You are on page 1of 7

Comparative Effectiveness of Two Oil AdjuvantInactivated Avian Influenza

H9N2 Vaccines
Author(s): Walid H. Kilany, Abdel-Hamid I. Bazid, Ahmed Ali, Ayman H. El-Deeb, Mohamed A. Zain
El-Abideen, Magdy El Sayed and Magdy F. El-Kady
Source: Avian Diseases, 60(1s):226-231.
Published By: American Association of Avian Pathologists
DOI: http://dx.doi.org/10.1637/11145-050815-Reg
URL: http://www.bioone.org/doi/full/10.1637/11145-050815-Reg

BioOne (www.bioone.org) is a nonprofit, online aggregation of core research in the biological, ecological, and
environmental sciences. BioOne provides a sustainable online platform for over 170 journals and books published
by nonprofit societies, associations, museums, institutions, and presses.
Your use of this PDF, the BioOne Web site, and all posted and associated content indicates your acceptance of
BioOnes Terms of Use, available at www.bioone.org/page/terms_of_use.
Usage of BioOne content is strictly limited to personal, educational, and non-commercial use. Commercial
inquiries or rights and permissions requests should be directed to the individual publisher as copyright holder.

BioOne sees sustainable scholarly publishing as an inherently collaborative enterprise connecting authors, nonprofit publishers, academic institutions, research
libraries, and research funders in the common goal of maximizing access to critical research.
AVIAN DISEASES 60:226231, 2016

Comparative Effectiveness of Two Oil AdjuvantInactivated Avian Influenza


H9N2 Vaccines
Walid H. Kilany,AF Abdel-Hamid I. Bazid,B Ahmed Ali,C Ayman H. El-Deeb,D Mohamed A. Zain El-Abideen,AE
Magdy El Sayed,E and Magdy F. El-KadyC
A
Reference Laboratory for Veterinary Quality Control on Poultry Production (RLQP), Animal Health Research Institute, P.O. Box, 264, Dokki,
Giza 12618, Egypt
B
Virology Department, Faculty of Veterinary Medicine, Sadat City University, Menoufia 32897, Egypt
C
Poultry Diseases Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef 65211, Egypt
D
Virology Department, Faculty of Veterinary Medicine, Cairo University, Giza 12511, Egypt
E
Middle East for Veterinary Vaccine Company, Second Industrial Area, El-Salhya El-Gededa, El-Sharqia 44671, Egypt
Received 13 May 2015; Accepted 5 February 2016; Published ahead of print 8 February 2016

SUMMARY. Low pathogenic avian influenza H9N2 virus infection has been an important risk to the Egyptian poultry industry
since 2011. Economic losses have occurred from early infection and co-infection with other pathogens. Therefore, H9N2
vaccination of broiler chicks as young as 7 days old was recommended. The current inactivated H9N2 vaccines (0.5 ml/bird)
administered at a reduced dose (0.25 ml/bird) do not guarantee the delivery of an effective dose for broilers. In this study, the
efficacy of the reduced-dose volume (0.3 ml/bird), compared with the regular vaccine dose (0.5 ml/bird) of inactivated H9N2
vaccines using two different commercially available adjuvants, was investigated. The vaccines were prepared from the local H9N2
virus (Ck/EG/114940v/NLQP/11) using the same antigen content: 300 hemagglutinating units. Postvaccination (PV) immune
response was monitored using the hemagglutination inhibition test. At 4 wk PV, both vaccinated groups were challenged using
the homologous H9N2 strain at a 50% egg infective dose (EID50) of 106 EID50/bird via the intranasal route. Clinical signs,
mortality, and virus shedding in oropharyngeal swabs were monitored at 2, 4, 6, and 10 days postchallenge (DPC). The reduced-
dose volume of vaccine induced a significantly faster and higher immune response than the regular volume of vaccine at 2 and 3
wk PV. No significant difference in virus shedding between the two vaccine formulas was found (P $ 0.05), and both vaccines
were able to stop virus shedding by 6 DPC. The reduced-dose volume of vaccine using a suitable oil adjuvant and proper antigen
content can be used effectively for early immunization of broiler chicks.

RESUMEN. Eficacia comparativa de dos vacunas contra la influenza aviar H9N2 inactivadas y emulsionadas en aceite.
La infeccin por el virus de la influenza aviar de baja patogenicidad H9N2 ha sido un riesgo importante para la industria avcola
egipcia desde el ao 2011. Las prdidas econmicas se han producido por esta infeccin y por la coinfeccin con otros patgenos.
Por lo tanto, se recomienda la vacunacin contra este virus H9N2 en pollos de engorde tan temprano como a los siete das de edad.
Las vacunas inactivadas actuales H9N2 (0.5 ml/ave) administradas con una dosis reducida (0.25 ml/ave) no garantizan la aplicacin
de una dosis eficaz para pollos de engorde. En este estudio se investig la eficacia del volumen reducido de la dosis (0.3 ml/ave), en
comparacin con la dosis de vacuna regular (0.5 ml/ave) de las vacunas inactivadas H9N2 utilizando dos adyuvantes diferentes
disponibles en el mercado. Las vacunas se prepararon a partir del virus H9N2 local (Pollo/EG/114940v/NLQP/11), utilizando el
mismo contenido de antgeno: 300 unidades hemaglutinantes. Despus de la vacunacin, la respuesta inmune se determin
mediante la prueba de inhibicin de la hemaglutinacin. A las cuatro semanas despus de la vacunacin, ambos grupos vacunados
fueron desafiados con la cepa H9N2 homloga con una dosis de 106 dosis infectantes para embrin de pollo (EID50) por ave,
a travs de la ruta intranasal. Los signos clnicos, la mortalidad y la diseminacin del virus mediante hisopos orofarngeos se
determinaron a los dos, cuatro, seis y diez das despus del desafo. La vacuna con volumen reducido de dosis indujo una respuesta
inmune significativamente ms rpida y ms alta que el volumen regular de la vacuna a las dos y tres semanas despus de la
vacunacin. No se encontraron diferencias significativas en la diseminacin del virus entre las dos frmulas de vacunas (P $ 0.05) y
ambas vacunas fueron capaces de detener la propagacin del virus por el da seis despus del desafo. Se puede utilizar eficazmente el
volumen de la dosis reducida de la vacuna para la inmunizacin temprana de pollos de engorde usando el adyuvante oleoso y el
contenido de antgeno adecuados.
Key words: AIV, LPAI H9N2, SPF chickens, inactivated vaccine
Abbreviations: AI 5 avian influenza; AIV 5 AI virus; DPC 5 days post challenge; EID50 5 50% egg infective dose;
G1 5 genotype 1; HA 5 hemagglutinin; HI 5 hemagglutination inhibition; HPAI 5 highly pathogenic avian influenza; IL 5 inter-
leukin; ISA70 5 Montanide ISA 70 VG adjuvant; ISA71 5 Montanide ISA 71 VG adjuvant; LPAI 5 low pathogenic avian influ-
enza; NA 5 neuraminidase; PV 5 postvaccination; RT-PCR 5 reverse transcriptase; PCR; SPF 5 specific pathogen free

Avian influenza virus (AIV) is one of the most devastating viral dis- the internal proteinsprincipally nucleoproteins and the M1 matrix
eases in the poultry industry and has a worldwide distribution. AIV protein. All AIVs are type A. There are two surface glycoproteins:
infects many species of domesticated and wild birds (23). AIV is an hemagglutinin (HA) and neuraminidase (NA). AIVs are classified
enveloped virus that belongs to the Orthomyxoviridae family and has into subtypes according to the combination of 18 HA and 11 NA
an eight-segmented single-stranded negative sense RNA genome. molecules (27). AIVs are classified into two distinct pathogenic
AIVs are categorized into three distinct types, A, B, and C, based on groups: highly pathogenic avian influenza (HPAI) and low pathogenic
avian influenza (LPAI) (24). LPAI H9N2 viruses isolated from poultry
F
Corresponding author. E-mail: walidhamdy78@yahoo.com in Middle Eastern countries give rise, in these countries, to four

226
Inactivated H9N2 vaccine dose reduction 227
distinct genetic groups: A, B, C, and D. All four groups belong to the SPF eggs; Koom Oshiem, Fayoum, Egypt) and was confirmed to be free
lineage (14). of other biological contaminants, including avian influenza H5N1, avian
The LPAI H9N2 virus was first reported in Egypt at a commercial leukosis subgroup J, infectious bursal disease virus, avian reovirus, infec-
quail farm (10). The current endemic situation of both LPAI H9N2 tious bronchitis virus, Newcastle disease virus, chicken infectious anemia,
and HPAI H5N1 viruses in commercial chicken farms in Egypt have avian encephalomyelitis virus, adenoviruses, egg drop syndrome (EDS76)
virus, avian mycoplasma, and Salmonella spp.
raised concern of the potential appearance of a reassortment virus and
Reverse transcriptase PCR and HA gene sequencing. The viral RNA
increased human zoonotic risk (2,16). Further economic losses in the
was extracted from harvested allantoic fluids, by the BiofluxH viral RNA Mini
poultry industry ensued when coinfection of LPAI H9N2 occurred Spin column kit (BioFlux, Hangzhou China), following the manufacturers
with other respiratory infections (9), causing increased severity and instructions. Reverse transcriptase (RT)-PCR was used for HA gene amplifi-
high mortality rates in Egyptian broiler chickens. cation using specific oligonucleotide primers (22). The final reaction volume
In addition to biosecurity measures, the practice of vaccination is was 25 ml; including a 7-ml RNA template; 5 ml of 56 RT-PCR enhancer;
an important tool in the prevention and control of avian influenza 2.5 ml of 106 RT-PCR buffer; 1.5 ml of each forward and reverse primers;
infection in chickens. Several experimental studies have demonstrated 1.25 ml of hot master Taq polymerase; 1 ml of Super pure dNTPs; 0.25 ml
that inactivated avian influenza (AI) vaccines are capable of inducing of RNasin; 0.25 ml of Quant RTase; and 4.75 ml of RNase-free water. Ther-
antibody response, which aids in the protection of the infected birds mocycling RT-PCR conditions were 50 for 15 min, then 95 for 15 min,
(5,6). Because of the widespread nature of the LPAI H9N2 viruses followed by 35 cycles at 95 for 15 sec, then another 30 sec at 47 and 1
and their zoonotic potential, vaccination of susceptible birds with min at 65 . Final extension was performed at 65 for 10 min. Amplified
RT-PCR products were purified using the PCR purification KitH (Thermo
inactivated oil emulsion vaccine was employed to control the disease
Scientific, Waltham, MA), following the manufacturers instructions and
(19). Despite improper practices during the administration of the were then sent for sequencing by Macrogen Inc. (the Republic of Korea).
H9N2 vaccine in the small broiler poultry sector (1), and despite pos- A BLAST search was conducted (http://www.ncbi.nlm.nih.gov/BLAST).
sible coinfection with other poultry viruses (e.g., infectious bronchitis Sequence comparisons and phylogenetic relationships through a bootstrap
virus) (9), only minor genetic changes were observed as detected by of 1000 trials were determined with the MEGA version 6 program using
HA gene analysis of recently isolated viruses (9,17). the Clustal W alignment algorithm (25). Additionally, nucleotide and amino
Limited literature is available on the efficacy of H9N2 inactivated acid identities were determined using GeneiousH 7.1.3 (Biomatters Ltd.,
oil emulsion commercial vaccines being used in Egypt. Full-dose AI New Zealand).
H9N2 vaccination in broiler chicks was recommended to be adminis- Virus inactivation and vaccine formulation. The LPAI H9N2 virus
tered as early as 7 days of agewith a booster doseto obtain high harvest inactivation was conducted using 0.2% formalin at 37 C for 24 hr
and homogeneous antibody titers (3). Yet, the use of the full dose of (Sigma, St. Louis, MO). Two different oil emulsioninactivated vaccines
0.5 ml/bird is not applicable to young broiler chicks; on the other were used in this experiment. The first was designated Vaccine A, contain-
hand, the reduction of dose volume without considering the antigen ing the local (H9N2) isolate at a concentration of 300 hemagglutinating
units per dose and blended with Montanide ISA 70 VG adjuvant
load may hinder obtaining the required protection. The suggested (ISA70) at a dose of 0.5 ml/bird. The second vaccine was designated Vac-
alternative is to reduce the vaccine dose volume while maintaining cine B, containing the same antigen load but blended with Montanide
antigen dose and using efficacious immune stimulant vaccine adju- ISA 71 VG adjuvant (ISA71) at a dose of 0.3 ml/bird. The antigen aque-
vants. The MontanideTM (SEPPIC, Paris, France) adjuvants have ous phase was pre-emulsified with either ISA70 or ISA71 at a 30:70 ratio
been widely used in veterinary applications. Montanide ISA 70 and (w/w) at 1000 rpm using the Silverson L5M high shear laboratory mixer
ISA 71 VG are mineral oilbased adjuvants that have been proven (Silverson Machines Inc., Buckinghamshire, UK). The speed of the mixer
to improve cellular response, with only minimal side effects associated was then increased to 3000 rpm and maintained for 30 min for the emul-
with other mineral oil emulsions (8). Studies on immunization with sification of 10,000 doses of the vaccine. During the emulsion formula-
low doses of Eimeria profilin plus the ISA 70 VG or ISA 71 VG sub- tion, the temperature of the mixture was maintained between 18 and 22
unit vaccine showed that Eimeria profilin plus ISA 71 VG reduced C. The three different vaccine batches were inactivated H9N2 ISA70 vac-
fecal oocyst shedding and enhanced serum antibody levels and gene cine batches (A1, A2, and A3) and inactivated H9N2 ISA71 vaccine
batches (B1, B2, and B3).
transcripts encoding interleukin (IL)-2, IL-10, IL-17A, and interfer-
Birds and experimental design. Seventy SPF chickens 2 wk of age
on- in intestinal intraepithelial lymphocytes. Such studies also
were randomly divided into seven groups, including six experimental
showed increased infiltration of lymphocytes, especially CD8+ lym- and one control group (10 chicks/group; Table 1). Each group was reared
phocytes, at the site of immunization in chickens (12). separately in chicken isolators at the Laboratory Animal Research Unit of
The present work was conducted to study the efficacy of dose vol- ME VACH Company (El-Sharqia, Egypt). Feed and water were available
ume reduction (from 0.5 to 0.3 ml/dose) of inactivated H9N2 ad libitum. At 2 wk of age, birds in Groups A1A3 were vaccinated with
vaccines prepared from the A/CK/Eg/114940v/NLQP/2011(H9N2) the H9N2 AI vaccine A (0.5 ml/bird) via the intramuscular route in the
virus, utilizing the Montanide ISA 71 adjuvants, and compare this thigh muscle. The birds in Groups B1B3 were vaccinated with H9N2
with the alternate utilization of Montanide ISA 70 adjuvants. Com- vaccine B by the same route, but at a dose of 0.3 ml/bird. Finally, the
parisons were made in terms of immunity and reduction of viral unvaccinated birds (control Group 7) were inoculated with an equal vol-
replication and shedding in broiler chickens challenged with ume of placebo vaccine (normal saline). Sera were collected on a weekly
LPAI H9N2. basis, for 4 wk postvaccination (PV) and at 14 days postchallenge (DPC).
Hemagglutination inhibition test. The hemagglutination inhibition
(HI) test was carried out according to the World Organization for Animal
MATERIALS AND METHODS Health Manual (20) to monitor the postvaccination humoral immune
response for each vaccine batch using the homologous HA antigen. Brief-
Virus. The virus LPAI H9N2 virus [A/CK/Eg/114940v/NLQP/2011 ly, twofold serial dilutions of sera were mixed with four hemagglutination
(H9N2)] (GenBank Q440373) was isolated from a broiler flock, in (HA) units of H9N2 AI antigen. The HI titer was determined using a
Menofia governorate, Egypt, in 2011 at the Reference Laboratory for Vet- 1% red blood cell suspension, which was collected from at least three
erinary Quality Control on Poultry Production, Animal Health Research SPF chickens (8 wk of age) reared in our lab animal facility.
Institute, Dokki, Giza, Egypt. This virus was selected to be representative Challenge experiment. The challenge experiment was conducted in
of circulating LPAI virus (H9N2) in Egypt. The selected virus was propa- Biosafety Level 3 chicken isolators, at ME VAC and according to the
gated using specific-pathogen-free (SPF) embryonated chicken eggs (Nile ME VAC guidelines on animal research ethics. Because of the limited
228 W. H. Kilany et al.

Table 1. Postvaccination HI mean antibody titers and seroconversion rates in SPF chickens immunized with different formulas of inactivated A/
H9N2 vaccines against the homologous A/CK/Eg/114940v/NLQP/11(H9N2) antigen.
PostvaccinationB Postchallenge
14 days
Week 1 Week 2 Week 3 Week 4 postchallenge
Vaccine typeA Batch no. Mean % Sero. Mean % Sero. Mean % Sero. Mean % Sero. Mean % Sero.
A 1 1.7 0.9 a 20 4.3 0.9 a 100 8 0.7 a 100 9.5 0.5 a 80 10 0.4 100
2 1.7 0.9 a 20 4.4 0.7 a 100 7.7 0.5 a 100 9.6 0.5 a 100
3 1.6 1.1 a 20 3.9 0.6 a 100 7.6 0.7 a 100 9.5 0.7 a 100
B 1 1.6 1.1 a 20 5.2 0.8 b 100 9.1 0.7 b 100 9.8 0.4 a 100 10 0 100
2 1.5 1.4 a 30 4.9 1.1 b 100 8.4 0.5 b 100 9.8 0.4 a 100
3 1.5 1.2 a 20 4.8 0.8 b 100 8.3 0.7 b 100 9.5 0.5 a 100
A
Vaccine A: inactivated H9N2 ISA70 vaccine batches (A1, A2, and A3) 0.5 ml/dose; vaccine B: inactivated H9N2 ISA71 vaccine batches (B1, B2,
and B3) 0.3 ml/dose.
B
Mean HI antibody titers are log2 SD; % Sero. 5 seroconversion rate 5 (no. of birds in a group with HI titers $ 23/no. of birds in a group) 6
100. Titers in the same column followed by different lowercase letters are statistically different (P , 0.05).

number of Level 3 isolators, the challenge experiment was conducted for nonvaccinated group showed mild respiratory signs and depression.
only two vaccinated groups (A1, B1) and one nonvaccinated group (C) The virus shedding titers in both vaccinated groups at 2 and 4
using the homologous LPAI H9N2 virus, at 4 wk PV. The challenged DPC were significantly lower than the nonvaccinated challenge group
birds received a 10-ml dose at 106 50% egg infective dose (EID50) via by at least 23 log EID50. Both vaccines were able to stop virus shed-
the intranasal route. At 2, 4, 6, and 10 DPC, oropharyngeal swabs ding by 6 DPC. The challenge virus was detected in 30% and 10% of
were collected from each challenged bird separately for virus detection
and titration.
challenged birds in Group A (vaccinated with ISA70) at 2 and 4
Viral isolation and detection. Oropharyngeal swabs were collected in
DPC, respectively; whereas 20% and 10% of challenged birds in
a 1-ml phosphate-buffered saline, pH 7.2, containing gentamycin (50 g/ Group B (vaccinated with ISA71) showed detectable virus shedding
ml) and nystatin (1000 U/ml) (20). Swab samples were vortexed and then at 2 and 4 DPC, respectively (Table 2).
centrifuged at 2000 rpm for 10 min at 4 C to pellet the debris. The
supernatants of individual samples were tested via virus titration to moni-
tor virus shedding in 10-day SPF embryonated chicken eggs. Infective DISCUSSION
dose (EID50/ml) was then calculated (21).
Statistical analysis. Statistical analysis was performed using SPSS ver- AI is one of the most devastating viral diseases in the poultry indus-
sion 13. The nonparametric Kruskal-Wallis test was used for the compar- try and has a worldwide distribution (23). Since the first H9N2 LPAI
ison of vaccinated and unvaccinated groups. Differences were considered outbreak in Egypt (10), several outbreaks have occurred. This has
significant at P , 0.01. The Mann-Whitney U-test was also used for the caused significant health problems and great economic losses. In
comparison between groups. A P , 0.05 was considered statistically Egypt, the main adopted AI control measure is vaccination against
significant.
both A/H5N1 and A/H9N2. The endemic situation of both viruses,
as well as other poultry viruses, has complicated the current vaccina-
tion programs in all poultry sectors. Several experimental studies have
RESULTS demonstrated that inactivated water in oil emulsion vaccines are capa-
ble of inducing antibody response to protect poultry (5,19). Howev-
Phylogenetic and sequence analysis of the AIV strain. The phyloge- er, the need for potent vaccines with full antigen dose that could be
netic tree and sequence analysis of the partial sequence of the HA used during an early stage of life has increased (3). To reduce dose
gene showed that the strain Ck/EG/114940v/NLQP/11(H9N2) volume, a potent adjuvant to enhance immunogenicity of the antigen
has genetic traits similar to those of the currently circulating viruses was deemed important (4). A perfect oil adjuvant should enhance the
belonging to the G1-like lineages circulating in Egypt (Fig. 1). Minor immune response and reduce the number of immunizations required.
evolution was observed, with no significant differences in the HA The Montanide ISA71 adjuvants have been shown to stimulate both
gene. The nucleotide identity showed that the virus was 99.2% simi- humoral and cell-mediated immune response in different animal spe-
lar to the recent Egyptian H9N2 strains. cies, including chickens (12,13). In this study, the efficacy of a
Postvaccination humoral immune response. In general, there were reduced dose volume of vaccine (from 0.5 to 0.3 ml) containing inac-
no significant differences (P $ 0.05) between the postvaccination tivated H9N2 vaccines and formulated with Montanide ISA71 was
immune responses in the three different batches of each vaccine. compared with a regular dose of vaccine (i.e., 0.5 ml/bird) formulated
The vaccinated birds showed detectable HI antibody titers by week with Montanide ISA70 adjuvant.
2 PV in all groups of both vaccine batches ($4 log2). Vaccine B The phylogenetic analysis of the HA gene sequence showed that
showed faster and higher immune response than Vaccine A batches the selected LPAI H9N2 virus strain had genetic and antigenic traits
by weeks 2 and 3 PV (P # 0.05). However, this significant difference similar to those of the field viruses currently circulating in Egypt (Fig.
disappeared by week 4 PV (Table 1). 1), with a 99.2% nucleotide identity. This high genetic similarity
Challenge experiment in SPF chickens. To evaluate the protective with the recent Egyptian H9N2 LPAI and further vaccine efficacy
efficiency of both vaccine groups, A1 and B1 vaccinated SPF chickens tests conducted on SPF chickens indicated that the virus can be
were challenged with the same LPAI H9N2 at week 4 PV. The mean used as an effective vaccine, with no need for modification.
log2 HI titers of the A1 and B1 groups at age of challenge were 9.5 The two formulated H9N2 vaccines using Montanide ISA70 and
0.4 and 9.8 0.5 log2, respectively (Table 1). Neither vaccinated ISA71 developed a protective immune response in vaccinated SPF
challenged groups showed clear clinical signs, whereas the birds by week 2 PV using a single-dose regime. Previous studies
Inactivated H9N2 vaccine dose reduction 229

Fig. 1. Phylogenetic evolutionary analyses of A/H9N2 viruses (conducted through a bootstrap trial of 1000 using MEGA version 6).

have shown that AI vaccine emulsified with Montanide ISA70 oil immunity in chickens vaccinated with low doses of Eimeria profilin
adjuvant provided good protection to immunized chickens against subunit vaccines (12).
H9N2 infection (7). Although the pathogenicity of H9N2 was previously reported (18),
In this study, the antibody levels elicited by reduced ISA71 (0.3 ml) the role of secondary agents in the pathogenesis of AIV H9N2,
vaccine were faster and higher than with a regular dose (0.5 ml) of including environmental and other pathogens, cannot be ignored
ISA70 vaccine at as early as weeks 2 and 3 PV (P # 0.05). These results (11,19). Because of the low pathogenic nature of H9N2 in the
indicate that the dose volume reduction adds to the benefits of early absence of complicating agents, quantitation of viral replication and
immunization against infection. This might be attributable to the shedding in infected birds were used for the evaluation of both vac-
reduction of oil volume used, having the effect of accelerating the anti- cines in this study. It has been documented that an effective vaccine
gen absorption rate. On the other hand, the stimulatory effect of the not only prevents clinical disease, but it also reduces viral shedding
oil adjuvant used cannot be ignored. Montanide ISA71 has been compared with unvaccinated control birds, thereby decreasing the
shown to enhance both serum antibody levels and cell-mediated likelihood of transmission of the virus to new susceptible hosts (24).
230 W. H. Kilany et al.

Table 2. Mean titer of virus shedding and number of sheddersB of different vaccine formulas and nonvaccinated SPF chickens challenged with A/
A

CK/Eg/114940v/NLQP/11(H9N2).
Vaccine AC Vaccine BD Unvaccinated challengedE
DPC Titer (mean SD) No. shedders (pos/total) Titer (mean SD) No. shedders (pos/total) Titer (mean SD) No. shedders (pos/total)
2 2.5 0.7 3/10 2.4 0.5 2/10 4.4 0.5 10/10
4 1.5 1/10 1.9 1/10 4.4 1.1 7/10
6 0 0/10 0 0/10 3.6 0.5 3/10
10 0 0/10 0 0/10 0 0/10
A
Titers expressed as EID50/ml SD.
B
No. of positive (pos) samples/total positive tested samples; shedding rate 5 number of shedder birds/total tested birds in the group.
C
Vaccine A: inactivated H9N2 ISA70 vaccine (0.5 ml/dose) challenged with A/CK/Eg/114940v/NLQP/11(H9N2) virus at 6 log EID50/bird.
D
Vaccine B: inactivated H9N2 ISA71 vaccine (0.3 ml/dose) challenged with A/CK/Eg/114940v/NLQP/11(H9N2) virus at 6 log EID50/bird.
E
Unvaccinated challenged group challenged with A/CK/Eg/114940v/NLQP/11(H9N2) virus at 6 log EID50/bird.

The results of virus isolation and viral shedding in both vaccines indi- Hafez. Isolation of H9N2 avian influenza virus from bobwhite quail (Colinus
cated that the vaccine-challenged chickens were protected against viral virginianus) in Egypt. Arch. Virol. 157:11671172. 2012.
shedding. There was no significant difference in the virus shedding 11. Haghighat-Jahromi, M., K. Asasi, H. Nili, H. Dadras, and A. H.
Shooshtari. Coinfection of avian influenza virus (H9N2 subtype) with infec-
titers. The percentage of shedder birds in the ISA71 vaccinated group
tious bronchitis live vaccine. Arch. Virol. 153:651655. 2008.
however was lower than that in the ISA70 group, although not statisti- 12. Jang, S. I., H. S. Lillehoj, S. H. Lee, K. W. Lee, E. P. Lillehoj, F.
cally significant. The ability of inactivated oil emulsion vaccines to pro- Bertrand, L. Dupuis, and S. Deville. Montanide ISA 71 VG adjuvant
tect chickens against AI clinical disease, both in experimental (7) and enhances antibody and cell-mediated immune responses to profilin subunit
field conditions (26), has previously been documented. The endemic antigen vaccination and promotes protection against Eimeria acervulina and
situation of H9N2 virus in Egypt could be due to the shortage of vacci- Eimeria tenella. Exp. Parasitol. 127:178183. 2010.
nation coverage in different poultry sectors (15). Not only is an effective 13. Jang, S. I., H. S. Lillehoj, S. H. Lee, K. W. Lee, M. S. Park, G. R.
Bauchan, E. P. Lillehoj, F. Bertrand, L. Dupuis, and S. Deville. Immunoen-
mass vaccination strategy required for proper AI control in endemic
hancing effects of Montanide ISA oil-based adjuvants on recombinant coccid-
countries, but focus also should be put on using potent vaccines that ia antigen vaccination against Eimeria acervulina infection. Vet. Parasitol.
are capable of inducing an early postvaccination immune response. 172:221228. 2010.
To conclude, although both vaccine formulas (ISA70 and ISA71) 14. Kandeil, A., R. El-Shesheny, A. M. Maatouq, Y. Moatasim, M. M.
were potent and lower virus shedding was observed, the dose-reduced Shehata, O. Bagato, A. Rubrum, K. Shanmuganatham, R. J. Webby, M. A.
inactivated vaccine with a potent adjuvant showed good efficacy, as Ali, and G. Kayali. Genetic and antigenic evolution of H9N2 avian influenza
observed by earlier and higher immune responsesas early as 2 wk viruses circulating in Egypt between 2011 and 2013. Arch. Virol. 159:
PV. Thus it may be stated that the dose-reduced vaccine could be 28612876. 2014.
15. Monne, I., H. A. Hussein, A. Fusaro, V. Valastro, M. M. Hamoud,
used effectively to achieve earlier protection, especially in broiler
R. A. Khalefa, S. N. Dardir, M. I. Radwan, I. Capua, and G. Cattoli.
chicks in endemic countries. H9N2 influenza A virus circulates in H5N1 endemically infected poultry
population in Egypt. Influenza Other Respir. Viruses. 7:240243. 2013.
REFERENCES 16. Naeem, K., N. Siddique, M. Ayaz, and M. A. Jalalee. Avian influenza
1. Abdelwhab, E. M., and A. S. Abdel-Moneim. Epidemiology, ecology in Pakistan: outbreaks of low- and high-pathogenicity avian influenza in
and gene pool of influenza A virus in Egypt: will Egypt be the epicentre of Pakistan during 20032006. Avian Dis. 51:189193. 2007.
17. Naguib, M. M., A. S. Arafa, M. F. El-Kady, A. A. Selim, V. Gunalan,
the next influenza pandemic? Virulence 6:618. 2015.
2. Alexander, D. J. An overview of the epidemiology of avian influenza. S. Maurer-Stroh, K. V. Goller, M. K. Hassan, M. Beer, E. M. Abdelwhab,
Vaccine 25:56375644. 2007. and T. C. Harder. Evolutionary trajectories and diagnostic challenges of
3. Amer, M. M., A. S. Hammouda, A. H. Amin, K. M. El-Bayomi, and potentially zoonotic avian influenza viruses H5N1 and H9N2 co-circulating
S. A. N. Nasr. Comparative study on immunogenicity of commercially avail- in Egypt. Infect. Genet. Evol. 34:278291. 2015.
able inactivated oil adjuvant avian influenza H5N1 and H5N2 vaccines in 18. Nili, H., and K. Asasi. Natural cases and an experimental study of
broiler chicks. Glob. Vet. 8:618624. 2012. H9N2 avian influenza in commercial broiler chickens of Iran. Avian Pathol.
4. Aucouturier, J., S. Deville, C. Perret, I. Vallee, and P. Boireau. Assess- 31:247252. 2002.
ment of efficacy and safety of various adjuvant formulations with a total solu- 19. Nili, H., and K. Asasi. Avian influenza (H9N2) outbreak in Iran. Avi-
ble extract of Trichinella spiralis. Parasite 8:S126S132. 2001. an Dis. 47:828831. 2003.
5. Bublot, M., F. X. Le Gros, D. Nieddu, N. Pritchard, T. R. Mickle, and 20. [OIE] World Organisation for Animal Health. Chapter 2.3.4. Avian
D. E. Swayne. Efficacy of two H5N9-inactivated vaccines against challenge influenza. In: Manual of Diagnostic Tests and Vaccines for Terrestrial Ani-
with a recent H5N1 highly pathogenic avian influenza isolate from a chicken mals. [Internet]. 2012 [modified 2012 May; cited 2015 April] Available
in Thailand. Avian Dis. 51:332337. 2007. from: http://www.oie.int/fileadmin/Home/eng/Health_standards/tahm/2.
6. Capua, I., and D. J. Alexander. Avian influenza vaccines and vaccina- 03.04_AI.pdf
tion in birds. Vaccine 26(Suppl 4):D70D73. 2008. 21. Reed, L. J., and H. Muench. A simple method of estimating fifty per
7. Choi, J. G., Y. J. Lee, Y. J. Kim, E. K. Lee, O. M. Jeong, H. W. cent endpoints. Am. J. Epidemiol. 27:493497. 1938.
Sung, J. H. Kim, and J. H. Kwon. An inactivated vaccine to control the 22. Shany, S. A. Further studies on the current situation of avian influen-
current H9N2 low pathogenic avian influenza in Korea. J. Vet. Sci. 9: za in Egypt. Ph.D. thesis. Beni-Suef University, Beni-Suef, Egypt. pg.
6774. 2008. 227. 2015.
8. Dupuis, L., S. Ascarateil, J. Aucouturier, and V. Ganne. SEPPIC vac- 23. Stallknecht, D. E., and S. M. Shane. Host range of avian influenza
cine adjuvants for poultry. Ann. N. Y. Acad. Sci. 1081:202205. 2006. virus in free-living birds. Vet. Res. Commun. 12:125141. 1988.
9. Eid , A. M. Studies on viral respiratory problems prevalent in broiler 24. Swayne, D. E., and D. A. Halvorson. Influenza. In: Diseases of poul-
chickens. M.Sc. Thesis. Beni-Suef University, Beni-Suef, Egypt. pg. 249. 2015. try, 12th ed. Y. M. Saif, H. J. Barnes, A. M. Fadl, J. R. Glisson, L. R.
10. El-Zoghby, E. F., A. S. Arafa, M. K. Hassan, M. M. Aly, A. Selim, W. McDougald, and D. E. Swayne, eds. State University Press, Ames, IA. pp.
H. Kilany, U. Selim, S. Nasef, M. G. Aggor, E. M. Abdelwhab, andH. M. 153184. 2008.
Inactivated H9N2 vaccine dose reduction 231
25. Tamura, K., G. Stecher, D. Peterson, A. Filipski, and S. Kumar. 27. Tong, S., Y. Li, P. Rivailler, C. Conrardy, D. A. Castillo, L. M.
MEGA6: Molecular Evolutionary Genetics Analysis version 6.0. Mol. Biol. Chen, S. Recuenco, J. A. Ellison, C. T. Davis, I. A. York, A. S. Turmelle,
Evol. 30:27252729. 2013. D. Moran, S. Rogers, M. Shi, Y. Tao, M. R. Weil, K. Tang, L. A.
26. Terregino, C., A. Toffan, M. S. Beato, R. De Nardi, A. Drago, and I. Rowe, S. Sammons, X. Xu, M. Frace, K. A. Lindblade, N. J. Cox, L. J.
Capua. Conventional H5N9 vaccine suppresses shedding in specific-patho- Anderson, C. E. Rupprecht, and R. O. Donis. A distinct lineage of
gen-free birds challenged with HPAI H5N1 A/chicken/Yamaguchi/7/2004. influenza A virus from bats. Proc. Natl. Acad. Sci. U. S. A. 109:4269
Avian Dis. 51:495497. 2007. 4274. 2012.

You might also like