You are on page 1of 9

Research Article

Tissue Inhibitor of Metalloproteinases-1 Promotes Liver Metastasis


by Induction of Hepatocyte Growth Factor Signaling
1 1 4 1 6
Charlotte Kopitz, Michael Gerg, Obul Reddy Bandapalli, Dilek Ister, Caroline J. Pennington,
1 1 7 5 8
Stephanie Hauser, Christin Flechsig, Hans-Willi Krell, Dalibor Antolovic, Keith Brew,
9 2 3 2
Hideaki Nagase, Manfred Stangl, Claus W. Hann von Weyhern, Björn L.D.M. Brücher,
4 10 6 1
Karsten Brand, Lisa M. Coussens, Dylan R. Edwards, and Achim Krüger
1
Institut für Experimentelle Onkologie und Therapieforschung, 2Chirurgische Klinik, and 3Institut für Allgemeine Pathologie und
Pathologische Anatomie, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany; 4Institute of Pathology
and 5Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany; 6School of Biological Sciences, University
of East Anglia, Norwich, Norfolk, United Kingdom; 7Roche Diagnostics GmbH, Penzberg, Germany; 8Department of
Biomedical Science, Florida Atlantic University, Boca Raton, Florida; 9The Kennedy Institute of Rheumatology
Division, Imperial College School of Medicine, London, United Kingdom; and 10Department of Pathology
and Comprehensive Cancer Center, University of California, San Francisco, California

Abstract these facts, the historical view of MMPs and cancer was that they
Balanced expression of proteases and their inhibitors is one exerted proinvasive and prometastatic activity by mere ECM
prerequisite of tissue homeostasis. Metastatic spread of tumor remodeling (1). Thus, it was hypothesized that therapeutic broad-
cells through the organism depends on proteolytic activity and spectrum inhibition of MMPs would result in antimetastatic
is the death determinant for cancer patients. Paradoxically, activity (1). Indeed, under certain experimental circumstances,
overexpression of the endogenous broad-spectrum metalloprotei-
increased expression of tissue inhibitor of metalloproteinases-
1 (TIMP-1), a natural inhibitor of several endometalloprotei- nase [including a disintegrin and metalloproteinase-10 (ADAM-10);
ref. 4] inhibitor tissue inhibitor of metalloproteinases-1 (TIMP-1;
nases, including matrix metalloproteinases and a disintegrin
ref. 5) can reduce tumor cell invasion (6, 7).
and metalloproteinase-10 (ADAM-10), in cancer patients is
In contrast to these observations are other results, which are
negatively correlated with their survival, although TIMP-1
actually in agreement with clinical studies showing a correlation
itself inhibits invasion of some tumor cells. Here, we show that
between elevated TIMP-1 expression and poor prognosis in many
elevated stromal expression of TIMP-1 promotes liver metas-
human cancer types (5, 8), where elevated levels of TIMP-1 either
tasis in two independent tumor models by inducing the
had no effect on metastasis (9) or led to increased incidence
hepatocyte growth factor (HGF) signaling pathway and
(10, 11) or growth (12) of primary tumors. These effects of TIMP-1
expression of several metastasis-associated genes, including
on cancer development have been attributed to its known
HGF and HGF-activating proteases, in the liver. We also found
proproliferative, antiapoptotic (5), or proangiogenic (13) activities,
in an in vitro assay that suppression of ADAM-10 is in
which are necessary but not sufficient for metastasis.
principle able to prevent shedding of cMet, which may be one
Tumor cell invasion and metastasis are the primary determi-
explanation for the increase of cell-associated HGF receptor
nants of the survival of cancer patients (13). Signaling pathways
cMet in livers with elevated TIMP-1. Similar TIMP-1–associ-
leading to the expression of proteolytic enzymes have been
ated changes in gene expression were detected in livers of
suggested to be key mediators of invasive growth (14). Hepatocyte
patients with metastatic colorectal cancer. The newly identi-
growth factor (HGF/scatter factor) signaling regulates a multitude
fied role of TIMP-1 to create a prometastatic niche may also
of downstream prometastatic effector molecules, such as urokinase-
explain the TIMP-1 paradoxon. [Cancer Res 2007;67(18):8615–23]
type plasminogen activator (uPA), uPA receptor (uPAR), plasminogen
activator inhibitor-1 (PAI-1), and MMPs (15–17). HGF stimulates
Introduction metastasis of diverse tumor cells (15) and elevated levels of
Metastasis and progressive scattering of tumor cells throughout circulating HGF (18) or aberrant activity of cMet (14) has been
tissues is the main cause of organ failure and subsequent death detected in patients with many cancer types, including non-
of cancer patients. Proteolytic remodeling of components of the Hodgkin’s lymphoma (19) and colorectal cancer (14). Although
extracellular matrix (ECM) in tissue interstitium is a prerequisite of elevated TIMP-1 expression in these patients correlates with
the metastatic process (1). Matrix metalloproteinases (MMP) have decreased survival, a link between TIMP-1 and metastasis-
long been thought to be major regulators of the pericellular promoting pathways has previously not been described. Here,
protease-dependent steps of tumor progression (2). Altered we address this issue and report that elevated stromal levels of
expression profiles of some MMPs have been correlated with poor TIMP-1 induce HGF signaling, leading to enhanced expression of
clinical prognosis for several human tumor types (3). Based on other metastasis-promoting genes. Consequently, susceptibility
of the liver for metastasis of tumor cell lines of different origin was
shown to be increased, assigning a new role to TIMP-1 as a factor
Note: Supplementary data for this article are available at Cancer Research Online creating a prometastatic niche.
(http://cancerres.aacrjournals.org/).
Requests for reprints: Achim Krüger, Institut für Experimentelle Onkologie und
Therapieforschung, Klinikum rechts der Isar der Technischen Universität München,
Ismaninger Str. 22, D-81675 Munich, Germany. Phone: 49-89-4140-4463; Fax: 49-89- Materials and Methods
4140-6182; E-mail: achim.krueger@lrz.tu-muenchen.de.
I2007 American Association for Cancer Research. Cell culture and adenoviruses. HEK 293 (20), HT1080lacZ-K15 (21),
doi:10.1158/0008-5472.CAN-07-0232 L-CI.5s (22), and 293T cells (23) were cultured as described previously.

www.aacrjournals.org 8615 Cancer Res 2007; 67: (18). September 15, 2007
Cancer Research

NIH3T3 cells were cultured similarly to HT1080lacZ-K15 (21). Amino acids adenoviral shuttle plasmid pKA1 (26). Recombinant adenoviruses were
1 to 126 of the human TIMP-1 cDNA (N-TIMP-1; ref. 24) were cloned into generated, purified, and plaque titered as described previously (26). As
pSecTag2/Hygro A (Invitrogen). A DNA fragment consisting of Ign-leader control, Addl70-3, an adenovirus without transgene, was used (27).
sequence and N-TIMP-1 cDNA or full-length human TIMP-1 cDNA Functionality of adenoviral constructs was tested in vitro (data not shown).
(pGEMhTIMP-1; ref. 25), respectively, was obtained and cloned into the Adenoviral infection in vitro was done as described previously (26).
Experimental metastasis assays. Pathogen-free, athymic CD1nu/nu mice
(Charles River) were injected i.v. with 2  109 plaque-forming units of
Addl70-3, AdTIMP-1, and AdN-TIMP-1, respectively, or vehicle (1% glycerin
in PBS) alone. Three days later, 5,000 lacZ-tagged murine T-cell lymphoma
cells (L-CI.5s; ref. 22) or 1  106 lacZ-tagged human fibrosarcoma cells
(HT1080lacZ-K15; ref. 21) were injected i.v. Alternatively, inoculation of
tumor cells preceded the adenoviral transfer by 2 days (Fig. 1C). Mice were
sacrificed and their livers and lungs were removed at 6 days (L-CI.5s) or 21
days (HT1080lacZ-K15) after inoculation. The right lobe of each lung and
three samples of each liver, with a volume of f0.5 cm3, were snap frozen in
liquid nitrogen for biochemical analysis, and left lung lobes and remaining
liver were stained with 5-bromo-4-chloro-3-indolyl-h-D-galactopyranoside
(X-Gal; Roche Diagnostics) as described (22). Blue multicellular foci on
the surface of the livers (L-CI.5s) or lungs (HT1080lacZ-K15) were counted.
Total metastasis burden was assessed by quantitative real-time PCR
(RT-PCR) for lacZ. All animal experiments were done in compliance with
the guidelines of the Tierschutzgesetz des Landes Oberbayern.
Immunostaining. Fixed sections of livers of the different groups were
paraffin embedded for immunostaining. Dewaxing, rehydration, blocking,
and antigen retrieval of liver sections (4 Am) were done as described (28).
Sections were incubated with primary antibodies [proliferating cell nuclear
antigen (PCNA) antibody, 1:2,000, Novocastra Laboratories Ltd.; cMet
antibody, 1:200, Santa Cruz Biotechnology] for 20 h at 4jC and washed in
TBS, 0.1% (v/v) Tween 20. StreptAB Complex/HRP Duett Mouse/Rabbit kit
(Dako Cytomation) and 3,3¶-diaminobenzidine (DAB) substrate (DAKO
Liquid DAB+ Substrate-Chromogen Solution, DAKO Corp.) were used for
detection. Sections were counterstained with Mayer’s hemalaun (Merck).
For detection of cMet activation, unfixed fresh liver samples were

Figure 1. TIMP-1–mediated reduction of formation of macrometastases but


induction of liver-specific secondary spread of tumor cells. A and B, CD1nu/nu
mice received the respective adenovirus 3 d before inoculation of L-CI.5s (A )
or HT1080lacZ-K15 cells (B ), respectively. Six or 21 d, respectively, after tumor
cell inoculation, mice were sacrificed and livers and lungs were removed.
A, top left, number of X-Gal–stained L-CI.5s macrometastases (>0.2 mm) over
the whole liver surface was reduced by overexpression of human TIMP-1
or N-TIMP-1 compared with the virus control group. Columns, mean; bars, SE.
Addl70-3: 67.60 F 7.73, n = 5; AdTIMP-1: 38.50 F 8.14, n = 6, P = 0.03;
AdN-TIMP-1: 32.83 F 8.32, n = 6, P = 0.009. Close-up pictures of the surfaces
of representative X-Gal–stained L-CI.5s metastasis-bearing livers (bottom )
show increased micrometastasis in livers with elevated levels of TIMP-1
or N-TIMP-1. Top right, the total L-CI.5s tumor burden in livers overexpressing
the TIMP-1 variants was significantly increased compared with the virus
control as detected by quantitative RT-PCR of lacZ in metastasis-bearing livers.
Columns, mean of relative lacZ mRNA amount versus 18S RNA; bars, SE.
Addl70-3: 0.22 F 0.04, n = 3; AdTIMP-1: 1.99 F 0.80, n = 4, P = 0.044;
AdN-TIMP-1: 1.96 F 0.76, n = 4, P = 0.041. B, significant reduction of HT1080
lung metastasis by overexpression of human TIMP-1. Top left, columns,
mean of the number of X-Gal–stained metastases over the whole surface of the
left lung lobe; bars, SE. Addl70-3: 38.1 F 16.1, n = 16; AdTIMP-1: 2.5 F 1.4,
n = 10; P = 0.018. Significant increase of HT1080 metastasis in livers with
elevated levels of TIMP-1. lacZ mRNA and 18S RNA levels were detected in
total RNA of HT1080lacZ-K15 metastasis-bearing livers by quantitative RT-PCR.
Top right, columns, mean of relative lacZ mRNA amount versus 18S RNA;
bars, SE. Addl70-3: 0.016 F 0.002; AdTIMP-1: 0.040 F 0.007, n = 3, P = 0.04.
Bottom, close-up pictures of the surfaces of representative X-Gal–stained
HT1080 metastasis-bearing livers. C, effects of TIMP-1 elevation after tumor
cell inoculation on liver metastasis. CD1nu/nu mice received the respective
adenovirus 2 d after inoculation of L-CI.5s. Six days after tumor cell inoculation,
mice were sacrificed and livers were removed and X-Gal stained. The number of
macrometastases (>0.2 mm) was counted on the whole surface of the liver.
Top left, columns, mean; bars, SE. Addl70-3: 45 F 4, n = 4; AdTIMP-1: 44 F 3,
n = 4. Total metastasis burden was assessed by quantitative RT-PCR of the
marker gene lacZ. Top right, columns, mean of relative lacZ mRNA versus
18S RNA; bars, SE. Addl70-3: 0.29 F 0.16, n = 4; AdTIMP-1: 2.25 F 0.62,
n = 4. Bottom, X-Gal–stained livers. Representative pictures of each group.
Bar , 200 Am. Asterisks, macrometastases; arrows, micrometastases.

Cancer Res 2007; 67: (18). September 15, 2007 8616 www.aacrjournals.org
TIMP-1 Promotes Liver Metastasis via HGF Signaling

embedded in Tissue-Tek OCT compound (Sakura Finetek), shock frozen on 99 nmol/L; c-Jun NH2-terminal kinase (JNK) 1, 12 nmol/L; JNK3, 2 nmol/L;
dry ice, and stored at 80jC until sectioning. Sections were dried, fixed in p38a, 10 nmol/L; p38h, 18 nmol/L; other kinases (254 tested), >1,000 nmol/L;
ice-cold acetone, dried at room temperature, rehydrated in TBS, blocked data not shown; ref. 32].
with 3% goat serum in TBS-Tween 20, and incubated with phosphorylated Knockdown of ADAM-10 in NIH3T3 cells. Oligodeoxynucleotides
Met (Tyr1349) antibody [Cell Signaling Technology; 1:100 in antibody diluting coding for small hairpin RNA interference (shRNAi) directed against murine
buffer (DAKO)] overnight at 4jC. Counterstaining was done with 100 ng/mL ADAM-10 (small interfering RNA sequence: AACCTACGAATGAAGAGG-
4¶,6-diamidino-2-phenylindole (DAPI; AppliChem). Sections were mounted GAC) were designed and obtained from Eurogenetec. Annealed shRNAis
with Moviol (Merck). Green fluorescent signal was displayed using were cloned into pSiren-RetroQ (Clontech). Recombinant retroviruses were
stereomicroscope Axiovert 135 (Zeiss) with integrated digital camera generated as described (23) by transient cotransfection of 293T cells with
disposing the AxioVision LE 4.2 software (Zeiss Vision GmbH). pHIT60 (23), pHCMV-G (33), and pSiren-RetroQ coding for anti-ADAM-10
HGF ELISA. Livers were perfused with 100 mL PBS before excision, shRNAi. Infection of NIH3T3 cells was done according to standard protocols
sample preparation, purification (HiTrap heparin column, Amersham), and (23). Transduced cells were selected with 20 Ag/mL puromycin (Clontech).
ELISA [rat HGF ELISA (Institute of Immunology Co. Ltd.)] were done cMet ELISA. For detection of membrane-bound cMet, NIH3T3 cells
following the manufacturers’ instructions. This kit detects mouse HGF transduced with the different viral constructs were resuspended in 0.1 mol/L
equally well (manufacturers’ manual). The ELISA was done in duplicates Tris, 10 mmol/L EDTA, and 0.1% Triton X-114 (AppliChem) at pH 7.5,
and results were expressed as picogram HGF per milligram extracted liver. supplemented with 10 AL Protease Inhibitor Cocktail (Sigma). Subsequently,
In situ zymography and MMP-9 activity assay. In situ zymography was cells were incubated for 5 min on ice and 5 min at 37jC and centrifuged
done and documented as described previously (29). To detect the origin of for 5 min at 300  g at 30jC. The lower phase containing the membrane
gelatinolytic activity, serial sections were overlaid with DQ-gelatin proteins was used for ELISA. For detection of cMet shedding, transduced
(Molecular Probes; ref. 29) containing either 10 Amol/L E64 (AppliChem), NIH3T3 cells were incubated with serum-free medium for 36 h. Total and
10 Amol/L aprotinin (AppliChem), or 400 Amol/L 1,10-phenanthroline cMet protein in membrane fractions and supernatants were quantified by
(AppliChem). MMP-9 activity was quantified by MMP-9 activity assay bicinchoninic acid assay and cMet ELISA (Mouse HGF R/c-MET ECD
(Amersham) according to the manufacturer’s instructions. The antibody DuoSet, R&D Systems), respectively, following the manufacturers’ instruc-
used in this assay binds human and murine species of MMP-9 tions. Assays were done in triplicates.
(manufacturer’s information). MMP-9 activity in 80 Ag total protein was Statistical analysis. Data were tested for normal distribution and
determined. Assay was done in triplicates. normally distributed data were tested by Student’s t test, and where the
Patient material. Liver tissue of 14 patients with metastatic colorectal normality test failed, Mann-Whitney test was applied. P < 0.05 was
cancer (7 males and 7 females; mean age, 61.0 F 2.3 years) was obtained considered significant.
from patients who had signed informed consent for scientific study of their
tumors and surrounding tissues at the Department of Surgery of the
Results
Klinikum rechts der Isar or of the University Hospital Heidelberg. Tissue
samples were harvested under identical, rigorous procurement protocol Reduction of experimental macrometastases but induction
and snap frozen in liquid nitrogen immediately after surgical resection. of scattered micrometastases in the liver by TIMP-1. To achieve
‘‘Normal’’ liver tissue was harvested from zones 2 cm distant from visible elevated TIMP-1 expression in the host, we transduced full-length
liver metastases. Three experienced pathologists from two independent cDNAs of TIMP-1 by i.v. inoculation of adenoviral vectors into
institutions used cryodissected sections for confirmation of histologic mice. Efficient adenoviral transduction of liver cells was deter-
identity. All samples were stored at 80jC until use. mined by quantitative RT-PCR (Supplementary Fig. S1). Mainte-
RNA isolation, reverse transcription, design of primer and probes,
nance of transgene overexpression achieved by the adenoviral
and quantitative RT-PCR. RNA isolation and reverse transcription were
vectors until the end of all following metastasis assays was verified
done as described (28). Design of primers and probes for murine uPA,
murine uPAR, murine PAI-1, murine matriptase, murine MMP-9, murine (Supplementary Fig. S1). To test the effects of elevated stromal
MMP-2, and murine ADAM-10 was described previously (28, 30, 31). The TIMP-1 on tumor cell invasion, we used the well-established
lacZ primer and probes were designed and obtained from Applied syngeneic in vivo metastasis assay with lacZ-tagged lymphoma
Biosystems ( forward primer: 5¶-CAAGCCGTTGCTGATTCGA-3¶; reverse cells (L-CI.5s), which form hepatic metastatic foci following i.v.
primer: 5¶-GCTCATCCATGACCTGACCAT-3¶; probe: 5¶-FAM-ACCGTCAC- injection. Colonies that we defined as macrometastases in livers
GAGCATCAT-nFQ-3¶). All other quantitative RT-PCRs were done with reached the cutoff size of 0.2 mm in this assay at day 6 after tumor
inventoried primers and probes from Applied Biosystems. Quantitative RT- cell inoculation. At this time point, the combination of successful
PCR was done as described previously (28). extravasation right after tumor cell inoculation and proliferation of
Inhibition of HGF signaling by a specific kinase inhibitor. Three
the extravasated tumor cells for at least six days resulted in
days before i.v. tumor cell inoculation, four mice were transduced with
multicellular foci of this size. Detected micrometastases (<0.2 mm)
adenoviral vectors. Mice received 100 mg/kg of 2-(2,6-dichloro-4-(2-
hydroxyethoxy)phenyl-4-(3-benzyloxyphenyl)-5-(2-[3-hydroxypropyl-ami- resulted from additionally invaded tumor cells if their number
no]pyrimidin-4-yl)-N-H-imidazole (cMet inhibitor), with high affinity for exceeds the 5,000 inoculated tumor cells.
cMet [dissolved in 6% (v/v) Tween 80 with two equivalents HCl], i.p. 1 h Three days after adenoviral gene transfer, TIMP-1–transduced
before and 1 h after tumor cell inoculation. I.p. treatment with the cMet and control mice were challenged with L-CI.5s tumor cells. The
inhibitor was continued daily (100 mg/kg/d). As control, mice were treated pattern of metastasis in TIMP-1–transduced livers was significantly
with vehicle alone. Three days after tumor cell inoculation, mice were altered compared with the control virus–transduced livers:
sacrificed and livers were removed and X-Gal stained as described Elevated levels of TIMP-1 correlated with significant reduction of
previously (22). Inhibition of cMet by this inhibitor was measured by an the number of macrometastases (by 43%, P = 0.003; Fig. 1A, top
ELISA-type assay: Lysate of human colon adenocarcinoma HT29 cells was
left). However, on the level of micrometastases, no decrease but a
exposed to a microtiter plate with an antihuman HGF receptor antibody
drastic increase of scattered infiltrating tumor cells was detected in
(BAF 358 ATP phosphorylation; R&D Systems). ATP phosphorylation of the
receptor kinase (cMet kinase) was detected in the presence or absence of livers with elevated TIMP-1 (Fig. 1A). In one photographic frame
the test compounds using a phosphorylated tyrosine mouse IgG (Santa (1.3 mm  1.5 mm), already 623 micrometastatic foci at the liver
Cruz Biotechnology). The IC50 value for cMet inhibition is 2 nmol/L. Kinase surface were visible (Fig. 1A, bottom), showing that the number of
inhibition activity against other kinases was measured as described by scattered micrometastases in the entire liver by far exceeded the
Fabian et al. [IC50 values: cyclin-dependent kinase 2, 290 nmol/L; HER4, number of inoculated cells (5,000). Quantification of the tumor cell

www.aacrjournals.org 8617 Cancer Res 2007; 67: (18). September 15, 2007
Cancer Research

tag lacZ revealed an overall increase of total metastasis burden in


livers transduced with TIMP-1 cDNA compared with the control
(P = 0.044; Fig. 1A, top right), reflecting induction of micro-
metastasis.
To determine whether inhibition of macrometastasis but
promotion of micrometastasis was a function of the metal-
loproteinase-inhibitory activity of TIMP-1, we investigated the
effect of the N-terminal domain of human TIMP-1 (N-TIMP-1)
harboring the metalloproteinase-inhibitory bioactivity on liver
metastasis. Indeed, elevated levels of N-TIMP-1 significantly
suppressed macrometastasis by 51% (P = 0.009; Fig. 1A) and
promoted micrometastasis (P = 0.041; Fig. 1A), indicating that
N-TIMP-1 is sufficient to evoke the described effects.
We used lacZ-tagged human fibrosarcoma cells (HT1080lacZ-
K15) that typically form large pulmonary metastatic foci and only
few single tumor cells as micrometastases, but no macrometa-
stases, in livers (26) to exclude that the TIMP-1 effect was cell type
dependent. After challenge of CD1nu/nu mice with HT1080lacZ-
K15, significant reduction of lung metastasis by 93% was observed
in TIMP-1–transduced animals compared with mice with virus
control (P = 0.018; Fig. 1B, top left). Again, in livers with elevated
TIMP-1 levels, a significant increase of the metastatic burden was
found (P = 0.04; Fig. 1B, top right). No induction of HT1080lacZ-K15
micrometastasis was detected in lungs (data not shown). In the
above experiments, no differences in metastasis between control
virus–transduced mice and mice treated with vehicle alone were
found, excluding effects of viral transduction itself (data not
shown).
Effects of TIMP-1 elevation on liver metastasis after tumor
cell inoculation. To determine whether TIMP-1 affects liver
metastasis on two different levels, [i.e., on the level of initial
manifestation and subsequent growth (leading to formation of
macrometastases) or subsequent infiltration (micrometastases)],
we inoculated L-CI.5s cells into CD1nu/nu 2 days before adenoviral
transfer of TIMP-1 cDNA. The number of macrometastatic colonies
was not affected compared with the control (Fig. 1C, top left).
However, we still observed induction of micrometastasis compared
with the control (Fig. 1C, bottom), resulting in a significantly
increased total metastasis burden in livers with elevated TIMP-1
(Fig. 1C, top right). This indicates that micrometastases take
advantage of the TIMP-1–modulated host microenvironment. In
contrast, macrometastasis formation was only reduced when
TIMP-1 was already elevated in the host at the time point of
extravasation right after tumor cell inoculation (Fig. 1A). Figure 2. Induction of hepatocyte proliferation and HGF signaling by
Proliferation activity of TIMP-1–induced scattered liver elevated levels of TIMP-1. A, PCNA staining of L-CI.5s metastasis-bearing
micrometastases. To assess if the increased presence of invaded liver sections (4 Am) 9 d after adenoviral gene transfer and 6 d after
L-CI.5s inoculation. Representative sections. Asterisks, PCNA-positive
L-CI.5s cells in the liver was a function of TIMP-1–induced hepatocytes; arrows, PCNA-positive micrometastases. B, left, HGF protein
proliferation, we determined their proliferative status by examining level 9 d after gene transfer and 6 d after L-CI.5s inoculation. HGF
PCNA expression. Indeed, infiltrating T-cell lymphoma cells in protein in pools of three livers per group was quantified by HGF ELISA
(Addl70-3: 30.3 pg/mg; AdTIMP-1: 75.8 pg/mg; AdN-TIMP-1: 80.6 pg/mg).
livers with elevated TIMP-1 or N-TIMP-1 were proliferatively active Right, HGF protein in metastasis-free livers 3 d after gene transfer.
(Fig. 2A), indicating a proproliferative effect of the TIMP-1– HGF protein in pools of three livers per group was quantified by ELISA
modulated environment on these cells, accounting for the increase (Addl70-3: 25.2 pg/mg; AdTIMP-1: 41.1 pg/mg; AdN-TIMP-1: 45.7 pg/mg).
C, increased cMet expression in AdTIMP-1–transduced livers. Paraffin-
of total metastasis burden in the liver. A proproliferative
embedded sections of L-CI.5s metastasis-bearing livers 9 d after
environment was further indicated by the presence of proliferating adenoviral gene transfer and 6 d after tumor cell inoculation were
hepatocytes in these groups (Fig. 2A, arrows). In the control livers, immunohistochemically analyzed for cMet localization. Bottom inset,
proliferation was restricted to the macrometastases (Fig. 2A, left). stronger staining for cMet was found within metastases compared with liver
parenchyma; top inset, controls without primary antibody for each slide.
TIMP-1 induces HGF signaling in the liver. To elucidate a Black arrows, additionally stained micrometastases. D, TIMP-1–induced
molecular mechanism responsible for the TIMP-1–induced pro- activation of cMet. Cryosections of metastasis-bearing (top ) and
proliferative and proinvasive environment in the liver, we tested for metastasis-free (bottom ) livers, respectively, were immunohistochemically
analyzed for phosphorylated cMet (phospho cMet ). Counterstaining was
expression of HGF, also known as scatter factor. Elevated levels of done with DAPI. Asterisks, lumen of blood vessels; arrows, staining of
HGF correlated with the TIMP-1–induced scattered metastatic phosphorylated cMet. Bar , 100 Am.

Cancer Res 2007; 67: (18). September 15, 2007 8618 www.aacrjournals.org
TIMP-1 Promotes Liver Metastasis via HGF Signaling

phenotype (Fig. 2B, left). In fact, increase of HGF expression was gelatinolytic activity in macrometastatic foci in Addl70-3–trans-
already a component of the TIMP-1–altered liver environment as it duced livers but not in foci of AdTIMP-1–transduced animals
could already be detected 3 days after adenoviral gene transfer (i.e., (Fig. 4A, left). As this was inhibitable by the canonical metal-
at the time point of tumor cell challenge; Fig. 2B, right). In addition, loproteinase inhibitor 1,10-phenanthroline (Fig. 4B, left), we showed
cMet, the tyrosine kinase receptor of HGF, was elevated throughout that the achieved TIMP-1 levels indeed inhibited MMP activity
the tissue of metastasis-bearing livers transduced by either TIMP-1 in vivo. No MMP-9–derived proteolytic activity was detected in
species (Fig. 2C) and in macrometastases (Fig. 2C, insets). livers with elevated TIMP-1 (Fig. 4C), indicating the expected
Elevated expression of either TIMP-1 variant led to markedly functional activity of TIMP-1 to inhibit proMMP-9 activation.
increased HGF signaling, as detected by immunohistochemical The gelatinolytic activity detectable in TIMP-1–elevated liver
staining of phosphorylated cMet throughout the liver parenchyma parenchyma distant from metastases (Fig. 4A, right) was by and
(Fig. 2D). In control animals, activated cMet was very low even in large metalloproteinase independent, as it was only slightly
macrometastases (Fig. 2D, top). Importantly, this TIMP-1–mediated inhibitable by 1,10-phenanthroline (Fig. 4B). Addition of broad-
induction of HGF signaling was already present 3 days after TIMP-1 spectrum cysteine protease inhibitor E64 (34) or broad-spectrum
gene transfer (Fig. 2D, bottom), at the time point of tumor cell serine protease inhibitor aprotinin (35) to the in situ zymography
challenge, suggesting that a HGF pathway-connected prometastatic DQ-overlay revealed the remaining gelatinolytic activity as partly
niche has been formed. derived from serine and cysteine proteases (Fig. 4D). Elevated levels
Causal relationship between HGF signaling and TIMP-1– of N-TIMP-1 had the same effects (data not shown). These data
induced scattering of experimental liver metastasis. In TIMP- indicate that increased expression of TIMP-1 in the liver led to an
1–transduced animals, the scattered metastasis phenotype was increase of metalloproteinase-independent proteolytic activity.
already apparent 3 days after inoculation of L-CI.5s cells (Fig. 3A, TIMP-1–induced expression of metastasis-related genes in
top). To investigate whether HGF signaling was essential for the liver. Induction of HGF signaling and nonmetalloproteinase
formation of scattered metastasis, we first transduced mice with proteolytic activity in liver parenchyma by TIMP-1 suggested an
AdTIMP-1 or Addl70-3 virus, treated the mice with a cMet altered HGF signaling-related gene expression favoring metastasis.
inhibitor, and challenged with L-CI.5s cells. Kinase inhibition Therefore, we used quantitative RT-PCR to characterize the
prevented formation of micrometastasis (Fig. 3A, bottom), indicat- expression of HGF-inducible genes, HGF-activating proteases, as
ing that HGF signaling was essential for TIMP-1–induced micro- well as gelatinolytic proteases. Elevation of TIMP-1 led to increase
metastatic spread. The TIMP-1–induced significant increase of of mRNA expression of uPA, uPAR, PAI-1, tissue plasminogen
total metastasis burden compared with the control (P < 0.001; activator (tPA), matriptase, MMP-9, MMP-2, ADAM-10, cathepsin
Fig. 3B) was significantly reduced by cMet inhibition (P < 0.001; G, and neutrophil elastase in the tumor-free liver (Fig. 5). Elevated
Fig. 3B). The number of multicellular metastatic foci was signifi- levels of N-TIMP-1 had the same effects (data not shown).
cantly reduced by elevated levels of TIMP-1 compared with the To test whether these TIMP-1–regulated changes in gene
control (P = 0.048; Fig. 3C), indicating that formation of macro- expression are of more general relevance, we measured TIMP-1
metastasis is not affected by HGF signaling. mRNA expression in human liver tissue samples distant from
Overexpression of TIMP-1 reduces metalloproteinase activ- colorectal metastases. In normal tissue, elevated expression of
ity and increases activity of other proteases in the liver. As TIMP-1 (relative mRNA expression: 98.92 F 19.84, n = 8) was
proteolytic activity is one prerequisite of metastasis, we next associated with increased mRNA expression of uPA, uPAR,
assessed whether TIMP-1–related changes in metastasis rely on matriptase, MMP-9, MMP-2, ADAM-10, cathepsin G, and neutro-
modulated proteolytic activity. In situ zymography revealed phil elastase compared with livers with significantly lower TIMP-1

Figure 3. Reduction of TIMP-1–induced micrometastatic spread by cMet inhibition. A to C, CD1nu/nu mice were transduced with AdTIMP-1 or Addl70-3. Three
days later, mice were treated with a cMet-specific kinase inhibitor [2-(2,6-dichloro-4-(2-hydroxyethoxy)phenyl-4-(3-benzyloxyphenyl)-5-(2-[3-hydroxypropyl-amino]-
pyrimidin-4-yl)-N-H-imidazole] or vehicle alone and challenged by inoculation of L-CI.5s cells. Three days after tumor cell inoculation, mice were sacrificed and livers
were removed and X-Gal stained. A, pictures of the surface of representative livers of each treatment group. Bar, 0.1 mm. B, total metastasis burden in the liver
as detected by quantitative RT-PCR of lacZ. Columns, mean; bars, SE. Addl70-3/vehicle: 0.059 F 0.021; AdTIMP-1/vehicle: 0.728 F 0.233; Addl70-3/cMet inhibitor:
0.040 F 0.016; AdTIMP-1/cMet inhibitor: 0.051 F 0.030; all n = 3. C, number of multicellular metastatic foci was counted on the whole surface of X-Gal–stained livers.
Columns, mean; bars, SE. Addl70-3/vehicle: 55 F 10; AdTIMP-1/vehicle: 36 F 2; Addl70-3/cMet inhibitor: 42 F 9; AdTIMP-1/cMet inhibitor: 25 F 6; all n = 5.

www.aacrjournals.org 8619 Cancer Res 2007; 67: (18). September 15, 2007
Cancer Research

Figure 4. Reduction of gelatinase activity but increase


of non-MMP proteolytic activity by overexpression of
TIMP-1 detected by in situ zymography. A, B, and
D, cryosections of L-CI.5s metastasis-bearing livers,
transduced by either AdTIMP-1 or Addl70-3, were overlaid
with agarose containing DQ-gelatin (A). Parallel sections
were overlaid with the same solution supplemented
with 1,10-phenanthroline (B), E64, and aprotinin (D ),
respectively. Counterstaining was done with DAPI. White
line, boundaries of metastases; asterisks, lumen of blood
vessels; arrows, areas of micrometastases. C, inhibitory
activity of TIMP-1 in the liver 3 d after adenoviral gene
transfer was tested using a MMP-9 activity assay.
Columns, mean; bars, SE. Addl70-3: 100 F 23.23%, n = 4;
AdTIMP-1: 0 F 0%, n = 4.

expression (relative mRNA expression: 21.22 F 1.89, n = 6, P < TIMP-1 indeed inhibited cMet shedding, as reduced levels of
0.001; Fig. 5). This difference also correlated with shorter time soluble cMet were detected in supernatant of cells overexpressing
interval between detection of primary tumor and liver metastases TIMP-1/N-TIMP-1 compared with the control (both P V 0.033;
[26.83 F 5.81 months (n = 6) versus 13.75 F 2.62 months (n = 8); Fig. 6D). The elevated level of cell-associated cMet in cells
P = 0.049]. overexpressing TIMP-1/N-TIMP-1 correlated with increased phos-
Inhibition of cMet shedding by TIMP-1. Interestingly, phorylation of cMet (Supplementary Fig. S3).
although cMet protein was increased in livers with elevated Activity of ADAM-10 regulates cMet shedding. Knockdown of
TIMP-1 (Fig. 2C), mRNA expression of cMet was not up-regulated ADAM-10 mRNA expression in NIH3T3 cells by 88% (Fig. 6A) led to
in these livers, neither at the time point of tumor cell inoculation a significant increase of membrane-bound cMet protein compared
(Fig. 5) nor in metastasis-bearing livers (Supplementary Fig. S2). with the control (P = 0.018; Fig. 6B), whereas cMet mRNA expres-
Reduced shedding of cMet by TIMP-1–induced inhibition of sion was not enhanced (P = 0.2; Fig. 6C). ADAM-10 knockdown was
metalloproteinase activity may account for the accumulation of verified on protein level (Supplementary Fig. S4). A significant
cMet protein, but it was thus far unknown that TIMP-1 can inhibit reduction of shed cMet in the supernatant of cells with reduced
its shedding. As this hypothesis cannot be addressed in vivo, we ADAM-10 expression was detected (P = 0.021; Fig. 6D), indicating
chose a murine nontumorigenic cell line expressing inhibitable that depletion of ADAM-10 was sufficient to accumulate cMet on
metalloproteinases as well as cMet, thus providing all prerequisites the cell surface. In addition, suppression of ADAM-10 mRNA
for a proof-of-concept study. Overexpression of either TIMP-1 or expression significantly increased cMet signaling (Supplementary
N-TIMP-1 in NIH3T3 cells (Fig. 6A) resulted in significantly Fig. S3).
augmented membrane-bound cMet protein in these cells com- Together, these data suggest a molecular pathway regulated by
pared with the virus control (both P V 0.026; Fig. 6B), although RNA TIMP-1 that could lead to a microenvironment with higher
expression of cMet was not increased (both P > 0.33; Fig. 6C). susceptibility to metastases.

Cancer Res 2007; 67: (18). September 15, 2007 8620 www.aacrjournals.org
TIMP-1 Promotes Liver Metastasis via HGF Signaling

Discussion of TIMP-1 (i.e., induction of scattered invasion of tumor cells in


This study shows that host-derived TIMP-1 can promote liver liver parenchyma). The bulk of these micrometastases cannot
derive from augmented extravasation of the L-CI.5s cells because
metastasis by induction of HGF signaling. Until recently, the dominant
the number of the proliferative active and scattered micro-
concept of natural inhibitors of tumor-associated proteases focused
metastases by far exceeded the number of inoculated tumor cells.
on their capacity to repress activity of ECM remodeling enzymes,
Previous studies have reported that, even when metalloproteinases
such as metalloproteinases. This view has undergone revision based
are required for metastasis in a particular tumor model, TIMP-1
on increased awareness of ‘‘degradome’’ functions of metalloprotei-
does not always suppress metastasis (9, 37), supporting the idea
nases and the tumor microenvironment and recognition of their
that TIMP-1 possesses a broad range of biological activities, some
participation during many stages of tumor progression (3). of which may be independent of its metalloproteinase-inhibitory
The original hypothesis that TIMP-1 overexpression would yield function (11, 38–40). Those variations of antitumorigenic efficacy of
a reduction in the number of successfully extravasated tumor cells TIMP-1 have been suggested to depend on the concentrations of
into a target organ was based on numerous reports on the TIMP-1 (10, 41) or related to the proproliferative and antiapoptotic
importance of MMPs for metastasis in many tumor models (36). In features of TIMP-1 (11, 12, 38).
agreement with these and our previous studies indicating the The new aspect is that we found a proinvasive feature of TIMP-1
importance of gelatinase inhibition for formation of macrometa- when elevated in the liver based on TIMP-1–induced stimulation
stases of the lymphoma model (28, 29), elevated host levels of of HGF signaling and downstream expression of metastasis-
TIMP-1 prevented macrometastases only when it was already promoting genes. The N-terminal domain of TIMP-1, which includes
increased at the time point of tumor cell inoculation. This confirms the metalloproteinase-inhibitory domain, was sufficient to exhibit
the already known effect of metalloproteinase inhibition by TIMP-1 this effect, indicating that broad-spectrum inhibition of metal-
on the level of extravasation. loproteinases is responsible for this scattering. In agreement with
In contrast to this inhibitory action on initial invasion, we reveal this notion, treatment with a synthetic broad-spectrum metal-
here for the first time an unexpected metastasis-promoting feature loproteinase inhibitor also leads to liver-specific promotion of

Figure 5. Common TIMP-1–associated alterations in gene expression in mouse and human. Mice were transduced with either control virus (Addl70-3) or AdTIMP-1.
Three days later, mice were sacrificed and livers were removed. Normal liver samples of patients with metastatic colorectal cancer were obtained at least 2 cm distant
from colorectal liver metastases during curative surgical interventions. Total RNA was isolated from murine and human liver samples and gene expression was
assessed by quantitative RT-PCR. Human samples were initially tested for TIMP-1 expression and then divided into two subgroups according to their TIMP-1
expression levels (low TIMP-1: 21.22 F 1.89, n = 6; high TIMP-1: 98.92 F 19.84, n = 8; P < 0.001). All samples were tested for uPA, uPAR, PAI-1, tPA, matriptase,
PCNA, MMP-2, MMP-9, ADAM-10, cathepsin G (Cath G), neutrophil elastase (NE ), and Met mRNA expression. Columns, mean of gene expression in murine
[Addl70-3 (n = 4) versus AdTIMP-1 (n = 3): uPA: 100 F 16.3% versus 193.3 F 20.3%; uPAR: 100 F 15.3% versus 557.9 F 63.2%; PAI-1: 100 F 21.2% versus
5,083.0 F 3,052.9%; tPA: 100 F 18.4% versus 1,673.8 F 465.2%; matriptase: 100 F 4.4% versus 451.5 F 33.8%; PCNA: 100 F 82.7% versus 554.6 F 80.3%; MMP-9:
100 F 47.2% versus 494.1 F 34.6%; MMP-2: 100 F 45.9% versus 289.4 F 63.2%; ADAM-10: 100 F 35.7% versus 542.8 F 397.4%; cathepsin G: 100 F 52.6%
versus 2,040.0 F 721.1%; neutrophil elastase: 100 F 66.0% versus 1,024.3 F 541.1%; Met: 100 F 9.3% versus 109.6 F 8.9%] and human (low TIMP-1 versus high
TIMP-1: uPA: 100 F 16.6% versus 615.5 F 61.2%; uPAR: 100 F 20.7% versus 691.6 F 131.1%; PAI-1: 100 F 15.7% versus 107.1 F 182%; tPA: 100 F 10.8%
versus 116.9 F 18.6%; matriptase: 100 F 18.1% versus 213.0 F 24.4%; PCNA: 100 F 24.3% versus 231.7 F 50.7%; MMP-9: 100 F 29.5% versus 2,418.6 F 396.4%;
MMP-2: 100 F 30.6% versus 695.3 F 590.9%; ADAM-10: 100 F 11.1% versus 150.6 F 33.9%; cathepsin G: 100 F 31.9% versus 379.5 F 107.8%; neutrophil
elastase: 100 F 75.5% versus 1621.6 F 339.2%; Met: 100 F 17.0% versus 111.5 F 27.6%) livers; bars, SE. *, P < 0.05.

www.aacrjournals.org 8621 Cancer Res 2007; 67: (18). September 15, 2007
Cancer Research

metastasis in several xenograft models as well as in the syngeneic niche than in the liver parenchyma. However, several studies
T-cell lymphoma model (42, 43). In addition, in the present study, revealed that inhibition or overexpression of proteases or
promotion of liver metastasis was found, whereas lung metastasis protease families evolves different effects on metastasis to liver
was significantly reduced. Either TIMP-1 was not able to induce versus lung (26, 42–44). This leads to the hypothesis that
HGF signaling in the lung or induction of HGF signaling in the lung metastasis to the liver requires a different repertoire of proteases
tissue has different effects on the formation of a premetastatic than metastasis to the lung and that the different microenviron-
ments present in these organs react differentially on changed
proteolysis. The underlying mechanism of the organ specificity of
the prometastatic effect of TIMP-1 will need to be determined in
future studies.
Upon elevation of TIMP-1, we observed augmented HGF as well as
cMet protein in livers. Simultaneous increase of HGF and functional
cell-associated cMet can explain the overall increase of HGF/cMet
signaling. Elevated presence of the active form of HGF, which is
required for signaling (15), can derive from the observed up-
regulation of the HGF-activating proteases uPA and matriptase
(45, 46) in the liver. Active matriptase and uPA (also activated by
matriptase; ref. 46) can lead to an additional increase of proteolytic
activity by initiating a protease activation cascade, including the
activation of plasminogen by uPA and of some pro-MMPs by active
plasmin (47). Elevated levels of TIMP-1 in the liver also increased
expression of cathepsin G, also a gelatinolytic enzyme (48), as well as
expression of neutrophil elastase, which also enhances plasminogen
activation (49). Such proteases are likely the source of the increased
nonmetalloproteinase-derived proteolytic activity observed in me-
tastasis-bearing liver sections with augmented TIMP-1 levels in situ.
Interestingly, the TIMP-1–provoked gene expression signature of the
experimental model resembled in some instances (matriptase, uPA,
uPAR, PCNA, MMP-9, and cathepsin G) the signature observed in
liver samples of colorectal cancers in patients with elevated levels of
TIMP-1, which is a hint on general molecular mechanisms provoked
by elevated stromal TIMP-1. In the future, stable knockdown of
cMet in host and/or tumor cells will further characterize the role of
TIMP-1–induced cMet signaling in liver metastasis.
A link between metalloproteinase inhibition and enhancement
of HGF/cMet signaling was revealed by an in vitro biochemical
assay: Elevated levels of TIMP-1/N-TIMP-1 preserved activatable
cMet on cell surfaces by reduced cMet shedding. Interestingly,
suppression of ADAM-10 also prevented shedding of cMet,
indicating that ADAM-10 is involved in the regulation of cMet
shedding. Sheddases are already known for their capacity to
regulate cell surface–associated tyrosine kinase receptors (50).
These data indicate that preservation of cMet is induced by TIMP-1

Figure 6. Preservation of cell-associated cMet by either TIMP-1 overexpression


or ADAM-10 knockdown. NIH3T3 cells were infected either with Addl70-3
(NIH/Addl70-3 ), AdTIMP-1 (NIH/AdTIMP-1 ), AdN-TIMP-1 (NIH/AdN-TIMP-1 all
adenoviruses with a multiplicity of infection of 100), with a retroviral vector coding
for shRNAi targeting murine ADAM-10 (NIH-shADAM-10 ), or with a retroviral
vector encoding scrambled shRNAi (NIH-scr ). A, quantitative RT-PCR of
human TIMP-1 (huTIMP-1 ; left ) and murine ADAM-10 (muADAM-10 ; right ),
respectively, verified overexpression of TIMP-1/N-TIMP-1 and suppression of
ADAM-10. Columns, mean of relative huTIMP-1 and muADAM-10 mRNA,
respectively, versus 18S RNA; bars, SE. B, membrane preparations of
transduced cells were tested for the presence of cMet by ELISA. Elevated levels
of either TIMP-1 variant (left ) or reduced levels of ADAM-10 (right ) led to
increased membrane-bound cMet protein compared with the controls. C, murine
cMet (mu-cMet ) expression in cells with either increased expression of TIMP-1/
N-TIMP-1 (left ) or reduced expression of ADAM-10 (right ), respectively, was
assessed by quantitative RT-PCR. Columns, mean of murine cMet mRNA
versus 18S RNA; bars, SE. D, quantification of soluble cMet. FCS-free culture
medium of transduced NIH3T3 cells was tested by cMet ELISA. Columns, mean
of soluble cMet in culture medium; bars, SE. ELISAs were done in triplicates.
Results of three independent experiments.

Cancer Res 2007; 67: (18). September 15, 2007 8622 www.aacrjournals.org
TIMP-1 Promotes Liver Metastasis via HGF Signaling

or N-TIMP-1 and may be a consequence of direct inhibition of penultimate step of cancer evolution and are one explanation for
ADAM-10 and thereby loss of its sheddase activity (4) or indirectly the association between tumor aggressiveness and increased levels
by inhibition of ADAM-10 activation. of TIMP-1 in cancer patients.
Many tumor cell lines respond to elevated levels of HGF (scatter
factor) with increased invasive behavior (15, 16). Indeed, the altered Acknowledgments
host environment fed back to the inoculated tumor cell lines,
Received 1/18/2007; revised 6/22/2007; accepted 7/11/2007.
which expressed functional cMet (Supplementary Fig. S5). Thus, Grant support: Deutsche Forschungsgemeinschaft grant SFB 469 (A. Krüger),
increased scattering of tumor cells in the liver parenchyma seems European Union Framework Programme 6, project LSHC-CT-2003-503297, Cancer-
degradome (A. Krüger, K. Brand, and D.R. Edwards), and NIH grant AR40994 (K. Brew
to be based on a reaction of the tumor cells to the TIMP-1– and H. Nagase).
provoked change of homeostasis in the host. Identification of this The costs of publication of this article were defrayed in part by the payment of page
TIMP-1–provoked prometastatic niche reveals a novel role for charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
TIMP-1 during cancer progression and illuminates the intricacy of We thank Dr. Susanne Schaten and Katja Honert (both from Technische Universität
protease and protease inhibitor networks that regulate the München) for expert technical support.

References 18. Kuroi K, Toi M. Circulating angiogenesis regulators 34. Barrett AJ, Kembhavi AA, Brown MA, et al. L-trans-
in cancer patients. Int J Biol Markers 2001;16:5–26. Epoxysuccinyl-leucylamido(4-guanidino)butane (E-64) and
1. Overall CM, Lopez-Otin C. Strategies for MMP 19. Hsiao LT, Lin JT, Yu IT, et al. High serum hepatocyte its analogues as inhibitors of cysteine proteinases including
inhibition in cancer: innovations for the post-trial era. growth factor level in patients with non-Hodgkin’s cathepsins B, H and L. Biochem J 1982;201:189–98.
Nat Rev Cancer 2002;2:657–72. lymphoma. Eur J Haematol 2003;70:282–9. 35. Fritz H, Wunderer G. Biochemistry and applications
2. Overall CM, Kleifeld O. Tumour microenvironment— 20. Hitt M, Bett AJ, Addison CL, Prevec L, Graham FL. of aprotinin, the kallikrein inhibitor from bovine organs.
opinion: validating matrix metalloproteinases as drug Techniques for human adenovirus vector construction Arzneimittelforschung 1983;33:479–94.
targets and anti-targets for cancer therapy. Nat Rev and characterization. In: Adolph KW, editor. Viral gene 36. Coussens LM, Werb Z. Matrix metalloproteinases and
Cancer 2006;6:227–39. techniques. San Diego: Academic Press; 1995. p. 13–30. the development of cancer. Chem Biol 1996;3:895–904.
3. Egeblad M, Werb Z. New functions for the matrix 21. Schweinitz A, Steinmetzer T, Banke IJ, et al. Design of 37. Soloway PD, Alexander CM, Werb Z, Jaenisch R.
metalloproteinases in cancer progression. Nat Rev novel and selective inhibitors of urokinase-type plas- Targeted mutagenesis of Timp-1 reveals that lung tumor
Cancer 2002;2:161–74. minogen activator with improved pharmacokinetic invasion is influenced by Timp-1 genotype of the tumor
4. Amour A, Knight CG, Webster A, et al. The in vitro properties for use as antimetastatic agents. J Biol Chem but not by that of the host. Oncogene 1996;13:2307–14.
activity of ADAM-10 is inhibited by TIMP-1 and TIMP-3. 2004;279:33613–22. 38. Chesler L, Golde DW, Bersch N, Johnson MD.
FEBS Lett 2000;473:275–9. 22. Krüger A, Schirrmacher V, von Hoegen P. Scattered Metalloproteinase inhibition and erythroid potentiation
5. Chirco R, Liu XW, Jung KK, Kim HR. Novel functions of micrometastases visualized at the single-cell level: are independent activities of tissue inhibitor of metal-
TIMPs in cell signaling. Cancer Metastasis Rev 2006;25: detection and re-isolation of lacZ-labeled metastasized loproteinases-1. Blood 1995;86:4506–15.
99–113. lymphoma cells. Int J Cancer 1994;58:275–84. 39. Porter JF, Shen S, Denhardt DT. Tissue inhibitor of
6. Elezkurtaj S, Kopitz C, Baker AH, et al. Adenovirus- 23. Soneoka Y, Cannon PM, Ramsdale EE, et al. A metalloproteinase-1 stimulates proliferation of human
mediated overexpression of tissue inhibitor of metal- transient three-plasmid expression system for the cancer cells by inhibiting a metalloproteinase. Br J
loproteinases-1 in the liver: efficient protection against production of high titer retroviral vectors. Nucleic Acids Cancer 2004;90:463–70.
T-cell lymphoma and colon carcinoma metastasis. Res 1995;23:628–33. 40. Kong Y, Poon R, Nadesan P, et al. Matrix metal-
J Gene Med 2004;6:1228–37. 24. Wei S, Chen Y, Chung L, Nagase H, Brew K. Protein loproteinase activity modulates tumor size, cell motility,
7. Brand K. Cancer gene therapy with tissue inhibitors of engineering of the tissue inhibitor of metalloproteinase and cell invasiveness in murine aggressive fibromatosis.
metalloproteinases (TIMPs). Curr Gene Ther 2002;2: 1 (TIMP-1) inhibitory domain. In search of selective Cancer Res 2004;64:5795–803.
255–71. matrix metalloproteinase inhibitors. J Biol Chem 2003; 41. Yamauchi K, Ogata Y, Nagase H, Shirouzu K. Inhibition
8. Hammer JH, Basse L, Svendsen MN, et al. Impact of 278:9831–4. of liver metastasis from orthotopically implanted colon
elective resection on plasma TIMP-1 levels in patients 25. Parkes R, Meng QH, Siapati KE, McEwan JR, cancer in nude mice by transfection of the TIMP-1 gene
with colon cancer. Colorectal Dis 2006;8:168–72. Hart SL. High efficiency transfection of porcine vascular into KM12SM cells. Surg Today 2001;31:791–8.
9. Krüger A, Fata JE, Khokha R. Altered tumor growth cells in vitro with a synthetic vector system. J Gene Med 42. Krüger A, Soeltl R, Sopov I, et al. Hydroxamate-type
and metastasis of a T-cell lymphoma in Timp-1 2002;4:292–9. matrix metalloproteinase inhibitor batimastat promotes
transgenic mice. Blood 1997;90:1993–2000. 26. Kopitz C, Anton M, Gansbacher B, Krüger A. liver metastasis. Cancer Res 2001;61:1272–5.
10. Goss KJ, Brown PD, Matrisian LM. Differing effects of Reduction of experimental human fibrosarcoma lung 43. Della Porta P, Soeltl R, Krell HW, et al. Combined
endogenous and synthetic inhibitors of metalloproteinases metastasis in mice by adenovirus-mediated cystatin C treatment with serine protease inhibitor aprotinin and
on intestinal tumorigenesis. Int J Cancer 1998;78:629–35. overexpression in the host. Cancer Res 2005;65:8608–12. matrix metalloproteinase inhibitor batimastat (BB-94) does
11. Rhee JS, Diaz R, Korets L, Hodgson JG, Coussens LM. 27. Jones N, Shenk T. Isolation of deletion and substitu- not prevent invasion of human esophageal and ovarian
TIMP-1 alters susceptibility to carcinogenesis. Cancer tion mutants of adenovirus type 5. Cell 1978;13:181–8. carcinoma cells in vivo. Anticancer Res 1999;19:3809–16.
Res 2004;64:952–61. 28. Arlt M, Kopitz C, Pennington C, et al. Increase in 44. Tester AM, Waltham M, Oh SJ, et al. Pro-matrix
12. Guedez L, McMarlin AJ, Kingma DW, Bennett TA, gelatinase-specificity of matrix metalloproteinase inhib- metalloproteinase-2 transfection increases orthotopic
Stetler-Stevenson M, Stetler-Stevenson WG. Tissue itors correlates with antimetastatic efficacy in a T-cell primary growth and experimental metastasis of MDA-
inhibitor of metalloproteinase-1 alters the tumorigenic- lymphoma model. Cancer Res 2002;62:5543–50. MB-231 human breast cancer cells in nude mice. Cancer
ity of Burkitt’s lymphoma via divergent effects on tumor 29. Krüger A, Arlt MJ, Gerg M, et al. Antimetastatic Res 2004;64:652–8.
growth and angiogenesis. Am J Pathol 2001;158:1207–15. activity of a novel mechanism-based gelatinase inhibi- 45. Mars WM, Zarnegar R, Michalopoulos GK. Activation
13. Wurtz SO, Schrohl AS, Sorensen NM, et al. Tissue tor. Cancer Res 2005;65:3523–6. of hepatocyte growth factor by the plasminogen
inhibitor of metalloproteinases-1 in breast cancer. 30. Banke IJ, Arlt MJ, Pennington C, et al. Increase of activators uPA and tPA. Am J Pathol 1993;143:949–58.
Endocr Relat Cancer 2005;12:215–27. anti-metastatic efficacy by selectivity- but not affinity- 46. Lee SL, Dickson RB, Lin CY. Activation of hepatocyte
14. Trusolino L, Comoglio PM. Scatter-factor and optimization of synthetic serine protease inhibitors. Biol growth factor and urokinase/plasminogen activator by
semaphorin receptors: cell signalling for invasive Chem 2003;384:1515–25. matriptase, an epithelial membrane serine protease.
growth. Nat Rev Cancer 2002;2:289–300. 31. Palosaari H, Pennington CJ, Larmas M, Edwards DR, J Biol Chem 2000;275:36720–5.
15. Jiang WG, Martin TA, Parr C, Davies G, Matsumoto K, Tjaderhane L, Salo T. Expression profile of matrix 47. Reuning U, Magdolen V, Wilhelm O, et al. Multifunc-
Nakamura T. Hepatocyte growth factor, its receptor, and metalloproteinases (MMPs) and tissue inhibitors of tional potential of the plasminogen activation system in
their potential value in cancer therapies. Crit Rev Oncol MMPs in mature human odontoblasts and pulp tissue. tumor invasion and metastasis [review]. Int J Oncol
Hematol 2005;53:35–69. Eur J Oral Sci 2003;111:117–27. 1998;13:893–906.
16. Fujiuchi Y, Nagakawa O, Murakami K, Fuse H, 32. Fabian MA, Biggs WH III, Treiber DK, et al. A small 48. Capodici C, Berg RA. Cathepsin G degrades dena-
Saiki I. Effect of hepatocyte growth factor on invasion molecule-kinase interaction map for clinical kinase tured collagen. Inflammation 1989;13:137–45.
of prostate cancer cell lines. Oncol Rep 2003;10:1001–6. inhibitors. Nat Biotechnol 2005;23:329–36. 49. Machovich R, Owen WG. An elastase-dependent
17. Tacchini L, Matteucci E, De Ponti C, Desiderio MA. 33. Yee JK, Miyanohara A, LaPorte P, Bouic K, Burns JC, pathway of plasminogen activation. Biochemistry 1989;
Hepatocyte growth factor signaling regulates trans- Friedmann T. A general method for the generation of 28:4517–22.
activation of genes belonging to the plasminogen high-titer, pantropic retroviral vectors: highly efficient 50. Blobel CP. ADAMs: key components in EGFR
activation system via hypoxia inducible factor-1. Exp infection of primary hepatocytes. Proc Natl Acad Sci signalling and development. Nat Rev Mol Cell Biol
Cell Res 2003;290:391–401. U S A 1994;91:9564–8. 2005;6:32–43.

www.aacrjournals.org 8623 Cancer Res 2007; 67: (18). September 15, 2007

You might also like