wall imag-
heterozygous females includes presentations
ing, perfusion imaging, and MR spectroscopy
ranging from asymptomatic to symptom severity
are discussed.
similar to males. Generally, disease in females is
characterized by less kidney involvement but sim-
ilar prevalence of stroke or TIA. Patients usually
Keywords
have their first stroke between 20 and 50 years of
Angiography • Functional imaging • Vessel
age, with males presenting a decade earlier than
wall imaging • Autoimmune vasculitis •
females.
RCVS • PRES • Infectious vasculitis •
Takayasu’s • Moyamoya • CADASIL
Clinical Presentation

Introduction Early clinical symptoms include acroparesthesia,


present in 60–80 % of classically affected chil-
This chapter discusses a plethora of autoimmune, dren, waning in adulthood; anhidrosis leading to
infectious, and not easily categorized CNS and heat and exercise intolerance; angiokeratoma;
systemic diseases, which result in neurological corneal opacities; gastrointestinal symptoms
symptoms primarily as a result of their effects on including abdominal pain, diarrhea, and vomiting;
the intracranial vasculature. We aim to review the and tinnitus. Cardiac, renal, and central nervous
current clinical and pathological understanding of system abnormalities often present outside of
the diseases and to discuss standard, and, where childhood. Affected adolescent males have
applicable, advanced imaging methods currently shown decreased heart rate variability suggesting
employed in the clinical setting. autonomic dysfunction. Cardiac findings in adults
33 Miscellaneous Vascular Diseases 753

include left ventricular hypertrophy, arrhythmia, periventricular white matter. There is evidence of
and angina. Renal manifestations include end-arterial ischemic lesions, more prominent in
microalbuminuria and proteinuria in the second the posterior territories. Dystrophic calcification is
and third decades progressing to azotemia in the seen in the basal ganglia, pulvinar of the thalamus,
fifth to sixth decades. Symptoms due to CNS and subcortical arcuate fibers of the white
involvement, including headache, vertigo, cogni- matter [6].
tive impairment, and ischemic strokes, usually
present in adulthood.
MRI Findings

Pathophysiology Structural MRI findings show T2 hyperintense


white matter lesions corresponding to the distri-
Artery-associated complications give rise to bution seen on CT. Ischemic lesions, predomi-
nephropathy and CNS. The etiology of the nantly in the vertebrobasilar territory, are also
vasculopathy is not entirely clear, although sev- seen. Pulvinar T1 hyperintensity has been
eral mechanisms have been postulated. These suggested as a very specific finding for Fabry’s
include deposition of glycolipid moieties, such disease, present in about 30 % of patients over the
as globotriaosylceramide (Gb3) in the lysosomes age of 50 [6]. This pulvinar T1 hyperintensity
of smooth muscle and endothelial cells; an does not show signal loss on fat-saturated imag-
increase in prothrombotic factors and inflamma- ing. Additionally, signal drop seen in the pulvinar
tory mediators; and altered cerebrovascular flow. on susceptibility weighted imaging corresponds
In addition to lysosomal deposition, Gb3 is also with the degree of calcification on CT suggesting
found in Schwann cells, in dorsal root ganglia, that mineralization of the pulvinar is responsible
and in CNS neurons. for pulvinar T1 hyperintensity (Fig. 1). The intra-
Fabry patients show increased soluble cranial vessels are dolichoectatic [7].
intercellular adhesion molecule-1 (sICAM-1), Several studies have shown increased white
soluble vascular cell adhesion molecule-1 matter diffusivity on diffusion-weighted and dif-
(sVCAM-1), and macrophage 1 antigen MAC-1 fusion tensor imaging, predating structural
levels suggesting an enhanced inflammatory state. changes. A recent MR study comparing white
Increased levels of plasminogen activator inhibi- matter lesion load, white matter diffusivity
tor, and reduced thrombomodulin concentrations changes, and arterial diameters, but not the T1
compared to controls, suggest an enhanced coag- hyperintense pulvinar sign, has shown that
ulant state [3]. increased basilar artery diameter is the best dis-
Several PET studies have shown evidence of criminator between patients with Fabry’s disease
cerebral hyperperfusion, particularly in the poste- and normal controls [8].
rior cerebral circulation [4]. This is in contradis-
tinction to classic leukoaraiosis, which is usually
caused by hypoperfusion. Studies have also
shown disordered autoregulation which may con- PRES
tribute to the white matter disease [5].
The term posterior reversible encephalopathy syn-
drome is a clinical and radiologic syndrome with
CT Findings specific imaging findings, characterized by
reversible, mostly posterior territory, vasogenic
Hypodensities, corresponding to white matter edema, which is the end point of a myriad of
lesions, can be seen in the deep and different etiologies.
754 M.H. Machnowska and R.I. Aviv

Fig. 1 Sagittal T1-WI and


SWI images (a, b) showing
pulvinar T1 hyperintensity
and SWI hypointensity
(arrowheads) in a 30-year-
old male with Fabry’s
disease (Images courtesy of
D. Mandell)

Other names used to describe the clinical or showing that about 30 % of patients who develop
radiologic syndrome include hypertensive symptoms and imaging findings compatible with
encephalopathy and reversible posterior PRES are not hypertensive or demonstrate cere-
leukoencephalopathy. Most authors prefer the bral hypoperfusion. Further, several of the under-
term posterior reversible encephalopathy because lying diseases or agents associated with the
up to 30 % of patients are not hypertensive at development of PRES affect endothelial cell
presentation, and imaging changes are often also function [9].
present in the cortex and not limited to the white
matter. Symptoms include headaches, decreased
alertness, altered mental functioning, seizures, Pathology
and visual loss occasionally attributable to cortical
blindness. The white matter and cortical edema tends to
The clinical and imaging findings have concentrate in the parieto-occipital regions.
been associated with preeclampsia/eclampsia, Pathology reports of biopsy specimens have
posttransplant disorders, renal failure, immune shown findings of white matter vacuolation, con-
suppression, infection and sepsis, autoimmune sistent with white matter edema often with mild
disease, chemotherapeutic agents, and numerous inflammatory reaction and, less commonly, find-
other miscellaneous conditions. ings of neuronal necrosis and petechial hemor-
rhages [10]. Autopsy specimens have shown
fibrinoid necrosis of arterioles with thrombosis
Pathophysiology of arterioles and capillaries, as well as parenchy-
mal microinfarcts and petechial hemorrhages
Although several theories of PRES etiology thought to be secondary to vascular changes.
exist, two main hypotheses are likely. Occasionally, pathology specimens have revealed
The original theory suggests that hypertension evidence of white matter pallor, attributed to
leads to autoregulatory vasoconstriction and demyelination and astrocytosis [11].
hypoperfusion, ischemia, and edema. A more More recent pathology reports have revealed
popular theory suggests that severe hypertension evidence of perivascular T-cell trafficking, endo-
results in breakthrough of the autoregulatory thelial activation, endothelial immunoreactivity to
mechanism leading to edema. tumor necrosis factor-alpha, robust endothelial
There has been a recent resurgence of interest VEGF mRNA transcription, and VEGF protein
in the original theory, as a result of several studies expression [9].
33 Miscellaneous Vascular Diseases 755

Imaging Findings often seen. Thalamic, basal ganglia, and


brainstem involvement has been described.
CT Findings are most commonly associated with
CT typically demonstrates focal regions of sym- hypointensity or isointensity on diffusion-
metric, predominantly subcortical white matter weighted imaging with concomitantly increased
parieto-occipital hypodensity. The frontal lobes, ADC values. Foci of predominantly cortical dif-
cerebellum, and inferior temporal-occipital lobes fusion restriction have been described. These may
may also be affected. The distribution does not represent ischemia and irreversible injury. Revers-
conform to a vascular territory but is often seen in ible cortical diffusion restriction has also been
watershed regions. The cortex may be involved. described in the setting of seizure activity, pre-
sumably related to excitotoxic edema [12] (Fig. 2)
MRI Contrast enhancement has been shown in
MRI shows T2 prolongation in the subcortical 16–30 %. Petechial and subarachnoid hemor-
white matter and cortex almost always involving rhages (SAH) occur in 15 % of lesions
the parieto-occipital lobes. Isolation solely to the [10]. FLAIR sulcal hyperintensity may be second-
posterior brain regions is rare, however, and ary to SAH but can also occur secondary to pro-
involvement of other brain regions including the teinaceous exudate. Diffusion tensor imaging has
frontal lobes, temporal lobes, and cerebellum is revealed regions of reversible increased
anisotropy [13].

Fig. 2 PRES encephalopathy. (a, b) Hyperintensity is confirms vasogenic edema in the distribution of T2
seen on T2-WI images with subcortical and cortical hyperintensity (black arrow). (f) MRV is normal (Images
involvement in the parieto-occipital regions (arrowheads). courtesy of M. Shroff)
(c, d) DWI is normal due to T2 wash out. (e) ADC map
756 M.H. Machnowska and R.I. Aviv

Catheter Angiography with acute onset, severe headaches and may be


Angiographic studies range from normal to find- accompanied by other neurological signs. Spon-
ings of vasospasm evidenced by arterial taneous resolution of the vasoconstriction is usu-
narrowing that can be peripheral, central, or dif- ally seen in 1–3 months [17]. In its broadest
fuse. Arterial beading has also been described. definition, the term encompasses disorders of
Peripheral vasospasm is often difficult to charac- numerous etiologies characterized by reversible
terize on MRA/CTA imaging and is better visual- vasospasm which do not show histologic hall-
ized with catheter angiography [14]. marks of vasculitis. RCVS has been known in
the literature by multiple names including Call-
Perfusion Studies Fleming syndrome, benign angiopathy of the
CTP studies, performed in hypertensive patients, central nervous system, thunderclap headache
have shown increased cerebral blood flow, with reversible vasospasm, and migrainous
increased cerebral blood volume, and decreased angiopathy. Reversible vasospasm in the setting
mean transit time allowing for differentiation of known etiologic factors such as drug-induced
between PRES and cerebrovascular accident [15]. vasculopathy, hypercalcemia, unruptured saccular
Hypoperfusion, however, has been demon- aneurysm, dissection, postcarotid endarterectomy,
strated using Tc99m-HMPAO SPECT in women and postpartum angiopathy is also included in the
with eclampsia, performed within 48 h of umbrella term of RCVS [17]. Known etiologic
delivery [16]. factors account for up to 25–60 % of cases of
Several recent MR perfusion studies have also RCVS [18].
shown decreased perfusion in PRES [14]. Find-
ings have generally shown decreased cerebral
blood flow and volume in affected regions, with Epidemiology
some variability of mean transit time.
The discrepancy of these perfusion findings is The age of onset of RCVS peaks at around
not well understood. It may relate to the timing of 45 years of age and is more common in females,
the perfusion study with respect to disease onset. with a 2.6:1 preponderance in a French study to
Animal experiments have shown that vasocon- 10:1 in Taiwanese study [18].
striction occurs within the limits of
autoregulation, at which point there is break-
through of the autoregulatory mechanism Clinical
resulting in hyperperfusion and breakdown of
the blood-brain barrier. In animal studies, this Presentation is similar to aneurismal rupture and
has been followed by vasodilation and characterized by rapid onset thunderclap head-
hypoperfusion. Alternatively, authors postulate ache, peaking in less than 1 min and recurring
that abnormal radiopharmaceutical uptake could over 1–2 weeks.
represent luxury perfusion in the setting of loss of Diagnostic criteria were defined by Calabrese
autoregulation and vascular tone, rather than true and include: [17]
hyperperfusion.
1. Transfemoral angiography or indirect (CT or
MRI) angiography documenting segmental
cerebral artery vasoconstriction.
RCVS 2. No evidence of aneurismal subarachnoid
hemorrhage.
Reversible cerebral vasoconstriction syndrome 3. Normal or near-normal cerebrospinal fluid
(RCVS) is defined as a group of disorders that analysis (protein level <80 mg/dL, white
result from prolonged but reversible vasoconstric- blood cell count <10/μL, normal glucose
tion of the cerebral arteries. It is usually associated level).
33 Miscellaneous Vascular Diseases 757

Fig. 3 Reversible cerebral vasoconstriction syndrome. A results revealed no evidence of an inflammatory process.
36-year-old female presented with an acute thunderclap The patient was diagnosed with RCVS, and steroids were
headache. (a) CT shows occipital parenchymal (arrow) withheld. (d) MRI T2-WI study performed 3 months later
and subarachnoid hemorrhage. (b, c) CTA performed the shows residual hemosiderin deposition in the left occipital
same day shows beading within middle cerebral artery lobe. (e) Concurrent MRA shows complete resolution of
(MCA) and anterior cerebral artery (ACA) territories arterial narrowing
(arrowheads). Lumbar puncture and serum biochemistry

4. Severe, acute headache, with or without addi- Occasionally, there may be a small amount of
tional neurological signs or symptoms. subarachnoid hemorrhage overlying the cortical
5. The diagnosis cannot be confirmed until revers- surface or in the parenchyma. In 10–39 % of
ibility of the angiographic abnormalities is patients, findings may result in ischemic or hem-
documented within 12 weeks of onset. If death orrhagic parenchymal strokes [19] (Fig. 3a, d).
occurs before the follow-up studies are com- Reversible brain edema, compatible with PRES,
pleted, autopsy should rule out conditions such is seen in about 10 % of patients [19].
as vasculitis, intracranial atherosclerosis, and CTA and MRA usually show segmental
aneurismal subarachnoid hemorrhage, which beading and intermittent dilatation of vessels
can also manifest with headache and stroke. (string of beads) [17] (Fig. 3b, c). Initial
noninvasive angiography may be negative or
CSF analysis must be performed to rule out show distal vessel irregularities. Maximum
subarachnoid hemorrhage in patients presenting vessel constriction involving the proximal
with thunderclap headache, with normal imaging arterial beds usually occurs 18–22 days following
findings. CT and MRI are usually normal. headache onset, often at the time of headache
758 M.H. Machnowska and R.I. Aviv

resolution, and may persist for several weeks. Moyamoya


This has led some authors to speculate that it is
actually the vascular dilation, and not constric- Moyamoya syndrome is an occlusive cerebrovas-
tion, which is responsible for pain and microvas- cular disease which affects the terminal internal
cular rupture resulting in hemorrhage [20]. The carotid arteries and proximal circle of Willis ves-
gold standard in diagnosis remains catheter angi- sels. Associated engorgement of the lenticulostriate
ography, which has shown a sensitivity of 100 % and thalamoperforating leads to the moyamoya or
compared to noninvasive methods (MRA and “puff of smoke” appearance on angiography.
CTA) which showed sensitivities of around Moyamoya disease refers to the idiopathic form
75 % [19]. of the arteriopathy; moyamoya syndrome refers to
Recent studies have shown an association an association with other pathologic processes
between RCVS and vertebral or carotid artery including neurofibromatosis, sickle cell anemia,
dissection, particularly in postpartum females, Down syndrome, and prior cranial irradiation.
and it may be prudent to screen the neck vessels,
particularly in patients who present with neck
pain [19]. Epidemiology
The foremost mimicker of RCVS is primary
angiitis of the CNS (PACNS), while the clinical Incidence of moyamoya disease in Japan has
symptoms are usually more gradual in onset in range up to 0.94/100,000 of the population, with
angiitis. Headache is usually subacute and pro- increasing incidence reported in recent years
gressive, rather than thunderclap type, and may attributed to better diagnostic measures
be accompanied by transient neurological deficits [22]. Rates of moyamoya syndrome in the United
eventually progressing to stroke. Asymptomatic States and Europe are generally one tenth those in
infarcts are often seen on initial MR studies. Japan [23]. The disease has a bimodal distribution
Angiographic findings may lead to clinical confu- with peaks in the first and fourth decades of life
sion due to their similarities in the two [23]. Both symptomatic and asymptomatic forms
pathologies. of the disease have a slight female to male pre-
Distinguishing features are that in at least 70 % dominance of about 2:1 [22].
of cases, MRI will be normal in RCVS, while it Children generally present with ischemic
will be abnormal in PACNS in up to 90 % of events such as TIAs or strokes, while adult
cases showing multiple, deep, or superficial patients more commonly present with hemor-
infarcts of different ages. CSF analysis should be rhage. The disease accounts for 6 % of strokes
normal in RCVS and abnormal in over 95 % of in children [24].
PACNS cases. Irregular, eccentric, and asymmet- Clinical associations with moyamoya include
ric occlusions without interspersed dilations skull base, sellar, and suprasellar tumors and optic
are more suggestive of a vasculitis rather than nerve gliomas either idiopathically or as a result of
RCVS [18]. However, such subtle angiographic radiation treatment, Fanconi anemia, sickle cell
differences may be difficult to appreciate, anemia and other hemoglobinopathies, neurofi-
particularly due to the limits in spatial resolution bromatosis type I, progeria, inflammatory factors
of noninvasive angiography. Vessel wall such as anticardiolipin syndrome, protein S eleva-
imaging, which utilizes a single inversion tion, SLE, neuro-Behcet’s, Kawasaki disease;
recovery-prepared fast spin echo black blood collagen vascular disorders such as Marfan, con-
T1-WI sequence prior to and following genital cardiac disease, renal artery stenosis,
contrast administration, has shown promise in infectious diseases such as tuberculosis,
distinguishing the two entities [21]. Vessel wall leptospirosis, tonsillitis, atherosclerosis, and
enhancement is seen with inflammatory fibromuscular dysplasia (Fig. 4). These clinical
processes affecting the vessel lumen and is absent associations can be seen in the setting of unilateral
in RCVS. disease.
33 Miscellaneous Vascular Diseases 759

Forty to 65 % of adults present with hemor-


rhage usually localized to the basal ganglia and
ventricular system. Hemorrhages are often recur-
rent and more common in females [27].
Hemodynamic studies, discussed below, have
shown a decreased cerebral blood flow in children
with moyamoya in comparison to age-matched
controls and adults with the disease, an increased
cerebral blood volume suggesting a chronic state
of vasodilation, and an increased oxygen extrac-
tion fraction and significantly impaired cerebro-
vascular reactivity [28]. These findings may help
to explain why ischemia is the most common
presenting symptom in children.

Pathophysiology
Fig. 4 Conventional angiogram showing left MCA steno-
sis in a child with neurofibromatosis representing Moyamoya disease is characterized by complex
moyamoya syndrome (Courtesy of M. Shroff, G. DeVeber)
channels of medium- and small-sized muscular
arteries branching off the circle of Willis, anterior
Genetics choroidal arteries, internal carotid arteries, and
posterior cerebral arteries and connecting to the
The familial form of disease accounts for about distal portions of the anterior and middle cerebral
15 % of patients. arteries. These channels give rise to numerous,
To date one gene locus has been implicated in dilated or stenotic, tortuous perforating vessels
moyamoya: the ring finger protein 213 (RNF213) that correspond to the lenticulostriate and
gene located on chromosome 17q has been iden- thalamoperforating vessels. Dilated vessels are
tified as a susceptibility gene for moyamoya dis- more common in younger patients, while stenotic
ease. Other gene foci identified by chromosomal vessels in older patients.
linkage analyses include 3p24.2-26, 6q25, 8q23, Microscopic findings in stenotic vessels are
and 12p12. The inheritance pattern is thought to characterized by intimal thickening, thickening
be autosomal dominant with incomplete and duplication of the elastic lamina, fibrosis of
penetrance [25]. the tunica media, and fibroelastosis of the wall.
The media is thinned, and there is a decrease in the
external diameter of the vessel [29]. Dilated ves-
Clinical Presentation sels show fibrosis of the media, attenuation of the
wall, and moderate increase in elastic fibers
Children with moyamoya typically present with [29]. Lipid deposits have been found, but are
evidence of TIAs (40 %) or infarction (40 %) thought to relate to atherosclerosis.
[26]. Motor disturbances vary from weakness to Hemorrhage is attributed to rupture of the per-
paralysis of the extremities. Other presentations forating vessels, aneurismal rupture, or very rarely
include headaches, seizures, and decreasing rupture of dilated collateral arteries on the brain
cognitive function. Paroxysmal symptoms have surface. Frequency of intracranial aneurysms is
been seen in the setting of elevated body temper- increased in patients with moyamoya. Aneurysms
atures, exercise or episodes of crying, and hyper- can be saccular or dissecting and occur centrally,
ventilation leading to hypocapnia and most frequently in the basilar arteries or
vasoconstriction. peripherally [29].
760 M.H. Machnowska and R.I. Aviv

Imaging hemorrhage or atrophy and infarction in the ante-


rior circulation territory.
Angiography
Catheter angiography remains the gold standard CTA
in moyamoya diagnosis. While MRA is an accept- CTA is a powerful tool in the diagnosis of
able alternative in diagnosing moyamoya, cathe- suspected vascular abnormalities in the setting of
ter angiography is recommended in adult surgical intracerebral hemorrhage. It can be used for the
candidates, with pediatric cases considered on an assessment of intracranial stenoses and the identi-
individual basis [30]. The vascular picture is char- fication of aneurysms and areas of contrast
acterized by stenosis of the supraclinoid distal enhancement. It has also been used as a method
ICA, the proximal MCA and ACA branches, and for neurosurgical planning and assessment of
the development of dilated striate arteries. EC-IC bypass.
Involvement of the posterior cerebral artery
(PCA) is seen in about 30 % of patients, usually MRI
at the late stage of disease. MRI findings include atrophy, particularly in the
Six angiographic stages are described: [31] anterior circulation territory. Evidence of prior
infarcts or hemorrhage can be seen on FLAIR and
1. Narrowing of the carotid termination. gradient-recalled echo imaging. Asymptomatic
2. Dilation of the proximal ACA and MCA with microbleeds, in the periventricular deep white mat-
initial moyamoya blush and dilation of the ter, have been seen in up to 44 % of patients [32].
perforating moyamoya vessels. Diffusion-weighted imaging will detect acute
3. Stenosis of the ACA and MCA with intensifi- infarcts. The flow voids at the skull base should be
cation of the moyamoya blush. carefully scrutinized for evidence of ICA and
4. Minimization of the moyamoya blush with exten- proximal ACA/MCA diminution. Increased flow
sion of internal carotid artery (ICA) occlusion to voids can also be seen within the basal cisterns,
the junction of the posterior communicating artery basal ganglia, and around the circle of Willis
(PComm). The constituent vessels are thinned, consistent with increased collateral circulation
and there is a concomitant increase in the collat- (Fig. 5a).
eral vessels from the extracranial carotid arteries. Increased intersulcal FLAIR signal
5. Further reduction of the moyamoya and com- corresponding to increased contrast in the sub-
plete disappearance of the main arteries arising arachnoid space on enhanced T1-WI images has
from the ICA. Occlusion of the ICA can reach been termed the “ivy sign” and is felt to represent
as far as C2. Persisting increase in ECA collat- slow flow in dilated pial collaterals (Fig. 5b).
eral circulation.
6. Disappearance of the moyamoya, with intra- MRA
cranial supply from the ECA and potentially According to the guidelines for diagnosis and
vertebrobasilar system. treatment of moyamoya disease, MRA can pro-
vide a definitive diagnosis when the following
Angiography can be used to identify aneu- findings are fulfilled on time of flight (TOF) imag-
rysms, define a specific transdural anastomotic ves- ing using a scanner of at least 1.5-T.
sel, and, in very rare cases, identify other vascular
anomalies. Investigation of the ECA system for 1. Stenosis or occlusion of the terminal portion of
stenoses is also recommended and may have impli- the intracranial internal carotid artery or prox-
cations for treatment success rates [30] (Fig. 5). imal portion of the anterior and/or middle cere-
bral arteries.
CT 2. Abnormal vascular networks in the basal
Unenhanced CT may be completely normal or ganglia. When two or more visible flow voids
show subarachnoid and intraparenchymal in the basal ganglia are present at least
33 Miscellaneous Vascular Diseases 761

Fig. 5 Moyamoya. (a, b)


Axial FLAIR images show
multiple flow voids in the
basal ganglia (white
arrowheads), white matter
leukoaraiosis in the
watershed regions, and the
ivy sign in the right
hemispheric sulci (arrow).
(c) MRA shows numerous
leptomeningeal and
lenticulostriate collaterals
(black arrowheads), which
show retrograde filling of
the distal right MCA on
conventional angiography,
(d) (Courtesy of M. Shroff,
G. DeVeber)

unilaterally on MRI, they can be deemed as of cerebrovascular reactivity which is defined as a


representing an abnormal vascular network. change in blood flow per unit change in stimulus,
3. Bilaterality of findings 1 and 2. such as carbon dioxide. In the setting of a down-
stream stenosis, maintenance of normal cerebral
In cases of unilaterality, the guideline authors blood flow is possible due to a relaxation of vas-
consider a quasi-moyamoya or moyamoya cular tone, resulting in normal baseline perfusion
syndrome [33]. parameters. Analogous to the myocardial stress
test, a cerebrovascular reactivity study uncovers
Functional and Hemodynamic Studies the compensatory vasodilatory state by stressing
In general, hemodynamic studies show decreased its limits. The strengths and limitations of the
cerebral blood flow, increased mean transit time, currently utilized functional and hemodynamic
and increased cerebral blood volume in the studies are discussed below.
affected hemisphere, coupled with an increased PET is the only imaging modality that can
oxygen extraction fraction [30]. The cerebral met- measure oxygen extraction fraction and the cere-
abolic rate of oxygen is often decreased, reflecting bral metabolic rate of oxygen consumption. Its
the influence of limited cerebral blood flow, par- limitations include the need for an on-site cyclo-
ticularly in ischemic cases [34]. Many perfusion tron due to the rapid decay of 15O (2 min half-life)
studies are performed at baseline and following a and a PET scanner, the availability of which is less
vasodilatory challenge using acetazolamide or than MR, CT, or SPECT. The whole body radia-
end-tidal CO2 manipulation. The use of a tion exposure is 0.5–2.0 mSv, depending on the
vasodilatory challenge allows for the calculation quality of the PET camera and protocol. Increased
762 M.H. Machnowska and R.I. Aviv

oxygen extraction fraction is linked to progression effects of deoxyhemoglobin normally cause


of disease and reversed with EC-IC bypass. dephasing of protons in local tissues, reducing
CT and MR are relatively easily accessible, but the returned BOLD signal. This method not only
technically difficult in moyamoya patients due to shows patients in whom CVR is exhausted but
the lack of availability of a reliable arterial input also those with a paradoxical response or steal
function in the diseased anterior circulation vessels, phenomenon in patients with uncompensated
occasionally necessitating the placement of ROIs moyamoya disease. CVR defects may also reverse
in posterior circulation vessels [35]. The presence with EC-IC bypass.
of multiple diseased vessels precludes the accurate Proton MR spectroscopy has shown reduced
comparison of concentration time curves derived levels of choline, creatine, and N-acetylaspartate
from one major vessel to those derived in the in the affected brain of moyamoya patients with
microvascular territories supplied by several differ- relative increase 6 months following revasculari-
ent diseased vessels. Flow maps will only be accu- zation surgery [39].
rate for the territory of the vessel in which the
arterial input function was positioned, but not for
those tissues fed by other diseased vessels [36]. A Cerebral Autosomal Dominant
recent study of 16 patients showed poor angio- Arteriopathy with Subcortical Infarcts
graphic stage correlation with normalized baseline and Leukoencephalopathy (CADASIL)
CT perfusion parameters (with respect to ipsilat-
eral cerebellar hemisphere), but significant corre- Cerebral autosomal-dominant arteriopathy with
lation with percent changes of CBF during an subcortical infarcts and leukoencephalopathy is a
acetazolamide challenge [37]. genetic small artery disease that leads to dementia
SPECT uses the single-pass lipophilic isotopes and early death.
99m
technetium and 123iodine that cross the blood- Its main clinical features include migraine with
brain barrier. The use of single-pass isotopes permits aura, ischemic events, mood disturbances, and
an estimation of blood flow, due to first pass depo- cognitive impairment. Other presentations such
sition through the microcirculation, without provid- as apathy or seizures have also been described.
ing quantitative measurements. Measurements are Dementia and dependence are prominent features
weighted to tracer delivery/deposition, but immune of the end stages of the disease [40].
to the input function problem of CT and MR perfu-
sion studies. While technetium isotope use alone
requires a 24 h interval between baseline and acet- Epidemiology
azolamide challenge, the combination of technetium
and iodine allows the study to be performed in a The prevalence of the disease is reported as 2–4
single sitting using separate windowing for the per 100,000. The mean age of disease onset is in
99m
technetium and 123iodine keV [27]. the mid third decade, with earlier discovery of the
A recently developed MRI technique uses disease in patients presenting with migraine
blood oxygenation level-dependent contrast (mean age 28 years) than those with stroke
(BOLD) imaging while manipulating the (mean age 41 years) [41]. The median age of
end-tidal CO2 (ETCO2) through a rebreathing death is reported as 65 years for males and
circuit [38]. This method does not rely on input 71 years for females, with 80 % of patients depen-
functions or pharmacologic agents and provides dent by the time of death [40].
spatial and quantitative maps of percent BOLD
signal change per mm Hg change in ETCO2. It can
be performed on all 1.5 and 3-T MRI systems with Genetics
echoplanar capability. In normal tissues, BOLD
signal increases with hypercapnea due to a wash- CADASIL is a genetic disorder due to mutations
out of deoxyhemoglobin. The paramagnetic in the extracellular domain of the Notch 3 gene
33 Miscellaneous Vascular Diseases 763

Fig. 6 CADASIL Axial


FLAIR images show
widespread T2 signal
hyperintensity in the
periventricular, deep, and
subcortical white matter,
volume loss, temporal pole
involvement (arrows), and
subcortical lacunar infarcts
(arrowhead)

located on chromosome 19. The gene contains Microbleeds are seen in 25–69 % of patients
33 exons, but only mutations in exons 2–24 and were present most frequently in the thalami,
occur in CADASIL, 60 % occurring in exons cortical/subcortical regions, white matter, pons,
3 and 4. and basal ganglia [44, 45]. Number does not cor-
relate with white matter lesion burden, but is
increased with age. Similarly, dilated Virchow-
Pathology Robin spaces, mainly in the lentiform nuclei and
subcortical white matter of the temporal lobes, are
Macroscopic examination shows diffuse myelin seen significantly more frequently in CADASIL
pallor sparing the U-fibers. Lacunar infarcts are patients than age-matched controls and show a
seen in the white matter and basal ganglia. Pontine correlation with increasing age, not white matter
involvement is most common in the brainstem, lesion burden [46].
with relative sparing of the medulla. Microscopic Recent studies have shown a correlation with
examination reveals a classic non-amyloid granu- the disease burden at diagnosis and severity of
lar osmiophilic material in the media extending progression [44].
into the adventitia of small- and medium-sized
leptomeningeal arteries.
Fibrosis of the adventitia and sometimes dupli-
cation and hyalinosis of the lamina are described. CARASIL
Loss of vascular smooth muscle cells occurs but to
a lesser extent than in Binswanger’s dementia Cerebral autosomal recessive arteriopathy with
[42]. Apoptosis in layers 3 and 5 of the cortex is subcortical infarcts and leukoencephalopathy is a
associated with increased severity of white matter recently recognized single gene disorder caused
lesions [43]. by mutations in the HTRA1 gene. Its prevalence is
currently unknown, with approximately
50 patients described in the literature to date,
MRI Imaging mostly from Japan with a 3:1 male to female ratio.
Its main clinical manifestations are ischemic
MR imaging shows areas of FLAIR stroke, progressive dementia, early alopecia, and
hyperintensity in the white matter, pons, and spondylosis deformans. Ischemic stroke occurs in
external capsules (Fig. 6). about half of patients and the onset of dementia is
764 M.H. Machnowska and R.I. Aviv

in the fourth decade (mean 32 years). Alopecia, Clinical Symptoms


seen in 90 % of patients, usually presents in the Most frequent symptoms include headache,
third decade, but may be seen in adolescence. altered cognition, focal neurological manifesta-
Low back pain is seen in 80 % of patients, with tions, stroke, and visual symptoms [48]. Onset is
an onset between 20 and 40 years. usually gradual and progressive.
CT shows diffuse white matter hypodensity, The largest series includes 101 patients, show-
volume loss, and corticospinal tract Wallerian ing no statistically significant correlation with
degeneration. Similarly, MRI studies show T2 male gender and increased mortality rate or higher
hyperintense periventricular and deep white mat- disability scores (modified Rankin scale 4–6).
ter lesions, with some sparing of U-fibers. Lesions Instead, presentation with a focal neurological
subsequently extend into basal ganglia, thalami, deficit rather than headache, cognitive impairment
brainstem, and cerebellum. Temporal lobe versus headache or constitutional symptoms,
involvement may be seen, similar to cerebral infarction versus no infarction, and
CADASIL [47]. large-vessel versus small-vessel involvement sig-
nificantly correlated with increased mortality.
The disease in children may more often present
with seizures [49].
Vasculitis
Pathology
PACNS Biopsy of the brain or spinal cord is considered
the gold standard. Biopsy results are often nega-
Primary angiitis of the CNS (PACNS) is a rare, tive in 25–50 % of cases [48]. Including
potentially severe, form of vasculitis that involves leptomeninges in the biopsy results in slightly
the small- (200–300 μm in diameter) and medium- higher diagnostic yields [49].
sized (500 μm) vessels of the brain and spinal Three patterns are recognized: granulomatous,
cord. PACNS is also referred to as granulomatous lymphocytic, and necrotizing. Findings include
angiitis, granulomatous giant cell angiitis, inflammatory infiltrates of giant cells, plasma
noninfectious granulomatous angiitis, and iso- cells, lymphocytes in the vessel wall accompanied
lated angiitis. Its current name reflects the fact by necrosis, or granuloma formation. A sparse
that many cases have a non-granulomatous perivascular mononuclear cell infiltrate is
histology. insufficient for diagnosis as it can be seen in
many other conditions. Necrotizing vasculitis is
Epidemiology associated with the destruction of the internal
PACNS has an estimated incidence of 2.4 per elastic lamina.
1,000,000 inhabitants, with a slight female pre- CSF analysis is abnormal in 80–90 % of cases.
dominance [48, 49]. Although recent large cohort ESR and CRP are more likely to be elevated in
studies state a slight female predominance, cau- children [49].
tion must be taken in interpreting this data, as
several studies include patients with angio- Imaging
graphic, but not pathologic, proof of disease and, CT imaging will show nonspecific findings of
therefore, may occasionally include patients with vasculitis such as infarcts or hemorrhages and is
benign angiopathy of the CNS, who were once less sensitive than MRI.
characterized as a subset of PACNS with a more MRI imaging is abnormal in nearly all cases,
favorable prognosis and have since been even those that are angiography or biopsy nega-
reclassified as RCVS. Earlier studies showed tive, and most commonly shows cortical and sub-
either a slight male predominance of 4:3 or no cortical infarcts, which are frequently multiple
sex predilection. The median age of onset is about and frequently involve both cerebral hemispheres.
50 years [49]. Infarctions may occur in a large or perforator
33 Miscellaneous Vascular Diseases 765

Fig. 7 PACNS. Superior


cerebellar artery (SCA)
infarct shown on DWI (a)
secondary to stenosis of the
right SCA shown on the
right vertebral artery
injection (b) (Courtesy of
M. Shroff, G. DeVeber)

Fig. 8 PACNS. (a) DWI showing right MCA lenticulostriate territory infarction. (b, c) MR and digital subtraction
angiography show beading and marked stenosis of the right M1 and M2 vessels

artery distribution. Intracranial hemorrhage Angiography


occurs in about one tenth of patients [48]. Contrast The classic angiographic signs of vasculitis are
enhancement of the meninges, perivascular alternating areas of stenosis and dilation (beading)
spaces, and parenchymal mass lesions may occur involving both large and small vessels. A recent
[48]. Although nonspecific, negative MRI and study showed angiographic abnormalities in 90 %
negative CSF sampling are thought to have a of patients with PACNS [48]. MRA and CTA
high negative predictive value for the presence show similar findings, although with a lower sen-
of vasculitis. The specificity of both tests is esti- sitivity owing to the limited spatial resolution of
mated at 36 % (Figs. 7 and 8). 500 μm, below the threshold of detecting small-
High-resolution pre- and postcontrast T1-WI vessel vasculitides.
black blood imaging of the vessel walls has been
used to distinguish vasculitic processes from ath-
erosclerosis and RCVS (Fig. 9). Although this is a Takayasu
novel technique, preliminary results have shown
concentric luminal enhancement in vasculitis, Epidemiology – 1–2/1,000,000, with more reports
eccentric enhancement in atherosclerosis, and initially in South East Asia, but recent reports also
lack of enhancement in RCVS. in the western world.
766 M.H. Machnowska and R.I. Aviv

Fig. 9 Vessel wall imaging shows thickening (a, white arrowheads) and resultant collateralization through ECA
arrow) and enhancement (b, white arrowhead) of the cav- branches (black arrows). The patient presented with left
ernous ICA walls in a patient with rheumatoid arthritis and temporal lobe hemorrhage on SWI (e)
SLE. Marked bilateral ICA stenosis (c, d) (black

Pathology Clinical
This is a large-vessel vasculitis with predominant In addition to systemic symptoms of fever
involvement of the aorta and great vessels. Histo- and weight loss, the main neurological
logically, findings are those of a chronic large- symptoms attributable to Takayasu include
vessel vasculitis characterized by lymphocytic, dizziness, headache, vertigo, vision loss,
NK cell, neutrophil, and macrophage infiltration memory loss, TIAs, strokes, hypertensive
in the outer thirds of the media and adventitia. encephalopathy, and increased intracranial
Giant cells and granulomas have been described. pressure [50].
The elastic fibers are fragmented, and there can be CNS findings have also included
loss of smooth muscle cells, medial weakening, subarachnoid hemorrhage, most commonly due
and vascular dilation. Reactive fibrosis and to aneurismal rupture, but also non-aneurismal in
aneurismal formation is seen. etiology.
33 Miscellaneous Vascular Diseases 767

Fig. 10 A 6-year-old female with Takayasu’s arteritis. on the transverse ultrasound image (c). The patient
Coronal MR angiogram shows aneurismal dilation of the presented with concurrent posterior reversible leukoence-
right ICA (a, arrowhead). Axial T1-WI, contrast-enhanced phalopathy felt to be a result of renal artery involvement
image shows wall enhancement and a crescentic (d) (Courtesy of M. Shroff)
intraluminal filling defect in the right ICA (b) corroborated

MRI compatible with posterior reversible encephalop-


Arterial stenosis is the most common finding, athy [50] (Fig. 10).
although dilation, occlusion, and aneurysm for-
mation can be seen. In the head and neck, the Special Procedures
subclavian and common carotid arteries are most Panarteritis is seen in two thirds of patients with
often involved. There may be rare involvement of Takayasu’s. Angiographic classification is based
intracranial vessels. Initial experience using seg- on the segment involved.
mented k-space inversion recovery prepared gra-
dient echo MR pulse sequences to obtain delayed Type I: aortic arch and its branches
enhancement images of the aortic wall, suggests a Type II: descending thoracic and abdominal aorta
correlation between wall enhancement and active Type III: aortic arch and thoracoabdominal aorta
inflammation [51]. MRI may also show evidence Type IV: pulmonary artery in addition to the afore-
of ischemia, white matter lesions, and findings mentioned types
768 M.H. Machnowska and R.I. Aviv

Type V: coronary artery in addition to the other include bitemporal headaches, jaw claudication,
types[52]. scalp tenderness, and most severely vision loss.
The latter occurs irreversibly in 10–15 % of
Angiography cannot distinguish between patients, due to acute anterior optic neuritis, reti-
active and chronic inflammation. nal artery occlusion, or retrobulbar optic neuritis
PET labeled with fluorodeoxyglucose (FDG) is [57]. Systemic symptoms such as fever, weight
more sensitive than MRI for demonstrating the early loss, and polymyalgia also occur.
stages of Takayasu’s, with increased uptake in the
vessel wall seen during active inflammation [53]. Imaging
Rates of positive temporal artery biopsy are
50–90 % owing to the segmental nature of the
Giant Cell Arteritis disease. Some authors claim the sensitivity of
imaging, if it includes not only the temporal arter-
The disease occurs almost exclusively in those ies but also the aorta and supra-aortic arteries, is
over 50, with highest prevalence in the seventh superior to biopsy [58].
decade. Women are affected two to three times
more commonly than men. The disease incidence Ultrasound
is around 10–29/100,000 [54]. Color Doppler ultrasound was the first imaging
modality used to assess temporal arteritis and
Genetics currently has the highest imaging resolution of
Many small cohort genetic studies have looked at all imaging techniques used for diagnosis.
associations between genes encoding cytokines T axial and lateral resolution is 0.1 mm using
and their receptors, adhesion molecules, and high-frequency probes. Doppler ultrasound can
chemokines; however, the most consistent associ- assess arterial stenoses, wall edema, and blood
ation is between GCA and the MHC class II flow velocities through the affected segments [59].
molecules, specifically carriage of the
HLA-DRB1*04 alleles. The latter has also been CTA
associated with resistance to corticosteroid treat- CTA may be helpful as a quick way of evaluating
ment and greater risk of visual manifestation. the aorta and great vessels in patients with known
temporal artery involvement. A recent study
showed a 67 % occurrence of aortitis in newly
Pathophysiology
diagnosed GCA patients [60]. Similarly to con-
The disease is a panarteritis, involving all layers of
ventional angiography, its role in evaluating tem-
the vessel wall, the basis of which is an antigen-
poral arteritis is limited.
driven inflammatory response [55]. The disease
stimulus is currently unknown. It is thought to
MRI/MRA
activate adventitial dendritic cells, which release
3-T MRI has been quite helpful in the assessment
chemoattractants to recruit CD4+ T cells to the
of the temporal artery walls using contrast-
adventitia-media border. T cells release INF-γ
enhanced fat-saturated spin echo T1-WI images,
and thereby lead to attraction and differentiation
achieving a sensitivity of 80.6 % and specificity of
of macrophages resulting in giant cell infiltration
97 % in a recent study [61].
and granuloma formation in the intima-media
junction. Inflammatory cytokine release by mac-
rophages promotes systemic inflammation [56].
Polyarteritis Nodosa

Clinical Polyarteritis nodosa (PAN) is a necrotizing vascu-


Because of the predominant involvement of exter- litis, affecting small- and medium-sized arteries,
nal carotid arteries and their branches, symptoms never arterioles, capillaries, or venules. This is in
33 Miscellaneous Vascular Diseases 769

contradistinction to microscopic polyangiitis fever, conjunctivitis, rash, lymphadenopathy, irri-


(MAP), which has recently emerged in the litera- tability, oromucosal erythema, limb edema, and
ture, as a separate entity affecting mainly small skin peeling classically over the tips of the fingers
vessels. MAP has been shown to be an and toes. Coronary artery aneurysms are present
antineutrophil cytoplasmic antibody (ANCA)- in 25 %. It generally presents in children less than
associated vasculitis, associated more commonly 5 years of age. CNS involvement has been
with myeloperoxidase (MPO) ANCA, but described in 0.4–4 % of cases and includes facial
manifesting as a pure vasculitis without granu- palsy, seizures, cerebral artery stenosis, and
loma formation. moyamoya syndrome. In a small recent study,
PAN is characterized as a hyaline-like necrosis seven out of ten patients with Kawasaki disease
of the media and internal elastic lamina, with exten- showed CSF pleocytosis [64].
sion of an inflammatory reaction to the arterial wall
and adjacent tissues. A substantial portion of PAN
patients are carriers of the HBV virus with recent Churg-Strauss Syndrome
reports describing HCV association. HBV-PAN is
shown to have greater gastrointestinal organ Like MAP and Wegener’s, Churg-Strauss is an
involvement, with some improvement in symp- ANCA-associated small to medium vessel necro-
tomatology following antiretroviral therapy [62]. tizing vasculitis, often preceded by asthma and
eosinophilia manifesting similar to Wegener’s
Clinical Manifestations with granuloma formation. Unlike Wegener’s,
As with other systemic vasculitides, the disease is however, ANCA positivity is present in only
characterized by constitutional symptoms of fevers about 40 % compared to 90 % of cases and is
and weight loss, but also livedo reticularis, myal- associated more commonly with MPO-ANCA
gias, weakness or leg tenderness, and peripheral (pANCA). It has also been referred to as allergic
neuropathies. The most common organ of involve- granulomatosis or allergic granulomatosis with
ment is the peripheral nervous system. CNS man- polyangiitis.
ifestations have been reported in 20–45 % of
patients and manifest mainly as encephalopathy, Epidemiology
focal neurological deficits, and seizures [63]. The prevalence rates of Churg-Strauss range
between 2 and 22/million, suggesting the disease
Epidemiology is about two to eight times less common than
PAN generally presents in the fourth to sixth MPA or Wegener’s [65].
decades of life without gender preference. Its inci-
dence is estimated at 2–3/100,000. Genetics
Positive associations are found with the human
Imaging leukocyte antigen HLA-DRB1*07 genotype.
CT and MRI have shown cortical and subcortical A strong association with IL10 promoter polymor-
infarcts. Angiography has shown segmental phisms has been detected in Churg-Strauss [66].
nonspecific narrowing of middle-sized and small-
sized arteries, consistent with vasculitis, although Clinical
may be normal. Subarachnoid hemorrhage and intra- Asthma is present in about 90 % of patients with
cranial aneurysms are also reported although rare. Churg-Strauss syndrome [67]. Chest findings are
characterized by migratory infiltrates and symp-
tomatic cardiac involvement which occurs in up to
Kawasaki Disease 47 % of patients. Eosinophilic myocardial infil-
tration may progress to fibrosis and left ventricular
Kawasaki disease is an acute febrile mucocutane- dysfunction resulting in myocardial infarction and
ous lymph node syndrome with classic findings of arrhythmia as the main clinical features.
770 M.H. Machnowska and R.I. Aviv

Peripheral neuropathy is seen in about half of highest incidences reported in Northern Europe.
patients [67]. CNS involvement is seen in The usual age of diagnosis is in the fifth and sixth
10–30 % of patients and manifests as seizures, decades, but the disease can occur in the pediatric
stroke, and encephalopathy. population or present as late as the eighth
decade [69]. Two forms exist: the proteinase
Pathophysiology 3-antineutrophil cytoplasmic autoantibody
Multiple cell types contribute to the pathogenesis (PR3-ANCA) (cANCA)-associated form pre-
of Churg-Strauss. These include predominantly dominates in Caucasians in the Northern Hemi-
CD4+ Th2 (helper T cells) and eosinophils. sphere and the perinuclear myeloperoxidase
Circulating levels of eosinophils parallel disease (MPO-ANCA)-associated form predominates in
activity. Recent research has also shown the pres- Caucasian and non-Caucasians of the Southern
ence of Th1 and Th17 T-cell responses and indi- Hemisphere.
rect evidence for the role of B cells [68]. ANCA positivity is seen in over 90 % of
Wegener’s patients compared to 40 % of Churg-
Pathology Strauss patients [66].
The small-vessel necrotizing vasculitis with possi-
ble granuloma formation is nonspecific and often Clinical
indistinguishable from other forms of ANCA- Pulmonary and kidney involvement is most char-
associated vasculitides. Eosinophilic extravascular acteristic of Wegener’s. Pulmonary symptoms
tissue infiltration of virtually any organ character- include cough, dyspnea, thoracic pain, and
izes the early phase of disease. Nerve biopsy may hemoptysis and reflect the involvement of the
demonstrate epineural necrotizing vasculitis, eosin- entire respiratory tract. Kidney involvement usu-
ophilic infiltrates, and lymphocytes. ally manifests as microscopic hematuria, elevated
serum creatinine, and proteinuria ultimately
MRI Findings resulting in renal failure. Peripheral nervous sys-
Granulomatous involvement of the parenchyma tem involvement and cranial neuropathies are
and meninges can be seen and is usually T2 more common than central nervous system
hypointense and enhancing and indistinguishable involvement and are seen in 20–50 % [70].
from other granulomatous processes such as Sinonasal and orbital involvement is seen in
Wegener’s, sarcoid, and tuberculosis. Patients about 50 % of patients. CNS involvement is seen
may present with infarct and hemorrhage. There in about 8 % of patients [70]. CNS manifestations
are a few case reports of aneurismal subarachnoid are due to sino-orbital spread of disease, CNS
hemorrhage attributed to vasculitic changes of the vasculitis, or granulomatous lesions. CNS symp-
disease. toms result from pachymeningeal thickening, vas-
culitis, and least likely pituitary involvement
thought to result from contiguous spread. Symp-
Wegener’s toms include headaches, meningismus, seizures,
encephalopathy, and stroke [71].
Wegener’s disease is a c-ANCA-associated nec-
rotizing small-vessel vasculitis with predomi- Genetics
nantly airway, lung, and kidney involvement. The key candidate genes are the MHC complex
The disease is currently referred to as necrotizing II-associated HLA-DRB1 genes on chromosome
granulomatosis with polyangiitis or necrotizing 6, particularly the extended DPB1*0401/
granulomatosis. RXRB03 haplotype. Another association has
been made with the PTPN22 polymorphism
Epidemiology 620 W in ANCA positive Wegener’s patients,
Wegener’s granulomatosis is rare with reported which results in disruption of the PTPN22 protein
prevalence rates of 23.7–156.5/million, with responsible for T-cell receptor signaling. This may
33 Miscellaneous Vascular Diseases 771

result in enhanced T-cell activation with subse-


quent greater humoral abnormalities [72].

Pathophysiology
The disease is thought to result from activation of
cytokine primed neutrophils by ANCA resulting in
the release of proteolytic enzymes, reactive oxygen
species, and proinflammatory mediators [73]. The
disease is characterized by vasculitis and granu-
loma formation suggesting the presence of T-cell
hyperactivity. The causative agents resulting in
granuloma formation are unknown [73].
Several etiologic environmental factors have
been implicated in the development of WG and
include crystalline silica, organic solvents, cad-
mium, volatile hydrocarbons, and pesticides [73].

Imaging Findings
CNS imaging findings include pachymeningeal
thickening, which is nonspecific and cannot be
differentiated from granulomatous infections, Fig. 11 Pachymeningeal thickening along the right ante-
neoplastic involvement by benign and malignant rior convexity in a 10-year-old patient with Wegener’s
granulomatosis (Courtesy of M. Shroff)
entities, hypertrophic pachymeningitis, and
noninfectious granulomatous diseases such as sar-
coid (Fig. 11). sinonasal undifferentiated carcinoma (SNUC),
Pituitary involvement is rare, most often affect- melanoma, esthesioneuroblastoma, and NUT
ing the posterior with absence of the neurohypo- midline carcinoma. Osteitis can be mimicked by
physeal “bright spot,” but can lead to pituitary gland ossifying fibroma, osteoma, Paget disease, fibrous
enlargement with homogenous or heterogenous dysplasia, and sinonasal sarcoid [75] (Fig. 12).
enhancement and infundibular thickening. The
most common clinical finding is diabetes insipidus.
Less common involvement of the anterior pituitary Systemic Lupus Erythematosus
may manifest as hyperprolactinemia or panhypopi-
tuitarism [74]. Vasculitis is not well characterized Systemic lupus erythematosus (SLE) is a heterog-
by angiography due to small-vessel involvement. enous autoimmune disease with widespread
Orbital involvement, better characterized with MRI systemic involvement and well-recognized neuro-
than CT, can be seen in 50 % of patients and present psychiatric syndromes.
as both intra- and extraconal masses.
Sinonasal involvement is characterized by
bone involvement which may manifest as bone Epidemiology
destruction, usually of the septum and sinus walls,
sclerosing osteitis, bony thickening, and nodular Recent large cohort studies of the disease suggest
mucosal thickening [75]. Septal destruction may the prevalence of neuropsychiatric lupus
ultimately result in a “saddle-nose” deformity (NPSLE) including both major and minor criteria
[70]. Sinonasal findings have to be differentiated to be somewhere between 30 % and 40 % [76, 77].
from other diseases. The bony destruction can be NPSLE prevalence in children is similar to that in
mimicked by T-cell lymphoma, toxic etiologies adults. CNS disease accounts for about 5–7 %
(cocaine), sinonasal squamous cell carcinoma, mortality in SLE patients.
772 M.H. Machnowska and R.I. Aviv

Fig. 12 Sinonasal
manifestations of
Wegener’s including
erosion of the nasal bone (a)
necessitating nasal bone
prosthesis (b)

Clinical Presentation Pathology

Recognized neuropsychiatric syndromes include Of the cases of lupus vasculitis, more than 60 %
major manifestations such as seizure, cerebrovas- demonstrate leukocytoclastic inflammation, 30 %
cular disease, acute confusional state, psychosis, vasculitis with cryoglobulinemia, and about 6 %
and myelopathy. Minor CNS complaints also rec- systemic vasculitis resembling polyarteritis nodosa.
ognized as part of the neuropsychiatric syndrome
spectrum by the American College of Rheumatol-
ogy (ACR) include headache, anxiety, mild forms Imaging
of depression, and cognitive dysfunction. About
16 % of patients develop at least one of the The most common but also most nonspecific man-
major criteria: seizures, cerebrovascular disease, ifestation of SLE is focal T2 hyperintensities in
myelopathy, optic neuritis, aseptic meningitis, the subcortical and periventricular white matter.
and psychosis. About 50–60 % of the These usually do not contrast enhance and may
19 ACR-recognized neuropsychiatric syndromes occasionally show diffusion restriction. The
are thought to be a result of metabolic distur- hyperintensities are poorly differentiated from
bances, infections, or drug effects, including pos- microangiopathic disease seen with increasing
terior reversible encephalopathy syndrome, rather age or with increased vascular risk factors, such
than direct disease activity on the CNS [78]. as hypertension and diabetes mellitus, multiple
sclerosis, migraine, sarcoidosis, and HIV enceph-
alopathy. In 12 % of SLE patients, MRI has shown
Pathophysiology cortical T2 hyperintensities, associated with dif-
fusion restriction, similar to those seen with sei-
The etiology of CNS disease in SLE is multifac- zures [80]. Large-vessel infarction is usually
torial and includes cardioembolism from left- embolic in origin and associated with bland endo-
sided valvular disease, a prothrombotic state due carditis. Bilateral middle cerebellar peduncle
to antiphospholipid antibody (aPL), thrombotic infarcts have been reported as a presenting feature
thrombocytopenic purpura, antibody-mediated of SLE [81].
injury, and vasculitis. Thromboembolism is the Forty to 50 % of SLE patients with neuropsy-
predominant cause of cerebrovascular events chiatric symptom show normal MRI studies
[79]. Antiphospholipid is associated with ische- [80]. Several metabolic and functional
mic stroke, venous thrombosis, and TIA. imaging techniques have been used to identify
33 Miscellaneous Vascular Diseases 773

brain changes undetectable by conventional decrease of cell-mediated immunity to VZV.


imaging. These include magnetization transfer Both primary and secondary CNS vasculitis may
imaging (MTI), proton magnetic resonance be preceded by a rash in about one half to two
spectroscopy (MRS), and PET. MTI shows thirds of patients, although the onset of CNS find-
reduced magnetization transfer ratios in ings may occur several months following cutane-
patients with NPSLE indicative of subclinical ous findings [88].
damage, likely due to axonal damage [82]. MRS While a pure VZV-related encephalitis exists,
shows lower N-acetyl-aspartate (NAA) to creatine it is far less common than VZV-associated vascu-
(Cr) ratios reflecting axonal damage and reduced litis. The vasculitis most commonly involves large
neuronal-axonal density. Increased choline (Cho) and small arteries leading to large-vessel and per-
to creatine (Cr) ratios reflect inflammation and are forator territory infarcts and well-demarcated
associated with gliosis, vasculopathy, and edema. ischemic foci at the gray-white matter junctions.
Ratios are elevated in the active phase of disease In a recent study, a combination of large- and
and chronically increased in SLE patients with small-vessel involvement was seen in 50 % of
moderate to severe cognitive impairment patients, followed by small arterial involvement
[83]. MRS changes have been shown to herald (37 %) and large-vessel involvement (13 %)
the future appearance of white matter lesions in [88]. Based on feline studies, the disease spread
normal appearing brain matter [84]. is thought to be through ipsilateral trigeminal gan-
FDG-PET studies show hypometabolism in the glion afferents to the anterior circulation arteries,
parieto-occipital white matter in 60–80 % of i.e., the carotid terminus and MCA, and from the
patients [85]. cervical dorsal root ganglia to the vertebrobasilar
system [89]. PCR techniques have detected
viral DNA in the media of cerebral arteries,
Special Procedures while histopathologic evaluation has shown
multinucleated giant cells and herpes virions in
Lupus vasculitis occurs in about 10 % of patients smooth muscle cells of the MCA. The small-
and in most cases affects only small-caliber ves- vessel vasculopathy often yields lesions with
sels [86]. Nonspecific small-vessel occlusion may intranuclear Cowdry A inclusions in glia
be seen. Large-vessel occlusion can be seen in (a hallmark of herpes infection), central cavita-
active disease and has been associated with tion, and macrophage influx [90].
moyamoya disease. Patients with systemic MRI appearances show gray matter
middle-sized vessel involvement have the worst hyperintensity, swelling, and cortical and subcor-
outcomes. Large-vessel occlusion carries a 13 % tical white matter ischemic foci or hemorrhages.
recurrent stroke risk. Focal ectasias and intracra- The predominant angiographic abnormalities
nial aneurysm formation have also been reported. reveal segmental stenosis or occlusion of proxi-
mal vessels. Vessel wall imaging may show arte-
rial wall enhancement in the acute phase. There
Infectious Vasculitides are rare reports of aneurysm formation, subarach-
noid hemorrhage, vascular ectasia, and dissection
Varicella Zoster Vasculitis [90] (Fig. 13).

Varicella zoster virus (VZV) is a human herpes


virus manifesting as a primary infection HIV Vasculitis
(chickenpox) or secondary reactivation (shingles).
Primary infection usually occurs in children and is HIV causes predominantly a large-vessel vasculi-
responsible for about 31 % of strokes in the young tis, although cases of penetrating, small, and
[87], while reactivation is more common in immu- medium arterial involvement are reported. Vessels
nocompromised or older individuals, due to a show disruption of the internal elastic lamina and
774 M.H. Machnowska and R.I. Aviv

Fig. 13 Varicella zoster vasculitis in a 6-year-old child wall enhancement in the left carotid terminus (c) (Courtesy
resulting in left lenticulostriate territory infarcts shown on of M. Shroff, G. DeVeber)
DWI (a, b). Postcontrast T1-WI vessel wall imaging shows

subsequent media layer disruption [91]. Vessel Tuberculous Vasculitis


narrowing, occlusion, fusiform dilation, and aneu-
rysm formation have been described. The diagno- Tuberculous meningitis classically manifests with
sis should only be made after exclusion of exudative basilar infiltrates, resulting in an infil-
concomitant VZV, CMV, and syphilitic- or trative vasculitis, which proceeds from the adven-
toxoplasma-associated vasculitis. titia of vessels to their inner layers, and affecting
predominantly the terminal carotids; proximal
MCA, ACA, and PCA; and basilar arteries. Pro-
Bacterial Vasculitis liferative vasculitis, characterized by intimal pro-
liferation, and necrotizing vasculitis have also
Bacterial infection can cause vascular injury been described. Vascular arterial narrowing has
through several mechanisms. Direct extension been reported in the inferior aspect of vessels
from adjacent mastoiditis or sinusitis with or and may be limited to regions surrounding
without associated osteomyelitis can result in tuberculomas (Fig. 14).
vessel irregularity, narrowing, occlusion, or fusi-
form aneurysm formation. Basal vessels, includ-
ing the carotid arteries, are most often affected Fungal Vasculitis
with less common involvement of peripheral
vessels. Fungal infection can be directly spread through
Meningitis, cerebritis, abscess, empyema, and concomitant sinonasal infection, as in the case of
septic emboli can also result in vessel irregularity. zygomycoses, or hematogeneously spread, partic-
A retrospective study of 87 pneumococcal men- ularly in the immunocompromised host, as in the
ingitis patients found arterial and venous infarc- case of aspergillus infection in Western countries.
tion in 22 % and 10 % of cases, respectively [92]. Vasculitis can occur in two patterns: the tubercu-
Proposed mechanisms for large and medium lous type, indistinguishable from tuberculosis, or
arterial narrowing in meningitis include encroach- the hyphal type. The latter results from thrombosis
ment of large vessels by subarachnoid inflamma- and inflammation caused by angioinvasive hyphal
tory exudate; invasion of the vessel wall by elements and results in hemorrhagic infarctions,
inflammatory cells with resulting intimal thicken- subarachnoid hemorrhage, abscesses, or mycotic
ing, focal stenosis, dilatation, and edema; and aneurysms. A high index of suspicion for fungal
bland vasospasm [93]. infection is required, particularly in the case of
33 Miscellaneous Vascular Diseases 775

Fig. 14 A 15-year-old male with TB meningitis. (a, b) vermis suggestive of a basal meningitis. (d) MRA shows
DWI images shows multiple basal ganglia and posterior narrowing of the right A1 segment (Courtesy of M Shroff,
fossa infarcts. (c) Abnormal T2 signal is noted at the G. DeVeber)
margins of the interpeduncular cistern and in the cerebellar

patients with hematologic malignancies and bone reported between 20 and 421/100,000 [94]. The
marrow transplant recipients who present with age of onset is usually in the third decade, with no
hemorrhagic parenchymal CNS lesions [93]. gender predilection for the systemic disease, and a
slight (2.6:1) male predominance seen in CNS
disease [95].
Neuro-Behcet’s Disease Pediatric involvement is rare, but can occur,
and a more severe course of disease is seen in men
Behcet’s disease is a multisystem vasculitis which younger than 25.
classically presents a triad of oral and genital
ulcerations with uveitis. CNS involvement is Etiology
seen in 5–10 % of patients. The etiology of Behcet’s disease is
currently unknown. An association has been
Epidemiology made with a higher prevalence of HLA B5
Behcet’s disease follows the “Silk Road,” an (51) in Behcet’s patients, particularly in highly
ancient trading route dating back to the second endemic areas, but viral, bacterial, genetic, and
century BC, spanning from the Mediterranean to other autoimmune etiologies have also been
Japan. Its prevalence in the Mediterranean is implicated [95].
776 M.H. Machnowska and R.I. Aviv

Fig. 15 A 6-year-old male with Behcet’s disease. (a) CT enhanced T1-WI images show an ill-defined T2
shows left middle cerebellar peduncle hypodensity and hyperintense, ring enhancing lesion with peripheral
associated mass effect. (b, c) FLAIR and contrast- edema, and mild mass effect (Courtesy of M. Shroff)

Clinical manifestations seldom coexist. Parenchymal


CNS involvement is usually preceded by the clas- lesions are characterized by T1 hypointensity
sic triad of uveitis and genital and oral ulceration. and T2 hyperintensity on MRI. Contrast enhance-
Neuro-Behcet’s syndrome (NBS) can present as ment and greater cranial and caudal extent of T2
focal or multifocal CNS dysfunction resembling signal abnormality are observed in the acute and
multiple sclerosis. A subset of NBS patients dis- subacute phases. The mesencephalic-thalamic
plays symptoms of intracranial hypertension junction is the most common site of CNS involve-
attributable to dural venous sinus thrombosis. ment. Other regions of involvement include the
Peripheral nervous system involvement is rare, pontobulbar region; the hypothalamus-thalamus,
and greater than half of the patients are basal ganglia; and least commonly the
moderately to severely disabled 10 years periventricular, deep white matter and subcortical
following symptom onset. white matter. Pontobulbar lesions tend to involve
the tegmentum and superior cerebellar peduncles
Pathology and always spare the red nuclei. Cortical lesions
Large-vessel arterial and venous involvement is are seldom seen [95] (Fig. 15).
seen in up to 35 % of cases, with venous involve- Large-vessel involvement includes stenosis
ment seen about eight times more commonly than and occlusion and rare aneurismal formation.
arterial involvement in the CNS [96]. Pathologi- Venous sinus thrombosis is seen in 30 % of
cally, a mononuclear vascular and perivascular patients. In addition to the dural venous sinuses,
infiltrate is noted. There is disruption of the inter- the superior vena cava, subclavian veins, and jug-
nal elastic lamina and absence of intimal and ular veins can be involved.
medial layers. Fibrinoid necrosis has been
observed. Venous occlusion, thrombophlebitis,
and thrombosis are the most common findings; Summary
however, arterial occlusion and cerebral aneurysm
formation have been described. The diseases affecting intracranial vessels
discussed in this chapter often result in transient
Imaging or permanent deficits not infrequently affecting
Parenchymal NBS occurs in about 75 % of cases younger patients. When faced with work up for
and carries a worse prognosis than venous throm- stroke in the young, imaging can prove invaluable
bosis, the other NBS manifestation. The two in aiding diagnosis and treatment. Specific
33 Miscellaneous Vascular Diseases 777

findings such as extensive temporal white matter 11. Wilson SE, de Groen PC, Aksamit AJ, Wiesner RH,
involvement in CADASIL, the angiographic Garrity JA, Krom RA (1988) Cyclosporin A-induced
reversible cortical blindness. J Clin Neuroophthalmol
“puff of smoke” in moyamoya, or pulvinar 8(4):215–220
hyperintensity in Fabry’s disease can lead to 12. Kassem-Moussa H, Provenzale JM, Petrella JR, Lewis
rapid diagnosis of disease. Emerging new DV (2000) Early diffusion-weighted MR imaging
methods, such as vessel wall imaging, can help abnormalities in sustained seizure activity. AJR Am J
Roentgenol 174(5):1304–1306
detect the presence of inflammation either to 13. Mukherjee P, McKinstry RC (2001) Reversible poste-
guide diagnosis, biopsy, or treatment. Functional rior leukoencephalopathy syndrome: evaluation with
imaging methods are becoming significantly more diffusion-tensor MR imaging. Radiology 219(3):
important in determining the optimal time for 756–765
14. Bartynski WS, Boardman JF (2008) Catheter angiog-
treatment, particularly in the case of moyamoya, raphy, MR angiography, and MR perfusion in posterior
allowing for earlier detection of tissue at risk than reversible encephalopathy syndrome. AJNR Am J
standard anatomical imaging methods. Neuroradiol 29(3):447–455
15. Wartenberg KE, Parra A (2006) CT and CT-perfusion
findings of reversible leukoencephalopathy during
triple-H therapy for symptomatic subarachnoid
References hemorrhage-related vasospasm. J Neuroimaging
16(2):170–175
1. Rolfs A, Fazekas F, Grittner U et al (2013) Acute 16. Naidu K, Moodley J, Corr P, Hoffmann M (1997)
cerebrovascular disease in the young: the stroke in Single photon emission and cerebral computerised
young fabry patients study. Stroke 44(2):340–349 tomographic scan and transcranial doppler sono-
2. Brouns R, Thijs V, Eyskens F et al (2010) Belgian fabry graphic findings in eclampsia. Br J Obstet Gynaecol
study: prevalence of fabry disease in a cohort of 1000 104(10):1165–1172
young patients with cerebrovascular disease. Stroke 17. Calabrese LH, Dodick DW, Schwedt TJ, Singhal AB
41(5):863–868 (2007) Narrative review: reversible cerebral vasocon-
3. DeGraba T, Azhar S, Dignat-George F et al (2000) striction syndromes. Ann Intern Med 146(1):34–44
Profile of endothelial and leukocyte activation in 18. Ducros A (2014) Reversible cerebral vasoconstriction
fabry patients. Ann Neurol 47(2):229–233 syndrome. Handb Clin Neurol 121:1725–1741
4. Moore DF, Kaneski CR, Askari H, Schiffmann R 19. Ducros A, Boukobza M, Porcher R, Sarov M,
(2007) The cerebral vasculopathy of fabry disease. Valade D, Bousser MG (2007) The clinical and radio-
J Neurol Sci 257(1–2):258–263 logical spectrum of reversible cerebral vasoconstriction
5. Hilz MJ, Kolodny EH, Brys M, Stemper B, Haendl T, syndrome. A prospective series of 67 patients. Brain
Marthol H (2004) Reduced cerebral blood flow 130(Pt 12):3091–3101
velocity and impaired cerebral autoregulation 20. Ducros A, Fiedler U, Porcher R, Boukobza M, Stapf C,
in patients with fabry disease. J Neurol 251(5): Bousser MG (2010) Hemorrhagic manifestations of
564–570 reversible cerebral vasoconstriction syndrome: fre-
6. Moore DF, Ye F, Schiffmann R, Butman JA (2003) quency, features, and risk factors. Stroke 41(11):
Increased signal intensity in the pulvinar on 2505–2511
T1-weighted images: a pathognomonic MR imaging 21. Mandell DM, Matouk CC, Farb RI et al (2012) Vessel
sign of fabry disease. AJNR Am J Neuroradiol wall MRI to differentiate between reversible
24(6):1096–1101 cerebral vasoconstriction syndrome and central ner-
7. Fellgiebel A, Mazanek M, Whybra C et al (2006) Pat- vous system vasculitis: preliminary results. Stroke
tern of microstructural brain tissue alterations in fabry 43(3):860–862
disease: a diffusion-tensor imaging study. J Neurol 22. Baba T, Houkin K, Kuroda S (2008) Novel epidemio-
253(6):780–787 logical features of moyamoya disease. J Neurol
8. Fellgiebel A, Keller I, Marin D et al (2009) Diagnostic Neurosurg Psychiatry 79(8):900–904
utility of different MRI and MR angiography measures 23. Smith ER, Scott RM (2012) Spontaneous occlusion of
in fabry disease. Neurology 72(1):63–68 the circle of willis in children: pediatric moyamoya
9. Bartynski WS (2008) Posterior reversible encephalop- summary with proposed evidence-based practice
athy syndrome, part 2: controversies surrounding path- guidelines. A review. J Neurosurg Pediatr 9(4):
ophysiology of vasogenic edema. AJNR Am J 353–360
Neuroradiol 29(6):1043–1049 24. Nagaraja D, Verma A, Taly AB, Kumar MV,
10. Bartynski WS (2008) Posterior reversible encephalop- Jayakumar PN (1994) Cerebrovascular disease in chil-
athy syndrome, part 1: fundamental imaging and clin- dren. Acta Neurol Scand 90(4):251–255
ical features. AJNR Am J Neuroradiol 29(6): 25. Mineharu Y, Takenaka K, Yamakawa H et al (2006)
1036–1042 Inheritance pattern of familial moyamoya disease:
778 M.H. Machnowska and R.I. Aviv

autosomal dominant mode and genomic imprinting. level-dependent magnetic resonance imaging.
J Neurol Neurosurg Psychiatry 77(9):1025–1029 J Neurosurg 103(2):347–355
26. Smith ER, Scott RM (2008) Progression of disease in 39. Shimizu H, Shirane R, Fujiwara S, Takahashi A,
unilateral moyamoya syndrome. Neurosurg Focus Yoshimoto T (1997) Proton magnetic resonance spec-
24(2):E17 troscopy in children with moyamoya disease. Clin
27. Burke GM, Burke AM, Sherma AK, Hurley MC, Neurol Neurosurg 99(Suppl 2):S64–S67
Batjer HH, Bendok BR (2009) Moyamoya disease: 40. Opherk C, Peters N, Herzog J, Luedtke R, Dichgans M
a summary. Neurosurg Focus 26(4):E11 (2004) Long-term prognosis and causes of death in
28. Lee M, Zaharchuk G, Guzman R, Achrol A, Bell- CADASIL: a retrospective study in 411 patients.
Stephens T, Steinberg GK (2009) Quantitative hemo- Brain 127(Pt 11):2533–2539
dynamic studies in moyamoya disease: a review. 41. Desmond DW, Moroney JT, Lynch T, Chan S, Chin SS,
Neurosurg Focus 26(4):E5 Mohr JP (1999) The natural history of CADASIL: a
29. Yamashita M, Oka K, Tanaka K (1983) Histopathology pooled analysis of previously published cases. Stroke
of the brain vascular network in moyamoya disease. 30(6):1230–1233
Stroke 14(1):50–58 42. Okeda R, Arima K, Kawai M (2002) Arterial changes
30. Bacigaluppi S, Dehdashti AR, Agid R, Krings T, in cerebral autosomal dominant arteriopathy with sub-
Tymianski M, Mikulis DJ (2009) The contribution of cortical infarcts and leukoencephalopathy (CADASIL)
imaging in diagnosis, preoperative assessment, and in relation to pathogenesis of diffuse myelin loss of
follow-up of moyamoya disease: a review. Neurosurg cerebral white matter: examination of cerebral medul-
Focus 26(4):E3 lary arteries by reconstruction of serial sections of an
31. Suzuki J, Takaku A (1969) Cerebrovascular autopsy case. Stroke 33(11):2565–2569
“moyamoya” disease. Disease showing abnormal 43. Viswanathan A, Gray F, Bousser MG, Baudrimont M,
net-like vessels in base of brain. Arch Neurol 20(3): Chabriat H (2006) Cortical neuronal apoptosis in
288–299 CADASIL. Stroke 37(11):2690–2695
32. Kikuta K, Takagi Y, Nozaki K et al (2005) Asymptom- 44. van den Boom R, Lesnik Oberstein SA, Ferrari MD,
atic microbleeds in moyamoya disease: T2*-weighted Haan J, van Buchem MA (2003) Cerebral autosomal
gradient-echo magnetic resonance imaging study. dominant arteriopathy with subcortical infarcts and
J Neurosurg 102(3):470–475 leukoencephalopathy: MR imaging findings at differ-
33. Research Committee on the Pathology and Treatment ent ages–3rd-6th decades. Radiology 229(3):683–690
of Spontaneous Occlusion of the Circle of Willis, 45. Dichgans M, Holtmannspotter M, Herzog J, Peters N,
Health Labour Sciences Research Grant for Bergmann M, Yousry TA (2002) Cerebral microbleeds
Research on Measures for Infractable Diseases (2012) in CADASIL: a gradient-echo magnetic resonance
Guidelines for diagnosis and treatment of imaging and autopsy study. Stroke 33(1):67–71
moyamoya disease (spontaneous occlusion of the cir- 46. Cumurciuc R, Guichard JP, Reizine D, Gray F, Bousser
cle of Willis). Neurol Med Chir (Tokyo) 52(5): MG, Chabriat H (2006) Dilation of virchow-robin
245–266 spaces in CADASIL. Eur J Neurol 13(2):187–190
34. Nariai T, Matsushima Y, Imae S et al (2005) Severe 47. Fukutake T (2011) Carasil. Brain Nerve 63(2):99–108
haemodynamic stress in selected subtypes of patients 48. Salvarani C, Brown RD Jr, Calamia KT et al (2007)
with moyamoya disease: a positron emission tomogra- Primary central nervous system vasculitis: analysis of
phy study. J Neurol Neurosurg Psychiatry 76(5): 101 patients. Ann Neurol 62(5):442–451
663–669 49. Giannini C, Salvarani C, Hunder G, Brown RD (2012)
35. Sakamoto S, Ohba S, Shibukawa M, Kiura Y, Arita K, Primary central nervous system vasculitis: pathology
Kurisu K (2006) CT perfusion imaging for childhood and mechanisms. Acta Neuropathol 123(6):759–772
moyamoya disease before and after surgical revascu- 50. Ringleb PA, Strittmatter EI, Loewer M et al (2005)
larization. Acta Neurochir (Wien) 148(1):77–81, Cerebrovascular manifestations of Takayasu arteritis
discussion 81 in Europe. Rheumatology (Oxford) 44(8):1012–1015
36. Aviv R, Letourneau-Guillon L, Symons S, Fox A 51. Desai MY, Stone JH, Foo TK, Hellmann DB, Lima JA,
(2013) Other arteriopathies. In: Naidich T, Castillo M, Bluemke DA (2005) Delayed contrast-enhanced MRI
Cha S, Smirniotopoulos J (eds) Imaging of the brain. of the aortic wall in Takayasu’s arteritis: initial experi-
Saunders Elsevier, Philadelphia, pp 451–469 ence. AJR Am J Roentgenol 184(5):1427–1431
37. Kang KH, Kim HS, Kim SY (2008) Quantitative cere- 52. Gulati A, Bagga A (2010) Large vessel vasculitis.
brovascular reserve measured by acetazolamide- Pediatr Nephrol 25(6):1037–1048
challenged dynamic CT perfusion in ischemic adult 53. Walter MA, Melzer RA, Schindler C, Muller-Brand J,
moyamoya disease: initial experience with angio- Tyndall A, Nitzsche EU (2005) The value of [18F]
graphic correlation. AJNR Am J Neuroradiol 29(8): FDG-PET in the diagnosis of large-vessel vasculitis
1487–1493 and the assessment of activity and extent of disease.
38. Mikulis DJ, Krolczyk G, Desal H et al (2005) Preop- Eur J Nucl Med Mol Imaging 32(6):674–681
erative and postoperative mapping of cerebrovascular 54. Gonzalez-Gay MA, Vazquez-Rodriguez TR, Lopez-
reactivity in moyamoya disease by using blood oxygen Diaz MJ et al (2009) Epidemiology of giant cell
33 Miscellaneous Vascular Diseases 779

arteritis and polymyalgia rheumatica. Arthritis Rheum assessment and management. Curr Opin Rheumatol
61(10):1454–1461 26(1):16–23
55. Gonzalez-Gay MA, Garcia-Porrua C, Gonzalez- 69. Almouhawis HA, Leao JC, Fedele S, Porter SR (2013)
Juanatey C, Miranda-Filloy JA, Blanco R, Llorca J Wegener’s granulomatosis: a review of clinical features
(2013) Biopsy-proven giant cell arteritis patients with and an update in diagnosis and treatment. J Oral Pathol
coronary artery disease have increased risk of aortic Med 42(7):507–516
aneurysmal disease and arterial thrombosis. Clin Exp 70. Mujagic S, Sarihodzic S, Huseinagic H, Karasalihovic
Rheumatol 31(1 Suppl 75):S94 Z (2011) Wegener’s granulomatosis of the paranasal
56. Ly KH, Regent A, Tamby MC, Mouthon L (2010) sinuses with orbital and central nervous system
Pathogenesis of giant cell arteritis: more than just an involvement-diagnostic imaging. Acta Neurol Belg
inflammatory condition? Autoimmun Rev 9(10): 111(3):241–244
635–645 71. Seror R, Mahr A, Ramanoelina J, Pagnoux C, Cohen P,
57. Gonzalez-Gay MA, Garcia-Porrua C, Llorca J Guillevin L (2006) Central nervous system involve-
et al (2000) Visual manifestations of giant cell arteritis. ment in Wegener granulomatosis. Medicine (Balti-
Trends and clinical spectrum in 161 patients. Medicine more) 85(1):54–65
(Baltimore) 79(5):283–292 72. Jagiello P, Aries P, Arning L et al (2005) The PTPN22
58. Schmidt WA (2013) Imaging in vasculitis. Best Pract 620W allele is a risk factor for Wegener’s
Res Clin Rheumatol 27(1):107–118 granulomatosis. Arthritis Rheum 52(12):4039–4043
59. Schmidt WA, Kraft HE, Vorpahl K, Volker L, 73. Manna R, Cadoni G, Ferri E et al (2008) Wegener’s
Gromnica-Ihle EJ (1997) Color duplex ultrasonogra- granulomatosis: an update on diagnosis and therapy.
phy in the diagnosis of temporal arteritis. N Engl J Med Expert Rev Clin Immunol 4(4):481–495
337(19):1336–1342 74. Yong TY, Li JY, Amato L et al (2008) Pituitary involve-
60. Prieto-Gonzalez S, Arguis P, Garcia-Martinez A ment in Wegener’s granulomatosis. Pituitary 11(1):
et al (2012) Large vessel involvement in biopsy-proven 77–84
giant cell arteritis: prospective study in 40 newly diag- 75. Lohrmann C, Uhl M, Warnatz K, Kotter E, Ghanem N,
nosed patients using CT angiography. Ann Rheum Dis Langer M (2006) Sinonasal computed tomography in
71(7):1170–1176 patients with Wegener’s granulomatosis. J Comput
61. Bley TA, Uhl M, Carew J et al (2007) Diagnostic value Assist Tomogr 30(1):122–125
of high-resolution MR imaging in giant cell arteritis. 76. Borowoy AM, Pope JE, Silverman E et al (2012) Neu-
AJNR Am J Neuroradiol 28(9):1722–1727 ropsychiatric lupus: the prevalence and autoantibody
62. Segelmark M, Selga D (2007) The challenge of man- associations depend on the definition: results from the
aging patients with polyarteritis nodosa. Curr Opin 1000 faces of lupus cohort. Semin Arthritis Rheum
Rheumatol 19(1):33–38 42(2):179–185
63. Provenzale JM, Allen NB (1996) Neuroradiologic 77. Unterman A, Nolte JE, Boaz M, Abady M,
findings in polyarteritis nodosa. AJNR Am J Shoenfeld Y, Zandman-Goddard G (2011) Neuropsy-
Neuroradiol 17(6):1119–1126 chiatric syndromes in systemic lupus erythematosus:
64. Korematsu S, Uchiyama S, Miyahara H et al (2007) a meta-analysis. Semin Arthritis Rheum 41(1):1–11
The characterization of cerebrospinal fluid and serum 78. Steup-Beekman GM, Zirkzee EJ, Cohen D et al (2013)
cytokines in patients with Kawasaki disease. Pediatr Neuropsychiatric manifestations in patients with sys-
Infect Dis J 26(8):750–753 temic lupus erythematosus: epidemiology and radiol-
65. Gibelin A, Maldini C, Mahr A (2011) Epidemiology ogy pointing to an immune-mediated cause. Ann
and etiology of Wegener granulomatosis, micro- Rheum Dis 72(Suppl 2):ii76–ii79
scopic polyangiitis, churg-strauss syndrome and 79. Hanly JG, Walsh NM, Sangalang V (1992) Brain
goodpasture syndrome: vasculitides with frequent pathology in systemic lupus erythematosus.
lung involvement. Semin Respir Crit Care Med 32(3): J Rheumatol 19(5):732–741
264–273 80. Luyendijk J, Steens SC, Ouwendijk WJ et al (2011)
66. Wieczorek S, Holle JU, Epplen JT (2010) Recent pro- Neuropsychiatric systemic lupus erythematosus: les-
gress in the genetics of wegener’s granulomatosis and sons learned from magnetic resonance imaging. Arthri-
churg-strauss syndrome. Curr Opin Rheumatol 22(1): tis Rheum 63(3):722–732
8–14 81. Okamoto K, Tokiguchi S, Furusawa T et al (2003) MR
67. Comarmond C, Pagnoux C, Khellaf M et al (2013) features of diseases involving bilateral middle cerebel-
Eosinophilic granulomatosis with polyangiitis (churg- lar peduncles. AJNR Am J Neuroradiol 24(10):
strauss): clinical characteristics and long-term 1946–1954
followup of the 383 patients enrolled in the french 82. Bosma GP, Steens SC, Petropoulos H et al (2004)
vasculitis study group cohort. Arthritis Rheum Multisequence magnetic resonance imaging study of
65(1):270–281 neuropsychiatric systemic lupus erythematosus.
68. Mahr A, Moosig F, Neumann T et al (2014) Eosino- Arthritis Rheum 50(10):3195–3202
philic granulomatosis with polyangiitis (churg- 83. Axford JS, Howe FA, Heron C, Griffiths JR (2001)
strauss): evolutions in classification, etiopathogenesis, Sensitivity of quantitative (1)H magnetic resonance
780 M.H. Machnowska and R.I. Aviv

spectroscopy of the brain in detecting early neuronal 90. Gilden D, Cohrs RJ, Mahalingam R, Nagel MA (2009)
damage in systemic lupus erythematosus. Ann Rheum Varicella zoster virus vasculopathies: diverse clinical
Dis 60(2):106–111 manifestations, laboratory features, pathogenesis, and
84. Appenzeller S, Li LM, Costallat LT, Cendes F (2007) treatment. Lancet Neurol 8(8):731–740
Neurometabolic changes in normal white matter may 91. Gutierrez J, Ortiz G (2011) HIV/AIDS patients with
predict appearance of hyperintense lesions in systemic HIV vasculopathy and VZV vasculitis: a case series.
lupus erythematosus. Lupus 16(12):963–971 Clin Neuroradiol 21(3):145–151
85. Popescu A, Kao AH (2011) Neuropsychiatric systemic 92. Kastenbauer S, Pfister HW (2003) Pneumococcal men-
lupus erythematosus. Curr Neuropharmacol 9(3): ingitis in adults: spectrum of complications and prog-
449–457 nostic factors in a series of 87 cases. Brain 126(Pt 5):
86. D’Cruz D (1998) Vasculitis in systemic lupus 1015–1025
erythematosus. Lupus 7(4):270–274 93. Chow FC, Marra CM, Cho TA (2011) Cerebrovascular
87. Askalan R, Laughlin S, Mayank S et al (2001) disease in central nervous system infections. Semin
Chickenpox and stroke in childhood: a study of fre- Neurol 31(3):286–306
quency and causation. Stroke 32(6):1257–1262 94. Mat C, Yurdakul S, Sevim A, Ozyazgan Y, Tuzun Y
88. Nagel MA, Cohrs RJ, Mahalingam R et al (2008) The (2013) Behcet’s syndrome: facts and controversies.
varicella zoster virus vasculopathies: clinical, CSF, Clin Dermatol 31(4):352–361
imaging, and virologic features. Neurology 70(11): 95. Kocer N, Islak C, Siva A et al (1999) CNS involvement
853–860 in neuro-behcet syndrome: an MR study. AJNR Am J
89. Saito K, Moskowitz MA (1989) Contributions from the Neuroradiol 20(6):1015–1024
upper cervical dorsal roots and trigeminal ganglia to 96. Koc Y, Gullu I, Akpek G et al (1992) Vascular involve-
the feline circle of Willis. Stroke 20(4):524–526 ment in Behcet’s disease. J Rheumatol 19(3):402–410
Vascular Loop Syndromes
34
Divyata R. Hingwala and Kesavadas Chandrasekharan

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 781 Vascular loop syndromes are disorders usually
associated with vascular compression of the
Trigeminal Neuralgia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 782
Clinical Features, Classification, and Definition . . . . 782
specific cranial nerves. Neuroimaging may
Etiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 782 play an important role in the preoperative diag-
Pathogenesis and Pathophysiology . . . . . . . . . . . . . . . . . . 782 nosis of these syndromes. MRI can detect the
Necessity for Neuroimaging in TN . . . . . . . . . . . . . . . . . . 782 vascular loops compressing the cranial nerves
Neuroimaging Techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . 782
Neuroimaging Findings in TN . . . . . . . . . . . . . . . . . . . . . . . 783
(fifth, seventh, ninth, eighth, and fourth) and
can also rule out secondary causes. The aim of
Hemifacial Spasm . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 784 this chapter is to outline the clinical features
Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 784
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 785 and pathogenesis of neurovascular syndromes
Role of Neuroimaging in HFS . . . . . . . . . . . . . . . . . . . . . . . 785 and give an overview of neuroimaging tech-
Neuroimaging Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 785 niques and findings.
Other Rare Manifestations of Vascular
Loop Syndrome and Their Imaging Features . . . . 786 Keywords
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 787 Trigeminal neuralgia • Hemifacial spasm •
Glossopharyngeal neuralgia • Superior oblique
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 787
myokymia

Introduction

Vascular loop syndromes are disorders usually


associated with vascular compression of the spe-
cific cranial nerves. This compression can cause
paroxysmal attacks of pain or abnormal move-
D.R. Hingwala (*)
ments in the distribution of the affected nerves
NM Medical Centre, Mumbai, Maharashtra, India
e-mail: divyatahingwala@gmail.com [1]. Neuroimaging may play an important role in
the preoperative diagnosis of these syndromes.
K. Chandrasekharan
NM Medical Centre, Mumbai, Maharashtra, India The aim of this chapter is to outline the clinical
features and pathogenesis of neurovascular syn-
Sree Chitra Tirunal Institute for Medical Sciences and
Technology, Trivandrum, Kerela, India dromes and give an overview of neuroimaging
e-mail: chandkesav@yahoo.com techniques and findings.
# Springer Science+Business Media New York 2016 781
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_9
782 D.R. Hingwala and K. Chandrasekharan

Trigeminal Neuralgia Pathogenesis and Pathophysiology

Clinical Features, Classification, Trigeminal neuralgia is caused by demyelination


and Definition of the trigeminal sensory fibers within the
trigeminal nerve root [8]. Walter Dandy was the
Trigeminal neuralgia (TN) has been defined by the first to observe that the nerve is compressed by
International Association for the Study of Pain as an overlying blood vessel [9], and it has been
sudden, usually unilateral, severe, brief, stabbing, confirmed by various authors including
recurrent episodes of pain in the distribution of Hamlyn [10] that vascular compression at the
one or more branches of the trigeminal nerve root entry zone (REZ) is the cause of TN in
[2]. Specific clinical criteria have been described 80–90 % of the cases. The REZ of the trigeminal
like characteristic, stereotyped patterns of pain nerve is the transition zone between the central
attacks, no objective neurological deficit, and no myelin (formed by oligodendrocytes) and periph-
other identified causes of pain [3]. eral myelin (formed by Schwann cells) and is
According to the latest classification of the located approximately 1–2.5 mm from the
International Headache Society, TN is classified pons [11].
as classic and symptomatic. Classic TN (CTN) is Pathological studies have demonstrated
distinguished from symptomatic TN (STN) by the demyelinated axons which are in direct apposition
absence of an established etiology and clinically with few intervening glial processes [8]. This
evident neurologic deficit [3]. allows the generation of ectopic impulses and
This pain is localized to the trigeminal nerve non-synaptic ephaptic transmission of impulses
distribution. It often affects the maxillary (V2) or [12]. The pulsatility of overlying arteries may
mandibular (V3) divisions of the trigeminal nerve contribute to the demyelination and abnormal
or a combination of adjacent divisions. The topo- impulse generation [8]. Nerve fibers subserving
graphic distribution of pain is determined by the light touch and pain are the nearest in proximity at
location of the vascular loop. Clinical stimuli the REZ. This theory further explains how light
within specific trigger zones or specific activities touch provokes pain [4].
like chewing, shaving, or brushing teeth may pre-
cipitate pain attacks. Occasionally, in the period
following a pain attack, the patient may be refrac- Necessity for Neuroimaging in TN
tory to further pain [4].
Neuroepidemiological studies have projected Studies have found that STN and CTN cannot be
the incidence of TN to be between 4 and 5 per reliably differentiated on the basis of clinical fea-
100,000 per year. It is more frequent with advanc- tures, trigeminal reflex, and trigeminal nerve-
ing age and in females as compared with evoked potentials [13, 14], though younger age
males [5]. and abnormal trigeminal nerve-evoked potentials
may suggest the presence of a secondary cause
[15]. The role of MRI is not only to exclude
Etiology secondary causes but also to depict vascular
loops causing TN.
CTN is caused by neurovascular compression (see
section “Pathophysiology”). Potential etiologies
of STN include multiple sclerosis plaques, tumors Neuroimaging Techniques
(schwannomas, meningiomas, epidermoids, etc.),
and abnormalities of the skull base [6]. Stretching CT is inadequate for evaluating a patient with
of the trigeminal nerve in Chiari I malformation is vascular loop syndromes [16]. Conventional
also a rare cause of trigeminal neuralgia [7]. MRI images may be useful for excluding
34 Vascular Loop Syndromes 783

nonvascular causes of HFS. However, these may vessel is the anterior inferior cerebellar artery, the
fail to satisfactorily delineate the neurovascular site of conflict is inferior or medial (Fig. 1). Con-
relationships [17]. flict at the lateral aspect of the nerve is extremely
Useful techniques [18] include a heavily uncommon. The pattern of venous conflict is
T2-WI sequence like Constructive Interference variable [22].
in Steady State (CISS, Siemens) or 3D Fast Imag- Other vascular causes of TN include aneu-
ing with Steady State (FIESTA, GE) sequence in rysms and vascular malformations [dural arterio-
which the CSF is bright and other cisternal struc- venous fistulas and arteriovenous malformations
tures like nerves, arteries, and veins appear dark. (including microAVMs)] (Fig. 2) [24].
MR angiography is also very useful to delineate According to increasing severity, MRI findings
the culprit artery due to superb contrast between are graded as Grade I (simple contact
the artery and CSF, but the nerves are not well between nerve and vessel), Grade II (displace-
defined as compared with the CSF [19]. These ment/distortion of nerve roots by vessel), and
sequences have high spatial resolution due to Grade III (indentation and thinning of nerve
thin contiguous slices. Contrast-enhanced high- root) [25].
resolution 3D T1-WI sequences provide addi- The sensitivity and specificity of high-
tional delineation of the arterial and venous struc- resolution MRI for identification of neurovascular
tures causing neurovascular conflict and other conflict varied from 52–100 % to 29–93 %,
enhancing lesions like AVM, tumors, and devel- respectively. Because of the inconsistency of the
opmental venous anomalies. results, according to the practice parameters pre-
scribed by the AAN, there is insufficient evidence
to support or refute the usefulness of MRI to
Neuroimaging Findings in TN identify vascular contact in CTN or to indicate
the most reliable MRI technique.
An intraoperative finding of arterial or venous Other imaging indicators of TN include an
compression of the trigeminal nerve root is seen atrophy of the trigeminal nerve, a small ipsilateral
in 90–95 % of the patients with CTN as reported cerebellopontine angle cistern, a short length of
in a series of patients who undergo microvascular the cisternal segment of the trigeminal nerve, and
decompression (MVD) [20, 21]. a narrower trigeminopontine angle [25, 26].
The most commonly implicated vessels are the Apart from neurovascular conflict, neuroimag-
SCA, AICA, and the petrosal vein [22]. The supe- ing detected other structural causes in 10–18 % of
rior cerebellar artery is by far the most common cases [27].
artery seen compressing the trigeminal nerve, The pitfalls of MRI include partial volume
seen in about 75 % of the cases. Other arteries effects due to the small size of structures being
associated with compression include the anterior imaged and a false-positive rate of 14–21 % of
inferior cerebellar artery (9.6 %), posterior infe- vascular compression in asymptomatic
rior cerebellar artery (0.7 %), vertebral artery individuals [28].
(1.6 %), basilar artery (0.7 %), and labyrinthine In addition to anatomical imaging, diffusion
artery (0.2 %). Multivessel conflict may also be tensor imaging (DTI) can objectively quantify
seen [23]. the microstructural changes in the affected nerve.
Venous causes of compression include the Coregistering anatomic and DTI images can
transverse pontine vein (identification of which enhance the localization of small structures. The
may change the surgical approach) [24]. fractional anisotropy in the ipsilateral nerve has
The site of neurovascular conflict between the been described to be reduced as compared with
superior cerebellar artery and trigeminal nerve is the contralateral side. ADC values remain
superior, superomedial, or medial. If the culprit unchanged [29].
784 D.R. Hingwala and K. Chandrasekharan

Fig. 1 A sixty-eight-year-old female patient with left tri- reconstruction (e, f) show AICA loop (yellow arrow) cross-
geminal neuralgia. Axial CISS images (a, b), axial time-of- ing inferomedial to the left trigeminal nerve and causing
flight MRA source images (c, d), and coronal lateral displacement and thinning of the REZ

Fig. 2 Other vascular causes of trigeminal neuralgia: (a) anomaly in the vicinity of the trigeminal nucleus (Drawing
posterior fossa arteriovenous malformation, (b) brainstem adapted from Harsha et al. [24])
cavernoma, (c) microAVM, and (d) developmental venous

muscles innervated by the ipsilateral facial


Hemifacial Spasm nerve, predominantly the periorbital or perioral
facial musculature [30]. Clinically, it needs to be
Clinical Features differentiated from other causes of facial dyskine-
sias like blepharospasm, tics, myokymia, focal
Hemifacial spasm (HFS) is a peripheral move- seizures, or psychogenic conditions. Usually uni-
ment disorder clinically characterized by parox- lateral, it may be bilateral in about 0.6–5 % cases
ysms of tonic or clonic contractions involving [31]. While this condition is not life threatening,
34 Vascular Loop Syndromes 785

the clinical significance of diagnosing and treating incidence of hypertension and its related changes
this disorder lies in its disabling nature [32]. in the vasculature.
HFS has a higher incidence in Asians, proba-
bly due to the smaller size of the posterior fossa.
Pathophysiology HFS can also rarely be caused by other vascu-
lar abnormalities like vertebrobasilar ectasia
Dandy in 1932 [9], Campbell et al. in 1947 [33], (Fig. 3) and fusiform aneurysms at the vertebral
and Gardner in 1962 [34] had initially reported artery (VA)–posterior inferior cerebellar artery
that vascular compressions of the fifth and seventh (PICA) junction [41] and of the VA [42]. The
cranial nerves were possible causes of trigeminal VA fusiform aneurysm may also compress the
neuralgia and hemifacial spasm. contralateral facial nerve, causing HFS [43]. How-
In 1977, Jannetta et al. [35] were the first to ever, in the presence of aneurysms, the possibility
describe the site of compression of these nerves. of coexistence of additional causative vessels like
They reported that vascular compression of the PICA or AICA adjacent to the aneurysm should
seventh cranial nerve at the REZ was a cause of be ruled out.
HFS. They further elaborated that the
compression of the nerve must be perpendicular
to its course. Peripheral vessels and vessels run- Role of Neuroimaging in HFS
ning parallel to the nerve were previously not
thought to be associated with HFS. The REZ of HFS is mostly related to neurovascular compres-
the facial nerve is the point where the cells sion of the facial nerve [32, 44]. Microvascular
forming the myelin sheath change from the central decompression is the surgical treatment of choice
oligodendroglia to the central Schwann cells, for providing symptomatic relief to the patients
making it most susceptible to irritation by [45]. The decision to perform this surgery is based
mechanical stimulation such as vascular on clinical history and examination. However,
compression [36]. rarely, another cause like a tumor or a demyelin-
Other authors have described a far more distal ating disorder may be detected [46]. Preoperative
location of compression midway between the imaging is thus important to rule out a
REZ and the internal acoustic meatus (IAM) or nonvascular cause. In addition, imaging of the
at the IAM associated with HFS [37]. neurovascular relationships at the REZ and delin-
This compression is seen generally on the ante- eating the responsible vessel may be important in
rior surface of the facial nerve and is associated the surgical planning to prevent/minimize
with typical HFS [38]. Rarely, it can occur at the intraoperative complications like hearing loss,
rostral and posterior surface of the nerve and is facial paresis, and meningitis [47]. Imaging can
then associated with atypical HFS [39]. also aid in patient counseling as the detection of a
Configuration variation of the facial- small compressing artery or vein may be associ-
vestibulocochlear nerve complex may also be ated with a higher postoperative recurrence rate of
associated with the AICA compressing the distal symptoms [48].
portion of the facial nerve. In the case report
described by Kawashima et al., the facial nerve
arose 5 mm away from the vestibulocochlear Neuroimaging Findings
nerve without forming a complex with it and
was strongly bent posteriorly [40]. On imaging, we have to identify the course of the
Patients with young-onset HFS (<30 years) facial nerve and follow it from its origin, along the
account for up to 6 % of the patients overall. cisternal segment and up to the intracanalicular
This subgroup may present with similar clinical segment for any compression, contact, or inden-
features and imaging findings as the older sub- tation. The exact location of these findings on the
group. However, the older subgroup has more circumference of the nerve has to be documented.
786 D.R. Hingwala and K. Chandrasekharan

Fig. 3 A patient with left


hemifacial spasm. Axial
CISS (a) and high-
resolution T2 (b) images
and axial time-of-flight MR
angiography source images
show a tortuous left
vertebral artery (yellow
arrow) compressing the left
seventh to eighth nerve
complex

In addition, the dominance of the vertebrobasilar Tan et al. [51], the MRI finding of contralateral
system and rotation of the brainstem, if any, are NVC has a poor predictive value for the presence
noted. Finally, as described above, other findings of bilateral symptoms. However, patients with
like aneurysms, arteriovenous malformations, and bilateral symptoms have a significantly higher
nonvascular causes like tumors have to be incidence of presence of contralateral NVC in
excluded. addition to the ipsilateral NVC. Also, in patients
Neurovascular contact has been found in with bilateral symptoms, there is a correlation
86–90 % [49] of the patients on MRA and defor- between the severity of HFS symptoms and the
mity of the nerves in 56 % of the cases. The MRA degree of neurovascular compression at the REZ.
findings correspond with the surgical findings in
87 % of the cases. A false-negative rate of about
15 % has also been observed [50]. CISS sequence Other Rare Manifestations of Vascular
has a sensitivity of 100 % for demonstrating Loop Syndrome and Their Imaging
neurovascular compression [18]. Features
Neurovascular compression of the facial nerve
may also be seen in asymptomatic controls and in Glossopharyngeal neuralgia is a rare condition
the contralateral asymptomatic side in patients characterized by severe, unilateral paroxysmal
with unilateral HFS. Thus, this imaging finding pain in the oropharynx, ear, or both locations,
may not always lead to symptoms. According to triggered by specific activities like swallowing,
34 Vascular Loop Syndromes 787

chewing, or coughing. It may also be manifested glossopharyngeal neuralgia: similarities and differ-
by other sensory phenomena like palatal myoclo- ences, Rochester, Minnesota, 1945–1984. Neuroepi-
demiology 10:276–281
nus, syncope, or rarely even cardiac arrest. MRI 6. Gronseth G, Cruccu G, Alksne J, Argoff C, Brainin M,
can depict vascular compression of the Burchiel K, Nurmikko T, Zakrzewska JM (2008) Prac-
glossopharyngeal nerve at the supraolivary tice parameter: the diagnostic evaluation and treatment
fossette (especially by the posterior inferior cere- of trigeminal neuralgia (an evidence-based review)
report of the Quality Standards Subcommittee of the
bellar artery) [52]. American Academy of Neurology and the European
Superior oblique myokymia is an unusual Federation of Neurological Societies. Neurology
movement disorder with tonic or phasic abnormal 71:1183–1190
eye movements caused by vascular compression 7. Papanastassiou AM, Schwartz RB, Friedlander RM
(2008) Chiari I malformation as a cause of
of the trochlear nerve. High-resolution MRI can trigeminal neuralgia: case report. Neurosurgery 63(3):
demonstrate arterial contact at the root entry zone E614–E615
of the affected trochlear nerve [53]. 8. Love S, Coakham HB (2001) Trigeminal neuralgia:
Vascular compression of the vestibulocochlear pathology and pathogenesis. Brain 124:2347–2360
9. Dandy WE (1934) Concerning the cause of trigeminal
nerve has been suggested as a cause of tinnitus neuralgia. Am J Surg 24:447–455
and/or vertigo by many authors [54]. However, 10. Hamlyn PJ, King TT (1992) Neurovascular compres-
other authors [55] have refuted these findings. sion in trigeminal neuralgia: a clinical and anatomic
study. J Neurosurg 76:948–954
11. Peker S, Kurtkaya O, Uzun I, Pamir MN (2006) Micro-
anatomy of the central-peripheral myelin transition
Conclusion zone of the trigeminal nerve. Neurosurgery 59
(2):354–359
Vascular loop syndromes (TN and HFS) are 12. Smith KJ, McDonald WI (1980) Spontaneous and
evoked activity due to central demyelinating lesion.
caused by vascular compression at the REZ and Nature 286(5769):154–155
focal demyelination. These are often refractory 13. Cruccu G, Biasiotta A, Galeotti F, Iannetti GD,
conditions that may require surgical management Truini A, Gronseth G (2006) Diagnostic accuracy of
of the neurovascular conflict. Neuroimaging trigeminal reflex testing in trigeminal neuralgia.
Neurology 66(1):139–141
(MRI) plays an important role in the evaluation 14. Mursch K, Schafer M, Steinhoff BJ, Behnke – Mursch
of these patients. The entire cisternal course of the J (2002) Trigeminal evoked potentials and sensory
affected nerve should be imaged in multiple deficits in atypical facial pain – a comparison with
planes to verify the presence of neurovascular results in trigeminal neuralgia. Funct Neurol
17(3):133–136
conflict. 15. Bradley WG Jr, Waluch V, Yadley RA, Wycoff RR
(1984) Comparison of CT and MR in 400 patients
with suspected disease of the brain and cervical spinal
References cord. Radiology 152(3):695–702
16. Kent DL, Larson EB (1988) Magnetic resonance imag-
ing of the brain and spine. Is clinical efficacy
1. Langner S, Schroeder HW, Hosten N, Kirsch M (2012) established after the first decade? Ann Intern Med
Diagnosing neurovascular compression syndromes. 108(3):402–424
Rofo 184(3):220–228 17. Papanagiotou P, Grunwald IQ, Politi M, Struffert T,
2. Merskey H, Bogduk N (1994) Classification of chronic Ahlhelm F, Reith W (2006) Vascular anomalies of the
pain: descriptions of chronic pain syndromes and def- cerebellopontine angle. Radiologe 46(3):216–222
initions of pain terms. IASP Press, Seattle, pp 59–71 18. Tarnaris A, Rendowen S, Coakham HB (2007) A com-
3. Headache Classification Subcommittee of the Interna- parison of magnetic resonance angiography and con-
tional Headache Society (2004) The international clas- structive interference in steady state-three-dimensional
sification of headache disorders. 2nd edition. Fourier transformation magnetic resonance imaging in
Cephalalgia 24(1):126–136 patients with hemifacial spasm. Br J Neurosurg
4. Prasad S, Galetta S (2009) Trigeminal neuralgia histor- 21(4):375–381
ical notes and current concepts. Neurologist 15 19. Sindou M, Howeidy T, Acevedo G (2002) Anatomic
(2):87–94 observations during microvascular decompression for
5. Katusic S, Williams DB, Beard CM, Bergstralh EJ, idiopathic trigeminal neuralgia with correlations
Kurland LT (1991) Epidemiology and clinical features between topography of pain and site of the
of idiopathic trigeminal neuralgia and
788 D.R. Hingwala and K. Chandrasekharan

neurovascular conflict: prospective study in a series of treatment of hemifacial spasm. Operative techniques
579 patients. Acta Neurochir (Wien) 144(1):1–12 and results in 47 patients. J Neurosurg 47(3):321–328
20. Sindou M, Leston J, Decullier E, Chapuis F (2007) 36. Tomii M, Onoue H, Yasue M, Tokudome S, Abe T
Microvascular decompression for trigeminal neuralgia: (2003) Microscopic measurement of the facial nerve
long-term effectiveness and prognostic factors in a root exit zone from central glial myelin to peripheral
series of 362 consecutive patients with clear-cut Schwann cell myelin. J Neurosurg 99(1):121–124
neurovascular conflicts who underwent pure decom- 37. Ryu H, Yamamoto S, Sugiyama K, Uemura K,
pression. J Neurosurg 107(6):1144–1153 Miyamoto T (1998) Hemifacial spasm caused by vas-
21. Meaney JMF, Miles JB, Nixon TE, Whitehouse GH, cular compression of the distal portion of the facial
Ballantyne ES, Eldridge PR (1994) Vascular contact nerve. Report of seven cases. J Neurosurg 88
with the fifth cranial nerve at the pons in patients with (3):605–609
trigeminal neuralgia: detection with 3D FISP imaging. 38. Ho SL, Cheng PW, Wong WC, Chan FL, Lo SK, Li LS,
AJR 163(6):1447–1452 Tsang KL, Leong LL (1999) A case-controlled
22. Jannetta PJ (1967) Arterial compression of the trigem- MRI/MRA study of neurovascular contact in
inal nerve at the pons in patients with trigeminal neu- hemifacial spasm. Neurology 53(9):2132–2139
ralgia. J Neurosurg 17:159–180 39. Barker FG II, Jannetta PJ, Bissonette DJ, Shields PT,
23. Seoane ER, Rhoton AL Jr (1999) Suprameatal exten- Larkins MV, Jho HD (1995) Microvascular decom-
sion of the retrosigmoid approach: microsurgical anat- pression for hemifacial spasm. J Neurosurg 82
omy. Neurosurgery 44(3):553–560 (2):201–210
24. Harsha KJ, Kesavadas C, Chinchure S, Thomas B, 40. Kawashima M, Yamada M, Sato S, Oka H, Fujii K,
Jagtap S (2012) Imaging of vascular causes of trigem- Matsushima T (2009) Hemifacial spasm caused by
inal neuralgia. J Neuroradiol 39(5):281–289 vascular compression of the distal portion of the facial
25. Kress B, Rasche D, Fiebach J, Tronnier V, Sartor K, nerve associated with configuration variation of the
Stippich C (2004) MR volumetry of the trigeminal facial and vestibulocochlear nerve complex. Turk
nerve in patients with unilateral facial pain. RoFo Neurosurg 19(3):269–275
176:719–723 41. Choi SK, Rhee BA, Park BJ, Lim YJ (2008)
26. Ha SN, Kim SH, Yoo EH, Han IB, Shin DA, Cho KG, Hemifacial spasm caused by fusiform aneurysm at
Chung SS, Park YS (2012) Patients with idiopathic vertebral artery posterior inferior cerebellar artery junc-
trigeminal neuralgia have a sharper-than-normal tion. J Korean Neurosurg Soc 44(6):399–400
trigeminal-pontine angle and trigeminal nerve atrophy. 42. Uchino M, Nomoto J, Ohtsuka T, Kuramitsu T (2005)
Acta Neurochir 154(9):1627–1633 Fusiform aneurysm of the vertebral artery presenting
27. Goh BT, Poon CY, Peck RH (2001) The importance of with hemifacial spasm treated by microvascular
routine magnetic resonance imaging in trigeminal neu- decompression. Acta Neurochir (Wien) 147
ralgia diagnosis. Oral Surg Oral Med Oral Pathol Oral (8):901–903
Radiol Endod 92:424–429 43. Nakagawa I, Takayama K, Kurokawa S, Wada T,
28. Tash AR, Sze G, Leslie DR (1989) Trigeminal neural- Nakagawa H, Kichikawa K, Nakase H (2011)
gia: MR imaging features. Radiology 172(3):767–770 Hemifacial spasm due to contralateral aneurysmal
29. Liu Y, Li J, Butzkueven H, Duan Y, Zhang M, Shu N, compression of the facial nerve successfully treated
Li Y, Zhang Y, Li K (2013) Microstructural abnormal- with endovascular coil embolization: case report.
ities in the trigeminal nerves of patients with trigeminal Neurosurgery 69(3):E768–E772
neuralgia revealed by multiple diffusion metrics. Eur J 44. Jannetta P (1970) Microsurgical exploration and
Radiol 82(5):783–786 decompression of the facial nerve in hemifacial
30. Wang A, Jankovic J (1998) Hemifacial spasm: clinical spasm. Curr Top Surg Res 2:217–220
findings and treatment. Muscle Nerve 21 45. Chung SS, Chang JH, Choi JY, Chang JW, Park YG
(12):1740–1747 (2001) Microvascular decompression for hemifacial
31. Tan E-K, Jancovik J (1999) Bilateral hemifacial spasm: spasm: a long-term follow-up of 1,169 consecutive
a report of five cases and a literature review. Mov cases. Stereotact Funct Neurosurg 77(1–4):190–193
Disord 14:345–349 46. Galvez-Jimenez N, Hanson MR, Desai M (2001)
32. Tan EK, Fook-Chong S, Lum SY, Lim E (2004) Botu- Unusual causes of hemifacial spasm. Semin Neurol
linum toxin improves quality of life in hemifacial 21(1):75–83
spasm: validation of a questionnaire (HFS-30). 47. Coakham H (2000) The microneurosurgical treatment
J Neurol Sci 219(12):151–155 of trigeminal neuralgia, hemifacial spasm and
33. Campbell E, Keedy C (1947) Hemifacial spasm: a note glossopharyngeal neuralgia. In: Robertson HC, Rob-
on the etiology in two cases. J Neurosurg 4:342–347 ertson JH (eds) Cranial base surgery. Churchill Living-
34. Gardner WJ (1962) Concerning the mechanism of tri- stone, London
geminal neuralgia and hemifacial spasm. J Neurosurg 48. Engh JA, Horowitz M, Burkhart L, Chang YF, Kassam
19:947–958 A (2005) Repeat microvascular decompression for
35. Jannetta PJ, Abbasy M, Maroon JC, Ramos FM, Albin hemifacial spasm. J Neurol Neurosurg Psychiatry 76
MS (1977) Etiology and definitive microsurgical (11):1574–1580
34 Vascular Loop Syndromes 789

49. Fukuda H, Ishikawa M, Okumura R (2003) Demon- 53. Yousry I, Dieterich M, Naidich TP, Schmid UD,
stration of neurovascular compression in trigeminal Yousry TA (2002) Superior oblique myokymia:
neuralgia and hemifacial spasm with magnetic reso- magnetic resonance imaging support for the
nance imaging comparison with surgical findings in neurovascular compression hypothesis. Ann Neurol
60 consecutive cases. Surg Neurol 59(2):93–100 51(3):361–368
50. Tan EK, Chan LL (2006) Young onset hemifacial 54. McDermott AL, Dutt SN, Irving RM, Pahor AL,
spasm. Acta Neurol Scand 114(1):59–62 Chavda SV (2003) Anterior inferior cerebellar artery
51. Tan E-K, Chan LL (2004) Clinico-radiologic correla- syndrome: fact or fiction. Clin Otolaryngol 28
tion in unilateral and bilateral hemifacial spasm. (2):75–80
J Neurol Sci 222(1–2):59–64 55. Gultekin S, Celik H, Akpek S, Oner Y, Gumus T,
52. Hiwatashi A, Matsushima T, Yoshiura T, Tanaka A, Tokgoz N (2008) Vascular loops at the
Noguchi T, Togao O, Yamashita K, Honda H (2008) cerebellopontine angle: is there a correlation with tin-
MRI of glossopharyngeal neuralgia caused by nitus? Am J Neuroradiol 29(9):1746–1749
neurovascular compression. AJR 191(2):578–581
Migraine
35
Mark Kruit

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 792 This chapter focuses on diagnostic clinical neu-
Pathophysiology of Migraine . . . . . . . . . . . . . . . . . . . . . . . . 793 roimaging in migraine. In most migraine cases,
Diagnostic Neuroimaging Indications
patient history, details of symptoms, and careful
in Migraine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 794 clinical neurological examination are together
AAN Recommendations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 794 the most important tools in diagnosing and
EFNS Guidelines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795 treating migraine, and consequently, there is
Summarized Recommendations . . . . . . . . . . . . . . . . . . . . . 795
mostly no need for further laboratory tests or
Migraine and Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 796 neuroimaging. In selected non-acute headache
Migrainous Infarction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 796 cases, neuroimaging is warranted, and recom-
Migraine as a Risk Factor for Clinical
Ischemic Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 803 mendations are provided. Imaging in acute
Migraine as a Risk Factor for Clinical Hemorrhagic headaches is not subject of this chapter.
Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 804 Good understanding of the migraine patho-
Migraine as a Risk Factor for Subclinical Stroke . . . 805 physiology allows better interpretation of neu-
Migraine and T2 Hyperintensities on MRI . . . . . . . 807 roimaging findings notably when patients
White Matter Lesions in Migraine (WMLs) . . . . . . . . 807 present acutely, in or outside an attack. The
“Subclinical” MRI Findings in the Individual neuroradiological findings relevant in under-
Migraine Patient . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 811 standing the complex relationship between
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 811 migraine and stroke will therefore be discussed,
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 812
including details on findings during migraine
aura. On MRI scans in migraine patients, fre-
quently, white matter hyperintensities and (cer-
ebellar) infarcts can be present. Knowledge of
the epidemiological findings that have set
migraine today also as a risk factor for (progres-
sive) subclinical brain lesions is relevant in the
everyday neuroradiological practice.

Keywords
Migraine disorders • Headache • Pathophysi-
M. Kruit (*)
ology • MRI • Stroke • Aura • White matter
Department of Radiology, Leiden University Medical
Center, Leiden, The Netherlands lesions • Infarct • Cerebellar infarcts
e-mail: m.c.kruit@lumc.nl

# Springer Science+Business Media New York 2016 791


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_24
792 M. Kruit

Introduction Table 1 Diagnostic criteria for migraine without and with


aura [3]
Migraine is a common, multifactorial, Migraine without aura
multiphasic, disabling primary headache disorder. A. At least 5 attacks fulfilling criteria B–D
Effects on the brain (structure, neurochemistry, B. Headache attacks lasting 4–72 h (untreated or
unsuccessfully treated)
function) and neurovascular system have been
C. Headache has at least two of the following
widely documented during the different phases characteristics
of migraine. a. Unilateral location
The disorder is typically characterized by b. Pulsating quality
recurrent attacks of disabling headache that last c. Moderate or severe pain intensity
4–72 h and are accompanied by nausea or d. Aggravation by or causing avoidance of routine
vomiting and photophobia and phonophobia physical activity (e.g., walking or climbing stairs)
(migraine without aura). The pain is typically D. During headache, at least one of the following
located unilaterally, of moderate to severe a. Nausea and/or vomiting
intensity, and has mostly a pulsating character. b. Photophobia and phonophobia
The headache phase may be preceded by a E. Not attributed to another disorder
premonitory phase (occurring hours or days Previously used terms: common migraine, hemicrania
simplex
before the headache) and followed by a
Migraine with aura
resolution phase. In the premonitory and resolu- A. At least 2 attacks fulfilling criteria B–D
tion phases, patients often experience various B. Aura consisting of at least one of the following, but
symptoms, including hyper- or hypoactivity, no motor weakness
food craving, yawning, fluid retention, and a. Fully reversible visual symptoms including positive
depression [1]. features (e.g., flickering lights, spots, or lines) and/or
Up to one third of patients also have neurolog- negative features (i.e., loss of vision)
b. Fully reversible sensory symptoms including
ical aura symptoms (migraine with aura): these
positive features (i.e., pins and needles) and/or negative
focal neurological symptoms usually precede or features (i.e., numbness)
sometimes accompany the headache, and c. Fully reversible dysphasic speech disturbance
most often affect the visual field, but may also C. At least two of the following
involve the sensory or motor system, or can affect a. Homonymous visual symptoms and/or unilateral
speech. Aura symptoms usually develop gradu- sensory symptoms
ally over 5–20 min, last for less than 60 min, and b. At least one aura symptom develops gradually over
5 min and/or different aura symptoms occur in
are fully reversible. Headache with the features of
succession over 5 min
migraine without aura usually follows the aura c. Each symptom lasts 5 and 60 min
symptoms. Less commonly, headache lacks D. Headache fulfilling criteria B–D for migraine
migrainous features or is completely absent (aura without aura begins during the aura or follows aura
without migraine) [2]. Migraine is diagnosed within 60 min
according to the International Classification of E. Not attributed to another disorder
Headache Disorders (ICHD, 2nd edition, 2005; Previously used terms: Classic or classical migraine,
ophthalmic, hemiparesthetic, hemiplegic or
Table 1) [1].
aphasic migraine, migraine accompagnée, complicated
Epidemiologic studies described that at least migraine
12 % of the general population suffers from reg-
ular migraine attacks [4]. Women are roughly
three times more often affected than men.
Migraine patients suffer a median of 12 migraine disorders [5] and has been estimated to be the
attacks per year; 25 % have at least two attacks per most costly neurological disorder in the
month. Migraine is rated by the World Health European countries, largely explained by days
Organization among the most disabling chronic missed from work and reduced productivity at
35 Migraine 793

work [6]. Acute attack treatments are in less than Migraine Aura: Cortical Spreading
half of the patients fully satisfactory, and more Depression
effective and better-tolerated prophylactic agents The pathophysiological mechanism behind the
to prevent attacks are needed. Mainstream migraine aura symptoms is cortical spreading
migraine prophylactic treatments have been depression (CSD). This is a transient depression
largely based on serendipitous observations and of activity in neural tissue and is often preceded by
presumed class effects and have disappointing a brief burst of action potential firing, which
efficacy and tolerability. slowly propagates in brain tissue [19–21]. CSD
most often involves the occipital lobe, leading to
visual symptoms in about 30 % of patients. Dur-
Pathophysiology of Migraine ing the CSD, regional brain hyper- and
hypoperfusion (supposed to remain above ische-
Understanding of the pathophysiology of mic thresholds), reductions of blood–brain barrier
migraine starts in acknowledging that migraine integrity, and plasma extravasation have been
is not simply a disease of intermittently occurring described (see below in section “Neuroimaging
pain, but that it relates to processes that over time Findings in (Prolonged) Aura”). It is unknown
affect the brain, or act as triggers on a predisposed why CSD occurs episodically, although tempo-
brain. These processes seem to lead to increased rary changes in excitability seem to be crucial,
sensitivity or hyperexcitability of brain areas, and likely already develop gradually in hours to
allowing headache and aura to develop days before the symptoms present. The potential
paroxysmally [7]. The mechanisms behind such role of CSD in activating the migraine headache is
triggering processes are poorly understood, but compelling in animal experiments but still contro-
are likely multifactorial, and involve several versial in humans [22].
brain structures. Glutamatergic-mediated neuro-
nal hyperexcitability seems an important trigger- Role of Neuroimaging in Understanding
ing mechanism [8], but environmental factors the Pathophysiology of Migraine
(e.g., physiologic factors, stress, weather), comor- Effects on the brain (structure, neurochemistry,
bid depression, and obesity are also of function) and neurovascular system have been
influence [9]. widely documented during the different phases
of migraine, and neuroimaging has played a sig-
Mechanisms Behind Migraine Headache nificant role in the current understanding of path-
The migraine headache results from activation of ophysiological processes behind migraine.
the trigeminovascular system (TGVS), which However, these processes are still only partially
consists of neural connections between dural understood, are likely multifactorial, and involve
blood vessels and trigeminal brainstem nuclei several brain structures.
[10]. Functional imaging illustrated dynamic Neuroimaging has contributed significantly to
involvement of brainstem areas in migraine the current neuroscientific knowledge on struc-
[11–15]. In the ictal phase, nociceptive (pain) tural and functional brain changes during the
signals are projected from the TGVS to the hypo- ictal phase (aura and headache) of migraine. It is
thalamus, the thalamus, and higher CNS centers. however largely unknown which parts of the brain
These signals are normally controlled by a net- structurally, functionally, or biochemically
work involving the hypothalamus and brainstem change earlier on, during the premonitory phase.
areas (periaqueductal grey matter and ventral teg- The thalamus, hypothalamus, and probably other
mental area) [16, 17]. Paroxysmal dysfunction of deep brain and brainstem structures seem to play a
(parts of) this network leads to ineffective role. Further research focusing on such early
antinociceptive modulation and vascular control, changes in the premonitory phase may provide
resulting in the ictal symptoms (pain and auto- insight in when and why brainstem nuclei and
nomic symptoms) [18]. pain networks become paroxysmally
794 M. Kruit

dysfunctional, how the trigeminovascular system Furthermore, the current resource-restricted med-
becomes activated, and what pathophysiological ical environment more and more requires
changes precede and characterize the aura symp- evidence-based justification of diagnostic
toms. Given the diagnostic focus of this chapter, imaging.
the neuroscientific neuroimaging findings (based
on, e.g., voxel-based morphometry, DTI, MRS,
etc.) behind parts of migraine pathophysiology are
considered out of scope here and will therefore not AAN Recommendations
be further discussed.
In 2000, the quality standards subcommittee of the
American Academy of Neurology (AAN)
Diagnostic Neuroimaging Indications published an evidence-based guideline on the role
in Migraine of neuroimaging in patients with headache [24], to
assist the physicians in making appropriate choices
The diagnosis of primary headaches is exclusively in diagnostic workup. A total of 28 studies (from
a clinical task based on history taking and careful 1966 to 1998) were reviewed. Based on reported
neurologic examination. In cases of acute head- rates of “abnormalities related to headache that
ache (like primary thunderclap headache or after may require further action” (like acute cerebral
trauma, etc.) or suspected symptomatic headache, infarct, neoplastic disease, hydrocephalus, aneu-
the need for neuroimaging is mostly evident and rysm, or arteriovenous malformation) combined
will not further be discussed in this chapter. For with data from patient history and neurological
non-acute headache, as applies to most migraine examination, the following symptoms were identi-
patients, neuroimaging is overused and is only in fied to significantly increase the odds of finding a
selected cases considered to be appropriate. significant abnormality on neuroimaging in
CT scans and notably MRI scans are frequently patients with non-acute headache [24]:
requested and performed in migraine patients who
seek medical help. Often this is driven by patient’s • Rapidly increasing headache frequency
anxiety about having an underlying pathologic • History of lack of coordination
condition, or to improve patient overall satisfac- • History of localized neurologic signs or a his-
tion and medical care. The fact that radiological tory such as subjective numbness or tingling
examinations are not particularly invasive or • History of headache causing awakening from
uncomfortable reduces thresholds further. sleep (although this can occur with migraine
However, notably, in patients presenting with and cluster headache)
typical primary headaches, the very low likeli-
hood of detecting explanatory underlying diseases Based on these findings, the following AAN
that change treatment or diagnosis must be con- recommendations for non-acute headache were
sidered. Combined results of imaging studies formulated:
(CT and MRI) in over 3,700 headache patients
(not exclusively “typical migraine”) together • Consider neuroimaging in patients with an
show a low yield of about 0.4 % in migraine unexplained abnormal finding on the neuro-
patients [23]. In “typical migraine” patients, the logic examination (Grade B).
yield is likely to be even lower. • Consider neuroimaging in patients with atypi-
Potential risk of “unnecessary imaging” is the cal headache features or headaches that do not
discovery of an incidental finding, which eventu- fulfill the strict definition of migraine or other
ally needs further diagnostic workup of follow-up. primary headache disorders (or have some
Further, potential problems include false-positive additional risk factor, such as immune defi-
studies, false reassurance from an inadequate ciency), when a lower threshold for neuroim-
study, allergic reaction to contrast agent, etc. aging may be applied (Grade C).
35 Migraine 795

• Neuroimaging is not usually warranted in Table 2 Recommendation on the use of diagnostic neu-
patients with migraine and a normal neurologic roimaging in non-acute headache
examination (Grade B). A. Consider neuroimaging in non-acute headache
patients with “red flags”
Unexplained abnormal findings on neurologic
examination
EFNS Guidelines (Atypical) headaches that do not fulfill ICHD-II
criteria for primary headaches
The European Federation of Neurological Socie- Additional risk factors (e.g., immune deficiency,
ties (EFNS) published a similar guideline for the tumors, etc.)
management of non-acute headache (revision in History of (associated) seizures
2010) [25], also mainly based on review of Recent changes in headache pattern
published evidence. This EFNS guideline Nonvisual (e.g., sensory or motor) or atypical aura
pattern
includes the following summarized statements
B. Neuroimaging is not usually warranted in patients
relevant for migraine (grade B recommendations): with typical migraine with or without aura and
normal neurological examination
• In adult and pediatric patients with migraine, C. When neuroimaging is warranted, MRI is method
with no recent change in pattern, no history of of primary choice
seizures, and no other focal neurological signs
or symptoms, the routine use of neuroimaging
is not warranted.
• Exception of these rules should be made in the
diagnosis of trigeminal autonomic headaches recommendations from this meta-analysis are
and headaches that are aggravated by exertion consistent with the guidelines by AAN and
or a Valsalva-like maneuver [23]. EFNS, although additional recommendations
• In patients with atypical headache patterns, a were included to perform imaging in
history of seizures, or neurological signs or cases (1) with nonvisual of aura (sensory or
symptoms or symptomatic illness such as motor), (2) an aura that has changed in character,
tumors, acquired immunodeficiency syndrome or (3) an aura that cannot be clearly described as
(AIDS), and neurofibromatosis, MRI may be typical of migraine aura.
indicated. Since a 2007 case series demonstrated that
• When neuroimaging is warranted, the most even cluster headache with a typical time
sensitive method should be used, and MRI pattern and an excellent response to typical
(and not CT) is recommended in these cases. treatment can still be caused by underlying
• With a normal unenhanced MRI, in the structural pathology such as a pituitary tumor,
absence of other diseases and suspicion on patients with trigeminal autonomic headaches
metastasis/vasculitis/etc., there is no need for should be considered to undergo
additional scanning with gadolinium. neuroimaging [26].
• There is no role for conventional Röentgen In summary, patient history, details of symp-
techniques. toms, and careful clinical neurological examina-
• Digital subtraction angiography is not appro- tion are together the most important tools in
priate in the screening of patients with head- diagnosing and treating migraine. In most patients
ache for intracranial disease. with non-acute headache, this will lead to a reli-
able diagnosis (applying the ICHD criteria) and
do not require any further laboratory tests or
Summarized Recommendations neuroimaging.
Table 2 summarizes the combined recommen-
In the recent meta-analysis by Detsky et al., data dations on the use of neuroimaging in non-acute
from over 3,700 patients were included [23]. The headache patients.
796 M. Kruit

Migraine and Stroke Table 3 Diagnostic criteria for migrainous infarction [3]
A. The present attack in a patient with migraine with aura
Accumulating evidence from the last 3–4 decades is typical of previous attacks except that one or more aura
has expanded the spectrum of neurovascular symptoms persist for >60 min
pathology linked to migraine. Initial case reports B. Neuroimaging demonstrates ischemic infarction in a
relevant area
of “migrainous stroke” were followed by retro-
C. Not attributed to another disorder
spective and prospective mostly hospital-based
case-control studies assessing the prevalence of
clinical ischemic and hemorrhagic stroke in
migraine patients, showing a consistent associa- stroke is assumed to be directly and causally
tion between migraine with aura and stroke; the related to an acute migraine attack.
association with migraine without aura is less Because it is often impossible by clinical
evident [27, 28]. MRI studies have further identi- examination alone to differentiate between TIA,
fied that migraine is also associated with markers prolonged aura, and migrainous infarction, MRI
of small vessel disease, including (progressive) (notably with diffusion weighting) today plays a
white matter lesions (WMLs), brainstem T2 key role in the diagnosis of migrainous infarction.
hyperintensities, posterior circulation subclinical The current ICHD-II criteria strictly define
infarcts, and microbleeds [29–31]. And finally, migrainous infarction as ischemic stroke that
reports from epidemiological studies on associa- occurs when during a typical migraine with aura
tions between migraine and coronary events [32, attack, one or more migrainous aura symptoms
33] and all-cause mortality [34] further illustrate persist longer than 60 min, with neuroimaging
the broad spectrum of lesions associated with proof of an associated ischemic brain lesion in
migraine, likely to be explained via complex an appropriate region and absence of other under-
relationships [35]. lying causes (Table 3).
The relationship between migraine and stroke From this definition, it is indirectly evident that
is complex, and the following paragraphs will migraine patients who present with “prolonged
expand on different aspects of this relationship; aura symptoms” require an appropriate neuroim-
neuroimaging examples will illustrate how aging workup with MRI and, when an acute ische-
migraine patients with (suspicion of) hemorrhagic mic lesion is present, additional diagnostic
or ischemic stroke may present. workup to exclude other underlying disorders. In
First, migrainous infarction and its imaging cases of coexisting other causes (like cardiac
appearances will be described, to be followed by arrhythmia, coagulation disorders, embolism
a paragraph on “aura-related” neuroimaging find- through a patent foramen ovale, cervical artery
ings, because the clinical symptoms of either dissection), the diagnosis then has to be (changed
“infarction” or “aura” are often very similar in to) ischemic stroke coexisting with migraine. The
the acute and subacute moments of presentation. same applies when in a patient with a history of
migraine without aura, an ischemic lesion
develops during or after a migraine attack. In
Migrainous Infarction other cases, when criteria are not completely ful-
filled, ischemic stroke in a migraine patient may
Kurth et al. suggested that probably the first report be categorized as cerebral infarction of other
on migrainous infarction came from Féré, who causes presenting with symptoms resembling
described a patient with migraine who died after migraine with aura [3].
2 months of headache, visual disturbances, and In the past decades, the diagnostic criteria for
hemiplegia [35, 36]. Various case reports of migrainous infarction have been changed (ICHD-
migrainous infarction have been published since I vs. ICHD-II), and studies inconsistently applied
then and have made clear that migraine can act as the criteria. This explains probably the relatively
a direct cause of ischemic stroke. In such cases, wide range (0.8–3.4 per 100,000) of reported
35 Migraine 797

annual incidences and points at a probable amount was relatively favorable. Although no studies
of overdiagnosis [37]. Although this implies that have systematically examined the appearance of
migrainous infarction is a rare condition, which is migrainous infarcts, notably when supraten-
further illustrated by Wolf et al. who estimated torially, from a number of case reports, it is
that it accounts for approximately two among suggested that the ischemic insults predominantly
1,000 “overall” strokes per year [38], it needs to may affect the cortex. Similarly, cortical ischemia
be considered that migrainous infarction predom- that crosses different vascular territories may also
inantly affects younger patients. In that age cate- point at a migrainous infarct mechanism, but this
gory, migrainous infarction was estimated to is probably an infrequent finding, since in the
account for 13 % of first-ever ischemic study by Wolf et al., no such “crossing” lesions
strokes [39]. were identified, neither on DWI nor on PWI
[38]. However, aura-related hypoperfusion (see
Neuroimaging Findings in Migrainous below) quite typically seems to cross territories.
Infarction The underlying mechanisms of migrainous
The largest series of migrainous infarction cases infarction are unknown, but are probably related
to date are reported by Wolf et al. (17 cases) [38] to CSD-related changes, including
and Laurell et al. (33 cases) [40]. In both reports, hypoperfusion, and changes in blood–brain bar-
patients underwent an appropriate stroke workup, rier permeability (which might lead to an exacer-
and diagnoses according to ICHD-II criteria were bation of local cellular injury caused by ischemia).
reported. Table 4 shows the main study findings. Together with factors predisposing to
Both studies reported a clear predominance of coagulopathy and release of vasoactive neuropep-
infarcts in the posterior circulation, supporting tides, further changes in cerebral hemodynamics,
previous observations. The low age at stroke arterial thrombosis, and infarctions may be
onset is a further key finding in both studies; explained [41].
therefore, when treating a young patient In the Figs. 1, 2, 3, and 4, case descriptions
presenting with stroke, migrainous infarction illustrate various presentations and appearances of
should be kept in mind. In both studies, outcome migrainous infarcts. In Fig. 5, a case with “cere-
bral infarction presenting with symptoms resem-
Table 4 Recent case series of migrainous infarction bling migraine with aura” is described and
Laurell illustrates that it can be difficult to apply a correct
et al. [40] Wolf et al. [38] and meaningful diagnosis, given the strict ICHD-
Cases n = 33 (all n = 17 (n = II criteria.
ICHD-II) 11 ICHD-II)a
M:F 39 %:61 % 23 %:77 % Neuroimaging Findings
Age at stroke 19–76, median 20–71, mean
onset 39 years 45 years
in (Prolonged) Aura
Posterior 82 % 71 %
According to the ICHD-II criteria, a diagnosis of
circulation persistent aura without infarct can be applied
Cerebellum 21 % 6% when aura symptoms remain present longer than
Multiple lesions 41 % 1 week and when there is no neuroradiological
Family history of 75 % 24 % evidence of ischemia. This is a rare condition that
migraine seems to affect genetic forms of migraine (like
Patent foramen 40 % 65 % familial hemiplegic migraine) somewhat more
ovale
often.
ICHD-II = International Classification of Headache Dis-
In “regular” migraine with aura patients,
orders, Second Edition [3]
a
n = 6 had a history of MO (thus not fulfilling the ICHD-II aura symptoms incidentally also persist longer
criteria in the strict sense) and presented with first-ever than 60 min. When more than one aura
neurologic symptoms compatible with MA and concomi- symptom is present (e.g., visual and sensory
tant migraine headaches
symptoms together or in succession) for each
798 M. Kruit

Fig. 1 Bilateral occipital migrainous infarction. A were no other abnormalities. CTA of the cervical and
33-year-old female patient with migraine without aura intracranial arteries was negative (not shown). There
from childhood and visual aura attacks since age 21. She were no other underlying causes. Follow-up MRI after
presented with usual visual aura symptoms but with now a 3 weeks showed only minimal residual hyperintensity on
persisting visual field defect, and persisting positive scin- the FLAIR images, consistent with nearly normalization of
tillating scotoma, accompanied by migraine headache. No the ischemic foci. Images: a1 and a2 FLAIR images; a3 and
other neurological signs and symptoms. Family history for a4 corresponding B1000 diffusion-weighted images in the
migraine with aura was positive. An MRI scan performed acute setting; b1 and b2 FLAIR images after 3 weeks
after 1 day of symptoms revealed bilaterally in the occipital follow-up
lobe small cortical areas of diffusion restriction. There

type, 60 min may be accepted. When aura persists related effects on the brain tissue and
longer, this might point at migrainous neurovascular system that will be discussed in
infarction, although most often symptoms still the next paragraphs.
spontaneously normalize, and probably, only inci- Cutrer et al. [42] and Sanchez del Rio et al. [43]
dentally, patients will be scanned. Imaging studies studied spontaneous migraine episodes with
in such “non-infarct” cases by definition do not perfusion-weighted MRI, including six patients
show ischemic changes, but various case reports studied during regular (not prolonged) visual
and series have described other CSD- or aura- aura, within 31  6 min after the onset of visual
35 Migraine 799

Fig. 2 Bilateral occipital and thalamic migrainous communicating arteries were not identified. Whole brain
infarction. A 61-year-old female patient, with long history perfusion CT (b) demonstrated reduced CBV and CBF and
of migraine with aura, presented with persisting left upper prolonged MTT and TTP values in the right occipital lobe,
quadrant visual field defect that had developed during a but also to a less degree in the left occipital lobe. MRI after
regular migraine with visual aura attack. Initial noncontrast one day (c) confirmed recent bilateral infarction, with signs
CT (a) dubiously showed some reduced gray-white matter of hemorrhagic transformation on the left side (arrow
differentiation in the right occipital lobe. CTA (not shown) head), but also identified right-sided thalamic infarction.
showed normal caliber of the carotids, the vertebrobasilar In the following diagnostic workup, no other underlying
system and the posterior cerebral arteries; the posterior causes were identified

symptoms. In all studies, perfusion deficits were MTT. Several small series and case reports have
observed in the occipital visual cortex from which been published since then, mostly with consistent
the hemifield defect was originating. Maximum findings of mild regional hypoperfusion, uni- or
measured changes were 37 % decrease in CBF, bilaterally affecting overlapping vascular
33 % decrease in rCBV, and 82 % increase in territories.
800 M. Kruit

Fig. 3 Bilateral cerebellar migrainous infarction. A was performed after few hours and showed bilateral cere-
45-year-old male patient with migraine with aura and bellar hypodensities consistent with bilateral cerebellar
visual aura attacks since age 35, with an average attack infarction. A subsequent MRI scan (T2 images, lower
frequency of 2 years, presented with usual visual aura row) confirmed the presence of three cerebellar and one
symptoms that lasted longer than normal, and were accom- vermian infarct. MRA of the cervical and intracranial
panied by sensory symptoms over his whole body, and arteries was negative (not shown). No other underlying
diplopia and dysarthria. Non-enhanced CT (upper row) causes were identified

Förster et al. reported a prospective study in There was no clear association between clinical
which patients with suspected acute ischemic symptoms and location of perfusion changes.
stroke were evaluated. In this study, 33 patients There were no diffusion-weighted imaging
with a final diagnosis of migraine with aura were (DWI) abnormalities related to the aura symp-
compared with age-matched patients with a final toms, and there were no vessel occlusions or ste-
diagnosis of acute ischemic stroke [44]. As a noses on MRA. In comparison with acute
consequence of the study methodology, in this ischemic stroke patients, the aura patients more
cohort, the number of patients with “rare” aura often had hypoperfusion involving more than one
symptoms (like hemihypesthesia, hemiparesis, territory and less increased TTP and MTT ratios.
aphasia) was overrepresented, as well as “acute A few reports pointed at the occurrence of
onset” of symptoms. In 54 % (n = 18) of migraine “crossed-cerebellar diaschisis” in cases with
with aura patients, PWI showed hypoperfusion aura-related perfusion changes. Dodick
that was involving more than the PCA territory et al. reported a migraine patient with typical
in all but one of the patients, although the PCA attacks of sensory aura during 30–60 min
territory was predominantly involved in 61 % of followed by headache, who showed reversible
cases. In seven patients (39 %), the hypoperfusion reduction in CBF in the left cerebral hemisphere
extended to the parietal, temporal, or frontal lobe. and associated crossed-cerebellar diaschisis
35 Migraine 801

Fig. 4 Subacute right occipital migrainous infarct. A the start of symptoms and showed a subacute occipital
58-year-old male patient with migraine with aura and corticosubcortical infarct on the right: hyperintensity on
visual and sometimes sensory aura attacks since childhood FLAIR and T2 slices (open arrows); cortical diffusion
presented with a history of recent persisting visual aura restriction and signs of cortical necrosis (arrows). Note a
symptoms for >1 week, followed by partial spontaneous small lipoma in the vermis, initially diagnosed as suspi-
recovery. MRI was performed to exclude ischemia or other cious for an AVM (arrowhead)
underlying causes. The scan was performed 12 days after

(hypoperfusion in the right cerebellar hemisphere) ischemic lesions in the cerebellum in migraine
during a typical attack on a brain SPECT scan patients (see later).
[45]. Iizuka et al. observed a case with prolonged Besides flow alterations, a variety of other
aura crossed-cerebellar hyperperfusion on brain imaging findings have been described on brain
SPECT scan on day 2 after symptom onset, imaging during (prolonged) migraine aura that
which was explained by the authors as a conse- all together may be explained by temporary
quence of uncoupled hyperperfusion with low changes in blood–brain barrier function that are
function in the left cerebral hemisphere probably secondary to aura-related cortical
(corresponding to the neurological deficits). Both spreading depression and/or spreading
the crossed hyperperfusion and hypoperfusion hypoperfusion. A number of reports described
illustrate the possibility of associated flow (and “vasogenic leakage,” that may present as regional
metabolic) alterations distant to and opposite of sulcal hyperintensity on native FLAIR MRI
the primary site of disturbances, which could be images [46], as (delayed) subarachnoid (sulcal),
relevant in understanding the occurrence of silent leptomeningeal, or cortical gadolinium
802 M. Kruit

Fig. 5 (continued)
35 Migraine 803

Fig. 5 Cerebral infarction presenting with symptoms diffusion-weighted images; c, sagittal and coronal FLAIR
resembling migraine with aura. A 46-year-old female images). Additional diagnostic workup remained negative
patient with migraine with aura and regular attacks of for other underlying causes. The patient recovered
(left-sided) visual aura since age 22, woke up in the morn- completely, although a follow-up MRI scan after 1 year
ing with right-sided hemiparesis and aphasia, with associ- (d, FLAIR images) showed postischemic cortical paren-
ated migraine-like headache, followed in the hospital by chymal loss. The cortically restricted infarct was consid-
unilateral pulsating headache with nausea and vomiting, ered somewhat unusual for ordinary MCA stroke and
resembling her prior migraine attacks. The noncontrast CT might have been due to aura-related CSD. However,
in the acute setting showed slight hypodensity (not shown). because the infarction was not clearly temporally related
An MRI scan 1 day later confirmed a cortically restricted to a migraine with aura attack that was similar to previous
zone of cytotoxic edema in the left MCA territory involv- attacks, the diagnosis was cerebral infarction presenting
ing parts of the frontoparietal and insular cortex, consistent with symptoms resembling migraine with aura and not
with her symptoms (a, T2-weighted images; b, B1000 migrainous infarction

enhancement on FLAIR or T1-weighted MRI Over the past four decades, many observa-
scans [47–50] or as increased vascular permeabil- tional studies, hospital-based stroke case-control
ity on perfusion-weighted MRI [51]. In few cases studies, and population-based studies have evalu-
with (prolonged) aura, symptoms may be associ- ated the association between migraine and clinical
ated with reversible cortical swelling and ischemic stroke. Meta-analyses [27, 53] of these
hyperintense signal changes on FLAIR images, studies summarized that migraine patients are
distributed along the regional cortical ribbon about at doubled increased risk, which notably
corresponding to the symptoms, without clear applies to patients with migraine with aura.
diffusion restriction (vasogenic edema) [52]. Sch€urks calculated a pooled relative risk of 1.7
(95 % CI 1.3–2.3) for migraineurs compared to
controls [27]. From eight studies, data specified
Migraine as a Risk Factor for Clinical by migraine subtype was available, resulting in a
Ischemic Stroke pooled relative risk of 2.2 (95 % CI: 1.5–3.0) for
patients with migraine with aura and 1.2 (95 % CI:
Besides the (rare) presentation of “migrainous 0.9–1.7) for patients with migraine without aura,
infarction” (i.e., directly related to a migraine compared to controls. A higher migraine fre-
attack), migraine patients have also a higher quency seems also to further increase the risk of
chance to present with an ischemic or hemor- ischemic stroke. Due to the lower prevalence of
rhagic stroke unrelated to a migraine attack. migraine in men, the association between
804 M. Kruit

migraine and ischemic stroke is less certain Oygarden et al. suggested that the higher fre-
in men. quency of smaller lesions in migraineurs with aura
The risk is highest in women, notably at youn- may point at a higher vulnerability to damage
ger age (<45 years), and in this group, the risk following minor ischemic insults (similarly as
further increases (up to 10x increased risk) with was described in experiments in mice with famil-
concurrent use of oral contraceptives. In individ- ial hemiplegic migraine) or that ischemia in
ual studies, effects of concurrent hypertension and migraineurs may simultaneously lead to a small
heavy smoking showed a greater than multiplica- “clinically silent” ischemic lesion (that normally
tive effect on the risk [54, 55]. Associations with would not be noticed) and ischemia-triggered
markers of endothelial dysfunction and factors CSD resulting in “clinical” stroke like neurologi-
linked to prothrombotic or proinflammatory con- cal deficits leading to hospital admission [58].
ditions have also been suggested as explanatory Although the earlier study results were criti-
for the increased stroke risk in migraine cized (e.g., because of potential misclassification,
[56]. Genetic factors, including polymorphisms referral bias, lack of neuroimaging proof, etc.), the
in the MTHFR, ACE, MEPE, and IRX4 genes, consistent findings in several studies over the
have been linked with both migraine and ischemic years have migraine set today as an acknowledged
stroke, but further studies need to assess the path- ischemic stroke risk factor. It has to be noted that
ophysiological relevance of such findings. in absolute terms, stroke in young women with
migraine (estimated at 5.5/100,000 annually) [59]
Topographic and Pathophysiological remains rare, although at the same time, in this age
Considerations group, migraine should be considered as an
Few structured data exist on the topography of important risk factor.
non-migrainous infarcts in migraine patients. In a
large series of 3,500 patients with acute stroke,
130 (3.7 %) had active migraine, and about 50 % Migraine as a Risk Factor for Clinical
of these were <45 years of age. In the younger Hemorrhagic Stroke
patients, posterior circulation involvement (55 %)
was characteristic [57]. From case reports and Data on a possible association between migraine
small series, it was also suggested that the occipital and hemorrhagic stroke have remained inconsis-
lobe and/or the posterior cerebral artery territory tent for years. However, in 2013, Sacco
seem to be overrepresented in migraine patients et al. meta-analyzed a total of four case-control
with clinical ischemic stroke. Recently, Oygarden and four cohort studies that together included
et al. retrospectively investigated whether the 320,539 individuals in whom 1,600 intracerebral
lesion pattern on diffusion-weighted MRI scans or subarachnoid hemorrhages occurred
was different between migraineurs (with and with- [28]. Migraine patients were found to be at signif-
out aura; n = 196) and non-migraineurs (n = 720) icantly increased risk for hemorrhagic stroke
with clinical ischemic stroke [58]. In the migraine (OR 1.48; 95 % CI 1.2–1.9; P = 0.001). There
group, younger patients and women were overrep- was no clear difference between migraine with
resented, and infarcts were more often due to aura (OR 1.6; 95 % CI 0.9–3.0; P = 0.13) or
cardioembolism and less often due to small vessel migraine without aura (OR 1.4; 95 % CI
disease than in the control group. Migraine patients 0.7–2.62 P = 0.3), male and female patients, or
presented more often with symptoms from the younger and older subjects with migraine,
posterior circulation and had more cortical although Kuo et al. reported higher risks of
(OR 1.8 CI: 1.3–2.5), small (OR 1.9 CI: migraine with aura in male patients and patients
1.04–3.5), cerebellar (P = 0.026), and occipital <45 years of age [60]. A limitation in the review
infarcts (13.3 % vs. 8.6 %; P = 0.05). Migraine by Sacco et al. was that subarachnoid and intrapar-
patients with infarcts had a three times higher enchymatous hemorrhages could not be sepa-
chance to have a patent foramen ovale (PFO). rated. Next to ischemic stroke, migraine patients
35 Migraine 805

are thus likely also at increased risk for hemor- The higher risk for subjects with higher attack
rhagic stroke, although mechanisms likely differ frequency may point at migraine attack-related
and remain to be elucidated, and subgroups most mechanisms. During and after migraine attacks,
at risk still need to be identified. sluggish cerebral flow below an ischemic thresh-
old has been described [42, 43, 62–64]. A
decrease in brain perfusion pressure (e.g., during
Migraine as a Risk Factor for Subclinical migraine) theoretically affects the clearance and
Stroke destination of embolic particles; narrowing of the
arterial lumen and endothelial abnormalities stim-
Posterior Circulation Infarcts ulate formation of thrombi; occlusive thrombi
The population-based cross-sectional CAMERA further reduce blood flow and brain perfusion.
MRI study (n = 435) assessed whether migraine Because the deep cerebellar territories have a pat-
cases were at increased risk of several types of tern of progressively tapering arteries with only
“silent” (or subclinical) brain lesions and whether few anastomoses present, they are likely to be
certain areas of the brain were particularly vulner- particularly vulnerable to hypoperfusion-related
able [61]. None of the participants reported a borderzone infarct mechanisms. This
history of stroke or transient ischemic attack or hypoperfusion-related concept matches the find-
showed relevant abnormalities at standard neuro- ings of previous studies in which the small cere-
logical examination. bellar borderzone infarcts, in particular when
In the migraine with aura group, 8 % of sub- multiple, were strongly associated with severe
jects had one or more posterior circulation occlusive and/or (artery-to-artery) embolic dis-
infarcts. This was significantly higher compared ease based on vertebrobasilar atherosclerosis,
to controls (0.7 %; P = 0.005) and migraineurs likely to result in hypoperfusion and infarction.
without aura (2 %). The highest risk was found in The population-based AGES-Reykjavik study
MA with 1 attack per month (OR, 15.8; 95 % CI, (n = 4,689) confirmed these findings and also
1.8–140). Infarct size ranged from 2 to 21 mm. reported a significantly higher prevalence of cer-
Most lesions were located in the cerebellum ebellar infarcts on MRI scans in female migraine
(Fig. 6), typically in a borderzone location. The with aura patients at late life (23 % vs. 15 %; P <
average number of lesions per subject was 1.8. 0.001) [65]. In that study, there was no increased
Although migraine-related clinical strokes seem risk for cortical or subcortical infarcts on MRI for
to have a predilection for the occipital lobes (see migraine patients.
above), in the CAMERA study, none of the pos- In the 9-year follow-up CAMERA 2 study,
terior circulation infarcts were in the occipital 66 % of the original sample was rescanned with
lobes. the same MRI scanners and protocols. None of the
Migraine patients with posterior circulation infarcts present at baseline had disappeared. Only
infarcts were significantly older, but cardiovascu- in the migraine group new posterior circulation
lar risk factors were not more prevalent, and the infarcts had occurred (5 % vs. 0 %; P = 0.07) [31].
presence of these lesions was not significantly
associated with supratentorial brain changes, Other Population-Based Evidence
such as WMLs. These two observations suggest for Silent Infarcts in Migraine
that the lesions are not atherosclerotic in origin. In the EVA study (Epidemiology of Vascular Age-
The combination of vascular distribution, deep ing; n = 780), migraine with aura patients (n =
borderzone location, shape, size, and imaging 17) had over a threefold increased risk (OR 3.4;
characteristics on MR imaging makes it likely 95 % CI 1.2–9.3) compared to controls (n = 617)
that the lesions have an infarctious origin. The for any infarct on MRI, and there was a suggestion
most likely etiologic mechanism seems to be that migraine with aura patients were at increased
hypoperfusion and/or embolism, rather than ath- risk for multiple infarcts (OR 3.7; 95 % CI
erosclerosis or small vessel disease. 0.8–17) [66]. In this study, most infarcts were
806 M. Kruit

Fig. 6 Cerebellar infarcts in migraine with aura patients. Corresponding T2-weighted (left) and FLAIR (right) MRI
images showing (multiple) cerebellar infarcts (arrowheads) in three migraine with aura patients from the CAMERA study

located outside the cerebellum or brainstem, and patients had a significantly higher risk of subclin-
the number of patients was unfortunately too ical brain infarcts (OR 2.1; 95 % CI 1.0–4.2),
small for further statistical testing of specific which was even higher in migraineurs
infarct locations like the cerebellum (5.9 % without aura (OR 2.6; 95 % CI 1.3–5.5).
vs. 2.8 %) and thalamus (11.8 % vs. 2.1 %) in In the groups of participants >75 years of
migraine with aura versus control participants. age, 30 % of migraineurs had at least
The MRI substudy of the NOMAS study 1 infarct on MRI, compared to 15 % of controls.
(Northern Manhattan Study) included n = In this study, infarcts were found most commonly
546 racial/ethnically diverse population-based in the white matter (13 %) and cerebellum
participants; 65 % were Hispanic. Migraine (10 %) [67].
35 Migraine 807

In summary, an association between migraine WMLs in Migraine: The CAMERA-Study


and subclinical infarcts is well established by now. 1 and 2
Similarly as for clinical ischemic stroke, notably, Because the clinic-based studies mentioned above
migraine with aura patients are at highest risk. The suffered from methodological difficulties, and
exact mechanisms and potential consequences might have evaluated a more than average severe
need further evaluation and research. The poste- subgroup of migraine patients (due to their
rior circulation territory is probably most vulner- “clinic-based” origin), the cross-sectional popula-
able in migraine patients. tion-based CAMERA MRI study was carried out
(see above in paragraph “Migraine as a Risk Fac-
tor for Clinical Ischemic Stroke”). Among women
Migraine and T2 Hyperintensities of the CAMERA cohort, the risk of having a high
on MRI deep WML load (top 20th percentile of the distri-
bution of deep WML load, Fig. 7) was increased
White Matter Lesions in Migraine in migraineurs compared to controls (OR 2.1;
(WMLs) 95 % CI 1.0–4.1). This risk was higher in those
with higher attack frequency (1 attack/per
Already from the early days of MRI, reports exist month; OR 2.6; 95 % CI 1.2–5.7) and was similar
on the presence of WMLs on T2-weighted images among women with migraine with and without
in migraine patients. Initial clinic-based studies aura. Among men, prevalence of deep WMLs
presented data on WMLs in migraine patients, but did not differ between controls and migraineurs.
results were often uncontrolled, inconsistent, or No association was found between severity of
conflicting. Some studies evaluated migraine sub- periventricular WMLs and migraine, irrespective
types, also with inconsistent results. One study of gender or migraine frequency or subtype.
found that migraine attack frequency was related The finding of higher risks in those with a
to an increased prevalence of WMLs. Despite the higher attack frequency was suggestive of a
various limitations and discrepancies in those MRI potential causal relationship between migraine
studies, a meta-analysis [29] summarized the attacks or attack severity and the development of
results of seven case-control studies and reported WMLs. Therefore, the 9-year follow-up CAM-
that migraine patients were at 4 increased risk ERA 2 study (n = 286, mean age 57  8 years)
(OR: 3.9; 95 % CI 2.3–6.7) for WMLs, regardless was carried out to test for accumulative effects of
of comorbidities like cardiovascular risk factors, recurrent attacks and to study the underlying
demyelinating disease, inflammatory conditions, mechanisms and possible cognitive
and valvular heart disease. consequences [31].

Fig. 7 Focal small- to medium-sized deep WMLs in a 47-year-old female migraine patient
808 M. Kruit

Fig. 8 FLAIR images showing preexisting (arrowhead) and newly developed (arrows) deep WMLs after 9 years of
follow-up in a 37-year-old (at baseline) female patient with migraine without aura

In the follow-up study, there were again no WMLs in Migraine: Other Population-
differences in WMLs between male participants Based Evidence
with migraine versus controls. However, deep In the longitudinal EVA study (n = 780; mean age
WML volume was higher in female migraineurs 69  3), participants with a history of any severe
compared to controls (P = .04), and their deep headache (including migraine) were at increased
WML progression was more severe (77 % risk of higher WML volumes (OR 2.0, 95 % CI
vs. 60 %; P = .02), which was highest in migraine 1.3–3.1) compared to controls, with similar find-
without aura (83 %). Multivariate logistic regres- ings for deep and periventricular WMLs [66]. In
sion showed that migraine was independently participants with migraine with aura, the associa-
associated with deep WML progression (OR 2.1; tions with WMLs were strongest.
95 % CI 1.0–4.1; P = .04). The increase in total A subset of participants of the ARIC study
deep WML volume was explained by an (Atherosclerosis Risk in Communities cohort
increased number of new lesions, rather than by study; n = 1,028) received two MRI examina-
increase in size of preexisting lesions. Figure 8 tions, between 8 and 12 years apart. The authors
shows an example of incident lesions in a showed also in this study that migraine without
female migraine patient after 9 years of aura (OR 1.9; 95 % CI 1.0–3.4) is associated with
follow-up. The mean size of individual WMLs in cross-sectional analysis. However, they
hyperintensities at follow-up did not differ failed to demonstrate that migraine was associated
between participants with migraine and controls. with WML volume progression. Differences in
Among females with migraine, deep WMLs lesion–quantification methodology, in the defini-
appeared to be more diffusely distributed com- tion of progression, in the older age, size, and
pared to controls (Fig. 9). Hypertension and dia- other characteristics of the cohort, etc., might
betes were not associated with a higher incidence explain why the findings seem to contrast with
of DWMH progression. Exploratory analyses the findings of the CAMERA 2 study.
showed no association of number of migraine Unexpectedly, in the NOMAS study (n = 546;
attacks, migraine attack duration, migraine fre- two thirds of participants was >60 years of age),
quency, type of attack, migraine therapy, with no association between WML volume and
deep WML progression. migraine or its subgroups was found. The authors
35 Migraine 809

Fig. 9 Topography and progression of deep WMLs in maps (normalized for differences in group size; controls,
female migraine patients and controls. Baseline and N = 52; migraine with aura, N = 75; migraine without
follow-up deep WMLs are projected on transparent 3D aura, N = 57)

suggested that no difference was detected because to tissue damage. Reversible MRI abnormalities
of the high burden of other cardiovascular risk during migraine aura, including areas of increased
factors in the racially diverse older cohort. vasogenic leakage [52] and evidence of
blood–brain barrier dysfunction in prolonged
Pathophysiological Mechanisms aura [47, 48, 50], seem to illustrate the possibility
Although the pathologic substrate of WMLs in of direct effects to the brain during attacks, most
migraine remains unknown, the most likely histo- likely first acting on the level of the microvascu-
logical substrate of these abnormalities is incom- lature [71], and possibly being enhanced by other
plete infarction with changes such as gliosis and factors like matrix metalloproteinase nine-
demyelination [68]. The causative mechanisms, dependent cascade mechanism, that may increase
however, remain largely unknown. Different the risk of local tissue damage [72].
explanatory options have been described. On the other hand, because the progression of
Attack-related mechanisms may play a role, as WMLs and occurrence of infarcts in the CAM-
was suggested by the higher risk of WMLs in the ERA cohort were not dependent on persisting
CAMERA 1 study and as seems likely given the migraine activity, and also because of the attack-
occurrences of migrainous infarcts. During unrelated increased risk of clinical ischemic
attacks, reduced blood flow in large and/or small strokes in migraineurs, attack-unrelated factors
arteries [43], possibly in combination with vaso- also seem to play a role. With increasing age,
constriction or activation of the clotting system/ when attacks generally diminish, other systemic
platelets, might lead to formation of local thrombi. migraine “disease-related” conditions leading to
Alternatively, local tissue changes during WMLs are possibly increasing and likely compli-
migraine attacks, such as excessive neuronal acti- cate the detection of attack-related mechanisms.
vation, neurogenic inflammation, neuropeptide Attack-unrelated factors could, e.g., include
and cytokine release [69], or excitotoxity [70], chronic procoagulatory or proinflammatory
may occur, and such changes may directly lead changes due to endothelial dysfunction [73, 74],
810 M. Kruit

Fig. 10 Infratentorial hyperintense lesions (IHLs) in developed lesions (a, arrowheads), additional lesions (b,
migraine. T2-weighted images of baseline and 9-year open arrows), or increases in size (c, arrows), compared to
follow-up of three migraine patients from the CAMERA baseline
study, showing increasing loads of IHLs: either newly

elevated homocysteine levels [75], or recurrent [31]. Figure 10 shows typical examples of T2
paradoxical (micro-) emboli due to right-to-left hyperintensities in the brainstem and their pro-
shunts [76]. In addition, population-based evi- gression over time.
dence that migraine (with aura) is associated Typically, brainstem lesions were located in the
with a higher cardiovascular risk profile might dorsal basis pontis, adjacent to the tegmentum, at
contribute to the risk of (ischemic) brain the level of, and slightly cranial to, the entry zone
lesions [77]. of the trigeminal nerve. In nearly all cases, lesions
were located bilaterally, sometimes extending to
Brainstem Lesions the midline; none reached the surface of the pons.
In the CAMERA 1 study, also infratentorial Hyperintensities seem to involve the
hyperintense lesions (IHLs) were significantly pontocerebellar fibers, the pontine nuclei, or the
more prevalent in migraine patients (4.4.%) com- nucleus reticularis tegmenti pontis, and, in some
pared with controls (0.7 %; P = 0.04) [61]. The cases, parts of the corticopontine (pyramidal tract)
majority of the IHLs were located in the pons. The or medial lemniscus fibers. These anatomical
CAMERA 2 study showed that after 9 years locations are supplied by the anteromedial and
follow-up, the prevalence of IHLs remained anterolateral groups arising from the basilar
higher in women with versus without migraine artery.
(21 % vs. 4 %; adjusted OR, 6.5; 95 % CI, Earlier reports in non-migraineurs linked
1.5–28.3; P = .01) and that they also more often pontine IHLs to patients with cardiovascular
showed progression of IHLs (15 % vs. 2 %; risk factors, leukoaraiosis, lacunar infarcts,
adjusted OR, 7.7; 95 % CI, 1.0–59.5; P = .05) and poor clinical outcome after stroke
35 Migraine 811

[78]. Histopathologically, pontine IHLs corre- in adults with migraine does therefore generally
spond to myelin pallor and reactive astrocytosis. not need further medical attention.
The pathophysiology of these lesions is assumed Incidentally, WMLs are observed on MRI
to be comparable as for lesions in subcortical scans in relatively young patients, e.g., below
arteriosclerotic encephalopathy, i.e., ischemia sec- age 40. In such cases, number, location, aspect,
ondary to small artery sclerosis. There is a good and distribution of the lesions have to be carefully
correlation between MRI findings and histopa- evaluated, and based on the images and clinical
thology [79]. Similar hyperintense brainstem history together, the likelihood of other diseases
lesions are also frequent in CADASIL, a disorder that are associated with white matter lesions, such
in which migraine with aura is often the as multiple sclerosis, vasculitis, CADASIL,
presenting symptom [80]. In CADASIL, MELAS, coagulation disorders, cardiac abnor-
decreased cerebral perfusion secondary to malities, etc., has to be considered. “Migraine” is
changes in the wall of cerebral arteries leads to part of this differential diagnostic list, but other
early damage of the white matter. Regions that are options should never be disregarded.
irrigated by the longest perforating arteries are Similarly, the identification of one or more
most vulnerable to hypoperfusion. This is partic- silent cerebellar infarcts on an MRI scan can’t
ularly the case for the central part of the pons. directly be attributed to “migraine,” although
Similarly, as discussed above for DWMLs, these findings – with the data from the CAMERA
migraine attacks might be causally related to the study, showing 8 % of patients with migraine with
brainstem lesions, because repeated or prolonged aura from the general population affected – are not
reduced perfusion has been described in migraine so unusual in migraine patients with aura. In such
attacks, although not specifically in the pons cases, if the observation is incidental, no direct
[64]. Alternatively, attack-unrelated “systemic” clinical consequence seems to be necessary. If
factors could also explain the association (see future research indicates that there is a risk of
above), may be in combination with suggested progression of number or size of lesions or if
impaired adaptive cerebral hemodynamic mecha- there are associated functional consequences,
nisms in the posterior circulation of migraine additional study of causes and evaluation of use-
patients. fulness of preventive therapy has to be initiated.
However, a patient who presents with an acute
cerebellar infarct, irrespective of the size of the
“Subclinical” MRI Findings lesion or presence of migraine, has to be screened
in the Individual Migraine Patient for (embolic) sources of the infarction and treated
accordingly.
The knowledge from hospital-based and
population-based studies that migraine is an inde-
pendent risk factor for DWMLs, IHLs, and sub- Summary
clinical infarcts does not imply that when such
lesions are identified on a brain MRI scan in a Migraine is a very prevalent disorder. Knowledge
migraine patient, migraine is “the cause.” In rou- of the migraine pathophysiology allows better
tine neuroradiological practice, T2 hyperintense interpretation of neuroimaging findings in
lesions in the white matter are frequently encoun- migraine patients, who may present both in acute
tered in subjects above age 50 but also appear in situations and in regular outpatient settings.
younger individuals. No studies found clear pref- A migraine patient may present during a
erential locations or distribution patterns for migraine attack with neurologic deficits. Normal
WMLs migraine patients. Therefore, in most neurologic aura symptoms need to be differenti-
patients with migraine and WMLs, the lesions ated from “prolonged aura” and ischemic stroke.
will remain “aspecific.” The identification of a When a clinical diagnosis is not possible, urgent
limited number of small- to medium-sized lesions neuroimaging is mandatory. Neuroimaging may
812 M. Kruit

show heterogeneous changes during “prolonged 6. Linde M, Gustavsson A, Stovner LJ, Steiner TJ,
aura” that mostly can be differentiated from find- Barré J, Katsarava Z, Lainez JM, Lampl C, Lantéri-
Minet M, Rastenyte D, Ruiz de la Torre E, Tassorelli C,
ings in true ischemia. If ischemia is identified Andrée C (2012) The cost of headache disorders in
during a migraine attack, this might be either due Europe: the Eurolight project. Eur J Neurol
to true migrainous infarction, due to ischemic 19:703–711. doi:10.1111/j.1468-1331.2011.03612.x
stroke coexisting with migraine, or due to cerebral 7. Sprenger T, Borsook D (2012) Migraine changes the
brain: neuroimaging makes its mark. Curr Opin Neurol
infarction of other causes presenting with symp- 25:252–262. doi:10.1097/WCO.0b013e3283532ca3
toms resembling migraine with aura. 8. Ligthart L, De VB, Smith AV, Ikram MA, Amin N,
Migraine patients, notably those with aura and Hottenga JJ, Koelewijn SC, Kattenberg VM, de Moor
in the younger age category, are also at increased MH, Janssens AC, Aulchenko YS, Oostra BA, de Geus
EJ, Smit JH, Zitman FG, Uitterlinden AG, Hofman A,
risk of clinical ischemic of hemorrhagic stroke Willemsen G, Nyholt DR, Montgomery GW, Terwindt
unrelated to a migraine attack. The exact GM, Gudnason V, Penninx BW, Breteler M, Ferrari
mechanisms behind these associations are still MD, Launer LJ, van Duijn CM, van den Maagdenberg
unknown, although attack-unrelated “systemic” AM, Boomsma DI (2011) Meta-analysis of genome-
wide association for migraine in six population-based
proinflammatory, prothrombotic, endothelial, car- European cohorts. Eur J Hum Genet 19:901–907
diac, and genetic factors probably play a role and 9. Breslau N, Lipton RB, Stewart WF, Schultz LR, Welch
may be part of “migraine as a disease.” Migraine KM (2003) Comorbidity of migraine and depression:
is associated with subclinical infarcts, investigating potential etiology and prognosis. Neurol-
ogy 60:1308–1312
supratentorial, and infratentorial T2 hyperintense 10. Goadsby PJ, Lipton RB, Ferrari MD (2002) Migraine –
lesions on MRI. The higher prevalence of such current understanding and treatment. N Engl J Med
lesions is probably explained by similar patho- 346:257–270
physiological mechanisms. Migraine is part of 11. Weiller C, May A, Limmroth V, Juptner M, Kaube H,
Vonschayck R, Coenen HH, Diener HC (1995) Brain-
the differential diagnostic list of WMLs and cere- stem activation in spontaneous human migraine
bellar infarcts, but other options should never be attacks. Nat Med 1:658–660
disregarded. 12. Cao Y, Aurora SK, Nagesh V, Patel SC, Welch KMA
Neuroimaging in migraine patients is overused (2002) Functional MRI-BOLD of brainstem structures
during visually triggered migraine. Neurology
in non-acute situations; recommendations have 59:72–78
been formulated to identify patient in whom neu- 13. Sánchez del Rio M, Linera JA (2004) Functional neu-
roimaging is warranted. roimaging of headaches. Lancet Neurol 3:645–651
14. May A (2009) New insights into headache: an
update on functional and structural imaging
findings. Nat Rev Neurol 5:199–209. doi:10.1038/
nrneurol.2009.28
References 15. Stankewitz A, May A (2011) Increased limbic and
brainstem activity during migraine attacks following
1. Olesen J (2004) The international classification of olfactory stimulation. Neurology 77(5):476–482.
headache disorders, 2nd edn (ICDH-II). J Neurol doi:10.1212/WNL.0b013e318227e4a8
Neurosurg Psychiatry 75:808–811. doi:10.1136/ 16. Miranda-Cardenas Y, Rojas-Piloni G, Martinez-
jnnp.2003.031286 Lorenzana G, Rodriguez-Jimenez J, Lopez-Hidalgo M,
2. Goadsby PJ (2001) Migraine, aura, and cortical spread- Freund-Mercier MJ, Condes-Lara M (2006) Oxytocin
ing depression: why are we still talking about it? and electrical stimulation of the paraventricular hypo-
AnnNeurol 49:4–6 thalamic nucleus produce antinociceptive effects that
3. Olesen J (2006) International classification of headache are reversed by an oxytocin antagonist. Pain
disorders, second edition (ICHD-2): current status and 122:182–189
future revisions. Cephalalgia 26:1409–1410. 17. Holden JE, Farah EN, Jeong Y (2005) Stimulation of
doi:10.1111/j.1468-2982.2006.01214.x the lateral hypothalamus produces antinociception
4. Launer LJ, Terwindt GM, Ferrari MD (1999) The prev- mediated by 5-HT1A, 5-HT1B and 5-HT3 receptors
alence and characteristics of migraine in a population- in the rat spinal cord dorsal horn. Neuroscience
based cohort: the GEM study. Neurology 53:537–542 135:1255–1268
5. Menken M, Munsat TL, Toole JF (2000) The global 18. Akerman S, Holland PR, Goadsby PJ (2011) Dience-
burden of disease study: implications for neurology. phalic and brainstem mechanisms in migraine. Nat Rev
Arch Neurol 57:418–420 Neurosci 12:570–584. doi:10.1038/nrn3057
35 Migraine 813

19. Ayata C (2009) Spreading depression: from serendipity 33. Bigal ME, Kurth T, Santanello N, Buse D, Golden W,
to targeted therapy in migraine prophylaxis. Robbins M, Lipton RB (2010) Migraine and cardio-
Cephalalgia 29:1095–1114 vascular disease: a population-based study. Neurology
20. Welch KM, Cao Y, Aurora S, Wiggins G, Vikingstad 74:628–635. doi:10.1212/WNL.0b013e3181d0cc8b
EM (1998) MRI of the occipital cortex, red nucleus, 34. Gudmundsson LS, Scher AI, Aspelund T, Eliasson JH,
and substantia nigra during visual aura of migraine. Johannsson M, Thorgeirsson G, Launer L, Gudnason
Neurology 51:1465–1469 V (2010) Migraine with aura and risk of cardiovascular
21. Lauritzen M (1994) Pathophysiology of the migraine and all cause mortality in men and women: prospective
aura. The spreading depression theory. Brain 117 cohort study. BMJ 341:c3966. doi:10.1136/bmj.c3966
(Pt 1):199–210 35. Kurth T, Chabriat H, Bousser M-G (2012) Migraine
22. Zhang X, Levy D, Kainz V, Noseda R, Jakubowski M, and stroke: a complex association with clinical impli-
Burstein R (2011) Activation of central trigemino- cations. Lancet Neurol 11:92–100. doi:10.1016/
vascular neurons by cortical spreading depression. S1474-4422(11)70266-6
Ann Neurol 69:855–865 36. Féré C (1883) Note sur un cas de migraine ophtalmique
23. Detsky ME, McDonald DR, Baerlocher MO, à accès répétés suivis de mort. Rev Med 3:194–201
Tomlinson GA, McCrory DC, Booth CM (2006) 37. Bousser M-G (2004) Estrogens, migraine, and
Does this patient with headache have a migraine or stroke. Stroke 35:2652–2656. doi:10.1161/01.
need neuroimaging? JAMA 296:1274–1283. STR.0000143223.25843.36
doi:10.1001/jama.296.10.1274 38. Wolf ME, Szabo K, Griebe M, Förster A, Gass A,
24. Silberstein SD (2000) Practice parameter: evidence- Hennerici MG, Kern R (2011) Clinical and MRI
based guidelines for migraine headache (an evidence- characteristics of acute migrainous infarction.
based review): report of the Quality Standards Sub- Neurology 76:1911–1917. doi:10.1212/
committee of the American Academy of Neurology. WNL.0b013e31821d74d5
Neurology 55:754–762 39. Arboix a, Massons J, García-Eroles L, Oliveres M,
25. Sandrini G, Friberg L, Coppola G, Jänig W, Jensen R, Balcells M, Targa C (2003) Migrainous cerebral infarc-
Kruit M, Rossi P, Russell D, Sanchez Del Rìo M, tion in the Sagrat Cor Hospital of Barcelona stroke
Sand T, Schoenen J (2011) Neurophysiological tests registry. Cephalalgia 23:389–394
and neuroimaging procedures in non-acute headache 40. Laurell K, Artto V, Bendtsen L, Hagen K, Kallela M,
(2nd edition). Eur J Neurol 18:373–381. doi: 10.1111/ Meyer EL, Putaala J, Tronvik E, Zwart JA, Linde M
j.1468-1331.2010.03212.x (2011) Migrainous infarction: A Nordic multicenter
26. Wilbrink LA, Ferrari MD, Kruit MC, Haan J (2009) study. Eur J Neurol 18:1220–1226. doi: 10.1111/
Neuroimaging in trigeminal autonomic cephalgias: j.1468-1331.2011.03364.x
when, how, and of what? Curr Opin Neurol 41. Kruit MC, Launer LJ, Ferrari MD, van Buchem M
22:247–253. doi:10.1097/WCO.0b013e32832b4bb3 (2005) Infarcts in the posterior circulation territory
27. Schurks M, Rist PM, Bigal ME, Buring JE, Lipton RB, in migraine. The population-based MRI CAMERA
Kurth T (2009) Migraine and cardiovascular disease: study. Brain 128:2068–2077. doi:10.1093/brain/
systematic review and meta-analysis. BMJ 339:b3914. awh542
doi:10.1136/bmj.b3914 42. Cutrer FM, Sorensen a G, Weisskoff RM,
28. Sacco S, Ornello R, Ripa P, Pistoia F, Carolei A (2013) Ostergaard L, Sanchez del Rio M, Lee EJ, Rosen BR,
Migraine and hemorrhagic stroke: a meta- Moskowitz M a (1998) Perfusion-weighted imaging
analysis. Stroke 44(11):3032–3038. doi:10.1161/ defects during spontaneous migrainous aura. Ann
STROKEAHA.113.002465 Neurol 43:25–31. doi:10.1002/ana.410430108
29. Swartz RH, Kern RZ (2004) Migraine is associated 43. Sanchez del Rio M, Bakker D, Wu O, Agosti R,
with magnetic resonance imaging white matter abnor- Mitsikostas D, Ostergaard L, Wells W, Rosen B,
malities: a meta-analysis. Arch Neurol 61:1366–1368. Sorensen G, Moskowitz M, Cutrer F (1999) Perfusion
doi:10.1001/archneur.61.9.1366 weighted imaging during migraine: spontaneous visual
30. Kruit M, Launer L, Van Buchem M, Terwindt G, aura and headache. Cephalalgia 19:701–707.
Ferrari M (2005) MRI findings in migraine. Rev doi:10.1046/j.1468-2982.1999.019008701.x
Neurol (Paris) 161:661–665 44. Förster A, Wenz H, Kerl HU, Brockmann MA, Groden
31. Palm-Meinders IH, Koppen H, Terwindt GM, Launer C (2014) Perfusion patterns in migraine with aura.
LJ, Konishi J, Moonen JME, Bakkers JTN, Hofman P Cephalalgia 34:11 870–876
a M, van Lew B, Middelkoop H a M, van Buchem M a, 45. Dodick DW, Roarke MC (2008) Crossed cerebellar
Ferrari MD, Kruit MC (2012) Structural brain changes diaschisis during migraine with prolonged aura: a pos-
in migraine. JAMA 308:1889–1897. doi:10.1001/ sible mechanism for cerebellar infarctions. Cephalalgia
jama.2012.14276 28:83–86. doi:10.1111/j.1468-2982.2007.01458.x
32. Kurth T, Gaziano JM, Cook NR, Logroscino G, Diener 46. Gómez-Choco M, Capurro S, Obach V (2008)
H-C, Buring JE (2006) Migraine and risk of cardiovas- Migraine with aura associated with reversible sulcal
cular disease in women. JAMA 296:283–291. hyperintensity in FLAIR. Neurology 70:2416–2418.
doi:10.1001/jama.296.3.283 doi:10.1212/01.wnl.0000314693.57386.f0
814 M. Kruit

47. Iizuka T, Sakai F, Suzuki K, Igarashi H, Suzuki N 61. Kruit MC, van Buchem M, Hofman PM, Bakkers JTN,
(2006) Implication of augmented vasogenic leakage Terwindt GM, Ferrari MD, Launer LJ, Zaidat OO
in the mechanism of persistent aura in sporadic hemi- (2004) Migraine as a risk factor for subclinical brain
plegic migraine. Cephalalgia 26:332–335. lesions. JAMA 291:427–434. doi:10.1001/
doi:10.1111/j.1468-2982.2005.01025.x jama.291.4.427
48. Iizuka T, Sakai F, Yamakawa K, Suzuki K, Suzuki N 62. Woods RP, Iacoboni M, Mazziotta JC (1994) Brief
(2004) Vasogenic leakage and the mechanism of report: bilateral spreading cerebral hypoperfusion dur-
migraine with prolonged aura in Sturge-Weber syn- ing spontaneous migraine headache. N Engl J Med
drome. Cephalalgia 24:767–770. doi:10.1111/j.1468- 331:1689–1692
2982.2004.00769.x 63. Olesen J, Friberg L, Olsen TS, Iversen HK, Lassen NA,
49. Lanfranconi S, Corti S, Bersano A, Costa A, Prelle A, Andersen AR, Karle A (1990) Timing and topography
Sciacco M, Bresolin N, Ghione I (2009) Aphasic of cerebral blood flow, aura, and headache during
and visual aura with increased vasogenic leakage: an migraine attacks. Ann Neurol 28:791–798
atypical migrainosus status. J Neurol Sci 285:227–229. 64. Bednarczyk EM, Remler B, Weikart C, Nelson AD,
doi:10.1016/j.jns.2009.06.009 Reed RC (1998) Global cerebral blood flow, blood
50. Smith M, Cros D, Sheen V (2002) Hyperperfusion with volume, and oxygen metabolism in patients with
vasogenic leakage by fMRI in migraine with prolonged migraine headache. Neurology 50:1736–1740
aura. Neurology 58:1308–1310 65. Scher AI, Gudmundsson LS, Sigurdsson S,
51. Rotstein DL, Aviv RI, Murray BJ (2012) Migraine with Ghambaryan A, Aspelund T, Eiriksdottir G, van
aura associated with unilateral cortical increase in vas- Buchem M a, Gudnason V, Launer LJ (2009) Migraine
cular permeability. Cephalalgia 32:1216–1219. headache in middle age and late-life brain infarcts.
doi:10.1177/0333102412462286 JAMA 301:2563–2570. doi:10.1001/jama.2009.932
52. Resnick S, Reyes-Iglesias Y, Carreras R, Villalobos E 66. Kurth T, Mohamed S, Maillard P, Zhu Y-C, Chabriat H,
(2006) Migraine with aura associated with reversible Mazoyer B, Bousser M-G, Dufouil C, Tzourio C
MRI abnormalities. Neurology 66:946–947. (2011) Headache, migraine, and structural brain lesions
doi:10.1212/01.wnl.0000203646.05540.ec and function: population based epidemiology of vas-
53. Etminan M, Takkouche B, Isorna FC, Samii A (2005) cular ageing-MRI study. BMJ 342:c7357. doi:10.1136/
Risk of ischaemic stroke in people with migraine: sys- bmj.c7357
tematic review and meta-analysis of observational stud- 67. Monteith T, Gardener H, Rundek T, Dong C,
ies. BMJ 330:63. doi:10.1136/bmj.38302.504063.8F Yoshita M, Elkind MSV, DeCarli C, Sacco RL, Wright
54. Chang CL, Donaghy M, Poulter N (1999) Migraine CB (2014) Migraine, white matter hyperintensities,
and stroke in young women: case–control study. The and subclinical brain infarction in a diverse
World Health Organisation Collaborative Study of Car- community: the northern Manhattan study.
diovascular Disease and Steroid Hormone Contracep- Stroke 45:1830–1832. doi:10.1161/
tion. BMJ 318:13–18 STROKEAHA.114.005447
55. Tzourio C, Kittner SJ, Bousser MG, Alperovitch A 68. Rocca MA, Colombo B, Pratesi A, Comi G, Filippi M
(2000) Migraine and stroke in young women. (2000) A magnetization transfer imaging study of the
Cephalalgia 20:190–199 brain in patients with migraine. Neurology 54:507–509
56. Tietjen G (2009) Migraine as a systemic vasculopathy. 69. Goadsby PJ (1997) Pathophysiology of migraine: a dis-
Cephalalgia 29:987–996 ease of the brain. In: Goadsby PJ, Silberstein SD (eds)
57. Milhaud D, Bogousslavsky J, Van Melle G, Liot P Headache. Butterworth-Heinemann, Boston, pp 5–25
(2001) Ischemic stroke and active migraine. Neurology 70. Eggers AE (2001) New neural theory of migraine. Med
57:1805–1811 Hypotheses 56:360–363
58. Oygarden H, Kvistad CE, Bjørk M, Thomassen L, 71. Ghabriel MN, Lu MX, Leigh C, Cheung WC, Allt G
Waje-Andreassen U, Naess H (2014) Diffusion- (1999) Substance P-induced enhanced permeability of
weighted lesions in acute ischaemic stroke patients dura mater microvessels is accompanied by pro-
with migraine. Acta Neurol Scand Suppl 129:41–46. nounced ultrastructural changes, but is not dependent
doi:10.1111/ane.12236 on the density of endothelial cell anionic sites. Acta
59. WHO Collaborative Study of Cardiovascular Disease Neuropathol 97:297–305
and Steroid Hormone Contraception (1996) Ischaemic 72. Gursoy-Ozdemir Y, Qiu J, Matsuoka N, Bolay H,
stroke and combined oral contraceptives: results of an Bermpohl D, Jin H, Wang X, Rosenberg GA, Lo EH,
international, multicentre, case–control study. WHO Moskowitz MA (2004) Cortical spreading depression
Collaborative Study of Cardiovascular Disease and activates and upregulates MMP-9. J Clin Invest
Ste. Lancet 348:498–505 113:1447–1455. doi:10.1172/JCI200421227.The
60. Kuo C-Y, Yen M-F, Chen L-S, Fann C-Y, Chiu Y-H, 73. Lee ST, Chu K, Jung KH, Kim DH, Kim EH, Choe VN,
Chen H-H, Pan S-L (2013) Increased risk of hemor- Kim JH, Im WS, Kang L, Park JE, Park HJ, Park HK,
rhagic stroke in patients with migraine: a population- Song EC, Lee SK, Kim M, Roh JK (2008) Decreased
based cohort study. PLoS One 8:e55253. doi:10.1371/ number and function of endothelial progenitor cells in
journal.pone.0055253 patients with migraine. Neurology 70:1510–1517
35 Migraine 815

74. Tietjen GE, Herial N a, White L, Utley C, Kosmyna JM, study. Neurology 64:614–620. doi:10.1212/01.
Khuder S a (2009) Migraine and biomarkers of endothe- WNL.0000151857.43225.49
lial activation in young women. Stroke 40:2977–2982. 78. Mantyla R, Pohjasvaara T, Vataja R, Salonen O,
doi:10.1161/STROKEAHA.109.547901 Aronen HJ, Standertskjold-Nordenstam CG, Kaste M,
75. Hamed S a (2009) The vascular risk associations with Erkinjuntti T (2000) MRI pontine hyperintensity after
migraine: relation to migraine susceptibility and pro- supratentorial ischemic stroke relates to poor clinical
gression. Atherosclerosis 205:15–22. doi:10.1016/j. outcome. Stroke 31:695–700
atherosclerosis.2008.10.016 79. Pullicino P, Ostrow P, Miller L, Snyder W, Munschauer
76. Schwedt TJ, Demaerschalk BM, Dodick DW (2008) F (1995) Pontine ischemic rarefaction. Ann Neurol
Patent foramen ovale and migraine: a quantitative sys- 37:460–466
tematic review. Cephalalgia 28:531–540 80. Chabriat H, Mrissa R, Levy C, Vahedi K, Taillia H,
77. Scher AI, Terwindt GM, Picavet HSJ, Verschuren Iba-Zizen MT, Joutel A, Tournier-Lasserve E, Bousser
WMM, Ferrari MD, Launer LJ (2005) Cardiovascular MG (1999) Brain stem MRI signal abnormalities in
risk factors and migraine: the GEM population-based CADASIL. Stroke 30:457–459
Moyamoya Disease (Spontaneous
Occlusion of the Circle of Willis) 36
Akira Yamamoto, Tomohisa Okada, and Jun C. Takahashi

Contents Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 827


An Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 827
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 819 Ischemic Symptoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 827
Epidemiological Features . . . . . . . . . . . . . . . . . . . . . . . . . . 819 Hemorrhage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 828
Geographical Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . . 819 Headache . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 829
Gender and Age Distribution . . . . . . . . . . . . . . . . . . . . . . . . 819 Involuntary Movement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 831

Familial Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . 821 Diagnostic Evaluation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 831


Current Diagnostic Criteria
Pathogenesis of Moyamoya Disease . . . . . . . . . . . . . . . 821 of Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 831
Cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 821 Definitive Moyamoya Disease and Probable
Genetic Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 822 Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 832
CT and CT Angiography in Moyamoya Disease . . . . 832
Asymptomatic Moyamoya Disease . . . . . . . . . . . . . . . . 822
MRI/MRA Diagnosis of Moyamoya Disease . . . . . . . 834
Unilateral Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . 824 FLAIR Ivy Sign . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 834
Feasibility of 3-T MRI/MRA
Quasi-moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . 825 in Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 836
Systemic Vessel Involvement Susceptibility Weighted Imaging in Moyamoya
of Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 825 Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 837

Pathological Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 827 Staging by Conventional Angiography


in Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 838
Angiographical Staging Progression
of Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 840
Aneurysm in Moyamoya Disease . . . . . . . . . . . . . . . . . . . 841
Roles of Morphological and Hemodynamic
Functional Imaging for MMD . . . . . . . . . . . . . . . . . . . . . 842
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 842
Staging of Moyamoya Disease by MRI/MRA . . . . . . 844
Altered Perfusion Status in MMD . . . . . . . . . . . . . . . . 845
A. Yamamoto (*) • T. Okada Evaluation of Perfusion Status . . . . . . . . . . . . . . . . . . . . 846
Department of Diagnostic Imaging and Nuclear Medicine, Oxygen-15 PET . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 847
Graduate School of Medicine, Kyoto University, SPECT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 847
Sakyo-ku, Kyoto, Japan CT Perfusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 848
e-mail: yakira@kuhp.kyoto-u.ac.jp; MR Perfusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 848
tomokada@kuhp.kyoto-u.ac.jp Arterial Spin Labeling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 848
J.C. Takahashi
Department of Neurosurgery, National Cerebral and
Cardiovascular Center, Suita City, Osaka, Japan
e-mail: juntak@ncvc.go.jp

# Springer Science+Business Media New York 2016 817


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_25
818 A. Yamamoto et al.

CBF Changes After Bypass Surgery . . . . . . . . . . . . . . 849 Abstract


Visualization of Lenticulostriate Moyamoya is a disease affecting the cerebral
Artery (LSA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 850 arteries, which predisposes individuals to
Treatment of Moyamoya Disease . . . . . . . . . . . . . . . . . . 851 recurrent ischemic attack in association with
Treatment for Ischemic Moyamoya Disease . . . . . . . . 851 progressive steno-occlusive change of the
Treatment for Hemorrhagic Moyamoya Disease . . . . 855 intracranial internal carotid arteries and their
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 857 proximal branches. The disorder is typically
characterized by a reduction in blood flow in
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 858
the major vessels of the anterior circulation of
the brain. This results in the development of a
compensatory collateral vasculature around
the stenosed vessels, near the apex of the inter-
nal carotid, on the cortical surface,
leptomeninges, and branches of the external
carotid artery and the base of the skull. In
advanced cases, the posterior circulation is
also involved, including the basilar and poste-
rior cerebral arteries. While it was first
described as a “hypoplasia of the bilateral
internal carotid arteries,” the characteristic
appearance of the network of abnormally
dilated collateral vessels on conventional angi-
ography was compared to “a puff of cigarette
smoke” (Arch Neurol 20(3):288–899, 1969) or
“moyamoya” in Japanese. The designation
“spontaneous occlusion of the circle of Willis”
was recently suggested as an alternative by the
International Classification of Diseases (ICD).
Moyamoya disease patients are predominantly
found in Eastern Asian countries such as Japan
and the Republic of Korea. Moyamoya disease
patients show bimodal distribution, adult type
and pediatric type. Symptom of adult
moyamoya patients can be either ischemic or
hemorrhagic; nevertheless, pediatric patients
usually present with ischemia.
In ischemic moyamoya disease, the thera-
peutic effect of surgical revascularization
(extracranial-intracranial bypass) has been
well established. Bypass surgery can improve
the impaired cerebral hemodynamic state and
decrease the recurrent ischemic events. Man-
agement of the hemorrhagic moyamoya dis-
ease, on the other hand, presents a serious
challenge. Despite the extremely high rate of
rebleeding attacks, no therapeutic method has
been established until recently. In 2013, the
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 819

Japan Adult Moyamoya (JAM) Trial, a multi- this chapter, state-of-the-art description, symp-
centered prospective randomized controlled tom, epidemiology, diagnosis, and treatment of
trial, was completed. It has been revealed that moyamoya disease based on this latest guideline
bypass surgery significantly decreases the rate will be discussed (Fig. 1).
of rebleeding attacks and improve the patients’
prognosis during the following 5 years. This
epoch-making study is expected to establish a Epidemiological Features
guiding principle for the treatment of hemor-
rhagic moyamoya disease. Geographical Distribution

East Asian countries feature the highest incidence


Keywords of moyamoya disease, more particularly Japan
Moyamoya disease • MRI • MRA • SWI • and the Republic of Korea. A study conducted in
Perfusion • ASL • Direct bypass • STA-MCA Japan reported the total annual number of
anastomosis • Indirect bypass • SPECT • PET • moyamoya disease patients at 3,900, with preva-
Intracranial hemorrhage • JAM trial • Preg- lence and annual incidence rates of 3.16 and 0.35
nancy • Delivery per 100,000, respectively, in 1994. The female-to-
male ratio was 1.8 and 10 % of the patients had a
family history of the disease. The distribution of
Introduction the age at onset has two peaks: a dominant peak at
5 years of age and a more modest peak around the
Moyamoya disease represents a unique cerebro- late 20s to 30 years of age [4]. A recent review of
vascular disorder characterized by steno- regional differences shows that incidences per
occlusive change at the terminal portion of the 100,000 patient-years ranged in Japan from 0.35
internal carotid artery (ICA) and a fine vascular to 0.94 (95 % CI 0.69–1.19) and in the USA from
network, the so-called “moyamoya” vessels. This 0.05 ( 0.04–0.12) in Iowa to 0.17 ( 0.06–0.40)
fine vascular network forms a collateral pathway in Hawaii and were 0.41 (0.28–0.54) in Nanjing,
and compensates for the reduced cerebral blood China, and 0.02 (0.003–0.04) in Taiwan. The
flow due to the steno-occlusion occurring in the female-to-male ratio ranged from 1.1 (0.9–1.5)
major arteries. The disease shows a differential in Nanjing to 2.8 (1.2–6.1) in Iowa. Proportions
incidence and predominantly affects Eastern with cerebral hemorrhage as the initial presenta-
Asia. Its clinical features include ischemia and tion were 56 % in China, 52 % in Taiwan, 29 % in
hemorrhage, epilepsy, headache, etc. While Hawaii, 21 % in Japan, and 10 % in Iowa. Patients
young patients usually present with ischemia, with childhood onset presented most often with
adult patients present either ischemia or hemor- ischemia (more than 75 %) in all regions [5].
rhage. Evidence suggests that revascularization
surgery is highly efficient at preventing an ische-
mic event; in addition, the latest report [2] sug- Gender and Age Distribution
gests its effect on prevention of hemorrhagic
events. Due to a familial occurrence of 10–15 %, MMD has two peak ages of onset, initially at
intense investigations were made towards the 5 years of age (pediatric or juvenile type) and
identification of gene candidates, which led to subsequently at 30–50 years of age (adult type).
the identification of several genetic loci associated Peak age in male is 10–14 years and 35–49 years,
with the disease. Nevertheless, its pathogenesis whereas peak in female is 20–24 years and 50–54
remains largely unknown. years [6]. In 2003, the total number of patients
Recently, for the diagnosis and treatment of treated in Japan was estimated at 7,700. The
moyamoya disease (spontaneous occlusion of female-to-male ratio was 1.8, and a family history
the circle of Willis) have been updated [3]. In was found in 12.1 % of patients. The prevalence
820 A. Yamamoto et al.

Fig. 1 Conventional digital subtraction angiography of view (d–f) show steno-occlusive change of the terminal
the left internal carotid arteries of a pediatric moyamoya portion of the left ICA and moyamoya vessels in the
disease patient. Anterior-posterior view (a–c) and lateral perforator artery of the basal ganglia

rate was calculated at 6.03 per 100,000, and the surveys. The female-to-male ratio was 2.18 and
annual rate of newly diagnosed cases was 0.54 per the age of onset occurred in two peaks with a
100,000. In 2009, an analysis of the regional different trend: the highest peak arose between
all-inclusive epidemiological data obtained in 45 and 49 years, and the second occurred between
Hokkaido, a major island of Japan counting a 5 and 9 years. A familial history was observed in
population of 5.63 million [7], reported a preva- 15.4 % of patients. These epidemiological fea-
lence of 10.5 patients per 100,000 and an annual tures differ significantly from the data obtained
incidence of 0.94 per 100,000, both of which in previous studies. However, the higher detection
greatly exceeded the results of the previous and prevalence reported may not reflect an actual
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 821

increase in the incidence of moyamoya disease, While parents presented the onset of symptoms at
but may rather be due to the increased availability the age of 22–36 (mean 30.7 [7.5] years), their
of noninvasive diagnostic tools, such as MRI/MR offspring presented symptoms much earlier,
angiography. within 5–11 years of age (mean 7.2 [2.7] years).
Moyamoya disease patients’ age distribution This phenomenon is called clinical anticipation.
shows characteristic bimodal pattern, pediatric Clinical anticipation was reported in triplet repeat
and adult patients. The precise mechanism for disease; however, triplet repeat has not been
this bimodal distribution has not been determined. reported in moyamoya disease patients. This indi-
In the pediatric patients, the rapidly progressing cates a strong genetic association with a female
maturation of the brain may precipitate deficiency predominance in cases of familial moyamoya
of a balance with cerebral blood flow maturation, disease [9].
and as a consequence, patients progress to
develop ischemic attack. However, still many
things are unknown about pediatric normal devel- Pathogenesis of Moyamoya Disease
opment of cerebral blood flow. This lack of bal-
ance between maturation of the brain and Cytokines
development of cerebral blood flow might not be
able to explain the full mechanism of the devel- Most studies about cytokines in moyamoya dis-
opment of ischemic attack in pediatric moyamoya ease patients have been performed by the exami-
disease patients. In adult moyamoya disease nation of cerebrospinal fluid (CSF) sampled from
patients, hemorrhagic symptom can occur more the subarachnoid space during surgical therapy.
commonly than in pediatric patients. In adult Angiogenetic cytokines such as basic fibroblast
moyamoya patients, prevalence of ischemic and growth factor (b-FGF), hepatocyte growth factor,
hemorrhagic symptom is almost the same. and transforming growth factor (TGF) showed
Another unsolved issue of moyamoya disease increased concentrations in the CSF as well as in
is why the number of patients between these pedi- other surgical specimens such as the arterial wall
atric and adult moyamoya patients is small. About specimens and the meningeal specimens. Among
this issue, ischemic symptom which commonly these cytokines, angiogenetic cytokine b-FGF has
occurs in pediatric period may become unlikely been thought to be related with several findings in
to occur because a balance between brain matura- moyamoya disease: steno-occlusive lesion in the
tion and cerebral blood flow will be achieved. circle of Willis, the development of moyamoya
After getting over this dangerous pediatric period, vessels and dilatation of the cortical small arteri-
moyamoya disease patient may become symp- oles, and angiogenesis after surgical indirect
tomatic due to a decrease of cerebral blood flow synangiosis therapy. Reportedly, b-FGF induces
by arteriosclerosis in the adult period. the proliferation of vascular endothelial cells,
which may precipitate the stenosis of the major
arteries. On the contrary, b-FGF has also an
Familial Moyamoya Disease angiogenetic and dilatation effect on the small
arteries, which may explain the development of
An epidemiological feature of moyamoya disease moyamoya vessels and the dilated pial arterioles
is the high incidence of familial occurrence, which on the cortex in moyamoya disease [10].
accounts for about 15 % of patients [8]. In patients The role of cytokine abnormality in moyamoya
with familial incidence, the ratio of women to men disease has not yet been totally determined. Ele-
was reported to equal 5.0, as opposed to only 1.6 vation of cytokines in the CSF may simply reflect
in sporadic cases; the mean (SD) age of onset was the response of cytokines to hypo-oxygenation
11.8 (11.7) years in familial cases vs. 30.0 (20.9) status in moyamoya disease. In this scheme, ele-
years in sporadic cases. Out of eight parent- vated concentrations of cytokines may simply be a
offspring pairs, all were paired with the mother. consequence of hypo-oxygenation but not a direct
822 A. Yamamoto et al.

pathogenesis of moyamoya disease. Validation Asymptomatic Moyamoya Disease


study for the elevation mechanism of cytokine
levels will be necessary [10]. In recent years, asymptomatic cases of moyamoya
disease and moyamoya disease manifesting with
nonspecific symptoms only, such as headache,
Genetic Factors have been reported. “Asymptomatic” patients
with moyamoya disease have been defined as
In cases of familial moyamoya disease, the gene those who have experienced neither ischemic
loci 3p24-p26 and 8q23 have been identified nor hemorrhagic episodes since birth. According
using genome-wide analysis, while loci 6q25 to previous reports, clues to the diagnosis of
and 17q25 were determined using chromosomal asymptomatic moyamoya disease included
search. nonspecific symptoms such as tension-type head-
Cases of unilateral moyamoya disease ache, dizziness, head trauma, etc. Some patients
progressing to bilateral moyamoya disease have were incidentally diagnosed using MRI/MRA
been well known, as well as progression of major which was performed for brain checkup or scru-
artery stenosis on the contralateral side to the tiny for the disease because of family history of
initial disease. It has thus been suspected that moyamoya disease [15]. The increase in the
major artery stenosis, unilateral moyamoya dis- number of asymptomatic patients could be attrib-
ease, and bilateral moyamoya disease were among uted, at least partially, to the current increasing
a spectrum of relational phenomena, based on availability of MRI/MRA examination in
genetic susceptibility. In addition, familial Japan. In an all-inclusive survey of moyamoya
moyamoya disease has been known to be an auto- disease in Hokkaido, the authors reported that
somal dominantly inherited disease with incom- asymptomatic patients comprised 17.8 % of the
plete penetrance, and various stages of the disease 267 newly registered patients with moyamoya
are thus observed in familial moyamoya disease disease between 2002 and 2006 [7]. From this
patients. From these points, a combination of observation, the prevalence of asymptomatic
internal genetic factors and external environmen- moyamoya disease may be much higher than pre-
tal factors has been believed to be relevant for viously thought [16].
disease occurrence or progression. These mecha- The reported imaging finding in asymptomatic
nisms of moyamoya disease have been reported to moyamoya disease patients has been limited in
be relevant with three heterogeneities, disease number. Kuroda et al. reported cerebral infarction
heterogeneity, genetic heterogeneity, and locus in 16 out of 77 (20.8 %) involved hemispheres in
heterogeneity [11]. asymptomatic moyamoya disease patients
Recently, a genome-wide association study [15]. About 40 % of the involved hemispheres
identified ring finger protein (RNF)213 (http:// had a moderate or severe reduction of cerebral
omim.org/entry/613768) as the first moyamoya perfusion reserve in asymptomatic moyamoya
disease gene [12]. Also, another genome-wide disease [15]. The annual risk for ischemic or hem-
linkage analysis by assuming the inheritance pat- orrhagic stroke was estimated at 3.2 %. Disease
tern of moyamoya disease as autosomal dominant progression was associated with ischemic events
mode with incomplete penetrance and whole or silent infarction in four of five patients. Also,
genome-exome analysis provided evidence asymptomatic abnormal findings were identified
suggesting the involvement of RNF213 in genetic in another three patients [15].
susceptibility to moyamoya disease [13]. Further Asymptomatic moyamoya disease may thus be
studies are ongoing to clarify the biochemical closely related to the development of asymptom-
function and pathological role of RNF213 in atic cerebral infarction. This observation may be
moyamoya disease [14]. consistent with the findings that steno-occlusive
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 823

Fig. 2 A case of asymptomatic adult moyamoya patient. (b) and T2-WI image (c) shows both stenosis of ICA.
Conventional digital subtraction angiography lateral view Previous T2-WI image (d) shows apparent flow void of
image shows stenosis of ICA and moyamoya vessels at the bilateral MCA compared to (c). MIP image of TOF-MRA
frontal area and basal area. (a) Source image of TOF-MRA shows bilateral ICA stenosis apparently (e)

arterial change in adult moyamoya disease moyamoya disease deteriorates from an asymp-
patients may progress in both the anterior and tomatic to symptomatic. It might be feasible to
posterior circulation, in both bilateral and unilat- monitor asymptomatic moyamoya disease
eral disease types, and in both symptomatic and patients by less invasive imaging methods such
asymptomatic patients [17]. The natural course of as MRI/MRA for disease progression and ische-
asymptomatic moyamoya disease is still not fully mic and/or hemorrhagic stroke findings [16, 18].
understood [3]. Therefore, it is important to know Recently, in Japan, asymptomatic moyamoya
that moyamoya disease progression may occur registry project (AMORE) has been started.
asymptomatically and can suddenly cause ische- By achievements of this study, progression mech-
mic or hemorrhagic stroke even in asymptomatic anism and the natural course of asymptomatic
patients. It is currently unknown how and when moyamoya disease might be elucidated (Fig. 2).
824 A. Yamamoto et al.

Unilateral Moyamoya Disease concurrent unilateral moyamoya disease in addi-


tion to 43 patients with definitive moyamoya dis-
Based on the current diagnostic guideline [3], ease and suggested the possibility that the disease
unilateral moyamoya disease in adult patient is was inherited by the same autosomal dominant
diagnosed as probable moyamoya disease and inheritance pattern. From this observation, unilat-
refers to the presence of unilateral steno-occlusive eral moyamoya disease with a family history can
change of the terminal portion of the internal also be considered as a subtype of moyamoya
carotid arteries accompanied by the formation of disease. In addition, unilateral moyamoya disease
moyamoya vessels around that region. These uni- without a positive family history should be distin-
lateral changes may occur concurrently with other guished from definitive bilateral moyamoya
underlying diseases, such as hyperthyroidism, disease [20].
intracranial arteriovenous malformation, The symptoms of unilateral moyamoya disease
Down’s syndrome, Apert syndrome, von are the same as those of definitive bilateral
Recklinghausen’s disease (neurofibromatosis moyamoya disease. Ischemic symptoms, hemor-
type 1), postirradiation therapy of the brain, sys- rhagic symptoms, concurrent cerebral aneurysm,
temic lupus erythematosus, and Sjögren’s syn- involuntary movement, and headache may be
drome; in these situations, the patient’s condition noted.
is classified as quasi-moyamoya disease and not The reported frequency of progression from
as unilateral moyamoya disease [3]. In children, unilateral to bilateral moyamoya disease varies
unilateral moyamoya disease associated with ste- from 10 % to 39 % [21]. Each study cohort con-
nosis of the terminal portion of the internal carotid sists of a limited number of patients; therefore,
arteries should be considered as definitive progression rate from unilateral to bilateral dis-
moyamoya disease and not as probable ease may be influenced by the number of patients.
moyamoya disease [19]. Progression to bilateral moyamoya disease may
The frequency of unilateral moyamoya disease not only affect pediatric patients but also adult
was reported as 10.6 % within 2,635 patients with patients. The statistically significant risk factors
moyamoya disease, including initially diagnosed for progression to bilateral disease have been
and re-diagnosed patients [3]. A family history is reported to be the presence of equivocal or mild
occasionally present in patients with unilateral stenotic changes in the terminal portion of the
moyamoya disease. An analysis of 15 families ICA, the middle cerebral artery (MCA), or the
having a family history of moyamoya disease in anterior cerebral artery (ACA) of the contralateral
3 or more generations revealed 5 patients with side [22] (Fig. 3).

Fig. 3 A case of unilateral moyamoya disease patient vessels are not prominent in the cistern both on T2-WI
shows right ICA stenosis on MRA. MIP image of image and TOF-MRA source image (b, c)
TOF-MRA shows right ICA stenosis (a). Moyamoya
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 825

Quasi-moyamoya Disease of 7,941 moyamoya disease patients and 0.34


patients per 100,000 populations. Annual inci-
Quasi-moyamoya disease refers to the presence of dence of quasi-moyamoya disease is 0.11 patients
steno-occlusive change of the terminal portion of per 100,000 populations.
the ICA accompanied by an abnormal vascular Symptom of quasi-moyamoya disease may be
network in association with an underlying dis- epilepsy, headache, or asymptomatic. Concurrent
ease. Even in cases with unilateral lesions, if an presence of symptoms associated with mental
underlying disease is present, the condition can be retardation due to underlying disease and symp-
considered as quasi-moyamoya disease. Unilat- toms associated with cerebrovascular disorder
eral moyamoya disease in adult patient without may cause a relatively complicated clinical con-
underlying disease should be considered as prob- dition for the precise diagnosis. Cerebral conven-
able moyamoya disease and should be differenti- tional digital subtraction angiography can show
ated from quasi-moyamoya disease. Quasi- various findings from those very similar to defin-
moyamoya disease has been reported to affect itive moyamoya disease to be rather different,
people of all races. Concurrent occurrence with such as atherosclerotic lesions. Pathological find-
underlying congenital disease is more frequent in ings also vary according to the underlying disease.
children; however, concurrent occurrence with Treatment of quasi-moyamoya disease is like that
acquired underlying disease is more frequent in of definitive moyamoya disease. The effect of
adults [3]. revascularization on the prevention of rebleeding
The following illnesses have been reported as in patients with quasi-moyamoya disease has not
underlying diseases: atherosclerosis, autoimmune yet been clarified. In quasi-moyamoya disease,
disease (systemic lupus erythematosus, unilateral disease may progress to bilateral dis-
antiphospholipid antibody syndrome, periarteritis ease. The nature of the underlying diseases influ-
nodosa, and Sjogren’s syndrome), meningitis, von ences the prognosis of patients with quasi-
Recklinghausen’s disease (neurofibromatosis type I), moyamoya disease (Fig. 4).
brain tumors, Down’s syndrome, head injury, irra-
diation, hyperthyroidism, stenocephaly, Turner’s
syndrome, Alagille syndrome, Williams syn- Systemic Vessel Involvement
drome, Noonan’s syndrome, Marfan syndrome, of Moyamoya Disease
tuberous sclerosis, Hirschsprung’s disease, glyco-
gen storage disease type I, Prader-Willi syndrome, In addition to intracranial arterial steno-occlusive
Wilms tumor, primary oxalosis, sickle cell dis- lesions associated with moyamoya disease, steno-
ease, Fanconi’s anemia, spherocytosis, eosino- occlusive lesions of extracranial arteries including
philic granuloma, type II plasminogen renal, coronary, pulmonary, mesenteric, and
deficiency, leptospirosis, pyruvate kinase defi- peripheral arteries have also been reported
ciency, protein S deficiency, protein C deficiency, [23]. Among these extracranial arteries,
fibromuscular hyperplasia, osteogenesis moyamoya disease most commonly affects the
imperfecta, polycystic kidney, oral contracep- renal artery. The prevalence of renal artery
tives, and poisoning (cocaine poisoning) [3]. involvement in moyamoya disease patients ranges
Epidemiology of quasi-moyamoya disease was from 5 % to 8 %. In most cases of moyamoya
reported in the annual report of the Research disease, in which the renal artery was involved,
Committee on Spontaneous Occlusion of the Cir- renal artery stenosis mainly involves the proximal
cle of Willis (Moyamoya Disease) of the Ministry one-third of the main branch. Severe stenosis case
of Health, Labour and Welfare, Japan 2011 (http:// can precipitate renovascular hypertension.
mhlw-grants.niph.go.jp/niph/search/NIDD02.do? Although the definitive cause and pathogenesis
resrchNum=201128175A). Prevalence of quasi- of moyamoya disease remain unclear, pathologic
moyamoya disease is reported as 5.4 % in a total studies of the steno-occlusive lesions of the
826 A. Yamamoto et al.

Fig. 4 Images of a quasi-moyamoya disease (neurofibro- flow voids in the cistern. (b, c) Conventional digital sub-
matosis type I, von Recklinghausen disease) patient are traction angiography images (d, e) show steno-occlusive
shown. MIP image of TOF-MRA (a) shows steno- change and moyamoya vessels more apparently. On T2-WI
occlusive change of bilateral ICA. Source image of image, right optic nerve mass lesion and high-signal area in
TOF-MRA shows cisternal moyamoya vessels as high- the right cerebellar peduncle are apparent; those are char-
signal spots, which are also apparent on T2-WI image acteristic lesion of neurofibromatosis type I (f)
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 827

cerebral vessels typically reveal fibrous thicken- disease develop transient ischemic attack (TIA) or
ing of the intima with a small amount of lipid cerebral infarction, whereas about half of adult
deposition. Inflammatory cell infiltration is not patients develop intracranial bleeding, and half
noted in the vascular walls, and the internal elastic develop TIA or cerebral infarction or both
lamina is well preserved. Similar histological [28]. The symptoms and clinical course may
findings of intimal fibrous thickening in the extra- change according to the age and the disease type
cranial vessels have been reported in autopsy manifested at the initial attack, and a wide range of
cases of moyamoya disease [23]. severity of symptoms have been noted, such as
TIAs and hemorrhagic stroke which resulted in
permanent neurological deficits.
Pathological Features According to the recent increase in the avail-
ability of MRI/MRA, increasing numbers of
Pathological evaluation of involved vessels in patients have been reported, who were inciden-
moyamoya disease has shown that the outer diam- tally diagnosed as having moyamoya disease dur-
eters of the terminal portions of the relevant ICA ing the asymptomatic stage [15] or with only
are markedly diminished. In addition to outer minor complaint such as headache [3]. In pediatric
diameter change, fibrocellular thickening of the population, acute infantile hemiplegia is one of
intima, an irregular undulation (waving) of the the most important symptoms of moyamoya dis-
internal elastic lamina, and attenuation of the ease. Benign familial nocturnal alternating hemi-
media have been reported in a histological finding plegia of childhood has been also reported to be a
in the vascular wall of the ICA [24, 25]. Data from possible symptom of moyamoya disease.
recent studies have suggested that caspase-3- The frequency of each initial symptom in the
dependent apoptosis might be associated with 1,127 definitive moyamoya disease patients reg-
these histological changes in the vascular wall of istered until 2000 is presented in Table 1 for the
moyamoya disease patients [26]. patients with the hemorrhagic type and ischemic
Moyamoya vessels consist of dilated perforat- type (infarction type, TIA type, and frequent TIA
ing arteries, which can show a wide range of type) of initial attack. For both types, muscle
histological findings, including fibrin deposits in weakness, consciousness disturbance, headache,
the wall, fragmented elastic lamina, attenuated speech disorder, and sensory disturbance were the
media, and the formation of microaneurysms. In most frequent; however, the incidence of con-
the early stage of the disease (stage I according to sciousness disturbance and headache was higher.
Suzuki’s angiographic classification), moyamoya Incidence of muscle weakness was lower for
vessels are rarely observed. Steno-occlusive patients with the hemorrhagic-type than for the
change of the arterial lumen and subsequent ischemic-type initial attacks ( p < 0.01) [3].
thrombosis can also be seen in the moyamoya
vessels [27]. Therefore, these histological changes
might have close association with the onset or Ischemic Symptoms
progression of ischemic or hemorrhagic stroke in
moyamoya disease. Moyamoya disease patients usually show cerebral
ischemic change in the anterior territory of the
ICA, particularly in the frontal lobe. Therefore,
Clinical Features most patients with symptomatic moyamoya dis-
ease may present with focal neurological deficits,
An Overview such as dysarthria, aphasia, or hemiparesis. How-
ever, moyamoya disease patients might also occur
The clinical features of moyamoya disease show with other relatively atypical symptoms such as
substantial difference between pediatric and adult consciousness disturbance, sensory disturbance,
patients. Most pediatric patients with moyamoya visual symptoms, or involuntary movements,
828 A. Yamamoto et al.

Table 1 Initial symptom of moyamoya disease (n = ventilation. Pediatric moyamoya disease patients
1,127) sometimes develop TIA when slow waves appear
Hemorrhagic Ischemic again during EEG scans. This reappearance of
Initial symptom type (%) type (%) slow waves is pathognomonic for pediatric
Muscle weakness 58.6 79.8a moyamoya disease patients and is known as
Consciousness 70.4a 14.1 rebuild-up phenomenon [32]. Recently, this
disturbance
rebuild-up phenomenon has been reported to be
Headache 64.6a 18.8
Seizure 8.5 8.0
induced by post-hyperventilation hypoxia com-
Psychiatric 8.7 2.5 bined with a hyperventilation-induced reduction
symptom in cerebral blood flow and originates in the deep
Speech disorder 24.5 20.1 cortical sulci, where the cerebral perfusion reserve
Sensory 18.4 19.3 is diminished [33]. Rebuild-up phenomenon can
disturbance completely disappear after effective surgical
Involuntary 3.3 3.0 revascularization [33] (Fig. 5).
movement
Intellectual 5.3 6.2
disturbance
Visual impairment 2.0 3.2 Hemorrhage
Visual field defect 3.9 5.0
Reprinted with permission from Guidelines for Diagnosis About a half of adult moyamoya disease patients
and Treatment of Moyamoya Disease (Spontaneous Occlu- develop intracranial hemorrhage, though hemor-
sion of the Circle of Willis). #The Japan Neurosurgical rhagic symptom is rare in pediatric moyamoya
Society [3] patients [24]. Also, hemorrhagic symptom has
a
Significantly more frequent as compared with the others
( p < 0.05) been known to be more common in female
patients than male patients. Hemorrhagic symp-
tom patients show more severe symptom and
particularly in children [29, 30]. Some pediatric deteriorated outcome than ischemic symptom
patients develop intellectual impairment owing to patients.
frontal lobe ischemia, infarction, or both [31]. In Two main and one additional causes of intra-
adult moyamoya disease patients, cognitive dys- cranial hemorrhage in moyamoya disease have
function, such as short-term memory disturbance, been assumed: (1) rupture of dilated, fragile
irritability, or agitation, has been reported. moyamoya vessels or (2) rupture of saccular aneu-
Patients who present with these atypical symp- rysms in the circle of Willis especially in the
toms may be misdiagnosed with psychiatric dis- posterior part. For the first cause, the rupture
orders, such as schizophrenia, depression, or may be due to persistent hemodynamic stress on
personality disorder. the moyamoya vessels and occurs mainly in the
In pediatric patients, ischemic attacks are typ- basal ganglia, thalamus, or periventricular region,
ically induced by hyperventilation, for example, which is common with basal moyamoya vessels.
when crying or playing with a pinwheel. Electro- Intraventricular hemorrhage is more frequent in
encephalogram (EEG) findings in pediatric moyamoya disease patients than other origin hem-
moyamoya patients are characteristic. In healthy orrhagic patients [34]. Peripheral aneurysms in
children, high-amplitude slow waves are induced the collateral vessels or moyamoya vessels might
during hyperventilation, which will disappear be identified on conventional digital subtraction
after hyperventilation stops (an event known as cerebral angiography [35].
build-up phenomenon). However, in pediatric For the second cause, rupture of saccular aneu-
moyamoya disease patients, the slow waves rysms located around the circle of Willis occurs
appear again after a recovery of normal most commonly at the basilar artery bifurcation or
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 829

Fig. 5 3-T MR images of a pediatric moyamoya patient in the bilateral basal ganglia and thalamus. FLAIR image
are shown. On T2-WI image (a), moyamoya vessels are (c) shows tiny high-signal area in the bilateral occipital
apparent in the bilateral basal ganglia. On the source image lobe (FLAIR ivy sign) and chronic infarction
of time-of-flight MRA (b), moyamoya vessels are apparent

at the junction of the basilar artery and the supe- brain cortical surface, although this is rare [37]
rior cerebellar artery. The vertebrobasilar system (Figs. 6 and 7).
has an important role in providing collateral cir-
culation for steno-occlusive change of bilateral
internal carotid arteries in moyamoya disease Headache
patients. Consequently, hemodynamic stress
probably may precipitate the formation of a Recently, headache is also considered to be one of
saccular aneurysm predominantly in the posterior the common clinical presentations of pediatric
part of the circle of Willis or vertebrobasilar moyamoya disease patients. In many clinical
system. Rupture of a saccular aneurysm in the cases, pediatric patients complaining headache
vertebrobasilar system can cause subarachnoid may not be considered as having moyamoya dis-
hemorrhage [36]. Therefore, in the imaging ease, and other origins of headache such as men-
diagnosis of moyamoya disease patients, ingitis or brain tumor will be looked for. Headache
scrutiny for aneurysm especially in the can be seen in about 20–30 % of moyamoya
vertebrobasilar system is important. 3-T disease patients. Specific type of headache for
MRI/MRA examination has superior signal-to- moyamoya disease patients has not been reported;
noise ratio, higher spatial resolution, and less however, patients can show migraine in the more
invasiveness and, therefore, is suitable for clinical severely affected side. Also, patients with alter-
routine follow-up examination for moyamoya dis- nating hemiplegia of childhood showing migraine
ease patients [18]. and hemiplegia have been reported
There is increasing reports that adult [38]. Moyamoya disease patients may complain
moyamoya disease patients might present sub- of headache before and after revascularization
arachnoid hemorrhage over the cerebral cortex surgery. Preoperative headache in moyamoya dis-
despite the absence of an intracranial aneurysm ease patients was considered to be related to
[37]. A third cause of intracranial hemorrhage in hypoperfusion or hypo-oxygenation because it
adult moyamoya disease patients is rupture of the has been reported that headaches disappeared
dilated collateral pial superficial arteries on the after revascularization surgery [39].
830 A. Yamamoto et al.

Fig. 6 In a case of patient presenting with headache, stenosis of the left ICA and MCA (d). Arterial spin labeling
unenhanced CT scan image shows subtle high-density perfusion-weighted image shows decreased flow in the left
area in the left frontal lobe surface with obliteration of the hemisphere. (e) Diagnosis of probable moyamoya disease
left frontal lobe sulci. (a) FLAIR (b) and SWI (c) images was confirmed by a conventional digital subtraction angi-
show superficial high signal and low signal in the left ography (f, g)
frontal lobe, respectively. TOF-MRA MIP image shows
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 831

Fig. 7 Difference of appearance of hemorrhage among comes from effect of 180 refocus pulse used in T2-WI
different MRI sequences. T2-WI image (a), T2-star- image. In addition, MiniP image of SWI shows hemor-
weighted image, and MiniP image of SWI of hemorrhage rhage as more pronounced low-signal area compared to
in the right basal ganglia are shown. T2-star-weighted T2-star-weighted image. This difference comes from the
image shows (b) hemorrhage as more pronounced (c) fact that SWI utilizes phase information to enhance the
low-signal area compared to T2-WI image. This difference local susceptibility difference

Involuntary Movement hypothesis is supported by the fact that revascu-


larization surgery is associated with improvement
Involuntary movements are relatively rare symp- of the involuntary movements or at least a signif-
toms in moyamoya disease patients, and their icant decrease in their frequency [40].
frequency is estimated about 5 % [40]. Association
of various involuntary movement disorders with
moyamoya disease has been reported in the liter- Diagnostic Evaluation
ature. The spectrum of these involuntary move-
ments includes chorea, choreoathetosis, Current Diagnostic Criteria
dyskinesia, dystonia, limb shaking, and epilepsia of Moyamoya Disease
partialis continua. Some of them may occur in a
paroxysmal manner, while others can be triggered 1. Cerebral angiography is considered essential
by the initiation of some movement for the diagnosis and must show at least the
(“kinesigenic” manner) [41], by some particular following findings [3, 42]:
form of exercise, or by various types of hyperven- i. Stenosis or occlusion of the terminal por-
tilation maneuvers. The involuntary movements tion of the intracranial ICA or proximal
are usually transient, ranging in duration from portions of the anterior and/or the MCA
several seconds to a few months, and rarely ii. Abnormal vascular networks in the vicinity
constant [40]. of the occlusive or stenotic lesions in the
The main mechanism of onset of these invol- arterial phase
untary movements is thought to be cerebral iii. Bilaterality of findings (i) and (ii)
hypoperfusion induced by steno-occlusive change 2. However, when magnetic resonance imaging
of the internal carotid arteries in the circle of (MRI) and magnetic resonance angiography
Willis. Limb shaking is typically triggered by (MRA) findings meet all of the following
diffuse hemispheric hypoperfusion caused by criteria, cerebral angiography can be omitted:
severe stenosis of the ICA. Any activities induc- i. MRA shows stenosis or occlusion of the
ing hyperventilation, such as singing and crying, terminal portion of the intracranial ICA or
may precipitate the involuntary movement, proximal portions of the anterior and/or
because of induced cerebral hypoperfusion. This the MCA.
832 A. Yamamoto et al.

ii. MRA shows abnormal vascular networks Definitive Moyamoya Disease


in the basal ganglia. and Probable Moyamoya Disease
Note: When two or more visible flow
voids are present in the basal ganglia on Moyamoya disease should be classified as defin-
MRI, at least unilaterally, they can be itive or probable based on the abovementioned
deemed as representing an abnormal vas- items (1) to (4) [3]. For definitive moyamoya
cular network. disease, all criteria listed in (1) or (2) and in
iii. Bilaterality of findings (i) and (ii). (3) should be fulfilled. In pediatric patients, how-
3. Moyamoya disease is an illness of unknown ever, the criteria in item (1) or (2) (i) and (ii), on
etiology. The differential diagnosis of this one side, and visible stenosis around the terminal
disease includes similar cerebrovascular portion of the internal carotid arteries, on the other
lesions associated with the following underly- side, are sufficient for a definitive moyamoya
ing diseases, which should, therefore, be disease diagnosis. For probable moyamoya dis-
excluded: (i) atherosclerosis, (ii) autoimmune ease, all criteria are fulfilled except item (1) (iii)
disease, (iii) meningitis, (iv) brain tumors, and/or item (2) (iii) among the criteria of (1) or
(v) Down’s syndrome, (vi) von (2) and (3).
Recklinghausen’s disease, (vii) head injury,
(viii) cerebrovascular lesions after head irradi-
ation, and (ix) others. CT and CT Angiography in Moyamoya
4. Pathological findings that can be used as refer- Disease
ences for the diagnosis:
i. Thickening of the arterial intima, mainly CT scan is one of the primary survey modalities
in the terminal portion of the internal carotid for patients suspected of stroke; however, any
arteries, and narrowing or blockage of the definitive diagnostic information of moyamoya
lumen caused by this change, usually bilat- disease may not be obtained by this modality. In
eral. Occasionally, lipid deposits are also advanced stages of moyamoya disease, chronic
present in the thickened intima. cerebral infarction and unusual cerebral atrophy
ii. Arteries such as the anterior, middle, and can be seen on CT scan; however, definitive diag-
posterior cerebral arteries forming the cir- nosis cannot be done [43]. On unenhanced CT
cle of Willis occasionally show varying scan, the steno-occlusive change and develop-
degrees of stenosis or occlusion associated ment of moyamoya vessels are not apparent. For
with fibrocellular thickening of the intima, the assessment of cerebral vessels, contrast-
waviness of the internal elastic lamina, and enhanced CT scan and CT angiography is neces-
thinning of the media. sary, and they can precisely show the steno-
iii. Numerous small vascular channels (perfo- occlusive change and development of moyamoya
rating and anastomotic branches) can be vessels. During the diagnosis process of
seen around the circle of Willis. moyamoya disease, findings of unenhanced CT
iv. Pia mater may also show reticular con- scan of stroke is not enough to exclude moyamoya
glomerates of small vessels (Fig. 8). disease, and further examination such as
MRI/MRA or conventional angiography is
Reprinted with permission from Guidelines necessary [43].
for Diagnosis and Treatment of Moyamoya Unenhanced CT scan is a reliable imaging
Disease (Spontaneous Occlusion of the modality when the diagnosis of an ischemic or
Circle of Willis). #The Japan Neurosurgical hemorrhagic stroke is suspected. In adult
Society [3]. moyamoya disease patients, hemorrhage is
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 833

Fig. 8 In a pediatric moyamoya disease patient, right vertebral artery digital subtraction angiography images are shown.
Anterior-posterior view (a–c) and lateral view (d–f) show collateral moyamoya vessels from vertebrobasilar systems
834 A. Yamamoto et al.

common such as intraventricular hemorrhage, (without ionizing radiation exposure) compared


cerebral hemorrhage, and subarachnoid hemor- to conventional angiography.
rhage due to rupture of moyamoya vessels or Cerebral angiography is essential for a defini-
aneurysms. In any cases of supratentorial cerebral tive diagnosis of moyamoya disease [3]. MRI/
hemorrhage seen on unenhanced CT scan, hemor- MRA can also be used for a definitive diagnosis
rhage due to moyamoya disease must be consid- when the following findings are fulfilled on
ered as a differential diagnosis, and further TOF-MRA imaging conducted using a scanner
examination such as contrast-enhanced CTA is with a static magnetic field strength of more than
recommended [43]. 1.5-T (especially 3-T MR scanner is preferable):
The recent advancement of multi-detector row
computed tomography (MDCT) scanners has 1. On MRA, stenosis or occlusion of the terminal
enabled high-resolution three-dimensional recon- portion of the intracranial ICA or proximal
struction. CTA and MRA were compared in terms portion of the anterior and/or middle cerebral
of the steno-occlusive changes exhibited in each arteries
vessel. CTA and MRA scores were assigned on 2. On MRA, abnormal vascular networks in the
the basis of the severity of occlusive changes in basal ganglia
the ICA, MCA, ACA, and posterior cerebral Note: When two or more visible flow voids in
artery (PCA). CTA scores were significantly cor- the basal ganglia are present at least unilaterally
related with MRA scores, and their scores were in on MRI, they can be deemed as representing an
complete agreement in 39.6 %. The mean CTA abnormal vascular network.
score was significantly lower than the mean MRA 3. Bilaterality of findings (1) and (2)
score. CTA using MDCT is having a higher spatial
resolution, therefore, can be a more reliable Moyamoya disease stage classification has
method than MRA for diagnosing moyamoya dis- also been reported based on the MR findings
ease, particularly in emergency cases [44] (Figs. 9 (Table 2). In this classification system, the
and 10). stage is determined by simply assigning
scores to the MRA findings and then totaling the
scores. The stage classification using this
MRI/MRA Diagnosis of Moyamoya method corresponds well to the conventional clas-
Disease sification based on angiography and has been
reported to show high sensitivity and
In moyamoya disease patients, anterior circula- specificity [45].
tion (steno-occlusive change of the ICA and col-
lateral moyamoya vessels), posterior circulation
(steno-occlusive change of the PCA and FLAIR Ivy Sign
leptomeningeal anastomosis collateral vessels
from the posterior circulation), and transdural Fluid-attenuated inversion recovery (FLAIR)
anastomosis collateral vessels such as ethmoidal image has been widely used for the diagnosis of
moyamoya vessels and vault moyamoya vessels stroke and other cerebral abnormalities with a
should be evaluated to monitor the hemodynamic high capability of lesion detection by nulling the
state of moyamoya disease patients. Time-of- CSF signal with an employment of sophisticated
flight (TOF) MRA method is more suitable than inversion pulse [46]. Characteristic imaging find-
phase contrast MRA or other MRA methods ing on FLAIR has been reported in moyamoya
because TOF-MRA can show the steno-occlusive disease patients [46]. On FLAIR image,
change of the major artery and development of moyamoya disease patients may show linear or
moyamoya vessels simultaneously and more pre- serpentine high signal in the sulci along the corti-
cisely. MRI/MRA examination can provide the cal surface, which is believed to represent dilated
significant diagnostic information less invasively leptomeningeal collateral vessels [46], and this
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 835

Fig. 9 CT scan image of an adult moyamoya patient spots. (a, b) In the same patient, MRA (d) and T2-WI
shows chronic infarction as low-density area (c). images (e) show steno-occlusive change of bilateral ICA
Contrast-enhanced CT angiography images show and moyamoya vessels. Final diagnosis was confirmed by
moyamoya vessels in the cisternal area as tiny high-density conventional digital subtraction angiography (f, g)

finding is called “FLAIR ivy sign” because of its including moyamoya disease. This mechanism of
appearance resembling an ivy on the wall. This high signal on FLAIR image may at least partly
sign is frequently observed in distal areas with share with the mechanism of findings intra-arterial
decreased perfusion caused by vascular stenosis high signal on FLAIR ivy sign may partly share
836 A. Yamamoto et al.

Fig. 10 In a case of unilateral moyamoya disease, contrast-enhanced CT angiography image shows steno-occlusive
change of the right ICA and surrounding moyamoya vessels in the cistern (a, b)

the mechanism of intra-arterial high signal scanner in the diagnosis of various neurological
observed in acute cerebral infarction on FLAIR disorders. The capability of higher spatial
[47]. The mechanism of FLAIR ivy sign is also resolution and signal-to-noise ratio of 3-T MRI
applicable to the deep medullary vessel area in attributable to higher static magnetic field
moyamoya disease patients, and linear high signal strength is also valuable for evaluating fine imag-
ivy sign was also reported in the deep white ing findings in moyamoya disease patients.
matter [48]. Moyamoya vessels have been reported to be
FLAIR ivy sign can change its appearance better delineated with MRA at 3-T than at
responding to the change of hemodynamic status 1.5-T [52]. For evaluation of petechial hemor-
and, therefore, is useful for the evaluation of rhage in moyamoya disease patients, which rep-
hemodynamic status in moyamoya disease resents an important factor for prognosis,
patients. Its presence correlates highly with susceptibility weighted imaging (SWI) has been
reduced CVR indicating misery perfusion area, reported to offer better detection at 3-T than 1.5-T
and regional arteriocapillary circulation time was MRI [53]. In the routine clinical situation,
elongated [49] but was decreased after bypass MRI/MRA findings also have been used for the
surgery, correlated with improved hemodynamic diagnosis of moyamoya disease, such as cisternal
status observed by 123I-IMP-SPECT [50, 51] moyamoya vessel flow voids and FLAIR ivy
(Figs. 11, 12, 13 and 14). sign [46].
Among these fine imaging findings of
moyamoya disease, the cisternal moyamoya ves-
Feasibility of 3-T MRI/MRA sels visualized as a distinct feature of moyamoya
in Moyamoya Disease disease on 3-T MR might be beneficial for diag-
nosing moyamoya disease. Cisternal moyamoya
3-T MRI scanner has already been widely used in vessels show low-signal flow voids on T2-WI
clinical hospitals and has been reported to produce image and high signal on TOF-MRA [18]. Com-
predominance compared to 1.0-T/1.5-T MR parison between existing MRI/MRA diagnosis
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 837

Table 2 Classification and scoring based on the magnetic radiation exposure) of MRI/MRA than conven-
resonance angiography (MRA) findings [45] tional angiography [18] (Figs. 15, 16, and 17).
MRA findings Score
Internal carotid Normal 0
artery Stenosis of C1 1 Susceptibility Weighted Imaging
Discontinuity of the C1 2 in Moyamoya Disease
signal
Invisible 3
Cerebral hemorrhage is one of the main symptoms
Middle cerebral Normal 0
artery of moyamoya disease especially in adult patients.
Stenosis of M1 1
Petechial hemorrhage in moyamoya disease
Discontinuity of the M1 2
signal patients is considered as an important factor for
Invisible 3 prognosis. On MRI, local inhomogeneous suscep-
Anterior cerebral Normal A2 and its distal 0 tibility effect due to hemorrhagic component such
artery A2 and its distal signal 1 as hemosiderin can become a diagnostic clue for
decrease cerebral hemorrhage. Such local susceptibility
Invisible 2 inhomogeneity effect is more apparent on gradient
Posterior cerebral Normal P2 and its distal 0 echo T2-WI image than T2-WI image. Also, this
artery P2 and its distal signal 1 susceptibility effect is more apparent with 3-T MR
decrease
scanner than with 1.0-T/1.5-T MR scanner. In
Invisible 2
addition, recent development of MRI technology
A total score of ICA to PCA is calculated individually for
both sides including 3-T MR scanner and image acquisition
sequence produced more sophisticated sequence
MRA total score MRA stage for the detection of hemorrhage; susceptibility
0–1 1 weighted imaging (SWI) has been introduced. In
2–4 2 SWI calculation of its final image from gradient
5–7 3 echo T2WI magnitude image, component of
8–10 4 phase mask information is utilized, which can
Reprinted with permission from Guidelines for Diagnosis enhance local susceptibility inhomogeneity effect
and Treatment of Moyamoya Disease (Spontaneous Occlu- more prominent than conventional T2WI image
sion of the Circle of Willis). #The Japan Neurosurgical
Society [3]. MRA stage 1 identified using the above [53]. T2WI image at 3-T found cerebral
approach corresponds to stages I and II of the angiographic microbleeds in 44 % of MMD patients compared
classification, stage 2 corresponds to stage III, stage 3 cor- with 5.8 % in control subjects [54]. They are
responds to stage IV, and stage 4 corresponds to stages V detected significantly more on SWI than T2WI
and VI
image at 3-T [53]. SWI can detect more hemor-
rhagic lesion in moyamoya disease patients than
conventional gradient echo T2WI images.
Not only for hemorrhage detection, SWI also
criteria and the proposed criteria using cisternal can show fine deep medullary veins apparently in
moyamoya vessels was performed. Diagnostic the deep white matter, which has never been visu-
accuracy was 62.5 % with the existing MR criteria alized on other image sequences such as T2WI
and 97.5 % with the proposed MR criteria. The image [55]. Evaluation of deep medullary veins in
proposed MR criteria were more sensitive than the moyamoya disease patients by SWI has been
existing MR criteria, but less specific than the reported, and symptomatic patients show more
existing MR criteria. By using this proposed prominent appearance of deep medullary veins
MRI/MRA criteria, diagnostic accuracy of than asymptomatic patients. Reportedly, promi-
moyamoya disease will be improved, and this nent deep medullary vein appearance correlated
criterion should be suitable for pediatric patients with less cerebral blood flow and cerebral vascu-
because of the less invasiveness (without ionizing lar reserve (Figs. 18 and 19).
838 A. Yamamoto et al.

Fig. 11 3-T MR images of a pediatric moyamoya patient FLAIR ivy sign. Also, collateral vessels in the deep white
are shown. On FLAIR image (b, c), dilated collateral matter are noted (b). Infarction was noted in the bilateral
vessels in the cortical surface are noted as so-called parietal cortical area

Fig. 12 3-T MR images of


an adult moyamoya disease
patient are shown. (a) MIP
image of TOF-MRA shows
bilateral stenosis of ICA and
moyamoya vessels. (b)
FLAIR image shows intra-
sulcal high-signal-intensity
area representing dilated
collateral vessels. This
finding is known as “FLAIR
ivy sign.” (c) Also, FLAIR
image at lower level shows
linear high-signal-intensity
area in the deep white
matter. This deep medullary
vessel high signal on
FLAIR also represents
dilated collateral vessels.
Infarction is apparent in the
same slice

modality due to its higher spatial resolution and


Staging by Conventional Angiography capability of hemodynamics information
in Moyamoya Disease (Table 3).
However, as mentioned in the above sections,
When the original concept of moyamoya disease as far as the depiction of the steno-occlusive
was introduced, digital subtraction angiography change of the circle Willis is concerned, other
(DSA) is still the most reliable diagnostic
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 839

Fig. 13 FLAIR ivy sign


and cerebral perfusion. A
man in his 60s lost
consciousness when
driving. Stroke was
suspected, and MRI/MRA
was conducted. (a) MRA
shows severe stenosis at
bilateral ICA tops. There is
no apparent asymmetry.
(b–d) FLAIR images at
different levels show no
asymmetry at a glance.
However, a careful look at
the right Sylvian fissure and
frontal sulci shows
serpentine high signal,
called “FLAIR ivy sign.” It
is considered to indicate
maximally dilated and
slowly flowing arterial
branches. (e) IMP-SPECT
at rest shows frontoparietal
hypoperfusion bilaterally
without apparent
asymmetry. (f) IMP-SPECT
after acetazolamide or
vasodilator infusion shows
reduced CBF at the right
frontal lobe where “FLAIR
ivy sign” is observed. (g)
Cerebrovascular reserve
map (CVR) or percent CBF
increase map decreased
flow reserve at the right
frontal lobe. Other areas
show normal CBF increase
of more than 30 %,
represented as yellow to red
colors

modalities such as TOF-MRA and three- circulation is not always clear even with state-of-
dimensional CTA with best quality are quite com- the-art MRA/MRI due to temporal and spatial
patible with DSA. However, the depiction of the resolution and signal-to-noise ratio limitation. In
fine moyamoya vessels and the collateral addition, 3D-CTA essentially shows the
840 A. Yamamoto et al.

Fig. 14 FLAIR ivy sign pre- and post-bypass surgery. A reduced CBF at the right temporo-occipital area. FLAIR
man in his 40s had headache and paresthesia. (a) MRA ivy sign is observed at the corresponding area (d), which
shows bilateral ICA and right PCA stenosis. (b) Rest and disappeared after right STA-MCA bypass surgery (e)
(c) post-acetazolamide infusion IMP-SPECT shows

morphological aspect of the vasculature that is not subtraction angiography may be necessary
necessarily identical to the true circulation of the (Fig. 20).
blood flow (lack of information of
hemodynamics).
In addition, the collateral circulations such as Angiographical Staging Progression
basal moyamoya and transdural anastomosis of of Moyamoya Disease
the meningeal artery including the vault
moyamoya and ethmoidal moyamoya may not Angiographically, moyamoya disease progresses
be well demonstrated on other imaging modali- along with the six stages introduced by Suzuki
ties. In addition, a microaneurysm associated with [1, 56]. Firstly, stenosis appears in the terminal
moyamoya vessels, which is supposed to be the portion of the ICA and the proximal portion of the
cause of the intracerebral and intraventricular ACA and MCA bilaterally. Development of an
hemorrhage, may not be well demonstrated on extensive collateral network at the base of the
other imaging modalities. For the diagnosis of brain along with the classic “puff of smoke”
these small microaneurysms, conventional digital appearance on conventional angiography is seen
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 841

Fig. 15 3-T MR images of a pediatric moyamoya patient MIP image of 3D-TOF-MRA shows moyamoya vessels
are shown. On T2-WI axial image (a), moyamoya vessels and steno-occlusive change of bilateral terminal portion of
in the sylvian valley are apparent as collection of tiny flow the internal carotid arteries. (d) Chronic infarction was
voids. The source image of time-of-flight MR angiography apparent on both T2-WI image (a) and FLAIR image (c)
(b) shows moyamoya vessels as high signal intensities.

in the intermediate stage. “Basal moyamoya” ophthalmic artery to the ACA branches
includes abnormal dilatation of the perforating [57]. Finally, the collateral flow from the dural
arteries, such as the lenticulostriate artery and arteries to the pial arteries develops, and this path-
the thalamoperforating artery, in the basal ganglia way is known as “vault moyamoya” [57]. Most of
and thalamus. The steno-occlusive process also the patients present with intermediate stages and
involves the posterior circulation, including the are treated promptly; therefore, the advanced
basilar and posterior cerebral arteries. Total occlu- stage is nowadays rarely seen.
sion of the ICA terminal results in the disappear-
ance of “basal moyamoya,” and simultaneous
development of other collateral circulation is Aneurysm in Moyamoya Disease
observed in the advanced stage. “Ethmoidal
moyamoya” involves dilatation of the anterior In adult moyamoya disease patients, prevalence of
and posterior ethmoidal arteries, which also func- aneurysm has been reported as 14 % [58]. How-
tion as a collateral pathway, mainly from the ever, in pediatric moyamoya disease patients,
842 A. Yamamoto et al.

Fig. 16 3-T MR images of


an adult moyamoya disease
patient are shown. T2-WI
image (a), TOF-MRA
image (b), and MIP image
of TOF-MRA show
moyamoya vessels in the
cistern and bilateral ICA
stenosis. Also, chronic
infarction is apparent in the
left temporal lobe (a)

aneurysm is rare. Aneurysms in moyamoya dis-


ease can be categorized as (1) aneurysm occurring Roles of Morphological
in the circle of Willis and (2) small aneurysm and Hemodynamic Functional Imaging
occurring in the peripheral or moyamoya vessels. for MMD
In the former, contrary to the common prevalence
of aneurysm, higher prevalence has been reported Diagnosis
in the posterior circulation especially
vertebrobasilar artery systems bifurcation. It has Conventional angiography is the gold standard for
been reported that aneurysm was rarely seen in the the diagnosis of moyamoya disease (MMD)
MCA and ACA. In the latter, spontaneous regres- [48, 59]. It clearly visualizes arterial stenosis and
sion or size decrease after revascularization sur- moyamoya vessels (MMVs) and enables correct
gery has been reported; therefore, evaluation, but there may be some difficulty when
pseudoaneurysm due to the rupture of moyamoya it is conducted in children. Moreover, conven-
vessels has been hypothesized as a cause. tional angiography may have potentially serious
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 843

Fig. 17 3-T MR images of


an adult moyamoya disease
patient are shown. (a) Time-
of-flight MRA source
image and (b) T2-weighted
image show cisternal
moyamoya vessels
apparently. (c) MIP image
of TOF-MRA shows
bilateral ICA stenosis

adverse effects [60]. Noninvasive imaging diag- comparable to conventional method [62] and col-
nosis is preferable, and angiography using CT and laterals [63], but it still requires infusion of con-
MR (CTA and MRA, respectively) has been much trast material and radiation exposure is inevitable.
investigated for the diagnosis of MMD. Recent More recently, a 3-T MRI system is widely
progress in noninvasive imaging methods has available and has superiority to 1.5-T for the diag-
improved diagnostic capability. Guidelines for nosis of various neurological disorders. The
the diagnosis and treatment of spontaneous occlu- higher spatial resolution and signal-to-noise ratio
sion of the circle of Willis (i.e., MMD) from the (SNR) at 3-T are useful for evaluating the fine
Ministry of Health and Welfare, Japan, have vascular changes of MMD. MMVs can be better
included MRA as a diagnostic method delineated with MRA at 3-T than at 1.5-T
[3, 19]. Time-of-flight (TOF) MR angiography at [52]. Sawada et al. [18] focused on cisternal
1.5-T could accurately depict stenosis around MMVs compared with MMVs at the basal
80 % with some overestimation. Moyamoya ves- ganglia, which is adopted in the existing MR
sels (MMVs) were better visualized with MRI criteria, and reported that detection of cisternal
than MRA, and MRI diagnosis had 92 % sensi- MMVs on T2-WI and MRA could attain 97.5 %
tivity and 100 % specificity at 1.5-T [61]. CTA is sensitivity with slightly deteriorated specificity of
accurate and has high diagnostic capability 95 % at 3-T. MMD is a progressive disorder, and
844 A. Yamamoto et al.

Fig. 18 (a) CT scan image of an adult moyamoya patient low-signal-intensity area more apparently. Magnitude
shows low-density area in the right basal ganglia. (b) 3-T image (d) and SWI (e) and MiniP image (f) are shown.
MRI of T2-WI image shows low signal intensity in the On SWI and MiniP image, hemorrhage is the most appar-
same area, indicating that there might be hemosiderin due ent. Also, on SWI, deep medullary veins are apparent as
to hemorrhage. (c) T2WI image shows this hemorrhage as low-signal-intensity stripe

serial evaluation is mandatory. Imaging measure- cerebral arteries. MRA scores were significantly
ments had better be noninvasive and repeatable correlated with the conventional angiographic
with high precision, hopefully without radiation staging (coefficient = 0.77). Four grades based
exposure or contrast material administration. on the MRA scoring system correlated well with
Suzuki’s stages with high sensitivity and specific-
ity, and is a reliable alternative to conventional
Staging of Moyamoya Disease by staging, especially for children.
MRI/MRA At the same time, we have to be aware
that disparity is sometimes observed between
For this purpose as well, conventional angiogra- angiography staging and clinical severity. Even
phy has been the reference standard, but MRA when bilateral ICAs are heavily stenotic or
scoring has been proposed [45]. Scores were occluded, ample collateral vessels may keep the
assigned from 0 (normal) to 10 (most severe) for patient asymptomatic, whereas patients with
changes from normal, stenosis, discontinuity, and angiographically confirmed mild stenosis may
invisibility of the ICA, the horizontal portion of have TIA attacks. Simple angiographic
the MCA, and the anterior and the posterior evaluation may not be sufficient to estimate future
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 845

Fig. 19 Difference of appearance of hemorrhage among refocus pulse used in T2-WI image. In addition, MiniP
different MRI sequences. T2-WI image (a), T2WI image, image of SWI shows hemorrhage as more pronounced
and MiniP image of SWI of hemorrhage in the left tempo- low-signal area compared to T2WI image (c). This differ-
ral lobe are shown. T2WI image shows hemorrhage as ence comes from the fact that SWI uses phase information
more pronounced low-signal area compared to T2-WI to enhance the local susceptibility difference
image (b). This difference comes from the effect of 180

Table 3 Suzuki’s stage classification of moyamoya dis-


ease by conventional angiography [3] Altered Perfusion Status in MMD
I Narrowing of the carotid fork
II Initiation of the moyamoya (dilated major cerebral The cerebral artery has an autoregulation mecha-
artery and a slight moyamoya vessel network) nism, and CBF stays constant for changes in cere-
III Intensification of the moyamoya (disappearance of bral perfusion pressure (CPP) within the range of
the middle and anterior cerebral arteries and thick
and distinct moyamoya vessels) 50–150 mmHg. When stenosis or occlusion of a
IV Minimization of the moyamoya (disappearance of cerebral vessel around the circle of Willis reduces
the PCA and narrowing of individual moyamoya CPP beyond the autoregulatory capacity, CBF
vessels) begins to decrease. When CPP reduction is not
V Reduction of the moyamoya (disappearance of all so severe, CBV increases and oxygen extraction
the main cerebral arteries arising from the internal
fraction (OEF) increases. This compensatory
carotid artery system, further minimization of the
moyamoya vessels, and an increase in the reaction maintains cerebral metabolic rate of oxy-
collateral pathways from the external carotid artery gen (CMRO2) and brain function. Further reduc-
system) tion of CPP, however, cannot be compensated and
VI Disappearance of the moyamoya (disappearance of may result in infarction.
the moyamoya vessels, with cerebral blood flow
derived only from the external carotid artery and
There is another index of compensatory mech-
the vertebrobasilar artery systems) anism, called cerebrovascular reserve (CVR). It
Reprinted with permission from Guidelines for Diagnosis reflects the capacity to further dilate vessels
and Treatment of Moyamoya Disease (Spontaneous Occlu- from static condition, when CPP is decreased or
sion of the Circle of Willis). #The Japan Neurosurgical vasodilator such as acetazolamide (ACZ) or CO2
Society [3]
is administered. Vessels are fully dilated in
severely ischemic condition, and little
morbidity of the MMD patients. Perfusion status additional capacity of dilatation remains,
has to be examined more in detail including infor- which is reflected as decreased CVR. In the
mation on the affected regions and local worst, “steal phenomenon” is observed where
hemodynamics. flow is diverted to dilated vessels within a
846 A. Yamamoto et al.

Fig. 20 MIP image of TOF-MRA shows stenosis of ICA basal area. Ethmoidal moyamoya vessels in the frontal
(a). Conventional digital subtraction angiography (b, c) skull base area are also apparent only on conventional
shows same stenosis more precisely. In addition, conven- DSA image
tional DSA image shows finer moyamoya vessels in the

non-impaired region. In patients with unilateral ischemic attacks (TIA), infarction associated
ICA or MCA occlusion, strokes recurred in with TIA (I/TIA), permanent deficit with infarc-
35 % and 6 % of cases with and without regional tion (PD), and non-symptomatic (NS) and com-
CVR reduction for a follow-up period of pared these types with normal controls using 15O-
24 months [64]. CVR is an important predictor labeled PET tracers. In NS, cerebral blood flow
of future morbidity. (CBF) was preserved, but cerebral blood volume
CBF and CVR difference in age has to be taken (CBV) was increased without increase in oxygen
into considerations for evaluation of perfusion extraction fraction (OEF). In patients with TIA,
status. Normal CBF is known to correlate nega- CBF was preserved, but CBV showed further
tively with age. When young, CBF at rest shows increment. If they got infarction (I/TIA), CBF
hyperperfusion at the frontal region, and CVR is was largely decreased by around 10 ml/min/
also very high. Both CBF and CVR decline with 100 ml and OEF was increased. When they got
aging. The images and measured values depend PD, all of OEF, CBV, and CBF were decreased
on the method used for the measurement (Fig. 21). (matched perfusion status).
MMD is graded based on digital subtraction
angiography (DSA), which has limited clinical
availability. Czabanka et al. [66] proposed a grad-
Evaluation of Perfusion Status ing system included the following: for DSA, ste-
nosis/occlusion = 1 point, stenosis/occlusion +
There are several conditions of cerebral perfusion intracranial compensation = 2 points, and steno-
associated with MMD. Nariai et al. [65] sorted sis/occlusion + intracranial compensation +
MMD patients out into those with transient extracranial-intracranial compensation = 3 points
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 847

Fig. 21 MRA and cerebral perfusion. Pre- and postoper- improved. (c) After successive left STA-MCA bypass sur-
ative changes in TOF-MRA (left column) and IMP-SPECT gery. Although right bypass is better visualized this time
perfusion at rest (right column) by bypass surgery. (a) indicating further improvement in perfusion on the right
Before bypass surgery. Collateral circulation from the MCA area, the left MCA area shows higher perfusion than
right PCA and MCA, resulting in less perfusion on the the other side. Note the relative flow decrease in the cere-
right hemisphere. (b) After right STA-MCA bypass sur- bellum caused by the cerebral flow increase
gery. The bypass is well visualized, and hypoperfusion is

and, for MRI, no sign of ischemia = 0 point, signs Oxygen-15 PET


of ischemia = 1 point, and CVR capacity > 5 %
= 0 points and < 5 % = 2 points. MMD grade I This modality is considered most accurate and has
referred to 1–2 points, grade II to 3–4, and grade comprehensive ability to estimate multiple hemo-
III to 5–6 points. In an analysis of 80 hemispheres dynamic parameters: CBF, CBV, OEF (oxygen
of MMD patients, they were graded as mild extraction fraction), and therefore CMRO2 (cere-
(grade I, 18 %), moderate (grade II, 44 %), and bral metabolic rate of oxygen). This modality has
severe (grade III, 39 %) with clinical symptoms in been widely used to study patients with MMD
21 %, 63 %, and 93 %, respectively. This grading [65, 68, 69]. However, 15O-PET examination sys-
system allows stratification for clinical symptoms tem is not readily available, and the short half-life
in MMD patients. time of 15O (2 min) requires a cyclotron that is
Mismatched perfusion condition has to be cor- very expensive.
rectly evaluated. Until recently, PET (positron
emission tomography), SPECT (single-photon
emission tomography), and Xe-CT have been SPECT
used. CT perfusion and MR imaging including
DSC and ASL can now be used for cerebral Availability of SPECT is much higher than PET.
hemodynamic analysis [67]. Each has its advan- However, quantitation of CBF is affected by scan
tages and limitations, which should be well condition as well as the tracer used for measure-
understood. ment, and spatial resolution is much lower than
848 A. Yamamoto et al.

PET. In spite of these limitations, MMD has been multi-detector row CT that can cover the whole
much studied with SPECT [70–72]. Additionally brain [81].
to rest CBF, CVR (cerebrovascular reserve) can
be measured by using acetazolamide (ACZ,
17 mg/kg) to dilate the cerebrovasculature. MR Perfusion
Three tracers are currently used for brain
123
perfusion SPECT: I-N-isopropyl-p- As in CT, MR perfusion can be conducted
99m
iodoamphetamine (IMP), Tc-ethyl cysteinate using gadolinium contrast agent, called
dimer (ECD), and 99mTc-hexamethyl propylene dynamic susceptibility contrast (DSC) MR perfu-
amine oxime (HMPAO). Among them, sion imaging [67], where first-pass signal
IMP is used for quantitative scan, called change is analyzed and semiquantitative results
“QSPECT” [73–75]. In this scan protocol, rest can be obtained. The area size of signal
and ACZ-loaded scans can be successively change shows CBV, and peak height shows
conducted. Rest scan begins at the onset of IMP CBF, and MTT is calculated as CBV/CBF. As in
infusion that lasts for a minute. After around CT, quantitation is conducted using AIF (and
10 min, an arterial blood sample is collected to VOF). Signal intensity after contrast administra-
calibrate arterial input function. ACZ is infused at tion is logarithmically proportional to about
20 min, and scan lasts to 28 min later. After 2 min 5 mMol/L at 1.5-T, but this linear relationship is
intermission, i.e., at 30 min, a second scan is not true when the concentration becomes higher
initiated by infusing another dose of IMP and [82]. There are several models and
lasts to 58 min. Quantitation is not conducted in implementations for this analysis, and results
a pediatric case. may be different with frequent effects of bolus
delay [80]. Although the administered amount is
much smaller than iodine CT contrast material,
CT Perfusion free form of gadolinium is highly toxic and rarely
causes fatal event in patients with renal insuffi-
Xenon-enhanced CT was much used in the former ciency called “nephrogenic systemic fibrosis” or
time, but more recently, perfusion CT is NSF [83].
conducted by dynamic intravenous infusion of
iodine contrast material using a first-pass
tracer model. With the advent of multi-detector Arterial Spin Labeling
row CT that does not require table movement
to cover the central part or the entire brain, There is another MR perfusion method called
this method is gaining popularity and has been “arterial spin labeling” or ASL [84, 85]. It sup-
used in MMD cases [76–78]. Higher availability presses the signal from stationary tissues and
of MDCT scanners than RI scanners is advanta- detects blood signal that flows into the brain.
geous. By using arterial input function (AIF) The blood is magnetically labeled upstream at
and venous output function (VOF), the skull base and used as an endogenous contrast
quantitative measurement of CBF, CBV, BAT material, and no contrast material administration
(bolus arrival time), or MTT (mean transit is required. For better background signal suppres-
time) can be conducted [79]. However, sion, brain images are acquired as a pair of images
cautions have to be paid, because the results with and without labeling, and subtraction is
can be affected by the analysis methods [80]. conducted. Due to this process, motion causes a
Radiation exposure is the most critical issue, large misregistration artifact. Recently, however,
but modern dose-reduction methods make ASL imaging can be conducted in as short as
this method more attractive. CT perfusion 1 min on a 3-T MRI system (Fig. 22). Because
imaging and analysis of the whole brain there is no need to use exogenous contrast mate-
can now be conducted by introduction of rial, ASL measurement can be conducted
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 849

Fig. 22 Arterial spin labeling (ASL) for perfusion mea- reduced perfusion called “arterial spin labeling” or ASL
surement. (a) Severe stenosis is observed at bilateral MCA (c). In this case, arterial signal is prominent on the left
origin. (b) IMP-SPECT after acetazolamide infusion MCA area as high signal surrounded by low signal,
shows reduced CBF at the left MCA area, which is hard which means that arterial flow is very slow on the
to infer from the MRA. There is another method to detect corresponding area

repeatedly [86], which is beneficial especially for


the children. Labeling can also be conducted to a CBF Changes After Bypass Surgery
specific artery. Saida et al. applied this method to a
bypassed artery and found relatively low flow in Quantitative hemodynamics enables better selec-
the early postoperative period, which later tion of patients for revascularization surgery and
increased [87]. Post-bypass perfusion can also be also postoperative evaluation of the clinical
hyperemic in the early period, and noninvasive outcome. Cerebral arterial bypass surgery
ASL can be applied to multiphase observations. improves CBF and CVR, and the latter is more
CBF measured by ASL had relatively weak prominent [90]. It also decreases CBV and OEF
correlation with that by rest IMP-SPECT. How- [68, 72]. Early increase in CBF after revasculari-
ever, it was much higher with ACZ IMP-SPECT zation can be detected as an asymmetry ratio of
and CVR, suggesting that ASL could reflect the the operative side compared with the other side
currently and potentially dangerous zones for using IMP-SPECT and ASL [91]. It can be symp-
ischemia [88]. Determining the presence and ade- tomatic hyperperfusion, which is a potential com-
quacy of collateral blood flow is important in plication of bypass surgery. In a review of 82 adult
MMD. ASL can also identify collaterals in MMD patients with 99 surgeries, 39 patients
MMD patients compared with DSA in high sen- (47 sides, 47 %) suffered from transient neurolog-
sitivity and specificity over 0.8 [89]. ASL is use- ical deterioration due to hyperperfusion from 1 to
ful, but one drawback is that ASL is highly 9 days after operation (median 2 days), which was
sensitive to delay in arterial arrival time. Labeled sustained for 1–14 days (median 7 days) [92,
blood may not reach the brain parenchyma and 93]. If symptom is severe, MR scans with
stay within the proximal artery. It looks as if flow T2WI or susceptibility weighted imaging (SWI)
is increased on the more severely impaired side. would be the primary choice to exclude subarach-
Caution has to be paid on the serpentine high noid hemorrhage, and addition of an ASL scan
signal within the artery (Fig. 23). would be helpful.
850 A. Yamamoto et al.

Fig. 23 ASL dependency


on post-labeling delay time
(PLDT). (a) An MMD
patient after bilateral
STA-MCA bypass surgery.
(b) ASL with PLDT of
1,800 ms. Intra-arterial
signal is prominent, and
severe CBF reduction can
be assumed. However, (c)
CVR measured by
IMP-SPECT shows no
apparent reduction. (d) ASL
with PLDT of 2,600 ms
suggests much better
perfusion status. ASL is
very easy to use, but caution
has to be paid to transit
artifact caused by prolonged
arterial transit time

In a retrospective review of 73 MMD children


who had surgical revascularization due to strokes Visualization of Lenticulostriate
or TIAs, three children (4.1 %) had recurrent Artery (LSA)
strokes, and TIAs were improved in about 80 %
for follow-up periods of 34 (5–166) months Dilatation of the LSA is visualized as moyamoya
[94]. Pediatric MMD patients at 6–12 months vessels. Occlusion of this small artery causes
after indirect bypass operation, preserved CVR one-third of all strokes and infrequent hemorrhage
observed by SPECT, had better outcome than due to aneurysm formation [95]. For LSA
those with decreased CVR resulting in remaining visualization, CT angiography can be used. It
neurological deficit and ischemic attacks on allows subtle changes such as diminished visual-
follow-up [71]. However, it is not necessarily ization of the LSA [96]. In MRA, TOF-MRA can
matched with clinical outcomes. When CVR visualize some LSA branches. It visualizes LSA
improvement is not observed, close monitoring as “white-blood”; there is another option of LSA
would be required especially when patients have visualization as “black-blood.” One of such
worsening symptoms. method is called flow-sensitive black-blood
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 851

Fig. 24 TOF-MRA
vs. flow-sensitive black-
blood (FSBB) MRA. A case
of unilateral MMD. (a)
TOF-MRA shows severe
left MCA stenosis. (b)
FSBB-MRA at 1.5-T shows
focal stenosis with distal
patency. Note that the
lenticulostriate artery is
more prominent on the
left side

(FSBB) MRA, which can visualize more of the


LSA branches than TOF-MRA [97]. This Treatment of Moyamoya Disease
relatively new method found reduced visualiza-
tion of LSA branches in lacunar stroke Treatment for Ischemic Moyamoya
patients [98]. Disease
Another interesting option is a hybrid of
opposite-contrast (HOP) MRA that combines Surgical Treatment
TOF and FSBB contrasts using a dual-echo
three-dimensional gradient echo sequence Indication
[99]. Black-blood signal is subtracted Surgical revascularization improves hemody-
from white-blood signal making better namic impairment and reduces ischemic attacks
depiction of arteries and suppression of the back- in patients with ischemic moyamoya disease
ground. In cases with major trunk, [101]. Therefore, indications of surgical treatment
HOP-MRA better visualized branches distal to should be first examined in the patients
steno-occlusion of the major trunk artery, which manifesting with cerebral ischemic attacks. Chil-
was considered to represent collaterals and dren and adults with moyamoya disease who suf-
would contribute much as well in MMD [100] fer from cerebral infarction or frequent TIAs
(Figs. 24 and 25). caused by definite hemodynamic impairment are
good candidates for surgery.
852 A. Yamamoto et al.

Fig. 25 FSBB-MRA of a
normal subject at 3-T. The
lenticulostriate artery is
much better depicted at 3-T
than at 1.5-T

Single-photon emission computed tomogra- 2. Adult cases: Either one of the following
phy (SPECT) is widely used to measure regional approaches is effective: direct bypass proce-
cerebral blood flow (rCBF) and detect hemody- dures or a combination of procedures including
namic failure. SPECT studies obtained after intra- direct bypass. Indirect bypass procedures only
venous administration of acetazolamide can are not recommended because of inferior
evaluate cerebrovascular reserve capacity which effectiveness.
is an excellent indicator of hemodynamic failure
[102]. Positron emission computer tomography Direct Bypass
(PET) using 15O tracer can evaluate rCBF, Direct bypass is a procedure for directly
regional cerebral blood volume (rCBV), regional establishing the blood flow pathway from extra-
cerebral metabolic rate of oxygen (rCMRO2), and cranial arteries to intracranial cortical arteries
regional oxygen extraction fraction (rOEF). At using the microvascular anastomotic technique
present, PET study is thought to be a gold standard (Fig. 26). Superficial temporal artery-to-middle
to detect cerebral hemodynamic failure. cerebral artery (STA-MCA) anastomosis is the
most popular and has been proven to be effective
Selection and Application of Surgical in improving cerebral circulation in moyamoya
Revascularization Procedures disease [104, 105]. It can be combined with vari-
Since the 1970s, numerous surgical techniques ous indirect procedures. Direct bypass provides an
have been proposed, which can be divided into immediate improvement of hemodynamic failure,
two categories: direct anastomotic bypass but it requires sophisticated skills, because the
and indirect bypass. Active debates on the selec- walls of the recipient cortical arteries are
tion of these bypass procedures were once com- extremely fragile in moyamoya disease.
mon in the previous century, but a consensus on
this issue has recently emerged, as clearly stated in Indirect Bypass
the new guidelines published in 2009 Principally, indirect methods are based on the
(in Japanese) [103]. idea that neovascularization can be induced
from the extracranial arteries to the cortical
1. Pediatric cases: Either one of the following arteries by putting the vascular-rich tissues.
approaches is effective at improving hemody- A variety of indirect revascularization procedures
namic impairment: direct bypass procedures or – such as encephalo-myo-synangiosis (EMS),
indirect bypass procedures. encephalo-galeo-synangiosis (EGS),
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 853

Fig. 26 Direct bypass procedures for a 3-year-old female dura mater. (d) Donor STA and recipient MCA. (e)
with moyamoya disease. (a) Skin incision and line of the Microanastomosis is completed. (f) Confirmation of the
craniotomy. (b) STA is dissected from the inner surface of bypass flow (white arrows) using fluorescent dye
the skin flap. (c) Brain surface is exposed by opening the

encephaloduroarteriosynangiosis (EDAS), and surgery. Repeated radiological studies are


omentum transplantation – have been described recommended during the postoperative acute
[106–109]. Although indirect revascularization is period to detect subdural or epidural hematomas
technically easy, the success of this strategy because most patients with ischemic moyamoya
depends on the natural neovascularization capa- disease are under the effect of preoperative
bility of the patient’s brain; in some cases, post- antiplatelet drugs.
operative collateral formation is insufficient and Transient neurological events mimicking TIAs
the progression of cerebral ischemia cannot be are frequently observed after direct bypass pro-
halted [105, 110]. It is well known that neovascu- cedures with no evidence of infarction apparent in
larization tends to be insufficient in adult patients. MR imaging. Although their pathophysiology has
long remained a mystery, recent postoperative
Perioperative Management SPECT studies have revealed that these events
Regardless of the kinds of the bypass procedure, can be caused by local hyperperfusion
perioperative management is extremely impor- [111]. Because management of hyperperfusion is
tant. Throughout the period, adequate intravenous completely different from that of hypoperfusion
drip is mandatory to avoid dehydration. Because (ischemia), the postoperative routine CBF study is
intraoperative hypercapnia, hypocapnia, and essential (Figs. 27 and 28).
hypotension can increase the risk of ischemic
complications, anesthesiologists are required to Medical Treatment
strictly control PaCO2 levels within the normal Patients only with mild hemodynamic failure
range and to avoid systemic hypotension during can be treated conservatively. However, it must
854 A. Yamamoto et al.

Fig. 27 Effect of STA-MCA bypass in ischemic MCA territory. (c) Preoperative 123I-IMP-SPECT showing
moyamoya disease. (a, b) The anteroposterior view (a) the reduced CBF in the bilateral frontal lobe. (d) Postop-
and the lateral view (b) of the left external carotid angio- erative 123I-IMP-SPECT showing the increased CBF in the
gram obtained after left STA-MCA bypass. The blood flow left frontal lobe
via the STA (black arrows) covers the wide area of the

Fig. 28 Postoperative hyperperfusion. (a) Intraoperative confirmation of the bypass flow (yellow arrows). (b) 123I-IMP-
SPECT on postoperative day 7 showing local hyperperfusion (white arrows)
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 855

Fig. 29 Intracranial bleeding patterns of hemorrhagic Intraventricular hemorrhage. (d) Subcortical hemorrhage.
moyamoya disease. (a) Thalamic hemorrhage. (b) (e) Subarachnoid hemorrhage
Putaminal hemorrhage with ventricular perforation. (c)

be remembered that moyamoya disease is a pro- Treatment for Hemorrhagic Moyamoya


gressive disease and the hemodynamic failure can Disease
deteriorate later. When medical treatment is
selected, oral administration of aspirin or More than one-half of all adult patients with
clopidogrel is recommended for both adults moyamoya disease present with intracranial hem-
and children. Care must be taken, however, to orrhage [112]. Such bleeding attacks, which are
avoid excessively long-term treatment with potentially fatal, seriously affect the patient’s
antiplatelet drugs in adults because it can prognosis [113]. Typically, the hemorrhage occurs
increase the risk of intracranial bleeding. Hemor- in the thalamus and basal ganglia and frequently
rhagic transformation of adult ischemic involves perforation to the lateral and third ven-
moyamoya disease is not uncommon. Therefore, tricles. In rare instances, subcortical or subarach-
these drugs should be discontinued when the noid hemorrhage can also be observed (Fig. 29).
patients are free from ischemic attacks for It is speculated that chronic hemodynamic
several years. stress might induce pathological change in the
856 A. Yamamoto et al.

collateral moyamoya vessels and the following because the rate of rebleeding attacks is extremely
phenomena cause bleeding attacks [114]: high [115, 116]. A survey by Nishimoto
et al. revealed that 33 % of 175 patients with
1. Rupture of fragile and maximally dilated hemorrhagic moyamoya disease experienced a
moyamoya vessels rebleeding attack [115]. Moreover, Kobayashi
2. Rupture of microaneurysms formed in the et al. reported the annual rebleeding rate to be
moyamoya vessels 7.09 % [116].
Although these hemorrhagic attacks cause seri-
Management of the Acute Period ous permanent neurological deficits and the mas-
The patient should be treated with special consid- sive bleeding can be life-threatening, no
eration for the pathophysiology of moyamoya therapeutic method for preventing rebleeding
disease. Although blood pressure should be attacks has been established. No evidence exists
strictly controlled to prevent rebleeding during that treatment of hypertension reduces the
the acute period, excessive reduction of blood rebleeding rate in hemorrhagic moyamoya disease.
pressure and dehydration must be avoided At present, bypass surgery is the only promis-
because hemodynamic failure can coexist. Acute ing strategy. It is well known that, in ischemic
hydrocephalus should be treated by emergent ven- moyamoya disease, reductions in moyamoya ves-
tricular drainage to reduce the intracranial pres- sels can often be detected by angiography after
sure and maintain adequate cerebral perfusion direct bypass surgery (Fig. 30) [117]. Because tiny
pressure. Massive intracerebral bleeding requires moyamoya vessels are the source of the bleeding,
removal of the hematoma through a craniotomy. the rate of hemorrhagic attacks can possibly be
Ruptured microaneurysms in tiny collateral ves- decreased by reducing this hemodynamic stress
sels are usually treated conservatively because and consequently reducing the size and number of
direct excision and intravascular treatment are moyamoya vessels. As a result, a hypothesis has
both difficult. emerged that direct anastomotic bypass surgery
prevents future rebleeding; in fact, some authors
Prevention of Rebleeding in the have reported the effectiveness of direct bypass
Chronic Stage for hemorrhagic moyamoya disease [118]. On the
Management of the hemorrhagic type of other hand, some reports indicate no significant
moyamoya disease presents a serious challenge reduction in the rebleeding rate following

Fig. 30 Reduction of moyamoya vessels after STA-MCA bypass. The reduced size of the moyamoya vessels is
bypass in adult patients (ischemic type). (a) A left internal evident. (c) A left external carotid angiogram obtained
carotid angiogram obtained before surgery. The after STA-MCA bypass. The blood flow via the STA
moyamoya vessels are remarkably well developed. (b) A (arrow) covers approximately two-thirds of the MCA ter-
left internal carotid angiogram obtained after STA-MCA ritory (red arrowheads)
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 857

revascularization surgery [113, 119]; therefore, women with moyamoya disease was conducted
surgical treatment of adult hemorrhagic in Japan [122]. This survey targeted the experi-
moyamoya disease remains controversial. ences of 270 perinatal medical centers during the
To resolve these issues, the Japan Adult 5 years. It cited 64 deliveries comprising 59 cases
Moyamoya (JAM) Trial was undertaken in Japan of previously diagnosed moyamoya disease and
in 2001 [2, 120]. This multicentered prospective 5 cases of moyamoya disease newly diagnosed as
randomized controlled trial seeks to determine a result of perinatal stroke. The results of this
whether “direct” bypass surgery such as survey indicate that the incidence of perinatal
STA-MCA anastomosis affects the prognosis stroke is low in pregnant women previously diag-
and incidence of recurrent bleeding attacks. nosed with moyamoya disease. Bypass surgery
Patients who fulfilled all the clinical and radiolog- had been performed on 58 % of the patients before
ical requirements underwent a computer- pregnancy. Only one case of cerebral hemorrhage
generated randomization scheme and were during pregnancy was reported in which the
assigned to receive either the best medical care maternal prognosis was poor. Although caesarian
to modify risk factors or the best medical care plus section was mainly employed for women
extracranial-intracranial direct bypass. Eighty previously diagnosed with moyamoya disease
patients were enrolled in the trial, and a 5-year (76 %), no attacks were observed during either
follow-up of all the patients was completed in caesarian sections or vaginal deliveries;
2013. Kaplan-Meier survival analysis revealed therefore, no evidence exists that vaginal delivery
that direct bypass surgery significantly decreased should be avoided. On the other hand, serious
the rate of both all adverse events and rebleeding cerebrovascular events (three cases of
attacks during the following 5 years [2]. These cerebral hemorrhage and two cases of cerebral
results strongly suggest that the newly established ischemia) occurred in patients who had not been
bypass flow can change the hemodynamic state of diagnosed with moyamoya disease before preg-
the collateral vessels and lessen their overstress. nancy, and one patient with intracerebral
While subanalyses of the JAM Trial are now hemorrhage died.
underway, this epoch-making study will establish Although most pregnant patients known to
a guiding principle for treatment of hemorrhagic have moyamoya disease can deliver safely, the
moyamoya disease. risk of pregnancy and delivery in women with
moyamoya disease should not be underestimated.
Special Topics Moyamoya Disease and Serious cerebrovascular events occur in patients
Pregnancy with moyamoya disease undiagnosed before preg-
It is not uncommon for such patients to become nancy. Poor prognosis is mostly likely to result
pregnant and give birth. In general, pregnancy is from cerebral hemorrhage. All physicians have an
known to increase the risk of cerebrovascular dis- obligation to provide accurate information and
ease [121]. No evidence exists that the risk of ensure appropriate management of patients with
stroke increases during pregnancy in patients with moyamoya disease. Rapid formulation of guide-
moyamoya disease; however, there have been spo- lines on the management of pregnancy and deliv-
radic case reports and reviews of pregnant ery in women with moyamoya disease is
moyamoya patients presenting with stroke. At pre- mandatory.
sent, no guidelines have been issued for managing
pregnancy in patients diagnosed with moyamoya
disease, and even recommendations regarding Summary
delivery method (natural labor, painless labor
with spinal epidural anesthesia, or caesarian sec- 1. Moyamoya disease has an unknown origin and
tion) seem to differ from one hospital to the next. shows steno-occlusive change of the terminal
In 2008, the first nationwide survey on the portion of the ICA with development of
management of pregnancy and delivery in moyamoya vessels.
858 A. Yamamoto et al.

2. Diagnosis of moyamoya disease should be Neurosurg Psychiatry 79(8):900–904, PubMed


done with conventional angiography; how- PMID: 18077479. Epub 2007/12/14. eng
8. Yamauchi T, Houkin K, Tada M, Abe H (1997) Famil-
ever, MRI/MRA can become a substitute for ial occurrence of moyamoya disease. Clin Neurol
it in some situations. Neurosurg 99(Suppl 2):S162–S167, PubMed PMID:
3. Bypass surgery is the essential strategy for 9409429. Epub 1997/12/31. eng
ischemic moyamoya disease. 9. Nanba R, Kuroda S, Tada M, Ishikawa T, Houkin K,
Iwasaki Y (2006) Clinical features of familial
4. Recent randomized controlled trial has moyamoya disease. Child’s Nerv Syst 22
revealed the effectiveness of “direct” bypass (3):258–262, PubMed PMID: 16195874. Epub
surgery. 2005/10/01. eng
5. Although most pregnant patients known to 10. Houkin K, Yoshimoto T, Abe H, Nagashima K,
Nagashima M, Takeda M et al (1998) Role of basic
have moyamoya disease can deliver safely, fibroblast growth factor in the pathogenesis of
the risk of pregnancy and delivery with moyamoya disease. Neurosurg Focus 5(5):e2,
moyamoya disease should not be PubMed PMID: 17112205. Epub 2006/11/23. eng
underestimated. 11. Mineharu Y, Takenaka K, Yamakawa H, Inoue K,
Ikeda H, Kikuta KI et al (2006) Inheritance pattern
of familial moyamoya disease: autosomal dominant
mode and genomic imprinting. J Neurol Neurosurg
Psychiatry 77(9):1025–1029, PubMed PMID:
References 16788009. Pubmed Central PMCID: PMC2077755.
Epub 2006/06/22. eng
1. Suzuki J, Takaku A (1969) Cerebrovascular 12. Kamada F, Aoki Y, Narisawa A, Abe Y,
“moyamoya” disease. Disease showing abnormal Komatsuzaki S, Kikuchi A et al (2011) A genome-
net-like vessels in base of brain. Arch Neurol 20 wide association study identifies RNF213 as the first
(3):288–299, PubMed PMID: 5775283 moyamoya disease gene. J Hum Genet 56(1):34–40,
2. Miyamoto S, Yoshimoto T, Hashimoto N, Okada Y, PubMed PMID: 21048783. Epub 2010/11/05. eng
Tsuji I, Tominaga T et al (2014) Effects of 13. Liu W, Morito D, Takashima S, Mineharu Y,
extracranial-intracranial bypass for patients with hem- Kobayashi H, Hitomi T et al (2011) Identification of
orrhagic moyamoya disease: results of the Japan RNF213 as a susceptibility gene for moyamoya dis-
Adult Moyamoya Trial. Stroke 45(5):1415–1421, ease and its possible role in vascular development.
PubMed PMID: 24668203. Eng PLoS One 6(7):e22542, PubMed PMID: 21799892.
3. Research Committee on the Pathology and Treatment Pubmed Central PMCID: PMC3140517. Epub 2011/
of Spontaneous Occlusion of the Circle of Willis, 07/30. eng
Health Labour Sciences Research Grant for Research 14. Houkin K, Ito M, Sugiyama T, Shichinohe H,
on Measures for Infractable Diseases, Hashimoto N Nakayama N, Kazumata K et al (2012) Review
et al (2012) Guidelines for diagnosis and treatment of of past research and current concepts on the etiology
moyamoya disease (spontaneous occlusion of the cir- of moyamoya disease. Neurol Med Chir 52
cle of Willis). Neurol Med Chir ;52(5):245–266. (5):267–277, PubMed PMID: 22688062. Epub
PubMed PMID: 22870528. Epub 2012/08/09. eng 2012/06/13. eng
4. Wakai K, Tamakoshi A, Ikezaki K, Fukui M, 15. Kuroda S, Hashimoto N, Yoshimoto T, Iwasaki Y
Kawamura T, Aoki R et al (1997) Epidemiological (2007) Radiological findings, clinical course, and
features of moyamoya disease in Japan: findings from outcome in asymptomatic moyamoya disease: results
a nationwide survey. Clin Neurol Neurosurg 99(Suppl of multicenter survey in Japan. Stroke 38
2):S1–S5, PubMed PMID: 9409395. Epub 1997/12/ (5):1430–1435, PubMed PMID: 17395863. Epub
31. eng 2007/03/31. eng
5. Kleinloog R, Regli L, Rinkel GJ, Klijn CJ (2012) 16. Kuroda S (2010) Asymptomatic moyamoya disease.
Regional differences in incidence and patient charac- In: Cho B-K, Tominaga T (eds) Moyamoya disease
teristics of moyamoya disease: a systematic review. update. Springer, Tokyo, pp 336–340
J Neurol Neurosurg Psychiatry 83(5):531–536, 17. Kuroda S, Ishikawa T, Houkin K, Nanba R,
PubMed PMID: 22378916. Epub 2012/03/02. eng Hokari M, Iwasaki Y (2005) Incidence and clinical
6. Kuriyama S, Kusaka Y, Fujimura M, Wakai K, features of disease progression in adult moyamoya
Tamakoshi A, Hashimoto S et al (2008) Prevalence disease. Stroke 36(10):2148–2153, PubMed PMID:
and clinicoepidemiological features of moyamoya 16179571. Epub 2005/09/24. eng
disease in Japan: findings from a nationwide epide- 18. Sawada T, Yamamoto A, Miki Y, Kikuta K, Okada T,
miological survey. Stroke 39(1):42–47, PubMed Kanagaki M et al (2012) Diagnosis of moyamoya
PMID: 18048855. Epub 2007/12/01. eng disease using 3-T MRI and MRA: value of cisternal
7. Baba T, Houkin K, Kuroda S (2008) Novel epidemi- moyamoya vessels. Neuroradiology 54
ological features of moyamoya disease. J Neurol
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 859

(10):1089–1097, PubMed PMID: 22349623. Epub (4):454–460, PubMed PMID: 3760953. Epub 1986/
2012/02/22 10/01. eng
19. Fukui M (1997) Guidelines for the diagnosis and 31. Kuroda S, Houkin K, Ishikawa T, Nakayama N,
treatment of spontaneous occlusion of the circle of Ikeda J, Ishii N et al (2004) Determinants of intellec-
Willis (‘moyamoya’ disease). Research Committee tual outcome after surgical revascularization in pedi-
on Spontaneous Occlusion of the Circle of Willis atric moyamoya disease: a multivariate analysis.
(Moyamoya Disease) of the Ministry of Health and Child’s Nerv Syst 20(5):302–308, PubMed PMID:
Welfare, Japan. Clin Neurol Neurosurg 99(Suppl 2): 15045517. Epub 2004/03/27. eng
S238–S240, PubMed PMID: 9409446. Epub 1997/ 32. Kodama N, Aoki Y, Hiraga H, Wada T, Suzuki J
12/31 (1979) Electroencephalographic findings in
20. Houkin K, Abe H, Yoshimoto T, Takahashi A (1996) children with moyamoya disease. Arch Neurol 36
Is “unilateral” moyamoya disease different from (1):16–19, PubMed PMID: 420596. Epub 1979/01/
moyamoya disease? J Neurosurg 85(5):772–776, 01. eng
PubMed PMID: 8893713. Epub 1996/11/01. eng 33. Kuroda S, Kamiyama H, Isobe M, Houkin K, Abe H,
21. Kawano T, Fukui M, Hashimoto N, Yonekawa Y Mitsumori K (1995) Cerebral hemodynamics and “re-
(1994) Follow-up study of patients with “unilateral” build-up” phenomenon on electroencephalogram in
moyamoya disease. Neurol Med Chir 34 children with moyamoya disease. Child’s Nerv Syst
(11):744–747, PubMed PMID: 7533268. Epub 11(4):214–219, PubMed PMID: 7621482. Epub
1994/11/01. eng 1995/04/01. eng
22. Kelly ME, Bell-Stephens TE, Marks MP, Do HM, 34. Irikura K, Miyasaka Y, Kurata A, Tanaka R, Fujii K,
Steinberg GK (2006) Progression of unilateral Yada K et al (1996) A source of haemorrhage in adult
moyamoya disease: a clinical series. Cerebrovasc patients with moyamoya disease: the significance of
Dis (Basel, Switzerland) 22(2–3):109–115, PubMed tributaries from the choroidal artery. Acta Neurochir
PMID: 16685122. Epub 2006/05/11. eng 138(11):1282–1286, PubMed PMID: 8980730. Epub
23. Cheong HI, Choi Y (2010) Systemic arterial involve- 1996/01/01. eng
ment in moyamoya disease. In: Cho B-K, Tominaga T 35. Kuroda S, Houkin K, Kamiyama H, Abe H (2001)
(eds) Moyamoya disease update. Springer, Tokyo, pp Effects of surgical revascularization on peripheral
126–131 artery aneurysms in moyamoya disease: report of
24. Fukui M, Kono S, Sueishi K, Ikezaki K (2000) three cases. Neurosurgery 49(2):463–467, PubMed
Moyamoya disease. Neuropathology 20(Suppl): PMID: 11504126. discussion 7–8. Epub 2001/08/16.
S61–S64, PubMed PMID: 11037190. Epub 2000/ eng
10/19. eng 36. Kawaguchi S, Sakaki T, Morimoto T, Kakizaki T,
25. Oka K, Yamashita M, Sadoshima S, Tanaka K (1981) Kamada K (1996) Characteristics of intracranial
Cerebral haemorrhage in moyamoya disease at aneurysms associated with moyamoya disease. A
autopsy. Virchows Arch A Pathol Anat Histol 392 review of 111 cases. Acta Neurochir 138
(3):247–261, PubMed PMID: 7269227. Epub 1981/ (11):1287–1294, PubMed PMID: 8980731. Epub
01/01. eng 1996/01/01. eng
26. Takagi Y, Kikuta K, Sadamasa N, Nozaki K, Hashi- 37. Osanai T, Kuroda S, Nakayama N, Yamauchi T,
moto N (2006) Caspase-3-dependent apoptosis in Houkin K, Iwasaki Y (2008) Moyamoya disease
middle cerebral arteries in patients with moyamoya presenting with subarachnoid hemorrhage localized
disease. Neurosurgery 59(4):894–900, discussion -1. over the frontal cortex: case report. Surg Neurol 69
PubMed PMID: 17038954. Epub 2006/10/14. eng (2):197–200, PubMed PMID: 18261650. Epub 2008/
27. Yamashita M, Oka K, Tanaka K (1983) Histopathol- 02/12. eng
ogy of the brain vascular network in moyamoya dis- 38. Verret S, Steele JC (1971) Alternating hemiplegia in
ease. Stroke 14(1):50–58, PubMed PMID: 6823686. childhood: a report of eight patients with complicated
Epub 1983/01/01. eng migraine beginning in infancy. Pediatrics 47
28. Kuroda S, Houkin K (2008) Moyamoya disease: cur- (4):675–680, PubMed PMID: 5089756. Epub 1971/
rent concepts and future perspectives. Lancet Neurol 04/01. eng
7(11):1056–1066, PubMed PMID: 18940695. Epub 39. Shirane R, Fujimura M (2010) Headache in
2008/10/23. eng moyamoya disease. In: Cho B-K, Tominaga T (eds)
29. Karasawa J, Touho H, Ohnishi H, Miyamoto S, Moyamoya disease update. Springer, Tokyo,
Kikuchi H (1993) Cerebral revascularization using pp 110–113
omental transplantation for childhood moyamoya dis- 40. Nogawa S, Suzuki N (2010) Involuntary movement.
ease. J Neurosurg 79(2):192–196, PubMed PMID: In: Cho B-K, Tominaga T (eds) Moyamoya disease
8331399. Epub 1993/08/01. eng update. Springer, Tokyo, pp 126–131
30. Miyamoto S, Kikuchi H, Karasawa J, Nagata I, 41. Gonzalez-Alegre P, Ammache Z, Davis PH,
Ihara I, Yamagata S (1986) Study of the posterior Rodnitzky RL (2003) Moyamoya-induced paroxys-
circulation in moyamoya disease. Part 2: visual dis- mal dyskinesia. Mov Disord 18(9):1051–1056,
turbances and surgical treatment. J Neurosurg 65 PubMed PMID: 14502675. Epub 2003/09/23. eng
860 A. Yamamoto et al.

42. Nishimoto A, Takeuchi S (1968) Abnormal cerebro- 53. Mori N, Miki Y, Kikuta K, Fushimi Y, Okada T,
vascular network related to the internal carotid arter- Urayama S et al (2008) Microbleeds in moyamoya
ies. J Neurosurg 29(3):255–260, PubMed PMID: disease: susceptibility-weighted imaging versus T2-
5684407. Epub 1968/09/01. eng weighted imaging at 3 Tesla. Invest Radiol 43
43. Houkin K, Iihoshi S, Mikami T (2010) Overview of (8):574–579, PubMed PMID: 18648257. Epub
image diagnosis of moyamoya disease. In: Cho B-K, 2008/07/24. eng
Tominaga T (eds) Moyamoya disease update. 54. Kikuta K, Takagi Y, Nozaki K, Hanakawa T, Okada T,
Springer, Tokyo, pp 141–149 Mikuni N et al (2005) Asymptomatic microbleeds in
44. Sugino T, Mikami T, Ohtaki S, Hirano T, Iihoshi S, moyamoya disease: T2-weighted gradient-echo
Houkin K et al (2013) Assessment of moyamoya magnetic resonance imaging study. J Neurosurg 102
disease using multidetector row computed tomogra- (3):470–475, PubMed PMID: 15796381. Epub 2005/
phy. J Stroke Cerebrovasc Dis 22(5):644–649, 03/31
PubMed PMID: 22365710. Epub 2012/03/01. eng 55. Horie N, Morikawa M, Nozaki A, Hayashi K,
45. Houkin K, Nakayama N, Kuroda S, Nonaka T, Suyama K, Nagata I (2011) “Brush Sign” on
Shonai T, Yoshimoto T (2005) Novel magnetic reso- susceptibility-weighted MR imaging indicates the
nance angiography stage grading for moyamoya dis- severity of moyamoya disease. AJNR Am J
ease. Cerebrovasc Dis 20(5):347–354, PubMed Neuroradiol 32(9):1697–1702, PubMed PMID:
PMID: 16131804. Epub 2005/09/01. eng 21799039. Epub 2011/07/30. eng
46. Maeda M, Tsuchida C (1999) “Ivy sign” on fluid- 56. Hayashi K, Nagata I (2010) Progression of
attenuated inversion-recovery images in childhood moyamoya disease. In: Cho B-K, Tominaga T (eds)
moyamoya disease. AJNR Am J Neuroradiol 20 Moyamoya disease update. Springer, Tokyo,
(10):1836–1838, PubMed PMID: 10588105. Epub pp 118–125
1999/12/10 57. Suzuki J, Kodama N (1983) Moyamoya disease – a
47. Toyoda K, Ida M, Fukuda K (2001) Fluid-attenuated review. Stroke 14(1):104–109, PubMed PMID:
inversion recovery intraarterial signal: an early sign of 6823678. Epub 1983/01/01. eng
hyperacute cerebral ischemia. AJNR Am J 58. Kodama N, Suzuki J (1978) Moyamoya disease asso-
Neuroradiol 22(6):1021–1029, PubMed PMID: ciated with aneurysm. J Neurosurg 48(4):565–569,
11415892. Epub 2001/06/21. eng PubMed PMID: 632880. Epub 1978/04/01. eng
48. Takanashi J (2011) Moyamoya disease in children. 59. Currie S, Raghavan A, Batty R, Connolly DJ,
Brain Dev 33(3):229–234, PubMed PMID: Griffiths PD (2011) Childhood moyamoya disease
20932697. Epub 2010/10/12. eng and moyamoya syndrome: a pictorial review. Pediatr
49. Kawashima M, Noguchi T, Takase Y, Ootsuka T, Neurol 44(6):401–413, PubMed PMID: 21555050.
Kido N, Matsushima T (2009) Unilateral hemispheric Epub 2011/05/11
proliferation of ivy sign on fluid-attenuated inversion 60. Maki Y, Enomoto T (1988) Moyamoya disease.
recovery images in moyamoya disease correlates Child’s Nerv Syst 4(4):204–212, PubMed PMID:
highly with ipsilateral hemispheric decrease of cere- 3048657. Epub 1988/08/01
brovascular reserve. AJNR Am J Neuroradiol 30 61. Yamada I, Suzuki S, Matsushima Y (1995)
(9):1709–1716, PubMed PMID: 19713323. Epub Moyamoya disease: comparison of assessment with
2009/08/29. eng MR angiography and MR imaging versus conven-
50. Kawashima M, Noguchi T, Takase Y, Nakahara Y, tional angiography. Radiology 196(1):211–218,
Matsushima T (2010) Decrease in leptomeningeal ivy PubMed PMID: 7784569. Epub 1995/07/01. eng
sign on fluid-attenuated inversion recovery images 62. Tsuchiya K, Makita K, Furui S (1994) Moyamoya
after cerebral revascularization in patients with disease: diagnosis with three-dimensional CT angiog-
moyamoya disease. AJNR Am J Neuroradiol 31 raphy. Neuroradiology 36(6):432–434, PubMed
(9):1713–1718, PubMed PMID: 20466798. Epub PMID: 7991085. Epub 1994/08/01
2010/05/15. eng 63. Kikuchi M, Asato M, Sugahara S, Nakajima K,
51. Mori N, Mugikura S, Higano S, Kaneta T, Sato M, Nagao K et al (1996) Evaluation of surgically
Fujimura M, Umetsu A et al (2009) The formed collateral circulation in moyamoya
leptomeningeal “ivy sign” on fluid-attenuated inver- disease with 3D-CT angiography: comparison with
sion recovery MR imaging in moyamoya disease: a MR angiography and X-ray angiography.
sign of decreased cerebral vascular reserve? AJNR Neuropediatrics 27(1):45–49, PubMed PMID:
Am J Neuroradiol 30(5):930–935, PubMed PMID: 8677026. Epub 1996/02/01
19246527. Epub 2009/02/28. eng 64. Ogasawara K, Ogawa A, Yoshimoto T (2002) Cere-
52. Fushimi Y, Miki Y, Kikuta K, Okada T, Kanagaki M, brovascular reactivity to acetazolamide and outcome
Yamamoto A et al (2006) Comparison of 3.0- and 1.5- in patients with symptomatic internal carotid or mid-
T three-dimensional time-of-flight MR angiography dle cerebral artery occlusion: a xenon-133 single-
in moyamoya disease: preliminary experience. Radi- photon emission computed tomography study. Stroke
ology 239(1):232–237, PubMed PMID: 16467209. 33(7):1857–1862, PubMed PMID: 12105366. Epub
Epub 2006/02/10. eng 2002/07/10
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 861

65. Nariai T, Matsushima Y, Imae S, Tanaka Y, Ishii K, Kaku Igaku 49(2):49–58, PubMed PMID: 22779347.
Senda M et al (2005) Severe haemodynamic stress in Epub 2012/07/12
selected subtypes of patients with moyamoya disease: 74. Hatazawa J, Iida H, Shimosegawa E, Sato T,
a positron emission tomography study. J Neurol Murakami M, Miura Y (1997) Regional cerebral
Neurosurg Psychiatry 76(5):663–669, PubMed blood flow measurement with iodine-123-IMP auto-
PMID: 15834024. Pubmed Central PMCID: radiography: normal values, reproducibility and sen-
1739646. Epub 2005/04/19. eng sitivity to hypoperfusion. J Nucl Med 38
66. Czabanka M, Pena-Tapia P, Schubert GA, Heppner (7):1102–1108, PubMed PMID: 9225798. Epub
FL, Martus P, Horn P et al (2011) Proposal for a new 1997/07/01
grading of moyamoya disease in adult patients. 75. Iida H, Itoh H, Nakazawa M, Hatazawa J,
Cerebrovasc Dis (Basel, Switzerland) 32(1):41–50, Nishimura H, Onishi Y et al (1994) Quantitative map-
PubMed PMID: 21576942. Epub 2011/05/18 ping of regional cerebral blood flow using iodine-
67. Tanaka Y, Nariai T, Nagaoka T, Akimoto H, 123-IMP and SPECT. J Nucl Med 35
Ishiwata K, Ishii K et al (2006) Quantitative evalua- (12):2019–2030, PubMed PMID: 7989987. Epub
tion of cerebral hemodynamics in patients with 1994/12/01
moyamoya disease by dynamic susceptibility contrast 76. Kang KH, Kim HS, Kim SY (2008) Quantitative
magnetic resonance imaging–comparison with posi- cerebrovascular reserve measured by acetazolamide-
tron emission tomography. J Cereb Blood Flow challenged dynamic CT perfusion in ischemic adult
Metab 26(2):291–300, PubMed PMID: 16049427. moyamoya disease: initial experience with angio-
Epub 2005/07/29 graphic correlation. AJNR Am J Neuroradiol 29
68. Ikezaki K, Matsushima T, Kuwabara Y, Suzuki SO, (8):1487–1493, PubMed PMID: 18499785. Epub
Nomura T, Fukui M (1994) Cerebral circulation and 2008/05/24
oxygen metabolism in childhood moyamoya disease: 77. Rim NJ, Kim HS, Shin YS, Kim SY (2008) Which
a perioperative positron emission tomography study. CT perfusion parameter best reflects cerebrovascular
J Neurosurg 81(6):843–850, PubMed PMID: reserve?: correlation of acetazolamide-challenged CT
7965114. Epub 1994/12/01 perfusion with single-photon emission CT in
69. Kanno I, Uemura K, Higano S, Murakami M, Iida H, Moyamoya patients. AJNR Am J Neuroradiol 29
Miura S et al (1988) Oxygen extraction fraction at (9):1658–1663, PubMed PMID: 18617583. Epub
maximally vasodilated tissue in the ischemic brain 2008/07/12
estimated from the regional CO2 responsiveness mea- 78. Sakamoto S, Ohba S, Shibukawa M, Kiura Y, Arita K,
sured by positron emission tomography. J Cereb Kurisu K (2006) CT perfusion imaging for childhood
Blood Flow Metab 8(2):227–235, PubMed PMID: moyamoya disease before and after surgical revascu-
3125186. Epub 1988/04/01 larization. Acta Neurochir 148(1):77–81, discussion
70. Saito N, Nakagawara J, Nakamura H, Teramoto A PubMed PMID: 16184319. Epub 2005/09/27
(2004) Assessment of cerebral hemodynamics in 79. Wintermark M, Maeder P, Verdun FR, Thiran JP,
childhood moyamoya disease using a quantitative Valley JF, Schnyder P et al (2000) Using 80 kVp
and a semiquantitative IMP-SPECT study. Ann Nucl versus 120 kVp in perfusion CT measurement of
Med 18(4):323–331, PubMed PMID: 15359926. regional cerebral blood flow. AJNR Am J Neuroradiol
Epub 2004/09/14 21(10):1881–1884, PubMed PMID: 11110541. Epub
71. So Y, Lee HY, Kim SK, Lee JS, Wang KC, Cho BK 2000/12/08
et al (2005) Prediction of the clinical outcome of 80. Kudo K, Christensen S, Sasaki M, Ostergaard L,
pediatric moyamoya disease with postoperative Shirato H, Ogasawara K et al (2013) Accuracy and
basal/acetazolamide stress brain perfusion SPECT reliability assessment of CT and MR perfusion anal-
after revascularization surgery. Stroke 36 ysis software using a digital phantom. Radiology 267
(7):1485–1489, PubMed PMID: 15947261. Epub (1):201–211, PubMed PMID: 23220899. Pubmed
2005/06/11 Central PMCID: 3606546. Epub 2012/12/12
72. Touho H, Karasawa J, Ohnishi H (1996) Preoperative 81. Zhang J, Wang J, Geng D, Li Y, Song D, Gu Y (2013)
and postoperative evaluation of cerebral perfusion Whole-brain CT perfusion and CT angiography
and vasodilatory capacity with 99mTc-HMPAO assessment of moyamoya disease before and after
SPECT and acetazolamide in childhood Moyamoya surgical revascularization: preliminary study with
disease. Stroke 27(2):282–289, PubMed PMID: 256-slice CT. PLoS One 8(2):e57595, PubMed
8571424. Epub 1996/02/01 PMID: 23451248. Pubmed Central PMCID:
73. Iida H, Nakagawara J, Hatazawa J, Suzuki M, 3579776. Epub 2013/03/02
Takahashi M, Shimosegawa E et al (2012) Estimation 82. Morkenborg J, Pedersen M, Jensen FT, Stodkilde-
of the arterial blood radioactivity concentration using Jorgensen H, Djurhuus JC, Frokiaer J (2003) Quanti-
the whole body-to-arterial blood partition coefficients tative assessment of Gd-DTPA contrast agent from
and the cross-calibration factor in 123I- signal enhancement: an in-vitro study. Magn Reson
iodoamphetamine SPECT – towards a noninvasive Imaging 21(6):637–643, PubMed PMID: 12915195.
clinical protocol with the QSPECT-DTARG method. Epub 2003/08/14
862 A. Yamamoto et al.

83. Nardone B, Saddleton E, Laumann AE, Edwards BJ, 93. Hwang JW, Yang HM, Lee H, Lee HK, Jeon YT, Kim
Raisch DW, McKoy JM et al (2014) Pediatric JE et al (2013) Predictive factors of symptomatic
nephrogenic systemic fibrosis is rarely reported: a cerebral hyperperfusion after superficial temporal
RADAR report. Pediatr Radiol 44(2):173–180, artery-middle cerebral artery anastomosis in adult
PubMed PMID: 24057195. Epub 2013/09/24 patients with moyamoya disease. Br J Anaesth 110
84. Detre JA, Subramanian VH, Mitchell MD, Smith DS, (5):773–779, PubMed PMID: 23274781. Epub 2013/
Kobayashi A, Zaman A et al (1990) Measurement of 01/01
regional cerebral blood flow in cat brain using 94. Ng J, Thompson D, Lumley JP, Saunders DE,
intracarotid 2H2O and 2H NMR imaging. Magn Ganesan V (2012) Surgical revascularisation for
Reson Med 14(2):389–395, PubMed PMID: childhood moyamoya. Child’s Nerv Syst 28
2345518. Epub 1990/05/01 (7):1041–1048, PubMed PMID: 22570164. Epub
85. Kuwabara Y, Ichiya Y, Sasaki M, Yoshida T, 2012/05/10
Masuda K, Matsushima T et al (1997) Response to 95. Cai X, Han S, Feske SK, Chou SH (2013) Pearls and
hypercapnia in moyamoya disease. Cerebrovascular oy-sters: small but consequential: intracerebral hem-
response to hypercapnia in pediatric and adult patients orrhage caused by lenticulostriate artery aneurysm.
with moyamoya disease. Stroke 28(4):701–707, Neurology 80(9):e89–e91, PubMed PMID:
PubMed PMID: 9099182. Epub 1997/04/01 23439707. Pubmed Central PMCID: 3598457. Epub
86. Aoyama K, Fushimi Y, Okada T, Miyasaki A, Taki H, 2013/02/27
Shibamoto K et al (2012) Detection of symptomatic 96. Gotoh K, Okada T, Satogami N, Yakami M,
vasospasm after subarachnoid haemorrhage: initial Takahashi JC, Yoshida K et al (2012) Evaluation of
findings from single time-point and serial measure- CT angiography for visualisation of the
ments with arterial spin labelling. Eur Radiol 22 lenticulostriate artery: difference between normoten-
(11):2382–2391, PubMed PMID: 22653285. Epub sive and hypertensive patients. Br J Radiol 85(1019):
2012/06/02 e1004–e1008, PubMed PMID: 22744324. Pubmed
87. Saida T, Masumoto T, Nakai Y, Shiigai M, Central PMCID: 3500797. Epub 2012/06/30
Matsumura A, Minami M (2012) Moyamoya disease: 97. Gotoh K, Okada T, Miki Y, Ikedo M, Ninomiya A,
evaluation of postoperative revascularization using Kamae T et al (2009) Visualization of the
multiphase selective arterial spin labeling MRI. J lenticulostriate artery with flow-sensitive black-
Comput Assist Tomogr 36(1):143–149, PubMed blood acquisition in comparison with time-of-flight
PMID: 22261785. Epub 2012/01/21. eng MR angiography. J Magn Reson Imaging 29
88. Noguchi T, Kawashima M, Irie H, Ootsuka T, (1):65–69, PubMed PMID: 19097113. Epub 2008/
Nishihara M, Matsushima T et al (2011) Arterial 12/20
spin-labeling MR imaging in moyamoya disease 98. Okuchi S, Okada T, Ihara M, Gotoh K, Kido A,
compared with SPECT imaging. Eur J Radiol 80(3): Fujimoto K et al (2013) Visualization of
e557–e562, PubMed PMID: 21315533. Epub 2011/ lenticulostriate arteries by flow-sensitive black-blood
02/15. eng MR angiography on a 1.5 T MRI system: a compar-
89. Zaharchuk G, Do HM, Marks MP, Rosenberg J, ative study between subjects with and without stroke.
Moseley ME, Steinberg GK (2011) Arterial spin- AJNR Am J Neuroradiol 34(4):780–784, PubMed
labeling MRI can identify the presence and intensity PMID: 23064597. Epub 2012/10/16
of collateral perfusion in patients with moyamoya 99. Kimura T, Ikedo M, Takemoto S (2009) Hybrid of
disease. Stroke 42(9):2485–2491, PubMed PMID: opposite-contrast MR angiography
21799169. Pubmed Central PMCID: 3164217. Epub (HOP-MRA) combining time-of-flight and flow-
2011/07/30. eng sensitive black-blood contrasts. Magn Reson Med
90. Lee HY, Paeng JC, Lee DS, Lee JS, Oh CW, Cho MJ 62(2):450–458, PubMed PMID: 19526496. Epub
et al (2004) Efficacy assessment of cerebral arterial 2009/06/16. eng
bypass surgery using statistical parametric mapping 100. Tsuchiya K, Kobayashi K, Nitatori T, Kimura T,
and probabilistic brain atlas on basal/acetazolamide Ikedo M, Takemoto S (2010) Hybrid of opposite-
brain perfusion SPECT. J Nucl Med 45(2):202–206, contrast MRA of the brain by combining time-of-
PubMed PMID: 14960636. Epub 2004/02/13. eng flight and black-blood sequences: initial experience
91. Sugino T, Mikami T, Miyata K, Suzuki K, Houkin K, in major trunk stenoocclusive diseases. J Magn Reson
Mikuni N (2013) Arterial spin-labeling magnetic res- Imaging 31(1):56–60, PubMed PMID: 20027573.
onance imaging after revascularization of moyamoya Epub 2009/12/23. eng
disease. J Stroke Cerebrovasc Dis 22(6):811–816, 101. Miyamoto S, Akiyama Y, Nagata I, Karasawa J,
PubMed PMID: 22721824. Epub 2012/06/23 Nozaki K, Hashimoto N et al (1998) Long-term out-
92. Zhao WG, Luo Q, Jia JB, Yu JL (2013) Cerebral come after STA-MCA anastomosis for moyamoya
hyperperfusion syndrome after revascularization sur- disease. Neurosurg Focus 5(5):e5, PubMed PMID:
gery in patients with moyamoya disease. Br J 17112208
Neurosurg 27(3):321–325, PubMed PMID: 102. Nakagawara J, Takeda R, Suematsu K, Nakamura J
23461748. Epub 2013/03/07 (1997) Quantification of regional cerebral blood flow
36 Moyamoya Disease (Spontaneous Occlusion of the Circle of Willis) 863

and vascular reserve in childhood moyamoya disease 112. Nishimoto A (1979) Moyamoya disease (author’s
using [123I]IMP-ARG method. Clin Neurol transl). Neurol Med Chir 19(3):221–228, PubMed
Neurosurg 99(Suppl 2):S96–S99, PubMed PMID: PMID: 86959
9409415. Epub 1997/12/31. eng 113. Yoshida Y, Yoshimoto T, Shirane R, Sakurai Y (1999)
103. Research Comittee on Spontaneous Occlusion of the Clinical course, surgical management, and long-term
Circle of Willis (Moyamoya Disease) (2009) outcome of moyamoya patients with rebleeding after
Research on intractable diseases of the Ministry of an episode of intracerebral hemorrhage: an extensive
Health LaW, Japan. Recommendations for the Man- follow-up study. Stroke 30(11):2272–2276, PubMed
agement of Moyamoya Disease. A Statement from PMID: 10548656
Research Committee on Spontaneous Occlusion of 114. Takahashi JC, Miyamoto S (2010) Moyamoya dis-
the Circle of Willis (Moyamoya Disease). Surg ease: recent progress and outlook. Neurol Med Chir
Cereb Stroke 37(5):321–337. Japanese 50(9):824–832, PubMed PMID: 20885116
104. Karasawa J, Kikuchi H, Furuse S, Kawamura J, 115. Nishimoto A, Ueda K, Honma Y (eds) (1982)
Sakaki T (1978) Treatment of moyamoya disease Follow-up study on outcome of the occlusion of
with STA-MCA anastomosis. J Neurosurg 49 the circle of Willis. Proceedings of the Research
(5):679–688, PubMed PMID: 712390 Committee on Spontaneous Occlusion of the
105. Miyamoto S, Kikuchi H, Karasawa J, Nagata I, Circle of Willis. Ministry of Health and Welfare,
Yamazoe N, Akiyama Y (1988) Pitfalls in the surgical Tokyo
treatment of moyamoya disease. Operative tech- 116. Kobayashi E, Saeki N, Oishi H, Hirai S, Yamaura A
niques for refractory cases. J Neurosurg 68 (2000) Long-term natural history of hemorrhagic
(4):537–543, PubMed PMID: 3351581 moyamoya disease in 42 patients. J Neurosurg 93
106. Karasawa J, Kikuchi H, Furuse S, Sakaki T, Yoshida (6):976–980, PubMed PMID: 11117870
Y (1977) A surgical treatment of “moyamoya” dis- 117. Houkin K, Kamiyama H, Abe H, Takahashi A,
ease “encephalo-myo synangiosis”. Neurol Med Chir Kuroda S (1996) Surgical therapy for adult
17(1 Pt 1):29–37, PubMed PMID: 74029. Epub 1977/ moyamoya disease. Can surgical revascularization
01/01. eng prevent the recurrence of intracerebral hemorrhage?
107. Takeuchi S, Abe H, Ozawa T, Tanaka R (2000) Sur- Stroke 27(8):1342–1346, PubMed PMID: 8711799.
gical treatment for moyamoya disease. Surgical effect Epub 1996/08/01. eng
of encephalo-galeo-synangiosis on moyamoya dis- 118. Kawaguchi S, Okuno S, Sakaki T (2000) Effect of
ease. Surg Cereb Stroke 28:98–103, Japanese direct arterial bypass on the prevention of future
108. Matsushima Y, Fukai N, Tanaka K, Tsuruoka S, stroke in patients with the hemorrhagic variety of
Inaba Y, Aoyagi M et al (1981) A new surgical treat- moyamoya disease. J Neurosurg 93(3):397–401,
ment of moyamoya disease in children: a preliminary PubMed PMID: 10969936
report. Surg Neurol 15(4):313–320, PubMed PMID: 119. Fujii K, Ikezaki K, Irikura K, Miyasaka Y, Fukui M
7245020 (1997) The efficacy of bypass surgery for the patients
109. Karasawa J, Kikuchi H, Kawamura J, Sakai T (1980) with hemorrhagic moyamoya disease. Clin Neurol
Intracranial transplantation of the omentum for cere- Neurosurg 99(Suppl 2):S194–S195, PubMed PMID:
brovascular moyamoya disease: a two-year follow-up 9409436
study. Surg Neurol 14(6):444–449, PubMed PMID: 120. Miyamoto S (2004) Study design for a prospective
7221855 randomized trial of extracranial-intracranial bypass
110. Nakagawa Y, Abe H, Kamiyama H, Sawamura Y, surgery for adults with moyamoya disease and hem-
Gotoh S, Kashiwaba T (1988) Revascularization sur- orrhagic onset–the Japan Adult Moyamoya Trial
gery for 50 patients with moyamoya disease. In: Group. Neurol Med Chir 44(4):218–219, PubMed
Suzuki J (ed) Advances in surgery for cerebral stroke. PMID: 15185765. Epub 2004/06/10. eng
Springer, Tokyo, pp 141–149 121. Wiebers DO (1985) Ischemic cerebrovascular com-
111. Fujimura M, Kaneta T, Mugikura S, Shimizu H, plications of pregnancy. Arch Neurol 42
Tominaga T (2007) Temporary neurologic deteriora- (11):1106–1113, PubMed PMID: 2864911
tion due to cerebral hyperperfusion after superficial 122. Takahashi JC, Ikeda T, Iihara K, Miyamoto S (2012)
temporal artery-middle cerebral artery anastomosis in Pregnancy and delivery in moyamoya disease: results
patients with adult-onset moyamoya disease. Surg of a nationwide survey in Japan. Neurol Med Chir
Neurol 67(3):273–282, PubMed PMID: 17320638. 52(5):304–310, PubMed PMID: 22688067
Epub 2007/02/27. eng
Brain Death Imaging
37
M. Sawicki, Joanna Wojczal, Bozena Birkenfeld, and
Lech Cyrylowski

Contents Prerequisites for Blood Flow Studies


in the Diagnosis of Brain Death . . . . . . . . . . . . . . . . . . . . . 872
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 866 Catheter Cerebral Angiography . . . . . . . . . . . . . . . . . . . . . 872
The Role of Imaging Tests in Determination CT Angiography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 874
of Brain Death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 867 Perfusion CT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 878
Xenon CT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 880
Pathophysiological Mechanism of Cerebral MR Angiography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 881
Circulatory Arrest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 867 Transcranial Doppler Sonography . . . . . . . . . . . . . . . . . . . 881
Nuclear Medicine Methods in the Diagnosis
Imaging of Structural Changes in
of Brain Death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 886
Brain Death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 869
CT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 869 Limitations of Blood Flow Studies
MRI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 869 in the Diagnosis of Brain Death . . . . . . . . . . . . . . . . . . . 889
Functional Imaging of Brain Death . . . . . . . . . . . . . . . 871 Future Trends: Looking for an Ideal
Ancillary Test . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 889
Imaging of Cerebral Circulatory Arrest . . . . . . . . . . 872
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 892
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 893

M. Sawicki (*)
Department of Diagnostic Imaging and Interventional
Radiology, Pomeranian Medical University, Szczecin,
Poland
e-mail: msaw@pum.edu.pl; msaw3108@gmail.com
J. Wojczal
Department of Neurology, Medical University of Lublin,
Lublin, Poland
e-mail: jwojczal@poczta.onet.pl
B. Birkenfeld
Department of Nuclear Medicine, Pomeranian Medical
University, Szczecin, Poland
e-mail: birka@pum.edu.pl
L. Cyrylowski
CT and MRI Clinic, “Nasz Doktor” Health Care
Institution, Szczecin, Poland
MRI Clinic, “Euromedic” Health Care Institution,
Szczecin, Poland
e-mail: blecyr@hotmail.com

# Springer Science+Business Media New York 2016 865


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_26
866 M. Sawicki et al.

spectroscopy • Diffusion-weighted MRI •


Abstract
Functional MRI
Brain death is defined as the irreversible cessa-
tion of functioning of the entire brain, includ-
ing the brain stem. Brain death is principally
Introduction
established using clinical criteria including
coma, absence of brain stem reflexes, and loss
The concept of death is an important part of med-
of central drive to breathe, assessed with apnea
icine since the beginning of its history. In modern
test. In situations in which clinical testing can-
medicine, the death of a human is identified with
not be performed or when uncertainty exists
the death of his/her brain. The earliest description
about the reliability of its parts due to
of brain death was “coma dépassé” (beyond
confounding conditions, ancillary tests (i.e.,
coma) by Mollaret and Goulon in 1959. The first
imaging studies) may be useful. The objective
formalized definition of brain death referred to as
of ancillary tests in the diagnosis of brain death
irreversible coma was by the Ad Hoc Committee
is to demonstrate the absence of cerebral elec-
of the Harvard Medical School in 1968. The def-
trical activity (EEG and evoked potentials) or
inition has evolved over the next years, since there
cerebral circulatory arrest. In clinical practice
has been a greater understanding of neuronal
catheter cerebral angiography, perfusion scin-
function and mechanisms of brain cell injury.
tigraphy, transcranial Doppler sonography, CT
Nowadays, brain death is defined as the irrevers-
angiography, and MR angiography are used.
ible cessation of functioning of the entire brain,
Other methods, like perfusion CT, xenon CT,
including the brain stem with two pathways to
MR spectroscopy, diffusion-weighted MRI,
reach that point: (i) permanent absence of circula-
and functional MRI, are being studied as
tion and (ii) subsequent to a catastrophic brain
potentially useful in the diagnosis of brain
injury [1].
death.
The definition and diagnosis of brain death are
Among these tests, blood flow and perfu-
of particular importance in view of the medical
sion studies are most commonly applied. The
and technological advances in supporting,
diagnosis of brain death can be confirmed
repairing, or replacing failing organs. The confir-
when a lack of blood flow or tissue perfusion
mation of brain death, synonymous with the dec-
in the whole brain is documented.
laration of death, is a necessary condition for the
However, the application of blood flow and
termination of futile life support and potential
perfusion tests to the diagnosis of brain death is
procurement of organs for transplantation. There-
limited by variable vulnerability of individual
fore, diagnosis of brain death is one of the main
brain structures to ischemia. Besides there are
points of interest in intensive and critical care and
known factors predisposing to dissociation
transplantation medicine.
between brain death and total cessation of cere-
Brain death is principally established using
bral circulation. These factors include skull
clinical criteria defined as diagnostic testing
decompression, infratentorial localization,
based on direct, measurable observation or exam-
and hypoxic-ischemic mechanism of brain
ination of the patient. Because the clinical criteria
injury.
vary in detail between countries, the minimum
acceptable clinical standard has been agreed to
Keywords
test for the cessation of brain function [1] – see
Brain death • Cerebral circulatory arrest •
Table 1.
Ancillary test • Catheter angiography • Com-
To confirm brain death according to clinical
puted tomographic angiography • Perfusion
criteria, prerequisites should be fulfilled prior to
CT • Xenon CT • Transcranial Doppler sonog-
the application of the diagnostic tests – see
raphy • Perfusion scintigraphy • Magnetic res-
Table 2. Most countries mandate that the clinical
onance imaging • MR angiography • MR
37 Brain Death Imaging 867

Table 1 Minimum acceptable clinical standard for the Table 3 Confounding conditions that may impede or
cessation of brain function invalidate the clinical diagnosis of brain death
1. Coma (excluding spinal cord-mediated reflexes) 1. Trauma to the eyes
2. Absence of brain stem reflexes 2. Trauma to the middle and/or inner ears
(a) Pupils mid-position or greater and absent pupillary 3. Nerve or muscle dysfunction or neuromuscular
light reflex (fixed dilated pupils) blockade potentially accounting for unresponsiveness
(b) Corneal 4. Presence of atypical responses
(c) Gag/pharyngeal 5. Unresuscitated shock
(d) Cough/tracheal 6. Acquired or therapeutic hypothermia
(e) Vestibulo-ocular (“cold caloric”) 7. Severe metabolic and endocrine abnormality that is
(f) Loss of central drive to breathe, assessed with apnea difficult to reverse (e.g., glucose, acid-base, electrolytes)
test 8. Clinically significant drug intoxications (e.g.,
alcohol, barbiturates, sedatives, hypnotics)
9. Inability to perform apnea test
10. High cervical spine injury
Table 2 Preconditions: patient-related prerequisites that 11. Primary infratentorial brain injury
should be fulfilled prior to application of diagnostic tests 12. Young children
1. Established etiology and/or structural lesion capable of
causing death by neurological criteriaa
2. Reduced consciousness (as measured by GCS 3–5) brain death may allow for avoidance of protracted
3. Evidence for progressing loss of brain stem function stays in the intensive care unit (ICU) and poten-
a
Neuroimaging studies are useful for assessing the severity tially expedite organ donation before tissue via-
of brain injury
bility becomes a concern. In some protocols,
ancillary tests are used to shorten the duration of
diagnosis be confirmed by more than one physi- the observation period.
cian, either two or three. The objective of ancillary tests in the diagnosis
of brain death is to demonstrate the absence of
cerebral electrical activity (EEG and evoked
The Role of Imaging Tests potentials) or cerebral circulatory arrest. Cur-
in Determination of Brain Death rently, only catheter cerebral angiography and
perfusion scintigraphy are widely accepted as
There are known circumstances during which a ancillary tests in the determination of cerebral
clinical examination to confirm brain death may circulatory arrest. However, in clinical practice
become unreliable or cannot be completely transcranial Doppler sonography (TCD), CT angi-
performed. These confounding conditions that ography (CTA), and MR angiography (MRA) are
may impede or invalidate clinical diagnosis of used as well. Other methods, like perfusion CT
brain death are listed in Table 3. (PCT), xenon CT (XeCT), MR spectroscopy
In situations in which clinical testing cannot be (MRS), diffusion-weighted MRI (DWI), and
performed or when uncertainty exists about the functional MRI (fMRI), are being studied as
reliability of its parts due to confounding condi- potentially useful in the diagnosis of brain death.
tions, ancillary tests (i.e., imaging studies) may be
useful. Ancillary tests are applied as complemen-
tary to clinical testing that otherwise, for any Pathophysiological Mechanism
reason, cannot be conducted or is unreliable. Fur- of Cerebral Circulatory Arrest
thermore, the usage of ancillary tests can reduce
the time taken to confirm the diagnosis and Cerebral blood flow (CBF) is continuously regu-
increase its confidence in all cases of suspected lated by cerebral perfusion pressure (CPP) and
brain death regardless of the presence of cerebrovascular resistance (CVR). CPP is mea-
confounding conditions. Early determination of sured as a difference between the entry pressure,
868 M. Sawicki et al.

mean arterial pressure (MAP), and the exit pres- spreads proximally progressively involving big-
sure – intracranial pressure (ICP). According to ger arteries. As ICP reaches the diastolic pressure,
the Poiseuille equation: the end-diastolic cerebral flow completely ceases.
At this stage CBF is deeply compromised,
CPP MAP  ICP although still sufficient to preserve a viability of
CBF ¼ ¼ :
CVR CVR the brain. When ICP exceeds the diastolic pres-
sure approaching MAP, the circulatory arrest
The normal CBF at rest is maintained within a occurs in the compressed cerebral capillaries and
range of 45–60 ml/100 g of brain tissue/min. A venules. At this time larger arteries stay patent,
brain injury is accompanied by cerebral and their elastic walls are distended by the blood
edema. Because the skull bones limit the intracra- pushed forward by the heartbeat in systolic phase
nial volume, this “mass effect” leads to the (antegrade flow). In diastole a similar blood vol-
intracranial hypertension. The process spreads ume is pressed out of the collapsed arteries (retro-
from the region of primary injury gradually grade flow). Such an oscillating, to-and-fro
involving the whole brain, and the intracranial movement of the blood provides the zero-net
vessels become compressed but preserve their CBF. This is an early phase of cerebral circulatory
patency. Initially, CVR is kept almost constant arrest leading to necrosis of neurons within 10–15
by the activated autoregulation mechanisms min. With further ICP elevation, the oscillations
releasing blood vessel tone. However, this phe- of blood column in the proximal arteries become
nomenon is capable to preserve a sufficient blood weaker. If ICP exceeds the level of systolic pres-
supply as long as CPP exceeds approx. 50 mmHg sure, a blood movement in the cerebral vessels
– see Fig. 1. ceases completely, while oscillation can be still
Further elevation of ICP results in an increase observed in extracranial segments of the internal
of CVR. The capillaries as the most susceptible carotid arteries (ICAs) and vertebral arteries
vessels are primarily affected; then the process (VAs).

Fig. 1 Relationship between cerebral blood flow (CBF) capable to preserve almost constant CBF as long as CPP
and cerebral perfusion pressure (CPP). CPP is measured as exceeds approx. 50 mmHg. Further elevation of ICP
a difference between the entry pressure, mean arterial results in deterioration of cerebral blood flow leading to
pressure (MAP), and the exit pressure, intracranial pressure cerebral circulatory arrest
(ICP). In increasing ICP the cerebral autoregulation is
37 Brain Death Imaging 869

Imaging of Structural Changes in Common MRI findings in echo-planar scans


Brain Death are variable edema, including diffuse gyral swell-
ing, most prominent in the acute phase, diffuse
CT cerebral white matter injury, diffuse cortical high
signal intensity, and tonsillar herniation
Non-contrast-enhanced CT (NECT) of the head is [4–6]. Some authors observed also transtentorial
useful for assessing the severity of brain injury in herniation [4, 7, 8]. Less frequent finding is focal
the suspicion of brain death. The study may reveal edema in the upper spinal cord [5].
the features of cerebral edema including a gener- Poor gray/white matter differentiation after the
alized hypodensity of brain parenchyma, diagnosis of brain death is not commonly
decreased differentiation between the gray and observed. Matsumura et al. [8] found significant
white matter (GM:WM ratio), sulcal effacement, increasing of the ratio of signal intensities
absence of the basal cisterns, and signs of brain between white and gray matters on PD- and
herniation – see Fig. 2. It was shown that a midline T2-WI images but decreased on T1-WI images.
shift >10 mm, absence of the ambient cistern, and These phenomena may be related to different vul-
GM:WM ratio <1.18 predict progression to brain nerabilities of white and gray matter to anoxic
death. Although the study is ubiquitous, noninva- injury – see Fig. 3a, c.
sive, non-time-consuming, and non-cost consum- They also showed the absence of normal flow
ing, it lacks sufficient diagnostic value as an voids in MR echo-planar images. Moreover, with
ancillary test. serial MR before and after the diagnosis of brain
death, they demonstrated disappearance of flow
voids in cavernous portions of the ICAs after the
MRI diagnosis. The disappearance of flow voids in the
ICA correlates with nonfilling of the ICA revealed
The advent of magnetic resonance imaging has with intra-arterial digital subtraction arteriogra-
improved diagnostic possibilities in brain death, due phy [8]. Sohn et al. [9] using 3-T MRI demon-
to several sophisticated techniques offered by MRI. strated that tonsillar herniation and loss of intra-
In the early 1990s, first reports demonstrated arterial flow signal voids on T2-WI are highly
the potential role of MRI in the diagnosis of brain sensitive and specific findings in patients with
death, including MR spectroscopy [2, 3]. brain death.

Fig. 2 CT findings in a
23-year-old man after
cardiac arrest who
presented with clinical signs
of brain death: (a, b) 5 mm
MPRs in axial plane show
diffuse edema, sulcal
effacement, ventricular
narrowing, low GM/WM
differentiation, and
diminution of the basal
cisterns
870 M. Sawicki et al.

Fig. 3 MR findings in a 42-year-old man after cardiac 5,000 ms, TE = 86 ms) shows restricted diffusion in
arrest who presented with clinical signs of brain death. lentiform nuclei and cortex. Cortical and deep GM are
First examination performed several hours after resuscita- more frequently affected because neurons are more vulner-
tion: (a) 5 mm axial T2-WI image (TR = 3,713 ms, TE = able to hypoxia than oligodendroglia or astrocytes; (e, f)
104 ms) shows moderate increase in signal of lentiform SWI (TR = 73 ms, TE = 47 ms) shows multiple and
nuclei consistent with edema; (b) DWI (TR = 5,000 ms, branching low signal intensities extending through the
TE = 86 ms) shows restricted diffusion in lentiform nuclei. cerebral hemisphere parallel or perpendicular to the outer
Second examination performed the next day: (c) 5 mm wall of both lateral ventricles (arrowhead, transcerebral
axial T2-WI image (TR = 3,713 ms, TE = 104 ms) vein sign) and abnormal low signal intensities in both
shows massive edema involving the basal ganglia and cerebral hemisphere cortical areas (arrows, bilateral corti-
cortex with narrowing of ventricles; (d) DWI (TR = cal vein sign)

Additional signs include the bilateral Diffusion-Weighted Imaging


transcerebral vein sign, defined as branching Lövblad et al. first demonstrated the usefulness of
structures extending through cerebral hemi- diffusion-weighted images (DWI) in the diagnosis
spheres parallel or perpendicular to the outer of brain death. They found a significant drop in the
wall of a lateral ventricle, and the bilateral cortical apparent diffusion coefficient (ADC) values com-
vein sign, defined as visualization of cortical veins pared with those of normal controls, and these obser-
of cerebral hemispheres. Both signs can be easily vations have been later confirmed by other authors
demonstrated with T2*WI gradient-recalled echo [7, 10, 11]. With DWI and ADC mapping, it is
(GRE) and susceptibility-weighted images (SWI) possible to identify areas corresponding to cytotoxic
[9] – see Fig. 3e, f. edema and ischemic damage – see Fig. 3b, d.
37 Brain Death Imaging 871

Selcuk et al. [11] observed greater decrease in To our best knowledge, no other study on
ADC values in white matter than in gray application of MRS in human brain death is
matter for both the cerebral and cerebellar available.
hemispheres. The sensitivity and specificity as
well as positive and negative predictive
cutoff values of ADC to distinguish patients Functional Imaging of Brain Death
with brain death from controls were 100 % in
their study, but they recommend to The future perspectives of imaging of the brain in
determine ADC cutoff values for different comatose patients may be linked to functional
MRI scanners and DWI protocols. They MRI (fMRI) with blood oxygen level-dependent
believe that a 100 % sensitivity and specificity in (BOLD) signal. In 2009 Boly et al. [13] first
their results indicates a potential role of ADC reported that fMRI showed no significant func-
maps as confirmatory test in diagnosis of brain tional connectivity in a brain death patient in
death. contrary to a vegetative state patient with pre-
However, Luchtmann et al. evaluating the served cortico-cortical connectivity. In 2010
reliability of DWI in the diagnosis of brain Monti et al. [14] successfully elaborated new
death concluded that effects of pseudonorma- methodology, which can be applied in fMRI in a
lization of ADC values (after 5–28 days after patient without conscience. With imagery tasks
brain ischemic damage) and possible susceptibil- they were able to establish functional and interac-
ity artifacts due to microhemorrhages preclude a tive communication with a minority of patients
leading role of DWI in the diagnosis of brain who met the behavioral criteria for a vegetative
death [10]. state after serious brain injury, detecting residual
cognitive function in them. For this reason fMRI
MR Spectroscopy probably will provide a method for detecting min-
Other sophisticated technique offered by MRI is imal cognitive ability in patients without a behav-
MR spectroscopy (MRS), allowing to study ioral response and could be considered a
metabolic changes in brain tissue. In previously complementary tool for existing diagnostic
used method of 31P MRS, the most striking methods. Hence, it can be believed that fMRI will
feature was a complete absence of adenosine increase significantly our understanding of con-
triphosphate (ATP) and domination of intense science disorders in patients with severe brain dam-
inorganic phosphate signal [2, 3]. Kato et al. [3] age and differentiate with the highest accuracy
observed also an absence of phosphocreatine and brain death from a vegetative state or chronic coma.
stated that MRS may be helpful in diagnostics of From the theoretical point of view, MRI might
brain death particularly in children, when other seem an ideal confirmatory test in brain-dead
examinations are not sufficient to establish patients because it is able to prove the cessation
defined diagnosis. of cerebral blood flow (MRA) and to demonstrate
Falini et al. [12] performing sequential MR the irreversible loss of cerebral bioelectrical activ-
imaging and proton (1H) MRS in a patient ity (fMRI). However, practical application of MRI
with severe hypoxic-ischemic brain injury in these patients is limited because transportation
found that observed structural and and monitoring of ventilated patients to an MRI
biochemical changes reflected the known evolu- scanner can be challenging, and the procedure is
tion of brain degeneration after asphyxial time-consuming. On the other hand, in chosen
injury, particularly the massive decrease of corti- cases MRI may add certainty to the diagnosis of
cal N-acetylaspartate in the acute phase, brain death in addition to clinical findings. Further
suggesting the severity of the neuronal damage, extensive studies are needed to establish a poten-
and the subsequent progressive increase of cho- tial role of MRI techniques, including MRA,
line related to the delayed degeneration of white DWI, MRS, and fMRI, in the diagnostic algo-
matter. rithm for brain death.
872 M. Sawicki et al.

Imaging of Cerebral Circulatory Arrest The pendulum movement of the contrast is


accompanied by its slow antegrade propagation
Prerequisites for Blood Flow Studies and intracranial stasis. The contrast is gradually
in the Diagnosis of Brain Death eliminated from the intracranial vessels through
the arteriovenous shunts and the interrupted
It is recommended that blood flow studies be blood-brain barrier. Noteworthy the superior sag-
performed when it is impossible to complete the ittal sinus and the transverse sinuses may fill from
clinical criteria defined in Table 1. Before these the extracranial vessels through the emissary
tests are performed, at least two particular clinical veins. With further increasing ICP, the contrast
criteria must be met: column is stopped in more proximal arterial seg-
ments. A total intracranial nonfilling occurs when
1. An established etiology capable of causing the ICP reaches systolic blood pressure.
neurological death in the absence of reversible
conditions capable of mimicking neurological Technique of Catheter Cerebral
death Angiography in the Diagnosis of Brain
2. Deep unresponsive coma Death
Catheter cerebral angiography for the determination
Prior to the testing, unresuscitated shock and of brain death is performed with an injection of
hypothermia must be corrected. iodinated contrast material to reach both anterior
The blood flow studies are not confounded by and posterior cerebral circulation. The propagation
drugs or metabolic disorders, so they are reliable of contrast is assessed in a filming sequence acquired
in cases of barbiturate overdose. with the usage of DSA technique. The duration of
the acquisition varies among national guidelines, but
it should not be shorter than 15 s to confirm cerebral
Catheter Cerebral Angiography circulatory arrest (personal experience).
The national guidelines permit two different
Catheter cerebral angiography is considered as a techniques of contrast injection:
reference method in the diagnosis of majority of
cerebrovascular pathologies including cerebral 1. Aortocervical arteriography – injection from
circulatory arrest. For the diagnosis of brain death, the aortic arch
it was used as the first imaging modality in 1953. 2. Selective 4-vessel cerebral angiography,
The study involves a follow-up of iodinated performed with a selective catheterization of
contrast dynamics after its intravascular injection both ICAs or the common carotid arteries
on time-resolved images made with the usage of (CCAs) and VAs
the digital subtraction technique (DSA).
While performing the selective angiography,
Catheter Cerebral Angiography Findings one must take into account that the contrast
in Intracranial Hypertension and Brain injected under high pressure into the ICA or VA
Death may be pushed forward to intracranial arteries
At raising ICP the cerebral arteries and veins fill resulting in an artifactual intracranial filling. This
with contrast with increasing delay in comparison can be avoided if the injection from the aortic arch
to extracranial vessels of the head. At the early is applied. Moreover, the selective technique
stage of cerebral circulatory arrest, nonfilling of requires higher manual skills and experience
the cortical arterial branches and the deep cerebral than aortic arch angiography. Finally, the risk of
veins (the internal cerebral vein, ICV; the great procedure-related complications is higher for
cerebral vein, GCV; and the straight sinus) occurs. selective technique. Because both techniques are
At this stage an oscillation of contrast column in characterized by equally high accuracy, the aortic
the proximal intracranial arteries may be detected. arch angiography is a preferred option.
37 Brain Death Imaging 873

Fig. 4 Catheter angiography findings in a 74-year-old slow propagation of contrast is visible in extracranial parts
woman after traumatic brain injury who presented with of the ICAs (arrowheads) and the VAs (arrows); (c) 15 s
clinical signs of brain death. Aortic arch injection of after injection contrast reaches cavernous segments of the
30 ml of contrast at flow rate of 15 ml/s. (a) 3 s after ICAs (arrowheads). Venous outflow through the external
injection branches of the external carotid arteries are filled jugular veins is observed (arrows). No intracranial filling is
(empty arrows), while filling of the internal carotid arteries noted. These findings are consistent with the diagnosis of
(arrowheads) and the vertebral arteries (arrows) is limited brain death
to their proximal extracranial parts; (b) 10 s after injection

Intravenous angiography can be applied in 1. Filling of external carotid arteries confirming a


the determination of cerebral circulatory proper contrast delivery
arrest as well [15]. However, when performing 2. No filling of ICAs beyond the level of the
the exam with i.v. injection, one must consider anterior clinoid process
the jugular and dural venous sinus reflux of 3. No filling of VAs beyond their dural
contrast. Such finding is not indicative of pre- penetration
served cerebral circulation [15]. Another disad- 4. No filling of the internal cerebral veins [16]
vantage of this technique is a low concentration (see Fig. 4)
of contrast achieved in the head and neck vessels
due to the effect of dilution. This could make the However, delayed opacification of proximal
detection of possible intracranial opacification segments of the cerebral arteries with contrast
equivocal. Therefore arterial injection is stasis and without deep venous filling does not
preferred. preclude the diagnosis of brain death – see Fig. 5.

Criteria for the Diagnosis of Brain Death Usefulness of Catheter Cerebral


in Catheter Cerebral Angiography Angiography in the Diagnosis of Brain
Cerebral circulatory arrest can be documented by Death
catheter cerebral angiography if all of the follow- Catheter angiography is widely accepted as a
ing conditions are met: reference test in the diagnosis of brain death.
874 M. Sawicki et al.

Fig. 5 Catheter angiography findings in an 18-year-old (arrowhead) is limited to its proximal extracranial part;
man after traumatic brain injury who presented with clin- (b) 12 s after injection stasis of contrast is visible in the
ical signs of brain death. Selective injection of 8 ml of M1 segment of the middle cerebral artery (arrowhead) and
contrast at flow rate of 6 ml/s to the left common carotid A1 and A2 segments of the anterior cerebral artery (arrow).
artery. (a) 3 s after injection branches of the external carotid No intracranial venous outflow is noted. These findings are
artery are filled (arrow), while filling of the ICA consistent with the diagnosis of brain death

Despite this several disadvantages limit its applica- reports of preserved blood flow in the posterior
tion. The catheterization of aortic arch, especially fossa with cerebral circulatory arrest in the anterior
ICAs and VAs, requires high expertise. The exam circulation [15, 18]. This points to the protective
must be performed in the angiographic lab; thus, function of the cerebellar tentorium, which causes
transportation of a critically ill patient is necessary. an uneven increase in ICP. In all false-negative
The procedure is invasive, accompanied by the risk exams, patients met all clinical criteria of brain
of serious complications, such as an injury of a death, and in all cases intracranial circulation was
catheterized vessel or vasospasm. Besides catheter markedly slowed.
angiography is the most costly procedure among
the blood flow tests. Potential risk of a damage of
transplantable organs caused by iodinated contrast, CT Angiography
although postulated, has not been confirmed yet.
Moreover, it was revealed that contrast medium CTA for the determination of brain death was
administration to donors does not affect kidney proposed in 1998 [19]. Despite the fact that tech-
graft function after transplant [17]. No false- nical protocol or evaluation criteria have not been
positive results of catheter angiography in the diag- standardized yet, CTA is considered the most
nosis of brain death have been reported (i.e., angi- promising ancillary test.
ography revealed cerebral circulatory arrest, yet the
patient survived), which means the 100 % specific- CT Angiography Findings in Brain Death
ity. The sensitivity is close to 100 % as well. There Unfortunately CTA findings were not studied in
are casuistic reports of false-negative results in non-brain-dead patients with intracranial hyper-
cases in which residual preservation or restoration tension. The earliest sign of cerebral circulatory
of intracranial blood flow was observed after ICP arrest in CTA is a lack of opacification of deep
was relieved through some decompressive mecha- veins – the ICVs and the GCV. The sensitivity of
nism, e.g., craniectomy or skull fractures (see this finding in the diagnosis of brain death in CTA
“Limitations” section) [15]. There are several is 98–100 % [19–23]. A lack of opacification of
37 Brain Death Imaging 875

Fig. 6 CTA findings in a 50-year-old man with traumatic noted. Opacification of the superficial temporal artery
brain injury and right-sided craniectomy presented with (empty arrow) indicates that the contrast was injected
signs of brain death: 10 mm MIP in coronal plane (a) and properly. In some cases of brain death, the stasis of contrast
VRT (b) with a delay of 60 s after injection show in the posterior circulation may be observed due to the
opacification of the basilar artery (arrows). No filling of protective function of cerebellar tentorium
the anterior cerebral circulation or deep venous outflow is

Fig. 7 CTA findings in a 22-year-old woman with a brain VRT shows opacification of the right pericallosal artery
stem ischemic stroke and a right-sided craniectomy who (arrow). No filling of the basilar artery or deep venous
presented with signs of brain death on clinical examina- outflow is noted. In some cases of brain death, the stasis
tion. (a) 10 mm MIP in coronal plane with a delay of 60 s of contrast in the pericallosal arteries may be observed
after injection shows opacification of M1 and M2 segments because of higher CPP in these arteries in comparison to
of the middle cerebral arteries (arrowheads) and A1 seg- cortical branches of the middle cerebral arteries
ments of the anterior cerebral arteries (arrow); (b) sagittal

cortical branches of the middle cerebral arteries sensitivity is 85–94 % and 79 %, respectively [19,
(MCA-M4) is slightly less sensitive indicator of 21]. The least sensitive finding of brain death is a
brain death with the sensitivity of 86–100 % [19, lack of opacification of cortical branches of the
21, 22]. The basilar artery (BA) and cortical anterior cerebral arteries (ACA-A3, the
branches of the posterior cerebral arteries pericallosal arteries) with the sensitivity of 64 %
(PCA-P2) are more frequently opacified in brain- [21] – see Fig. 7. Such a sequence is explained by
dead patients than MCA-M4 – see Fig. 6. Their the highest susceptibility of cortical branches of
876 M. Sawicki et al.

Fig. 8 CTA findings consistent with the diagnosis of brain shows slow propagation of contrast to the level of petrous
death: (a) VRT 20 s after contrast injection shows filling of segment of the right ICA and cavernous segment of the left
the ICAs (arrows) and the VAs (arrowheads) limited to ICA (arrows). Vertebral arteries are opacified to the level
their proximal extracranial parts. Opacification of extracra- of foramen magnum, before their dural penetration (arrow-
nial arteries (empty arrows) indicates that the contrast was heads). Venous outflow through external jugular veins is
injected properly; (b) VRT 60 s after contrast injection observed (empty arrows). No intracranial filling was noted

the MCA to intracranial hypertension and the 1. Non-enhanced scanning as a reference for
lowest CPP in these arteries. In the course of assessing vascular opacification.
cerebral circulatory arrest, proximal segments of 2. Early post-contrast scanning for assessing
the cerebral arteries frequently show delayed intra- and, more important, extracranial vascu-
opacification for some time. These vessels appear lar opacification. This phase is started approx.
as thin and faint. The intracranial stasis of contrast 20 s after the beginning of injection.
is observed with its extremely slow elimination Opacification of branches of external carotid
through the arteriovenous shunts and extravasa- arteries – the superficial temporal or the facial
tion due to the interrupted blood-brain barrier. The arteries – indicates that the contrast was
latest sign of cerebral circulatory arrest is intracra- injected correctly, and there are no hemody-
nial nonfilling – see Fig. 8. namic abnormalities causing a delay of con-
trast delivery to the vessels of head and neck.
Technique of CT Angiography To ensure scanning is performed during the
in the Diagnosis of Brain Death phase of optimal contrast enhancement, sev-
The technique of CTA involves a rapid, intrave- eral strategies can be applied. A fixed delay is
nous administration of iodinated contrast in the the easiest but least favorable option since it
bolus at a flow rate of 3–5 ml/s. Volume of con- does not adjust for individual variations in
trast medium used for CTA for the diagnosis of contrast dynamics. Two more advanced strate-
cerebral circulatory arrest is 60–120 ml. The con- gies, test bolus injection and bolus tracking
trast can be followed by the infusion of 30–40 ml technique, can be applied to tailor the contrast
of normal saline at the same rate (i.e., saline flush) injection to the individual patient.
to push the contrast medium forward and to opti- 3. Late post-contrast scanning for assessing intra-
mize contrast enhancement. cranial vascular opacification. This phase is
For the diagnosis of brain death, at least three started 60 s after the beginning of injection,
acquisitions should be performed: with a delay of 40 s to the early phase. The
37 Brain Death Imaging 877

necessity of performing the late phase of CTA Table 4 Criteria for the diagnosis of brain death in CT
in the diagnosis of cerebral circulatory arrest angiography
is motivated by a possible delayed Sensitivity
intracranial opacification in increased ICP. Ter- Criteria Lack of opacification of (%)
mination of the study on the early phase may Intracranial ICA beyond the level of 86–92a
nonfilling the anterior clinoid
result in a failure to recognize delayed intra- [24, 25] process
cranial filling producing a false-positive result. VA beyond their dural
penetration
Intermediate post-contrast scans may be added ICV, GCV, and the
between the early and the late phase for a better straight sinus
follow-up of the contrast dynamics. 10 point [26] BA 54–70
Right and left PCA-P2
Practical Tips Right and left ACA-A3
(pericallosal artery)
The superficial temporal artery (STA) is routinely
Right and left MCA-M4
used for assessing sufficient contrast delivery. But Right and left ICV
in some cases, both STAs may be surgically GCV
ligated during bilateral temporal craniectomy. In 7 point Right and left ACA-A3 52–100
such cases the facial artery can be used as a good [19–21, 27] (pericallosal artery)
alternative. Right and left MCA-M4
In patients with suspected brain death, CT Right and left ICV
images often show subarachnoid hemorrhage GCV
(SAH) or high-attenuation areas along the basal 4 point [21, Right and left 86–97
cisterns and cortical sulci, mimicking SAH. The 22] MCA-M4b
Right and left ICV
latter is described as pseudo-SAH phenomenon,
which is a synergistic result of distention of In the 10-, 7-, and 4-point scale, one point is noted for each
nonopacified vessel in the late phase. Cerebral circulatory
congested superficial veins due to elevated intra- arrest is diagnosed with the score 10, 7, or 4 points
cranial pressure and severe brain edema. Both true accordingly
a
SAH and pseudo-SAH impede the assessment of Based on the opacification in the early phase
b
intracranial filling considering that cerebral ves- According to the 4-point scale, opacification of 1 or 2 cor-
tical branches of the MCA on the same side does not
sels in intracranial hypertension are usually abnor- exclude the diagnosis of cerebral circulatory arrest, pro-
mally thin and faintly opacified. Therefore vided a lack of opacification of the ICVs
cerebral vessels should be assessed on pre- and
post-contrast scans.
Another potential difficulty associated with Usefulness of CT Angiography
CTA assessment is a consequence of frequently in the Diagnosis of Brain Death
found midline shift and marked ventricular dis- CTA is far more widely available than any other
placement. This may cause problems with identi- blood flow test. The test is noninvasive, techni-
fication of the dislocated deep cerebral veins on cally uncomplicated, and non-time-consuming.
routinely used axial images. These vessels can be However, CTA cannot be applied at the bedside,
far easier localized on sagittal maximum intensity while a transport of the ICU patient is always
projections (MIPs). insecure. Potential risk of a damage of transplant-
able organs caused by iodinated contrast was
Criteria for the Diagnosis of Brain Death addressed above. The accuracy of CTA in the
in CT Angiography diagnosis of brain death has not been reliably
There are no widely accepted criteria of CTA in studied yet. In particular, specificity of the method
the diagnosis of brain death. Four methods of has not been evaluated with non-brain-dead
evaluation have been proposed so far; they are patients with intracranial hypertension as
summarized in Table 4 and Fig. 9. controls. However, there are no reports of the
878 M. Sawicki et al.

Fig. 9 Evaluation criteria of CTA in the diagnosis of diagnosis of brain death; (b) in the 7-point scale, positive
cerebral circulatory arrest. A point is given for each vessel result (score = 7) is recorded with a lack of opacification of
without opacification: (a) positive result in the 10-point the bilateral ACA-A3, the bilateral MCA-M4, the bilateral
scale (score = 10) confirming the diagnosis of brain death ICV, and the GCV. Scores from 0 to 6 are classified as
is recorded when the following vessels are not opacified, negative results; (c) positive result in the 4-point scale
the bilateral PCA-P2, the BA, the bilateral ACA-A3, the (score = 4) is recorded when the bilateral MCA-M4 and
bilateral MCA-M4, the bilateral ICV, and the GCV. Scores the bilateral ICV are not opacified. Scores from 0 to 3 are
from 0 to 9 are classified as negative results excluding the classified as negative results

false-positive CTAs involving a multiphase scan- injury. The diagnosis of brain death is a new
ning. Skull decompression, primary infratentorial application of PCT [28, 29].
process, and hypoxic-ischemic injury after car- PCT studies are obtained by monitoring the
diac arrest are predisposing factors to false- first pass of an iodinated contrast agent through
negative results of CTA (see “Limitations” sec- the cerebral vasculature. Changes in tissue atten-
tion) [21, 25, 27, 28]. uation that occur in the brain after the contrast
injection are measured. Post-processing of the
PCT data allows the generation of color-coded
Perfusion CT maps of various perfusion parameters, including
cerebral blood flow (CBF), cerebral blood volume
PCT is commonly used to detect brain ischemia in (CBV), mean transit time (MTT), and the time to
different clinical conditions, i.e., stroke, TIA, vas- peak (TTP) – the time from the start of contrast
culitis, vasospasm, carotid stenosis, and traumatic injection to the time of maximum enhancement.
37 Brain Death Imaging 879

Perfusion CT Findings in Brain Death superior sagittal sinus or torcular Herophili.


The normal mean CBF is 45–60 ml/100 g/min. In some brain-dead patients, filling of the intra-
This value is approx. 50 % higher for the gray cranial veins may be insufficient to provide a
matter and 50 % lower for the white matter. The proper VOF. Poor placement of venous out-
cerebral autoregulation mechanisms maintain the flow region can result in an overestimation of
blood flow at almost constant level, provided the CBV and CBF.
CPP exceeds 50 mmHg. When an elevation of the Delay of arterial inflow and venous outflow
ICP is high enough to cause a decrease of CPP PCT protocols are usually designed for
below 50 mmHg, CBF gradually decreases. With acquisition duration of 40–45 s, principally to
CBF 20 ml/100 g/min, disturbances of neuronal minimize radiation dose. However, in brain-
electrical function develop. Neurological symp- dead patients, intracranial time-attenuation
toms occur at this stage as a result of physiologic curve (TAC), if detectable, is flat and low. In
paralysis of neurons and EEG becomes flatlined. such cases the standard series would not cover
In such conditions near-normal concentrations of the entire TAC. This may preclude calculation
extracellular potassium and tissue ATP are of diagnostic PCT maps.
detected that reflect preservation of energy metab- Scan range
olism and ion pumping thereby viability of neu- In the diagnosis of cerebral circulatory
rons. This state of ischemia is potentially arrest, the scanned volume must include the
reversible if the normal blood flow is restored whole brain. Standard CT scanners (16–64
within several hours. CBF 10 ml/100 g/min detector rows) cover 2–9,6 cm in z-axis,
causes a failure of energy metabolism and ion which is not enough for the diagnosis of
pumping represented by an increase of extracel- whole-brain death. Such a limitation does not
lular potassium and diminishing of tissue ATP apply to cutting-edge scanners with 128–320
concentration. This is consistent with cerebral detector rows offering the possibility of
circulatory arrest leading to necrosis of neurons 9,6–16 cm coverage with acceptable temporal
within several minutes. It should be emphasized resolution.
that there are significant differences in CBF
between the gray and white matter as well as Criteria for the Diagnosis of Brain Death
differences in vulnerability to ischemia between in Perfusion CT
individual brain structures. The threshold values of PCT parameters for brain
infarct are well known. It is widely accepted that
Technique of Perfusion CT CBF <10 ml/100 g/min, CBV <2 ml/100 g, and
in the Diagnosis of Brain Death MTT >8 s are insufficient to preserve metabolic
Several technical issues need to be resolved when processes of neurons and lead to neuronal necrosis
performing PCT for the diagnosis of brain death: if maintained above 5–8 min – see Fig. 10. How-
ever, the PCT diagnostic criteria for cerebral cir-
Arterial input function (AIF) selection culatory arrest have not been established yet. An
To ensure the accuracy of results, one needs extrapolation of the values for stroke on the deter-
to select an artery with early and high contrast mination of brain death may not be appropriate
enhancement. This may be impossible in some because of the technical issues listed above.
brain-dead patients without detectable intracra-
nial filling. Wrong selection of AIF may result Usefulness of Perfusion CT
in an overestimation of MTT and an underes- in the Diagnosis of Brain Death
timation of CBF. The major advantage of PCT is the ability to
Venous output function (VOF) selection quantify a capillary blood flow, which is a key
AIF is normalized with respect to the factor in the diagnosis of brain death. Angio-
venous output function. The venous region of graphic tests and TCD are capable to show the
interest (ROI) is usually placed over the blood flow in large and medium vessels, while it
880 M. Sawicki et al.

Fig. 10 Perfusion CT findings in a patient presenting (arrow), and extracranial part of the ICA, proximal to the
clinical signs of brain death: (a) CBF map in axial plane petrous part (empty arrow). Reduction of blood flow below
and (b) CBF map in sagittal plane shows blood flow 10 ml/100 g/min is noted in an entire volume of the brain
limited to torcular Herophili (arrowhead), sigmoid sinus consistent with neuronal necrosis

does not always correspond with true blood sup- diffuses rapidly into the brain. Diffusion into the
ply to the brain parenchyma. The cerebral arterio- brain is determined by blood flow and solubility
venous shunts and the phenomenon of ineffective of the Xe within different brain compartments.
reperfusion in hypoxic-ischemic brain injury after Patients inhale a mixture of 26–32 % Xe mixed
cardiac arrest are known examples of such dis- with O2 over a period of approximately 4 min.
crepancy. The test is ubiquitous, non-time-con- Processing of the data allows generation of CBF
suming, not costly, and noninvasive. maps, from which quantitative data can be
On the other hand, the study cannot be extracted.
performed at the bedside, and the unsafe transport It is known that CBF values less than
of the ICU patient is necessary. Besides this, sev- 10 ml/100 g/min correlate with eventual neuronal
eral principal technical issues (mentioned above) necrosis. CBF between 10 and 20 ml/100 g/min
must be resolved before the wide implementation has been associated with potentially reversible
of PCT as an ancillary test. The post-processing of neurological deficits. It was shown that CBF <5
PCT data is not as simple as in angiographic ml/100 g/min is required in order to confirm the
methods and requires expertise in interpretation diagnosis of brain death [30, 31]. However, there
of perfusion studies. Finally, the test is not stan- is insufficient evidence to determine the accuracy
dardized, and there are no reliable case-control of XeCT as an ancillary test in the determination
studies assessing its diagnostic value. of brain death.
The method is fast and easy to perform and
simple in interpretation, and the accessory
Xenon CT equipment for commercial CT scanner is inexpen-
sive. A minor drawback is that XeCT quantifies
Xenon (Xe) is an inert gas that is soluble in both only a single parameter – CBF. A major disadvan-
water and lipid. X-ray attenuation by Xe is similar tage is the lack of standardization and
to that of iodine, so Xe can be used as a contrast unavailability of xenon for medical applications.
agent in conjunction with CT (XeCT). The gas The test is available only in few centers around the
dissolves rapidly in blood after inhalation and world.
37 Brain Death Imaging 881

MR Angiography systolic arterial pressure, cerebral perfusion will


cease. Due to elasticity of the arterial wall and the
Magnetic resonance imaging of vessels (MRA, compliance of the vasculature distal to the record-
MR angiography) can be performed with several ing site, such cerebral circulatory arrest is associ-
techniques, from which time-of-flight (TOF) ated with Doppler evidence of oscillatory
imaging is widely accepted as a technique for movement of blood in the large arteries at the
the examination of the intracranial circulation. It base of the brain. However, the net forward flow
is based on the flow-related signal enhancement of volume is zero. With time the oscillations
the blood entering into the volume of interest. decrease in amplitude of spectral spikes until no
TOF is completely a noninvasive method because pulsations are detectable. This development cor-
there is no need to administer contrast agents. relates with a more proximal demonstration of the
Other advantages include high spatial resolution, angiographic flow arrest [32]. At the time of
relative short imaging time, and high signal-to- angiographic flow arrest at the internal carotid
noise ratio. TOF imaging, however, has some artery (ICA), the TCD shows an oscillating flow
technical limitations, e.g., reduced sensitivity to pattern in the middle cerebral artery, because the
slow blood flow as well as saturation of the signal contrast medium progresses slowly toward the
when vessels run parallel to the imaging slice brain. From the clinical experience from cardiac
orientation, what subsequently makes the blood arrest, such cerebral ischemia of about 10–15 min
flow undetectable, even when it is not severely in vivo at normal body temperature leads to irre-
impaired. versible total loss of brain function [33].
On MR angiograms in brain-dead patients, a
unique finding is the absence of any cerebral Transcranial Doppler Findings
blood flow above the level of the supraclinoid in Cerebral Circulatory Arrest
portions of the ICAs and in the vertebrobasilar 1. Increasing pulsatility of the velocity wave-
arteries [4, 5]. form: if the velocity at the end of diastole is
Karantanas et al. [5], who performed also MR zero, then the ICP has reached the diastolic
venography, demonstrated lack of visualization of blood pressure. Forward flow persists in sys-
veins and sinuses in the most but not all patients tole. Therefore, this stage does not correspond
with defined brain death. On the other hand, veins to cerebral circulatory arrest.
and sinuses were visualized in all MR venograms 2. Oscillating flow: cessation of cerebral perfu-
performed in comatose patients in whom the sion has been reached when forward and
follow-up did not reveal brain death in the next reverse flows are nearly equal (biphasic, oscil-
12 months. lating, and net zero flow, i.e., the ICP equals or
Luchtmann et al. [6] also documented that exceeds the systolic blood pressure). Equality
another technique, contrast-enhanced angio-MR, of both flow components is reached if the area
may be considered conclusive in detecting of ces- under the envelope of the positive and negative
sation of cerebral blood flow due to its extremely deflection is the same. This finding correlates
high sensitivity and specificity. with the angiographic appearance of cerebral
circulatory arrest [34].
3. Systolic spikes: with further reduction of the
Transcranial Doppler Sonography blood movement, only a very short velocity
peak can be seen (systolic spike). The systolic
Transcranial Doppler in Increased spike pattern is highly characteristic for cere-
Intracranial Pressure bral circulatory arrest. In this stage it can be
Extensive death of brain tissue causes extreme assumed that the slower and longer-lasting
increase of ICP. When the ICP equals the diastolic reverse flow component (see oscillating flow)
arterial pressure, the brain is perfused only in is obscured due to high-pass filtering. All
systole, and with further increase of ICP over the Doppler devices use high-pass filters in order
882 M. Sawicki et al.

Fig. 11 Progressive deterioration of TCD waveform observed during changes leading to cerebral circulatory arrest
(Modified from Hassler et al. [32]); DBP diastolic blood pressure, VB vascular bed

to eliminate artifacts from wall movements. 1. Tachycardia must not exceed 120/min.
For the diagnosis of cerebral circulatory arrest, 2. Systolic blood pressure must not be less than
this filter must be set at its lowest level (e.g., 90 mmHg (in some countries mean arterial
50 Hz). blood pressure must not be less than 80 mmHg).
4. No flow signals: as the intracranial pressure 3. The patient must be normocapnic (PCO2 must
increases further and the flow obstruction not be below the normal range).
becomes more proximal, no flow signals in
the basal cerebral arteries can be detected. Fail- These conditions may vary in some minor
ure to detect flow signals can also result from details in national guidelines; however, they are
ultrasonic transmission problems. In such a justified by influence on a Doppler spectrum
case the extracranial findings, including those shape.
of the vertebral arteries (VA), represent an The technical prerequisites of TCD in cerebral
important criterion for the diagnosis, or one circulatory arrest include [38]:
must confirm the disappearance of the flow
signal (the baseline examination before cere- 1. The examination must be performed by highly
bral circulatory arrest with sufficient bone win- skilled certified examiner.
dow must be available) [32, 33, 35] – see 2. High-pass filter should be 50 Hz.
Fig. 11. 3. It is advised that gain be increased.
4. It is advised that sample volume be set
Clinical Prerequisites of Transcranial 15 mm [38] (in guidelines in some countries,
Doppler sample volume should be lower than 10 mm
Besides the prerequisites that apply to all [36, 37], and in the opinion of the authors, the
other blood flow tests, there are some special sample volume depends on the device used
clinical conditions for Doppler examination [transcranial Doppler or transcranial duplex]
[33, 36–38]: and the bone window penetrability).
37 Brain Death Imaging 883

Fig. 12 Systolic spikes in


the middle cerebral artery,
insonation depth 55 mm.
Spikes’ duration is
<200 ms and velocity <50
cm/s

5. Envelope should be switched off. 3. Systolic spikes are sharp unidirectional veloc-
6. A portable, 2 MHz, pulsed-wave Doppler ity signals in early systole of less than 200 ms
ultrasonographic instrument is preferred [34, duration and less than 50 cm/s peak systolic
39]; however, in some countries transcranial velocity and without a flow signal during the
duplex scanner is permitted [36, 37]. remaining cardiac cycle. Transitory patterns
between oscillating flow and systolic spikes
Criteria of Sonographic Findings may be seen [33].
to Confirm Cerebral Circulatory Arrest 4. The diagnosis established by the intracranial
According to the World Federation examination must be confirmed by the extra-
of Neurology [33] cranial bilateral recording of the common
Cerebral circulatory arrest can be confirmed if the carotid artery (CCA), ICA, and vertebral artery
following extra- and intracranial Doppler sono- (VA); they are allowed to be performed by
graphic findings have been recorded and duplex device – Fig. 14.
documented both intra- and extracranially and 5. The lack of a signal during transcranial
bilaterally on two examinations at an interval of insonation of the basal cerebral arteries is not
at least 30 min. a reliable finding because this can be due to
transmission problems. But the disappearance
1. Systolic spikes or oscillating flow in any cere- of intracranial flow signals in conjunction with
bral artery which can be recorded by bilateral typical extracranial signals can be accepted as
transcranial insonation of the ICA and MCA, proof of circulatory arrest [33, 40].
respectively, and any branch or other artery 6. Ventricular drains or large openings of the skull
which can be recorded (anterior and posterior like decompressive craniectomy possibly
circulation) – Fig. 12. interfering with the development of the ICP
2. Oscillating flow is defined by signals with are not present.
forward and reverse flow components in one
cardiac cycle exhibiting almost the same area
under the envelope of the waveform (to-and-fro Once reverberating flow is identified, it should
movement) [33]. Some German guidelines be confirmed in the both MCAs and the BA and
permit the velocity of the retrograde (diastolic) monitored for 30 min to exclude effects of tran-
component of biphasic (oscillating) flow to not sient intracranial pressure increase. In such a case
be greater than one third of the antegrade the repetition of TCD examination on monitored
(systolic) velocity of the flow [38] – Fig. 13. artery after 30 min is not necessary [40].
884 M. Sawicki et al.

Fig. 13 Oscillating flow in


the middle cerebral artery.
Insonation depth 55 mm.
All diastolic flow is in
opposite direction to
systolic antegrade flow

Fig. 14 Biphasic
(oscillating) flow observed
in extracranial segment of
the internal carotid artery.
Usually it overlaps partly
with spectrum from the
external carotid artery
(4 MHz CW probe)

Practical Tips Some kind of oscillating flow can be seen in


Middle cerebral artery examination should be the aorta or common carotid artery dissections or
performed in depth 55–65 mm, avoiding internal in severe aortal valve insufficiency. It can be eas-
carotid artery bifurcation area, where overlapping ily distinguished in all branches of the aorta or
spectra of middle cerebral and anterior cerebral common carotid artery by longer duration of the
arteries can be observed. It is especially important systolic complex than in systolic spikes [36, 38].
because oscillating flow in both arteries can
mimic normal bifurcation two-directional flow Usefulness of TCD in Cerebral Circulatory
[38] – Fig. 15. Arrest Confirmation
It is common that systolic spikes can vary in TCD can be used as a screening tool to determine
velocity with respiratory action [41] – Fig. 16. the timing for other confirmatory tests such as
Sometimes triphasic flow can be seen, with nuclear brain scanning or angiography. It is of
small early diastole antegrade flow. It is not con- particular value in patients with high barbiturate
sidered to be a sign of irreversible circulatory levels when clinical examination and electroen-
arrest – Fig. 17. cephalography are unreliable [39, 42]. A variety
37 Brain Death Imaging 885

Fig. 15 Bifurcation of the


internal carotid artery at the
depth of 65 mm. This area
should be avoided in
confirmation of cerebral
circulatory arrest, because
overlapping spectra with
oscillating flow in the
middle cerebral artery and
the anterior cerebral artery
can mimic “normal flow”

Fig. 16 Systolic spikes


often change velocity with
respiratory movements. The
middle cerebral artery.
Insonation depth 55 mm

Fig. 17 Triphasic flow is


not consistent with cerebral
circulatory arrest. It is often
observed in patients with
large skull defects. The
middle cerebral artery.
Insonation depth 55 mm
886 M. Sawicki et al.

of clinical conditions may lead to brain death. is reported to be virtually 100 % [35, 43,
TCD is indicated when the clinical examination 44]. There have been several case reports of
indicates brain death or is unreliable. The test may patients with demonstrable CBF on TCD who
determine positive end-diastolic flow and rule out were clinically brain-dead. In these cases the
cerebral circulatory arrest or may demonstrate pathology was limited to the cerebellum or brain
reverberating flow and confirm clinical findings. stem leaving blood flow in the anterior cerebral
It is extremely important to document arterial circulation relatively intact. The cases of a small
blood pressure during TCD examination to rule reperfusion in anterior cerebral circulation after
out transient arrest of cerebral circulation due to hypoxic-ischemic injury due to cardiac arrest
hypotension [36, 40]. were reported after some days. For this reason it
Patients at risk of brain death may need base- is important that patients show clinical signs of
line TCD examination before clinical changes brain death before performing TCD [35], personal
become apparent. A baseline TCD will help to experience]. The same is true in patients with
establish the presence of temporal windows and great skull defects, in whom the preservation of
end-diastolic flow. Subsequent TCD studies can orthograde diastolic flow can persist, despite of
be performed hours or days later with greater fulfilling all clinical criteria of brain death
confidence, and intracranial flow changes will be diagnosis [33].
easier to interpret. For example, absent There are no reports in the available literature
end-diastolic flow in both MCAs and the BA in of a child or adult patient “surviving” who dem-
a patient with clinical brain death may onstrated bilateral signals of oscillating flow or
indicate incomplete arrest (extremely high resis- systolic spikes in the MCA and ICA for at least
tance to flow with some residual brain perfusion) half an hour [33].
or complete arrest (vessel wall distention In the meta-analysis [44] including ten studies
during systole with no brain perfusion). Although of the usage of TCD in brain death, two false-
velocities may be minimal with the latter, positive results were reported, but in both patients
confusion may arise if temporal windows were brain stem function did show brain death shortly
not established beforehand. In this situation, it is thereafter. In the first case respiratory movements
prudent to wait and repeat TCD 1–2 h later. Pro- in response to an apnea test persisted for some
gression to arrest will manifest as reverberating hours after the first TCD signals of cerebral circu-
flow, and a complete arrest will make identifica- latory arrest were detected. The second clinically
tion of any signals more difficult (asonic brain-dead patient, with cerebral circulatory arrest
arteries) [40]. by TCD in the basilar artery and the circle of
TCD can effectively shorten the time to con- Willis, confirmed by angiography, became iso-
firm the clinical diagnosis of brain death that is of electric only several hours later.
great importance when organ donation is planned
[36]. It is a preferable test due to noninvasiveness
and portability. Nuclear Medicine Methods
Main disadvantages of TCD include lack of in the Diagnosis of Brain Death
sufficient bone window that occurs in about
10 % patients, confounding noises in intensive Brain scintigraphy is one of the reference methods
care unit setting during bedside examination, dif- among others used for the diagnosis of brain
ficulties with keeping parameters of patient’s death. Dynamic brain scintigraphy with non-
blood pressure and heart rate in sufficient levels brain-binding agent Tc99m-DTPA has been used
to perform TCD examination, and lack of well- for a long time. After bolus intravenous injection
trained, experienced examiners [36, 38, 42]. of the tracer brain vascular flow is estimated.
Although the sensitivity of TCD for the diag- DTPA does not cross the blood-brain intact barrier
nosis of brain death is 91.2–100 %, the specificity so intracranial blood flow is seen in normal
37 Brain Death Imaging 887

Fig. 18 Planar brain


Tc99m-HMPAO images.
Absence of tracer
accumulation in the brain
tissue consistent with brain
death

patients. The absence of intracranial blood flow is Tc99m-HMPAO/Tc99m-ECD brain study


considered brain death. The disadvantage of this should be performed according to the guidelines
tracer is that it provides rather low-resolution [47, 48]. Proper labeling of the tracer and injec-
brain vascular flow [45]. tion time according to manufacturer’s instruction
Development of new radiopharmaceuticals is crucial. Before injection quality control of the
Tc99m-HMPAO (hexamethylpropyleneamine tracer labeling is necessary. Yield of labeling
oxime) and Tc99m-ECD (ethyl cysteinate dimer), for Tc99m-HMPAO and for Tc99m-ECD should
which are brain specific, gave new input to the role be very high to avoid misinterpretation of the
of nuclear medicine methods in the determination study. The mode of acquisition can be planar or
of brain death. Both radiopharmaceuticals are lipo- SPECT (single-photon emission computed
philic agents and after intravenous injection cross tomography). Immediately after the tracer injec-
the blood-brain barrier and are accumulated pro- tion dynamic acquisition can be done, and it can
portionally to the blood flow in normal gray matter add additional information about brain blood
– brain cells of cerebrum, cerebellum, and brain flow. Planar (anterior, posterior and lateral) scin-
stem. So not only blood flow but brain parenchyma tigraphy is enough to make the diagnosis, but
is seen in normally functioning brain. If there is no additional SPECT or SPECT/CT is preferable
blood flow, there is no radiotracer accumulation in [49] – see Figs. 18 and 19.
the brain tissue (“hollow skull phenomenon”). Sometimes delayed images are helpful to visu-
After tracer injection there is a very quick accumu- alize if brain blood flow is present or absent.
lation of the tracer in a functioning brain. It makes SPECT acquisition can give better visualization
Tc99m-HMPAO/Tc99m-ECD brain study much of perfusion in posterior fossa and brain stem
more reliable than investigations estimating only structures. There can be different patterns of
blood flow. The injected radiopharmaceuticals are radiotracer accumulation in the brain. According
safe, with no side effects to the patients, and do not to existing standards, the only pattern which
interact with medications used for patients’ allows diagnosing brain death is lack of accumu-
treatment [46]. lation in the cerebrum and cerebellum. Some
888 M. Sawicki et al.

Fig. 19 SPECT/CT brain Tc99m-HMPAO images of a patient with intracerebral hemorrhage with ventricular involve-
ment and subsequent brain death. CT component, upper row; SPECT, lower row

authors report that lack of tracer accumulation in physiologically accumulated in viable brain tissue
the cerebrum is enough to diagnose brain death. can be considered as excellent tracer for estima-
Positron emission tomography (PET) agent tion of brain viability. The quality and spatial
18-fluorodeoxyglucose (FDG) which is resolution of images are superior to SPECT but
37 Brain Death Imaging 889

lack of instant availability of the tracer, logistic impossible) to define uniform diagnostic criteria
problems made it did not gain the widespread of cerebral circulatory arrest for the whole brain.
use [49]. There are known factors predisposing to disso-
Brain perfusion scintigraphy in most patients ciation between brain death and total cessation of
investigated due to suspicion of brain death gives cerebral circulation. Their common feature is that
proper diagnosis; is reliable, safe, reproducible, they cause a disproportional increase of the ICP,
and noninvasive; and can be repeated in which in certain areas of the brain may be insuf-
inconclusive cases [24]. There was no false- ficient to cease the cerebral circulation. There are
positive study reported. The study should be several mechanisms causing a local decrease of
performed in accredited nuclear medicine depart- the ICP including craniectomy, ventricular drain-
ment having experience in performing brain per- age, skull fracture, open fontanels, and unfused
fusion scintigraphy. Brain death scintigraphy is sutures in neonates and infants. The regionally
helpful in patients considered as possible organ decreased ICP may result in preservation or res-
donors to document the lack of blood flow. But toration of residual cerebral blood flow at the site
according to guidelines, the brain perfusion scin- of decompression, while in the remaining parts of
tigraphy alone is not sufficient to diagnose the the brain, the cessation of blood flow is observed,
brain death even if there is absence of perfusion as presented in Fig. 20 [15, 25]. Similar problem
[47]. It should be correlated with other findings can be met in some cases of infratentorial injuries
like clinical examination, cerebral angiography, (e.g., thrombosis of the BA or brain stem hemor-
EEG, or others. rhage) in which the rising of ICP in infratentorial
compartment precedes its increase in
supratentorial region due to protective function
Limitations of Blood Flow Studies of the cerebellar tentorium. In such cases the
in the Diagnosis of Brain Death blood flow in cerebral hemispheres ceases with
marked delay in comparison to posterior circula-
The limitations of blood flow ancillary tests can be tion [35] – see Fig. 21. The opposite sequence is
divided into the technical and clinical ones. The known as well, when cessation of blood flow in
technical limitations were discussed above. supratentorial region precedes cerebral circulatory
In the early days of research on brain death, arrest in the basilar artery [15, 18]. Diagnostic
there was a prevailing notion based on the autop- difficulties are also encountered in cases of
sies that the clinical diagnosis of brain death is hypoxic-ischemic brain injuries after cardiac
associated with totally necrotic brain described as arrest when the ineffective restoration of cerebral
the so-called respirator brain. However, recent blood flow is observed [28, 35] – see Fig. 22. It
neuropathologic findings show widespread areas should be noted that in all abovementioned situa-
of normal to near-normal neuronal areas in one tions, clinical criteria of brain death are met. It is
third to two thirds of patients who meet the clin- obvious that the situation when the residual per-
ical criteria of brain death [50]. Thus, normally or fusion is revealed in a patient meeting the clinical
hypoperfused areas frequently exist in the brain criteria of brain death leads to diagnostic confu-
which lost its all functions and thereby is defined sion and delays the diagnosis of brain death.
as the dead brain. Therefore, total cerebral circu-
latory arrest is not a necessary condition for a loss
of all brain functions. Given that the blood flow
test must demonstrate global cerebral circulatory Future Trends: Looking for an Ideal
arrest to confirm brain death, this goal may be Ancillary Test
unattainable in some cases.
Furthermore, it is known that individual brain Although a number of ancillary tests were intro-
structures show variable vulnerability to ische- duced to the diagnosis of brain death, none of
mia. This makes it difficult (presumably them is ideal. A potentially ideal method should
890 M. Sawicki et al.

Fig. 20 CT angiography and perfusion findings in a shows filling of the deep cerebral veins (arrowhead). CTA
71-year-old man after aneurysmal SAH and a bilateral findings preclude the diagnosis of brain death. Perfusion
craniectomy who presented with signs of brain death on CT-CBF (d) and CBV (e) maps show CBF of 15–20
clinical examination: (a, b) VRTs 60 s after contrast injec- ml/100 g/min and CBV 2–2.5 ml/100 g in cortical areas
tion show filling of cortical branches of the MCAs (arrow- at the regions of craniectomies (arrows) and in the right
heads) and the ACAs (arrows). No opacification of the basal ganglia (empty arrows). These values are above the
basilar artery or intracranial parts of the vertebral arteries is thresholds for necrosis, consistent with deep ischemia
noted (empty arrows); (c) VRT 60 s after contrast injection

Fig. 21 CT angiography and perfusion findings in a noted (arrowheads). CTA findings preclude the diagnosis
34-year-old woman with brain stem hemorrhage with a of brain death. (c) CT perfusion. CBF map shows values
frontal craniotomy and ventricular drainage who presented consistent with necrosis in the brain stem (asterisk), mid-
with signs of brain death on clinical examination: (a) 5 mm brain, cerebellum, and occipital lobes. However, the blood
NECT in axial plane shows massive edema, sulcal efface- flow is preserved in the basal ganglia and temporal and
ment, ventricular narrowing, low GM/WM differentiation, frontal lobes. In infratentorial brain injuries, the blood flow
and hematoma in the brain stem (arrow); (b) VRT 60 s after ceases initially in the posterior circulation, while perfusion
contrast injection shows filling of cortical branches of the in the supratentorial compartment may be preserved for
MCAs and the ACAs (arrows). No opacification of the some time
basilar artery or intracranial parts of the vertebral arteries is
37 Brain Death Imaging 891

Fig. 22 CTA findings in a


58-year-old man after
cardiac arrest who
presented with signs of
brain death on clinical
examination. NECT: (a)
5 mm MPR in axial plane
shows diffuse edema and
hypodense basal ganglia
(arrows) and midbrain
(arrowhead); (b) 5 mm
MPR in axial plane shows
sulcal effacement and
hypodense cortex (arrows).
Cortical and deep GM is
more frequently affected
because neurons are more
vulnerable to hypoxia than
oligodendroglia or
astrocytes. CT angiography:
(c) VRT 60 s after contrast
injection shows filling of
cortical branches of the
MCAs and the ACAs
(arrowheads); (d) filling of
the deep cerebral veins is
preserved (arrow). CTA
findings preclude the
diagnosis of brain death

Table 5 Comparison of the selected blood flow studies in the diagnosis of brain death
Technical Interpretation Specificity Sensitivity
Test Availability Feasibility simplicity simplicity Noninvasiveness (%) (%)
Catheter  +  +  100 100
angiography
CTA +++ + ++ + + 100b 52–97a
PCT ++ + ++ ++ + 100b 89–100a
XeCT  + + ++ + 100 67–100
MRA +  ++ + + 100 100
DWI +  + + + 100 89–100
TCD  +/   + 100 91–100
Nuclear  + + ++ + 100 100
scan
a
Depending on the evaluation criteria used
b
Reliable case-control studies are missing
892 M. Sawicki et al.

Fig. 23 Time-resolved dynamic CT angiography propagation of contrast with weak opacification of


(4D CTA) in a patient with clinical signs of brain death: MCA-M1 (arrowheads) and the basilar artery (empty
(a) VRT 10 s after injection shows opacification of extra- arrow). Extracranially contrast outflow through the super-
cranial arteries (arrows) indicating proper contrast deliv- ficial temporal veins is noted (arrows). No intracranial
ery. No filling of the ICAs or the VAs is visible; VRT 25 s venous outflow is noted
(b) and 45 s (c) after injection show extremely slow

be characterized by 100 % specificity (the test In recent years new modalities were proposed,
does not reveal cerebral circulatory arrest in non- e.g., the bispectral index scale monitor (mathe-
brain-dead cases), sensitivity close to 100 %, matical algorithm of EEG), measuring of venous
ubiquity, noninvasiveness, technical and interpre- oxygen saturation in the jugular bulb and brain
tational simplicity, and portability – see Table 5. tissue oxygenation.
It seems that catheter angiography and TCD
reached the peak of their technical capabilities in
the diagnosis of brain death. This does not apply Summary
to CT considering the recent introduction of
ultrafast, wide-detector scanners. Such machines Brain death is defined as the irreversible cessa-
are capable to perform a time-resolved dynamic tion of functioning a the brain stem. Brain death
CTA (4D CTA) – see Fig. 23. Combination of the is principally established using clinical criteria
4D CTA with the whole-brain PCT using a single including coma, absence of brain stem reflexes,
contrast injection seems to be the most promising and loss of central drive to breathe, assessed with
alternative for the future. The issue of portability apnea test. In situations in which clinical testing
of CT may be resolved in the next years as first cannot be performed or when uncertainty exists
portable CT scanners are already on the market. about the reliability of its parts due to
37 Brain Death Imaging 893

confounding conditions, ancillary tests Radiol 12(11):2710–2716. doi:10.1007/s00330-002-


(i.e., imaging studies) may be useful. The objec- 1336-z
6. Luchtmann M, Beuing O, Skalej M, Kohl J, Serowy S,
tive of ancillary tests in the diagnosis of brain Bernarding J, Firsching R (2014) Gadolinium-
death is to demonstrate the absence of cerebral enhanced magnetic resonance angiography in brain
electrical activity (EEG and evoked potentials) or death. Sci Rep 4:3659. doi:10.1038/srep03659
cerebral circulatory arrest. In clinical practice 7. Lövblad K-O, Basssetti C (2000) Diffusion-weighted
magnetic resonance imaging in brain death. Stroke J
catheter cerebral angiography, perfusion scintig- Cereb Circ 31(2):539–542. doi:10.1161/01.STR.31.2.539
raphy, transcranial Doppler sonography, CT 8. Matsumura A, Meguro K, Tsurushima H, Komatsu Y,
angiography, and MR angiography are used. Kikuchi Y, Wada M, Nakata Y, Ohashi N, Nose T
Other methods, like perfusion CT, xenon CT, (1996) Magnetic resonance imaging of brain death.
Neurol Med Chir 36(3):166–171
MR spectroscopy, diffusion-weighted MRI, and 9. Sohn CH, Lee HP, Park JB, Chang HW, Kim E, Kim E,
functional MRI, are being studied as potentially Park UJ, Kim HT, Ku J (2012) Imaging findings of
useful in the diagnosis of brain death. brain death on 3-tesla MRI. Korean J Radiol 13(5):
Among these tests, blood flow and perfusion 541–549. doi:10.3348/kjr.2012.13.5.541
10. Luchtmann M, Bernarding J, Beuing O, Kohl J, Bondar I,
studies are most commonly applied. The diagno- Skalej M, Firsching R (2013) Controversies of diffusion
sis of brain death can be confirmed when a lack of weighted imaging in the diagnosis of brain death. J Neu-
blood flow or tissue perfusion in the whole brain is roimaging 23(4):463–468. doi:10.1111/jon.12033
documented. 11. Selcuk H, Albayram S, Tureci E, Hasiloglu ZI,
Kizilkilic O, Cagil E, Kocer N, Islak C (2012)
However, the application of blood flow and Diffusion-weighted imaging findings in brain death.
perfusion tests to the diagnosis of brain death is Neuroradiology 54(6):547–554. doi:10.1007/s00234-
limited by variable vulnerability of individual 011-0912-9
brain structures to ischemia. Besides there are 12. Falini A, Barkovich AJ, Calabrese G, Origgi D,
Triulzi F, Scotti G (1998) Progressive brain failure
known factors predisposing to dissociation after diffuse hypoxic ischemic brain injury: a serial
between brain death and total cessation of cerebral MR and proton MR spectroscopic study. AJNR Am J
circulation. These factors include skull decom- Neuroradiol 19(4):648–652
pression, infratentorial localization, and hypoxic- 13. Boly M, Tshibanda L, Vanhaudenhuyse A,
Noirhomme Q, Schnakers C, Ledoux D, Boveroux P,
ischemic mechanism of brain injury. Garweg C, Lambermont B, Phillips C, Luxen A,
Moonen G, Bassetti C, Maquet P, Laureys S (2009)
Functional connectivity in the default network during
References resting state is preserved in a vegetative but not in a
brain dead patient. Hum Brain Mapp 30(8):
1. WHO (2012) International guidelines for the determina- 2393–2400. doi:10.1002/hbm.20672
tion of death – phase I. Canadian Blood Services http:// 14. Monti MM, Vanhaudenhuyse A, Coleman MR,
www.who.int/patientsafety/montreal-forum-report.pdf Boly M, Pickard JD, Tshibanda L, Owen AM, Laureys
2. Aichner F, Felber S, Birbamer G, Luz G, Judmaier W, S (2010) Willful modulation of brain activity in disor-
Schmutzhard E (1992) Magnetic resonance: a nonin- ders of consciousness. N Engl J Med 362(7):579–589.
vasive approach to metabolism, circulation, and mor- doi:10.1056/NEJMoa0905370
phology in human brain death. Ann Neurol 32(4): 15. Braun M, Ducrocq X, Huot JC, Audibert G,
507–511. doi:10.1002/ana.410320405 Anxionnat R, Picard L (1997) Intravenous angiogra-
3. Kato T, Tokumaru A, O’Uchi T, Mikami I, Umeda M, phy in brain death: report of 140 patients. Neuroradi-
Nose K, Eguchi T, Hasegawa M, Okuyama K (1991) ology 39(6):400–405
Assessment of brain death in children by means of P-31 16. Heran MK, Heran NS, Shemie SD (2008) A review of
MR spectroscopy: preliminary note. Work in progress. ancillary tests in evaluating brain death. Can J Neurol
Radiology 179(1):95–99. doi:10.1148/radiology.179. Sci 35(4):409–419
1.2006312 17. Grosse K, Brauer B, Kucuk O, Kruger R, Polster F,
4. Ishii K, Onuma T, Kinoshita T, Shiina G, Grauhan O, Wesslau C (2006) Does contrast medium
Kameyama M, Shimosegawa Y (1996) Brain death: administration in organ donors affect early kidney graft
MR and MR angiography. AJNR Am J Neuroradiol function? Transplant Proc 38:668–669. doi:10.1016/j.
17(4):731–735 transproceed.2006.01.034
5. Karantanas AH, Hadjigeorgiou GM, Paterakis K, 18. Kricheff II, Pinto RS, George AE, Braunstein P, Korein
Sfiras D, Komnos A (2002) Contribution of MRI and J (1978) Angiographic findings in brain death. Ann N
MR angiography in early diagnosis of brain death. Eur Y Acad Sci 315:168–183
894 M. Sawicki et al.

19. Dupas B, Gayet-Delacroix M, Villers D, Antonioli D, tomography in diagnosis of brain death. Neuroradiology
Veccherini MF, Soulillou JP (1998) Diagnosis of brain 55(9):1061–1069. doi:10.1007/s00234-013-1210-5
death using two-phase spiral CT. AJNR Am J 30. Ashwal S, Schneider S, Thompson J (1989) Xenon
Neuroradiol 19(4):641–647 computed tomography measuring cerebral blood
20. Bohatyrewicz R, Sawicki M, Walecka A, Walecki J, flow in the determination of brain death in children.
Rowinski O, Bohatyrewicz A, Kanski A, Ann Neurol 25(6):539–546. doi:10.1002/ana.
Czajkowski Z, Krzysztalowski A, Solek-Pastuszka J, 410250603
Zukowski M, Marzec-Lewenstein E, Wojtaszek M 31. Pistoia F, Johnson DW, Darby JM, Horton JA,
(2010) Computed tomographic angiography and per- Applegate LJ, Yonas H (1991) The role of xenon CT
fusion in the diagnosis of brain death. Transplant Proc measurements of cerebral blood flow in the clinical
42(10):3941–3946. doi:10.1016/j.transproceed.2010. determination of brain death. AJNR Am J Neuroradiol
09.143 12(1):97–103
21. Frampas E, Videcoq M, de Kerviler E, Ricolfi F, 32. Hassler W, Steinmetz H, Pirschel J (1989) Transcranial
Kuoch V, Mourey F, Tenaillon A, Dupas B (2009) CT Doppler study of intracranial circulatory arrest. J
angiography for brain death diagnosis. AJNR Am J Neurosurg 71(2):195–201. doi:10.3171/jns.1989.71.
Neuroradiol 30(8):1566–1570. doi:10.3174/ajnr. 2.0195
A1614 33. Ducrocq X, Hassler W, Moritake K, Newell DW, von
22. Leclerc X, Taschner CA, Vidal A, Strecker G, Reutern GM, Shiogai T, Smith RR (1998) Consensus
Savage J, Gauvrit JY, Pruvo JP (2006) The role of opinion on diagnosis of cerebral circulatory arrest
spiral CT for the assessment of the intracranial circula- using Doppler-sonography: task force group on cere-
tion in suspected brain-death. J Neuroradiol J bral death of the Neurosonology Research Group of the
Neuroradiol 33(2):90–95 World Federation of Neurology. J Neurol Sci 159(2):
23. Sawicki M, Bohatyrewicz R, Safranow K, Walecka A, 145–150
Walecki J, Rowinski O, Solek-Pastuszka J, 34. Wijdicks EF (1995) Determining brain death in adults.
Czajkowski Z, Guzinski M, Burzynska M, Wojczal J Neurology 45(5):1003–1011
(2014) Computed tomographic angiography criteria in 35. Hadani M, Bruk B, Ram Z, Knoller N, Spiegelmann R,
the diagnosis of brain death—comparison of sensitivity Segal E (1999) Application of transcranial Doppler
and interobserver reliability of different evaluation ultrasonography for the diagnosis of brain death. Inten-
scales. Neuroradiology 56(8):609–620. doi:10.1007/ sive Care Med 25(8):822–828
s00234-014-1364-9 36. Lovrencic-Huzjan A, Vukovic V, Gopcevic A,
24. Berenguer CM, Davis FE, Howington JU (2010) Brain Vucic M, Kriksic V, Demarin V (2011) Transcranial
death confirmation: comparison of computed tomo- Doppler in brain death confirmation in clinical practice.
graphic angiography with nuclear medicine perfusion Ultraschall Med 32(1):62–66. doi:10.1055/s-0029-
scan. J Trauma 68(3):553–559. doi:10.1097/ 1245237 (Stuttgart, Germany: 1980)
TA.0b013e3181cef18a 37. Segura T, Calleja S, Irimia P, Tembl JI, Spanish Society
25. Escudero D, Otero J, Marques L, Parra D, Gonzalo JA, of Neurosonology (2009) Recommendations for the
Albaiceta GM, Cofino L, Blanco A, Vega P, Murias E, use of transcranial Doppler ultrasonography to deter-
Meilan A, Roger RL, Taboada F (2009) Diagnosing mine the existence of cerebral circulatory arrest as
brain death by CT perfusion and multislice CT angiog- diagnostic support for brain death. Rev Neurosci 20
raphy. Neurocrit Care 11(2):261–271. doi:10.1007/ (3–4):251–259
s12028-009-9243-7 38. Widder B, Görtler M, Widder G (2004) Doppler- und
26. Combes JC, Chomel A, Ricolfi F, d’Athis P, Freysz M Duplexsonographie der hirnversorgenden Arterien, 6th
(2007) Reliability of computed tomographic angiogra- edn. Springer, Berlin
phy in the diagnosis of brain death. Transplant Proc 39. Wijdicks EF, Varelas PN, Gronseth GS, Greer DM,
39(1):16–20. doi:10.1016/j.transproceed.2006.10.204 American Academy of Neurology (2010) Evidence-
27. Quesnel C, Fulgencio JP, Adrie C, Marro B, Payen L, based guideline update: determining brain death in
Lembert N, El Metaoua S, Bonnet F (2007) Limitations adults: report of the Quality Standards Subcommittee
of computed tomographic angiography in the diagnosis of the American Academy of Neurology. Neurology 74
of brain death. Intensive Care Med 33(12):2129–2135. (23):1911–1918. doi:10.1212/
doi:10.1007/s00134-007-0789-6 WNL.0b013e3181e242a8
28. Shankar JJ, Vandorpe R (2013) CT perfusion for con- 40. Alexandrov AV, Sloan MA, Tegeler CH, Newell DN,
firmation of brain death. AJNR Am J Neuroradiol Lumsden A, Garami Z, Levy CR, Wong LK,
34(6):1175–1179. doi:10.3174/ajnr.A3376 Douville C, Kaps M, Tsivgoulis G, American Society
29. Sawicki M, Bohatyrewicz R, Safranow K, Walecka A, of Neuroimaging Practice Guidelines C (2012) Practice
Walecki J, Rowinski O, Solek-Pastuszka J, standards for transcranial Doppler (TCD) ultrasound.
Czajkowski Z, Marzec-Lewenstein E, Motyl K, Part II. Clinical indications and expected outcomes. J
Przybyl W, Czarnecka A (2013) Dynamic evaluation Neuroimaging 22(3):215–224. doi:10.1111/j.1552-
of stasis filling phenomenon with computed 6569.2010.00523.x
37 Brain Death Imaging 895

41. Ducrocq X, Braun M, Debouverie M, Junges C, comparison between SPECT and contrast angiography.
Hummer M, Vespignani H (1998) Brain death and Crit Care Med 33(9):2068–2073
transcranial Doppler: experience in 130 cases of brain 46. Zuckier L, Kolano J (2008) Radionuclide studies in the
dead patients. J Neurol Sci 160(1):41–46 determination of brain death: criteria, concepts, and
42. White H, Venkatesh B (2006) Applications of controversies. Semin Nucl Med 38(4):262–273.
transcranial Doppler in the ICU: a review. Intensive doi:10.1053/j.semnuclmed.2008.03.003
Care Med 32(7):981–994. doi:10.1007/s00134-006- 47. Donohoe KJ, Agrawal G, Frey KA, Gerbaudo VH,
0173-y Mariani G, Nagel JS, Shulkin BL, Stabin MG, Stokes
43. de Freitas GR, Andre C (2006) Sensitivity of MK (2012) SNM practice guideline for brain death
transcranial Doppler for confirming brain death: a scintigraphy 2.0. J Nucl Med Technol 40(3):198–203.
prospective study of 270 cases. Acta Neurol Scand doi:10.2967/jnmt.112.105130
113(6):426–432. doi:10.1111/j.1600-0404.2006. 48. Donohoe KJ, Frey KA, Gerbaudo VH, Mariani G,
00645.x Nagel JS, Shulkin B (2003) Procedure guideline for
44. Monteiro LM, Bollen CW, van Huffelen AC, brain death scintigraphy. J Nucl Med 44(5):846–851
Ackerstaff RG, Jansen NJ, van Vught AJ (2006) 49. Sinha P, Conrad GR (2012) Scintigraphic confirmation
Transcranial Doppler ultrasonography to confirm of brain death. Semin Nucl Med 42(1):27–32.
brain death: a meta-analysis. Intensive Care Med 32 doi:10.1053/j.semnuclmed.2011.07.007
(12):1937–1944. doi:10.1007/s00134-006-0353-9 50. Wijdicks EF, Pfeifer EA (2008) Neuropathology of
45. Munari M, Zucchetta P, Carollo C, Gallo F, De brain death in the modern transplant era. Neurology
Nardin M, Marzola MC, Ferretti S, Facco E (2005) 70(15):1234–1237. doi:10.1212/01.wnl.0000289762.
Confirmatory tests in the diagnosis of brain death: 50376.b6
Part VII
Veins Imaging
Jugular Vein Thrombosis
38
Nicholas A. Koontz, Richard H. Wiggins III, and
Lubdha M. Shah

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 900 Jugular vein thrombosis (JVT) is a not uncom-
mon and serious finding encountered by radi-
Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 900
ologists. While identifying thrombus within
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 902 the internal jugular vein (IJV) is usually not a
Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 903 diagnostic dilemma, a thorough evaluation of
the surrounding structures is critical due to the
Etiologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 904
Iatrogenic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 904 sundry etiologies, potentially devastating com-
Neoplastic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 906 plications (including pulmonary embolism,
Infectious . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 908 septic emboli, and dural sinus thrombosis),
Traumatic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 915
and high association with neoplasm. In this
Hematologic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 915
Rheumatologic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 916 chapter, we discuss the typical anatomy of the
Gynecologic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 917 IJV and the pathophysiology, clinical presenta-
Endocrine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 918 tion, and potential etiologies of JVT (including
Idiopathic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 918
iatrogenic, neoplastic, infectious, traumatic,
JVT in Pediatric Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 918
hematologic, rheumatologic, gynecologic,
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 919 endocrine, and idiopathic causes, as well as
CT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 919
US . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 921 pediatric-specific considerations). Multimodality
MRI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 923 imaging findings of JVT (computed tomography,
Nuclear Medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 924 magnetic resonance imaging, ultrasound, nuclear
Angiography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 927 medicine, and angiography) and the diagnostic
Diagnostic Pitfalls . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 928 pitfalls are also reviewed, utilizing an image-rich,
False-Negative Misinterpretation . . . . . . . . . . . . . . . . . . . . 929 comprehensive review of the medical literature.
False-Positive Misinterpretation . . . . . . . . . . . . . . . . . . . . . 929
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 931 Keywords
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 931 CT • MRI • Vascular venous imaging • Jugular
vein

N.A. Koontz • R.H. Wiggins III • L.M. Shah (*)


Department of Radiology, University of Utah Health
Sciences Center, Salt Lake City, UT, USA
e-mail: Nick.Koontz@hsc.utah.edu; Richard.
Wiggins@hsc.utah.edu; Lubdha.shah@hsc.utah.edu

# Springer Science+Business Media New York 2016 899


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_42
900 N.A. Koontz et al.

Introduction Table 1 Causes of jugular vein thrombosis


Iatrogenic
Jugular vein thrombosis (JVT) is a not uncommon Central venous access device (CVAD)
pathologic process characterized by thrombus for- Cardioverter defibrillator/pacemaker
mation within the internal jugular vein (IJV). This Ventriculoatrial shunt
review will focus on the extracranial IJV. Postoperative
Although JVT is a relatively straightforward Cervical manipulation
diagnosis to make with imaging, the challenge Neoplastic
lies in determining the etiology and identifying Mass effect
associated pathology and complications. While Primary tumor
Lymphadenopathy
most commonly encountered in ill patients
Direct invasion
with thrombogenic central venous catheters or
Migratory thrombophlebitis (Trousseau syndrome)
postoperatively in patients following neck
Posttreatment (chemotherapy and/or radiation therapy)
dissections, sundry etiologies exist beyond the
Infectious
obvious iatrogenic sources and are Intravenous drug abuse (IVDA)
frequently dependent upon the patient profile Human necrobacillosis (Lemierre syndrome)
(Table 1). Otomastoiditis
Given the many etiologies of JVT, it is impor- Postauricular abscess
tant for radiologists to consider the clinical con- Cytomegalovirus
text with this imaging finding, as well as the Traumatic
appearance on different imaging modalities. In Blunt trauma
doing so, one can suggest an appropriate, Penetrating trauma
patient-specific, clinically relevant differential Hematologic
diagnosis and add real clinical value to a radio- Activated protein C (APC) resistance
logic report. A comprehensive search for the eti- Congenital
ology of JVT will often rely on a thorough review Factor V Leiden thrombophilia
of the electronic medical record and clinical con- Factor V Cambridge mutation
sultation, as well as scrutinizing other radiologic Acquired
Antiphospholipid antibodies (APLAs)
studies for imaging clues. The diagnosis of JVT,
Oral contraceptives
like so many other diagnoses encountered in diag-
Paraneoplastic
nostic imaging, is an investigational starting point
Thalidomide
and not simply a dead-end observation.
Hemophilia therapies
Factor VIII inhibitor bypass activity (FEIBA)
Activated recombinant factor VII (rVIIa)
Rheumatologic
Anatomy Rheumatoid arthritis
Systemic lupus erythematosus
The paired IJVs serve as the primary conduit for Silk road disease (Behçet disease)
venous return not only from the superficial parts Gynecologic
of the face and neck but also from the intracranial Assisted reproductive techniques (ART)
circulation to the right atrium of the heart (Fig. 1). In vitro fertilization
Functionally, the IJVs are a continuation of the Ovarian hyperstimulation syndrome (OHSS)
sigmoid sinuses below the level of the jugular Pregnancy
foramina in the skull base. The left IJV is gener- Endocrine
ally smaller than the right, and there is a unilateral Tertiary or secondary hypercortisolism (Cushing
syndrome)
valve in most cases [1].
Thyroid carcinoma
At its cranial terminus is a normal, focal dila-
Goiter
tation of the IJV, which is known as the superior
(continued)
38 Jugular Vein Thrombosis 901

Table 1 (continued)
Idiopathic
Pediatric patients
Iatrogenic
CVAD
Intravenous immunoglobulin (IVIg) infusion
Infectious
Human necrobacillosis (Lemierre syndrome)
Mastoiditis
Neoplastic
Hematologic
Traumatic
Congenital heart disease
Cystic fibrosis

Fig. 2 Typical appearance of the superior jugular


bulb (black open arrow), a normal focal dilatation of
the IJV

bulb (Figs. 1 and 2). The superior jugular bulb


typically lies below the floor of the internal audi-
tory canal (IAC), but those extending above the
floor of the IAC are considered “high-riding.”
Although a high-riding jugular bulb is a clinically
silent finding, it may be at increased risk of injury
during mastoidectomy. In comparison, a dehis-
cent jugular bulb occurs when rarefaction of the
osseous covering allows extension of the jugular
bulb into the middle ear cavity. One may also
encounter a jugular bulb diverticulum variant,
which is a tubular projection of the jugular bulb
into the adjacent skull base without extension into
the middle ear.
Below the skull base, the IJV resides within the
carotid space, deep into the sternocleidomastoid
muscle. In the suprahyoid neck, the IJV is typi-
cally located posterolateral to the internal carotid
artery (ICA) (Fig. 3), overlying the rectus capitis
Fig. 1 Coronal reconstruction of CECT of the neck dem-
onstrates a normal appearance of the right IJV (black lateralis, and the glossopharyngeal and hypoglos-
arrow), which extends from the confluence of the subcla- sal nerves pass between the ICA and IJV, while
vian and brachiocephalic veins (white asterisk) centrally to the vagus nerve lies posteriorly. In the infrahyoid
the sigmoid sinus peripherally. Normal, focal dilatations at
neck, the IJV is typically located lateral and
the cranial and caudal extremes of the IJV are known as the
superior jugular bulb (black open arrow) and inferior jug- anterolateral to the internal carotid and common
ular bulb (white open arrow), respectively carotid arteries (Fig. 4). However, the exact
902 N.A. Koontz et al.

Fig. 3 Normal anatomy of


the IJV in the suprahyoid
neck. Axial CECT (a) and
axial T1-WI +C (b) at the
level of the nasopharynx
demonstrate the IJV (white
arrow) to be located lateral
to the ICA (white open
arrow)

Fig. 4 Normal anatomy of the IJV in the infrahyoid neck.


Axial CECT at the level of the glottic larynx demonstrates
the IJVs (white arrows) to be located lateral to the internal
carotid arteries (white open arrows). While typically
located lateral or anterolateral to the ICA in the infrahyoid
neck, the location of the IJV can be variable

location of the IJV with respect to the carotid


artery is somewhat variable, particularly at the
extremes of age [2]. At its caudal terminus, the Fig. 5 Coronal CECT image demonstrates the inferior
jugular bulb (white open arrow), a normal, focal dilatation
IJV joins the subclavian vein and forms the of the IJV just before its confluence with the jugular vein
brachiocephalic vein. Just proximal to its termi-
nation is a second dilatation, the inferior jugular
bulb (Figs. 1 and 5). Numerous venous tributaries Pathophysiology
commonly feed into the IJV, including the facial
vein, retromandibular vein, superior thyroid vein, The pathophysiology of JVT is rooted in the well-
and middle thyroid vein, although there is great known Virchow triad. This is named after, but
variation and overlap of tributary venous return. probably not actually described (at least in its
38 Jugular Vein Thrombosis 903

Table 2 High-risk patient populations for development


of JVT
Central venous access device (CVAD)
Static Blood Intravenous drug abuse (IVDA)
Flow Malignancy

venous access devices (CVADs) are at increased


risk of JVT due to static venous flow along the
Hyper- Endothelial catheter as well as due to endothelial injury from
coaguability Injury catheter placement. Patients with a history of
intravenous drug abuse (IVDA) are at increased
risk of JVT from either caustic, chemical endo-
thelial injury, or endothelial injury from IVDA-
associated blood-borne infection. Patients with
Fig. 6 An abnormality in one or more of these variables underlying neoplasm may be at increased risk
increases the risk of developing a thrombus. Areas of for JVT formation due to a systemic, cancer-
intersection of the circles in this Venn diagram reflect
increased risk of thrombosis. At highest risk are patients induced hypercoagulable state or from compres-
who fall into all three categories sion of the vessel lumen by cervical lymphade-
nopathy or primary tumor, resulting in static flow
and impaired venous return. These few examples
current understanding), by Rudolf Ludwig Karl illustrate how the components of the Virchow
Virchow (1821–1902), the so-called father of triad contribute to thrombosis of this ordinarily
modern pathology; this model posits that three resilient vessel. Keeping these basic principles in
broad categories of factors are at play in the devel- mind during image interpretation can be quite
opment of vascular thrombosis: static blood flow, helpful in deciphering the etiology of JVT in
endothelial injury, and hypercoagulability [3] specific clinical settings.
(Fig. 6). An abnormality of one or more of these
variables increases the risk of developing a throm-
bus. In its native state, the IJV, like all other veins Clinical Presentation
of the head and neck, is an unlikely culprit to
develop thrombus, due in large part to its paucity Just as the etiologies of JVT are diverse and
of valves and the aid of gravity in venous return numerous, so too can be the clinical presentation
while the body is in an upright state [4]. While in (Table 3). Presentation may range from
the supine position, the IJVs serve as the primary completely asymptomatic (incidental) to palpable
venous drainage pathway for the brain; in the erect neck lump, dysphagia, cervical edema, upper
position, these veins collapse and flow via alter- extremity edema, erythrocyanosis, varicose
nate vertebral venous pathways [5]. For these superficial collateral veins, pulmonary embolism,
reasons, spontaneous JVT is rare and must raise or frank sepsis, depending on the etiology, acuity,
clinical concern for an underlying culprit causing and extent of disease [6, 7]. Patients more com-
static blood flow, endothelial injury, or monly present with unilateral disease, although
hypercoagulability. bilateral disease is not infrequently encountered
However, certain patient populations are at (Fig. 7). During the acute and subacute phases
increased risk of altering the dynamics of the (typically within 10–14 days of presentation) of
Virchow triad, shifting them toward a jugular vein thrombophlebitis, patients may expe-
prothrombotic state and subsequent JVT rience diffuse edema and tenderness overlying the
(Table 2). For example, patients with central sternocleidomastoid muscle. Patients are often
904 N.A. Koontz et al.

Table 3 Clinical presentation of jugular vein thrombosis


Acute phase (first 2 weeks)
Asymptomatic
Palpable lump
Neck pain
Dysphagia
Cervical or upper extremity edema
Erythrocyanosis
Varicose collateral veins
Pulmonary embolism
Sepsis
Chronic phase (>2 weeks)
Palpable, cord-like mass
Varicose collateral veins

Fig. 8 Postoperative NECT in a 31-year-old man with


recent left retrosigmoid craniectomy (black open arrow)
for resection of a vestibular schwannoma demonstrates
high density within the left IJV (black arrow),
corresponding to “downhill thrombus” propagating from
the thrombosed sigmoid sinus

Etiologies

In order to systematically cover the many possible


etiologies for JVT, it is helpful to first consider the
diagnosis from a “bird’s-eye view,” compartmen-
talizing the potential sources into several general
categories – iatrogenic, neoplastic, infectious,
traumatic, hematologic, rheumatologic, gyneco-
logic, endocrine, and idiopathic – which will be
discussed individually. Potential etiologies of JVT
in the pediatric population will also be discussed.

Fig. 7 Sixty-year-old man with left ventricular access Iatrogenic


device (LVAD) who presents with altered mental status
concerning for acute cerebral infarct. Coronal reformat of
CTA of the neck demonstrates occlusive thrombus in the Given the widespread use of CVADs, the preva-
left IJV (white arrow) and near-complete occlusive throm- lence of head and neck surgery, and the increased
bus in the right IJV (white open arrow) use of transvenous medical devices, iatrogenic
sources are the most common causes of JVT in
suspected of having an abscess. In the chronic modern medicine [8–10]. IJV occlusion (typically
phase after the inflammation subsides, thrombus non-thrombotic) is infrequently seen postopera-
may persist within the internal jugular vein, tively following head and neck surgery
resulting in a palpable, cord-like mass and in (Figs. 8–15). Among iatrogenic causes, the vast
many cases total occlusion. majority of JVT is due to central venous catheters,
38 Jugular Vein Thrombosis 905

Fig. 9 Volumetric T1-WI +C in the same patient as Fig. 8


demonstrates a low signal intensity filling defect (white
open arrow) within the enlarged left jugular bulb,
corresponding with acute postoperative “downhill” JVT
Fig. 11 Coronal maximal intensity projection (MIP) from
that had propagated from the sigmoid sinus
2D time-of-flight (TOF) MRV in the same patient as
Figs. 8, 9, and 10 shows no flow-related enhancement
within the distal left transverse sinus, sigmoid sinus, jugu-
lar bulb, and visualized IJV, corresponding with “down-
hill” thrombus following left retrosigmoid craniectomy

often resulting in “uphill thrombosis” that propa-


gates into the IJV from the downstream subcla-
vian or brachiocephalic veins (Fig. 16). At-risk
patient populations include those with chronic
indwelling central venous catheters, as might be
utilized for chemotherapy or frequent blood
draws, and critically ill neonates requiring central
venous access. JVT as a complication of other
transvenous biomedical devices, such as
cardioverter defibrillators, pacemakers, and
ventriculoatrial shunts, is seen less commonly but
has been reported in the medical literature [9, 10].
As previously described, most iatrogenic causes
of JVT center around catheter-/device-induced
venous stasis and endothelial injury, although it is
important to consider superimposed hypercoa-
Fig. 10 Coronal 2D TOF image in the same patient as gulability in these typically ill patients.
Figs. 8 and 9 demonstrates apparent filling defect (white A large group of patients with increased sus-
arrow) in the right IJV which is due to turbulent blood flow ceptibility to iatrogenic JVT is hemodialysis
and should not be confused with true thrombus. In equiv-
(HD) patients, as they tend to have numerous,
ocal cases, further evaluation with dynamic post-contrast
MR venography or second-look ultrasound can be consid- long-term CVADs placed over a lifetime
ered for clarification (Fig. 17). Studies have demonstrated that in HD
906 N.A. Koontz et al.

Fig. 12 Axial diffusion-


weighted imaging (a) and
ADC map (b) in the same
patient as Figs. 8, 9, 10, and
11 demonstrate the JVT to
be hyperintense to
surrounding osseous
structures on both
sequences

Another subset of patients at risk for IJV


thrombosis or occlusion is that group who has
undergone neck surgery, most commonly follow-
ing neck dissection for head and neck cancer.
While IJV patency is dependent on the type and
extent of neck dissection (with intentional sacri-
fice of the IJV performed in some cases), true
thrombosis or complete occlusion following
IJV-sparing neck dissection is uncommon
[12]. However, a diminution in IJV caliber is
expected following neck dissection, reaching a
nadir in the immediate postoperative period and
frequently returning to near-normal caliber by
3 months [12]. Uncommonly, JVT can be seen
following carotid endarterectomy, presumably
due to prolonged intraoperative retraction of
the IJV.
Fig. 13 Axial T1-WI C demonstrates the left JVT Lastly, there are reports of JVT following cer-
(white open arrow) to be isointense to brain parenchyma
vical manipulation, including chiropractic and
and slightly hyperintense to skeletal muscle
deep tissue massage. Other than case reports, the
literature evidence for or against cervical
patients, JVT risk increases with both duration of manipulation-induced IJV thrombosis is certainly
catheter placement and total lifetime number of lacking. The mechanism of thrombus formation in
catheters placed, with subsequent JVT reported in these rare cases is not well elucidated, although it
2–64 % of cases based on older studies [8, 11]. might theoretically reflect a combination of
The underlying renal insufficiency itself is asso- pressure-induced venous stasis and traumatic
ciated with hemostatic abnormalities, and HD endothelial injury.
adds to these disturbances with turbulent flow,
high shear stress, and contact of blood to artificial
surfaces. This leads to activation of the coagula- Neoplastic
tion cascade, which is the target of anticoagulation
agents, such as unfractionated heparin and low- Like lower extremity deep vein thrombosis
molecular-weight heparin. (DVT), JVT is commonly associated with
38 Jugular Vein Thrombosis 907

Fig. 14 Axial T1-WI +C (a) and axial T2-WI FS (b) in a that the thrombus is isointense to slightly hyperintense to
31-year-old man with recent left retrosigmoid craniectomy brain parenchyma on T1-WI +C and T2-WI FS and dem-
for resection of a vestibular schwannoma demonstrates onstrates a thin rim of peripheral enhancement on post-
“downhill” JVT (white open arrow) that has propagated contrast imaging due to enhancement of engorged vasa
from iatrogenic thrombosis of the left sigmoid sinus. Note vasorum

Fig. 15 Sagittal SPGR-C


imaging through the
bilateral IJVs in the same
patient demonstrates the
normal, low signal (white
arrow) within the patent
right IJV (a) and increased
signal (white open arrow)
within the thrombosed left
IJV (b)

malignancy, with coincidence reported up to factor Xa, C-reactive protein, carcinoma


10–35 % in some series [7, 13] (Figs. 18–21). mucins, tumor-associated cysteine proteinase, or
Mechanistically, neoplasm-related thrombosis of oncogene activation), or (c) iatrogenic conse-
the IJV results from (a) compression/invasion of quences of cancer treatment (cf. Iatrogenic)
the vessel lumen by lymphadenopathy or primary [4, 14, 15].
tumor, resulting in static blood flow, (b) a com- Among all cancer patients, treatment-related
plex, incompletely understood cancer-induced causes are the most common etiology of JVT;
hypercoagulable state (often referred to as migra- these include CVADs, prolonged immobility,
tory thrombophlebitis or Trousseau syndrome) intravascular administration of caustic chemother-
that results in increased levels or activity of sev- apeutic agents, postsurgical vascular injury, and
eral procoagulant factors (including tissue factor, postradiation therapy vascular complications.
fibrinogen, factor VIIa, factor VIII, factor IXa, Nonetheless, the presence of JVT, especially in
908 N.A. Koontz et al.

Fig. 16 Digital subtraction angiography (DSA) of the


right upper extremity in a 45-year-old woman with “uphill
thrombosis” of the right IJV demonstrates filling defects
(black arrows) within the right IJV, subclavian vein, right
brachiocephalic vein, left brachiocephalic vein, and supe-
rior vena cava. A potentially thrombogenic central venous
catheter is present (black open arrow)

cancer patients, should prompt the interpreting


radiologist to carefully assess the cervical lymph
Fig. 17 Coronal MIP from CTA of the neck in a 60-year-
node stations for metastatic lymphadenopathy and old critically ill patient requiring hemodialysis delineates
to assess the deep spaces of the neck for a lesion the nonocclusive thrombus in the right IJV (white open
exerting mass effect on the thrombosed IJV. While arrow), as well as a potentially thrombogenic right internal
most often due to underlying head and neck can- jugular venous catheter (white arrowhead)
cer (such as squamous cell carcinoma of the
aerodigestive tract or thyroid malignancy), cervi- Infectious
cal chain lymphadenopathy causing JVT can also
be seen in widely disseminated metastatic disease Prior to the widespread use of antibiotics in the
of any number of primary malignancies 1940s, infection was by far the most common
[16–18]. The most common of these primary etiology of JVT. However, in current medical
malignancies include pancreatic, lung, stomach, practice infectious sources of JVT are quite rare,
and ovarian cancer, which may contribute to JVT attributed mainly to IVDA (Figs. 22–24) and spo-
due to increased factor VIII and thromboplastin radic cases of typically oropharyngeal infections
levels. Additionally, nonmalignant neoplasms, resulting in Lemierre syndrome. IVDA increases
such as lymphangioleiomyomatosis, can lead to the risk of JVT for several reasons: (1) shooters
JVT due to local mass effect and resulting venous typically do not employ sterile technique, thus
stasis. Both contrast-enhanced CT (CECT) and risking blood-borne infection; (2) traumatic endo-
ultrasound (US) are useful modalities for assess- thelial injury from the needle encourages throm-
ment of the neck masses, with US offering the bosis; (3) infectious (mycotic) endothelial damage
opportunity for image-guided fine needle aspira- by blood-borne organisms results in
tion or core needle biopsy of suspicious masses or prothrombotic infiltration and congestion of the
lymphadenopathy. adventitia (and subsequently the other layers of
38 Jugular Vein Thrombosis 909

Fig. 20 Sagittal reconstruction from CECT in the same


patient as Figs. 18 and 19 demonstrates thrombus (white
Fig. 18 Coronal reconstruction from a CECT of the cer- open arrow) within the left IJV, as well as the bulky,
vical soft tissues in a 47-year-old man with laryngeal necrotic, conglomerate nodal metastatic disease (white
squamous cell carcinoma (SCCa) and necrotic, conglom- arrow)
erate nodal metastases (white arrow) demonstrates marked
mass effect upon the left IJVand direct invasion with tumor
thrombus (white open arrow)

Fig. 19 Axial CECT in the same patient as Fig. 18 dem- Fig. 21 Axial CECT in the same patient as Figs. 18, 19,
onstrates the necrotic conglomerate nodal metastases and 20 demonstrates soft-tissue density tumor thrombus
(white arrow) with extranodal spread of tumor, which invading the left IJV (white open arrow) and encasing the
completely obliterates the left IJV ICA (white arrowhead)
910 N.A. Koontz et al.

Fig. 22 Axial CECT in a 50-year-old woman with acute


catheter-associated JVT and septic thrombophlebitis, who
presented with 6 days of right-sided neck pain, swelling, and
fever. Image demonstrates complete occlusion of an enlarged
right IJV with central low-attenuation filling defect (white Fig. 24 Sagittal reconstruction of CECT in the same
arrow), prominent enhancement of the vasa vasorum (white patient as Figs. 22 and 23 demonstrates complete occlusion
open arrow), perivascular edema (white asterisks), subtle intra- of an enlarged right IJV with central low-attenuation filling
muscular enhancement (black arrowhead) within an enlarged, defect (white arrow), prominent enhancement of the vasa
edematous right sternocleidomastoid muscle (black arrow), vasorum (white open arrow), and perivascular edema
and multiple venous bypass collaterals (white arrowheads) (white asterisks)

the vein wall) by inflammatory cells; and


(4) IVDA-associated deep neck infection contrib-
utes to altered blood flow dynamics, including
venous stasis [19]. Other infectious causes of the
deep spaces of the head and neck, such as post-
surgical infection (Figs. 25–33), acute
otomastoiditis, or postauricular abscess, may
also contribute to the development of JVT, albeit
much less commonly. Additionally, an increased
risk of JVT in immunocompetent patients with
severe acute cytomegalovirus infection has been
described, a link which may be under-recognized
and underreported.

Lemierre Syndrome
Fig. 23 Coronal reformat of CECT in the same patient as
A unique subset of JVT that warrants special
Fig. 22 demonstrates complete occlusion of an enlarged right consideration is Lemierre syndrome (LS), also
IJV with central low-attenuation filling defect (white arrow), known as human necrobacillosis or anaerobic
prominent enhancement of the vasa vasorum (white open postanginal sepsis. Although not originally
arrow), perivascular edema (white asterisks), intramuscular
phlegmon/abscess (black arrowhead) within an enlarged,
described by French bacteriologist André
edematous right sternocleidomastoid muscle (black arrow), Lemierre (1875–1956), this eponymous distinc-
and prominent venous bypass collateral (white arrowhead) tion was awarded to him as a result of his detailed
38 Jugular Vein Thrombosis 911

Fig. 25 Axial CECT in a


76-year-old woman with
recent anterior cervical
fusion presents with neck
pain and fever. Image
reveals central filling defect
in the left IJV (white
arrowhead) compatible
with JVT. Additional
thrombus was seen in the
left ventral epidural venous
plexus (black arrow) and
left-sided muscular
collateral veins (white open
arrow)

Fig. 26 Coronal reconstruction of the NECT data in the


same patient as Fig. 25 better delineates the irregular,
low-attenuation filling defect (white asterisk) seen eccen- Fig. 27 Coronal reconstruction of the NECT data in the
trically within the left IJV as well as peripheral wall same patient as Figs. 25 and 26 better delineates the irreg-
enhancement corresponding to engorged vasa vasorum ular, low-attenuation filling defect (white asterisk) seen
(white arrowhead) eccentrically within the left IJV as well as peripheral wall
enhancement corresponding to engaged vasa vasorum
(white arrowhead)

description of systemic manifestations of Bacillus


funduliformis (now called Fusobacterium septic embolic disease (typically to the lungs),
necrophorum) in 1936. Classically, LS is charac- internal jugular vein thrombophlebitis, and isola-
terized by a constellation of clinical findings that tion of bacteria (typically F. necrophorum) from
include pharyngitis within the preceding 4 weeks, the blood or another sterile body site.
912 N.A. Koontz et al.

Fig. 28 Axial CECT at the


C1 level demonstrates a
filling defect (black arrow)
within the left ventral
epidural space corresponds
with additional thrombus

Fig. 30 Axial T1-WI +C demonstrates filling defect


(white arrow) within the ventral left epidural space at
Fig. 29 Sagittal T1-WI +C demonstrates filling defect C1–C2, compatible with thrombus in the epidural venous
within a left-sided muscular collateral vein (white arrow) plexus
as well as peripheral enhancement within the vessel
resulting from enhancement of the vasa vasorum
ear/mastoid (2 %), larynx (2 %), dental (1 %),
Although most commonly associated with paranasal sinuses (1 %), and orbit (1 %), approx-
F. necrophorum (57 %) or other Fusobacterium imately 25 % of cases originate in the chest or
species (33 %), LS has also been reported with lower respiratory tract [20]. Extraordinarily rare
Staphylococcus aureus (including methicillin- cases of LS originating from the gastrointestinal
resistant), Streptococcus, Peptostreptococcus, tract, genitourinary tract, and musculoskeletal
Proteus, Bacteroides, Klebsiella, and system have also been reported. Presenting symp-
Porphyromonas species [20]. While LS is most toms typically relate to site of involvement, most
often associated with a head and neck site of commonly sore throat, neck pain, or neck mass.
infection (~75 %), including the tonsils (37 %), Several series have described LS to primarily
pharynx/upper respiratory tract (30 %), middle affect previously healthy children, adolescents,
38 Jugular Vein Thrombosis 913

Fig. 31 Axial T1-WI +C in the same patient as Fig. 30


demonstrates peripherally enhancing fluid collections
(black open arrows) in the prevertebral soft tissues, com- Fig. 33 Axial T2-WI in the same patient as Fig. 32 illus-
patible with small abscesses trates a filling defect within the left IJV (white arrowhead)
which is slightly hyperintense to skeletal muscle. The
patient has recently undergone anterior cervical fixation
(white arrow)

progression often advances in a predictable, step-


wise manner through these phases, it is not unex-
pected to find cases where metastatic
complications, such as pulmonary disease, pre-
sent before JVT or JVT appears before
parapharyngeal disease. Early intervention with
prolonged (multi-week) course of antibiotics is
the current therapeutic standard [21, 22]. Thera-
peutic anticoagulation remains controversial,
often reserved for patients with underlying
prothrombotic disorder, secondary cerebral
infarction, or associated cavernous sinus throm-
bosis [21]. Surgical intervention is typically lim-
ited to abscess drainage with IJV ligation rarely
performed in current clinical practice.
Fig. 32 Axial T1-WI +C demonstrates a filling defect
within the left IJV (white arrowhead), which is isointense Perhaps as a result of the widespread use of
to skeletal muscle. The patient has recently undergone antimicrobial agents starting in the mid-1940s, LS
anterior cervical fixation (white arrow) became a vanishingly rare clinical entity through
the 1980s, receiving the moniker of “forgotten
and young adults, with an equal to slightly disease” by many physicians [20, 21, 23].
increased male-to-female predominance [20, 21]. However, since then, there has been a progressive
LS is characterized by four distinct pathophys- increase in the reported LS cases in the medical
iologic phases: primary infection, invasion of the literature. If there is an increased incidence
parapharyngeal (lateral pharyngeal) space, inter- of LS, it may be the result of increased anti-
nal jugular vein thrombophlebitis, and metastatic biotic resistance, evolving antibiotic prescribing
(septic embolic) disease [22]. While disease patterns of physicians, or a reduced number of
914 N.A. Koontz et al.

tonsillectomies. However, it is unclear whether


the increase in reported cases is a true increase in
the incidence of LS or increased detection due to
improved medical imaging [20, 21].
First-line imaging evaluation of suspected LS
is best accomplished with CECT of the neck and
chest, which provides a rapid, comprehensive
evaluation of the primary site of infection (usually
the tonsils), the adjacent deep tissues of the neck,
the IJVs, associated cervical chain lymphadenop-
athy, and assessment for metastatic/septic embolic
disease involving the pulmonary parenchyma
(Figs. 34 and 35). CECT can accurately identify
tonsillar and peritonsillar edema, phlegmon, and
abscess associated with the primary site of infec-
tion as well as elucidate filling defects within the
Fig. 34 Axial CECT at the level of the oropharynx in a IJVs and tributary vessels and perivascular edema
20-year-old man who presents with right-sided neck pain, (thrombophlebitis). Magnetic resonance imaging
fever, and odynophagia demonstrates classic imaging find- (MRI) and magnetic resonance venography
ings of Lemierre syndrome, including nonocclusive throm-
(MRV) can provide useful adjunctive imaging,
bus in the right IJV (white open arrow), edema and
phlegmon replacing the normal triangular-shaped fat of particularly for evaluation of intracranial exten-
the right parapharyngeal space (white arrowhead), and sion of disease. Additionally, duplex Doppler
tonsillopharyngitis with enlarged bilateral palatine tonsils ultrasound can rapidly identify JVT but is limited
(white arrows) with striated enhancement, the so-called
for assessment of secondary complications.
“tiger stripe” sign

Fig. 35 Coronal
reconstruction of the CECT
in the same 20-year-old
patient with Lemierre
syndrome in Fig. 34
demonstrates the
nonocclusive thrombus in
the right IJV (black open
arrow) as well as associated
enlarged, reactive
jugulodigastric lymph
nodes (white arrowhead)
38 Jugular Vein Thrombosis 915

Fig. 36 Axial CECT in a 25-year-old man with gunshot


wound to the left neck demonstrates posttraumatic JVT
(black open arrow) as well as obvious signs of penetrating
trauma, including subcutaneous emphysema (white arrow-
head) and multiple ballistic fragments (black arrows)

Fig. 37 Sagittal reconstruction of CECT of the neck in


Traumatic the same patient as Fig. 36 demonstrates
nonocclusive thrombus in the left IJV (black
Reports of posttraumatic JVT in the medical liter- open arrow), diffuse subcutaneous emphysema (white
ature are exceedingly rare with only a few case arrowhead), and extensive streak artifact from ballistic
fragments (black arrow)
reports documented, more likely due to the mun-
dane nature of traumatic etiologies rather than a
truly low incidence (Figs. 36–38). The few
reported cases have all involved nonpenetrating
trauma to the head or neck, one of which demon- alterations of the Virchow triad that shift patients
strated concurrent thrombosis of the sigmoid toward a prothrombotic state. Underlying
sinus (suggesting propagation of dural sinus hypercoagulability of various etiologies can play
thrombosis into the downstream IJV) [24, 25]. In a critical role in tipping this balance toward JVT,
the authors’ experience, JVT associated with pen- which has been reported in patients with activated
etrating trauma is not uncommon. The true inci- protein C (APC) resistance. APC resistance,
dence of posttraumatic JVT is unknown but is which is thought to account for somewhere
likely higher than the paucity of literature reports between 20 % and 50 % of all thrombotic events
might suggest. The presence of JVT in a trauma in general, may be either congenital (hereditary
patient certainly warrants close inspection of the factor V Leiden or factor V Cambridge gene
dural venous sinuses, especially in the setting of mutations) or may be acquired (resulting from
skull base fractures (Fig. 39). antiphospholipid antibodies, oral contraceptive
use, paraneoplastic syndromes, or due to thalido-
mide use) [26]. Interestingly, JVT has also been
Hematologic reported in patients with hemophilia, which seems
counterintuitive given their underlying bleeding
As previously described (cf. Pathophysiology), disorder. While thrombotic disease in hemophilia
the propensity to develop JVT is entrenched in patients is typically related to concomitant
916 N.A. Koontz et al.

Fig. 38 Coronal
reconstruction from CECT
of the neck in the same
patient as Figs. 36 and 37
performed approximately
6 h later demonstrates
interval progression of
posttraumatic JVT, now
with complete occlusion
and gradual tapering (black
open arrow) of the column
of refluxed iodinated
contrast that retrograde fills
the proximal left IJV

Fig. 39 Axial CT in bone algorithm (a) reveals an oblique extension through the jugular foramen (black arrow).
fracture through the right aspect of the occipital bone in a Axial CECT (b) shows filling defects in the right IJV
pediatric patient who fell from a jungle gym. There is (open black arrows), which are compatible with
diastasis of the occipitomastoid suture (white arrow) and nonocclusive, posttraumatic JVT

vascular catheters, it can also be treatment related, Rheumatologic


seen following therapeutic administration of fac-
tor eight (VIII) inhibitor bypassing activity Uncommonly, rheumatologic disease may result
(FEIBA) and activated recombinant factor VII in JVT. While not strictly limited to patients with
(rFVIIa). underlying rheumatologic disease, the presence of
38 Jugular Vein Thrombosis 917

prothrombotic antiphospholipid antibodies incredibly rare in patients undergoing ART,


(APLAs), such as anticardiolipin antibody and occurring in 0.08–0.11 % of ART treatment cycles
lupus anticoagulant (a misnomer given its [30]. As part of ART, patients receive exogenous
procoagulant effects in vivo), is associated with gonadotropins and gonadotropin-releasing drugs
an increased risk of thromboses (venous greater in order to bolster the development and number of
than arterial) [27]. While seen commonly in ovarian follicles, which can be complicated by
patients with rheumatoid arthritis and systemic OHSS. The OHSS may be severe in 1–2 % of
lupus erythematosus, the prevalence of APLAs patients, resulting in significant ovarian enlarge-
within the general population is estimated ment, ascites, abdominal bloating, nausea, pleural
between 1 % and 5 %; moreover, they are effusions, hypoalbuminemia, hypercoagulability,
observed in 10 % of patients with DVT or pulmo- venous thromboembolism (VTE), renal failure,
nary embolism and in up to 42 % of patients with hypovolemic shock, respiratory distress, and
stroke [28]. Whether APLAs are truly a cause or even death [31].
an effect (or both) of thrombosis is not entirely While the majority of ART patients who develop
clear. Nonetheless, the presence of APLAs should JVT or UEDVT have preceding OHSS, studies
be considered when venous thrombosis occurs in have found that a sizable minority (up to 29 % in
atypical sites like the IJV. one series) do not have clinically apparent OHSS
An extremely rare rheumatologic disorder asso- [30]. The exact mechanism by which ART contrib-
ciated with thrombosis is Behçet disease, which utes to hypercoagulability (and subsequent JVT) is
demonstrates vascular involvement in approxi- not completely understood and is further compli-
mately 25 % of cases. Rarely, Behçet disease has cated by the variability in concurrent OHSS. The
been implicated in spontaneous JVT [29]. Behçet pathogenesis of thrombotic disease with OHSS
patients are at an increased risk of catheter- or likely results from hormonal effects of human cho-
venipuncture-associated endothelial injury and rionic gonadotropin (hCG) and estrogen, which
may benefit from vascular evaluation with nonin- lead to increased vascular permeability, leukocyte
vasive magnetic resonance angiography (MRA) or margination, hemoconcentration, thrombocytosis,
MRV in order to prevent iatrogenic vascular com- and reduced fibrinolysis [31, 32]. Coupled
plications (such as posttraumatic pseudoaneurysm with immobility and recumbent positioning (due
or catheter-related venous thrombosis). to abdominal discomfort from ascites), use
of CVADs, as well as any underlying inherited
thrombophilic disorders (cf. Hematologic), it
Gynecologic becomes readily apparent how these women may
be tipped toward a prothrombotic state that results in
Nonneoplastic gynecologic etiologies make up a JVT [32].
small, but important source of JVT and are Unassisted pregnancy (without ART) can cer-
observed in three patient subgroups: (1) women tainly be viewed as a hypercoagulable state, and
undergoing in vitro fertilization or other assisted spontaneous JVT has been reported rarely in oth-
reproductive techniques (ART) therapy, erwise healthy pregnancies [30]. Nonetheless, the
(2) women with ovarian hyperstimulation syn- presence of thrombotic disease within the IJV,
drome (OHSS), and (3) and pregnant women. subclavian vein, or upper extremity deep veins
One of the striking features of JVT of gynecologic in pregnancy should warrant further investigation
etiology is that it frequently occurs in otherwise into underlying thrombophilia.
healthy women of reproductive age rather than the Lastly, the diagnosis of JVT of gynecologic
stereotypical chronically ill (cancer, hemodialy- etiology warrants careful evaluation of thrombus
sis, etc.) or elderly patients that more commonly extension, as well as assessment for complica-
develop JVT. tions. For reasons that are not entirely clear, preg-
Firstly, it is important to note that JVT or upper nant women with JVT appear to have a
extremity deep vein thrombosis (UEDVT) is predilection for thrombus propagation or
918 N.A. Koontz et al.

pulmonary embolism even in the face of “appro- such as a goiter, can lead to JVT due to mass effect
priate” therapy with heparin or low-molecular- and altered blood flow dynamics.
weight heparin (LMWH), a risk reported between
14 % and 20 % [30]. The mortality rate of
gynecologic-related JVT is unknown, but given Idiopathic
the risk of complications, particularly in gravid
women, inpatient hospital management is In most scenarios with knowledge of the clinical
advised. In this special patient population, therapy context, a causative etiology of JVT will be deter-
requires the use of heparin or LMWH, as warfarin mined in the majority of affected patients. An
crosses the placental barrier and is contraindicated extremely rare entity, spontaneous (idiopathic)
in pregnancy. JVT should be viewed as a diagnosis of exclusion.
There are no well-established guidelines or con-
firmed evidence in the literature to suggest what
Endocrine percentage of patients lack a discrete etiology for
JVT following appropriate clinical work-up.
True endocrine causes of JVT are rare. As previ- However, given the high association with under-
ously discussed, OHSS and ART are important lying malignancy, caution is advised when con-
endocrine-related gynecologic causes of JVT, sidering a diagnosis of idiopathic JVT.
which can affect women of childbearing age. Far
less commonly, it has been reported that patients
with Cushing syndrome (CS) have an increased JVT in Pediatric Patients
risk of VTE, including JVT, which is probably
due to increased levels of factor VIII, factor IX, Venous thrombosis (in particular JVT) in children
von Willebrand factor, fibrinogen, and plasmino- is uncommon and most frequently due to CVADs
gen activator inhibitor-1 [33]. There also appears and infectious etiologies. Other etiologies of JVT in
to be a greater risk of postoperative VTE follow- pediatric patients include postsurgical mass effect
ing transsphenoidal resection of pituitary lesions from cervical or upper mediastinal masses, hema-
in ACTH-dependent CS as compared to ACTH- tologic disorders, trauma, sequelae of congenital
independent CS patients [33]. heart disease, and post-intravenous immunoglobu-
JVT can also be associated with endocrine neo- lin infusion [37, 38] (Figs. 39 and 40). A potential
plasia. Numerous cases of thyroid carcinoma association between cystic fibrosis and venous
directly invading the IJV have been reported in the thrombosis has also been reported.
literature [34–36]. In such cases, increased vascu- Perhaps even more so in children than in
larity within the thrombus may be demonstrated on adults, central venous catheters are the leading
color or power Doppler US [34]. Similarly, intra- culprit for causing JVT [38] (Fig. 41). There is
clot enhancement on CT or MRI should raise con- wide variation in the reported incidence of
cern for tumor thrombus in this patient population catheter-related venous thrombosis in children,
rather than bland thrombus. Additionally, 131I, but concurrent CVAD use has been reported in
67
Ga, and 18FDG uptake can be seen within thyroid up to 60 % of children and up to 90 % of neonates
cancer tumor thrombus, although 18FDG avidity is with venous thrombosis [38]. Studies have found
not specific to thyroid carcinoma or a neoplastic that nearly 90 % of these DVTs involve the IJV,
process [35, 36]. Clinically, tumor thrombus in thy- roughly paralleling the distribution of central
roid carcinoma patients portends a poor prognosis, venous catheter location [37]. The sensitivity
but the actual clinical presentation may vary from (20 %) and specificity (86 %) of clinical diagnosis
an asymptomatic incidental finding to superior vena (i.e., patient history and physical exam) of DVT in
cava syndrome [36]. Lastly, benign thyroid masses, these patients is lacking, thus underscoring the
38 Jugular Vein Thrombosis 919

Fig. 40 Coronal CECT (a) reveals a filling defect com- air (white asterisk) overlying the right cerebral hemisphere.
patible with an intraluminal thrombus in the right trans- Coronal CECT (b) shows extension of the sigmoid sinus
verse and sigmoid sinuses (white arrows) in this recently thrombus into the right IJV (black arrowhead)
postoperative patient as noted by the extra-axial fluid and

A serious, potentially fatal cause of JVT is


Lemierre syndrome, starting from pharyngitis in
a previously healthy child (Fig. 42). JVT has also
been reported in children following mastoiditis.

Imaging

CT

Contrast-enhanced CT (CECT) is a reliable means


of evaluating JVT or, as is often the case, identi-
fying incidental JVT. Just as the clinical presenta-
tion of JVT varies with chronicity of thrombus, so
too does the imaging appearance (Table 4). With
acute JVT, CECT may demonstrate an enlarged
Fig. 41 Digital subtraction angiography in a pediatric IJV with an intraluminal filling defect (Fig. 43).
patient shows a filling defect (black open arrow) in the
proximal right IJV as well as multiple venous collaterals Clot attenuation is variable depending on the state
(black arrows) typical of chronic, catheter-associated JVT of the hemoglobin, but it can be hyperdense in the
acute phase, possibly masking the finding against
importance of radiologic imaging [37]. Due to its the backdrop of intravascular contrast. For this
noninvasive technique and lack of ionizing radia- reason, it is helpful to broaden the windowing on
tion, US remains the modality of choice for imag- the PACS monitors, effectively allowing the radi-
ing pediatric patients in whom there is a concern ologist to “see through” the contrast, similar to
for JVT. evaluating a CT angiogram of the chest for pul-
As previously described (cf. “Infectious”), monary emboli (Fig. 44). While bland (i.e., not
infectious causes of JVT can affect children. associated with tumor) thrombus does not
920 N.A. Koontz et al.

Fig. 42 Axial CECT (a)


demonstrates a peripherally
enhancing, centrally
hypodense collection (black
asterisk) centered in the left
palatine tonsil. Axial CECT
(b) shows obscuration of
the musculofascial planes
between the carotid,
retropharyngeal, and
prevertebral spaces due to
phlegmon (black open
arrows). The left IJV is
partially thrombosed with a
residual crescent of luminal
opacification (white
arrowhead)

Table 4 CT findings of jugular vein thrombosis (JVT) of true thrombus enhancement must be viewed
Acute JVT with suspicion for underlying tumor, although
NECT the finding is not entirely pathognomonic
Frequently normal (Figs. 45 and 46). In the acute phase, there is
Occasionally high-density thrombus commonly perivascular soft tissue edema with
Secondary finding of retropharyngeal edema obscuration of the musculofascial planes and sur-
CECT rounding fat stranding. Additionally, edema/fluid
Filling defect within an enlarged internal jugular within the retropharyngeal space may be seen.
vein (IJV) In the setting of chronic JVT, there is typically
Thin rim of contrast enhancement at periphery a decrease or resolution of the associated
of clot
perivascular edema, as well as a return to normal
Rare enhancing thrombus (bland or tumor
thrombus) or even subnormal IJV luminal diameter (Fig. 47).
Secondary finding of retropharyngeal edema Bland thrombus commonly remains
Chronic JVT nonenhancing and the burden of thrombus is
NECT often better delineated. Recanalization of the
Prominent bypass collaterals thrombus with internal channels of contrast
Return to normal or subnormal IJV caliber enhancement can be seen in some cases, so one
Variable density clot, typically decreases with age must exercise caution when definitively reporting
+/ Resolution of associated edema enhancing clot as tumor thrombus. Prominent
CECT venous collaterals may serve as a physiologic
Filling defect within subnormal to normal size IJV clot bypass and their presence argues for chronic-
+/ Enhancement within thrombus ity of thrombus (Fig. 48).
Recanalization channels Due to the timing of the contrast bolus on
Tumor thrombus
conventional CECT, mixing artifact within the
Prominent bypass collaterals
IJV can occur, which may mimic thrombus
+/ Resolution of associated edema
(Fig. 49). In such cases, secondary findings may
provide a useful discriminator to distinguish
between artifact and true pathology. CT angiogra-
typically enhance, a thin rim of contrast enhance- phy/venography can be considered if findings on
ment can be seen at the periphery of the thrombus, conventional CECT remain equivocal.
secondary to opacification of the engorged vasa JVT can also be diagnosed prospectively on non-
vasorum. In the setting of malignancy, the finding contrast-enhanced CT (NECT), although the
38 Jugular Vein Thrombosis 921

Fig. 43 Axial CECT (a) and coronal reconstruction from including a central filling defect (white arrows) and
CECT (b) of the neck in a 45-year-old man with right-sided marked enhancement of the vasa vasorum (white open
neck pain and swelling for a few days demonstrate an acute arrows)
JVT with complete occlusion of an enlarged right IJV,

Fig. 44 Coronal chest CECT (a, b) in a pediatric patient right JVT. Broadening of the window-level settings on
with a thrombogenic right IJV catheter. The narrowing of PACS (b) reveals the underlying right JVT (white arrow),
the window-level settings on PACS masks the underlying a subtle “corner finding” on this examination

sensitivity is suboptimal. As above, thrombus may US


be hyperdense in the acute phase, but clot attenua-
tion often decreases as it ages. For this reason, true Ultrasound (US) is an excellent means of evalu-
thrombus visualization on NECT is often limited. ating for JVT (Table 5). As it is noninvasive,
Instead, radiologists must rely more heavily on sensitive, quick, (relatively) inexpensive, porta-
secondary signs to make the diagnosis of JVT on ble, and lacking ionizing radiation, it can be
NECT, such as perivascular and retropharyngeal argued that duplex ultrasonography should be
edema in the acute phase and bypass collaterals in the first-line modality for evaluating JVT, partic-
the chronic phase. ularly in children, who are more radiation
922 N.A. Koontz et al.

Fig. 45 Coronal T1-WI –C (a) and T1-WI +C (b) in an Tubular low signal intensity within the tumor corresponds
81-year-old woman with extensive left IJV tumor throm- with high-velocity flow voids (white open arrow), typical
bus from a glomus jugulare paraganglioma. Coronal of a paraganglioma. Coronal T1-WI +C (b) demonstrates
T1-WI –C (a) demonstrates expansion of the entire left avidly enhancing paraganglioma tumor (white arrows)
IJV (partially out of plane of the image) with tumor (black with high-velocity flow voids (open white arrows) filling
arrows) that is slightly hyperintense to skeletal muscle. the left IJV

Fig. 46 Axial T1-WI –C (a) and T2-WI FS (b) in the same whereas tubular hypointense structures represent high-
patient as Fig. 45. Axial T1-WI –C image (a) at the level of velocity flow voids (white open arrow), which comprise
the oropharynx demonstrates expansion of the left IJV with the “pepper.” (b) Axial T2-WI at the level of the orophar-
tumor (white arrow) that is slightly hyperintense to skeletal ynx demonstrates the mass (white arrow) to be
muscle. Focal areas of high intrinsic T1 signal (black hyperintense to skeletal muscle with multiple internal
arrowheads) correspond with subacute hemorrhage, high-velocity flow voids (white open arrows)
manifesting as the “salt” of this “salt and pepper” lesion,

sensitive than adults are. In the acute phase of of the clot in cases of nonocclusive thrombus.
JVT, grayscale US shows an enlarged, incom- Absence of the normal biphasic wave form on
pletely compressible IJV filled with hypoechoic spectral Doppler analysis is a useful secondary
thrombus (Fig. 50). Color or power Doppler US imaging feature.
demonstrates absent flow within bland thrombus, In the chronic phase of JVT, decreased luminal
but intravascular flow can be seen at the margins caliber and increased venous collaterals can be
38 Jugular Vein Thrombosis 923

Fig. 47 Axial CECT of the neck in a 57-year-old woman


with chronic right JVT demonstrates occlusive thrombus
(white arrow) in a narrow caliber IJV as well as enhance-
ment of the vasa vasorum (white open arrow) and a prom-
inent right external jugular vein (white arrowhead) serving
as a bypass collateral

seen on US, just as is seen with CT. Color and


power Doppler modes can be helpful for demon- Fig. 48 Coronal reformat of CECT of the neck in the same
patient as Fig. 47 demonstrates occlusive thrombus (white
strating venous collaterals. The imaging charac- arrow) in a narrow caliber IJV as well as enhancement of
teristics of the clot also change with time, with the vasa vasorum (white open arrow) and a prominent right
chronic thrombus typically demonstrating external jugular vein (white arrowhead) serving as a
increased echogenicity on grayscale US. bypass collateral. The tip of a potentially thrombogenic
central venous catheter can be seen inferiorly as a “corner
finding” (black arrow)

MRI
within an enlarged IJV is typical of acute throm-
Less commonly performed for primary detection bus (Figs. 9, 14, 29, 32, and 33). As with CECT, a
of thrombus, MRI is a useful adjunct modality for thin rim of peripheral contrast enhancement can
assessment of JVT (Table 6). Signal intensity of be seen at the margin of the IJV lumen in acute
the thrombus is dependent upon its molecular JVT secondary to engorged vasa vasorum. In
contents, particularly the state of hemoglobin. chronic JVT, post-contrast T1-WI reveals a nor-
On non-contrast T1-weighted imaging (T1-WI), mal or subnormal-sized IJV with non-enhancing
JVT demonstrates isointensity in the acute phase filling defect in most cases of bland thrombus.
and increases in signal intensity in the subacute Recanalization channels within the clot may be
phase due to increasing methemoglobin, which is seen. Just as with CT, the presence of enhancing
first intracellular and then extracellular (Figs. 13 clot in the setting of tumor-related JVT should
and 15). Adding fat saturation to the T1-WI will raise the concern for vascular invasion and
increase the conspicuity of the intraluminal tumor thrombus. Also just like CT, the presence
thrombus by nulling the signal intensity from the of prominent venous collaterals is a useful dis-
perivascular fat. On post-contrast T1-WI, criminator for determining the chronicity of a clot.
non-enhancing low signal intensity thrombus On T2-weighted imaging (T2-WI), thrombus can
924 N.A. Koontz et al.

Fig. 49 Mixing artifact mimicking JVT. CECT of the there is incomplete opacification of the more proximal IJV
neck in a 51-year-old woman demonstrates an apparent (c) in keeping with mixing of contrast-opacified and non-
filling defect in the left IJV (white arrow) that looks like contrast-opacified blood. In equivocal cases, delayed-
a nonocclusive thrombus on axial image (a). However, phase imaging or second-look ultrasound are useful for
careful review of the coronal images (b) demonstrates the characterization
“filling defect” to have a more feathery appearance, and

Table 5 Ultrasound findings of jugular vein thrombosis MR venography (MRV) can be utilized for
(JVT) evaluating the extent of thrombus. Traditionally,
Acute JVT this has been performed utilizing two-dimensional
Enlarged, incompletely compressive internal jugular time-of-flight (TOF) imaging which does not
vein (IJV) require administration of intravenous contrast.
Hypoechoic luminal content – thrombus On TOF MRV, a saturation pulse is applied to
Absent flow within thrombus on color or power
diminish signal from the stationary soft tissues
Doppler
and, importantly, from the inflowing arterial blood.
May maintain flow at periphery if nonocclusive
thrombus The lack of flow-related enhancement at the level of
Absent biphasic wave form on spectral Doppler the JVT manifests as segmental signal absence
Chronic JVT within the region of the IJV (Figs. 10 and 11).
Decreased luminal caliber A potential pitfall of TOF MRV is subacute
Prominent bypass collaterals thrombus, which contains methemoglobin and
Increased echogenicity of thrombus consequently demonstrates intrinsic high signal.
Internal flow within thrombus secondary to Dynamic contrast-enhanced MRV (e.g., 3D spoiled
recanalization/organization gradient echo (SPGR) technique) has become more
widely used and is less susceptible to this sort of
diagnostic misinterpretation.
be bright in the hyperacute phase, becoming
hypointense in the acute phase due to the
deoxyhemoglobin (Fig. 14). The T2 signal inten- Nuclear Medicine
sity becomes more hypointense as the clot ages.
Adding fat saturation to conventional fast spin Prior to the widespread use of cross-sectional (CT,
echo, T2-WI is particularly useful for assessing MR, or US) imaging for assessment of upper or
secondary findings of JVT, including edema lower extremity venous thrombosis, radionuclide
within the perivascular soft tissues and the venography was a relatively common practice.
retropharyngeal space. Marked low signal inten- With this technique, a radioactive blood pool
sity susceptibility artifact (“blooming” artifact) (i.e., 99mTc-labeled red blood cells) or venous
can be seen within the thrombus on T2* blood flow (i.e., 99mTcO4 , 99mTc-MAA, or
99m
gradient-recalled echo (GRE) imaging. Tc-DTPA) agent is used to evaluate for
38 Jugular Vein Thrombosis 925

Fig. 50 Grayscale ultrasound (a) of the right IJV in the thrombus. Color Doppler ultrasound (b) of the right IJV in
longitudinal plane demonstrates an eccentric, echogenic, the longitudinal plane illustrates flow in the vessel around
intraluminal filling defect (white arrow) in an incompletely the small intraluminal thrombus
compressible vessel. This is concerning for a nonocclusive

Table 6 MRI findings of jugular vein thrombosis (JVT) patency of an affected venous channel. Reduced
radiotracer activity within a vein relative to the
T1-WI –C FS
Acute – isointense thrombus
contralateral side or relative to an ipsilateral con-
Early subacute – hyperintense due to intracellular tiguous venous segment is suggestive of venous
methemoglobin thrombosis, as is delayed transit time, venous
Late subacute – hyperintense due to extracellular reflux, and increased activity within collateral
methemoglobin channels [39]. While this technique is utilized
T1-WI +C primarily for detection of DVT involving the
Acute extremities, it theoretically could be applied for
Hypo-/isointense thrombus in enlarged internal the evaluation of the deep veins of the neck and
jugular vein (IJV)
might be considered in cases when cross-sectional
+/ Thin rim of peripheral enhancement
imaging cannot be performed or is nondiagnostic.
Chronic
Filling defect within subnormal to normal caliber
Currently relegated to a historical footnote in
IJV DVT imaging, nuclear medicine studies may be
+/ Thrombus enhancement making somewhat of a comeback. While cross-
Organized clot sectional imaging modalities are the first-line
Tumor thrombus modality for making the diagnosis of JVT, new
T2-WI molecular imaging techniques may prove valu-
Hyperacute – hyperintense thrombus able for determining the acuity of a thrombus.
Acute – hypointense thrombus The basic concept is simple: find and exploit a
Early subacute – hypointense thrombus molecular marker that is present in acute throm-
Late subacute – hyperintense due to extracellular bus, but which is not found in organized clot.
methemoglobin
Several radiopharmaceuticals have been targeted,
Chronic – diminishing T2 signal as clot ages
including radiolabeled fibrinogen, platelets,
GRE
Marked low signal intensity
antiplatelets, antifibrin, fibrinolytic enzymes
Often “blooming” artifact (streptokinase and urokinase), soluble fibrin, frag-
MRV ment E1, plasmin, tissue plasminogen activator,
Acute – filling defect in IJV heparin, and multiple synthetic peptides [40]. Cur-
Chronic – filling defect with bypass collaterals rently, one such agent, 99mTc-apcitide (AcuTect,
Diatide Inc, Londonderry, NH), a GPIIb/IIIa
926 N.A. Koontz et al.

Fig. 51 A forty-six-year-old with history of pancreatic to background hepatic activity but slightly increased com-
adenocarcinoma and catheter-associated thrombus in the pared to blood pool activity. Lack of FDG uptake within
right IJV. Coronal reconstruction of CECT (a) demon- the clot itself argues against tumor thrombus. Incidental
strates occlusive filling defect in the right IJV (white note is made of a radiolabeled pulmonary embolus in the
arrow) with enhancement of the vasa vasorum (white left lung (black arrowhead on b and c), the so-called “hot
open arrow). Coronal fused FDG-PET/CT image (b) dem- clot” sign. This is due to accidental tagging of catheter-
onstrates a rim of increased FDG avidity at the periphery of associated thrombus at time of intravenous 18F-FDG
the clot (white arrowhead), corresponding to the engorged administration, which subsequently embolized to the lung
vasa vasorum and surrounding perivascular inflammation. (Image courtesy of Steven Westphal, M.D., Indiana Uni-
Coronal FDG-PET image (c) demonstrates that the periph- versity School of Medicine, Department of Radiology and
eral FDG avidity (white arrowheads) is similar in activity Imaging Sciences)

receptor antagonist, is approved by the Food and heterogeneous etiologies: postsurgical, catheter-
Drug Administration for clinical use, and other associated, posttraumatic, ART- or pregnancy-
similar agents remain under investigation associated, APLA-associated, or even malignancy-
[41]. While the efficacy of such molecular agents associated hypercoagulability [43]. Given lack
has not been reported specific to JVT (solely of specificity of FDG-PET for both metabolically
reported for extremity DVT), it stands to reason active and non-metabolically active thrombi,
that the mechanism of action (e.g., binding of JVT encountered on FDG-PET studies requires
activated platelets in acute thrombosis) should diligent review of associated anatomic imaging
allow for detection of acute thrombus in the IJV. and correlation with clinical history.
More commonly, JVT is encountered as an inci- Detection of IJV tumor thrombus by 131I scin-
dental finding on nuclear medicine studies tigraphy for papillary thyroid carcinoma and 67Ga
performed for other reasons. In such cases, radio- scintigraphy for anaplastic thyroid carcinoma has
tracer accumulation along the lateral neck localizes been reported [35, 36]. Radioiodine-avid throm-
to the thrombus within the IJV. This is probably bus has a high specificity for well-differentiated
encountered with greatest frequency on FDG-PET/ (papillary, follicular, or mixed origin) thyroid neo-
CT, but one must keep in mind that FDG accumu- plasm but not for anaplastic or medullary thyroid
lation is nonspecific to etiology, occurring in areas carcinoma due to lack of iodine concentration in
with neoplasm, infection, and inflammation [42, 43] these tumors. While the finding of 67Ga accumu-
(Figs. 51 and 52). On the other hand, a significant lation in the IJV should raise concern for JVT, its
portion of JVT are not metabolically active on specificity for anaplastic thyroid carcinoma as a
FDG-PET, reflecting bland thrombus of primary tumor of origin is questionable.
38 Jugular Vein Thrombosis 927

Fig. 52 A forty-six-year-old with history of pancreatic avidity at the periphery of the clot (white arrowhead),
adenocarcinoma and catheter-associated thrombus in the corresponding to the engorged vasa vasorum and surround-
right IJV. Axial CECT image (a) demonstrates occlusive ing perivascular inflammation. Lack of FDG uptake within
filling defect in the right IJV (white arrow) with enhance- the clot itself argues against tumor thrombus (Image cour-
ment of the vasa vasorum (white open arrow). Axial tesy of Steven Westphal, M.D., Indiana University School of
FDG-PET image (b) demonstrates a rim of increased FDG Medicine, Department of Radiology and Imaging Sciences)

Angiography

Retrograde jugular venography was originally


described by Swedish authors in the early 1960s
and pioneered throughout the remainder of the
decade. Prior to the introduction of cross-sectional
imaging, it was utilized primarily for evaluation of
carotid space and base of skull tumors but is also
efficacious for detection of JVT (Fig. 53). A sen-
sitive, but invasive procedure typically employing
common femoral vein puncture and placement of
an angiographic catheter in the internal jugular
vein, conventional retrograde venography is
rarely employed for primary detection of JVT in
current clinical practice. Additionally, retrograde
venography runs the risk of dislodging the throm-
bus with subsequent downstream embolization if
not performed carefully.
Fig. 53 Digital subtraction angiography (DSA) of the left
Presently, JVT is identified by interventional upper extremity in a 45-year-old woman with “uphill
radiologists during the exchange of thrombosis” of the left IJV demonstrates filling defects
malfunctioning central venous catheters due to (black arrows) within the left subclavian and
brachiocephalic veins with no retrograde filling of the
the high frequency of catheter-associated throm-
completely occluded left IJV. Numerous IJV bypass col-
bus (Fig. 41). The actual imaging finding of JVT laterals (black open arrows) are also seen (Image courtesy
on conventional venography parallels that of of Perry P.Ng, MBBS, University of Utah Health Sciences,
cross-sectional modalities with a discrete filling Department of Radiology)
defect found within the IJV, which may be occlu-
sive or nonocclusive. As observed on CT, MRI, Although successful endovascular interven-
and/or US, acute clot tends to enlarge the vessel, tional treatments for JVT have been reported,
whereas chronic JVT typically results in a smaller medical management with anticoagulation is
caliber vessel with prominent venous bypass favored (Fig. 54). In a small study of 12 patients
collaterals. with symptomatic, occlusive JVT, catheter-
928 N.A. Koontz et al.

Fig. 54 (a) Digital subtraction angiography (DSA) just demonstrates marked improvement in opacification of the
prior to left IJV suction thrombectomy demonstrates filling left IJV with minimal residual filling defects (black arrow)
defects (black arrows) within the left IJV as well as numer- as well as reduction in venous shunting through numerous
ous bypass collaterals (black open arrows). The suction bypass collaterals (black open arrows) (Image courtesy of
thrombectomy catheter is noted (white open arrow in both Perry P.Ng, MBBS, University of Utah Health Sciences,
a and b). DSA following suction thrombectomy (b) Department of Radiology)

directed thrombolysis utilizing recombinant tissue facets of medical imaging, a few broad categories
plasminogen activator was shown to be safe and of errors may account for misdiagnosis: failures in
effective for treatment of JVT regardless of phys- systems-based practice, errors of perception,
ical length of clot [44]. Thrombolysis was more errors of judgment, and technical limitations/arti-
effective in patients with acute thrombus (less facts. For instance, a simple clinical history, such
than 2 weeks in age), and 50 % of treated patients as “neck pain and swelling – concern for JVT,”
demonstrated a patent IJV at 2-month follow-up. would be extremely helpful for directing the
However, IJV patency was maintained in only interpreting radiologist to search for JVT. Unfor-
25 % of patients with persistent CVAD use tunately, the presentation of JVT is variable and
[44]. Successful mechanical thrombectomy of the diagnosis may not be considered by the refer-
JVT has also been reported in small patient ring clinician. Furthermore, even if the clinician is
populations with several devices, including the suspicious of JVT, it is certainly not uncommon
use of the Oasis mechanical thrombectomy sys- for pertinent history to become truncated along
tem (Medi-Tech/Boston Scientific, Watertown, the workflow pathway from the ordering clinician
MA), rotatable pigtail catheters, and thromboas- to the interpreting radiologist (a failure in
piration techniques [45]. Further investigation systems-based practice). Misinterpretation of
into the safety, efficacy, and durable results of imaging studies may be due to lack of specific
endovascular interventional therapy of JVT will attention to the appearance of the IJV (error of
be necessary to determine its place in the manage- perception), which lies toward the edge of the
ment of this disorder. images rather than in the center of the reader’s
focus. Furthermore, the proximal and distal por-
tions of the IJV are at the beginning and end of an
Diagnostic Pitfalls imaging series, respectively, and may be inadver-
tently overlooked. To avoid misdiagnosis of JVT,
While identifying the finding of JVT is often it is important to include venous structures in the
straightforward (especially when one remembers search pattern when reviewing studies. Some-
to look for it), there are several pitfalls that may times the radiologist recognizes the findings of
result in the misdiagnosis of JVT. As with other JVT, such as increased luminal signal on MRI,
38 Jugular Vein Thrombosis 929

but interprets the pathologic findings as some- present as a filling defect within the IJV. While a
thing benign, such as slow flow (error in judg- filling defect is a common appearance of throm-
ment). Lastly, all imaging modalities have bus, a chronically thrombosed IJV can demon-
intrinsic technical limitations and artifacts that strate homogeneous enhancement because of
may obfuscate the diagnosis of JVT, and it is capillary channels within the clot.
important to be aware of these to improve diag-
nostic accuracy. Although most of the literature
description of missed diagnoses (i.e., false- False-Positive Misinterpretation
negative studies) and misdiagnoses (i.e., false-
positive studies) of venous thrombosis in neuro- Although very difficult to call prospectively on
imaging involves the dural venous sinuses and NECT, especially in the absence of secondary
cerebral veins, the technical reasons such misin- findings such as edema in the surrounding soft
terpretations occur are also applicable to JVT tissues or bypass collaterals, hyperdense throm-
imaging. bus may occasionally be identified. However, one
must keep in mind that hyperdense clot within the
IJV may be masked if a patient has an elevated
False-Negative Misinterpretation hematocrit, which may be due to dehydration or a
hypercoagulable status. Hemoconcentration has
On unenhanced CT, it may be challenging to been shown to correlate with CT attenuation
delineate the tubular lumen of the IJV from the [47]. Absolute Hounsfield unit values greater
surrounding soft tissue structures, particularly than 65, Hounsfield unit to hematocrit ratios
lymph nodes. Additionally, evolution of greater than 1.7, and venoarterial Hounsfield unit
intraluminal blood products may cause the throm- value differences greater than 15 are useful
bus to appear isodense to adjacent soft tissues adjuncts to NECT of the head for the diagnosis
rather than hyperdense. Occasionally on CECT, of dural venous thrombosis [48]. Although correl-
a high-density thrombus may not be visible ative values have not been reported with regard to
against the background of contrast opacification, JVT, it stands to reason that similar findings
wall enhancement (vasa vasorum), or partial should be seen. Lastly, in pediatric patients, one
recanalization of the IJV [46]. There may also be must keep in mind their physiologically normal,
a false-negative study if the timing of scanning higher hematocrit, which could mimic thrombus.
after contrast administration is inappropriate. For When in doubt, one should seek other clues that
example, if the CECT is performed too soon after the hyperdensity is related to hemoconcentration
contrast administration, there will be decreased rather than true thrombosis, such as homogeneity
contrast resolution between the thrombus and the of the hyperattenuation and symmetry of involve-
opacified blood. ment of all other venous channels.
On MRI, the challenge of diagnosing JVT lies On CECT, one of the most common reasons for
in the fact that the signal intensity of thrombus can a false-positive finding of JVT is the so-called
simulate normal blood flow. For example, acute mixing artifact, which is caused by mixing of
thrombus may appear hypointense on T2-WI, contrasted and non-contrasted blood and results
simulating a flow void of a patent vessel. As the in the appearance of a filling defect [49]. Mixing
blood products evolve into methemoglobin, the artifact typically demonstrates a swirling, irregu-
thrombus may appear hyperintense on T1-WI and lar, or discontinuous configuration, while true
TOF MRV. As a word of caution, TOF MRV thrombus typically is well defined, linear, and
should always be read in conjunction with the contiguous (Figs. 49 and 55). Although this usu-
other MRI sequences to avoid misinterpreting ally does not result in a major diagnostic dilemma,
the hyperintense signal in the thrombus as flow- delayed-phase imaging or second-look US are
related enhancement. Lastly, on contrast- useful options to resolve true thrombus from
enhanced T1-WI, one typically expects JVT to such a pseudolesion in difficult cases. Other
930 N.A. Koontz et al.

Fig. 55 Coronal reconstruction of CECT of the neck Fig. 56 Axial T1-WI +C at the level of the right superior
demonstrates the typical appearance of “mixing” artifact. jugular bulb exhibits a focal intraluminal hypointensity,
The appearance of feathery, swirling low attenuation which is suggestive of a filling defect (white arrowhead).
within the bilateral IJVs (white open arrows) is dependent However, this artifactual pseudolesion is caused by turbu-
on the timing of the contrast bolus and results from mixing lent flow and can easily be misdiagnosed as JVT. In equiv-
of contrast-opacified and non-contrast-opacified blood. ocal cases, SPGR +C or CTV can be performed for
This should not be mistaken for thrombus, and in equivocal clarification
cases, delayed-phase imaging or second-look ultrasound
can be considered for further characterization

common reasons for the false-positive diagnosis signal from inflowing protons. In the coronal
of JVT include vascular compression and surgical plane, this can be seen in the IJV and distal sig-
ligation. In the neck, the IJV is a thin-walled, moid sinus. Phase-contrast venography can be
pliable structure, which may be extrinsically com- helpful to differentiate between true occlusion
pressed by surgical or radiation treatment-related and slow flow as it demonstrates blood flow char-
scar or by a cervical soft tissue mass, including acteristics, including directionality, and is not
lymphadenopathy, which can give the appearance affected by methemoglobin hyperintensity.
of occlusion. Alternatively, it may be ligated in Lastly, there is often asymmetry of the path-
certain types of surgical dissections. One must ways of cerebral venous return, including the
keep in mind that the absence of contrast dural venous sinuses and internal jugular veins,
opacification of the IJV does not equate to throm- which can be seen in up to 25 % of normal sub-
bosis, and it is important to follow the IJV through jects and should not be confused for underlying
its course to identify the location of external com- thrombotic pathology [50] (Fig. 57). Although the
pression and return to normal luminal caliber. depiction of venous collateral flow has been
On MRI, false-positive cases can be seen with described as an indirect diagnostic parameter in
increased IJV signal intensity due to slow flow, patients with cerebral and jugular venous throm-
turbulent flow, in-plane flow, and entry-flow phe- bosis, complex variability of cerebrovenous anat-
nomenon on spin echo sequences, most com- omy may be confounding and require a
monly (Fig. 56). In-plane flow phenomenon multimodality (CTV, MRV, US, or catheter angi-
refers to flow parallel to the plane of acquisition, ography) approach to reach the appropriate
which results in loss of signal due to saturation of diagnosis.
38 Jugular Vein Thrombosis 931

References
1. Valecchi D, Bacci D, Gulisano M et al (2010) Internal
jugular vein valves: an assessment of prevalence, mor-
phology and competence by color Doppler echography
in 240 healthy subjects. Ital J Anat Embryol
115(3):185–189
2. Malcom GE, Raio CC, Poordabbagh AP, Chiricolo GC
(2008) Difficult central line placement due to variant
internal jugular vein anatomy. J Emerg Med
35:189–191
3. Dickson BC (2004) Venous thrombosis: on the
history of Virchow’s triad. Univ Tor Med J 81
(3):166–171
4. Pata YS, Ünal M, G€ ulhan S (2008) Internal jugular
vein thrombosis due to distant malignancies: two case
reports and literature review. J Laryngol Otol
122:318–320
5. Gisolf J, van Lieshout JJ, van Heusden K, Pott F, Stok
WJ, Karemaker JM (2004) Human cerebral venous
outflow pathway depends on posture and central
venous pressure. J Physiol 560(Pt 1):317–327
6. Volturo GA, Repeta RJ (2001) Non-lower extremity
deep vein thrombosis. Emerg Med Clin North Am
19(4):877–893
Fig. 57 Coronal maximum intensity projection image 7. Gbaguidi X, Janvresse A, Benicou J, Cailleux N,
from a dynamic (time-resolved) MRA of the neck demon- Levesque H, Marie I (2011) Internal jugular vein
strates anatomic variation of the right IJV, which is focally thrombosis: outcome and risk factors. Q J Med
narrowed (white arrow). This benign variant is not uncom- 104:209–219
monly seen and should not be confused for JVT 8. Yardim H, Erkoc R, Soyoral YU, Begenik H, Avcu S
(2012) Assessment of internal jugular vein thrombosis
due to central venous catheter in hemodialysis patients:
a retrospective and prospective serial evaluation with
ultrasonography. Clin Appl Thromb Hemost 18(6):
662–665
Conclusion 9. Arhi CS, Buchanan MA, Allen SA, Pickles J (2010)
Internal jugular vein thrombosis secondary to a perma-
JVT is a not uncommon and serious finding nent cardiac pacemaker: an unusual case of lateral neck
swelling. J Laryngol Otol 124:916–918
encountered by imagers. Although not usually a 10. Yurtseven T, Erşahin Y, Kitiş Ö, Mutluer S (2005)
diagnostic dilemma, the sundry etiologies (iatro- Thrombosis and thrombophlebitis of the internal
genic, neoplastic, infectious, traumatic, hemato- jugular vein as a very rare complication of ventricu-
logic, rheumatologic, gynecologic, endocrine, loatrial shunt. Clin Neurol Neurosurg 107:144–146
11. Agraharkar M, Isaacson S, Mendelssohn D et al (1995)
and idiopathic), the potentially devastating com- Percutaneously inserted silastic jugular hemodialysis
plications (such as pulmonary embolism, septic catheters seldom cause jugular vein thrombosis.
emboli, and dural sinus thrombosis), and the high ASAIO J 41(2):169–172
association with neoplasm warrant additional 12. Harada H, Omura K, Takeuchi Y (2003) Patency and
caliber of the internal jugular vein after neck dissection.
investigation into the etiology and associated Auris Nasus Larynx 30:269–272
complications on the part of the radiologist. 13. Sheikh MA, Topoulos AP, Deitcher SR (2002) Isolated
While CECT remains the first-line modality for internal jugular vein thrombosis: risk factors and natu-
imaging JVT, both MRI/MRV and duplex Dopp- ral history. Vasc Med 7:177–179
14. Vormittag R, Simanek R, Ay C et al (2009) High factor
ler US can add important information and play a VIII levels independently predict venous thromboem-
valuable role in imaging evaluation of these bolism in cancer patients. Arterioscler Thromb Vasc
patients. Biol 29:2176–2181
932 N.A. Koontz et al.

15. Kanz R, Vukovich T, Vormittag R et al (2011) Throm- 32. Arya R, Shehata HA, Patel RK et al (2001) Internal
bosis risk and survival in cancer patients with elevated jugular vein thrombosis after assisted conception ther-
C-reactive protein. J Thromb Haemost 9:57–63 apy. Br J Haematol 115:153–155
16. Cheang PP, Fryer J, Ayoub O, Singh V (2004) Sponta- 33. Stuijver DJF, van Zaane B, Feelders RA et al (2011)
neous bilateral internal jugular vein thrombosis: a sign Incidence of venous thromboembolism in patients with
of metastasis. J Laryngol Otol 118:570–572 Cushing’s syndrome: a multicenter cohort study. J Clin
17. Chen M-H, Chang PM-H, Chen P-M et al (2010) Endocrinol Metab 96:3525–3532
Prolonged facial edema is an indicator of poor progno- 34. Panzironi G, Rainaldi R, Ricci F, Casale A, Macciucca
sis in patients with head and neck squamous cell car- MDV (2003) Gray-scale and color Doppler findings in
cinoma. Support Care Cancer 18:1313–1319 bilateral internal jugular vein thrombosis caused by
18. Veras E, Srodon M, Neijstrom ES, Ronnett BM (2009) anaplastic carcinoma of the thyroid. J Clin Ultrasound
Metastatic HPV-related cervical adenocarcinomas 31(2):111–115
presenting with thromboembolic events (Trousseau 35. Yoshimura M, Kawamato A, Kakasone K et al (2003)
syndrome): clinicopathologic characteristics of Gallium-67 accumulation to the tumor thrombus in
2 cases. Int J Gynecol Cancer 28:134–139 anaplastic thyroid cancer. Ann Nucl Med 17
19. Lin D, Reeck JB, Murr AH (2004) Internal jugular vein (8):689–691
thrombosis and deep neck infection from intravenous 36. Mugunthan N, Kumar AR, Thomas P (2010) Tumor
drug use: management strategy. Laryngoscope thrombus in the great veins from papillary carcinoma
114:56–60 of the thyroid: 131-I scan findings. J Clin Endocrinol
20. Karkos PD, Asrani S, Karkos CD et al (2009) Metab 95:2527–2528
Lemierre’s syndrome: a systematic review. Laryngo- 37. Hanslik A, THom K, Haumer M et al (2008) Incidence
scope 119:1552–1559 and diagnosis of thrombosis in children with short-term
21. Wright WF, Shiner CN, Ribes JA (2012) Lemierre central venous lines of the upper venous system. Pedi-
syndrome. South Med J 105(5):283–288 atrics 122:1284–1291
22. Chirinos JA, Lichtenstein DM, Garcia J, Tamariz LJ 38. Male C, Kuhle S, Mitchell L (2003) Diagnosis of
(2002) The evolution of Lemierre syndrome: report of venous thromboembolism in children. Semin Thromb
2 cases and review of the literature. Medicine Hemost 29:377–390
81(6):458–465 39. Khosa F, Otero HJ, Prevedello LM, Rybicki FJ, Di
23. O’Brien WT, Lattin GE, Thompson AK (2006) Salvo DN (2010) Imaging presentation of venous
Lemierre syndrome: an all-but-forgotten disease. AJR thrombosis in patients with cancer. AJR Am J
Am J Roentgenol 187:W324 Roentgenol 194:1099–1108
24. Duke BJ, Ryu RK, Brega KE, Coldwell DM (1997) 40. Taillefer R (2001) Radiolabeled peptides in the detec-
Traumatic bilateral jugular vein thrombosis: case report tion of deep venous thrombosis. Semin Nucl Med
and review of the literature. Neurosurgery 41 31(2):102–123
(3):680–683 41. Bates SM, Lister-James J, Julian JA, Taillefer R, Moyer
25. Caplan JM, Kalpey Z, Gates J (2008) Closed traumatic BR, Ginsberg JS (2003) Imaging characteristics of a
head injury: dural sinus and internal jugular vein novel technetium Tc 99m-labeled platelet glycoprotein
thrombosis. Emerg Med J 25(11):777–778 IIb/IIIa receptor antagonist in patients with acute deep
26. Mnejja M, Hammami B, Bougacha L et al (2011) Inter- vein thrombosis or a history of deep vein thrombosis.
nal jugular vein thrombosis by protein C activated Arch Intern Med 163:452–456
resistance. Eur Ann Otorhinolaryngol Head Neck Dis 42. Kikuchi M, Yamamoto E, Shiomi Y, Kikuchi M,
128:269–271 Yamamoto E, Shiomi Y et al (2004) Internal and exter-
27. Provenzale JM, Ortel TL, Allen NB (1998) Systemic nal jugular vein thrombosis with marked accumulation
thrombosis in patients with antiphospholipid anti- with FDG. Br J Radiol 77:888–890
bodies: lesion distribution and imaging findings. AJR 43. Sopov V, Bernstine H, Stern D, Yefremov N, Sosna J,
Am J Roentgenol 170:285–290 Groshar D (2009) The metabolic spectrum of venous
28. Misita CP, Moll S (2005) Antiphospholipid antibodies. thrombotic disorders found on PET/CT. AJR Am J
Circulation 112:e39–e44 Roentgenol 193:W530–W539
29. Danaci M, Akpolat T, Koyuncu M, Unal R, Belet U 44. Chang R, Horne MK, Mayo DJ, Doppman JL (1996)
(2000) The advantages of MRI and MRA for diagnos- Pulse-spray treatment of subclavian and jugular venous
ing Behçet’s disease and internal jugular vein throm- thrombi with recombinant tissue plasminogen activa-
bosis. Comput Med Imaging Graph 24:121–124 tor. J Vasc Interv Radiol 7(6):845–851
30. Chan WS, Ginsberg JS (2006) A review of upper 45. Tajima H, Murata S, Kumazaki T, Ichikawa K, Tajiri T,
extremity deep vein thrombosis in pregnancy: Yamamoto Y (2004) Successful interventional treat-
unmasking the ‘ART’ behind the clot. J Thromb ment of acute internal jugular vein thrombosis. AJR
Haemost 4:1673–1677 Am J Roentgenol 182:467–469
31. Fleming T, Sacks G, Nasser J (2012) Internal jugular 46. Provenzale JM, Kranz PG (2011) Dural sinus throm-
vein thrombosis following ovarian hyperstimulation bosis: sources of error in image interpretation. AJR Am
syndrome. Aust N Z J Obstet Gynaecol 52:87–90 J Roentgenol 196(1):23–31
38 Jugular Vein Thrombosis 933

47. Black DF, Rad AE, Gray LA, Campeau NG, Kallmes 49. Takhtani D (2005) CT neuroangiography: a glance at
DF (2011) Cerebral venous sinus density on the common pitfalls and their prevention. AJR Am J
noncontrast CT correlates with hematocrit. AJNR Am Roentgenol 185:772–783
J Neuroradiol 32:1354–1357 50. Ayanzen RH, Bird CR, Keller PJ, McCully FJ,
48. Besachio DA, Quigley EP 3rd, Shah LM, Salzman KL Theobald MR, Heiserman JE (2000) Cerebral MR
(2013) Noncontrast computed tomographic Hounsfield venography: normal anatomy and potential
unit evaluation of cerebral venous thrombosis: a quan- diagnostic pitfalls. AJNR Am J Neuroradiol 21
titative evaluation. Neuroradiology 55(8):941–945 (1):74–78
Imaging of Cerebral Venous and Sinus
Thrombosis 39
Jennifer Linn

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 936 Cerebral venous and sinus thrombosis (CVST)
constitutes a rare but important cause of stroke
Subtypes of CVST . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 936
with an annual incidence of approximately
Clinical Signs and Symptoms . . . . . . . . . . . . . . . . . . . . . . 938 three to four cases per one million adults. In
Laboratory Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 938 recent decades, the clinical outcome of CVST
has significantly improved, largely due to
Prognosis and Outcome . . . . . . . . . . . . . . . . . . . . . . . . . . . . 938
improved neuroimaging methods. However,
Imaging Features of CVST . . . . . . . . . . . . . . . . . . . . . . . . 939 the average interval from symptom onset to
Direct and Indirect Neuroimaging Signs . . . . . . . . . . . . 939
Digital-Subtraction Angiography (DSA) . . . . . . . . . . . . 939 definite diagnosis is still 7 days. This is mainly
Magnetic Resonance Imaging . . . . . . . . . . . . . . . . . . . . . . . 940 due to its subacute onset and its unspecific
Computed Tomography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 945 clinical presentation. To avoid delayed diagno-
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 950 sis, it is essential to consider CVST as a differ-
ential diagnosis in patients presenting with
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 951
unspecific neurological symptoms, particularly
headache, other signs of elevated intracranial
pressure, and seizures.
Magnetic resonance imaging (MRI) and
computed tomography (CT) with CT angiog-
raphy provide excellent techniques for the
diagnosis of sinus or deep cerebral venous
thrombosis. The diagnosis of cortical venous
thrombosis is particularly challenging and
requires T2*WI MRI sequences. Knowledge
of potential diagnostic pitfalls and artifacts is
essential to avoid misinterpretation of MRI
or CT.

Keywords
Cerebral Venous thrombosis • Sinus thrombo-
J. Linn (*)
sis • Imaging • MRI • CT
Institute of Neuroradiology, University Hospital Carl
Gustav Carus, Dresden, Dresden
e-mail: jennifer.linn@uniklinikum-dresden.de

# Springer Science+Business Media New York 2016 935


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_29
936 J. Linn

Introduction or more risk factors are present, while in 15–20 %


of all patients, no predisposing factors can be
Cerebral venous and sinus thrombosis (CVST) is identified [2].
a relatively rare but important cause of stroke with
an estimated annual incidence of three to four
cases per one million in adults and approximately
seven cases per one million in children. Although Subtypes of CVST
young and middle-aged women are most com-
monly affected, CVST can occur in all age groups Based on the location of the thrombosis, i.e., the
and in both sexes [1]. affected venous structures, three subtypes of
The most important predisposing factors and CVST should be distinguished, which differ with
causes of CVST include (acquired or congenital) regard to their clinical presentation and prognosis
coagulation disorders, intake of oral contraceptive (Fig. 1). In addition, the value of the different
drugs, pregnancy, lactation, and ear, nose, and imaging modalities and MRT sequences differs
throat infections. In over 40 % of patients, two considerably depending on the site of the

Fig. 1 Three subtypes of cerebral venous and sinus Galen and its tributaries, indicated in blue in b), and corti-
thrombosis (CVST). Based on the affected venous struc- cal venous thrombosis (= thrombosis of the superficial
tures, the following subtypes of CVST can be distin- cortical veins including the anastomotic vein of Labbé
guished: dural sinus thrombosis (= thrombotic occlusion indicated in yellow in c). The straight sinus (arrow in b)
of one or more dural sinuses, indicated in red in a), deep is included with the deep cerebral veins by most authors.
cerebral venous thrombosis (= thrombosis of the vein of Arrows in (c) indicate the anastomotic vein of Labbé
39 Imaging of Cerebral Venous and Sinus Thrombosis 937

Fig. 2 Deep cerebral


venous thrombosis. T2*WI
gradient-echo images of a
27-year-old female patient
with thrombosis of the
internal cerebral veins
(normal white arrows in
b and c), the vein of Galen
(normal white arrow in d),
the basal vein of Rosenthal
(black arrow in c), and their
tributaries (arrows in a,
dotted white arrow, and
black arrow in d) as well as
of the straight sinus (dotted
arrows in c and d). Dotted
arrow in (b) indicates the
right septal vein and black
arrow in (b) indicates the
left thalamic vein. The
venous clot is delineated as
linear hypointense signal
within the affected veins on
T2*WI images (a–d). Note
that imaging quality is
impaired due to motion
artifacts in this patient
presenting with impaired
consciousness

thrombosis [3]. Combination of the different sub- The term cortical venous thrombosis (CVT)
types is relatively common. indicates thrombosis of the superficial cortical
The most prevalent type of CVST is dural veins including the anastomotic vein of Labbé
sinus thrombosis (or sinus thrombosis, SVT), (Fig. 1). CVT most commonly affects the frontal
which refers to thrombotic occlusion of one or cortical veins, followed by the parietal veins.
more dural sinuses. The superior sagittal sinus is Comparable to isolated deep venous thrombosis,
most commonly affected, followed by the trans- isolated CVT seems to be rather rare and has only
verse sinus [2]. Deep cerebral venous thrombosis been described in case reports and small case
(DVT) affects the internal cerebral veins, vein of series. In the wide majority of cases, concomitant
Galen, and/or the basal veins of Rosenthal and SVT is present, which typically involves the supe-
their tributaries. The straight sinus is included rior sagittal sinus. It is hypothesized that in those
with the deep cerebral veins by most authors. combined cases, CVT develops secondary to
Involvement of the deep cerebral veins is present superior sagittal sinus thrombosis by retrograde
in approximately 10 % of all patients with CVST spread of thrombotic material from the sinus into
and is often accompanied by sinus thrombosis. the cerebral veins draining into it [5].
Isolated thrombosis of the deep cerebral veins is Evaluation of the cortical veins is challenging
much less common [4] (Fig. 2). because the cortical veins, unlike the dural sinuses
938 J. Linn

and deep cerebral veins, show considerable intra- concomitant parenchymal changes, and the inter-
and interindividual variations regarding their val from symptom onset to diagnosis [10, 11].
number, diameter, and anatomic course. This
complicates the interpretation of neuroimaging
studies, particularly the analysis of angiographic Laboratory Findings
datasets. Within the last decade T2*WI gradient-
echo sequences have been found to be very sen- Although normal D-dimer levels have a high neg-
sitive for the detection of CVT and currently rep- ative predictive value for suspected CVST, false-
resent the diagnostic gold standard for the negative results can be observed, particularly in
diagnosis of this specific subtype of CVST (for patients with isolated thrombosis of the deep cere-
details see below) [5–7]. The application of these bral veins. It has been shown that D-dimer levels
sequences in the diagnostic work-up of CVST may be normal in up to 25 % of cases, presumably
resulted in an increased number of reported CVT due to the relatively small volume of thrombotic
cases in recent years. Today it is widely believed material [12].
that cortical vein involvement is much more com-
mon in CVST than previously thought and that
isolated cortical vein thrombosis might not be as Prognosis and Outcome
rare as previously thought but was often missed
prior to the wide use of T2*WI sequences in The outcome of CVST has improved significantly
suspected CVST. in recent decades, largely due to improvements in
neuroimaging, which enable promptly diagnosis
and treatment with intravenous or low-molecular-
weight heparin. To date, CVST has a very good
Clinical Signs and Symptoms prognosis when diagnosed early, with complete
recovery achieved in 80 % of patients [13]. How-
In contrast to arterial ischemic stroke, CVST usu- ever, it must be mentioned that the average time
ally presents with a more subacute clinical onset interval from symptom onset to definitive diagno-
with variable, rather unspecific, and heteroge- sis is still 7 days, mainly due to the diverse and
neous symptoms. More or less severe headache unspecific clinical presentation of the disease and
constitutes the most common initial symptom of its delayed or subacute clinical onset, especially in
CVST, which is observed in 75–90 % of all the small subset of patients with normal D-dimer
patients. Besides, patients often present with levels [1].
other signs of increased intracranial pressure Delayed diagnosis correlates with a poorer
such as dizziness, nausea, and visual disturbances. clinical outcome in CVST, which is observed in
Additional symptoms of CVST depend basically approximately 10–15 % of patients. Patients with
on the location of the thrombosis, i.e., on the deep cerebral venous thrombosis, males, and
subtype of CVST [1, 8]. patients who only present with signs of increased
Involvement of the deep cerebral veins often intracranial pressure are particularly prone to
results in an altered level of consciousness, which delayed diagnosis of CVST [4, 14]. Other risk
is present in over 70 % of DVT cases [8] (Fig. 2), factors that are associated with a poor outcome
while cortical venous thrombosis frequently pre- include advanced age, secondary intracerebral
sents with focal or generalized seizures or focal hemorrhage due to venous congestion, and
neurological deficits as, e.g., hemiparesis, apha- involvement of the deep cerebral veins or right
sia, or hemianopsia. In cases with isolated throm- transverse sinus [4, 14]. In addition, outcome is
bosis of the deep or cortical veins, symptoms of worse in patients in whom central nervous system
increased intracranial pressure may be absent infection or an intracerebral tumor is found as
[9]. Other important factors that determine clinical predisposing cause of CVST [4]. Although
presentation are patient age, the presence of involvement of the deep cerebral veins is one of
39 Imaging of Cerebral Venous and Sinus Thrombosis 939

the most important risk factors or a poor or even Particularly in cases with bilateral rather symmet-
fatal course of the disease, it has been shown that ric parenchymal signal abnormalities in the thal-
even in this specific subtype the outcome has ami and/or basal ganglia, suspicion for deep
considerably improved within the recent decades. cerebral venous thrombosis should be high. The
A study on 32 patients with isolated or combined presence of secondary intracerebral hemorrhage
thrombosis of the deep cerebral veins reports a at initial diagnosis correlates with more severe
good clinical outcome with a modified Rankin initial symptoms and with a poorer outcome [4].
Scale (mRS) of 2 or higher indicating little or no
functional disability in 75 % of patients of DVT
patients after prompt treatment with intravenous Digital-Subtraction Angiography (DSA)
or subcutaneous low-molecular-weight heparin.
Twenty-five percent of patients deteriorated with While DSA has once been considered the gold
progressing coma, even though most of them standard for CVST diagnosis, it no longer plays a
received venous endovascular recanalization significant role in the diagnosis of this disease to
therapy [12]. date but has been largely replaced by noninvasive
imaging modalities, i.e., CTA and MRA. How-
ever, due to its higher spatial and temporal reso-
Imaging Features of CVST lution, DSA is still superior to MRA and CTA
with regard to dynamic information and can
Direct and Indirect Neuroimaging Signs yield important additional information particu-
larly in collateral venous drainage. This might
Basically, both direct and indirect signs of CVST justify its diagnostic use in difficult cases.
can be identified on neuroimaging regardless of Some authors hypothesize that DSA might
the site of the thrombosis. In this regard, “direct yield a higher sensitivity for cortical venous
signs” indicate imaging abnormalities, which are thrombosis compared to noninvasive imaging
caused directly by the thrombotic material within modalities, yet systematic data to support this
the affected veins and/or sinus. Direct signs hypothesis is not available. In patients with iso-
include both “positive” visualization of the throm- lated cortical vein thrombosis, DSA may show an
bus on unenhanced CT or MRI and “negative” “absent” cortical vein and/or a partially opacified
visualization of the thrombotic material as a filling vein with an abrupt cutoff. However, as men-
defect in digital-subtraction angiography (DSA), tioned above, the value of these direct signs of
CT angiography (CTA), or other contrast- CVST is considerably limited by the large
enhanced images or absence of a flow signal in interindividual variations regarding both the num-
flow-sensitive venous MR angiography (MRA). ber and anatomic location of the cortical veins.
On the contrary, the term “indirect signs” refers Therefore, diagnosis of cortical venous thrombo-
to brain parenchymal changes developing second- sis based on DSA mainly relies on indirect signs.
ary to the thrombotic occlusion, e.g., venous The latter include dilated cortical veins adjacent to
edema or venous infarction, subarachnoid hemor- the site of thrombosis (“corkscrew vessels”),
rhage, and parenchymal hemorrhage due to which indicate collateral venous drainage, evi-
venous congestion. In contrast to arterial ischemic dence of a focal delay in venous drainage, as
stroke, edema or infarction caused by CVST does well as signs of venous congestion in the brain
not conform to arterial territories but crosses ter- parenchyma drained by the occluded vein. The
ritory boundaries. Thrombosis of the dural sinus limitations of DSA for the evaluation of cortical
and/or cortical veins commonly results in venous veins likewise apply to noninvasive angiographic
edema or infarction in subcortical white matter or techniques including CTA and MRA.
the cerebral cortex. If the deep cerebral veins are Besides its role as an additional diagnostic
affected, edema or venous infarction typically measure in difficult cases, DSA also is of potential
involves the thalamus and basal ganglia [3]. value as a therapeutic tool in selected patients,
940 J. Linn

Table 1 Stage- and sequence-dependent variability of clot signal characteristics (Modified from Linn and Br€
uckmann
[3])
Signal Acute stage (0–5 Subacute stage (6–15 Chronic stage (>15
MRI sequence intensity days)a days)a days)a
T1w Hyperintense 30 % 71 % 39 %
Isointense 68 % 29 % 54 %
Hypointense 2% 0% 7%
T2w, PDw, Hyperintense 25 % 52 % 43 %
FLAIR Isointense 10 % 32 % 45 %
Hypointense 65 % 16 % 12 %
Numbers printed in bold letters indicate the most common clot signal intensity in the respective stage
T1w T1-weighted spin-echo sequence
T2w T2-weighted spin-echo sequence
PDw proton-density-weighted spin-echo sequence
FLAIR fluid-attenuated inversion recovery-weighted sequence
a
Indicates days after symptom onset

which are considered for interventional treatment Awareness of the significant time- and sequence-
such as local intra-arterial thrombolysis or dependent variations of thrombus signal intensity
mechanical recanalization [13]. Although these is essential for the correct interpretation of MRI
invasive treatment options have been shown to be datasets in patients with suspected CVST, which
effective in single cases and small case series [15, requires a great deal of experience in order to
16], systematic data on their value in CVST are avoid diagnostic and technical pitfalls.
lacking, and they are currently widely regarded as
last therapeutic option in otherwise devastating cases. Spin-Echo T1-, T2-, Proton-Density-,
and Fluid-Attenuated Inversion Recovery
(FLAIR)-Weighted Sequences
Magnetic Resonance Imaging In the acute phase of CVST (days 0–5 from symp-
tom onset), the thrombotic material typically pre-
Today MRI including MR angiography is com- sents with low signal intensity on T2-WI and a
monly regarded the diagnostic gold standard in relative isointense signal on T1-WI spin-echo
suspected CVST although CTA is increasingly sequences (Fig. 3). As these signal characteristics
reported to be equivalent to MR angiography very much resemble those of regular flow voids in
(see below) [1, 13]. patent venous structures, thrombus detection early
in the course of the disease is hampered on these
Direct Signs of CVST on MRI sequences. This fact significantly limits the sensi-
On MRI, CVST presents with rather complex tivity of spin-echo MR sequences, especially dur-
MRI sequence-dependent signal abnormalities, ing the initial days after symptom onset.
which also vary significantly between the differ- During the subacute phase (days 6–15), the
ent stages of the disease. Depending on the age of venous clot becomes increasingly hyperintense
the thrombotic material, an acute, subacute, and in both T2- and T1-WI spin-echo sequences [17]
chronic stage of CVST can be distinguished which (Fig. 4).
all present with different signal characteristics Although these signal patterns are found in the
which are indicated in Table 1. On the contrary, majority of cases, thrombotic signal intensity
the location of the thrombus, i.e., the subtype of might vary considerably in the individual case
CVST, plays a minor role, as the signal character- (Table 1). On FLAIR and proton-density
istics of the thrombotic material are similar regard- sequences, signal characteristics of the thrombotic
less of whether the dural sinuses, the deep cerebral material are very similar to those found on T2-WI
veins, or the cortical veins are involved [3]. spin-echo sequences [5] (Figs. 3 and 4).
39 Imaging of Cerebral Venous and Sinus Thrombosis 941

Fig. 3 MRI findings in the acute phase of sinus thrombo- profoundly hypointense on T2*WI imaging (c). It shows
sis. A 35-year-old female patient with thrombosis of the no hyperintense signal on diffusion-weighted imaging (d)
right transverse sinus. 3-T MRI was performed 3 days after and delineates as lack of regular flow signal on maximum
symptom onset. The venous clot (arrows) is partially iso- intensity projection (MIP) reconstruction (e) and source
and partially hyperintense on T1-WI imaging (a), iso- to images (f) of venous time-of-flight MR angiography
slightly hypointense on FLAIR sequence (b), and

Fig. 4 MRI findings in the


subacute phase of sinus
thrombosis. A 76-year-old
female patient with
thrombosis of the left
transverse and sigmoid
sinuses. 1.5-T MRI was
performed 2 weeks after
symptom onset. The venous
clot (arrows) is
hyperintense on T1-WI (a),
T2-WI (b), and FLAIR
imaging (d) and delineates
as lack of regular flow signal
on the source image of
venous MR angiography (c)
942 J. Linn

Fig. 5 Hyperintense signal


of venous clot on diffusion-
weighted imaging (DWI). A
27-year-old female patient
with deep cerebral venous
thrombosis. DWI images
(a, b) performed within the
acute phase of the disease
show a hyperintense signal
of the venous clot (arrows
in a and b). On
susceptibility-weighted
imaging (SWI, c), the
thrombotic material is
profoundly hypointense
(normal white arrows in c).
Note also the hypointense
normal flow signal in
regular veins due to the
deoxyhemoglobin fraction
of venous blood (e.g.,
dotted white arrows in c). In
contrast, regular arteries
with oxyhemoglobin are
delineated as hyperintense
signal on this sequence
(e.g., black arrows in c)

One has to be aware of several potential It has predominately been observed in the sub-
sources of artifacts and pitfalls on MRI. As an acute stage of CVST. In this stage, the thrombus is
example, a hyperintense signal from normal typically also hyperintense on T2-, T1-, and
venous flow might mimic a thrombus on T1-WI FLAIR-weighted images and thus can usually be
spin-echo sequences, if those are acquired without easily identified without additional information
adequate flow compensation. Its proneness to arti- provided by DWI. However, there is initial evi-
facts constitutes a slight disadvantage of MRI dence that thrombus signal characteristics on DWI
compared to CT with CTA and requires expert might serve as a prognostic factor in CVST. In a
knowledge to avoid misinterpretation in pilot study by Favrole et al., the presence of
suspected CVST [3, 17]. restricted diffusion in the clot was associated
with low rates of recanalization [19]. The value
Diffusion-Weighted Imaging (DWI) of this observation should be investigated in fur-
Venous clot might present with a hyperintense ther studies.
signal on B1000 images of diffusion-weighted
MR sequences and with a correspondingly T2*WI Gradient-Echo Sequences
decreased apparent diffusion coefficient (ADC) and Susceptibility-Weighed Imaging
on ADC maps, which indicates restricted diffu- On T2*WI gradient-echo MRI sequences, para-
sion within the thrombus itself (Fig. 5). magnetic substances as, e.g., thrombotic material
The sensitivity of this sign ranges from approx- induce considerable susceptibility artifacts, which
imately 5 % to 40 % in different studies [5, 18]. results in a profound hypointense signal. Recent
39 Imaging of Cerebral Venous and Sinus Thrombosis 943

Fig. 6 T2*WI imaging in cortical venous thrombosis. A ideal for the depiction of venous edema (arrows in d–f),
73-year-old patient with isolated left frontoparietal cortical while the T2*WI sequence is best suited to detect subse-
venous thrombosis. T2*WI gradient sequences (a–c) show quence hemorrhagic changes (black arrows in b and c).
a profound hypointense signal in the affected cortical veins Note that extensive preexisting microangiopathic white
(white arrows in a–c). Secondary parenchymal changes, matter lesions were present in this patient (black asterisks
i.e., indirect signs of cerebral venous thrombosis, are also in f)
present. Axial T2 (d, e) and coronal FLAIR (f) images are

studies have shown that T2*WI sequences are of T2*WI imaging is of particular diagnostic
considerable additional value in the diagnosis of yield in patients with suspected cortical vein
CVST, particularly in the acute phase of the dis- thrombosis and has been shown to be significantly
ease, when the thrombus is still isointense on superior to all other MRI sequences and CTA with
T1-WI sequences [20–22]. T2*WI gradient-echo regard to the evaluation of the cortical veins
imaging delineates the acute thrombus as a homo- (Fig. 6). To date, T2*WI sequences are considered
geneous, markedly hypointense tubular structure, the gold standard for the diagnosis of isolated or
which allows its detection early after symptom combined cortical venous thrombosis. Thus, it is
onset (Figs. 2 and 3). strongly recommended to implement this
With regard to the thrombus signal characteris- sequence in the standard MRI protocol in
tics later in the course of the disease, findings are suspected CVST [5–7].
somewhat less consistent. Some authors report a However, one has to keep in mind that T2*WI
persistence of the hypointense signal of the venous sequences might also yield false-positive or
clot for several months in a considerable percent- (rarely) false-negative results. Chronic convexity
age of cases [21]. In contrast, others observed a subarachnoid hemorrhage or cortical superficial
relatively strong time dependence of the siderosis might mimic cortical venous thrombosis
hypointense signal, which they observed in 90 % on T2*WI images. Cortical superficial siderosis is
of cases during the first week but in only 9–32 % of defined as a hypointense linear signal within the
cases during the subacute or chronic phases [22]. superficial layers of the cerebral cortex, and
944 J. Linn

represents an important MRI biomarker of cere- considerably prone to artifacts, and one should be
bral amyloid angiopathy, but should not be aware of the following potential pitfalls [26]:
misinterpreted as a hypointense venous clot [23, Artificial flow gaps on venous TOF-MRA
24]. To avoid false-positive diagnoses, a linear might result from slow blood flow or from
hypointense structure on T2*WI GRE images in-plane flow parallel to the acquisition plane.
should only be regarded as direct signs of venous Within the non-dominant transverse sinus (typi-
clot if it (1) conforms to the anatomic course of a cally the left one), such artificial flow gaps are
venous structure, (2) results in a prominent observed in over 30 % of patients [26, 27]. In
blooming effect which exceeds the regular diam- addition, a hypo- or aplastic transverse sinus,
eter of the affected vessel, and (3) shows a round which is frequently found as a variant, might
cross section in slices perpendicular to its course easily be misinterpreted as a sinus thrombosis, if
[23, 24]. venous MRA is interpreted solely [17, 26]
The susceptibility effect of paramagnetic sub- (Fig. 7).
stances on conventional T2*WI sequences can be Finally, if imaging is performed within the
further enhanced by either combining both mag- subacute stage of the disease, a T1-hyperintense
nitude and phase information of the MRI signal venous clot might simulate regular flow on
using adequate post-processing steps or by apply- TOF-MRA [27]. To avoid misinterpretation of
ing multiple echo times. These approaches are TOF-MRA, it is strongly recommended to care-
referred to as susceptibility-weighted imaging fully analyze not only the three-dimensional min-
(SWI) or susceptibility-weighted angiography imum-intensity projection reconstructions but
(SWAN). Susceptibility-weighted images are also the source images.
even more sensitive than conventional T2*WI Besides TOF-MRA, phase-contrast venous
sequences for detecting subtle abnormalities in MRA, contrast-enhanced venous MRA, and
magnetic susceptibility. They allow an excellent contrast-enhanced MPRAGE (magnetization-
visualization of the normal cerebral venous prepared rapid acquisition gradient-echo)
anatomy, based on the fact that venous blood is sequences are also used to evaluate CVST. The
dark on SWI due to its high deoxyhemoglobin contrast-enhanced techniques are much less prone
fraction. Venous clot should appear even more to artifacts than flow-sensitive techniques. New
profoundly hypointense on these sequences. In MRA techniques such as time-resolved MRA
single cases SWI has yielded promising results might be of additional value for the diagnosis of
with regard to diagnosis of CVST [3, 25] CVST. As an example, the combination of
(Fig. 5). Yet, it has to be considered that – as dynamic and static three-dimensional MRA,
mentioned before – regularly perfused veins also which is known as combined, four-dimensional
appear hypointense on these sequences, which venous MRA, has been shown to be superior to
might hamper their interpretation (Fig. 5). There- TOF-MRA with regard to the evaluation of the
fore, larger studies are needed to determine the cerebral venous system [28], but further studies
value of susceptibility-weighted imaging in the are required to evaluate its diagnostic yield in
diagnosis of CVST. CVST. Regarding the evaluation of the cortical
veins, it should be taken into account that basi-
Venous MR Angiography (MRA) cally all venous MRA techniques are hampered by
Different technical approaches are used in imag- the same limitations as DSA and CT angiography,
ing the cerebral veins and sinuses on MRI. Under namely, by the considerable anatomic variations
routine clinical conditions, venous time-of-flight of these vessels.
(TOF-) MRA is most widely used. It is a
noncontrast-enhanced, flow-sensitive technique Indirect Signs of CVST in MRI
in which the thrombus is delineated negatively MRI constitutes the most sensitive modality for
as the absence of regular flow in the affected detecting secondary parenchymal changes in
vascular segment (Fig. 3). Venous TOF-MRA is CVST, i.e., indirect signs of venous thrombosis.
39 Imaging of Cerebral Venous and Sinus Thrombosis 945

Fig. 7 Hypoplastic transverse sinus mimicking sinus lumen. Careful inspection of the source images of the
thrombosis on venous MR angiography. Patient with a angiography (b) and of the other MR sequences helps to
hypoplastic proximal left transverse sinus, which might exclude the thrombosis. On the source image of the angi-
be misinterpreted as sinus thrombosis on maximum inten- ography, flow within the hypoplastic part of the sinus can
sity projection (MIP) reconstruction of the venous MR be depicted (arrows in b). There are no signal abnormali-
angiography (a). Distal from the confluence with the left ties indicative of venous clot on FLAIR (c) or T2*WI
vein of Labbé, the distal transverse sinus shows a regular sequences (d)

Venous edema and venous infarction are best Computed Tomography


delineated on T2-, proton-density-, and/or
FLAIR-weighted sequences, while T2*WI Unenhanced CT
gradient-echo sequences ideally visualize second- Due to its wide availability, its cost-effectiveness, and
ary hemorrhagic changes due to venous conges- the short examination time, unenhanced CT still
tion (Fig. 6). constitutes the imaging method of first choice in the
Diffusion-weighted sequences yield variable emergency setting in most clinical institutions. On
and heterogeneous findings in the brain paren- unenhanced CT, an acute thrombus might present as
chyma in CVST and might indicate cytotoxic or a homogeneous hyperdense structure. This direct
vasogenic edema or a mixture of both in individ- sign of thrombosis is referred to as cord sign or
ual cases. Clinical studies as well as studies in dense vein sign [31]. Both terms are used rather
animal models provided evidence that cytotoxic inconsistently in the literature on CVST. It might be
edema precedes vasogenic edema in CVST, which recommended to use the term “cord sign” if throm-
is reflected by a decreased apparent diffusion bosis affects the dural sinuses and the term “dense
coefficient (ADC value) at early stages of throm- vein sign” in case of suspected thrombosis of the
bosis, followed by a normalization or increase in cerebral veins. A cord sign or dense vein sign is most
ADC value later during the course of the disease commonly observed in the acute stage of CVST, i.e.,
[29, 30] (Fig. 8). during the first week after symptom onset (Fig. 9).
946 J. Linn

Fig. 8 Venous edema on T2- and diffusion-weighted diffusion-weighted sequence (b), there are mixed signal
images. A 27-year-old female patient with thrombosis of intensities within the thalami. Parts of the edema show a
the internal cerebral veins (same patient as shown in hyperintense signal (e.g., arrows in b), with corresponding
Fig. 2). FLAIR image (a) shows symmetrical edema within low signal on apparent diffusion coefficient (ADC) maps
the thalami (normal arrows in a) and the caudate nuclei (c, arrows), indicative of restricted diffusion
(dotted arrows in a), bilaterally. On B1000 images of the

During the second week the thrombus first On unenhanced CT, several potential pitfalls
becomes isodense and later hypodense. Data on may lead to false-positive or false-negative find-
the sensitivity and specificity of the cord sign for ings on the presence of a cord sign or dense vein
diagnosis of sinus thrombosis varies in recent sign. First of all, partial-volume effects, which are
studies between 65 % and 73 % for sensitivity caused by the adjacent skull, may mimic a false-
and 97 % and 100 % for specificity [31, 32]. The positive cord sign in the dural sinuses or a dense
sensitivity of the dense vein sign for cortical vein vein sign in the cortical veins. Evaluation of the
thrombosis has been found to be very low (25 %) deep cerebral veins is not hampered considerably
[5]. In contrast, its diagnostic yield in thrombosis by this effect owing to their deep location. Regular
of the deep cerebral veins seems to be much flow in patent veins might also appear slightly
higher with a reported sensitivity of 100 % and a hyperdense on noncontrast CT and lead to false-
specificity of 99 % compared to MRI as the gold positive results, particularly in patients with ele-
standard [31] (Fig. 10). vated hematocrit or high hemoglobin levels, as it
39 Imaging of Cerebral Venous and Sinus Thrombosis 947

Fig. 9 Cord sign on unenhanced CT. A 34-year-old male angiography (b, arrows) and as a profound hypointense
patient with thrombosis of the proximal left transverse structure on T2*WI images (c, arrows). Note the sharp
sinus. The venous clot delineated hyperdense on margin of the thrombus (asterisks in b and c). There is
unenhanced CT (a), a direct sign of thrombosis, which is regular flow in the distal part of the sinus (dotted arrows in
referred to as cord sign (arrows in a). The venous clot is b and c)
visualized as a filling defect in the affected sinus on CT

has been shown that sinus attenuation correlates Contrast-Enhanced Computed


moderately with hematocrit and hemoglobin Tomography
levels [33, 34] (Fig. 11). To date, contrast-enhanced CT plays only a minor
Quantitative assessment of the sinus density role in the diagnosis of CVST owing to the wide
might be helpful. For patent sinuses, a mean den- availability of CTA. If contrast-enhanced CT is
sity of approximately 50 Hounsfield units performed, a so-called “empty delta sign” might
(HU) with a standard deviation of 7.5 HU has be delineated in CVST patients. This finding indi-
been reported [34], while cutoff value of cates a direct sign of sinus thrombosis, which is
70 Hounsfield units (HU) seems to be indicative typically found in the superior sagittal sinus on
of a thrombosis [33]. If imaging is not performed slices perpendicular to the course of the sinus and
within the acute phase of CVST, i.e., within the refers to a triangular area with an enhancing
first week, the thrombotic material may appear periphery and a non-enhancing center [35]. This
isodense again, yielding a false-negative result. imaging appearance of the affected sinus is due to
In summary, regarded the limited sensitivity of the fact that the sinus walls show strong contrast
unenhanced CT, this method alone does not allow enhancement while the thrombotic material
to reliably detect a CVST but is useful as a screen- within the lumen of the sinus does not enhance
ing examination which should be followed by significantly after administration of intravenous
CTA or MRI if clinical suspicion is high. contrast agent (Fig. 12).
948 J. Linn

Fig. 10 Dense vein sign on unenhanced CT. A 27-year- arrow in b), and the straight sinus (dotted arrows in
old female patient with thrombosis of the internal cerebral a and b). This direct sign of venous thrombosis on
veins (same patient as shown in Figs. 2 and 8). On unenhanced CT is referred to as “dense vein sign.” On
unenhanced CT (a, b) the venous clot delineates as sagittal reconstructions of the CT angiography (c), the
hyperdense tubular structures within the internal cerebral venous clot is visualized as a filling defect in the affected
veins (normal arrows in a), the vein of Galen (normal veins (c, arrows)

CT Angiography as a filling defect in the affected venous structures


CT angiography (CTA) performed on modern and vessels on CTA (Fig. 13). As a major advan-
multidetector-row CT scanners enables delineation tage compared to flow-sensitive TOF-MRA, CTA
of the dural sinuses and cerebral veins at relatively is almost not susceptible to flow artifacts. This is
high spatial resolution and allows for three- particularly important if anatomic variations are
dimensional multi-planar reconstructions and/or present in a patient, as, e.g., a hypoplastic left
maximum intensity projections of the cerebral transverse sinus. CTA is of particular value in the
venous system. This technology is increasingly emergency setting due to its short examination
considered equivalent or even superior to MR angi- time, its wide availability, and its ability to simul-
ography, particularly with regard to the evaluation taneously delineate the arterial and the venous
of smaller veins, the inferior sagittal sinus, and the cerebral system in one scan if applied with an
non-dominant transverse sinus. CVST is visualized appropriate scan delay [36].
39 Imaging of Cerebral Venous and Sinus Thrombosis 949

Fig. 11 False-positive cord sign on unenhanced abnormalities indicative of venous clot on T2*WI
CT. Patient with an elevated hematocrit resulting in a sequences (e, arrows). Venous MR angiography (f) con-
relative high density in the dural sinuses on unenhanced firms regular venous flow in the transverse and superior
CT (a–c, e.g., arrows). There is a normal flow void on the sagittal sinuses (e.g., arrows in f)
T2-WI image (d, arrows), and there are no signal

However, the value of CTA in CVST seems to


differ considerably depending on the
localization of the thrombus, i.e., of subtype of
CVST. With regard to sinus thrombosis, it
has been demonstrated that multi-slice CTA is
equally sensitive and specific as MRI [37,
38]. On the contrary, CTA showed a very
low sensitivity for the detection of cortical vein
thrombosis due to similar limitations as
mentioned above for DSA. Therefore, CTA can-
not be considered suitable to diagnose or
exclude isolated or combined cortical vein
thrombosis of the cortical [5]. Data on the delin-
eation of the deep cerebral veins on CTA are
promising; however, systematic studies on the
diagnostic value of this technology for the detec-
tion of deep venous thrombosis are currently
lacking (Fig. 10).
Fig. 12 Empty delta sign on contrast-enhanced CT. A Although CTA is less prone to artifacts com-
45-year-old patient with thrombosis of the superior sagittal pared to MRI, some potential pitfalls have to be
sinus. On contrast-enhanced CT, the thrombosis results in a considered, as they might result in misinterpreta-
so-called empty delta sign (arrow). This direct sign of sinus
thrombosis refers to a triangular area with an enhancing
tion (particularly false-negative results) of CTA.
periphery and a non-enhancing center and is due to the fact Especially in the acute phase of the disease (days
that the sinus walls show strong contrast enhancement 0–5 after symptom onset), a very hyperdense
while the thrombotic material within the lumen of the thrombus (corresponding to a cord sign or dense
sinus does not enhance significantly
950 J. Linn

Fig. 13 Sinus thrombosis on CT angiography. A 45-year- venous clot is visualized as a filling defect in the affected
old patient with thrombosis of the superior sagittal sinus sinus (a–c, arrows)
(same patient as shown in Fig. 12). On CT angiography, the

vein sign on unenhanced CT) might mimic nor- effective dose of an unenhanced cranial CT
mal contrast filling of the affected venous struc- scan), radiation exposure is only regarded as a
ture on CTA and yield false-negative results. To minor disadvantage of CTA compared to
avoid this source of misinterpretation, CTA scans MRI [39].
should always be interpreted together with
unenhanced CT scans, which directly demonstrate
the cord sign or dense vein sign. This might be Summary
particularly important with regard to the evalua-
tion of the deep cerebral veins on CTA, but further To date, MRI including venous MR angiography
data on this issue are needed. and a T2*WI gradient-echo sequence is consid-
In contrast to fresh thrombotic material, an ered the gold standard for the diagnosis of CVST.
organized, chronic venous clot can show contrast Its correct interpretation requires a detailed
enhancement on CTA instead of causing a filling knowledge of the complex time- and sequence-
defect, which might also lead to a false-negative dependent signal characteristics of the thrombotic
interpretation. Partial-volume effects from the material. Diagnosis of isolated or combined
enhancing walls of small veins or from the adja- involvement of the cortical veins in CVST is
cent skull might obscure a filling defect and rep- particularly challenging and requires T2*WI
resent another source of false-negative results. sequences which delineate the venous clot as
False-positive interpretation of CTA can be hypointense tubular structure.
caused by prominent arachnoid granulations Unenhanced CT together with multidetector-
(pacchionian granulations), which might result in row CT angiography provides a very fast, widely
a filling defect similar to the one caused by throm- available, and cost-effective alternative technique
botic vessel occlusion. In most cases, however, which is increasingly considered equivalent to
they can rather easily be distinguished from MRI in suspected sinus thrombosis and deep cere-
thrombotic material based on their typical round bral venous thrombosis, but not in thrombosis of
or oval shape, smooth margins, and cerebrospinal the cortical veins.
fluid isodensity [37]. Radiologists should be aware of a variety of
As the effective radiation dose of a potential pitfalls of both MRI and CT techniques,
multidetector-row CTA scan at 120 kV is less which might yield false-positive or false-negative
than 1 mSv (and thus even less than the mean results regarding the presence of a CVST.
39 Imaging of Cerebral Venous and Sinus Thrombosis 951

References 17. Teasdale E (2000) Cerebral venous thrombosis: mak-


ing the most of imaging. J R Soc Med 93:234–237
1. Stam J (2005) Current concepts: thrombosis of the 18. Lövblad KO, Bassetti C, Schneider J et al (2001)
cerebral veins and sinuses. N Engl J Med Diffusion-weighted MRI in cerebral venous thrombo-
352:1791–1798 sis. Cerebrovasc Dis 11:169–176
2. Ferro JM, Bacelar-Nicolau H, Rodrigues T, ISCVT and 19. Favrole P, Guichard JP, Crassard I et al (2004)
VENOPORT investigators et al (2009) Risk score to Diffusion-weighted imaging of intravascular clots in
predict the outcome of patients with cerebral vein and cerebral venous thrombosis. Stroke 35:99–103
dural sinus thrombosis. Cerebrovasc Dis 28:39–44 20. Selim M, Fink J, Linfante I et al (2002) Diagnosis of
3. Linn J, Br€uckmann H (2010) Cerebral venous and cerebral venous thrombosis with echo-planar T2*-
dural sinus thrombosis: state-of-the-art imaging. Clin weighted magnetic resonance imaging. Arch Neurol
Neuroradiol 20:25–37 59:1021–1026
4. Ferro JM, Canhao P, Stam J, ISCVT Investigators 21. Idbaih A, Boukobza M, Crassard I et al (2006) MRI of
et al (2004) Prognosis of cerebral vein and dural sinus clot in cerebral venous thrombosis. High diagnostic
thrombosis: results of the International Study on Cere- value of susceptibility-weighted images. Stroke
bral Vein and Dural Sinus Thrombosis (ISCVT). 37:991–995
Stroke 35:664–670 22. Leach JL, Strub WM, Gaskill-Shipley MF (2007)
5. Linn J, Michl S, Bochmann K et al (2010) Cortical vein Cerebral venous thrombus signal intensity and suscep-
thrombosis: the diagnostic value of different imaging tibility effects on gradient recalled-echo MR imaging.
modalities. Neuroradiology 52:899–911 AJNR Am J Neuroradiol 28:940–945
6. Boukobza M, Crassard I, Bousser MG et al (2009) MR 23. Linn J, Herms J, Dichgans M et al (2008) Subarachnoid
Imaging features of isolated cortical vein thrombosis: hemosiderosis and superficial cortical hemosiderosis in
diagnosis and follow-up. AJNR Am J Neuroradiol cerebral amyloid angiopathy. AJNR Am J Neuroradiol
30:344–348 29:184–186
7. Ihn YK, Jung WS, Hwang SS (2013) The value of T2*- 24. Linn J, Halpin A, Demaerel P et al (2010) Prevalence
weighted gradient-echo MRI for the diagnosis of cere- of superficial siderosis in patients with cerebral amy-
bral venous sinus thrombosis. Clin Imaging 37:446–450 loid angiopathy. Neurology 74:1346–1350
8. Ameri A, Bousser MG (1992) Cerebral venous throm- 25. Kawabori M, Kuroda S, Kudo K et al (2009)
bosis. Neurol Clin 10:87–111 Susceptibility-weighted magnetic resonance imaging
9. Jacobs K, Moulin T, Bogousslavsky J et al (1996) The detects impaired cerebral hemodynamics in the supe-
stroke syndrome of cortical vein thrombosis. Neurol- rior sagittal sinus thrombosis – case report. Neurol Med
ogy 47:376–382 Chir (Tokyo) 49:248–251
10. Ferro JM, Canhão P, Bousser MG, Stam J, 26. Ayanzen RH, Bird CR, Keller PJ et al (2000) Cerebral
Barinagarrementeria F, ISCVT Investigators (2005) MR venography: normal anatomy and potential diag-
Cerebral vein and dural sinus thrombosis in elderly nostic pitfalls. AJNR Am J Neuroradiol 21:74–78
patients. Stroke 36:1927–1932 27. Lafitte F, Boukobza M, Guichard JP et al (1997) MRI
11. Ferro JM, Lopes MG, Rosas MJ, Fontes J, and MRA for diagnosis and follow-up of cerebral
VENOPORT Investigators (2005) Delay in hospital venous thrombosis (CVT). Clin Radiol 52:672–679
admission of patients with cerebral vein and dural 28. Meckel S, Reisinger C, Bremerich J et al (2010) Cere-
sinus thrombosis. Cerebrovasc Dis 19:152–156 bral venous thrombosis: diagnostic accuracy of com-
12. Pfefferkorn T, Crassard I, Linn J et al (2009) Clinical bined, dynamic and static, contrast-enhanced 4D MR
features, course and outcome in deep cerebral venous venography. AJNR Am J Neuroradiol 31:527–535
system thrombosis: an analysis of 32 cases. J Neurol 29. Ducreux D, Oppenheim C, Vandamme X et al (2001)
256:1839–1845 Diffusion-weighted imaging patterns of brain damage
13. Diener HC, Putzki N (eds) (2008) Diagnostic and ther- associated with cerebral venous thrombosis. AJNR Am
apeutic guidelines in neurology, 4th edn. Thieme, J Neuroradiol 22:261–268
Stuttgart 30. Srivastava AK, Kalita J, Haris M et al (2009) Radio-
14. Girot M, Ferro JM, Canhão P, ISCVT Investigators logical and histological changes following cerebral
et al (2007) Predictors of outcome in patients with venous sinus thrombosis in a rat model. Neurosci Res
cerebral venous thrombosis and intracerebral hemor- 65:343–346
rhage. Stroke 38:337–342 31. Linn J, Pfefferkorn T, Ivanicova K et al (2009)
15. Guo XB, Song LJ, Guan S (2014) Endovascular treat- Noncontrast CT in deep cerebral venous thrombosis
ment of chronic, recurrent headache secondary to and sinus thrombosis: comparison of its diagnostic
chronic cerebral venous sinus thrombosis. J Stroke value for both entities. AJNR Am J Neuroradiol
Cerebrovasc Dis 23:560–563 30:728–735
16. Guo XB, Fu Z, Song LJ, Guan S (2013) Local throm- 32. Roland T, Jacobs J, Rappaport A et al (2010)
bolysis for patients of severe cerebral venous sinus Unenhanced brain CT is useful to decide on further
thrombosis during puerperium. Eur J Radiol imaging in suspected venous sinus thrombosis. Clin
82:165–168 Radiol 65:34–39
952 J. Linn

33. Black DF, Rad AE, Gray LA et al (2011) Cerebral multislice CT. A single-bolus, monophasic protocol.
venous sinus density on noncontrast CT correlates Acta Radiol 42:560–562
with hematocrit. AJNR Am J Neuroradiol 37. Linn J, Ertl-Wagner B, Seelos KC et al (2007) Diag-
32:1354–1357 nostic value of multidetector-row CT angiography in
34. Fanous R, Leung A, Karlik S (2010) Quantitative the evaluation of thrombosis of the cerebral venous
assessment of the superior sagittal sinus on unenhanced sinuses. AJNR Am J Neuroradiol 28:946–952
computed tomography. Eur J Radiol 75:336–342 38. Gaikwad AB, Mudalgi BA, Patankar KB et al (2008)
35. Virapongse C, Cazenave C, Quisling R et al (1987) The Diagnostic role of 64-slice multidetector row CT scan
empty delta sign: frequency and significance in and CT venogram in cases of cerebral venous throm-
76 cases of dural sinus thrombosis. Radiology bosis. Emerg Radiol 15:325–333
162:779–785 39. Ertl-Wagner B, Hoffmann RT, Bruening R et al (2004)
36. Klingebiel R, Zimmer C, Rogalla P et al (2001) Assess- Multi-slice CT angiography of the brain at various
ment of the arteriovenous cerebrovascular system by kilovoltage settings. Radiology 231:528–535
The Role of Diagnostic Imaging
Techniques for Detection 40
of Extracranial Venous System
Abnormalities Associated with Central
Nervous System Disorders

Kresimir Dolic and Robert Zivadinov

Contents Invasive Imaging Modalities . . . . . . . . . . . . . . . . . . . . . . . . 971


Multimodal Imaging Approach . . . . . . . . . . . . . . . . . . . . . . 974
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 954
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 974
Anatomy and Function of the Cerebral
Venous System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 956 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 975
Extracranial Cerebral Venous Drainage Pathway:
Neck Veins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 957
IJV Valves . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 957
Other Neck Veins Serving as Collaterals
for Cerebral Venous Drainage . . . . . . . . . . . . . . . . . . . . . . . 957
Extracranial Cerebral Venous Drainage Pathway:
Abdominal and Thoracic Veins . . . . . . . . . . . . . . . . . . . . . . 958
Extracranial Venous Abnormalities Versus
Developmental Variants . . . . . . . . . . . . . . . . . . . . . . . . . . . . 960
Diagnostic Imaging and the Diagnosis
of Extracranial Venous Abnormalities . . . . . . . . . . . . 960
Noninvasive Imaging Modalities . . . . . . . . . . . . . . . . . . . . 960

K. Dolic (*)
Buffalo Neuroimaging Analysis Center, Department of
Neurology, University at Buffalo, State University of
New York, Buffalo, NY, USA
Clinical Department of Diagnostic and Interventional
Radiology, Clinical Hospital Center Split, University of
Split, Split, Croatia
e-mail: kdolic@bnac.net
R. Zivadinov
Buffalo Neuroimaging Analysis Center, Department of
Neurology, University at Buffalo, State University of
New York, Buffalo, NY, USA
MRI Clinical Translational Research Center, University at
Buffalo, State University of New York, Buffalo, NY, USA
e-mail: rzivadinov@bnac.net; rz4@buffalo.edu

# Springer Science+Business Media New York 2016 953


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_12
954 K. Dolic and R. Zivadinov

insufficiency • Cerebral venous system •


Abstract
Neck veins • Jugular vein reflux • Doppler
The extracranial venous system is complex and
sonography • Magnetic resonance venography
variable. In the last decade, it has been repeat-
• Computed tomography venography • Cathe-
edly shown that the presence and severity of
ter venography • Intravascular ultrasound •
jugular vein reflux is associated with a number
Plethysmography • Multimodal imaging
of central nervous system (CNS) disorders
such as transient global amnesia, transient
monocular blindness, cough headache, pri-
Introduction
mary exertional headache, and more recently
Alzheimer’s disease and aging. A newly pro-
The main function of the venous system is to
posed vascular condition, named chronic cere-
return blood to the heart from the periphery and
brospinal venous insufficiency (CCSVI), has
maintain cardiac output. It represents complex,
triggered recently intense interest in better
low-pressure, freely communicating network of
understanding the role of extracranial venous
vessels, which contains 75 % of the body’s circu-
abnormalities and their developmental vari-
lating blood volume [1, 2]. The extracranial
ants. Their relationship to intracranial CNS
venous system is a complex three-dimensional
pathology, especially in patients with multiple
(3D) structure, not widely examined and only
sclerosis (MS), is poorly understood at this
partially understood, that is often asymmetric
time. So far there is no established invasive or
and represents significantly more variability than
noninvasive diagnostic imaging modality that
intra- and extracranial arterial anatomy [2, 3]. The
can serve as a “gold standard” for the detection
role of arterial supply abnormalities in relation to
of these venous abnormalities/developmental
the pathology of central nervous system (CNS)
variants. The use of noninvasive diagnostic
disorders is well defined in comparison with
imaging techniques such as Doppler sonogra-
extracranial venous abnormalities which in the
phy (DS) remains controversial; however, con-
past were mostly acknowledged as
sensus guidelines and standardized protocols
non-pathological findings due to the lack of inves-
are emerging. The use of magnetic resonance
tigations aimed to define their nature and clinical
venography (MRV) and phase-contrast imag-
significance [4, 5]. More recently, it has been
ing is gaining an increasing interest as an alter-
hypothesized that these venous abnormalities are
native noninvasive diagnostic approach.
likely to be truncal venous malformations [6]
Further, catheter venography (CV) and intra-
characterized by intraluminal defects (such as
vascular ultrasound (IVUS) are becoming
flaps, webs, septums, membranes, and malformed
important diagnostic tools for confirming the
valves) or by extraluminal anomalies represented
presence and severity of extracranial venous
by stenoses of the venous wall ([2, 5, 7]; Table 1).
pathology. Most likely, a multimodal imaging
Recently several CNS disorders such as tran-
approach will ultimately be the most compre-
sient global amnesia, transient monocular blind-
hensive means for screening, diagnostic, as
ness, leukoaraiosis, dementia, normal-pressure
well as monitoring purposes. Further research
hydrocephalus, cough headache, and primary
is needed to determine the spectrum and prev-
exertional headache have been linked to the pres-
alence of these extracranial venous abnormal-
ence and severity of uni- or bilateral jugular
ities/developmental variants and to compare
venous reflux [2, 8–12]. Further, more recently
the imaging findings with pathological
the introduction of a composite criteria-based vas-
examinations.
cular condition named chronic cerebrospinal
venous insufficiency (CCSVI) generated an
Keywords intense interest in better understanding the role
Multiple sclerosis • Extracranial venous abnor- of the extracranial venous system in the patho-
malities • Chronic cerebrospinal venous physiology of CNS disorders, especially in
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 955

Table 1 Proposed classification of internal jugular vein diagnosis is based mainly on color Doppler
(IJV) abnormalities on Doppler sonography (Adapted from sonography (DS) findings in the extracranial
Zivadinov and Chung [5])
(neck) and intracranial veins by assessing five
Doppler sonography venous hemodynamic (VH) criteria (with cutoff
classification Types/examples
of 2 positive criteria used for diagnosis of
Intraluminal Web: multiple septae and/or
structural venous flaps located in a cluster CCSVI) [17, 18]. From the time it was first men-
abnormality: This is Flap: thin linear echogenic tioned, the CCSVI theory has not ceased to pro-
an echogenic structure structure extending from voke controversy and attention in the scientific
extending from the endothelial lining of vein wall community and the media, mostly because the
endothelial lining of Septum: thin linear echogenic
the vein wall with/ sensitivity and specificity results originally
structure extending from
without the presence of reported [14, 17] were not reproduced by other
endothelial lining of vein wall
functional and attached to it at both ends. groups [19–21]. Also, the reliability of using DS
abnormality. These Septum may extend across a in the diagnosis of CCSVI is questionable without
abnormalities include vein to attach on opposing
web, flap, septum, proper training and has been the focal point of
sides or attach on same side
membrane, and recent statements from various societies [19, 22,
Membrane: membranous
malformed valve
structure almost occluding the
23]. Further, the CCSVI concept did not take into
entire diameter of the vein account the physiological variations of the IJV in
Malformed valve: diameter [24].
dysdynamic or fibrous valve One of the main criticisms of the CCSVI con-
Extraluminal Stenosis: cross-sectional area cept arose from the use of endovascular proce-
structural venous (CSA) measurement of 3
dures to unblock potentially stenotic IJV and
abnormality: This is a mm2
restriction of the azygos veins in an open-label fashion without
Annulus: circumferential
venous wall or thickened vein wall that is previously establishing: (a) diagnostic imaging
stenosis. These restricting the vein from fully modalities and protocols that will serve as a
abnormalities include expanding with respiratory or “gold standard” for the detection of extracranial
stenosis and annulus positional changes
venous anomalies and (b) safety as well as effi-
Functional venous Reflux/bidirectional flow:
abnormality: This is present in the IJV for more
cacy of the endovascular procedures in properly
an abnormal cerebral than 0.88 s with the head at designed randomized, double-blinded, sham-con-
venous outflow in the 90 and 0 trolled studies, as well as the potential usefulness
presence of a structural Paradox: vein wall not of endovascular treatment of vein pathology
venous anomaly. reacting to respiratory phase;
These abnormalities
related to CCSVI [5, 25–27]. So far there have
noncompliant
include reflux/ been several case reports concerning patients who
No flow: no color flow noted
bidirectional flow, in vein, despite deep breaths had serious side effects after angioplasty for
paradox, and no flow CCSVI like IJV stent thrombosis requiring open
thrombectomy, stent migration, aneurismal vein
dilatation, cranial nerves neuropathy, as well as
multiple sclerosis (MS) [13, 14]. This returned some reports of lethal cases [2, 28, 29].
possible vascular etiology of MS lesions to the Furthermore, classification, existence, and
forefront of MS research. interpretation of venous abnormalities are ques-
CCSVI is characterized by impaired brain and tionable, given the fact that the same can be found
spinal cord venous drainage due to outflow among healthy populations and patients with
obstruction in the extracranial venous system, other CNS disorders [11, 12, 30, 31, 32, 33]. At
mostly related to anomalies in the internal jugular this time, it remains unclear whether extracranial
(IJV) and azygos veins [14–16]. It was hypothe- venous abnormalities represent an acquired
sized that these venous anomalies may cause alter- pathology or developmental variants.
ations to blood flow that eventually result in iron It is pretty clear that there is an urgent need to
deposition, degeneration of neurons, and charac- define and validate the spectrum of extracranial
teristic brain injury patterns. The CCSVI venous abnormalities and their relationship to
956 K. Dolic and R. Zivadinov

neurodegenerative diseases. Since there is still no than 50 % of cases, depending on the configura-
established noninvasive or invasive diagnostic tion of torcular Herophili. In 20 % of cases, one
imaging modality that can serve as a “gold stan- transverse sinus drains the SSS in total (most often
dard/benchmark” for the detection of extracranial on the right side), and the other one drains the
venous anomalies, it is most likely that only a straight sinus, which collects blood from the deep
multimodal imaging approach will represent the venous system [5, 37–39].
most comprehensive means for the screening, The cavernous sinus extends from the superior
diagnosis, as well as monitoring for these venous orbital fissure to the petrous apex, which receives
abnormalities ([2, 9, 23, 34]). orbital venous and middle cranial fossa drainage.
From the cavernous sinus, blood drains
posterolaterally along the superior petrosal sinus
Anatomy and Function of the Cerebral into the transverse sinus and inferior-laterally
Venous System along the inferior petrosal sinus into the sigmoid
sinus [5, 40]. The deep cerebral venous system
It is important to first appreciate the anatomy and drains the deep WM and the regions surrounding
function of the cerebral venous drainage system in the lateral and third ventricles or the basal cistern.
order to understand the potential role of the extra- Three veins unite just behind the interventricular
cranial venous system in diseases of the CNS and foramen of the Monro to form the internal cerebral
aging. The venous system contains approximately vein (s). These include the choroid vein, septal
70 % of the circulating blood volume, with vein, and thalamostriate vein. The vein of Galen is
approximately three-quarters of it within small a short (1–2 cm long), thick vein that passes
veins and venules [35, 36]. Cerebral venous sys- posterosuperiorly behind the splenium of corpus
tem drainage is composed of two systems: the callosum in the quadrigeminal cistern. The vein of
superficial and the deep venous system (Fig. 1). Galen receives the internal cerebral vein, the basal
The superficial system drains blood from the cor- veins of Rosenthal, and the posterior fossa veins
tex and superficial white matter (WM) by cortical and then drains to the anterior end of the straight
veins, collected by dural sinuses. There are two sinus where this unites with the inferior sagittal
important dural sinuses: the superior sagittal sinus sinus. The main collecting vein for the deep
(SSS) draining dorsolaterally and the cavernous venous system is the straight sinus, which
sinus draining antero-ventrally. The transverse receives the venous blood from the vein of
sinus then drains the SSS equally on both sides Galen and flows into the transverse sinus (most
in only 20 % of cases and asymmetrically in more often into the left side). The basal vein of

Fig. 1 Intracranial venous


system anatomy of dural
sinuses, cortical veins, deep
intracerebral veins, and
cavernous sinus (The figure
was reproduced with
permission from the
Radiology Assistant
website: (http://www.
radiologyassistant.nl/en/
p4befacb3e4691/cerebral-
venousthrombosis.html))
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 957

Rosenthal is an important collateral pathway for drainage system within the craniocervical junc-
the internal cerebral veins and the vein of Galen. tion region, which includes the anterior condylar
Venous blood flow can bypass the straight sinus confluent (ACC) and its tributes. Numerous anas-
by connecting with the superficial Sylvian vein tomoses of the ACC make it a crossroad between
via the deep Sylvian vein [38, 39, 41]. Venous the cavernous sinus, dural sinuses of the posterior
drainage of the posterior fossa mainly depends on fossa, IJVs, and posterior cervical outflow tract
the galenic system and the petrosal system and, to (vertebral venous system and deep cervical veins)
a lesser extent, the tentorial veins and the trans- [5, 36, 42, 43].
verse sinuses.

IJV Valves
Extracranial Cerebral Venous Drainage
Pathway: Neck Veins Although there are anatomical variations, the
valves are generally located about 0.5 cm above
The cerebral venous drainage pathway continues the union of the subclavian vein and IJVs at the
via neck veins, mainly the IJV, vertebral venous lower limit of the jugular bulb, which are shown in
system, and deep cervical veins (veins in cervical 96.8 % of the general population. They make IJVa
soft tissue) (Fig. 2) [5, 35, 38]. buffer zone between large central veins and the
The IJVs are the largest veins in the neck and cerebral venous system. The IJV valves are gen-
are generally considered to be the most important erally thought to prevent the backflow of venous
cerebral venous outflow pathways. Venous drain- blood and backward venous pressure into the
age of the superficial and deep cerebral venous cerebral venous system during conditions where
system is via the transverse sinuses to the sigmoid the central venous pressure or intrathoracic pres-
sinuses, which then drain into the IJV [5, 32, sure is increased, such as chest compression
36]. The inferior petrosal sinus, a major drainage during external cardiopulmonary resuscitation
route collecting blood from the cavernous sinus, and severe or repetitive cough and straining
communicates with the basilar plexus, anterior [5, 44–46]. The pressure gradient across compe-
and lateral condylar veins, anterior condylar con- tent IJV valves can be as high as 100 mmHg.
fluence, and vertebral venous plexus before Without competent IJV valves, a sustained or
draining into the IJVs. The IJVs then join with prolonged retrograde-transmitted venous pressure
the subclavian veins to form the brachiocephalic via IJVs might impair cerebral venous drainage
vein (BV). The confluence of the bilateral BV is and lead to neurological deficits. For example, IJV
the superior vena cava, which ultimately drains valve incompetence has been associated with
venous blood into the heart. Several tributaries in encephalopathy after cardiopulmonary resuscita-
the neck also drain into the IJVs. These bilateral tion [5, 47].
IJV branches will interconnect with each other at
the midline to form anastomosing plexi that can
serve as collateral channels to maintain adequate Other Neck Veins Serving as Collaterals
venous drainage when the principal pathways are for Cerebral Venous Drainage
obstructed [5].
The vertebral venous system consists of two When there is narrowing of the principal path-
components; one is the vertebral venous plexus ways of the extracranial venous system, collateral
and the other is the vertebral vein (VV). The veins probably represent physiological variations
vertebral venous plexus can be subdivided as of the venous system that may play a compensa-
internal (posterior and anterior internal vertebral tory role [2, 13]. The extra-jugular venous drain-
plexus) and external (posterior and anterior exter- age system for cerebral venous drainage mainly
nal vertebral plexus). The IJVs can also exhibit consists of the vertebral venous system and deep
anastomosis with the other extracranial venous cervical veins [5, 36, 40, 48, 49]. The external
958 K. Dolic and R. Zivadinov

Fig. 2 Illustration depicting the predominant veins and was reproduced with permission from Lazzaro MA, Zaidat
sinuses involved in the craniocervical venous outflow. OO, Mueller-Kronast N, Taqi MA, Woo D Endovascular
Venous narrowing is depicted at locations of interest in therapy for chronic cerebrospinal venous insufficiency in
chronic cerebrospinal venous insufficiency. (The figure multiple sclerosis. Front Neurol 2011, 2:44)

jugular vein (EJV) and anterior jugular vein the anterior (jugular venous system) and the
(AJV), compared with the IJV, are located super- posterior (vertebral and other deep neck venous
ficially in the neck. They serve as collaterals and system), and different patterns of collateral estab-
become prominent (enlarged lumen) when the lishment may reflect the location and severity of
main cerebral venous drainage pathways (IJV venous outflow obstruction [5, 51, 52].
and VV) are compromised. EJV is formed by the
confluence of the posterior branch of the posterior
facial vein and the posterior auricular vein [5, 50]. Extracranial Cerebral Venous Drainage
It usually terminates into the confluence of the Pathway: Abdominal and Thoracic
subclavian vein and IJV. The AJV receives blood Veins
from superficial veins, such as EJVs, facial veins,
or IJVs. They usually end in the subclavian vein There is communication of vertebral venous
or EJV. Bilateral AJVs may communicate via the plexus system with the deep thoracic and lumbar
jugular venous arch (JVA), which is located just veins, intercostal veins, as well as the hemiazygos
above the sternum. The JVA receives tributaries and azygos veins. Abnormalities in these abdom-
from the thyroid gland via inferior thyroid veins. inal and thoracic veins may impair venous drain-
In summary, venous collaterals in the neck include age from the vertebral venous system, which
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 959

Anastomosis ant.
Ant.
cardinal veins
cardinal
vein

Common Supracardinal
cardinal vein vein

Post.
cardinal vein Hepatic
segment
int. vena cava

Subcardinal
vein

Int. Jugular V.
(Distal segment of Ext. Jugular V.
Precardinal Vein)
Subclavian V.
Innominate (right) V. Innominate (left) V.
(Brachiocephalic Vein)

Sup. Vena Cava


(Proximal segment of
Coronary sinus
Precardinal/Anterior
Cardinal Vein)

Azygos V. Accessory Hemiazygos V.


(The most proximal segment of
Right Postcardinal Vein with
distal extension by Right
Supercardinal Vein)

Hemiazygos V.

Fig. 3 Paired anterior cardinal veins form common cardi- atrophies to become the base of the “coronary sinus” of
nal veins with paired posterior cardinal veins, draining the heart as displayed. The right anterior cardinal
centrally into the sinus venosus (sinus horns) as depicted (precardinal) vein proximal to the right brachiocephalic
(top). Paired anterior cardinals soon form an anastomosis vein forms the superior vena cava (SVC) with the common
between them; the connection grows from the left to the cardinal and terminal/proximal segment of the posterior
right anterior cardinal vein to form the left brachiocephalic cardinal (postcardinal) vein (The figure was reproduced
(innominate) vein (bottom). The left anterior cardinal vein with permission from Lee BB: Venous embryology: the
distal (cranial) to the anastomosis becomes the “left inter- key to understanding anomalous venous conditions.
nal jugular vein,” while the left anterior cardinal vein Phlebolymphology 2012, 4:170–181)
proximal to the brachiocephalic anastomosis regresses/

serves as an important collateral for cerebral and developmental stages of the abdominal/tho-
venous drainage. The hemiazygos arch is racic blood vessels can be quite variable (Fig. 3).
connected with the left renal vein that represents For example, in some rare variations, the azygos
a major outflow route for shunting blood into the vein also drains thoracic veins, bronchial veins,
inferior vena cava. Ultimately, the azygos vein and even gonadal veins. The vein is so named
serves as the final venous blood collector and because it has no symmetrically equivalent vein
drains into the superior vena cava. The anatomy on the left side of the body [5, 6].
960 K. Dolic and R. Zivadinov

Extracranial Venous Abnormalities change, respiration, cardiac function,


Versus Developmental Variants hypovolemia, and hydration status even by the
pulsation of nearby arteries [2, 50, 53, 54]. The
The venous system development through stages current definition (narrowing of >50 %) with
may be associated with a number of developmen- respect to the proximal adjacent vein segment is
tal variants that do not necessarily represent path- mainly derived from observations in the arterial
ological findings. This is the reason why there is system [5, 55, 56]. Even less is known about the
still disagreement about the physiological range main drainage routes of the spine, namely, the
of hemodynamic measurements in these veins azygos venous system and its pathophysiology.
including determination of normal or abnormal When performing imaging of the extracranial
function. It has been hypothesized that these venous system, it is almost impossible to take all
abnormalities may be related to embryologic of the above factors into account, regardless of the
developmental arrest, aging, or other imaging modality utilized. While there is common
comorbidities [2, 6, 32]. Probably, even more knowledge among radiologists about how to per-
important is to establish what constitutes a signif- form diagnostic imaging and intervention on the
icant narrowing of the extracranial venous system extracranial arteries, no established standardized
with hemodynamic consequences for the intracra- guidelines for detection of extracranial venous
nial venous drainage. abnormalities currently exist. This chapter sum-
So far, there have been reported a huge range of marizes current knowledge regarding the advan-
possible abnormalities in the extracranial veins tages and disadvantages of both noninvasive and
draining cerebral venous blood flow. These can be invasive imaging modalities for the detection of
classified as structural/morphological, hemody- those extracranial venous pathologies (Tables 2
namic/functional, and those determined only by and 3).
the composite criteria and use of multimodal imag-
ing (Table 1) [2, 9]. For example, structural/mor-
phological venous abnormalities can be divided Noninvasive Imaging Modalities
into those creating narrowing or occlusion and
those causing abnormal distensibility. On the Doppler Sonography
other hand, hemodynamic functional venous
abnormalities represent an abnormal cerebral Description of Imaging Techniques
venous outflow in the presence or absence of a DS is clinically the most useful technique for
structural venous anomaly in the extracranial detecting, localizing, and evaluating peripheral
veins. Finally, because it is almost impossible to venous obstruction and venous valvular incompe-
determine the prevalence of a single structural/mor- tence [57, 58]. The sensitivity and specificity of
phologic or hemodynamic/functional venous venous DS for symptomatic proximal deep vein
abnormality, regardless of the imaging modality thrombosis exceeds 90 % [59, 60]. Spectral anal-
or methodology utilized, the need for use of com- ysis of the DS signal is used to confirm the pres-
posite criteria by uni- or multimodal imaging ence or absence of flow and indicates its direction
modalities of the extracranial venous system is and the patterns. Spectral analysis of DS signal
emerging [23]. and color DS are used to confirm the presence of
reflux. Several DS studies have shown a relation-
ship between IJV drainage abnormalities, charac-
Diagnostic Imaging and the Diagnosis terized by JVR and specific neurological diseases
of Extracranial Venous Abnormalities of undetermined etiology along with a higher
prevalence of brain WM hyperintensities in elderly
Unlike the carotid artery, the vascular wall of the people ([8, 12, 32, 61]). An increased prevalence of
IJVs is much more flexible with a variable lumen JVR, dilated vessel lumen, and slowed flow veloc-
diameter which can be influenced by postural ity in the left IJV, as well as decreased
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 961

Table 2 Advantages and disadvantages of noninvasive diagnostic methods for diagnosis of extracranial venous
anomalies (Adapted from Dolic et al. [2])
Noninvasive
diagnostic
methods Advantages Disadvantages
Doppler Noninvasive No standardized guidelines
sonography Without ionizating radiation Operator dependent
Less expensive Time-consuming (60–120 min)
High resolution Blinding procedures are challenging
Real-time information Cannot perform global view of the veins (limited
Sensitive to detect flow changes, intra- and window)
extraluminal abnormalities Misidentification of the veins
Ability to measure velocity Influenced by hydration status
Possible control of respiratory phases
Magnetic Noninvasive No real-time informations
resonance Without ionizating radiation Cannot detect intraluminal abnormalities
venography Well-established method Low specificity of conventional MRV techniques
Operator independent Influenced by hydration status
Less time-consuming than DS Azygos vein examination needs technical
Provide global view of intra- and improvements due to important artifacts
extracranial venous system (breathing, heart movements, etc.)
Easy to blind Underestimate the vascular caliber
Ability to measure flow and velocity with “Snapshot” nature
advanced technique (phase-contrast MRV)
Global view of collateral veins
Can be performed without contrast
(pregnancy, allergy, etc.)
Plethysmography Noninvasive Time-consuming method
Provides valuable information regarding the Higher false-positive rate due to venous
impact of reflux and obstruction on overall compression arising from incorrect patient
venous function positioning or the action of extrinsic masses
Can monitor the dynamics of venous disease Low resolution
over time and evaluation of treatment
outcomes

Table 3 Advantages and disadvantages of invasive diagnostic methods for diagnosis of extracranial venous abnormal-
ities (Adapted from Dolic et al. [2])
Invasive diagnostic
methods Advantages Disadvantages
Catheter Considered the gold standard Invasive method
venography Real-time information can be obtained by Ionizating radiation
using contrast Cannot be performed without contrast (allergy,
Ability to measure pressure toxicity, etc.)
Provide “road map” for planning Operator dependent
endovascular procedures Time-consuming (>45 min)
Can be complemented by use of more Cannot detect intraluminal abnormalities
sophisticated criteria (time to empty contrast No global view of veins and collaterals
from vein or wasting of the balloon) No standardized definition of significant
Vein stenoses
Intravascular Offers 360 view of the vessel’s wall from Invasive method
ultrasound the inside Lack of experience – no standardize protocols
Can detect intraluminal abnormalities Ring-down artifacts
Easily access all parts of IJVs in comparison Geometric distortion – from imaging in an
with DS oblique plane
Provides more accurate assessment of vein Size of IVUS probe – limitation in the imaging
stenosis and wall thickness than CV and DS of severe stenosis
962 K. Dolic and R. Zivadinov

Fig. 4 Examples of chronic cerebrospinal venous insuffi- CSA measurement of 3 mm2; (e, f) reflux/bidirectional
ciency venous hemodynamic criteria on Doppler sonogra- flow directed toward the brain for a duration of >0.88 s in
phy. (a) Flap anomalies noted in internal jugular vein (IJV) the right IJV in the supine position (E demonstrates reflux
lumen; (b) annulus in the left IJV: circumferential thick- using color flow, while F demonstrates reflux using spec-
ened vein wall that is restricting the vein from fully tral analysis – waveform noted above baseline for greater
expanding with respiratory or positional changes; (c) than 0.88 s) (The figure was reproduced with permission
thrombus noted in IJV; (d) severe stenosis of left IJV: from Dolic et al. [2])

time-averaged mean velocity of bilateral IJV was disorders, emphasizes the need for more quantita-
found in those over 70 years of age ([12, 32]). tive and reproducible measures for the integration
Because of the advantages of the DS in of morphological and functional anomalies. These
detecting intraluminal venous pathology, it was include blood flow as well as velocity and blood
initially promoted as a method of choice for volume measurements that could be potentially
screening of extracranial venous abnormalities more reliable in assessing the degree of venous
and developmental variants, indicative of CCSVI outflow obstruction in the IJVs and azygos vein
[17, 62]. The diagnosis of CCSVI is both based on (Fig. 5). Monti et al. performed quantitative evalu-
hemodynamic and imaging findings that utilize ation of cerebral venous blood outflow in the
DS to study the deep cerebral veins, the IJVs, supine and sitting positions and found significant
and the VVs in both erect and supine positions. alterations in cerebral venous outflow in MS
Recent findings suggest that the majority of patients in comparison with healthy controls and
CCSVI pathology is confined to the intraluminal patients with other neurological diseases ([64, 65]).
portion of extracranial veins which can be While the value of these CCSVI VH criteria in
visualised with high resolution B-mode imaging detecting venous abnormalities or developmental
while visible “stenoses” (Fig. 4) or extraluminal variants is uncertain and operator dependent, no
venous abnormalities most likely develop more other validated criteria have been proposed at this
frequently with the progression of the disease or time [23]. Recently, the International Society for
age [7, 56, 63]. Neurovascular Disease (ISNVD) developed a
The prevalence of CCSVI and JVR, as well as more comprehensive consensus document that
their relationship to clinical findings in CNS included the participation of more than
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 963

Fig. 5 Example of velocity (a) and volume (b) measurement over 4 s phase in internal jugular vein (IJV) (The figure was
reproduced with permission from Dolic et al. [2])

60 international experts in DS imaging [23]. DS direction without background Doppler noise. But
was proposed as a standardized screening tool for because of its limited availability, QDP technology
determining CCSVI status. The protocol proposes can only be considered as an additional criterion
also the use of quantitative measures for the def- for diagnosis of CCSVI.
inition of functional anomalies such as blood flow Also recently, the European Society of
velocity and volume (Fig. 5) that could be poten- Neurosonology and Cerebral Hemodynamics
tially more reliable in assessing the degree of (ESNCH) expressed considerable concerns regard-
venous outflow obstruction in the IJVs. It also ing the accuracy of the proposed criteria for CCSVI
refines originally proposed VH criteria. A novel in MS [19] and proposed the central blinded DS
technology called Quality Doppler Processing reading as part of a recent multicenter Italian
(QDP) [62, 66] designed to insonate the veins at CoSMo study investigating the prevalence of
the base of the skull may become useful in the near CCSVI in MS patients, healthy controls, and
future, because it is difficult to visualize the supe- patients with other neurologic diseases (OND) [67].
rior petrosal sinus, the inferior petrosal sinus, and Dynamic contrast-enhanced exams can poten-
the contralateral inferior and superior petrosal tially increase the value of DS by overcoming the
sinuses. Because insonation of the petrosal sinuses operator-dependent analysis of the ultrasonic sig-
requires a Doppler angle close to 90 , detection of nal [68]. Unlike the diffusible agents commonly
reflux in the form of bidirectional flow can be made used in MR imaging and computed tomography
only using a multi-angle Doppler system. The (CT), DS contrast agents are true intravascular
QDP technology enables the operator to determine agents that remain entirely within the vascular
the direction of blood flow within the examined space and possess intravascular characteristics
cerebral veins and make an adjustment of the pulse similar to those of red blood cells. This technique
repetition frequency (PRF) to clearly visualize the allows registration of changes in ultrasonic signal
964 K. Dolic and R. Zivadinov

intensity during and after intravenous injection of interindividual variation of the cerebral venous
a contrast agent. Time dependence of enhance- anatomy [72].
ment can be shown in a time-intensity curve DS also has limits regarding extracranial vein
(TIC) where the average signal from a region of characterization, since findings can be influenced
interest (ROI) is plotted as a function of time. by hydration status. It is a very time-consuming
Tissue and vascular uptake, transit times, and method, and visualization of the central veins,
washout of the contrast agent can also be obtained particularly in the thorax and abdomen, is often
from a TIC [69, 70]. Mancini et al. used contrast- limited and cannot give the global view of vein
enhanced ultrasonography of the internal jugular anatomy. Although it can detect extracranial col-
vein with TIC analysis and revealed significantly lateral veins which are probably associated with
reduction of washout rate in IJVs in MS patients CCSVI, it is not technically feasible to follow the
compared to healthy controls [68]. complete course of the collateral veins which can
be more easily visualized with the use of magnetic
Advantages resonance venography (MRV), computed tomog-
DS has the advantage among other diagnostic raphy venography (CTV), or CV. Other pitfalls in
techniques of being noninvasive, providing DS imaging include the misidentification of veins.
high-resolution images with real-time dynamic Additionally, overlying bone and muscle may
information such as flow and velocity, showing prevent continuous imaging (cannot visualize
intraluminal as well as extraluminal abnormali- suitably the confluence of the IJVs and the sub-
ties and developmental variants (Fig. 4), and clavian vein because the clavicle commonly
being considerably less expensive than other blocks direct visualization). Similarly, the cervical
noninvasive imaging techniques. DS imaging part of IJV and the jugular bulb cannot be visual-
can also be readily applied in the follow-up ized by DS because of the limited acoustic win-
period of subjects undergoing endovascular dow resulting from the spine, mandible, and skull.
treatment because it can recognize the associated So far, none of the recently published DS studies
complications (residual stenosis, restenosis, or have reproduced the originally reported CCSVI
venous thrombosis) (Fig. 4) [2, 5, 13, 22, 71]. prevalence, regardless of diagnostic DS method
DS can also assess the hemodynamic conse- utilized [5, 48, 71, 74, 75].
quences of outflow derangement, while B-mode
ultrasound detects structural venous intraluminal Magnetic Resonance Venography
anomalies (Fig. 4) [22, 71].
Description of Imaging Techniques
Disadvantages During the past decade, catheter-based digital
The main criticism of the recommended DS pro- subtraction angiography, as the preferred method
tocol is that its reproducibility depends on the for imaging of the intracranial venous anatomy,
training level and skills of the operator and it is has been increasingly supplanted by MRV, an
not easy to be blinded and standardized in either a often overlooked and underappreciated noninva-
research or clinical setting [7, 19, 72, 73]. More- sive and safe method for the evaluation of head
over, the value of the CCSVI VH criteria is and neck veins [76, 77]. In the absence of better
controversial because they combine functional noninvasive techniques for the imaging of the
and structural intra- and extracranial venous dural venous sinuses, well-known and
abnormalities/developmental variants in a single documented pitfalls associated with flow-
binary composite. Further, the assessment of the sensitive MR techniques have been tolerated [34,
second CCSVI criterion (reflux in deep cerebral 56, 78–81]. Furthermore, simple protocols that
veins) (Fig. 6) is particularly controversial incorporate two-dimensional time-of-flight
because the direction of the blood flow in veins (2D-TOF) pulse sequence have already improved
connecting cortical with deep veins may vary their accuracy for the diagnosis of deep venous
considerably as a consequence of the physiologic thrombosis involving the femoral, popliteal, or
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 965

Fig. 6 Example of reflux in the deep cerebral veins using Quality Doppler Profile (QDP). Doppler profile on opposite
sides of baseline (The figure was reproduced with permission from Dolic et al. [2])

iliac veins [82]; however, experience with MRV presence of venous abnormalities and their rela-
techniques in the cervical veins remains still tionship to CCSVI in MS.
limited.
The CCSVI concept triggered several MRV Time of Flight
studies that emphasized the value of MRV as a TOF venography has the advantage of simplicity
comprehensive, noninvasive, and relatively because no special pulse sequences are required,
operator-independent technique which provided and this technique is available on nearly every
a 3D structural assessment of the intra- and extra- MRI system. TOF pulse sequences are spoiled
cranial vasculature for the potential identification gradient-echo or gradient-echo acquisitions
of stenosis and quantification of blood flow performed sequentially, i.e., all “phase-encode”
through major veins [7, 48, 56, 78, 79, 81, steps are played out in a single slice before mov-
83–90]. But so far almost none of the MRV stud- ing on to the next slice, which results in much
ies found significant differences in extracranial greater suppression of stationary tissue [34, 77,
vein morphology between MS patients and con- 82]. It also has the advantage of avoiding the need
trols. It could be hypothesized that those so-called for the use of contrast agents, and it remains the
abnormalities in venous outflow likely reflect nor- technique of choice in the evaluation of the pregnant
mal developmental anatomic variants. Further, the patient with suspected dural sinus thrombosis [34,
findings of these studies suggest also that MRV 78]. Furthermore, the accompanying conventional
morphologic information by itself may be insuffi- MR study is more sensitive in terms of the detection
cient to allow conclusions to be drawn about the of cortical venous infarction than a CT [91].
966 K. Dolic and R. Zivadinov

Fig. 7 Example of normal and abnormal flow in internal left internal jugular vein on axial 2D time of flight (b). The
jugular vein on magnetic resonance venography. Normal figure was reproduced with permission from Dolic et al. [2]
flow in both internal jugular veins (a) and abnormal flow in

The axial orientation of the acquisition allows for imparted upon the moving spins to distinguish
high in-plane resolution, which is ideal for cross- flowing blood from the surrounding stationary
sectional area (CSA) measurements of the veins tissue, thus providing information regarding
(Fig. 7). However, the TOF sequence is easily both anatomy and flow (Fig. 8). The major
affected by motion artifacts, especially from the advantage of PC-MRI angiography is excellent
patient’s breathing, swallowing, snoring, or head background suppression as well as quantitative
motion [87, 92]. Relative insensitivity to in-plane determination of blood velocities [93, 94]. How-
flow is another limitation of the TOF technique. ever, it requires long imaging times and a prior
Regarding the direction of flow, the optimal acqui- estimate of blood flow velocity. Furthermore, it
sition plane lies orthogonal, which is inefficient may also be more sensitive to signal loss due to
from the standpoint of acquisition time and not turbulence or intravoxel dephasing. So far there
always achievable. Although it has a higher spatial are only a few studies that used PC-MRI to quan-
resolution, 2D TOF may overestimate stenosis in tify venous flow in MS patients compared to
the setting of turbulent or slow flow [78]. All in all, healthy controls [92, 95, 96]. More studies are
standard conventional MRV techniques are more needed to validate the venous flow at the upper
prone to artifacts than phase-contrast MRV and neck level on an adequate number of age- and
multidirectional TOF angiography [34, 56]. These gender-matched healthy controls with heteroge-
techniques can potentially alleviate some of the neous age groups.
usual MRV artifacts and provide more detailed
flow information. One obvious improvement is to Contrast-Enhanced Techniques
image at higher field strength, such as 3 T, because Contrast-enhanced (CE) MRV, 3D time-resolved
this increases signal-to-noise ratio and better char- imaging of contrast kinetics (TRICKS) angiogra-
acterizes slow flow. phy, is a noninvasive and safe method for the
evaluation of head and neck veins, without the
Phase-Contrast Imaging attendant risks of conventional angiography. It is
In contrast to TOF techniques, which rely mainly preferred over TOF angiography because contrast
on flow-related enhancement for producing vas- medium reduces the T1 relaxation time of blood
cular images, phase-contrast MR angiography and virtually eliminates the effect of saturation
(PC-MRA) uses velocity-induced phase shifts ([2, 97, 98]; Fig. 9).
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 967

Fig. 8 Example of internal


jugular vein pathology on a c
cine phase-contrast MRI
study. The regions of
interest (ROIs) outlined are
the internal jugular veins.
These ROIs were used to
measure the flow through
these vessels. An example
showing the flow
quantification magnitude
image in stenotic (a) and b d
normal IJV (b) and the flow
quantification on phase
images of the same IJVs (c,
d). Graph showing the
differences in velocity
between stenotic and
non-stenotic IJV (e) (The
figure was reproduced with
permission from Dolic
et al. [2])

Instantaneous Velocity Over Cardiac Cycle


e in the Right Internal Jugular
0
0 5 10 15 20 25 30 35

−5
Instantaneous Velocity (cm/s)

−10

−15

Representative_Stenotic_Velocity
−20
Representative_Nonstenotic_Velocity

−25
Instantaneous Phase of Cardiac Cycle

CE MRV is probably the most widely used In contrast to MRA, the limitation of CE MRV is
technique and is essentially identical to 3D CE that maximal contrast enhancement achieved in
MR angiography, employing a 3D spoiled veins is typically lower than arteries because the
gradient-echo sequence in conjunction with a contrast bolus is more dilute by the time it reaches
bolus of gadolinium-based contrast. Vascular con- the venous system [99]. To improve background
trast results from the T1-shortening effects of gad- suppression and emphasize vascular signal, fat
olinium on adjacent water protons and has saturation can be added to a 3D spoiled gradient-
relatively little dependence on inflow effects. echo sequence with a small increase in acquisition
968 K. Dolic and R. Zivadinov

Fig. 9 Example of normal and abnormal flow morphol- enhanced 3D time-resolved imaging of contrast kinetics
ogy in internal jugular vein on magnetic resonance venog- (TRICKS) (The figure was reproduced with permission
raphy. Normal (a, b) flow morphology in both and from Dolic et al. [2])
abnormal (c, d) flow in the left internal jugular vein on

time. 3D reconstruction of CE MRV data is some- evaluation of not only anatomic stenoses but also
what less straightforward than MR angiography their impact on venous waveforms. It is based on
reconstruction since the vein/background contrast the principle that moving protons change phase in
is lower and there is usually arterial as well as proportion to their velocity. By enabling a quali-
venous enhancement [2, 91, 100]. tative assessment of the presence and direction of
Veins can have variable MR imaging signal collateral circulation, velocity-encoded cine MR
intensity due to entry slice phenomenon, imaging provides information about the presence
in-plane flow, and flow turbulence effects and and severity of obstruction. The technique has
can have variable enhancement. The maximum been most extensively used for the evaluation of
intensity projection (MIP) volumetric reconstruc- patterns of blood flow in the thoracic aorta,
tions of these sequences often underestimate the including the characterization of abnormal flow
vascular caliber, especially when there are seg- patterns associated with pathologic disorders such
ments with decreased flow (velocity or volume) as ascending aortic aneurysm and dissection
[2, 91]. [101]. Recent studies have explored the use of
Disadvantages of CE MRV include the 4D flow imaging for other areas of vascular anat-
expense of the contrast agent as well as contrast omy and pathology, including intracranial arterial
toxicity and patient discomfort in obtaining and venous blood flow [79]. With its detailed
antecubital venous access. In case of dural sinus characterization of complex, dynamic blood-
thrombosis however, confident early diagnosis of flow patterns and its ability to quantify flow, the
this common and treatable disease can dramati- technique could supplement both current nonin-
cally reduce patient morbidity. vasive and invasive imaging of intra- and extra-
cranial vascular pathologic disorders. The
4D Flow Imaging diagnostic and monitoring value of 4D flow imag-
Another promising MR technique is cine velocity- ing of venous flow anomalies, indicative of
encoded phase-contrast 4D flow that may permit CCSVI, is currently lacking.
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 969

Advantages or may result from diminished flow above true


In relation to DS, the advantages are driven by stenoses commonly located at the confluent
MRV being a noninvasive technique, less time- region of the veins. Additionally, it cannot satis-
consuming, and less operator dependent. MRV factorily evaluate the azygos and hemiazygos
can also depict, easily and globally, the anatomy veins [2, 48, 78, 85, 103].
and morphology of the intra and extracranial Unlike DS, with most MR scanners, data can
venous system [2, 34, 102]. MRV is also only be collected in the supine position, although
extremely useful in detecting collateral veins, some scanners can do an upright scanning as well.
which probably represent physiological variations In a study that used positional MR imaging to
of the venous system that may play a compensa- describe the influence of positional changes on
tory role when there are more venous extracranial the cerebral venous outflow, it has been found
abnormalities present because they bypass that IJV strictures are a common finding in healthy
blocked veins and thereby reduce resistance to controls in the supine position without relevance
drainage [17, 56]. in the erect position, which questions the validity
The assessment of the possible prominence or of the DS VH criterion 5 (lack of collapse of the
collateralization of the extracranial veins in the IJV in upright posture) for the diagnosis of
neck by MRV is an important diagnostic step in CCSVI. It is obvious that this criterion (to study
examining the status of the venous system. Fur- the change in flow in the IJVs from supine to
ther, thoracic central veins are largely inaccessible sitting position) cannot be studied with the con-
by DS, and MRV is an excellent technique for the ventional MR system [104].
assessment of axillary, jugular, subclavian, supe- CTV is another multi-planar imaging tech-
rior vena cava, and pulmonary veins. Addition- nique complementary to MRV in global venous
ally, CTV always requires the use of intravenous system evaluation but with better spatial resolu-
contrast, while many non-contrast methods are tion and possibility of direct assessment of the
available with MRV, making MRV the preferred azygos vein (morphology, caliber, course, and
technique in patients who also suffer from renal possible calcifications). Although it is more avail-
insufficiency or contrast allergy. CTV may also able and less expensive as well as time-consuming
require two or more acquisitions to adequately method, there are no case-control CTV studies in
capture contrast opacification of the veins, thereby MS or other neurological patients [2, 50, 56]. This
increasing the radiation dose [2, 34, 56]. is most likely due to the potential for radiation
exposure to controls and need for use of contrast
Disadvantages agent. Buffalo group gained preliminary experi-
Finally, MRV suffers from its “snapshot” nature. ence by using CTV as part of a multimodal diag-
An accurate depiction of these veins requires mul- nostic approach in a currently ongoing
tiple views and maneuvers such as inspiration and “Prospective Randomized Endovascular Therapy
expiration, flexion and extension, as well as rota- in MS (PREMiSe)” study (Fig. 10; [9, 105]).
tion imaging of the neck. Its main disadvantages
are the lack of MRV dynamism in real time and Plethysmography
lower resolution compared with DS and CV (can-
not evaluate intraluminal pathology such as the Description of Imaging Techniques
immobile valves, webs, septations, membranes, Plethysmography is the only existing practical
and duplications) and it is affected by the nature noninvasive modality for global physiologic eval-
of the veins themselves, which are prone to col- uation of extremity veins. Moreover, the use of
lapse under frequently encountered conditions, as strain-gauge or air plethysmography to diagnose
opposed to arteries. MRV often detects spurious venous thrombosis in the lower extremities has
stenoses that are not confirmed by CV, especially been well documented [89, 106–108]. By inflat-
in the lower parts of IJVs. These stenoses may ing a cuff on the thigh, the constriction of veins
represent transient phasic narrowings (functional) causes the venous volume to rise. When the cuff is
970 K. Dolic and R. Zivadinov

Fig. 10 Example of multimodality imaging of extracra- venography (e), intravascular sonography (f), and axial
nial neck veins in the Prospective Randomized computed tomography venography (g) all showing venous
Endovascular Therapy in MS (PREMiSe) study. Axial abnormality of the left internal jugular vein (narrowing)
2D time of flight (a), enhanced 3D time-resolved imaging (The figure was reproduced with permission from Dolic
of contrast kinetics (b, c), Doppler sonography (d), catheter et al. [2])

released, the sensor detects rapid venous runoff vasoactive agents [106]. The technique is rarely
and a return to the resting blood volume. If throm- used in the cervical region. Despite their value in
bosis is present, the plethysmography will detect a the anatomical localization of disease, imaging
delay in the emptying process. modalities such as DS and CV cannot assess the
The identification and assessment of venous global severity of reflux or obstruction.
obstruction by plethysmography is based on an Zamboni et al. recently showed that cervical
estimation of these two parameters: venous capac- plethysmography is much less prone to operator
itance and venous resistance. Quite recently, error compared to DS and has great potential to be
plethysmography has been used to measure endo- used as an inexpensive diagnostic tool for dem-
thelial function as well as the vascular response to onstrating extracranial venous anomalies and
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 971

development variants [109]. Another study, plac- less sensitive in revealing the exact nature of
ing a strain-gauge collar around CCSVI patients’ narrowed extracranial vein segments. The first
necks and tipping them from the upright (90 ) to finding to consider when evaluating a patient for
supine position (0 ) in a chair showed that hemo- CCSVI is the degree of narrowing within the vein
dynamics of the extracranial venous system are as seen on CV and the decision as to what consti-
greatly altered in CCSVI patients [110]. Further, tutes a significant stenosis. The concept of a sig-
Viola et al. used transcranial brain photoplethys- nificant obstruction when the vessel has been
mography to measure the increase in cerebral reduced to 50 % of its diameter (which corre-
blood volume during both IJV compression for sponds to a 75 % reduction in CSA) is derived
10s in sitting position and concluded that it can be mainly from observations in the arterial system
used as a potential marker for diagnosis of [85, 114, 115]. However, these criteria may not be
CCSVI [111]. applicable in the venous system because there are
Further research is needed in identifying cutoff some fundamental differences. One potential
values and the reproducibility of the test along issue is that the IJV can vary significantly in
with assessing intra- and interobserver variability. both size and symmetry with various factors,
including hydration status, cardiac output, respi-
Advantages ratory excursions, as well as head position that can
The advantage of plethysmography is that it is a account for some of the noted variability [2, 33,
noninvasive technique that provides valuable 114]. Thus, determination of the diameter of the
information regarding the impact of reflux and vein by CV is often arbitrary, and therefore it
obstruction on overall venous function and can under- or overestimates the proper size of the
provide a measure of calf muscle pump function balloon for the angioplasty. In light of the high
(strain-gauge plethysmography) [102, 104]. It is pressures necessary to dilate the stenosis, proper
complementary modality to DS and is also valu- sizing is crucial to avoid injury to the vein by
able for monitoring the dynamics of venous dis- over-dilatation or early recurrent stenosis by
ease over time and for the evaluation of treatment under-dilatation (Fig. 11).
outcomes [109, 110]. The recent results from Prospective Random-
ized Endovascular Treatment in MS (PREMiSe)
Disadvantages study also showed that CV may not be sensitive
Unfortunately, as with their invasive counterparts, enough to reveal the exact nature of narrowed vein
most of the noninvasive tests display the funda- segments [5, 9, 116]. Veroux et al. in a large CV
mental dichotomy of providing either anatomic or study found venous angioplasty to be effective in
hemodynamic information. Plethysmography can only a minority of MS patients with severe
be prone to a higher false-positive rate due to impaired IJV flow [117]. Further, in a recent
venous compression arising from incorrect patient published multicenter CV study, Traboulsee
positioning or the action of extrinsic masses et al. found that extracranial venous narrowing
[108]. It is also a time-consuming method. of greater than 50 % is a frequent finding in
patients with multiple sclerosis, unaffected sib-
lings, and unrelated controls, which also calls in
Invasive Imaging Modalities question the existence of CCSVI as a clinical
entity [55].
Catheter Venography Uncertainties related to the use of CV for
detection of extracranial venous abnormalities
Description of Imaging Techniques can be grouped into several domains: the pre-
CV is usually considered to be the “gold standard” ferred vascular access for CV, whether IVUS
for defining the degree of stenosis in the blood should accompany a routine CV evaluation for
vessels associated with altered blood flow [13, 15, CCSVI, whether a phasic stenosis (i.e., a
85, 112, 113]. However, it has been found to be narrowing which is transient or dynamic) should
972 K. Dolic and R. Zivadinov

Fig. 11 Catheter venography of azygos and internal jug- stenosis of the proximal right IJV (c) (The figure was
ular veins. Example of normal patent lumen of the azygos reproduced with permission from Dolic et al. [2])
vein (a) and left internal jugular vein (IJV) (b). Significant

be regarded as a normal variant or pathological, septa, and flaps that represent majority of CCSVI
and how to classify venographically obvious pathology. Further, there is the concern that an
CCSVI lesions [2, 9, 23, 116]. intraluminal anomaly such as septae may easily
be displaced out of the way by an inflated balloon
Advantages but upon deflation fall right back in its original
CV has several important advantages, including position and continue to functionally obstruct
the ability to perform pressure gradient measure- flow [115, 116]. It is possible to use very dilute
ments as well as to provide a helpful “road map” contrast and cone-downed images at high rate of
for planning endovascular procedures [2, 17, 115]. acquisition to pick up some of these intraluminal
It is a commonly performed low-risk procedure features, but they are generally harder to detect on
with a safety record established over decades of CV using conventional acquisition parameters
experience [27, 112, 118]. and contrast strengths. On the other hand, these
abnormalities can be seen with high-resolution
Disadvantages color DS or intravascular ultrasound (IVUS)
Its invasiveness, use of contrast agents, and radi- B-mode imaging [116]. In addition, malformed
ation exposure make it suboptimal as a routine and/or reversed valve cusps can be crossed by
screening tool in a clinical setting. It is also oper- the catheter and kept open artificially, thereby
ator dependent, only AP projection views are rou- preventing the documentation of stenosis.
tinely obtained, and stenosis assessment may
depend on the precise locations and rates of con- Intravascular Sonography
trast injection; CV can only show the collaterals
that drain the specific vein being injected without Description of Imaging Techniques
the possibility of showing global extracranial IVUS is an endoluminal CV-based DS technique
venous system at once, i.e., as with MRV or that offers a tomographic, 360 view of the ves-
CTV [2, 34, 116]. The display of extracranial sel’s wall from the inside [119]. The most com-
venous structures can be improved with additional mon indications for IVUS have been in the
injected contrast medium, more selective cathe- evaluation and treatment of arterial disease
terization, and additional projections [2, 116]. [120–122]. Its excellent resolution compared
Although CV is a luminogram, it brings little or with angiography has contributed to the under-
no data regarding the vessel’s intraluminal struc- standing of the pathophysiology and enhanced
tures, because of dense opacification of the lumen diagnosis of coronary artery disease achieving
with contrast, which obliterates subtle new milestones in interventional cardiology.
intraluminal structures like valve malformations, IVUS has been shown to provide a more accurate
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 973

Fig. 12 Example of intravascular ultrasound in the internal jugular vein. Normal patent lumen (a) and stenotic lumen (b)
with fibrotic wall (The figure was reproduced with permission from Dolic et al. [2])

assessment of vessel circumference and CSA and intraluminal venous anomalies in IJVs and azygos
thus is useful in detecting critical stenoses [123, vein and more accurate in measurement of steno-
124, 125]. While values for stenosis definition sis and wall thickness and allows for the explora-
used for CV (50 %) rely on a ratio between the tion of pulsatility in the veins [5].
stenotic segment diameter and a pre(non)-stenotic
vein which is more variable, the IVUS definition Advantages
is more strict (a lumen that embraces the IVUS The exploration of IJV valves is particularly well
probe for a critical stenosis) and does not refer to a seen on IVUS. Additionally, thrombus and dissec-
non-stenotic segment. It remains unclear at what tions are readily seen on IVUS. It can also show
level and with what criteria is there a significant the degrees of echogenicity, both of the vessel
hemodynamic effect of stenosis by either modal- wall and of the intraluminal thrombi, which may
ity [116]. Venous stenosis is currently measured indicate varying degrees of wall thickness and
using arterial criteria, which are clearly not opti- may correlate with the age of the thrombosis, an
mal. The hemodynamics of venous flow remain a important aspect of the vessel pathology that is not
major area of investigation, and better understand- possible to be determined with CV [80, 119].
ing will likely lead to a revision of stenosis Additionally, it provides an image with a
criteria. greater resolution of both lumen and wall (with
Although diagnostic experience is growing additional 3D features), providing better vessel
with the use of IVUS for investigation of both wall information. IVUS imaging may reflect
intra- and extracranial arteries, there is limited truly the size of stenotic lesions because it allows
literature regarding its use for the exploration of more complete and accurate assessment than is
venous vasculature in general, as well as specifi- possible with the use of CV examination
cally in relation to the investigation of venous [116]. Further, analysis of the vessel dimensions
abnormalities and developmental variants indica- allows a more accurate selection of balloon size,
tive of CCSVI (Fig. 12) [9, 116, 126, 127]. Recent thus reducing the risk of injury and providing a
studies showed that the IVUS assessment of IJVs more effective angioplasty. It provides cross-
and azygos vein can detect higher rates of venous sectional, in vivo visualization and the demonstra-
abnormalities than CV and that provides a diag- tion of the motility of small intraluminal struc-
nostic advantage over the “gold-standard” CV in tures, like bulging cusps as well as septum and
detecting extracranial venous abnormalities and webs, which cannot be optimally revealed by
developmental variants indicative of CCSVI traditional diagnostic methods [9, 116, 126]. It
[116, 126]. Also, PREMiSe study showed that has been shown that such venous pathology in
IVUS is more accurate in the detection of the iliac vein is unrecognized by CV and is well
974 K. Dolic and R. Zivadinov

visualized by IVUS [79]. Moreover, CV is inca- techniques were applied and compared [7, 17, 48,
pable of monitoring respiratory pulsatility which 62, 78, 83, 85, 100, 111, 116, 117, 118, 126, 128,
involves periods with reduced vessel diameter 129, 130]. The findings of these studies are
that can be investigated with IVUS [80]. extremely important to understand the true preva-
The advantages of IVUS compared with DS, lence of CCSVI, and the comparison of invasive
among others, include the sonographic penetra- vs. noninvasive imaging findings is especially
tion from within the vessel by excluding extravas- important in this endeavor. It is emerging that
cular soft tissues. It also assesses blood vessels not the prevalence of venous abnormalities and devel-
easily accessible by conventional DS, such as the opmental variants, indicative of CCSVI, is even
lower part of the IJV (behind the clavicle), upper higher when investigated with sophisticated inva-
part of the IJV, intracranial sinuses, and azygos sive imaging techniques [63, 116, 126, 127].
vein [9, 126]. Based on these recent findings, a multimodal
approach is recommended to determine whether
Disadvantages CCSVI exists as a clinical entity and not as an
Ring-down artifacts produced by acoustic oscilla- anatomic variant and to what extent it is present in
tions in the piezoelectric transducer that obscures various healthy and disease groups as well as MS
the near field result in an acoustic catheter size subtypes (Fig. 10; [23]). The introduction of more
larger than its physical size and may adversely quantitative criteria to describe extracranial
affect IVUS images. Geometric distortion can venous structural and hemodynamic functional
result from imaging in an oblique plane (not per- impairment in future multimodal approach studies
pendicular to the long axis of the vessel) [2, will be a significant improvement compared to the
81]. Furthermore, visible distortion of the image current binary CCSVI diagnosis.
can be due to another important artifact,
“nonuniform rotational distortion,” which arises
from uneven drag on the drive cable of the mechan- Summary
ical style catheters, resulting in cyclical oscillations
in rotational speed. The physical size of IVUS There is an urgent need to define and validate the
catheters (currently  1.0 mm) constitutes an spectrum of extracranial venous abnormalities
important limitation in the imaging of severe ste- and to establish reliable, diagnostic gold-standard
noses. Further depending on the probe, there is a test(s). Each noninvasive and invasive imaging
finite limit to IVUS resolution which rapidly modality has its own inherent advantages and
degrades beyond this particular radius typically disadvantages [2, 9]. Most likely, only multi-
10–12 mm [81]. In summary, the frequency of the modal imaging will eventually become the reli-
transducer, gain settings, depth of penetration, and able screening, diagnostic, and monitoring tool
focal depth are some of the factors that affect the for the assessment of the extracranial venous sys-
sensitivity of the IVUS imaging. tem [23]. The classification of the presence and
severity of extracranial venous abnormalities/
developmental variants by imaging and pathology
Multimodal Imaging Approach findings should be the first step in the determina-
tion of their role in the pathology of CNS disor-
The dramatic difference in prevalent findings ders and aging.
between different studies using noninvasive and The use of noninvasive methods to confirm
invasive imaging techniques emphasizes the presence of extracranial venous abnormalities
urgent need for the use of a multimodal imaging will be an essential step toward better understand-
approach for better understanding of the extracra- ing of its importance in general population and
nial venous abnormalities and developmental disease states. Regarding noninvasive techniques
variants indicative of CCSVI. In a number of there is still no consensus on DS protocols or use
recent studies, noninvasive and invasive imaging of MRV as alternative approach which could
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 975

ensure appropriate quality control for the determi- results of the PREMiSe pilot study. BMC Neurol
nation of extracranial venous abnormalities 13:151
10. Chuang YM, Hu HH (2005) Cough headache and
[23]. Thus, at present, the true prevalence of extra- thoracic inlet valvular competence in uremia. Eur
cranial venous abnormalities in MS patients and Neurol 53(2):78–80
other CNS disorders or healthy population has not 11. Chung CP, Hsu HY, Chao AC, Sheng WY, Soong
been adequately assessed. BW, Hu HH (2007) Transient global amnesia: cere-
bral venous outflow impairment-insight from the
Nevertheless, the concept of CCSVI has trig- abnormal flow patterns of the internal jugular vein.
gered an intense and rapid accumulation of Ultrasound Med Biol 33(11):1727–1735
knowledge over the past 4 years and has stimu- 12. Chung CP, Wang PN, Wu YH et al (2011) More
lated the need for further research to better under- severe white matter changes in the elderly with jugu-
lar venous reflux. Ann Neurol 69(3):553–559
stand the function and potential role of the 13. Zamboni P, Galeotti R, Menegatti E et al (2009) A
extracranial venous system. Only properly prospective open-label study of endovascular treat-
designed, safe, and ethical studies using noninva- ment of chronic cerebrospinal venous insufficiency.
sive and invasive imaging techniques IVUS J Vasc Surg 50(6):1348–1358, e1-3
14. Zamboni P, Galeotti R, Menegatti E et al (2009)
should be encouraged in collecting this longitudi- Chronic cerebrospinal venous insufficiency in
nal observational information. patients with multiple sclerosis. J Neurol Neurosurg
Psychiatry 80(4):392–399
15. Al-Omari MH, Al-Bashir A (2012) Internal jugular
References vein valve morphology in the patients with chronic
cerebrospinal venous insufficiency (CCSVI); angio-
1. Eberhardt RT, Raffetto JD (2005) Chronic venous graphic findings and schematic demonstrations. Rev
insufficiency. Circulation 111(18):2398–2409 Recent Clin Trials 7(2):83–87
2. Dolic K, Siddiqui AH, Karmon Y, Marr K, Zivadinov 16. Haacke EM (2011) Chronic cerebral spinal venous
R (2013) The role of noninvasive and invasive diag- insufficiency in multiple sclerosis. Expert Rev
nostic imaging techniques for detection of extra- Neurother 11(1):5–9
cranial venous system anomalies and developmental 17. Zamboni P, Menegatti E, Galeotti R et al (2009) The
variants. BMC Med 11:155 value of cerebral Doppler venous haemodynamics in
3. Burrows PE, Konez O, Bisdorff A (2003) Venous the assessment of multiple sclerosis. J Neurol Sci
variations of the brain and cranial vault. Neuroimag- 282(1–2):21–27
ing Clin N Am 13(1):13–26 18. Zamboni P, Morovic S, Menegatti E, Viselner G,
4. Dake MD, Zivadinov R, Haacke EM (2011) Chronic Nicolaides AN (2011) Screening for chronic cerebro-
cerebrospinal venous insufficiency in multiple sclero- spinal venous insufficiency (CCSVI) using ultrasoun-
sis: a historical perspective. Funct Neurol 26(4): d–recommendations for a protocol. Int Angiol
181–195 30(6):571–597
5. Zivadinov R, Chung CP (2013) Potential involve- 19. Baracchini C, Valdueza JM, Del Sette M et al (2012)
ment of the extracranial venous system in central CCSVI and MS: a statement from the European Soci-
nervous system disorders and aging. BMC Med ety of neurosonology and cerebral hemodynamics.
11:260 J Neurol 259(12):2585–2589
6. Lee BB, Laredo J, Neville R (2010) Embryological 20. Mayer CA, Pfeilschifter W, Lorenz MW et al (2011)
background of truncular venous malformation in the The perfect crime? CCSVI not leaving a trace in MS.
extracranial venous pathways as the cause of chronic J Neurol Neurosurg Psychiatry 82(4):436–440
cerebro-spinal venous insufficiency. Int Angiol 21. Oger J, Alkhawajah M (2010) CCSVI: hope, hype or
29:95–108 snake oil? Can J Neurol Sci 37(6):716
7. Dolic K, Marr K, Valnarov V et al (2012) Intra- and 22. Nicolaides AN, Morovic S, Menegatti E, Viselner G,
extraluminal structural and functional venous anom- Zamboni P (2011) Screening for chronic cerebrospi-
alies in multiple sclerosis, as evidenced by 2 noninva- nal venous insufficiency (CCSVI) using ultrasound:
sive imaging techniques. AJNR Am J Neuroradiol recommendations for a protocol. Funct Neurol
33(1):16–23 26(4):229–248
8. Chung CP, Cheng CY, Zivadinov R et al (2013) Jug- 23. Zivadinov R, Bastianello S, Dake M, et al (2014)
ular venous reflux and plasma endothelin-1 are asso- Recommendations for multimodal noninvasive
ciated with cough syncope: a case control pilot study. and invasive screening for detecting extracranial
BMC Neurol 13:9 venous abnormalities, indicative of chronic cerebro-
9. Zivadinov R, Karmon Y, Dolic K et al (2013) Multi- spinal venous insufficiency (CCSVI). A position
modal noninvasive and invasive imaging of extracra- statement of International Society for Neurovascular
nial venous abnormalities indicative of CCSVI: Disease (ISNVD). J Vasc Interv Radiol. Sep 22.
976 K. Dolic and R. Zivadinov

pii: S1051–0443(14)00746–5. doi:10.1016/j. normal function in humans. Brain Res Brain Res
jvir.2014.07.024 Rev 46(3):243–260
24. Van den Berg PJ, Visser LH (2013) The fluctuating 39. Schmidek HH, Auer LM, Kapp JP (1985) The cere-
natural course of CCSVI in MS patients and controls, bral venous system. Neurosurgery 17(4):663–678
a prospective follow-up. PLoS One 8(11):e78166 40. Caruso RD, Rosenbaum AE, Chang JK, Joy SE
25. Cortes Nino Mdel P, Tampieri D, Melancon D (2010) (1999) Craniocervical junction venous anatomy on
Endovascular venous procedures for multiple sclero- enhanced MR images: the suboccipital cavernous
sis? Mult Scler 16(7):771–772 sinus. AJNR Am J Neuroradiol 20(6):1127–1131
26. Mandato KD, Hegener PF, Siskin GP et al (2012) 41. Ono M, Rhoton AL Jr, Peace D, Rodriguez RJ (1984)
Safety of endovascular treatment of chronic cerebro- Microsurgical anatomy of the deep venous system of
spinal venous insufficiency: a report of 240 patients the brain. Neurosurgery 15(5):621–657
with multiple sclerosis. J Vasc Interv Radiol 23(1): 42. Andeweg J (1996) The anatomy of collateral venous
55–59 flow from the brain and its value in aetiological inter-
27. Petrov I, Grozdinski L, Kaninski G, Iliev N, Iloska M, pretation of intracranial pathology. Neuroradiology
Radev A (2011) Safety profile of endovascular treat- 38(7):621–628
ment for chronic cerebrospinal venous insufficiency 43. Epstein HM, Linde HW, Crampton AR, Ciric IS,
in patients with multiple sclerosis. J Endovasc Ther Eckenhoff JE (1970) The vertebral venous plexus as
18(3):314–323 a major cerebral venous outflow tract. Anesthesiology
28. Burton JM, Alikhani K, Goyal M et al (2011) Compli- 32(4):332–337
cations in MS patients after CCSVI procedures abroad 44. Akkawi NM, Agosti C, Borroni B et al (2002) Jugular
(Calgary, AB). Can J Neurol Sci 38(5):741–746 valve incompetence: a study using air contrast ultra-
29. Valdueza JM, Doepp F, Schreiber SJ et al (2013) sonography on a general population. J Ultrasound
What went wrong? The flawed concept of cerebrospi- Med 21(7):747–751
nal venous insufficiency. J Cereb Blood Flow Metab 45. Brownlow RL Jr, McKinney WM (1985) Ultrasonic
33(5):657–668 evaluation of jugular venous valve competence.
30. Beggs CB (2013) Venous hemodynamics in neuro- J Ultrasound Med 4(4):169–172
logical disorders: an analytical review with hydrody- 46. Silva MA, Deen KI, Fernando DJ, Sheriffdeen AH
namic analysis. BMC Med 11:142 (2002) The internal jugular vein valve may have a
31. Beggs C, Chung CP, Bergsland N et al (2013) Jugular significant role in the prevention of venous reflux:
venous reflux and brain parenchyma volumes in evidence from live and cadaveric human subjects.
elderly patients with mild cognitive impairment and Clin Physiol Funct Imaging 22(3):202–205
Alzheimer’s disease. BMC Neurol 13:157 47. Fisher J, Vaghaiwalla F, Tsitlik J et al (1982) Deter-
32. Chung CP, Lin YJ, Chao AC et al (2010) Jugular minants and clinical significance of jugular venous
venous hemodynamic changes with aging. Ultra- valve competence. Circulation 65(1):188–196
sound Med Biol 36(11):1776–1782 48. Dolic K, Marr K, Valnarov V et al (2011) Sensitivity
33. Zivadinov R, Marr K, Cutter G et al (2011) Preva- and specificity for screening of chronic cerebrospinal
lence, sensitivity, and specificity of chronic cerebro- venous insufficiency using a multimodal
spinal venous insufficiency in MS. Neurology 77(2): non-invasive imaging approach in patients with mul-
138–144 tiple sclerosis. Funct Neurol 26(4):205–214
34. Zivadinov R, Lopez-Soriano A, Weinstock-Guttman 49. Zamboni P, Galeotti R (2010) The chronic cerebro-
B et al (2011) Use of MR venography for character- spinal venous insufficiency syndrome. Phlebology
ization of the extracranial venous system in patients 25(6):269–279
with multiple sclerosis and healthy control subjects. 50. Escott EJ, Branstetter BF (2006) It’s not a cervical
Radiology 258(2):562–570 lymph node, it’s a vein: CT and MR imaging findings
35. Doepp F, Schreiber SJ, von Munster T, Rademacher J, in the veins of the head and neck. Radiographics
Klingebiel R, Valdueza JM (2004) How does the 26(5):1501–1515
blood leave the brain? A systematic ultrasound anal- 51. Deslaugiers B, Vaysse P, Combes JM et al (1994)
ysis of cerebral venous drainage patterns. Neuroradi- Contribution to the study of the tributaries and the
ology 46(7):565–570 termination of the external jugular vein. Surg Radiol
36. San Millan Ruiz D, Gailloud P, Rufenacht DA, Anat 16(2):173–177
Delavelle J, Henry F, Fasel JH (2002) The 52. Schummer W, Schummer C, Bredle D, Frober R
craniocervical venous system in relation to cerebral (2004) The anterior jugular venous system: variability
venous drainage. AJNR Am J Neuroradiol 23(9): and clinical impact. Anesth Analg 99(6):1625–1629
1500–1508 53. Beddy P, Geoghegan T, Ramesh N et al (2006)
37. Meder JF, Chiras J, Roland J, Guinet P, Bracard S, Valsalva and gravitational variability of the internal
Bargy F (1994) Venous territories of the brain. jugular vein and common femoral vein: ultrasound
J Neuroradiol 21(2):118–133 assessment. Eur J Radiol 58(2):307–309
38. Schaller B (2004) Physiology of cerebral venous 54. Schreiber SJ, Lurtzing F, Gotze R, Doepp F,
blood flow: from experimental data in animals to Klingebiel R, Valdueza JM (2003) Extrajugular
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 977

pathways of human cerebral venous blood drainage of CCSVI in multiple sclerosis (CoSMo study): ratio-
assessed by duplex ultrasound. J Appl Physiol 94(5): nale, design, and methodology. Neurol Sci 34(8):
1802–1805 1297–1307
55. Traboulsee AL, Knox KB, Machan L et al (2014) 68. Mancini M, Morra VB, Di Donato O et al (2012)
Prevalence of extracranial venous narrowing on cath- Multiple sclerosis: cerebral circulation time. Radiol-
eter venography in people with multiple sclerosis, their ogy 262(3):947–955
siblings, and unrelated healthy controls: a blinded, 69. Shalhoub J, Owen DR, Gauthier T, Monaco C, Leen
case–control study. Lancet 383(9912):138–145 EL, Davies AH (2010) The use of contrast enhanced
56. Zivadinov R, Galeotti R, Hojnacki D et al (2011) ultrasound in carotid arterial disease. Eur J Vasc
Value of MR venography for detection of internal Endovasc Surg 39(4):381–387
jugular vein anomalies in multiple sclerosis: a pilot 70. Staub D, Partovi S, Imfeld S et al (2013) Novel
longitudinal study. AJNR Am J Neuroradiol 32(5): applications of contrast-enhanced ultrasound imaging
938–946 in vascular medicine. Vasa 42(1):17–31
57. Meissner MH, Moneta G, Burnand K et al (2007) The 71. Thapar A, Lane T, Nicholas R et al (2011) Systematic
hemodynamics and diagnosis of venous disease. review of sonographic chronic cerebrospinal venous
J Vasc Surg 46(S):4S–24S insufficiency findings in multiple sclerosis. Phlebol-
58. Perone N, Bounameaux H, Perrier A (2001) Compar- ogy 26(8):319–325
ison of four strategies for diagnosing deep vein throm- 72. Doepp F, Paul F, Valdueza JM, Schmierer K,
bosis: a cost-effectiveness analysis. Am J Med Schreiber SJ (2010) No cerebrocervical venous con-
110(1):33–40 gestion in patients with multiple sclerosis. Ann
59. Goodacre S, Sampson F, Stevenson M et al (2006) Neurol 68(2):173–183
Measurement of the clinical and cost-effectiveness of 73. Menegatti E, Genova V, Tessari M et al (2010) The
non-invasive diagnostic testing strategies for deep reproducibility of colour Doppler in chronic cerebro-
vein thrombosis. Health Technol Assess 10(15): spinal venous insufficiency associated with multiple
1–168, iii-iv sclerosis. Int Angiol 29(2):121–126
60. Kassai B, Boissel JP, Cucherat M, Sonie S, Shah NR, 74. McDonald S, Iceton JB (2012) The use of Doppler
Leizorovicz A (2004) A systematic review of the ultrasound in the diagnosis of chronic cerebrospinal
accuracy of ultrasound in the diagnosis of deep venous insufficiency. Tech Vasc Interv Radiol 15(2):
venous thrombosis in asymptomatic patients. Thromb 113–120
Haemost 91(4):655–666 75. Zaniewski M, Kostecki J, Kuczmik W et al (2013)
61. Chung CP, Beggs C, Wang PN et al (2014) Jugular Neck duplex Doppler ultrasound evaluation for
venous reflux and white matter abnormalities in assessing chronic cerebrospinal venous insufficiency
Alzheimer’s disease: a pilot study. J Alzheimers Dis in multiple sclerosis patients. Phlebology 28(1):24–31
39(3):601–609 76. Prince MR, Sostman HD (2003) MR venography:
62. Zamboni P, Consorti G, Galeotti R et al (2009) unsung and underutilized. Radiology 226(3):630–632
Venous collateral circulation of the extracranial cere- 77. Vogt FM, Herborn CU, Goyen M (2005) MR venog-
brospinal outflow routes. Curr Neurovasc Res 6(3): raphy. Magn Reson Imaging Clin N Am 13(1):
204–212 113–129, vi
63. Lugli M, Morelli M, Guerzoni S, Maleti O (2012) The 78. Zaharchuk G, Fischbein NJ, Rosenberg J, Herfkens
hypothesis of patho-physiological correlation RJ, Dake MD (2011) Comparison of MR and contrast
between chronic cerebrospinal venous insufficiency venography of the cervical venous system in multiple
and multiple sclerosis: rationale of treatment. Phle- sclerosis. AJNR Am J Neuroradiol 32(8):1482–1489
bology 27(Suppl 1):178–186 79. Hope MD, Purcell DD, Hope TA et al (2009) Com-
64. Monti L, Menci E, Piu P et al (2014) A sonographic plete intracranial arterial and venous blood flow eval-
quantitative cutoff value of cerebral venous outflow in uation with 4D flow MR imaging. AJNR Am J
neurologic diseases: a blinded study of 115 subjects. Neuroradiol 30(2):362–366
AJNR Am J Neuroradiol Jul;35(7):1381–6. 80. Neglen P, Raju S (2002) Intravascular ultrasound scan
doi:10.3174/ajnr.A3864. Epub 2014 Mar 7 evaluation of the obstructed vein. J Vasc Surg 35(4):
65. Monti L, Menci E, Ulivelli M et al (2011) Quantita- 694–700
tive Colour Doppler Sonography evaluation of cere- 81. Finet G, Maurincomme E, Tabib A et al (1993) Arti-
bral venous outflow: a comparative study between facts in intravascular ultrasound imaging: analyses and
patients with multiple sclerosis and controls. PLoS implications. Ultrasound Med Biol 19(7):533–547
One 6(9):e25012 82. Kanne JP, Lalani TA (2004) Role of computed tomog-
66. Ciccone MM, Galeandro AI, Scicchitano P raphy and magnetic resonance imaging for deep
et al (2012) Multigate quality Doppler profiles and venous thrombosis and pulmonary embolism. Circu-
morphological/hemodynamic alterations in multiple lation 109(12 Suppl 1):I15–I21
sclerosis patients. Curr Neurovasc Res 9(2):120–127 83. Doepp F, Wurfel JT, Pfueller CF et al (2011) Venous
67. Comi G, Battaglia MA, Bertolotto A et al (2013) Ital- drainage in multiple sclerosis: a combined MRI and
ian multicentre observational study of the prevalence ultrasound study. Neurology 77(19):1745–1751
978 K. Dolic and R. Zivadinov

84. Haacke EM, Feng W, Utriainen D et al (2012) Patients J Magn Reson Imaging Aug;40(2):341–7.
with multiple sclerosis with structural venous abnor- doi:10.1002/jmri.24388.
malities on MR imaging exhibit an abnormal flow 97. Farb RI, Scott JN, Willinsky RA, Montanera WJ,
distribution of the internal jugular veins. J Vasc Interv Wright GA, terBrugge KG (2003) Intracranial venous
Radiol 23(1):60–68, e1-3 system: gadolinium-enhanced three-dimensional MR
85. Hojnacki D, Zamboni P, Lopez-Soriano A et al (2010) venography with auto-triggered elliptic centric-
Use of neck magnetic resonance venography, Doppler ordered sequence–initial experience. Radiology
sonography and selective venography for diagnosis of 226(1):203–209
chronic cerebrospinal venous insufficiency: a pilot 98. Liang L, Korogi Y, Sugahara T et al (2001) Evaluation
study in multiple sclerosis patients and healthy con- of the intracranial dural sinuses with a 3D contrast-
trols. Int Angiol 29(2):127–139 enhanced MP-RAGE sequence: prospective compar-
86. Utriainen D, Trifan G, Sethi S et al (2012) Magnetic ison with 2D-TOF MR venography and digital sub-
resonance imaging signatures of vascular pathology traction angiography. AJNR Am J Neuroradiol 22(3):
in multiple sclerosis. Neurol Res 34(8):780–792 481–492
87. Utriainen D, Feng W, Elias S, Latif Z, Hubbard D, 99. Hingwala DR, Thomas B, Kesavadas C,
Haacke EM (2012) Using magnetic resonance imag- Kapilamoorthy TR (2011) Suboptimal contrast
ing as a means to study chronic cerebral spinal venous opacification of dynamic head and neck MR angiog-
insufficiency in multiple sclerosis patients. Tech Vasc raphy due to venous stasis and reflux: technical con-
Interv Radiol 15(2):101–112 siderations for optimization. AJNR Am J Neuroradiol
88. Wattjes MP, van Oosten BW, de Graaf WL 32(2):310–314
et al (2011) No association of abnormal cranial 100. Zivadinov R, Ramanathan M, Dolic K et al (2011)
venous drainage with multiple sclerosis: a magnetic Chronic cerebrospinal venous insufficiency in multi-
resonance venography and flow-quantification study. ple sclerosis: diagnostic, pathogenetic, clinical and
J Neurol Neurosurg Psychiatry 82(4):429–435 treatment perspectives. Expert Rev Neurother 11(9):
89. Heijboer H, Buller HR, Lensing AW, Turpie AG, 1277–1294
Colly LP, ten Cate JW (1993) A comparison of real- 101. Hom JJ, Ordovas K, Reddy GP (2008) Velocity-
time compression ultrasonography with impedance encoded cine MR imaging in aortic coarctation: func-
plethysmography for the diagnosis of deep-vein tional assessment of hemodynamic events. Radio-
thrombosis in symptomatic outpatients. N Engl J graphics 28(2):407–416
Med 329(19):1365–1369 102. Glockner J (2010) Magnetic resonance venography.
90. Rosales M, Radeva P, Rodriguez-Leor O, Gil D Appl Radiol 39(6):36
(2009) Modelling of image-catheter motion for 3-D 103. Hartel M, Kluczewska E, Simka M, Ludyga T,
IVUS. Med Image Anal 13(1):91–104 Kostecki J, Zaniewski M (2011) Magnetic resonance
91. Ayanzen RH, Bird CR, Keller PJ, McCully FJ, venography of chronic cerebrospinal venous insuffi-
Theobald MR, Heiserman JE (2000) Cerebral MR ciency in patients with associated multiple sclerosis.
venography: normal anatomy and potential diagnostic Pol J Radiol 76(1):59–62
pitfalls. AJNR Am J Neuroradiol 21(1):74–78 104. Niggemann P, Seifert M, Forg A, Schild HH,
92. Feng W, Utriainen D, Trifan G, Sethi S, Hubbard D, Urbach H, Krings T (2012) Positional venous MR
Haacke EM (2012) Quantitative flow measurements angiography: an operator-independent tool to evalu-
in the internal jugular veins of multiple sclerosis ate cerebral venous outflow hemodynamics. AJNR
patients using magnetic resonance imaging. Rev Am J Neuroradiol 33(2):246–251
Recent Clin Trials 7(2):117–126 105. Siddiqui AH, Zivadinov R, Benedict RH et al (2014)
93. Alperin N, Hushek SG, Lee SH, Sivaramakrishnan A, Prospective randomized trial of venous angioplasty in
Lichtor T (2005) MRI study of cerebral blood flow MS (PREMiSe). Neurology Jul 29;83(5):441–9.
and CSF flow dynamics in an upright posture: the doi:10.1212/WNL.0000000000000638
effect of posture on the intracranial compliance and 106. Higashi Y, Yoshizumi M (2003) New methods
pressure. Acta Neurochir Suppl 95:177–181 to evaluate endothelial function: method for assessing
94. Stoquart-Elsankari S, Lehmann P, Villette A endothelial function in humans using a strain-gauge
et al (2009) A phase-contrast MRI study of physio- plethysmography: nitric oxide-dependent and -inde-
logic cerebral venous flow. J Cereb Blood Flow pendent vasodilation. J Pharmacol Sci 93(4):399–404
Metab 29(6):1208–1215 107. Janssen MC, Wollersheim H, Haenen JH, van Asten
95. Sundstrom P, Wahlin A, Ambarki K, Birgander R, WN, Thien T (1998) Deep venous thrombosis: a
Eklund A, Malm J (2010) Venous and cerebrospinal prospective 3-month follow-up using duplex scan-
fluid flow in multiple sclerosis: a case–control study. ning and strain-gauge plethysmography. Clin Sci
Ann Neurol 68(2):255–259 94(6):651–656
96. Macgowan CK, Chan KY, Laughlin S, Marrie RA, 108. Locker T, Goodacre S, Sampson F, Webster A, Sutton
Banwell B (2014) Cerebral arterial and venous AJ (2006) Meta-analysis of plethysmography and
blood flow in adolescent multiple sclerosis patients rheography in the diagnosis of deep vein thrombosis.
and age-matched controls using phase contrast MRI. Emerg Med J 23(8):630–635
40 The Role of Diagnostic Imaging Techniques for Detection of Extracranial Venous System. . . 979

109. Zamboni P, Menegatti E, Conforti P, Shepherd S, 120. Mintz GS, Popma JJ, Pichard AD et al (1996) Intra-
Tessari M, Beggs C (2012) Assessment of cerebral vascular ultrasound predictors of restenosis after per-
venous return by a novel plethysmography method. cutaneous transcatheter coronary revascularization.
J Vasc Surg 56(3):677–685, e1 J Am Coll Cardiol 27(7):1678–1687
110. Beggs C, Shepherd S, Zamboni P (2014) Cerebral 121. Miskolczi L, Guterman LR, Flaherty JD, Hopkins LN
venous outflow resistance and interpretation of cervi- (1996) Depiction of carotid plaque ulceration and
cal plethysmography data with respect to the diagno- other plaque-related disorders by intravascular sonog-
sis of chronic cerebrospinal venous insufficiency. raphy: a flow chamber study. AJNR Am J Neuroradiol
Phlebology 29(3):191–199 17(10):1881–1890
111. Viola S, Viola P, Fiorelli L, Buongarzone M, Litterio P 122. Potkin BN, Bartorelli AL, Gessert JM et al
(2013) Transcranial brain photoplethysmography to (1990) Coronary artery imaging with intravascular
study the venules of cerebral cortex in patients with high-frequency ultrasound. Circulation 81(5):
multiple sclerosis. Phlebology. Dec 13. [Epub ahead 1575–1585
of print] 123. Reid DB (1996) Intravenous IVUS: room with a view.
112. Ludyga T, Kazibudzki M, Simka M et al (2010) J Endovasc Ther 3(4):435
Endovascular treatment for chronic cerebrospinal 124. Reid DB, Diethrich EB, Marx P, Wrasper R (1996)
venous insufficiency: is the procedure safe? Phlebol- Intravascular ultrasound assessment in carotid inter-
ogy 25(6):286–295 ventions. J Endovasc Ther 3(2):203–210
113. Simka M (2011) Commentary: Safety of endovascular 125. Reid DB, Douglas M, Diethrich EB (1995) The clin-
treatment for CCSVI and future perspectives. ical value of three-dimensional intravascular ultra-
J Endovasc Ther 18(3):326–327 sound imaging. J Endovasc Ther 2(4):356–364
114. Werner JD, Siskin GP, Mandato K, Englander M, Herr A 126. Scalise F, Farina M, Manfredi M, Auguadro C,
(2011) Review of venous anatomy for venographic Novelli E (2013) Assessment of jugular endovascular
interpretation in chronic cerebrospinal venous insuffi- malformations in chronic cerebrospinal venous insuf-
ciency. J Vasc Interv Radiol 22(12):1681–1690; quiz 91 ficiency: colour-doppler scanning and catheter venog-
115. Zivadinov R, Weinstock-Guttman B (2012) Role of raphy compared with intravascular ultrasound.
venoplasty for treatment of multiple sclerosis: value Phlebology 28(8):409–417
of open-label studies and surrogate treatment out- 127. Sclafani SJ (2012) Intravascular ultrasound in the
comes. J Vasc Interv Radiol 23(10):1308–1310 diagnosis and treatment of chronic cerebrospinal
116. Karmon Y, Zivadinov R, Weinstock-Guttman B venous insufficiency. Tech Vasc Interv Radiol 15(2):
et al (2013) Comparison of intravascular ultrasound 131–143
with conventional venography for detection of extra- 128. Baracchini C, Perini P, Calabrese M, Causin F,
cranial venous abnormalities indicative of chronic Rinaldi F, Gallo P (2011) No evidence of chronic
cerebrospinal venous insufficiency. J Vasc Interv cerebrospinal venous insufficiency at multiple sclero-
Radiol 24(10):1487–1498, e1 sis onset. Ann Neurol 69(1):90–99
117. Veroux P, Giaquinta A, Perricone D et al (2013) Inter- 129. Baracchini C, Perini P, Causin F, Calabrese M,
nal jugular veins out flow in patients with multiple Rinaldi F, Gallo P (2011) Progressive multiple scle-
sclerosis:a catheter venography study. J Vasc Interv rosis is not associated with chronic cerebrospinal
Radiol 24(12):1790–1797 venous insufficiency. Neurology 77(9):844–850
118. Simka M, Latacz P, Ludyga T et al (2011) Prevalence 130. Simka M, Ludyga T, Latacz P, Kazibudzki M (2013)
of extracranial venous abnormalities: results from a Diagnostic accuracy of current sonographic criteria
sample of 586 multiple sclerosis patients. Funct for the detection of outflow abnormalities in the inter-
Neurol 26(4):197–203 nal jugular veins. Phlebology 28(6):285–292
119. Nissen SE, Yock P (2001) Intravascular ultrasound:
novel pathophysiological insights and current clinical
applications. Circulation 103(4):604–616
Part VIII
Pediatrics
Pediatric Vascular Malformations
41
Sachin K. Pandey and Darren B. Orbach

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 984 The appropriate classification and understand-
ing of the pathophysiology of vascular
Imaging Techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 984
malformations, both within and outside the
Extracranial Vascular Malformations . . . . . . . . . . . 985 central nervous system, is of the utmost impor-
Venous Malformations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 985 tance in the diagnosis and treatment of these
Capillary Malformations . . . . . . . . . . . . . . . . . . . . . . . . . . . 988
Lymphatic Malformations . . . . . . . . . . . . . . . . . . . . . . . . . . 988 entities. This is critical, both in determining the
Sinus Pericranii . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 994 appropriate types of treatment for given lesions
CNS Vascular Malformations . . . . . . . . . . . . . . . . . . . . 994
and in determining which lesions may require
Cavernous Malformation . . . . . . . . . . . . . . . . . . . . . . . . . . . 994 no treatment at all. Similarly, understanding of
Capillary Telangiectasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . 996 the pathology of a given entity informs the
Arteriovenous Malformations . . . . . . . . . . . . . . . . . . . . . . 997 goals of therapy. As highlighted in this chapter,
Vein of Galen Aneurysmal
Malformation (VGAM) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 999
many of these malformations may have pro-
Spinal Vascular Malformations . . . . . . . . . . . . . . . . . . . . . 1003 tean clinical and radiologic presentations. As
such, a multidisciplinary approach to the diag-
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1005
nosis and management of vascular
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1005 malformations is important. Multidisciplinary
medicine is at its core, reliant on precise and
clear communication, and thus precise utiliza-
tion of terminology of greater importance. The
multidisciplinary approach to management, as
well as the rarity of many of these
malformations and operator-dependent risk of
therapy, makes most vascular malformations
ideal candidates for therapy at specialized,
large-volume institutions.
S.K. Pandey (*)
Neurointerventional Radiology, Brigham and Women’s Keywords
Hospital, Boston, MA, USA
Venous malformations • Sclerotherapy • Cap-
e-mail: skpandey@partners.org
illary malformations • Lymphatic
D.B. Orbach
malformations • AVMs • Embolization •
Neurointerventional Radiology, Boston Children’s
Hospital, Harvard Medical School, Boston, MA, USA Sinus pericranii • Cavernous malformations •
e-mail: darren.orbach@childrens.harvard.edu Vein of Galen aneurysmal malformations
# Springer Science+Business Media New York 2016 983
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_31
984 S.K. Pandey and D.B. Orbach

Introduction Table 1 ISSVA classification scheme


Vascular malformations Vascular tumors
The classification of vascular anomalies has tradi- Low-flow malformations Benign
tionally been a source of confusion, in part, due to Lymphatic malformations Infantile
such factors as the rarity of many of these anom- hemangioma
alies and overlapping imaging and clinical mani- Venous malformations Congenital
hemangioma
festations. Additionally, vascular anomalies are
Capillary malformations Tufted angiomas
diagnosed and treated from a multispecialty per-
Spindle-cell
spective, and as such, each group of physicians hemangioma
involved may have its own inherited terminology. Epithelioid
As an example, a recent study noted that 71.3 % of hemangioma
publications from 2009 used the term hemangi- Pyogenic granuloma
oma from that year did so erroneously. High-flow malformations Locally aggressive/
The Mulliken-Glowacki classification scheme borderline
was based on the biological behavior and endo- Arteriovenous Kaposiform
malformations hemangioendothelioma
thelial characteristics of these anomalies [1] and
Arteriovenous fistulae Retiform
was the basis from which the current International hemangioendothelioma
Society for the Study of Vascular Anomalies Papillary
(ISSVA) classification system grew (Table 1). intralymphatic
While a detailed discussion is beyond the scope angioendothelioma
of this chapter, the scheme separates vascular Composite
hemangioendothelioma
anomalies into neoplasms and malformations
Kaposi sarcoma
and with the former category includes infantile
Complex malformations Malignant
and congenital hemangiomas, Kaposiform Two or more vascular Angiosarcoma
hemangioendothelioma, and others. Vascular malformations within the Epithelioid
malformations are distinguished from vascular same lesion hemangioendothelioma
neoplasms by the absence of increased endothelial
cellular mitosis. As such, vascular malformations
tend to grow in proportion to overall childhood extra-CNS vascular malformations. But it is
growth, with some increased growth during apparent from any overview of pediatric vascular
puberty and pregnancy, or as a result of inflam- malformations that the bridges between the two
mation, infection, or hemorrhage. These areas are real and of critical importance, at the
malformations are now separated into three cate- levels of anatomy, genetics, and treatment.
gories: low-flow vascular malformations, high-
flow vascular malformations, and combined or
complex vascular malformations. Imaging Techniques
Although historically, clinicians and investiga-
tors who focused on vascular malformations For extracranial vascular malformations, the diag-
involving the central nervous system (CNS) nosis is often highly suggested by an informed
have worked in “splendid isolation” from those physical exam (Is the lesion warm and pulsatile?
involved with extra-CNS lesions, it has become Is there local cutaneous ischemia, ulceration, or
increasingly evident that complete separation of hemorrhage? Is the lesion purplish and expansile
these areas is artificial and counterproductive. As with recumbency or Valsalva?), and imaging gen-
there are diagnostic, natural history, and manage- erally plays a complementary role. Conversely,
ment issues unique to CNS lesions, this chapter for intracranial malformations, imaging is often
has sections dedicated separately to CNS and the alpha and the omega of diagnosis.
41 Pediatric Vascular Malformations 985

The imaging tools generally available for the Extracranial Vascular Malformations
assessment of these entities are ultrasonography
(US), magnetic resonance imaging (MRI), mag- Venous Malformations
netic resonance angiography and venography
(MRA/MRV), computed tomography (CT), CT Venous malformations occur with an overall inci-
angiography (CTA), and digital subtraction angi- dence of approximately 1 per 10,000. They are
ography (DSA) [2]. generally soft, compressible lesions that are dark
For extracranial malformations, US is frequently in color. Recent years have seen identification of
the first-line imaging choice, due to lack of ionizing genetic loci responsible for both inherited venous
radiation, wide availability, ability to obtain real- malformations (via germ line mutation) [4] and
time flow dynamic information, and lack of need for sporadic VM (via somatic mutation) [5]. Regard-
sedation. Drawbacks to this modality include less of anatomic location, venous malformations
operatory dependence and difficulty in imaging can cause discomfort, often related to swelling or
intracranial intraosseous lesional extension. How- intralesional thrombosis. The classic presentation
ever, in addition to its intrinsic diagnostic utility, US on clinical exam is that of a fluctuant swelling
is extremely useful for image-guided therapy [3]. with dependent positioning or during crying or
CT and MRI both provide excellent evaluation Valsalva maneuver. Depending on the location
of the extent of malformations and their relation- of the lesion, other symptoms, such as difficulty
ship to adjacent anatomic structures. Benefits of feeding, exophthalmos, or visual blurring, may
CT include rapid time of acquisition, which can also occur.
limit the need for sedation. However, much of the Various schemata have been proposed for the
patient population with vascular malformations is classification of venous malformations: types
pediatric, and avoidance of unnecessary ionizing I–IV, based on the relationship of the lesion to
radiation is vital. As such, every effort should be surrounding venous drainage and the morphology
made to utilize acceptable imaging alternatives, of that drainage, and truncular versus extra-
and if CT is necessary, scans should be limited to truncular types, based on the embryological
the area of interest only. On similar grounds, stage at which the lesion was presumed to have
multiphasic scanning should be kept to a mini- developed [6]. Although most venous
mum. MRI and MRA provide excellent, poten- malformations are sporadic, inherited multiple
tially time-resolved, tissue and spatial resolution lesions can occur in the context of syndromes
of vascular malformations, without the need for such as blue rubber bleb nevus syndrome, mani-
ionizing radiation. The most significant drawback fest as numerous head, neck, and extremity
of MR, particularly in the pediatric setting, is the venous malformations with the possibility of
need for sedation in the majority of patients. new lesions potentially developing
Because of its temporal resolution, unmatched throughout life.
spatial resolution, and vessel selectivity, DSA has On imaging, venous malformations are com-
a critical role in evaluating high-flow vascular pressible lesions of heterogeneous echotexture on
malformations, that is, those with an arterial com- sonography (Fig. 1). Color Doppler interrogation
ponent. DSA allows for precise mapping of often reveals slow monophasic flow, if any. If
involved anatomy and is an intrinsic part of high-velocity or biphasic flow is encountered, an
catheter-based endovascular therapy. DSA rarely alternative diagnosis of high-flow vascular mal-
has a role in the workup or management of vas- formation should be entertained. CT can delineate
cular malformations that do not harbor an arterial the extent of involvement of the surrounding tis-
component and is rarely the first-line diagnostic sues and will also highlight phleboliths [3]. MRI
modality for any lesion, due to its invasive nature also offers excellent delineation of the extent of
and patients’ exposure to ionizing radiation. involvement of these malformations. Though VM
986 S.K. Pandey and D.B. Orbach

Fig. 1 A 24-year-old
female visible left cheek
“birthmark,” progressively
expanding since age 8, with
recent skin discoloration.
Enlargement with Valsalva
maneuvers was noted on
examination. Pre- (a) and
post- (b) contrast T1W
images demonstrate
moderate, heterogeneous
enhancement and T2W (c)
image demonstrates a
hyperintense, multiseptated
abnormality. Grayscale
ultrasound (d) demonstrates
a hypoechoic
heterogeneous lesion.
Clinical and imaging
characteristics were
consistent with a venous
malformation

may have varied appearance on conventional generally painful and, as such, is most often
T1-WI and T2-WI images due to the presence or performed under general anesthesia. Though easily
absence of associated thrombus, they are gener- manageable in most locations, post-sclerosis edema
ally hyperintense on T2-WI imaging and interme- must always be considered in approaching these
diate on T1-WI imaging. VM may demonstrate lesions. For example, orbital venous malformations
variable contrast enhancement (although nearly leave little room for post-therapeutic swelling, and
all do clearly enhance on delayed imaging) and a sclerotherapy in this location should be undertaken
lobulated contour. Contrast enhancement is with the ophthalmology team available for urgent
important in differentiating VM from decompressive lateral canthotomy, if necessary;
non-enhancing vascular malformations, such as other options include surgical resection with or
macrocystic lymphatic malformations. Internal without associated endovascular liquid embolic
signal voids on MRI may represent phleboliths occlusion. If there is no vision compromise, obser-
within the VM. Catheter-based angiography is vation of these lesions is a reasonable option as
not helpful in the workup of VM and has almost well. Similarly, venous malformations of the head
no diagnostic role. In cases where angiography and neck associated with the airway, particularly
has been done, delayed passage of contrast those with baseline airway compromise and the
through abnormal veins and sinusoidal spaces need for repetitive treatments, may necessitate tra-
has been described, though angiographic appear- cheostomy in conjunction with therapy. As men-
ance is more commonly normal [7]. tioned below, bleomycin is associated with less
Therapy for venous malformations had tradi- post-procedural swelling than other sclerosants
tionally been surgical [8], though sclerotherapy is and can be an excellent choice in locations such as
the current treatment of choice. Sclerotherapy is the orbit and airway.
41 Pediatric Vascular Malformations 987

Fig. 2 Lateral projection


from a direct percutaneous
angiogram of the left cheek
venous malformation (a)
demonstrates a large,
disorganized cluster of
venous pouches, ultimately
communicating with the
normal venous drainage
system. Frontal spot
radiograph (b) during
sclerosis with 3 % sodium
tetradecyl sulfate (in a 2:1:1
mixture with lipiodol and
air) demonstrates sclerosant
within the malformation

Sclerotherapy is generally performed under dose kept below 1 mL/kg (maximum of 60 mL)
sonographic and/or fluoroscopic guidance and given gradually, in small aliquots. As head
(Fig. 2) [9]. Results are best with localized venous and neck lesions are rarely voluminous enough to
malformations, and for very diffuse lesions, ther- require very large doses of sclerosant
apeutic goals generally focus around the most (as compared with the sometimes massive anom-
symptomatic portions of the malformation. One alies seen in truncal or limb lesions), the cardiac
important consideration is the identification of morbidity and pulmonary hypertension are rarely
rapid initial venous drainage. In those cases, man- seen in this setting. In addition, sclerosants includ-
ual compression of these draining veins for the ing alcohol cause hemolysis, which at a sizable
initial 2–3 min of sclerosis can be helpful. In some scale may lead to hemoglobinuria. This is typi-
cases, coil or liquid embolic occlusion of the cally managed by generous hydration, urinary
outlet from the malformation to the adjacent alkalinization, and close monitoring of urinary
draining veins can compartmentalize the lesion, output [9], so other than for treatment of the
ensuring that a large amount of sclerosant does not smallest lesions, placement of a Foley catheter
escape into the venous system. An additional for these procedures is of great utility.
technique for such situations is the double needle Given the potential soft tissue and nerve-
technique [10], whereby the malformation is related morbidity of ethanol sclerosis, many oper-
accessed with two needles, using one for ators prefer other sclerosants, such as detergent
sclerosant injection and the other as a agents. The most commonly used such agent is
low-pressure escape valve, to reduce the risk of sodium tetradecyl sulfate (STS), with others in
nontarget sclerosis. this class including polydocanol, sodium
The choice of sclerosing agent is dependent morrhuate, and ethanolamine. These are typically
both on availability in a given country and on mixed with contrast agents such as Ethiodol for
operator preference. Ethanol is the most potent procedural visibility. Foaming of the sclerosant
of the sclerosing agents but is likely associated plus Ethiodol plus air is reported to increase effi-
with the most serious potential morbidity; extrav- cacy, possibly due to increased surface area of the
asation can result in severe damage or necrosis of microbubbles generated for endothelial contact,
adjacent soft tissue and nerve injury. Doses as compared with a pure liquid [12].
exceeding 0.75 mL/kg invariably result in intoxi- Bleomycin, an antibiotic and antitumor agent
cation [11]. Though there is a risk of cardiovascu- originally derived from Streptomyces verticillus in
lar collapse with ethanol, this is rare and can be 1966, is also used as a sclerosing agent for venous
avoided with careful, limited use, with the total malformations. Given that bleomycin induces
988 S.K. Pandey and D.B. Orbach

minimal posttreatment edema in comparison with ipsilateral to the capillary malformation, the skin
other sclerosing agents, it can be of particular lesion itself generally conforms to a distribution
utility for lesions involving the airway. While tracking the trigeminal nerve subsegments. Up to
bleomycin is associated with possible pulmonary 25 % of all patients with a V1 distribution capil-
fibrosis, this morbidity is seen at much larger lary malformation have additional signs of Sturge-
systemic doses than are used with percutaneous Weber syndrome [13], while the association is
sclerosis, with a threshold of >400 units. By weaker with V2 and V3 distribution lesions. Neu-
comparison, the doses used for typical pediatric rological involvement in Sturge-Weber syndrome
interventions are 0.5–1 mg/kg, and cases of typically results in seizures, hemiplegia, and
bleomycin-associated pulmonary fibrosis have developmental delay.
not been reported in the context of vascular anom- The imaging evaluation of capillary
aly sclerotherapy. Despite this, many groups malformations is primarily focused on assessment
obtain baseline pulmonary function tests and for associated pathology in underlying tissue.
chest radiographs in patients undergoing Doppler ultrasound is of particular utility in
bleomycin sclerotherapy and then continue to confirming or excluding associated arteriovenous
monitor pulmonary function after treatment. shunting. Treatment of capillary malformations,
Although not helpful as a primary treatment aimed at improving cosmesis, is generally by way
agent, as mentioned above, coils can be a useful of pulsed-dye laser. Although only 15–20 % of
adjunct, in particular, for malformations with lesions clear completely. For capillary
rapid communication to adjacent veins. Liquid malformations with underlying AVM or other
embolic agents, such as acrylate glue (nBCA) or vascular anomalies, laser therapy will not have
ethylene vinyl copolymer (Onyx), although real utility, and repeated attempts at treatment
designed for the treatment of high-flow vascular most often lead to skin injury with little benefit.
malformations, can also be used to isolate venous
malformations from adjacent veins into which
they drain, allowing for safe and effective Lymphatic Malformations
sclerotherapy.
Lymphatic malformations affect between 0.02 %
and 0.05 % of the population, with half identified
Capillary Malformations at birth and nearly all by 2 years of age [14]; they
occur with equal incidence among females and
Capillary malformations are vascular lesions males. The pathogenesis of these malformations
involving the superficial dermal layers and occur is likely related to a defect occurring during
in 3 of every 1,000 newborns, darkening with age. embryonic lymphangiogenesis, whereby buds of
Histopathologically, capillary malformations rep- lymphatic tissue become sequestered during
resent ectatic blood vessels in relatively dener- development. Lymphatic malformations are most
vated dermis. The most important feature of often cutaneous in location, though 10 % may
these lesions, aside from cosmesis, is their associ- involve the viscera. However, they are particu-
ation with vascular syndromes such as Sturge- larly trans-spatial in nature, frequently involving
Weber syndrome, Klippel-Trenaunay syndrome, all types of soft tissue and bone within a region.
and Parkes Weber syndrome. In addition, it is Lymphatic endothelium may be confirmed patho-
commonly the case that capillary malformations logically with staining for the D2-40 antibody.
involve the skin over an underlying fast-flow Lymphatic malformations (LMs) can be clas-
lesion, such as AVM, and at times, may involve sified as microcystic, macrocystic, or mixed.
the skin over lymphatic or venous malformations Many authors suggest that cysts <2 cm be classi-
as well. fied as microcysts and those >2 cm as macrocysts,
In the case of Sturge-Weber, which involves while others advocate for classification based on
ocular and leptomeningeal abnormalities typical appearance: those cysts appearing
41 Pediatric Vascular Malformations 989

Fig. 3 A 3-year-old female with exodeviation and ptosis, heterogeneous left orbital malformation. Power Doppler
leading to discovery of an orbital macrocystic lymphatic ultrasound image (c) confirms a multiseptated cystic
malformation. Axial pre-contrast T1W image (a) and cor- lesion, with no evidence of internal hypervascularity on
onal T2W image (b) demonstrate a multiseptated, power Doppler interrogation

sonographically anechoic, with clearly delineated Imaging of lymphatic malformations is often


spaces and thin echogenic walls, are categorized focused on delineation of infiltrative components
as macrocystic, while those appearing as a near- or to help refine differential possibilities when
solid echogenic mass are classified as microcystic with dealing indeterminate cystic lesions. The
[9]. As the classification system has most rele- specific appearance is dependent upon the
vance in guiding treatment, this latter schema macrocystic, microcystic, or mixed nature of the
has utility in its favor. lesion. Macrocystic lesions have a multilocular
Macrocystic lymphatic malformations are most cystic sonographic appearance without evidence
commonly found in the axilla, chest, and neck and of internal flow (Fig. 3). Layering debris from
can frequently be identified on prenatal ultrasound. prior hemorrhage or infection may also be seen.
In that setting, important syndromic associations to Conversely, microcystic lymphatic malformations
consider include Down, Turner, or Noonan syn- typically appear as an echogenic mass due to the
drome. On examination, macrocystic density of acoustic interfaces within the malfor-
malformations are typically soft and compressible, mation. By CT, macrocystic LMs typically appear
although striking changes in size and firmness can as hypodense, septated masses [3]. MR imaging
occur with hemorrhage or infection. Mixed generally reveals hypointense T1-WI signal and
microcystic and macrocystic lymphatic hyperintense T2-WI signal with or without fluid-
malformations are common in the head and neck fluid levels. Post-contrast imaging may demon-
and can result in facial deformities; when involving strate mural or septal enhancement, but no
the tongue or pharynx, airway compromise may enhancement is seen within cystic spaces of the
occur, particularly in the setting of hemorrhage or malformation.
infection. Syndromic associations have also Therapy of LM is guided by the size, area of
recently been described with the CLAPO syn- involvement, and symptoms related to the malfor-
drome, with the initial description consisting of six mation. Treatment modalities for macrocystic
patients with lower lip capillary malformation, face malformations include sclerotherapy, in some
and neck lymphatic malformation, facial and limb cases laser therapy, and surgical excision
asymmetry, and partial or generalized overgrowth. (Fig. 4). Unfortunately, recurrence is not rare
Interestingly, periorbital lymphatic malformations after resection, and scarring and damage to adja-
have been associated with intracranial vascular cent nerves or blood vessels may occur. Sclero-
anomalies, including developmental venous anom- therapy of LM typically involves cannulation of
alies, cavernous malformations, and others [15]. each cyst, in which fluid is then aspirated and a
990 S.K. Pandey and D.B. Orbach

Fig. 4 A 3-year-old female


with left orbital macrocystic
lymphatic malformation.
Grayscale sonographic
image (a) demonstrates
percutaneous needle access
of the malformation, with
confirmation of
intralesional position noted
on spot lateral radiography
(b). Axial T2W images
prior to (c) and following
(d) sclerotherapy with
doxycycline 10 mg/mL
diluted in iodinated contrast
demonstrate interval
decrease in size of the
malformation (white
arrows)

similar volume of sclerosing agent is injected. OK-432 demonstrated that ~94 % of patients
Some lesions may be so large as to necessitate with macrocystic lymphatic malformations had
placement of a pigtail catheter which can remain regression or resolution of their malformations
in place for several days, facilitating repeat aspi- [16]. Alcoholic solution of zein is a combination
ration and injection. of corn protein (zein), oleum in ethanol, and
Sclerosing agents used for lymphatic sodium diatrizoate, also not available in the
malformations include ethanol, doxycycline, United States but used in Canada and Europe,
sodium tetradecyl sulfate, bleomycin, alcoholic with outcomes comparable to other agents [17].
solution of zein (Ethibloc), and OK-432 In the natural history of LM, superinfection is
(Picibanil) [9]. Doxycycline is the most com- frequent. As such, many practitioners give pro-
monly used of these agents, usually at a concen- phylactic antimicrobial therapy during and fol-
tration of 10 mg/mL, due to its low morbidity and lowing sclerotherapy, with some extending this
high efficacy; it is, however, painful on injection, for 7–10 days following treatment. Involution of
and requires general anesthesia, even in adults. In cysts is typically assessed at 4–6 weeks post-
neonates, doses greater than 150 mg in a single procedure, with staged treatments generally
treatment setting are avoided, due to risks of met- occurring at 6–8-week intervals.
abolic acidosis, hypoglycemia, and hemolytic Microcystic malformations are technically
anemia. OK-432 (Picibanil), derived from a more difficult to treat, tend to have poorer
low-virulence strain of Streptococcus pyogenes, response to therapy, and tend to recur after treat-
is not currently approved for use in the United ment more frequently than macrocystic LM
States. A large prospective randomized trial of [18]. Direct intralesional injection with
41 Pediatric Vascular Malformations 991

Table 2 Syndromic associations with high-flow vascular [22]. PTEN-associated arteriovenous lesions
malformations tend to respond poorly, if at all, to embolization.
PHACES In the head and neck, AVMs most frequently
Lumbar manifest with symptoms of hemorrhage, pain, or
Parkes Weber deformity. Particularly because total cure is so
CM-AVM elusive and can only be entertained for the
Cobb smallest of lesions, where complete resection
Osler-Weber-Rendu might be possible, therapeutic goals are typically
Wyburn-Mason
focused on symptomatic control, on preservation
PTEN hamartoma tumor syndrome
of critical functions such as vision, and on
cosmesis. There is no universal treatment algo-
rithm, with different teams not infrequently opting
pingyangmycin (a bleomycin variant), as well as for different embolic agents, frequency of treat-
radiofrequency ablation [19], has been described ment, modality of treatment, etc. In those few
as showing efficacy. cases where complete cure is the goal of therapy,
the best results seem to occur with preoperative
Extracranial AVM embolization followed by surgical resection.
In comparison to low-flow vascular The choice of embolic agent is dependent on
malformations, extracranial high-flow vascular the overall context of therapy for a given patient.
malformations are rare, with a slightly higher Specifically, preoperative embolization can be
prevalence in females. As is true for most vascular effectively performed with short-term embolic
malformations, AVMs generally grow in propor- agents such as PVA particles, Gelfoam, or
tion to the child, but they have been shown to Embospheres. While these provide excellent
grow rapidly during puberty, pregnancy, infec- devascularization for the purposes of reducing
tion, and trauma [20]. Some extracranial AVMs intraoperative blood loss, these are not durable
have been associated with mutations of the embolic agents, and thus, if the goal of treatment
RASA1 gene [21]. RASA1 encodes for a p120 is permanent (i.e., anything other than preopera-
Ras GTPase-activating protein which itself is tive embolization), liquid embolic agents, such as
involved in angiogenesis and cell adhesion, and ethylene vinyl alcohol copolymer (Onyx), etha-
children harboring mutations present with a com- nol, and n-butyl cyanoacrylate (nBCA) glue, are
bination of skin capillary stains and a propensity preferred. nBCA is a clear, liquid, monomeric
to develop AVMs throughout the body substance which is frequently mixed with tanta-
(CM-AVM). The PTEN gene on chromosome lum powder and Ethiodol for radiopacity. Expo-
10q has also been associated with AVMs. This sure to anions in blood causes the glue to
gene encodes a tumor suppressor protein involved polymerize and form a cast that is adherent to
in regulating angiogenesis, cell growth, and cell the endothelium, with the rate of polymerization
proliferation, and mutations are seen in multiple of the glue controllable by the glue-to-Ethiodol
eponymous syndromes, such as Bannayan-Riley- ratio. In addition to the acute inflammatory fibro-
Ruvalcaba and Cowden syndrome (Table 2). The sis caused by the glue, there is a delayed foreign-
preferred designation for syndromes resulting body giant cell granulomatous reaction
from such PTEN mutations is PTEN hamartoma [23]. Onyx is a liquid embolic agent that precipi-
tumor syndrome (PHTS). Interestingly, greater tates at its surface in contact with blood, while
than half of these patients have high-flow vascular maintaining a liquid core. Thus embolization
anomalies of the head and neck. However, these technique consists of the formation of a proximal
are unlike typical AVMs, in that they often have plug around the microcatheter tip, with further
ectopic fat within or adjacent to the malformation, infusion allowing for controlled delivery of the
disproportionate dilation of draining veins, and liquid embolic, with longer and more extensive
multiple non-contiguous sites of involvement embolization than is possible with nBCA. Onyx
992 S.K. Pandey and D.B. Orbach

Fig. 5 A 21-year-old male


with pulsatile tinnitus and
progressive enlargement of
a soft left auricular “mass.”
Axial fat-saturated proton
density (a) and T1W post-
contrast (b) MR images
demonstrate asymmetric
enlargement of the left
external ear with multiple
small flow voids. AP (c) and
lateral (d) projections from
a left external carotid
angiogram demonstrate
abnormal arteriovenous
shunting, consistent with a
left auricular arteriovenous
malformation

additionally causes less endothelial damage and to establish the diagnosis. Dedicated diagnostic
incites less inflammatory response than nBCA, catheter angiography is the routine standard of
resulting in improved handling of embolized care for intracranial lesions, where multiphasic
malformations during surgical resection. Some evaluation of flow dynamics to clearly identify
advocate ethanol, which results in endothelial arteriovenous shunting, precise delineation of
denudation, as the embolic treatment of choice arterial inflow, nidal vessels, and venous drainage
[24], though with potentially the greatest and superselective angiography for identification
morbidity risk. of normal parenchymal branches arising from
The sonographic appearance of arteriovenous feeding pedicles is critical [25]. In contradistinc-
malformations is characterized by multiple tion, for extracranial AVM, diagnostic angiogra-
hypoechoic vascular channels, without a well- phy can typically be performed as part of the first
defined soft tissue mass. Doppler analysis reveals session of embolization.
arterialized draining venous waveforms [3]. CT In terms of location within the head and neck,
angiographic imaging demonstrates increased cal- midface lesions, supplied by the internal maxil-
iber of feeding arteries and enlarged draining lary artery, are disproportionately represented.
veins. Similar findings are also noted on MRI, The cheek and ear are relatively common sites,
where the rapid flow may also manifest as abnor- with the latter generally confined to the outer ear
mal flow voids on spin-echo sequences. On all and possible extra-auricular spread; middle and
imaging modalities, the absence of an enhancing inner ear involvement virtually never occurs
soft tissue mass is critical in distinguishing these (Fig. 5) [26]. Because arterial supply to ear
lesions from others. DSA is virtually never needed AVMs is almost always from end arteries,
41 Pediatric Vascular Malformations 993

Fig. 6 A 16-year-old male with purple left perimandibular shunting with a primarily intraosseous left mandibular
discoloration and growth, with complaint of aching left arteriovenous malformation. Repeat lateral projection
mandibular pain with prolonged mastication. Coronal CT image from left external carotid angiography (c) and fron-
image (a) displayed in bone windows demonstrates abnor- tal spot radiograph (d) obtained following Onyx liquid
mal irregular lytic lesion in the left body of the mandible embolization demonstrate interval occlusion of the major-
with cortical disruption. Lateral projection from left exter- ity of the malformation
nal carotid angiography (b) demonstrates arteriovenous

complete endovascular embolization may posterior ethmoidal artery branches may also con-
unavoidably induce tissue necrosis and lead to tribute (Fig. 6). Given the intraosseous nature of
amputation. As such, ear AVMs are generally these malformations, surgical resection is chal-
observed until symptoms necessitate intervention. lenging and necessitates bony excision and recon-
Although amputation and plastic surgical recon- struction with graft. While some authors advocate
struction may be the likely end point, staged pal- for a combined embolization and surgical
liative embolizations can delay amputation for approach from the outset for these lesions, others
several years. have reported that most can be controlled with
Mandibular and maxillary arteriovenous embolization alone [27]. High-volume vascular
malformations pose unique risks for life- anomaly centers often use embolization as first-
threatening hemorrhage associated with dental line therapy for newly discovered maxillary or
eruptions or dental extractions. Arterial supply to mandibular AVM and to approach any loose
maxillary malformations is usually from internal tooth or required dental extraction with a further
maxillary branches, though ascending pharyngeal preparatory embolization, with extraction in the
branches, labial branches of the facial artery, and angiography suite.
994 S.K. Pandey and D.B. Orbach

Fig. 7 A 10-year-old boy with enlarging scalp mass, with subjacent superior sagittal sinus. Lateral projection from
expansion during Valsalva. Axial T2W MRI (a) and sagit- a right internal carotid artery angiogram in the venous
tal CTA (b) demonstrate a focal frontal calvarial defect phase (c) confirms sinus pericranii with demonstration of
with subcutaneous T2 hyperintense, contrast enhancing communication between the palpable malformation and
malformation appearing to communicate with the the underlying sinus

Sinus Pericranii Diagnosis of sinus pericranii can usually be


made on the basis of clinical exam. Confirmation
Sinus pericranii is a link between extracranial and of the presence of transosseous channels may be
intracranial vascular anomalies. Patients with this obtained with use of sonography. MR imaging,
condition have a transosseous venous bridge particularly sagittal and coronal high-resolution
allowing for direct flow between intracranial and contrast-enhanced sequences with fat suppres-
extracranial venous drainage pathways (Fig. 7). sion, is helpful in clearly localizing the bridging
The intracranial contributor flowing into the channels and also in providing a more global view
transosseous bridge may be a venous sinus (with of the overall intracranial venous drainage.
the superior sagittal sinus being by far the most Symptomatic cases of sinus pericranii can be
common) or a pial vein or developmental venous treated, either with sclerotherapy (taking care that
anomaly. Sinus pericranii is rare overall, most the sclerosant does not travel intracranially) or
commonly involves small venous channels, and resection. For cases where pial veins or a DVA
is infrequently symptomatic, with incidental dis- serves as the intracranial contributor to the bridg-
covery on imaging being the rule. When the con- ing vessel (rather than a venous sinus), catheter
dition does present clinically, the most common angiography is necessary in order to ensure that
manifestation is as a nonpulsatile mass that alternative drainage is available. Cases where the
expands with recumbency, crying, or Valsalva. sinus pericranii serves as an obligate venous
The extracranial, scalp portion of the sinus drainage pathway for a brain region cannot be
pericranii may have the mass-like, globular con- safely treated without incurring a high risk of
figuration of a venous malformation, rather than venous infarct.
the morphology of a tubular venous channel.
These cases are clinically apparent from birth
and are most commonly located frontally, running CNS Vascular Malformations
from the glabella up toward the hairline,
manifesting as a large, purplish, expansile lesion. Cavernous Malformation
Though in a minority of cases, sinus pericranii
may occur secondarily as a result of trauma, the Cavernous malformations (CMs) are well-
vast majority of cases are congenital, and they are circumscribed, sinusoidal endothelium-lined
frequently associated with intracranial develop- channels. As is true of other vascular
mental venous anomalies [28]. malformations, they are frequently referred to by
41 Pediatric Vascular Malformations 995

Fig. 8 A 14-year-old male with seizure disorder and left demonstrate a left parietal cavernous malformation (white
parietal spikes on EEG. Axial non-contrast CT scan of the arrows). Also noted is an associated development venous
head (a), axial T2W MRI (b), and axial minimum intensity anomaly (white arrowhead)
projection susceptibility-weighted image (c) all

other names such as cavernomas, cavernous hem- vein stenosis develop increased capillary bed
angiomas, and cavernous angiomas. The vascular pressure that in turn leads to microhemorrhage.
spaces in cavernous malformations are immature, This hemorrhage may induce recruitment of fibro-
lacking elastin and having minimal smooth mus- blasts which lay down fragile endothelialized ves-
cle [29], with a hallmark being absence of inter- sels. Although speculative and although clear
vening brain parenchyma. CMs are common, venous stenosis is not seen in most cases of
occurring in just under 1 % of the general popula- DVA with CM, support for this mechanism can
tion. The distribution of these malformations follows be derived from the observation of endothelial
the volume of the neuraxis, with 80–90 % being development at the site of chronic intracranial
supratentorial, 8–20 % being infratentorial, and hematomas [31].
approximately 3 % being spinal. The most common The natural history of CM has been controver-
clinical presentation of these malformations, in 36 % sial. Some of the earlier literature, written when
of cases, is seizures. Twenty-five percent of patients many believed that all cavernous malformations
present with hemorrhage, and the balance is discov- were congenital, considered the at-risk period to
ered incidentally on imaging. extend all the way from birth, thus mistakenly
While some CMs are congenital or present in lowering the calculated annual risk of hemor-
early childhood, de novo cavernous rhage. Further adding to confusion, some patients
malformations have been detected following radi- may experience clinical deterioration in the
ation therapy, prior brain biopsy, post-viral infec- absence of imaging-documented hemorrhage or
tions, and in certain familial cases. Cavernous other change. Meta-analysis data from prospec-
malformations frequently coincide with develop- tive databases suggests an overall rate of hemor-
mental venous anomalies (DVAs), with the latter rhage of 2.4 % per patient year. Interestingly, the
affecting approximately 4 % of the general popu- size of cavernous malformations appears to play
lation (Fig. 8). DVAs, as originally described by little or no role in risk of hemorrhage, while loca-
Lasjaunias and colleagues [30], represent variants tion has been shown to be more strongly corre-
of normal venous drainage, rather than true lated, with deep brain lesions and infratentorial
malformations. However, a causal relationship lesions having higher hemorrhagic risk
between DVAs and cavernous malformations has [29]. While some authors suggest that deeper elo-
been proposed, whereby DVAs with collector quent lesions are more likely to manifest with
996 S.K. Pandey and D.B. Orbach

clinical symptoms secondary to hemorrhage than prognosis, may merit more aggressive surgical
superficial non-eloquent lesions, the reasons for intervention. Conversely, resection of these deep
the positional dependence remain unclarified. brain malformations carries increased risk of surgi-
The imaging tool of choice for the diagnosis cal morbidity. This combination of considerations
and characterization of cavernous malformations has led to interest in stereotactic radiosurgery for
is MRI. On MRI, CMs appear as well-defined, deep brain cavernous malformations. However,
lobulated lesions with heterogeneous signal inten- several studies have suggested that the high inci-
sity on conventional T1-WI and T2-WI images, dence of hemorrhage- and radiation-related com-
often with a “popcorn”-like appearance. This het- plications, as much as sevenfold greater than with
erogeneity reflects mineralization, hemorrhage, arteriovenous malformation therapy, renders ste-
and fibrosis, all of which frequently coexist within reotactic radiosurgery inappropriate for all but the
a cavernous malformation. Gradient echo or most exceptional cases [33] of CM.
susceptibility-weighted images demonstrate
rim-like magnetic susceptibility, reflecting hemo-
siderin deposition in the surrounding brain paren- Capillary Telangiectasia
chyma. Zabramski and colleagues developed a
classification scheme for CM, based on the MRI Capillary telangiectasia represents a collection of
appearance. CT imaging may demonstrate focal abnormally dilated capillaries. Similar to cavern-
parenchymal hyperdensities, in keeping with min- ous malformations, they demonstrate paucity of
eralization or hemorrhage, though the appearance elastin and smooth muscle. Unlike cavernous
is less specific than that of MRI. Traditionally, malformations, however, these capillaries are
DSA has no role in the evaluation of cavernous interspersed with normal brain parenchyma.
malformations as they are angiographically Capillary telangiectasia occurs with an estimated
occult, but a recent report demonstrates visualiza- overall prevalence of 0.7 % and demonstrates
tion of a CM using cone-beam CT with intra- a predilection for pontine location, accounting
arterial angiographic injection, including delinea- for approximately 78 % of all cases [34]. Less
tion of an adjacent DVA, where none had been common locations include the basal ganglia
seen on MRI [32]. Despite the well-characterized and cerebral hemispheres. Clinically, the over-
appearance on MRI in most cases, a specific diag- whelming majority of these lesions are asymp-
nosis is sometimes not possible, particularly in the tomatic and are predominantly incidental
setting of acute hemorrhage, and in those situa- imaging findings.
tions, serial imaging to exclude other hemorrhagic The imaging modality of choice for character-
lesions is often helpful. Of particular note, early ization of capillary telangiectasia is MRI. These
postoperative imaging after resection of cavern- malformations most commonly demonstrate mild
ous malformations may be particularly challeng- enhancement on post-contrast MR imaging, with
ing, as the appearance can be very similar to the gradient echo and susceptibility-weighted MRI
preoperative imaging despite total surgical resec- demonstrating abnormal magnetic susceptibility
tion [29]. As such, postoperative imaging should (Fig. 9). Since telangiectasias are very rarely asso-
be interpreted with caution and delayed imaging ciated with hemorrhage, the magnetic susceptibil-
may also be helpful. ity is postulated to represent sluggish,
Surgical resection has been the mainstay of deoxygenated blood within the malformation
treatment for cavernous malformations. Given the rather than hemorrhagic blood products. The asso-
more benign course of superficial lesions, they are ciation of these malformations with developmen-
frequently observed clinically and radiographi- tal venous anomalies is weaker than that observed
cally. However, if a particular lesion can be with cavernous malformations, with only 11 %
shown to be an epileptogenic source, resection of demonstrating an associated DVA [35]. Similar
superficial lesions may be warranted. Deep brain to cavernous malformations, capillary telangiec-
cavernous malformations, owing to their worse tasia is angiographically occult.
41 Pediatric Vascular Malformations 997

Fig. 9 A 17-year-old male


with incidental finding of
pontine capillary
telangiectasia (white
arrows) on MRI done for
headaches. Axial T1W
images pre- (a) and post-
(b) contrast demonstrate
faint “brush-like”
enhancement. The
malformation is isointense
to pontine parenchyma on
axial T2W imaging (c) and
demonstrates classic
susceptibility artifact on
susceptibility-weighted
imaging (d)

These malformations generally require no may rupture. Additionally, the feeding arteries,
interventional therapy, reflecting their benign nat- subject to fast flow into a low-pressure outlet,
ural history, and the primary importance of imag- may develop flow-related aneurysms, also subject
ing for telangiectasia is specific diagnosis, so as to to hemorrhage (Fig. 11). Additionally, the sink
avoid unnecessary surgical intervention. Given effect of the malformation may cause a steal phe-
the incidental nature of their discovery in most nomenon resulting in ischemia to surrounding
cases and the benign natural history, routine brain parenchyma.
follow-up is generally not indicated [34]. Specific features of brain AVMs associated
with hemorrhagic presentation have been studied
in the pediatric population and include smaller
Arteriovenous Malformations nidal size, exclusively deep venous drainage,
and infratentorial location [36]. Although tradi-
As their name denotes, arteriovenous tional estimates place the annual risk of hemor-
malformations (AVMs) are characterized by direct rhage at between 2 % and 4 % per year,
connections between arteries and veins, historically, the cases that have come to clinical
circumventing the capillary bed (Fig. 10). attention are those that presented with hemor-
AVMs, in the great majority of cases, are thought rhage, likely artificially skewing the annualized
to be congenital, related to errors in vascular mor- risk upwards. In an attempt to delineate the natural
phogenesis. The abnormal arteriovenous shunting history, the recent ARUBA study was conducted,
that is the hallmark of these malformations leads where patients with unruptured brain AVMs were
to the subsequent pathology: the draining veins, randomized to treatment versus observation.
being subjected to abnormally pressurized flow, Although the study was halted because of an
998 S.K. Pandey and D.B. Orbach

Fig. 10 A 21-year-old female with headaches. AP (a) and malformation. The nidus (black arrows) receives left supe-
lateral (b) projections from a selective left vertebral angio- rior cerebellar and anterior-inferior cerebellar arterial sup-
gram, as well as 3-D volume-rendered reconstruction (c) of ply. There are two feeding artery aneurysms (arrowheads)
the same, demonstrate a left cerebellar arteriovenous associated with the superior cerebellar arterial pedicle

Fig. 11 Lateral projection


from a post-embolization
selective left vertebral
artery angiogram (a) and
spot lateral projection skull
radiograph post-
embolization (b). Onyx-18
liquid embolic material is
visualized (black arrow)
within the superior
cerebellar arterial pedicle,
with occlusion of the two
feeding artery aneurysms
seen preoperatively

improved outcome in the observational arm [37], arterial supply, while also blocking access routes
many observers criticized the study design. for additional endovascular therapy. Similarly,
Unlike aneurysmal subarachnoid hemorrhage, subtotal surgical resection also frequently leads
intracranial hemorrhage from arteriovenous to progression. Surgical risk has been linked to
malformations is less severe, though still with a the Spetzler-Martin grading system for adults
10 % risk of death. (Table 3). Though this has some relevance for
Treatment of brain AVMs is focused on oblit- the pediatric population, some factors within the
eration of the nidus. This may accomplished with system, such as determining cerebral eloquence
radiotherapy, surgically or endovascularly. and the focality of venous drainage within a
Regardless of whether the approach is surgical restricted brain volume, likely differ in children
or endovascular, general principles of arteriove- and adults [38].
nous malformation management include avoid- Stereotactic radiotherapy is also a commonly
ance of proximal feeding artery occlusion, as used therapeutic modality for brain AVMs, partic-
doing so allows the nidus to continue to recruit ularly for cases measuring less than 4 cm in size.
41 Pediatric Vascular Malformations 999

Table 3 Spetzler-Martin classification scheme for intra- [39]) includes Wyburn-Mason syndrome, or
cranial arteriovenous malformations Bonnet-Dechaume-Blanc syndrome, in which
Size <3 cm – 1 point orbital, periorbital, and retinal extracranial
3 cm–6 cm – 2 points AVMs track backwards along the optic pathways,
>6 cm – 3 points following the optic nerve to the chiasm, and back
Venous drainage Superficial – 0 points to the lateral geniculate region of the
Deep – 1 point thalamus [40].
Eloquence Non-eloquent – 0 points Hereditary syndromic associations with AVMs
Eloquent – 1 point
have also been described. Genetic defects in
Superficial venous drainage includes cortical veins and endoglin and ALK-1 binding proteins for
convexity sinuses. Eloquent locations include sensorimo-
tor cortex, language areas, visual cortex, hypothalamus, transforming growth factor-β result in endothelial
internal capsule, brainstem, cerebellar peduncles, or deep dysfunction and have been associated with AVMs
cerebellar nuclei in Osler-Weber-Rendu syndrome (or hereditary
hemorrhagic telangiectasia) (Fig. 12). In addition
Table 4 Modified Pollock-Flickinger score for predicting to the abovementioned association of mutations in
arteriovenous malformation obliteration with radiosurgery the RASA1 gene with extracranial CM-AVM,
Modified Pollock-Flickinger score RASA1 mutations have also been associated
AVM score = 0.1 (volume, mL) + 0.02 (age, year) + 0.5 with brain arteriovenous malformations and
(location) fistulae [21].
Location score = 0 for hemispheres/corpus callosum/cere-
bellum. Location score = 1 for basal ganglia/thalamus/
brainstem
Vein of Galen Aneurysmal
Malformation (VGAM)

The Spetzler-Martin scale is of less utility in VGAM represents specific choroidal types of
predicting outcomes for radiotherapy than it is intracranial arteriovenous lesions, manifest in
for surgical prognostication. As such, the young infants. Specifically, they are supplied by
Pollock-Flickinger score was derived and has choroidal arteries (anterior and posterior), with
been well correlated to outcomes in both adults venous drainage to the median prosencephalic
and children following radiotherapy (Table 4). vein of Markowski, an embryonic precursor to
Drawbacks to radiosurgery include the delayed the vein of Galen [41], with no development of a
nature of nidal occlusion, with a median time to true deep venous system in most cases (Fig. 13)
obliteration of approximately 3 years. Other con- [42]. Developmentally, the median prosence-
cerns include the risk of radiation necrosis involv- phalic vein is the primary venous drainage from
ing adjacent brain parenchyma, often presenting 6 to 11 weeks of gestational age, after which the
with significant delay, and increased risk of tumor median prosencephalic vein is partially assimi-
induction from the stochastic effects of radiation lated into the vein of Galen. However, when the
exposure, particularly in children. connections between choroidal arteries and the
Syndromic associations with AVMs are myr- median prosencephalic vein fail to regress by
iad, though rare. Nonhereditary segmental abnor- approximately 8 weeks gestational age, progres-
malities that involve AVMs are thought to relate to sive enlargement of the vein may occur, leading to
the fact that neural crest and adjacent mesoderm VGAM. Several similar appearing entities are
originate from a shared metamere. Thus, in situa- often confused with VGAM, including tectal
tions of developmental disruption prior to daugh- AVMs which drain into the vein of Galen, and
ter cell migration, malformations may be vein of Galen varices. As the treatment approach
segmental in distribution. This group of for these lesions is quite different than for VGAM,
cerebrofacial arteriovenous metameric syndromes a precise delineation of a given lesion’s anatomy
(CAMS, as named by Lasjaunias and colleagues is critical.
1000 S.K. Pandey and D.B. Orbach

Fig. 12 A 24-year-old
female with seizures,
epistaxis, and remote
history of cerebral abscess.
AP projection (a) from a
right vertebral artery
angiogram in mid-arterial
phase demonstrates three
distinct arteriovenous
malformations (black
arrowheads) with
associated early venous
drainage. Lateral projection
(b) from a right internal
carotid artery angiogram
demonstrates an additional
right parietal arteriovenous
malformation (black
arrowhead). Oblique
sagittal MIP (c) and 3-D
volume-rendered
reconstruction (d) from a
subsequent CT angiogram
of the chest help to confirm
the diagnosis of Osler-
Weber-Rendu syndrome
with identification of
pulmonary arteriovenous
malformation (white
arrowhead)

Fig. 13 A 6-month-old male with abnormal prenatal vein of Galen aneurysmal malformation. There is notable
imaging. Sagittal post-contrast T1W MR image (a), axial dilation of the median prosencephalic vein of Markowski
T2W MR image (b), and lateral projection from a left (white arrows), without communication to the normal deep
internal carotid angiogram (c) all demonstrate abnormal venous drainage system
41 Pediatric Vascular Malformations 1001

Fig. 14 Choroidal (a) and mural (b) architectural sub- median prosencephalic vein of Markowski (black arrows).
types of vein of Galen aneurysmal malformations. The The mural type demonstrates more direct abnormal drain-
choroidal type demonstrates numerous choroidal arterial age of feeding arteries to the lateral wall of the median
feeding arteries converging on a complex fistulous site prosencephalic vein of Markowski, without intervening
(black arrowhead), which in turn drains to the dilated complex fistulous bed

Nidal architecture of a VGAM can generally be symptoms is generally related to the angioarch-
divided into two primary subtypes: mural and itecture of the VGAM. Cardiac manifestations
choroidal (Fig. 14). The mural configuration, gen- stem from abnormally elevated return to the
erally better tolerated clinically, consists of direct right side of the heart, leading to right atrioven-
arteriovenous fistulas in the wall of the median tricular dilation and pulmonary hypertension
prosencephalic vein. Choroidal types generally [44]. High-output left-sided heart failure is com-
have more severe neonatal symptoms and involve monly present as well. Cardiac symptoms can
an interposed nidal network between the choroi- range from the very severe, including those
dal arteries and the large venous pouch. The pres- detected sonographically in utero and those
ence of a high-flow VGAM may interfere with requiring intervention in the first few days of
normal maturation of skull base venous sinuses life, to the milder, allowing for observation until
and the jugular bulbs, resulting in the persistence 4–5 months of age. Neurological symptoms are
of fetal occipital or marginal sinuses and incom- generally the result of venous congestion and
plete maturation of the jugular bulbs, and venous hydrocephalus. As focal neurological symptoms
outlet stenosis may portend extremely poor prog- are difficult to detect in newborns and young
nosis. In contrast, venous drainage may lead to the infants, macrocrania may be an early presenting
cavernous sinuses and thereafter to the facial and symptom. Other neurological manifestations may
scalp veins, often predictive of good prognosis include neurocognitive delay, which, when
(Fig. 15). In cases where outlet obstruction leads severe, may be clinically irreversible despite
to venous congestion, MRI may demonstrate embolization.
focal white matter lesions or diffuse destruction Endovascular embolization is the mainstay of
of the brain (the so-called melting brain) [42]. VGAM therapy (Fig. 16). The goals of VGAM
Clinical presentation of VGAM generally falls therapy are primarily to reduce or reverse cardiac
into two categories: those related to high-output symptoms and to prevent neurological manifesta-
cardiac failure and neurological symptoms related tions. As such, complete angiographic oblitera-
to venous congestion and hydrocephalus (Table 5) tion of the VGAM is not generally the end point
[43]. As noted previously, the severity of these of therapy as this may involve unnecessary
1002 S.K. Pandey and D.B. Orbach

Fig. 15 Lateral projection from an early (a) and late (b) venous drainage and extensive drainage to the cavernous
venous phase of a left internal carotid angiogram in a child sinus anteriorly. The later phase demonstrates the thalamic
with a vein of Galen aneurysmal malformation. The earlier (black arrowhead) and lateral perimesencephalic (white
phase demonstrates a “pseudophlebitic” appearance to the arrowhead) veins forming the common “epsilon” pattern
congested cortical veins. Note the absence of typical deep of venous drainage

Table 5 Bicetre neonatal evaluation score for vein of Galen aneurysmal malformations
Cardiac
Score Cerebral function function Respiratory function Hepatic function Renal function
0 Permanent Resistance to Assisted ventilation, with Abnormal Anuria
neurological medical therapy desaturation coagulation,
deficit transaminitis
1 Seizures Ventilation Assisted ventilation, with Moderate or Unstable
necessary normal saturation at FiO2 transient hepatic diuresis with
>25 % insufficiency medical therapy
2 Isolated Failure, Assisted ventilation, with Hepatomegaly Transient anuria
convulsion unstable with normal saturation at FiO2
medical therapy <25 %
3 Nonconvulsive, Failure, stable Tachypnea, unable to Normal Normal
episodic with medical finish a bottle
neurological signs therapy
4 Subclinical EEG Overload, with Tachypnea, able to finish N/A N/A
abnormalities no medical a bottle
therapy
5 Normal Normal Normal N/A N/A
Maximal score = 21

neurological risk. The trans-arterial route is pre- experienced centers [45], neonatal patients with
ferred by most practitioners to transvenous embo- VGAM have the highest rates of mortality and
lization, as it is associated with lower morbidity. neurological morbidity. If early heart failure is
Early heart failure may compel embolization not present, treatment is typically deferred to ~5
within the first few days of life, with reduction of months of age and is often staged over several
the shunt by approximately 30 %, generally months or longer. Noncardiac clinical scenarios
allowing for clinical abatement of cardiopulmo- necessitating early embolization include worsen-
nary symptoms for the first few months of life. ing ventriculomegaly or any neurodevelopmental
Despite impressive results in this population at concerns.
41 Pediatric Vascular Malformations 1003

Fig. 16 Axial T2W MR


images from a young child
with vein of Galen
aneurysmal malformation at
6 months (a) and 15 months
(b) of age. The dilation of
the median prosencephalic
vein of Markowski (white
arrow), the extent of
hydrocephalus, and the
resultant thinning of the
parieto-occipital lobes have
all markedly improved
following interval trans-
arterial Onyx and coil
embolization

Spinal Vascular Malformations anterior and posterior spinal arterial branches


(Fig. 17). Venous egress is usually diffuse, involv-
In an effort to understand and approach spinal ing both dorsal and ventral surfaces of the spinal
vascular malformations, numerous classification cord and with drainage extending cranially and
schemes have been developed, particularly for the caudally. As with cranial AVMs, features of
high-flow malformations. Early schemes classi- angioarchitectural weakness, such as feeding
fied high-flow spinal vascular malformations artery aneurysms, may occur. Therapy, always
into four subtypes. Type I lesions are spinal high risk for intramedullary lesions, is generally
dural arteriovenous fistulae; type II lesions are focused on complete surgical excision, facilitated
intramedullary “glomus” AVMs; type III lesions in select cases by preoperative embolization.
are metameric AVMs, sometimes involving the Intramedullary lesions may also extend into
entire spinal canal and paraspinal structures at a extramedullary tissues, corresponding to the type
given level; and type IV lesions are perimedullary III or metameric subtype of spinal vascular mal-
arteriovenous fistulae. While this scheme has formation (also known as Cobb syndrome). These
gained widespread use, alternatives and revisions malformations may involve multiple spinal levels
have been proposed, such as a scheme that focuses and extend axially to involve paraspinal tissues.
on the distinction between arteriovenous fistula This can also result in secondary osseous spinal
and malformation and then further classifying by changes, such as expansion of the spinal canal, as
axial location into intramedullary, well as laminar and pedicular erosion [47]. Type
extramedullary-intradural, extradural, or trans- III malformations most commonly manifest in
spatial [46]. children as progressive myelopathy and pain.
Intramedullary spinal AVMs may be either Given the infiltrative nature of these
compact or diffuse. The former, referred to as malformations, treatment can be particularly dif-
glomus AVMs, represent approximately 90 % of ficult and challenging and often focuses on
the intramedullary cases. Overall, spinal AVMs debulking for reduction of mass effect on the cord.
occur with approximately one-tenth the frequency Intramedullary lesions usually present with
of their intracranial counterparts. They most com- back or radicular pain, myelopathy, bowel or blad-
mon occur at the cervicomedullary junction and der dysfunction, and sensorimotor deterioration.
are frequently diagnosed in childhood or young Symptom onset may be sudden, in cases related to
adulthood (in contradistinction to spinal dural lesional hemorrhage. Unlike spinal dural arterio-
arteriovenous fistulae, which are much more com- venous fistulae, this hemorrhage may be
monly diagnosed later in life). Arterial supply to intramedullary as well as in the subarachnoid
intramedullary lesions is typically from multiple space. This latter point is of particular importance
1004 S.K. Pandey and D.B. Orbach

Fig. 17 A 16-year-old
male with worsening
bilateral lower extremity
weakness. Sagittal T2W
MR image (a) demonstrates
expansion of the
mid-thoracic spinal cord
with abnormal
intramedullary flow voids
and prominent
extramedullary flow voids.
AP projection from a left T7
artery angiogram (b)
demonstrates abnormal
arteriovenous shunting
related to an intramedullary
spinal arteriovenous
malformation

when considering that these malformations can Lesions in the posterior aspect of the cord are
commonly occur at the cervicomedullary junc- more amenable to safe resections, as are lesions
tion, and thus, in cases of intracranial subarach- in the filum terminale [47]. Embolization can have
noid hemorrhage, with no identifiable intracranial a role in select cases, either on its own or as a
etiology, a spinal AVM must always be preoperative measure. Often, the critical determi-
considered. nant of the feasibility of safe embolization is the
The mainstay of the initial imaging evaluation anatomic relationship of the malformation to the
of spinal malformations is MRI. T2-WI imaging anterior spinal arterial axis.
often demonstrates spinal cord signal High-flow spinal malformations without an
hyperintensity adjacent to the malformation, intervening nidus between arterial supply and
suggesting venous hypertension or developing venous egress are categorized as spinal arteriove-
myelomalacia. Flow-related signal voids that are nous fistulae. As with AVMs, these are categorized
part of the lesion itself and additionally dilated by anatomic location, namely, intradural or
flow voids along the surface of the cord, extradural. The latter is rare and usually involves
representing supply and draining vessels, are fre- radicular arterial branches communicating with the
quently present. However, digital subtraction epidural venous plexus [48], most commonly in the
angiography remains the gold standard in pre- cervical spine. If the pressurized epidural plexus
cisely characterizing spinal vascular does not transmit arterial pressure in retrograde
malformations. Additionally, angiography allows fashion to the perimedullary veins of the cord,
for characterization of normal vessels, such as the neurological symptoms as a result of venous con-
artery of Adamkiewicz, that must be preserved in gestion are unlikely. Rather, epidural fistulae usu-
the course of treatment (Fig. 18). ally become symptomatic as a result of local mass
It has long been known that the natural history effect from congested epidural venous plexus
of intramedullary spinal AVMs is poor, with more resulting in spinal canal stenosis [49], although
than one-third of young patients developing associated perimedullary venous communication
severe impairment after 3 years of evolution. and myelopathy has been reported [50].
41 Pediatric Vascular Malformations 1005

treatment of these entities. This is important both


in determining the appropriate types of treatment
for given lesions and in determining which lesions
may require no treatment at all. Similarly, under-
standing of the pathology of a given entity
informs the goals of therapy. As highlighted
above, many of these malformations may have
protean clinical and radiologic presentations. As
such, a multidisciplinary approach to the diagno-
sis and management of vascular malformations is
important. Multidisciplinary medicine is at its
core, reliant on precise and clear communication
and thus precise utilization of terminology of
greater importance. The multidisciplinary
approach to management, as well as the rarity of
many of these malformations and operator-
dependent risk of therapy, makes most vascular
malformations ideal candidates for therapy at spe-
cialized, large-volume institutions.
Fig. 18 AP image of a selective left T10 angiogram,
demonstrating supply to the artery radiculomedullaris
magna (artery of Adamkiewicz)
References
The more common type of arteriovenous fis-
tula is intradural, representing approximately 1. Mulliken JB, Glowacki J (1982) Hemangiomas and
vascular malformations in infants and children: a clas-
70 % of all spinal vascular malformations. These sification based on endothelial characteristics. Plast
develop in the lateral epidural space and the point Reconstr Surg 69(3):412–422
of fistulization in these cases is at the dural nerve 2. Kollipara R, Odhav A, Rentas KE, Rivard DC, Lowe
root sleeve, with communication between a radic- LH, Dinneen L (2013) Vascular anomalies in pediatric
patients: updated classification, imaging, and therapy.
ular artery and perimedullary veins. There is a Radiol Clin North Am 51(4):659–672
notable male predominance of these fistulae, as 3. Dubois J, Garel L (1999) Imaging and therapeutic
is the case for intracranial lateral epidural arterio- approach of hemangiomas and vascular malformations
venous fistulae. Symptoms related to these fistulae in the pediatric age group. Pediatr Radiol 29(12):
879–893
are generally related to venous hypertension and 4. Boon LM, Mulliken JB, Vikkula M, Watkins H,
vascular steal, with slow, progressive sensorimo- Seidman J, Olsen BR et al (1994) Assignment of a
tor myelopathy being the most common presenta- locus for dominantly inherited venous malformations
tion. This insidious nature may explain the to chromosome 9p. Hum Mol Genet 3(9):1583–1587
5. Uebelhoer M, Natynki M, Kangas J, Mendola A,
paucity of these entities in children, though a Nguyen HL, Soblet J et al (2013) Venous
likelier explanation is that these fistulae are malformation-causative TIE2 mutations mediate an
acquired later in life rather than congenital AKT-dependent decrease in PDGFB. Hum Mol Genet
malformation. 22(17):3438–3448
6. Lee BB, Baumgartner I, Berlien P, Bianchini G,
Burrows P, Gloviczki P et al (2014) Diagnosis and
treatment of venous malformations consensus docu-
Summary ment of the International Union of Phlebology (IUP):
updated 2013. Int Angiol: J Int Union Angiol
123:206–212
The appropriate classification and understanding 7. Konez O, Burrows PE (2002) Magnetic resonance of
of the pathophysiology of vascular malformations vascular anomalies. Magn Reson Imaging Clin N Am
is of the utmost importance in the diagnosis and 10(2):363–388, vii
1006 S.K. Pandey and D.B. Orbach

8. Kang GC, Song C (2008) Forty-one cervicofacial and other fast-flow vascular anomalies are caused by
vascular anomalies and their surgical RASA1 mutations. Hum Mutat 29(7):959–965
treatment–retrospection and review. Ann Acad Med 22. Tan WH, Baris HN, Burrows PE, Robson CD, Alomari
Singapore 37(3):165–179 AI, Mulliken JB et al (2007) The spectrum of vascular
9. Choi DJ, Alomari AI, Chaudry G, Orbach DB (2009) anomalies in patients with PTEN mutations: implica-
Neurointerventional management of low-flow vascular tions for diagnosis and management. J Med Genet
malformations of the head and neck. Neuroimaging 44(9):594–602
Clin N Am 19(2):199–218 23. Gemmete JJ, Pandey AS, Kasten SJ, Chaudhary N
10. Puig S, Casati B, Staudenherz A, Paya K (2005) Vas- (2013) Endovascular methods for the treatment of vas-
cular low-flow malformations in children: current con- cular anomalies. Neuroimaging Clin N Am 23(4):
cepts for classification, diagnosis and therapy. Eur J 703–728
Radiol 53(1):35–45 24. Lee BB, Baumgartner I, Berlien HP, Bianchini G,
11. Burrows PE, Mason KP (2004) Percutaneous treatment Burrows P, Do YS et al (2013) Consensus document
of low flow vascular malformations. J Vasc Interv of the International Union of Angiology (IUA)-2013.
Radiol 15(5):431–445 Current concept on the management of arterio-venous
12. Yamaki T, Nozaki M, Sakurai H, Takeuchi M, management. Int Angiol 32(1):9–36
Soejima K, Kono T (2008) Prospective randomized 25. Kalani MY, Albuquerque FC, Fiorella D, McDougall
efficacy of ultrasound-guided foam sclerotherapy com- CG (2013) Endovascular treatment of cerebral arterio-
pared with ultrasound-guided liquid sclerotherapy in venous malformations. Neuroimaging Clin N Am
the treatment of symptomatic venous malformations. 23(4):605–624
J Vasc Surg 47(3):578–584 26. Wu JK, Bisdorff A, Gelbert F, Enjolras O, Burrows PE,
13. Ch’ng S, Tan ST (2008) Facial port-wine stains – Mulliken JB (2005) Auricular arteriovenous malforma-
clinical stratification and risks of neuro-ocular involve- tion: evaluation, management, and outcome. Plast
ment. J Plast Reconstr Aesthet Surg 61(8):889–893 Reconstr Surg 115(4):985–995
14. Perkins JA, Manning SC, Tempero RM, Cunningham 27. Persky MS, Yoo HJ, Berenstein A (2003) Management
MJ, Edmonds JL Jr, Hoffer FA et al (2010) Lymphatic of vascular malformations of the mandible and maxilla.
malformations: review of current treatment. Laryngoscope 113(11):1885–1892
Otolaryngol Head Neck Surg 142(6):795–803, e1 28. San Millan Ruiz D, Gailloud P (2010) Cerebral devel-
15. Bisdorff A, Mulliken JB, Carrico J, Robertson RL, opmental venous anomalies. Childs Nerv Syst
Burrows PE (2007) Intracranial vascular anomalies in 26(10):1395–1406
patients with periorbital lymphatic and lymphati- 29. Rivera PP, Willinsky RA, Porter PJ (2003) Intracranial
covenous malformations. AJNR Am J Neuroradiol cavernous malformations. Neuroimaging Clin N Am
28(2):335–341 13(1):27–40
16. Smith MC, Zimmerman MB, Burke DK, Bauman NM, 30. Lasjaunias P, Burrows P, Planet C (1986) Developmen-
Sato Y, Smith RJ et al (2009) Efficacy and safety of tal venous anomalies (DVA): the so-called venous
OK-432 immunotherapy of lymphatic malformations. angioma. Neurosurg Rev 9(3):233–242
Laryngoscope 119(1):107–115 31. Maeder P, Gudinchet F, Meuli R, de Tribolet N (1998)
17. Dubois J, Garel L, Abela A, Laberge L, Yazbeck S Development of a cavernous malformation of the brain.
(1997) Lymphangiomas in children: percutaneous AJNR Am J Neuroradiol 19(6):1141–1143
sclerotherapy with an alcoholic solution of zein. Radi- 32. Radvany MG, Rigamonti D, Gailloud P (2014) Angio-
ology 204(3):651–654 graphic detection of cerebral cavernous malformations
18. Alomari AI, Karian VE, Lord DJ, Padua HM, Burrows with C-arm cone beam CT imaging in three patients.
PE (2006) Percutaneous sclerotherapy for lymphatic J Neurointerv Surg 6(2):e17
malformations: a retrospective analysis of patient- 33. Pollock BE, Garces YI, Stafford SL, Foote RL,
evaluated improvement. J Vasc Interv Radiol 17(10): Schomberg PJ, Link MJ (2000) Stereotactic radiosur-
1639–1648 gery for cavernous malformations. J Neurosurg
19. Kim SW, Kavanagh K, Orbach DB, Alomari AI, Mul- 93(6):987–991
liken JB, Rahbar R (2011) Long-term outcome of 34. Gross BA, Puri AS, Popp AJ, Du R (2013) Cerebral
radiofrequency ablation for intraoral microcystic lym- capillary telangiectasias: a meta-analysis and review of
phatic malformation. Arch Otolaryngol Head Neck the literature. Neurosurg Rev 36(2):187–193, discus-
Surg 137(12):1247–1250 sion 94
20. Liu AS, Mulliken JB, Zurakowski D, Fishman SJ, 35. Lee RR, Becher MW, Benson ML, Rigamonti D (1997)
Greene AK (2010) Extracranial arteriovenous Brain capillary telangiectasia: MR imaging appearance
malformations: natural progression and recurrence and clinicohistopathologic findings. Radiology
after treatment. Plast Reconstr Surg 125(4):1185–1194 205(3):797–805
21. Revencu N, Boon LM, Mulliken JB, Enjolras O, 36. Ellis MJ, Armstrong D, Vachhrajani S, Kulkarni AV,
Cordisco MR, Burrows PE et al (2008) Parkes Weber Dirks PB, Drake JM et al (2013) Angioarchitectural
syndrome, vein of Galen aneurysmal malformation, features associated with hemorrhagic presentation in
41 Pediatric Vascular Malformations 1007

pediatric cerebral arteriovenous malformations. analysis of the literature with proposal of a new classi-
J Neurointerv Surg 5(3):191–195 fication system. J Neurosurg Pediatr 12(3):293–306
37. Mohr JP, Parides MK, Stapf C, Moquete E, Moy CS, 44. Chevret L, Durand P, Alvarez H, Lambert V,
Overbey JR et al (2014) Medical management with or Caeymax L, Rodesch G et al (2002) Severe cardiac
without interventional therapy for unruptured brain failure in newborns with VGAM. Prognosis signifi-
arteriovenous malformations (ARUBA): a multicentre, cance of hemodynamic parameters in neonates
non-blinded, randomised trial. Lancet 383(9917): presenting with severe heart failure owing to vein of
614–621 Galen arteriovenous malformation. Intensive Care
38. Krings T, Geibprasert S, Terbrugge K (2010) Classifi- Med 28(8):1126–1130
cation and endovascular management of pediatric cere- 45. Berenstein A, Fifi JT, Niimi Y, Presti S, Ortiz R, Ghatan
bral vascular malformations. Neurosurg Clin N Am S et al (2012) Vein of Galen malformations in neonates:
21(3):463–482 new management paradigms for improving outcomes.
39. Bhattacharya JJ, Luo CB, Suh DC, Alvarez H, Neurosurgery 70(5):1207–1213, discussion 13–4
Rodesch G, Lasjaunias P (2001) Wyburn-Mason or 46. Spetzler RF, Detwiler PW, Riina HA, Porter RW
Bonnet-Dechaume-Blanc as Cerebrofacial Arteriove- (2002) Modified classification of spinal cord vascular
nous Metameric Syndromes (CAMS). A new concept lesions. J Neurosurg 96(2 Suppl):145–156
and a new classification. Interv Neuroradiol 7(1):5–17 47. Patsalides A, Knopman J, Santillan A, Tsiouris AJ,
40. Krings T, Geibprasert S, Luo CB, Bhattacharya JJ, Riina H, Gobin YP (2011) Endovascular treatment of
Alvarez H, Lasjaunias P (2007) Segmental spinal arteriovenous lesions: beyond the dural fistula.
neurovascular syndromes in children. Neuroimaging AJNR Am J Neuroradiol 32(5):798–808
Clin N Am 17(2):245–258 48. Kim LJ, Spetzler RF (2006) Classification and surgical
41. Raybaud CA, Strother CM, Hald JK (1989) Aneu- management of spinal arteriovenous lesions: arteriove-
rysms of the vein of Galen: embryonic considerations nous fistulae and arteriovenous malformations.
and anatomical features relating to the pathogenesis of Neurosurgery 59(5 Suppl 3):S195–S201, discussion
the malformation. Neuroradiology 31(2):109–128 S3–13
42. Lasjaunias P (2007) Vascular diseases in neonates, 49. Krings T, Geibprasert S (2009) Spinal dural arteriove-
infants and children: interventional neuroradiology nous fistulas. AJNR Am J Neuroradiol 30(4):639–648
management, 2nd edn 50. Silva N Jr, Januel AC, Tall P, Cognard C (2007) Spinal
43. Mortazavi MM, Griessenauer CJ, Foreman P, Bavarsad epidural arteriovenous fistulas associated with progres-
Shahripour R, Shoja MM, Rozzelle CJ et al (2013) sive myelopathy. Report of four cases. J Neurosurg
Vein of Galen aneurysmal malformations: critical Spine 6(6):552–558
Pediatric Stroke
42
Daniel Tibussek, Gabrielle deVeber, and Manohar Shroff

Contents Perinatal Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1030


Perinatal Arterial Ischemic Stroke . . . . . . . . . . . . . . . . . 1030
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1010 Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1030
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1010 Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1030
Clinical Features and Diagnostic Delays . . . . . . . . . . . 1011 Periventricular Venous Infarction . . . . . . . . . . . . . . . . . . 1031
Perinatal Hemorrhagic Stroke . . . . . . . . . . . . . . . . . . . . . . 1031
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012
Computed Tomography . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012 Pediatric Cerebral Sinovenous Thrombosis . . . . 1032
Magnetic Resonance Imaging . . . . . . . . . . . . . . . . . . . . . . 1013 Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1032
Conventional Angiography . . . . . . . . . . . . . . . . . . . . . . . . . 1015 Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1032
Patterns of Childhood Arterial Ischemic Stroke . . . 1015 Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1032
Imaging of CSVT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1032
Risk Factors of Arterial Ischemic Strokes . . . . . . 1019 Magnetic Resonance Imaging and Venography . . . 1033
Treatment of Arterial Ischemic Stroke . . . . . . . . . . . . . 1027 Computed Tomography . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1035
Follow-Up Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1035
Hemorrhagic Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1028
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1035
Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1029
Outcome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1035
Prognosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1029
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1037
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1037

D. Tibussek (*)
Children’s Stroke Program, Division of Neurology,
The Hospital for Sick Children, University of Toronto,
Toronto, ON, Canada
Department of General Pediatrics, Neonatalogy and
Pediatric Cardiology, University Children’s Hospital,
Heinrich-Heine University, D€usseldorf, Germany
e-mail: daniel.tibussek@sickkids.ca;
daniel.tibussek@gmx.net
G. deVeber
Children’s Stroke Program, Division of Neurology,
The Hospital for Sick Children, University of Toronto,
Toronto, ON, Canada
e-mail: gabrielledev@gmail.com;
gabrielle.deveber@sickkids.ca
M. Shroff
Department of Diagnostic Imaging, The Hospital for Sick
Children, University of Toronto, Toronto, ON, Canada
e-mail: manohar.shroff@sickkids.ca

# Springer Science+Business Media New York 2016 1009


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_32
1010 D. Tibussek et al.

Abstract
Introduction
Over the last two decades, childhood ischemic
Over the last two decades, childhood ischemic
stroke, including arterial ischemic stroke and
stroke, including arterial ischemic stroke and cere-
cerebral sinovenous thrombosis (CSVT), has
bral sinovenous thrombosis (CSVT), has been
been increasingly recognized. Despite the
increasingly recognized. Although childhood
widely held belief that the increased plasticity
arterial ischemic stroke (CAIS) is rare compared
of the young brain protects against the effects
with adults, the estimated incidence ranges from
of injury, early brain injury due to stroke com-
1.6 to 13 per 100,000 children [1] and appears to
monly results in significant long-term
be rising [2]. Children under 1 year of age are
impairment.
particularly at risk with a peak incidence in the
Considerable differences exist between
perinatal period [1].
stroke in childhood and adults. These include
Considerable differences exist between stroke
maturational changes of the vascular, coagula-
in childhood and adults. These include matura-
tion, and nervous systems in neonates, older
tional changes of the vascular, coagulation, and
infants, children, and adults. In addition, etiol-
nervous systems in neonates, older infants, chil-
ogy differs significantly from adulthood.
dren, and adults. In addition, etiology differs sig-
Presenting symptoms and clinical findings
nificantly from adulthood. Presenting symptoms
vary with the age of the child. Finally, imaging
and clinical findings vary with the age of the child
may be more challenging with high rates of
and are frequently nonspecific, particularly in
falsely negative CT scans and a large number
infants. Finally, imaging may be more challenging
of clinical stroke mimics to consider. Together
with high rates of falsely negative CT scans and a
with a general lack of awareness, these factors
large number of clinical stroke mimics to con-
frequently result in significant delays in recog-
sider. Together with a general lack of awareness,
nition and appropriate investigation and treat-
these factors frequently result in significant delays
ment. Due to different underlying mechanisms
in recognition and appropriate investigation and
and important developmental changes
treatment.
throughout childhood, neuroimaging and man-
Despite the widely held belief that the
agement decisions in pediatric stroke cannot be
increased plasticity of the young brain protects
simply extrapolated from adults.
against the effects of injury, early brain injury
MRI imaging plays a crucial role in pediat-
due to stroke commonly results in significant
ric stroke in order to detect and characterize an
long-term impairment. Recent studies suggest
ischemic lesion and exclude common stroke
that early stroke often leads to widespread motor,
mimics. It also provides valuable information
behavioral, and cognitive dysfunction [3, 4].
about the potential etiology and prognosis.
CAIS results in neurologic morbidity in over
This in turn guides further diagnostic steps
two-thirds of survivors with considerable socio-
and assists in treatment decisions.
economic burden. For children with stroke
In the near future, more sophisticated imag-
beyond the perinatal period, recurrence rate is up
ing techniques such as vessel wall imaging,
to 25 % and, without antithrombotic treatment,
DTI, and functional imaging techniques will
even higher [5].
likely emerge into the clinical arena and play
an important role in enhancing our understand-
ing of pediatric stroke.
Epidemiology
Keywords
Stroke • Children • Neonatal • Sinovenous Stroke in childhood has long been thought to be
thrombosis • Ischemia • Haemorrhagic • rare and relatively benign. However, in the past
Haemorrhagic • Arteriopathy • Imaging three decades, epidemiological studies have
42 Pediatric Stroke 1011

Fig. 1 Age distribution of 16


cases with childhood
arterial ischemic stroke (n = 14
96) (From Mallick et al. [1]
with permission) 12

Number of cases
10

0
<1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15
Age at stroke onset (completed years)

repeatedly demonstrated that, in childhood, stroke diagnosis to exclude. Although the majority of
is as common as brain tumors. A recent children with CAIS will not be eligible for tPA,
population-based study of arterial ischemic stroke an early diagnosis is still essential to guide imme-
in children (excluding neonates) found an inci- diate neuroprotective and urgent antithrombotic
dence rate of 1.6 per 100,000 per year [1]. A treatments. However, despite an increasing num-
significant effect of age on incidence was found, ber of international publications about CAIS and
with children under the age of 1 year being par- increased awareness of the entity over the last
ticularly at risk (4.14 per 100,000 per year). An decade, the diagnosis of stroke in children is still
incidence above average has also been reported in frequently delayed or missed. A diagnostic delay
adolescence (Fig. 1). of 24 h and more from symptom onset is not
Studies that included neonates have reported uncommon even when the child is brought to
higher incidence rates of 2–3 per 100,000 per medical attention within the first 6 h after symp-
year. Neonates accounted for 8–46 % of total tom onset [7]. Diagnostic delay is frequently max-
pediatric AIS cases across different studies, and imal in the period following hospital arrival.
perinatal stroke is as common as 1 in 2,500 live Besides a lack of awareness of CAIS among par-
births [6]. It is not completely understood why the ents and health professionals, a complex differen-
incidence rates of childhood arterial ischemic tial diagnosis with a wide spectrum of common
stroke vary with age, but different susceptibilities stroke mimics occurring in up to 30 % of apparent
related to different underlying risk factors with strokes contributes to the diagnostic confusion. In
age are likely relevant. Ethnicity background addition, in children with definite CAIS, a falsely
plays an important role in stroke risk with African negative CT scan, still often the first imaging
and Asian children having a significantly higher study performed, has been reported in up to
incidence of stroke than Caucasian children 84 % [7]. This has resulted in a move to urgent
[1]. This is partially due to sickle cell disease in MRI as initial imaging for CAIS.
African and moyamoya disease in Asian children. Acute hemiparesis is the most common
presenting focal deficit. However, a variety of
other focal neurological deficits have been
Clinical Features and Diagnostic Delays described in CAIS (Table 1). Unlike in adulthood,
seizures at stroke onset are frequent in children
Any acute onset of a focal neurological deficit in a [1]. Age-related differences in clinical presenta-
child is a medical emergency, with ischemic or tion have to be considered. Especially in younger
hemorrhagic stroke being the most urgent children, infants, and neonates where over
1012 D. Tibussek et al.

Table 1 Presenting signs and symptoms of arterial ische- Table 2 Advantages of MR imaging versus computed
mic stroke in different age groups (Adapted from Mallick tomography in childhood
et al. [1])
MR imaging Computed tomography
<1 1–5 6–10 11–15 Less widely available Widespread access
year (%) years (%) years (%) years (%)
Extended acquisition time; Very fast acquisition
Focal 75 89 70 91 sedation may be needed speed
features
No radiation Uses ionizing radiation
Hemiparesis 69 85 60 52
Diffusion-weighted Misses stroke diagnosis
Facial 25 47 40 39 imaging highly sensitive in the majority of
weakness and specific for early stroke children
Speech 13 32 40 48 Gradient-echo imaging Highly sensitive for
disturbances sensitive for detection of detection of acute
Diffuse 63 47 100 74 hemorrhage hemorrhage
features MR angiography (TOF) CT angiography requires
Decreased 60 36 50 39 does not require contrast intravenous contrast
conscious
TOF angiography is prone CT angiography provides
level
to flow artifacts more accurate anatomic
Headache 0 13 50 52 illustration of cerebral
Seizures 75 26 20 9 arterial system
Susceptibility-weighted Imaging of cerebral
images provide valuable venous system requires
information about cerebral contrast
venous system
three-quarters present with only seizures, symp- Mismatch studies (core Mismatch studies (core
tomatology can be very nonspecific and mislead- vs. penumbra) usually not vs. penumbra) usually
ing. CAIS may occur in the context of an acute available in pediatric not available in pediatric
centers centers
illness, such as meningitis or post cardiac surgery,
when clinical evaluation can be particularly
challenging.

posterior fossa. However, immediate access to an


Imaging MRI is not always feasible and additional sedation
or anesthesia may be required compared with CT
Urgent neuroimaging is indicated to establish the leading to delayed imaging. Therefore, CT still
diagnosis in every case of a child presenting with has a place in the acute diagnostic process. A
an acute neurological deficit that is not otherwise head CT scan can rule out intracranial hemorrhage
sufficiently explained. Neuroimaging will also help (ICH) and may identify subtle findings suggestive
to rule out complications, such as cerebral edema of stroke. As in adults the main CT findings are
or intracranial hemorrhages, and possibly establish areas of cortical–subcortical hypoattenuating
the etiology. Importantly, in young infants with an within a vascular territory, obscuration and loss
open fontanel, cranial ultrasound (CUS) is not reli- of gray matter–white matter differentiation in the
able and, due to wide operator variability and a basal ganglia, and demonstration of a
peripheral location of many infarcts, frequently “hyperdense artery sign” (Fig. 2). CT may also
missed the diagnosis of CAIS [8]. reveal hemorrhagic transformation or malignant
infarct edema. However, sensitivity to early ische-
mia is generally low [7] (Fig. 3). The presence of a
Computed Tomography persistent arterial occlusion on CTA can also sug-
gest the diagnosis.
MRI is superior to CT in detecting strokes in Modern CT techniques such as perfusion CT
children (Table 2). This is particularly true for are not readily available in pediatric radiology
early and small strokes and those located in the departments. Furthermore, its use in childhood is
42 Pediatric Stroke 1013

generally discouraged because of an excessive Magnetic Resonance Imaging


radiation dosage. Therefore, compared to adults,
the role of an acute head CT to diagnose CAIS in A high-priority MRI is the preferred imaging
the pediatric population is limited. technique for diagnosing acute stroke. Besides a
high sensitivity and specificity to identify early
strokes, identification of associated treatable con-
ditions and exclusion of stroke mimics (see
below) pose other important advantages. By com-
paring different MRI sequences, e.g., diffusion
weighted (DWI) and FLAIR, the age of ischemic
lesions from within 7 days to older can be esti-
mated. Other features including mass effect or
atrophy can also aid in estimating lesion age. In
older teenagers, if tPA is a therapeutic option
priority, 1 MRI is indicated (within 1 h) to confirm
CAIS versus other mimics. Otherwise an early
MRI as early as possible but within 4 h after
presentation is still recommended in order to ini-
tiate urgent antithrombotic and neuroprotective
therapy in those with definite CAIS. This is feasi-
ble with most children. A specific hospital stroke
protocol focusing on the most relevant findings of
interest is useful (Definite ischemia? Recent
vs. old lesion, bleeding? Arterial anatomy). A
stroke protocol MRI can usually be completed
Fig. 2 Hyperdense artery sign within 10 min (Fig. 4).

Fig. 3 CT versus MRI of the same patient, 4 and 6 h after presentation with acute hemiparesis, respectively. The CT had
been reported as normal
1014 D. Tibussek et al.

Fig. 4 Simplified stroke 1) Diffusion weighted imaging/Apparent diffusion coefficient


protocol. This approach
allows exclusion of a stroke 2) EPI gradient echo T2*
after a maximum of 10 min 3) Axial FLAIR (>2 yrs) or Axial T2 (<2 yrs)
on the MR table
4) Time of flight MRA of circle of Willis

Study is supervised by neuroradiologist online.

Additional sequences added


according to findings of 1) – 4)

- Fat saturated T1
- Vessel wall imaging
- MR venography

DWI is the gold standard for diagnosis of acute


CAIS and has been proven helpful identifying
ischemic lesions within hours in neonates and
within minutes in older children [9]. Acute ische-
mic lesions demonstrate restricted diffusion,
evidenced by hyperintensity on trace diffusion
images. A matching reduced signal on apparent
diffusion coefficient (ADC) map helps to discrim-
inate recent from old lesions with so-called T2
shine-through. T2-WI and fluid-attenuated
inverted recovery (FLAIR) images may show
acutely infarcted tissue within 12–24 h of injury,
and abnormalities will usually persist.
MRA of the intracranial and neck vessels help
to identify and characterize an underlying
arteriopathy. Vascular imaging is an essential Fig. 5 Twelve-year-old girl with recurrent episodes of
slurred speech and right arm weakness. MR angiography
part of cerebral imaging in all children with stroke
shows proximal M1 stenosis
(Fig. 5). Vascular imaging demonstrates an intra-
or extracranial cerebral arteriopathy in about fifty
percent of children presenting with an AIS [10]. flight but not routinely performed in most pediat-
However, technical limitations require consid- ric centers.
eration. Because MRA relies on flow signal, areas SWI has recently been proven to be a useful
of normal (areas of branching or tortuosity) or MR sequence for CAIS for detecting hemorrhagic
abnormal (areas of stenosis) turbulent flow can components within the region of infarction and
mimic occlusions especially in infancy when identifying and quantifying microbleeds. In adults
arteries are smaller. TOF-MRA has been reported these have been shown to predict the risk of hem-
to overestimate the severity and length of a steno- orrhagic transformation before initiating thrombo-
sis. In addition, MRA has its limitations in reso- lytic treatment. Preliminary data suggest that SWI
lution, interfering with its ability to identify may be helpful for visualization of the penumbra
medium- or small-sized vessel diseases [11]. Gad- in children [12]. Potential pitfalls in SWI interpre-
olinium-enhanced MRA is superior to time of tation have been nicely summarized recently [13].
42 Pediatric Stroke 1015

Conventional Angiography

Despite advances in noninvasive imaging tech-


niques, conventional angiography (CA) remains
the gold standard for detailed cerebral vascular
imaging. Cerebral CA is the most sensitive and
specific method of diagnosing most cerebral
arteriopathies, many of which may only be iden-
tifiable with CA. CA also provides a dynamic
view of the arterial perfusion including areas of
low flow or, for collateral vessels and widened
zones of perfusion into the ischemic territory,
possible artery-to-artery embolism sources and
distal narrowing or occlusion. CA may identify
characteristic features of inflammatory and
non-inflammatory vasculopathies such as dissec-
tion and is recommended when central nervous
system vasculitis is suspected, especially when
MR imaging is not conclusive. In addition, CA
Fig. 6 Three-year-old male patient with acute onset left
including both intracranial and extracranial arm and face weakness. Diffusion-weighted images
carotid arteries is crucial for presurgical planning revealed a right paramedian pontine ischemia
of revascularization, e.g., in cases of moyamoya
disease.
Therefore, CA should be considered in all chil- peripheral wedge-shaped infarct involving the
dren with AIS in whom an etiology for CAIS cerebral cortex and subjacent white matter in ter-
remains elusive after initial MRA or CTA imag- ritorial distributions.
ing. With pediatric expertise provided, the com- The stroke pattern may differ according to the
plication rate for diagnostic CA is low even in underlying etiology. As an example, large, iso-
children below 3 years of age [14]. However, lated lenticulostriate AIS is commonly associated
radiation remains a concern, and carefully with parainfectious or inflammatory intracranial,
weighing up the expected additional information unilateral arteriopathy including transient cerebral
and the risk of unnecessary exposure to radiation arteriopathy or post-varicella arteriopathy [16,
is warranted. 17]. Small lacunar infarcts are rare in childhood
age groups except in cardiac or sickle cell disease.

Patterns of Childhood Arterial Ischemic Posterior Circulation Strokes


Stroke The vertebrovascular or posterior circulation is
involved in up to 37 % of all CAIS cases [18],
Typical radiological features of AIS seen in chil- and boys are more commonly affected. Typical
dren are similar to those of young adults. How- posterior circulation stroke patterns include occlu-
ever, maturational changes that occur during brain sions of basilar artery and its branches (Fig. 6),
development need to be taken into account. Imag- proximal posterior cerebral artery (PCA) occlu-
ing patterns of CAIS will depend on the location sion with large-vessel territory infarction in the
and the size of the vessels involved. Most CAIS occipital and temporal lobes, and infarcts involv-
involve the territory of the middle cerebral artery ing the perforating artery territories in the thala-
(MCA), most frequently affecting the left hemi- mus and splenium. Multiple posterior circulation
sphere [15]. The classical imaging appearance of infarcts involving different territories are not
occlusion of a large cerebral artery is the uncommon (Fig. 7). Underlying arterial
1016 D. Tibussek et al.

Fig. 7 Five-year-old male with sudden onset headache and unsteady gate. Multifocal diffusion restriction in the posterior
circulation

dissection or arteriopathies are frequently identi- Table 3 Classification of border zone infarcts (From
fied [18]. Although basilar artery strokes can Mangla et al. [20] with permission)
potentially be life threatening, recent pediatric External (cortical) infarcts
data suggest a much better prognosis in children Frontal cortex (between the anterior and middle
compared with adults [18, 19]. Death rate for cerebral arteries)
childhood brain-stem infarction is below 10 %, Occipital cortex (between the middle and posterior
cerebral arteries)
and survivors frequently have good outcomes. In
Paramedian white matter (between the anterior and
contrast, recurrent ischemic events after posterior middle cerebral arteries)
circulation strokes have been described in up to Internal (subcortical) infarcts
50 % [18]. Between the lenticulostriate and middle cerebral
arteries
Watershed (Border Zone) Infarcts Between the lenticulostriate and anterior cerebral
In contrast to the majority of CAIS which is due to arteries
Between the Heubner and anterior cerebral arteries
thrombotic vascular occlusion of one artery,
Between the anterior choroidal and middle cerebral
apparently isolated focal ischemic lesions can arteries
also be caused by diffuse hypoperfusion. These Between the anterior choroidal and posterior cerebral
are usually located in typical watershed or border arteries
zones at the anterior, middle, and/or posterior
cerebral artery (PCA) junctions. External (corti-
cal) border zone infarcts can be differentiated hypoxic–ischemic events (Fig. 8). Differentiating
from those affecting internal border zones these from cortical embolic strokes is not always
(Table 3). In childhood, internal or “deep” border straightforward. Although watershed infarcts are
zone infarcts are frequently observed in children typically bilateral, they also can be asymmetric or
with congenital heart disease or after unilateral.
42 Pediatric Stroke 1017

Fig. 8 Neonate with congenital heart disease and neonatal seizures. Acute MRI revealed multifocal cortical diffusion
restrictions. A follow-up MRI 5 days later showed cortical laminar necrosis suggesting a hypoperfusion injury

Internal border zone infarcts, involving the transformation occurred in 19 (30 %), of
deep white matter, suggest occlusion at the distal whom only two were symptomatic. Increased
internal carotid or proximal anterior cerebral infarct volume was associated with a higher risk
artery (ACA) or MCA frequently occurring with of HT, whereas the risk was low with
chronic occlusive vasculopathies, including vasculopathies as the stroke mechanism. Use of
moyamoya (see below). anticoagulation and antiplatelet therapy is not a
significant risk factor for HT in children [21, 22].
Hemorrhagic Transformation When HT occurs, it is usually ECASS
Knowledge about hemorrhagic transformation Grade 1 (some of which may actually be laminar
(HT) in CAIS is limited. In a retrospective series necrosis) or Grade 2 and usually
of 61 children with AIS, hemorrhagic asymptomatic [22].
1018 D. Tibussek et al.

With any episode of neurological deterioration 1.3 % of more than 700 children treated for ische-
early after stroke, intracranial hemorrhage needs mic stroke [24]. However, in another smaller
to be considered and emergent neuroimaging is population-based study, 12.5 % (2 out of 16) of
indicated (Table 4). all pediatric ischemic strokes over two 1-year
periods developed malignant MCA
Malignant MCA Infarction infarctions [25].
Malignant MCA infarctions in the pediatric pop- Without decompressive surgery, death is fre-
ulation may not be as rare as previously thought. quent. Outcome data in children undergoing
In a recent multicenter hospital-based study, decompressive surgery however remains scarce,
malignant MCA infarctions were identified in and specific eligibility criteria for
hemicraniectomy in the pediatric population
Table 4 Temporal changes of hematoma on MR imaging have not yet been defined. However, available
(Adapted from Rasalkar and Chu [23] with permission) data suggest that pediatric survivors of malignant
Stage and Signal Signal MCA infarction and hemicraniectomy may have a
time from Hemoglobin intensity intensity more favorable functional outcome than those
hematoma stage on T1 on T2 without surgery [24]. Therefore, identification of
Hyperacute Oxy-Hb Gray White predictors of a malignant course of the MCA in
(<6 h)
the pediatric population would be of great
Acute Deoxy-Hb Gray Black
(6 h–3 importance.
days) In adults involvement of more than 50 % of the
Early Intracellular White Black MCA territory was found to be a reliable predic-
subacute meth-Hb tor. Preliminary pediatric data suggest that a high
(3–7 days)
pediatric ASPECT score (Fig. 9) and a high pedi-
Late Extracellular White White
subacute meth-Hb
atric NIHSS are predictive of malignant MCA.
(1–4 weeks) Large infarcts (>10 % of total brain volume)
Early Extracellular White White also predict poorer functional outcome in chil-
chronic (>4 meth-Hb with with dren. The modified pediatric ASPECT score cor-
weeks) hemosiderin black relates well with infarct volume (= percent of
rim rim
supratentorial brain volume) in children of differ-
Late Hemosiderin Black Black
chronic ent ages with excellent inter-rater reliability and
(months to validity. In addition, the modified ASPECTS
years) assigns points to small but important brain struc-
tures (e.g., PLIC), not just volumes, thereby

Fig. 9 Scoring areas for the modified pediatric C caudate, L lentiform, I insula, IC internal capsule,
ASPECTS. A1 proximal anterior cerebral artery (ACA) T thalamus, P2 superior portion of PCA territory, A2 distal
territory, P1 inferior portion of PCA territory, M MCA, ACA territory (From Beslow et al. [26] with permission)
42 Pediatric Stroke 1019

Table 5 Stroke mimics in childhood (Adapted from Table 6 Categorization of childhood arterial ischemic
Shellhaas et al. [28]) stroke risk factors (International Pediatric Stroke Study)
(Mackay et al. [10])
Migraine
Conversion disorders Arteriopathies
Seizures Cardiac disorders
Postictal paralysis Chronic systemic disorders
ADEM Prothrombotic states
Cerebellitis Acute systemic disorders
Tumor Chronic head and neck disorders
PRESa Acute head and neck disorders
Intoxication Risk factors for artheriosclerosis in adulthood
Intracranial abscess
AVM
Idiopathic intracranial hypertension
Periodic hypertensive episodes Risk Factors of Arterial Ischemic
Metabolic stroke Strokes
Subdural empyema
Moyamoya Risk factors associated with CAIS are diverse and
a
PRES posterior reversible encephalopathy syndrome differ substantially from adults in whom risk factors
act to promote atherosclerosis of the cerebral (or in
cardiogenic stroke, the coronary) arteries (Table 6).
incorporating location along with volume in the According to the data from the International Pedi-
score. Therefore, this easy to use score seems to be atric Stroke Study [10], preexisting risk factors were
a useful tool for estimating infarct volume [26]. present in almost 50 % of children presenting with
The permeability of the blood–brain barrier as acute ischemic stroke beyond the perinatal period.
assessed with MRI, PET, and SPECT also predicts These consisted mainly of congenital heart disease
a malignant course of MCA infarction. However, and other chronic childhood disorders. Following a
these techniques need further prospective evaluation. thorough etiologic evaluation, currently up to 90 %
of children will have at least one risk factor identi-
Stroke Mimics fied, with the majority having two or more identifi-
In the majority of children referred to tertiary able factors. Although causality is not always easy
centers with an acute focal neurological deficit, a to proof, the identification of an underlying etiology
stroke mimic will be diagnosed including is important as it may help to determine the risk of
migraine, seizures, Bell palsy, and conversion dis- stroke recurrence and lead to a more specific or even
orders [27]. Even when stroke is the suspected causal therapy [5]. Therefore, a systemic diagnostic
diagnosis prior to imaging, around 20 % will approach is always warranted. The results of neuro-
turn out to be other stroke mimics [28] (Table 5). imaging will guide further management.
It can be assumed that the number is even higher The most relevant risk factors of CAIS will be
in non-tertiary centers. Knowledge about these discussed in more detail:
conditions is therefore of great importance, and
establishing the diagnosis requires expertise in 1. Cerebral arteriopathies
both child neurology and pediatric neuroimaging. Up to 53 % of children with stroke who
Careful history taking and examination may help undergo vascular imaging have evidence of
to define a provisional diagnosis. However, MRI an underlying cerebral arteriopathy [29, 10].
will often be required to exclude a stroke with Since arteriopathies have been shown to be a
certainty and to establish the diagnosis of AIS in strong predictor of stroke recurrence, detailed
most cases [23]. Many of the conditions mimick- cerebral and cervical vascular imaging for
ing stroke require a specific treatment or further detection and classification of an arteriopathy
workup. is crucial in all children with CAIS [5, 16, 17].
1020 D. Tibussek et al.

Classification of childhood arteriopathies contrast MRI may indeed reveal gadolin-


includes six categories: ium wall enhancement supporting the
(A) Transient cerebral arteriopathy diagnosis of vasculitis. However, differen-
(B) Moyamoya tiation from normal periarterial enhance-
(C) Fibromuscular dysplasia ment is important. If associated with
(D) Dissection chicken pox in the preceding 12 months,
(E) Vasculitis the term “post-varicella angiopathy” is
(F) Congenital arteriopathies used [16]. There is now strong epidemio-
Among these, the four most commonly logical evidence to support a causal link
encountered will be described below, namely, between a cerebral arteriopathy and vari-
(A)–(D): cella zoster infection in the preceding year
(A) Transient cerebral arteriopathy [32] (Fig. 10).
Transient cerebral arteriopathy (TCA) is a Common imaging features of TCA are
nonprogressive, often reversible, unilat- [33]:
eral arteriopathy involving the anterior cir- 1. Unilateral arteriopathy of the large ves-
culation at the circle of Willis. It has been sels of the anterior circulation, typi-
recognized as a leading cause of CAIS cally affecting the distal ICA and
[29, 17]. Because the diagnosis of TCA proximal segments of the MCA
is confirmed by a reversible or stable and ACA.
course on repeated vascular imaging after 2. Focal or segmental stenosis or occlu-
6 months, the less specific term focal cere- sion of these vessels on conventional
bral arteriopathy (FCA) has been angiography. “Banding” or “striated”
suggested at baseline, when the further appearance is often described on initial
course is still unpredictable [30]. CA (Fig. 11). Features of other
However, recently in 79 children with arteriopathies, such as dissection or
initial imaging showing a unilateral moyamoya, are absent.
arteriopathy characteristic for TCA, 3. Dynamic nature during the first weeks
94 % evolved to definite TCA indicating and months with repeated imaging
that the diagnosis can be made with a often demonstrating worsening of arte-
high degree of confidence on baseline rial changes.
imaging [17]. In the latter study 6 % 4. A monophasic course over the long
evolved to other conditions, namely, term, with follow-up imaging after
moyamoya. However, in these children, 6 months confirming no further pro-
nearly all had visible moyamoya vessels gression, and then either partial or com-
at onset. More recently it was demon- plete resolution of the arteriopathy.
strated that intracranial dissection can In addition, infarcts are nearly always
also mimic TCA [31]. located in perforator territories within the
TCA has been presumed to be a result basal ganglia zone with cortical involve-
of inflammatory mechanisms. Therefore, ment limited to the overlying insular/tem-
it has been referred to as nonprogressive poral cortex. TCA classically affects
childhood primary angiitis of the central previously healthy school-aged children.
nervous system in the rheumatology liter- Importantly, the risk of stroke recurrence
ature [11]. This forms the rationale for is at least 18 % and is maximal in the
steroid treatment during the initial months initial few weeks after index stroke.
after stroke at some centers in combina- Many patients are left with some degree
tion with antithrombotic and, if post- of residual arterial stenosis. This high-
varicella, with acyclovir treatment. Arte- lights the importance of serial imaging in
rial wall imaging using high-resolution these cases.
42 Pediatric Stroke 1021

Fig. 10 Five-year-old boy who suddenly collapsed. vasculopathy. A history of chicken pox 2 months prior
Developed right-sided hemiparesis. MRI shows left basal implies a causal relationship
ganglia stroke as a typical location of strokes related to

occlusive disease of the terminal internal


carotid arteries or the other arteries around
the circle of Willis with prominent arterial
collateral circulation. The characteristic
appearance of a dilated network of
lenticulostriate perforating vessels leads
to the term “moyamoya” (“puff of
smoke” in Japanese). If moyamoya is
associated with an underlying
condition (e.g., sickle cell disease, Neuro-
fibromatosis Type 1, Down syndrome), it
is referred to as “moyamoya syndrome.”
Moyamoya disease and moyamoya syn-
drome occur in children and adults of all
ages. Rarely it can present in infancy
(Fig. 12). Moyamoya can be bilateral or
unilateral.
Fig. 11 Five-year-old boy, history of two episodes of
transient neurological symptoms over the past 2–3 days.
Common clinical presentations in
Prior MR showed right lentiform infarct. Conventional childhood include headaches, transient
angiography showing beading appearance of M1 segment ischemic attacks, and AIS. Symptomatic
of right MCA (arrow) ischemic episodes can often be triggered
by crying, blowing, coughing, or hyper-
(B) Moyamoya disease and moyamoya ventilation. Cognitive decline is another
syndrome concern. Intracranial hemorrhage, com-
Moyamoya is a rare progressive cerebro- mon in adults, is relatively rare in
vascular disorder characterized by childhood.
1022 D. Tibussek et al.

Fig. 12 Seven-month-old male with sickle cell disease. ACA territory. MRA revealed non-visualization of the
Presented with acute focal seizures and eye deviation to the right ACA and MCA. The patient was later diagnosed
right. Acute diffusion restriction involving the MCA and with moyamoya syndrome

Studies from Asia suggest that for moyamoya describing the presence
two-thirds of children will have symptom- of high signal intensity at the outer
atic progression at 5 years after diagnosis. edge of the cortical ribbon on FLAIR
The annual stroke risk in asymptomatic sequences, likely reflecting engorged
pediatric patients was estimated at 3.2 % pial vessels. However, artifact-related
[34]. Little is known about the natural causes of FLAIR hyperintensity have
course of moyamoya in non-Asian to be kept in mind, above all in children
children. receiving supplemental oxygen during
After diagnosis it is now widely MRI under anesthesia.
accepted that all affected children should In addition to focal infarction presumably
be referred for surgical evaluation. How- due to thrombotic occlusion in
ever, no strict criteria exist to define which established arterial territories,
child should undergo revascularization moyamoya also produces a second,
surgery. Imaging findings, including cere- very important mechanism for infarc-
brovascular reserve (CVR) studies, clini- tion, namely, hypoperfusion leading to
cal symptoms, and ischemic events (TIA, internal or deep watershed infarcts that
AIS), as well as neuropsychological may be subclinical. The typical imag-
assessment, should all be taken into ing pattern of these deep white matter
consideration. infarcts is a “string of pearls” appear-
ance (Fig. 14). Impaired cerebrovascu-
(a) Imaging of Moyamoya lar autoregulation appears to be an
The diagnosis of moyamoya can be easily important underlying mechanism for
made using MRI and MRA (Fig. 13). these infarcts [35]. Functional vascular
Besides visualization of unilateral or imaging can directly or indirectly deter-
bilateral stenosis of the terminal inter- mine inadequate perfusion and cere-
nal carotid arteries (ICAs) and the per- brovascular reactivity (CVR). In some
forator artery collateral network, centers these techniques are routinely
conventional MRI sequences may used in patients with moyamoya dis-
demonstrate the so-called ivy sign. ease and may assist in treatment deci-
This is a sensitive but nonspecific sign sions and help to predict the success of
42 Pediatric Stroke 1023

Fig. 13 Seven-year-old girl with a recent history of “complicated” migraine, including headache and unilateral
weakness and tingling. MRI revealed an ivy sign. Moyamoya disease was confirmed with conventional angiography

angiographic findings include stenosis


or occlusion at the distal ICA and/or the
origin of the anterior cerebral and middle
cerebral arteries on both sides. In addi-
tion, moyamoya vessels are characteris-
tic reflecting the collateral vascular
networks of the lenticulostriate perfora-
tors. CA also helps to visualize the
dynamics of the blood flow, collatera-
lization, and areas of cerebral hypo-
perfusion. Before revascularization it
also assists in displaying arteries poten-
tially available for bypass surgery
(superficial temporal artery, external
carotid artery).

(b) Sickle Cell Disease (SCD) and Moyamoya


Syndrome
Strokes affect 7–11 % of children with
Fig. 14 Same patient as Fig. 13: typical “string of pearls” sickle cell disease. Ischemic strokes
appearance indicating chronic deep (internal) border zone predominately affect young patients
infarcts between the ages of 2 and 5. Hemor-
rhagic strokes have the highest preva-
lence in patients between the ages of
reperfusion after revascularization 20 and 29 years with SCD. Silent
surgery. infarcts were identified in 21.8 % of
The gold standard to establish the diagnosis children between 6 and 19 years of
of moyamoya is conventional angiogra- age with SCD-SS in one study. Further
phy (Fig. 13). Characteristic studies have shown an estimated
1024 D. Tibussek et al.

cumulative incidence of silent infarct as increased stroke risk, and regular trans-
high as 37 % in patients with SCD by fusion therapy (target hemoglobin S
14 years of age [36]. <20–30 %) significantly reduces the
How SCD leads to cerebrovascular disease stroke risk in children with elevated
is only partly understood and likely mul- Doppler velocities. Additional stroke
tifactorial. The two favored mechanisms prevention strategies are currently
include a progressive internal carotid under evaluation including hydroxy-
moyamoya-type arteriopathy and an urea and nocturnal oxygen supplemen-
occlusion of small arteries by sickled tation. Aspirin should be considered,
cells and related thrombus resulting in especially in cases with large-vessel
randomly distributed multifocal small vasculopathy and/or recurrent stroke.
infarcts. Revascularization surgery may be suc-
In about 64 % of children with homozy- cessful in SCD-related moyamoya.
gous SCD, stroke is related to a large- (C) Fibromuscular dysplasia
vessel occlusive vasculopathy, usually FMD is a segmental vasculopathy of
moyamoya syndrome. The unknown etiology. The disease predomi-
supraclinoid ICA is most affected with nantly affects the renal arteries and the
less involvement of the proximal mid- internal carotid arteries. In contrast to
dle and anterior cerebral arteries. Pro- adults, where medial fibrodysplasia is
gression of this vasculopathy with common, intimal fibrodysplasia is the typ-
formation of moyamoya collaterals ical pathological finding in children
has been reported in up to 40 % of [39]. Involvement of other vessels can be
patients [37]. However, a small per- observed, including hepatic, celiac, mes-
centage of children have clinically enteric, axillary, and external iliac arteries.
apparent strokes without evidence of a Childhood FMD is believed to be rare and
large-vessel vasculopathy. poorly understood.
Up to 37 % of all children with sickle cell Characteristic angiographic findings
disease will have areas of signal abnor- have been described; however, they are
mality on MRI without a history of nonspecific. The most common finding is
acute neurological impairment indicat- the so-called arterial “string of beads”
ing silent ischemic infarction. Such appearance, characterized by multifocal
children develop progressive cognitive short stenoses (Fig. 11). However, this
decline as the lesions accumulate. finding is also well described in other
These lesions are typically small and arteriopathies of childhood [17]. There-
multiple and follow the distribution of fore, the diagnosis of FMD is challenging
the internal white matter border zones. and cannot be confirmed based solely on
In serial MRI with MRA, screening for angiographic features, renal arteriopathy,
presymptomatic cerebrovascular dis- or clinical findings. Histopathology is cur-
ease is now standard of care in SCD rently the only confirmative diagnostic
to detect silent infarcts secondary to tool for childhood fibromuscular dysplasia
small-vessel disease and possible [39]. As in other arteriopathies, there is a
moyamoya. risk of recurrence of ischemic stroke
Transcranial Doppler (TCD) screening to [39]. Preventive treatment, mainly aspirin,
measure flow and detect large-vessel will usually be indicated in cases of
arteriopathy and predict stroke risk is pediatric FMD.
now standard of care of children with (D) Cervical arterial dissection
SCD based on strong evidence Arterial dissection is likely to be
[38]. Flow rates above 200 cm/s predict underdiagnosed in children. It accounts
42 Pediatric Stroke 1025

for 14–20 % of CAIS cases and has been the neck or skull base will best visual-
reported in up to 50 % of posterior circu- ize the abnormal intramural signal. In
lation strokes [40]. Males are more com- addition, a tapered narrowing or even
monly affected. Besides head and neck occlusion of the dissected vessel at
trauma, several potentially causative fac- characteristic arterial locations may be
tors have been reported, including preced- visualized on MRA.
ing infections, inflammation, connective However, negative MR and CT imaging of
tissue disorders, and homocystinuria. the cervical arteries does not exclude the
Non-accidental trauma should be consid- diagnosis of ICAD. It is important to
ered as an important differential diagnosis, recognize that time-of-flight MRA can
especially in small children with miss dissection in up to 61 % of cases.
dissection. Especially in cases of presumed dissec-
In children, the dissecting event is usu- tion where the diagnosis could not be
ally followed by a latent period lasting days established by MRA or CTA, CA
or even weeks after trauma. New onset remains the gold standard. Typical
head or neck pain may be a presenting angiographic findings include an intimal
feature in children. Compared to adults, flap, a double lumen, or a long tapered
children with dissection have a high likeli- stenosis at characteristic locations (also
hood of presenting with an AIS as the first referred to as “flame-shaped” stenosis).
symptom of dissection. Recurrence rates The formation of a pseudoaneurysm will
are estimated at least 20 % [5, 40]. sometimes be identified on follow-up
(a) Imaging Features imaging (Fig. 15).
In posterior circulation dissection, verte- Follow-up imaging is always indicated
bral artery at the level of C1–C2 is the and will guide treatment decisions.
most typical location. Extracranial ICA Complete resolution will occur in the
dissections commonly occur near the majority of cases [40]. However, per-
origin of the ICA or at the distal sistence (23 %) or even progression
segment before or immediately after (10 %) has been reported.
entering the skull base [40]. Intracranial 2. Cardiac disorders
dissection involving the anterior Congenital heart disease is a risk factor for
circulation in children also occurs embolic stroke [42], especially in the setting of
and carries the risk of dissecting cardiac surgery, cardiac catheterization, extra-
aneurysms secondary to subarachnoid corporeal membrane oxygenation (ECMO),
hemorrhage [40]. and ventricular assist device (VAD) placement.
Neuroimaging is essential to establish the Cardiac diseases are responsible for about
diagnosis of dissection [41]. MRI will one-third of all AIS in children [43, 15].
detect ischemic lesions, which are However, ischemic brain lesions related to
commonly found at multiple locations, congenital heart disease are frequently asymp-
especially in the posterior circulation. tomatic and may be found on routine
In these cases MRI allows estimation preoperative MRI.
of the timing of the ischemic events. In addition to congenital heart disease,
Specific arterial wall MRI findings of acquired cardiac etiologies predispose to AIS,
dissection include an intramural hema- including infective endocarditis, cardiomyop-
toma that is demonstrated as enlarge- athy, valvular heart disease, arrhythmia, and
ment of the artery with an eccentrically atrial myxoma.
located crescent-shaped rim of abnor- AIS in multiple, distinct vascular
mal signal. Axial fat-suppressed, territories suggests a cardiac source. Infarcts
non-contrast T1-WI images through are usually thromboembolic with typical
1026 D. Tibussek et al.

Fig. 15 Ten-year-old boy with sudden neck pain and vertebral artery. Follow-up angiography after 3 months
dizziness during basketball game. Conventional angiogra- revealed a pseudoaneurysm
phy revealed a filling defect and irregular stenosis of right

peripheral infarcts involving the cortex and Table 7 Role of thrombophilia in childhood arterial
ischemic stroke and cerebral sinovenous system (Trenor
subcortical white matter of both the anterior and Michelson [44] with permission)
and posterior circulation. Vascular imaging
Odds ratio Odds ratio
may show abrupt occlusion(s) indicating (95 % (95 %
emboli. confidence confidence
Hypoperfusion patterns of infarction lim- interval) for interval) for
ited to watershed distributions are commonly Thrombophilia AIS CSVT
Two or more 18.75 6.12
seen after acute changes in cardiac output. The
genetic traits (6.49–54.14) (0.87–43.07)
role of PFO as a conduit for paradoxical embo-
Protein C 11.0 6.12
lism remains a concern, and to date the risk of deficiency (5.13–23.59) (0.87–43.07)
stroke in isolated childhood PFO remains Antiphospholipid 6.95 –
unknown. antibodies (3.67–13.14)
3. Prothrombotic conditions Lipoprotein 6.53 –
Inherited or acquired thrombophilia has been (a) elevation (4.46–9.55)
Factor V Leiden 3.70 2.74
identified in up to 50 % of children with
(2.82–4.85) (1.73–4.34)
AIS (Table 7). The overall impact of Antithrombin 3.29 18.41
thrombophilia in childhood AIS is still deficiency (0.70–15.48) (3.25–104.29)
unclear [44]. Prothrombotic disorders Factor II 2.60 1.95
likely play a role as additional “predisposing” G20210A (1.66–4.08) (0.93–4.07)
risk factors working in concert with other MTHFR 1.58 –
“triggering” factors at the time of AIS. thermolabile (1.20–2.08)
Protein S 1.49 5.27
They also predict increased risk of recurrence.
deficiency (0.32–6.92) (1.53–18.21)
Coagulation testing is generally recommended
in any child with AIS including children
with other identified risk factors. However,
accurate testing and interpretation of test 4. Genetics and stroke
results requires an experienced laboratory The role of genetic factors in causing child-
with well-established age-appropriate norma- hood stroke is increasingly recognized but
tive values. complex [45]. It is important to recognize that
42 Pediatric Stroke 1027

Fig. 17 Patient with ACTA2 mutation: symmetric prom-


inence of the distal cervical segments of the ICAs and
fusiform aneurysmal dilatation of the petrous and cavern-
ous segments. Symmetric stenoses and tapering noted at
the mid cavernous ICA segments and extending to the
Fig. 16 Example of a patient with PHACES syndrome carotid terminations, involving the anterior and middle
and multiple vascular abnormalities on MR angiography cerebral arteries with straightening of the major branches

certain genetic syndromes are associated with stroke-like episodes mimic AIS; however, the
an increased risk of strokes in childhood that is mechanism is metabolic rather than primarily
usually related to cerebral vasculopathies (e.g., ischemic [47].
Neurofibromatosis Type 1, PHACES (Fig. 16),
Alagille, Down, Williams, moyamoya).
More recently, monogenetic disorders asso- Treatment of Arterial Ischemic Stroke
ciated with arteriopathies have been identified,
among these COL4A1, ACTA2 (Fig. 17), and The main focus of early stroke management is
RNF213 in familial moyamoya cases. neuroprotection, prevention of stroke evolution,
Characteristic imaging patterns have been and recurrence and early rehabilitation. Treatment
described in some of these genetic disorders. of CAIS is based on consensus guidelines for
COL4A1 mutations may be a relatively acute and chronic treatment of CAIS [48–50].
common cause for a genetic small-vessel dis- Neuroprotection is a crucial mainstay of acute
ease affecting the brain, eyes, and kidneys, CAIS treatment. This includes adequate oxygen-
leading to intracerebral hemorrhage, cerebral ation, maintenance of normoglycemia, mainte-
calcification, and ischemic damage typically nance of blood pressure between the 50th and
with ante-, peri-, or postnatal onset. Character- 90th percentiles, and, importantly in children,
istically, but not exclusively, the disorder is aggressive seizure and fever control. Especially
associated with porencephaly. in the child with reduced level of consciousness,
Several inborn errors of metabolism predis- continuous EEG should be considered in order to
pose to AIS. Homocystinuria, a potentially detect nonconvulsive seizures.
treatable condition, can lead to both arterial Further treatment options for acute therapy of
and venous thrombosis and may be recognized CAIS include anticoagulation (ultra-fractionated
by clinical features and laboratory markers or low-molecular weight heparin) or antiplatelet
[46]. In mitochondrial diseases, including therapy with acetylsalicylic acid (ASA). No com-
mitochondrial encephalopathy, lactic acidosis, parative studies are available to prove superiority
and stroke-like episodes (Fig. 18), associated of one or the other drug.
1028 D. Tibussek et al.

Fig. 18 Example of a patient with proven MELAS syndrome who developed three stroke-like episodes within 6 months.
MRI revealed areas of diffusion restriction not matching arterial territories

While in adults strong evidence supports the There may be a role in selected children, for
usage of IV tPA, in CAIS no such evidence exists. example, an older child with life-threatening pos-
Safety concerns lead to the recommendation not terior circulation stroke. Recommendations for an
to use tPA in childhood stroke outside clinical optimal interdisciplinary approach to care of these
trials, even if a child presents within the 4.5 h children have been recently proposed [52]. A
window. However, despite safety concern, tPA is close collaboration of a neurointerventional team
increasingly administered to children, frequently with childhood stroke neurologists or, when not
outside the recommended 4.5 h window. Time to available, a team including a pediatric neurologist
treatment after onset up to 52 h has been and adult stroke neurologist, where at least one
reported [51]. member of the team has extensive pediatric stroke
Successful thrombectomy has been reported in experience, is required.
childhood basilar artery thrombosis [52]. How-
ever, severe complications have also been
reported [53]. Safety and efficacy are basically Hemorrhagic Stroke
unknown and such device use in children is not
FDA approved. Multiple published pediatric The incidence of hemorrhagic stroke (HS) in chil-
stroke guidelines all suggest avoidance of dren is similar to CAIS, approximately 1.4 per
endovascular procedures in the pediatric popula- 100,000 children per year. Intracerebral hemor-
tion [48–50]. It is crucial that interventional neu- rhage (ICH) is the most common type of hemor-
roradiologists be aware of this. Since the devices rhagic stroke and accounts for around 50 % of
for endovascular management of acute ischemic pediatric strokes.
stroke were designed for use in adults, safety is an Traumatic head injury, non-accidental injury,
even greater concern in the pediatric population. bleeding disorders, arteriovenous malformations
Also, the likelihood of a better outcome in most (Fig. 19), and hemorrhagic neoplasms are some of
children with CAIS treated conservatively under- the causes of hemorrhagic stroke in children [54].
lines the importance of avoiding endovascular The clinical presentation of HS in childhood is
treatments as a general rule. The use of thrombo- nonspecific, and symptomatology may be partic-
lytic therapies or thrombectomy in children with ularly misleading in young children and infants,
CAIS must be considered highly experimental. where altered mental status and convulsions are
42 Pediatric Stroke 1029

Fig. 19 Sixteen-year-old girl with sudden onset severe headache. CT angiography revealed a ruptured arteriovenous
malformation. AVM nidus involving the left thalamus and corona radiata

the most common presenting symptoms. Typical pediatric intensive care is always indicated. Any
signs in older children include headaches and further clinical deterioration should prompt a
sudden or rapidly progressive mental status follow-up CT scan to look for secondary lesions
changes with focal signs, whereas seizures are such as hydrocephalus, extension of an intracere-
only observed in 16 %. An important diagnostic bral hematoma, herniation, or vasospasm.
hint in all age groups is the sudden onset of focal
symptoms [54]. A high level of suspicion is there-
fore needed to allow early diagnosis and Prognosis
intervention.
Mortality in childhood hemorrhagic stroke is
about 23 %, usually as a result of the initial hem-
Imaging orrhage. Importantly, a recurrent cerebral bleed
resulted in death in 33 % of children in one
CT is generally considered the preferred acute study. This underlines the importance of a thor-
imaging of choice in children with suspected HS. ough etiological evaluation. More than 2/3 of
In a stable patient initial MRI may be consid- children will have motor impairment and/or cog-
ered. Blood-sensitive sequences such as SWI will nitive deficits at follow-up. Epilepsy developed in
identify hemorrhages. In addition, MRI may be 11 % of children.
helpful for differential diagnosis and identifica- Prognosis of HS is linked to the intrapar-
tion of an underlying etiology, such as AVM or enchymal component of the intraparenchymal
brain tumor. A thorough evaluation for vascular hemorrhage. Lesions including beyond 2 % of
anomalies is critical. If CTA and MRA do not total brain volume resulted in moderate disabil-
establish the cause of the HS, conventional angi- ities and [44] at or beyond 4 % in severe disability
ography needs to be considered. or death. Another reported predictor of poor out-
After the diagnosis has been established and come has been altered mental status within 6 h of
acute treatment initiated, close monitoring in hospital arrival.
1030 D. Tibussek et al.

Beslow et al. [55] recently demonstrated that a are initially well, and seizures tend to occur later
pediatric ICH score derived from volume of hem- in the first day than in neonates with
orrhage, infratentorial location, and presence of hypoxic–ischemic encephalopathy (HIE). Other
either hydrocephalus or herniation can predict the symptoms are nonspecific, including poor feed-
extent of disability or death. ing, hypotonia, and apnea. Presumed perinatal
ischemic stroke (PPIS) is a term that has been
introduced to characterize children, who typically
Perinatal Stroke present at the age of 3–6 months with early signs
of hemiparesis (e.g., early hand preference) or
The perinatal period is a focused high-risk period seizures [57].
for stroke. Perinatal strokes include arterials
ischemic stroke, periventricular venous infarc-
tion, hemorrhagic stroke, and CSVT. Strokes can Imaging
present acutely in the newborn period or later in
infancy with the evolution of chronic hemiplegic Perinatal arterial ischemic AISs are more typically
cerebral palsy as the brain matures. observed in the left hemisphere, mostly involving
Mortality of perinatal stroke is low, and fatal the MCA territory. A brain MRI after neonatal
cases are often related to an underlying disease. seizures is always indicated. In fact, a recent
However, the risk for long-term neurological def- study found that MRI after neonatal seizures con-
icits is significant, including unilateral spastic tribute to a final diagnosis in >90 % of cases, with
cerebral palsy (up to 50 %) and impairments in strokes being the second most common diagnosis
vision, cognition, language, and behavior [3, 56]. (12 %) after HIE [58].
The lifelong risk for epilepsy is increased. Recur- Serial cranial ultrasound is now routinely
rence of stroke in the child with a history of performed in most NICU. Ultrasound may reveal
perinatal stroke is rare. a wedge-shaped area of increased echogenicity
with a linear demarcation line. However, stroke
detection rates are not satisfactory, and further
Perinatal Arterial Ischemic Stroke imaging is required to exclude stroke [8].
MRI is the most sensitive imaging modality for
Perinatal AIS comprises approximately 25 % of detection of perinatal strokes. The most frequently
pediatric AIS, with the remainder in older infants used sequences include T1-WI, T2-WI, and
and children. Studies report an incidence of peri- diffusion-weighted imaging (DWI). As in older
natal strokes of approximately 1 per 1,600–5,000 age groups, DWI plays the most important role in
live births [6]. However, the fact that MRI is not the diagnosis of an acute PAIS. While an ischemic
routinely done in all neonates under intensive care lesion will be dark on ADC in an early stage, it
suggests that current estimates of incidence may be normalizes at 6–10 days (“pseudonorma-
too low. Also the frequency of delayed presentation lization”). Within the first week after PAIS, T2
of infants diagnosed beyond 6 months of age with will show high signal intensity in the affected
an emerging severe hemiparesis and chronic AIS cortex and white matter. Cortical highlighting is
indicates that silent perinatal infarcts having more often visible on T1 (Fig. 8). Typically a cystic
normal outcomes are likely missed frequently. evolution will be seen on follow-up.
Male neonates are at higher risk for stroke. An important imaging finding early after an
ischemic insult is the “acute Wallerian”
degeneration, characterized by restricted diffusion
Clinical Presentation on DWI at the level of the ipsilateral cerebral
peduncle. This finding has been identified as a
PAIS in the newborn often manifests with sei- reliable predictor of poor motor outcome [59]
zures, typically unilateral. Commonly the babies (Fig. 20).
42 Pediatric Stroke 1031

Fig. 20 Neonate with focal neonatal seizures due to


stroke. DWI revealed pre-Wallerian degeneration of the
corticospinal tract and corpus callosum

Fig. 21 Periventricular venous infarction noted on MRI in


a 3-month-old infant with early signs of hemiparesis
In children with PPIS neuroimaging shows
evidence of a remote, chronic infarction with
absent diffusion restriction, encephalomalacia, leukomalacia that show rather diffuse ex vacuo
gliosis, and atrophy of the cerebral peduncle enlargement of the posterior aspects of the lateral
reflecting chronic Wallerian degeneration. ventricles, MRI of patients with PVI shows a focal
Potential risk factors of perinatal strokes are gliosis at the site of infarct frequently with accom-
commonly identified and significantly differ panying blood products. In addition, the lateral
from older children. However, they are diverse ventricles typically show a very focal expansion
and the exact causal role is not always adjacent to the gliosis or a porencephalic cyst.
definable. Risk factors may be categorized in It is observed in term babies often without obvi-
prenatal/maternal (e.g., diabetes mellitus, thyroid ous perinatal risk factors. PVI represents a common
disease, infertility), peripartum (e.g., fetal distress, subgroup of presumed perinatal ischemic strokes
prolonged rupture of membranes, maternal fever), and is typically diagnosed among populations of
and neonatal conditions (e.g., low APGAR children with hemiplegic cerebral palsy [57].
scores, hypoglycemia, early onset bacterial
infection) [60].
Perinatal Hemorrhagic Stroke

Periventricular Venous Infarction Perinatal hemorrhagic stroke (PHS) is observed in


6.2 in 100,000 live births [38]. PHS is typically
Periventricular venous infarction (PVI) has been unifocal and unilateral (Fig. 21). Most newborn
described as a distinct and previously underappre- present with signs of encephalopathy (poor feed-
ciated stroke pattern predominantly seen in PPIS ing, apnea, respiratory distress) and seizures. Etiol-
[57]. PVI are typically localized in the ogies are difficult to identify and may be
periventricular white matter, sparing subcortical multifactorial [61]. While an underlying vascular
white matter, cortex, and basal ganglia, and occur malformation is often suggested, these are typically
in premature brain rather than term babies. Contrary not detectable, even on follow-up. Other risk
to MRI findings of patients with periventricular factors include thrombocytopenia, fetal distress,
1032 D. Tibussek et al.

and postmaturity. Bleeding disorders are rarely developing gradually over hours, days, or even
been identified. However, it is also important weeks. These include headache, papilledema, and
to recognize that approximately 20 % of appar- a decreased level of consciousness. Additional
ently normal newborns have small amounts of visual disturbances are present in 18 % of children
subdural, subarachnoid, or parenchymal hemor- (diplopia, visual field deficits, and progressive
rhages after birth. blindness due to prolonged compression of the
A recent study of hemorrhagic stroke in neo- optic nerves). Seizures are present in nearly half
nates suggested that hemorrhagic transformation of of children [63]. Focal deficits are a presentation
an arterial or venous infarction is the leading mech- of CSVT when focal hemorrhages or infarcts
anism of PHS [61]. Importantly, PHS is a well- accompany the thrombus.
documented complication of neonatal CSVT.

Risk Factors
Pediatric Cerebral Sinovenous
Thrombosis Risk factors for CSVT are age dependent. During
the first 28 days of life, these include maternal
Epidemiology fever/chorioamnionitis, hypoxic–ischemic injury,
dehydration, infection, and thrombophilia. Often,
Cerebral sinovenous thrombosis (CSVT) in child- neonates have more than one risk factor.
hood has been increasingly recognized during the In childhood CSVT, thrombosis results from a
past several decades. It accounts for one in four combination of systemic and local vascular
for cases of pediatric ischemic strokes, with an factors. Within individual patients, certain
incidence of around 1 per 100,000 [62]. The underlying risk factors including prothrombotic
highest incidence is found in neonates (2.6 per states may predispose to thrombosis, and other
100,000) [63]. Similar to AIS, important differ- states including acute illnesses or prothrombotic
ences in CSVT in children compared with adults medications act as triggering factors. Head and
have to be considered. neck infections including meningitis or otitis
media and mastoiditis are a common cause for
septic thrombophlebitis and CSVT among
Clinical Presentation toddlers.

The clinical presentation is influenced by the age


of the child, the extent and location of the throm- Imaging of CSVT
bus, and the presence or absence of associated
venous infarction. The diagnosis of CSVT in infants and children can
Seizures are the most common presentation in be challenging. Findings and radiologic appear-
neonates. Additional signs are usually nonspecific ances are more variable compared with adults.
and include lethargy, hypotonia, feeding Understanding of normal variations and possible
difficulties, respiratory distress, and apnea artifacts and correctly applying newer available
[63]. Focal neurological deficits are rare [63, 64]. techniques are crucial.
In severe cases, bulging fontanels, splaying of the MRI with venography (MRV) or CT venogra-
cranial sutures, and prominent scalp veins may be phy (CTV) are the methods of choice for
observed, indicating poor cerebral venous drain- investigation of CSVT. The diagnosis is
age and increased intracranial pressure. established by demonstration of a causal throm-
In older infants and children, signs of raised bus, a lack of flow in the affected cerebral veins,
intracranial pressure predominate, typically and possibly additional venous infarction or
42 Pediatric Stroke 1033

hemorrhage. Brain parenchymal injuries can Magnetic Resonance Imaging


result from venous congestions leading to and Venography
impaired parenchymal venous drainage, with
resultant venous hypertension, vasogenic and MR imaging is currently the diagnostic study of
cytotoxic edema, and possibly hemorrhagic choice in neonatal and childhood CSVT. MRI
venous infarction. with MRV is able to visualize flow, the actual
The superficial venous system is more thrombus, and areas of infarction and may also
frequently involved than the deep system with identify an underlying abnormality [65]. The
the transverse, superior sagittal, sigmoid, appearance of the thrombus on MRI is age depen-
and straight sinuses being the most common dent. An acute thrombus within a sinus is isodense
sites of CSVT. Multiple sites of obstruction or hyperintense with brain on T1-WI images and
within the sinovenous system at the time of diag- dark on T2-WI images. In the subacute stage, the
nosis may be present. In severe cases, the superfi- clot is isointense to brain on T1-WI images and
cial and deep systems are thrombosed in hypointense on T2-WI images. A chronic throm-
combination. bus becomes hyperintense on both T1-WI and
Nearly 60 % of pediatric patients with CSVT T2-WI images (Table 8).
have associated parenchymal infarcts Time-of-flight (TOF) MR venography is com-
[63]. Thrombosis of the superficial system will monly used to visualize sinuses and cerebral veins
cause infarcts in the cortex and white matter and (Fig. 22). However, since the signal is flow depen-
may be bilateral. A thrombosis located in the deep dent, it is prone to artifacts, particularly in neo-
venous system usually affects one or both thalami nates and infancy due to the slower venous flow
or the cerebellum [65]. and anatomically smaller veins. Flow gaps are
Intraventricular or thalamic hemorrhage in also common in the nondominant transverse
term babies strongly suggests sinovenous throm- sinus. In neonates, compression of posterior por-
bosis involving the deep system. tion of the superior sagittal sinus by the occipital
bone is another reason for a localized flow artifact.
Ultrasound Another commonly misinterpreted finding in neo-
The presence of an open fontanel allows radiolo- nates is subdural blood adjacent to the tentorium
gist and neonatologists to image the brain and and the torcula that can be confused with lateral
sinovenous system with ultrasound. Cranial ultra- sinus thrombosis. Imaging in different planes may
sound may detect a midline thrombus in the supe- be necessary to differentiate both conditions.
rior sagittal sinus. Susceptibility-weighted imaging (SWI) has
Doppler ultrasound can define absent or reduced been reported as another useful sequence in
flow in the sinovenous channels in CSVT. A uni- confirming the presence of CSVT and its
lateral thalamic hemorrhage or venous infarcts as complications. Deoxygenated hemoglobin within
well as intraventricular hemorrhages can also be cerebral veins serves as an intrinsic contrast that
detected by cranial ultrasound. Although cranial helps to visualize even tiny cerebral veins. Abnor-
ultrasound is highly operator dependent, it can be mal prominence of cerebral veins may be seen on
very useful and effective in skilled hands. Supple- SWI especially when a CSVT involves deep
mental cranial ultrasound with color and pulsed venous structures. This sign can be visible in
Doppler may also improve diagnostic accuracy in early CSVT. Blooming on SWI within
equivocal cases. One study showed that color thrombosed cortical veins may be another useful
Doppler ultrasound is highly specific for detecting imaging finding indicating cortical vein thrombo-
true negative cases of cerebral venous thrombosis in sis. Finally, SWI is the most sensitive pulse
clinically suspected neonates. In this study, excel- sequence for the detection of small areas of paren-
lent reading agreement was noted [66]. chymal hemorrhage.
1034 D. Tibussek et al.

Table 8 Appearance of thrombus at various stages on different MRI sequences (From Bracken et al. [67] with
permission)
Contrast-
Fluid- Susceptibility- enhanced Time-of-
Thrombus age T1-WI sensitive weighted T1-WI flight Contrast-enhanced
(stage) images images images images MRV MRV
<24 h SI$(#) SI(") +/SI# Filling Absent Filling defect
(hyperacute) defect flow
1–3 days (acute) SI$ SI# may SI# Filling Absent Filling defect
mimic normal defect flow
SI# flow void
3–14 days SI" Early subacute Early Late Late Late subacute SI" may
(subacute; early, SI# may subacute SI#. subacute subacute mimic normal flow
days 3–7; late, mimic normal Late subacute SI" may SI" may
days 3–7; late, flow void. may not show mimic mimic
days 7–14) Late subacute SI# normal normal
SI" flow flow
>14 days SI# SI# may May not show Chronic Absent Filling defect. Delay
(chronic) may mimic normal SI# clot may flow between contrast
mimic flow void enhance administration and
normal imaging may result in
flow thrombus enhancement
void
SI signal intensity, $ isointense, # hypointense, " hyperintense, (#) slightly hypointense, (") slightly hyperintense, +/
may or may not be

Fig. 22 Ten-year-old girl with a history of otitis media, right jugular bulb are noted. Corresponding bright signal
headache, and new onset blurred vision. On MRV flow on T2/FLAIR images (arrow)
gaps involving the right transverse and sigmoid sinus and
42 Pediatric Stroke 1035

Computed Tomography and fever, and prevention of recurrent seizures,


is essential. In older children treatment for
Computed tomography (CT), including CTV, is intracranial hypertension may be required.
still often the initial test used in children with This frequently includes acetazolamide and, if
suspected CSVT. Easy access, short scanning persistent, serial lumbar punctures with removal
time, and ability to detect acute hemorrhage are of CSF. Careful monitoring of headache and
some advantages. On the unenhanced CT, visual impairment, the degree of papilledema,
increased attenuation within cortical veins, and visual field defects is important. Rarely, in
superior and inferior sagittal sinuses, or the deep resistant cases, placement of a lumboperitoneal
midline venous structures should raise suspicion shunt or optic nerve sheath fenestration may be
of CSVT. A number of CT signs have been necessary.
described as suggestive of CSVT (cord sign, Available data from neonatal childhood studies
hyperdense dural sinus sign, empty delta sign); supports that anticoagulant therapy should be con-
however, sensitivity is insufficient. Normal cere- sidered in children and neonates with CSVT. The
bral veins and dural venous sinuses may be safety of anticoagulation therapy in children and
hyperdense in newborns due to physiological neonates with CSVT previously was supported by
polycythemia. numerous studies [68, 63]. Anticoagulation aims
However, in equivocal cases, particularly if deep to prevent the propagation of an existing thrombus
venous infarction or cortical venous thrombosis is and to enable the dissolution of existing thrombus
suspected, high-resolution CT venography as an by the activity of the fibrinolytic system. Without
endoluminal technique may be required. At CTV a anticoagulation propagation of the thrombus is
thrombus is seen as an area of non-enhancement documented in 30 %, associated with worse
within a venous structure. In addition, CTV may outcome [69].
help to clarify the venous anatomy in cases where However, in the treatment decisions, several
TOF MRV suggests absent flow. factors must be considered. These include the
extent and location of the thrombus, the presence
of intracranial hemorrhage, and the reversibility of
Follow-Up Imaging risk factors for the CSVT. If anticoagulants are not
given, it is crucial to reimage and assess for prop-
The timing and type of follow-up imaging after agation of the initial thrombus or evolution of
confirmed CSVT varies with the clinical status parenchymal venous infarcts 5–7 days later.
and type of therapy used in the individual patient.
When anticoagulation has been initiated, an early
follow-up CT or blood-sensitive MRI technique is Outcome
suggested to exclude hemorrhagic complications.
Further follow-up studies using MRV are indi- CSVT is a potentially life-threatening condition.
cated to evaluate whether the thrombus is resolv- In the Canadian Registry, a mortality rate of 8 %
ing or extending under therapy and before was found [63]. Recurrent CSVT or other sys-
termination of treatment. temic thromboses occur in 13 % of children with
CSVT. Recurrence may even occur despite
receiving initial anticoagulation therapy.
Treatment Few studies document long-term outcomes in
more detail. However, motor and cognitive
If applicable, specific primary treatment for all impairments, as well as epilepsy, can be fre-
reversible underlying risk factors, including mas- quently observed [64]. The presence of a cerebral
toiditis or meningitis, is indicated. In addition, venous infarction is one predictor of poor out-
neuroprotection, including maintenance of blood come, particularly when infarction is bilateral
pressure, correction of hyper- or hypoglycemia [63, 8].
1036 D. Tibussek et al.

Prediction of Outcomes from Pediatric demonstrate the ability of acute DWI changes
Stroke: The Role of Imaging within the descending corticospinal tracts to pre-
Despite the widely held belief that the increased dict poor motor outcome [59].
plasticity of the young brain protects against the The addition of studying brain plasticity to
effects of injury, there is now considerable evidence studies of stroke lesion characteristics in
that early brain injury results in significant long- predicting long-term motor outcome in childhood
term impairment. Recent studies provide evidence is a promising area of imaging research. Asym-
that early stroke-related brain injury commonly metry of diffusion tensor imaging (DTI) parame-
leads to widespread motor, sensory, behavioral, ters has been recently correlated with an increase
and cognitive dysfunction [70, 3, 71]. However, in severity of motor impairments after neonatal
outcomes are variable in all age groups. strokes. DTI-based tractography performed
Prediction of clinical outcomes of children 3 months after a perinatal stroke showed an excel-
with ischemic stroke is of major importance for lent positive predictive value in terms of later
clinical management including selection of more unilateral motor dysfunction [73].
aggressive therapeutic options, counseling of par- Novel methods of imaging classification of
ents, understanding the child’s health-related pediatric stroke are just emerging which combine
needs, and planning of rehabilitation and early type, location, and volume assessment,
intervention programs. including Perinatal Stroke Classification system
A number of cognitive outcome studies [57] and a pediatric modification of an adult
suggested worse outcomes after neonatal or peri- method, namely, the modified pediatric ASPECT
natal stroke compared to later in childhood. A scoring [74]. Recognizable patterns of presumed
U-shaped curve relating cognitive deficits with perinatal stroke may predict neurological morbid-
age is also reported. Lesions that occurred in ity in [57].
middle childhood (5–10 years old) led to better With the rapid progress and availability of
cognitive outcome than early lesions (0–5 years sophisticated brain imaging techniques, more
old) or late (10–18 years old). Age and brain mat- detailed analyses of type, localization, and extent
uration likely play an important role when looking of ischemic lesions as well as cerebral reorgani-
at vulnerability and repair mechanisms [4]. zation and functional consequences after cerebral
Available studies are somewhat inconsistent ischemia will be made possible.
with regard to stroke volume and location and Some imaging observations of yet unknown
cognitive outcome. Current evidence suggests significance may deserve further research.
that combined cortical and subcortical lesions A frequent finding in posterior circulation
are linked to a worse cognitive outcome [4]. strokes in neonates is a diffusion restriction of
Limited evidence suggests that motor outcome the optic radiation, sometimes also affecting the
may be predicted by type, localization, and extent corpus callosum (Fig. 23).
of the ischemic zone. In neonates concurrent A high percentage of these children suffering
involvement of basal ganglia, posterior limb of from visual field abnormalities suggests a func-
the internal capsule, and cerebral cortex predicted tional significance. Another example is cerebellar
hemiparesis compared with other patterns of atrophy associated with a contralesional pontine
injury. Kirton et al. [72] showed that an abnormal DWI signals that was recently demonstrated on
signal on diffusion-weighted MRI in the MRI after CAIS [75].
descending corticospinal tract (“acute Wallerian Post-stroke dystonia is a severe and common
degeneration”) was predictive of hemiparesis movement disorder affecting approximately 25 %
after neonatal stroke. Chronic Wallerian degener- of children after basal ganglia stroke. However,
ation was seen in all children with hemiparesis research focusing on imaging prediction of
[72] (Fig. 20). post-stroke movement disorders is not available.
When studying children with stroke outside the In the future, functional imaging techniques such
neonatal period, Domi et al. [59] were able to as resting state fMRI and other methods of
42 Pediatric Stroke 1037

In the near future more sophisticated imaging


techniques such as vessel wall imaging, DTI, and
functional imaging techniques will likely emerge
into the clinical arena and play an important role
in enhancing our understanding of pediatric
stroke.

References
1. Mallick AA, Ganesan V, Kirkham FJ et al (2014)
Childhood arterial ischaemic stroke incidence,
presenting features, and risk factors: a prospective
population-based study. Lancet Neurol 13(1):35–43
2. Agrawal N, Johnston SC, Wu YW et al (2009) Imaging
data reveal a higher pediatric stroke incidence than
prior US estimates. Stroke 40(11):3415–3421
3. Kirton A, deVeber G (2013) Life after perinatal stroke.
Stroke 44(11):3265–3271
4. Westmacott R, Askalan R, MacGregor D et al (2010)
Cognitive outcome following unilateral arterial
ischaemic stroke in childhood: effects of age at stroke
Fig. 23 Neonatal stroke involving the right posterior cir-
and lesion location. Dev Med Child Neurol
culation. On DWI highlighting of the optic radiation is
52(4):386–393
noted, likely representing acute Wallerian degeneration
5. Fullerton HJ, Wu YW, Sidney S et al (2007) Risk of
recurrent childhood arterial ischemic stroke in a
connectivity measurement will help to better under- population-based cohort: the importance of cerebro-
stand the underlying networks and to identify chil- vascular imaging. Pediatrics 119(3):495–501
6. Kirton A, deVeber G (2009) Advances in perinatal
dren at risk.
ischemic stroke. Pediatr Neurol 40(3):205–214
7. Srinivasan J, Miller SP, Phan TG et al (2009) Delayed
recognition of initial stroke in children: need for
Summary increased awareness. Pediatrics 124(2):e227–e234
8. Golomb MR, Dick PT, MacGregor DL et al (2003)
Cranial ultrasonography has a low sensitivity for
Important progress has been made during recent detecting arterial ischemic stroke in term neonates. J
years contributing to a better understanding of Child Neurol 18(2):98–103
pediatric stroke. It is now clear that significant 9. Groenendaal F, van der Ground J, Witkamp TD
et al (1995) Proton magnetic resonance spectroscopic
differences between neonates (birth-28 days),
imaging in neonatal stroke. Neuropediatrics
children (1 month–18 years), and adults need to 26:243–248
be considered when dealing with stroke patients. 10. Mackay MT, Wiznitzer M, Benedict SL et al (2011)
Due to different underlying mechanisms and Arterial ischemic stroke risk factors: the international
pediatric stroke study. Ann Neurol 69:130–140
important developmental changes throughout
11. Benseler S, Pohl D (2013) Childhood central nervous
childhood, neuroimaging and management deci- system vasculitis. Handb Clin Neurol 112:1065–1078
sions in pediatric stroke cannot be simply extrap- 12. Meoded A, Poretti A, Benson JE et al (2014) Evalua-
olated from adults. tion of the ischemic penumbra focusing on the venous
drainage: the role of susceptibility weighted imaging
Imaging, usually by means of MRI, plays a
(SWI) in pediatric ischemic cerebral stroke.
crucial role in pediatric stroke in order to detect J Neuroradiol 41(2):108–16
and characterize an ischemic lesion and exclude 13. Bosemani T, Verschuuren SI, Poretti A et al (2013)
common stroke mimics. It also provides valuable Pitfalls in susceptibility-weighted imaging of the pedi-
atric brain. J Neuroimaging 24:221–225
information about the potential etiology and prog-
14. Hoffman CE, Santillan A, Rotman L et al (2014) Com-
nosis. This in turn guides further diagnostic steps plications of cerebral angiography in children younger
and assists in treatment decisions. than 3 years of age. J Neurosurg Pediatr 13(4):414–419
1038 D. Tibussek et al.

15. deVeber G, Canadian Paediatric Ischemic Stroke Study arteriopathy in childhood arterial ischemic stroke. J
Group (2000) Canadian paediatric ischemic stroke reg- Child Neurol 26(9):1203–1206
istry: analysis of children with arterial ischemic stroke 32. Thomas SL, Minassian C, Ganesan V et al (2014)
(Abstract). Ann Neurol 48(3):514 Chickenpox and risk of stroke: a self-controlled case
16. Askalan R, Laughlin S, Mayank S et al (2001) series analysis. Clin Infect Dis 58(1):61–68
Chickenpox and stroke in childhood: a study of fre- 33. Mineyko A, Kirton A (2013) Mechanisms of pediatric
quency and causation. Stroke 32(6):1257–1262 cerebral arteriopathy: an inflammatory debate. Pediatr
17. Braun KP, Bulder MM, Chabrier S et al (2009) The Neurol 48(1):14–23
course and outcome of unilateral intracranial 34. Kurokawa T, Chen YJ, Tomita S et al (1985) Cerebro-
arteriopathy in 79 children with ischaemic stroke. vascular occlusive disease with and without the
Brain 132(Pt 2):544–557 moyamoya vascular network in children.
18. Mackay MT, Prabhu SP, Coleman L (2010) Childhood Neuropediatrics 16(1):29–32
posterior circulation arterial ischemic stroke. Stroke 35. Mikulis DJ, Krolczyk G, Desal H et al (2005) Preop-
41(10):2201–2209 erative and postoperative mapping of cerebrovascular
19. Goeggel SB, Ritter B, Gautschi M et al (2013) Basilar reactivity in moyamoya disease by using blood oxygen
artery stroke in childhood. Dev Med Child Neurol level-dependent magnetic resonance imaging. J
55(1):65–70 Neurosurg 103(2):347–355
20. Mangla R, Kolar B, Almast J et al (2011) Border zone 36. Bernaudin F, Verlhac S, Arnaud C et al (2011) Impact
infarcts: pathophysiologic and imaging characteristics. of early transcranial Doppler screening and intensive
Radiographics 31(5):1201–1214 therapy on cerebral vasculopathy outcome in a new-
21. Beslow LA, Smith SE, Vossough A et al (2011) Hem- born sickle cell anemia cohort. Blood 117(4):
orrhagic transformation of childhood arterial ischemic 1130–1140
stroke. Stroke 42(4):941–946 37. Prengler M, Pavlakis SG, Prohovnik I et al (2002)
22. Schechter T, Finkelstein Y, Ali M et al (2012) Sickle cell disease: the neurological complications.
Unfractionated heparin dosing in young infants: clini- Ann Neurol 51(5):543–552
cal outcomes in a cohort monitored with anti-factor Xa 38. Armstrong-Wells J, Grimes B, Sidney S et al (2009)
levels. J Thromb Haemost JTH 10(3):368–374 Utilization of TCD screening for primary stroke pre-
23. Rasalkar DD, Chu WC (2012) Imaging in children vention in children with sickle cell disease. Neurology
presenting with acute neurological deficit: paediatric 72(15):1316–1321
stroke mimics. Postgrad Med J 88(1045):639–648 39. Kirton A, Crone M, Benseler S et al (2013)
24. Smith SE, Kirkham FJ, deVeber G et al (2011) Out- Fibromuscular dysplasia and childhood stroke. Brain
come following decompressive craniectomy for malig- J Neurol 136(Pt 6):1846–1856
nant middle cerebral artery infarction in children. Dev 40. Rafay MF, Armstrong D, deVeber G et al (2006)
Med Child Neurol 53(1):29–33 Craniocervical arterial dissection in children: clinical
25. Rahme R, Jimenez L, Bashir U et al (2013) Malignant and radiographic presentation and outcome. J Child
MCA territory infarction in the pediatric population: Neurol 21(1):8–16
subgroup analysis of the Greater Cincinnati/Northern 41. Orman G, Tekes A, Poretti A et al (2013) Posttraumatic
Kentucky Stroke Study. Childs Nerv Syst 29(1): carotid artery dissection in children: not to be missed!
99–103 42. Giglia TM, Massicotte MP, Tweddell JS et al (2013)
26. Beslow LA, Vossough A, Dahmoush HM et al (2012) Prevention and treatment of thrombosis in pediatric
Modified pediatric ASPECTS correlates with infarct and congenital heart disease: a scientific statement
volume in childhood arterial ischemic stroke. Front from the American Heart Association. Circulation
Neurol 3:122 128(24):2622–2703
27. Mackay MT, Chua ZK, Lee M et al (2014) Stroke and 43. Ganesan V, Prengler M, McShane MA et al (2003)
nonstroke brain attacks in children. Neurology Investigation of risk factors in children with arterial
82(16):1434–1440 ischemic stroke. Ann Neurol 53(2):167–173
28. Shellhaas RA, Smith SE, O’Tool E et al (2006) Mimics 44. Trenor CC III, Michelson AD (2010) Thrombophilia
of childhood stroke: characteristics of a prospective and pediatric stroke. Circulation 121(16):1795–1797
cohort. Pediatrics 118(2):704–709 45. Munot P, Crow YJ, Ganesan V (2011) Paediatric
29. Amlie-Lefond C, Bernard TJ, Sebire G et al (2009) stroke: genetic insights into disease mechanisms and
Predictors of cerebral arteriopathy in children with treatment targets. Lancet Neurol 10(3):264–274
arterial ischemic stroke: results of the International 46. Bodensteiner JB, Hille MR, Riggs JE (1992) Clinical
Pediatric Stroke Study. Circulation 119(10): features of vascular thrombosis following varicella.
1417–1423 Am J Dis Child 146(1):100–102
30. Bernard TJ, Manco-Johnson MJ, Lo W et al (2012) 47. Mudd SH, Skovby F, Levy HL et al (1985) The natural
Towards a consensus-based classification of childhood history of homocystinuria due to cystathionine beta-
arterial ischemic stroke. Stroke 43(2):371–377 synthase deficiency. Am J Hum Genet 37(1):1–31
31. Dlamini N, Freeman JL, Mackay MT et al (2011) Intra- 48. Monagle P, Chalmers E, Chan A et al (2008)
cranial dissection mimicking transient cerebral Antithrombotic therapy in neonates and children:
42 Pediatric Stroke 1039

American College of Chest Physicians evidence-based 61. Bruno CJ, Beslow LA, Witmer CM et al (2014)
clinical practice guidelines (8th Edition). Chest Haemorrhagic stroke in term and late preterm neonates.
133(6 Suppl):887S–968S Arch Dis Child Fetal Neonatal Ed 99(1):F48–F53
49. Paediatric Stroke Working Group (2004) Stroke in 62. Mallick AA, O’Callaghan FJ (2010) The epidemiology
childhood: clinical guidelines for diagnosis, manage- of childhood stroke. Eur J Paediatr Neurol 14(3):
ment and rehabilitation. Clinical Effectiveness 197–205
and Evaluation Unit, Royal College of Physicians, 63. deVeber G, Andrew M, Adams C et al (2001) Cerebral
London sinovenous thrombosis in children. N Engl J Med
50. Roach ES, Golomb MR, Adams R et al (2008) Man- 345(6):417–423
agement of stroke in infants and children: a scientific 64. Jordan LC, Rafay MF, Smith SE et al (2010)
statement from a Special Writing Group of the Amer- Antithrombotic treatment in neonatal cerebral
ican Heart Association Stroke Council and the Council sinovenous thrombosis: results of the International
on Cardiovascular Disease in the Young. Stroke Pediatric Stroke Study. J Pediatr 156(5):704–710,
39(9):2644–2691 710.e1–710.e2
51. Amlie-Lefond C, deVeber G, Chan AK et al (2009) Use 65. Shroff M, deVeber G (2003) Sinovenous
of alteplase in childhood arterial ischaemic stroke: a thrombosis in children. Neuroimaging Clin N Am
multicentre, observational, cohort study. Lancet Neurol 13(1):115–138
8(6):530–536 66. Miller E, Daneman A, Doria AS et al (2012) Color
52. Ellis MJ, Amlie-Lefond C, Orbach DB (2012) Doppler US of normal cerebral venous sinuses in neo-
Endovascular therapy in children with acute ischemic nates: a comparison with MR venography. Pediatr
stroke: review and recommendations. Neurology Radiol 42(9):1070–1079
79(13 Suppl 1):S158–S164 67. Bracken J, Barnacle A, Ditchfield M (2013) Potential
53. Felker MV, Zimmer JA, Golomb MR (2010) Failure of pitfalls in imaging of paediatric cerebral sinovenous
a clot retrieval device in an adolescent stroke patient. thrombosis. Pediatr Radiol 43(2):219–231
Pediatr Neurol 43(6):435–438 68. Moharir MD, Shroff M, Stephens D (2010) Anticoag-
54. Lo WD (2011) Childhood hemorrhagic stroke: an ulants in pediatric cerebral sinovenous thrombosis: a
important but understudied problem. J Child Neurol safety and outcome study. Ann Neurol 67(5):590–599
26(9):1174–1185 69. Moharir MD, Shroff M, Pontigon AM et al (2011) A
55. Beslow LA, Ichord RN, Gindville MC et al (2014) prospective outcome study of neonatal cerebral
Pediatric intracerebral hemorrhage score: a simple sinovenous thrombosis. J Child Neurol 26(9):1137–1144
grading scale for intracerebral hemorrhage in children. 70. Ganesan V (2013) Outcome and rehabilitation after
Stroke 45(1):66–70 childhood stroke. Handb Clin Neurol 112:1079–1083
56. Westmacott R, MacGregor D, Askalan R et al (2009) 71. Murias K, Brooks B, Kirton A et al (2014) A review of
Late emergence of cognitive deficits after unilateral cognitive outcomes in children following perinatal
neonatal stroke. Stroke 40(6):2012–2019 stroke. Dev Neuropsychol 39(2):131–157
57. Kirton A, deVeber G, Pontigon AM et al (2008) Pre- 72. Kirton A, Shroff M, Visvanathan T et al (2007) Quan-
sumed perinatal ischemic stroke: vascular classifica- tified corticospinal tract diffusion restriction predicts
tion predicts outcomes. Ann Neurol 63(4):436–443 neonatal stroke outcome. Stroke 38(3):974–980
58. Osmond E, Billetop A, Jary S et al (2014) Neonatal 73. van der Aa NE, Leemans A, Northington FJ
seizures: magnetic resonance imaging adds value in the et al (2011) Does diffusion tensor imaging-based
diagnosis and prediction of neurodisability. Acta tractography at 3 months of age contribute to the pre-
Paediatr 103:820–826 diction of motor outcome after perinatal arterial ische-
59. Domi T, DeVeber G, Shroff M et al (2009) mic stroke? Stroke 42(12):3410–3414
Corticospinal tract pre-wallerian degeneration: a 74. Beslow LA, Kasner SE, Smith SE et al (2012) Concur-
novel outcome predictor for pediatric stroke on acute rent validity and reliability of retrospective scoring of
MRI. Stroke 40(3):780–787 the Pediatric National Institutes of Health Stroke Scale.
60. Van der AN, Benders M, Groenendaal F et al (2014) Stroke 43(2):341–345
Neonatal stroke: a review of the current evidence on 75. Mah S, deVeber G, Wei XC et al (2013) Cerebellar
epidemiology, pathogenesis, diagnostics and therapeu- atrophy in childhood arterial ischemic stroke: acute
tic options. Acta Paediatr 103(4):356–364 diffusion MRI biomarkers. Stroke 44(9):2468–2474
Congenital Vascular Syndromes and
Diseases 43
Sarah Milla, Jennifer Vaughn, and Nilesh K. Desai

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1041 There are several neurovascular diseases and
syndromes that are present at birth and may
Moyamoya Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1042
present in the pediatric age range. This chapter
PHACE Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1043 will describe the most common congenital
AVM/AVF Syndromes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1047 neurovascular entities and help to classify
Heriditary Hemorrhagic Telangiectasia . . . . . . . . . . . . 1047 them by dominant etiology (arterial or venous
Wyburn-Mason . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1049 abnormality) when known. The following enti-
Vein of Galen Malformation . . . . . . . . . . . . . . . . . . . . . . . 1050
Sturge-Weber Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . 1051
ties will be discussed: moyamoya, PHACE
syndrome, arteriovenous malformations and
Venous Malformations . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1054
associated syndromes, Sturge-Weber syn-
Developmental Venous Anomalies . . . . . . . . . . . . . . . . . 1054
drome, venous malformations, and mitochon-
Cavernous Malformations . . . . . . . . . . . . . . . . . . . . . . . . 1055 drial encephalopathy with lactic acidosis and
Mitochondrial Myopathy, Encephalopathy, stroke-like symptoms (MELAS).
Lactic Acidosis, and Stroke-Like Symptoms
(MELAS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1057
Keywords
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1059 Pediatric stroke • Congenital neurovascular
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1059 syndromes • Moyamoya • PHACE • Sturge
Weber • MELAS

Introduction
S. Milla (*)
Department of Radiology, Emory University School of
Medicine Egleston Hospital, Children’s Healthcare of Several neurovascular abnormalities have an
Atlanta, Atlanta, GA, USA underlying congenital or genetic etiology. Abnor-
e-mail: sarahsarvismilla@gmail.com malities during embryology and genetic predispo-
J. Vaughn sitions may cause significant neurovascular
Department of Radiology, Boston Children’s Hospital, abnormalities such as hemorrhage and stroke.
Boston, MA, USA
Patients may present asymptomatically for
e-mail: Jennifer.Vaughn@childrens.harvard.edu
screening, such as a patient with a facial heman-
N.K. Desai
gioma, while others present acutely after stroke or
Department of Radiology, Texas Children’s Hospital,
Baylor College of Medicine, Houston, TX, USA hemorrhage, such as MCA territory infarct in a
e-mail: ndesai26@gmail.com patient with moyamoya disease or rupture of an
# Springer Science+Business Media New York 2016 1041
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_33
1042 S. Milla et al.

arteriovenous malformation. When encountering


imaging being performed in infants and children
with neurologic symptoms, even as routine as
headache, knowledge of patient history or under-
lying syndromes, as well as the imaging evalua-
tion of the vasculature, parenchyma and
leptomeninges are important in making the appro-
priate and timely diagnosis of a neurovascular
disorder.

Moyamoya Disease

Background: Moyamoya arteriopathy manifests


as an acquired narrowing of the intracranial arter-
ies; typically involving the anterior circulation to
include the supraclinoid internal carotid arteries
(ICAs), the middle cerebral arteries (MCAs), and
the anterior cerebral arteries (ACAs). This
narrowing is a gradual process, which typically Fig. 1 “Ivy sign” within the leptomeninges on T2 FLAIR
results in enlargement and increasing number of (thin arrows) and white matter changes related to silent
small basilar and perforating arteries to create ischemia often seen in watershed areas (thick arrows)
collateral vasculature. Early diagnosis of
moyamoya arteriopathy is critical for preservation hyperintensity along the leptomeninges on T2
of neurologic function. FLAIR imaging typically in the MCA territory,
Moyamoya disease is defined as an idiopathic also known as the “ivy sign” (Fig. 1). This should
bilateral arteriopathy in the absence of a systemic be distinguished from the occasionally seen inci-
disorder. This term is distinguished from dental diffuse symmetric leptomeningeal
moyamoya syndrome, which is used when the hyperintensity related to a variety of causes, such
arteriopathy is found in the presence of a known as hyperoxygenation and propofol administration
disease, syndrome or unilateral arteriopathy. in sedated children. Another subtle sign is the loss
There are many pathologies and syndromes that of the normal flow void and decreased caliber of
predispose patients to developing arteriopathies. the distal ICAs and MCAs, as seen best on the T2
Most commonly encountered predispositions weighted images (Fig. 2). Often the T2 images or
include sickle cell anemia, Down’s syndrome, thin section T1 postcontrast images will show
neurofibromatosis type 1, and previous radiation multiple tiny basilar collateral vessels especially
to the area (e.g., radiation therapy for brain along the sylvian fissures, suprasellar cistern and
tumor). Less common associations include the perimesencephalic cistern. Other findings seen
Alagille syndrome, hyperthyroidism, congenital on conventional MR imaging are T2/FLAIR
cardiac anomalies, cervicofacial hemangiomas hyperintensities suggesting foci of gliosis in the
(see section “PHACE Syndrome”), and renal subcortical or deep white matter, particularly in
artery stenosis. Certain prothrombotic genes the ACA/MCA watershed distribution, consistent
have also been described as risk factors. with ischemic changes, often referred to as clini-
Imaging: Early findings of moyamoya cally silent infarcts (CSI) (Fig. 3). If moyamoya
arteriopathy can be subtle, so a heightened aware- arteriopathy is not detected and treated early, large
ness of these subtle findings on conventional MR territorial infarctions can occur.
imaging is critical. A subtle finding concerning for On MRA, narrowing of the distal ICAs and/or
moyamoya arteriopathy is curvilinear the proximal MCA/ACAs can be seen (Fig. 4).
43 Congenital Vascular Syndromes and Diseases 1043

Fig. 2 An 8-year-old with headaches. Decreased visuali-


zation of normal MCA flow voids on T2 (arrows)
Fig. 4 Time of flight MRA, anterior projection MIP, in a
10-year-old with moyamoya demonstrates the absence of
flow in MCAs (thin arrows) and multiple tiny collaterals
(thick arrows)

formation causing thin wispy densities to appear


like strands of smoke (Fig. 5).
TREATMENT: The treatment for moyamoya
is a revascularization procedure. Many centers are
utilizing indirect or direct techniques to create
collaterals or reestablish arterial blood supply to
the cerebral areas at risk for infarct. A current com-
mon technique is an indirect revascularization,
encephalo-duro-arterio-synangiosis (EDAS), which
can be combined with other techniques such as pial
synangiosis, dural inversion, or split dural tech-
niques in order to optimize collateral formation
(Fig. 6). Some centers may include perfusion
imaging techniques to determine presurgical and
postsurgical appearances.

Fig. 3 Same patient as Fig. 2. Eight-year-old with head- PHACE Syndrome


aches. T2 FLAIR image demonstrates a focus of chronic
white matter ischemia in the left ACA/MCA watershed
territory (arrow) The presence of an infantile hemangioma in the
head and neck region, particularly if in a segmen-
tal distribution, should raise the clinical concern
MRA may show basilar collateral formation. for associated intracranial and vascular abnormal-
Digital subtraction angiography will show these ities. The acronym PHACE (or PHACES) repre-
findings well, demonstrating the characteristically sents the spectrum of findings in this rare
described “puff of smoke” as the basilar collateral neurocutaneous condition.
1044 S. Milla et al.

Fig. 5 Eleven year old with new stroke found to have (arrowhead). (b). Follow-up 2 years later demonstrates
moyamoya. (a). At initial diagnosis, right ICA injection complete occlusion of MCA with formation of basilar
(lateral) projection, showing narrowing of MCA collaterals, aka “moyamoya vessels” (arrows)

Fig. 6 Early (a) and delayed (b) images from ECA injection (AP projection). 13-year-old with moyamoya, status post
revascularization with pial synangiosis using right superficial temporal artery (arrow)
43 Congenital Vascular Syndromes and Diseases 1045

Posterior fossa/intracranial abnormalities


Hemangioma (cervicofacial)
Arterial cerebrovascular abnormalities
Cardiac abnormalities and aortic coarctation
Eye abnormalities

PHACE association is listed in Online Mende-


lian Inheritance in Man (OMIM 606519) and may
be termed PHACES when referring to the associ-
ation with the involvement of Sternal cleft or
supraumbilical raphe abnormalities.
Patients with hemangiomas of the head or neck
often undergo evaluation for PHACE syndrome
with imaging examinations. It is important to
understand the diagnostic imaging criteria for
making the clinical diagnosis. MRI of the brain
with and without contrast to evaluate for structural
abnormalities as well as optimized MRA of the
aorta, neck and head with and without contrast are Fig. 7 Axial T2-weighted image in child with right facial
important for accurate determination of arterial hemangioma demonstrates ipsilateral cerebellar hypopla-
abnormalities commonly seen in PHACE sia, enlarged posterior fossa, and absence of ipsilateral
patients. internal carotid artery (arrow)

Posterior fossa/intracranial abnormalities – A


major criterion for PHACE syndrome is poste-
rior fossa abnormalities. This is most typified
by cerebellar hypoplasia. This may manifest as
unilateral cerebellar hemispheric hypoplasia,
usually ipsilateral to the cervicofacial heman-
gioma, however the hypoplasia may be bilat-
eral or involving the cerebellar vermis (Fig. 7).
Intracranial hemangiomas have typical imag-
ing appearances for infantile hemangiomas and
are considered minor criteria (Fig. 8). Other
structural anomalies, in particular midline and
neuronal migration abnormalities, are consid-
ered minor criteria.
Hemangioma – The infantile hemangioma is the
most common tumor in infancy and typically
presents within the first few weeks of life, has a Fig. 8 Three-month-old infant with bilateral facial hem-
angiomas with right posterior fossa hemangiomas, largest
rapid proliferative phase within the first year of
at the right porus acousticus (arrow)
life, then begins spontaneous regression. As
defined in a consensus statement published in
2009, PHACE syndrome requires a facial hem- two minor criteria or may have no hemangi-
angioma >5 cm (plus one major or two minor oma if two major criteria are present. Imaging
criteria). Possible PHACE syndrome may have features of hemangioma include significant T2
only one minor criterion or may include a hyperintensity, presence of flow voids, and
chest/neck hemangioma with one major or early arterial enhancement with persistent
1046 S. Milla et al.

Fig. 9 Six-month-old girl with bilateral facial masses. (a). homogeneous enhancement of the masses is seen on coro-
Coronal T2 fat sat image demonstrates hyperintense bilat- nal T1 fat saturated post contrast image
eral parotid masses with internal flow voids. (b). Intense

blood pool enhancement (Fig. 9). During the


proliferative phase, lesions are homogeneous
in enhancement and well defined. The presence
of an infantile cutaneous lesion with similar
intense enhancement but with infiltrative or
aggressive imaging features should question
the diagnosis of hemangioma and raise con-
cern for a kaposiform hemangioendothelioma.
Arterial cerebrovascular abnormalities –
Craniocervical arterial abnormalities are
major criteria in PHACE syndrome. Common
abnormalities include congenital absence,
excessive tortuosity, looping, kinking,
dolichoectasia, aneurysm or stenosis of a por-
tion of the carotid, vertebrobasilar, or intrace-
rebral vasculature (Fig. 10). Moyamoya
vasculopathy has also been reported. Persistent
embryonic arterial vasculature is also common,
with persistent trigeminal artery being a major
criterion for PHACE. While arterial strokes
have been reported, they are rare but reflect a Fig. 10 Axial image from noncontrast time of flight MRA
significant possible sequela of the syndrome. shows significant tortuosity of the left ICA
Cardiac abnormalities and aortic coarctation –
Aortic arch anomalies are a major criterion,
with dysplasia and coarctation being the classic presence of a right-sided aortic arch are minor
appearance (Fig. 11). Aberrant subclavian ori- criteria.
gin and aortic aneurysm are also considered Eye abnormalities – Posterior segment abnormal-
major criteria. Ventricular septal defects and ities are major criteria, specifically persistent
43 Congenital Vascular Syndromes and Diseases 1047

are typically assessed by individual case review.


Recommendations for routine follow-up imaging
relate to severity of arteriopathy and concern for
progression of disease and potential need for
treatment.

AVM/AVF Syndromes

In reference to the terminology of vascular


malformations, reference to ISSVA classification
is recommended. In general, arteriovenous
malformations are considered congenital disor-
ders, with underlying etiology of the malforma-
tion due to abnormal developmental
communications of arteries and veins. Classifica-
tion and discussion of AVMs and AVFs are
covered more extensively in Chapter “▶ Brain
Arteriovenous Malformations”.
Many children may have CNS abnormalities in
association with their syndrome/disease or as
reportable variant anomalies. The syndromes
Fig. 11 3D MRA reconstruction (posterior view) demon-
strating aortic dysplasia, kinking, and coarctation in an
below are highlighted because of their primary
infant with PHACES neurovascular manifestation.

hyperplastic primary vitreous (PHPV), retinal Heriditary Hemorrhagic Telangiectasia


vascular anomalies, morning glory disc anom-
aly, optic nerve hypoplasia, peripapillary Heriditary hemorrhagic telangiectasia (HHT),
staphyloma, and colobomas. Anterior segment also known as Rendu-Osler-Weber or Osler-
abnormalities are minor criteria, specifically Weber-Rendu disease, is an autosomal dominant
cataract, anterior coloboma, sclerocornea, and condition that manifests as mucocutaneous telan-
microphthalmia. giectasias and visceral arteriovenous
malformations. Central nervous system abnormal-
ities in HHT are less common than involvement of
TREATMENT: Multiple therapeutic options the body. Arteriovenous malformations and cav-
can be utilized for the management of infantile ernous malformations are the most common CNS
hemangiomas, including steroids, surgical resec- manifestations (Fig. 12). Dural arteriovenous fis-
tion, laser treatments, and most recently the use of tulas (AVFs) may be present. Commonly, CNS
beta blockers, in particular propranolol. Heman- findings may relate to sequela of systemic
gioma therapy may be related to size and location AVMs, particularly embolic strokes and abscesses
of the mass. Additional treatments are related to (Fig. 13).
the individual patient’s manifestations of PHACE Gene mutations have been identified, allowing
syndrome, particularly cardiovascular abnormali- subclassification of HHT. The most frequent
ties. While the cerebrovascular abnormalities are mutation is HHT type 1 (HHT1) caused by muta-
well-defined in the literature, the long-term course tion in ENG gene (chromosome 9q34). HHT2 has
and necessity for treatment and management of been mapped to ALK1 gene (12q), HHT3 (5q31),
intracranial arteriopathies are being evaluated and AND HHT4 (7p14), and juvenile polyposis/HHT
1048 S. Milla et al.

Fig. 12 Small AVM in a child with HHT. (a). Axial SWI showing the right frontal AVM (white arrow). (d).
shows blooming artifact in right frontal lobe. (b). Post Draining vein of AVM seen on subsequent image (black
contrast T1 weighted image shows associated focal arrow)
enhancement. (c). Arterial phase injection of right ICA

syndrome has been linked to mutation in SMAD4 strokes or abscesses from pulmonary AVMs,
gene (18q21). which are a more typical manifestation. Cerebral
HHT patients often present in childhood, with AVMs are less common, but are more typical in
clinically significant epistaxis (most common), ENG mutations than other known mutations.
evaluation of mucosal or skin telangiectasias, GI Imaging for HHT patients typically includes
bleeding, or visceral angiodysplasias (Fig. 14). workup and screening with MRI and MRA. Addi-
Screening for cerebral AVMs is often performed tion of susceptibility weighted imaging (SWI) or
in patients with genetic or clinical diagnosis of gradient echo sequences will maximize sensitivity
HHT. Neurologic symptoms are occasionally the to blood products. CTA is a useful tool, however,
presentation of HHT, typically from embolic in the era of limiting pediatric exposure to
43 Congenital Vascular Syndromes and Diseases 1049

Fig. 15 Left vertebral injection in a teenage HHT patient


Fig. 13 DWI shows a punctate embolic stroke in a teen- showing a left thalamic AVM supplied by a PCA branch
ager with HHT (arrow). This was not identified on conventional
MRI/MRA

ionizing radiation (As Low As Reasonably


Achievable- ALARA principle), the use of MRA
has increased. There are occasions when small
AVMs are not well seen on MRA and may only
be seen on digital subtraction angiography
(Fig. 15), which is still considered the gold stan-
dard for diagnosis and also allows potential for
treatment.

Wyburn-Mason

Wyburn-Mason syndrome (also known as


Bonnet-Dechaume-Blanc syndrome or retinoen-
cephalofacial angiomatosis) is classically
described as the presence of an AVM of the visual
pathway (typically retinal or orbital AVM), ipsi-
lateral midbrain AVM and often an ipsilateral
facial angioma. Wyburn-Mason studied patients
with retinal AVMs and found a strong association
with concomitant intracranial AVMs. The current
Fig. 14 Same patient as Fig. 13. Teenager with HHT.
classification does not necessitate all three find-
Coronal CT image demonstrates right-sided pulmonary
AVM and angiodysplasia within the spleen. Hemorrhagic ings of the classical description. The embryology
fluid is seen in the abdomen is thought to relate to a fetal abnormality of the
1050 S. Milla et al.

primitive mesoderm before 7 weeks gestational the arterial feeders and type of arterial to venous
age causing persistence of early fetal vascular communication. Type I–III are AV fistulas (Type
tissues shared by the developing optic cup and I: Small pure cisternal fistula between pericallosal
anterior neural tube. Abnormal vascular develop- arteries or posterior cerebral artery and vein; Type
ment leads to anomalous communications II: multiple fistulas between thalamoperforating
between arteries and veins, thus AVM formation. vessels and vein; Type III: high flow, mixed type
While midbrain AVMs are classically described, I and II) and Type IV being a true arteriovenous
hypothalamic/thalamic and basal ganglia AVMs malformation with a nidus and dominant draining
are also common. The ipsilateral facial angioma is vein. The Lasjaunias classification describes cho-
only seen in 30–50 % of patients classified with roidal and mural subtypes. The choroidal classifi-
Wyburn-Mason. cation accounts for the dominant percentage of
Initial presentation may be related to ocular cases. Choroidal type has numerous feeding arter-
and orbital involvement, including decreased ies including choroidal, pericallosal, and
visual acuity, proptosis, afferent pupillary defect, thalamoperforating arteries, which communicate
or pale optic disk. Retinal AVMs are the most with the anterior aspect of the embryonic median
common orbital finding in published cases and prosencephalic vein of Markowski. These patients
reportedly are more stable than intracranial typically present as neonates with shunt physiol-
AVMs. Neurologic symptoms may be mild to ogy and congestive heart failure because of
severe, ranging from headaches and retroorbital high flow dynamics without outflow restriction.
pain to hemiparesis and seizures. Treatment In contradistinction, the mural subtype typically
options may be reduced to embolization given has fewer arterial feeders and is associated with
location of central CNS AVMs. outflow restriction associated with stenosis of
dural sinuses and/or at the level of the jugular
foramen. If not prenatally diagnosed,
Vein of Galen Malformation these patients can present later in infancy, often
with hydrocephalus, developmental delay, or
The vein of Galen malformation (VOGM) forms seizures. Hypoxic ischemic injury to brain
in early fetal life and involves abnormal arterial parenchyma can occur due to shunting of
communication to the embryonic prosencephalic oxygenated blood away from tissue into venous
vein of Markowski, which is intimately associated drainage.
with the development of the vein of Galen, thus Due to prevalence of fetal sonography and fetal
the naming of the disorder. Embryologic study MRI, many VOGM cases are prenatally diag-
correlates this time period of choroid and mid- nosed. Sonographic imaging demonstrates a
brain arterial development with the prosence- hypoechoic rounded structure in the expected
phalic vein of Markowski, explaining the typical location of the vein of Galen which demonstrates
feeding arterial supply involving the anterior cere- avid color flow on Doppler imaging, confirming a
bral artery branches, anterior and posterior cho- dilated vascular structure. Waveform interroga-
roidal arteries (ACHA and PCHA), and the dorsal tion shows a dominant venous waveform, which
midbrain arterial plexus. The timing of early may be arterialized. Sagittal midline Doppler
embryologic venous development also correlates imaging typically demonstrates the varix and
to early fetal life, as most patients with VOGM often the associated persistant falcine sinus
will have drainage from the dilated vein into a (Fig. 16). Fetal MRI better demonstrates the arte-
persistent falcine sinus, with absence of or a rial supply and also better determines parenchy-
diminutive straight sinus. mal damage that may be present (Fig. 17).
Historically, two classifications have been uti- Postnatal imaging with sonography and MRI
lized to describe the characteristics of individual with MRA/MRV are routinely performed. Direct
anatomic arterial to venous connections. The angiography and interventional treatment are typ-
Yasargil classification is based on the location of ically necessary in VOGM, occasionally
43 Congenital Vascular Syndromes and Diseases 1051

Fig. 16 (a) Sagittal


Doppler on fetal ultrasound
and (b) sagittal and (c) axial
fetal brain MRI shows large
varix in the expected
location of the vein of Galen

Fig. 17 Postnatal MRI in newborn with vein of Galen malformation showing (a) varix, (b) arterial feeders, and (c) MRV
showing persistent falcine sinus (arrow)

necessitating urgent intervention in the neonatal Sturge-Weber Syndrome


period in the “choroidal” type, if congestive heart
failure and shunt physiology do not respond to Sturge-Weber syndrome (SWS), also known as
medical management. encephalotrigeminal angiomatosis, is a
1052 S. Milla et al.

neurocutaneous syndrome classically involving a location, larger areas of involvement, and/or bilat-
facial port-wine stain (PWS, also known as nevus eral PWS have suggested higher risks for SWS,
flammeus) associated with ipsilateral reportedly from 20 % to 70 % depending on the
leptomeningeal angiomatosis and often ipsilateral study population. Most patients with SWS will
glaucoma from ocular choroid involvement. This have infantile spasms which progress to seizure
classic triad is considered Type I utilizing the disorder. Between 75 % and 100 % of classic
Roach Scale, with Type II being facial PWS and SWS patients will have seizures, with more exten-
often glaucoma without CNS involvement. Type sive and bilateral disease portending a worse
III consists of intracranial leptomeningeal prognosis. Developmental delay and migraine-
angiomatosis without facial or orbital involve- like headaches are also very common in SWS
ment. Recent studies suggest SWS and patients. More recently, endocrine problems,
nonsyndromic PWS correlation with a somatic such as central hypothyroidism and growth hor-
mosaic mutation of the Guanine Nucleotide Bind- mone deficiency, have been reported as well.
ing Protein, Q polypeptide (GNAQ) gene on Contrast-enhanced MRI is now the standard of
chromosome 9q21. care study to initially evaluate for the presence of
Intracranial manifestation of Sturge-Weber intracranial abnormalities in SWS. Intracranial
syndrome is thought to be related to the lack of findings reflect the underlying pathophysiology
normal development of cortical draining veins of SWS. The lack of normal cortical veins with
and proliferation of pial capillaries and small the proliferation of leptomeningeal capillaries and
venous channels during early fetal life. This is small veins leads to slow flow and increased
postulated to be embryologic in etiology due to enhancement in the leptomeninges, which is best
similar timing of the development of the ectoderm demonstrated by postcontrast T2 FLAIR and
and the vascular development of the related por- postcontrast T1 techniques (Fig. 18). An enlarged
tion of the neural tube at that time. The term and avidly enhancing choroid plexus is com-
encephalotrigeminal is due to the suggested cor- monly seen within the ipsilateral lateral ventricle
relation of trigeminal nerve distribution of the due to similar vascular malformation (Fig. 19).
facial port-wine stain with intracranial involve- Developmental venous anomalies and dilated
ment. Correlation has been suggested of V1 (oph- medullary veins may be present as well, due to
thalmic branch) facial distribution with ipsilateral underdevelopment of the normal cortical venous
occipital lobe involvement, V2 (maxillary branch) drainage. This lack of normal cortical venous
with parietal lobe involvement, and V3 (mandib- drainage is thought to cause venous stasis and
ular branch) with frontal lobe involvement. How- ischemia to the subcortical white matter and
ever, there has been recent research suggesting within the cortex itself. Over time, these ischemic
that the distribution of the port-wine stain may changes can lead to subcortical and cortical calci-
not be related to trigeminal nerve development fications and parenchymal atrophy. As time pro-
but rather the intrinsic pattern of facial vascular gresses, these changes become more obvious.
development. Multisegment facial port-wine stain Occasionally, involvement of the globe will be
with ipsilateral lobar involvement is commonly visualized on MRI with buphthalmos or choroidal
seen in more extensive intracranial cases. Approx- enhancement. CT studies will demonstrate corti-
imately 10–20 % of patients have bilateral facial cal/subcortical calcification and atrophy in
and intracranial involvement. Occipital-parietal affected areas over time (Fig. 20). These calcifi-
involvement is most common. cations on radiography and subsequently CT have
The diagnosis of a facial port wine stain is been referred to as “gyral” or “tramtrack.” PET
typically made after birth, and appropriately, par- studies will often show areas of reduced FDG
ents are counseled about the possibility of SWS. uptake in affected areas. In dynamic MR perfu-
While a facial PWS is common, reportedly up to sion studies, the involved areas of the brain dem-
1 in 300 patients, approximately 8 % of patients onstrate abnormal perfusion and mean transit
with facial PWS will have SWS. Forehead times thought to be due to impaired venous
43 Congenital Vascular Syndromes and Diseases 1053

Fig. 18 MRI in two infants with Sturge-Weber. (a). portion of the right frontal lobe. (b). Postcontrast FLAIR
Postcontrast T1 weighted image in an infant with bilateral image in an infant with right sided port-wine stain showing
port-wine stains showing extensive abnormal bilateral ipsilateral leptomeningeal enhancement
leptomeningeal enhancement with relative sparing of a

Fig. 19 MRI of an 8-month-old with right-sided port- Postcontrast T1 weighted image shows leptomeningeal
wine stain. (a) Gradient echo and (b) precontrast T1 enhancement (thin arrow) as well as ipsilateral choroid
shows right parietal calcification (thin arrows). (c) plexus enlargement (thick arrow)

drainage. MR spectroscopic findings support the being investigated. In patients with seizures and
theory of ongoing ischemic change in affected focal SWS in a noneloquent area, surgical
areas. resection may be performed. In patients with
Treatment depends on clinical symptomatol- more extensive unilateral lobar SWS, a hemi-
ogy, location, and extent of disease. Treatment spherectomy may be performed early in child-
with antiepileptic medication is important in the hood to allow the contralateral normal
management of patients with seizures. Aspirin has hemisphere to take over functions from the
been suggested to be helpful and its use in SWS is affected hemisphere.
1054 S. Milla et al.

Fig. 20 (a) Soft tissue and (b) bone windows from a CT scan in a 4-year-old with bilateral port-wine stains,
demonstrating cortical and subcortical calcifications and prominent left hemiatrophy

DVAs are typically incidentally noted on imag-


Venous Malformations ing. They can be well seen on contrast-enhanced
studies and have a “Medusa-like” appearance,
Developmental Venous Anomalies with contrast enhancement of the medullary
veins converging into the larger draining vein
Developmental venous anomalies (DVAs) are and subsequently into the superficial or deep
common venous malformations or perhaps ana- venous system (Fig. 21). The imaging appearance
tomic variations of the venous drainage system, is classic and similar on CT and MRI when thin-
reported in up to 2.5 % of the population in slice imaging techniques are utilized. Digital sub-
autopsy study. DVAs consist of a network of traction angiography also has a similar appear-
small medullary veins that converge into a larger ance. On noncontrast studies, DVAs may be
draining vein, which then communicates with the difficult to see on CT, and may be faintly visible
superficial or deep venous system. Although not on MRI, best seen on susceptibility weighted
distinctly known, it is thought that they may form imaging (SWI) sequences. Recent study of pedi-
due to maldevelopment of superficial or deep atric DVAs suggests that up to 11.6 % of DVAs
local veins or possibly relate to localized variation not associated with cavernous malformations had
in capillary venous pressures. They may be seen adjacent T2/FLAIR signal abnormality and 4.1 %
in patients with other vascular anomalies, such as had parenchymal atrophy. These findings adjacent
cerebral cavernous malformations, but are most to DVAs have also been reported in adults and
commonly seen without any other associated con- may be related to the altered hemodynamics found
dition. While DVAs themselves are thought to be adjacent to DVAs or possibly associated venous
benign, cases of venous thrombosis of DVAs with hypertension. As mentioned previously, rarely
infarctions of associated parenchyma have been DVAs may thrombose, and precontrast linear T1
described in patients with inflammatory condi- hyperintensity of a DVA should be concerning for
tions, dehydration, diabetes, and pregnancy. thrombosis. In these cases, diffusion weighted
43 Congenital Vascular Syndromes and Diseases 1055

Fig. 21 (a, b) Two-contrast enhanced T1 images illustrate the “medusa like” appearance of an incidental developmental
venous anomaly (arrows) in the right cerebellar hemisphere of a 19-month-old female

Fig. 22 (a) Coronal SWI and (b) coronal postcontrast T1 demonstrate heterogeneous appearance of a left frontal
cavernous malformation. (c) Sagittal postcontrast T1 illustrates associated developmental venous anomaly (arrow)

imaging should be evaluated for evidence of endothelium, and are prone to hemorrhage.
ischemia. CCMs can be seen in sporadic or familial forms.
A sporadic CCM is typically solitary, small,
and is often associated with an adjacent develop-
Cavernous Malformations mental venous anomaly (DVA) (Fig. 22). Familial
forms (fCCMs) have been linked to autosomal
Cerebral cavernous malformations (CCMs, also dominant heterozygous germline mutations in
known as cavernous angiomas and informally as the CCM1 (approximately 50 % of familial
cavernomas) are abnormal capillary-venous com- cases), CCM2, or CCM3 genes. These proteins
munications, appearing as sinusoidal-shaped are important for control of endothelial permeabil-
blood vessels lined with a single layer of ity and are also important in angiogenesis.
1056 S. Milla et al.

scan that may show acute parenchymal and/or


intraventricular hemorrhage if the CCM has
bled. In this acute scenario, further imaging is
typically performed with CT angiogram to deter-
mine if the hemorrhage is due to an AVM or a
cavernous malformation, as they are the most
common causes of acute hemorrhage in children
in the absence of trauma. In the absence of acute
hemorrhage, noncontrast CT may detect an inci-
dental well-defined blood density lesion with or
without calcification. Imaging finding such as
lack of edema and stability over time mitigate
against acute hemorrhage. CTA may show an
associated developmental venous anomaly with-
out any additional suggestion of abnormal arterial
structures, which may help make the diagnosis.
MRI is the most specific imaging modality to
diagnose CCM with the imaging appearance
depending on the stage of hemorrhage associated
Fig. 23 Sixteen-year-old with CCM3/PDCD10 mutation.
SWI demonstrates numerous cavernous malformations, as
with the lesion, e.g., hyperintensity on T1
evidenced by foci of hemosiderin blooming. Right frontal weighted imaging suggests subacute hemorrhage.
artifact from prior surgeries Gradient echo and susceptibility weighted imag-
ing (SWI) sequences highlight blooming artifacts
Patients with fCCM demonstrate multiple cavern- from hemorrhage which are classic to the diagno-
ous malformations with the number of lesions sis. Similarly, a T2 hypointense peripheral ring
increasing over time. These patients can have around the lesion reflects deposition from hemo-
hundreds of lesions (Fig. 23). It is thought that siderin from remote hemorrhage. Internal lesional
fCCM lesions may not have the same association T2 hypointensity and hyperintensity may come
with DVAs as with sporadic CCM lesions. A from various states of hemoglobin breakdown.
history of craniospinal radiation from CNS Imaging appearance on T1, T2, and FLAIR have
tumor also may increase the likelihood of devel- often been described as “popcorn-like” due to the
oping multiple cavernous malformations. heterogeneous appearance in the central core of
Sporadic CCM patients often present after an the lesion (Fig. 24). FLAIR signal abnormality is
episode of hemorrhage, with an acute neurologic commonly seen surrounding a CCM after acute or
symptom such as seizure or loss of consciousness. subacute hemorrhage, which typically resolves
Occasionally, these patients are diagnosed over time. After contrast administration, many
because of a history of headaches, either poten- CCMs do not demonstrate enhancement, while
tially related to prior hemorrhage or incidentally others may demonstrate punctate areas of
found. fCCM patients may present symptomati- enhancement, possibly due to internal capillary
cally or may have known family history with telangiectasia within the CCM. Enhancement of
lesions found on screening. Cases of de novo an associated DVA is again helpful to make the
mutations of fCCM genes, as well as children of diagnosis of CCM. In patients with fCCM, lesions
asymptomatic carriers, have been documented. If demonstrate typical CCM characteristics, but over
more than one CCM is found in a patient, there is time, new lesions typically appear. Lesions are
approximately an 80 % chance of fCCM gene seen throughout the supra and infratentorial
mutation. brain. Brainstem and spinal lesions are most wor-
As the presentation may be due to acute neu- risome, as they tend to have significant morbidity
rologic event, patients may have a noncontrast CT after hemorrhage. While spinal cavernous
43 Congenital Vascular Syndromes and Diseases 1057

Fig. 24 MRI in 16-year-old with CCM3 mutation. (a) malformations. (b) T2 weighted image demonstrates “pop-
Axial T1 image demonstrates multiple foci of T1 corn” heterogeneity and shape of the cavernous
hyperintensity representing hemorrhage within cavernous malformations

malformations may occur with similar imaging maternal. Manifestation is variable, and clinical
characteristics, they are less common than presentation often includes severe headache, nau-
CCMs, even in fCCM patients. sea/vomiting, and neurologic symptoms (“stroke-
In patients with solitary cavernous like” episodes) such as ataxia, hemianopsia,
malformations, surgical resection is often hemiparesis, and cortical deafness. Presentation
entertained, particularly in the setting of recurrent is often in the second decade of life, and paired
hemorrhage. In fCCM families, surgery is often clinical presentation and imaging appearance
not considered prophylactically due to number of should suggest the diagnosis. Blood and CSF
lesions, but may be performed as indicated after lactate levels are elevated in the acute presenta-
evidence of hemorrhage. tion. Confirmation of the diagnosis with muscle
biopsy, looking for ragged-red fibers, COX nega-
tive fibers, and mitochondrial inclusions with sub-
Mitochondrial Myopathy, sequent genetic testing is routine.
Encephalopathy, Lactic Acidosis, While the term “stroke-like” is utilized, imag-
and Stroke-Like Symptoms (MELAS) ing characteristics are fairly similar to typical
stroke imaging appearance. The biggest differ-
MELAS is a rare mitochondrial disorder, which ence between the appearance in MELAS and clas-
can be caused by mutation in one of many genes, sic stroke findings is the lack of distinct vascular
with approximately 80 % of cases due to mutation territory correlating with the ischemic area. The
in the MT-TL1 gene. Other involved genes may most common locations for ischemia in MELAS
include MT-ND1, MT-ND5, MT-TH, and MT-TV. are parietal and occipital cortex, however any lobe
These mutations impair the ability of mitochon- can be affected and grey matter involvement can
dria to make proteins, use oxygen, and produce be seen. CT may show loss of grey white differ-
energy, yet it is still unclear exactly how these entiation in area of cortical involvement (Fig. 25).
mutations cause strokes. As this relates to mito- Basal ganglia calcification may be seen. In the
chondrial DNA, the inheritance pattern is acute phase, cortical edema on MRI is
1058 S. Milla et al.

Fig. 25 Ten-year-old with seizure. Genetic testing con- hemisphere posteriorly concerning for stroke. (b) Axial
firmed MELAS. (a) Axial CT with hyperdensity FLAIR and (c) DWI demonstrate abnormal signal and
representing abnormal mineralization in globi palladi and reduced diffusion in the left occipital and posterior tempo-
subtle loss of grey white differentiation in the left cerebral ral lobes consistent with acute stroke

Fig. 26 Eight-year-old female with abnormal vision and loss in right temporal and occipital lobes and loss of grey
headache with subacute right temporal occipital and acute white differentiation in left occipital lobe. (b) DWI dem-
left occipital infarcts. (a) Axial T2 weighted image show- onstrated restricted diffusion consistent with acute ische-
ing cortical and white matter hyperintensity with volume mia in left occipital lobe

demonstrated with T2/FLAIR hyperintensity and examinations, there can be evolution of previ-
reduced diffusion noted on DWI, consistent with ously seen findings with normalization of signal
cytotoxic edema (Fig. 26). Some studies report and/or increased cortical T1 hyperintensity with
T2/FLAIR abnormalities with associated development of cortical atrophy. Additionally, on
increased diffusion on DWI, which suggests serial imaging, new areas may develop acute
vasogenic edema in some cases. In serial T2/FLAIR and DWI abnormalities. MRAs in
43 Congenital Vascular Syndromes and Diseases 1059

these patients are normal. MR Spectroscopy • Vein of Galen malformations are typically AV
shows lactate peaks in areas of ischemia, but fistula with complications of hypoxic injury
may also show lactate in areas that may not have due to a steal phenomenon.
any other abnormality on conventional imaging, • Most patients with port-wine stains do not have
including over CSF containing spaces. These intracranial abnormalities; however, close
parenchymal areas that appear normal on conven- attention to the postcontrast T1 and
tional T2/FLAIR and DWI yet have abnormal postcontrast T2 FLAIR sequences are useful
lactate on MRS may indicate areas which will in detecting the pial angiomatosis classically
later develop abnormal MRI findings. This obser- seen in Sturge-Weber syndrome.
vation on MRS and the lack of vascular territories • Cavernous malformations are commonly asso-
corresponding to the ischemic areas should raise ciated with developmental venous abnormali-
the diagnosis of MELAS on initial presentation ties. SWI sequence is optimal in the detection
and guide clinicians to clinical diagnosis by lac- of cavernous malformations.
tate levels, muscle biopsy, and genetic workup. • MELAS is a genetic syndrome that causes
Reports of improvement after treatment with strokes or stroke like symptoms not distinctly
L-arginine, an amino acid involved in endothelial- in typical arterial distributions. Lactate peaks
dependent vascular relaxation, have been in CSF or in normal appearing parenchyma
published. Additional supportive therapies may be helpful in making the diagnosis.
including vitamin regimens, coenzyme CoQ10,
and riboflavin have been utilized; however, no
consensus treatment regimen has currently been References
identified.
1. Metry D et al. (2009) Consensus statement on diagnos-
tic criteria for PHACE syndrome. Pediatrics
124:1447–1456
Summary 2. Hess CP, Fullerton HJ, Metry DW, Drolet BA, Siegel
DH, Auguste KI, Gupta N, Haggstrom AN, Dowd CF,
Frieden IJ, Barkovich AJ (2010) Cervical and intracra-
• Many congenital neurovascular abnormalities nial arterial anomalies in 70 patients with PHACE
present during childhood. syndrome. AJNR Am J Neuroradiol 31(10):
• The spectrum of congenital neurovascular 1980–1986
3. Michelle L, Bayer et al. (2013) Congenital cardiac,
abnormalities can often be categorized into aortic arch, and vascular bed anomalies in PHACE
arterial or venous disorders. syndrome (from the International PHACE Syndrome
• There are known genetic mutations that cause Registry). Am J Cardiol 112(12):1948–1952
many of the congenital neurovascular 4. Dayani PN, Sadun AA (2007) A case report of
Wyburn-Mason syndrome and review of the literature.
syndromes. Neuroradiology 49(5):445–456
• Moyamoya disease is idiopathic, while 5. Guey S, Tournier-Lasserve E, Hervé D, Kossorotoff M
moyamoya syndrome is associated with a (2015) Moyamoya disease and syndromes: from genet-
known underlying disease or syndrome. The ics to clinical management. Appl Clin Genet 8:49–68
6. Beth A, Drolet et al. (2013) Initiation and use of pro-
“ivy sign” or watershed ischemic changes pranolol for infantile hemangioma: report of a consen-
should prompt evaluation of ICA/MCA T2 sus conference. Pediatrics 131(1):128–140
flow voids. 7. Ito H, Mori K, Kagami S (2011) Neuroimaging of
• Infants with hemangiomas of the head or neck stroke-like episodes in MELAS. Brain Dev
33:283–288
should be evaluated for PHACES association 8. Linscott LL, Leach JL, Zhang B, Jones BV (2014)
with MRI/MRA. Brain parenchymal signal abnormalities associated
• Most AVMs and AVFs are sporadic; however, with developmental venous anomalies in children and
there are syndromes that should be considered, young adults. AJNR Am J Neuroradiol 35(8):
1600–1607
particularly in the presence of other 9. Meyers PM, Halbach VV, Barkovich AJ (2012) Anom-
angiodysplasias of other organs. alies of cerebral vasculature: diagnostic and
1060 S. Milla et al.

endovascular considerations. In: Barkovich AJ, 14. Smith ER, Scott RM (2010) Moyamoya: epidemiol-
Raybaud C (eds) Pediatric neuroimaging, 5th edn. ogy, presentation, and diagnosis. Neurosurg Clin N Am
Lippincott Williams & Wilkins, Philadelphia 21(3):543–551
10. Nozaki T, Nosaka S, Miyazaki O, Makidono A, 15. Sudarsanam A, Ardern-Holmes SL (2014) Sturge-
Yamamoto A, Niwa T, Tsutsumi Y, Aida N, Weber syndrome: from the past to the present. Eur J
Masaki H, Saida Y (2013) Syndromes associated with Paediatr Neurol 18(3):257–266
vascular tumors and malformations: a pictorial review. 16. Vanaman MJ, Hervey-Jumper SL, Maher CO (2010)
Radiographics 33(1):175–195 Pediatric and inherited neurovascular diseases.
11. Pascual-Castroviejo I (1978) Vascular and nonvascular Neurosurg Clin N Am 21(3):427–441
intracranial malformations associated with external 17. Waelchli R, Aylett SE, Robinson K, Chong WK,
capillary hemangiomas. Neuroradiology 16:82–84 Martinez AE, Kinsler VA (2014) New vascular
12. Raybaud C (2010) Normal and abnormal embryology classification of port-wine stains: improving
and development of the intracranial vascular system. prediction of Sturge-Weber risk. Br J Dermatol
Neurosurg Clin N Am 21:399–426 171(4):861–867
13. Smith ER (2012) Moyamoya arteriopathy. Curr Treat 18. www.OMIM.org (Online Mendelian Inheritance in
Options Neurol 14(6):549–556 Man)
Part IX
Spine
Spinal Vascular Imaging: Technique
44
Maria Isabel Vargas, Fabrice Bing, Joanna Gariani,
and Jean-Louis Dietemann

Contents Preoperative Vascular Imaging


of Spinal Vascular Tumors and Malformations . . . . 1074
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1064 Abdominal Aortic Aneurysm, Thoracoabdominal
Vascular Anatomy of the Spine . . . . . . . . . . . . . . . . . . 1064 Aortic Aneurysm, and Aortic Dissection:
Arterial Blood Supply . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1064 Does Preoperative Imaging of the SC
Venous Drainage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1066 Blood Supply Present an Interest? . . . . . . . . . . . . . . . . . 1075

Arteriography and CT Angiography Magnetic Resonance Imaging (MRI)


of the Vasculature of the SC: Techniques, and Magnetic Resonance
Results, Advantages, and Disadvantages . . . . . . . . 1067 Angiography (MRA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1078
Arteriography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1067 Clinical Indications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1078
Angio-CT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1069 Technical Protocol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1079
Patterns to Know in Vascular Lesions . . . . . . . . . . . . . . 1080
Spine and Spinal Cord Vascularization Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1083
Imaging: Main Indications . . . . . . . . . . . . . . . . . . . . . . . 1071 Imaging of Spinal Vascular Malformations . . . . . . . . 1086
Spinal Surgery and Percutaneous
Spinal Procedures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1074 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1089
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1089

M.I. Vargas (*)


Department of Neuroradiology, Geneva University
Hospitals, Geneva, Switzerland
e-mail: maria.i.vargas@hcuge.ch
F. Bing • J.-L. Dietemann
Department of Radiology, University Hospital of
Strasbourg, Strasbourg, France
e-mail: fabrice.bing@chru-strasbourg.fr;
jean-louis.dietemann@chru-strasbourg.fr
J. Gariani
Department of Radiology, Geneva University Hospitals,
Geneva, Switzerland
e-mail: joanna.gariani@hcuge.ch

# Springer Science+Business Media New York 2016 1063


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_13
1064 M.I. Vargas et al.

allows the visualization of vessels and the analysis


Abstract
of the spinal cord and other structures such as
In this chapter, the principal imaging tech-
bones, meninges, etc.
niques of vascular pathologies of the spine
Performing vascular MRI techniques at the
and spinal cord are described, including com-
level of spinal structures is more challenging
puted tomography angiography (CTA), mag-
than at the level of the brain due to the small size
netic resonance imaging (MRI), magnetic
of the anatomical structures of the spinal cord and
resonance angiography (MRA), and digital
spinal vessels.
subtraction angiography (DSA) as well as the
Firstly in this chapter, the anatomy of the spinal
technical protocols, the different vascular pat-
vasculature is described. Secondly, different tech-
terns to recognize, and the clinical indications
niques such as DSA, CTA, and MRI are presented
where these techniques can be applied.
with their advantages and disadvantages for the
MRI is the technique of choice for the first-
exploration of the spinal vessels. Thirdly, the main
line approach of vascular lesions and their
indications of vascular spine imaging are discussed
follow-up. It is the only non-radiating tech-
as well as the different imaging patterns and a brief
nique that allows visualization of the spinal
description of the principal vascular lesions.
cord and spinal vessels. It also permits the
Finally, the treatment of spinal vascular
differential diagnosis with other nonvascular
malformations requires imaging of very high
diseases. Currently, DSA is used mainly for
quality for the exact determination of the lesion
therapeutic purposes and for detailed analysis
anatomy, the systematic imaging of the main
particularly in small vascular malformations.
arteries supplying the spinal cord before surgical
Finally, the advantages of each technique will
or endovascular repair of aortic disease, or for
be illustrated with clinical examples.
percutaneous spinal procedures and surgery of
the spine.
Keywords
Spine • Spinal cord • Technical MR • Angio-
MR • CT • Angiography • DTI • Diffusion •
Vascular Anatomy of the Spine
Tumors • Arteriovenous malformations •
Ischemia
Arterial Blood Supply

The blood supply of the spinal cord (SC) is


Introduction formed by longitudinal vessels extending along
the axis of the SC from the foramen magnum to
The visualization of vessels supplying the spinal the conus medullaris. Three longitudinal arteries
cord requires high spatial and temporal resolution are described: one anterior spinal artery running in
imaging. For that reason and due to the complex- the anterior median fissure of the SC and two
ity of the spinal cord vasculature, spinal digital posterior (or posterolateral) spinal arteries,
subtraction angiography (DSA) is still considered located on the posterolateral surface of the
the gold standard for evaluating spinal arteries. SC. The anterior spinal artery supplies the two
However, the improvement in technology of non- anterior thirds of the SC, and the posterior spinal
invasive imaging modalities such as computed artery supplies the posterior third. At the surface
tomography angiography (CTA) and magnetic of the SC, a transversal pial plexus provides a
resonance angiography (MRA) allows examina- communication between these main longitudinal
tion of spinal vessels with good results in the axes. The intrinsic SC vascular system is formed
reported series. by a central system fed by the sulcal arteries
Currently, magnetic resonance imaging (MRI) (centrifugal system) and a peripheral system
is the first-line study for the diagnosis of vascular forming a pial network, giving perforating arteries
anomalies in the spine and spinal cord because it (centripetal system) [1, 2].
44 Spinal Vascular Imaging: Technique 1065

Table 1 Arterial anatomy of the SC at the contact of the SC, to join the anterior and
Name of the artery [2] Vascularization posterior spinal arteries.
Radicular Nerve root, dura mater The junction of the anterior radiculomedullary
(or radiculomeningeal) artery with the ASA forms the characteristic hair-
arteries (0.2–0.8 mm) pin configuration, located on the midline, identi-
Anterior Join the ASA (0.2–0.5 fied on high-resolution DSA or CTA coronal
radiculomedullary arteries mm) and supply the two
(6–8) and the great anterior thirds of the SC reconstruction. The posterior radiculomedullary
radiculomedullary artery artery also gives this hairpin turn, but the junction
(0.5–1.0 mm) with the PSA is off the midline.
Posterior Join the posterior and The metameric supplementation of the ASA
radiculomedullary arteries posterolateral spinal
and PSA is not regular. Cervicothoracic,
(11–16) (0.1–0.4 mm) arteries and supply the
posterior third of the SC midthoracic, and thoracolumbar regions present
anatomic variations of the SC vasculature
(Table 2). At the upper thoracic level, a dominant
radiculomedullary artery, described by von Haller
Along their course, spinal arteries receive con- in 1754, exists most of the time. This artery was
tributions from radiculomedullary arteries, which observed in 86 % of patients with conventional
arise from segmental arteries (intercostal and lum- angiography [4].
bar arteries) in the majority of cases. Segmental The great radiculomedullary artery (GRA) or
arteries originate from the posterior wall of the Adamkiewicz artery, described by Adamkiewicz,
aorta. They can present an ascending portion, originates from the segmental (intercostal or lum-
usually described at the thoracic level, and hence bar) arteries [5]. The course and morphology of
vascularize a higher metamer: in that case, the the GRA are described as stable: in 70 % of cases,
name of the artery is the name of the level the GRA originates on the left side, frequently at
vascularized, not the level of its origin. At the the T8-L1 vertebral level [6]. Adamkiewicz
level of the transverse process, segmental arteries described three anatomical types of the spinal
divide into a ventroparietal branch (which sup- vascular system [5]. Firstly, the ASA presents an
plies the transverse process and the rib) and a uninterrupted course from the cervical portion to
dorso-spinal branch. At the level of the neural the lumbar level, with supplementation coming
foramen, the dorso-spinal branch divides into a from intercostal arteries and the GRA. The second
muscular branch (which supplies the paraspinal system presents the same configuration, but the
muscles) and a radicular artery, dividing into ante- ASA is poorly developed. In the third one, the
rior and posterior branches when entering the ASA is interrupted, mostly at the thoracic level.
spinal canal. At each level, radicular arteries The second and third systems present the highest
(also called radiculomeningeal arteries) supply risk of SC ischemia after clamping radicular arter-
the bony walls of the spinal canal. ies and consequently the GRA. Biglioli
Concerning the radicular supply and the super- et al. reported an anatomic study on 51 adult
ficial SC arteries, Lasjaunias classification differ- cadavers without aortic disease, showing continu-
entiates three types of spinal radicular arteries: ity between the vertebral artery, the anterior spinal
radicular, radiculopial, and radiculomedullary artery, and the GRA [7]. This result supports that
arteries [3]. Thron [2] proposed a different ana- the sacrifice of segmental arteries during surgery
tomical classification of the spinal radicular arter- or endovascular treatment of aortic disease may be
ies (Table 1). Radicular arteries giving branches justified without the risk of SC injury [8, 9]. How-
for the spinal arteries are called radiculomedullary ever, this anatomic continuity may become a dan-
arteries: 6–8 anterior and 11–16 posterior gerous stealing pathway for the SC, explaining
radiculomedullary arteries are usually described. paraplegia occurring after surgery when
The branches of the radiculomedullary artery prolonged cross-clamping of the aorta is
divide into ascending and descending branches performed [7].
1066 M.I. Vargas et al.

Table 2 Main feeders of the SC blood supply at the different spine levels
Anterior feeders (for
ASA) Posterior feeders (for PSA) Main feeder and particularity
Cervical Intracranial section Vertebral artery (Fig. 2) Artery of the cervical enlargement (level
region of the vertebral C5-C6) arising from the vertebral artery
arteries (Fig. 2) PICA Radiculomedullary arteries arising from the
deep and ascending cervical arteries and the
vertebral artery
Variation: anastomoses with the occipital and
ascending pharyngeal artery (external carotid
artery). Artery of the cervical enlargement can
originate from the costocervical and thyrocervical
trunk. Duplication of the anterior spinal artery is
possible
Thoracic Supreme and Two dorsal feeders One radiculomedullary artery (level T4-T5)
region posterior intercostal entering the spine above arising from the intercostal artery
arteries and below the GRA One great anterior radiculomedullary artery
(level T9 to T12 on the left side (75 %)) (Fig. 1)
Variation: two (48 %) or three (7 %) anterior
radiculospinal artery [13]
Discontinuity of the ASA is possible
Lumbar Lumbar arteries Artery of the lumbar enlargement
region Anastomosis: at the level of the conus medullaris,
connection of the anterior and posterior spinal
arteries forming a basket shape anastomotic
network
Variation: duplication of the radiculomedullary
artery
Sacral Lateral sacral artery and iliolumbar artery from the Variation: spinal arteries caudal to the aortic
region internal iliac artery bifurcation (rare)
Medial sacral artery
GRA great radiculomedullary artery, PICA posterior inferior cerebellar artery, ASA anterior spinal artery, PSA posterior
spinal artery

Venous Drainage – The intrinsic veins are divided into sulcal and
radial veins.
Understanding the extensive anastomosing venous – The extrinsic or superficial venous system is
channels draining the SC and the spine is manda- composed of anterior and posterior spinal
tory before a surgical or endovascular approach of veins.
spinal vascular malformations (SVM). In fact, • The anterior median spinal vein (diameter
abnormal visibility of the spinal veins is one of 0.4–1.5 mm) [2] runs with the ASA and
the main signs of arteriovenous fistula and can be continues to the filum terminale (vein of
identified either on CTA or DSA. Dilatation of the the filum terminale).
veins will motivate the radiologist to find the path- • One posterior median, the greatest spinal
ological communication between the artery and the vein, is accompanied by two posterolateral
vein: for that purpose, DSA remains the most sen- veins, located next to the posterolateral spi-
sible imaging modality. nal arteries.
One of the major characteristics of the spinal
venous system is its variability, described as being
greater than that of the arterial system [1]. The The extrinsic network system is in contact with
venous system is composed of an intrinsic and the spinal pia matter. Spinal veins are character-
extrinsic system: ized by a high anatomical variability: longitudinal
44 Spinal Vascular Imaging: Technique 1067

veins can be replaced by a secondary network. The radiculomedullary veins communicate


Spinal veins drain into the anterior (numbers with the epidural venous plexus, into which the
8–20) [10] and posterior (numbers 5–10) [2] intradural veins are draining. Intradural and epi-
radiculomedullary veins that drain into the dural veins are separated by valves at the level of
paravertebral and intervertebral plexuses. These the dura. Groen et al. [12] differentiate three levels
venous plexuses drain in the segmental veins of intercommunicating veins:
draining into the ascending lumbar veins, azygos
system, and the pelvic venous plexuses. At the – The internal vertebral venous plexus, formed
thoracolumbar level, the great anterior radiculo- by intradural and epidural veins, communicat-
medullary vein (GRV; diameter 1.5–2.0 mm) may ing with the intracranial venous system and
be mistaken for the GRA, but the angle of the draining in the external vertebral veins
junction of the GRV with the median vein is – The external vertebral venous plexus, located
described as more obtuse than the hairpin config- around the vertebra
uration of the artery [11] (Fig. 1). – The basivertebral veins, within the vertebra

Arteriography and CT Angiography


of the Vasculature of the SC:
Techniques, Results, Advantages,
and Disadvantages

Arteriography

Technique
Spinal DSA can be performed without general
anesthesia, usually reserved for children and
patients who cannot cooperate or lie flat. The
pain induced by a tumoral vertebral lesion has to
be evaluated during the consultation with the radi-
ologist and the anesthesiologist: in fact, a painful
lesion can motivate the sedation of the patient, as
the patient may present difficulties to remain
immobile during the image acquisition. Most of
the time, local anesthesia with intravenous
midazolam and fentanyl is enough. Drugs are
injected for the comfort of the patients, as they
will be asked to hold their breath during the pro-
cedure to decrease artifacts. After local anesthesia,
a 4 or 5 F sheet introducer is placed in the femoral
Fig. 1 DSA, AP view. Fifty-year-old man, suspicion of
artery. Catheterization of segmental arteries is
arteriovenous fistula. Injection into the L2 left lumbar
artery: opacification at the arterial phase (a) and venous performed from the higher to the lower level,
phase (b). (a) The great radiculomedullary artery (GRA) mostly with a Cobra C2 (Terumo) or a Simmons
(double arrow), its hairpin turn (star), and the anterior 1 (Terumo) catheter. When the ostium of a seg-
spinal artery (ASA) (arrow) are opacified. (b) The great
mental artery is catheterized, manual injection of
radiculomedullary vein (GRV) (double arrow), the hairpin
turn (star), and the spinal vein (arrow) can present the same contrast material is performed. To catheterize the
aspect as the arterial distribution lower ostium, the radiologist has to gently retrieve
1068 M.I. Vargas et al.

Fig. 2 DSA, lateral view (a, b) and AP view (c). Fifty- arrow) comes from V4 at the level of the posterior cere-
five-year-old man presenting a renal tumor cervical spine bellar artery (PICA) (arrow head). The star shows the
metastasis. Injection in the right vertebral artery. The ante- space occupied by the spinal cord, between the ASA and
rior spinal artery (ASA) (black arrow) takes its origin at the the PSA
level of V3, and one posterior spinal artery (PSA) (white

the catheter without turning it until he gets a new the spinal lesion (Table 3). Except for the diagno-
stabilization, considering the ostia of the segmen- sis of arteriovenous fistula, which may require a
tal arteries as ladder rungs. Hence, it is global visualization of the spine and SC blood
recommended to begin the catheterization of seg- supply, the arterial exploration of a spinal lesion
mental arteries from one side before doing the concerns the level of the lesion and one or two
other side. levels above and below.
Bone relief of the spine is essential to get a
good projection of the spinal artery: the spinous Advantages and Disadvantages
process has to project between the pedicles on the DSA presents a higher spatial and temporal reso-
anteroposterior (AP) view. The GRA is well rec- lution than CTA and MRA. Moreover, angiogra-
ognized thanks to its hairpin configuration that phy may be the first step before an endovascular
projects on the midline, at the level of the junction treatment such as embolization of an arteriove-
with the ASA. If the ASA mostly presents as a nous fistula or preoperative embolization of ver-
straight vessel situated on the midline in young tebral tumors. Concerning the vascular disease of
patients (Fig. 3), it can present loops in older the spine, DSA remains the best diagnostic imag-
patients (Fig. 4). The radiologist has to be precise ing tool for the anatomic comprehension and ther-
on the level of the segmental artery giving rise to apeutic planification. Even if new generation CT
the GRA. The posterolateral radiculomedullary scanners can improve the anatomic comprehen-
artery presents a smaller course with a lateral sion of arteriovenous fistula, cases reporting the
projection. During the injection of a segmental feasibility of CT scanner in dynamic assessment
artery, a blush of the vertebra is clearly identified of spinal fistula remain anecdotal [14].
(Fig. 3). Difficulties to catheterize segmental DSA presents the main disadvantage of being
arteries can be explained by an agenesia (Fig. 5) invasive, time consuming, complicated, and con-
or thrombus of the artery in the case of an ather- sequently associated with a risk of SC ischemia
omatous aorta. [15]. Moreover, DSA exposes patients to poten-
Vessels that may supply the spinal arteries have tially high doses of radiation and contrast. The
to be known and explored according to the level of risk of paraplegia after arteriography was
44 Spinal Vascular Imaging: Technique 1069

Fig. 4 DSA, AP view. Seventy-year-old patient, suspicion


of arteriovenous fistula. Catheterization of the left T10
intercostal artery. GRA (double arrow), the hairpin (star),
and the ASA (arrow) are clearly identified. Note the loops
Fig. 3 DSA, AP view. Thirty-five-year-old woman, sus- of the ASA, on the contrary to case described on Fig. 3
picion of medullary arteriovenous fistula. The GRA (dou-
ble arrow) is visible after injection into the left T9
intercostal artery. The hairpin turn (star) and the ASA contrast agents, it can be assumed that modern
(arrow) run on the midline. The intercostal artery presents spinal DSA carries a very low risk of neurologic
a short ascension and divides in a ventral branch and a
and systemic complications [18].
dorso-spinal branch. Enhancement of the vertebra body
(brace) and of the paraspinal muscles (white arrow) is
clearly identified
Angio-CT

estimated to be 1.2 % in a series of 480 patients, Technique


which is significant compared with the 3–5 % Many reports propose protocols of injection and
occurrence of paraplegia after aortic surgery in technical points to visualize the segmental artery
a surgical series [16]. However, these high rates giving rise to the GRA. Many case series describe
of complications are described for patients different times of injection, aortic triggering, and
suffering from dissection or aneurysm of the contrast agents for different aortic diseases.
aorta. Other retrospective case series reported a Detection rates of the GRA are variable, ranging
minimal rate of neurologic complications and from 18 % to 100 % [22–29] (Table 5). This
emphasized the important role that spinal DSA variability may be attributable to the patient’s
still presents for the diagnosis of spinal vascular hemodynamic characteristics, the type of aortic
malformations or preoperative spine surgery disease explored, and technical factors such as
imaging (Table 4) [17, 18]. Hence, with the use imaging delay, volume, and flow of contrast
of new endovascular technologies and nonionic medium. Authors tried to determine the optimal
1070 M.I. Vargas et al.

Fig. 5 DSA, AP view with (a) and without (b) subtrac- arrow) is opacified. This artery presents an angulation and
tion. Thirty-three-year-old woman. Preoperative imaging a hairpin configuration (star) as the ASA, but is localized
for an intercostal tumor. Injection into the left T8 intercos- off the midline (image b). The PSA is visible (arrow)
tal artery. The posterior radiculomedullary artery (double

Table 3 Arteries supplying the spine and potentially the Some authors have evaluated the interest of
SC depending on regions multidetector-row CT angiography with intra-
Upper cervical Vertebral artery arterial contrast injection (IA-CTA) to depict the
spine Ascending cervical artery GRA. Authors reported very high percentages of
Deep cervical artery visualization of the GRA and were able to precise
Occipital arterya the origin of the GRA in almost all cases [29, 32].
Cervicothoracic Vertebral artery Only one study attempted to compare the sensi-
spine Thyrocervical and costocervical tivity of CT angiography and conventional angi-
trunks
ography in the visualization of the GRA before
Supreme intercostal artery
Intercostal arteries
thoracoabdominal aorta repair for aneurysms
Bronchial arterya or dissection [32]. The CT angiography was
Lumbosacral Lumbar arteries performed after IA (IA-CTA) injection of
spine Iliolumbar artery contrast, and a conventional angiography was
Medial sacral artery done just after the CT. It was possible to visualize
Lateral sacral artery the GRA in 90 % with IA-CTA. Moreover, in
a
Artery that may supply less frequently the spine 23 % of the patients, the GRA was visualized
with the IA-CTA but not with conventional
angiography.
New algorithms such as model-based iterative
protocols for the detectability of the GRA. reconstruction (MBIR) have been proposed to
Nakyama et al. proposed a contrast medium injec- improve the quality and spatial resolution
tion using an imaging delay of 18 s after triggering [33]. Nishida et al. [34] reported a series of
and an iodine dose of 720 mg l/kg body weight 34 patients presenting different aortic diseases
[30]. Dual-phase CT angiography with a catheter with an improvement of the detectability of the
inserted into the right atrium has been evaluated GRA in comparison with CT reconstruction algo-
for preoperative visualization of GRA in patients rithms (adaptive statistical iterative reconstruction
with aortic aneurysms [31]. Results were promis- and filtered back projection). This improvement
ing, but the problem of the double dose of radia- of delineation of the ASA has been reported in a
tion and contrast medium remains (Table 5). short series of ten patients [35].
44 Spinal Vascular Imaging: Technique 1071

Table 4 Case series of spinal DSA


Number of Localization of the Complications
Study patient Indication GRA (%) (%)
Forbes 1988 96 AVM (88), tumor (8) NA 3.7 systemic
[19] 2.2 neurologic
Savader 1993 50 Thoracoabdominal aortic aneurysm NA 4.6
[20]
Heinemann 46 Aneurysm and chronic dissection 65 0
1998 [21]
Kieffer 2002 487 Aortic aneurysm 86 0.8 neurologic
[15] 0.4 renal failure
0.6 groin
complication
Williams 2004 151 Aortic aneurysm and dissection 43 0
[14]
Chen 2011 [18] 302 Spinal vascular malformation (most 97.4 1 systemic
common indication) 0.7 groin
complication
Nair 2013 [17] 228 Spinal tumor embolization NA 0

Ou et al. [36] reported a series of 40 children However, CTA involves ionizing radiation,
(mean age, 7.5  5 years) explored with CT angi- and the exploration of the abdominal cavity is
ography for the assessment of the GRA. The GRA associated with a relatively high effective dose
was visualized in 95 % of the children. One dis- of 20 mSv [37]. The larger the abdomen, the
advantage of CT angiography is the artifacts gen- lower the contrast-to-noise ratio. In consequence,
erated by the proximity of bone structures. This it is possible that the high rate of detection of the
problem may be encountered and overcome with GRA, mostly reported in the Japanese population,
MRI because the bone shows no signal. is not the same with Caucasian patients.
It is difficult to find a dedicated timing for the
Advantages and Disadvantages visualization of the GRA (Table 5). High concen-
CTA presents the advantage of being noninvasive tration of contrast is mandatory, but the difficulty
in comparison with conventional arteriography. is to determine the exact timing of the acquisition,
New generations of scanner allow very fast acqui- as the enhancement of the GRA is visible for
sition, with an excellent spatial resolution, which approximately 10 s. After that very precise tem-
is necessary for the visualization of the vessels of poral window, the image is polluted by venous
the SC. Moreover, rates of detection of the GRA drainage, which can be mistaken for the SC arte-
reported are extremely high, even if there is a rial supply. This drawback is more obvious in the
publication bias and no comparison with arteriog- case of a spinal vascular malformation.
raphy most of the time. For aortic disease, CTA
allows a precise visualization of an aneurysm or a
dissection (extent, diameter, type of aneurysm, or
dissection). CTA is logistically more accessible Spine and Spinal Cord Vascularization
than MRI in most institutions. Moreover, the Imaging: Main Indications
availability of the software Osirix, dedicated to
DICOM image, allows the detection of the GRA The knowledge of the main arteries supplying the
with good agreement in comparison to the stan- SC, in particular the GRA, is one of the major
dard workstation [24]. points of spinal surgical planning. The aim is to
1072

Table 5 Main case series reporting the success rates of detection of the GRA and the technical aspects of injection with CTA
GRA
Aortic luminal visualization Continuity GRA
Study N= Indication MDCT Contrast material Injection attenuation (%) and aorta
Zhao 2009 99 Thoracic aortic disease 16-row Iohexol 120–150 mL; IV 358  62 UH 52 NA
[25] 350 mgI/mL; 4–5 mL/s
Clarençon 30 Aneurysm and dissection 16-row Iodixanol 120 mL; IA NA 90 84
2013 [32] 320 mg/mL; 15 Ml/S;
Nojiri 2007 27 Aneurysm (13) and dissection 4-row Iomeprol 400; iopamidol IA 4.5–7 s after 100 90 for the detection
[29] (14) 370 or iohexol 300; start of inj of the level: 61.5
100 ml;4–5 ml/s (A); 120 ml;7 for the exact
ml/s (D) continuity
Ou 2007 40 Congenital disease of the aorta 16-row Iohexol 300; 1.5 ml/kg; 2.5–3 IV 85 UH 95 95
[36] ml/s
Uotani 2008 32 Aneurysm (21) and dissection 16-row Iopamidol 370; 100 mL;5 IV 140–160 UH IACT:94.1 87.5
[22] (11) mL/s (15) and IVCT:60 55.6
IA (17)
Kudo 2003 19 Liver disease 4-row Iohexol 350; 100 mL;5 mL/s IV NA 68 NA
[28]
Nakayama 80 Aneurysms 64-row Iohexol 540 mg/mL; 6 mL/s IV 150UH 15 s 56.3 NA
2008 [28] 18 s
Iohexol 720 mg/mL; 6 mL/s 21 s
18 s
Boll 2006 100 40 Iomeprol 400 mg/mL;80 mL; IV 150 100 100
[38] 3.5 mL/s
M.I. Vargas et al.
44

Utsunomiya 80 Aneurysms 64 Iomeprol 300 mg/mL;100 mL; IV 250 75 50 (best protocol)


2008 [27] 5 mL/s
350 mg/mL;100 mL; 5 mL/s 80
300 mg/mL;100 mL; 3.5 mL/s 45
350 mg/mL;100 mL; 3.5 mL/s 50
Yoshioka 30 Dissection (12) 16-row Iopamidol 370 mg/mL; IV 200 83 58
2006 [27]a TAA (18) 2 mL/kg; 3.5 mL/s
Yoshioka 30 Dissection (10) 4-row Iopamidol IV 130 80 62.5
2003 [39]a TAA (20) 370 mg/mL;2.5 mL/kg; 3 Ml/s
Nishida 34 TAAA (6) 64 Iopamidol 370; 100 mL; IV 250 90b NA
2014 [34] Follow up TAAA surgical 5 mL/s
grafting (9), endovascular repair
Spinal Vascular Imaging: Technique

(15), chronic dissection (3),


Takayashu (1)
Amako 2012 110 TAA and TAAA 8 and Iopamidol 370; 120 mL; IV (right Two 100 90
[31] 16-row 8 mL/s atrium) acquisitions:
20 and 55 s
after the first
injection
Takase 2002 70 TAA (20) NA Iopamidol 300; 100 mL; IV 85 90 % 31.7
[23] AAA (16) 3.5 mL/s
Dissection (19)
Atherosclerosis (15)
a
MRA and CTA study: the depiction of the GRA is reported to be better with MRA
b
Score of 3 and 4 of the scale of depiction of the GRA proposed by the authors
1073
1074 M.I. Vargas et al.

Fig. 6 DSA, AP view with (a) and without (b) subtraction. Catheterization of the left T6 intercostal artery. A longitudinal
anastomosis coming from T6 supplies the level below (arrow). No left T5 intercostal artery was found for that patient

avoid neurologic complications, such as paraple- There is no interest to systematically search the
gia, anesthesia, loss of bladder and bowel control, GRA at levels far from the operative site. How-
and sexual dysfunction, described after the liga- ever, longitudinal anastomoses allow a new blood
ture of the GRA or the segmental artery giving the supply for the SC: hence, ablation of vertebras and
GRA. Radiculomedullary arteries have to be segmental arteries giving rise to spinal artery is of
clearly identified before an embolization. How- course not necessarily associated with paraplegia.
ever, the interest of searching for a radiculo- Murakami et al. reported their experience in total
medullary artery before endovascular or surgical en bloc spondylectomy associated with an inter-
treatment of aortic disease or even before spine ruption of the GRA. Fifteen cases in which the
surgery continues to be a controversial subject. GRA was described on DSA and ligated did not
Indications of such preoperative imaging are present alteration of their neurologic function.
discussed. They concluded that surgeons are allowed to sac-
rifice up to three pairs of segmental arteries and
that the presence of the GRA at the level of the
Spinal Surgery and Percutaneous surgery is not a contraindication for surgery
Spinal Procedures (Fig. 7) [40].

Concerning the vascular preoperative imaging of


spinal tumors, selective catheterization of seg- Preoperative Vascular Imaging
mental arteries feeding the tumor allows the visu- of Spinal Vascular Tumors
alization of spinal arteries, which can be helpful and Malformations
for surgical planification. In these cases, the aim of
DSA is to identify a radiculomedullary artery at Imaging of Spinal Vascular Tumors
the level concerned by the surgery or before a The interest of arteriography is not only to search
percutaneous treatment (Figs. 6 and 7). Koshino for a radiculomedullary artery but also to perform
et al. showed that the GRA does not specially an embolization in case of hypervascular
originate from big segmental artery and proposed tumors. Even if the embolization of thyroid
that all the segmental arteries have to be preserved carcinoma [41–43] or in general [44] remains
during surgery [6]. controversial, the embolization of metastasis of
44 Spinal Vascular Imaging: Technique 1075

Fig. 7 DSA, AP view with (a) and without (b) subtrac- schwannoma, surgery was canceled, and a cryotherapy
tion. Thirty-three-year-old woman presenting a was performed, taking precautions not to injure the inter-
schwannoma at the level of left T10 (double star). Injection costal artery. Hairpin turn (star) and ASA (arrow) are
into the left T10 intercostal artery. Because of the presence identified. Note the mass effect on the intercostal artery
of the GRA (double arrow) coming at the level of the which is upraised by the lesion

renal cell carcinoma before surgery has been Abdominal Aortic Aneurysm,
reported to be useful [45, 46]. A prospective ran- Thoracoabdominal Aortic Aneurysm,
domized controlled study is warranted to precise and Aortic Dissection: Does
the benefits of preoperative embolization for spi- Preoperative Imaging of the SC
nal tumors. Blood Supply Present an Interest?
Thiex et al. reported no statistical difference in
intraoperative blood loss between partial and Spinal cord ischemia after surgical aortic repair
complete transarterial tumor embolization and endovascular treatment of aortic disease
[47]. Interestingly, the authors reported a poor (Fig. 10) has been largely reported [53, 54]. Para-
correlation between MRI enhancement and angio- plegia and paraparesis rates for descending tho-
graphic vascularity. In consequence, spinal DSA racic aneurysms and thoracoabdominal aneurysm
may bring new information on the tumor vascu- resection are about 1 % and between 5 % and
larity which could motivate the realization of the 10 %, respectively [55]. The incidence of spinal
embolization. The interventional radiologist has injury after treatment with endovascular grafts is
to be sure that the segmental artery providing the between 3 % and 12 % and may be due to the
tumor does not supply a radiculomedullary artery occlusion of the GRA associated with a drastic
before embolization: in that case, it is considered decrease of the residual collateral blood supply
as an absolute contraindication to embolization [56]. This hypothesis is comforted by an article
(Fig. 8) [17, 47–51]. describing more SC ischemia with the number or
It is necessary to explore levels above and the length of aortic stent grafts [57].
below the segmental artery that have to be closed: Aneurysm and dissection have to be distin-
in fact, collateral supplies coming from the rich guished as the repercussion of these different aor-
anastomotic network may explain accidental tic mural diseases on the SC blood supply is
embolization of a radiculomedullary artery different. In fact, the mural thrombus of the aneu-
(Fig. 9) [52]. rysm is associated with an occlusion of the
1076 M.I. Vargas et al.

Fig. 8 DSA, AP view with (a) and without (b) subtrac- Radiograph of resected specimen of T3, T4, and T5. The
tion. Thirty-year-old woman presenting paravertebral radiculomedullary artery has been resected, and the patient
chondroblastoma. Catheterization of the left T4 intercostal did not present postoperative spinal cord ischemia (Image
artery at the level of the lesion (vascular blush, double courtesy of Pr Steib, Service de Chirurgie du Rachis, CHU
star). As a radiculomedullary artery (double arrow) was de Strasbourg)
injected, no preoperative embolization was performed. (c)

segmental arteries, and collateral channels devel- The systematic imaging exploration of the seg-
oped allowing blood supply for the SC. On the mental arteries in patients undergoing
contrary, in the case of dissection, most of the thoracoabdominal repair can be criticized, as
segmental arteries remain patent, and no collateral there is a controversy on the necessity for the
network develops. Hence the surgical or reimplantation of the intercostal and lumbar arter-
endovascular occlusion of the segmental artery ies [58]. Heinmann reported a series of 46 patients
giving the GRA may acutely disrupt the spinal treated for thoracoabdominal aneurysms and
cord vascularization. Williams et al. reported a chronic dissection and precised that preoperative
series of preoperative selective intercostal arteri- selective angiography was helpful in the
ography in 131 patients undergoing treatment for planification of the surgery [21]. When the sur-
the dissection or aneurysm of the aorta. SC ische- geon knows the level of the GRA and the appro-
mia was observed in 4.6 %, and all were observed priate branch is within the aortic segment to be
in patients with dissection [14]. replaced, reimplantation of the segmental artery is
44 Spinal Vascular Imaging: Technique 1077

to the longitudinal anastomoses between segmen-


tal arteries and the collateral circulation of the
thoracic wall allowing the perfusion of the SC
[61]. This fully efficient collateral circulation has
also been demonstrated in a postmortem study of
the spinal blood supply after the occlusion of
radicular arteries [62]. The first arterial system
described by Adamkiewicz presents a low risk of
SC ischemia, as many radiculomedullary arteries
supply an uninterrupted spinal artery.
The interest of preoperative DSA and its rela-
tionship to postoperative neurologic complica-
tions in patients operated for TAAA have been
evaluated. Savader et al. [20] reported a study
with two groups of 50 patients, one with positive
spinal artery and one without spinal artery
identified on DSA. The prevention of spinal
ischemia consists of the maintenance of SC
circulation and spinal protection. For the group
of patients with positive spinal artery, spinal
protection was performed, and the authors report
Fig. 9 DSA, AP view. Thirty-five-year-old woman, sus- no significant difference between the two groups
picion of medullary arteriovenous fistula (same patient as in the occurrence of neurologic complications.
Fig. 3). Injection into the right L1 intercostal artery. A They concluded that DSA reduced the risk of
radiculomedullary artery (double arrow) is opacified
through anastomoses coming from the left T12 intercostal neurologic complications, even if the rate of
artery supplying a spinal artery (arrow). This case illus- morbidity of DSA in that series was very high
trates the rich interconnecting longitudinal extravertebral (4.6 %) [20].
network between segmental arteries Feeding arteries of the SC have been explored
before and after thoracic endovascular aortic
indicated. When the level of the GRA is unknown repair (EVAR). Von Tengg-Kobligk
but with a risk to be included in the ill segment of et al. evaluated the visibility of posterior intercos-
the aorta, more aggressive techniques to preserve tal arteries, dorsal branches, and the GRA with CT
the blood flow in the segmental artery are angiography, before and after EVAR [63]. The
recommended [21]. Nijenhuis [59] insisted on authors showed that the majority of the segmental
the necessity of preoperative visualization of the arteries were not occluded after EVAR, due to
GRA, showing that no decrease in SC function retrograde perfusion. On 17 patients followed,
was observed when the clamped aorta did not one GRA was not visible after EVAR, but no
concern the segmental supplier of the GRA. On neurological events were observed. Amabile
the contrary, a 32% decline in SC function was et al. reported a series of 67 patients with lesions
observed if the supplier of the GRA was located of the descending thoracic aorta treated by stent
inside the cross-clamped area [59]. graft and showed that the length of aorta covered
However, case series showing very low rates of was the only independent predictive factor for SC
SC ischemia without segmental artery ischemia [57]. The authors identified a threshold
reimplantation and with several sacrificed seg- of 205 mm above which the risk of SC ischemia is
mental arteries have been published [60]. More- predictable with a sensitivity of 80 % and a spec-
over, the occlusion of the segmental artery giving ificity of 92.5 %. Delayed SC ischemia has also
rise to the spinal artery is obviously not systemat- been described. Etz et al. reported this rare situa-
ically associated with spinal ischemia: this is due tion, concerning 10 on 858 patients operated for
1078 M.I. Vargas et al.

Fig. 10 Ischemia. VRCT reformatting shows an aortic aneurysm (a); unfortunately, the patient becomes paraplegic after
surgery in rapport with ischemia of the spinal cord appearing as a high signal on T2 (b) with restricted diffusion (c, d)

thoracoabdominal aneurysms, has the conse- interesting results of MRA used for the preopera-
quence of subtle hemodynamic differences tive localization of the GRA in thoracoabdominal
[64]. In those cases, radiculomedullary arteries aortic aneurysm patients: comparing CTA and
may have been injured, but collaterals may have MRA, the authors showed that both techniques
allowed a fragile blood supply of the SC, until the allowed the localization of the GRA but that MRA
blood pressure decreases, revealing the insuffi- was more favorable because of a higher contrast-
ciency of the vascularization. to-noise ratio. Moreover, the quality of the image
In conclusion, these case series comfort the was independent of patient thickness. On con-
idea that systematic CTA or DSA exploration trary, Zao et al. reported that the detection rate of
before surgery or endovascular treatment of aortic the ASA and the Adamkiewicz artery (AKA) with
disease is not necessary. Methods to protect the CTA was influenced by the vertebral mass index
SC from ischemia have been proposed (Table 6). and the contrast-to-noise ratio [25]. Vargas
This emphasizes the theory of many collaterals et al. [65] showed the utility of MRI in the analysis
rather than a single dominant radiculomedullary of vascular lesions (such as cavernomas and
artery supplying the SC [8]. hemangioblastomas), in the surgical planning of
lesions, as the first-line method of analysis of
vascular malformations and for planification of
Magnetic Resonance Imaging (MRI) DSA in patients with vascular malformations.
and Magnetic Resonance Angiography
(MRA)
Clinical Indications
The other noninvasive vascular imaging of the SC
blood supply is magnetic resonance angiography Until the last decade, the visualization of vessels
(MRA). Nijenhuis et al. [37, 59] reported of the spine by MRI was difficult due to their
44 Spinal Vascular Imaging: Technique 1079

Table 6 Etiology of SC ischemia and methods to protect surgery, for example, in the case of the surgery
the spinal cord from ischemia during and after surgery of of aortic aneurysms (Fig. 10), endovascular treat-
the aorta
ment in the case of a thrombus (Fig. 11), spinal
Etiology of SC Coverage of the segmental tumors (Figs. 12 and 13) localized between Th8
ischemia artery with stent graft
and L1, or surgery for scoliosis and/or kyphosis.
Clamping of the aorta and
reperfusion injury with The difficulties that remain in some cases
cytotoxic metabolites include the differentiation between arteries and
released and flushed back in veins, the visualization of small shunts, and the
the spinal arteries visualization of normal AKA and accessory spinal
Clamping of the aorta and
cord arteries.
steal of the blood flow
Clamping of the aorta
In ischemia [67], spinal cord anomalies are
resulting in an increase in visible early only on diffusion imaging, but unfor-
intracranial pressure and a tunately, the implicated vessel is not detectable by
drop in SC perfusion, MRA (Fig. 10).
consequence of a rise in the
perfusion of the brain
On the other hand, the radiographer has to be
Systematic hypotension very familiar with the MR technique because
Embolization timing is crucial.
Internal iliac artery The reformatting of this sequence is time con-
circulation suming but is indispensible for understanding the
Methods to protect the Identification and vascular malformations.
SC from ischemia reimplantation of the
segmental arteries giving the
spinal arteries
Systemic hypothermia Technical Protocol
Adjunctive pharmacologic
therapy The technical protocol recommended is a T2-WI
CSF drainage sagittal (TE 120, TR 3,800, 3.5 mm slice thick-
ness) and axial (TE 120, TR 3,800, 3 mm slice
thickness), T1-WI sagittal (TE 10, TR
600, 3.5 mm thickness), diffusion tensor imaging
small size. Actually, the use of high-field MR in (DTI) with 30 directions, sagittal CISS (construc-
clinical routine imaging allows the realization of tive interference steady state), FIESTA (fast imag-
dynamic MRA and the analysis of different ing employing steady-state acquisition) or 3D T2
malformations. SPACE sequence (sampling perfection with appli-
The real advantage of 3D dynamic vascular cation optimized contrasts using different flip
sequences is the capacity to noninvasively evalu- angle evolutions) (TE 5.9 ms; TR 11.9 ms, slices
ate the shunt location and sometimes feeding 72, slice thickness 700.00 μm), and fat-saturated
arteries and drainage veins which is extremely T1-WI in the sagittal (TE 12, TR 400, 3.5 mm
helpful to guide invasive conventional digital slice thickness) and axial (TE 12, TR 400, 3 mm
angiography. thickness) planes after the administration of con-
This technique is extremely useful for the fine trast in the case of tumors and an angio-MR (three
analysis, characterization, and classification of MRA acquisitions) with late high-resolution
different vascular lesions; currently, we can iden- sequences are added in cases of suspicion of a
tify and analyze normal vessels of the spinal cord dural fistula or an arteriovenous
such as AKA (Fig. 11) as well as pathological malformation [65].
arteries in the case of arteriovenous The MRA sequence consists of three MRA
malformations [66] and dural fistulas. acquisitions based on a 3D contrast-enhanced
The localization of AKA is an essential data for gradient echo sequence sequentially performed
surgical planning of the spine and vascular after a bolus injection of an intravascular contrast
1080 M.I. Vargas et al.

Fig. 11 Visualization of AKA. Patient with extensive VR reformatting (b). The MRI shows no anomaly of the
thrombus of the aorta, renal, and first segment of iliac conus medullaris and hyperintensity of the aorta due to the
arteries very well illustrated by the coronal MPR (a) and thrombus (c). MRA illustrates the AKA (d)

agent. The sequence is fat saturated, and therefore no rectangular FOV, 72 slices, 0.6 mm slice thick-
it is not centric reordered. A test bolus is then ness. The sequence was also performed systemat-
performed in order to determine the arrival time ically at the isocenter.
of the contrast agent in the feeding arteries of the When your machine does not allow vascular
spinal cord, and the acquisition is started ade- sequences, CISS, FIESTA, 3D SPACE, and
quately so that the acquisition of the k-space cen- CUBE sequences permit a better visualization of
ter matches the bolus arrival time. For the the enlarged pial vessels around the spinal cord.
arterial and venous 3D gradient echo MR Finally, MPR reformatting and thin MIP are
sequence, the parameters were TE/TR/flip realized for all acquisitions.
angle = 1.52 ms/3.98 ms/20 , a matrix size
of 384, pixel size = 1.2  1.0  0.9 mm,
FOV = 380 mm, no rectangular FOV, 88 slices, Patterns to Know in Vascular Lesions
0.9 mm slice thickness. The phase encoding direc-
tion is in the craniocaudal direction to avoid flow The following systematic approach is proposed to
and motion artifacts from the ascending facilitate the diagnosis of vascular malformations.
hyperintense aorta projecting on the spinal cord. The following checklist of signs must be used
The sequence is acquired at the isocenter and for all vascular studies of the spinal cord:
requires 1 min for a single phase.
For the high-resolution late enhanced 3D 1. First, centromedullary high signal of the spi-
gradient echo MR sequence, the parameters nal cord on T2-WI: this can be localized in the
were TE/TR/flip angle = 1.89 ms/4.95 ms/20 , medullary conus, thoracic, or thoracolumbar
two averages, a matrix size of 512, pixel and cervical level. The anomaly may vary
size = 0.7  0.6  0.6 mm, FOV = 330 mm, and reflects venous hypertension (Fig. 14).
44 Spinal Vascular Imaging: Technique 1081

Fig. 12 Localization of AKA for presurgical planning heterogeneous on T2 (a) with strong enhancement on T1
of a schwannoma. Note the displacement of the spinal and the AKA is localized below the lesion (b)
cord well demonstrated by DTI (c): The lesion is

2. The presence or absence of vessels: They can veins, the vascular transverse structures to the
be located within or around the spinal cord. extradural space at root level, and possible
3. The presence of blood in the subarachnoid enhancement of the spinal cord reflect a break-
space or a hematoma within the spinal cord. down of the blood-spinal cord barrier [68] and
4. An enhancement of the spinal cord due to a should evoke the diagnosis of a dural fistula
breakdown of the blood-spinal cord barrier in (Fig. 14). The shunt itself between the vein and
cases of venous hypertension or arterial ische- a radicular artery is sometimes difficult to visu-
mia or due to the presence of dilated spinal alize by MRI.
cord vessels, in the extradural space or in the 2. If the MRI shows a ball (nidus) of
paravertebral tissues. intramedullary or intra- and perimedullary loca-
tion, with hyperintensity and the
The analysis of signs is tricky and their combi- enlarged perimedullary vessels which may
nation may cause the following different patterns: be associated with subarachnoid hemorrhage or
a hematoma, the most probable diagnosis is an
1. Centromedullary high signal of the spinal cord arteriovenous malformation of the spinal cord
associated with thin perimedullary corkscrew (Fig. 15). If it is in contact with the bone,
dilated vessels in rapport with dilated pial remodeling may result.
1082 M.I. Vargas et al.

Fig. 13 Hemangioblastoma. MRI shows the vascular the mass very well illustrated by DTI and the enlarged
lesion localized in the spinal cord with a perimedullary vessels around the mass. MRA is used to make a cartogra-
extension. Note the displacement of the spinal cord by phy for presurgical planning

Fig. 14 Dural fistula. Note the high signal of the thoracic spinal cord and the enlarged perimedullary vessels without any
anomaly on the DTI. MRA and axial slices nicely show the dural fistula similar to DSA

3. Another pattern consists of dilated vessels diagnosis is an arteriovenous fistula of the spi-
associated with a hyperintensity of the spinal nal cord. It is important to note that this diag-
cord due to a communication between the pos- nosis by MRI is difficult due to the size of the
terior and anterior spinal artery with the veins shunt (1–2 mm) and can be confounded with a
via direct fistulas, located close to the conus dural fistula. If the arteriovenous fistula of the
medullaris; in this case, the most probable spinal cord is larger, the diagnosis is easier, and
44 Spinal Vascular Imaging: Technique 1083

Fig. 15 Arteriovenous malformation. MR reveals demonstrated in the MRA reconstructions (c),


intramedullary flow voids representing feeding arteries, corresponding to what is seen on DSA (d)
nidus, and draining veins (a, b), note the feeder, nicely

in the axial plane, you can see the vascular The imaging of spinal vascular malformations
structures as pointed by a pencil that embrace will be described in the second part of this chapter
the anterior and posterior spinal cord. including their origins, clinical signs, pathophys-
4. If you observe a rounded intramedullary lesion, iology, treatment, and new imaging techniques in
well defined with heterogeneous central T1 and tumors and vascular malformations.
T2 signal (the signal anomaly will depend on the
stage of hemoglobin degradation) and a periph-
eral hypointense rim on T1 and particularly on T2 Tumors
images, you must think of a cavernoma. Never-
theless, in the case of recent bleeding, MRI pat- Tumors may be divided by their location into
tern can change, and the lesion appears intradural (hemangioblastoma, cavernoma) and
hyperintense on T2 and isointense or hypointense extradural tumors (hemangiomas, angiolipoma).
on T1. The “popcorn” appearance is the most Spinal cavernomas (Fig. 16), the correct
characteristic, and the T2 gradient echo sequences definition of these lesions are “hamartomas”
are essential and show a drop of signal due to a and are not true tumors however are in this
magnetic susceptibility effect. Contrast enhance- section because are angiographically occult
ment is absent or minimal (Fig. 16). and often produce a mass effect. Represent
5. The last pattern regarding spinal vascular 5 % of lesions in adults and 1 % in children
malformations is related to aggressive bone [70–73]. They can be sporadic, familial
hemangioma. MRI shows a bone lesion in the (multiple), or acquired (radiation induced).
vertebral body which may extend to the pedi- Mutations in genes (CCM1, CCM2, and
cles, heterogeneously hypointense on T1, CCM3) have been identified [70, 74–76]. The
hyperintense on T2, and STIR sequences with incidence is about 5 % in adults [70, 72, 77]
remodeling of the walls of the vertebra, asso- and 1 % in children [70, 75] of all spinal
ciated with an important enhancement and epi- vascular malformations [78]. These are more
dural or paravertebral tissue mass. The most common in women than men with a ratio of
common complication is spinal cord 2:1 [79] and become symptomatic between the
compression [69]. third and sixth decade of life.
1084 M.I. Vargas et al.

Fig. 16 Cavernoma. Fusiform lesion of the spinal cord, other lesions in the brain. Cavernomas are visible on mor-
hyperintense at the center with a hypointense rim on T2 phologic sequences and silent in MRA and conventional
without enhancement associated with high signal around angiography; they do not require MRA
the lesion due to a cavernoma with bleeding. Note also

The symptoms can vary from asymptomatic, suspected and has not bled, DSA can be useful
insidious, or sudden, the latter when the lesion with the use flat-panel catheter angiotomography
bleeds. (FPCA): the aim is to characterize “cryptic venous
The most common symptoms are a sensorimo- anomalies” by the analysis of the abnormal spinal
tor deficit, progressive paraparesis, or sudden veins [80].
paraplegia in a young adult. Pattern number 4 describes the imaging char-
The location in order of frequency is thoracic acteristics of cavernoma in MRI (Fig. 16).
(50 %), cervical (40 %), and spinal cord and at the The advances in techniques such as diffusion
medullary cone (10 %) [73]. tensor imaging and tractography can demonstrate
Cavernomas are angiographically silent and the relationship between the vascular malforma-
composed of dilated capillaries without a patho- tion and spinal cord tracts, which can be useful for
logical arterial supply. The scanner shows only surgical planning [81]. A brain MRI should be
calcified cavernomas; plain radiography is unnec- part of the imaging protocol of this type of pathol-
essary. MRI is the reference imaging method and ogy to exclude multiple cavernomas.
may be performed in an emergency setting when Radiation-induced cavernomas may be associ-
the onset of symptoms is sudden, due to cavern- ated with signs of postradiation myelitis with a
ous bleeding. hyperintense signal on T2 which may extend over
Spinal DSA is indicated in case of medullary several vertebral levels.
hemorrhage, which can reveal cavernomas as well Spinal cavernomas are usually intra-axial, but
as glomerular AVM [77]. When a cavernoma is there are reports in the literature that describe an
44 Spinal Vascular Imaging: Technique 1085

epidural location with extension to the paraspinal In hemangiomas that have a fatty involution,
muscles and foraminal nerve roots [82–93]. further investigation is unnecessary. However,
The differential diagnosis of intramedullary aggressive hemangiomas require a thorough
cavernomas is mainly with lesions likely to have review and monitoring, and when they become
a hemorrhagic component such as ependymoma symptomatic, treatment is usually necessary.
and hemorrhagic metastases and more rarely with Hemangiomas have a fairly characteristic
astrocytomas and hemangioblastomas. appearance on radiographs: round, well-defined
Standard treatment may be abstention and fol- lesions with thick vertical trabeculae.
low up by MRI if asymptomatic or surgical resec- In case of doubt, a CT scan with millimetric
tion if symptomatic [70, 73, 94, 95]. slices and high resolution is performed; it is the
Spinal hemangioblastoma (Fig. 13) is a large method of choice for the analysis of bone structure
vascular mass in conventional angiography with a usually not requiring injection of contrast. If the
high vascular nodule; generally, preoperative lesion shows a typical appearance, i.e., that of a
embolization is not necessary. Spinal hemangio- well-defined lesion, with a stippled appearance,
blastoma represents 2–10 % of intramedullary often with fatty infiltration, other investigations
spinal cord tumors [96–98]. The most frequent are unnecessary. If a deformation of the posterior
location is intramedullary with some lesions wall and enhancing epidural soft tissue mass is
presenting an exophytic component. Lesions visualized on CT, an MRI is essential for better
may also be exclusively intramedullary in loca- demonstration of the extension to the spinal canal
tion or in a minority of cases extramedullary and to evaluate possible spinal cord compression.
[96]. Spinal cord hemangioblastomas are associ- Magnetic resonance appearance of a
ated with von Hippel-Lindau disease in 25 % [99] nonaggressive hemangioma is hyperintense on
and are typically formed by a nodule and/or a cyst, T1-WI and T2-WI with no or very discrete con-
both hyperintense on T2; the nodule is isointense trast enhancement in the periphery on T1-WI
to the spinal cord and the cyst slightly sequence with FAT SAT.
hyperintense compared to cerebrospinal fluid The aggressive vertebral hemangioma has a
(CSF). After contrast, the nodule enhances prevalence of 10 % and a predilection for young
strongly and enhancement of the cyst wall is women at the thoracic level [100].
rare. MRA clearly illustrates the enlarged vessels The aggressive hemangioma is hypointense or
in the periphery of the tumor and is useful for the heterogeneous in T1 and hyperintense in T2, with
presurgical planning, as well as DTI, to guide the significant contrast enhancement. Tissue mass is
neurosurgeon and provide a vascular map in order frequently associated; the most common compli-
to avoid the enlarged vessels and bleeding. cation is spinal cord compression by the mass
Hemangiomas are the most common spine which requires emergency treatment.
tumor; they are often asymptomatic and are usu- Vertebroplasty may be associated with
ally an incidental finding. These lesions may be alcoholization of the epidural component and sur-
solitary or multiple; clinical manifestations are gical decompression only when necessary.
due to compression by the hemangioma, Vertebroplasty strengthens the vertebral body
hemorrhage, or a compression fracture of the ver- and decreases the risk of bleeding [69]. In some
tebra. Symptoms go from back pain to acute cases, a presurgical embolization is also used.
paraplegia. Malignant hemangioma has not been Multiple aggressive hemangiomas can be
reported. The thoracic spine is usually found in the “PTEN hamartoma tumor
involved, and the hemangioma is located within syndrome” [101].
the vertebral body, but they can extend to the Spinal angiolipoma Are rare tumors mainly
neural arch. located in the epidural space and more rarely
The differential diagnosis has to be made with intradural with a vascular enhancing component
a solitary plasmacytoma, metastasis, fatty islands, and a fatty part. Clinical symptoms are slowly
and Paget’s disease. progressive. MRI demonstrates a fatty epidural
1086 M.I. Vargas et al.

mass associated with a soft tissue enhancing com- The classification of SVM proposed by Krings
ponent. Fat saturation is required to confirm the et al. presents the advantage of being based on the
vascular part of the tumor. vascular anatomy of the SC [77]. The distinction
is made between arteriovenous malformation
(AVM) fed by radiculomedullary arteries and
Imaging of Spinal Vascular dural arteriovenous fistula (DAVF) fed by
Malformations radiculomeningeal arteries.
SVM are classified as inborn lesions –
If MRI is the first imaging modality to perform represented by AVM – and DAVF considered as
when a spinal vascular malformation (SVM) is acquired lesions. Epidemiology and clinical pre-
suspected, spinal DSA is mandatory to precise sentation of these SVM are different.
the type of SVM and to decide the therapy. As Spinal cord AVMs are high-flow
the enhancement of the veins is very fast, it is intramedullary and/or perimedullary
difficult to precise the anatomy of the SVM with malformations. Three types are described: glo-
noninvasive imaging modalities. CTA can be use- merular, fistulous, and juvenile. This distinction
ful to guide DSA in the aim to more rapidly is based on the presence (glomerular) or absence
identify the feeding artery of spinal vascular (fistulous) of a nidus. The juvenile type presents
malformations or fistulas, but it cannot be a sub- both fistulous and glomerular compartments, with
stitute [102]. CTA seems performant in the detec- an extension to the neighboring tissues (dura,
tion of dilated perimedullary veins but, as for vertebral body, and paravertebral muscles)
MRI, is not precise enough to determine the [77]. DSA shows typical feeding by multiple
exact type of fistula. Moreover, CTA cannot branches coming from both anterior and posterior
exclude the possibility of multiple feeders [103]. spinal arteries. The malformation can also be

Fig. 17 DSA, AP view.


Eight-month-old boy
presenting an
intramedullary hematoma
revealed by a paraplegia.
There is a familial context
of hereditary hemorrhagic
telangiectasia. Injection into
the left T12 intercostal
artery ((a) arterial phase and
(b) venous phase). (a) A
dilated ASA (arrow) is
directly connected to a
perimedullary venous
pouch. No nidus on MRI
was visible. Diagnosis: pial
arteriovenous fistula. (b)
The dilated venous pouch
drains into an anterior spinal
vein (arrow), which drains
into the intradural (double
arrow) and extradural
(arrowhead) plexuses.
Drainage in the plexuses
and the vein is orthograde
(Image courtesy of Pr Roy,
Service de Radiologie,
CHU de Montréal)
44 Spinal Vascular Imaging: Technique 1087

Fig. 18 Sixty-year-old man presenting hypoesthesia of a radiculomeningeal artery. A retrograde opacification of a


the legs and sphincter disturbance. Vertebral fracture was radiculomedullary vein (arrow) and the hairpin turn of the
known and the patient was initially addressed for vein (star) which connects with other perimedullary veins
vertebroplasty. (a) MRI, sagittal T2 spin echo. MRI (white arrow) are shown. Drainage from the shunt con-
shows increased signal within the conus and multiple cerns the intradural plexus also (double arrow). The etiol-
flow voids along the spinal cord (white arrow). (b) DSA, ogy of the progressive myelopathy is a dural
AP view. Injection into the right L1 lumbar artery. The arteriovenous fistula (Image courtesy of Dr Bessou, Ser-
intradural point of fistulization (double star) is supplied by vice de Radiologie, CHU de Bordeaux)

alimented by one feeder, but always coming from multiple fistulas in the same patient are extremely
a spinal artery (Fig. 17). An intramedullary hema- rare [68, 104].
toma which can extend into the subarachnoid DAVF presents in most cases as a progressive
space is common. myelopathy resulting in weakness of the lower
Dural arteriovenous fistula (DAVF) is an extremities, paresthesia, back pain, and urinary
intradural shunt between a radiculomeningeal symptoms. These small arteriovenous fistulas
artery (coming from segmental arteries) and a reverse the flow of the root veins, which drain
radicular vein; a traumatic or iatrogenic origin is into the intradural spinal cord venous system,
common. Spinal DSA is necessary to precise the thereby resulting in venous hypertension and cen-
location of the fistula (Figs. 18 and 19). tral edema of the spinal cord, a stagnant blood
DAVF represent 70 % of spinal arteriovenous flow and disruption of the blood–brain barrier.
malformations [68, 77, 104, 105] and are five This translates as edema on MRI with a
times more common in men [104] and may be hyperintense signal on T2 or STIR which extends
associated with neurofibromatosis [106]. Most fis- far beyond the fistula itself. Once [101] the fistula
tulas are located at the thoracolumbar level [68]; is treated, the largest part of the signal abnormality
1088 M.I. Vargas et al.

Fig. 19 Seventy-four-year-old man presenting dyses- through a perimedullary vein (retrograde flow) explaining
thesia in both feet since 1 year. Vesical sphincter distur- the cone edema and the symptoms. Diagnosis: epidural
bance since 6 months. (a) MRI, axial T2 spin echo. A left fistula draining intradurally. (d) Spine X-ray, after embo-
anterior epidural lesion (double star) is described. (b, c) lization with glue of the epidural fistula. The venous pouch
Spinal DSA, AP view. (b) Injection into the left L1 lumbar is filled with glue (double star). (e) Non-enhanced CT at
artery shows an arterial connection (arrow) with an epidu- the level of the epidural venous pouch, after embolization.
ral venous poach (double star). (c) Arterial hyperselective The hypersignal corresponds to the glue inside the venous
injection at the entry of the venous poach shows that the (double star) (Image courtesy of Pr Weill, Service de
venous pouch extends on three vertebral levels and drains Radiologie, CHU de Montréal)
44 Spinal Vascular Imaging: Technique 1089

disappears, but it is possible to find a small abnor- 3. Lasjaunias P, Berenstein A, Ter Brugge K (2002)
mal residual signal after several months related to Clinical vascular anatomy and variations, Surgical
neuroangiography. Springer, New York
venous infarction. 4. Gailloud P (2013) The artery of von Haller: a constant
A very rare presentation of dural fistulas can be anterior radiculomedullary artery at the upper thoracic
Foix–Alajouanine syndrome, also known as sub- level. Neurosurgery 73(6):1034–1043
acute necrotizing myelitis, which was reported for 5. Skalski JH, Zembala M (2005) Albert Wojciech
Adamkiewicz: the discoverer of the variable vascu-
the first time by Foix and Alajouanine. Young larity of the spinal cord. Ann Thorac Surg
patients developed rapid progressive and upward 80(5):1971–1975
paralysis and even death. The autopsy revealed 6. Koshino T, Murakami G, Morishita K et al (1999)
wide areas of necrosis in the lumbar and lower Does the Adamkiewicz artery originate from the
larger segmental arteries? J Thorac Cardiovasc Surg
thoracic cord and the medullary cone with severe 117(5):898–905
and incomplete destruction of the gray and white 7. Biglioli P, Roberto M, Cannata A et al (2004) Upper
matter. and lower spinal cord blood supply: the continuity of
the anterior spinal artery and the relevance of the
lumbar arteries. J Thorac Cardiovasc Surg 127(4):
1188–1192
Summary 8. Griepp RB, Ergin MA, Galla JD et al (1996) Looking
for the artery of Adamkiewicz: a quest to minimize
MRI is the first-line study of spinal vascular paraplegia after operations for aneurysms of the
descending thoracic and thoracoabdominal aorta.
malformations. CT may play a role in the evalua- J Thorac Cardiovasc Surg 112(5):1202–1213; discus-
tion of some pathologies, such as bone hemangi- sion 13–15
omas, as well as in the evaluation of aortic 9. Biglioli P, Spirito R, Roberto M et al (2000) The
aneurysms before surgery. anterior spinal artery: the main arterial supply of the
human spinal cord–a preliminary anatomic study.
Finally, with the emergence of high fields and J Thorac Cardiovasc Surg 119(2):376–379
new noninvasive vascular imaging techniques, 10. Lasjaunias P, Berenstein A (1990) Functional vascu-
the detection of vascular malformations becomes lar anatomy of the brain, spinal cord and spine,
easier and their localization more accurate. Surgical neuroangiography. Springer, New York
11. Bowen BC, DePrima S, Pattany PM et al (1996) MR
Dynamic sequences allow an approach to the angiography of normal intradural vessels of the
dynamic study obtained with arteriography thoracolumbar spine. AJNR Am J Neuroradiol
although it is still difficult to differentiate between 17(3):483–494
arteries and veins especially if they are small and 12. Groen RJ, du Toit DF, Phillips FM et al (2004) Ana-
tomical and pathological considerations in percutane-
due to temporal resolution. ous vertebroplasty and kyphoplasty: a reappraisal
Angiography is the second-line study and the of the vertebral venous system. Spine (Phila Pa
next step to precise the type of malformation and 1976) 29(13):1465–1471
for the treatment. A multidisciplinary approach is 13. Bert S, Iyriboz AT, Barret F et al (1995) An angio-
graphic study of spinal vascularization at the thoracic-
needed to determine which vascular lumbar level. J Neuroradiol 22(1):12–19
malformations will receive endovascular treat- 14. Williams GM, Roseborough GS, Webb TH
ment and which will be treated by surgery. et al (2004) Preoperative selective intercostal angiog-
raphy in patients undergoing thoracoabdominal aneu-
rysm repair. J Vasc Surg 39(2):314–321
15. Kieffer E, Fukui S, Chiras J et al (2002) Spinal cord
arteriography: a safe adjunct before descending
References thoracic or thoracoabdominal aortic aneurysmectomy.
J Vasc Surg 35(2):262–268
1. Santillan A, Nacarino V, Greenberg E et al (2012) 16. Jacobs MJ, Mess W, Mochtar B et al (2006) The value
Vascular anatomy of the spinal cord. J Neurointerv of motor evoked potentials in reducing paraplegia
Surg 4(1):67–74 during thoracoabdominal aneurysm repair. J Vasc
2. Thron AK (2008) Vascular anatomy of the spine and Surg 43(2):239–246
spinal cord. In: Hurst RW, Rosenwasser RH (eds) 17. Nair S, Gobin YP, Leng LZ et al (2013) Preoperative
Interventional neuroradiology. Informa Healthcare, embolization of hypervascular thoracic, lumbar, and
New York, pp 39–55 sacral spinal column tumors: technique and outcomes
1090 M.I. Vargas et al.

from a single center. Interv Neuroradiol 19(3): patients with aortic aneurysm. Kurume Med J
377–385 58(4):117–125
18. Chen J, Gailloud P (2011) Safety of spinal angiogra- 32. Clarencon F, Di Maria F, Cormier E et al (2013) Com-
phy: complication rate analysis in 302 diagnostic parison of intra-aortic computed tomography angiog-
angiograms. Neurology 77(13):1235–1240 raphy to conventional angiography in the presurgical
19. Forbes G, Nichols DA, Jack CR Jr et al (1988) Com- visualization of the Adamkiewicz artery: first results
plications of spinal cord arteriography: prospective in patients with thoracoabdominal aortic aneurysms.
assessment of risk for diagnostic procedures. Radiol- Neuroradiology 55(11):1379–1387
ogy 169(2):479–484 33. Nelson RC, Feuerlein S, Boll DT (2011) New itera-
20. Savader SJ, Williams GM, Trerotola SO et al (1993) tive reconstruction techniques for cardiovascular
Preoperative spinal artery localization and its relation- computed tomography: how do they work, and what
ship to postoperative neurologic complications. Radi- are the advantages and disadvantages? J Cardiovasc
ology 189(1):165–171 Comput Tomogr 5(5):286–292
21. Heinemann MK, Brassel F, Herzog T et al (1998) The 34. Nishida J, Kitagawa K, Nagata M et al (2014) Model-
role of spinal angiography in operations on the tho- based iterative reconstruction for multi-detector row
racic aorta: myth or reality? Ann Thorac Surg 65(2): CT assessment of the Adamkiewicz artery. Radiology
346–351 270(1):282–291
22. Uotani K, Yamada N, Kono AK et al (2008) Preoper- 35. Machida H, Tanaka I, Fukui R et al (2013) Improved
ative visualization of the artery of Adamkiewicz by delineation of the anterior spinal artery with model-
intra-arterial CT angiography. AJNR Am based iterative reconstruction in CT angiography: a
J Neuroradiol 29(2):314–318 clinical pilot study. AJR Am J Roentgenol
23. Takase K, Sawamura Y, Igarashi K et al (2002) Dem- 200(2):442–446
onstration of the artery of Adamkiewicz at multi- 36. Ou P, Schmit P, Layouss W et al (2007) CT angiogra-
detector row helical CT. Radiology 223(1):39–45 phy of the artery of Adamkiewicz with 64-section
24. Melissano G, Bertoglio L, Civelli Vet al (2009) Dem- technology: first experience in children. AJNR Am
onstration of the Adamkiewicz artery by J Neuroradiol 28(2):216–219
multidetector computed tomography angiography 37. Nijenhuis RJ, Jacobs MJ, Jaspers K et al (2007) Com-
analysed with the open-source software OsiriX. Eur parison of magnetic resonance with computed tomog-
J Vasc Endovasc Surg 37(4):395–400 raphy angiography for preoperative localization of the
25. Zhao SH, Logan L, Schraedley P et al (2009) Assess- Adamkiewicz artery in thoracoabdominal aortic aneu-
ment of the anterior spinal artery and the artery of rysm patients. J Vasc Surg 45(4):677–685
Adamkiewicz using multi-detector CT angiography. 38. Boll DT, Bulow H, Blackham KA et al (2006) MDCT
Chin Med J (Engl) 122(2):145–149 angiography of the spinal vasculature and the artery of
26. Yoshioka K, Niinuma H, Ogino Y et al (2006) Three- Adamkiewicz. AJR Am J Roentgenol 187(4):
dimensional demonstration of the collateral circula- 1054–1060
tion to the artery of Adamkiewicz with 16-row 39. Yoshioka K, Niinuma H, Ohira A et al (2003) MR
multislice computed tomography. Ann Thorac Surg angiography and CT angiography of the artery of
81(2):749 Adamkiewicz: noninvasive preoperative assessment
27. Utsunomiya D, Yamashita Y, Okumura S et al (2008) of thoracoabdominal aortic aneurysm. Radiographics
Demonstration of the Adamkiewicz artery in patients 23(5):1215–1225
with descending or thoracoabdominal aortic aneu- 40. Murakami H, Kawahara N, Tomita K et al (2010)
rysm: optimization of contrast-medium application Does interruption of the artery of Adamkiewicz dur-
for 64-detector-row CT angiography. Eur Radiol ing total en bloc spondylectomy affect neurologic
18(11):2684–2690 function? Spine (Phila Pa 1976) 35(22):
28. Kudo K, Terae S, Asano T et al (2003) Anterior spinal E1187–E1192
artery and artery of Adamkiewicz detected by using 41. Smit JW, Vielvoye GJ, Goslings BM (2000) Emboli-
multi-detector row CT. AJNR Am J Neuroradiol zation for vertebral metastases of follicular thyroid
24(1):13–17 carcinoma. J Clin Endocrinol Metab 85(3):989–994
29. Nojiri J, Matsumoto K, Kato A et al (2007) The 42. Court C, Noun Z, Gagey O et al (2000) Surgical
Adamkiewicz artery: demonstration by intra-arterial treatment of metastases from thyroid cancer in the
computed tomographic angiography. Eur axial skeleton. A retrospective study of 18 cases.
J Cardiothorac Surg 31(2):249–255 Acta Orthop Belg 66(4):345–352
30. Nakayama Y, Awai K, Yanaga Y et al (2008) Optimal 43. Prabhu VC, Bilsky MH, Jambhekar K et al (2003)
contrast medium injection protocols for the depiction Results of preoperative embolization for metastatic
of the Adamkiewicz artery using 64-detector CT angi- spinal neoplasms. J Neurosurg 98(2 Suppl):156–164
ography. Clin Radiol 63(8):880–887 44. Kobayashi K, Ozkan E, Tam A et al (2012) Preoper-
31. Amako M, Yamamoto Y, Nakamura K et al (2011) ative embolization of spinal tumors: variables affect-
Preoperative visualization of the artery of ing intraoperative blood loss after embolization. Acta
Adamkiewicz by dual-phase CT angiography in Radiol 53(8):935–942
44 Spinal Vascular Imaging: Technique 1091

45. Wilson MA, Cooke DL, Ghodke B et al (2010) Ret- in thoracic and thoracoabdominal aortic aneurysm
rospective analysis of preoperative embolization of surgery. J Vasc Surg 45(1):71–77; discussion 77–78
spinal tumors. AJNR Am J Neuroradiol 31(4): 60. Etz CD, Halstead JC, Spielvogel D et al (2006) Tho-
656–660 racic and thoracoabdominal aneurysm repair: is
46. Robial N, Charles YP, Bogorin I et al (2012) Is pre- reimplantation of spinal cord arteries a waste of
operative embolization a prerequisite for spinal time? Ann Thorac Surg 82(5):1670–1677
metastases surgical management? Orthop Traumatol 61. Nakai M, Shimizu S, Ochi Y et al (2009)
Surg Res 98(5):536–542 Thoracodorsal artery as a collateral source to the
47. Thiex R, Harris MB, Sides C et al (2013) The role of artery of Adamkiewicz after endovascular aneurysm
preoperative transarterial embolization in spinal repair for descending thoracic aortic aneurysm. Eur J
tumors. A large single-center experience. Spine J Vasc Endovasc Surg 37(5):566–568
13(2):141–149 62. Maliszewski M, Ladzinski P, Aleksandrowicz R
48. Jones KM, Gobin P, Liu A-H (2003) Embolisation of et al (1999) Occlusion of radicular arteries – reasons,
spinal tumors. Oper Tech Neurosurg 6(3):156–162 consequences and anastomotic substitution pathways.
49. Smith TP, Gray L, Weinstein JN et al (1995) Preop- Spinal Cord 37(10):710–716
erative transarterial embolization of spinal column 63. von Tengg-Kobligk H, Bockler D, Jose TM
neoplasms. J Vasc Interv Radiol 6(6):863–869 et al (2007) Feeding arteries of the spinal cord at CT
50. Shi HB, Suh DC, Lee HK et al (1999) Preoperative angiography before and after thoracic aortic
transarterial embolization of spinal tumor: emboliza- endografting. J Endovasc Ther 14(5):639–649
tion techniques and results. AJNR Am J Neuroradiol 64. Etz CD, Luehr M, Kari FA et al (2008) Paraplegia
20(10):2009–2015 after extensive thoracic and thoracoabdominal aortic
51. O’Reilly GV, Kleefield J, Klein LA et al (1989) aneurysm repair: does critical spinal cord ischemia
Embolization of solitary spinal metastases from occur postoperatively? J Thorac Cardiovasc Surg
renal cell carcinoma: alternative therapy for spinal 135(2):324–330
cord or nerve root compression. Surg Neurol 65. Vargas MI, Nguyen D, Viallon M et al (2010)
31(4):268–271 Dynamic MR angiography (MRA) of spinal vascular
52. Djindjian R, Cophignon J, Rey A et al (1973) diseases at 3 T. Eur Radiol 20(10):2491–2495.
Superselective arteriographic embolization by the doi:10.1007/s00330-010-1815-6
femoral route in neuroradiology. Study of 50 cases. 66. Wang D, Yang N, Zhang P et al (2013) The diagnosis
II. Embolization in vertebromedullary pathology. of spinal dural arteriovenous fistulas. Spine.
Neuroradiology 6(3):132–142 doi:10.1097/BRS.0b013e31828a38c4
53. Estrera AL, Miller CC 3rd, Huynh TT et al (2001) 67. Vargas MI, Delavelle J, Jlassi H et al (2008) Clinical
Neurologic outcome after thoracic and applications of diffusion tensor tractography of the
thoracoabdominal aortic aneurysm repair. Ann spinal cord. Neuroradiology 50(1):25–29
Thorac Surg 72(4):1225–1230; discussion 30–31 68. Krings T, Geibprasert S (2009) Spinal dural arterio-
54. Coselli JS, LeMaire SA, Miller CC 3rd et al (2000) venous fistulas. AJNR Am J Neuroradiol 30(4):
Mortality and paraplegia after thoracoabdominal aor- 639–648. doi:10.3174/ajnr.A1485
tic aneurysm repair: a risk factor analysis. Ann Thorac 69. Ide C, Gangi A, Rimmelin A et al (1996) Vertebral
Surg 69(2):409–414 haemangiomas with spinal cord compression: the
55. Griepp RB, Griepp EB (2007) Spinal cord perfusion place of preoperative percutaneous vertebroplasty
and protection during descending thoracic and with methyl methacrylate. Neuroradiology 38(6):
thoracoabdominal aortic surgery: the collateral net- 585–589
work concept. Ann Thorac Surg 83(2):S865–S869; 70. Kharkar S, Shuck J, Conway J et al (2007) The natural
discussion S90–S92 history of conservatively managed symptomatic
56. Weigang E, Hartert M, Siegenthaler MP et al (2006) intramedullary spinal cord cavernomas. Neurosurgery
Perioperative management to improve neurologic 60(5):865–872. doi:10.1227/01.NEU.0000255437.3
outcome in thoracic or thoracoabdominal aortic 6742.15; discussion 65–72
stent-grafting. Ann Thorac Surg 82(5):1679–1687 71. Deutsch H, Jallo GI, Faktorovich A et al (2000) Spi-
57. Amabile P, Grisoli D, Giorgi R et al (2008) Incidence nal intramedullary cavernoma: clinical presentation
and determinants of spinal cord ischaemia in stent- and surgical outcome. J Neurosurg 93(1 Suppl):
graft repair of the thoracic aorta. Eur J Vasc Endovasc 65–70
Surg 35(4):455–461 72. Spetzger U, Gilsbach JM, Bertalanffy H (1995) Cav-
58. Fereshetian A, Kadir S, Kaufman SL et al (1989) ernous angiomas of the spinal cord clinical presenta-
Digital subtraction spinal cord angiography in tion, surgical strategy, and postoperative results. Acta
patients undergoing thoracic aneurysm surgery. Neurochir (Wien) 134(3–4):200–206
Cardiovasc Intervent Radiol 12(1):7–9 73. Babu R, Owens TR, Karikari IO et al (2013) Spinal
59. Nijenhuis RJ, Jacobs MJ, Schurink GW et al cavernous and capillary hemangiomas in adults.
(2007) Magnetic resonance angiography and Spine 38(7):E423–E430. doi:10.1097/BRS.0b013e3
neuromonitoring to assess spinal cord blood supply 18287fef7
1092 M.I. Vargas et al.

74. Bergametti F, Denier C, Labauge P et al (2005) Muta- 88. Hatiboglu MA, Iplikcioglu AC, Ozcan D (2006) Epi-
tions within the programmed cell death 10 gene cause dural spinal cavernous hemangioma. Neurol Med
cerebral cavernous malformations. Am J Hum Genet Chir 46(9):455–458
76(1):42–51. doi:10.1086/426952 89. Kurose K, Kishi H, Sadatoh T (1989) Spinal epidural
75. Guzeloglu-Kayisli O, Amankulor NM, Voorhees J cavernous hemangioma. Case report. Neurol Med
et al (2004) KRIT1/cerebral cavernous malformation Chir 29(6):538–542
1 protein localizes to vascular endothelium, 90. Minh NH (2005) Cervicothoracic spinal epidural cav-
astrocytes, and pyramidal cells of the adult human ernous hemangioma: case report and review of the
cerebral cortex. Neurosurgery 54(4):943–949; discus- literature. Surg Neurol 64(1):83–85. doi:10.1016/j.
sion 49 surneu.2004.10.030; discussion 85
76. Labauge P, Brunereau L, Coubes P et al (2001) 91. Padolecchia R, Acerbi G, Puglioli M et al (1998)
Appearance of new lesions in two nonfamilial cere- Epidural spinal cavernous hemangioma. Spine
bral cavernoma patients. Eur Neurol 45(2):83–88. 23(10):1136–1140
doi:10.1159/000052100 92. Richardson RR, Cerullo LJ (1979) Spinal epidural
77. Krings T, Mull M, Gilsbach JM et al (2005) Spinal cavernous hemangioma. Surg Neurol 12(3):266–268
vascular malformations. Eur Radiol 15(2):267–278. 93. Rachinger J, Buslei R, Engelhorn T et al (2006)
doi:10.1007/s00330-004-2510-2 Intradural-extramedullary cavernous hemangioma of
78. Biondi A, Clemenceau S, Dormont D et al (2002) the left motor root C7 – case report and update of the
Intracranial extra-axial cavernous (HEM) angiomas: literature. Zentralbl Neurochir 67(3):144–148.
tumors or vascular malformations? J Neuroradiol doi:10.1055/s-2006-933362
29(2):91–104 94. Lefranc F, Baleriaux D, Brotchi J (2007)
79. Pechstein U, Zentner J, Van Roost D et al (1997) Intramedullary cavernomas: personal series of
Surgical management of brain-stem cavernomas. 24 cases. Neurochirurgie 53(2–3 Pt 2):203–207.
Neurosurg Rev 20(2):87–93 doi:10.1016/j.neuchi.2007.02.009
80. Pearl MS, Chen JX, Gregg L et al (2012) Angio- 95. Liang JT, Bao YH, Zhang HQ et al (2011) Manage-
graphic detection and characterization of “cryptic ment and prognosis of symptomatic patients with
venous anomalies” associated with spinal cord cav- intramedullary spinal cord cavernoma: clinical article.
ernous malformations using flat-panel catheter J Neurosurg Spine 15(4):447–456. doi:10.3171/
angiotomography. Neurosurgery 71(1 Suppl 2011.5.SPINE10735
Operative):125–132 96. Imagama S, Ito Z, Wakao N et al (2011) Differentia-
81. Vargas MI, Delavelle J, Jlassi H et al (2008) Clinical tion of localization of spinal hemangioblastomas
applications of diffusion tensor tractography of the based on imaging and pathological findings. Eur
spinal cord. Neuroradiology 50(1):25–29. Spine J Off Publ Eur Spine Soc Eur Spinal Deform
doi:10.1007/s00234-007-0309-y Soc Eur Sect Cerv Spine Res Soc 20(8):1377–1384.
82. Rovira A, Rovira A, Capellades J et al (1999) Lumbar doi:10.1007/s00586-011-1814-6
extradural hemangiomas: report of three cases. AJNR 97. Browne TR, Adams RD, Roberson GH (1976)
Am J Neuroradiol 20(1):27–31 Hemangioblastoma of the spinal cord. Review and
83. Hemalatha AL, Ravikumar T, Chamarthy NP report of five cases. Arch Neurol 33(6):435–441
et al (2013) A pure epidural spinal cavernous heman- 98. Parsa AT, Lee J, Parney IF et al (2004) Spinal cord and
gioma – with an innocuous face but a perilous behav- intradural-extraparenchymal spinal tumors: current
iour!! J Clin Diagn Res JCDR 7(7):1434–1435. best care practices and strategies. J Neurooncol
doi:10.7860/JCDR/2013/6030.3159 69(1–3):291–318
84. Zhong W, Huang S, Chen H et al (2012) Pure spinal 99. Mechtler LL, Nandigam K (2013) Spinal cord
epidural cavernous hemangioma. Acta Neurochir tumors: new views and future directions. Neurol
(Wien) 154(4):739–745. doi:10.1007/s00701-012- Clin 31(1):241–268. doi:10.1016/j.ncl.2012.09.011
1295-3 100. Fox MW, Onofrio BM (1993) The natural history and
85. Shin JH, Lee HK, Rhim SC et al (2001) Spinal epi- management of symptomatic and asymptomatic ver-
dural cavernous hemangioma: MR findings. tebral hemangiomas. J Neurosurg 78(1):36–45.
J Comput Assist Tomogr 25(2):257–261 doi:10.3171/jns.1993.78.1.0036
86. Aoyagi N, Kojima K, Kasai H (2003) Review of 101. Jenny B, Radovanovic I, Haenggeli CA et al (2007)
spinal epidural cavernous hemangioma. Neurol Med Association of multiple vertebral hemangiomas and
Chir 43(10):471–475; discussion 76 severe paraparesis in a patient with a PTEN
87. Hara K, Hidaka K (2006) Cavernous hemangioma hamartoma tumor syndrome. Case report. J
growing continuously from the spinal epidural space Neurosurg 107(4 Suppl):307–313. doi:10.3171/
into the paravertebral muscles: case report. No PED-07/10/307
Shinkei Geka Neurol Surg 34(5):521–525
44 Spinal Vascular Imaging: Technique 1093

102. Si-jia G, Meng-wei Z, Xi-ping L et al (2009) The 104. Thron A (2001) Spinal dural arteriovenous fistulas.
clinical application studies of CT spinal angiography Radiologe 41(11):955–960
with 64-detector row spiral CT in diagnosing 105. Krings T (2010) Vascular malformations of the spine
spinal vascular malformations. Eur J Radiol and spinal cord: anatomy, classification, treatment.
71(1):22–28 Klin Neuroradiol. doi:10.1007/s00062-010-9036-6
103. Yamaguchi S, Nagayama T, Eguchi K et al (2010) 106. Kamiyama K, Endo S, Horie Y et al (1985) Neurofi-
Accuracy and pitfalls of multidetector-row computed bromatosis associated with intra- and extracranial
tomography in detecting spinal dural arteriovenous aneurysms and extracranial vertebral arteriovenous
fistulas. J Neurosurg Spine 12(3):243–248 fistula. No Shinkei Geka Neurol Surg 13(8):875–880
Spinal Vascular Anatomy
45
Tibor Becske and Eytan Raz

Contents Keywords
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1095
Artery of adamkiewicz • Artery of lazorthes •
Radiculomedullary • Spinal vasculature
Arteries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1096
Veins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1101
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1103 Introduction

The purpose of this chapter is to summarize the


spinal vascular anatomy. After a detailed descrip-
tion of the segmental anatomy, the spinal cord
supply will ensue with description of the radiculo-
medullary arteries, the anterior spinal artery, and
the spinal veins. Part of the text, the concept and
the figures in this chapter are used with permission
from Maksim Shapiro, from the website www.
neuroangio.org.
The neural tube/spinal cord can be conceptual-
ized as a cylindrical mass of tissue. For a short
period of time after closure of the neural tube,
nutritional support can be adequately provided
by simple diffusion. When limits of diffusion are
exceeded, dedicated vascular supply to the spinal
cord and adjacent tissues is established via seg-
mental (metameric) vessels arising from the dor-
sal aorta. The human embryo is in fact subdivided
into 31 somites, each corresponding to a develop-
ing metameric segment, that ultimately gives rise
to all endo-, ecto-, and mesodermal derivatives.
Each somite is supplied by those paired segmental
T. Becske (*) • E. Raz (*) arteries originating from the dorsal aorta. Each
Neurointerventional Radiology Section, Department of segmental vessel supports its endodermal, ecto-
Radiology, NYU Langone Medical Center, New York, NY,
USA dermal, and mesodermal tissue. Longitudinal
e-mail: eytan.raz@gmail.com anastomoses are then established between
# Springer Science+Business Media New York 2016 1095
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_14
1096 T. Becske and E. Raz

segmental vessels, thereby giving rise to new ves- originate from any of the above described longi-
sels extending craniocaudally along the length of tudinal vessels, although most commonly, the
the cord. A grid-like pattern is hence established dominant cervical radiculomedullary branch
which is beginning to resemble mature (artery of Lazorthes) originates from the lower
arrangement. vertebral artery. The upper thoracic spine is a
But, as longitudinal vessels develop, hemody- transitional zone where segmental arteries are
namic need for having a segmental radiculo- supplied by the supreme intercostal artery, which
medullary vessel at each level is diminished. corresponds to the continuation of the
Most segmental vessels diminish to supply only pre-transverse anastomosis. As the cord continues
the region of segmental nerve root. The cord is to enlarge, perforator or sulco-comissural
now supplied by the “anterior spinal” artery ven- branches emerge, which unless pathologically
trally. A loose network of arteries with two enlarged, they are below limits of angiographic
roughly dominant channels forms the posterior resolution.
spinal arteries. Finally, only several segmental
vessels remain to supply the cord. These are the
radiculomedullary vessels (feeding nerve root and Arteries
cord) of the adult. Other segmental vessels persist
as purely radicular (supplying only nerve roots). The following discussion is visually recapitulated
The successive transversely oriented segmen- in Fig. 1, from the website www.neuroangio.org.
tal arteries anastomose through a number of Throughout the following description, all upper-
paraspinal vessels longitudinally arranged along case letters given in parentheses refer to legends in
the axis of the spine. These longitudinal channels Fig. 1.
include the prevertebral artery situated adjacent to The aorta (A) (Fig. 2) gives rise to the proto-
the anterolateral aspect of the vertebral body, the typical spinal segmental artery, lumbar or thoracic
pre-transverse anastomoses anterior to the trans- (B), which after the origin, continues posterior
verse process, as well as the post-transverse spinal and laterally along the vertebral body. Examples
anastomotic arcade which extends craniocaudally of these segmental arteries are shown in Figs. 2, 3,
along both sides of the spinal processes. In the 4, 5, 6, and 7. At the junction between the verte-
lower lumbosacral spine, a homologous arrange- bral body and the transverse process, the segmen-
ment is recognizable – with the median sacral tal artery gives rise to the dorsal spinal trunk, also
artery representing the continuation of the aorta called dorsal spinal artery, which quickly bifur-
and internal iliac arteries considered the homo- cates into dorsal (K) and ventral divisions (H).
logues of the paravertebral longitudinal arcades, The segmental artery continues as either an inter-
respectively – supplying the corresponding lower costal or muscular artery (F). The ventral division
lumbar and sacral metamers. of the dorsal spinal artery (K) traverses the neural
The same segmental arrangement is present in foramen and supplies all adjacent neural, dural,
the upper thoracic and cervical spine: the ascend- and osseous structures. It gives off anterior epidu-
ing cervical artery which corresponds to the ral branches which form a characteristic,
prevertebral artery, the vertebral artery (coursing diamond-shaped retrocorporeal arcade
in the cervical osseous homologue of the trans- vascularizing the ventral epidural space and serv-
verse process) which is homologous with the ing as a potential collateral conduit to the contra-
pre-transverse anastomotic artery, and the deep lateral segmental artery (Fig. 8). Branches to the
cervical artery with the post-transverse dorsal epidural space form a less consistently
anastomosis. visualized epidural arcade. The continuation of
As a consequence of this vascular disposition, the ventral branch becomes associated with the
the contributions to the anterior spinal axis can corresponding nerve root sleeve, where it
45 Spinal Vascular Anatomy 1097

Fig. 1 Schematics of the arterial supply to spinal cord division dorsal spinal artery; N – dural branch of the
(Courtesy of Dr. Maksim Shapiro from his website, with ventral division dorsal spinal artery; O – radiculopial
permission, www.neuroangio.org). A – aorta; B – segmen- artery; P – radiculomedullary artery; Q – anterior spinal
tal artery; Ba – intersegmental arterial anastomosis; C – artery; R – mesh-like pial arterial network; S, T – posterior
prevertebral anastomotic network; D – direct vertebral spinal artery; U, V – pial arterial network
body feeding arteries; E – dorsal spinal artery; F – inter- (a.k.a. vasocorona) anastomoses between anterior and pos-
costal/muscular artery; G – pretransverse anastomotic net- terior spinal arterial systems, W – sulcocommissural artery,
work; H – dorsal division of the dorsal spinal artery; I – X – rami perforantes of the peripheral (centripetal) system,
post-transverse anastomotic network; J – muscular Y – central (centrifugal) system of sulcal arteries, originat-
branches of the post-transverse anastomotic network; K – ing from pial network of the cord; altogether, the pial
ventral division of the dorsal spinal artery; Ka – radicular network and rami perforantes (R+Y) are called the
artery; La – ventral epidural arcade; Lb – dorsal epidural vasocorona or corona vasorum; Z – rami cruciantes
arcade; M – nerve root sleeve dural branch of the ventral (a.k.a. crux vasculosa, a.k.a. rami anastomotici arcuati)

provides radiculodural branches to supply the lamina, forming a post-transverse longitudinal


regional dura and ultimately penetrates the sleeve anastomosis (I) close to the spinous processes.
to become the radicular artery, supplying anterior The most effective transverse anastomosis
and posterior nerve roots and at discrete levels between left- and right-sided vessels is
contributing to the vascularization of the spinal represented by the retrocorporeal arcade within
cord as radiculomedullary (P) or radiculopial the anterior epidural space (Fig. 8).
arteries (O). The arteries supplying neural structures within
The dorsal division of the dorsal spinal artery the dural sheath can be divided into three groups:
(H) passes posteriorly beneath the ipsilateral (a) small radicular arteries, below angiographic
transverse process and along the outer surface of resolution, which are branches of the dorsal spinal
1098 T. Becske and E. Raz

Fig. 2 Aortogram demonstrating the segmental branches arising from the aorta. Notice the different orientation of the
segmental branches going from the thoracic to the lumbar region

Fig. 3 This is a left L3 pedicle injection in arterial phase subtracted (a) and unsubtracted (b)
45 Spinal Vascular Anatomy 1099

Fig. 4 This is a left T11 pedicle injection in arterial phase spinal artery. The dominant radiculomedullary contribu-
subtracted (a) and unsubtracted (b). There is a well visible tion to the anterior spinal artery comes more often from the
dominant radiculomedullary contribution to the anterior left side between T9 and T12

artery, which are present at nearly every level, and and quite smaller in the mid-thoracic area (0.1–04
which supply the nerve root. At some levels, the mm). The anterior spinal artery (Figs. 4 and 9) is
territory of these arteries is not limited to the nerve supplied by multiple radiculomedullary arteries,
root, in which case the vessel is termed radiculo- most commonly from the proximal cervical ver-
medullary, when supplying the anterior spinal tebral artery (artery of Lazorthes) – but can orig-
axis, or radiculopial when supplying posterior inate instead from anterior cervical, deep cervical
spinal axis. supreme intercostal arteries. At thoracic and lum-
The anterior spinal artery (Q) is a form of bar levels, radiculomedullary vessels arise from
longitudinal anastomosis between multiple trans- the ventral division of the segmental artery
verse radicular arteries. In the adult form, the (Figs. 4 and 9). The main thoracolumbar contrib-
anterior spinal artery is a longitudinal channel utory to the anterior spinal artery, the artery of
located in the anterior median sulcus, with the Adamkiewicz arises from the lower thoracic or
caliber varying with regional hemodynamic upper lumbar region, between T9 and L1 most
requirements, being larger in the cervical commonly from the left side. The confluence of
(0.2–0.5 mm) and lumbar regions (0.5–0.8 mm) the radiculomedullary artery and anterior spinal
1100 T. Becske and E. Raz

Fig. 5 This is a left T12 pedicle injection in arterial phase contribution to the anterior spinal artery is again seen,
unsubtracted (a) and subtracted (b). Notice the well visible which is related to anastomotic connections between the
epidural diamond-shaped arterial plexus (retrocorporeal T11 and T12 pedicles
arcade). Also partial visualization of the radiculomedullary

artery typically has a classical hairpin-turn shape and angiographically identifies the spinal cord
(Fig. 9). conus.
The posterior spinal artery axis does not exist The intrinsic spinal cord vasculature was
as a continuous channel but a discontinuous chan- subdivided into centrifugal and centripetal com-
nel beyond several adjacent levels supplied by ponents. The anterior spinal artery gives rise to
multiple radiculopial arteries, the larger visualized multiple sulco-comissural vessels, which pene-
angiographically as hairpin-like vessels, similar to trate deep within the anterior median sulcus to
the one supplying the anterior spinal artery, but vascularize the central gray matter, below DSA
slightly off-midline. resolution. The sulco-commissural arteries give
Arterial supply of conus medullaris consists of rise to a network of small vessels, which radiate
a characteristic “basketlike” arrangement, centrifugally from central cord to the peripheral
reflecting the confluence of the anterior spinal spinal cord. The sulco-commissural distribution is
artery with the paired posterior spinal classically described as covering the anterior
arteries, via prominent anastomotic channels two-thirds of the cross-sectional area of the spinal
called rami cruciantes (Fig. 10). The visualization cord, while the centripetally directed rami
of this vascular basket is highly desirable as perforantes, originating from the pial vascular
part of a complete spinal angiogram meshwork covering the surface of the spinal
45 Spinal Vascular Anatomy 1101

Fig. 6 This is a left supreme intercostal pedicle injection in arterial phase unsubtracted (a) and subtracted (b)

cord, cover the posterior third. This arrangement The intramedullary network consists of two
creates a watershed zone within the cord paren- components, the sulcal and the radial veins. The
chyma between centripetal and centrifugal sulcal component predominantly drains the terri-
systems. tory of gray matter in a centripetal fashion, with
outflow in the sulco-commissural veins, while the
radial veins drain centrifugally toward the surface
of the cord. Eventually, both systems empty into
Veins the superficial spinal cord veins. The main longi-
tudinal veins are the anterior and posterior spinal
The spinal venous system consists of three differ- veins which are located in the subpial space. The
ent components: (a) intrinsic venous network of anterior midline vein is located behind the artery
the spinal cord parenchyma, (b) extrinsic system in the anterior median sulcus, while the posterior
consisting of superficial spinal cord veins and median spinal vein is located in the dorsal median
radicular veins, and (c) extradural venous plexus, fissure. The longitudinal veins are the largest
consisting of veins in the epidural plexus, sur- perimedullary vessels, measuring up to 1.5 mm,
rounding the vertebral bodies and the paraspinal and are the vessels most likely to be seen on the
musculature. postcontrast spine MRI of normal patients. The
1102 T. Becske and E. Raz

anterior and posterior spinal veins are connected


by multiple transverse veins surrounding the cord,
as well as transmedullary channels running
through the spinal cord. The superficial venous
blood drains in the epidural venous plexus via the
radicular veins, which have a hairpin shape, sim-
ilar to the shape of the arteries (for this reason it is
not possible to discern between the radicular sup-
ply to an anterior spinal artery and a dominant
radicular vein on a non-dynamic spine MRA
study). The radicular veins represent the weaker
segment of the venous system since there is no
redundancy, while that is not the case for both the
intrinsic spinal cord system and the extrinsic
extradural veins. The radicular veins travel along
the nerve roots across the subarachnoid space and
enter the dural nerve root sleeve at a shallow
angle, an arrangement which acts as a functional
valve, preventing venous reflux into the cord
when there is increased central venous pressure.
The radicular veins drain in the epidural plexus,
which extends all the way continuously from the
base of the skull to the sacrum and via segmental
Fig. 7 This is a median sacral artery pedicle injection in
arterial phase, subtracted view
intervertebral (foraminal) veins is connected with
the azygos and hemiazygos veins.

Fig. 8 This is a left L2 pedicle injection in arterial phase unsubtracted (a) and subtracted (b). Notice the well visible
diamond-shaped retrocorporeal arcade
45 Spinal Vascular Anatomy 1103

Fig. 9 This is a left T7 pedicle injection in arterial phase, Fig. 10 This image shows the typical angiographic
subtracted view. Notice again the radiculomedullary con- appearance of the conus medullaris. The basket-like
tribution to the anterior spinal artery arrangement reflects the confluence between the anterior
and posterior spinal arteries

References
Pref by Guy Lazorthes English translation by Irvin I
1. Adamkiewicz AW (1881) Die BlutgefaBe des Kricheff. University Park Press, Baltimore, p 482
mencshlichen Ruckenmarks. I. Die GefaBe der 4. Lazorthes G, Gouaze A, Djindjian R (1973) Vasculari-
Ruckenmarks substanz. Sitz. Ber. Akad. Wiss. Wien, zation et pathologie vasculaire de la moelle epiniere.
Math. nat. Kl., Berlin, pp 469–502 Masson, Paris
2. Adamkiewicz AW (1882) Die BlutgefaBe des 5. Lasjaunias PL, Berenstein A, Raybaud C (1987) Surgi-
mencshlichen Ruckenmarks. II. Die GefaBe der cal neuroangiography. Springer, Berlin/New York
Ruckenmarks-oberflashe. Sitz. Ber. Akad. Wiss. Wien, 6. Lasjaunias PL, Berenstein A, Brugge KG (2001) Surgical
Math. nat. Kl., pp 101–130 neuroangiography, 2nd edn. Springer, Berlin/New York
3. Djindjian R (1970) Angiography of the spinal cord. In: 7. Shapiro M (2012) www.neuroangio.org. [cited; Cere-
Hurth M (ed) Angiography of the spinal cord/by R brospinal neurovascular information source]. Available
Djindjian, with collaboration of M Hurth [and others] from www.neuroangio.org
Spinal Vascular Malformations and
Treatment 46
Srinivasan Paramasivam

Contents Abstract
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1106 Spinal vascular malformations are rare, but a
diverse group of neurovascular pathologies
Spinal Vascular Anatomy . . . . . . . . . . . . . . . . . . . . . . . . . 1106
including arteriovenous malformations
Spinal Vascular Malformation Classification . . . 1107 (AVMs), cavernous malformations, and dural
Type I: Dural Arteriovenous Fistula . . . . . . . . . . . . . . . 1107 arteriovenous fistulas (dAVFs) that occur due
Type II: Intramedullary Arteriovenous
Malformation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1109 to developmental derangement of the vascular
Type III: Juvenile Arteriovenous Malformation . . . 1113 system. Though classified variously, the com-
Type IV: Intradural Perimedullary Arteriovenous monly followed one is anatomic classification
Fistula . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1116 based on the morphology of the malformation
Cavernous Malformation (Cavernomas) . . . . . . . . . . . 1119
to which added are cavernomas and spinal
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1121 vascular tumors. Pathophysiologic mecha-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1122 nisms leading to clinical symptoms include
intramedullary or subarachnoid hemorrhages,
arterial ischemia, progressive venous conges-
tion resulting in progressive myelopathy,
space-occupying nature of the malformation
and its venous drainage, and circulatory
steal phenomenon. Most spinal vascular
malformations come to clinical attention by
nonspecific symptomatology that may be
acute, subacute, or chronic. The initial clinical
diagnosis is challenging and it is based on
MRI; however, for better understanding and
planning therapeutic strategy spinal angiogram
is necessary. Once diagnosed, most spinal vas-
cular malformations need treatment, as the nat-
ural history suggests progressive stepwise
neurological deterioration. Early diagnosis
S. Paramasivam (*) and timely management in symptomatic
Mount Sinai Roosevelt Hospital, Hyman Newman Institute
patients can result in improvement or stabiliza-
for Neurology and Neurosurgery, Centre for Endovascular
Surgery, New York, NY, USA tion of clinical condition. Treatment should be
e-mail: kpsvasan@hotmail.com performed in specialized centers. Except spinal
# Springer Science+Business Media New York 2016 1105
L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_45
1106 S. Paramasivam

cord cavernomas and spinal vascular tumors, spine, including the vertebral bodies, paraspinal
all other vascular disorders are initially man- muscles, dura, nerve roots, and spinal cord with
aged by endovascular embolization, as it is the blood. Radicular arteries are the first branches of
least invasive but technically challenging and the dorsal division of the segmental arteries. At
demanding procedure. For cases where embo- each level the radiculomeningeal artery arises
lization is technically not possible or when from the radicular artery and supplies the dural
they fail embolization treatment, surgical treat- and the nerve root. From these radicular arteries
ment is an available option. In select cases a also arise the radiculomedullary and radiculopial
combined therapy might be sensible. Early arteries that follow the anterior or posterior nerve
diagnosis and prompt treatment are essential root to reach the anterior or posterior surface of
to alter the natural course and result in clinical the cord, where they form the anterior or posterior
improvement. spinal arteries [1]. The radiculomedullary feeders
are not present at every level, and their location is
Keywords highly variable and is not predictable.
Spinal vascular malformation • Arteriovenous The anterior and posterior spinal arteries con-
malformation • Dural arteriovenous fistula • stitute a superficial longitudinal anastomosing
Perimedullary arteriovenous fistula • system [2]. The anterior spinal artery originates
Cavernoma • Juvenile arteriovenous malfor- from the two vertebral arteries intracranially and
mation • AVM • Myelopathy • Paraplegia • unites in the midline to travel along the anterior
Endovascular embolization • Spinal MRI • sulcus for the entire length of the spinal cord. The
Spinal angiogram • Spinal hemorrhage paired posterolateral spinal arteries originate from
the pre-atlantal part of the vertebral artery or from
the posteroinferior cerebellar artery and run on the
Introduction posterior surface of the spinal cord as a network
for the entire length. The posterior and anterior
Spinal vascular malformations are developmental arterial systems are interconnected at the level of
derangements of the vascular system more specif- the cone via the “basket” anastomosis and via
ically interconnections between the arteries and transverse anastomoses by the pial network of
veins resulting in abnormal flow that may result in the vasocorona. They are reinforced by the ante-
secondary features of vascular enlargement, aneu- rior and posterolateral radiculomedullary arteries
rysm, and varix formation. With the spinal canal at different segmental levels [3]. Most commonly,
being a closed space, the clinical symptoms may the anterior radiculomedullary artery at T9 level is
be due to mass effect from the enlarged vessels, the largest called arteria radiculomedullaris
excessively high flow in the malformation magna aka the Adamkiewicz artery. The anterior
resulting in vascular steal and rarely cardiac fail- radiculomedullary artery follows the spinal nerve
ure, and/or elevated venous pressure with low root to ascend to the segmental level on the ante-
blood flow and back pressure ischemia due to rior surface of the spinal cord and branches in a
venous congestion. typical way with the ascending limb continuing in
the direction of the radicular artery, while the
descending limb does a hairpin curve before join-
Spinal Vascular Anatomy ing the anterior spinal artery forming a diamond-
shaped defect along the anterior spinal axis
Spinal vascular anatomy is highly variable but [1]. The substance of the spinal cord is supplied
predictable. Segmental arteries arising from vari- by the intrinsic network of vessels that include
ous sources that include the vertebral and deep sulco-commissural arteries arising from the ante-
cervical arteries in the neck, the posterior inter- rior spinal artery and perforating arteries arising
costal arteries in the thorax, and the lumbar arter- from the circumferential vasocorona derived from
ies in the abdomen provide vascular supply to the the anterior and paired posterolateral arteries [2].
46 Spinal Vascular Malformations and Treatment 1107

The rich longitudinal and transverse anastomosis cavernous malformations and vascular tumors
protects the spinal cord from ischemia. Extradural added for completeness:
interconnections between segmental arteries also
provide rich collateral network. Type I: Dural arteriovenous fistula (dAVF)
The venous drainage of the spinal cord is via Type II: Intramedullary arteriovenous malformation
the radially arranged intrinsic spinal cord veins Type III: Juvenile AVM
and superficial fine network of vessels along Type IV: Intradural perimedullary AVF
with the longitudinal veins that form the extrinsic Intramedullary cavernous malformations and spi-
spinal cord veins. These veins more or less follow nal vascular tumors include hemangioma,
the arteries (i.e., the anterior and posterior median hemangioblastoma, metastatic tumors, aneurys-
spinal vein) but have many anastomoses that cre- mal bone cyst, osteoblastoma, angiosarcoma,
ate a network with commonly more than one hemangiopericytoma, angiofibroma,
anterior and posterior vein [3]. They may also angiolipoma, and hemangioendothelioma.
use the roots as a vehicle to reach the epidural
plexus and the extraspinal veins and plexus with a Spinal vascular malformations are variable in
reflux-impeding mechanism within the dura mater their epidemiology, clinical presentation and mor-
[4]. It is important to note that the transition of a phology, imaging characteristics, natural history,
median vein into a radicular vein shows the same and management. In general the term angioarch-
hairpin shape as the artery. At the superior cervical itecture signifies the nature of the feeding arteries
part, they can run through the occipital foramen to (radiculomeningeal versus radiculopial or
connect to the vertebral plexus or the inferior radiculomedullary), the morphology of the spinal
dural sinus. Drainage of blood from the spine AV shunt proper (nidus versus fistula, intranidal
occurs through the valveless internal and external aneurysms, etc.), and the nature of the draining
venous vertebral plexus, which is connected to the veins (medullary, perimedullary, radicular, epidu-
azygos and hemiazygos venous systems [5]. ral, or parachordal). The morphology of a high-
flow vascular malformation and the nature of the
draining veins may change with time as the
Spinal Vascular Malformation shunting causes increasing venous ectasias,
Classification rerouting of blood, changes in the intranidal archi-
tecture, or recruitment of additional arterial
Several classification schemes have been pro- feeders, and this usually is associated with
posed for spinal vascular malformations change in clinical symptoms and imaging
[6–8]. Genetically they are classified as (1) hered- characteristics.
itary, (2) genetic nonhereditary, and (3) sporadic
[9]. Hereditary lesion like hereditary hemorrhagic
telangiectasia caused by mutation in the vascular Type I: Dural Arteriovenous Fistula
germ cell results in multifocal vascular
malformations including spinal vascular A spinal dAVF is an abnormal communication
malformation along with cutaneous capillary between the radiculomeningeal artery in the
malformations. Genetic nonhereditary lesions nerve root sleeve and a radicular vein with retro-
caused by somatic mutations share developmental grade flow into the perimedullary venous system,
metameric links, such as the spinal arteriovenous causing venous hypertension and congestion of
metameric syndromes that include Cobb syn- the spinal cord. The general belief is that the
drome, Klippel-Trenaunay syndrome, and Parkes glomerulus of Manelfe, a ball-like structure, is
Weber syndrome. Sporadic malformations believed to regulate regional venous and CSF
include most other spinal vascular malformations. pressures and to be the site of fistula formation
The following four-type system is the clini- [10]. The fistula typically consists of a single hole,
cally relevant and most commonly used, with even when multiple feeders produce a “busy”
1108 S. Paramasivam

appearance suggestive of a nidus. The fistulous diagnosis: 23 months [16]. The site of the fistula
connection may drain centrifugally into the exter- has no relationship with the patient’s symptom-
nal venous paraspinal network, in which case the atology because it is the spinal cord vascular con-
patient may be asymptomatic. However, drainage gestion, rather than fistula itself, that produces the
into a radiculomedullary vein, and the longitudi- clinical syndrome.
nal venous network responsible for draining the The common symptoms include paraparesis,
spinal cord, results in venous congestion of the followed by back pain that may radiate to the
spinal cord. lower legs, hypesthesia, paresthesias, impotence,
and sphincter disturbances [16, 17]. Patients may
Epidemiology report worsening of symptoms with exertion (neu-
Spinal dAVFs are the most frequent spinal vascu- rogenic claudication) or with certain postural
lar malformation, representing approximately changes [15]. Less commonly, patients may have
70 % of all spinal vascular malformations acute onset of symptoms or periodic remissions as
[11]. The exact etiology is unknown and it is the disease progresses. In rare instances, spinal
presumed to be an acquired lesion. Spinal dAVF can present with subarachnoid hemorrhage.
dAVFs have been associated with infection, syrin- Without therapy, spinal dAVF invariably results in
gomyelia, spine trauma, and surgery irreversible paraplegia or even quadriplegia.
[12–14]. Most fistulas affect the thoracolumbar
region with 85 % of lesions below T6 and 100 % Imaging
of lesions below T3 [15]. Most fistulas affect
middle-aged men (40–60 year) with a male/ MRI
female ratio of 5:1. MRI is the screening procedure of choice, and
characteristic findings in spinal dAVF are cord
Pathophysiology edema and dilated perimedullary vessels
Spinal dAVFs are located on the dural sleeve of [16]. T2-weighted (T2-WI) images typically
the spinal nerve root where a radiculomeningeal show poorly marginated centromedullary cord
artery lies in close proximity to the radicular hyperintensity that extends over multiple seg-
vein. The radiculomeningeal artery normally sup- ments [17]. A hypointense rim surrounds the
plies the nerve root and the meninges and may congested cord and most likely reflects deoxygen-
give rise to a radiculomedullary artery that sup- ated blood within the congested capillaries
plies the spinal cord, and the radicular vein drains [18]. There may be contrast enhancement from
into the veins of the spinal canal. Shunting in the chronic venous congestion in the post-
dAVF results in high flow in the valveless spinal gadolinium T1-weighted (TI-WI) images
perimedullary veins through the radicular veins [19]. With disease progression, the cord will
resulting in diminished AV pressure gradient become atrophic. On T2-WI imaging the dilated
thereby impeding the drainage of normal spinal and coiled perimedullary veins appear as flow
veins leading to venous congestion with voids in most cases. Post-gadolinium T1-WI
intramedullary edema. The slow flow in the cap- images are useful in identifying low-flow dAVF
illaries with stasis in the radiculomedullary arter- and differentiating from CSF pulsation artifact by
ies results in chronic hypoxia and progressive a typical salt-and-pepper appearance.
myelopathy. For localization of the fistula, noninvasive
diagnostic techniques, such as contrast-enhanced
Clinical Presentation MR angiography with relative fast acquisition
The clinical symptoms resulting from congestive protocols, have been developed [20, 21]. Apart
myelopathy are not specific, often slowly progres- from identifying the segmental level of the fistula,
sive, and even insidious, so patients present with it is also necessary to localize radiculomedullary
very long histories of vague but progressive arteries especially the Adamkiewicz artery. This
symptoms. Mean duration of symptoms prior to prior knowledge can help to prevent neurologic
46 Spinal Vascular Malformations and Treatment 1109

complications when performing therapeutic pro- fistula (Figs. 1 and 2). At the end of embolization,
cedures. To depict and separate the submillimeter- the collateral circulation of the dural involved
sized spinal cord arteries from the spinal cord region must be examined for the presence of nat-
veins, a simultaneously high spatial and temporal ural arterial collaterals, ipsilateral above and
resolution should be achieved. A large below the level of the fistula as well contralateral
craniocaudal field of view must be used because at the same level of the spinal dAVF. The success
the location of the fistula is highly variable rate of endovascular therapy varies from 25 % to
irrespective of the findings previously obtained 75 % and recurrence is not uncommon. Up to
by conventional MRI. Contrast-enhanced MR 80 % of patients have clinical improvement of
angiography with large field of view can depict various degrees to clinical stabilization following
the spinal cord arteries and veins [21]. embolization. Complications of embolization
include inadvertent closure of the spinal radiculo-
Angiography medullary artery mostly due to failure to recog-
Digital subtraction angiography (DSA) is the gold nize. Inadvertent deposition of liquid embolic
standard test for spinal dAVF [17]. Localization of material into the perimedullary veins beyond the
the spinal dural AVF can be very difficult. Selec- nidal-venous junction can result in thrombosis of
tive angiography of the segmental artery harbor- the spinal cord veins. In cases where endovascular
ing the AVF depicts the early venous filling and embolization is not possible, placement of a metal
retrograde flow of the opacified radiculo- coil in the major feeding artery can facilitate
medullary veins into the spinal canal. An exten- intraoperative fluoroscopic localization of the
sive network of dilated tortuous perimedullary fistula [23].
veins is often visible. When a fistula is found, Surgical ligation is performed by coagulation
the adjacent levels should be imaged as well and division of the intradural radicular vein that
because of the possibility of multiple radicular receives blood from the shunt resulting in visible
feeding arteries; most fistulas are located in the change in venous turgor and color of the
thoracic and lumbar regions. In 10 % of cases, the arterialized venous plexus. This is more invasive
sacral arteries are involved [22] and rarely, intra- but a relatively simple and effective intervention
cranial dural AVFs may drain inferiorly and except for sacral fistulas. The key to successful
mimic spinal dural AVFs clinically and on MRI. treatment is precise localization of the fistula.
Neither the location of the pathologic vessels nor Complete occlusion of the fistula by either
the location of the intramedullary imaging abnor- method stops disease progression, but only
malities on MRI seems to be related to level of the two-thirds of the patients have improvement in
fistula Figs. 1 and 2. motor symptoms, while about one-third show
improvement of their sensory disturbances. In
Management most patients, impotence and sphincter distur-
The natural history of untreated spinal dAVFs is bances do not recover. In patients with no
generally thought to be poor with progressive improvement within 4–6 weeks after treatment,
neurological deficit. The spinal dAVF may be repeat MRI imaging should be done. MRI
treated either by surgery or endovascular changes may or may not completely reverse; if
obliteration. persistent flow voids are visualized, it warrants
In most patients, the fistula may be obliterated another angiographic evaluation.
by super selective catheterization of the feeding
radiculomeningeal artery and obliteration of the
fistula using liquid embolic agents like n-butyl Type II: Intramedullary Arteriovenous
cyanoacrylate (n-BCA) and Onyx. The embolic Malformation
agent must pass the nidus and reach and occlude
the proximal segment of the draining vein to pre- Intramedullary arteriovenous malformation
vent subsequent intradural collateral filling of the (AVM) consists of malformation within the
1110

Fig. 1 Middle-aged man with subacute onset of progressive paraparesis with bladder n-BCA embolization with obliteration of the proximal segment of the vein just distal to
and bowel involvement had hyperintense signal in the MRI T2-WI (arrowhead) with the fistula (Black Arrow in f). Control angiogram reveals total obliteration of the dural
enlarged perimedullary veins (White Arrow) (a). T4 intercostal artery angiogram reveals arteriovenous fistula (g). Three months later MRI reveals significant improvement in
supply to the dural arteriovenous fistula (b–e). Super selective angiogram shows the signal changes in the cord (arrowhead in h) and total obliteration of the dorsal
exact location of the fistula with multiple feeders (Black Arrow). The fistula is treated by perimedullary veins
S. Paramasivam
46
Spinal Vascular Malformations and Treatment

Fig. 2 Right subclavian angiogram shows the cervical dural arteriovenous fistula g) the anatomic location of the fistula in relation to the bone. The embolization
(arrow in a) supplied by a dural branch from thyrocervical trunk. Super selective performed with n-BCA penetration into the proximal portion of the vein just distal to
angiograms (b–e) reveal the anatomic architecture of the dural fistula. Arrowhead in the fistula (h) (Arrow). Control angiogram reveals total obliteration of the fistula (i).
(c) shows the perimedullary venous drainage that drains intracranially. Arrow in (d, e) Posttreatment MRI reveals complete obliteration of the fistula as evidenced by absence
represents the proximal segment of the vein just distal to the fistula that needs to be of enlarged perimedullary veins (J)
targeted for embolization. Intra-procedural DYNA CT with contrast injection shows (f,
1111
1112 S. Paramasivam

substance of the spinal cord supplied by the to the intracranial cavity and may cause
radiculomedullary and radiculopial arteries and headaches and disturbances of consciousness.
drained by the pial veins. They include spinal Rehemorrhage is more common with spinal
cord glomerular AVMs with compact or diffuse AVMs than in the brain. It constitutes about
nidus and AV fistulas that may be intramedullary 10 % in the first month and 40 % in the first year
or perimedullary in location. after the event [28]. In adults, the correlation
between angioarchitecture and hemorrhage is
Epidemiology found as suggested by arterial, nidal aneurysm,
Intramedullary AVM is the second most common pial venous ectasia and congestion, and associated
spinal vascular lesion, accounting for about 20 % arteriovenous fistulas.
of spinal vascular lesions. There is slight male Progressive symptoms are mostly nonspecific
predominance and they commonly come to with features of hypesthesia, paresthesia, weak-
clinical attention in the second and third ness, bladder and bowel dysfunction, impotence,
decade of life [24]. They are most commonly and diffuse back and muscle pain. These symp-
located in the thoracolumbar region followed by toms can have a stuttering course sudden worsen-
the cervical region. Syndromes associated ing followed by some improvements over time.
with intramedullary spinal AVMs include neuro- The causes for deterioration are multiple. Among
fibromatosis and the Rendu-Osler-Weber, the well-accepted causes are intraspinal hemor-
Klippel-Trenaunay-Weber, and Parkes Weber rhage, thrombosis within the AVM, arachnoiditis,
syndromes [25]. and increased venous pressure. AVMs may
become symptomatic by venous congestion
Pathophysiology alone, in the absence of intraparenchymal or sub-
Glomerular spinal AVMs (nidal AVMs) are more arachnoid hemorrhage [29].
frequent of spinal cord AVMs. They are charac- Location of the AVM does have influence on
terized by a vascular nidus located intramedullary the clinical presentation: cervical lesions typically
and have multiple feeding arteries derived from present as progressive sensory deficits, whereas
both the anterior and posterior spinal arterial sys- thoracic AVMs most commonly present as hem-
tems. They drain into the dilated spinal cord veins. orrhage (hematomyelia or subarachnoid hemor-
They are distributed throughout the spinal cord: rhage) and often show progressive myelopathy.
about 30 % in the cervical region but most com- Lesions of the conus and filum terminale
monly found in the thoracolumbar region com- manifest as progressive myelopathy or acute
prising of 70 % that includes conus medullaris nonhemorrhagic paraplegia.
AVMs [24], a special category of spinal cord Aggravating factors including maneuvers that
AVMs that are attributed to an abnormality during increase intra-abdominal or intrathoracic pressure
neurulation and may be associated with a tethered such as the Valsalva maneuver and bending often
cord [26]. Conus medullaris AVMs possess mul- increase medullary venous pressure. Pregnancy is
tiple arterial feeders and are extensive. known to aggravate symptomatology by increas-
ing the intra-abdominal pressure, increased intra-
Clinical Presentation vascular volume, hormonal changes, and added
Spinal AVMs may present acutely in most cases. stress on the venous circulation during delivery.
Hemorrhage is the most common presentation and Trauma as a precipitating factor for symptoms is
more so in children than adults [15, 27]. It may be not well supported.
either subarachnoid or intraparenchymal hemor- Apart from neurologic symptoms, spinal
rhage. The typical syndrome of spinal hemorrhage deformities, and complications of the spinal
gives rise to severe pain, which frequently starts in cord, dysfunctions including urinary tract infec-
the back and rapidly spreads to the rest of the back tions, respiratory infections, and decubitus ulcer-
and nuchal area and to the legs. In severe hemor- ations are taken into consideration in determining
rhage in the cervical region, the blood can spread the morbidity and final outcome of these patients.
46 Spinal Vascular Malformations and Treatment 1113

Imaging Management
The natural history of spinal AVMs is unknown.
MRI In general, symptomatic spinal AVMs presenting
On MRI the glomerular spinal AVMs show focal with hemorrhage or progressive neurologic deficit
dilatation of the cord in the region of the lesion warrant treatment to ameliorate symptoms and
with conglomerate dilated, perimedullary, and improve the outcome. Due to the eloquence of
intramedullary vessels. There may be an area of spinal cord and complex nature of spinal AVM
low signal intensity around the nidus on T1-WI architecture, any modality of treatment procedure
and T2-WI corresponding to hemosiderin deposi- has a relatively higher complication risk. Manage-
tion. The enlarged vessels appear as flow voids on ment strategies have to be individualized based on
T2-WI imaging and as mixed hyperintense to the available expertise.
hypointense tubular structures on T1-weighted The therapy of choice for all spinal cord AVMs
(T1-WI) imaging (depending on their flow veloc- is endovascular embolization. Embolization is
ity and direction). The edema due to venous con- performed after careful analysis of the AVM by
gestion appears as cord swelling with selective spinal angiography and by using an
intramedullary hyperintensity on T2-WI embolic agent appropriate to the specific
[29]. Intraparenchymal hemorrhages demonstrate angioarchitecture. Proximal arterial occlusion
variable signal intensity on T1-WI and T2-WI will not obliterate the fistula, because of collateral
based on the age of hemorrhage. recanalization from the radiculopial network.
MRI plays a significant role in defining the Obliteration of proximal venous segment should
location of the AVM in relation to the spinal be the target of embolization. In most cases partial
cord and dura and can also distinguish between embolization is achieved with complete oblitera-
low-flow and high-flow forms of AVMs. As the tion rates ranging from 25 % to 30 % [28, 32]. Par-
small arterial feeders and draining veins escape tial obliteration may be an adjunct to surgery; in
detection on non-contrast MRI, contrast- general, partial embolization improves the out-
enhanced fast MR angiography is capable of come, with reduced rehemorrhage risk and
detecting the main arterial feeder of improved prognosis in most patients. Transient
glomerular-type AVMs. Contrast-enhanced MRI and permanent complication rates following
is useful to detect subtle venous dilatations embolization range from 10.6 % to 14 %
[30, 31]. [28, 32] (Figs. 3 and 4).
Surgical management requires appropriate
Angiography case selection. Surgically accessible lesions with
Catheter angiography is the gold standard evalu- compact nidus can be completely resected with a
ation for spinal cord AVMs [24]. A complete cure rate of up to 94 % of cases [33].
angiogram characterizes all feeding vessels, Radiosurgery may be helpful in selected
nidal configuration, flow characteristics, patients as preliminary reports are encouraging.
intranidal aneurysms, and venous drainage with However, it carries a risk of collateral acute and
or without venous ectasias. It is essential for treat- chronic radiation-induced injury to the surround-
ment plan. Selective catheterization and injection ing spinal cord [34].
of numerous arteries are essential to fully charac-
terize a spinal AVM. The feeding vessels may
arise from respective or distant radiculomedullary Type III: Juvenile Arteriovenous
arteries supplying the anterior or posterior spinal Malformation
arteries. The sources as far afield as the occipital,
ascending pharyngeal, vertebral, ascending and Type III lesions aka spinal arteriovenous
deep cervical, supreme intercostal, intercostal, metameric syndrome (SAMS) are complex
lumbar, and lateral and median sacral arteries AVMs involving structures derived from two or
[28] (Figs. 3 and 4). more embryonic layers. These AVMs involve the
1114

Fig. 3 Intramedullary AVM with perimedullary venous drainage cranially (a–c) has perimedullary venous system (arrow in g). Selective partial embolization of the AVM
gliotic changes in the spinal cord. DSA reveals supply from the left vertebral artery was performed and control angiogram reveals significant reduction in the supply to the
through anterior and posterolateral spinal arteries (d) and dominant supply from the left malformation from the thyrocervical trunk with preservation of the anterior spinal axis
radiculomedullary artery arising from the thyrocervical trunk supplying the anterior (h). There is residual supply to the malformation from the left vertebral artery (i)
spinal artery (e, f). The AVM has a dominant cranial venous drainage into the
S. Paramasivam
46 Spinal Vascular Malformations and Treatment 1115

Fig. 4 Young patient with cervical intramedullary AVM Note the venous ectasia (arrow in c). Super selective
with fistula presented with cervical subarachnoid hemor- angiogram reveals the fistulous component of the AVM
rhage due to perimedullary large venous ectasia. DSA with venous ectasia (e, f). Embolization of the AVM
examination shows anterior spinal artery from the right performed with n-BCA, which is deposited in the AVM
vertebral artery supplying part of the AVM (a, b). The and ectatic venous sac (g, h). Post-embolization control
dominant supply to the AVM is from the posterolateral angiogram reveals complete obliteration of the AVM (i–l)
spinal artery arising from the left vertebral artery (c, d).

spinal cord, bony structures, paraspinal Epidemiology


musculature, subcutaneous tissue, and skin in Type III lesions constitute about 6 % of all spinal
the same segment. The portion of the nidus vascular malformations. They are equally located
involving the spinal cord typically has neural tis- in the cervical, thoracic, and lumbar or conus
sue within its interstices. Cobb syndrome is by medullaris regions. There is a 2:1 male to female
characterized skin, bone, and spinal cord involve- ratio. They come to clinical attention in childhood
ment aka cutaneous vertebral medullary and early adulthood. They may be multiple in as
angiomatosis. high as 19 % of patients. They may be associated
1116 S. Paramasivam

with Klippel-Trenaunay and Parkes Weber Management


syndromes [28]. These lesions are extremely difficult to treat. The
management of each individual case is tailored
Pathophysiology based on the presentation. Endovascular emboli-
Type III lesions are AVMs that involve derivatives zation can be targeted at spinal cord lesions if
of all embryonic layers and consist of extensive symptomatic as done for nonmetameric lesions.
vascular malformation embedded within the tis- Extraspinal lesions are treated by embolization if
sue. The segmented mesodermal origin of the needed; the key is to understand the pathophysi-
endothelial cells establishes the topographic link ology of the clinical presentation and preserve the
between these segmentally related targets. The spinal cord vascular supply arising from the
spinal cord AVM is metameric if the myelomere radiculomedullary arteries. Although resection of
involved corresponds to that of the nerve the lesion by combining staged embolization
involved. The extraspinal and limb lesions are followed by surgery has been reported, it is asso-
usually high-flow arteriovenous fistulas supplied ciated with fatal complications and such an
by the segmental arteries like the intercostal or approach is not generally aimed for [35, 36].
lumbar arteries and corresponding segmental
arteries. The cutaneous lesions are fistulas of cap-
illary or arterial type with high-flow Type IV: Intradural Perimedullary
characteristics. Arteriovenous Fistula

Clinical Features Type IV lesions are intradural perimedullary AV


The clinical presentation may be due to spinal cord, fistulas located on the pial surface of the spinal
paraspinal, limb, or cutaneous lesions. The typical cord, usually on the anterior or lateral surface.
presentation is in children or young adults with pain They consist of a fistula between a spinal cord
and/or myelopathy [28]. Spinal cord involvement artery or arteries and the coronal venous plexus,
may cause progressive myeloradiculopathy due to and there is often ectasia of the vein at the artery-
venous congestion, compression due to enlarged to-vein transition site. They are a spectrum of pial
medullary veins, and spinal hemorrhage. The AV malformations ranging from the most com-
paraspinal vascular malformations can present with mon single giant fistulae supplied by anterior or
steal syndrome due to high-flow AVF. The cutane- posterolateral spinal arteries to larger, multiple-
ous vascular malformations are readily apparent and vessel fistulae supplied by the anterior and pos-
often present with a bruit. The comorbid conditions terolateral spinal arteries.
such as spinal deformities such as kyphosis and
scoliosis along with complications common to spi- Epidemiology
nal cord dysfunction such as urinary tract infections, Type IV lesion accounts are detected in relatively
respiratory infections, and decubitus ulcerations young patients (20–25 years) without sex predom-
must be taken into consideration in determining inance and account for 20 % of all spinal vascular
the morbidity and final outcome of these patients. lesions [15, 37]. An association with split cord
malformation and with Rendu-Osler-Weber and
Imaging Cobb syndrome is reported [38].
Multimodality imaging is the key in complete
analysis of these types of malformations. MRI is Pathophysiology
essential to look for the anatomic extent of the Type IV lesions occur most commonly at the
disease, the cord edema, and the intradural venous thoracolumbar junction, particularly at the
engorgement. Catheter angiography assessment conus, and, to a lesser extent, in the upper cervical
can be difficult as only a part of the lesion can be region [25]. Fistulous AVMs are characterized by
seen with injection of contrast into any particular direct AV shunts located superficially on the spi-
artery (Fig. 5). nal cord. There is usually no intramedullary
46 Spinal Vascular Malformations and Treatment 1117

Fig. 5 (continued)
1118 S. Paramasivam

Fig. 5 Type III juvenile metameric AVM involving pre- around with feeders arising from vertebral arteries (d–g),
dominantly the mesodermal element. The child presented dorsal (arrow) and ascending cervical artery (arrowhead)
with progressive weakness of all four limbs with predom- (h, i), and external carotid artery branches (j). Partial
inant lower motor neuron-type weakness in the upper embolization with Onyx and n-BCA (k, l) to reduce the
limbs with sensory disturbances. MRI revealed a meta- venous congestion was performed and posterior surgical
meric AVM with venous congestion of the cord with decompression to relieve pressure on the cord was
edema (b, c). The cervical spine X-ray reveals bony performed (o). Residual flow to the malformation is visu-
destruction due to AVM (a). The DSA examination reveals alized in the angiogram (m, n). Patient’s condition
supply to the AVM in the vertebra and the soft tissue stabilized

component and no intervening nidus. Fistulous development of large variceal pouches due to
spinal AVMs fed by the ventral spinal arterial high shunt volume. They may be associated with
system are subpial while fistulous spinal AVMs hereditary hemorrhagic telangiectasia (HHT).
fed by the dorsal spinal arterial system are sub-
arachnoid in location. Fistulous spinal AVMs may Clinical Features
be in micro- and macrofistulous forms. The clinical presentation depends on the micro- or
Microfistulous AVMs (mAVFs) comprise macrofistulous forms and the location. The
about 75 % and have low shunt volume and are mAVFs present during adulthood; the thoracic
mostly seen in adults, fed by the radiculo- mAVFs present with hemorrhage (two-third
medullary arteries and drained by the superficial intraparenchymal and one-third subarachnoid);
perimedullary veins. cervical, conus, and cauda equina mAVFs present
Macrofistulous AVMs (MAVFs) are typically with slowly progressive, asymmetric myeloradi-
found in children due to high-flow nature comes culopathy of the conus and cauda equina [37]. The
to clinical attention much earlier in life. There is MAVFs present earlier in life and the common
remodeling of the arteries and veins including presentation is progressive myeloradiculopathy.
46 Spinal Vascular Malformations and Treatment 1119

Imaging walled capillaries and sinusoids with no interven-


MRI characteristically demonstrates large flow ing parenchymal tissue, without the identifiable
voids [38] that comprise the enlarged arterial feeder feeding artery or specific draining vein.
and the dilated perimedullary venous channels and
associated cord changes. However, low-flow Epidemiology
microfistulous forms may not be apparent on regu- Cavernomas are distributed along the entire
lar MRI [39], while contrast-enhanced fast MR neuraxis and represent 5–12 % of all spinal vas-
angiography is capable of detecting the feeding cular lesions [42]. Women are affected more than
arteries. Apparent diffusion coefficient (ADC) men with a ration of 2:1. They usually come to
values may be reduced, indicating edema, and it clinical attention in the middle age with a peak in
may normalize after treatment [40]. Catheter angi- the fourth decade but may be found in all age
ography is the gold standard and should be done if a groups. Familial occurrence, coexistent meta-
type IV lesion is suspected but not seen on MRI. meric vascular nevi, and concurrent intracranial
Angiography is necessary to make a clear diagnosis lesion are described in as many as 40 % of patients
and understand the architecture of a perimedullary [43]. As in the brain, they are frequently associ-
fistula and plan therapy (Figs. 6 and 7). ated with a venous angioma. The thoracic spine is
more often affected than the cervical spine.
Management
Type IV fistulous lesions are rare, and estimations Pathophysiology
of the natural history of untreated lesions suggest Spinal cavernomas may cause symptoms acutely
progression from myelopathy to paraplegia within by intramedullary or subarachnoid hemorrhage or
5–7 years, and a high incidence of repeated hemor- chronically with progressive myelopathy
rhage in patients presenting with hemorrhage [15, resulting from lesion expansion by capillary dila-
25, 37]. Prompt diagnosis and treatment of tation or repetitive small hemorrhages in or
perimedullary fistulae is mandatory with the objec- around the lesion or due to toxic effects of degra-
tive of treatment being occlusion of the fistula. Most dation products of the intramedullary blood.
lesions can be effectively obliterated; endovascular
embolization targeted at the arteriovenous transition Clinical Presentation
leads to complete exclusion of the lesion with relief Spinal cavernomas may cause acute, recurrent,
of the venous congestion and clinical improvement and/or progressive myelopathy. The acute clinical
(Figs. 6 and 7). In cases where the embolization presentation is usually acute pain followed by
appears to be associated with higher risk or is sensorimotor deficit indicating new hemorrhage
deemed to be technically infeasible because of within or around the lesion. Slowly progressive
small feeders or tortuous long feeding arteries sensorimotor deficit and bladder and bowel dis-
such as in filum terminale lesions, surgery should turbance likely reflect repeated episodes of minor
be considered. The biggest challenge with surgery hemorrhage or toxic effects of blood products on
is the localization of the fistulous location, which is the surrounding spinal cord tissue. The course of
usually embedded, in the congested veins. symptoms is highly variable and the natural his-
The clinical outcome appears to be somewhat tory is far from clear. Assessments of the annual
variable, with partial neurological improvement or rate of symptomatic rehemorrhage rates range
no change occurring in the majority of patients, and from 0 % to 66 % [44].
worsening in a small minority of patients [37, 41].
Pathology
Grossly, spinal cavernomas are discrete, well-
Cavernous Malformation (Cavernomas) circumscribed, lobulated, red-purple lesions.
Microscopically, they are identical to those in the
Spinal cord cavernomas are intramedullary vascu- brain and are composed of dilated, thin-walled
lar malformations composed of dilated thin- capillaries that have a simple endothelial lining
1120 S. Paramasivam

Fig. 6 Child with


paraplegia revealed a spinal
pial arteriovenous fistula
with dilated perimedullary
vein (a). DSA examination
revealed an enlarged
radicular artery and anterior
spinal artery that supply the
fistula on the pial surface
(b) with enlarged
perimedullary veins
draining into the veins
intracranially (c). Super
selective catheterization of
the fistula (d) with coiling
of the venous sac (e) to
reduce the flow in the fistula
followed by embolization
of the fistula using n-BCA
(f) resulted in successful
obliteration of the fistula

with variably thin fibrous adventitia and may have well-defined, circumscribed lesions of varying
calcifications. In the spinal cord around the size. On T2-WI they characteristically display a
cavernoma, residua of previous hemorrhage may hypointense rim due to susceptibility artifacts
be present, including scarring, collections of from hemosiderin deposits [45].
hemosiderin-laden macrophages, variable degrees The inhomogeneous central core represents
of gliosis, and edema. hemorrhage in different stages of evolution giving
a “popcorn” appearance to the center [42]. The
Imaging size of the cavernoma and its precise relation to
CT is of limited value as in most other spinal the surface of the cord is best visualized on
pathologies; only acute hemorrhagic lesions and T1-WI, and T2-WI tend to overestimate the size
calcified lesions are detected and others remain of the cavernoma, because of the hemosiderin
indetectable. On MRI, cavernomas appear as ring. If multiple, cavernomas are readily picked
46 Spinal Vascular Malformations and Treatment 1121

Fig. 7 Young boy with progressive pial arteriovenous axis to supply the fistula (e, f). Super selective catheteriza-
fistula (a–c) was treated surgically initially with proximal tion up to the fistulous point and coil embolization of the
occlusion of the radicular artery at T4 level (clip in d). He fistula were performed with preservation of the anterior
had recurrence of the symptoms, as the fistula was not spinal axis (g, h). Follow-up angiogram 6 months later
treated. Repeat angiogram reveals the reconstitution of reveals persistent obliteration of the fistula with preserva-
the anterior spinal artery from above and below along the tion of the anterior spinal axis (i–k)

up with MRI. Spinal cord cavernomas may also Surgical resection is the treatment of choice and
mimic spinal cord tumors such as glioma, can be performed safely using meticulous micro-
ependymoma, or anaplastic astrocytoma, espe- surgical technique. Careful localization,
cially if they are large (Fig. 8). myelotomy, and meticulous dissection are key to
The current noninvasive imaging techniques good functional outcome. Endovascular treat-
cannot distinguish acute spinal cord hemorrhage ments of cavernomas are inappropriate.
due to solitary cavernoma or a micro-AVM. In
such a situation, in spite of the low yield, spinal
angiography is essential to pick up AVM, as Summary
cavernomas are angiographically occult.
Spinal vascular malformations are a group of var-
Management ied disorders due to developmental derangement
Asymptomatic cavernomas are usually followed of the vascular system that may present acutely
by serial imaging studies and not treated. The due to hemorrhage or subacutely due to venous
natural history is highly variable and the consen- hypertension or vascular steal with nonspecific
sus is to treat the symptomatic cavernomas. neurologic symptomatology making diagnosis
1122 S. Paramasivam

Fig. 8 Cervical spinal cord


cavernoma that came to
clinical attention due to
hemorrhage. T2-WI
displays a hypointense rim
due to susceptibility
artifacts from hemosiderin
deposits. The central core
represents hemorrhage in
different stages of evolution
(a–c)

challenging for neurologists, neurosurgeons, and need treatment as the natural history is suggestive
neuroradiologists. Spinal vascular malformations of progressive stepwise neurological deteriora-
are rare but treatable causes of progressive spinal tion. Early diagnosis and timely management in
cord symptoms. The pathophysiology of many symptomatic patients may result in improvement
spinal vascular malformations is determined by or stabilization of clinical conditions. Treatment
their venous drainage. Elevated intravenous pres- should be performed in specialized centers.
sure impedes blood flow, leading to stasis, pro- Except spinal cord cavernomas, all other vascular
gressive venous congestion, congestive venous disorders of the spine are initially managed by
edema, and cord ischemia. endovascular embolization as it is the least inva-
When spinal vascular diseases are suspected, sive but technically demanding. For cases that fail
MRI is the first diagnostic modality to identify the embolization, surgical treatment is an available
lesion and rule out potential differential diagnoses option. In select cases a combined therapy might
like acute cord compression, tumor, degenerative be sensible.
diseases of the spine, myelitis, etc. Initial diagno-
sis depends on demonstration of pathologic flow
voids, cord edema, or spinal hemorrhage and pro- References
vides an initial assessment of which malformation
1. Thron A (1988) Vascular anatomy of the spinal cord:
to expect. Contrast-enhanced fast-spin echo MR neuroradiological investigations and clinical syn-
angiography of the spinal cord constitutes a help- dromes. Springer, Berlin
ful adjunct to noninvasive diagnostics because it 2. Lasjaunias PL, Berenstein A, ter Brugge K (2004)
may demonstrate pathologic AV shunts and help Surgical neuroangiography. Clinical vascular anatomy
and variations, vol 1, 2nd edn. Springer, Berlin
to guide selective spinal arteriography. 3. Grunwald I, Thron A, Reith W (2001) Spinal angiog-
When neurologic symptoms and MRI suggest raphy: vascular anatomy, technique and indications.
a vascular malformation, spinal angiography is Radiologe 41:961–967 [in German]
the next and gold standard test to define the type 4. Thron A, Otto J, Schroeder JM (1990) Functional
anatomy of the dural segment of spinal cord draining
of vascular malformation and decide on the veins: a histological and microangiographic study. In:
appropriate therapy. Most spinal vascular du Boulay G (ed) Symposium neuroradiologicum.
malformations that come to clinical attention Springer, London, p 323
46 Spinal Vascular Malformations and Treatment 1123

5. Mull M, Thron A (2006) Spinal infarcts. In: von 21. Lee TT, Gromelski EB, Bowen BC, Green BA (1998)
Kummer R, Back T (eds) Magnetic resonance imaging Diagnostic and surgical management of spinal dural
in ischemic stroke. Springer, Berlin, pp 251–267 arteriovenous fistulas. Neurosurgery 43:242–246, dis-
6. Borden JA, Wu JK, Shucart WA (1995) A proposed cussion 246–247
classification for spinal and cranial dural arteriove- 22. Partington MD, Rufenacht DA, Marsh WR, Piepgras
nous fistulous malformations and implications for DG (1992) Cranial and sacral dural arteriovenous fis-
treatment. J Neurosurg 82:166–179. doi:10.3171/ tulas as a cause of myelopathy. J Neurosurg
jns.1995.82.2.0166 76:615–622. doi:10.3171/jns.1992.76.4.0615
7. Spetzler RF, Detwiler PW, Riina HA, Porter RW 23. Britz GW, Lazar D, Eskridge J, Winn HR (2004)
(2002) Modified classification of spinal cord vascular Accurate intraoperative localization of spinal
lesions. J Neurosurg 96:145–156 dural arteriovenous fistulae with embolization coil:
8. Zozulya YP, Slin’ko EI, Al Q II (2006) Spinal arterio- technical note. Neurosurgery 55:252–254, discussion
venous malformations: new classification and surgical 254–255
treatment. Neurosurg Focus 20:E7 24. Niimi Y, Berenstein A (1999) Endovascular treatment
9. Krings T et al (2007) Imaging in spinal vascular dis- of spinal vascular malformations. Neurosurg Clin N
ease. Neuroimaging Clin N Am 17:57–72. Am 10:47–71
doi:10.1016/j.nic.2007.01.001 25. Djindjian M, Djindjian R, Hurth M, Rey A, Houdart R
10. Manelfe C, Lazorthes G, Roulleau J (1972) Arteries of (1977) Spinal cord arteriovenous malformations and
the human spinal dura mater. Acta Radiol Diagn the Klippel-Trenaunay-Weber syndrome. Surg Neurol
13:829–841 8:229–237
11. Gilbertson JR, Miller GM, Goldman MS, Marsh WR 26. Hurst RW, Bagley LJ, Marcotte P, Schut L, Flamm ES
(1995) Spinal dural arteriovenous fistulas: MR and (1999) Spinal cord arteriovenous fistulas involving
myelographic findings. AJNR Am J Neuroradiol the conus medullaris: presentation, management,
16:2049–2057 and embryologic considerations. Surg Neurol
12. Finsterer J, Bavinzski G, Ungersbock K (2000) Spinal 52:95–99
dural arteriovenous fistula associated with syringomy- 27. Tobin WD, Layton DD (1976) The diagnosis and nat-
elia. J Neuroradiol 27:211–214 ural history of spinal cord arteriovenous
13. Asakuno K, Kim P, Kawamoto T, Ogino M (2002) malformations. Mayo Clin Proc 51:637–646
Dural arteriovenous fistula and progressive conus 28. Berenstein A, Lasjaunias P, TerBrugge KG (2004)
medullaris syndrome as complications of lumbar Surgical neuroangiography: clinical and endovascular
discectomy. Case report. J Neurosurg 97:375–379 treatment aspects in adults, vol 2.2, 2nd edn. Springer-
14. Vankan Y, Demaerel P, Heye S, Van Calenbergh F, van Verlag, Germany, pp 737–872
Loon J, Maleux G, Wilms G (2004) Dural arteriove- 29. Kataoka H, Miyamoto S, Nagata I, Ueba T, Hashimoto
nous fistula as a late complication of upper cervical N (2001) Venous congestion is a major cause of neu-
spine fracture. Case report. J Neurosurg 100:382–384 rological deterioration in spinal arteriovenous
15. Rosenblum B, Oldfield EH, Doppman JL, Di Chiro G malformations. Neurosurgery 48:1224–1229, discus-
(1987) Spinal arteriovenous malformations: a compar- sion 1229–1230
ison of dural arteriovenous fistulas and intradural 30. Doppman JL, Di Chiro G, Dwyer AJ, Frank JL,
AVM’s in 81 patients. J Neurosurg 67:795–802. Oldfield EH (1987) Magnetic resonance imaging of
doi:10.3171/jns.1987.67.6.0795 spinal arteriovenous malformations. J Neurosurg
16. Atkinson JL et al (2001) Clinical and radiographic 66:830–834. doi:10.3171/jns.1987.66.6.0830
features of dural arteriovenous fistula, a treatable 31. Thron A, Mull M, Reith W (2001) Spinal arteriovenous
cause of myelopathy. Mayo Clin Proc 76:1120–1130. malformations. Radiologe 41:949–954
doi:10.4065/76.11.1120 32. Rodesch G, Lasjaunias P, Berenstein A (1993) Embo-
17. Koch C, Kucinski T, Eckert B, Rother J, Zeumer H lization of arteriovenous malformations of the spinal
(2003) Spinal dural arteriovenous fistula: clinical and cord. In: Valavanis A (ed) Interventional neuroradiol-
radiological findings in 54 patients. Rofo ogy. Springer, Berlin, p 135
175:1071–1078. doi:10.1055/s-2003-40925 33. Connolly ES Jr, Zubay GP, McCormick PC, Stein BM
18. Hurst RW, Grossman RI (2000) Peripheral spinal cord (1998) The posterior approach to a series of glomus
hypointensity on T2-weighted MR images: a reliable (type II) intramedullary spinal cord arteriovenous
imaging sign of venous hypertensive myelopathy. malformations. Neurosurgery 42:774–785, discussion
AJNR Am J Neuroradiol 21:781–786 785–776
19. De Marco JK, Dillon WP, Halback VV, Tsuruda JS 34. Sinclair J, Chang SD, Gibbs IC, Adler JR Jr
(1990) Dural arteriovenous fistulas: evaluation with (2006) Multisession CyberKnife radiosurgery for
MR imaging. Radiology 175:193–199. doi:10.1148/ intramedullary spinal cord arteriovenous malformations.
radiology.175.1.2315480 Neurosurgery 58:1081–1089. doi:10.1227/01.
20. Huffmann BC, Gilsbach JM, Thron A (1995) Spinal NEU.0000215891.25153.BA, discussion 1081–1089
dural arteriovenous fistulas: a plea for neurosurgical 35. Menku A, Akdemir H, Durak AC, Oktem IS (2005)
treatment. Acta Neurochir 135:44–51 Successful surgical excision of juvenile-type spinal
1124 S. Paramasivam

arteriovenous malformation in two stages following coefficient values – case report. Neurol Med Chir
partial embolization. Minim Invasive Neurosurg 46:559–562
48:57–62. doi:10.1055/s-2004-830184 41. Hida K, Iwasaki Y, Goto K, Miyasaka K, Abe H (1999)
36. Spetzler RF, Zabramski JM, Flom RA (1989) Manage- Results of the surgical treatment of perimedullary arte-
ment of juvenile spinal AVM’s by embolization and riovenous fistulas with special reference to emboliza-
operative excision. Case report. J Neurosurg tion. J Neurosurg 90:198–205
70:628–632. doi:10.3171/jns.1989.70.4.0628 42. Deutsch H, Jallo GI, Faktorovich A, Epstein F (2000)
37. Mourier KL, Gobin YP, George B, Lot G, Merland JJ Spinal intramedullary cavernoma: clinical presentation
(1993) Intradural perimedullary arteriovenous fistulae: and surgical outcome. J Neurosurg 93:65–70
results of surgical and endovascular treatment in a 43. Cohen-Gadol AA, Jacob JT, Edwards DA, Krauss WE
series of 35 cases. Neurosurgery 32:885–891, discus- (2006) Coexistence of intracranial and spinal cavern-
sion 891 ous malformations: a study of prevalence and natural
38. Halbach VV, Higashida RT, Dowd CF, Fraser KW, history. J Neurosurg 104:376–381. doi:10.3171/
Edwards MS, Barnwell SL (1993) Treatment of giant jns.2006.104.3.376
intradural (perimedullary) arteriovenous fistulas. Neu- 44. Sandalcioglu IE, Wiedemayer H, Gasser T, Asgari S,
rosurgery 33:972–979, discussion 979–980 Engelhorn T, Stolke D (2003) Intramedullary spinal
39. Dillon WP, Norman D, Newton TH, Bolla K, Mark A cord cavernous malformations: clinical features and
(1989) Intradural spinal cord lesions: Gd-DTPA- risk of hemorrhage. Neurosurg Rev 26:253–256.
enhanced MR imaging. Radiology 170:229–237. doi:10.1007/s10143-003-0260-2
doi:10.1148/radiology.170.1.2909101 45. Weinzierl MR, Krings T, Korinth MC, Reinges MH,
40. Inoue T, Takahashi T, Shimizu H, Matsumoto Y, Gilsbach JM (2004) MRI and intraoperative findings
Takahashi A, Tominaga T (2006) Congestive in cavernous haemangiomas of the spinal cord.
myelopathy due to cervical perimedullary arteriove- Neuroradiology 46:65–71. doi:10.1007/s00234-003-
nous fistula evaluated by apparent diffusion 1072-3
Spinal Cord Infarction and Differential
Diagnosis 47
Srikanth R. Boddu, Alessandro Cianfoni, Kyung-Wha Kim,
Mohammad Amin Banihashemi, Emanuele Pravatà,
Y. Pierre Gobin, and Athos Patsalides

Contents Ischemic Spinal Cord Infarction . . . . . . . . . . . . . . . . . . . 1132


Hemorrhagic Spinal Cord Infarction . . . . . . . . . . . . . . . 1138
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1127 Spinal Venous Infarction . . . . . . . . . . . . . . . . . . . . . . . . . . . 1138
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1127 Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1139
History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1127 Concepts of Primary and Secondary injury . . . . . . . . 1140
Spinal Venous Infarction . . . . . . . . . . . . . . . . . . . . . . . . . . . 1142
Arterial Anatomy of the Spine . . . . . . . . . . . . . . . . . . . 1127 Imaging Correlation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1142
Segmental Arteries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1128
Radicular Arteries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1128 Clinical Presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1143
Ischemic Spinal Cord Infarction . . . . . . . . . . . . . . . . . . . 1143
Arterial Supply of Spinal Cord . . . . . . . . . . . . . . . . . . 1128 Hemorrhagic Spinal Cord Infarction . . . . . . . . . . . . . . . 1145
Superficial Arterial System of the Spinal Cord . . . . 1128 Venous Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1145
Intrinsic Arterial System . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1131
Imaging Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1145
Watershed Territories of the Spinal Cord . . . . . . . 1131 Evolution of Spine Imaging . . . . . . . . . . . . . . . . . . . . . . . . 1145
Longitudinal Anastomosis . . . . . . . . . . . . . . . . . . . . . . . . . . 1131 Standard Spine Protocol . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1145
Conventional MRI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1145
Etiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1132
Diffusion-Weighted Imaging (DWI)
of the Spinal Cord . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1146
Typical MRI Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1147
Associated Vertebral Body Infarction . . . . . . . . . . . . . . 1149
S.R. Boddu (*) • M.A. Banihashemi • Holistic Approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1149
Y.P. Gobin • A. Patsalides Advanced Techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1149
Department of Neurological Surgery, Weill Cornell Follow-Up Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1149
Medical Center, New York, NY, USA
e-mail: srb9017@med.cornell.edu;
mob2009@med.cornell.edu; yug2001@med.cornell.edu;
atp9002@med.cornell.edu
A. Cianfoni
Department of Radiology, Medical University of South
Carolina, Charleston, SC, USA
e-mail: cianfoni@musc.edu
K.-W. Kim
New York Presbyterian Hospital, Weill Cornell Medical
College of Cornell University, New York, NY, USA
e-mail: kyk2003@nyp.org
E. Pravatà
Neuroradiology Department, Neurocenter of Italian
Switzerland, Lugano, Switzerland
e-mail: emanuele.pravata@gmail.com

# Springer Science+Business Media New York 2016 1125


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6_30
1126 S.R. Boddu et al.

Differential Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1150 Abstract


Multiple Sclerosis (MS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1150 Spinal cord infarction is a rare disease and
Idiopathic Transverse Myelitis (ITM) . . . . . . . . . . . . . . 1151
Acute Disseminated Encephalomyelitis
constitutes one of the acute spinal emergencies.
(ADEM) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1152 In comparison to its cerebral counterpart, the
Spinal Cord Arteriovenous Shunts . . . . . . . . . . . . . . . . . 1153 spinal cord infarction has extremely low inci-
Spinal Cord Neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1156 dence, possibly related to the abundance of the
Role of Angiography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1158 arterial anastomosis and low incidence of the
Challenges in Spinal Angiography . . . . . . . . . . . . . . . . . 1158 atherosclerosis in the spinal arteries. Since the
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1169 first spinal cord infarction reported in early
Spinal Cord Ischemia Secondary to Embolism . . . . 1170 nineteenth century, there has been remarkable
Spinal Cord Ischemia Secondary progress in the understanding of this disease
to Hypoperfusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1170
Treatment of Underlying Cause . . . . . . . . . . . . . . . . . . . . 1171 entity. However, the fact that there is no
Role of High-Dose Steroids . . . . . . . . . . . . . . . . . . . . . . . . 1171 established standard of care treatment as of
Surgical Decompression . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1171 today highlights the complexity and challeng-
Prognosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172 ing nature of this disease. In this chapter the
Short-Term Outcome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172 arterial anatomy of the spinal cord,
Long-Term Outcome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172 etiopathogenesis, and clinical presentation of
Future Direction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172 the cord infarction with specific emphasis on
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1173
the imaging findings and differential diagnosis
are discussed. The role of catheter angiography
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1173
was specifically addressed along with the
merits and pitfalls of the noninvasive angiog-
raphy. The chapter concludes a discussion on
the current and potential future treatment
strategies.

Keywords
Watershed territory • Thrombolysis • Myelitis •
Infarction • Digital subtraction angiography •
Embolization • Signal-to-noise ratio • Diffu-
sion-weighted imaging • Fast spin echo • Gra-
dient echo • Diffusion tensor imaging • Fiber
tractography • Echo-planar imaging • Apparent
diffusion coefficient • Anterior spinal artery •
Posterior spinal artery • Segmental artery •
Radicular artery • Artery of Adamkiewicz •
Multiple sclerosis • Neuromyelitis optica • Idi-
opathic transverse myelitis • Acute dissemi-
nated encephalomyelitis • Acute
polyneuropathy • Subacute combined degener-
ation • Neurosarcoidosis • Neuro-Behcet’s dis-
ease • AIDS-associated myelopathy •
Radiation myelitis
47 Spinal Cord Infarction and Differential Diagnosis 1127

Introduction States, this would amount to 5,000–8,000 cases of


SCI a year [8]. However, none of these series
Spinal cord infarction is a diverse entity broadly include post-aortic surgery patients, which is cur-
categorized into vascular syndromes based on rently the most common etiology [7, 9, 10]
etiology and clinical presentation. Acute spinal for SCI.
cord ischemia constitutes 5–8 % of all acute
myelopathies and only 1–2 % of all vascular neu-
rological pathologies [1]. Still spinal cord infarc- History
tion (SCI) represents one of the most common
causes of acute non-compressive myelopathies Autopsy reports consistent with spinal cord
[2]. Despite extensive diagnostic efforts, infarction, referred to as spinal softening, had
35–60 % [1, 3] of acute spontaneous spinal cord been published as early as 1831 [11]. At that
syndromes remain etiologically ambiguous. Thor- time spinal cord infarction cases were categorized
ough understanding of the etiology, pathophysi- as acute myelitis. The earliest clinical description
ology, limitations of the conventional imaging, of this condition, consistent with spinal cord syn-
and poor prognosis from delayed treatment is drome, was described in literature in 1842
vital in the care of SCI patients. Recent advances [12]. Prior to 1866 it was widely believed that
in spinal cord imaging such as diffusion-weighted the underlying etiology of such case was inflam-
imaging (DWI), lessons learnt from the extensive mation, as identified by autopsy reports that
evidence of the brain stroke thrombolysis, and showed a “softening” of the spinal cord. Bastian
cutting-edge neurointerventional techniques may et al. first described the term “softening” for brain
define an effective early treatment of spinal cord tissue in 1866, and later in 1882 it was suggested
ischemia in the near future. by the same author that softening of the spinal
cord was caused by thrombosis of spinal vessels
rather than inflammation of the cord, although this
Epidemiology etiology was not widely accepted for more than a
decade after its description [5].
Spinal cord infarction (SCI) is a rare entity with
limited epidemiological data. In a largest autopsy
series of 3,737 consecutive postmortem studies Arterial Anatomy of the Spine
during 1909–1958, only 11 cases of SCI are
reported, of which nine are ischemic and two A detailed knowledge of the vascular anatomy of
hemorrhagic in nature [4]. The absence of com- the spine and spinal cord is a prerequisite for
mon etiological factors such as atherosclerosis understanding the pathophysiology of spinal
and hypertensive vascular disease in these cord infarction. A thorough understanding on vas-
11 patients is debatable for its completeness and cular anatomy helps to better understand how
its representation of the current population. This lifesaving treatments of thoracoabdominal aortic
low number may be due to the lack of routine aneurysm and intra-arterial embolization proce-
examination of the spinal cord in autopsies, as dures can turn into a risk factor for the spinal
19 cases of SCI in a 6-year interval were reported cord infarction. Also this is vital for planning of
in the early twentieth century [5]. In another series safe surgical and endovascular interventions. Dig-
of autopsies of asymptomatic patients from the ital subtraction angiography is the gold standard
1970s, an incidental rate of 3 % was reported for spinal vascular imaging. Increasingly, mag-
[6]. Currently the incidence of spinal cord infarc- netic resonance angiography (MRA) with time-
tion is estimated to account for 1.2 % of all stroke resolved imaging has gained widespread accep-
admissions in the United States [7]. Based on tance in the localization of the spinal vascular
available data on stroke admissions in the United malformations and promising results in the
1128 S.R. Boddu et al.

detection of large vascular structures, such as the by the level from which they originate. Segmental
artery of Adamkiewicz. arteries supply all of the dorsolateral tissues of a
single metamere, except the spinal cord. There are
extensive anastomoses between segmental arter-
Segmental Arteries ies with important connections both above and
below a given level, as well as contralaterally.
The arterial supply to the spinal column, The segmental arteries divide into three major
paraspinal muscles, dura, nerve roots, and spinal trunks: (i) lateral or ventral (posterior intercostal
cord derives from the segmental arteries. The seg- or lumbar artery), (ii) middle or dorsal (muscular
mental arteries in the thoracic and upper lumbar and cutaneous branches), and (iii) medial or
spine originate in pairs from the posterior aspect spinal [13].
of the descending aorta adjacent to the spinal
column. The segmental arteries usually include
one pair of supreme or superior intercostal arteries Radicular Arteries
(above T3), nine pairs of posterior intercostal
arteries (T3–T11), one pair of subcostal arteries Radicular artery along with the anterior and
(T12), and four pairs of lumbar arteries (L1–L4) posterior spinal arteries branches out from the
[13]. The superior/supreme intercostal artery medial (spinal) trunk, after entering the spinal
arises from aorta or costocervical trunk or verte- canal at intervertebral foramen. Further at each
bral artery. Segmental arteries in the lower lumbar nerve root level, the radicular artery ramifies
and sacral region originate from branches of the [14] into:
internal iliac artery (mainly the iliolumbar and
lateral sacral arteries) and the median sacral artery 1. Radiculoradial artery – supplies both the dura
(branch of the aorta at the level of the bifurcation), and the nerve roots.
providing arterial supply to the L5 vertebra and 2. Radiculomeningeal artery – supplies only
the sacrum. The distribution of the segmental the dura.
arteries is summarized in Table 1. 3. Radiculomedullary artery – supplies the spinal
It is important to remember that the segmental cord. Unlike other two branches, radiculo-
arteries are named by the level they supply and not medullary branches are present only at some
nerve root levels.
Table 1 Summary of distribution of segmental arteries
Cervical spine
Vertebral arteries
Deep and ascending cervical arteries
Arterial Supply of Spinal Cord
Thoracic and upper lumbar spine
Subclavian arteries via deep cervical artery
1. Superficial or external arterial system
T1 and T2 segmental/intercostal arteries 2. Deep or intrinsic arterial system
Aorta
Thoracic segmental arteries (intercostal)
One pair of superior/supreme intercostal artery Superficial Arterial System
(above T3) of the Spinal Cord
One pair at each vertebral level from T3–T12
Lumbar segmental arteries At the surface of the cord, two arterial systems can
One pair at each vertebral level from L1–L4 be described (Fig. 1):
Lower lumbar spine and sacrum
Internal iliac artery (iliolumbar and lateral sacral 1. Longitudinal arterial trunks: extend along the
arteries)
long axis of the spinal cord and are constituted
Median sacral artery (branch of the aorta)
by one anterior spinal artery and two posterior
47 Spinal Cord Infarction and Differential Diagnosis 1129

via the anterior radiculomedullary arteries in order


to maintain adequate blood flow to the entire
spinal cord. As a result, the ASA should not be
thought of as a single straight artery but rather as a
consecutive series of anastomotic vascular
loops [16].
ASA receives major contribution from the
radiculomedullary arteries at three regions:
cervicothoracic, mid-thoracic, and thoracolumbar,
with watershed areas at the border of each region,
especially in the upper thoracic region. The most
important anterior radiculomedullary arteries and
most easily recognized in angiography are the
artery of cervical enlargement in the cervical and
upper thoracic region and artery radiculome-
dullaris magna, also known as the artery of
Fig. 1 Arterial supply of the spinal cord: (1) anterior
Adamkiewicz (AKA) in the lower thoracic and
spinal artery, (2) posterior spinal artery, (3) spinal trunk
of segmental artery, (4) anterior radiculomedullary artery, lumbar region (Fig. 2).
(5) posterior radiculomedullary artery, (6) central (sulcal) The mid-thoracic spinal cord is more vulnera-
arteries, (7) vasocorona ble to ischemic compromise as there is
minimal collateral supply to the spinal cord
(or posterolateral) spinal arteries, which superior to the junction of the AKA with the
receive flow from the anterior and posterior ASA. The ASA continues caudally past the
radiculomedullary arteries, respectively conus as an insignificant branch to the filum
2. Pial plexus: covers the periphery of the spinal terminale; the more important, functional contin-
cord uations of the ASA are the “rami cruciantes” that
surround the conus and provide robust “basket”
Longitudinal Arterial Trunks anastomoses with the posterior spinal
arteries [17].
Anterior Spinal Artery
The anterior spinal artery (ASA) is typically Posterior Spinal Arteries
formed at the level of the foramen magnum by The two posterior spinal arteries (diameter <0.5
the confluence of descending branches of the mm) also originate at the level of the foramen
intracranial segments of the vertebral arteries magnum by branches of the ipsilateral
(above the posterior inferior cerebellar artery vertebral or posterior inferior cerebellar arteries
(PICA)). The descending branches from both ver- and travel along the posterolateral surface of the
tebral arteries may join at the C2–C4 level [15] spinal cord, receiving supply at various levels
and travel along the anterior sulcus of the spinal from the posterior radiculomedullary arteries
cord to the conus medullaris. ASA has variable (Fig. 3). 11–16 feeders contribute to the posterior
caliber (diameter 0.2–0.8 mm); it is thinnest in the spinal artery (PSA) at various levels through the
thoracic cord and thickest in the region of the spine. The largest posterior radiculomedullary
conus medullaris [15]. The ASA supplies the artery often enters below the level of the
anterior two-thirds of the spinal cord tissue AKA [15].
(including the anterior horns and the Throughout its course, each PSA gives off
spinothalamic and corticospinal tracts) by central branches that supply the posterior third of the
and pial branches. spinal cord, including the posterior columns, dor-
The ASA is usually continuous, and during its sal gray matter, and superficial dorsal aspect of the
long course, it receives additional arterial supply lateral columns of the spinal cord.
1130 S.R. Boddu et al.

Fig. 2 Anterior spinal artery: DSA demonstrating the thoracolumbar segment, artery of Adamkiewicz with typ-
midline anterior spinal artery (ASA) with contribution ical hairpin appearance (a, b). Note the vertebral artery
from the dominant radiculomedullary branch in the contribution to ASA in the cervical cord segment (c, d)

Radiculomedullary Arteries angiography is the artery radiculomedullaris


At several levels, the radicular artery gives branches magna, also known as the artery of Adamkiewicz
that follow the anterior and/or posterior nerve roots (AKA). It has a diameter of 0.5–1.0 mm [15] and
to supply the spinal cord, called the radiculo- almost always arises in the thoracolumbar region,
medullary arteries. The average number of anterior between T8 and L2 in 75 % of cases [16,
radiculomedullary arteries is 6 (range 2–14) [18, 21–23]. It has, however, been identified as supe-
19], whereas the number of posterior radiculo- riorly as T5 and as inferiorly as L4 [19]. In 80 % of
medullary arteries varies from 11 to 16 [15]. cases, it is found in the left side. The AKA forms
the classic “hairpin” loop when it reaches the ASA
Radiculomedullary Artery of Cervical and gives off a thin ascending branch and a larger
Enlargement descending branch.
There are 2–3 anterior radiculomedullary arteries
in the cervical region. The most important supply Pial Plexus
to the cervical ASA is usually located between the Besides the direct connections between the ASA
C4–C8 levels of the spinal cord and is termed the and PSAs at the conus medullaris, there is an
artery of cervical enlargement [20], often a branch extensive arterial network on the entire surface
of the deep cervical artery, and usually accom- of the spinal cord formed by effective anastomo-
panies the C6 nerve root. ses between the transverse and oblique branches
from the ASAs and PSAs. This superficial rich
Artery of Adamkiewicz (AKA) anastomotic network is termed the pial plexus
The most important anterior radiculomedullary (vasocorona) and has been visualized on
artery and the one most easily recognized in microangiograms of the spinal cord [15]. This
47 Spinal Cord Infarction and Differential Diagnosis 1131

pial plexus at the periphery of the spinal cord and


course into the white matter centripetally.

Watershed Territories
of the Spinal Cord

The concept of watershed areas has been applied


to the areas of relative poor vascularity in the rich
spinal cord vascular network. As detailed in the
anatomy section, the spinal cord has rich vascu-
larity contributed from the longitudinal and cir-
cumferential networks. The blood supply of the
cord thus depends on the integrity of half a dozen
radicular arteries so far as the anterior three-
quarters of the cord is concerned and on a slightly
greater number of posterior radicular arteries for
the nourishment of the posterior third.
Given the relative paucity of these networks in
certain regions, there exists “watershed terri-
tories” in both the networks.

Longitudinal Anastomosis
Fig. 3 Posterior spinal artery: DSA demonstrating the
posterior spinal artery (PSA) in the lower thoracic cord.
Note the paramedian location of the PSA compared to ASA Although radicular arteries accompany the spinal
and the sharper angle of the hairpin curve nerves at each level, the radiculomedullary
branches supplying the spinal cord are present
network is responsible for vascularizing the only at few levels. The average number of anterior
periphery of the spinal cord. radiculomedullary arteries is 6 (range 2–14) [18,
19], whereas the number of posterior radiculo-
medullary arteries varies from 11 to 16 [15]. The
Intrinsic Arterial System superior cord segment involving cervical and
upper thoracic cord (up to T2/T3) is usually sup-
The spinal cord parenchyma is supplied by the plied by vertebral arteries and 2–3 anterior
intrinsic arterial system, which is subdivided into radiculomedullary arteries, including the artery
a central (centrifugal) system and a peripheral of cervical enlargement [20], often a branch of
(centripetal or vasocorona) system [13]. The cen- the deep cervical artery, and usually accompanies
tral system is comprised of the central arteries the C6 nerve root. Similarly 2–3 anterior radiculo-
(sulcal or sulcocommisural arteries; diameter medullary arteries, including the artery of
0.06–0.40 mm), which originate from the ASAs Adamkiewicz (AKA), usually supply the inferior
and travel into the anterior median fissure, pene- cord segment involving the lower thoracic and
trate into the cord either on the left or right, and lumbar cord. The intermediate cord segment
branch centrifugally, mainly within the gray mat- involving the mid-thoracic cord (T4–T8) is a
ter [19]. The peripheral system (vasocorona; poorly vascularized segment with paucity of the
diameter 0.1–0.2 mm) [19] consists of small per- radicular arteries, although occasional radicular
forators (rami perforantes) that originate from the artery is present at T7 level. In addition to the
1132 S.R. Boddu et al.

overlapping areas between the upper, mid, and myelopathy, which demonstrated lesions located
lower cord segments, the entire mid-thoracic throughout the spinal cord in nine cases and
cord due to its paucity of the radiculomedullary restricted only to the lumbosacral cord in four
arteries is considered as an anatomical “watershed patients.
territory” of the spinal cord. High metabolic demands and the large number
of neurons in the gray matter make it more vul-
Circumferential Anastomosis nerable to ischemia than the white matter
Within the intrinsic arterial system, there is anas- [24]. The spinal cord blood flow analysis in pri-
tomosis between the central (centrifugal) system mates showed a higher blood supply to the gray
and the peripheral (centripetal or vasocorona) sys- matter (57.6 +/ 2.3 ml/100 g/min), due to the
tem [13]. The sulcal artery is a branch of the much higher metabolic rate compared to the white
anterior spinal artery, which, centrally oriented, matter (10.3 +/ 0.2 ml/100 g/min) [24]. Similarly
runs through the anterior median sulcus, enters primate studies’ total flows in cervical, upper tho-
the cord, and supplies most of the gray matter racic, and lumbar areas were 14.9 + 1.4, 10.4 
and the adjacent white matter. The peripheral sys- 0.8, and 19.7  1.2 ml/min/100 g, respectively
tem (vasocorona) [19] consists of small perfora- [24]. The lowest total flow is observed in the
tors (rami perforantes) that originate from the pial thoracic cord; in part, this difference has been
plexus at the periphery of the spinal cord and attributed to the relative decrease in the gray mat-
course into the white matter centripetally. Located ter at this level.
in between, there exists a border zone that is
supplied by both the central and peripheral
arteries. Etiology

Ischemic Vulnerability of the Spinal Cord Spinal cord infarction (SCI) can be categorized
Historically, the literature has supported the into three groups (Table 2):
notion of a spinal cord “watershed zone” of ische-
mic vulnerability centered at the mid-thoracic 1. Ischemic infarction
level (T4–T8 level) [24, 25]. However, Duggal 2. Hemorrhagic infarction
et al. showed that >95 % of cardiac arrest and 3. Venous infarction
hypotensive patients with associated ischemic
myelopathy had predominant involvement of the
lumbosacral level followed by cervical level with Ischemic Spinal Cord Infarction
relative sparing of thoracic levels. None of the
examined patients in their study had neuronal This is the most common type of spinal cord
necrosis limited to the thoracic level only. Authors infarction. The common etiological factors in
concluded that, although the thoracic spinal cord this group are included in two distinct groups:
may be the anatomic watershed zone with respect hypoperfusion and embolism. The individual
to regional blood supply, the lumbosacral cord risk factors are discussed in detail below:
neurons appear to be more susceptible to
ischemia. Hypoperfusion Related
Neuronal necrosis restricted to the lumbosacral
level of the spinal cord was first reported by Gilles Aortic Surgery
and Nag [26] in 6 neonates suffering transient Paraplegia as a complication of aortic surgery in
cardiopulmonary arrest. These findings were humans was first reported in 1956 by Adams
later confirmed in a larger series by Azzarelli et al. [36]. Surgical repair of thoracic and
and Roessmann [27] in their material from thoracoabdominal aortic aneurysms and aortic
16 patients, including 11 adults with ischemic dissections (Fig. 4) are the most common etiology
47 Spinal Cord Infarction and Differential Diagnosis 1133

Table 2 Etiology of spinal cord infarction for SCI [7, 9, 10]. Although this complication is
Ischemic infarction reported both after open and endovascular repair
Hypoperfusion of ruptured aneurysm, its incidence is lower by
Aortic surgery using endovascular techniques, partly related to
Aortic dissection the patient selection bias [37].
Systemic hypotension Variable incidence of the SCI after
Thrombosis of abdominal aortic aneurysm endovascular stent graft has been reported up to
Atherosclerosis of segmental arteries 10 % [38], but it could be as low as 0.6 % [39] and
Vertebral artery dissection as high as 28 % in high-risk cases [40]. In a
Mechanical injury (surfer’s myelopathy) multicenter comparison trial, the reported inci-
Abdominal/pelvic surgeries dence of SCI after endovascular repair of
Spinal surgery or intervention
descending aortic aneurysms has a risk of 3 %
Embolism
versus 14 % for open surgery [41]. SCI incidence
Thromboembolism
is much lower for endovascular repair of the
Cardiac arrhythmias
abdominal, rather than thoracic aorta [42]. In the
Valvular heart disease
Therapeutic embolization (spinal, renal, and
setting of trauma and open repair, the rate of spinal
bronchial) cord ischemia is about 7 % when cross-clamping
Aortic catheterization is used alone. This can be decreased to 3 % by
Hypercoagulable conditions [28, 68] using shunts for distal perfusion and cardiopul-
Fibrocartilaginous embolism monary bypass [37].
Septic (infection) embolus In a large case series, the incidence of paraple-
Hemorrhagic infarction gia as a result of thoracoabdominal aneurysm
Subarachnoid hemorrhage repair was reported as high as 21 %, whereas the
Arterial dissection aneurysmal repair confined to only the abdominal
Ruptured spinal aneurysm aorta was 1 % [43]. Spinal cord ischemia has been
Dural arteriovenous fistula (DAVF) reported with iliac aneurysm repair as well
Perimedullary arteriovenous malformation (AVM)
[44]. Hypoperfusion in the abdominal aorta with
Anticoagulation
prolonged clamping [45], declamping syndrome
Bleeding diathesis
after declamping of the infrarenal aorta and spinal
Epidural/subdural hemorrhage
cord tamponade [46], and increased pressure of
Spontaneous rupture of spinal DAVF, perimedullary
AVM [29] the cerebrospinal fluid (CSF) during clamping
Intramedullary hemorrhage – hematomyelia [47] are the main risk factors in the pathogenesis
Spinal angiomas [29] of the spinal cord ischemia following the aortic
Ruptured spinal AVM surgery.
Cavernoma Spinal cord ischemia may also happen in pro-
Spinal tumors cedures involving the ligation of radicular arter-
Venous infarction ies, such as spinal tumor resection, scoliosis
Spinal DAVF, perimedullary AVM correction, or pneumonectomy [48].
Cord compression from extramedullary lesions
Tumor (e.g., pancreatic) embolus [30] Atherosclerosis
Decompression sickness (air embolus) The link between atherosclerosis and spinal cord
Miscellaneous
ischemia has been considered since the late nine-
Radiation injury [31–33]
teenth century [49]. Atherosclerosis is minimal in
Vasculitis [31–33]
the spinal arterial network when compared to the
Epidural anesthesia [7, 9, 25]
rest of the body. The anterior spinal artery is the
Substance abuse (e.g., cocaine) [34]
Drug induced (e.g., sildenafil citrate) [35]
most affected, while other arteries in this system
may undergo fibrosis and thickening instead.
1134 S.R. Boddu et al.

Fig. 4 Spinal cord infarction following aortic surgery: a T2-WI hyperintensity in the central gray matter T7/T8
69-year-old male with Marfan’s syndrome, status post- level (c, d) and extends distally to the conus (inferiorly
graft repair of the ascending aorta (a) following type A involves both the central and peripheral aspects of the
dissection, presented with paraplegia following surgery. entire distal cord). SCI in the anterior and posterior spinal
Note the bilateral iliac artery dissections. (b) MRI – arteries with cord swelling noted inferiorly (c, e)

There is both ascending and descending flows Aortic Dissection


within both the anterior and posterior spinal arte- Spinal cord ischemia as a possible cause of weak-
rial systems. This allows for collateral circulation ness in the limbs as a result of impaired perfusion
in the event of occlusion [50]. While moderate to of the spinal cord aortic dissection had been
severe aortic atherosclerosis was observed in known since at least the late nineteenth century
86 % in an autopsy series of SCIs, no meaningful [53]. Dissecting thoracoabdominal aortic aneu-
correlation has been observed between degree of rysms, especially those with a left-sided false
aortic atherosclerosis, hypertension, or coronary lumen, are associated with a higher incidence of
artery disease and occurrence of lumbosacral spinal cord ischemia, because in 85 % of the
ischemia [51]. In a case series of 1,000 autopsies, population, the anterior spinal artery arises from
28 patients who had atherosclerotic emboli from the left lower intercostal vessels [46]. Based on
the abdominal aorta to visceral organs were iden- the Stanford classification of the aortic dissection:
tified, and the spinal cord was dissected. In
12 cases atheromatous emboli were evident in Stanford type A aortic dissection – dissection
the spinal arteries, most commonly in the anterior involving the ascending aorta, regardless of
spinal artery. However, only one had a spinal cord the site of primary intimal tear [54] (Fig. 5).
infarction, showing that apparently good collat- Neurological symptoms can be present in 29 %
eral circulation and lack of general atherosclerosis upon presentation. Spinal cord ischemia can be
prevents infarction, unless hypoperfusion leads to present preoperatively in about 1–9 %. In a
decompensation [52]. study showing a preoperative incidence rate
47 Spinal Cord Infarction and Differential Diagnosis 1135

Fig. 5 SCI following aortic dissection: a 71-year-old addition to the T2-WI hyperintensity (c, d), notice the
male: status post-dissection of the ascending aorta (a, b). post-contrast enhancement in the area of infarction (e, f)
Subacute spinal cord infarction involving the central gray suggestive of subacute nature (>5 days) of this infarction
matter extends from T11 to the conus medullaris. In

of 1 %, this rate increased to 4 % involved area in 95 % of cases (Fig. 6). Ischemic


postoperatively [55]. damage limited to only the thoracic region was
Stanford type B aortic dissection – consists of all seen in only 7.6 % of cases and never in isolation,
aortic dissections except type A. Spinal cord whereas lumbosacral involvement was the most
ischemia is present in about 3 % of this common affected area, with nearly 70 % of cases
type [56]. of spinal cord ischemia showing only involve-
ment of this region. This is potentially due to
Systemic Hypotension higher metabolic demand and a large number of
Based on the autopsy series of 145 fatal cases of neurons in the lumbosacral region [51]. Similar
cardiac arrest or severe systemic hypotension, SCI predisposition of lumbar spinal cord for SCI is
was documented in 46 % of cases [51]. Although also documented in the neonates [57].
widely understood that the mid-thoracic is the
watershed area, and presumably most susceptible Surfer’s Myelopathy
to ischemic injury, the thoracic area was spared, This is a rare acute nontraumatic myelopathy,
and the lumbar area was the most commonly affecting first time/inexperienced surfers.
1136 S.R. Boddu et al.

Fig. 6 Anterior spinal artery territory infarct: a 6-year-old T2 hyperintense signal in the anterior spinal artery territory
girl with gastroenteritis presented with sudden onset of from T6 to the conus (a–c) in anterior spinal artery syn-
lower extremity weakness, thoracic pain, loss of rectal drome. Subsequent DSA shows patency of AKA (d) and
tone, and mid-thoracic sensory loss. MRI shows diffuse PSA (e)

Thompson et al. first described this entity in 2004 with hypercoagulable state and deep venous
[58]. The exact etiology is unknown. Prolonged thrombosis) [58].
spinal hyperextension while in prone posture and The thoracic spinal cord is most commonly
repetitive flexion and extension are the main pre- affected in the surfer’s myelopathy [60]. The vari-
cipitating factors resulting in spinal cord arterial able levels of thoracic spinal cord involvement may
insufficiency and watershed ischemia [58]. Pro- be explained by the variable origins of the artery of
posed vascular mechanisms of injury include Adamkiewicz, arising from T5–T8 in 15 %,
avulsion of perforating vessels, vasospasm of the T9–T12 in 75 %, and L1–L2 in 10 % of the popu-
artery of Adamkiewicz, and transient ischemia in lation [60]. The outcome is variable and does not
areas of borderline perfusion as a result of tension correlate with the extent or intensity of the signal
on the spinal cord with hyperextension [59]. abnormality on the MRI [61]. Most patients had a
Other possible mechanisms include inferior complete or near-complete recovery; residual com-
vena cava obstruction and fibrocartilaginous plete paraplegia has been reported [58, 59, 61].
embolism. The former may occur secondary to
compression by the liver while lying prone over Embolism Related
the surfboard aggravated by concomitant
prolonged Valsalva maneuver during paddling, Thromboembolic Infarction
resulting in increased retrograde venous pressure The main embolic source for the SCI is cardiac,
in the epidural venous plexuses leading to infarc- well described in the literature with diverse
tion [59]. Potential risk factors that may predis- pathology such as atrial fibrillation, atrial myx-
pose surfers to ischemic cord injury include body oma [62], mitral valve disease [31], and patent
habitus (thin with underdeveloped musculature), foramen ovale [63]. Spinal angiogram may dem-
dehydration (related to travel and prolonged hours onstrate the abrupt termination of the anterior or
at the beach), and long-distance travel (associated posterior spinal arteries (Fig. 7).
47 Spinal Cord Infarction and Differential Diagnosis 1137

Fig. 7 Embolic spinal cord infarction: a 66-year-old Note the abrupt termination of the AKA (arrow) on subse-
female presented to ED with acute onset bilateral lower quent DSA suggesting an embolic occlusion (d) (Courtesy
extremity motor and sensory deficits. MRI: T2-WI Dr. Kim Nelson and Tibor Becske, NYU Medical Center,
hyperintensity extends from T11 to the conus medullaris New York, NY)
involving the central gray matter with cord swelling (a–c).

Fibrocartilaginous Embolism outcome, while those of thrombotic or other


Fibrocartilaginous embolism (FCE) was first embolic causes often improve over time.
described as a cause of spinal cord infarction in It is thought to occur when, due to increased
1961 [64]. A history of heavy lifting or trauma to intra-disk pressure, fragments of the intervertebral
the spine may be a clue to this etiology, but this disk are pushed into the disk vasculature or the
may not always be the case. Nearly 70 % of cases bone marrow and venous sinuses of vertebral
are young women, in contrast to thrombotic or bodies. Supporting this is the finding of fibrocar-
other embolic etiology, predominantly affecting tilaginous emboli in the bone marrow and vascu-
men. Three characteristics distinguish FCE from lar supply of the affected regions in autopsies [65,
other thrombotic or embolic causes of SCI 66]. Seventy percent involve the upper cervical
[65]. First, their presenting symptom is that of cord, with some extending to the lower medulla
the sudden severe pain in the region of the spine oblongata or the lower thoracic, which explains
that is affected, whereas in thrombotic and other the high fatality of these cases. The presence of
embolic causes, this is hardly ever the case. Sec- cord swelling and collapsed intervertebral disk at
ond, after the onset of pain, there is a period with the corresponding level indicates this
no neurological symptoms. Over the course of etiology [65].
15 min to 48 h, symptoms of neurological deficit
progress, indicating the evolution of a spinal Infection
stroke, whereas in thrombotic and other embolic Syphilis was diagnosed in a majority of cases of
etiologies, numbness and weakness of the legs spinal cord ischemia in a report from 1902
begin at onset and at the same time as pain, if [5]. This was prior to the introduction of penicillin
present. Finally, these cases have a very poor as a cure. Later case series show the most common
1138 S.R. Boddu et al.

identified etiology to be of aortic origin, with infarction (Fig. 8). Cervical spinal cord ischemia
many as a complication of surgery on the aorta has been described in association with spinal
[9]. Bacterial meningitis [67] and mucormycosis subarachnoid hemorrhage in patients
[68] are reported in the literature as an etiology following dissection of the vertebrobasilar system
for SCI. or ruptured vascular malformations of the
spinal cord [77]. Disruption of the blood supply
Iatrogenic Embolization due to the involvement of radiculomedullary
The spinal cord ischemia has been reported as a arteries is thought to be the underlying
complication related to therapeutic embolization. mechanism in the acute setting [78]. Micro- and
This is particularly reported with bronchia artery macrocirculatory failure secondary to subarach-
embolization for hemoptysis between 1.4 % and noid hemorrhage may result in delayed ischemic
6.5 % [69]. Many other iatrogenic procedures are deficits. Vasospasm has shown to be the most
reported to cause spinal cord infarction including likely mechanism causing secondary injury in
but not limited to renal artery embolization [70], experimental animal models of spinal cord
intercostal embolization in hemoptysis [71], inter- injury [79].
costal artery embolization in traumatic hemor- Although different lesions predispose to hem-
rhage [72], endoscopic variceal sclerotherapy orrhage in different compartments (Table 2), their
[73], ligation [74], regional anesthesia, and pain strict localization is not possible. The hemorrhage
injections [75]. tends to extend across multiple compartments
depending on the amount and site of hemorrhage
Spinal Angiography and Embolization and may result in subdural and epidural hematoma
Angiography of the thoracic or abdominal in addition to the subarachnoid hemorrhage.
branches of the aorta rarely results in spinal cord Prolonged mass effect from the extra-axial hema-
ischemia. Spinal angiography is more challenging toma in addition to vascular disruption related to
in patients with atherosclerotic aorta and involvement of the radiculomedullary feeders, cir-
thoracoabdominal aneurysm. Spinal cord infarc- culatory failure, and vasospasm might result in
tion as a complication of angiography was spinal cord ischemia in these scenarios [78]. Con-
documented as early as 1949 and reported on in ditions such as anticoagulation and bleeding
the decade thereafter [76]. However, with diathesis may cause hemorrhage in either
increased experience and better techniques and compartments.
equipment in this field, complications have been
lowered. In a series of 134 consecutive spine
angiograms in 63 patients published in 1988, Spinal Venous Infarction
three neurological complications were reported
(incidence 2.2 %), all of which resolved in a This less common etiology is usually associated
matter of 1 day to 1 week [76]. In more recent with spinal dural arteriovenous malformations
series on spinal angiography and embolization (Fig. 9) secondary to venous congestion [80] and
from high-volume centers [77] including authors’ is discussed in multiple subsequent sections of
experience with 220 spinal angiograms and this chapter. It may also be a complication of
180 embolizations [78], no neurological compli- deep sea diving, in which gaseous occlusion of
cations were reported. the spinal venous plexus has been postulated as a
mechanism of injury in spinal decompression
myelopathy [9]. Early recompression reverses
Hemorrhagic Spinal Cord Infarction symptoms in about three-fourths of patients [81,
82]. Kim et al. [83] stated that the hemorrhagic,
Spinal subarachnoid hemorrhage (SAH) accounts nonhemorrhagic, and embolic venous infarction
for 1 % of all SAH. However, this is the most could not be differentiated with certainty on clin-
common form of hemorrhagic spinal cord ical grounds. Hemorrhagic venous infarctions
47 Spinal Cord Infarction and Differential Diagnosis 1139

Fig. 8 Spinal cord infarction from SAH: a 54-year-old swelling (c, d). Also notice the tracking of the extra-axial
female presented with acute subarachnoid hemorrhage hemorrhage from posterior fossa along the anterior border
(SAH) and quadriplegia. CT – posterior fossa predominant of the spinal cord with dorsal displacement (c). DSA dem-
SAH with intraventricular extension and hydrocephalus (a, onstrates a small anterior spinal artery aneurysm (arrow),
b). MRI – posterior decompression of the cervical spine as a culprit of SAH, pre- (e, f) and post- (g) successful coil
with residual diffuse T2-WI hyperintense cord signal embolization projections
involving the central gray matter with edema and cord

were associated with marked pain at the time of


onset and progressed rapidly; survival time was Pathophysiology
relatively short. Nonhemorrhagic infarctions were
less often painful and evolved slowly. Survival Understanding of the biomolecular changes that
time was longer than in hemorrhagic infarction. occur during and after spinal cord infarction (SCI)
Embolic venous infarction was associated with is essential for all those involved in the care of
pain of abrupt onset. As the initial presentation these patients. Extensive research over the past
does not correlate with severity of the disease, 30–40 years has focused on elucidating acute
treatment with hyperbaric chamber should be ini- spinal cord injury including both primary and
tiated promptly in decompression sickness, even secondary mechanisms, slowly unraveling the
in those with mild symptoms [82]. Also, the complex pathophysiologic processes such as
potential association of malignancy and venous roles of calcium, free radicals, sodium, excitatory
spinal cord infarction is well recognized, particu- amino acids, vascular mediators, and apoptosis.
larly in the case of pancreatic neoplasm [30]. These concepts of primary and secondary
1140 S.R. Boddu et al.

Fig. 9 Spinal dural arteriovenous fistula (DAVF): a blood vessels over cord surface (b). Reconstructions from
42-year-old male with progressive paraplegia. MRI – dif- the contrast-enhanced time-resolved MRA demonstrate the
fuse serpiginous flow voids over the cord surface with spinal DAVF (c). This is confirmed on the subsequent
associated T2-WI hyperintense cord edema and swelling spinal DSA (d), supplied by the right T10 radiculo-
(a). Contrast administration reinforces the visualization of medullary artery

mechanisms and their duality in acute spinal cord arrest, thromboembolism, hypovolemic shock,
injury have since also been embraced in the under- and hemorrhage with associated compression.
standing of the pathophysiology of spinal and Similar to the brain, spinal cord vessels
cerebral ischemia, subarachnoid hemorrhage, autoregulate according to SBP and PaCO2, but
and head trauma [84]. the spinal cord perfusion is more directly affected
by changes in systemic blood pressure. The
“watershed region” of the spinal cord in the
Concepts of Primary and Secondary mid-thoracic region (T4–T8) is anatomically
injury more susceptible for the ischemic injury. In case
of spinal arteriovenous malformations, the
Fehlings proposed a two-step process involving shunting of the arterial blood away from the spinal
primary and secondary mechanisms in acute spi- cord (arterial steal phenomenon) or venous hyper-
nal cord injury [85]. Allen first postulated the tension may result in hypoperfusion. The hypoxia
concept of a secondary mechanism in 1911 and hypoperfusion resulting from the primary
based on canine studies [86, 87]. mechanisms trigger the secondary mechanism.

Primary Mechanism Secondary Mechanisms


The primary mechanism involves the initial pre- Over the past 40 years, various theories of sec-
cipitating event such as atherosclerosis, cardiac ondary mechanisms of SCI have undergone a
47 Spinal Cord Infarction and Differential Diagnosis 1141

process of maturation. In the 1970s, Demopoulos suggested that the ischemic response to cord
et al. [88] proposed the free radical hypothesis, as injury is mediated both by the loss of
crucial to the injury process. Ten years later the autoregulation and by relative constriction of the
focus shifted onto the role of calcium, opiate resistance vessels [98]. Ischemia may play a role
receptors, and lipid peroxidation. Research in the in the formation of local cord edema, although
twenty-first century has suggested apoptosis, whether edema formation is injurious in itself or
intracellular protein synthesis inhibition, and an epiphenomenon is still unclear [84].
glutaminergic mechanisms, among a myriad of Because of differences in relative vascularity,
pathophysiologic pathways that mediate second- the central gray matter, along with adjacent white
ary injury mechanisms. There is considerable evi- matter, is more severely affected by acute cord
dence that the primary mechanical injury initiates injury than peripheral white matter [99]. Primate
a cascade of secondary injury mechanisms [85, studies have shown that the gray matter has higher
89–93]. blood supply than the white matter due to the
Although there are many mediators of second- much higher metabolic rate. Also, the flow was
ary injury, vascular mechanisms, free radicals, more variable with lower values in the dorsal
and more recently apoptosis have received much horns and higher values in the central gray and
attention. anterior horns, compared to the more uniform
flow in the white matter [100]. The blood flow to
Free Radicals the corticospinal tracts was considerably higher
Free radicals are highly reactive molecules that than the rest of the white matter, suggestive of
possess an extra electron in the outer orbit. Free higher metabolic activity in these tracts and
radicals most commonly form from molecular increased predisposition to the ischemia [101].
oxygen. There is good evidence for the early The intrinsic arterial blood supply to the spinal
occurrence and pathophysiologic importance of cord is directly proportional to the cross-sectional
oxygen free radical formation with cell membrane area of the gray matter, which is most abundant in
lipid peroxidation in central nervous system the thoracolumbar segment [102]. Therefore, in
injury [88]. Severe uncontrolled production of normal conditions, the blood flow to the spinal
the hydroxyl radicals (HO) from dissociation of cord is highest in the thoracolumbar segment,
hydrogen peroxide (H2O2) can cause lipid perox- which has the highest concentration of central
idation, impairment of phospholipid-dependent arteries owing to the relative abundance of gray
enzymes, disruption of ionic gradients, and ulti- matter. Consequently, the same segment is the
mately the lysis of cell membrane [94, 95]. Lipid most vulnerable to hypoperfusion, particularly
peroxidation may also play a role in postictal the gray matter that corresponds to the lumbar
hypoperfusion after initial precipitating event enlargement of the spinal cord. Arterial infarcts
and may result in SCI [94]. of the posterior spinal artery territory are rare
compared with the anterior spinal artery territory.
Vascular Mechanisms The scarcity of this condition is most likely due to
The changes that occur in spinal cord blood flow extensive anastomosis of the posterior spinal
after an acute spinal cord injury can be divided artery [103] and relative resistance of the posterior
into systemic and local. column to the ischemia due to its inherent lowest
Local effects: Following acute cord injury, a blood flow of all the tracts in the spinal cord [101].
major reduction in blood flow occurs at the site of White matter perfusion typically decreases
ictus, i.e., embolus or hemorrhagic compression, within 5 min of an acute cord injury and begins
possibly precipitated by vasospasm or vasoactive to return to normal within 15 min. It remains
amines [96, 97]. Based on animal studies, thereafter near-normal pre-injury values during
autoregulation is intact during the initial the first 24 h. In contradistinction, in the central
60–90 min after cord injury but is then lost coin- gray matter, numerous microhemorrhages typi-
cident with the onset of ischemia. It has been cally occur, as early as 5 min after an acute cord
1142 S.R. Boddu et al.

injury. Perfusion is relatively absent 1 h after an [118], seen in areas of wallerian degeneration and
injury, and this state remains so for at least the first detectable between 24 h and 3 weeks post
24 h. This vascular standstill has been confirmed injury [116].
using microangiography [97, 104], fluorescent
tracer studies [105], and the operating microscope
[106] and plays a major role in the devastating Spinal Venous Infarction
outcomes from acute cord injury. Alterations in
endothelial cell function causing an increase in Spinal cord veins are valveless. However, an anti-
vascular permeability and edema formation have reflux mechanism has been described at the dural
been well documented [107–109]. exit of radicular veins. It consists of an increased
Systemic effects: After an acute SCI, an imme- smooth muscle fibers and a Z-shaped course of
diate but transient increase in mean arterial blood the radicular vein when it crosses the dura to
pressure may occur followed by profound hypo- drain spinal cord venous blood in the epidural
tension and bradycardia [110]. The reason for this veins [14]. In case of spinal dural arteriovenous
transient hypertension is unknown but may be fistulas, the arterialized vein drains the
mediated by both the thoracic sympathetic gangli- fistula blood flow toward the spinal cord, and the
ons and the adrenal glands [111]. Acute cord increased pressure is transmitted to the
injury is one of the causes of neurogenic shock intrinsic cord veins in retrograde fashion. This
[112], typically being related to the magnitude and results in decreased intramedullary arteriovenous
severity of the cord injury. gradient and hypoxia, loss of autoregulation, and
Of all purported mechanisms of secondary disruption of blood-cord barrier and, if left
injury, the vascular hypothesis has considerable untreated, results in subacute necrotic
weight, with biochemical, angiographic, histo- myelopathy.
pathologic, and clinical support for its key role
in damage after acute SCI.
Imaging Correlation
Apoptosis
Apoptosis also called programmed cell death Ischemia in the distribution of the anterior spinal
occurs in a wide variety of disease states in artery rapidly initiates vasogenic and cytotoxic
eukaryotic cells. Apoptosis is recognized as edema in the gray matter with subsequent increase
occurring in utero as a form of neuronal cell in mobile water. This is manifested by a focal
death during embryonic development [113] and increase in proton density and a prolongation of
is also now thought to play a role in many post- the T2 relaxation time, which appears as distinct
developmental disorders of the central nervous increase of signal intensity on the long TR pulse
system, including ischemia [114]. Apoptosis is sequences.
an active process that is characterized by cell The ischemia/signal abnormalities start in the
shrinkage, chromatin aggregation, and nuclear anterior horns of the gray matter and, with increas-
pyknosis [115]. In the spinal cord, apoptosis was ing severity, spread posteriorly to involve the pos-
first identified in 1995 as occurring in rats [116] terior horns. Ultimately, the ischemic changes and
and, more recently, in the human spinal cord corresponding MR signal abnormalities extend
[117]. This is a tightly regulated process with a laterally to the posterolateral funiculi of the spinal
sequence of activation steps that require energy cord, including the crossed corticospinal tracts. In
and specific macromolecular synthesis as de novo severe cases, the whole cross section of the spinal
gene transcription [115]. A family of cysteine cord is infarcted. There seems to be a correlation
proteins, the caspases, is thought to play an impor- between the distribution of the MR signal abnor-
tant role in apoptosis, especially caspase-3 malities and the severity of the clinical symptom-
[84]. Oligodendrocytes are the major cell type in atology seen in spinal cord ischemia [46]
compressive cord injury that undergo apoptosis (Table 3).
47 Spinal Cord Infarction and Differential Diagnosis 1143

Table 3 Summary of secondary injury in acute spinal of spinal cord infarcts [135]. TIA manifest as
cord infarction weakness of the lower limbs precipitated by phys-
Vascular changes [92, 97, 119] ical activity or as “drop attacks” for cervical cord
Ischemia ischemia.
Impaired autoregulation
Neurogenic shock, hemorrhage
Microcirculatory derangements Ischemic Spinal Cord Infarction
Vasospasm
Thrombosis
Based on international standards for neurological
Ionic derangements [93, 120]
and functional classification of spinal cord
Increased intracellular calcium
injury proposed by the American Spinal Injury
Increased extracellular potassium
Association (ASIA) and International Medical
Increased sodium permeability
Neurotransmitter accumulation
Society of Paraplegia (IMSOP), ASCIS can be
Catecholamines [121] (e.g., norepinephrine, categorized into four groups [137]. The clinical
dopamine) syndromes are:
Excitotoxic amino acids [122, 123] (e.g., glutamate)
Arachidonic acid release [124] 1. Anterior spinal artery syndrome (ASAS)
Free radical [124] and eicosanoid production [125, 2. Posterior spinal artery syndrome (PSAS)
126] 3. Brown-Sequard syndrome (BSS)
Lipid peroxidation [125, 126] 4. Complete spinal cord transaction (CSCT)
Endogenous opioids and cytokines [127, 128]
Edema [129]
Anterior Spinal Artery Syndrome (ASAS)
Decreased adenosine triphosphate production [130]
This is the most common presentation of ASCIS,
Apoptosis [131–133]
affecting the anterior two-thirds of the spinal
cord that is supplied by one anterior spinal
artery (Fig. 10). Cervical anterior cord infarction
manifests with diffuse or radicular neck pain
Clinical Presentation followed by quadriparesis, associated with anal
or bladder sphincter dysfunction and sensory
Acute spinal cord infarction syndrome (ASCIS) loss of pain, pinprick, and temperature modalities.
can result in various clinical manifestations Proprioception and vibration sensations, which
based on the vasculature affected and etiology, are usually spared in anterior cord syndrome,
but has common symptoms. Bilateral can be affected in anterior cervical lesion due to
weakness, sensory loss, back pain at onset, and involvement of medial lemniscus. Later on, flac-
urinary retention were the most common cidity and areflexia give way to spasticity and
symptoms at the time of presentation in a hyperreflexia. Patients present with acute
prospectively collected series of 28 patients interscapular pain, followed by paraparesis or
[134]. The most commonly affected location is paraplegia, urinary and rectal incontinence or
the thoracolumbar level, and paraparesis com- retention, and a sensory loss to pain and temper-
prises of 20–25 % of all spinal cord infarction ature most common at the T4 level. Lumbosacral
cases [135]. Pain and sensory changes occur region is affected by lesion in the radicular artery
first, followed by weakness within minutes to of Adamkiewicz. Extensive lesions can be seen
hours, which peaks in 12 h [135]. Almost with flaccid paraplegia and autonomic dysfunc-
invariably spinal cord infarction presents with tion, including respiratory compromise with cer-
pain, at the level of the lesion, one retrospective vical lesions. Rarely, one segmental branch of
study noting pain in 72.7 % of 23 patients anterior spinal cord artery can result in
[136]. Transient ischemic attacks (TIA) rarely monoparesis or hemiparesis, instead of
precede spinal cord infarct, comprising of 10 % paraparesis [138].
1144 S.R. Boddu et al.

Fig. 10 Anterior spinal artery (ASA) territory infarction. and white matter secondary to occlusion of the ASA (a).
T2-WI hyperintense signal abnormality involving the ante- Pictorial representation of the ASA territory infarction (b)
rior two-thirds of the spinal cord including both gray matter

Fig. 11 Posterior spinal artery (PSA) territory infarction. corresponding MRI on the left with subtle focal T2-WI
Schematic representation of the posterior cerebral artery hyperintensity in the right PSA territory of the spinal cord
(PCA) territory infarction on the right. Note the

Posterior Spinal Artery Syndrome (PSAS) deficit (vibration and proprioception) below the
Posterior cord infarction is rare due to the lesion and contralateral decrease in pain and tem-
rich anastomotic network (Fig. 11). Posterior perature sensation a few segments below the
column and dorsal horns are affected, lesion.
manifested as total anesthesia and areflexia at the
level of lesion. Affected patients have deficit in Central Spinal Cord Syndrome (CSCS)
vibratory and proprioception deficits below the Central cord syndrome may theoretically be a man-
lesion. ifestation of spinal cord ischemia, but commonly
result from cervical spondylosis, trauma, syringo-
Brown-Sequard Syndrome (BSS) myelia, or other tumors. Location is the cervical or
Central sulcal artery occlusion can cause Brown- upper thoracic regions, involving the central por-
Sequard syndrome, which typically presents with tions of the spinal cord. The hallmark sign is char-
ipsilateral weakness and posterior column sensory acterized by weakness in the arms, with relative
47 Spinal Cord Infarction and Differential Diagnosis 1145

sparing of the legs, due to the latter’s corresponding [46]. Following acute presentation, MRI should be
corticospinal tract in lateral part of spinal cord. performed on emergency basis to confirm the diag-
Affected patients also have varying degree of sen- nosis, delineate pattern of SCI, and rule out differ-
sory loss below the lesion and urinary ential diagnosis such as cord compression from
retention [139]. traumatic, degenerative, or neoplastic processes
and intramedullary lesions such as acute inflamma-
tion, hemorrhage, and congestive ischemia due to
Hemorrhagic Spinal Cord Infarction vascular malformations.

Hemorrhagic infarction presents with sudden


onset of pain, usually at the level of hemorrhage. Evolution of Spine Imaging
Meningeal irritation is a typical finding. Patients
may have associated headache, cranial neuropa- Low signal-to-noise ratio (SNR) and prolonged
thies, and decreased level of consciousness imaging times while scanning the entire spine are
depending on extent and distribution of the hem- the main challenges of the spine imaging. Introduc-
orrhage. The evaluation for spinal SAH typically tion of the phased-array coil technology [141] and
follows negative intracranial workup. increased field strength has increased the baseline
SNR, a major improvement in spine imaging,
which has inherently limited SNR. Similarly intro-
Venous Hypertension duction of the parallel imaging [142, 143] and
improvement of multielement radiofrequency
This is the most common presentation in SDAVF. (RF) coils have not only enhanced the SNR of
Patients present with progressive lower extremity high-resolution MR imaging over a large
weakness (>50 %), sensory deficits (30 %) craniocaudal extent but also dramatically shortened
including paresthesia, sensory loss and hyperes- the imaging times.
thesia, gait disturbance, back pain (24 %), bowel
and bladder disturbance, and impotence. Once
motor or sensory deficits appear, severe disability Standard Spine Protocol
ensues over 6 months to 2 years.
Regular structural MR imaging sequences for
diagnosis of the cord infarction include
Imaging Findings multiplanar T1- and T2-WI fast spin-echo (FSE),
gradient-echo (GRE), and contrast-enhanced
Magnetic resonance imaging (MRI) is the primary (CE) acquisitions. Diffusion-weighted imaging
imaging modality of choice for the diagnosis of (DWI) has increasingly shown as a promising
suspected spinal cord infarction due to its superior technique in the evaluation of the acute spinal
spatial and contrast resolution for differentiation of cord syndrome [144, 145] and used as a part of
the intraspinal soft tissues and detection of intrinsic the standard protocol in the evaluation of acute
cord pathology [140] (Fig. 12). Also MRI is most non-compressive myelopathy.
sensitive in the detection of the marrow changes of
the adjacent vertebral bodies [140]. Perhaps the
most important role of MRI in both ischemic and Conventional MRI
hemorrhagic SCIs is to rule out alternative diagno-
sis. MRI not only confirms the evidence of spinal The sensitivity of conventional MRI is limited,
cord infarction but also may provide information as particularly in the first several hours. The reported
to the underlying etiology. The extent of signal sensitivity of T2-WI cord signal abnormality in
abnormalities correlates well with the severity of SCI ranges between 45 % and 73 % depending in
the clinical findings and the potential for recovery part on timing of the scan [1, 9, 10,
1146 S.R. Boddu et al.

Fig. 12 Normal MR imaging of the spinal cord. Homo- T2-WI hyperintensity and post-contrast enhancement
geneous cord signal, no intrinsic T2 hyperintensity of the (arrows) in the middle of the posterior vertebral border
cord or cord swelling or serpiginous flow voids on the cord represent a vertebral vasculature, a normal finding. Note
surface (a, c, d). No vascular enhancement either intrinsi- the exquisite spatial and soft tissue contrast resolution
cally within the cord or vascular enhancement over the offered by the MRI in spine imaging
cord following contrast administration (b). The focal

146, 147]. Hence the initial scan may either con- spinal cord in 1991 [148]. By using a standard EPI
firm the diagnosis or be a normal study and serve as sequence and a spinal coil, Stepper and Lovblad
a baseline for future follow-up imaging. Isolated [149] demonstrated areas of hyperintensity on
finding of T2 signal abnormality, even with DWI (ischemic region) corresponding to a
restricted diffusion, is not specific for ischemia decrease of the local ADC (75 % of normal
and can be seen in transverse myelitis and other values) when compared with non-affected spinal
intrinsic cord pathologies. These findings, confined cord. The time course of the changes in the ADC
to a vascular territory, are more specific for SCI. values corresponds to what has been reported in
the brain [150].

Diffusion-Weighted Imaging (DWI)


of the Spinal Cord Technical Challenges
Several technical difficulties for the application of
Historical Aspects the most frequently available DWI technique, i.e.,
DWI of the spinal cord is technically challenging, single-shot echo-planar imaging (EPI), to spinal
but is currently feasible. Hajnal et al. are the first cord imaging include motion artifacts caused by
to publish the DW images of the human cervical physiological movement of the spinal cord and
47 Spinal Cord Infarction and Differential Diagnosis 1147

surrounding structures (e.g., cerebrospinal fluid during the first 24 h which limits the sensitivity
pulsatile flow, swallowing, respiration, and car- of this finding [80].
diac motion), susceptibility artifacts caused by A finding of restricted diffusion on diffusion-
the presence of bone and cerebrospinal fluid inter- weighted images (DWI-MRI) reported to be sig-
faces, and the low signal-to-noise ratio caused by nificantly more sensitive than standard T2
the small pixel dimensions required for appropri- images, appearing as early as 3–8 h after the
ate visualization of the spinal cord ictus, but the limited evidence precludes a
[151]. Improved imaging techniques such as nav- numerical estimate of its sensitivity [144, 151,
igated interleaved EPI or parallel imaging- 152, 158]. Studies have shown that restricted
enhanced EPI provide superior resolution and diffusion can be seen in SCI as early as 3–8 h
adequate resistance to motion artifacts compared following an ictus [152–154]. Moderate
to the conventional EPI [140]. More recently, line swelling of the cord is noted in the acute phase
scan diffusion imaging (LSDI) has been reported with onset of T1-WI hypointensity in the cord
as new advancement in the DWI of the spinal from days 5–7 [80]. Contrast enhancement is
cord [151]. noted in the subacute stage, typically after
5 days, and persists for up to 3 weeks after onset
Clinical Role [159]. Similar to the cerebral edema, venous
Increasingly, DWI has shown to be highly sensi- hypertension from spinal DAVF without cord
tive to the early detection of ischemic lesions in infarction gives pure extracellular edema, thus
spinal cord and thus can improve the diagnostic increasing T2-WI signal intensity but with normal
capabilities especially in the acute stage findings on DWI.
[152–154]. DWI is also a very useful
technique in differentiating diagnosis of subacute
infarction. After all, hyperintensities on T2-WI Stages of Spinal Cord Infarction
images are not specific for ischemia and can also The abnormal signal intensities in SCI typically
be seen in inflammatory diseases and tumors start in the territory of the anterior spinal artery
[155]. DWI will have increasingly important role and spread to the adjacent central white matter.
for ischemic diseases of the spinal cord not only in The patterns of MR signal abnormality with
early diagnosis but also in differentiating increasing signal changes reflect a temporal
diagnosis [155]. sequence of ischemic changes that can often be
recognized as distinct, separate stages. However,
the MR appearance is frequently a mixture of
Typical MRI Findings patterns, particularly in the patients with com-
pleted spinal cord stroke.
The typical MRI findings in spinal cord infarction Depending on the extent of involvement on
include isolated pencillike area of T2 MRI, spinal cord ischemia can be categorized
hyperintensity involving the centromedullary into four patterns [46]:
region with sparing of the anterior rim, often
encompassing more than two vertebral segments Anterior Horn Ischemia
on sagittal images [2, 156], and bilateral Results in abnormal T2-WI hyperintense signal
hyperintensities that are mostly confined to the involving the anterior horns of the gray matter
anterior horn area, leading to the typical “snake giving an “owl’s eye” appearance (Fig. 13a).
eyes or owl’s eyes” configuration on the axial
acquisitions [156, 157]. Although the T2
hyperintensities are seen in 90 % of patients with Central Gray Matter Ischemia
SCI, these are nonspecific [156]. Further in the T2-WI hyperintense signal abnormality involving
acute phase, only 50 % of the patients show a both anterior and posterior horns of the gray mat-
demarcation of T2 hyperintensity of the spine ter (Fig. 13b).
1148 S.R. Boddu et al.

Fig. 13 (a) Anterior horn infarction. Schematic represen- matter infarction on the right. Note the corresponding MRI
tation of the anterior horn infarction on the right. Note on the left depicting the diffuse T2-WI hyperintensity
the corresponding MRI on the left with subtle focal involving the central gray and white matter of the spinal
T2-WI hyperintensity in the anterior horns of the spinal cord. (d) Cross-sectional infarction. Schematic representa-
cord. (b) Central gray matter infarction. Schematic repre- tion of the cross-sectional infarction on the right. Note the
sentation of the central gray matter infarction on the right. corresponding MRI on the left with diffuse focal T2-WI
Note the corresponding MRI on the left demonstrating hyperintensity involving the cross-sectional area of the
focal T2-WI hyperintensity in the central gray matter of spinal cord. Note the thin rim of preserved normal cord
the spinal cord. (c) Central gray and white matter infarc- parenchyma secondary to the perfusion from peripheral
tion. Schematic representation of the central gray and white pial plexus

Gray and White Matter Ischemia Cross Section Ischemia


T2-WI hyperintense signal abnormality involving T2-WI hyperintense signal abnormality involving
the entire gray matter and the adjacent white mat- the entire cross section of the spinal cord involv-
ter (Fig. 13c). ing both gray matter and white matter (Fig. 13d).
47 Spinal Cord Infarction and Differential Diagnosis 1149

Associated Vertebral Body Infarction Advanced Techniques

A finding of vertebral body infarction on MRI is a Advanced techniques such as diffusion tensor
useful indicator of vascular nature of the imaging (DTI) and fiber tractography (FT) are
acute spinal cord syndrome [160–163]. However, increasingly investigated in the cord infarction.
this finding is not specific, seen in only 4–35 % of These techniques have technical challenges limit-
patients with spinal cord infarction [1, 146, ing the image quality, related to the highly mag-
164]. SCI may not always be accompanied by netically inhomogeneous material surrounding
vertebral body infarction, because arterial the spinal canal, small size of the spinal structures,
occlusion may be located distal to those long craniocaudal extent of the spinal canal, pul-
vessels supplying the vertebral body or sation effects of cerebrospinal fluid (CSF) and
because of the good collateral blood supply of blood, and motion degradation from respiration,
the vertebra [160]. Cheng et al. [164] swallowing, and patient movement. In patients
showed that concomitant spinal cord and with cervical cord injury, fractional anisotropy
vertebral body infarction is highly associated (FA) of DTI and FA showed better correlation
with aortic pathology, which highlights the with motor function and clinical findings such as
common blood supply segmental arteries neurological impairments compared to the con-
supplying the metamere and radiculomedullary ventional MRI [167]. DTI imaging of the spinal
branches of the radicular artery (branch of spinal cord is currently only a research tool, but prelim-
trunk of the segmental artery) feeding the spinal inary studies have shown that it holds consider-
cord [13]. able promise in predicting the severity of spinal
cord injury [168].
Useful Clues on MRI to the Underlying
Etiology
1. Intervertebral disk disease at the level of
infarction: possible fibrocartilaginous Follow-Up Imaging
embolism
2. Spondylotic disease at the level of Unlike cerebral infarct, onset of T2 hyperintense
infarction: possible compressive myelopathy signal abnormality in the spinal cord infarction is
[164] variable from 8 to 72 h, and studies reported
3. Multiple serpiginous flow voids on the surface normal MRI findings at 48 h following ictus
of the spinal cord: spinal cord vascular [157, 169]. High index of clinical suspicion war-
malformations [165] rants follow-up imaging. However, up to 14 % of
patients with suspected SCI have normal follow-
up MRI scans [10, 166].
Holistic Approach Repeated or follow-up DWI may also help in
differentiating diagnosis. There seems to be more
In a patient with suspected SCI, the MRI signal sensibility on DW images than on T2-WI images
abnormalities alone are nonspecific. Care should during signal changes within the course of dis-
be taken to interpret these findings in conjunction ease, and DWI may be useful in monitoring the
with history, clinical findings, and CSF analysis. progress of the disease [155]. Serial MR studies
Although CSF analysis in spinal cord infarction with contrast showed sequential changes of
is typically normal, but there can be pleocytosis enhancement similar to those seen in cerebral
(rarely more than 100 WBC) and an infarcts [169]. Understanding of characteristic
elevated protein (usually less than 119 mg/dl) MR findings and evolutional changes at different
[7, 9, 10, 166]. stages of SCIs and follow-up using serial MRI
1150 S.R. Boddu et al.

with contrast are vital in the early diagnosis The list of differential diagnosis is summarized
and more effective in the management of the in the table. Main differential diagnoses are
patients. discussed below.

Differential Diagnosis Multiple Sclerosis (MS)

The differential diagnosis of spinal cord infarction Multiple sclerosis is a chronic inflammatory
is broad and holds multiple entities under the roof demyelinating disease of the central nervous sys-
of “acute nontraumatic myelopathy.” These can tem (CNS). The frequent involvement of spinal
be categorized into (a) inflammation, cord in MS is shown up to 99 % in autopsy series
(b) infection, (c) compressive myelopathy, [170]. 70–80 % of the patients with MS have
(d) venous congestion related to vascular T2-WI cord signal abnormalities [171]. MS
malformations such as spinal dural arteriovenous plaques are best demonstrated on the T2-WI
fistula (SDAVF), and (e) tumor. Distinction of SCI acquisitions, with signal characteristics of T2-WI
from “SCI mimics” is challenging especially in hyperintensity and T1-WI iso-hypointensity
subacute stage. After all, hyperintensities on [172], similar to cord infarction. Cord swelling is
T2-WI images are not specific for ischemia and a usual feature of the relapsing-remitting form of
can also be seen in inflammatory diseases and MS [173, 174], and similarly cord atrophy with
tumors [155]. axonal loss is a recognized form of MS
Sudden onset of symptoms may support the [175]. Post-contrast enhancement of the demye-
diagnosis of medullary ischemia. In addition, lination lesions is described due to the associated
due to the characteristics of blood supply in breakdown of the blood-brain barrier, although
the spinal cord, infarction lesions are confined to significantly lower than their brain
arterial territories and usually lie in the anterior counterpart [176].
or posterior part of the spinal cord, whereas However, the MS lesions differ from SCI in
inflammatory or neoplastic medullary lesions pattern of cord involvement and extent of CNS
often occupy or affect the whole medulla. distribution. MS plaques present as well-
Detection of synchronous lesions in the brain circumscribed foci of T2-WI hyperintensity with
may isolate some of the differential diagnosis asymmetric involvement of the cord. They are
(Table 1). characteristically peripherally located, are less
DW MRI will have increasingly important role than two vertebral segments in length, and occupy
for ischemic diseases of the spinal cord not only in less than half the cross-sectional area of the cord.
early diagnosis but also in differentiating diagno- On sagittal acquisitions, they are located centrally,
sis [155]. Marked hyperintensity on DWI may anteriorly, and dorsally. On axial images, plaques
provide another indicator leading to the specific are located in the lateral segments and have wedge
diagnosis of spinal medullary ischemia. Mild shape with the basis at the cord surface or round
hyperintensity was likely to be seen on DW shape if there is no contact with the cord
images of myelitis or tumors, suggesting the pos- surface [172].
sibility of separating ischemic from inflammatory Unlike SCI, MS predominantly involves cer-
cord lesions and intramedullary tumors by the vical cord (62 %) and usually demonstrates
different range of ADC values [153, relapsing-remitting disease course [172]. Simulta-
155]. Repeated or follow-up DWI may also help neous imaging of the brain to detect synchronous
in differentiating diagnosis. There seems to be lesions helps to differentiate MS from cord infarc-
more sensibility on DW images than on T2-WI tion. Up to 92 % of MS patients with cord lesions
images during signal changes within the course of have associated brain lesions [177] (Fig. 14).
disease, and DWI may be useful in monitoring the Detection of synchronous lesions in brain and
progress of the disease [155]. spinal cord is useful in differentiating the MS
47 Spinal Cord Infarction and Differential Diagnosis 1151

Fig. 14 Multiple sclerosis: a 41-year-old female cord (b, d, f). The thoracic cord lesion shows post-contrast
presenting with relapsing-remitting course of neurological enhancement (e, f) suggestive of active demyelination
deficits. MRI – multiple T2-WI hyperintense demyelin- plaque. Simultaneous brain MRI shows multiple intracra-
ation plaques involving the cervical (a, b) and thoracic nial white matter lesions with distribution of multiple scle-
(c, d) spinal cord. MS plaques are peripherally located rosis involving the corpus callosum and peritrigonal
and occupy less than half the cross-sectional area of the distribution (g–i)

from other inflammatory and cerebrovascular dis- ITM has reported a prevalence of 15.6 % in a
eases with an accuracy of 93–95 % [178–180]. retrospective study involving 288 patients with
transverse myelitis [185]. ITM predominantly
affects the middle-aged adults. The term “trans-
Idiopathic Transverse Myelitis (ITM) verse” is self-explanatory describing the position
of the inflammation, that is, across the width of the
Transverse myelitis (TM) is predominantly spinal cord. Thoracic cord is most commonly
immune system mediated [181]. This may exist involved [182]. The criteria proposed by the
as an isolated idiopathic entity or as a part of the Transverse Myelitis Consortium Working Group
systemic process with reported incidence in for idiopathic transverse myelitis [186] include:
patients with vasculitis, neurosarcoidosis, multi-
ple sclerosis, and systemic lupus erythematosus 1. Bilateral sensory, motor, or autonomic spinal
[182] and following vaccination [183, 184] and cord dysfunction
acute gastrointestinal and respiratory 2. Defined sensory level with bilateral signs and
illness [181]. symptoms
1152 S.R. Boddu et al.

Fig. 15 Idiopathic
transverse myelitis: a
23-year-old female with
abrupt onset paraplegia.
Diffuse T2-WI
hyperintense cord signal
abnormality with edema
and cord swelling (a–c).
Note the signal abnormality
does not follow a specific
arterial territory and
involves more than
two-thirds of the cross-
sectional area of the spinal
cord (b, c). No associated
intracranial lesions are
demonstrated on the MRI
brain performed
simultaneously

3. Proof of spinal cord inflammation on MRI or differentiating these two entities. A history of
CSF recent vaccination or viral illness may suggest
4. Duration between symptoms’ onset to maxi- ITM, but these are often absent. Symptoms isolated
mum intensity: few hours to 21 days to the anterior spinal artery territory suggest infarc-
5. Exclusion of extra-axial compressive etiology tion over inflammation, but this is not definitive.
Progression of symptoms over at least a few hours,
Typical MR findings of ITM [182] include symptoms and MRI lesions that extend beyond a
focal centrally located T2-WI hyperintensity, usu- vascular territory, gadolinium enhancement on the
ally occupying more than two-thirds of the spinal initial presentation, and cerebrospinal fluid find-
cord cross-sectional area (88 %), signal abnormal- ings of pleocytosis or elevated IgG levels favor
ity that extends for more than three to four verte- transverse myelitis over infarction [188].
bral segments in length (53 %), variable presence
of cord swelling or expansion (47 %), and patchy
or diffuse post-contrast enhancement (47–53 %) Acute Disseminated Encephalomyelitis
[182, 185]. The variability in the duration of clin- (ADEM)
ical onset, extent, and pattern of cord signal abnor-
mality and given up to 40 % of ITM cases can have Acute disseminated encephalomyelitis (ADEM)
normal MR imaging [187] make it quite challeng- is a non-vasculitic inflammatory demyelinating
ing to differentiate ITM from cord infarction disorder of the central nervous system, most likely
(Fig. 15). A detailed history of associated systemic triggered by the autoimmune response to the mye-
conditions, through clinical examination and lin basic protein [172]. This usually affects the
close monitoring of the clinical condition in com- prepubertal children, in contrary to the SCI. The
bination with CSF analysis, may be helpful in thoracic cord is most commonly affected. The
47 Spinal Cord Infarction and Differential Diagnosis 1153

Fig. 16 Acute disseminated encephalomyelitis (ADEM): distribution (a–d). Presence of intracranial lesions in the
an 8-year-old boy with history of recent viral illness brachium pontis (e), thalamus (f), deep white matter (g),
presented with weakness in all four limbs and flaccid and centrum semiovale (h) on brain MRI performed simul-
reflexes. MRI: T2-WI hyperintensity involving the cervical taneously. Child was diagnosed with ADEM
and thoracic cord with no specific arterial territory

onset of ADEM usually occurs in the wake of a cortical lesions [192, 193]. The associated brain
clearly identifiable febrile prodromal illness or lesions explain the frequent clinical manifesta-
immunization and in association with prominent tions of brain stem signs and ataxia in ADEM
constitutional signs and encephalopathy of vari- [194] along with routine motor and sensory defi-
able degree [189], feature that are uncommon in cits seen in the context of acute myelopathy.
cord infarction. MRI findings of the spinal cord Rarely, ADEM lesions may contain areas of
are nonspecific with large T2-WI hyperintense hemorrhage, suggestive of herpes simplex virus
and T1-WI hypointense lesions extended over 2 (HSV-2) encephalomyelitis [193, 195], which
long segment of the spinal cord with associated can mimic the hemorrhagic cord infarction.
cord expansion [190] (Fig. 16). Spinal cord
involvement is noted in 71 % of patients, and all
patients with spinal involvement have cerebral Spinal Cord Arteriovenous Shunts
involvement and features of myelopathy
[191]. Thus, simultaneous cerebral imaging to Spinal cord arteriovenous shunts (SCAVS) are a
detect brain lesions helps to differentiate this rare heterogeneous group of vascular
entity from cord ischemia. The typical signs of malformations presenting with similar MR imag-
ADEM described in literature are involvement of ing findings mimicking the ischemic and/or hem-
the basal ganglia, thalamus, brain stem, and orrhagic spinal cord infarction.
1154 S.R. Boddu et al.

Fig. 17 Spinal epidural arteriovenous fistula (DAVF): a swelling (a–c) suggestive of spinal vascular malformation.
77-year-old male with chronic back problems and prior Also note the extensive degenerative changes in the lumbar
spinal surgery presented with new onset right lower spine and prior surgery (a). This patient had extensive
extremity weakness following L4/L5 injection. MRI – search for SVM on DSA that was localized to the left
multiple serpiginous flow voids over the cord surface internal iliac artery (d) on the second spinal DSA
with associated T2-WI hyperintense cord edema and

Spinal dural arteriovenous fistula (SDAVF) is a enhancement in subacute stage and cord atrophy
close differential diagnosis for the spinal cord in chronic stage [200]. However, additional
infarction (Fig. 17). SDAVF constitutes 70 % of T2-WI findings of dilated perimedullary vessels
all spinal vascular malformations (SVMs), com- manifested as tortuous flow voids [197], and a
monly affecting middle-aged (40–60 years) hypointense rim related to deoxygenated blood
males. It usually presents as slowly progressive within the dilated capillary vessels surrounding
neurological deficits; however, acute onset of the the congestive cord edema [201] is characteristic
disease and progressive deterioration interrupted of SDAVF and helps to differentiate the two enti-
by remissions have been described [196, 197]. If ties. If the shunt volume is small, the
left untreated, it results in irreversible paraplegia perimedullary vessels might only be seen after
or tetraplegia. The increased spinal venous hyper- contrast enhancement. The dilated perimedullary
tension decreases the arteriovenous pressure gra- vessels are seen as filling defects in the continuous
dient resulting in impairment of the normal spinal contrast column in myelography (Fig. 18). It is
venous drainage and resulting in venous conges- important to remember that neither the location of
tion with intramedullary edema [198, 199] and the pathologic flow voids nor the intramedullary
end stage with spinal cord infarction. imaging findings correspond to the localization of
On MRI diffuse edema from congestion results the fistula [80].
in centromedullary T2-WI hyperintensity of the The arteriovenous malformations (AVMs)
cord involving multiple segments and mimics the carry increased risk of intraparenchymal and
cord infarction [197]. Both entities have similar subarachnoid hemorrhage (Fig. 19) in addition
findings of cord swelling and contrast to venous congestion, compared to
47 Spinal Cord Infarction and Differential Diagnosis 1155

Fig. 18 Myelographic
demonstration of spinal
DAVF: a 48-year-old male
with known
spondyloarthropathy
evaluated with
myelography for lower limb
symptoms. Sagittal (a) and
coronal (b) myelography
reformats demonstrating
multiple serpiginous filling
defects over the cord
surface, suggestive of
underlying spinal vascular
malformation. Subsequent
DSA (c) confirmed
spinal DAVF

Fig. 19 Spinal intramedullary arteriovenous malforma- cord infarction. Spinal DSA: intramedullary AVM at C5,
tion (AVM): a 31-year-old male presented with sudden feeders from the ASA, right vertebral artery (e, f), left
onset quadriparesis. MRI: heterogeneous, predominantly ascending cervical artery from thyrocervical trunk (g).
T2-WI hyperintense signal (a, b) with patchy post-contrast Note the right cervical vertebral artery dissection with
enhancement (c, d), which can be seen in subacute spinal small pseudoaneurysm (e, f)
1156 S.R. Boddu et al.

Fig. 20 Ependymoma: a 64-year-old male with progres- mass lesion of the conus medullaris with cord edema and
sive weakness of the bilateral lower limbs. MRI – hetero- swelling (a, c). Note the patchy post-contrast enhancement
geneous predominantly T2 hyperintense intramedullary (b, d)

SDAVF [80]. The AVMs along with cavernous mass effect, and discrete borders mimicking the
malformation can mimic hemorrhagic cord infarc- cord infarction. Ependymomas, astrocytomas,
tion. The presence of intramedullary hemorrhage and hemangioblastomas are the three most fre-
makes MRI findings more complex with heteroge- quently seen primary intramedullary tumors in
neous T1-WI and T2-WI signal abnormalities and the adult population [205]. Majority of these
associated T2-WI peripheral hypointense rim from tumors are usually seen in the cervicothoracic
the blood products [202, 203]. However, the pres- region with some variability in the histologic
ence of intra- and perimedullary flow voids in type [206, 207]. The myxopapillary subtype,
AVMs [80] and typical intramedullary mulberry- which makes up nearly 30 % of all spinal
like appearance, representing hemorrhage in differ- ependymomas, is found almost exclusively in
ent stages [204], is a helpful clue to differentiate the cauda equina and filum terminale [208]
these diagnoses from isolated spinal cord infarction. (Fig. 20). The mean size of ependymomas
corresponds to height of 3–4 vertebral bodies
and 5–6 vertebral bodies for astrocytomas
Spinal Cord Neoplasms [205]. Ependymomas are more centrally located
compared to the eccentric location of the astrocy-
Primary and secondary tumors of the spinal cord tomas and hemangioblastomas. Diffuse avid post-
result in extensive intramedullary T2-WI contrast enhancement with smooth margins is
hyperintensity with associated cord expansion, noted in ependymomas and lesser extent of
47 Spinal Cord Infarction and Differential Diagnosis 1157

Fig. 21 Hemangioblastoma: a 46-year-old male with progressive worsening of lower limb weakness. MRI – large
intramedullary mass lesion, predominantly cystic (a–c) with enhancing mural nodule (arrow) on post-contrast images (d)

enhancement and nodular tumor margins in astro- cervical cord is the most frequent site for ISCM,
cytomas [206, 209]. Hemangioblastomas are although they have reported at all levels of the
either solitary or multiple (von Hippel-Lindau spinal cord [214]. ISCM are associated with dis-
disease) [210] with an intensely enhancing mural proportionate vasogenic edema involving the long
nodule and a large cystic component [206] segments of the spinal cord resulting in T2-WI
(Fig. 21). Hemangioblastoma larger than 24 mm hyperintensity and cord expansion, which may
in size is frequently associated with flow voids on mimic cord infarction [215]. However, the metas-
MRI [207]. These tumors are slow growing and tasis usually shows ringlike or homogeneous
often present with features of osseous remodeling, intense enhancement following contrast adminis-
such as canal expansion and scalloping of the tration [205]. Moreover, most patients have
posterior vertebral bodies [211], which are not known malignancy and systemic metastasis with
seen in acute scenario of cord infarction. In addi- frequent concomitant brain metastasis or
tion, slower clinical onset, diffuse or nodular post- leptomeningeal carcinomatosis at the time of
contrast enhancement, and presence of ISCM diagnosis [216].
intramedullary cystic components favor neoplasm Rarely, an acute ASA infarction of the bilateral
over infarction. anterior horns may be mimicked by a polio viral
Intramedullary spinal cord metastases (ISCM) infection. Such appearance is related to the
are rare with reported frequency of 0.9–1.2 % peculiar viral tropism underlying this
based on the autopsy series from cancer patients condition [217]. Vaccine-associated paralytic
[212, 213]. Lung cancer is the most common poliomyelitis cases have been also reported occa-
source followed by metastasis from the breast, sionally in the pediatric population [218] (Tables 4
kidney, melanoma, and lymphoma [214]. The and 5).
1158 S.R. Boddu et al.

Table 4 List of differential diagnoses for spinal cord tumors [227] (Figs. 31 and 32) to minimize the
infarction risk of SCI. Certainly spinal digital subtraction
A. Inflammation/demyelination angiography (SpDSA) is the ultimate imaging
1. Multiple sclerosisa technique for diagnosing, localizing, and classify-
2. Devic’s neuromyelitis opticaa ing spinal vascular lesions [80].
3. Idiopathic transverse myelitis The congestive myelopathy from spinal dural
4. Acute disseminated encephalomyelitisa arteriovenous fistulas or intracranial dural arterio-
5. Acute polyneuropathy (Guillain-Barré syndrome) venous fistulas with venous reflux into the brain
6. Subacute combined degeneration
stem and spinal cord can mimic spinal cord infarc-
7. Neurosarcoidosisa
tion (Fig. 33). It is proven beyond doubt that the
8. Neuro-Behcet’s diseasea (Fig. 22)
extent and site of the cord signal abnormality or
9. AIDS-associated myelopathya
surface flow void do not correlate with the loca-
10. Radiation myelitis
B. Infection
tion of the SDAVF [80]. The fact that the SDAVF
1. Bacterial myelitis constitutes 70 % of all SVMs [80] highlights the
2. Viral myelitis limitation of the conventional MRI in accurate
3. Fungal myelitis localization of the spinal vascular malformations.
4. Tuberculous myelitis On the other hand, with intrinsic cord pathology
5. Toxoplasmosis such as intramedullary vascular malformation,
C. Compressive myelopathy cavernoma, or intrinsic cord tumor, conventional
1. Epidural/subdural hematoma (Fig. 23) MRI can accurately localize the site of pathology;
2. Epidural/subdural abscess (Fig. 24) however, it fails to offer additional crucial infor-
3. Acute disk herniation with cord compression mation regarding the vascular supply of these
4. Epidural/dural based mass lesions and metastasis lesions that is vital in their treatment
(Figs. 25, 26, and 27)
planning [172].
D. Spinal cord arteriovenous shunts (SCAVS)
(Figs. 28, 29, and 30)
E. Neoplastic
1. Primary intramedullary tumors Challenges in Spinal Angiography
2. Intramedullary spinal cord metastasis
3. Leptomeningeal carcinomatosisa The main challenges in the spinal angiography in
a
Simultaneous brain imaging may provide useful diagnos- general irrespective of the imaging modality are
tic information in differentiating these entities from spinal the following:
cord infarction
1. Differentiation of submillimeter to millimeter
normal spinal cord arteries from veins (ASA,
Role of Angiography 0.2–0.8 mm; AKA, 0.5–1.0 mm; PSA, <0.5
mm) [15] and their relation to the adjacent
The principal role of catheter angiography in structures such as the spinal cord and verte-
suspected spinal cord infarction (SCI) is to con- brae, requiring high spatial resolution.
firm the differential diagnosis of spinal vascular 2. Characterization of SVMs, identification of
malformations (SVMs) and evaluate the patency multiple submillimeter feeding arteries, and
of the anterior and posterior spinal arteries and separation of the inflowing arterial feeders
localization of the dominant radiculomedullary from the outflowing draining veins that
artery (artery of Adamkiewicz). Accurate locali- demand high temporal resolution. Although
zation of the artery of Adamkiewicz is crucial in veins have relatively large caliber (0.4–1.5
the diagnosis and treatment planning of spinal mm) [15], the great anterior radiculomedullary
vascular lesions [225] and thoracoabdominal aor- vein (GARV), which is the largest vein
tic aneurysm repair [226] and for planning the draining the anterior thoracolumbar spinal
preoperative embolization of hypervascular spinal cord, is easily mistaken for the AKA due to
47 Spinal Cord Infarction and Differential Diagnosis 1159

Table 5 Summary of main differential diagnosis for SCI and useful clues
Differential diagnosis Highlights
1. Multiple sclerosis (MS) Peripheral in location
Less than two vertebral segments in length
Less than half cross-sectional area of the cord
>90 % incidence of associated intracranial lesions
Relapsing and remitting clinical course
2. Spinal cord neoplasm Invariable cord expansion present
Diffuse or nodular contrast enhancement
Extensive peritumoral edema
Associated cystic changes
Slower clinical onset
Remodeling of adjacent osseous structures
3. Idiopathic transverse myelitis Lesion centrally located
(ITS) 3–4 segments in length
Occupying >2/3 of cord cross-sectional area
No associated intracranial lesions
Onset not quite as sudden
4. Spinal cord arteriovenous Prominent enlarged serpentine pial veins on cord surface with cord swelling and
shunts (SCAVS) edema
May rarely show only cord expansion and edema on T2-WI, without prominent
vessels
Hypointense rim related to deoxygenated blood within the dilated capillary
vessels surrounding the congestive cord edema
Neither the location of the pathologic flow voids nor the intramedullary imaging
findings correspond to the localization of the fistula
Gradual progression
5. Guillain-Barré syndrome [219] Back pain, progressive, fairly symmetric muscle weakness accompanied by
(GBS) absent or depressed deep tendon reflexes
Ascending symptoms and development of upper motor neuron signs over time
No definite sensory level
6. Acute disseminated Usually affect prepubertal children
encephalomyelitis (ADEM) Commonly involves thoracic cord
Preceded by a prodromal febrile illness or immunization
Synchronous brain lesions: basal ganglia, thalamus, brain stem, and cortical
lesions
7. Subacute combined Vitamin B12 deficiency. Gradual onset
degeneration (SACD) [220] Associated pernicious anemia or neuropsychiatric symptoms
Most commonly affects posterior columns, followed by anterolateral and anterior
tracts
Bilateral hyperintense T2-WI hyperintense areas in posterior columns of the
cervical and thoracic cord
Typically no post-contrast enhancement
Clinical and imaging improvement after B12 supplementation
8. AIDS-associated myelopathy Vacuolar myelopathy. Gradual onset
[221, 222] Edematous swelling of the myelin in the absence of demyelination or
inflammation
Begins in the mid-low thoracic cord and extends rostral with disease progression
Bilateral symmetrical T2-WI hyperintensity in dorsal and lateral columns
extending over multiple segments
Typically no post-contrast enhancement
9. Radiation myelitis [211, 223] Acute: transient myelopathy or Lhermitte’s sign or acute paraplegia
Chronic: lower motor neuron disease of extremities and chronic progressive
myelitis
Prior radiation to the spinal cord with no active compression
(continued)
1160 S.R. Boddu et al.

Table 5 (continued)
Differential diagnosis Highlights
3–40 months latent period
Acute: long segment T2-WI hyperintensity, cord swelling, and post-contrast
enhancement
Chronic: cord atrophy and cystic
10. Compressive myelopathy Postoperative period, known malignancy, or degenerative spine disease
[224] Hematoma or abscess in the epidural or subdural location
Usually has a more subacute presentation with evolution over time; this
presentation may be obscured in the perioperative period
Postoperative setting; the differential diagnosis of paraplegia includes an epidural
hematoma, especially if the patient has had either a lumbar drain or epidural
anesthesia

Fig. 22 Neuro-Behcet’s disease: a 36-year-old male with predominantly unilateral hyperintense lesions (c–e) with
history of oral ulcerations presented with progressive increased diffusivity (f) involving the posterior limb of the
weakness of all limbs, lower limbs more than upper internal capsule, thalamus, cerebral peduncle, and pons
limbs. MRI: T2-WI hyperintense lesion in the cervical along the course of the corticospinal tracts, typical features
cord involving two-thirds of the cross-sectional area and seen in CNS involvement of Behcet’s disease
cord swelling (a, b). Simultaneous MRI brain shows
47 Spinal Cord Infarction and Differential Diagnosis 1161

Fig. 23 Epidural hematoma: a 62-year-old female with hyperintense (a) and T1-WI hyperintense signal (b) com-
history of thrombocytopenia presenting with back pain and pared to adjacent cord. Faint post-contrast enhancement (c)
increasing leg weakness. MRI – anterior epidural hema- suggests subacute nature of this hematoma
toma in the lumbar region (arrows) with T2-WI iso-/

its comparable spatial course and location architecture of the malformation, superior demon-
[228] (Fig. 34). stration of the nidal and venous aneurysms,
3. Coverage of a large area of potential fistulous enhance distinction of pathologic vessels in rela-
communication from the skull base, perhaps tion to the spinal cord (perimedullary versus
including the posterior fossa and caudally to intramedullary) [233], and offer a 3D relationship
the level of internal iliac arteries which requires of the vasculature to the bone [234]. However,
a large field of view (FOV), which itself chal- DSA is time-consuming which challenges patient
lenges the high spatial resolution. compliance and has small risk of complications
inherent to its invasive nature. Performing spinal
Spinal Digital Subtraction Angiography DSA under general anesthesia helps to obtain
Spinal digital subtraction angiography (SpDSA) better quality images by dedicated timing of
with its high spatial and temporal resolution is the apnea and overcomes the limitations related to
gold standard for the study of spinal vascular patient compliance. So a noninvasive spinal vas-
anatomy, detection of SVMs, localization of the cular imaging method such as spinal CTA or
AKA, and differentiation of embolic spinal cord MRA that can substitute for or guide DSA to
infarction from a non-embolic entity. The sensi- shorten acquisition time and reduce the complica-
tivity of spinal angiography in detecting the AKA tions is highly desirable in clinical practice.
is very high and approaches 100 % [23,
229–232]. The three-dimensional rotational spi- Spinal Multidetector CT Angiography
nal angiography (3D RSA) performed as a sup- The advent of multidetector spiral CT has enabled
plement to DSA was reported to better delineate very short acquisition time and increased scan
the complex, extremely tortuous vascular range and high spatial and temporal resolutions.
1162 S.R. Boddu et al.

Fig. 24 Epidural abscess: a 56-year-old female with pro- enhancement and dural thickening on post-contrast imag-
gressive worsening of back pain and lower limb weakness. ing (b, c). Note the underlying diskitis in the lower cervical
MRI: T2-WI hyperintense anterior epidural collection in region at C6/C7 and C7/T1 with associated large
the upper thoracic cord (a) with peripheral rim prevertebral abscess (d)

Fig. 25 Meningioma: an
84-year-old female with
chronic left lower limb
weakness. MRI – focal
dural-based mass lesion
dorsally over the conus
medullaris. The lesion is
isointense to the cord on
T2-WI sequence with
associated cord
compression and intrinsic
cord signal abnormality (a).
The lesion shows mild post-
contrast enhancement (b)
47 Spinal Cord Infarction and Differential Diagnosis 1163

Fig. 26 Nerve sheath tumor: a 34-year-old male with cord compression and intrinsic T2-WI hyperintense signal
worsening left lower limb weakness. MRI – heterogeneous (arrow) abnormality (a, b). Note the post-contrast
predominantly cystic extra-axial lesion over the left enhancement (c, d) in the solid component of the tumor
anterolateral aspect of the conus medullaris with associated (arrows)

All these features increased the feasibility of CT Spinal Magnetic Resonance Angiography
spinal angiography. MDCT angiography takes In recent years, CE-MRA has come up with many
only seconds, has submillimeter spatial resolu- innovative developments, including the 3D MRA
tion, and can even be performed in dynamic fash- and time-resolved MRA (TR-MRA). Conven-
ion [235]. Takase et al. reported that MDCT tional MR angiography techniques, including
(four-detector row) could clearly visualize the contrast-enhanced time-of-flight (TOF) and
normal medullary arteries and veins [236]. Lai phase-contrast angiography (PCA), offered suffi-
et al. using a 16-detector-row MDCT reported cient spatial resolution but were not able to depict
good correlation between CT angiography and the normal arteries of the spinal cord [241]. With
DSA for SDAVFs [237, 238]. Si-jia et al. using a contrast-enhanced TOF MR angiography, only
64-detector-row MDCT for CT angiography normal (large) veins in the spinal cord were
published similar results. Authors have admitted depicted to some extent [242]. For PCA, no
the role of CTA as a quite valuable screening reports on the feasibility of depicting normal ves-
exam before DSA with guiding effect on DSA, sels were published.
thus reducing the amount of time, contrast, and Contrast-enhanced 3D MR angiographic
radiation involved in DSA [237–239]. approaches appear successful for the localization
CTA offers superior spatial and temporal reso- of the AKA at 1.5T field strength. Current
lution over MRA. The wider scan range offered contrast-enhanced MR angiography techniques
by CTA would be a lot more time-consuming for claim success rates for the detection of the AKA
MRA [240]. ranging from 69 % to 100 % [22, 226,
1164 S.R. Boddu et al.

Fig. 27 Vertebral metastasis with epidural extension: a resulting in cord edema, and swelling on sagittal T2-WI
64-year-old female presenting with worsening quadriple- (a) and post-contrast images (b–d). Note the large pleural
gia in the lower limbs. MRI – vertebral metastasis involv- effusion in the right hemithorax (*) and underlying primary
ing C4–C6 and T5 with pathological compression fractures lung tumor in the left hemithorax (#)
of C5 and T5, epidural soft tissue, cord compression

Fig. 28 Cavernoma. Intramedullary vascular lesion with cord edema, and faint post-contrast enhancement. Cervical
lobulated T2-WI hyperintense signal (popcorn pattern), (a–c) and lower thoracic (d–f) cavernomas from two dif-
rim of hypointensity related to hemosiderin, adjacent ferent patients are included
47 Spinal Cord Infarction and Differential Diagnosis 1165

Fig. 29 Posterior spinal artery aneurysm: a 39-year-old showed T1 hyperintense focal hematoma in the distribu-
male presented with headache, back pain, and diplopia. tion of left PSA (b, c). Further evaluation with DSA
MRI brain showed subarachnoid hemorrhage at the con- showed a tiny left PSA aneurysm as a culprit for the local
vexity sulci (a) with no intracranial aneurysm. MRI spine hematoma and diffuse SAH

243, 244]. The approach uses a strong bolus, of centric k-sampling technique of a bolus-shaped
which mainly the first passage is exploited for signal time course, the contrast between arteries
imaging during a period of 20–40 s [226]. The and veins increases with vessel caliber [244] and
technique relies on intravenous contrast adminis- is, therefore, better for larger (typically extradural)
tration and spoiled gradient-echo pulse sequences than for smaller (intradural) vessels.
with ultrashort TE and TR for most effective Ali et al. [245] used the newer technology of
signal intensity from T1 shortening of the gado- dynamic multiphase time-resolved MRA in
linium and suppression of the non-enhanced back- 11 patients with suspected SVMs. The authors
ground tissue signal intensity, respectively. The correctly diagnosed six vascular lesions and
main advantage of the strong bolus technique is were able to localize within one vertebral level
that it may provide separation of intradural arter- in 5 of the 6 cases. More recently Saindane
ies and veins. However, as the technique uses et al. [246] also used CE-TR-MRA for the initial
1166 S.R. Boddu et al.

Fig. 30 Spinal epidural arteriovenous fistula: a 70-year- sacral branches of the iliosacral arteries (b) and single
old female with progressive paraplegia being evaluated for epidural draining vein (d). Patient had successful Onyx
spinal canal stenosis demonstrates edema and swelling of embolization of the bilateral arterial feeders with venous
the conus medullaris with multiple flow voids over the penetration resulting in complete obliteration of the fistula
spinal cord surface on T2-WI MRI (a). Further evaluation (c). Mild improvement of the paraplegia is noted immedi-
with DSA confirms the sacral epidural arteriovenous fistula ately following the procedure
at L5/S1 level with dominant arterial feeders from the left

evaluation of suspected SDAVFs, with high con- of diagnostic information offered, conventional
cordance with DSA. Authors showed that in cases angiography is unmatched. Although CT angiog-
where the CE-TR-MRA is able to localize an raphy and MR angiography provide excellent ana-
arterial feeder, the number of catheterized vessels tomic definition of intracranial vascular lesions
and total contrast dose can be decreased during comparable to that of DSA, these modalities
DSA, with attention to the suspected 1 segmen- have not been well developed for the evaluation
tal levels. However, if the CE-TR-MRA study of spinal vascular lesions. Indeed, the size of
does not confidently identify an arterial feeder or feeding vessels to these malformations is usually
is of suboptimal quality, a full spinal DSA is beyond the resolution of these modalities. The
recommended. spatial resolution (0.2 mm) and temporal resolu-
tion (0.25 s) of catheter cerebral angiography
Spinal DSA Versus CTA and MRA remain unparalleled. CTA may be approaching
It is not unreasonable to question, what is the this for some applications, with potentially
justification to use DSA over these noninvasive 0.4-mm spatial resolution and 0.5-s temporal res-
techniques? With regard to accuracy and breadth olution, whereas MRA remains a little further
47 Spinal Cord Infarction and Differential Diagnosis 1167

Fig. 31 Filum terminale paraganglioma: a 74-year-old (c). Note multiple flow voids over the cord surface (b) that
female evaluated for new onset bilateral lower limb weak- enhance following contrast administration (c). Time-
ness. MRI – focal extradural mass lesion in the filum resolved MRA (d) confirms the vascular nature of this
terminale with T2-WI hyperintensity (a), T1-WI isointense pathologically confirmed paraganglioma
(b) signal to the cord and avid post-contrast enhancement

away with regard to 1-mm spatial resolution and techniques. Selective arterial injections in DSA
2–3-s temporal resolution on original acquisition. offer superior contrast resolution (2,000–7,000
Thus, conventional angiography remains the clear HU) compared to contrast administration from
winner in these measures of performance [247]. the peripheral venous access in CTA or MRA
This variation of spatial and temporal resolu- [233]. These factors combined with cross-
tion reflects DSA comparable sensitivity of CTA sectional imaging technique-specific limitations,
and MRA for the feeding arteries and fistulas of such as artifacts, translate into clinically meaning-
SDAVF [239], against other SVMs. This is ful differences in accuracy between these nonin-
because the feeding arteries of SDAVF are the vasive techniques (CTA and MRA) and
radiculomedullary arteries arising from intercostal conventional angiography [247].
arteries, the diameter of which is relatively larger. The risk of ionizing radiation and injection of
However, their major limitation is in the detection iodinated contrast material is shared by both CTA
of all the feeding arteries in SVMs (low spatial and DSA, where MRA takes a clear edge. Both
resolution) and most importantly the arterialized CTA and MRA require tedious post-processing of
draining vein [239] (low temporal resolution), source images, requiring patience and
which is the primary treatment target. Further the experience [228].
inferior resolution of CTA and MRA limits pre- MRA is limited by long acquisition time, lim-
cise distinction between the intramedullary AVM ited scanning range, and lower spatial resolution
and perimedullary AVF, which is crucial in the compared with MDCT [239]. The small scan
treatment planning. range explains the limitation of arterial feeder
Contrast resolution is another important dis- detection for SDAVF [240, 246, 248, 249] and
criminator of DSA and noninvasive angiography repetition of MRA in published series [240],
1168 S.R. Boddu et al.

Fig. 32 Hypervascular vertebral body metastasis: a obvious tumor blush (b, e), which was successfully oblit-
78-year-old female with history of renal cell carcinoma erated by the particle embolization (c, f) minimizing the
and back pain presented with sudden onset paraplegia intraoperative blood loss. Remember, the torrential
from vertebral metastasis. Patient underwent surgical intraoperative blood loss from these hypervascular tumors
decompression (surgical clips in a, d) that was precluded without preoperative embolization may precipitate
by the extensive blood loss. Subsequent preoperative hypovolemia and spinal cord infarction
embolization of this hypervascular metastasis shows

highlighting the possibility of the feeder source Spinal DSA (SpDSA) is a highly skilled pro-
being outside the limited field of view. In general, cedure and is technically challenging. Unfortu-
however, the imaging quality of MR-based nately, the safety of this procedure has a poor
modalities is easily affected by motion degrada- historical reputation, and there are few current
tion secondary to respirations, especially in the reports on complication rates. The assumptions
thoracolumbar region, and the spatial resolution that SpDSA poses a high risk of neurologic com-
does not yet approach the sensitivity or anatomi- plications, requires general anesthesia, and
cal detail provided by standard DSA [249]. Of exposes patients to dangerous doses of radiation
course, the major merit of MRA is not using and contrast remain prevalent, despite being
ionization radiation, so the research of MRA has largely based on historical data [250,
rich prospect. As MRI continues to improve, it is 251]. These perceptions are maintained, in part,
likely that in the near future it will play a major by the scarcity of contemporary literature on this
role in the diagnosis of vascular lesions of the subject [76, 231]. Neuroradiologists relatively
spinal cord. less experienced in catheter angiography will
47 Spinal Cord Infarction and Differential Diagnosis 1169

Fig. 33 Intracranial DAVF with vertebral venous drain- (a, b) suggestive of underlying vascular shunt. DSA con-
age (type 5): a 72-year-old male with worsening quadri- firms the posterior fossa DAVF with arterial supply from
plegia. MRI – diffuse edema and swelling involving the the meningohypophyseal trunk and venous drainage into
craniocervical junction and cervical cord with multiple the vertebral venous plexus (c–e) resulting in venous con-
flow voids predominantly over the dorsal cord surface gestion and cord edema

further skew the imaging in the direction of CTA provide guidance for DSA. Despite the improve-
and MRA, irrespective of the accuracy of these ments in technology discussed above, neither
noninvasive techniques relative to catheter angi- cross-sectional imaging modality, CTA or MRA,
ography [252]. More recent evidence based on currently approaches the necessary degree of sen-
302 spinal angiograms showed no intra- or post- sitivity and spatial resolution of standard DSA to
procedure neurological complications and neph- rule out spinal vascular lesions. Therefore, DSA
rotoxicity, which reflects the effect of modern remains the “gold standard” in the diagnosis of
advances in endovascular technology [253] and these often-elusive spinal vascular lesions
emphasizes that SpDSA in the light of modern [234]. 3D RSA is a useful adjunct to conventional
technological advances performed by experienced DSA and helps to provide the 3D relationship of
practitioners in an appropriate patient population the vasculature to the bone to better distinguish
is relatively a safe procedure. between arterial and venous anatomy.

Summary
In summary, conventional MRI is the initial Treatment
screening investigation in the evaluation of spinal
vascular malformation, a common differential Ischemia if left untreated becomes progressively
diagnosis for SCI. The main purpose of spinal worse over the first few hours [96]. The primary
CTA and MRA is to display the feeding arteries mechanism of injury reaches plateau by 24 h,
and fistulas of vascular malformations and beyond which the injury from the secondary
1170 S.R. Boddu et al.

Spinal Cord Ischemia Secondary


to Embolism

Thrombolysis for acute spinal cord ischemia is


still evolving and has only been reported in liter-
ature a few times. Intravenous thrombolysis with
recombinant tissue plasminogen activator was
reported in one patient. This case was quickly
diagnosed as it happened as a complication of an
aortography evaluating a fusiform abdominal
aneurysm and therefore was within a 3-h treat-
ment window. The patient made a remarkable
recovery and was discharged 72 h after with
almost no deficits [254]. Three other cases were
reported, which were treated by introducing strep-
tokinase and steroids selectively via endovascular
therapy. Three injections were given in 1-week
intervals. All patients recovered with no to mini-
mal neurological (one with mild spasticity of the
lower extremities and another with minimal sen-
sory impairment) [255]. The main obstacle to such
interventions is timeliness, as there are contrain-
dications to using thrombolytic therapy [256] and
excluding these, as well as diagnosing spinal cord
Fig. 34 Great anterior radiculomedullary vein (GARV).
In spite of the relatively large caliber, the great anterior ischemia, may take beyond the optimal therapeu-
radiculomedullary vein (GARV), which is the largest vein tic window.
draining the anterior thoracolumbar spinal cord, is easily
mistaken for the artery of Adamkiewicz (AKA) due to its
comparable spatial course and location. This highlights the
importance of high temporal resolution and real-time imag- Spinal Cord Ischemia Secondary
ing of DSA in differentiating the small spinal arteries and to Hypoperfusion
veins
The hypotension/hypoperfusion can be a primary
mechanism dominates the picture [92]. This high- mechanism caused by myocardial infarction and
lights the importance of acute phase treatment in internal hemorrhage or may be part of secondary
the first 24 h in reversing the primary mechanism mechanism of cord damage. The precipitating
and minimizing the damage from the secondary event should be addressed immediately. Specific
mechanism. In addition to the supportive mea- management guidelines have been developed to
sures, the current treatment strategies for acute limit or reverse neurological deficits in spinal cord
spinal cord ischemia are broadly categorized ischemia after aortic surgery and thoracic
based on the primary mechanism of injury, i.e., endovascular aortic repair (TEVAR) [257]. There
embolism versus hypotension/hypoperfusion. is a lesser chance of reversing this pathology when
The primary aim is to treat the primary cause as the patient awakes from anesthesia with neurolog-
soon as possible and restoration of normal spinal ical deficit, rather than the findings developing
cord circulation. Early resuscitation and support- after a period of normal neurological function
ive measures help to minimize the spinal cord postoperatively. Measures such as increasing sys-
damage from the secondary pathophysiological tolic blood pressure to 95 mmHg and decreasing
mechanisms and may improve the long-term CSF pressure to 10 mmHg can result in symptom
outcome. relief and lead to recovery in a majority of affected
47 Spinal Cord Infarction and Differential Diagnosis 1171

patients with delayed onset symptoms of SCI after Meticulous search for spinal artery aneurysms
aortic surgery [258]. Alternatively preemptive should be carried out in the evaluation of spinal
measures can include intraoperative neurophysio- SAH. Extrinsic compression on the spinal cord
logic monitoring, which can identify SCI, while triggering ischemic changes such as epidural
the patient is still under general anesthesia. hematoma, abscess, extramedullary tumors, and
Lumbar CSF drain placement should be metastasis needs timely surgical decompression
considered prior to starting the vasopressors in and/or radiotherapy. In the event of epidural
the event of suspected SCI to maintain CSF pres- abscess, thorough scrutiny should be made for
sure between 8 and 12 mmHg. These measures diskitis/osteomyelitis as well as the source of sys-
have been shown to mitigate and, at times, reverse temic sepsis. Finally, always check for the syn-
the neurological sequelae in both intra- and post- chronous pathologies such as cervical disk and
operative scenarios. Other measures include intramedullary SVM.
epidural cooling, sequential sacrifice of intercostal
vessels before aneurysm excision, and the use
of distal aortic perfusion during these Role of High-Dose Steroids
procedures [257].
Following acute spinal trauma, administration of
high-dose methylprednisolone within 3 h is more
Treatment of Underlying Cause beneficial than within a 3–8-h window in patients
with complete and incomplete spinal cord inju-
The improvement of the neurological function ries. High-dose steroids may improve spinal cord
following the removal of hematomyelia was blood flow and microvascular perfusion [110,
documented as early as 1911 in canine studies 261] as well as clinical neurologic recovery after
[86]. Authors speculated that there was a putative experimental spinal cord injury [262]. They may
“biochemical factor” that was present in the hem- also provide some cytoprotection through the
orrhagic necrotic material at the epicenter of inhibition of lipid peroxidation, facilitate spinal
hematomyelia and that may be instigating ongo- cord impulse generation, and inhibit
ing damage [87]. Hemorrhage may promote prostaglandin-induced vasoconstriction [263].
ischemia [259] or induce thrombosis via platelet Because lipid peroxidation begins within the first
aggregation which in turn may precipitate ische- 5 min after an acute cord injury, administration of
mia [132]. Hence the prompt decompression of high-dose steroid should occur as close to the time
the intrinsic spinal hematoma is vital in minimiz- of injury as possible for maximal efficacy. Given
ing the subsequent injury. The precipitating fac- the similarity of pathophysiological concepts in
tors for hematomyelia such as SVMs and spinal secondary mechanism of injury between acute
tumors should be promptly treated to prevent spinal cord injury and cord infarction, there may
recurrent hemorrhage. be a beneficial role for high-dose steroids in cord
For treatment of venous hypertension, com- infarction.
plete obliteration of SVMs should be performed
with surgery or endovascular embolization. Using
permanent liquid embolic material (glue, Onyx), Surgical Decompression
the results of embolization and surgery are similar
in safety and efficacy [260]. The critical factors The timing of decompression of the neural ele-
for good outcome in endovascular treatment ments and, in particular, the efficacy of early
includes the identification of the fistula preopera- decompression (within 24 h) in improving neuro-
tively, avoidance of temporary embolic material logic recovery are still a matter of debate [264,
(PVA particles), and occlusion of draining vein. If 265]. A meta-analysis of studies of early decom-
inadvertent embolization of ASA is a concern, pression from 1966 through 2000 [266] showed
surgery is definitely the best choice [260]. that surgery performed within 24 h produced a
1172 S.R. Boddu et al.

significant improvement in neurologic recovery no need for urinary catheterization at the time of
compared with late surgery but concluded that hospital discharge [134]. In a series of 36 patients,
the evidence was not strong and that early surgery the outcome at the time of discharge showed 57 %
could be considered only as a practice option. confined to wheelchair, 25 % partly mobile with
Starting from this framework, a recent prospective ambulatory aids, and 18 % fully mobile.
multicentric study [267] demonstrated that the Advanced patient age is a negative prognostic
odds of achieving a 2 AIS grade improvement factor for the functional outcome. Absent muscle
are 2.8 times higher in patients undergoing early contraction at the time of admission has increased
surgical decompression (within 24 h). However, a risk of poor outcome and wheelchair bound
recent meta-analysis [268] reported a lack of sta- [147]. In a series of 57 patients with ASCIS,
tistical robustness of the articles examined; there- logistic regression analysis showed that poor
fore, the relationship between early surgery and grades at presentation (ASIA: A and B) and
better neurological outcome is still to be female gender are independent predictors of poor
demonstrated. functional outcome [1].

Prognosis Long-Term Outcome

Spinal cord infarction carries poor prognosis with The long-term functional outcome of ASCIS
permanent and disabling sequelae. The mortality depends on the degree of the initial neurological
rate of SCI based on published case series ranges deficits, especially the motor deficits [1]. Based on
from 9 % to 23 % [1, 10, 147, 166]. SCI with a case series with a mean follow-up of 3 years,
cardiac arrest, aortic rupture or dissection, and improvement in outcome is possible long after
high cervical involvement carries high risk of being discharged from the hospital. About 40 %
mortality [147]. Among the survivors of the of patients will show some kind of improvement
acute episode, respiratory complications are the [9]. As an example although about 40 % of
leading cause of death [269]. The leading reported patients are wheelchair bound upon discharge, at
secondary complications are typically pressure least 40 % of these will be able to walk indepen-
sores, chills, and urinary sepsis, atelectasis, pneu- dently in the future [10]. In patients with venous
monia, and deep vein thrombosis [84]. Prolonged congestion related to SVMs, motor function is
immobilization results in joint contractures and more likely to improve compared to sensory and
heterotopic calcification [269]. When comparing bladder function (60 % vs. 30 %) [271]. Impotence
tetraplegics and paraplegics, an increased inci- rarely improves. The presence of a sensory level is
dence of urinary tract infections and pressure a bad prognostic marker, suggesting irreversible
sores is seen in the former [270]. The social and cord damage [272].
economic impact of SCI extends to involve not
just the patient and the immediate family but the
community and society at large [84]. Future Direction

One should not forget the small footsteps in the


Short-Term Outcome evolution of thrombolysis for cerebral stroke. Cer-
tainly the fascinating journey across intravenous
Initial neurological assessment following Ameri- thrombolysis, intra-arterial thrombolysis,
can Spinal Injury Association (ASIA) classifica- mechanical thrombectomy, revolutionary contri-
tion proves to be the best predictor of prognosis bution from imaging, and incredible teamwork
[1, 147]. Approximately 50 % of patients have was not instantaneous. With high index of suspi-
favorable short-term outcome, defined by ability cion, better coordination, modern imaging, and
to walk without or with one assistive device and interventional techniques, spinal thrombolysis
47 Spinal Cord Infarction and Differential Diagnosis 1173

could become a treatment of choice in appropriate 6. Slager UT, Webb AT (1973) Pathologic findings in the
patients. Early diagnosis is the key for spinal spinal cord. Arch Pathol 96(6):388–394
7. Sandson TA, Friedman JH (1989) Spinal cord infarc-
thrombolysis. Early initiation of resuscitation tion. Report of 8 cases and review of the literature.
measures to maintain the local perfusion and min- Medicine (Baltimore) 68(5):282–292
imize secondary cord damage may widen the cur- 8. Hirtz D, Thurman DJ, Gwinn-Hardy K, Mohamed M,
rent 3-h therapeutic window for intra-arterial Chaudhuri AR, Zalutsky R (2007) How common are
the “common” neurologic disorders? Neurology
thrombolysis. Further research studies in this 68(5):326–337. doi:10.1212/01.wnl.0000252807.
area should be encouraged as a contribution 38124.a3
toward evidence-based medicine. 9. Cheshire WP, Santos CC, Massey EW, Howard JF Jr
(1996) Spinal cord infarction: etiology and
outcome. Neurology 47(2):321–330. doi:10.1212/
WNL.47.2.321
Summary 10. Robertson CE, Brown RD Jr, Wijdicks EFM,
Rabinstein AA (2012) Recovery after spinal cord
Thorough understanding of the arterial anatomy infarcts: long-term outcome in 115 patients.
Neurology 78(2):114–121. doi:10.1212/WNL.
and the rich intrinsic vascular anastomosis includ- 0b013e31823efc93
ing the anterior spinal artery is vital in the under- 11. Abercrombie J (1831) Pathological and practical
standing of the spinal cord infarction. High index researches on diseases of the brain and the spinal
of suspicion, awareness of the spinal cord infarc- cord. Carey & Lea, Philadelphia
12. Tweedie A, Gerhard W (1842) A system of practical
tion clinical syndromes, and early magnetic reso- medicine comprised in a series of original disserta-
nance imaging with diffusion-weighted tions. Lea & Blanchard, Philadelphia
acquisition in conjunction with the clinical assess- 13. Santillan A, Nacarino V, Greenberg E, Riina HA,
ment and cerebrospinal fluid analysis help to dif- Gobin YP, Patsalides A (2012) Vascular anatomy of
the spinal cord. J Neurointerv Surg 4(1):67–74.
ferentiate the spinal cord infarction from its doi:10.1136/neurintsurg-2011-010018
mimics. Catheter angiography helps to differenti- 14. Manelfe C, Lazorthes G, Roulleau J (1972) Arteries
ate between the embolic versus hypotensive of the human spinal dura mater. Acta Radiol Diagn
infarctions. Early diagnosis, maintaining the (Stockh) 13:829–841
15. Thron A (1988) Vascular anatomy of the spinal cord.
local perfusion thus preventing the free radical- Neuroradiological investigations and clinical
mediated secondary damage, and timely throm- syndromes. Springer, New York
bolysis in appropriate patients are the fundamen- 16. Hong MK-Y, Hong MK-H, Pan W-R, Wallace D,
tals of spinal cord infarction treatment. Ashton MW, Taylor GI (2008) The angiosome terri-
tories of the spinal cord: exploring the issue of preop-
erative spinal angiography. Laboratory investigation.
J Neurosurg Spine 8(4):352–364. doi:10.3171/SPI/
References 2008/8/4/352
17. Lasjaunias P, Berenstein A, TerBrugge K (2001) Clin-
1. Nedeltchev K, Loher TJ, Stepper F et al (2004) Long- ical vascular anatomy and variations. In: Surgical
term outcome of acute spinal cord ischemia syn- neuroangiography, vol 1. Springer, Berlin
drome. Stroke 35(2):560–565. doi:10.1161/01. 18. Piscol K (1972) Blood supply of the spinal cord and
STR.0000111598.78198.EC its clinical importance. Schriftenr Neurol 8:1–91
2. de Seze J, Stojkovic T, Breteau G et al (2001) Acute 19. Brockstein B, Johns L, Gewertz BL (1994)
myelopathies clinical, laboratory and outcome pro- Blood supply to the spinal cord: anatomic and
files in 79 cases. Brain 124(8):1509–1521. physiologic correlations. Ann Vasc Surg 8(4):
doi:10.1093/brain/124.8.1509 394–399
3. Martinelli V, Comi G, Rovaris M et al (1995) Acute 20. Lazorthes G, Poulhes J, Bastide G, Roulleau J,
myelopathy of unknown aetiology: a clinical, neuro- Chancholle AR (1958) Arterial vascularization of
physiological and MRI study of short- and long-term the spine; anatomic research and applications in
prognostic factors. J Neurol 242(8):497–503 pathology of the spinal cord and aorta.
4. Woollam DH, Millen JW (1955) The arterial supply Neurochirurgie 4(1):3–19
of the spinal cord and its significance. J Neurol 21. Koshino T, Murakami G, Morishita K, Mawatari T,
Neurosurg Psychiatry 18(2):97–102 Abe T (1999) Does the Adamkiewicz artery originate
5. Singer D (1902) The pathology of so-called acute from the larger segmental arteries? J Thorac
myelitis. Brain 25(2):332–340 Cardiovasc Surg 117(5):898–905
1174 S.R. Boddu et al.

22. Hyodoh H, Shirase R, Akiba H et al (2007) Double- aortic aneurysm repair. J Vasc Surg 40(4):703–709.
subtraction maximum intensity projection MR angi- doi:10.1016/j.jvs.2004.07.032; discussion 709–710
ography for detecting the artery of Adamkiewicz and 40. Messé SR, Bavaria JE, Mullen M et al (2008) Neuro-
differentiating it from the drainage vein. J Magn logic outcomes from high risk descending thoracic
Reson Imaging 26(2):359–365. doi:10.1002/ and thoracoabdominal aortic operations in the era of
jmri.21024 endovascular repair. Neurocrit Care 9(3):344–351.
23. Charles YP, Barbe B, Beaujeux R, Boujan F, Steib J-P doi:10.1007/s12028-008-9104-9
(2011) Relevance of the anatomical location of the 41. Bavaria JE, Appoo JJ, Makaroun MS et al (2007)
Adamkiewicz artery in spine surgery. Surg Radiol Endovascular stent grafting versus open surgical
Anat 33(1):3–9. doi:10.1007/s00276-010-0654-0 repair of descending thoracic aortic aneurysms in
24. Garland H, Greenberg J, Harriman DG (1966) Infarc- low-risk patients: a multicenter comparative trial.
tion of the spinal cord. Brain J Neurol 89(4):645–662 J Thorac Cardiovasc Surg 133(2):369–377.
25. Sliwa JA, Maclean IC (1992) Ischemic myelopathy: a doi:10.1016/j.jtcvs.2006.07.040
review of spinal vasculature and related clinical syn- 42. Setacci F, Sirignano P, De Donato G et al (2010)
dromes. Arch Phys Med Rehabil 73(4):365–372 Endovascular thoracic aortic repair and risk of spinal
26. Gilles FH, Nag D (1971) Vulnerability of human cord ischemia: the role of previous or concomitant
spinal cord in transient cardiac arrest. Neurology treatment for aortic aneurysm. J Cardiovasc Surg
21(8):833–839 (Torino) 51(2):169–176
27. Azzarelli B, Roessmann U (1977) Diffuse “anoxic” 43. Crawford ES, Crawford JL, Safi HJ et al (1986)
myelopathy. Neurology 27(11):1049–1052 Thoracoabdominal aortic aneurysms: preoperative
28. Rothman SM, Nelson JS (1980) Spinal cord infarc- and intraoperative factors determining immediate
tion in a patient with sickle cell anemia. Neurology and long-term results of operations in 605 patients.
30(10):1072–1076 J Vasc Surg 3(3):389–404
29. Houdart R, Djindjian R (1966) Angiomas of the spi- 44. Gloviczki P, Cross SA, Stanson AW et al (1991)
nal cord. Proc R Soc Med 59(8):787–790 Ischemic injury to the spinal cord or lumbosacral
30. Satran R (1988) Spinal cord infarction. Stroke plexus after aorto-iliac reconstruction. Am J Surg
19(4):529–532 162(2):131–136
31. Whiteley AM, Hauw JJ, Escourolle R (1979) A path- 45. Ross RT (1985) Spinal cord infarction in disease and
ological survey of 41 cases of acute intrinsic spinal surgery of the aorta. Can J Neurol Sci 12(4):289–295
cord disease. J Neurol Sci 42(2):229–242 46. Mawad ME, Rivera V, Crawford S, Ramirez A,
32. Shakir RA, Sulaiman K, Kahn RA, Rudwan M (1990) Breitbach W (1990) Spinal cord ischemia after resec-
Neurological presentation of neuro-Behçet’s syn- tion of thoracoabdominal aortic aneurysms: MR find-
drome: clinical categories. Eur Neurol 30(5):249–253 ings in 24 patients. AJNR Am J Neuroradiol
33. Gibb WR, Urry PA, Lees AJ (1985) Giant cell arteritis 11(5):987–991
with spinal cord infarction and basilar artery throm- 47. Brusoni B, Colombo A, Merlo L, Marchetti G, Longo
bosis. J Neurol Neurosurg Psychiatry 48(9):945–948 T (1978) Hemodynamic and metabolic changes
34. Sawaya GR, Kaminski MJ (1990) Spinal cord infarc- induced by temporary clamping of the thoracic
tion after cocaine use. South Med J 83(5):601–602 aorta. Eur Surg Res 10(3):206–216
35. Walden JE, Castillo M (2012) Sildenafil-induced cer- 48. Hurst RW, Haskal ZJ, Zager E, Bagley LJ, Flamm ES
vical spinal cord infarction. AJNR Am J Neuroradiol (1998) Endovascular stent treatment of cervical inter-
33(3):E32–E33. doi:10.3174/ajnr.A2628 nal carotid artery aneurysms with parent vessel pres-
36. Adams HD, van Geertruyden HH (1956) Neurologic ervation. Surg Neurol 50(4):313–317; discussion 317.
complications of aortic surgery. Ann Surg 144(4): Available at http://www.ncbi.nlm.nih.gov/pubmed/
574–609. Available at http://www.ncbi.nlm.nih. 9817452. Accessed 20 Mar 2012
gov/pmc/articles/PMC1465537/. Accessed 20 Mar 49. Hughes JT, Brownell B (1966) Spinal cord ischemia
2014 due to arteriosclerosis. Arch Neurol 15(2):189–202
37. Xenos ES, Abedi NN, Davenport DL et al (2008) 50. Tubbs RS, Blouir MC, Romeo AK, Mortazavi MM,
Meta-analysis of endovascular vs open repair for Cohen-Gadol AA (2011) Spinal cord ischemia and
traumatic descending thoracic aortic rupture. atherosclerosis: a review of the literature. Br J
J Vasc Surg 48(5):1343–1351. doi:10.1016/j.jvs. Neurosurg 25(6):666–670. doi:10.3109/02688697.
2008.04.060 2011.578774
38. Amabile P, Grisoli D, Giorgi R, Bartoli J-M, Piquet P 51. Duggal N, Lach B (2002) Selective vulnerability of
(2008) Incidence and determinants of spinal cord the lumbosacral spinal cord after cardiac arrest and
ischaemia in stent-graft repair of the thoracic aorta. hypotension. Stroke 33(1):116–121. doi:10.1161/
Eur J Vasc Endovasc Surg 35(4):455–461. hs0102.101923
doi:10.1016/j.ejvs.2007.11.005 52. Slavin RE, Gonzalez-Vitale JC, Marin OS (1975)
39. Maldonado TS, Rockman CB, Riles E et al (2004) Atheromatous emboli to the lumbosacral spinal
Ischemic complications after endovascular abdominal cord. Stroke 6(4):411–415
47 Spinal Cord Infarction and Differential Diagnosis 1175

53. Coleman J (1898) Dissecting aneurysm. Trans R Semin Intervent Radiol 29(3):241–244. doi:10.1055/
Acad Med Irel 16(1):116–124 s-0032-1326936
54. Tracci C, Cherry KJ Jr (2012) The aorta. In: 70. Gang DL, Dole KB, Adelman LS (1977) Spinal cord
Townsend CM (ed) Sabiston textbook of surgery. infarction following therapeutic renal artery emboli-
Saunders/Elsevier, Philadelphia, pp 1697–1724 zation. JAMA 237(26):2841–2842
55. Gaul C, Dietrich W, Friedrich I, Sirch J, Erbguth FJ 71. Vujic I, Pyle R, Parker E, Mithoefer J (1980) Control
(2007) Neurological symptoms in type A aortic dis- of massive hemoptysis by embolization of intercostal
sections. Stroke 38(2):292–297. doi:10.1161/01. arteries. Radiology 137(3):617–620. doi:10.1148/
STR.0000254594.33408.b1 radiology.137.3.7444046
56. Suzuki T, Mehta RH, Ince H et al (2003) Clinical 72. Ozoilo K, Stein M (2013) Paraplegia complicating
profiles and outcomes of acute type B aortic dissec- embolization for bleeding intercostal artery in pene-
tion in the current era: lessons from the International trating trauma. Inj Extra 44(8):70–73. doi:10.1016/j.
Registry of Aortic Dissection (IRAD). Circulation injury.2013.05.015
108(Suppl 1):II312–II317. doi:10.1161/01.cir. 73. Heller SL, Meyer JR, Russell EJ (1996) Spinal cord
0000087386.07204.09 venous infarction following endoscopic sclerotherapy
57. Sladky JT, Rorke LB (1986) Perinatal hypoxic/ische- for esophageal varices. Neurology 47(4):1081–1085
mic spinal cord injury. Pediatr Pathol 6(1):87–101 74. Tofuku K, Koga H, Yamamoto T, Yone K, Komiya S
58. Thompson TP, Pearce J, Chang G, Madamba J (2004) (2008) Spinal cord infarction following endoscopic
Surfer’s myelopathy. Spine 29(16):E353–E356 variceal ligation. Spinal Cord 46(3):241–242.
59. Chang CWJ, Donovan DJ, Liem LK et al (2012) doi:10.1038/sj.sc.3102092
Surfers’ myelopathy: a case series of 19 novice 75. Neal JM, Bernards CM, Hadzic A et al (2008) ASRA
surfers with nontraumatic myelopathy. Neurology practice advisory on neurologic complications in
79(22):2171–2176. doi:10.1212/WNL.0b013e31 regional anesthesia and pain mMedicine. Reg Anesth
827595cd Pain Med 33(5):404–415. doi:10.1016/j.rapm.2008.
60. Shamji MF, Maziak DE, Shamji FM, Ginsberg RJ, 07.527
Pon R (2003) Circulation of the spinal cord: an impor- 76. Forbes G, Nichols DA, Jack CR Jr et al (1988) Com-
tant consideration for thoracic surgeons. Ann Thorac plications of spinal cord arteriography: prospective
Surg 76(1):315–321 assessment of risk for diagnostic procedures.
61. Nakamoto BK, Siu AM, Hashiba KA et al (2013) Radiology 169(2):479–484. doi:10.1148/radiology.
Surfer’s myelopathy: a radiologic study of 23 cases. 169.2.3174997
AJNR Am J Neuroradiol 34(12):2393–2398. 77. Gonzalez LF, Zabramski JM, Tabrizi P, Wallace RC,
doi:10.3174/ajnr.A3599 Massand MG, Spetzler RF (2005) Spontaneous spinal
62. Hirose G, Singer P, Bass NH (1971) Successful treat- subarachnoid hemorrhage secondary to spinal aneu-
ment of posthypoxic action myoclonus with carba- rysms: diagnosis and treatment paradigm. Neurosur-
mazepine. JAMA 218(9):1432–1433 gery 57(6):1127–1131; discussion 1127–1131
63. Mori S, Sadoshima S, Tagawa K, Iino K, Fujishima M 78. Krishna V, Lazaridis C, Ellegala D et al (2012) Spinal
(1993) Massive spinal cord infarction with multiple cord infarction associated with subarachnoid hemor-
paradoxical embolism: a case report. Angiology rhage. Clin Neurol Neurosurg 114(7):1030–1032.
44(3):251–256 doi:10.1016/j.clineuro.2012.01.037
64. Naiman JL, Donohue WL, Prichard JS (1961) Fatal 79. Anthes DL, Theriault E, Tator CH (1996) Ultrastruc-
nucleus pulposus embolism of spinal cord after tural evidence for arteriolar vasospasm after spinal
trauma. Neurology 11:83–87 cord trauma. Neurosurgery 39(4):804–814
65. Tosi L, Rigoli G, Beltramello A (1996) Fibrocarti- 80. Krings T, Lasjaunias PL, Hans FJ et al (2007)
laginous embolism of the spinal cord: a clinical and Imaging in spinal vascular disease. Neuroimaging
pathogenetic reconsideration. J Neurol Neurosurg Clin N Am 17(1):57–72. doi:10.1016/j.nic.2007.
Psychiatry 60(1):55–60 01.001
66. Srigley JR, Lambert CD, Bilbao JM, Pritzker KP 81. Blatteau J-E, Gempp E, Simon O et al (2011) Prog-
(1981) Spinal cord infarction secondary to nostic factors of spinal cord decompression sickness
intervertebral disc embolism. Ann Neurol in recreational diving: retrospective and multicentric
9(3):296–301. doi:10.1002/ana.410090315 analysis of 279 cases. Neurocrit Care 15(1):120–127.
67. Mathew P, Todd NV, Hadley DM, Adams JH (1993) doi:10.1007/s12028-010-9370-1
Spinal cord infarction following meningitis. Br J 82. Aharon-Peretz J, Adir Y, Gordon CR, Kol S, Gal N,
Neurosurg 7(6):701–704 Melamed Y (1993) Spinal cord decompression sick-
68. Von Pohle WR (1996) Disseminated mucormycosis ness in sport diving. Arch Neurol 50(7):753–756
presenting with lower extremity weakness. Eur Respir 83. Kim RC, Smith HR, Henbest ML, Choi BH (1984)
J 9(8):1751–1753 Nonhemorrhagic venous infarction of the spinal cord.
69. Brown AC, Ray CE (2012) Anterior spinal cord Ann Neurol 15(4):379–385. doi:10.1002/ana.
infarction following bronchial artery embolization. 410150413
1176 S.R. Boddu et al.

84. Sekhon LH, Fehlings MG (2001) Epidemiology, spinal cord trauma. J Neurosurg 50(2):198–206.
demographics, and pathophysiology of acute spinal doi:10.3171/jns.1979.50.2.0198
cord injury. Spine 26(24 Suppl):S2–S12 99. Wolman L (1965) The disturbance of circulation in
85. Fehlings MG, Sekhon LH (2000) Cellular, ionic and traumatic paraplegia in acute and late stages: a path-
biomolecular mechanisms of the injury process. In: ological study. Paraplegia 2:213–226. doi:10.1038/
Tator CH, Benzel EC (eds) Contemporary manage- sc.1964.39
ment of spinal cord injury: from impact to rehabilita- 100. Sandler AN, Tator CH (1976) Regional spinal cord
tion. American Association of Neurological blood flow in primates. J Neurosurg 45(6):647–659.
Surgeons, New York, pp 33–50 doi:10.3171/jns.1976.45.6.0647
86. Allen A (1911) Surgery of experimental lesion of 101. Blisard KS, Follis F, Wong R, Miller KB, Wernly JA,
spinal cord equivalent to crush injury of fracture dis- Scremin OU (1995) Degeneration of axons in the
location of spinal column. A preliminary report. corticospinal tract secondary to spinal cord ischemia in
JAMA 57:878–880 rats. Paraplegia 33(3):136–140. doi:10.1038/sc.1995.30
87. Allen A (1914) Remarks on the histopathological 102. Suh T, Alexander L (1939) Vascular system of
changes in the spinal cord due to impact an experi- the human spinal cord. Arch Neurol Psychiatry
mental study. J Nerv Ment Dis 31:141–147 1939(41):660–676
88. Demopoulos H, Flamm ES, Seligman M (1979) 103. Grassner L, Klausner F, Wagner M et al (2014) Acute
Membrane perturbations in central nervous system and chronic evolution of MRI findings in a case
injury: theoretical basis for free radical damage and of posterior spinal cord ischemia. Spinal Cord
a review of the experimental data. In: Popp AJ 52:S23–S24. doi:10.1038/sc.2013.165
(ed) Neural trauma. Raven Press, New York, 104. Fried LC, Goodkin R (1971) Microangiographic
pp 63–78 observations of the experimentally traumatized spinal
89. Anderson DK, Hall ED (1993) Pathophysiology of cord. J Neurosurg 35(6):709–714. doi:10.3171/
spinal cord trauma. Ann Emerg Med 22(6):987–992 jns.1971.35.6.0709
90. Collins WF (1983) A review and update of experi- 105. Dohrmann GJ, Wick KM (1973) Intramedullary
ment and clinical studies of spinal cord injury. Para- blood flow patterns in transitory traumatic paraplegia.
plegia 21(4):204–219. doi:10.1038/sc.1983.34 Surg Neurol 1(4):209–215
91. Dusart I, Schwab ME (1994) Secondary cell death 106. Assenmacher DR, Ducker TB (1971) Experimental
and the inflammatory reaction after dorsal traumatic paraplegia. The vascular and pathological
hemisection of the rat spinal cord. Eur J Neurosci changes seen in reversible and irreversible spinal-cord
6(5):712–724 lesions. J Bone Joint Surg Am 53(4):671–680
92. Tator CH (1991) Review of experimental spinal cord 107. Griffiths IR, Miller R (1974) Vascular permeability to
injury with emphasis on the local and systemic circu- protein and vasogenic oedema in experimental con-
latory effects. Neurochirurgie 37(5):291–302 cussive injuries to the canine spinal cord. J Neurol Sci
93. Young W, Koreh I (1986) Potassium and calcium 22(3):291–304
changes in injured spinal cords. Brain Res 108. Hsu CY, Hogan EL, Gadsden RH Sr, Spicer KM, Shi
365(1):42–53 MP, Cox RD (1985) Vascular permeability in exper-
94. Hall ED, Wolf DL (1987) Post-traumatic spinal cord imental spinal cord injury. J Neurol Sci 70(3):
ischemia: relationship to injury severity and physio- 275–282
logical parameters. Cent Nerv Syst Trauma 109. Stewart WB, Wagner FC (1979) Vascular permeabil-
4(1):15–25 ity changes in the contused feline spinal cord. Brain
95. Hall ED, Braughler JM (1982) Effects of intravenous Res 169(1):163–167
methylprednisolone on spinal cord lipid peroxidation 110. Young W, Flamm ES (1982) Effect of high-dose
and Na+ + K+)-ATPase activity. Dose-response anal- corticosteroid therapy on blood flow, evoked poten-
ysis during 1st hour after contusion injury in the cat. tials, and extracellular calcium in experimental spinal
J Neurosurg 57(2):247–253. doi:10.3171/jns.1982. injury. J Neurosurg 57(5):667–673. doi:10.3171/
57.2.0247 jns.1982.57.5.0667
96. Fehlings MG, Tator CH, Linden RD (1989) The effect 111. Young W, DeCrescito V, Tomasula JJ, Ho V (1980)
of nimodipine and dextran on axonal function and The role of the sympathetic nervous system in pressor
blood flow following experimental spinal cord injury. responses induced by spinal injury. J Neurosurg
J Neurosurg 71(3):403–416. doi:10.3171/jns.1989. 52(4):473–481. doi:10.3171/jns.1980.52.4.0473
71.3.0403 112. Atkinson PP, Atkinson JL (1996) Spinal shock. Mayo
97. Tator CH, Fehlings MG (1991) Review of the sec- Clin Proc 71(4):384–389. doi:10.1016/S0025-6196
ondary injury theory of acute spinal cord trauma with (11)64067-6
emphasis on vascular mechanisms. J Neurosurg 113. Henderson CE (1996) Programmed cell death in the
75(1):15–26. doi:10.3171/jns.1991.75.1.0015 developing nervous system. Neuron 17(4):579–585
98. Senter HJ, Venes JL (1979) Loss of autoregulation 114. Namura S, Zhu J, Fink K et al (1998) Activation and
and posttraumatic ischemia following experimental cleavage of caspase-3 in apoptosis induced by
47 Spinal Cord Infarction and Differential Diagnosis 1177

experimental cerebral ischemia. J Neurosci 129. Wagner FC Jr, Stewart WB (1981) Effect of trauma
18(10):3659–3668 dose on spinal cord edema. J Neurosurg
115. Hockenbery D (1995) Defining apoptosis. Am J 54(6):802–806. doi:10.3171/jns.1981.54.6.0802
Pathol 146(1):16–19 130. Anderson DK, Means ED, Waters TR, Spears CJ
116. Crowe MJ, Bresnahan JC, Shuman SL, Masters JN, (1980) Spinal cord energy metabolism following
Beattie MS (1997) Apoptosis and delayed degenera- compression trauma to the feline spinal cord.
tion after spinal cord injury in rats and monkeys. Nat J Neurosurg 53(3):375–380. doi:10.3171/jns.1980.
Med 3(1):73–76 53.3.0375
117. Emery E, Aldana P, Bunge MB et al (1998) Apoptosis 131. Casha S, Yu WR, Fehlings MG (2001) Oligodendrog-
after traumatic human spinal cord injury. J Neurosurg lial apoptosis occurs along degenerating axons and is
89(6):911–920. doi:10.3171/jns.1998.89.6.0911 associated with FAS and p75 expression following
118. Li GL, Brodin G, Farooque M et al (1996) Apoptosis spinal cord injury in the rat. Neuroscience 103
and expression of Bcl-2 after compression trauma to rat (1):203–218
spinal cord. J Neuropathol Exp Neurol 55(3):280–289 132. De la Torre JC (1981) Spinal cord injury. Review of
119. Tator CH, Koyanagi I (1997) Vascular mechanisms in basic and applied research. Spine 6(4):315–335
the pathophysiology of human spinal cord injury. 133. Lou J, Lenke LG, Ludwig FJ, O’Brien MF (1998)
J Neurosurg 86(3):483–492. doi:10.3171/jns.1997. Apoptosis as a mechanism of neuronal cell death
86.3.0483 following acute experimental spinal cord injury.
120. Agrawal SK, Fehlings MG (1996) Mechanisms of Spinal Cord 36(10):683–690
secondary injury to spinal cord axons in vitro: role 134. Masson C, Pruvo JP, Meder JF et al (2004) Spinal
of Na+, Na(+)-K(+)-ATPase, the Na(+)-H+ exchanger, cord infarction: clinical and magnetic resonance
and the Na(+)-Ca2+ exchanger. J Neurosci 16(2): imaging findings and short term outcome. J Neurol
545–552 Neurosurg Psychiatry 75(10):1431–1435. doi:10.1136/
121. Osterholm JL, Mathews GJ (1972) Altered norepi- jnnp.2003.031724
nephrine metabolism, following experimental spinal 135. Geldmacher D (2012) Vascular diseases of the ner-
cord injury. 2. Protection against traumatic spinal cord vous system: spinal cord vascular disease. In: Daroff
hemorrhagic necrosis by norepinephrine synthesis RB, Bradley WG (eds) Bradley’s neurology in clini-
blockade with alpha methyl tyrosine. J Neurosurg cal practice, 6th edn. Saunders/Elsevier, Philadelphia,
36(4):395–401. doi:10.3171/jns.1972.36.4.0395 pp 1096–1099
122. Agrawal SK, Fehlings MG (1997) Role of NMDA 136. Cheng M-Y, Lyu R-K, Chang Y-J et al (2008)
and non-NMDA ionotropic glutamate receptors in Spinal cord infarction in Chinese patients. Clinical
traumatic spinal cord axonal injury. J Neurosci features, risk factors, imaging and prognosis.
17(3):1055–1063 Cerebrovasc Dis 26(5):502–508. doi:10.1159/
123. Faden AI, Simon RP (1988) A potential role for 000155988
excitotoxins in the pathophysiology of spinal cord 137. Maynard FM Jr, Bracken MB, Creasey G et al (1997)
injury. Ann Neurol 23(6):623–626. doi:10.1002/ International standards for neurological and func-
ana.410230618 tional classification of spinal cord injury. American
124. Demopoulos HB, Flamm ES, Pietronigro DD, Spinal Injury Association. Spinal Cord 35(5):
Seligman ML (1980) The free radical pathology and 266–274
the microcirculation in the major central nervous sys- 138. Misulis K (2012) Hemiplegia and monoplegia. In:
tem disorders. Acta Physiol Scand Suppl 492:91–119 Daroff RB, Mazziotta JC (eds) Bradley’s neurology
125. Hall ED, Yonkers PA, Horan KL, Braughler JM in clinical practice, 6th edn. Saunders/Elsevier,
(1989) Correlation between attenuation of Philadelphia, p 277
posttraumatic spinal cord ischemia and preservation 139. Dobkin B, Havton L (2012) Paraplegia and spinal
of tissue vitamin E by the 21-aminosteroid U74006F: cord syndromes. In: Bradley’s neurology in clinical
evidence for an in vivo antioxidant mechanism. practice, 6th edn. Saunders/Elsevier, Philadelphia, pp
J Neurotrauma 6(3):169–176 286–289
126. Hung TK, Albin MS, Brown TD, Bunegin L, Albin R, 140. Vertinsky AT, Krasnokutsky MV, Augustin M,
Jannetta PJ (1975) Biomechanical responses to open Bammer R (2007) Cutting-edge imaging of the
experimental spinal cord injury. Surg Neurol 4(2): spine. Neuroimaging Clin N Am 17(1):117–136.
271–276 doi:10.1016/j.nic.2007.01.003
127. Faden AI, Jacobs TP, Holaday JW (1982) Compari- 141. Roemer PB, Edelstein WA, Hayes CE, Souza SP,
son of early and late naloxone treatment in experi- Mueller OM (1990) The NMR phased array. Magn
mental spinal injury. Neurology 32(6):677–681 Reson Med 16(2):192–225
128. Faden AI, Jacobs TP, Smith MT (1984) Evaluation of 142. Griswold MA, Jakob PM, Heidemann RM
the calcium channel antagonist nimodipine in exper- et al (2002) Generalized autocalibrating partially
imental spinal cord ischemia. J Neurosurg 60(4): parallel acquisitions (GRAPPA). Magn Reson Med
796–799. doi:10.3171/jns.1984.60.4.0796 47(6):1202–1210. doi:10.1002/mrm.10171
1178 S.R. Boddu et al.

143. Pruessmann KP, Weiger M, Scheidegger MB, 157. Weidauer S, Nichtweiss M, Lanfermann H, Zanella
Boesiger P (1999) SENSE: sensitivity encoding for FE (2002) Spinal cord infarction: MR imaging and
fast MRI. Magn Reson Med 42(5):952–962 clinical features in 16 cases. Neuroradiology
144. Thurnher MM, Bammer R (2006) Diffusion-weighted 44(10):851–857. doi:10.1007/s00234-002-0828-5
MR imaging (DWI) in spinal cord ischemia. Neuro- 158. Beslow LA, Ichord RN, Zimmerman RA, Smith SE,
radiology 48(11):795–801. doi:10.1007/s00234-006- Licht DJ (2008) Role of diffusion MRI in diagnosis of
0130-z spinal cord infarction in children. Neuropediatrics
145. Marcel C, Kremer S, Jeantroux J, Blanc F, Dietemann 39(3):188–191. doi:10.1055/s-0028-1093335
J-L, De Sèze J (2010) Diffusion-weighted imaging in 159. Mull M, Thron A (2006) Spinal infarcts. In: Magnetic
noncompressive myelopathies: a 33-patient prospec- resonance imaging in ischemic stroke. Springer,
tive study. J Neurol 257(9):1438–1445. doi:10.1007/ Berlin, pp 251–267
s00415-010-5538-z 160. Faig J, Busse O, Salbeck R (1998) Vertebral body
146. Novy J, Carruzzo A, Maeder P, Bogousslavsky J infarction as a confirmatory sign of spinal cord ische-
(2006) Spinal cord ischemia: clinical and imaging mic stroke: report of three cases and review of the
patterns, pathogenesis, and outcomes in 27 patients. literature. Stroke 29(1):239–243
Arch Neurol 63(8):1113–1120. doi:10.1001/ 161. Mikulis DJ, Ogilvy CS, McKee A, Davis KR, Ojeman
archneur.63.8.1113 RG (1992) Spinal cord infarction and fibrocarti-
147. Salvador de la Barrera S, Barca-Buyo A, Montoto- lagenous emboli. AJNR Am J Neuroradiol 13(1):
Marqués A, Ferreiro-Velasco ME, Cidoncha-Dans M, 155–160
Rodriguez-Sotillo A (2001) Spinal cord infarction: 162. Yuh WT, Marsh EE, Wang AK et al (1992) MR
prognosis and recovery in a series of 36 patients. imaging of spinal cord and vertebral body infarction.
Spinal Cord 39(10):520–525. doi:10.1038/sj. AJNR Am J Neuroradiol 13(1):145–154. Available
sc.3101201 at http://www.ajnr.org/content/13/1/145. Accessed
148. Hajnal JV, Doran M, Hall AS et al (1991) MR imag- 18 Mar 2014
ing of anisotropically restricted diffusion of water in 163. Haddad MC, Aabed al-Thagafi MY, Djurberg H
the nervous system: technical, anatomic, and patho- (1996) MRI of spinal cord and vertebral body infarc-
logic considerations. J Comput Assist Tomogr tion in the anterior spinal artery syndrome. Neurora-
15(1):1–18 diology 38(2):161–162
149. Stepper F, Lövblad KO (2001) Anterior spinal artery 164. Cheng M-Y, Lyu R-K, Chang Y-J et al (2009) Con-
stroke demonstrated by echo-planar DWI. Eur Radiol comitant spinal cord and vertebral body infarction is
11(12):2607–2610. doi:10.1007/s003300100926 highly associated with aortic pathology: a clinical and
150. Schlaug G, Siewert B, Benfield A, Edelman RR, magnetic resonance imaging study. J Neurol
Warach S (1997) Time course of the apparent diffu- 256(9):1418–1426. doi:10.1007/s00415-009-5126-2
sion coefficient (ADC) abnormality in human stroke. 165. Hurst WR (2009) Vascular disorders of the spine and
Neurology 49(1):113–119 spinal cord. In: Atlas SW (ed) Magnetic resonance
151. Nogueira RG, Ferreira R, Grant PE et al (2012) imaging of the brain and spine. Wolters Kluwer
Restricted diffusion in spinal cord infarction demon- Health/Lippincott Williams & Wilkins, Philadelphia
strated by magnetic resonance line scan diffusion 166. Masson C, Leys D, Meder JF, Dousset V, Pruvo JP
imaging. Stroke 43(2):532–535. doi:10.1161/ (2004) Spinal cord ischemia. J Neuroradiol 31(1):
STROKEAHA.111.624023 35–46
152. K€uker W, Weller M, Klose U, Krapf H, Dichgans J, 167. Chang Y, Jung T-D, Yoo DS, Hyun JK (2010) Diffu-
Nägele T (2004) Diffusion-weighted MRI of spinal sion tensor imaging and fiber tractography of patients
cord infarction–high resolution imaging and time with cervical spinal cord injury. J Neurotrauma
course of diffusion abnormality. J Neurol 27(11):2033–2040. doi:10.1089/neu.2009.1265
251(7):818–824. doi:10.1007/s00415-004-0434-z 168. Rajasekaran S, Kanna RM, Shetty AP, Ilayaraja V
153. Bammer R, Fazekas F, Augustin M et al (2000) (2012) Efficacy of diffusion tensor anisotropy indices
Diffusion-weighted MR imaging of the spinal cord. and tractography in assessing the extent of severity of
AJNR Am J Neuroradiol 21(3):587–591 spinal cord injury: an in vitro analytical study in calf
154. Gass A, Back T, Behrens S, Maras A (2000) MRI of spinal cords. Spine J 12(12):1147–1153. doi:10.1016/
spinal cord infarction. Neurology 54(11):2195 j.spinee.2012.10.032
155. Zhang J, Huan Y, Qian Y, Sun L, Ge Y (2005) 169. Luo C-B, Chang F-C, Teng MM-H, Chen S-S, Lirng
Multishot diffusion-weighted imaging features in spi- J-F, Chang C-Y (2003) Magnetic resonance
nal cord infarction. J Spinal Disord Tech 18(3): imaging as a guide in the diagnosis and follow-up
277–282 of spinal cord infarction. J Chin Med Assoc 66(2):
156. Fortuna A, Ferrante L, Acqui M, Trillò G (1995) 89–95
Spinal cord ischemia diagnosed by MRI. Case report 170. Ikuta F, Zimmerman HM (1976) Distribution of
and review of the literature. J Neuroradiol plaques in seventy autopsy cases of multiple sclerosis
22(2):115–122 in the United States. Neurology 26(6 PT 2):26–28
47 Spinal Cord Infarction and Differential Diagnosis 1179

171. Lycklama G, Thompson A, Filippi M et al (2003) diagnostic criteria. Neurology 65(12):1950–1953.


Spinal-cord MRI in multiple sclerosis. Lancet Neurol doi:10.1212/01.wnl.0000188896.48308.26
2(9):555–562 186. Transverse Myelitis Consortium Working Group
172. Thurnher MM, Cartes-Zumelzu F, Mueller-Mang C (2002) Proposed diagnostic criteria and nosology of
(2007) Demyelinating and infectious diseases of the acute transverse myelitis. Neurology 59(4):499–505
spinal cord. Neuroimaging Clin N Am 17(1):37–55. 187. Scotti G, Gerevini S (2001) Diagnosis and differential
doi:10.1016/j.nic.2006.12.002 diagnosis of acute transverse myelopathy. The role of
173. Bachmann S, Kesselring J (1998) Multiple sclerosis neuroradiological investigations and review of the
and infectious childhood diseases. Neuroepi- literature. Neurol Sci 22(Suppl 2):S69–S73
demiology 17(3):154–160 188. Nance JR, Golomb MR (2007) Ischemic spinal cord
174. Bot JCJ, Blezer ELA, Kamphorst W et al (2004) The infarction in children without vertebral fracture.
spinal cord in multiple sclerosis: relationship of high- Pediatr Neurol 36(4):209–216. doi:10.1016/j.
spatial-resolution quantitative MR imaging findings pediatrneurol.2007.01.006
to histopathologic results. Radiology 233(2): 189. Alper G (2012) Acute disseminated encephalomyeli-
531–540. doi:10.1148/radiol.2332031572 tis. J Child Neurol 27(11):1408–1425. doi:10.1177/
175. Evangelou N, DeLuca GC, Owens T, Esiri MM 0883073812455104
(2005) Pathological study of spinal cord atrophy in 190. Singh S, Prabhakar S, Korah IP, Warade SS, Alexan-
multiple sclerosis suggests limited role of local der M (2000) Acute disseminated encephalomyelitis
lesions. Brain 128(Pt 1):29–34. doi:10.1093/brain/ and multiple sclerosis: magnetic resonance imaging
awh323 differentiation. Australas Radiol 44(4):404–411
176. Tench CR, Morgan PS, Jaspan T, Auer DP, 191. Khong P-L, Ho H-K, Cheng P-W, Wong VCN,
Constantinescu CS (2005) Spinal cord imaging in mul- Goh W, Chan F-L (2002) Childhood acute dissemi-
tiple sclerosis. J Neuroimaging 15(4 Suppl):94S–102S. nated encephalomyelitis: the role of brain and spinal
doi:10.1177/1051228405283292 cord MRI. Pediatr Radiol 32(1):59–66. doi:10.1007/
177. Bot JCJ, Barkhof F, Lycklama à Nijeholt G s00247-001-0582-6
et al (2002) Differentiation of multiple sclerosis 192. Callen DJA, Shroff MM, Branson HM et al (2009)
from other inflammatory disorders and cerebrovascu- Role of MRI in the differentiation of ADEM from MS
lar disease: value of spinal MR imaging. Radiology in children. Neurology 72(11):968–973. doi:10.1212/
223(1):46–56. doi:10.1148/radiol.2231010707 01.wnl.0000338630.20412.45
178. Paty DW, Oger JJ, Kastrukoff LF et al (1988) MRI in 193. Baum PA, Barkovich AJ, Koch TK, Berg BO (1994)
the diagnosis of MS: a prospective study with com- Deep gray matter involvement in children with acute
parison of clinical evaluation, evoked potentials, disseminated encephalomyelitis. AJNR Am J
oligoclonal banding, and CT. Neurology 38(2): Neuroradiol 15(7):1275–1283
180–185 194. Pittock SJ, Lucchinetti CF (2006) Inflammatory trans-
179. Fazekas F, Offenbacher H, Fuchs S et al (1988) verse myelitis: evolving concepts. Curr Opin Neurol
Criteria for an increased specificity of MRI interpre- 19(4):362–368. doi:10.1097/01.wco.0000236615.
tation in elderly subjects with suspected multiple 59215.d3
sclerosis. Neurology 38(12):1822–1825 195. Apak RA, Köse G, Anlar B, Turanli G, Topaloğlu H,
180. Barkhof F, Filippi M, Miller DH et al (1997) Com- Ozdirim E (1999) Acute disseminated encephalomy-
parison of MRI criteria at first presentation to predict elitis in childhood: report of 10 cases. J Child Neurol
conversion to clinically definite multiple sclerosis. 14(3):198–201
Brain 120(Pt 11):2059–2069 196. Thron A, Caplan L (2003) Vascular malformations
181. Kerr DA, Ayetey H (2002) Immunopathogenesis of and interventional neuroradiology of the spinal cord.
acute transverse myelitis. Curr Opin Neurol 15(3): In: Neurological disorderscourse and treatment.
339–347 Academic, Amsterdam, pp 517–528
182. Choi KH, Lee KS, Chung SO et al (1996) Idiopathic 197. Koenig E, Thron A, Schrader V, Dichgans J (1989)
transverse myelitis: MR characteristics. AJNR Am J Spinal arteriovenous malformations and fistulae: clin-
Neuroradiol 17(6):1151–1160 ical, neuroradiological and neurophysiological find-
183. Larner AJ, Farmer SF (2000) Myelopathy following ings. J Neurol 236(5):260–266
influenza vaccination in inflammatory CNS disorder 198. Hurst RW, Kenyon LC, Lavi E, Raps EC, Marcotte P
treated with chronic immunosuppression. Eur J (1995) Spinal dural arteriovenous fistula: the pathol-
Neurol 7(6):731–733 ogy of venous hypertensive myelopathy. Neurology
184. Bakshi R, Mazziotta JC (1996) Acute transverse 45(7):1309–1313
myelitis after influenza vaccination: magnetic reso- 199. Criscuolo GR, Oldfield EH, Doppman JL (1989)
nance imaging findings. J Neuroimaging 6(4): Reversible acute and subacute myelopathy in patients
248–250 with dural arteriovenous fistulas. Foix-Alajouanine
185. De Seze J, Lanctin C, Lebrun C et al (2005) Idiopathic syndrome reconsidered. J Neurosurg 70(3):354–359.
acute transverse myelitis: application of the recent doi:10.3171/jns.1989.70.3.0354
1180 S.R. Boddu et al.

200. De Marco JK, Dillon WP, Halback VV, Tsuruda JS Neurochir (Wien) 146(12):1347–1354. doi:10.1007/
(1990) Dural arteriovenous fistulas: evaluation with s00701-004-0386-1; discussion 1354
MR imaging. Radiology 175(1):193–199. 215. Watanabe M, Nomura T, Toh E, Sato M, Mochida J
doi:10.1148/radiology.175.1.2315480 (2006) Intramedullary spinal cord metastasis: a clini-
201. Hurst RW, Grossman RI (2000) Peripheral spinal cord cal and imaging study of seven patients. J Spinal
hypointensity on T2-weighted MR images: a reliable Disord Tech 19(1):43–47. doi:10.1097/01.bsd.
imaging sign of venous hypertensive myelopathy. 0000188661.08342.2a
AJNR Am J Neuroradiol 21(4):781–786 216. Mut M, Schiff D, Shaffrey ME (2005) Metastasis to
202. Gomori JM, Grossman RI, Yu-Ip C, Asakura T (1987) nervous system: spinal epidural and intramedullary
NMR relaxation times of blood: dependence on field metastases. J Neurooncol 75(1):43–56. doi:10.1007/
strength, oxidation state, and cell integrity. J Comput s11060-004-8097-2
Assist Tomogr 11(4):684–690 217. Haq A, Wasay M (2006) Magnetic resonance imaging
203. Krings T, Geibprasert S (2009) Spinal dural arterio- in poliomyelitis. Arch Neurol 63(5):778.
venous fistulas. AJNR Am J Neuroradiol 30 doi:10.1001/archneur.63.5.778
(4):639–648. doi:10.3174/ajnr.A1485 218. Ferraz-Filho JRL, dos Santos TU, de Oliveira EP,
204. Weinzierl MR, Krings T, Korinth MC, Reinges MHT, Souza AS (2010) MRI findings in an infant with
Gilsbach JM (2004) MRI and intraoperative findings in vaccine-associated paralytic poliomyelitis. Pediatr
cavernous haemangiomas of the spinal cord. Neurora- Radiol 40(Suppl 1):S138–S140. doi:10.1007/
diology 46(1):65–71. doi:10.1007/s00234-003-1072-3 s00247-010-1650-6
205. Balériaux DL (1999) Spinal cord tumors. Eur Radiol 219. Hui AC, Wong KS, Fu M, Kay R (2000) Ischaemic
9(7):1252–1258 myelopathy presenting as Guillain-Barré syndrome.
206. Lowe GM (2000) Magnetic resonance imaging of Int J Clin Pract 54(5):340–341
intramedullary spinal cord tumors. J Neurooncol 220. Hemmer B, Glocker FX, Schumacher M, Deuschl G,
47(3):195–210 L€ucking CH (1998) Subacute combined degenera-
207. Chu BC, Terae S, Hida K, Furukawa M, Abe S, tion: clinical, electrophysiological, and magnetic res-
Miyasaka K (2001) MR findings in spinal hemangio- onance imaging findings. J Neurol Neurosurg
blastoma: correlation with symptoms and with angio- Psychiatry 65(6):822–827
graphic and surgical findings. AJNR Am J 221. Thurnher MM, Post MJ, Jinkins JR (2000) MRI of
Neuroradiol 22(1):206–217 infections and neoplasms of the spine and spinal cord
208. Wippold FJ 2nd, Smirniotopoulos JG, Moran CJ, in 55 patients with AIDS. Neuroradiology
Suojanen JN, Vollmer DG (1995) MR imaging of 42(8):551–563
myxopapillary ependymoma: findings and value to 222. McArthur JC, Brew BJ, Nath A (2005)
determine extent of tumor and its relation to Neurological complications of HIV infection. Lancet
intraspinal structures. AJR Am J Roentgenol Neurol 4(9):543–555. doi:10.1016/S1474-4422(05)
165(5):1263–1267. doi:10.2214/ajr.165.5.7572515 70165-4
209. Miyazawa N, Hida K, Iwasaki Y, Koyanagi I, Abe H 223. Fitzgerald RH Jr, Marks RD Jr, Wallace KM (1982)
(2000) MRI at 1.5 T of intramedullary ependymoma Chronic radiation myelitis. Radiology 144(3):
and classification of pattern of contrast enhancement. 609–612. doi:10.1148/radiology.144.3.6808557
Neuroradiology 42(11):828–832 224. SreeHarsha CK, Rajasekaran S, Dhanasekararaja P
210. Richard S, Campello C, Taillandier L, Parker F, (2006) Spontaneous complete recovery of paraplegia
Resche F (1998) Haemangioblastoma of the central caused by epidural hematoma complicating epidural
nervous system in von Hippel-Lindau disease. French anesthesia: a case report and review of literature.
VHL Study Group. J Intern Med 243(6):547–553 Spinal Cord 44(8):514–517. doi:10.1038/sj.sc.
211. Bou-Haidar P, Peduto AJ, Karunaratne N (2009) Dif- 3101869
ferential diagnosis of T2 hyperintense spinal cord 225. Gobin YP (1997) Classification and endovascular
lesions: part B. J Med Imaging Radiat Oncol 53 treatment of spinal cord arteriovenous malformations
(2):152–159. doi:10.1111/j.1754-9485.2009.02067.x and fistulas. J Stroke Cerebrovasc Dis 6(4):282–286
212. Chason JL, Walker FB, Landers JW (1963) Metastatic 226. Yamada N, Okita Y, Minatoya K et al (2000) Preop-
carcinoma in the central nervous system and dorsal erative demonstration of the Adamkiewicz artery by
root ganglia. A prospective autopsy study. Cancer magnetic resonance angiography in patients with
16:781–787 descending or thoracoabdominal aortic aneurysms.
213. Costigan DA, Winkelman MD (1985) Intramedullary Eur J Cardiothorac Surg 18(1):104–111.
spinal cord metastasis. A clinicopathological study of doi:10.1016/S1010-7940(00)00412-7
13 cases. J Neurosurg 62(2):227–233. doi:10.3171/ 227. Shi HB, Suh DC, Lee HK et al (1999) Preoperative
jns.1985.62.2.0227 transarterial embolization of spinal tumor: emboliza-
214. Kalayci M, Cağavi F, G€ ul S, Yenid€unya S, Açikgöz B tion techniques and results. AJNR Am J Neuroradiol
(2004) Intramedullary spinal cord metastases: diag- 20(10):2009–2015. Available at http://www.ajnr.org/
nosis and treatment – an illustrated review. Acta content/20/10/2009. Accessed 10 Mar 2014
47 Spinal Cord Infarction and Differential Diagnosis 1181

228. Backes WH, Nijenhuis RJ (2008) Advances in spinal 241. Pattany PM, Saraf-Lavi E, Bowen BC (2003) MR
cord MR angiography. AJNR Am J Neuroradiol angiography of the spine and spinal cord. Top Magn
29(4):619–631. doi:10.3174/ajnr.A0910 Reson Imaging 14(6):444–460
229. Fereshetian A, Kadir S, Kaufman SL et al (1989) 242. Bowen BC, DePrima S, Pattany PM, Marcillo A,
Digital subtraction spinal cord angiography in Madsen P, Quencer RM (1996) MR angiography of
patients undergoing thoracic aneurysm surgery. normal intradural vessels of the thoracolumbar spine.
Cardiovasc Intervent Radiol 12(1):7–9 AJNR Am J Neuroradiol 17(3):483–494
230. Kieffer E, Richard T, Chiras J, Godet G, Cormier E 243. Nijenhuis RJ, Jacobs MJ, Jaspers K et al (2007) Com-
(1989) Preoperative spinal cord arteriography in parison of magnetic resonance with computed tomog-
aneurysmal disease of the descending thoracic and raphy angiography for preoperative localization of the
thoracoabdominal aorta: preliminary results in Adamkiewicz artery in thoracoabdominal aortic aneu-
45 patients. Ann Vasc Surg 3(1):34–46 rysm patients. J Vasc Surg 45(4):677–685.
231. Kieffer E, Fukui S, Chiras J, Koskas F, Bahnini A, doi:10.1016/j.jvs.2006.11.046
Cormier E (2002) Spinal cord arteriography: a safe 244. Jaspers K, Nijenhuis RJ, Backes WH (2007) Differ-
adjunct before descending thoracic or entiation of spinal cord arteries and veins by time-
thoracoabdominal aortic aneurysmectomy. J Vasc resolved MR angiography. J Magn Reson Imaging
Surg 35(2):262–268 26(1):31–40. doi:10.1002/jmri.20940
232. Williams GM, Roseborough GS, Webb TH, Perler 245. Ali S, Cashen TA, Carroll TJ et al (2007) Time-
BA, Krosnick T (2004) Preoperative selective inter- resolved spinal MR angiography: initial clinical expe-
costal angiography in patients undergoing rience in the evaluation of spinal arteriovenous
thoracoabdominal aneurysm repair. J Vasc Surg shunts. AJNR Am J Neuroradiol 28(9):1806–1810.
39(2):314–321. doi:10.1016/j.jvs.2003.09.039 doi:10.3174/ajnr.A0639
233. Prestigiacomo CJ, Niimi Y, Setton A, Berenstein A 246. Saindane AM, Boddu SR, Tong FC, Dehkharghani S,
(2003) Three-dimensional rotational spinal angiogra- Dion JE (2014) Contrast-enhanced time-resolved
phy in the evaluation and treatment of vascular MRA for pre-angiographic evaluation of suspected
malformations. AJNR Am J Neuroradiol 24(7): spinal dural arterial venous fistulas. J Neurointerv
1429–1435. Available at http://www.ajnr.org/con Surg 7:135–140. doi:10.1136/neurintsurg-2013-
tent/24/7/1429. Accessed 8 Mar 2014 010981
234. Ropper AE, Lin N, Gross BA et al (2012) Rotational 247. Kaufmann TJ, Kallmes DF (2008) Diagnostic cere-
angiography for diagnosis and surgical planning in bral angiography: archaic and complication-prone
the management of spinal vascular lesions. Neurosurg or here to stay for another 80 years? AJR Am J
Focus 32(5):E6. doi:10.3171/2012.1.FOCUS11254 Roentgenol 190(6):1435–1437. doi:10.2214/AJR.
235. Matsumoto M, Kodama N, Endo Y et al (2007) 07.3522
Dynamic 3D-CT angiography. AJNR Am J 248. Mull M, Nijenhuis RJ, Backes WH, Krings T,
Neuroradiol 28(2):299–304 Wilmink JT, Thron A (2007) Value and limitations
236. Takase K, Sawamura Y, Igarashi K et al (2002) Dem- of contrast-enhanced MR angiography in spinal arte-
onstration of the artery of Adamkiewicz at multi- riovenous malformations and dural arteriovenous fis-
detector row helical CT. Radiology 223(1):39–45. tulas. AJNR Am J Neuroradiol 28(7):1249–1258.
doi:10.1148/radiol.2231010513 doi:10.3174/ajnr.A0612
237. Lai P-H, Pan H-B, Yang C-F et al (2005) Multi- 249. Luetmer PH, Lane JI, Gilbertson JR, Bernstein MA,
detector row computed tomography angiography in Huston J 3rd, Atkinson JLD (2005) Preangiographic
diagnosing spinal dural arteriovenous fistula: initial evaluation of spinal dural arteriovenous fistulas with
experience. Stroke 36(7):1562–1564. doi:10.1161/ elliptic centric contrast-enhanced MR angiography and
01.STR.0000170048.94718.07 effect on radiation dose and volume of iodinated con-
238. Lai PH, Weng MJ, Lee KW, Pan HB (2006) trast material. AJNR Am J Neuroradiol 26(4):711–718
Multidetector CT angiography in diagnosing type I 250. Eddleman CS, Jeong H, Cashen TA et al (2009)
and type IVA spinal vascular malformations. AJNR Advanced noninvasive imaging of spinal vascular
Am J Neuroradiol 27(4):813–817 malformations. Neurosurg Focus 26(1):E9.
239. Si-jia G, Meng-wei Z, Xi-ping L et al (2009) The doi:10.3171/FOC.2009.26.1.E9
clinical application studies of CT spinal angiography 251. Di Chiro G, Wener L (1973) Angiography of the
with 64-detector row spiral CT in diagnosing spinal spinal cord. A review of contemporary techniques
vascular malformations. Eur J Radiol 71(1):22–28. and applications. J Neurosurg 39(1):1–29.
doi:10.1016/j.ejrad.2008.04.005 doi:10.3171/jns.1973.39.1.0001
240. Farb RI, Kim JK, Willinsky RA et al (2002) Spinal 252. Kaufmann TJ, Huston J 3rd, Mandrekar JN, Schleck
dural arteriovenous fistula localization with a tech- CD, Thielen KR, Kallmes DF (2007) Complications
nique of first-pass gadolinium-enhanced MR angiog- of diagnostic cerebral angiography: evaluation of
raphy: initial experience. Radiology 222(3):843–850. 19,826 consecutive patients. Radiology 243(3):
doi:10.1148/radiol.2223010826 812–819. doi:10.1148/radiol.2433060536
1182 S.R. Boddu et al.

253. Chen J, Gailloud P (2011) Safety of spinal angiogra- Injury 36(Suppl 2):B13–B26. doi:10.1016/j.
phy: complication rate analysis in 302 diagnostic injury.2005.06.011
angiograms. Neurology 77(13):1235–1240. 266. La Rosa G, Conti A, Cardali S, Cacciola F, Tomasello
doi:10.1212/WNL.0b013e3182302068 F (2004) Does early decompression improve neuro-
254. Restrepo L, Wityk RJ, Grega MA et al (2002) Diffu- logical outcome of spinal cord injured patients?
sion- and perfusion-weighted magnetic resonance Appraisal of the literature using a meta-analytical
imaging of the brain before and after coronary artery approach. Spinal Cord 42(9):503–512. doi:10.1038/
bypass grafting surgery. Stroke 33(12):2909–2915. sj.sc.3101627
Available at http://www.ncbi.nlm.nih.gov/pubmed/ 267. Fehlings MG, Vaccaro A, Wilson JR et al (2012)
12468790. Accessed 26 Apr 2012 Early versus delayed decompression for traumatic
255. Baba H, Tomita K, Kawagishi T, Imura S (1993) cervical spinal cord injury: results of the Surgical
Anterior spinal artery syndrome. Int Orthop Timing in Acute Spinal Cord Injury Study
17(6):353–356 (STASCIS). PLoS One 7(2):e32037. doi:10.1371/
256. Jauch EC, Saver JL, Adams HP Jr et al (2013) Guide- journal.pone.0032037
lines for the early management of patients with acute 268. Van Middendorp JJ, Hosman AJF, Doi SAR (2013)
ischemic stroke: a guideline for healthcare profes- The effects of the timing of spinal surgery after trau-
sionals from the American Heart Association/Ameri- matic spinal cord injury: a systematic review and
can Stroke Association. Stroke 44(3):870–947. meta-analysis. J Neurotrauma 30(21):1781–1794.
doi:10.1161/STR.0b013e318284056a doi:10.1089/neu.2013.2932
257. McGarvey ML, Cheung AT, Szeto W, Messe SR 269. DeVivo MJ, Kartus PL, Stover SL, Rutt RD, Fine PR
(2007) Management of neurologic complications of (1989) Cause of death for patients with spinal cord
thoracic aortic surgery. J Clin Neurophysiol 24 injuries. Arch Intern Med 149(8):1761–1766
(4):336–343. doi:10.1097/WNP.0b013e31811ec0b0 270. Young JS (1979) Spinal cord injury: associated gen-
258. Cheung AT, Weiss SJ, McGarvey ML et al (2002) eral trauma and medical complications. Adv Neurol
Interventions for reversing delayed-onset postopera- 22:255–260
tive paraplegia after thoracic aortic reconstruction. 271. Patsalides A, Knopman J, Santillan A, Tsiouris AJ,
Ann Thorac Surg 74(2):413–419; discussion 420-421 Riina H, Gobin YP (2011) Endovascular treatment of
259. Wallace MC, Tator CH, Frazee P (1986) Relationship spinal arteriovenous lesions: beyond the dural fistula.
between posttraumatic ischemia and hemorrhage in AJNR Am J Neuroradiol 32(5):798–808.
the injured rat spinal cord as shown by colloidal doi:10.3174/ajnr.A2190
carbon angiography. Neurosurgery 18(4):433–439 272. Da Costa L, Dehdashti AR, terBrugge KG (2009)
260. Kirsch M, Berg-Dammer E, Musahl C, Bäzner H, Spinal cord vascular shunts: spinal cord vascular
K€uhne D, Henkes H (2013) Endovascular manage- malformations and dural arteriovenous fistulas.
ment of spinal dural arteriovenous fistulas in Neurosurg Focus 26(1):E6. doi:10.3171/
78 patients. Neuroradiology 55(3):337–343. FOC.2009.26.1.E6
doi:10.1007/s00234-013-1134-0
261. Anderson DK, Means ED, Waters TR, Green ES
(1982) Microvascular perfusion and metabolism in
injured spinal cord after methylprednisolone treat- Suggested Reading
ment. J Neurosurg 56(1):106–113. doi:10.3171/
jns.1982.56.1.0106 Krings T, Geibprasert S (2009) Spinal dural arteriovenous
262. Anderson DK, Saunders RD, Demediuk P et al (1985) fistulas. AJNR Am J Neuroradiol 30(4):639–648
Lipid hydrolysis and peroxidation in injured spinal Krings T, Lasjaunias PL, Hans FJ et al (2007) Imaging in
cord: partial protection with methylprednisolone or spinal vascular disease. Neuroimaging Clin N Am
vitamin E and selenium. Cent Nerv Syst Trauma 17(1):57–72
2(4):257–267 Krishna V, Lazaridis C, Ellegala D et al (2012) Spinal cord
263. Hall ED, Braughler JM (1982) Glucocorticoid mech- infarction associated with subarachnoid hemorrhage.
anisms in acute spinal cord injury: a review and ther- Clin Neurol Neurosurg 114(7):1030–1032
apeutic rationale. Surg Neurol 18(5):320–327 Lasjaunias P, Berenstein A, TerBrugge K (2001) Clinical
264. Fehlings MG, Tator CH (1999) An evidence-based vascular anatomy and variations. In: Surgical
review of decompressive surgery in acute spinal cord neuroangiography, vol 1. Springer, Berlin
injury: rationale, indications, and timing based on Nair S, Gobin YP, Leng LZ et al (2013) Preoperative
experimental and clinical studies. J Neurosurg embolization of hypervascular thoracic, lumbar, and
91(1 Suppl):1–11 sacral spinal column tumors: technique and outcomes
265. Fehlings MG, Perrin RG (2005) The role and timing from a single center. Interv Neuroradiol 19(3):377–385
of early decompression for cervical spinal cord injury: Patsalides A, Knopman J, Santillan A, Tsiouris AJ,
update with a review of recent clinical evidence. Riina H, Gobin YP (2011) Endovascular treatment of
47 Spinal Cord Infarction and Differential Diagnosis 1183

spinal arteriovenous lesions: beyond the dural fistula. Setacci F, Sirignano P, De Donato G et al (2010)
AJNR Am J Neuroradiol 32(5):798–808 Endovascular thoracic aortic repair and risk of spinal
Saindane AM, Boddu SR, Tong FC, Dehkharghani S, Dion cord ischemia: the role of previous or concomitant
JE (2014) Contrast-enhanced time-resolved MRA for treatment for aortic aneurysm. J Cardiovasc Surg
pre-angiographic evaluation of suspected spinal dural (Torino) 51(2):169–176
arterial venous fistulas. J Neurointerv Surg 7:135–140 Thurnher MM, Cartes-Zumelzu F, Mueller-Mang C (2007)
Santillan A, Nacarino V, Greenberg E, Riina HA, Gobin Demyelinating and infectious diseases of the spinal
YP, Patsalides A (2012) Vascular anatomy of the spinal cord. Neuroimaging Clin N Am 17(1):37–55
cord. J Neurointerv Surg 4(1):67–74
Index

A Arteriotomy, 197–198
Acetazolamide, 1035 Arteriovenous malformations, 79, 262, 264, 1028, 1081,
Acetylsalicylic acid, 1027 1086
ACTA2, 1027 Arteriovenous malformations (AVMs), 79, 262–264, 397
Acute disseminated encephalomyelitis (ADEM), 1152–1153 Artery of Adamkiewicz (AKA), 1129–1130
Acute hemiparesis, 1011 Artery-to-artery embolism, 1015
Acute polyneuropathy, 1158 Artifacts, 586, 588
AIDS associated myelopathy, 1159 Ascending pharyngeal artery, 91
Alagille, 1027 Asymptomatic Carotid Atherosclerosis Study (ACAS), 192
Amyloid angiopathy, 396 Asymptomatic ICAS, 206
Anastomosis, 294, 296–298, 300 Atherosclerosis, 96–97, 171–172, 174
Ancillary test, 867 AVM, 991, 1029
Anesthetic management, 540 Axial fat-suppressed, 1025
Aneurysms, 199–200, 393–394, 397, 522
definition of, 552
iron age, 554–563 B
modern era, 571–577 Balloon remodeling, 554–563, 579
renaissance, 563–568 Basilar artery fenestration, 165–166
stone age, 553 Basilar artery strokes, 1016
Angiogenesis, 304, 306–307, 316, 318 Bell palsy, 1019
Angiography, 294–295, 297, 299, 1089 Bernoulli’s principle, 52–53
CT, 623–625 Black-blood, 100–101, 130
digital subtraction, 627–629 Bleeding disorders, 1028
super-selective angiography, 631 Blister aneurysm, 522–531
Angio-MR, 1079 Blister-like aneurysm, 552, 574
Anoxia, 332, 356–359 Blood blister aneurysm, 522
Anterior cerebral artery variations, 25 Blood vessels, 304–307
Anterior spinal artery (ASA), 1128, 1129, 1134 Bone infarction, 443, 455–456
Anticoagulant therapy, 1035 Bow Hunter’s syndrome, 184–188
Anticoagulation, 1027 Brain arteriovenous malformation (bAVM)
Antiplatelet therapy, 1027 angioarchitecture, 629
Antithrombotic therapy, 1011, 1013 arterial aneurysms, 630–631
Aortic arch, 88 computed tomography, 623–625
Apparent diffusion coefficient (ADC), 942, 945, 1014, definition, 605–606
1030, 1146, 1150 digital subtraction angiography, 627–629
Arachnoid granulations, 950 embolization, 633–634
Arrhythmia, 1025 embryology, 612–614
Arterial dissection, 1024–1025 epidemiology, 611–612
Arterial ischemic stroke, 1010 hemorrhage risk, 608–611
Arterial spin labeling (ASL), 460, 848–849 magnetic resonance imaging, 625–627
Arterial tortuosity syndrome, 147 microsurgical resection, 633
Arterial vasospasm, 510 natural history, 606
Arterials ischemic stroke, 1030 nidal size, 631
Arteriopathies, 1014, 1016, 1024 pathophysiology, 614–622

# Springer Science+Business Media New York 2016 1185


L. Saba, E. Raz (eds.), Neurovascular Imaging,
DOI 10.1007/978-1-4614-9029-6
1186 Index

Brain arteriovenous malformation (bAVM) (cont.) Cerebral sinovenous thrombosis (CSVT), 1010, 1030
radiotherapy, 634–635 Cerebral vascular malformations, 720
venous drainage, 631 Cerebral vasculitis, 698, 700, 703
Brain Cerebral venous and sinus thrombosis (CVST), 936
amphibians, 9 Cerebral venous system, 956–957
birds, 10 Cerebrovascular disease, 223, 227
death, 58, 866–892 Cerebrovascular reactivity, 1022
fishes, 6, 8 Cerebrovascular reserve (CVR), 1022
hemorrhage, 552 Childhood arterial ischemic stroke (CAIS), 1010
ischemia, 470–471 Choroidal stage, 15–16
mammals, 10–12 Chronic cerebrospinal venous insufficiency (CCSVI),
reptiles, 9 954–955, 962–965, 968, 971–974
retraction, 541 Churg-Strauss syndrome, 769
tumor, 1029 Circle of Willis, 48
Brainstem infarction, 1016 Clip inspection, 543
Bypass surgery, 1023 Clipping, 506, 513, 553, 556, 563, 574
Clip-wrapping, 526–527
Clot retrievers, 427–428
C Coagulopathy, 241
Capillary malformations, 988 Cognitive decline, 1024
Capillary telangiectasias, 742 Cognitive dysfunction, 1036
Cardiac catheterization, 1025 Cognitive outcome, 1036
Cardiac surgery, 1025 Coil(ing), 504, 554, 571
Cardiomyopathy, 1025 Coil embolization, 584–586, 591
Carotid aneurysms, 154–155 COL4A1, 1027
Carotid artery, 97–98 Collateral circulation, 208, 215, 294, 300
stenting, 108 Collateralization, 1023
Carotid artery dissection (CAD), 116–135 Color Doppler ultrasound, 1033
Carotid artery surgery Common carotid artery, 88–89, 93
aneurysms, 199–200 Complications, vasospasm and post-treatment, 589–599
carotid body tumours, 198–199 Computed tomography (CT), 332, 353, 920, 1011
pre-operative study, 192–195 Computed tomography angiography (CTA),
technique of, 196–198 391, 585–586, 589–590, 599, 623, 948,
Carotid body tumours, 198–199 Congenital heart disease, 1019, 1025
Carotid cavernous fistula (CCF), 658–680 Congenital neurovascular syndromes
Carotid endarterectomy, 98, 106, 108 AVM/AVF syndromes, 1047–1051
Carotid stenosis, 143 cavernous malformations, 1055, 1057
Carotid-vertebrobasilar variant, 165–167 MELAS, 1057, 1059
Carotidynia, 156–157 moyamoya disease, 1042–1043
Cavernoma, 720 PHACE syndrome, 1043–1047
Cavernous hemangiomas, 720 Sturge Weber syndrome, 1051–1053
Cavernous malformations (CMs), 266–267, 994–996 venous malformations, 1054–1055
Central nervous system vasculitis, 285, 693 Connective tissue disorders, 147, 150, 1025
Cerebral amyloid angiopathy, 244–248 Continuous EEG, 1027
Cerebral aneurysm, 583–586, 590 Continuous flushing technique, 69
Cerebral arteriopathies, 1015 Contrast-enhanced CT, 947
Cerebral autosomal dominant arteriopathy with subcortical Contrast-enhanced venous MRA, 944
infarcts and leukoencephalopathy (CADASIL), Conventional angiography (CA), 1015, 1023
762–763 Conversion disorders, 1019
Cerebral autosomal recessive arteriopathy with subcortical Cord sign, 945, 1035
infarcts and leukoencephalopathy (CARASIL), Corkscrew vessels, 939
763–764 Corpus callosum, 1036
Cerebral fat embolism, 449 Cortical venous drainage, 647
Cerebral hemodynamics, 223 Cortical venous reflux, 665
Cerebral infarction, 694, 698 Cortical venous thrombosis (CVT), 937
Cerebral, ischemia, 710 Cortical-subcortical hypoattenuating, 1012
Cerebral microbleeds, 251 Cranial ultrasound (CUS), 1012, 1030
Cerebral microhemorrhages, 247 Craniocervical artery dissection (CCAD), 44–45
Index 1187

Crescent-shaped rim, 1025 E


CREST trial, 192 Eagle syndrome, 157–158
Crossed cerebellar diaschisis, 800 ECASS, 1017
Cryptogenic stroke, 108–109 Echo-planar imaging (EPI), 1146
CT angiography. See Computed tomography angiography EFNS-guidelines, 795
(CTA) Ehler Danlos syndrome, 147
CT venography (CTV), 1032, 1035 Embolectomy, 432–433
CTA. See Computed tomography angiography (CTA) Emboli, 331–332, 357
Cytotoxic edema, 945 Embolization, 633–634, 991
iatrogenic, 1138
spinal angiography and, 1138
D transarterial, 650
D-dimer, 938 transvenous, 648, 650
Decompressive surgery, 1018 Embryonic Carotid-Basilar system anastomoses, 90
Deep cerebral veins, 938 Empty delta sign, 947, 1035
Deep cerebral venous thrombosis (DVT), 937 End diastolic velocity (EDV), 193
Dehydration, 1032 Endarterectomy, 196–198
Delivery, 857 Endocarditis, 1025
Dense vein sign, 945 Endovascular embolization, 671, 673, 1106, 1113
Descending corticospinal tract, 1036 Endovascular procedures, 1028
Detachable balloon, 553–554, 579 Endovascular techniques, 584–586
Developmental venous anomaly (DVA), 733–736 Endovascular therapy, 530–531
Diagnostic delay, 1011 Endovascular treatment, 426–430, 432
Diffusion, 1079 Epidural hematomas, 405–407
Diffusion tensor imaging (DTI), 457, 459, 1036, European Carotid Surgery Trial (ECST), 192–193
1084–1085, 1149 Evidence based medicine, 416–418
Diffusion weighted imaging (DWI), 942, 944, 1013–1014, External carotid artery (ECA), 89–92, 294–295,
1030, 1127 297, 299
spinal cord, 1146–1150 External-internal carotid arteries anastomoses, 92
Diffusion-weighted MRI, 870 Extracorporeal membrane oxygenation (ECMO), 1025
Diffusion-weighted sequences, 945 Extracranial internal carotid artery, 44
Digital subtraction angiography (DSA), 122, 585, Extra-cranial venous abnormalities, 954–955, 960, 962,
588, 939–940, 1127, 1158, 1161 971, 973–974
Digital subtraction cervico-cerebral angiography (DSA)
catheter and wire, 69–70
patient preparation, 68 F
preprocedural assessment, 68 Fabry’s disease, 752–753
procedure, 68–69 Facial artery, 91
projections, 70–83 Fast spin-echo (FSE), 1145
vascular access, 69 Fat supressed T1 sequence, 124
Diplopia, 1032 Fetal distress, 1031
Direct bypass, 852 Fetal posterior cerebral artery, 18
Direct signs, 939 Fiber tractography (FT), 1149
Dissection, 1015–1016 Fibromuscular dysplasia (FMD), 42–43
Dominant anterior choroidal artery, 19 angiography, 141–142
Doppler effect, 49, 50, 53 natural history and clinical features, 140–141
Doppler ultrasound, 1033 types, 141
Double lumen, 1025 Flame shape stenosis, 1025
Down’s syndrome, 1021 Flow, blood, 306–307, 309, 311
Duplications, 24 Flow disruptors, 579
Dural arteriovenous fistulas (DAVFs), 264, 397 Flow-diverters (FDs), 529, 571–579
Dural sinus thrombosis, 937 Fluid-attenuated inverted recovery (FLAIR), 1014
Dynamic Contrast Enhanced Computed Tomography Focal cerebral arteriopathy (FCA), 1020
(DCE-CT), 311 Fractional flow, 219, 227
Dynamic relaxivity contrast enhanced MRI (DRCE-MRI), Frontotemporal craniotomy, 544
310–311 Functional MRI (fMRI), 871
Dynamic susceptibility contrast enhanced MRI (DSCE- Fusiform, 570, 574
MRI), 308–310 Fusion patterns variations, basilar artery, 20–24
1188 Index

G spinal cord, 1127


Gamma knife surgery (GKS), 634–635 spinal venous, 1138–1139, 1142
GDCs, 555, 571 thromboembolic, 1136
Giant cell arteritis (GCA), 41–42, 144 venous, 1133
Global brain hypoxia, 471, 486, 492 Infection, 117
Glossopharyngeal neuralgia, 786–787 Inflammatory, 1015
Glue, 650 Integrin, 319–321
Gradient-echo (GRE), 1145, 1165 Internal carotid artery (ICA), 89, 294–295, 299
Guidelines, for tPA administration, 417 occlusion, 58–60
Internal maxillary artery, 92
Interventional treatment, 940
H Intimal fibrodysplasia, 1024
Headache, 1032 Intimal flap, 1025
Hemicraniectomy, 1018 Intra-arterial thrombolysis, 940
Hemifacial spasm (HFS), 784–786 Intracardiac shunt, 63
Hemiparesis, 1030 Intracranial aneurysms, 60, 74, 258, 522–523,
Hemiplegic cerebral palsy, 1030–1031 525, 527–528, 531, 536
Hemoglobin, 385, 388 bone resection, 541
Hemolytic anemia, 441 brain retraction, 541
Hemorrhagic neoplasm, 276–281 complication, 512–513, 546
Hemorrhagic stroke, 445, 447, 1011, 1028, 1030 conservative management, 508
clinical presentation, 385 clip inspection, 543
contrast enhanced MRA, 393–394 craniotomy selection, 545
magnetic resonance imaging, 387, 390 endovascular treatment, 504, 506
non-contrast computed tomography, 385–386 epidemiology, 498
vascular malformations, 397 follow-up procedure, 547
Hemorrhagic transformation, 1012 imaging, 501–503
of infarct, 270–274 permanent clipping, 542
Herniation, 1029 postoperative care, 545–546
High resolution magnetic resonance imaging (HR-MRI), prognostic factors, 513
129 surgical clipping, 536
HII. See Hypoxic ischemic injury (HII) surgical treatment, 506–508
Homocystinuria, 1025, 1027 subarachnoid dissection, 541
Hydrocephalus, 508–510, 1029 surgical clipping, 537
Hyperdense artery sign, 1012 symptoms, 499–500
Hyperdense dural sinus sign, 1035 temporary clipping, 542–543
Hypertension, 395 vascular control, 542
Hypertensive hemorrhage, 234–238 Intracranial arterial stenosis, 219, 224
Hypoperfusion, 1016 Intracranial atherosclerosis (ICAS), 206–207, 209
Hypoxia, 304, 306–307, 316, 332, 356–359 Intra-cranial dissection, 1025
Hypoxic ischemic injury (HII), 470–472, 476, 478–486, Intracranial dural fistula
488–492 classification, 643–645
Hypoxic-ischemic encephalopathy (HIE), 1030 imaging, 647–648
natural history, 646–647
physiopathology, 642–643
I symptoms, 646
Idiopathic transverse myelitis (ITM), 1151–1152 treatment, 648–655
Incidence, 1010–1011 Intracranial hemorrhage, 608, 611, 665–666, 828, 855,
Indirect bypass, 852–853 1012, 1035
Indirect signs, 939 Intracranial hypertension, 1035
Indocyanine green (ICG) fluorescent videoangiography, Intracranial pressure, 1032
543 Intracranial revascularization, 433
Infarct(s), 370–373, 376, 379 Intramural hematoma, 1025
Infarction Intraoperative digital subtraction angiography (DSA), 543
hemorrhagic spinal cord, 1138 Intravenous fibrinolysis (IVF), 425, 430–431
hemorrhagic, 1133 Ischemia, 331, 345, 1075–1077, 1079
ischemic spinal cord, 1132–1136, 1143 Ischemic stroke, 96, 98, 108, 206–207, 425–429, 445
ischemic, 1133 Ivy sign, 1022
Index 1189

J MRA. See Magnetic resonance angiography (MRA)


Japan Adult Moyamoya (JAM) Trial, 857 MRI with venography (MRV), 1032
Jugular vein, 900 Multi-contrast MRI, 102
Jugular vein reflux, 954 Multi-detector computed tomography-angiography
(MDCTA), 195
Multiple sclerosis (MS), 955, 962–966, 969, 971,
J 974, 1150–1151
Kawasaki disease, 769 Mural hematoma, 116, 118
Myelitis
acute, 1127
L transverse, 1146, 1151
Labyrinthine hemorrhage (LH), 456 Myelopathy, 1112, 1119
Labyrinthitis ossificans (LO), 456 Myxoma, 1025
Lactic acidosis, 1027
Lacunar infarct, 330, 355–356
Lindegaard ratio, 56 N
Lingual artery, 91 Neck veins, 957, 966, 970
Loss of gray matter–white matter differentiation, 1012 Neoplasms, 1028
Lumbo-peritoneal shunt, 1035 Neural tube/spinal cord, 1095
Lymphatic malformations, 988–991 Neuro-Behcet’s disease, 775, 1158
Neurofibromatosis, 150–151
Neurofibromatosis type1, 1021, 1027
M Neuroimaging, 208
Magnetic resonance angiography (MRA), 391–392, Neuromyelitis optica, 1158
394, 585–586, 588–589, 595, 625, 831, 834, Neuroprotection, 1027, 1035
844, 881, 940 Neuroprotective therapy, 1011, 1013
Magnetic resonance imaging (MRI), 96, 103, 337, 355, Neurosarcoidosis, 1151, 1158
831, 836–837, 923 Neurovasculitides, 684–687, 695, 698, 700, 705, 707, 714
Malignant infarct, 1012 Non-accidental injury, 1028
Marfan syndrome (MS), 150 Non-accidental trauma, 1025
Mastoiditis, 1032, 1035 Non-atherosclerotic vascular disease
Mechanical index (MI), 64 craniocervical artery dissection, 44–45
Mechanical recanalization, 940 extracranial internal carotid artery, 44
Meningitis, 1035 fibromuscular dysplasia, 42–43
Methemoglobin, 389, 393 giant cell arteritis, 41–42
Micro-Doppler ultrasonography, 543 moyamoya, 43–44
Microsurgery, 633 Takayasu arteritis, 40–41
Microsurgical clipping, 528, 584 Non-contrast T1-weighted images, 1025
Middle cerebral artery, 1015 Non-convulsive seizures, 1027
Migraine, 1019 Non-progressive childhood primary angiitis of the central
characterization, 792 nervous system, 1020
diagnostic criteria without and with aura, 792 Non-traumatic intracranial hemorrhage
MRI scans, 811 cerebral amyloid angiopathy, 244, 248
neuroimaging indications, 794–795 coagulopathy, 241
pathophysiology, 793–794 CT and MRI, 239
and stroke, 796, 807 GRE T2* images, 248
T2 hyperintensities, 807–811 hemorrhagic neoplasm, 276–281
Migrainous infarction, 796–797, 803 hemorrhagic transformation of infarct, 270–274
Mitochondrial encephalopathy, 1027 hypertensive hemorrhage, 234–238
Mitochondrial myopathy, Encephalopathy, Lactic nontraumatic subarachnoid hemorrhage, 251–262
Acidosis, and Stroke-like symptoms (MELAS), radiation-induced disease, 283
1057–1059 susceptibility weighted imaging, 248–251
Moyamoya, 1011, 1015, 1024, 1027 vascular malformations, 262–270
Moyamoya disease (MMD), 758, 819, 858, 1042–1043 vasculitis, 285
Moyamoya syndrome, 43–44, 449–451 venous thrombosis, 274–276
MR angiography. See Magnetic resonance angiography Non-traumatic subarachnoid hemorrhage, 251–262
(MRA), North American Symptomatic Carotid Endarterectomy
MR spectroscopy (MRS), 460, 871 Trial (NASCET), 192–193
1190 Index

O Propagation, 1035
Occipital artery, 91 Prothrombotic states, 1032
Onyx, 650 Pseudoaneurysm, 1025
Ophthalmic artery, 49, 60 Pseudonormalization, 1030
Optic nerve sheath fenestration, 1035 Pterional craniotomy, 545
Optic radiation, 1036 Puff of smoke, 1021
Osteomyelitis, 456
Otitis media, 1032
Q
Quantitative assessment, 947
P Quantitative MRI (qMRI), 460–461
Pacchionian granulations, 950
Papilledema, 1032, 1035
Parainfectious, 1015 R
Paraplegia, 1108 Radiation, 1013, 1015
Parenchymal changes, 939 microhemorrhage, 283
Parenchymal hemorrhage, 939 myelitis, 1159
Parenchymal infarcts, 1033 vasculopathy, 283
Partially-thrombosed aneurysm, 570, 574 Radiation-induced microangiopathy, 287
Patent foramen ovale, 63 Radicular artery, 1128, 1130–1131, 1143, 1149
Peak systolic velocity (PSV), 193 Recurrence, 121, 207, 216
Pediatric ASPECT score, 1018, 1036 Rehabilitation, 1027
Pediatric ICH score, 1030 Resistive index (RI), 53
Pediatric NIHSS, 1018 Revascularization, 1015
Pediatric stroke, 1057, 1059 Reversible cerebral vasoconstriction syndrome (RCVS), 756
Perfusion, 845, 847 Risk factors, 1011, 1019
imaging, 223, 459–460 Risk stratification, 207–208, 229
scintigraphy, 889 RNF213, 1027
Perinatal hemorrhagic stroke, 1031 Rupture, 499, 504
Perinatal period, 1030
Perinatal strokes, 1030
Periventricular leukomalacia, 1031 S
Periventricular venous infarction, 1030 Sclerotherapy, 987
Permanent clipping, 543 Segmental artery, 1128
Permeability, endothelial membrane, 306, 308–310 anastomose, 1096
Persistent trigeminal artery, 19 Seizures, 1011, 1019
PFO, 1026 Serial lumbar punctures, 1035
PHACE syndrome, 1027, 1043, 1047 Shamblin’s classification, 198
Phase-contrast venous MRA, 944 Sickle cell disease (SCD), 57–58, 440, 463, 1011, 1021
Plaque imaging, 205, 229 Signal-to-noise ratio (SNR), 1145, 1147
Plaque rupture, 98, 107 Silent cerebral infarct (SCI), 447, 449
Polyarteritis nodosa (PAN), 768 Single photon emission computed tomography (SPECT),
Polycythemia, 1035 461, 847–848, 852
Positron emission tomography (PET), 308, 316, 318–319, Sinus pericranii (SP), 744, 993–994
461, 847 Sinus thrombosis, 937
Posterior auricular artery, 92 Sloan ratio, 56
Posterior circulation, 169, 180–181 Spastic cerebral palsy, 1030
Posterior circulation stroke, 1015 Spinal angiogram, 1105
Posterior Reversible Encephalopathy Syndrome (PRES), Spinal cord, 1075, 1079, 1081, 1086
454, 753 vascular neurophylogeny, 3–4
Posterior spinal artery (PSA), 1129, 1141, 1144 Spine, 1068
Post-stroke dystonia, 1036 arteries, 1096, 1101
Post-varicella angiopathy, 1020 veins, 1101–1102
Prechoroidal stage, 13, 15 Spontaneous intracranial hemorrhage, 244. See also Non-
Pregnancy, 857 traumatic intracranial hemorrhage
Presumed perinatal ischemic stroke, 1030 Stenosis, 1014
Primary angiitis of CNS (PACNS), 764 Stent(s), 135, 524–526, 529–531, 563, 591, 593
Proatlantal artery, 20 retrievers, 427, 429–430
Index 1191

String of beads, 1024 Time of flight, 392–393


String of pearls, 1022 Time-resolved MRA, 944
Stroke, 172–176, 299, 364, 376–377, 443–444, 449, 606, TOF-MRA, 1014
612, 685, 693 Torcula, 1033
mimics, 1011, 1013 Tortuosity, 1014
Stroke-like episodes, 1027 tPA, 1011, 1013, 1028
Sturge Weber syndrome (SWS), 1051, 1053 Tractography, 1036
Sub acute combined degeneration, 1158–1159 Transcranial Doppler (TCD), 448, 461
Subarachnoid dissection, 541 screening, 1024
Subarachnoid hemorrhage, 498, 523–526, 528–531, 939 Transforaminal approach, 49
complications, 508, 512 Transient cerebral arteriopathy, 1015, 1020
DSA-negative, 503 (see also Intracranial aneurysm) Transient ischemic attack, 206
Subarachnoid hemorrhage (SAH), 385, 404–405 Transtemporal approach, 48
Subclavian steal syndrome, 185 Traumatic head injury, 1028
Subdural hematomas, 405, 407 Trigeminal autonomic headaches, 795
Superficial temporal artery, 92 Trigeminal neuralgia, 781, 783
Superficial temporal artery-to-middle cerebral artery Tumors, 1068, 1074
(STA-MCA) anastomosis, 852, 857 Turbulent flow, 1014
Superior oblique myokymia, 787
Superior sagittal sinus, 937
Superior thyroid artery, 91 U
Surface coils, 129 Ultrasound, 1033
Susceptibility-weighted angiography (SWAN), 944 Unenhanced CT, 946
Susceptibility weighted imaging (SWI), 837, 845,
977, 1014, 1029, 1033
Susceptibility-weighted magnetic resonance imaging V
(SWI) Valvular heart disease, 1025
capillary telangiectasias, 742 Varicella zoster virus (VZV), 773
cerebral cavernous malformation, 731 Vascular anomalies, 1029
DVA, 739 Vascular disorder, 606. See also Brain arteriovenous
Symptomatic ICAS, 206 malformation
Systemic lupus erythematosus (SLE), 771 Vascular neuroembryology, 12
accessory MCA, 18–19
adult pattern, 17
T choroidal stage, 15–16
T2 shine-through, 1014 dominant anterior choroidal artery, 19
T2-weighted, 1014 duplications, 24
T2*WI gradient-echo MRI sequences, 942 fetal posterior cerebral artery, 18
Takayasu arteritis (TA), 40–41, 142, 144 fusion patterns variations, basilar artery, 20, 24
criteria for diagnosis, 40 persistent hypoglossal artery, 19
etiology, 40 persistent trigeminal artery, 19
treatment, 41 prechoroidal stage, 13
types, 40 proatlantal artery, 20
Tandem occlusions, 430, 437 6 weeks, 16
Technic MR, 1080 7 weeks, 16–17
Temporal arteritis. See Giant cell arteritis (GCA) 9 weeks, 17
Temporary clipping, 542 Vascular neurophylogeny, spinal cord, 3–4
Territorial distributions, 1015 Vascular territories, 364, 367, 378
Thalami, 939 Vascular venous imaging, 919
Thalamic hemorrhage, 1033 Vasculitis, 142–146
Thermal index, 64 Vasculopathy, 451–454, 1015
Thrombectomy, 426–430, 1028 Vasogenic edema, 945
Thrombocytopenia, 1031 Vaso-occlusion, 441
Thromboembolic, 1025 Vasospasm, 48, 54, 57, 1029
Thrombolysis, 1170, 1172 Vein of Galen aneurysmal malformation, 999, 1002
Thrombophilia, 1026 Venous edema, 939
Thrombophlebitis, 1032 Venous infarction, 939
Thrombus, 1032 Venous malformations, 985, 988
1192 Index

Venous thrombosis, 274–276, 399 W


Venous time-of-flight (TOF-) MRA, 944 Wallerian degeneration, 1031, 1036
Ventricular assist device, 1025 Watershed infarct, 330, 353
Vertebral artery, 88–92, 172 Watershed territory, 1132
dissection, 177 Watershed/border zones, 1016
fenestration, 166 White matter lesions (WMLs), 807, 811
stenosis, 172, 177 Williams, 1027
Visual field deficits, 1032, 1035 Wrapping, 526–527

You might also like