* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download New Strategies for Vaccine Development - Initial Set Up
Adaptive immune system wikipedia , lookup
Molecular mimicry wikipedia , lookup
Vaccination policy wikipedia , lookup
Polyclonal B cell response wikipedia , lookup
Hygiene hypothesis wikipedia , lookup
Innate immune system wikipedia , lookup
Whooping cough wikipedia , lookup
Herd immunity wikipedia , lookup
Psychoneuroimmunology wikipedia , lookup
Childhood immunizations in the United States wikipedia , lookup
Hepatitis B wikipedia , lookup
Immunocontraception wikipedia , lookup
New Strategies for Vaccine Development Liubov M. Lobanova [email protected] Program in Vaccinology and Immunotherapeutics School of Public Health and Vaccine and Infectious Disease Organization University of Saskatchewan Saskatoon Canada For: Advanced Virology (VTMC 833) © Liubov M. Lobanova Abstract Introduction Reverse vaccinology approach Virus-like particles DNA vaccines Improving immunogenicity through formulation Improving immunogenicity by including immune modulatory adjuvants Improving immunogenicity by using next-generation delivery strategies Viral vectors Prime-boost strategy Conclusions References Abstract Vaccination remains the most successful strategy for preventing and controlling infectious diseases. In many cases, conventional vaccines, in particular live attenuated vaccines, inactivated microorganisms and subunit vaccines have been successful at inducing protective immunity, mainly humoral immunity, to disease-causing pathogens. However, it has become apparent that certain highly dangerous pathogens, that resist the humoral immunity, are not readily controlled 1 by conventional vaccination approaches. Recent knowledge of host-pathogen interactions, received from fundamental research in immunology, molecular-cellular biology and vaccinology provides understanding that the induction of T-cell immunity is required for optimal treatment against pathogens causing chronic infections. Although conventional live attenuated vaccines may induce cell-mediated immunity, there are serious safety issues regarding the application of this strategy for the development of vaccines against highly dangerous pathogens, such as human immunodeficiency virus (HIV). As a result of these problems, several new approaches to vaccine development have emerged. These approaches are distinguished by having advantages over conventional vaccine strategies. This review will focus on some of these new strategies developed to promote potent cellular immunity, including (i) virus-like particles, (ii) plasmid DNA vaccines, (iii) viral vectors, and (iv) prime-boost vaccination strategy. In addition, the reverse vaccinology approach, which facilitates systematic identification of new potential antigens of a pathogen, is discussed. Introduction The concept of vaccination was demonstrated over 200 years ago when Edward Jenner showed that prior exposure to cowpox could prevent infection by smallpox. Over the last century, the development and widespread use of vaccines against a variety of infectious agents have been a great triumph of medical science. We have vaccines for nearly thirty out of the more than seventy infectious diseases of humans. Among the greatest achievement of vaccination remains the eradication of smallpox. Other achievements are: virtual disappearance of previously disabling and lethal diseases such as diphtheria, tetanus, paralytic poliomyelitis, pertussis, measles, mumps, rubella and invasive Haemophilus influenzae type B. Hepatitis A and B are also being brought under control in an increasing number of countries. Conventional approaches, in particular attenuation of the pathogen, inactivation of the microorganism and creation of subunit preparations, have been very successful in the development of vaccines against infectious agents. However, there are some diseases for which attempts to develop vaccines using conventional approaches have failed. These include chronic infectious diseases such as acquired immunodeficiency syndrome (AIDS) and hepatitis C (HCV). Moreover, occasionally, a vaccine has to be withdrawn because of unexpected side 2 effects. For instance, an inactivated vaccine against measles was withdrawn because it had provoked atypical measles after natural infection (99). More recently, in October 1999, a simianhuman reassortant rotavirus vaccine was withdrawn due to intussusceptions and mortality of vaccinated infants (117). Therefore, there is a constant interest in developing new approaches to improve efficacy and safety of existing vaccines. There is also a continuous need for developing new vaccines to newly emerging pathogens, such as the severe acute respiratory syndrome virus (14) and hantavirus (26), as well as to pathogens for which no vaccines currently exist. Finally, development of therapeutic vaccines and vaccines against allergic and autoimmune diseases represents another new vaccine challenge. Achieving such ambitious goals certainly requires new approaches in vaccine development. This review explores some new approaches for vaccine development: reverse vaccinology, virus-like particles (VLPs), ‘naked’ DNA plasmids, viral vectors, and heterologous prime-boost strategy. Reverse vaccinology approach has emerged due to the need to identify potential vaccine candidates providing new solutions for those vaccines which have been difficult or impossible to develop, whereas the other approaches mentioned above have emerged because it appears that protection against infection by many agents, particularly viruses, requires generation of potent cell-mediated immunity (106). Successful vaccines are available for many viruses, but almost exclusively ones that produce acute, self-limited infection (74). The most important characteristic of these pathogens is that neutralizing antibodies protect against infection or disease. Conventional approaches (e.g., live attenuated vaccines, inactivated vaccines and subunit vaccines) have been employed successfully against these pathogens which elicit protective immune responses with a bias towards the induction of humoral immunity. In contrast, there are no successful vaccines available for viruses that cause chronic infections, such 3 as HIV, HCV and herpes simplex virus. It has been shown that CD8+ T cells are especially important for the control of HIV-1 and HCV infections (77, 106). In addition to CD8+ cell responses, CD4+ T cells have been found to be critical in the maintenance of adequate CD8+ T cell function and control of viremia in both HIV and HCV infection (64, 106). Live attenuated vaccines have been the most successful in induction of efficient B- and T-cell responses (59). However, there is much concern about the use of live attenuated vaccines against viruses that cause chronic infection. Besides, such vaccines are unlikely to induce protection as the immune response to the natural infection is not sufficient to eradicate the infection. Therefore, the development of vaccines against such pathogens is more challenging, since they would have to elicit more robust cytotoxic T lymphocyte (CTL) responses in addition to antibodies (71). This review will cover the approaches are being studied today which are based on eliciting strong cell-mediated immunity. Reverse vaccinology approach: The first revolution in vaccinology was the use of modern recombinant DNA technology to produce subunit vaccines. This approach involves propagation of a pathogen in the laboratory conditions, followed by identification of its components that are important in pathogenesis and immunity. Then, identified protective antigens are produced in a system for large-scale recombinant protein expression (Figure 1). While successful in some cases, for example the subunit vaccines against hepatitis B (6) and Bordetella pertussis (65), this strategy is limited to pathogens with known immunodominant protective antigens. The approach is time-consuming and is amenable to only those antigens which can be purified in high quantities. However, in some cases the most abundant proteins expressed by a pathogen are not suitable vaccine 4 candidates and genetic tools required for identification of less abundant components may be inadequate or not available. Moreover, this strategy is not applicable to non-cultivable microorganisms. Figure 1. Schematic representation of the conventional approach to vaccine development. This approach requires the pathogen to be grown in laboratory conditions, individual components to be identified and produced in a pure form, either directly from the microorganism or through recombinant DNA technology and then tested for their ability to induce immunity. The second revolution in vaccine development took place at the end of 20th century as a result of the use of the genomic technology. Currently, it became possible to determine the complete genome sequence of any microorganism in a few months at a low cost. As a result, the sequences of most of the pathogens are now available. This advantage is used by reverse vaccinology approach (142) which is shown schematically in Figure 2. The strategy utilizes computer analysis of available genome sequences to predict open reading frames (ORFs) as well as localization and possible function of proteins encoded by these ORFs. Two-dimensional gel electrophoresis together with mass spectrometry, DNA microarrays, in vivo expression technology provide more information on these potential antigens. For example, microarray 5 analysis is used to identify genes that are turned on during invasive infection (39), and thus, the proteins encoded by these genes can be considered as potential targets for vaccine development (156). It allows choosing, in a short period of time, many vaccine candidates including those proteins which are expressed at very low levels during infection. Next step is a high throughput expression of recombinant proteins and testing them in animal models. This approach reduces the time of vaccine development to 1-2 years, whereas the development of vaccines by conventional methods could take 5 or more years. An example of one of the first applications of reverse vaccinology approach is the identification of vaccine candidates against Neisseria meningitidis serogroup B (132). Potential antigens identified by computer programs were expressed in Escherichia coli as recombinant fusion proteins, purified and used to immunize animals. The resultant sera were examined for meningococcocidal activity in the presence of complement (132). Among the proteins with bactericidal activity, a novel surface-exposed oligomeric protein Neisseria adhesin A (NadA) was found to be present in most of the meningococcus B strains. As NadA elicited production of bactericidal antibodies that correlated with protection against the pathogen in the infant rat model it was considered as a promising vaccine candidate (32). Moreover, immunization with a multicomponent vaccine containing NadA and other protective antigens discovered by this approach, gave promising results in clinical phase I and II trials (61). While the vaccine is in clinical development, research efforts are focused on the structural characterization of the main vaccine antigens. For instance, a truncated soluble variant of NadA was subjected to extensive physico-chemical characterization in order to be formulated as part of the final drug product (97). 6 Figure 2. The reverse vaccinology approach to vaccine development. This approach utilizes available information about genomic sequences of pathogenic microorganisms. With the use of computer software, ORFs and possible localization and functions of proteins encoded by these ORFs are predicted. This is followed by a high-throughput cloning and expression of identified ORFs. Expressed proteins undergo screening in animal models to be tested for the immunogenicity and ability to protect against challenge. The success of the group B meningococcus paradigm facilitated the application of the reverse vaccinology strategy for the identification of vaccine candidates against other bacterial pathogens, such as Streptococcus pneumoniae (184), Porphyromonas gingivlis (150), Chlamydia preumoniae (112), Staphylococcous aureus (46) as well as to protozoan pathogens (63). However, in order to be successful against a more complex microorganism such as Streptococcus agalactiae this approach had to evolve giving the beginning of pan-genome reverse vaccinology and comparative reverse vaccinology (157). Pan-genome reverse analysis is focused on comparing many pathogenic strains of a species in order to identify antigens that could provide maximum coverage against different serotypes resulting in the development of a universal vaccine. The identification of vaccine candidates against Streptococcus agalactiae, a group B 7 streptococcus, is an example of successful application of multiple genomic approaches in vaccine design (98). Comparative reverse vaccinology approach involves comparative analysis of genomes of pathogenic and non-pathogenic strains of the same species with the goal of finding pathogenicity markers (69). This strategy reduces the number of vaccine candidates to express and test in the animal model and, consequently, reduces the time for the delivery of a vaccine as genes conserved in both pathogenic and non-pathogenic strains are excluded from the selection. Thus, the reverse vaccinology approach is aimed at the identification of potential vaccine candidates without the need for cultivating the pathogen, reducing the time and cost required for the identification of new vaccine candidates. Although this approach has been predominantly used for the development of vaccines against complex microorganisms, such as bacteria, vaccines against viral pathogens (hepatitis C and HIV) are currently under development with use of this approach (142). However, some limiting steps in reverse vaccinology include lack of algorithms that can be used to make a good correlation between antigens and their likely protective immune responses, and inability to identify non-protein antigens including polysaccharides or CD1-restricted antigens such as glycolipids (142). Virus-like particles: Virus-like particles represent another promising and viable strategy to the production of vaccines against many diseases. VLPs are a highly effective type of subunit vaccines that possess characteristics that make them a safe and effective vaccine platform. “Virus-like particles are highly organized spheres that self-assemble from virus-derived structural antigens” (93). VLPs 8 preparations are all based on the observation that expression of the capsid proteins of many viruses leads to the spontaneous assembly of particles that are structurally similar to authentic virus (4, 76, 123). Immunological features of viruses, such as repetitive surfaces, particulate structures and induction of innate immunity through activation of pathogen-associated molecular-pattern recognition receptors, make VLPs very effective in inducing potent B- and Tcell responses. The critical role of innate immune response induced by virus in directing the subsequent adaptive immune response of appropriate magnitude, quality and specificity has been highlighted by Gaucher and coauthors (59). Besides, the authors demonstrated that the sum of all immune arms is required for the long-lasting protection induced by yellow fever vaccine and integrated immune response constitutes the correlates of protection. Moreover, unique features of protective immune responses were identified, which can now be used as a basis for the development of novel vaccines. In practical terms, the fact that VLPs mimic the structure of virus particles usually means that VLPs elicit strong humoral response as it allows the adaptive immune system rapidly detect and respond to these repeated and ordered structures found on viral surfaces (11, 135). Moreover, the dense, repetitive nature of VLPs makes them particularly effective in inducing antibody responses, often without adjuvants (70). In addition, due to their structure and size VLPs are capable of diffusing to lymph nodes from the site of injection where they can interact directly with B-cells to trigger antibody responses (192). In addition to B-cell responses, VLPs also induce strong CD4 proliferative and CTL responses (116), which are the aim of current T-cell vaccine strategies. Features of VLPs, such as preferential targeting of dendritic cells (DCs), efficient delivery to both MHC class I and II molecules and the ability to directly activate the DC maturation, make them strong inducers of 9 CTL responses. Due to their particulate structure exogenous VLPs are readily taken up by antigen presenting cells (APCs), in particular by DCs, and presented effectively in the context of MHC class II molecules as well as they can be cross-presented on MHC class I molecules, thereby inducing a CTL response, which is essential for the clearance of intracellular pathogens such as viruses (115). For example, it has been reported that human papillomavirus (HPV) VLPs directly activate DCs leading to the expression of co-stimulatory molecules, cytokines necessary for activation of CTLs (85). Furthermore, some VLPs that retain receptor binding regions of cognate viruses can enter cells through their normal receptor and can be taken by DCs as exogenous antigens for class I presentation (13). Importantly, DC maturation and migration, essential for activating the innate and adaptive immune responses, are promoted by uptake of VLPs (54). It was shown that the stimulation of DCs does not require the virus replication but it requires an intact envelope of either an inactivated virus (50) or that of a VLP (13), or an intact non-enveloped VLP (187). This feature gives advantages to VLPs over the cognate live viruses for immune activation, because several viruses have an ability to interfere with the DC activation (125). Moreover, the self-adjuvanting effect of VLPs to target DCs is an important advantage over soluble antigens, which are less immunogenic and require the co-administration with adjuvants in several booster injections. However, not all VLPs can skew the immune response towards Th1, and the addition of innate immunity stimulators is required. This drawback can be easily overcome as VLPs have the ability to encompass immune stimulating adjuvants, such as Toll-like receptor (TLR) ligands, within their lumens. For instance, vaccination with CpG-loaded VLPs derived from the hepatitis B core antigen or the bacteriophage Qbeta was able to induce high frequencies of peptide-specific CD8+ T cells (164). TLR9 ligands have an adjuvant activity that directly affects B-cells to undergo isotype switching to Th1 (79). Similarly, single-stranded 10 RNA packaged within some VLPs can activate TLR7 present in B-cells and enhance B-cell responses and class switching to Th1 (79). Importantly, in addition to priming T-cell responses, CpG-loaded VLPs are capable of boosting them without a change in carrier in contrast to heterologous prime-boost strategy (155). Finally, in most of the cases VLPs lack the DNA or RNA viral genome, but have the authentic conformation of viral capsid proteins seen with attenuated virus vaccines, thus they represent a safer alternative to attenuated viruses. Therefore, all characteristics of VLPs described above have made them attractive vaccine candidates for many viral diseases as well as carrier molecules for epitopes for other pathogens (66) (Figure 3). Figure 3. Features of virus-like particles. The ability of VLPs to self-assemble allows incorporation of activators of innate immunity as well as plasmid DNA within their lumen and makes them a promising vehicle for delivering immune stimulating 11 adjuvants and genetic material into target cells. Due to their particulate structure VLPs are readily taken up by APCs and presented effectively in the context of MHC class I molecules, thereby inducing a CTL response. The dense, repetitive nature of VLPs makes them particularly effective in inducing antibody responses. Proteinaceous composition of VLPs permits chemical conjugation or genetic fusion, thereby allowing them to serve as carrier molecules for other pathogens. Figure created by using reference (129). The most straightforward application of VLPs is to use them for vaccination against the corresponding virus from which they are derived. Examples of such VLPs used for vaccines and vaccine development are hepatitis B virus, HPV, hepatitis E virus, influenza, hepatitis C virus, poliovirus, HIV, Ebola virus, Marburg virus, Norwalk virus, Rotavirus, SARS coronavirus VLPs (66). Recently, VLP-based vaccines for HBV and HPV have been licensed commercially. Recombivax (Merck & Co., Inc.) (104) and Engerix (GlaxoSmithKline (GSK)) (7), both were approved in the USA in 1980s. Gardasil (Merck) was approved in 2006 (55). In addition, VLPs can also be used to present foreign epitopes to the immune system. This can be achieved by genetic fusion or chemical conjugation. Genetic fusion of the protein transduction domain of HIV-1 Tat protein with the core gene of HBV provide an example of this approach (27). Alternatively, foreign vaccine proteins may be chemically conjugated to pre-formed VLPs. For example, this approach has been used in the production of HBV core antigen VLPs chemically conjugated with M2 peptide of influenza A virus (51). Chemical conjugation also allows nonprotein targets, such as glycans or other small haptens to be attached to VLPs. One recent example is the VLP-based vaccine against nicotine dependence. This vaccine was developed by covalently coupling nicotine to the surface of VLPs (103). Therefore, the ability of VLPs to serve as carriers of epitopes derived from either the parental virus or foreign sources has enhanced and broadened their potential as prophylactic and therapeutic vaccines. 12 This technological innovation, especially genetic fusion and chemical conjugation, has given the beginning of numerous chimeric VLP-based vaccines. Chimeric VLPs can act as carriers of immunologic epitopes derived from viral, microbial pathogens as well as they can be used to induce autoantibodies to disease-associated self-molecules involved in chronic diseases. For instance, VLPs derived from both double- and single-stranded DNA and RNA viruses encompassing 14 different families have been used for the production of chimeras (134). Among clinically tested vaccines based on VLPs carrying microbial epitopes are two anti-malaria vaccines, Malarivax (Apovia) (118) and RTS,S (GSK) (67). VLP display of self-antigens is used successfully to target molecules that are involved in the pathogenesis of a variety of chronic diseases, including Alzheimer’s disease (190), hypertension (5, 172), obesity (53) and certain cancers, specifically, epithelial cancers (58). Clinical proof of concept has been achieved with a VLP-based anti-angiotensin II vaccine against hypertension. This vaccine known as AngQb has been shown to reduce blood pressure in preclinical models of disease and in humans in a phase IIa clinical trial (5, 172). Therefore, ability of VLPs to carry heterologous epitopes made it possible to target antigens that were previously refractory to vaccine-based approaches. Recently, VLPs have been explored for their ability to serve as delivery vehicles for plasmid DNA (pDNA). The efficient packing of pDNA in VLPs derived from several viruses, such as papillomavirus (PV) (173), polyomavirus (30) and hepatitis E virus (165) was demonstrated. Touze and Coursaget showed higher gene transfer in diverse eukaryotic cell lines using papillomavirus like particles for DNA delivery compared with DNA alone or delivered with liposomes (173). Takamura et al. showed that VLPs derived from orally transmitted viruses, such as hepatitis E, were especially suitable for the oral application of DNA vaccines (165). In a mouse model, orally applied PV VLPs packed with pDNA encoding the HIV-1 Gag protein 13 showed the ability to induce Gag-specific memory CTLs and the generation of Gag-specific serum immunoglobulin G (IgG) and mucosal immunoglobulin A (IgA) antibodies (191). Thus, virus-like particles are a very promising vehicle for delivering genetic material into target cells. VLP technology has the potential for therapeutic vaccination as an economic and effective treatment option for chronic diseases. However, one of the drawbacks that may delay the application of this strategy is a low compositional and architectural consistency of VLPs produced using existing manufacturing methods. This problem creates a demand for the development of new large-scale bioprocesses having high yield, quick process time and reduced total cost. Future directions in manufacturing may include an in vitro chemical self-assembly of VLPs based on capsid components (66). DNA vaccines: Plasmid DNA vaccination was discovered over 10 years ago as a potent method to elicit humoral and cellular immune responses to foreign antigens (169). The simplicity and attractiveness of DNA vaccines were obvious, as they were represented by plasmid DNA which was easy to produce in laboratory bacterial strains at comparatively low cost, as well as easy to store and transport since plasmid DNA is chemically and biologically stable. Conventional plasmids used for vaccination are double-stranded DNA molecules, which can be subdivided into two functional units: an eukaryotic expression cassette (transcriptional unit) and a plasmid backbone (68) (Figure 4). The transcription unit consists of regulatory elements: eukaryotic promoter, enhancer and a polyadenylation signal required for the appropriate expression of the gene of interest in the target cell. An additional expression cassette for genes of immunomodulatory proteins may be inserted in order to enhance the immune response. The plasmid backbone 14 includes elements, such as antibiotic resistance genes and an origin of replication required for the production of plasmid DNA in bacterial cells. In addition, unmethylated CpG motifs, which stimulate the immune response in the host, can be incorporated into this functional unit (Figure 4). Figure 4. Schematic drawing of a plasmid DNA vector. The plasmid DNA can be divided into a transcriptional unit and a bacterial backbone. The transcriptional unit includes a promoter, which can drive high level of protein expression in eukaryotic cells, an ORF for a vaccine antigen, and transcription/termination sequences (Poly A). The bacterial backbone consists of a bacterial origin of replication and an antibiotic resistance gene. Unmethylated CpG motifs can be incorporated into plasmid DNA to stimulate immune responses in the host cell. 15 Figure 5. Mechanisms of antigen presentation after DNA immunization. Application of a plasmid DNA vaccine leads to: (A) Direct transfection of professional APCs resulting in antigen presentation to T cells; (B) Cross-priming, when antigens 16 generated by somatic cells can be taken up by professional APCs to prime T-cell responses. Figure created by using reference (68). The principle of DNA vaccination is based on the production of the antigen(s) of interest in vivo by cells of the vaccinated person or animal, whereby pathogen-derived genetic information combined with eukaryotic expression system is applied. It means that the in vivo synthesized antigens retain their native structures, including post-translational modifications, accurately mimicking their ‘natural’ counterpart. There are two main mechanisms by which the antigen encoded by plasmid DNA is processed and presented to elicit an immune response: direct transfection of professional APCs and cross-priming (68) (Figure 5A). When the gene of interest is transfected directly into professional APCs and expressed after transfer into the nucleus, the resulting protein is directed into the MHC class I pathway of antigen presentation. On the other hand, if the antigen is first secreted or released following expression in myocytes and then taken up by APCs, the protein is targeted to the MHC class II pathway of antigen presentation, although such antigens may also be able to ‘cross-over’ into the MHC class I pathway following internalization into APCs (cross-priming) (Figure 5B). In contrast to DNA vaccines, exogenous delivery of killed or subunit vaccines leads preferentially to the Th2 immune response, since the antigens are taken up by APCs and targeted into the MHC class II molecules. Therefore, the ability to present antigens through both MHC classes makes DNA vaccines able to stimulate both arms of the immune system simultaneously (24). Besides the intrinsic immunological advantages of DNA vaccines, there are other factors making these vaccines attractive. As DNA vaccine plasmids are non-live, non-replicating and non-spreading, there is little risk of either reversion to a disease-causing form or secondary infection. Moreover, DNA vaccines are highly flexible, encoding several types of genes 17 including viral or bacterial antigens, and immunological and biological proteins. In addition, the use of DNA approach avoids the risks linked to the manufacture of killed vaccines, as exemplified by the tainting of a polio vaccine with live polio virus owing to a production error (122). Due to their simplicity and versatility DNA vaccines became a promising strategy to develop immunity against several infectious diseases including HIV, tuberculosis, malaria, influenza and the newly emerging Ebola and SARS viruses (33, 42, 124, 141, 158). DNA immunization was used in a wide variety of model systems (72, 177, 178). As a result, four veterinary DNA vaccines have been approved for commercial sale by the respective regulatory agencies in the USA and Canada. One against West Nile virus in horses (37), one against infectious hematopoietic necrosis virus in schooled salmon (56), one for treatment of melanoma in dogs (17) and the most recent licensure, growth hormone releasing hormone product for foetal loss in swine (171). However, a much lower immunogenicity of DNA vaccines has been observed in higher primates and clinical trials in humans and high doses and/or multiple immunizations are required to induce protective immune responses (80, 90), which hampers the practical application of DNA vaccines. As a result of the limited immunogenicity of DNA vaccines, considerable efforts are underway to solve this problem. There are several ways in which antigen expression and immunogenicity can be improved for DNA vaccine platform: optimization of the transcriptional elements in the plasmid backbone with the aim to improve antigen expression levels; strategies to improve protein expression of the gene of interest, inclusion of adjuvants in the formulation or as immune modulators, and the development of new delivery systems for specific targeting and/or better DNA uptake (84). The latter two approaches influence the magnitude, the type of 18 immune response as well as they can facilitate antigen entry into DCs. Approaches that are aimed at targeting selected antigens to APCs, especially to DCs, have shown potential in vaccinating against disease in animal models and humans as targeting vaccines to DC appears to be essential for the induction and modulation of immune responses. Improving immunogenicity through formulation One current trend in DNA vaccine formulation is the use of biodegradable polymeric microparticles and liposomes (121). Polymeric delivery systems and liposomal delivery systems represent novel DNA delivery platforms that protect DNA from degradation and facilitate targeting to specific tissues, thereby increasing the transfection efficacy of target cells. It has been shown that microparticle constructs, such as poly (D,L-lactic-coglycolic acid) copolymers (PLGA), can be easily phagocytosed by DCs in vitro or in vivo (120, 179) and this can stimulate further DC maturation in the lymph node (139). The immune responses of DNA-loaded microparticles were investigated in mice, non-human primates and humans (82, 111, 126, 127, 181). It was shown that the parenteral administration of PLGA encapsulated DNA induces a significant CTL response (105). In addition, the oral administration of encapsulated DNA in PLGA microparticles elicited systemic and mucosal immune responses upon challenge (81). Moreover, polymeric matrices can be designed by the incorporation of ligands, such as folates, transferrin, antibodies and sugars, to enhance tissue targeting (108). Similar to polymers, liposomes are taken up by APCs and mediate MHC class I antigen presentation, and have been shown to induce cellular and humoral immunity (180, 182). Furthermore, immunogenicity of DNA vaccines can be improved by directly targeting the encoded vaccine protein to DCs in vivo. Nchinda et al. demonstrated that the incorporation of antigens into an antibody against the DC 19 endocytic receptor, DEC205, enhanced the antigen presentation by DCs in lymphoid tissues, thereby improving the efficacy of DNA vaccines (119). Improving immunogenicity by including immune modulatory adjuvants The DNA itself has adjuvant properties through CpG motifs. For instance, synthetic oligodeoxynucleotides containing unmethylated CpG motifs that are DNA specific sequences recognized by TLR9 can act as immune adjuvants in mice, as they boost the humoral and cellular responses to co-administered antigens (73). Moreover, the addition of many CpG motifs into a plasmid backbone has improved the immunogenicity of DNA vaccines inducing Th1-biased immune response (133). However, it has been recently shown that DNA vaccine immunogenicity has been attributed to its double-stranded structure, which activates TANK-binding kinase 1dependent innate immune signaling pathway in the absence of TLRs (31). In addition to self-adjuvanting effect of DNA, co-administration of genetic adjuvants such as cytokines, chemokines or co-stimulatory molecules is a separate approach in which genes are co-delivered in order to enhance the magnitude and type of desired immune responses to DNA vaccines. For example, the employment of Th1-associated cytokines such as interleukin (IL)-12 and IL-15 in non-human primates induced antigen specific cellular immune responses (29, 151). DNA vaccines co-injected with plasmids encoding Th2-inducing cytokines such as IL4 augmented antigen-specific humoral immune responses (87). Furthermore, proinflammatory cytokines have the ability to modulate immune responses to DNA vaccines. For instance, the granulocyte-macrophage colony-stimulating factor (GM-CSF) is capable of enhancing humoral and cellular immune responses (168). However, the timing of GM-CSF administration influences the Th1/Th2 immune response (83). Chemokines can also modulate the immune response and 20 protective immunity. Sin and colleagues observed that co-injection with IL-8 and RANTES plasmid DNAs dramatically enhanced antigen-specific Th1 type cellular immune responses and protection from lethal herpes simplex virus-2 challenge (159). In addition, co-delivery of the costimulatory molecules CD86 during DNA vaccination enhanced both helper and cytotoxic Tcell responses (2). Improving immunogenicity by using next-generation delivery strategies The direct injection of plasmid DNA into muscle or skin is still the most widely used. However, the biggest drawback of this delivery method is the low efficacy achieved in larger animals and humans. Therefore, development of physical delivery methods to increase the transfection efficiency of target cells has been a primary focus of research in more recent years. Physical methods for pDNA transfection comprise electroporation, ballistic needle-free delivery systems and microporation. Microporation involves use of hundreds of microneedles that enable topical immunization with naked plasmid DNA as they can bypass the stratum corneum, thereby delivering plasmid DNA to APCs of the skin. This induces stronger and less variable immune responses than via needle-based injections (109). Another physical method widely employed in DNA vaccination is particle-mediated epidermal delivery (PMED) also called the “gene gun”. In this procedure, DNA-coated microparticles composed of gold are accelerated to high velocity to penetrate cell membranes in the epidermis where a variety of cells, including the Langerhans cells, the APCs of the skin, can be directly transfected (130). As a result, the gene gun immunization have 10-100-fold more expression of the DNA-encoded protein than intramuscular vaccinations and 100-fold less DNA 21 is required for the same level of expression (12). The gene gun technique has been used to immunize nonhuman primates against a variety of diseases, including HIV, Ebola, Japanese encephalitis, hepatitis E and B, influenza, smallpox etc (52). Thus, PMED can induce higher antibody and /or CD8+ T cell responses in mice and monkeys with substantially lower doses of DNA in comparison to needle-based approaches. An alternative approach based on the use of electric pulses to transiently permeabilize cell membranes, thus permitting cellular uptake of plasmid DNA, is electroporation. It has been extensively studied in large animal species such as dogs, pigs, cattle and non-human primates to deliver DNA vaccines (10, 75, 148, 176). The potential of electroporation for DNA vaccination has been demonstrated by the increased protein expression and a robust stimulation of the immune response. Electroporation might allow for less frequent immunizations with the DNA vaccines, and can improve both cellular and humoral responses (149). Currently, the DNA platform represents almost one quarter of all gene therapy vector systems under clinical evaluation (84). The interest to further development of this vaccination strategy is strengthened by recent licenses in the area of animal health and by the improvements in immune potency reported in the non-human primate model systems. Viral Vectors: The idea of using viruses as gene-delivery systems to combat diseases stems from a documented immunogenicity and safety profile of the majority of existing live-attenuated vaccines. Attenuated live-virus vaccines appear to be the most effective immunogens that mimic natural infection. Another important attribute of these vaccines is high cost efficiency for mass 22 vaccination as they are optimal for large-scale production. Recent developments in genetic engineering coupled with the accumulated data on the nucleotide structure of viral genomes and functions of viral genes allowed precise manipulations of viral genomes cloned as bacterial plasmids (167) or bacterial artificial chromosomes (36). This set of diverse technologies is applicable to the generation of recombinant DNA and RNA viruses and is based on reverse genetics, which is the most powerful tool in modern virology and has the potential for a variety of applications, from the basic understanding of virus biology to the development of genetically engineered viral vaccines and therapeutic gene delivery systems. Reverse genetics approach can be applied for the construction of genetically attenuated vaccine strains (100) or development of live viral vectors that are engineered to induce immunity to an array of proteins encoded by genes inserted into the vector (22). One of the first licensed recombinant virus vaccines was represented by vaccinia virus carrying the gene encoding rabies virus glycoproteins. This vaccine reduced the spread of rabies virus in wildlife populations and also reduced the spill-over of virus infection from wildlife to domestic animals and humans (23, 96). Reverse genetics approach led to the development of a wide range of different DNA (1, 163) and RNA (20, 131, 186) viruses as vectors for efficient delivery of vaccine antigens. This strategy is especially attractive for the development of vaccines against highly dangerous pathogens, such as HIV, for which even a short course of replication of an attenuated live virus in the host is not acceptable due to the possibility of integration of vaccine genes or reversion of the vaccine strain to wild-type. The appropriate balance between safety and immunogenicity is a critical issue for the construction of any live attenuated vaccine. Inadequate safety is the main reason for retaining of many replication-competent viral vectors from entry into human clinical trials. In order to increase the safety of viral delivery vectors, the development of replication23 defective viruses produced in complementing cell systems was introduced (43). However, the abrogation of the ability to replicate in the vaccinated host is commonly associated with lower immune responses induced by replication-incompetent viral vectors. In this connection, replication-competent viral vectors that originate from commercial live-attenuated vaccines seem to be a promising perspective for the development of novel safe and immunogenic vaccines (20, 161). Major concerns associated with viral vector vaccines include the potential risk of changes in viral tropism induced by introduced modifications, and the presence of pre-existing antivector host immunity. The latter drawback can be managed by certain approaches including engineering of vaccine vectors from viral strains that have not circulated widely in host populations (101) or by mutating viral surface proteins in order to evade host neutralizing antibodies (147). Additional approach to circumvent the problem of pre-existing antivector immunity is to exploit the asymmetry in induction of systemic and mucosal immune responses. Specifically, if the mucosal immune system remains naïve after systemic immunization against certain virus, it might still be amenable to immunization with recombinant viral vector based on the same or related virus. For instance, mucosal administration of measles and measles-rubella vaccines was shown to be more efficient than subcutaneous administration in pre-immunized humans (16, 41). On the contrary, in the case of active mucosal immunity to a vector, subcutaneous vaccination may be an efficient way to overcome pre-existing immunity (48). However, the pre-existing immunity is not detrimental for some viral vectors, thus measles virus (MV) is able to induce cellular and humoral immunity after re-vaccination (92). 24 Currently, a variety of recombinant viral vectors are either under preclinical or early clinical investigation (89). Among the most promising viral vectors are adenovirus vectors (183, 189), adeno-associated virus (AAV) vectors (91), poxvirus vectors (62), alphavirus vectors (113, 145), MV (88), vesicular stomatitis virus (VSV) (78), poliovirus (35), and herpes virus (44). Each of these vectors has unique properties that should be considered for the selection of the optimal vector for the vaccination strategy of choice. A high level of antigen expression and efficient targeted delivery are the main goals in the process of the development of viral vector vaccines. Using the targeted vector delivery, the antigen-coding gene can be directed to professional APCs or to a specific tissue. For example, members of alphavirus genus and poxviruses are able to transduce DCs without inhibiting the immunostimulatory function of these APCs (114, 174). The transduction of DCs may elicit direct priming of an immune response. However, in the case of poxviruses it was shown that CTL responses against modified vaccinia virus Ankara were dominated by cross-priming in vivo, despite the ability of the virus to infect DCs (57). Adenoviruses (9), MV (15) and poliovirus (34) infect predominantly via mucosal surfaces. Vectors based on these viruses may therefore be effective for mucosal administration and induction of mucosal immunity. In addition, it was shown that VSV recombinants expressing the hemagglutinin protein of influenza virus (146) or the MV hemagglutinin (152) were able to induce protective immune responses after intranasal immunization in rodents. However, in the case of VSV as an intranasal vaccine vector potential neurotoxicity should be taken into consideration (143). A positive characteristic of VSV as a vaccine vector is that no proteins encoded by the VSV genome have the capacity to interfere with the host interferon 25 response (162). It seems likely that the cytokine response activated in target cells during the VSV infection will possesses an adjuvant activity for antigen-specific responses to vaccine antigens. Viral vectors are different in terms of their capacity for the insertion of foreign genetic material. Thus, non-enveloped viruses such as poliovirus, adenovirus, and AAV have a rigid capsid structure that does not allow incorporation of more genetic material than the size of wildtype virus genome. For example, adenoviral capsid can package ~105% of the adenovirus genome size. It means that a transgene of 3-4 kilo base pairs (kbp) can be packaged by a replication-competent E3-deleted adenovirus. Replication-defective vectors bearing additional deletions of essential E1 region in their genomes are able to accommodate up to 7.5 kbp of foreign DNA (140). Some viruses with larger genomes can tolerate larger insertions of foreign genetic material, but vector construction may in turn be more laborious. Viral vectors that replicate in the nucleus represent potential risk of incorporation of their genes into the host genome. Thus, vectors with entirely cytoplasmic replication cycle, such as poxviruses or MV, may be preferable to reduce the risk of integration. For some applications, especially in gene replacement therapy, integrating vectors, such as retroviruses, could be beneficial. Another important characteristic of viral vaccine vectors is that some of them can induce innate immune responses through pattern recognition receptors (PRRs), acting as adjuvants to the delivered antigens. This adjuvant effects are mediated through type I interferon production and an inflammatory response. It has been proposed that the more PRRs are activated, the more robust the immune response induced by the vaccine (136). A group of membrane-bound signaling PRRs is represented by an array of TLRs and the mannose receptor (8, 18). Each 26 member of the TLR family recognizes distinct pathogen-associated molecular pattern and mediates the activation of NF-kB and over signaling pathways (166). TLRs recognize several molecular patterns associated with viruses including surface glycoproteins (TLR1, 2 and 4) (19, 60) and nucleic acids (TLR3, 7, 8 and 9) (3, 40, 94, 175). TLR3 is a receptor for double-stranded (ds) RNA that transmits signals that activate NF-kB and IFN-b promoter (102). The replication cycle of many RNA viruses has a double-stranded RNA replication intermediate, which is able to activate innate immune response through the TLR3 pathway (144). TLR3 signaling may be of particular importance for the immune responses to viruses that do not directly infect DCs as it was shown to promote cross-priming of CD8+ T cells in response to dsRNA (154). The diversity of viruses and replication strategies employed by them creates unlimited perspectives for their use as gene-delivery vehicles to combat diseases. Although many problems associated with safety and efficient manufacturing of viral vectors remain to be addressed this strategy is one of the most promising in respect of development of a cure against unconquerable diseases, such as AIDS, malaria, tuberculosis, influenza, and various cancers. Prime-boost strategy: Commonly, schemes of vaccination include repeated administrations of the same vaccine (homologous boosting) to achieve protective humoral immune responses. However, immunity developed after the primary immunization by a vaccine antigen can impair effective antigen presentation after subsequent injections. This is believed to be a reason for a low cellular immune response induced by the use of homologous boosting. As a solution to this problem, the 27 sequential administration of a vaccine antigen using different antigen-delivery systems (heterologous boosting) was proposed and referred to as ‘prime-boosting’. This strategy evolved after the gene-based vaccines had emerged as promising alternatives to traditional vaccine strategies. The basic prime-boost strategy involves priming the immune system to an antigen delivered by one vector and then boosting this immunity by re-administration of the antigen in the context of a distinct vector. This vaccination scheme induces synergistic enhancement of immunity to the target antigen reflected in an increased number of antigen-specific T cells, selective enrichment of high avidity T-cells and increased efficacy against pathogen challenge (185). Recently, several studies have demonstrated the efficacy of prime-boost vaccination strategies in generating cellular immunity to a variety of pathogens. These include, Mycobacteriun tuberculosis (45, 137), HIV (21, 188) and simian immunodeficiency virus (153), malaria (28, 170), Leishmania (25, 138), Ebola virus (107), hepatitis C virus (38, 128), herpes simplex virus (49, 160), human papillomavirus (47, 95), hepatitis B virus (110), and Japanese encephalitis virus (86). Thus, heterologous prime-boost vaccination strategies with use of DNA vaccines and viral vectors generate high levels of both CD4+ and CD8+ T-cell mediated immunity, which is currently viewed as crucial for protection against a variety of pathogens, including HIV. Conclusions: Last decades have brought remarkable advances in the field of molecular biology and gene sequencing that help to uncover the mechanisms of interactions between microorganisms and the host immune system. This progress has significantly facilitated the development of rationally28 designed vaccines for both prophylaxis and therapy of a wide range of diseases. Currently, the field of vaccine development has a set of novel powerful tools. This arsenal of tools includes reverse vaccinology, which allows identification of potential vaccine antigens without the need for cultivating the pathogen, as well as a wide variety of vectors that allow increase and manipulation of the immune response towards the desired type of immunity. Some successful steps have been made to solve the key problems associated with the relatively low immunogenicity of DNA vaccines and pre-existing immunity to viral vectors. These steps include development of a DNA prime and recombinant viral vector boost strategy, which may help circumvent pre-existing immunity to the viral vector, and generation of different methods that improve the antigen expression by vaccine vectors and presentation of expressed antigens to the immune system. Some of these new vaccine strategies are being translated into clinical trials. References: 1. 2. 3. 4. Abe, S., K. Okuda, T. Ura, A. Kondo, A. Yoshida, S. Yoshizaki, H. Mizuguchi, D. Klinman, and M. Shimada. 2009. Adenovirus type 5 with modified hexons induces robust transgene-specific immune responses in mice with pre-existing immunity against adenovirus type 5. J Gene Med. Agadjanyan, M. G., M. A. Chattergoon, M. J. Holterman, B. Monzavi-Karbassi, J. J. Kim, T. Dentchev, D. Wilson, V. Ayyavoo, L. J. Montaner, T. Kieber-Emmons, R. P. Sekaly, and D. B. Weiner. 2003. Costimulatory molecule immune enhancement in a plasmid vaccine model is regulated in part through the Ig constant-like domain of CD80/86. J Immunol 171:4311-9. Alexopoulou, L., A. C. Holt, R. Medzhitov, and R. A. Flavell. 2001. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature 413:732-8. Almanza, H., C. Cubillos, I. Angulo, F. Mateos, J. R. Caston, W. H. van der Poel, J. Vinje, J. Barcena, and I. Mena. 2008. Self-assembly of the recombinant capsid protein of a swine norovirus into virus-like particles and evaluation of monoclonal antibodies cross-reactive with a human strain from genogroup II. J Clin Microbiol 46:3971-9. 29 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. Ambuhl, P. M., A. C. Tissot, A. Fulurija, P. Maurer, J. Nussberger, R. Sabat, V. Nief, C. Schellekens, K. Sladko, K. Roubicek, T. Pfister, M. Rettenbacher, H. D. Volk, F. Wagner, P. Muller, G. T. Jennings, and M. F. Bachmann. 2007. A vaccine for hypertension based on virus-like particles: preclinical efficacy and phase I safety and immunogenicity. J Hypertens 25:63-72. Andre, F. E. 1990. Overview of a 5-year clinical experience with a yeast-derived hepatitis B vaccine. Vaccine 8 Suppl:S74-8; discussion S79-80. Andre, F. E., and A. Safary. 1987. Summary of clinical findings on Engerix-B, a genetically engineered yeast derived hepatitis B vaccine. Postgrad Med J 63 Suppl 2:169-77. Apostolopoulos, V., and I. F. McKenzie. 2001. Role of the mannose receptor in the immune response. Curr Mol Med 1:469-74. Babiuk, L. A., and S. K. Tikoo. 2000. Adenoviruses as vectors for delivering vaccines to mucosal surfaces. J Biotechnol 83:105-13. Babiuk, S., C. Tsang, S. van Drunen Littel-van den Hurk, L. A. Babiuk, and P. J. Griebel. 2007. A single HBsAg DNA vaccination in combination with electroporation elicits long-term antibody responses in sheep. Bioelectrochemistry 70:269-74. Bachmann, M. F., and R. M. Zinkernagel. 1996. The influence of virus structure on antibody responses and virus serotype formation. Immunol Today 17:553-8. Barry, M. A., and S. A. Johnston. 1997. Biological features of genetic immunization. Vaccine 15:788-91. Barth, H., A. Ulsenheimer, G. R. Pape, H. M. Diepolder, M. Hoffmann, C. Neumann-Haefelin, R. Thimme, P. Henneke, R. Klein, G. Paranhos-Baccala, E. Depla, T. J. Liang, H. E. Blum, and T. F. Baumert. 2005. Uptake and presentation of hepatitis C virus-like particles by human dendritic cells. Blood 105:3605-14. Bartlam, M., Y. Xu, and Z. Rao. 2007. Structural proteomics of the SARS coronavirus: a model response to emerging infectious diseases. J Struct Funct Genomics 8:85-97. Bellanti, J. A., B. J. Zeligs, J. Mendez-Inocencio, M. L. Garcia-Garcia, R. IslasRomero, B. Omidvar, J. Omidvar, G. Kim, J. Fernandez De Castro, J. Sepulveda Amor, L. Walls, W. J. Bellini, and J. L. Valdespino-Gomez. 2004. Immunologic studies of specific mucosal and systemic immune responses in Mexican school children after booster aerosol or subcutaneous immunization with measles vaccine. Vaccine 22:1214-20. Bennett, J. V., J. Fernandez de Castro, J. L. Valdespino-Gomez, L. Garcia-Garcia Mde, R. Islas-Romero, G. Echaniz-Aviles, A. Jimenez-Corona, and J. SepulvedaAmor. 2002. Aerosolized measles and measles-rubella vaccines induce better measles antibody booster responses than injected vaccines: randomized trials in Mexican schoolchildren. Bull World Health Organ 80:806-12. Bergman, P. J., M. A. Camps-Palau, J. A. McKnight, N. F. Leibman, D. M. Craft, C. Leung, J. Liao, I. Riviere, M. Sadelain, A. E. Hohenhaus, P. Gregor, A. N. Houghton, M. A. Perales, and J. D. Wolchok. 2006. Development of a xenogeneic DNA vaccine program for canine malignant melanoma at the Animal Medical Center. Vaccine 24:4582-5. Beutler, B., Z. Jiang, P. Georgel, K. Crozat, B. Croker, S. Rutschmann, X. Du, and K. Hoebe. 2006. Genetic analysis of host resistance: Toll-like receptor signaling and immunity at large. Annu Rev Immunol 24:353-89. 30 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. Boehme, K. W., M. Guerrero, and T. Compton. 2006. Human cytomegalovirus envelope glycoproteins B and H are necessary for TLR2 activation in permissive cells. J Immunol 177:7094-102. Brandler, S., M. Lucas-Hourani, A. Moris, M. P. Frenkiel, C. Combredet, M. Fevrier, H. Bedouelle, O. Schwartz, P. Despres, and F. Tangy. 2007. Pediatric Measles Vaccine Expressing a Dengue Antigen Induces Durable Serotype-specific Neutralizing Antibodies to Dengue Virus. PLoS Negl Trop Dis 1:e96. Brave, A., A. Boberg, L. Gudmundsdotter, E. Rollman, K. Hallermalm, K. Ljungberg, P. Blomberg, R. Stout, S. Paulie, E. Sandstrom, G. Biberfeld, P. Earl, B. Moss, J. H. Cox, and B. Wahren. 2007. A new multi-clade DNA prime/recombinant MVA boost vaccine induces broad and high levels of HIV-1-specific CD8(+) T-cell and humoral responses in mice. Mol Ther 15:1724-33. Brave, A., K. Ljungberg, B. Wahren, and M. A. Liu. 2007. Vaccine delivery methods using viral vectors. Mol Pharm 4:18-32. Brochier, B., and P. P. Pastoret. 1993. Rabies eradication in Belgium by fox vaccination using vaccinia-rabies recombinant virus. Onderstepoort J Vet Res 60:469-75. Cardoso, A. I., M. Blixenkrone-Moller, J. Fayolle, M. Liu, R. Buckland, and T. F. Wild. 1996. Immunization with plasmid DNA encoding for the measles virus hemagglutinin and nucleoprotein leads to humoral and cell-mediated immunity. Virology 225:293-9. Carson, C., M. Antoniou, M. B. Ruiz-Arguello, A. Alcami, V. Christodoulou, I. Messaritakis, J. M. Blackwell, and O. Courtenay. 2009. A prime/boost DNA/Modified vaccinia virus Ankara vaccine expressing recombinant Leishmania DNA encoding TRYP is safe and immunogenic in outbred dogs, the reservoir of zoonotic visceral leishmaniasis. Vaccine 27:1080-6. Chandy, S., K. Yoshimatsu, R. G. Ulrich, M. Mertens, M. Okumura, P. Rajendran, G. T. John, V. Balraj, J. Muliyil, J. Mammen, P. Abraham, J. Arikawa, and G. Sridharan. 2008. Seroepidemiological study on hantavirus infections in India. Trans R Soc Trop Med Hyg 102:70-4. Chen, X., Y. Yu, Q. Pan, Z. Tang, J. Han, and G. Zang. 2008. Enhancement of cytotoxic T lymphocyte activity by dendritic cells loaded with Tat-protein transduction domain-fused hepatitis B virus core antigen. Acta Biochim Biophys Sin (Shanghai) 40:996-1004. Chinchilla, M., M. F. Pasetti, S. Medina-Moreno, J. Y. Wang, O. G. Gomez-Duarte, R. Stout, M. M. Levine, and J. E. Galen. 2007. Enhanced immunity to Plasmodium falciparum circumsporozoite protein (PfCSP) by using Salmonella enterica serovar Typhi expressing PfCSP and a PfCSP-encoding DNA vaccine in a heterologous prime-boost strategy. Infect Immun 75:3769-79. Chong, S. Y., M. A. Egan, M. A. Kutzler, S. Megati, A. Masood, V. Roopchard, D. Garcia-Hand, D. C. Montefiori, J. Quiroz, M. Rosati, E. B. Schadeck, J. D. Boyer, G. N. Pavlakis, D. B. Weiner, M. Sidhu, J. H. Eldridge, and Z. R. Israel. 2007. Comparative ability of plasmid IL-12 and IL-15 to enhance cellular and humoral immune responses elicited by a SIVgag plasmid DNA vaccine and alter disease progression following SHIV(89.6P) challenge in rhesus macaques. Vaccine 25:4967-82. Clark, B., W. Caparros-Wanderley, G. Musselwhite, M. Kotecha, and B. E. Griffin. 2001. Immunity against both polyomavirus VP1 and a transgene product induced 31 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. following intranasal delivery of VP1 pseudocapsid-DNA complexes. J Gen Virol 82:2791-7. Coban, C., S. Koyama, F. Takeshita, S. Akira, and K. J. Ishii. 2008. Molecular and cellular mechanisms of DNA vaccines. Hum Vaccin 4:453-6. Comanducci, M., S. Bambini, B. Brunelli, J. Adu-Bobie, B. Arico, B. Capecchi, M. M. Giuliani, V. Masignani, L. Santini, S. Savino, D. M. Granoff, D. A. Caugant, M. Pizza, R. Rappuoli, and M. Mora. 2002. NadA, a novel vaccine candidate of Neisseria meningitidis. J Exp Med 195:1445-54. Cox, K. S., J. H. Clair, M. T. Prokop, K. J. Sykes, S. A. Dubey, J. W. Shiver, M. N. Robertson, and D. R. Casimiro. 2008. DNA gag/adenovirus type 5 (Ad5) gag and Ad5 gag/Ad5 gag vaccines induce distinct T-cell response profiles. J Virol 82:8161-71. Crotty, S., and R. Andino. 2004. Poliovirus vaccine strains as mucosal vaccine vectors and their potential use to develop an AIDS vaccine. Adv Drug Deliv Rev 56:835-52. Crotty, S., C. J. Miller, B. L. Lohman, M. R. Neagu, L. Compton, D. Lu, F. X. Lu, L. Fritts, J. D. Lifson, and R. Andino. 2001. Protection against simian immunodeficiency virus vaginal challenge by using Sabin poliovirus vectors. J Virol 75:7435-52. Cui, H. Y., Y. F. Wang, X. M. Shi, T. Q. An, G. Z. Tong, D. S. Lan, L. He, C. J. Liu, and M. Wang. 2009. Construction of an infectious Marek's disease virus bacterial artificial chromosome and characterization of protection induced in chickens. J Virol Methods 156:66-72. Davidson, A. H., J. L. Traub-Dargatz, R. M. Rodeheaver, E. N. Ostlund, D. D. Pedersen, R. G. Moorhead, J. B. Stricklin, R. D. Dewell, S. D. Roach, R. E. Long, S. J. Albers, R. J. Callan, and M. D. Salman. 2005. Immunologic responses to West Nile virus in vaccinated and clinically affected horses. J Am Vet Med Assoc 226:240-5. Deng, Y., K. Zhang, W. Tan, Y. Wang, H. Chen, X. Wu, and L. Ruan. 2009. A recombinant DNA and vaccinia virus prime-boost regimen induces potent long-term Tcell responses to HCV in BALB/c mice. Vaccine 27:2085-8. Dhiman, N., R. Bonilla, D. J. O'Kane, and G. A. Poland. 2001. Gene expression microarrays: a 21st century tool for directed vaccine design. Vaccine 20:22-30. Diebold, S. S., T. Kaisho, H. Hemmi, S. Akira, and C. Reis e Sousa. 2004. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 303:1529-31. Dilraj, A., F. T. Cutts, J. F. de Castro, J. G. Wheeler, D. Brown, C. Roth, H. M. Coovadia, and J. V. Bennett. 2000. Response to different measles vaccine strains given by aerosol and subcutaneous routes to schoolchildren: a randomised trial. Lancet 355:798-803. Dobano, C., M. Sedegah, W. O. Rogers, S. Kumar, H. Zheng, S. L. Hoffman, and D. L. Doolan. 2009. Plasmodium: Mammalian codon optimization of malaria plasmid DNA vaccines enhances antibody responses but not T cell responses nor protective immunity. Exp Parasitol. Dudek, T., and D. M. Knipe. 2006. Replication-defective viruses as vaccines and vaccine vectors. Virology 344:230-9. Duke, C. M., C. A. Maguire, M. C. Keefer, H. J. Federoff, W. J. Bowers, and S. Dewhurst. 2007. HSV-1 amplicon vectors elicit polyfunctional T cell responses to HIV1 Env, and strongly boost responses to an adenovirus prime. Vaccine 25:7410-21. 32 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. Elvang, T., J. P. Christensen, R. Billeskov, T. Thi Kim Thanh Hoang, P. Holst, A. R. Thomsen, P. Andersen, and J. Dietrich. 2009. CD4 and CD8 T cell responses to the M. tuberculosis Ag85B-TB10.4 promoted by adjuvanted subunit, adenovector or heterologous prime boost vaccination. PLoS ONE 4:e5139. Etz, H., D. B. Minh, T. Henics, A. Dryla, B. Winkler, C. Triska, A. P. Boyd, J. Sollner, W. Schmidt, U. von Ahsen, M. Buschle, S. R. Gill, J. Kolonay, H. Khalak, C. M. Fraser, A. von Gabain, E. Nagy, and A. Meinke. 2002. Identification of in vivo expressed vaccine candidate antigens from Staphylococcus aureus. Proc Natl Acad Sci U S A 99:6573-8. Fiander, A. N., A. J. Tristram, E. J. Davidson, A. E. Tomlinson, S. Man, P. J. Baldwin, J. C. Sterling, and H. C. Kitchener. 2006. Prime-boost vaccination strategy in women with high-grade, noncervical anogenital intraepithelial neoplasia: clinical results from a multicenter phase II trial. Int J Gynecol Cancer 16:1075-81. Fischer, L., J. P. Tronel, C. Pardo-David, P. Tanner, G. Colombet, J. Minke, and J. C. Audonnet. 2002. Vaccination of puppies born to immune dams with a canine adenovirus-based vaccine protects against a canine distemper virus challenge. Vaccine 20:3485-97. Fotouhi, F., H. Soleimanjahi, M. H. Roostaee, and F. Behzadian. 2008. Enhancement of protective humoral immune responses against Herpes simplex virus-2 in DNAimmunized guinea-pigs using protein boosting. FEMS Immunol Med Microbiol 54:1826. Frank, I., M. Piatak, Jr., H. Stoessel, N. Romani, D. Bonnyay, J. D. Lifson, and M. Pope. 2002. Infectious and whole inactivated simian immunodeficiency viruses interact similarly with primate dendritic cells (DCs): differential intracellular fate of virions in mature and immature DCs. J Virol 76:2936-51. Fu, T. M., K. M. Grimm, M. P. Citron, D. C. Freed, J. Fan, P. M. Keller, J. W. Shiver, X. Liang, and J. G. Joyce. 2009. Comparative immunogenicity evaluations of influenza A virus M2 peptide as recombinant virus like particle or conjugate vaccines in mice and monkeys. Vaccine 27:1440-7. Fuller, D. H., P. Loudon, and C. Schmaljohn. 2006. Preclinical and clinical progress of particle-mediated DNA vaccines for infectious diseases. Methods 40:86-97. Fulurija, A., T. A. Lutz, K. Sladko, M. Osto, P. Y. Wielinga, M. F. Bachmann, and P. Saudan. 2008. Vaccination against GIP for the treatment of obesity. PLoS ONE 3:e3163. Gamvrellis, A., D. Leong, J. C. Hanley, S. D. Xiang, P. Mottram, and M. Plebanski. 2004. Vaccines that facilitate antigen entry into dendritic cells. Immunol Cell Biol 82:506-16. Garland, S. M., M. Hernandez-Avila, C. M. Wheeler, G. Perez, D. M. Harper, S. Leodolter, G. W. Tang, D. G. Ferris, M. Steben, J. Bryan, F. J. Taddeo, R. Railkar, M. T. Esser, H. L. Sings, M. Nelson, J. Boslego, C. Sattler, E. Barr, and L. A. Koutsky. 2007. Quadrivalent vaccine against human papillomavirus to prevent anogenital diseases. N Engl J Med 356:1928-43. Garver, K. A., S. E. LaPatra, and G. Kurath. 2005. Efficacy of an infectious hematopoietic necrosis (IHN) virus DNA vaccine in Chinook Oncorhynchus tshawytscha and sockeye O. nerka salmon. Dis Aquat Organ 64:13-22. 33 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. Gasteiger, G., W. Kastenmuller, R. Ljapoci, G. Sutter, and I. Drexler. 2007. Crosspriming of cytotoxic T cells dictates antigen requisites for modified vaccinia virus Ankara vector vaccines. J Virol 81:11925-36. Gathuru, J. K., F. Koide, G. Ragupathi, J. L. Adams, R. T. Kerns, T. P. Coleman, and P. O. Livingston. 2005. Identification of DHBcAg as a potent carrier protein comparable to KLH for augmenting MUC1 antigenicity. Vaccine 23:4727-33. Gaucher, D., R. Therrien, N. Kettaf, B. R. Angermann, G. Boucher, A. FilaliMouhim, J. M. Moser, R. S. Mehta, D. R. Drake, 3rd, E. Castro, R. Akondy, A. Rinfret, B. Yassine-Diab, E. A. Said, Y. Chouikh, M. J. Cameron, R. Clum, D. Kelvin, R. Somogyi, L. D. Greller, R. S. Balderas, P. Wilkinson, G. Pantaleo, J. Tartaglia, E. K. Haddad, and R. P. Sekaly. 2008. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med 205:3119-31. Georgel, P., Z. Jiang, S. Kunz, E. Janssen, J. Mols, K. Hoebe, S. Bahram, M. B. Oldstone, and B. Beutler. 2007. Vesicular stomatitis virus glycoprotein G activates a specific antiviral Toll-like receptor 4-dependent pathway. Virology 362:304-13. Giuliani, M. M., J. Adu-Bobie, M. Comanducci, B. Arico, S. Savino, L. Santini, B. Brunelli, S. Bambini, A. Biolchi, B. Capecchi, E. Cartocci, L. Ciucchi, F. Di Marcello, F. Ferlicca, B. Galli, E. Luzzi, V. Masignani, D. Serruto, D. Veggi, M. Contorni, M. Morandi, A. Bartalesi, V. Cinotti, D. Mannucci, F. Titta, E. Ovidi, J. A. Welsch, D. Granoff, R. Rappuoli, and M. Pizza. 2006. A universal vaccine for serogroup B meningococcus. Proc Natl Acad Sci U S A 103:10834-9. Gomez, C. E., J. L. Najera, M. Krupa, and M. Esteban. 2008. The poxvirus vectors MVA and NYVAC as gene delivery systems for vaccination against infectious diseases and cancer. Curr Gene Ther 8:97-120. Graham, S. P., Y. Honda, R. Pelle, D. M. Mwangi, E. J. Glew, E. P. de Villiers, T. Shah, R. Bishop, P. van der Bruggen, V. Nene, and E. L. Taracha. 2007. A novel strategy for the identification of antigens that are recognised by bovine MHC class I restricted cytotoxic T cells in a protozoan infection using reverse vaccinology. Immunome Res 3:2. Grakoui, A., N. H. Shoukry, D. J. Woollard, J. H. Han, H. L. Hanson, J. Ghrayeb, K. K. Murthy, C. M. Rice, and C. M. Walker. 2003. HCV persistence and immune evasion in the absence of memory T cell help. Science 302:659-62. Greco, D., S. Salmaso, P. Mastrantonio, M. Giuliano, A. E. Tozzi, A. Anemona, M. L. Ciofi degli Atti, A. Giammanco, P. Panei, W. C. Blackwelder, D. L. Klein, and S. G. Wassilak. 1996. A controlled trial of two acellular vaccines and one whole-cell vaccine against pertussis. Progetto Pertosse Working Group. N Engl J Med 334:341-8. Grgacic, E. V., and D. A. Anderson. 2006. Virus-like particles: passport to immune recognition. Methods 40:60-5. Guinovart, C., J. J. Aponte, J. Sacarlal, P. Aide, A. Leach, Q. Bassat, E. Macete, C. Dobano, M. Lievens, C. Loucq, W. R. Ballou, J. Cohen, and P. L. Alonso. 2009. Insights into long-lasting protection induced by RTS,S/AS02A malaria vaccine: further results from a phase IIb trial in Mozambican children. PLoS ONE 4:e5165. Gurunathan, S., D. M. Klinman, and R. A. Seder. 2000. DNA vaccines: immunology, application, and optimization*. Annu Rev Immunol 18:927-74. Hain, T., C. Steinweg, C. T. Kuenne, A. Billion, R. Ghai, S. S. Chatterjee, E. Domann, U. Karst, A. Goesmann, T. Bekel, D. Bartels, O. Kaiser, F. Meyer, A. 34 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. Puhler, B. Weisshaar, J. Wehland, C. Liang, T. Dandekar, R. Lampidis, J. Kreft, W. Goebel, and T. Chakraborty. 2006. Whole-genome sequence of Listeria welshimeri reveals common steps in genome reduction with Listeria innocua as compared to Listeria monocytogenes. J Bacteriol 188:7405-15. Harro, C. D., Y. Y. Pang, R. B. Roden, A. Hildesheim, Z. Wang, M. J. Reynolds, T. C. Mast, R. Robinson, B. R. Murphy, R. A. Karron, J. Dillner, J. T. Schiller, and D. R. Lowy. 2001. Safety and immunogenicity trial in adult volunteers of a human papillomavirus 16 L1 virus-like particle vaccine. J Natl Cancer Inst 93:284-92. Heeney, J. L. 2004. Requirement of diverse T-helper responses elicited by HIV vaccines: induction of highly targeted humoral and CTL responses. Expert Rev Vaccines 3:S53-64. Heppell, J., and H. L. Davis. 2000. Application of DNA vaccine technology to aquaculture. Adv Drug Deliv Rev 43:29-43. Higgins, D., J. D. Marshall, P. Traquina, G. Van Nest, and B. D. Livingston. 2007. Immunostimulatory DNA as a vaccine adjuvant. Expert Rev Vaccines 6:747-59. Hilleman, M. R. 1998. Six decades of vaccine development--a personal history. Nat Med 4:507-14. Hirao, L. A., L. Wu, A. S. Khan, A. Satishchandran, R. Draghia-Akli, and D. B. Weiner. 2008. Intradermal/subcutaneous immunization by electroporation improves plasmid vaccine delivery and potency in pigs and rhesus macaques. Vaccine 26:440-8. Hou, L., H. Wu, L. Xu, and F. Yang. 2009. Expression and self-assembly of virus-like particles of infectious hypodermal and hematopoietic necrosis virus in Escherichia coli. Arch Virol 154:547-53. Houghton, M., and S. Abrignani. 2005. Prospects for a vaccine against the hepatitis C virus. Nature 436:961-6. Iyer, A. V., B. Pahar, M. J. Boudreaux, N. Wakamatsu, A. F. Roy, V. N. Chouljenko, A. Baghian, C. Apetrei, P. A. Marx, and K. G. Kousoulas. 2009. Recombinant vesicular stomatitis virus-based west Nile vaccine elicits strong humoral and cellular immune responses and protects mice against lethal challenge with the virulent west Nile virus strain LSU-AR01. Vaccine 27:893-903. Jegerlehner, A., P. Maurer, J. Bessa, H. J. Hinton, M. Kopf, and M. F. Bachmann. 2007. TLR9 signaling in B cells determines class switch recombination to IgG2a. J Immunol 178:2415-20. Johnston, S. A., A. M. Talaat, and M. J. McGuire. 2002. Genetic immunization: what's in a name? Arch Med Res 33:325-9. Jones, D. H., S. Corris, S. McDonald, J. C. Clegg, and G. H. Farrar. 1997. Poly(DLlactide-co-glycolide)-encapsulated plasmid DNA elicits systemic and mucosal antibody responses to encoded protein after oral administration. Vaccine 15:814-7. Kaur, R., M. Rauthan, and S. Vrati. 2004. Immunogenicity in mice of a cationic microparticle-adsorbed plasmid DNA encoding Japanese encephalitis virus envelope protein. Vaccine 22:2776-82. Kusakabe, K., K. Q. Xin, H. Katoh, K. Sumino, E. Hagiwara, S. Kawamoto, K. Okuda, Y. Miyagi, I. Aoki, K. Nishioka, and D. Klinman. 2000. The timing of GMCSF expression plasmid administration influences the Th1/Th2 response induced by an HIV-1-specific DNA vaccine. J Immunol 164:3102-11. Kutzler, M. A., and D. B. Weiner. 2008. DNA vaccines: ready for prime time? Nat Rev Genet 9:776-88. 35 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. Lenz, P., P. M. Day, Y. Y. Pang, S. A. Frye, P. N. Jensen, D. R. Lowy, and J. T. Schiller. 2001. Papillomavirus-like particles induce acute activation of dendritic cells. J Immunol 166:5346-55. Li, P., R. B. Cao, Q. S. Zheng, J. J. Liu, Y. Li, E. X. Wang, F. Li, and P. Y. Chen. 2008. Enhancement of humoral and cellular immunity in mice against Japanese encephalitis virus using a DNA prime-protein boost vaccine strategy. Vet J. Lim, Y. S., B. Y. Kang, E. J. Kim, S. H. Kim, S. Y. Hwang, K. M. Kim, and T. S. Kim. 1998. Vaccination with an ovalbumin/interleukin-4 fusion DNA efficiently induces Th2 cell-mediated immune responses in an ovalbumin-specific manner. Arch Pharm Res 21:537-42. Liniger, M., A. Zuniga, T. N. Morin, B. Combardiere, R. Marty, M. Wiegand, O. Ilter, M. Knuchel, and H. Y. Naim. 2009. Recombinant measles viruses expressing single or multiple antigens of human immunodeficiency virus (HIV-1) induce cellular and humoral immune responses. Vaccine. Liniger, M., A. Zuniga, and H. Y. Naim. 2007. Use of viral vectors for the development of vaccines. Expert Rev Vaccines 6:255-66. Liu, M. A., and J. B. Ulmer. 2000. Gene-based vaccines. Mol Ther 1:497-500. Liu, T. H., J. Oscherwitz, B. Schnepp, J. Jacobs, F. Yu, K. B. Cease, and P. R. Johnson. 2009. Genetic vaccines for anthrax based on recombinant adeno-associated virus vectors. Mol Ther 17:373-9. Lorin, C., L. Mollet, F. Delebecque, C. Combredet, B. Hurtrel, P. Charneau, M. Brahic, and F. Tangy. 2004. A single injection of recombinant measles virus vaccines expressing human immunodeficiency virus (HIV) type 1 clade B envelope glycoproteins induces neutralizing antibodies and cellular immune responses to HIV. J Virol 78:14657. Ludwig, C., and R. Wagner. 2007. Virus-like particles-universal molecular toolboxes. Curr Opin Biotechnol 18:537-45. Lund, J., A. Sato, S. Akira, R. Medzhitov, and A. Iwasaki. 2003. Toll-like receptor 9mediated recognition of Herpes simplex virus-2 by plasmacytoid dendritic cells. J Exp Med 198:513-20. Mackova, J., J. Stasikova, L. Kutinova, J. Masin, P. Hainz, M. Simsova, P. Gabriel, P. Sebo, and S. Nemeckova. 2006. Prime/boost immunotherapy of HPV16-induced tumors with E7 protein delivered by Bordetella adenylate cyclase and modified vaccinia virus Ankara. Cancer Immunol Immunother 55:39-46. Mackowiak, M., J. Maki, L. Motes-Kreimeyer, T. Harbin, and K. Van Kampen. 1999. Vaccination of wildlife against rabies: successful use of a vectored vaccine obtained by recombinant technology. Adv Vet Med 41:571-83. Magagnoli, C., A. Bardotti, G. De Conciliis, R. Galasso, M. Tomei, C. Campa, C. Pennatini, M. Cerchioni, B. Fabbri, S. Giannini, G. L. Mattioli, A. Biolchi, S. D'Ascenzi, and F. Helling. 2009. Structural organization of NadADelta(351-405), a recombinant MenB vaccine component, by its physico-chemical characterization at drug substance level. Vaccine 27:2156-70. Maione, D., I. Margarit, C. D. Rinaudo, V. Masignani, M. Mora, M. Scarselli, H. Tettelin, C. Brettoni, E. T. Iacobini, R. Rosini, N. D'Agostino, L. Miorin, S. Buccato, M. Mariani, G. Galli, R. Nogarotto, V. Nardi Dei, F. Vegni, C. Fraser, G. Mancuso, G. Teti, L. C. Madoff, L. C. Paoletti, R. Rappuoli, D. L. Kasper, J. L. Telford, and 36 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. G. Grandi. 2005. Identification of a universal Group B streptococcus vaccine by multiple genome screen. Science 309:148-50. Martin, D. B., L. B. Weiner, P. I. Nieburg, and D. C. Blair. 1979. Atypical measles in adolescents and young adults. Ann Intern Med 90:877-81. Masic, A., L. A. Babiuk, and Y. Zhou. 2009. Reverse genetics-generated elastasedependent swine influenza viruses are attenuated in pigs. J Gen Virol 90:375-85. Mastrangeli, A., B. G. Harvey, J. Yao, G. Wolff, I. Kovesdi, R. G. Crystal, and E. Falck-Pedersen. 1996. "Sero-switch" adenovirus-mediated in vivo gene transfer: circumvention of anti-adenovirus humoral immune defenses against repeat adenovirus vector administration by changing the adenovirus serotype. Hum Gene Ther 7:79-87. Matsumoto, M., S. Kikkawa, M. Kohase, K. Miyake, and T. Seya. 2002. Establishment of a monoclonal antibody against human Toll-like receptor 3 that blocks double-stranded RNA-mediated signaling. Biochem Biophys Res Commun 293:1364-9. Maurer, P., G. T. Jennings, J. Willers, F. Rohner, Y. Lindman, K. Roubicek, W. A. Renner, P. Muller, and M. F. Bachmann. 2005. A therapeutic vaccine for nicotine dependence: preclinical efficacy, and Phase I safety and immunogenicity. Eur J Immunol 35:2031-40. McAleer, W. J., E. B. Buynak, R. Z. Maigetter, D. E. Wampler, W. J. Miller, and M. R. Hilleman. 1984. Human hepatitis B vaccine from recombinant yeast. Nature 307:17880. McKeever, U., S. Barman, T. Hao, P. Chambers, S. Song, L. Lunsford, Y. Y. Hsu, K. Roy, and M. L. Hedley. 2002. Protective immune responses elicited in mice by immunization with formulations of poly(lactide-co-glycolide) microparticles. Vaccine 20:1524-31. McMichael, A. J. 2006. HIV vaccines. Annu Rev Immunol 24:227-55. Mellquist-Riemenschneider, J. L., A. R. Garrison, J. B. Geisbert, K. U. Saikh, K. D. Heidebrink, P. B. Jahrling, R. G. Ulrich, and C. S. Schmaljohn. 2003. Comparison of the protective efficacy of DNA and baculovirus-derived protein vaccines for EBOLA virus in guinea pigs. Virus Res 92:187-93. Merdan, T., J. Kopecek, and T. Kissel. 2002. Prospects for cationic polymers in gene and oligonucleotide therapy against cancer. Adv Drug Deliv Rev 54:715-58. Mikszta, J. A., J. B. Alarcon, J. M. Brittingham, D. E. Sutter, R. J. Pettis, and N. G. Harvey. 2002. Improved genetic immunization via micromechanical disruption of skinbarrier function and targeted epidermal delivery. Nat Med 8:415-9. Miller, D. S., D. Boyle, F. Feng, G. Y. Reaiche, I. Kotlarski, R. Colonno, and A. R. Jilbert. 2008. Antiviral therapy with entecavir combined with post-exposure "primeboost" vaccination eliminates duck hepatitis B virus-infected hepatocytes and prevents the development of persistent infection. Virology 373:329-41. Minigo, G., A. Scholzen, C. K. Tang, J. C. Hanley, M. Kalkanidis, G. A. Pietersz, V. Apostolopoulos, and M. Plebanski. 2007. Poly-L-lysine-coated nanoparticles: a potent delivery system to enhance DNA vaccine efficacy. Vaccine 25:1316-27. Montigiani, S., F. Falugi, M. Scarselli, O. Finco, R. Petracca, G. Galli, M. Mariani, R. Manetti, M. Agnusdei, R. Cevenini, M. Donati, R. Nogarotto, N. Norais, I. Garaguso, S. Nuti, G. Saletti, D. Rosa, G. Ratti, and G. Grandi. 2002. Genomic approach for analysis of surface proteins in Chlamydia pneumoniae. Infect Immun 70:368-79. 37 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. Moran, T. P., J. E. Burgents, B. Long, I. Ferrer, E. M. Jaffee, R. M. Tisch, R. E. Johnston, and J. S. Serody. 2007. Alphaviral vector-transduced dendritic cells are successful therapeutic vaccines against neu-overexpressing tumors in wild-type mice. Vaccine 25:6604-12. Moran, T. P., M. Collier, K. P. McKinnon, N. L. Davis, R. E. Johnston, and J. S. Serody. 2005. A novel viral system for generating antigen-specific T cells. J Immunol 175:3431-8. Moron, G., P. Rueda, I. Casal, and C. Leclerc. 2002. CD8alpha- CD11b+ dendritic cells present exogenous virus-like particles to CD8+ T cells and subsequently express CD8alpha and CD205 molecules. J Exp Med 195:1233-45. Murata, K., M. Lechmann, M. Qiao, T. Gunji, H. J. Alter, and T. J. Liang. 2003. Immunization with hepatitis C virus-like particles protects mice from recombinant hepatitis C virus-vaccinia infection. Proc Natl Acad Sci U S A 100:6753-8. Murphy, T. V., P. M. Gargiullo, M. S. Massoudi, D. B. Nelson, A. O. Jumaan, C. A. Okoro, L. R. Zanardi, S. Setia, E. Fair, C. W. LeBaron, M. Wharton, and J. R. Livengood. 2001. Intussusception among infants given an oral rotavirus vaccine. N Engl J Med 344:564-72. Nardin, E. H., G. A. Oliveira, J. M. Calvo-Calle, K. Wetzel, C. Maier, A. J. Birkett, P. Sarpotdar, M. L. Corado, G. B. Thornton, and A. Schmidt. 2004. Phase I testing of a malaria vaccine composed of hepatitis B virus core particles expressing Plasmodium falciparum circumsporozoite epitopes. Infect Immun 72:6519-27. Nchinda, G., J. Kuroiwa, M. Oks, C. Trumpfheller, C. G. Park, Y. Huang, D. Hannaman, S. J. Schlesinger, O. Mizenina, M. C. Nussenzweig, K. Uberla, and R. M. Steinman. 2008. The efficacy of DNA vaccination is enhanced in mice by targeting the encoded protein to dendritic cells. J Clin Invest 118:1427-36. Newman, K. D., P. Elamanchili, G. S. Kwon, and J. Samuel. 2002. Uptake of poly(D,L-lactic-co-glycolic acid) microspheres by antigen-presenting cells in vivo. J Biomed Mater Res 60:480-6. O'Hagan, D. T., M. Singh, and J. B. Ulmer. 2006. Microparticle-based technologies for vaccines. Methods 40:10-9. Offit, P. A. 2005. The Cutter incident, 50 years later. N Engl J Med 352:1411-2. Oka, T., M. Yamamoto, K. Miyashita, S. Ogawa, K. Katayama, T. Wakita, and N. Takeda. 2009. Self-assembly of sapovirus recombinant virus-like particles from polyprotein in mammalian cells. Microbiol Immunol 53:49-52. Okada, M., Y. Kita, T. Nakajima, N. Kanamaru, S. Hashimoto, T. Nagasawa, Y. Kaneda, S. Yoshida, Y. Nishida, H. Nakatani, K. Takao, C. Kishigami, Y. Inoue, M. Matsumoto, D. N. McMurray, E. C. Dela Cruz, E. V. Tan, R. M. Abalos, J. A. Burgos, P. Saunderson, and M. Sakatani. 2009. Novel prophylactic and therapeutic vaccine against tuberculosis. Vaccine. Ostrowski, M., M. Vermeulen, O. Zabal, J. R. Geffner, A. M. Sadir, and O. J. Lopez. 2005. Impairment of thymus-dependent responses by murine dendritic cells infected with foot-and-mouth disease virus. J Immunol 175:3971-9. Otten, G. R., M. Schaefer, B. Doe, H. Liu, I. Srivastava, J. Megede, J. Kazzaz, Y. Lian, M. Singh, M. Ugozzoli, D. Montefiori, M. Lewis, D. A. Driver, T. Dubensky, J. M. Polo, J. Donnelly, D. T. O'Hagan, S. Barnett, and J. B. Ulmer. 2005. Enhanced potency of plasmid DNA microparticle human immunodeficiency virus vaccines in 38 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. rhesus macaques by using a priming-boosting regimen with recombinant proteins. J Virol 79:8189-200. Pai Kasturi, S., H. Qin, K. S. Thomson, S. El-Bereir, S. C. Cha, S. Neelapu, L. W. Kwak, and K. Roy. 2006. Prophylactic anti-tumor effects in a B cell lymphoma model with DNA vaccines delivered on polyethylenimine (PEI) functionalized PLGA microparticles. J Control Release 113:261-70. Pancholi, P., M. Perkus, N. Tricoche, Q. Liu, and A. M. Prince. 2003. DNA immunization with hepatitis C virus (HCV) polycistronic genes or immunization by HCV DNA priming-recombinant canarypox virus boosting induces immune responses and protection from recombinant HCV-vaccinia virus infection in HLA-A2.1-transgenic mice. J Virol 77:382-90. Pattenden, L. K., A. P. Middelberg, M. Niebert, and D. I. Lipin. 2005. Towards the preparative and large-scale precision manufacture of virus-like particles. Trends Biotechnol 23:523-9. Peachman, K. K., M. Rao, and C. R. Alving. 2003. Immunization with DNA through the skin. Methods 31:232-42. Perri, S., C. E. Greer, K. Thudium, B. Doe, H. Legg, H. Liu, R. E. Romero, Z. Tang, Q. Bin, T. W. Dubensky, Jr., M. Vajdy, G. R. Otten, and J. M. Polo. 2003. An alphavirus replicon particle chimera derived from venezuelan equine encephalitis and sindbis viruses is a potent gene-based vaccine delivery vector. J Virol 77:10394-403. Pizza, M., V. Scarlato, V. Masignani, M. M. Giuliani, B. Arico, M. Comanducci, G. T. Jennings, L. Baldi, E. Bartolini, B. Capecchi, C. L. Galeotti, E. Luzzi, R. Manetti, E. Marchetti, M. Mora, S. Nuti, G. Ratti, L. Santini, S. Savino, M. Scarselli, E. Storni, P. Zuo, M. Broeker, E. Hundt, B. Knapp, E. Blair, T. Mason, H. Tettelin, D. W. Hood, A. C. Jeffries, N. J. Saunders, D. M. Granoff, J. C. Venter, E. R. Moxon, G. Grandi, and R. Rappuoli. 2000. Identification of vaccine candidates against serogroup B meningococcus by whole-genome sequencing. Science 287:1816-20. Pontarollo, R. A., L. A. Babiuk, R. Hecker, and S. Van Drunen Littel-Van Den Hurk. 2002. Augmentation of cellular immune responses to bovine herpesvirus-1 glycoprotein D by vaccination with CpG-enhanced plasmid vectors. J Gen Virol 83:2973-81. Pumpens, P., and E. Grens. 2001. HBV core particles as a carrier for B cell/T cell epitopes. Intervirology 44:98-114. Pushko, P., T. M. Tumpey, F. Bu, J. Knell, R. Robinson, and G. Smith. 2005. Influenza virus-like particles comprised of the HA, NA, and M1 proteins of H9N2 influenza virus induce protective immune responses in BALB/c mice. Vaccine 23:57519. Querec, T., S. Bennouna, S. Alkan, Y. Laouar, K. Gorden, R. Flavell, S. Akira, R. Ahmed, and B. Pulendran. 2006. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. J Exp Med 203:413-24. Rahman, M. J., and C. Fernandez. 2009. Neonatal vaccination with Mycobacterium bovis BCG: potential effects as a priming agent shown in a heterologous prime-boost immunization protocol. Vaccine. Ramos, I., A. Alonso, J. M. Marcen, A. Peris, J. A. Castillo, M. Colmenares, and V. Larraga. 2008. Heterologous prime-boost vaccination with a non-replicative vaccinia 39 139. 140. 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. recombinant vector expressing LACK confers protection against canine visceral leishmaniasis with a predominant Th1-specific immune response. Vaccine 26:333-44. Randolph, G. J., K. Inaba, D. F. Robbiani, R. M. Steinman, and W. A. Muller. 1999. Differentiation of phagocytic monocytes into lymph node dendritic cells in vivo. Immunity 11:753-61. Randrianarison-Jewtoukoff, V., and M. Perricaudet. 1995. Recombinant adenoviruses as vaccines. Biologicals 23:145-57. Rao, S. S., D. Styles, W. Kong, C. Andrews, J. P. Gorres, and G. J. Nabel. 2009. A gene-based avian influenza vaccine in poultry. Poult Sci 88:860-6. Rappuoli, R. 2001. Reverse vaccinology, a genome-based approach to vaccine development. Vaccine 19:2688-91. Reiss, C. S., I. V. Plakhov, and T. Komatsu. 1998. Viral replication in olfactory receptor neurons and entry into the olfactory bulb and brain. Ann N Y Acad Sci 855:75161. Richer, M. J., D. J. Lavallee, I. Shanina, and M. S. Horwitz. 2009. Toll-like receptor 3 signaling on macrophages is required for survival following coxsackievirus B4 infection. PLoS ONE 4:e4127. Riezebos-Brilman, A., J. Regts, M. Chen, J. Wilschut, and T. Daemen. 2009. Augmentation of alphavirus vector-induced human papilloma virus-specific immune and anti-tumour responses by co-expression of interleukin-12. Vaccine 27:701-7. Roberts, A., L. Buonocore, R. Price, J. Forman, and J. K. Rose. 1999. Attenuated vesicular stomatitis viruses as vaccine vectors. J Virol 73:3723-32. Roberts, D. M., A. Nanda, M. J. Havenga, P. Abbink, D. M. Lynch, B. A. Ewald, J. Liu, A. R. Thorner, P. E. Swanson, D. A. Gorgone, M. A. Lifton, A. A. Lemckert, L. Holterman, B. Chen, A. Dilraj, A. Carville, K. G. Mansfield, J. Goudsmit, and D. H. Barouch. 2006. Hexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity. Nature 441:239-43. Roos, A. K., S. Moreno, C. Leder, M. Pavlenko, A. King, and P. Pisa. 2006. Enhancement of cellular immune response to a prostate cancer DNA vaccine by intradermal electroporation. Mol Ther 13:320-7. Rosati, M., A. Valentin, R. Jalah, V. Patel, A. von Gegerfelt, C. Bergamaschi, C. Alicea, D. Weiss, J. Treece, R. Pal, P. D. Markham, E. T. Marques, J. T. August, A. Khan, R. Draghia-Akli, B. K. Felber, and G. N. Pavlakis. 2008. Increased immune responses in rhesus macaques by DNA vaccination combined with electroporation. Vaccine 26:5223-9. Ross, B. C., L. Czajkowski, D. Hocking, M. Margetts, E. Webb, L. Rothel, M. Patterson, C. Agius, S. Camuglia, E. Reynolds, T. Littlejohn, B. Gaeta, A. Ng, E. S. Kuczek, J. S. Mattick, D. Gearing, and I. G. Barr. 2001. Identification of vaccine candidate antigens from a genomic analysis of Porphyromonas gingivalis. Vaccine 19:4135-42. Schadeck, E. B., M. Sidhu, M. A. Egan, S. Y. Chong, P. Piacente, A. Masood, D. Garcia-Hand, S. Cappello, V. Roopchand, S. Megati, J. Quiroz, J. D. Boyer, B. K. Felber, G. N. Pavlakis, D. B. Weiner, J. H. Eldridge, and Z. R. Israel. 2006. A dose sparing effect by plasmid encoded IL-12 adjuvant on a SIVgag-plasmid DNA vaccine in rhesus macaques. Vaccine 24:4677-87. 40 152. 153. 154. 155. 156. 157. 158. 159. 160. 161. 162. 163. 164. Schlereth, B., L. Buonocore, A. Tietz, V. Meulen Vt, J. K. Rose, and S. Niewiesk. 2003. Successful mucosal immunization of cotton rats in the presence of measles virusspecific antibodies depends on degree of attenuation of vaccine vector and virus dose. J Gen Virol 84:2145-51. Schulte, R., Y. S. Suh, U. Sauermann, W. Ochieng, S. Sopper, K. S. Kim, S. S. Ahn, K. S. Park, N. Stolte-Leeb, G. Hunsmann, Y. C. Sung, and C. Stahl-Hennig. 2009. Mucosal prior to systemic application of recombinant adenovirus boosting is more immunogenic than systemic application twice but confers similar protection against SIVchallenge in DNA vaccine-primed macaques. Virology 383:300-9. Schulz, O., S. S. Diebold, M. Chen, T. I. Naslund, M. A. Nolte, L. Alexopoulou, Y. T. Azuma, R. A. Flavell, P. Liljestrom, and C. Reis e Sousa. 2005. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature 433:887-92. Schwarz, K., E. Meijerink, D. E. Speiser, A. C. Tissot, I. Cielens, R. Renhof, A. Dishlers, P. Pumpens, and M. F. Bachmann. 2005. Efficient homologous prime-boost strategies for T cell vaccination based on virus-like particles. Eur J Immunol 35:816-21. Serruto, D., J. Adu-Bobie, B. Capecchi, R. Rappuoli, M. Pizza, and V. Masignani. 2004. Biotechnology and vaccines: application of functional genomics to Neisseria meningitidis and other bacterial pathogens. J Biotechnol 113:15-32. Serruto, D., L. Serino, V. Masignani, and M. Pizza. 2009. Genome-based approaches to develop vaccines against bacterial pathogens. Vaccine. Sheets, R. L., J. Stein, T. S. Manetz, C. Duffy, M. Nason, C. Andrews, W. P. Kong, G. J. Nabel, and P. L. Gomez. 2006. Biodistribution of DNA plasmid vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile virus is similar, without integration, despite differing plasmid backbones or gene inserts. Toxicol Sci 91:610-9. Sin, J., J. J. Kim, C. Pachuk, C. Satishchandran, and D. B. Weiner. 2000. DNA vaccines encoding interleukin-8 and RANTES enhance antigen-specific Th1-type CD4(+) T-cell-mediated protective immunity against herpes simplex virus type 2 in vivo. J Virol 74:11173-80. Soleimanjahi, H., M. H. Roostaee, M. J. Rasaee, F. Mahboudi, A. Kazemnejad, T. Bamdad, and K. Zandi. 2006. The effect of DNA priming-protein boosting on enhancing humoral immunity and protecting mice against lethal HSV infections. FEMS Immunol Med Microbiol 46:100-6. Somboonthum, P., H. Yoshii, S. Okamoto, M. Koike, Y. Gomi, Y. Uchiyama, M. Takahashi, K. Yamanishi, and Y. Mori. 2007. Generation of a recombinant Oka varicella vaccine expressing mumps virus hemagglutinin-neuraminidase protein as a polyvalent live vaccine. Vaccine 25:8741-55. Steinhoff, U., U. Muller, A. Schertler, H. Hengartner, M. Aguet, and R. M. Zinkernagel. 1995. Antiviral protection by vesicular stomatitis virus-specific antibodies in alpha/beta interferon receptor-deficient mice. J Virol 69:2153-8. Stolte-Leeb, N., K. Bieler, J. Kostler, J. Heeney, P. T. Haaft, Y. S. Suh, G. Hunsmann, C. Stahl-Hennig, and R. Wagner. 2008. Better protective effects in rhesus macaques by combining systemic and mucosal application of a dual component vector vaccine after rectal SHIV89.6P challenge compared to systemic vaccination alone. Viral Immunol 21:235-46. Storni, T., C. Ruedl, K. Schwarz, R. A. Schwendener, W. A. Renner, and M. F. Bachmann. 2004. Nonmethylated CG motifs packaged into virus-like particles induce 41 165. 166. 167. 168. 169. 170. 171. 172. 173. 174. 175. 176. 177. protective cytotoxic T cell responses in the absence of systemic side effects. J Immunol 172:1777-85. Takamura, S., M. Niikura, T. C. Li, N. Takeda, S. Kusagawa, Y. Takebe, T. Miyamura, and Y. Yasutomi. 2004. DNA vaccine-encapsulated virus-like particles derived from an orally transmissible virus stimulate mucosal and systemic immune responses by oral administration. Gene Ther 11:628-35. Takeda, K., and S. Akira. 2007. Toll-like receptors. Curr Protoc Immunol Chapter 14:Unit 14 12. Takeda, M., K. Takeuchi, N. Miyajima, F. Kobune, Y. Ami, N. Nagata, Y. Suzaki, Y. Nagai, and M. Tashiro. 2000. Recovery of pathogenic measles virus from cloned cDNA. J Virol 74:6643-7. Tan, B., H. Wang, L. Shang, and T. Yang. 2009. Coadministration of chicken GM-CSF with a DNA vaccine expressing infectious bronchitis virus (IBV) S1 glycoprotein enhances the specific immune response and protects against IBV infection. Arch Virol 154:1117-24. Tang, D. C., M. DeVit, and S. A. Johnston. 1992. Genetic immunization is a simple method for eliciting an immune response. Nature 356:152-4. Tartz, S., H. Russmann, J. Kamanova, P. Sebo, A. Sturm, V. Heussler, B. Fleischer, and T. Jacobs. 2008. Complete protection against P. berghei malaria upon heterologous prime/boost immunization against circumsporozoite protein employing Salmonella type III secretion system and Bordetella adenylate cyclase toxoid. Vaccine 26:5935-43. Thacker, E. L., D. J. Holtkamp, A. S. Khan, P. A. Brown, and R. Draghia-Akli. 2006. Plasmid-mediated growth hormone-releasing hormone efficacy in reducing disease associated with Mycoplasma hyopneumoniae and porcine reproductive and respiratory syndrome virus infection. J Anim Sci 84:733-42. Tissot, A. C., P. Maurer, J. Nussberger, R. Sabat, T. Pfister, S. Ignatenko, H. D. Volk, H. Stocker, P. Muller, G. T. Jennings, F. Wagner, and M. F. Bachmann. 2008. Effect of immunisation against angiotensin II with CYT006-AngQb on ambulatory blood pressure: a double-blind, randomised, placebo-controlled phase IIa study. Lancet 371:821-7. Touze, A., and P. Coursaget. 1998. In vitro gene transfer using human papillomaviruslike particles. Nucleic Acids Res 26:1317-23. Trevor, K. T., E. M. Hersh, J. Brailey, J. M. Balloul, and B. Acres. 2001. Transduction of human dendritic cells with a recombinant modified vaccinia Ankara virus encoding MUC1 and IL-2. Cancer Immunol Immunother 50:397-407. Triantafilou, K., G. Orthopoulos, E. Vakakis, M. A. Ahmed, D. T. Golenbock, P. M. Lepper, and M. Triantafilou. 2005. Human cardiac inflammatory responses triggered by Coxsackie B viruses are mainly Toll-like receptor (TLR) 8-dependent. Cell Microbiol 7:1117-26. van Drunen Littel-van den Hurk, S., S. L. Babiuk, and L. A. Babiuk. 2004. Strategies for improved formulation and delivery of DNA vaccines to veterinary target species. Immunol Rev 199:113-25. van Drunen Littel-van den Hurk, S., V. Gerdts, B. I. Loehr, R. Pontarollo, R. Rankin, R. Uwiera, and L. A. Babiuk. 2000. Recent advances in the use of DNA vaccines for the treatment of diseases of farmed animals. Adv Drug Deliv Rev 43:13-28. 42 178. 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. Wahren, B., and M. Brytting. 1997. DNA increases the potency of vaccination against infectious diseases. Curr Opin Chem Biol 1:183-9. Walter, E., D. Dreher, M. Kok, L. Thiele, S. G. Kiama, P. Gehr, and H. P. Merkle. 2001. Hydrophilic poly(DL-lactide-co-glycolide) microspheres for the delivery of DNA to human-derived macrophages and dendritic cells. J Control Release 76:149-68. Wang, D., M. E. Christopher, L. P. Nagata, M. A. Zabielski, H. Li, J. P. Wong, and J. Samuel. 2004. Intranasal immunization with liposome-encapsulated plasmid DNA encoding influenza virus hemagglutinin elicits mucosal, cellular and humoral immune responses. J Clin Virol 31 Suppl 1:S99-106. Wang, F., X. W. He, L. Jiang, D. Ren, Y. He, D. A. Li, and S. H. Sun. 2006. Enhanced immunogenicity of microencapsulated multiepitope DNA vaccine encoding T and B cell epitopes of foot-and-mouth disease virus in mice. Vaccine 24:2017-26. Wang, J., J. H. Hu, F. Q. Li, G. Z. Liu, Q. G. Zhu, J. Y. Liu, H. J. Ma, C. Peng, and F. G. Si. 2007. Strong cellular and humoral immune responses induced by transcutaneous immunization with HBsAg DNA-cationic deformable liposome complex. Exp Dermatol 16:724-9. Wang, J., L. Thorson, R. W. Stokes, M. Santosuosso, K. Huygen, A. Zganiacz, M. Hitt, and Z. Xing. 2004. Single mucosal, but not parenteral, immunization with recombinant adenoviral-based vaccine provides potent protection from pulmonary tuberculosis. J Immunol 173:6357-65. Wizemann, T. M., J. H. Heinrichs, J. E. Adamou, A. L. Erwin, C. Kunsch, G. H. Choi, S. C. Barash, C. A. Rosen, H. R. Masure, E. Tuomanen, A. Gayle, Y. A. Brewah, W. Walsh, P. Barren, R. Lathigra, M. Hanson, S. Langermann, S. Johnson, and S. Koenig. 2001. Use of a whole genome approach to identify vaccine molecules affording protection against Streptococcus pneumoniae infection. Infect Immun 69:15938. Woodland, D. L. 2004. Jump-starting the immune system: prime-boosting comes of age. Trends Immunol 25:98-104. Wu, K., G. N. Kim, and C. Y. Kang. 2009. Expression and processing of human immunodeficiency virus type 1 gp160 using the vesicular stomatitis virus New Jersey serotype vector system. J Gen Virol 90:1135-40. Yang, R., C. M. Wheeler, X. Chen, S. Uematsu, K. Takeda, S. Akira, D. V. Pastrana, R. P. Viscidi, and R. B. Roden. 2005. Papillomavirus capsid mutation to escape dendritic cell-dependent innate immunity in cervical cancer. J Virol 79:6741-50. Yu, S., X. Feng, T. Shu, T. Matano, M. Hasegawa, X. Wang, H. Ma, H. Li, Z. Li, and Y. Zeng. 2008. Potent specific immune responses induced by prime-boost-boost strategies based on DNA, adenovirus, and Sendai virus vectors expressing gag gene of Chinese HIV-1 subtype B. Vaccine 26:6124-31. Zakhartchouk, A. N., C. Pyne, G. K. Mutwiri, Z. Papp, M. E. Baca-Estrada, P. Griebel, L. A. Babiuk, and S. K. Tikoo. 1999. Mucosal immunization of calves with recombinant bovine adenovirus-3: induction of protective immunity to bovine herpesvirus-1. J Gen Virol 80 ( Pt 5):1263-9. Zamora, E., A. Handisurya, S. Shafti-Keramat, D. Borchelt, G. Rudow, K. Conant, C. Cox, J. C. Troncoso, and R. Kirnbauer. 2006. Papillomavirus-like particles are an effective platform for amyloid-beta immunization in rabbits and transgenic mice. J Immunol 177:2662-70. 43 191. 192. Zhang, H., R. Fayad, X. Wang, D. Quinn, and L. Qiao. 2004. Human immunodeficiency virus type 1 gag-specific mucosal immunity after oral immunization with papillomavirus pseudoviruses encoding gag. J Virol 78:10249-57. Zhang, S., R. Cubas, M. Li, C. Chen, and Q. Yao. 2009. Virus-like particle vaccine activates conventional B2 cells and promotes B cell differentiation to IgG2a producing plasma cells. Mol Immunol 46:1988-2001. 44