You are on page 1of 12

REVIEWS

Viruses as vaccine vectors for


infectious diseases and cancer
Simon J. Draper* and Jonathan L. Heeney‡
Abstract | Recent developments in the use of viruses as vaccine vectors have been facilitated
by a better understanding of viral biology. Advances occur as we gain greater insight into the
interrelationship of viruses and the immune system. Viral-vector vaccines remain the best
means to induce cellular immunity and are now showing promise for the induction of strong
humoral responses. The potential benefits for global health that are offered by this field reflect
the scope and utility of viruses as vaccine vectors for human and veterinary applications, with
targets ranging from certain types of cancer to a vast array of infectious diseases.

Reactogenicity
Twenty-five years have elapsed since the first published have frequently suffered from poor immunogenicity.
The capacity of a vaccine use of a recombinant virus to deliver antigens from Conversely, viral vaccine vectors that replicate in vivo
formulation to induce an another infectious agent for the purposes of vaccination. with a vigour that is similar to their wild-type parental
adverse reaction following Vaccinia virus (VACV) recombinant for the hepatitis B viruses are often highly immunogenic, but also carry
immunization.
surface antigen (HBsAg) was shown to express HBsAg the risk of recombination, reactogenicity or reversion
and, subsequently, to induce immunity in chimpanzees to virulence. The search for an optimal medium has
that was sufficient to protect against hepatitis B infec- driven renewed effort in the development of attenuated
tion1,2. Despite this promising early start and substantial replication-defective viral vectors, a strategy aimed at
further development, the list of licensed viral-vector maximizing immunogenicity and safety (FIG. 1).
vaccines for human use is short, in part owing to the
long, hard road to human licensure (for a review, see Attenuating viruses as vaccine vectors
REF. 3). Issues concerning the large-scale production One of the breakthroughs underpinning the viral vac-
and stability of such vaccines often require substantial cine field over the past decade has been the realization
scientific investment, and stringent safety requirements that replication competence and viral virulence do not
must be met for viruses that, in their natural state, have always correlate with immunological robustness. For
the potential to be human pathogens. This is especially example, VACV and its safer, highly attenuated (rep-
true for those viral vectors that remain replication com- lication-defective) cousins — modified vaccinia virus
petent or that, by their nature, persist in the host beyond Ankara (MVA)4–6 and New York attenuated vaccinia
the inflammatory period. Indeed, in the simplest sense virus (NYVAC)7 — have shown comparable and some-
there remains a range of safety that increases with atten- times even better immunogenicity in animal studies8–11.
uation, which historically had to be balanced with the Currently, a wide range of virus families are under intensive
*The Jenner Institute, belief that reduced replicative capacity would result in development as vaccine vectors for either human or vet-
University of Oxford, Old Road
Campus Research Building,
reduced immunogenicity. However, despite these chal- erinary use, including some that are replication competent
Oxford, OX3 7DQ, UK. lenges, arguably the most important factor driving the but also many that are specifically attenuated. TABLE 1 out-

Laboratory of Viral continued development of viral-vector vaccines is their lines the main virus families that are under development
Zoonotics, Department of promising immunogenicity, especially with some of as vectors and their associated diseases.
Veterinary Medicine,
nature’s most difficult antigens belonging to some of the
University of Cambridge,
Madingley Road, Cambridge, world’s toughest pathogens and cancers (TABLE 1). Immune evasion: implications for attenuation.
CB3 0ES, UK. In most cases, a replicating viral infection is very Historically, the attenuation of viruses has been empiri-
Correspondence to S.J.D. effective at eliciting robust immune responses in a host cal, through continuous passage in transformed cell lines
e‑mail: simon.draper@ndm. that may last for several years. This is in contrast to many or in atypical hosts, and through delivery by unnatu-
ox.ac.uk
doi:10.1038/nrmicro2240
recombinant antigens that are delivered either as subunit ral routes. Many viruses are complex pathogens, and
Published online DNA plasmids or proteins; although these are considered large DNA viruses such as poxviruses, adenoviruses
7 December 2009 to be reasonably safe (dependent on the adjuvant), they (AdVs) and herpesviruses have devoted much of their

62 | jANuArY 2010 | VOluMe 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | Viral-vector vaccines in development for human and veterinary use


Recombinant viral Target
vector
Veterinary Human
Adenovirus Avian influenza virus, Plasmodium falciparum, M. tuberculosis, influenza
Mycobacterium tuberculosis and virus, HIV-1 and hepatitis C virus
foot-and-mouth disease virus
Bacteriophage in Shigella None M. tuberculosis
Attenuated canarypox Equine influenza virus, West Nile virus HIV-1 and cancer
virus (WNV), rabies virus, feline leukaemia
virus and canine distemper virus
Fowlpox virus (FPV) Avian influenza virus, FPV and Cancer
Newcastle disease virus (NDV)
NDV Avian influenza virus and NDV None
Turkey herpesvirus Infectious bursal disease virus and None
Marek’s disease virus
Flavivirus YFV-17D WNV WNV, dengue virus and Japanese encephalitis
virus
Lentivirus None Melanoma and HIV-1
Measles virus None P. falciparum and human papilloma virus
Modified vaccinia virus Mycobacterium bovis P. falciparum, M. tuberculosis, influenza A virus, HIV-1
Ankara and colorectal, renal, lung and prostate cancer
New York attenuated None P. falciparum and HIV-1
vaccinia virus
Sendai virus None HIV-1
Vaccinia virus Rabies virus Cancer
YFV-17D, attenuated yellow fever virus strain 17D.

genomes to genes that allow them to evade host immune Empirical versus rational attenuation. Attenuated pox-
responses. Many such viral genes encode proteins that viruses were first used during the smallpox eradication
affect specific immune defences, including those campaign and have since been further developed and
that act on early innate pathways such as pathways involv- exploited as vaccine vectors. One of these, MVA, was
ing interferons12, pattern recognition receptors (Toll-like passaged more than 570 times in chick embryo fibrob-
receptors; Tlrs)13,14, chemokines15,16 and cytokines17, as lasts, lost approximately 15% of its genome and was
well as those that act on subsequent adaptive responses ultimately replication defective in mammalian cells4,21,22.
by altering major histocompatibility complex processing Conversely, NYVAC was derived from the Copenhagen
of viral peptides18–20. To date, most processes of attenu- isolate of VACV by the deletion of 18 different OrFs
ation — for instance, through continuous in vitro pas- through in vitro recombination, which caused replica-
sage in cell lines — are non-selective of the types of viral tion deficiency 7. Several recent studies have indicated
genes that are lost or become altered. ultimately, that MVA maintains a strong immunostimulatory
viral variants have been selected for their lack of del- capacity, perhaps even greater than that of NYVAC23–26,
eterious effects in small animals, such that they are no whereas a recent primate study of HIV-1 vaccine can-
longer pathogenic and do the host little or no harm. didates showed that these two vaccinia-derived vec-
However, viral genes that become lost or altered during tors induced qualitatively different types of immune
the nonspecific attenuation process may not necessarily responses to the encoded Gag–Pol–Nef (group-specific
be the most optimal in terms of making a virus better for antigen–polymerase–HIV-1 negative factor) and enve-
the immune presentation of recombinant inserts with lope (env) antigens — NYVAC induced a CD4+ T cell-
a more potent adjuvant effect. Given that many viral dominant response, whereas MVA induced a stronger
genomes are now well characterized, future efforts to CD8+ T cell response and accompanying CD4+ T cell
improve the adjuvant properties of viral vectors and responses27. The combined action of many viral pro-
to engineer them to induce different types of responses teins, some of which act on Tlr signalling pathways
will be based on targeting specific viral genes that dif- and other antigen-presenting cell functions, probably
ferentially affect the host’s immune system. For exam- accounts for these differences. until there are better
ple, knocking out viral genes that have evolved to reduce predictive assays for in vivo outcome, the process will
host antiviral immune responses could reduce immune remain largely empirical and dependant on the best
evasion properties and improve adjuvant-specific char- in vivo models to assess the immunostimulatory prop-
acteristics of certain viral vectors; however, anti-vector erties of each vector. Nonetheless, in the case of empir-
responses may preclude the subsequent re-use of that ically attenuated MVA, the rational deletion of genes
vector in an individual. can further enhance immunogenicity, with two reports

NATure reVIewS | MicRobiology VOluMe 8 | jANuArY 2010 | 63

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Initially, heterologous prime–boost protocols with


common vaccine inserts often used a DNA plasmid to
prime the immune system; however, more recently inter-
Genetic complexity and replication potential est has grown in the use of combinations of different
recombinant viral vectors and in how their sequence of
Alum, Smallpox, administration can influence the nature of the induced
HPV and Polio (salk) AdV,
MF59, VEE and yellow fever,
AS01 and
West Nile and MVA and
sindbis measles and immune response. Multiple approaches have now been
virus influenza AAV tested in both animal models and humans, including
CpG FluMist
DNA–MVA, DNA–NYVAC, fowlpox virus (FPV)–MVA,
Adjuvants Virus-like Inactivated Replication- Replicons Live virus
particle viruses defective vectors
influenza–MVA, AdV–MVA, heterologous AdV–AdV,
and DNA–Sendai virus, targeting a wide range of diseases
Figure 1 | A range of co-stimulation — from adjuvants
from malaria, HIV-1, tuberculosis (TB) and hepatitis C
to live viruses. Attenuated replication-defective viral
Nature Reviews | Microbiology
vectors have been developed as part of a strategy to through to cancers (TABLE 2). A consistent observa-
maximize immunogenicity and safety. They lie in the tion throughout all of these studies is the differential
middle of a range of complexity and replication ability of certain vectors to prime or boost responses.
potential, between vaccine technologies at both DNA vaccines, FPV and influenza virus are good prim-
extremes that have been licensed for human use. ing vectors, whereas poxviruses (MVA and NYVAC)
Although clinical vaccine development requires a are consistently able to boost T cell responses that are
long-term programme of work, the licensure of such primed by other means. A large body of data now indi-
Ex vivo interferon‑γ ELISPOT
An ELISPOT (enzyme-linked
vectored vaccines (if proved to be safe and effective) cates that, in general, recombinant AdV can prime and
immunosorbent spot) assay remains a wholly attainable and realistic goal. AAV, boost T cell and B cell responses remarkably well10,31,37–46.
that is used to measure adeno-associated virus; AdV, adenovirus; AS01, adjuvant
More recently, a combination of three unrelated vector
antigen-specific effector T cell system 01 from GlaxoSmithKline; HPV, human papilloma
responses. Typically, virus; MF59, oil-in-water emulsion adjuvant from Novartis; modalities expressing the same vaccine insert has been
splenocytes or peripheral MVA, modified vaccinia virus Ankara; VEE, Venezuelan used47,48. The assessment of these vectors has been aided
blood mononuclear cells are equine encephalitis virus; VLP, virus like particle. by a greater in-depth analysis, in particular using multi-
incubated with the antigen parameter flow cytometry 34, which allows the dissection
(peptide or protein) for
of the different T cell populations — both phenotypically
18 hours, and the cytokines
(typically interferon-γ) that are demonstrating that the deletion of B15R, which encodes and in terms of cytokine production and effector func-
released during that time by a soluble interleukin-1β (Il-1β) receptor, can increase tion — that are induced by such different vectors and
antigen-specific cells are the virus-specific memory CD8+ T cell response28,29. regimes27,37,49–53. Correlating the protective outcomes of
captured by a monoclonal Similarly, by reducing vector-specific gene expression ‘quality versus quantity’ of the T cell responses that are
antibody, thereby allowing
enumeration by a modified
of a recombinant VACV, the CD8+ T cell response to induced by these different immunization regimes and
enzyme-linked immunosorbent the vector itself can be decreased and that against the vector strategies remains a challenge to the field.
assay (ELISA) technique. transgene consequently enhanced30. Future efforts
are set to continue along these lines, aiming to delete Enhancing humoral immunity. More recent work has
Cultured interferon‑γ
more genes, alone and in combination, in an attempt established the use of prime–boost immunization
ELISPOT
A modified ex vivo interferon-γ to rationally enhance the immunogenicity of these regimes to induce B cell as well as T cell responses,
(IFN-γ) ELISPOT (enzyme-linked viral vectors. in particular AdV–MVA or heterologous AdV–AdV
immunosorbent spot) assay regimes39,40. The induction of both arms of the adap-
that involves a prolonged Enhancing immunogenicity tive immune response is likely to be beneficial for
period of in vitro culture of
lymphocytes before the IFN-γ
Prime–boost regimes. In the 1990s, excitement sur- protection against pathogens such as malaria para-
ELISPOT assay. Typically, cells rounded the advent of DNA plasmid vaccines. However, sites and many viruses, in contrast to mycobacteria,
are cultured for 10 days in the it soon became apparent that DNA vaccines by them- for which cellular immunity is probably the domi-
presence of antigen and selves were insufficiently immunogenic to generate T cell nant protective mechanism. AdV vectors seem to be
supplemented with
responses of a magnitude sufficient to protect against able to prime and boost B cell responses, in contrast
interleukin-2. Evidence
indicates that a different difficult diseases in humans31,32. with advances in the to MVA vectors, which are far better able to boost
population of T cells, most quantitative T cell assays that are used in clinical trials, the response than they are to prime it. This may be a
likely central memory T cells including the ex vivo interferon-γ ELISPOT (enzyme-linked result of prolonged high-level antigen expression fol-
that differentiate into effector immunosorbent spot) and cultured interferon-γ ELISPOT lowing AdV immunization that is more beneficial for
T cells during the culture
period, are measured by this
assays as well as multiparameter flow cytometry33,34, viral B cell priming, compared with a short burst of antigen
assay, as compared with the vectors resumed centre stage following the demonstration from MVA that is more suited for B cell boosting 54.
measurement of circulating of their enhanced potency for cellular immune responses Alternatively, studies on plasmacytoid dendritic cells
effector T cells that are compared with other vaccine types35,36. However, unlike have shown that a wave of cytokines, specifically type I
quantified by the immediate
DNA vaccines, after one or two administrations in vivo, interferon (IFN) and then Il-6, is essential for driv-
ex vivo ELISPOT.
host responses to the structural proteins of the viral vec- ing B cell differentiation into effector plasma cells in
Heterologous prime–boost tor itself become self-limiting, leading to diminishing response to influenza virus infection55. Conversely, the
Repeated immunization using responses against the vaccine insert when the vector is priming of transgene-specific antibody responses by
different vaccines; used to used for multiple immunizations. The development of AdV vectors seems to negatively correlate with type I
stimulate a better immune
response when a single
prime–boost protocols has been required to overcome IFN production 56,57, which is also associated with
application is not sufficient to these anti-vector responses while maximizing the host reduced transgene expression58. In the case of MVA,
induce a protective response. response to the vaccine insert. E3L confers resistance to type I IFN and is essential for

64 | jANuArY 2010 | VOluMe 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 2 | Viral-vector vaccines and prime–boost immunization regimes in clinical development for human use
Recombinant viral-vector Target pathogen Target antigen Vaccine details Developer clinical phase
immunization regime or disease
HAdV-5–HAdV-5 Plasmodium CSP and AMA1 Monocistronic vectors; NMRC and GenVec I/IIa
falciparum malaria HAdV-5 (del E1,E3,E4);
administered as a mixture
AdCh63–MVA P. falciparum malaria ME–TRAP, MSP1 Monocistronic vectors University of Oxford I/IIa
and AMA1 and Okairòs
HAdV-35 P. falciparum malaria CSP None Crucell I
MVA Mycobacterium 85A BCG–MVA (containing University of Oxford and IIb efficacy field
tuberculosis the gene encoding 85A) Emergent BioSolutions trial (South
Africa)
HAdV-35 M. tuberculosis 85A, 85B and 10.4 Fusion antigen; (r)BCG Aeras and Crucell I
prime, HAdV-35 boost,
AERAS-402
MVA Influenza A virus NP and M1 Fusion antigen University of Oxford I
HAdV-5 Influenza virus NP Intranasal administration GenVec I
Flavivirus YFV-17D (live WNV preM-Env Strain NY99 Sanofi Pasteur II
chimeric virus)
Flavivirus YFV-17D (live Dengue virus preM-Env Strains PUO-359, Sanofi Pasteur II
chimeric virus) (subtypes 1–4) PUO-218, PaH881 and
1228
Flavivirus YFV-17D (live Japanese encephalitis preM-Env Strain SA-14-14-2 Sanofi Pasteur III
chimeric virus) virus pre-registration
DNA–NYVAC HIV-1 Gag–Pol–Nef and Clade C EuroVacc I
Env
DNA–MVA HIV-1 Gag, Pol and Env Clade B GeoVax IIa
DNA–HAdV-5 HIV-1 Gag–Pol–Nef and HAdV-5 (del E1,E3,E4) VRC, NIAID (NIH) and II
Env GenVec
ALVAC containing the gene HIV-1 gp160 and gp120 ALVAC–HIV Sanofi Pasteur and III
encoding HIV-1 gp160, and vCP1521 prime and Global Solutions for
protein gp120 AIDSVAX–gp120 subtype Infectious Diseases
B/E boost
HAdV-5 and HAdV-6 HIV-1 Gag–Pol–Nef MRKAd5 and MRKAd6 Merck I
trigene vaccines
VACV and ALVAC or Cancer CEA TRICOM vectors NIH I/II
FPV with or without (pancarcinoma) co-express B7.1, ICAM1
combination therapy and LFA3
MVA Colorectal, renal and 5T4 (solid tumors) TroVax Oxford Biomedica I–III
prostate cancer
MVA Lung cancer MUC1 TG4010 vector Transgene SA IIb
co-expresses IL-2
Lentivirus Melanoma MART1 TCR immunotherapy Caltech and UCLA I
HAdV-6 and AdCh3 Hepatitis C virus NS3, NS4 and NS5 Fusion antigen Okairòs and the I
‘NSmut’mutation in NS5 University of Oxford
85a, mycobacterial mycolyl transferase 85A antigen; AdCh63, chimpanzee adenovirus serotype 63; ALVAC, attenuated canarypox virus; AMA1, apical-membrane
antigen 1; BCG, Mycobacterium bovis bacille Calmette–Guérin; CEA, carcinoembryonic antigen; CSP, circumsporozoite protein; del, deleted; Env, envelope protein;
FPV, fowlpox virus; Gag, group-specific antigen; gp160, glycoprotein 160; HAdV-5, human adenovirus serotype 5; ICAM1, intercellular adhesion molecule 1; IL-2,
interleukin-2; LFA3, lymphocyte function-associated antigen 3; M1, matrix 1 antigen; MART1, melanoma antigen recognized by T cells 1; ME–TRAP, thrombospondin-
related adhesion protein fused to a multi-epitope string; MSP1, merozoite surface protein 1; MUC1, mucin 1; MVA, modified vaccinia virus Ankara; Nef, HIV-1
negative factor; NIAID (NIH), National Institute of Allergy and Infectious Diseases (US National Institutes of Health); NMRC, US Naval Medical Research Center NP,
nucleoprotein antigen; NS, non-structural protein; NYVAC, New York attenuated vaccinia virus; preM, pre-membrane protein; (r)BCG, recombinant BCG; TCR,
T-cell receptor; TRICOM, triad of costimulatory molecules; UCLA, University of California, Los Angeles (USA); VACV, vaccinia virus; VRC, Dale and Betty Bumpers
Vaccine Research Centre; WNV, West Nile virus; YFV-17D, attenuated yellow fever virus strain 17D.

sustained growth in chick embryo fibroblasts, and an is observed with this vector. ultimately, a much better
E3L-knockout virus induces enhanced levels of chicken understanding of how cytokines can affect the induc-
type I IFN and Il-6 in cell culture59,60. If B cell priming tion of B cell responses by different virus families is
by MVA occurs in a similar manner to that induced by essential to improve the rational design of vectors and
the influenza virus, the E3L-mediated immune-evasion prime–boost strategies that can be tailored to induce
mechanism could explain the weak B cell priming that optimal antibody responses.

NATure reVIewS | MicRobiology VOluMe 8 | jANuArY 2010 | 65

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Circumventing pre-existing immunity. Viruses that antigens have been developed as vectors to deliver genes
commonly infect the human population bring with to specific cells in vivo for intracellular immunization75.
them the problem of pre-existing immunity. For cer- This is made possible in part by the minimal expression
tain viral vectors, such as those based on human AdVs of lentivirus-vector genes and in part because repeated
and herpesviruses, this can be a major obstacle. For in vivo administration (as in traditional immunization) is
example, a candidate HIV-1 vaccine based on the not the aim. Indeed, lentiviruses are currently being used
human AdV serotype 5 (HAdV-5) vector was recently to ‘engineer immunity’ for cancer and HIV-1 (REFS 76,77).
assessed in a Phase II trial that was conducted in Transduction of haematopoietic stem cells with lentivi-
individuals who were at high risk of HIV-1 infection. ruses containing T cell receptor genes has now entered
The trial was halted owing to lack of efficacy, but at clinical testing for cancer immunotherapy. Substantial
the time a nonsignificant trend was observed towards preclinical progress has also been made using these vec-
increased acquisition of HIV-1 infection in those indi- tors to transduce human CD34+ cells with B cell recep-
viduals with higher titres of pre-existing HAdV-5- tor genes encoding neutralizing monoclonal antibodies
neutralizing antibodies at the time of enrollment 52,61. against HIV-1 (REF. 76). Strategies for targeting lentiviral
This issue provoked intense debate about the use of vectors to specific cell types, such as skin dendritic cell
this commonly occurring human virus as a vaccine (DC) subsets, have also been developed; for example, a
vector, but despite this setback for the HAdV-5 vec- mutated version of the Sindbis virus fusogenic protein
tor platform other strategies are actively being pursued that retains the ability to bind DC-SIGN (dendritic cell-
to provide viable alternatives that can overcome pre- specific ICAM3-grabbing non-integrin) proteins has
existing vector immunity 62. These include modifying been developed78. The development of further specific
the vector to express heterologous adenovirus hexons63 targeting technologies could greatly enhance the rational
or using human adenoviruses of rarer serotypes40,44,64,65 or design of viral-vector vaccines that induce tailored and
those from other species such as chimpanzees 66–70. effective immune responses for the prevention of a wide
Importantly, all such new vectors, especially those range of infectious diseases and cancers.
exogenous to a species, will need to clear the regulatory,
safety and manufacturing hurdles. Advances in veterinary vaccines
The most successful applications of viral-vector vaccines
Empirical versus rational design of vectored vaccines. have undoubtedly been in the veterinary field. This is
with our expanding knowledge of immunology and in contrast to the development of vaccines for human
viral biology, a new era of science in vaccinology has been use, which entails more stringent regulatory require-
established that has provided a future basis for the rational ments, step-wise clinical trials in human volunteers and
development of viral-vector vaccines. Optimizing subunit a longer time to licensure and return on investment.
vaccine antigen design and understanding vector tropism, At least 12 viral-vector vaccines are currently licensed
including target antigen processing and presentation for veterinary use (TABLE 3), and many more are under
in vivo, is crucial to this approach. In the case of VACV, it development (for a review, see REFS 79,80). Nonetheless,
has been shown that the route of administration71, as well these technologies still face an uphill struggle against
as the poxviral promoter used to drive transgene expres- competition from classical inactivated vaccines, which
sion72, can influence the extent to which antigens are are often easier to produce. The challenges of control-
directly or cross-presented to T cells. Similarly, new data ling avian influenza in poultry are driven by econom-
indicate that orthopoxvirus structural antigens, which ics, and require mass vaccination strategies such as
are expressed late in infection and normally prevented administration in drinking water, by aerosol spray or by
from entering the direct presentation pathway owing to immunization in ovo81 (FIG. 2). Keeping the costs of these
an abortive infection in antigen-presenting cells, are also routes of administration low while maintaining vaccine
specifically hidden in viral factories, thereby escaping the efficacy poses considerable challenges for recombinant
cross-presentation pathway and hampering the induction viral-vector technologies. One successful approach has
of poxvirus-specific CD8+ T cell responses73. This is not been to use the new recombinant Newcastle disease
the case for foreign antigens driven by poxvirus early or virus (NDV), which provides a bivalent method for
late promoters, because these antigens are not usually tar- the control of two different avian pathogens at once
geted to such poxvirus factories. It is important that the by in ovo immunization82. New vaccine strategies for
consequences of such observations for vaccine-induced another important and highly contagious veterinary
T cell responses against the vector and the transgene are pathogen, foot-and-mouth disease virus (FMDV) 83,
better understood, and these data may begin to explain are being explored using replication-defective HAdV-5.
why poxvirus vectors such as MVA have been shown to Protective immunity has been elicited in cattle and
be effective at re-boosting immune responses in humans swine 7 days after a single immunization with recom-
12 months after a previous immunization with the same binant HAdV-5 expressing the FMDV capsid and pro-
vector 74. tease antigens84,85. unlike for inactivated, whole-virus
vaccines, the ability to distinguish infected from vacci-
‘Engineering’ immunity. In contrast to the formidable nated animals (DIVA; see BOX 1) using this vector vac-
complexities of the anti-vector immunity that is associ- cine platform, coupled with greatly increased biosafety,
ated with persistent DNA viruses such as adenoviruses, would make this effective technology highly desirable
engineered lentiviruses devoid of much of their structural for FMDV-free countries. Similarly, with respect to

66 | jANuArY 2010 | VOluMe 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 3 | Viral vector veterinary vaccines licensed for commercial use


Recombinant viral Target pathogen Target Target antigen brand name Distributor
vector species
ALVAC (plus tetanus toxoid Equine influenza virus Horses HA (Kentucky and ProteqFlu-Te (Europe) Merial
and Carbopol adjuvant) Newmarket strains) Recombitek (USA)
ALVAC West Nile Virus (WNV) Horses PreM–Env Recombitek Equine WNV Merial
ALVAC Rabies virus Cats Glycoprotein G Purevax Feline Rabies Merial
ALVAC Feline leukaemia virus (FeLV) Cats Env, Gag–Pol Purevax FeLV Merial
ALVAC Canine distemper virus Dogs HA and F RECOMBITEK rDistemper Merial
ALVAC Canine distemper virus Ferrets HA and F Purevax Ferret Distemper Merial
Fowlpox virus (FPV) Avian influenza virus and FPV Poultry H5 HA Trovac AI H5 Merial
FPV Newcastle disease virus (NDV) and FPV Poultry HN and F Vectormune FP-N Biomune
Vaccinia virus Rabies virus Wildlife Glycoprotein G Raboral Merial
NDV (LaSota strain) Avian influenza virus and NDV Poultry H5 HA NewH5 Avimex
Flavivirus YFV-17D WNV Horses preM-Env of WNV in PreveNile Intervet
(live chimeric virus) YFV-17D backbone
HVT (live chimeric virus) IBDV and Marek’s disease virus Poultry VP2 of IBDV in HVT Vaxxitek HVT + IBD Merial
backbone
ALVAC, attenuated canarypox virus; Env, envelope protein; Gag, group-specific antigen; F, fusion antigen; H5 HA, HA from influenza virus H5; HA, haemagglutinin;
HN, haemagglutinin–neuraminidase protein; HVT, Turkey herpesvirus; IBDV, infectious bursal disease virus; Pol, polymerase; preM, pre-membrane protein; VP2,
viral protein 2; YFV-17D, attenuated yellow fever virus strain 17D.

important respiratory pathogens of livestock such as first time in human clinical trials — a goal that, despite
bovine tuberculosis, recombinant HAdV-5 or MVA extensive efforts, has yet to be realized by DNA plas-
Pre‑erythrocytic‑stage vaccine vectors are being explored. These vectors, mid vaccine technologies alone. Key to this advance
malaria expressing the mycobacterial mycolyl transferase 85A has been the development of AdV-vectored vaccines
Plasmodium sporozoites antigen, have both been described as effective boosting — AdV has a remarkable ability to prime immune
invade hepatocytes and agents for the Mycobacterium bovis bacille Calmette– responses against a transgene that can be boosted to
develop over a period of
6–7 days. This time is known
Guérin (BCG) vaccine in cattle86–88. Challenge experi- high levels with a second vector that is recombinant
as the liver or pre-erythrocytic ments are now required to assess protection with these for the same antigen, typically MVA or a second
stage and precedes BCG prime–vector boost regimes in herds of immu- AdV of heterologous serotype. Similarly, both MVA
subsequent blood-stage nized cattle following direct exposure and also in a and AdV can substantially boost immune responses
infection. The liver stage is
natural transmission setting. If more effective than primed by other means, for example the BCG vaccina-
clinically silent and infected
persons do not show any signs the BCG vaccine alone, the use of vectored vaccines tion. A comparison of these two widely used vectors is
or symptoms of infection. to boost BCG-primed responses could hold promise provided in TABLE 4.
for the control of bovine TB, which is estimated to
Blood‑stage malaria cost the world approximately uS$3 billion per annum. Malaria vaccines. recent progress has been made
During the liver stage of
malaria infection, parasites
Importantly, the cattle TB setting continues to provide using a mixture of two HAdV-5 vectors expressing the
develop into merozoites in useful immunological insight for the design and testing pre-erythrocytic-stage malaria antigen circumsporozoite
infected liver cells. These of candidate human TB vaccines89. protein (CSP) or blood-stage malaria antigen apical mem-
rupture out into the blood after brane antigen 1 (AMA1)90. Following a single immu-
a period of 6–7 days and
Vaccines for human infectious disease nization in human volunteers, an impressive mean
undergo a continuous cycle of
asexual growth in erythrocytes, Viral-vector vaccines were originally developed to response to each antigen of 500–1,000 spot-forming units
lasting approximately induce protective T cell responses against intracellular (SFu) per million peripheral blood mononuclear cells
48 hours. pathogens. However, to date neither a viral-vector vac- (PBMCs) was measured by ex vivo IFN-γ elISPOT.
cine nor a vaccine that acts directly by T cell-mediated Depletion studies defined the response as a mixed CD8+
Spot‑forming unit
(SFU). The ELISPOT assay is
immunity (with the possible exception of BCG) has and CD4+ T cell response, with the CD8+ phenotype three
enumerated by a modified been licensed for human use. This situation is com- to five times as frequent as the CD4+ phenotype among
enzyme-linked immunosorbent pounded by the fact that these new approaches have IFN-γ-secreting responder cells, as measured by flow
assay (ELISA) technique. During been chosen for some of the most complex human cytometry (M. Sedegah and T. richie, personal com-
the development stage of the
pathogens for which conventional vaccine approaches munication). In an effort to circumvent the problem
assay, so-called ‘spots’ are
formed on the membrane of have not been effective. These include Plasmodium of pre-existing host responses to HAdV-5, recom-
the plates that are used. After falciparum malaria, Mycobacterium tuberculosis and binant adenoviruses of rare human serotypes or from
counting the spots, results are HIV-1. Nonetheless, substantial progress has been non-human primates are also under development as
routinely expressed as SFU per made with recombinant viral-vaccine technologies. A malaria vaccine candidates. Priming and boosting with
million cells, given that it is
possible to calculate this from
list of vectors that are currently in development for heterologous AdVs, such as HAdV-35 and HAdV-11,
the number of cells known to human use is presented in TABLE 2. Impressive levels of is being explored preclinically 44 and, encouragingly,
have been used in the assay T cell immunogenicity are now being reported for the a recombinant chimpanzee adenovirus serotype 63

NATure reVIewS | MicRobiology VOluMe 8 | jANuArY 2010 | 67

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

a antibody responses against blood-stage infection39, as


Intranasal Aerosolized well as T cell responses that were partially effective
+ Mucosal and lung immunity + Mucosal and lung immunity against the preceding liver-stage infection91.
– Safety issues in humans + Mass vaccination with nebulizer or
inhaler or by spraying animals Heterologous poxvirus and DNA–poxvirus immu-
– Safety issues in humans nization regimes established the value of prime–boost
Oral approaches in humans, achieving much stronger
+ Cheap mass vaccination
+ Mucosal immunity Intradermal T cell responses in clinical trials than single-vector
– Stability in the GI tract + Clinically relevant approaches31,38,92. Nonetheless, efficacy was limited,
+ Systemic immunity
– Needle required (new and so Phase IIa malaria challenge studies of these new
Intramuscular needle-free ‘vaccine patch’ AdV-vector vaccines, scheduled for 2009–2010, will be
+ Clinically relevant transcutaneous technologies important. even if successful, these promising vaccine
+ Systemic immunity are in development)
– Needle required candidates will face further challenges, including the
potential problems of high-level pre-existing immu-
Intraperitoneal nity to the HAdV-5 vector in the human (especially
Intravenous
+ Used for some candidate + Used for some candidate
African) population93. Furthermore, it remains to be
cancer vaccines cancer vaccines seen how immunogenic AdV vectors will be in the tar-
– Safety issues –Safety issues get population of young African children. In children
between 1 and 6 years of age, living in areas of high
b malaria transmission, reduced vaccine immunogenic-
ity has been reported in comparison with naturally
In ovo immune African adults and malaria-naive volunteers
+ Mass vaccination of animals
– Labour intensive in developed countries94.

Figure 2 | Possible routes of immunization for human and veterinary vaccines.


Nature Reviews | Microbiology Tuberculosis vaccines. Similar to the situation for
Potential advantages (+) and disadvantages (–) are shown for various possible routes malaria, highly promising vectored-vaccine pro-
of vaccination. a | Intramuscular immunization forms the mainstay approach to human grammes are under way for human TB. These remain
vaccination, with minor exceptions such as Mycobacterium bovis bacille Calmette–Guérin somewhat hindered in the absence of an experimental
(BCG) vaccine that are routinely given intradermally. Work is now progressing to human challenge model, although efforts are under way
develop needle-free transcutaneous devices such as patches coated with vaccine that to develop M. bovis BCG as a surrogate challenge for
can be self-applied in the absence of a trained individual — an obvious advantage for M. tuberculosis in humans86. Consequently, efficacy field
mass and emergency vaccination. Certain cancer vaccines may also be administered trials remain essential once candidate vaccines have been
intravenously, but in general the safety concerns remain high for this route. Mucosal
selected — a process that is both long and expensive. A
immunity may be enhanced in the gastrointestinal (GI) and respiratory tracts through
the use of oral or aerosolized vaccines. This may enhance protective immunity to BCG prime and MVA–85A (an MVA vector contain-
pathogens, for example against tuberculosis infection in the lungs or HIV infection in ing the gene encoding the mycobacterial mycolyl trans-
mucosal tissue. Recombinant adenovirus human serotype 41 (HAdV-41), a natural ferase 85A antigen) boost regimen is now entering into
enteric pathogen, may prove to be a more suitable vector for the oral route, given that a Phase IIb efficacy study in South Africa and has so far
it has been reported to stimulate immune responses under acidic conditions127. demonstrated high levels of T cell immunogenicity both
However, there is still much work to be done in terms of demonstrating and in the uK and Africa, and an excellent safety record in
maintaining vaccine safety, efficacy, stability and dosing before these routes of over 500 people including adults, adolescents, infants,
vaccination can be widely used in humans. Keeping the costs of using any of these less HIV-positive individuals and those who are latently
standard routes of administration in humans low while maintaining vaccine efficacy infected with M. tuberculosis 51,95–97. The TB vaccine field
and, more importantly, safety poses considerable challenges for recombinant
is also developing AdV-vector candidate vaccines and
viral-vector technologies, and much proof-of-concept research is still required.
b | Veterinary vaccines also make use of standard parenteral immunization routes using similar approaches to circumvent human anti-
(intradermal and intramuscular), but the use of alternative routes such as administration HAdV-5 responses by using rare human serotypes such
in drinking water, by aerosol spray or by immunization in ovo are driven by economics, as HAdV-35. An HAdV-35 vaccine candidate expressing
less stringent safety requirements in animals and the frequent need for mass a fusion of three TB antigens, 85A, 85B and 10.4 (REF. 98),
vaccination strategies. can boost T cell responses that are primed by either BCG
or a recombinant BCG in non-human primates99, estab-
lishing an approach that is being tested in humans100. A
(AdCh63) 70 expressing the pre-erythrocytic-stage mixed CD8+ and CD4+ T cell response has been reported
antigen thrombospondin-related adhesion protein in Phase I studies in both the uSA and South Africa137,
fused to a multi-epitope string (Me-TrAP) followed but the contribution of CD8+ T cell responses to human
by a boost with MVA elicits mean levels of >1,000 SFu immunity against TB remains unclear. For both leading
per million PBMCs against the malaria antigen in viral-vector vaccine candidates, preclinical studies are
human volunteers (A. Hill, personal communication). now actively pursuing the potential for aerosolized rather
Further preclinical studies have also reported the use than systemic delivery, possibly using nebulizers, to try to
of an AdV–MVA regime in mice to induce high-titre enhance the mucosal immunity that may be more pro-
antibody responses and potent T cell responses. This tective against M. tuberculosis infection50 (FIG. 2). Further
specific HAdV-5–MVA regime, using vectors that are preclinical data from non-human primates are necessary
recombinant for the blood-stage malaria antigen mero- if we are to address these outstanding immunological
zoite surface protein 1 (MSP1), induced protective questions.

68 | jANuArY 2010 | VOluMe 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Differentiation of infected from vaccinated animals are associated with protection, as some evidence sug-
gests106, then a viral-vector vaccine approach could
In the case of certain livestock infections, effective conventional vaccines are available prove highly cost-effective in comparison with sea-
but cannot be used without interfering with disease monitoring based on serological sonal influenza immunizations aimed at inducing
testing. Both natural infection and vaccination result in a seropositive animal. A classic
antibodies against the variant-specific surface coat
example is foot-and-mouth disease virus (FMDV) in cattle: although inactivated
antigens.
vaccines can be used effectively to control disease, they are not used routinely in
‘FMDV-free’ countries, because this would compromise a country’s disease-free status
and lead to substantial economic repercussions for international trade. FMDV vaccines HIV. Perhaps a greater challenge for vaccine devel-
have been used to reduce the spread of the disease during an outbreak, although the opment than the seasonal variability and antigenic
vaccinated animals must be subsequently slaughtered to enable re-establishment of drift of influenza virus is the constantly evolving HIV
that country’s FMDV-free status126. Other examples include bovine herpesvirus type 1 epidemic. T cell-inducing vaccines for HIV-1 aim to
infection of cattle (which leads to infectious bovine rhinotracheitis), classical swine fever reduce viral load and CD4+ T cell loss, prolong survival
and Aujeszky’s disease (caused by pseudorabies virus or suid herpesvirus 1) in pigs79. and reduce transmission107. renewed efforts are focused
One alternative to prevent this scenario is the development of vaccines that allow on poxvirus regimes using attenuated canarypox
the differentiation of infected from vaccinated animals (DIVA). This is achieved by the
virus (AlVAC), MVA or NYVAC. each of these poxvirus
use of subunit vaccines (for example, viral vectors expressing a single antigen from
vaccine vectors has been developed and evaluated
the pathogen) or the selective deletion of identified genes from conventional
attenuated vaccines. Vaccination and natural infection can therefore be distinguished for their ability to induce HIV-1-specific immune
serologically using suitable diagnostic assays that identify antibody responses against responses in humans. The most recent Phase III
an antigen in the vaccine versus those against an antigen that is deleted or absent HIV-1 clinical trial used a combination of AlVAC
from the vaccine, to which the animal is only exposed by natural infection with the and AIDSVAX (monomeric HIV-1 glycoprotein 120)
wild-type pathogen. in a large, community-based population in which
the risk of transmission is based on heterosexual life
style. Although the study found an encouraging trend
Flaviviruses. Yellow fever, a viral disease, is re-emerging towards prevention of HIV-1 infection (31.2% effi-
in naive humans owing to a shift in its epidemiology. A cacy), there was no benefit for post-infection virus
highly effective attenuated vaccine (YFV-17D) has been load or CD4+ T cell counts108. In preclinical efficacy stud-
used in humans for more than 70 years101. with the ies in non-human primates, control of virus load and
development of live attenuated flavivirus vectors based preservation of CD4+ T cell counts have been reported
on YFV-17D, a programme of rational development of when certain pox-based vectors have been used in het-
flavivirus vaccines has evolved102 in which the preM– erologous prime–boost combinations to boost DNA-
Env genes, encoding the flavivirus pre-membrane primed responses41,48. They are able to induce antibody
and envelope proteins, have been replaced with those responses against env and elicit T cell responses of
from west Nile virus, japanese encephalitis virus or varying levels. In humans, T cell responses are of a
dengue virus to generate chimeric vectors. It has been mixed CD4 + and CD8 + phenotype and are broadly
far more difficult to stably express non-flavivirus anti- multifunctional in terms of cytokine production37,109,110.
gens in these vectors, but insertion of non-flavivirus Nonetheless, the consensus remains that these vaccine
epitopes has been reported103, as has the stable inser- regimes need to be improved to increase the magnitude
tion of GFP 104. Following extensive and successful and breadth of the CD8+ and CD4+ T cell responses
safety and immunogenicity testing in Phase I–III trials, against antigens such as Gag and Pol, and to induce high-
the chimeric flavivirus for japanese encephalitis may titre neutralizing antibodies or high-avidity antibodies
well be the first viral-vector vaccine to be licensed for against the native trimeric env protein.
use in humans105. The development of HAdV-5 recombinants for
HIV-1 vaccination produced the most immunogenic
Influenza virus. The development of a cross-strain candidate vaccine to date, which was taken forward
influenza vaccine would be a major and long-sought into a human efficacy trial, designated STeP. The
achievement, especially given the continued seasonal HAdV-5 recombinants remained a leading approach
emergence of new influenza variants, the current for HIV-1 vaccine developers until the STeP trial
H1N1 influenza A virus pandemic and the possibility was terminated on the grounds of futility 52,61,111. even
of an outbreak of highly pathogenic avian influenza. though much stronger T cell responses have been
The induction of protective T cell responses against elicited in malaria and TB prime–boost vaccine pro-
conserved internal influenza antigens, rather than grammes in humans, the failure of the STeP trial was
variant-specific antibody responses against surface suggested by some as grounds for dismissal of the
haemagglutinin and neuraminidase antigens, is one concept of vaccines aimed at inducing T cell-mediated
approach that may stand a chance of success. Phase I immunity. encouragingly, however, after some initial
trials of a new candidate cross-strain influenza vac- despondency, discussion has refocused on the further
cine are now under way, using an MVA vector that optimization of vaccine immunogens and technolo-
expresses the highly conserved internal nucleoprotein gies62. Indeed, now that far higher T cell induction is
and matrix 1 (NP–M1) influenza A antigens. This vec- being reported for non-HIV infections, it is time to
tor can boost T cell responses in individuals who are match these in the HIV vaccine field. Vaccine develop-
presumably primed by natural exposure (S. Gilbert, ers for HIV-1 have already begun to focus on other, less
personal communication). If these T cell responses established, chimeric viral-vector technologies. Sendai

NATure reVIewS | MicRobiology VOluMe 8 | jANuArY 2010 | 69

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 4 | Comparison of human adenovirus and modified vaccinia virus Ankara viral vectors
Human adenovirus (HAdV) Modified vaccinia virus Ankara (MVA) Refs
Family Adenoviridae Poxviridae None
genus Mastadenovirus Orthopoxvirus None
Species Six species (A–F) Vaccinia virus None
Serotypes 51 HAdV serotypes Attenuated MVA and NYVAC None
Virion 60–90 nm, non-enveloped icosahedral particles with a 300 x 250 nm, enveloped, rounded or brick-shaped 64,128,
protein capsid (240 hexons and 12 pentons) virion of multiple forms: IMV, IEV, CEV and EEV 129
genome 34–43 kb double-stranded DNA with 15 transcriptional 178 kb double-stranded MVA DNA with 193 ORFs 64,130
units: E1–E5, IX, IVa2, L1–L5 and VA RNA and 177 genes (including 25 pseudogenes)
Tropism Broad tropism Broad tropism
cell receptors Coxsackie adenovirus receptor, heparin-sensitive Complex entry mechanisms 64,129,
receptor, CD46, CD80 and CD86 (subgroup B) 131,132
Vector generation Genomic plasmid with the deletion of one or more Attenuated by passage in CEFs (MVA) or by genetic 28,133,
essential genes (E1, E3 and E4) and the transgene deletion (NYVAC); the transgene is cloned into a 134
cloned into the deletion site; the vector is transfected plasmid shuttle vector and the vector is recombined in
into a packaging cell line (HEK293 or PER.C6), and a cell culture into the parental virus genome; a marker
pure recombinant virus is obtained gene allows selection of the recombinant virus
insert capacity ~7.5 kb in E1- and E3-deleted HAdV-5 At least 25 kb in MVA 134,135
Transgene promoters Cytomegalovirus, β-actin and elogation factor 1α from Natural or synthetic early and late poxviral 5,10,67,
humans, cytomegalovirus from Mus musculus and promoters, for example P7.5, SSP, mH5 and I1L 72,90,
ubiquitin 136
Key differences in virion, genome, vector tropism, vector generation and design of transgene inserts are described. CEFs, chick embryo fibroblasts; CEV,
cell-associated enveloped virus; EEV, extracellular enveloped virus; HAdV-5, human adenovirus serotype 5; IEV, intracellular enveloped virus; IMV, intracellular
mature virus; NYVAC, New York attenuated vaccinia virus; VA, virus-associated.

virus, cytomegalovirus (CMV), reovirus, herpes sim- effective immune responses (an issue shared with
plex virus and NDV are currently under evaluation by many infectious-disease vaccines), but has also tried
a range of groups, including the International AIDS to do so in the face of the unique and complex chal-
Vaccine Initiative (IAVI). The recent use of CMV as lenge of breaking tolerance against self-antigens —
a vector has shown that this vector has substantial which are, by definition, either weakly immunogenic
efficacy in the rigorous SIVMAC (simian immunode- or functionally non-immunogenic in an immuno-
ficiency virus of the macaque) rhesus macaque chal- logically compromised environment. Intriguingly,
lenge model, exceeding the suppression of virus load there seems to be hope for certain cancer vaccines in
that was observed with another study using HAdV-5 specific settings. The use of therapeutic viral-vector
(REFS 112,113). Alternative strategies will probably com- vaccines targeting the carcinoembryonic antigen (CeA),
bine the most immunogenic prime–boost regimes that in combination with local radiation, chemotherapy or
are currently available, such as AdV–MVA or heterolo- cytotoxic T lymphocyte protein 4 (CTlA4)-specific
gous AdV–AdV regimes or even triple combinations, monoclonal antibodies, has led to improved survival
to facilitate the recruitment of both T cell and B cell in some patients117. In this setting, an ‘antigen cascade’
responses 41,114. A recombinant HAdV-26–HAdV-5 has been observed, whereby T cell responses to the
regime has recently been described that can induce a CeA were noted not only post-vaccination but also as
T cell response against SIVMAC Gag in rhesus macaques de novo responses to other tumour-associated antigens
that is stronger, more polyfunctional and more pro- (TAAs), which were possibly induced following more
tective than the response induced by a homologous effective immune targeting of cancerous cells. MVA
HAdV-5 regime40. Indeed, a mean of >2,000 SFu per vectors expressing TAAs are also currently in advanced
million PBMCs was achieved in these macaques40, in clinical trials as an addition to first-line therapies for
Carcinoembryonic antigen
(CEA). An oncofetal membrane
comparison with around 300 SFu per million PBMCs renal, prostate and non-small-cell lung cancers118–120.
glycoprotein and member of on average in volunteers in the HAdV-5 homologous recent immunological analyses have indicated that
the immunoglobulin gene prime–boost STeP trial52. Future studies will reveal there is a significant correlation between the level of
superfamily. CEA is expressed whether these approaches can consistently improve activated natural killer cells at baseline and the subse-
in fetal gastrointestinal tissue,
on the immunological and viral control bench- quent efficacy in patients receiving chemotherapy plus
but is also overexpressed in
many human carcinomas of marks that were set by the combination of DNA and MVA–MuC1–Il-2 for lung cancer 121. The level of acti-
the colon, rectum, breast, HAdV-5 vaccine vectors in the SIVMAC rhesus macaque vated natural killer cells could potentially be used as
lung and pancreas. CEA can challenge model. a predictive biomarker for patients who might benefit
be used as a serological from viral-vector immunization combination therapy.
marker of malignancy and is a
tumour-associated antigen that
Cancer vaccines. The cancer vaccine field (for com- As with immune-compromising viral infections, the
is a widely used target for prehensive reviews, see REFS 115,116 ) has not only use of cancer vaccines in a therapeutic setting will
cancer vaccines. suffered the problem of inducing strong, durable, require the re-establishment of a functional and robust

70 | jANuArY 2010 | VOluMe 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

immune response that can be sustained in a potentially in improved antigen presentation, improved adjuvant
tolerogenic setting — a setting induced as part of the effects and intentional skewing of immune responses are
immune evasion strategy of some cancers. In this light, important new lines of investigation. New technologies
combination therapies hold the greatest hope in the include the easier manipulation of poxvirus genomes by
cancer vaccine field, although preclinical data from ‘recombineering’ in bacterial artificial chromosomes28,124,
mouse122 and non-human primate123 studies indicate and high-throughput transgene insertion into AdV vec-
that AdV vectors expressing candidate TAAs can break tors, including the use of bacterial tet operator elements
tolerance. It remains to be seen whether candidate in the transgene promoter to shut down the transcrip-
cancer vaccines based on AdV vectors can achieve tion of transgenes that may be toxic or deleterious to
even better immunogenicity in human trials than that viral growth125. The use of such technologies will be
observed with poxvirus-based immunotherapies. adopted for a range of different vector platforms, there-
fore allowing the screening of novel viral vaccine vectors
Future directions that remain safe but show improved immunogenicity for
Most viruses have evolved mechanisms to evade host the recombinant vaccine antigens that they deliver.
immune responses and to modulate the subsequent The development of viral-vector vaccines for use
inflammatory environment in different ways. These in animals and humans faces substantial challenges,
evasive attributes are contrary to the immunostimula- both in the scientific and regulatory arenas. However,
tory properties of ideal vaccine adjuvants. However, in these technologies are finally achieving the efficacy
the new era of genomics, specific viral genes encoding in animals and strong immune responses in humans
factors with immune-evasive properties are being tar- that have been sought after for so long, in particular
geted to enhance these immunostimulatory properties. in the fields of malaria, TB and influenza. The transla-
Not only is this new science in vaccinology using well- tion of these new technologies will facilitate vaccine
established viral vectors, but as new viruses are discov- development against some of the toughest challenges
ered and characterized their utility as vaccine vectors in the field, such as cancer and HIV-1. The licensure of
is also being explored. For instance, investigations are replication-deficient viral-vector vaccines is feasi-
under way using multiple strategies to find alternative ble, given that they lie between the two extremes of
vectors to circumvent the problem of pre-existing vec- protein-in-adjuvant vaccines and live viral vaccines
tor immunity. These vectors, combined with new vac- — both of which, after considerable effort, have been
cine prime–boost regimes that will induce stronger, licensed before. with the first chimeric flavivirus
broader and more durable effector responses, offer vaccine against japanese encephalitis virus pending
promising solutions to immunological problems. The regulatory approval, the road is paved for many other
emergence of many new vector systems must not dis- viral-vector vaccines in advanced clinical develop-
tract from the great opportunities to improve current ment. The next 25 years will prove a challenging but
vectors. Approaches that knock out viral genes and result exciting time for this renaissance in vaccine science.

1. Smith, G. L., Mackett, M. & Moss, B. Infectious in diversified prime and boost vaccine regimens to mechanism of virus modulation of the host response
vaccinia virus recombinants that express hepatitis B elicit therapeutic antitumor responses. Cancer Res. to infection. Cell 71, 153–167 (1992).
virus surface antigen. Nature 302, 490–495 (1983). 63, 7942–7949 (2003). 18. Blair, G. E. & Blair‑Zajdel, M. E. Evasion of the immune
2. Moss, B., Smith, G. L., Gerin, J. L. & Purcell, R. H. Live 10. Schneider, J. et al. Enhanced immunogenicity for system by adenoviruses. Curr. Top. Microbiol.
recombinant vaccinia virus protects chimpanzees CD8+ T cell induction and complete protective efficacy Immunol. 273, 3–28 (2004).
against hepatitis B. Nature 311, 67–69 (1984). of malaria DNA vaccination by boosting with modified 19. Dasgupta, A., Hammarlund, E., Slifka, M. K. & Fruh, K.
Along with reference 1, this is the first major study vaccinia virus Ankara. Nature Med. 4, 397–402 Cowpox virus evades CTL recognition and inhibits the
to use recombinant poxvirus as a potential vaccine (1998). intracellular transport of MHC class I molecules.
for another infectious disease. 11. Hirsch, V. M. et al. Patterns of viral replication J. Immunol. 178, 1654–1661 (2007).
3. Peter, G. et al. Lessons learned from a review of the correlate with outcome in simian immunodeficiency 20. Antoniou, A. N. & Powis, S. J. Pathogen evasion
development of selected vaccines. National Vaccine virus (SIV)‑infected macaques: effect of prior strategies for the major histocompatibility complex
Advisory Committee. Pediatrics 104, 942–950 immunization with a trivalent SIV vaccine in modified class I assembly pathway. Immunology 124, 1–12
(1999). vaccinia virus Ankara. J. Virol. 70, 3741–3752 (2008).
4. Mayr, A., Hochstein‑Mintzel, V. & Stickl, H. Passage (1996). 21. Meyer, H., Sutter, G. & Mayr, A. Mapping of deletions
history, properties, and applicability of the 12. Mann, B. A. et al. Vaccinia virus blocks Stat1‑ in the genome of the highly attenuated vaccinia virus
attenuated vaccinia virus strain, MVA. Infection 3, dependent and Stat1‑independent gene expression MVA and their influence on virulence. J. Gen. Virol. 72,
6–14 (1975). induced by type I and type II interferons. J. Interferon 1031–1038 (1991).
5. Sutter, G. & Moss, B. Nonreplicating vaccinia vector Cytokine Res. 28, 367–380 (2008). 22. Blanchard, T. J., Alcami, A., Andrea, P. & Smith, G. L.
efficiently expresses recombinant genes. Proc. Natl 13. Stack, J. et al. Vaccinia virus protein A46R targets Modified vaccinia virus Ankara undergoes limited
Acad. Sci. USA 89, 10847–10851 (1992). multiple Toll‑like‑interleukin‑1 receptor adaptors and replication in human cells and lacks several
6. Sutter, G., Wyatt, L. S., Foley, P. L., Bennink, J. R. & contributes to virulence. J. Exp. Med. 201, immunomodulatory proteins: implications for use as a
Moss, B. A recombinant vector derived from the host 1007–1018 (2005). human vaccine. J. Gen. Virol. 79, 1159–1167 (1998).
range‑restricted and highly attenuated MVA strain 14. Bowie, A. G. & Unterholzner, L. Viral evasion and 23. Lehmann, M. H. et al. Modified vaccinia virus ankara
of vaccinia virus stimulates protective immunity in subversion of pattern‑recognition receptor signalling. triggers chemotaxis of monocytes and early
mice to influenza virus. Vaccine 12, 1032–1040 Nature Rev. Immunol. 8, 911–922 (2008). respiratory immigration of leukocytes by induction of
(1994). This article highlights the interactions between CCL2 expression. J. Virol. 83, 2540–2552 (2009).
7. Tartaglia, J. et al. NYVAC: a highly attenuated strain of viruses and pattern-recognition receptors. 24. Waibler, Z. et al. Vaccinia virus‑mediated inhibition of
vaccinia virus. Virology 188, 217–232 (1992). 15. Alejo, A. et al. A chemokine‑binding domain in the type I interferon responses is a multifactorial process
8. Ramirez, J. C., Gherardi, M. M. & Esteban, M. Biology tumor necrosis factor receptor from variola (smallpox) involving the soluble type I interferon receptor B18
of attenuated modified vaccinia virus Ankara virus. Proc. Natl Acad. Sci. USA 103, 5995–6000 and intracellular components. J. Virol. 83,
recombinant vector in mice: virus fate and activation (2006). 1563–1571 (2009).
of B‑ and T‑cell immune responses in comparison with 16. Alcami, A. New insights into the subversion of the 25. Midgley, C. M., Putz, M. M., Weber, J. N. & Smith,
the Western Reserve strain and advantages as a chemokine system by poxviruses. Eur. J. Immunol. 37, G. L. Vaccinia virus strain NYVAC induces substantially
vaccine. J. Virol. 74, 923–933 (2000). 880–883 (2007). lower and qualitatively different human antibody
9. Hodge, J. W. et al. Modified vaccinia virus ankara 17. Alcami, A. & Smith, G. L. A soluble receptor for responses compared with strains Lister and Dryvax.
recombinants are as potent as vaccinia recombinants interleukin‑1β encoded by vaccinia virus: a novel J. Gen. Virol. 89, 2992–2997 (2008).

NATure reVIewS | MicRobiology VOluMe 8 | jANuArY 2010 | 71

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

26. Delaloye, J. et al. Innate immune sensing of modified 43. Yu, S. et al. Potent specific immune responses induced 62. Bradac, J. & Dieffenbach, C. W. HIV vaccine
vaccinia virus Ankara (MVA) is mediated by TLR2‑ by prime–boost–boost strategies based on DNA, development: Lessons from the past, informing the
TLR6, MDA‑5 and the NALP3 inflammasome. PLoS adenovirus, and Sendai virus vectors expressing gag future. IDrugs 12, 435–439 (2009).
Pathog. 5, e1000480 (2009). gene of Chinese HIV‑1 subtype, B. Vaccine 26, 63. Roberts, D. M. et al. Hexon‑chimaeric adenovirus
This study identifies innate immune signalling 6124–6131 (2008). serotype 5 vectors circumvent pre‑existing anti‑vector
pathways that are activated by MVA. 44. Lemckert, A. A. et al. Immunogenicity of heterologous immunity. Nature 441, 239–243 (2006).
27. Mooij, P. et al. Differential CD4+ versus CD8+ T‑cell prime–boost regimens involving recombinant This study demonstrates that modification of viral
responses elicited by different poxvirus‑based human adenovirus serotype 11 (Ad11) and Ad35 vaccine surface proteins can increase the efficacy of
immunodeficiency virus type 1 vaccine candidates vectors in the presence of anti‑Ad5 immunity. J. Virol. HAdV-5-based vectors.
provide comparable efficacies in primates. J. Virol. 82, 79, 9694–9701 (2005). 64. Tatsis, N. & Ertl, H. C. Adenoviruses as vaccine
2975–2988 (2008). 45. Folgori, A. et al. A T‑cell HCV vaccine eliciting effective vectors. Mol. Ther. 10, 616–629 (2004).
This work uses different viral vectors to show immunity against heterologous virus challenge in 65. Harro, C. et al. Safety and immunogenicity of the
that there is differential modulation of the chimpanzees. Nature Med. 12, 190–197 (2006). MRKAd5 and MRKAd6 HIV‑1 trigene vaccines alone
vaccine-induced CD4+ and CD8+ T cell responses. 46. Fattori, E. et al. Efficient immunization of rhesus and in combination in healthy adults. Clin. Vaccine
28. Cottingham, M. G. et al. Recombination‑mediated macaques with an HCV candidate vaccine by Immunol. 16, 1285–1292 (2009).
genetic engineering of a bacterial artificial heterologous priming–boosting with novel adenoviral 66. Tatsis, N. et al. Chimpanzee‑origin adenovirus vectors
chromosome clone of modified vaccinia virus Ankara vectors based on different serotypes. Gene Ther. 13, as vaccine carriers. Gene Ther. 13, 421–429 (2006).
(MVA). PLoS ONE 3, e1638 (2008). 1088–1096 (2006). 67. Sridhar, S. et al. Single‑dose protection against
29. Staib, C., Kisling, S., Erfle, V. & Sutter, G. Inactivation 47. Heeney, J. L. et al. A vaccine strategy utilizing a Plasmodium berghei by a simian adenovirus vector
of the viral interleukin 1β receptor improves CD8+ combination of three different chimeric vectors which using a human cytomegalovirus promoter containing
T‑cell memory responses elicited upon immunization share specific vaccine antigens. J. Med. Primatol. 29, intron, A. J. Virol. 82, 3822–3833 (2008).
with modified vaccinia virus Ankara. J. Gen. Virol. 86, 268–273 (2000). 68. Farina, S. F. et al. Replication‑defective vector based on
1997–2006 (2005). 48. Koopman, G. et al. Vaccine protection from CD4+ a chimpanzee adenovirus. J. Virol. 75, 11603–11613
30. Fischer, M. A., Tscharke, D. C., Donohue, K. B., T‑cell loss caused by simian immunodeficiency virus (2001).
Truckenmiller, M. E. & Norbury, C. C. Reduction of (SIV) mac251 is afforded by sequential immunization 69. Reyes‑Sandoval, A. et al. Single‑dose immunogenicity
vector gene expression increases foreign antigen‑ with three unrelated vaccine vectors encoding multiple and protective efficacy of simian adenoviral vectors
specific CD8+ T‑cell priming. J. Gen. Virol. 88, SIV antigens. J. Gen. Virol. 85, 2915–2924 (2004). against Plasmodium berghei. Eur. J. Immunol. 38,
2378–2386 (2007). A report on the efficacy of a combination of three 732–741 (2008).
31. McConkey, S. J. et al. Enhanced T‑cell immunogenicity different vectors to elicit vaccine protection against 70. Dudareva, M. et al. Prevalence of serum neutralizing
of plasmid DNA vaccines boosted by recombinant SIV in macaques. antibodies against chimpanzee adenovirus 63 and
modified vaccinia virus Ankara in humans. Nature 49. Darrah, P. A. et al. Multifunctional TH1 cells define a human adenovirus 5 in Kenyan children, in the context
Med. 9, 729–735 (2003). correlate of vaccine‑mediated protection against of vaccine vector efficacy. Vaccine 27, 3501–3504
32. Mwau, M. et al. A human immunodeficiency virus 1 Leishmania major. Nature Med. 13, 843–850 (2007). (2009).
(HIV‑1) clade A vaccine in clinical trials: stimulation of This work highlights the importance of measuring 71. Shen, X., Wong, S. B., Buck, C. B., Zhang, J. &
HIV‑specific T‑cell responses by DNA and recombinant antigen-specific T cell quality and not just quantity Siliciano, R. F. Direct priming and cross‑priming
modified vaccinia virus Ankara (MVA) vaccines in when analysing immune responses for correlates of contribute differentially to the induction of CD8+ CTL
humans. J. Gen. Virol. 85, 911–919 (2004). protection. following exposure to vaccinia virus via different
33. Keating, S. M. et al. Durable human memory T cells 50. Forbes, E. K. et al. Multifunctional, high‑level cytokine‑ routes. J. Immunol. 169, 4222–4229 (2002).
quantifiable by cultured enzyme‑linked immunospot producing Th1 cells in the lung, but not spleen, 72. Bronte, V. et al. Antigen expression by dendritic cells
assays are induced by heterologous prime boost correlate with protection against Mycobacterium correlates with the therapeutic effectiveness of a
immunization and correlate with protection against tuberculosis aerosol challenge in mice. J. Immunol. model recombinant poxvirus tumor vaccine. Proc. Natl
malaria. J. Immunol. 175, 5675–5680 (2005). 181, 4955–4964 (2008). Acad. Sci. USA 94, 3183–3188 (1997).
34. Lamoreaux, L., Roederer, M. & Koup, R. Intracellular 51. Beveridge, N. E. et al. Immunisation with BCG and 73. Tewalt, E. F. et al. Viral sequestration of antigen
cytokine optimization and standard operating recombinant MVA85A induces long‑lasting, subverts cross presentation to CD8+ T cells. PLoS
procedure. Nature Protoc. 1, 1507–1516 polyfunctional Mycobacterium tuberculosis‑specific Pathog. 5, e1000457 (2009).
(2006). CD4+ memory T lymphocyte populations. Eur. J. 74. Moorthy, V. S. et al. Phase 1 evaluation of 3 highly
35. Vuola, J. M. et al. Differential immunogenicity of Immunol. 37, 3089–3100 (2007). immunogenic prime–boost regimens, including a
various heterologous prime–boost vaccine regimens 52. McElrath, M. J. et al. HIV‑1 vaccine‑induced immunity 12‑month reboosting vaccination, for malaria
using DNA and viral vectors in healthy volunteers. in the test‑of‑concept Step Study: a case‑cohort vaccination in Gambian men. J. Infect. Dis. 189,
J. Immunol. 174, 449–455 (2005). analysis. Lancet 372, 1894–1905 (2008). 2213–2219 (2004).
36. Mooij, P. et al. Comparison of human and rhesus 53. Liu, J. et al. Magnitude and phenotype of cellular 75. Yang, L., Bailey, L., Baltimore, D. & Wang, P. Targeting
macaque T‑cell responses elicited by boosting with immune responses elicited by recombinant adenovirus lentiviral vectors to specific cell types in vivo. Proc.
NYVAC encoding HIV‑1 clade C immunogens. J. Virol. vectors and heterologous prime–boost regimens in Natl Acad. Sci. USA 103, 11479–11484 (2006).
83, 5881–5889 (2009). rhesus monkeys. J. Virol. 82, 4844–4852 (2008). 76. Luo, X. M. et al. Engineering human hematopoietic
37. Harari, A. et al. An HIV‑1 clade C DNA prime, NYVAC 54. Geiben‑Lynn, R., Greenland, J. R., Frimpong‑Boateng, stem/progenitor cells to produce a broadly neutralizing
boost vaccine regimen induces reliable, polyfunctional, K. & Letvin, N. L. Kinetics of recombinant adenovirus anti‑HIV antibody after in vitro maturation to human B
and long‑lasting T cell responses. J. Exp. Med. 205, type 5, vaccinia virus, modified vaccinia ankara virus, lymphocytes. Blood 113, 1422–1431 (2008).
63–77 (2008). and DNA antigen expression in vivo and the induction This article describes the basis of engineering
38. Webster, D. P. et al. Enhanced T cell‑mediated of memory T‑lymphocyte responses. Clin. Vaccine immunity with lentiviruses. These mediate gene
protection against malaria in human challenges by Immunol. 15, 691–696 (2008). transfer into human hematopoietic stem cells,
using the recombinant poxviruses FP9 and modified 55. Jego, G. et al. Plasmacytoid dendritic cells induce which can then be differentiated in vitro into B cells
vaccinia virus Ankara. Proc. Natl Acad. Sci. USA 102, plasma cell differentiation through type I interferon that secrete neutralizing HIV-1 antibodies.
4836–4841 (2005). and interleukin 6. Immunity 19, 225–234 (2003). 77. Garcia Casado, J. et al. Lentivector immunization
This paper illustrates the effect of heterologous 56. Xiang, Z. et al. Novel, chimpanzee serotype 68‑based induces tumor antigen‑specific B and T cell responses
prime–boost protocols in humans using viral-vector adenoviral vaccine carrier for induction of antibodies to in vivo. Eur. J. Immunol. 38, 1867–1876 (2008).
vaccines that lead to enhanced immunogenicity and a transgene product. J. Virol. 76, 2667–2675 (2002). 78. Yang, L. et al. Engineered lentivector targeting of
protection against malaria infection. 57. Hensley, S. E., Giles‑Davis, W., McCoy, K. C., Weninger, dendritic cells for in vivo immunization. Nature
39. Draper, S. J. et al. Effective induction of high‑titer W. & Ertl, H. C. Dendritic cell maturation, but not Biotech. 26, 326–334 (2008).
antibodies by viral vector vaccines. Nature Med. 14, CD8+ T cell induction, is dependent on type I IFN 79. Meeusen, E. N., Walker, J., Peters, A., Pastoret, P. P. &
819–821 (2008). signaling during vaccination with adenovirus vectors. Jungersen, G. Current status of veterinary vaccines.
This article highlights the ability of certain J. Immunol. 175, 6032–6041 (2005). Clin. Microbiol. Rev. 20, 489–510 (2007).
viral-vector vaccine combinations to elicite 58. Hensley, S. E. et al. Type I interferon inhibits antibody A summary of the current status of the viral
antibody as well as T cell responses. responses induced by a chimpanzee adenovirus vaccines that are in veterinary use.
40. Liu, J. et al. Immune control of an SIV challenge by a vector. Mol. Ther. 15, 393–403 (2007). 80. Poulet, H. et al. Development and registration of
T‑cell‑based vaccine in rhesus monkeys. Nature 457, 59. Hornemann, S. et al. Replication of modified vaccinia recombinant veterinary vaccines. The example of the
87–91 (2009). virus Ankara in primary chicken embryo fibroblasts canarypox vector platform. Vaccine 25, 5606–5612
This study describes the increased efficacy of requires expression of the interferon resistance gene (2007).
heterologous adenoviral prime–boost regimes over E3L. J. Virol. 77, 8394–8407 (2003). 81. Swayne, D. E. & Kapczynski, D. Strategies and
homologous regimes for eliciting protection against 60. Ludwig, H. et al. Role of viral factor E3L in modified challenges for eliciting immunity against avian influenza
a stringent SIV challenge in macaques, which vaccinia virus Ankara infection of human HeLa cells: virus in birds. Immunol. Rev. 225, 314–331 (2008).
suggests that such an approach should be adopted regulation of the virus life cycle and identification of 82. Steel, J. et al. A combination in‑ovo vaccine for avian
for HIV-1 vaccine development. differentially expressed host genes. J. Virol. 79, influenza virus and Newcastle disease virus. Vaccine
41. Shiver, J. W. et al. Replication‑incompetent adenoviral 2584–2596 (2005). 26, 522–531 (2008).
vaccine vector elicits effective anti‑immunodeficiency‑ 61. Buchbinder, S. P. et al. Efficacy assessment of a cell‑ 83. Kitching, P. et al. Global FMD control — is it an
virus immunity. Nature 415, 331–335 (2002). mediated immunity HIV‑1 vaccine (the Step Study): a option? Vaccine 25, 5660–5664 (2007).
42. Gherardi, M. M. et al. Prime–boost immunization double‑blind, randomised, placebo‑controlled, test‑of‑ 84. Pacheco, J. M., Brum, M. C., Moraes, M. P., Golde,
schedules based on influenza virus and vaccinia virus concept trial. Lancet 372, 1881–1893 (2008). W. T. & Grubman, M. J. Rapid protection of cattle
vectors potentiate cellular immune responses against Along with reference 52, this paper reports on the from direct challenge with foot‑and‑mouth disease
human immunodeficiency virus Env protein first Phase III clinical trial to use a T cell-based virus (FMDV) by a single inoculation with an
systemically and in the genitorectal draining lymph adenoviral vaccine vector in humans, and warns of adenovirus‑vectored FMDV subunit vaccine. Virology
nodes. J. Virol. 77, 7048–7057 (2003). the concerns of anti-HAdV-5 vector immunity. 337, 205–209 (2005).

72 | jANuArY 2010 | VOluMe 8 www.nature.com/reviews/micro

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

85. Pena, L. et al. Delivery of a foot‑and‑mouth disease 106. McMichael, A. J., Gotch, F. M., Noble, G. R. & Beare, 125. Stanton, R. J., McSharry, B. P., Armstrong, M.,
virus empty capsid subunit antigen with nonstructural P. A. Cytotoxic T‑cell immunity to influenza. N. Engl. J. Tomasec, P. & Wilkinson, G. W. Re‑engineering
protein 2B improves protection of swine. Vaccine 26, Med. 309, 13–17 (1983). adenovirus vector systems to enable high‑throughput
5689–5699 (2008). 107. Heeney, J. L. & Plotkin, S. A. Immunological correlates analyses of gene function. Biotechniques 45,
86. Minassian, A. M. & McShane, H. Tuberculosis of protection from HIV infection and disease. Nature 659–662, 664–668 (2008).
vaccines: present and future. Expert Rev. Respir. Med. Immunol. 7, 1281–1284 (2006). This article shows how toxic transgenes can be
2, 721–738 (2008). 108. Rerks‑Ngarm, S. et al. Vaccination with ALVAC and switched off during the production of
87. Vordermeier, H. M., Huygen, K., Singh, M., Hewinson, AIDSVAX to prevent HIV‑1 infection in Thailand. adenoviral-vector vaccines in cell culture, thereby
R. G. & Xing, Z. Immune responses induced in cattle N. Engl. J. Med. 19 November 2009 (doi: 10.1056/ enabling the growth and generation of recombinant
by vaccination with a recombinant adenovirus NEJMoa0908492). AdV vaccines encoding such toxic proteins.
expressing mycobacterial antigen 85A and The first HIV-1 vaccine Phase III clinical trial 126. Paton, D. J. et al. Application of non‑structural protein
Mycobacterium bovis BCG. Infect. Immun. 74, reporting a modest trend of protection from antibody tests in substantiating freedom from
1416–1418 (2006). infection. foot‑and‑mouth disease virus infection after
88. Vordermeier, H. M. et al. Cellular immune responses 109. Thongcharoen, P. et al. A phase 1/2 comparative emergency vaccination of cattle. Vaccine 24,
induced in cattle by heterologous prime–boost vaccine trial of the safety and immunogenicity of a 6503–6512 (2006).
vaccination using recombinant viruses and bacille CRF01_AE (subtype E) candidate vaccine: ALVAC‑HIV 127. Ko, S. Y. et al. Enhanced induction of intestinal cellular
Calmette‑Guerin. Immunology 112, 461–470 (2004). (vCP1521) prime with oligomeric gp160 (92TH023/ immunity by oral priming with enteric adenovirus 41
89. Buddle, B. M. et al. Cattle as a model for development LAI‑DID) or bivalent gp120 (CM235/SF2) boost. vectors. J. Virol. 83, 748–756 (2009).
of vaccines against human tuberculosis. Tuberculosis J. Acquir. Immune Defic. Syndr. 46, 48–55 (2007). 128. Gallego‑Gomez, J. C. et al. Differences in virus‑induced
(Edinb.) 85, 19–24 (2005). 110. Sandstrom, E. et al. Broad immunogenicity of a cell morphology and in virus maturation between MVA
90. Limbach, K. J. & Richie, T. L. Viral vectors in malaria multigene, multiclade HIV‑1 DNA vaccine boosted with and other strains (WR, Ankara, and NYCBH) of
vaccine development. Parasite Immunol. 31, heterologous HIV‑1 recombinant modified vaccinia vaccinia virus in infected human cells. J. Virol. 77,
501–519 (2009). virus Ankara. J. Infect. Dis. 198, 1482–1490 10606–10622 (2003).
91. Draper, S. J. et al. Recombinant viral vaccines (2008). 129. Smith, G. L., Vanderplasschen, A. & Law, M. The
expressing merozoite surface protein‑1 induce 111. Priddy, F. H. et al. Safety and immunogenicity of a formation and function of extracellular enveloped
antibody‑ and T cell‑mediated multistage protection replication‑incompetent adenovirus type 5 HIV‑1 vaccinia virus. J. Gen. Virol. 83, 2915–2931
against malaria. Cell Host Microbe 5, 95–105 (2009). clade B gag/pol/nef vaccine in healthy adults. Clin. (2002).
92. Hill, A. V. Pre‑erythrocytic malaria vaccines: towards Infect. Dis. 46, 1769–1781 (2008). 130. Antoine, G., Scheiflinger, F., Dorner, F. & Falkner, F. G.
greater efficacy. Nature Rev. Immunol. 6, 21–32 112. Hansen, S. G. et al. Effector memory T cell responses The complete genomic sequence of the modified
(2006). are associated with protection of rhesus monkeys from vaccinia Ankara strain: comparison with other
93. Nwanegbo, E. et al. Prevalence of neutralizing mucosal simian immunodeficiency virus challenge. orthopoxviruses. Virology 244, 365–396
antibodies to adenoviral serotypes 5 and 35 in the Nature Med. 15, 293–299 (2009). (1998).
adult populations of The Gambia, South Africa, and This paper describes the use of CMV as an HIV-1 131. Gaggar, A., Shayakhmetov, D. M. & Lieber, A. CD46 is
the United States. Clin. Diagn. Lab. Immunol. 11, vaccine vector in macaques and shows that this a cellular receptor for group B adenoviruses. Nature
351–357 (2004). vector can induce T cells with an effector memory Med. 9, 1408–1412 (2003).
94. Bejon, P. et al. A Phase 2b randomised trial of the phenotype that are associated with protection 132. Cheng, C. et al. Mechanism of Ad5 vaccine immunity
candidate malaria vaccines FP9 ME‑TRAP and MVA from a mucosal challenge with SIV. and toxicity: fiber shaft targeting of dendritic cells.
ME‑TRAP among children in Kenya. PLoS Clin. Trials 113. Wilson, N. A. et al. Vaccine‑induced cellular immune PLoS Pathog. 3, e25 (2007).
1, e29 (2006). responses reduce plasma viral concentrations after 133. Ghosh‑Choudhury, G., Haj‑Ahmad, Y., Brinkley, P.,
95. McShane, H. et al. Recombinant modified vaccinia repeated low‑dose challenge with pathogenic simian Rudy, J. & Graham, F. L. Human adenovirus cloning
virus Ankara expressing antigen 85A boosts BCG‑ immunodeficiency virus SIVmac239. J. Virol. 80, vectors based on infectious bacterial plasmids. Gene
primed and naturally acquired antimycobacterial 5875–5885 (2006). 50, 161–171 (1986).
immunity in humans. Nature Med. 10, 1240–1244 114. Casimiro, D. R. et al. Comparative immunogenicity in 134. Bett, A. J., Haddara, W., Prevec, L. & Graham, F. L.
(2004). rhesus monkeys of DNA plasmid, recombinant An efficient and flexible system for construction of
The first clinical study to demonstrate that immune vaccinia virus, and replication‑defective adenovirus adenovirus vectors with insertions or deletions in early
responses primed by BCG can be boosted using a vectors expressing a human immunodeficiency virus regions 1 and 3. Proc. Natl Acad. Sci. USA 91,
viral-vector system encoding an antigen from type 1 gag gene. J. Virol. 77, 6305–6313 8802–8806 (1994).
mycobacteria. (2003). 135. Smith, G. L. & Moss, B. Infectious poxvirus vectors
96. Hawkridge, T. et al. Safety and immunogenicity of a 115. Ward, S., Copier, J. & Dalgleish, A. Technical have capacity for at least 25 000 base pairs of foreign
new tuberculosis vaccine, MVA85A, in healthy adults challenges facing therapeutic cancer vaccines. Curr. DNA. Gene 25, 21–28 (1983).
in South Africa. J. Infect. Dis. 198, 544–552 Opin. Drug Discov. Devel. 11, 168–177 (2008). 136. Liu, X., Kremer, M. & Broyles, S. S. A natural vaccinia
(2008). 116. Emens, L. A. Cancer vaccines: on the threshold of virus promoter with exceptional capacity to direct
97. Sander, C. R. et al. Safety and immunogenicity of a success. Expert Opin. Emerg. Drugs 13, 295–308 protein synthesis. J. Virol. Methods 122, 141–145
new TB vaccine, MVA85A, in M. tuberculosis infected (2008). (2004).
individuals. Am. J. Respir. Crit. Care Med. 179, 117. Hodge, J. W., Guha, C., Neefjes, J. & Gulley, J. L. 137. Crucell & AERAS. Aeras and Crucell announce TB
724–733 (2009). Synergizing radiation therapy and immunotherapy for vaccine clinical trial in Kenya; promising vaccine
98. Radosevic, K. et al. Protective immune responses to a curing incurable cancers. Opportunities and candidate advances to Phase II safety study in South
recombinant adenovirus type 35 tuberculosis vaccine challenges. Oncology 22, 1064–1070; discussion Africa. Crucell [online], http://hugin.
in two mouse strains: CD4 and CD8 T‑cell epitope 1075, 1080–1081, 1084 (2008). info/132631/R/1260403/275810.pdf (2008).
mapping and role of gamma interferon. Infect. Immun. 118. Kaufman, H. L. et al. Phase II trial of modified vaccinia
75, 4105–4115 (2007). Ankara (MVA) virus expressing 5T4 and high dose
99. Magalhaes, I. et al. rBCG induces strong antigen‑ interleukin‑2 (IL‑2) in patients with metastatic renal Acknowledgements
specific T cell responses in rhesus macaques in a prime‑ cell carcinoma. J. Transl. Med. 7, 2 (2009). We thank A. Hill, M. Friede and G. Nabel for organizing the
boost setting with an adenovirus 35 tuberculosis 119. Ramlau, R. et al. A phase II study of Tg4010 recent meeting ‘Viral Vector Vaccines 2008’ in Hinxton, which
vaccine vector. PLoS ONE 3, e3790 (2008). (Mva‑Muc1‑Il2) in association with chemotherapy in stimulated the writing of this Review. We thank A. Hill for
100. Hoft, D. F. et al. A new recombinant bacille Calmette‑ patients with stage III/IV non‑small cell lung cancer. comments on a draft of the manuscript and G.J. Nabel for
Guerin vaccine safely induces significantly enhanced J. Thorac. Oncol. 3, 735–744 (2008). providing the drawing on which figure 1 is based. J.L.H. is
tuberculosis‑specific immunity in human volunteers. 120. Dreicer, R. et al. MVA‑MUC1‑IL2 vaccine currently sponsored by grants from the Bill and Melinda
J. Infect. Dis. 198, 1491–1501 (2008). immunotherapy (TG4010) improves PSA doubling Gates Foundation, USA, and the National Institutes of
101. Roukens, A. H. & Visser, L. G. Yellow fever vaccine: time in patients with prostate cancer with biochemical Health, USA.
past, present and future. Expert Opin. Biol. Ther. 8, failure. Invest. New Drugs 27, 379–386 (2009).
1787–1795 (2008). 121. Quoix, E. et al. Randomized, controlled phase IIb trial Competing interests statement
102. Pugachev, K. V., Guirakhoo, F., Trent, D. W. & Monath, evaluating the therapeutic vaccine TG4010 The authors declare no competing financial interests.
T. P. Traditional and novel approaches to flavivirus (MVA‑MUC1‑IL‑2) as an adjunct to chemotherapy in
vaccines. Int. J. Parasitol. 33, 567–582 (2003). patients with advanced non‑small cell lung cancer
103. McAllister, A., Arbetman, A. E., Mandl, S., (NSCLC). Ann. Oncol. 19, viii92 (2008).
Pena‑Rossi, C. & Andino, R. Recombinant yellow fever 122. Salucci, V. et al. CD8+ T‑cell tolerance can be broken by DATABASES
viruses are effective therapeutic vaccines for an adenoviral vaccine while CD4+ T‑cell tolerance is Entrez Gene: http://www.ncbi.nlm.nih.gov/gene
treatment of murine experimental solid tumors and broken by additional co‑administration of a Toll‑like B15R | E3L
pulmonary metastases. J. Virol. 74, 9197–9205 receptor ligand. Scand. J. Immunol. 63, 35–41 (2006). UniProtKB: http://www.uniprot.org
(2000). 123. Aurisicchio, L. et al. Immunogenicity and safety of a AMA1 | CEA | CSP | MSP1 | mycobacterial mycolyl
104. Bonaldo, M. C. et al. Construction and characterization DNA prime/adenovirus boost vaccine against rhesus transferase 85A
of recombinant flaviviruses bearing insertions CEA in nonhuman primates. Int. J. Cancer 120,
between E and NS1 genes. Virol. J. 4, 115 (2007). 2290–2300 (2007). FURTHER INFORMATION
105. Monath, T. P. et al. Clinical proof of principle for 124. Domi, A. & Moss, B. Engineering of a vaccinia virus Simon J. Draper’s homepage: http://www.jenner.ac.uk
ChimeriVax: recombinant live, attenuated vaccines bacterial artificial chromosome in Escherichia coli by IAVI: http://www.iavi.org
against flavivirus infections. Vaccine 20, 1004–1018 bacteriophage λ‑based recombination. Nature All linkS ARe AcTiVe in THe online PDF
(2002). Methods 2, 95–97 (2005).

NATure reVIewS | MicRobiology VOluMe 8 | jANuArY 2010 | 73

© 2010 Macmillan Publishers Limited. All rights reserved

You might also like