Main Geno

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Since January 2020 Elsevier has created a COVID-19 resource centre with

free information in English and Mandarin on the novel coronavirus COVID-


19. The COVID-19 resource centre is hosted on Elsevier Connect, the
company's public news and information website.

Elsevier hereby grants permission to make all its COVID-19-related


research that is available on the COVID-19 resource centre - including this
research content - immediately available in PubMed Central and other
publicly funded repositories, such as the WHO COVID database with rights
for unrestricted research re-use and analyses in any form or by any means
with acknowledgement of the original source. These permissions are
granted for free by Elsevier for as long as the COVID-19 resource centre
remains active.
ADVANCES IN VETERINARY MEDICINE, VOL. 41

Viral Vectors for Veterinary Vaccines

MICHAEL SHEPPARD

Animal Health Biological Discovery, Pfizer Central Research, Groton,


Connecticut 06340

I.Introduction and Background


II.Viral Vector Construction
III.Advantages and Disadvantages of Viral Vectors for Vaccine Delivery
IV. Construction of Safer Viral Vectors for Vaccine Delivery
A. Deletion of Nonessential Genes
B. Deletion of Essential Genes
C. Replication Limited Virus
V. Examples of Reported Viral Veterinary Vaccine Vectors
VI. CommerciallyAvailable Viral Vaccine Vectors for Veterinary Use
VII. Summary
References

I. Introduction and Background

Numerous reviews have described the use of viral vectors for pos-
sible vaccine delivery (e.g., Cavanagh, 1985; Sheppard and Fahey,
1989; Wray and Woodward, 1990; G r a h a m and Prevec, 1992; Boyle and
Heine, 1993; Hilleman, 1994; Martin, 1994; Dorner, 1995; Babiuk et
al., 1996; Perkus and Paoletti, 1996). However, in this review I will
focus solely on the use of viral vectors for delivery of veterinary vac-
cines. It is without question that vaccination plays an essential role in
veterinary medicine, providing the major and often the only prophyla-
tic approach for the control of infectious diseases. In spite of the vast
array of currently available vaccines veterinarians and the livestock
producers continue to express the need for vaccines that not only main-
tain the best features of killed or subunit vaccines (such as safety) as
well as the best features of conventional modified live vaccines (such as
145
Copyright 9 1999by Academic Press
All rights of reproduction in any form reserved.
1093-975X/99 $25.00
146 MICHAEL SHEPPARD

efficacy) but improve on them. As well as the need for continual im-
provement of vaccines there exists a need for new vaccines either to
new diseases (e.g., chicken anemia virus or porcine reproductive and
respiratory syndrome virus) or to old diseases for which vaccines are
not available or no longer meet the requirements of the end user (e.g.,
bovine virus diarrhea virus vaccines). As well as new vaccines there is
also need for vaccines with special features that allow potential cus-
tomers to design disease control programs that suit their specific needs
on top of offering greater safety and improved protection. The design
and construction of these new veterinary vaccines is a major challenge
facing the field ofvaccinology. With the continued demand of improving
vaccines and producing new ones it is easier for potential vaccine can-
didates to fail to meet the increased level of requirements that are
expected. The failure of some vaccines can result from problems associ-
ated with delivery, such as insufficient or no induction of the appropri-
ate protective immune response. The development of delivery systems
to produce vaccines that are more effective, offer greater safety, are
convenient to administer, and are compatible with customer practices
is part of the challenge for vaccinologists. The development of safe and
convenient live viral vectors for the delivery of veterinary vaccines is
one possible way of meeting some of these challenges. Recombinant
DNA technology has allowed more detailed characterization of the ge-
netic organization of many viruses to such an extent that regions suit-
able for insertion of foreign genetic material have been identified. This
has resulted in the development of numerous types of viral vectors
from a wide variety of viral families. Some of these viral vectors have
been developed with the potential for delivering and expressing gene(s)
from a foreign pathogen and so act as a vaccine vector (Table I). The
viral vector is often genetically attenuated or cannot complete its repli-
cation cycle in the animal to be immunized, and thus produces no
clinical disease. Although initially the majority of viral vector develop-
ment centered around poxviruses, especially vaccinia (Panicali and
Paoletti, 1982; Macket et al., 1982), it was not long before viral vector
development witnessed a virtual explosion in the types of viruses de-
veloped into vectors. These included herpeviruses (Post et al., 1982),
adenoviruses (Berkner and Sharp, 1982), retroviruses (Wei et al.,
1981), papoviruses (Southern and Berg, 1982), polyoma virus (Fried
and Ruley, 1982), picornaviruses (Kitson et al., 1991), Semliki Forest
virus (SFV; Zhou et al., 1994), Sindbis virus (Pugachev et al., 1995),
and even some plant viruses (Jagadish et al., 1996; Dalsgaard et al.,
1997).
VIRAL VECTORS FOR VETERINARY VACCINES 147

TABLE I

CHARACTERISTICS OF THE MORE COMMON VIRUS GROUPS USED AS VECTORS

Pox Herpes
Characteristics viruses Adenoviruses viruses Retroviruses

Genome 180-300 kb 30-45 kb 150-200 kb 9.2 kb


Max. Insert >30 kb >5 kb 30 kb 8 kb
Max. Titer 107-109 10s-1011 106-108 106-109
Administration Scarification/ Injection/aerosol/ Injection/water Injection
injection oral
Safety Problems Inflammation Latency Genomic
with insertion
immuno-
suppressed
Background
expression by Yes Yes Yes No
vector

II. Viral Vector Construction

Greater understanding of the structure and function of a wide range


of viruses at the genetic level has opened up ways of designing novel
viral vaccine vectors which should improve the quality and effective-
ness of some future vaccines as major prophylatic tools. Viral vaccine
vectors have really developed from a greater technological understand-
ing of viruses at the genetic level, where today they have become a
viable alternative strategy as one method for the delivery of vaccines.
The concept of viral vectors was first highlighted by Bernard Moss and
others in the early 1980s (Mackett et al., 1982; Panicali and Paoletti,
1982), where they showed that vaccinia virus could be engineered to
carry and express foreign genes (Panicali and Paoletti, 1982; Mackett
et al., 1982). From the time when Moss and others first demonstrated
that vaccinia virus could be developed as a vector for the expression of
foreign genes, the technology has been exploited to apply to a variety of
virus families as well as a variety of foreign genes including those that
encode antigens from pathogens. As a result both DNA and RNA vi-
ruses have been developed as viral vaccine vectors (Table I).
To produce viral vaccine vectors it is first necessary to study the
genome of the vector to a stage of understanding where at least one
region suitable for insertion of foreign genetic material has been iden-
148 MICHAEL SHEPPARD

tiffed. Second, genes from pathogens t h a t encode proteins t h a t will


induce an appropriate protective immune response and can be stably
integrated into the vector's genome and expressed need to be identi-
fied. Finally, it is necessary to insert the foreign gene(s) in such a way
as to ensure the correct and sufficient expression of the foreign gene(s).
The ideal viral vaccine vector would have all or at the very least
some of the following features:
9Safe and nonpathogenic for the vaccinate
9Evoke the appropriate protective immune response
9Single host or limited host range
9Stable genome
9No integration into the host genome
9Readily accessible region(s) for insertion of foreign genetic material
9Able to tolerate well insertion of foreign genetic material and expres-
sion of foreign gene(s)
9Convenient to deliver and fits with m a n a g e m e n t practices
9Relatively simple and cost effective to produce
9Limited background gene expression by the vector

III. Advantages and Disadvantages of Viral Vectors


for Vaccine Delivery

Live viral vectors offer several advantages for vaccine delivery com-
pared to killed, subunit, or conventional modified live vaccines. First,
because of the possibility of delivering divalent or even perhaps multi-
valent vaccines, using a single type of vector can result in a single
manufacturing process r a t h e r t h a n several and possibly even a single
vaccination r a t h e r t h a n several. Therefore, vectored vaccines have the
potential to be less expensive to the m a n u f a c t u r e r and the end user.
Because the foreign gene is being expressed in the cells of its n a t u r a l
host, it is expected t h a t any post-translational modifications required
will be correct and produce an authentic antigen, as opposed to Es-
cherichia coli or baculovirus systems (among others) t h a t do not al-
ways produce authentic foreign proteins. Depending on the vector se-
lected it may be possible to deliver the vectored vaccine more
conveniently to the m a m m a l or bird by spray or water or some other
means r a t h e r t h a n by needle injection. Such a mass administration
approach may be particularly relevant to the poultry industry. The
vector could also be constructed to deliver simultaneously an immu-
nomodulator (e.g., g a m m a interferon), which could modify the type or
VIRAL VECTORS FOR VETERINARYVACCINES 149

magnitude of the immune response to allow the vaccine to be success-


ful or more successful t h a n it would be otherwise. The vector only
expresses the antigens from the pathogen that are required to elicit a
protective i m m u n e response and therefore reduces or eliminates the
chance of disease by being exposed to the whole pathogen as with a
killed or modified live vaccine. Finally, the appropriate viral vectors
will induce both cell-mediated and humoral immune responses and in
some cases are particularly suitable for inducing a local immune re-
sponse in the mucosal surfaces.
One of the main disadvantages of using viral vectors for vaccine
delivery is t h a t like subunit vaccines each vector can only deliver one
or a relatively small n u m b e r of foreign antigens to the host animal and
therefore rely on those being able to elicit a completely protective im-
mune response. Also the only antigens that can be delivered are those
t h a t are encoded by nucleic acid. Thus such things as lipopolysac-
charides are not deliverable. With any vector, regardless of type, only a
limited amount of foreign genetic material can be inserted into the
vector's genome stably and expressed appropriately. One must always
be wary of altered tissue tropisms due to the expression of the foreign
gene(s). Of course the effectiveness of a viral vector is limited by preex-
isting i m m u n e response in the animal from prior exposure to the virus
used to construct the vector. Finally, as with all live vaccines there is
the problem of shelf life and compatibility with other vaccine prepara-
tions.

IV. Construction of Safer Viral Vectors for Vaccine Delivery

To produce viral vaccine vectors successfully it is necessary to ensure


t h a t the vector itself does not pose any disease t h r e a t to the animal
t h a t receives the vaccination or to the person delivering the vaccine to
the animal. Most often this is achieved by a t t e n u a t i n g the viral vector
in some way. Until recent times the means of generating a live attenu-
ated virus had been entirely empirical. This process usually involved
the passaging of the virus in cell culture or animals t h a t were not the
n a t u r a l host, followed by testing of the resulting viruses for decreased
virulence in the n a t u r a l host. The basis for attenuation is most often
unknown, and may be a result from as minor as a single base change,
and thus the chance of reversion back to virulence is always a possi-
bility. This type of traditional method for generating a live a t t e n u a t e d
virus is not necessarily the most attractive method for generating a
viral vaccine vector. With the advent of molecular biology and our
150 MICHAEL SHEPPARD

improved knowledge of viruses at the genetic level it is now possible to


generate live attenuated viruses with precise genetic changes, improv-
ing their safety and thus make them more suitable as vectors for vac-
cine delivery.

A. DELETION OF NONESSENTIAL GENES


A good example is the deletion of the thymidine kinase (TK) gene.
While the deletion of the TK gene has little or no effect on virus growth
in cell culture, TK deleted viruses can be significantly attenuated in
vivo ((Buller et al., 1985; Kit et al., 1985, 1986; Becker et al., 1986).
This feature has been exploited successfully for the development of live
attenuated herpesvirus vaccines (McGregor et al., 1985; Kit et al.,
1985; Marchioli et al., 1987; Moorman et al., 1990) as well as safer
herpesvirus and poxvirus vectors (e.g., Buller et al., 1985; Bayliss et
al., 1991; Mulder et al., 1994; Hu et al., 1997).

B. DELETION OF ESSENTIAL GENES


If an essenential gene is deleted from a virus, the virus can only grow
if the gene or gene product is provided in trans. This virus is phe-
notypically normal but genotypically defective and cannot replicate in
the host because the deleted gene product is not available. This type of
virus can replicate in vitro with the help of a genetically engineered
supporting cell line that expresses the deleted gene product. The stage
of the virus life cycle of which the gene product is required will govern
how far through the replication cycle a virus will proceed. In some
cases (e.g., if the essential deleted gene is required for virus penetra-
tion of the cell) the virus will complete a single round of replication in
the host but the progeny viruses will not be able to invade any other
cell. (Farrell et al., 1994; McLean et al., 1994; Peeters et al., 1994).
However, if the deleted essential gene is an early gene that is required
to activate other viral genes, then the number of viral proteins synthe-
sized may be limited and the viral genome may not be able to complete
even a single cycle of replication (Chen and Knipe, 1996; Brehm et al.,
1997; Da Costa et al., 1997).

C. REPLICATION LIMITED VIRUS


A third alternative, which has been exploited successfully, is to use a
virus that can only completely replicate in one species as a vector in
another species, where it cannot complete an entire cycle of replication
but can commence a replication cycle sufficiently to allow expression of
VIRAL VECTORS FOR VETERINARY VACCINES 151

the foreign gene (Tartaglia et al., 1992). The canarypox virus (CPV)
vector, termed ALVAC, has successfully been exploited to the degree of
commercial success. The CPV is restricted to avian cells only for pro-
ductive replication but can be used to vaccinate m a m m a l s where it can
elicit an immune response to the foreign gene product without complet-
ing an entire cycle of replication (Tartaglia et al., 1992, 1993; Taylor et
al., 1995). The h u m a n adenovirus type 5 (HAV-5) has also been exploit-
ed in a similar fashion to the CPV (Table III) but has the disadvantage
that this virus is a h u m a n pathogen and so has yet to be exploited
commercially.
Several other strategies are also available and in some cases have
been exploited successfully in order to generate safe viral vectors for
vaccine delivery. Table V (next section) provides a s u m m a r y of some of
these possible approaches.

V. Examples of Reported Viral Veterinary Vaccine Vectors

Even though there are a great many examples of viral vectors report-
ed in the literature since they were first described in 1982, the number
of publications reporting the use of viral vectors for veterinary vaccine
delivery is not that large. After searching for published papers that
describe viral vectors with veterinary vaccine applications, especially
those that could be described as purposely developed for veterinary
use, the obvious conclusion was that even though this research was
first described in 1982 the veterinary side is still in its infancy. Publica-
tions describing viral vectors for veterinary vaccine delivery can be
divided into several groups, which are represented in the following tables:
Table II, poxvirus vectors; Table III, adenovirus vectors; Table IV, her-
pesvirus vectors; and Table V, other virus vectors. Although these four
tables probably do not include every single publication describing viral
vectors for veterinary vaccine delivery they do describe the majority of
published papers and present the reader with an idea of the limited
amount of research that has occurred in this field during the last 15
years.

VI. Commercially Available Viral Vaccine Vectors


for Veterinary Use

At the time of writing this review only three viral vectored vaccines
for use in the veterinary field have been licensed for release. All three
are based on poxvirus vectors and the three vectors represent the
152 MICHAEL SHEPPARD

TABLE II
PoxvIRUS VECTORS FOR THE DELIVERY OF VETERINARY VACCINES

Test
Vector Pathogen Antigen animal Reference

CPV RHDV Capsid Rabbit Fischer et al., (1997)


RPV FPV/rabies VP2/G Cat Hu et al., (1997)
CPV/VV CDV F/HA Ferret Stephensen et al., (1997)
FPV NDV F/HN Chicken Taylor et al., (1996)
Myxoma Influenza HA Rabbit Kerr and Jackson (1995)
SPV PrV gp50/gp63 Swine van der Leek et al., (1994)
CPV FeLV env/gag Cat Tartaglia et al., (1993)
VV Rabies G Fox Brochier et al., (1991)
PPV NDV F Chicken Latellier et al., (1991)
FPV NDV HA/NA Chicken Boursnell et al., (1990a)
FPV NDV HN/F Chicken Boursnell et al., (1990b)
VV BLV env Rabbit Ohishi et al., (1990)
VV EHV-1 gpl3 Mouse Guo et al., (1989)
VV PrV gp50/63/I/X Mouse Kost et al., (1989)
FPV Rabies G Dog/cat Taylor et al., (1988)
VV FeLV env Cat Gilbert et al., (1987)
VV Rabies G Fox Blancou et al., (1986)
VV Rabies G Mouse Kieny et al., (1984)

Key: VV, vaccinia virus; FPV, fowl poxvirus; PPV, pigeon poxvirus; SPV swine poxvi-
rus; CPV, canary poxvirus; RHDV, rabbit hemorrhagic disease virus; CDV, canine dis-
temper virus; FPV, feline parvovirus; PrV, pseudorabies virus; FeLV, feline leukemia vi-
rus; NDV, Newcastle disease virus; BLV, bovine leukosis virus; EHV, equine herpes
virus.

TABLE III
ADENOVIRUS VECTORS FOR THE DELIVERY OF VETERINARY VACCINES

Vector Pathogen Antigen Test animal Reference

OAV ~ n e a ovis 45W Sheep Rothel et al., (1997)


HAV-5 PRCV Spike Swine Callebaut et al., (1996)
HAV-5 TGE Spike Swine Torres Iet al., (1996)
HAV-5 Rabies G Skunk Yarosh et al., (1996)
HAV-5 BCV HEG Cotton rat Bacca-Estrada et al., (1995)
HAV-5 FIV env Cat Gonin et al., (1995)
HAV-5 PRCV Spike Swine Callebaut et al., (1994)
HAV-5 PrV gD Swine Adam et al., (1994)
HAV-5 Rabies G Dog Prevec et al., (1990)
HAV-5 PrV gp50 Rabbit/mouse Eloit et al., (1990)

Key: OAV, ovine adenovirus; HAV-5, human adenovirus type 5; PRCV, porcine respira-
tory corona virus; TGE, transmissible gastroenteritis virus; BCV, bovine corona virus;
PrV, pseudorabies virus.
VIRAL VECTORS FOR VETERINARY VACCINES 153

TABLE IV
HERPES VIRUS VECTORS FOR THE DELIVERY OF VETERINARY VACCINES

Vector Pathogen Antigen Test animal Reference

HVT NDV HN/F Chicken Reddy et al., (1996)


HVT MDV gpAB Chicken Reddy et al., (1996)
FHV-1 FeLV env Cat Willemse et al., (1996)
PrV HCV gpE1 Swine Mulder et al., (1994)
HVT MDV gpB Chicken Ross et al., (1993)
BHV-1 PrV gpC Swine Kit et al., (1992)
FHV-1 FeLV gag/env Cat Wardley et al., (1992)
BHV-1 FMDV cp-epitopes Cattle M. Kit et al., (1991)
BHV-1 FMDV cp-epitopes Cattle S. Kit et al., (1991)
PrV HCV gpE1 Swine van Zijl et al., (1991)

Key: HTV, herpes virus of turkeys; FHV, feline herpes virus; BHV, bovine herpes virus;
PrV, pseudorabies virus; NDV, Newcastle disease virus; MDV, Marek's disease virus;
FMDV, foot-and-mouth disease virus; HCV, hog cholera virus.

evolution in poxvirus vector development. The first vector approved


was the vaccinia virus vector carrying the rabies G glycoprotein gene
(e.g., Kieny et al., 1984; Blancou et al., 1986; Brochier et al., 1991). In
terms of complying with the characteristics of a desirable vector for
vaccine delivery in the veterinary setting, this vector has the greatest
number of undesirable characteristics. However, it satisfied an unmet
need and as a result was released in various parts of the world. The
second vector to be licensed for release was the fowlpox virus vector.
This vector delivers the Newcastle disease virus HN and F genes and is
designed to vaccinate poultry (e.g., Boursnell et al., 1990a,b; Taylor et
al., 1996). While this vector has the desirable characteristic of only
replicating in poultry it also has some limitations that affect its use in
the field. The third vector licensed is the canarypox virus vector and
represents the state-of-the-art poxvirus vector. This vector was devel-
oped to deliver the HA and F genes of canine distemper virus and is the
most recently available of the three vector vaccines (e.g., Stephensen et
al., 1997).

VII. Summary

Whatever strategy is adopted for the development of viral vectors for


delivery of veterinary vaccines there are several key points to consider:
(1) Will the vectored vaccine give a delivery advantage compared to
TABLE V
OTHER VIRUS VECTORS FOR THE DELIVERY OF VETERINARY VACCINES

Vector Pathogen Antigen Test animal Reference

CPMV MEV VP2 epitope Mink Dalsgaard et al. (1997)


Poliovirus FMDV Epitopes Guinea pig Kitson et al. (1991)
Retrovirus NDV HN Chicken Morrison et al. (1990)
Retrovirus Influenza HA Chicken Hunt et al. (1988)

OTHER ALTERNATIVE VIRAL VECTORS THAT HAVE THE POTENTIAL FOR VETERINARY VACCINE DELIVERY

Amplicons VLPs SFV Sinbis Bacteriophage

Frenkel et al. (1994) Jagadish et al. (1996) Atkins et al. (1996) Pugachev et al. (1995) Bastien et al. (1997)
Smith et al. (1995) Porter et al. (1996) Mossman et al. (1996)
Fink et al. (1996) Roy (1996) Zhou et al. (1995)
Pechan et al. (1996) Schodel et al. (1994a) Zhou et al. (1994)
Starr et al. (1996) Schodel et al. (1994b)

Key: CPMV, cowpea mosaic virus; MEV, mink enteritis virus.


VIRAL VECTORS FOR VETERINARY VACCINES 155

w h a t ' s a l r e a d y a v a i l a b l e ? (2) Will t h e v e c t o r e d v a c c i n e give a m a n u f a c -


t u r i n g a d v a n t a g e c o m p a r e d to w h a t ' s a l r e a d y a v a i l a b l e ? (3) Will t h e
v e c t o r e d v a c c i n e p r o v i d e i m p r o v e d s a f e t y c o m p a r e d to w h a t ' s a l r e a d y
a v a i l a b l e ? (5) Will t h e v e c t o r e d v a c c i n e i n c r e a s e t h e d u r a t i o n of i m m u -
n i t y c o m p a r e d to w h a t ' s a l r e a d y a v a i l a b l e ? (6) Will t h e v e c t o r e d vac-
cine be m o r e c o n v e n i e n t to store c o m p a r e d to w h a t ' s a l r e a d y a v a i l a b l e ?
(7) Is t h e v e c t o r e d v a c c i n e c o m p a t i b l e w i t h o t h e r v a c c i n e s ? If t h e r e is
no o t h e r a l t e r n a t i v e a v a i l a b l e t h e n t h e a n s w e r to t h e s e q u e s t i o n s is
easy. H o w e v e r , if t h e r e a r e a l t e r n a t i v e v a c c i n e s a v a i l a b l e t h e n t h e
a n s w e r s to t h e s e q u e s t i o n s b e c o m e v e r y i m p o r t a n t b e c a u s e t h e an-
s w e r s will d e t e r m i n e w h e t h e r a v e c t o r e d v a c c i n e is m e r e l y a good
l a b o r a t o r y i d e a or a s u c c e s s f u l vaccine.

REFERENCES

Adam, M., Lepottier, M. F., and Eloit, M. (1994). Vaccination of pigs with replication-
defective adenovirus vectored vaccines: The example of pseudorabies. Vet. Microbiol.
42, 205-215.
Atkins, G. J., Sheahan, B. J., and Liljestrom, P. (1996). Manipulation of the Semliki
Forest virus genome and its potential for vaccine construction. Mol. Biotechnol. 5, 33-
38.
Babiuk, L. A., van Drunen Littel-van den Hurk, S., Tikoo, S. K., Lewis, P. J., and Liang,
X. (1996). Novel viral vaccines for livestock. Vet. Immunol. Immunopathol. 54, 355-
363.
Bacca-Estrada, M. E., Liang, X., Babiuk, L. A., and Yoo, D. (1995). Induction of mucosal
immunity in cotton rats to haemagglutinin-esterase glycoprotein of bovine corona-
virus by recombinant adenovirus. Immunology 86, 134-140.
Bastien, N., Trudel, M., and Simard, C. (1997). Protective immune responses induced by
the immunization of mice with a recombinant bacteriophage displaying an epitope of
the human respiratory syncytial virus. Virology 234, 118-122.
Bayliss, C. D., Peters, R. W., Cook, J. K. A., Reece, R. L., Howes, K., Binns, M. M., and
Boursnell, M. E. G. (1991). A recombinant fowlpox virus that expresses the VP2
antigen of infectious bursal disease virus induces protection against mortality caused
in the virus. Arch. Virol. 120, 193-205.
Becker, Y., Hadar, J., Taylor, E., Ben-Hur, T., Raibstein, I., Rosen, A., and Darai, G.
(1986). A sequence in HpaI P fragment of herpes simplex virus 1 DNA determines
intraperitoneal virulence in mice. Virology 149, 255-259.
Berkner, K. L., and Sharp, P. A. (1982). Preparation of adenovirus recombinants using
plasmids of viral DNA. In "Eukaryotic Viral Vectors" (Y. Gluzman, ed.), pp. 193-198.
Cold Spring Harbor Lab., Cold Spring Harbor, NY.
Blancou, J., Kieny, M. P., Lathe, R., Lecocq, J. P., Pastoret, P. P., Soulebot, J. P., and
Desmettre, P. (1986). Oral vaccination of the fox against rabies using a live recombi-
nant vaccinia virus. Nature 322, 373-375.
Boursnell, M. E. G., Green, P. F., Campbell, J. I. A., Deuter, A., Peters, R. W., Tomley,
F. M., Samson, A. C. R., Emmerson, P. T., and Binns, M. M. (1990a). A fowlpox virus
vaccine vector within insertion sites in the terminal repeats: Demonstration of the
efficacy using the fusion gene of Newcastle disease virus. Vet. Microbiol. 23, 305-316.
156 MICHAEL SHEPPARD

Boursnell, M. E. G., Green, P. F., Samson, A. C. R., Campbell, J. I. A. Deuter, A., Peters,
R. W., Millar, N. S., Emmerson, P. T., and Binns, M. M. (1990b). A recombinant
fowlpox virus expressing the hemagglutinin-neuraminidase gene of Newcastle dis-
ease virus (NDV). Protects chickens against by NDV. Virology 178, 297-300.
Boyle, D. B., and Heine, H. G. (1993). Recombinant fowlpox virus vaccines for poultry.
Immunol. Cell Biol. 71, 391-397.
Brehm, M. A., Bonneau, R. N., Knipe, D. M., and Tevethia, S. S. (1997). Immunization
with a replication-deficient mutant of herpes simplex virus type 1 (HSV-1) induces a
CD8 + cytotoxic T-lymphocyte response and confers a level of protection comparable to
that of wild-type HSV-1. J. Virol. 71, 3534-3544.
Brochier, B., Kieny, M. P., Costy, F., Coppens, P., Bauduin, B., Lecocq, J. P., Languet, B.,
Chappuis, G., Desmettre, P., Afiademanyo, K., Libois, R., and Pastoret, P. P. (1991).
Large-scale eradication of rabies using recombinant vaccinia-rabies vaccine. Nature
354, 520-522.
Buller, R. M. L., Smith, G. L., Cremer, K., Notkins, A., and Moss, B. (1985). Decreased
virulence of recombinant vaccinia virus expressed vectors is associated with a thy-
midine kinase negative phenotype. Nature 317, 813-815.
Callebaut, P., Pensaert, M., and Enjuanes, L. (1994). Construction of a recombinant
adenovirus for the expression of the glycoprotein S antigen of porcine respiratory
coronavirus. In "Coronaviruses" (H. Lande and J. F. Vantherot, eds.), pp. 469-470.
Plenum, New York.
Callebaut, P., Enjuanes, L., and Pensaert, M. (1996). An adenovirus recombinant expres-
sing the spike glycoprotein of porcine respiratory coronavirus is immunogenic in
swine. J. Gen. Virol. 77, 309-313.
Cavanagh, D. (1985). Viral and bacterial vectors of immunogens. Vaccine 3, 45-48.
Chen, Y. M., and Knipe, D. M. (1996). A dominant mutant form of the herpes simplex
virus ICP8 protein decreases viral late gene transcription. Virology 221, 281-290.
Da Costa, X. J., Bourne, N., Stanberry, L. R., and Knipe, D. M. (1997). Construction and
characterization of a replication-defective herpes simplex virus 2 ICP8 mutant strain
and its use in immunization studies in a guinea pig model of genital disease. Virology
232, 1-12.
Dalsgaard, K., Uttenthal, A., Jones, T. D., Xu, F., Merryweather, A., Hamilton, W. D. O.,
Langeveld, J. P. M., Boshuizen, R. S., Kamstrup, S., Lomonossoff, G. P., Porta, C.,
Vela, C., Casal, J. I., Meloen, R. H., and Rodgers, P. B. (1997). Plant-delivered vaccine
protects target animals against a viral disease. Nat. Biotechnol. 15, 248-252.
Dorner, F. (1995). An overview of vaccine vectors. Dev. Biol. Stand. 84, 23-32.
Eloit, M., Gilardi- Hebenstreit, P., Toma, B., and Perricaudet, M. (1990). Construction of
a defective adenovirus vector expressing the pseudorabies virus glycoprotein gp50
and its use as a live vaccine. J. Gen. Virol. 71, 2425-2431.
Farrell, H. E., McClean, C. S., Harley, C., Efstathiou, S., Inglis, S., and Minson, A. C.
(1994). Vaccine potential of a herpes simplex virus type 1 mutant with an essential
gene deleted. J. Virol. 68, 927-932.
Fink, D. J., DeLuca, N. A., Goins, W. F., and Glorioso, J. C. (1990). Gene transfer to
neurons using herpes simplex virus based vectors. Annu. Rev. Neurosci. 19, 265-287.
Fischer, L., LeGros, F.-X., Mason, P. W., and Paoletti, E. (1997). A recombinant ca-
narypox virus protects rabbits against a lethal rabbit hemorrhagic disease virus
(RHDV) challenge. Vaccine 15, 90-96.
Frenkel, N., Singer, O., and Kwong, A. D. (1994). Minireview: The herpes simplex virus
amplicon--a versatile defective virus vector. Gene Ther. 1, Suppl. 1, $40-$46.
VIRAL VECTORS FOR VETERINARY VACCINES 157

Fried, M., and Ruley, E. (1982). Use of polyoma virus vector. In "Eukaryotic Viral Vec-
tors" (Y. Gluzmann, ed.), pp. 67-70. Cold Spring Harbor Lab., Cold Spring Harbor,
NY.
Gilbert, J. H., Pedersen, N. C., and Nunberg, J. H. (1987). Feline leukemia virus enve-
lope protein expression encoded by a recombinant vaccinia virus: Apparent lack of
immunogenicity in vaccinated animals. Virus Res. 7, 49-67.
Gonin, P., Fournier, A., Oualikene, W., Moraillon, A., and Eloit, M. (1995). Immunization
trial of cats with a replication defective adenovirus type 5 expressing the ENV gene of
feline immunodeficiency virus. Vet. Microbiol. 45, 393-401.
Graham, F. L., and Prevec, L. (1992). Adenovirus-based expression vectors and recombi-
nant vaccines. In '~Vaccines: New Approaches to Immunological Problems" (R. W.
Ellis, ed.), pp. 363-430. Butterworth-Heinemann, Boston.
Guo, P. X., Goebel, S., Davis, S., Perkus, M. E., Languet, B., Desmettre, P., Allen, G., and
Paoletti, E. (1989). Expression in recombinant vaccinia virus of the equine her-
pesvirus 1 gene encoding glycoprotein 13 and protection of immunized animals. J.
Virol. 63, 4189-4198.
Hilleman, M. R. (1994). Recombinant vector vaccines in vaccinology. Dev. Biol. Stand. 82,
3-20.
Hu, L., Ngichabe, C., Trimarchi, C. V., Esposito, J. J., and Scott, F. W. (1997). Raccoon
poxvirus live recombinant feline panleukopenia virus VP2 and rabies virus glycopro-
tein bivalent vaccine. Vaccine 15, 1466-1472.
Hunt, L. A., Brown, D. W., Robinson, H. L., Naeve, C. W., and Webster, R. G. (1988).
Retrovirus-expressed hemagglutinin protects against lethal influenza virus infec-
tions. J. Virol. 62, 3014-3019.
Jagadish, M. N., Edwards, S. J., Hayden, M. B., Grusovin, J., Vandenberg, K., Schoofs,
P., Hamilton, R. C., Shukla, D. D., Kalnins, H., McNarmara, M., Haynes, J., Nisbet,
I. T., Ward, C. W., and Pye, D. (1996). Chimeric potyvirus-like particles as vaccine
carriers. Intervirology 39, 85-92.
Kerr, P. J., and Jackson, R. J. (1995). Myxoma virus as a vaccine vector for rabbits--
antibody levels to influenza virus hemagglutinin presented by a recombinant myxoma
virus. Vaccine 13, 1722-1726.
Kieny, M. P., Lathe, R., Drillen, R., Spehner, D., Skory, S., Schmitt, D., Wiktor, T.,
Koprowski, H., and Lecocq, J. P. (1984). Expression of rabies virus glycoprotein from a
recombinant vaccinia virus. Nature 312, 163-166.
Kit, M., Kit, S., Little, S. P., DiMarchi, R. D., and Gale, C. (1991). Bovine herpesvirus-1
(infectious bovine rhinotracheitis virus)-based viral vectors which expresses foot-and-
mouth disease epitopes. Vaccine 9, 564-572.
Kit, S., Kit, M., and Pirtle, E. C. (1985). Attenuated properties of thymidine kinase-
negative deletion mutant of pseudorabies virus. Am. J. Vet. Res. 46, 1359-1367.
Kit, S., Kit, M., and McConnell, S. (1986). Intramuscular and intravaginal vaccination of
pregnant cows with thymidine kinase-negative, temperature-resistant infectious
bovine rhinotracheitis virus (bovine herpesvirus 1). Vaccine 4, 55-61.
Kit, S., Kit, M., DiMarchi, R. D., Little, S. P., and Gale, C. (1991). Modified-live infectious
bovine rhinotracheitis virus vaccine expressing monomer and dimer forms of foot-
and-mouth disease cupsid protein epitopes on surface of hybrid virus particles. Arch.
Virol. 120, 1-17.
Kit, S., Otsuka, H., and Kit, M. (1992). Expression of porcine pseudorabies virus genes by
a bovine herpesvirus-1 (infectious bovine rhinotracheitis virus) vector. Arch. Virol.
124, 1-20.
158 MICHAEL SHEPPARD

Kitson, J. D. A., Burke, K. L., Pullen, L. A., Belsham, G., and Almond, J. W. (1991).
Chimeric polioviruses that include sequences derived from two independent antigenic
sites of foot-and-mouth disease virus (FMDV) induce neutralizing antibodies against
FMDV in guinea pigs. J. Virol. 65, 3068-3075.
Kost, T. A., Jones, E. V., Smith, K. M., Reed, A. P., Brown, A. L., and Miller, T. J. (1989).
Biological evaluation of glycoproteins mapping to two distinct mRNAs within the
BamHI fragment 7 of pseudorabies virusi expression of the coding regions by vaccinia
virus. Virology 171, 365-376.
Letellier, C., Burny, A., and Meulemans, G. (1991). Construction of a pigeonpox virus
recombinant: expression of the Newcastle disease virus (NDV) fusion glycoprotein
and protection of chickens against NDV challenge. Arch. Virol. 118, 43-56.
Mackett, M., Smith, G. L., and Moss, B. (1982). Vaccinia virus: A selectable eukaryotic
cloning and expression vector. Proc. Natl. Acad. Sci. USA 79, 7415-7419.
Marchioli, C. C., Yancey, R. J., Wardley, R. C., Thomsen, D. R., and Post, L. E. (1987). A
vaccine strain of pseudorabies virus with deletions in the thymidine kinase and gly-
coprotein X genes. Am. J. Vet. Res. 48, 1577-1583.
Martin, S. J. (1994). Vaccine design: Future possibilities and potentials. Biotechnol. Adv.
12, 619-624.
McGregor, S., Easterday, B. C., Kaplan, A. S., and Ben-Porat, T. (1985). Vaccination of
swine with thymidine kinase-deficient mutants of pseudorabies virus. Am. J. Vet. Res.
46, 1494-1497.
McLean, C. S., Erturk, M., Jennings, R., Ni Challanain, D., Minson, A. C., Duncan, I.,
Boursnell, M. E. G., and Inglis, S. C. (1994). Protective vaccination against primary
and recurrent disease caused by herpes simplex virus (HSV) type 2 using a genetically
disabled HSV-1. J. Infect. Dis. 170, 1100-1109.
Moorman, R. J. M., deRover, T., Briaire, J., Peters, B. P. H., Gielkens, A. L. J., and van
Oirschot, J. T. (1990). Inactivation of the thymidine kinase gene of a gI deletion
mutant of pseudorabies virus generates a safe but still highly immunogenic vaccine
strain. J. Gen. Virol. 71, 1591.
Morrison, T., Hinshaw, V. S., Sheerer, M., Cooley, A. J., Brown, D., McQuain, C., and
McGinnes, L. (1990). Retroviral expressed hemagglutinin-neuraminidase protein pro-
tects chickens from Newcastle disease virus induced disease. Microbiol. Pathol. 9,
387-396.
Mossman, S. P., Bex, F., Berglund, P., Arthos, J., O'Neil, S. P., Riley, D., Maul, D. H.,
Bruck, C., Momin, P., Burny, A., Fultz, P. N., Mullins, J. I., Liljestrom, P., and Hoover,
E. A. (1996). Protection against lethal simian immunodeficiency virus SIVsmmPBjl4
disease by a recombinant Semliki Forest virus gpl60 vaccine and by a gpl20 subunit
vaccine. J. Virol. 70, 1953-1960.
Mulder, W. A. M., Priem, J., Glazenburg, K. L., Wagenaar, F., Gruys, E., Gielkens, A. L.
J., Pol, J. M. A., and Kimman, T. G. (1994). Virulence and pathogenesis of non-virulent
and virulent strains of pseudorabies virus expressing envelope glycoprotein E1 of hog
cholera virus. J. Gen. Virol. 75, 117-124.
Ohishi, K., Suzuki, H., Maruyama, T., Yamamoto, T., Funahashi, S., Miki, K., Ikawa, Y.,
and Sugimoto, M. (1990). Induction of neutralizing antibodies against bovine leukosis
virus in rabbits by vaccination with recombinant vaccinia virus expressing bovine
leukosis virus envelope glycoprotein. Am. J. Vet. Res. 51, 1170-1173.
Panicali, D., and Paoletti, E. (1982). Construction of poxviruses as cloning vectors: Inser-
tion of the thymidine kinase gene from herpes simplex virus into the DNA of infec-
tious vaccinia virus. Proc. Natl. Acad. Sci. USA 79, 4927-4931.
VIRAL VECTORS FOR VETERINARY VACCINES 159

Pechan, P. A., Fotaki, M., Thompson, R. L., Dunn, R., Chase, M., Chiocca, E. A., and
Breakefield, X. O. (1996). A novel 'piggyback' packaging system for herpes simplex
virus amplicon vectors. Hum. Gene Ther. 7, 2003-2013.
Peeters, B., Bouma, A., deBruin, T., Moorman, R., Gielkens, A., and Kimman, T. (1994).
Non-transmissible pseudorabies virus gp50 mutants: A new generation of safe live
vaccines. Vaccine 12, 375-380.
Perkus, M. E., and Paoletti, E. (1996). Recombinant virus as vaccination carrier of
heterologous antigens. In "Concepts in Vaccine Development" (S. H. E. Kaufman, ed.),
pp. 379-422. de Gruyter, Berlin.
Porter, D. C., Melsen, L. R., Compans, R. W., and Morrow, C. D. (1996). Release of virus-
like particles from cells infected with poliovirus replicons which express human im-
munodeficiency virus type 1 Gag. J. Virol. 70, 2643-2649.
Post, L. E., Norrild, B., Simpson, T., and Roizman, B. (1982). Chicken ovalbumin gene
fused to a herpes simplex virus a promoter and linked to a thymidine kinase gene is
regulated like a viral gene. Mol. Cell. Biol. 2, 233-240.
Prevec, L., Campbell, J. B., Christie, B. S., Belbeck, L., and Graham, F. L. (1990). A
recombinant human adenovirus vaccine against rabies. J. Infect. Dis. 161, 27-30.
Pugachev, K. V., Mason, P. W., Shope, R. E., and Frey, T. K. (1995). Double-subgenomic
sindbis virus recombinants expressing immunogenic proteins of Japanese encepha-
litis virus induce significant protection in mice against lethal JEV infection. Virology,
212, 587-594.
Reddy, S. K., Sharma, J. M., Ahmad, J., Reddy, D. N., McMillen, J. K., Cook, S. M., Wild,
M. A., and Schwartz, R. D. (1996). Protective efficacy at a recombinant herpesvirus of
turkeys as an in ovo vaccine against Newcastle and Marek's disease in specific-patho-
gen-free chickens. Vaccine 14, 469-477.
Ross, L. J., Binns, M. M., Typers, P., Pastorek, J., Zelnik, V., and Scott, S. (1993).
Construction and properties of a turkey herpesvirus recombinant expressing the Mar-
ek's disease virus homologue of glycoprotein B of herpes simplex virus. J. Gen. Virol.
74, 371-377.
Rothel, J. S., Boyle, D. B., Both, G. W., Pye, A. D., Waterkeyn, J. G., Wood, P. R., and
Lightowlers, M. W. (1997). Sequential nucleic acid and recombinant adenovirus vac-
cination induces host-protective immune responses against Taenia ovis infection in
sheep. Parasite Immunol. 19, 221-227.
Roy, P. (1996). Genetically engineered particulate virus-like structures and their use as
vaccine delivery systems. Intervirology 39, 62-71.
Schodel, F., Peterson, D., Hughes, J., and Milich, D. (1994a). Hepatitis B virus core
particles as a vaccine carrier moiety. Int. Rev. Immunol. 11, 153-165.
Schodel, F., Wirtz, R., Peterson, D., Hughes, J., Warren, R., Sudoff, J., and Milich, D.
(1994b). Immunity to malaria elicited by hybrid hepatitis B virus core particles carry-
ing circumsporozoite protein epitopes. J. Exp. Med. 180, 1037-1046.
Sheppard, M., and Fahey, K. J. (1989). Herpesviruses and adenoviruses as potential
vectors for the poultry industry. Aust. Vet. J. 66, 421-423.
Smith, R. L., Geller, A. I., Escudero, K. W., and Wilcox, C. L. (1995). Long-term expres-
sion in sensory neurons in tissue culture from herpes simplex virus type 1 (HSV-1)
promoters in an HSV-l-derived vector. J. Virol. 69, 4593-4599.
Southern, P., and Berg, P. (1982). Mammalian cell transformation with SV40 vector. In
"Eukaryotic Viral Vectors" (Y. Gluzman, ed.), pp. 41-45. Cold Spring Harbor Lab.,
Cold Spring Harbor, NY.
Starr, P. A., Lim, F., Grant, F. D., Trask, L., Lang, P., Yu, L., and Geller, A. I. (1996). Long-
160 MICHAEL SHEPPARD

term persistence of defective HSV-1 vectors in the rat brain is demonstrated by reac-
tivation of vector gene expression. Gene Ther. 3, 615-623.
Stephensen, C. B., Welter, J., Thaker, S. R., Taylor, J., Tartaglia, J., and Paoletti, E.
(1997). Canine distemper virus (CDV) infection of ferrets as a model for testing Mor-
billivirus vaccine strategies: NYVAC- and ALVAC-based CDV recombinants protect
against symptomatic infection. J. Virol. 71, 1506-1513.
Tartaglia, J., Perkus, M. E., Taylor, J., Norton, E. K., Audonnet, J.-C., Cox, W. I., Davis,
S. W., VanderHoeven, J., Meignier, B., Riviere, M., Languet, B., and Paoletti, E.
(1992). NYVACP: A highly attenuated strain of vaccinia virus. Virology 188, 217-232.
Tartaglia, J., Jarrett, O., Neil, J. C., Desmettre, P., and Paoletti, E. (1993). Protection of
cats against feline leukemia virus by vaccination with a canarypox virus recombinant,
ALVAC-FL. J. Virol. 67, 2370-2375.
Taylor, J., Weinberg, R., Languet, B., Desmettre, P., and Paoletti, E. (1988). Recombinant
fowlpox virus inducing protective immunity in non-avian species. Vaccine 6, 497-503.
Taylor, J., Meignier, B., Tartaglia, J., Languet, B., VanderHoeven, J., Franchini, G.,
Trimarchi, C., and Paoletti, E. (1995). Biological and immunogenic properties of a
canarypox-rabies recombinant, ALVAC-RG (vCP65) in non-avian species. Vaccine 13,
539-549.
Taylor, J., Christensen, L., Gettig, R., Goebel, J., Bouquet, J. F., Mickle, T. R., and
Paoletti, E. (1996). Efficacy of a recombinant fowlpox-based Newcastle disease virus
vaccine candidate against velogenic and respiratory challenge. Avian Dis. 40, 173-
180.
Torres, J. M., Alonso, C., Ortega, A., Mittal, S., Graham, F., and Enjuanes, L. (1996).
Tropism of human adenovirus type 5-based vectors in swine and their ability to
protect against transmissible gastroenteritis coronavirus. J. Virol. 70, 3770-3780.
van der Leek, M. L., Feller, J. A., Sorensen, G., Isaacson, W., Adams, C. L., Borde, D. J.,
Pfeiffer, N., Tran, T., Moyer, R. W., and Gibbs, E. P. (1994). Evaluation of swinepox
virus as a vaccine vector in pigs using Aujeszky's disease (pseudorabies) virus gene
insert coding for glycoproteins gp50 and gp63. Vet. Rec. 134, 13-18.
van Zijl, M., Wensvoort, G., deKluyver, E., Hulst, M., van der Gulden, H., Gielkens, A.,
Berns, A., and Moormann, R. (1991). Live attenuated pseudorabies virus expressing
envelope glycoprotein E1 of hog cholera virus protects swine against both pseudo-
rabies and hog cholera. J. Virol. 65, 2761-2765.
Wardley, R. C., Berlinski, P. J., Thomsen, D. R., Meyer, A. L., and Post, L. E. (1992). The
use of feline herpesvirus and baculovirus as vaccine vectors for the gag and env genes
of feline leukemia virus. J. Gen. Virol. 73, 1811-1818.
Wei, C.-M., Gibson, M., Spear, P. G., and Scolnick, E. M. (1981). Construction and isola-
tion of a transmissible retrovirus containing the src gene of Harvey murine sarcoma
virus and the thymidine kinase gene of herpes simplex virus type 1. J. Virol. 39, 935-
944.
Willemse, M. J., van Schooneveld, S. H. B., Chalmers, W. S. K., and Sondermeijer, P. J. A.
(1996). Vaccination against feline leukemia using a new feline herpesvirus type I
vector. Vaccine 14, 1511-1516.
Wray, C., and Woodward, M. S. (1990). Biotechnology and veterinary science: Production
of veterinary vaccines. Rev. Sci. Tech. Off. Int. Epizoot. 9, 779-794.
Yarosh, O. K., Wandeler, A. I., Graham, F. L., Campbell, J. B., and Prevec, L. (1996).
Human adenovirus type 5 vectors expressing rabies glycoprotein. Vaccine 14, 1257-
1264.
Zhou, X., Berglund, P., Rhodes, G., Parker, S. E., Jondal, M., and Liljestrom, P. (1994).
VIRAL VECTORS FOR VETERINARY VACCINES 161

Self replicating Semliki Forest virus RNA as recombinant vaccine. Vaccine 12, 1510-
1514.
Zhou, X., Berglund, P. Zhao, H., Lilijestrom, P., and Jondal, M. (1995). Generation of
cytotoxic and humoral immune responses by nonreplicative recombinant Semliki For-
est virus. Proc. Natl. Acad. Sci. USA 92, 3009-3013.

You might also like