141
Views
6
CrossRef citations to date
0
Altmetric
Review

Current progress in the development of a prophylactic vaccine for HIV-1

&
Pages 9-26 | Published online: 22 Dec 2010

Abstract

Since its discovery and characterization in the early 1980s as a virus that attacks the immune system, there has been some success for the treatment of human immunodeficiency virus-1 (HIV-1) infection. However, due to the overwhelming public health impact of this virus, a vaccine is needed urgently. Despite the tireless efforts of scientist and clinicians, there is still no safe and effective vaccine that provides sterilizing immunity. A vaccine that provides sterilizing immunity against HIV infection remains elusive in part due to the following reasons: 1) degree of diversity of the virus, 2) ability of the virus to evade the hosts’ immunity, and 3) lack of appropriate animal models in which to test vaccine candidates. There have been several attempts to stimulate the immune system to provide protection against HIV-infection. Here, we will discuss attempts that have been made to induce sterilizing immunity, including traditional vaccination attempts, induction of broadly neutralizing antibody production, DNA vaccines, and use of viral vectors. Some of these attempts show promise pending continued research efforts.

Introduction

Since its discovery and characterization in the early 1980s as a virus that attacks the immune system, leaving patients unable to fight off opportunistic infections, there has been an ebb and flow of effective treatments and hope as scientists continue to search for ways to eradicate human immunodeficiency virus-1 (HIV-1) from the human population similar to what has been accomplished in the case of smallpox. The majority of the effort and nearly all of the success has come in the area of patient treatment rather than inhibition of contraction or spread of the virus. A class of treatments, antiretroviral therapies (ARTs) and later highly active antiretroviral therapies (HAARTs), has been the mainstay of disease control during the last 15 years. Notwithstanding the increased life span of patients, increased time to full-blown AIDS, and decreased contraction of opportunistic infections and AIDS-related diseases (ie, non-Hodgkin’s lymphoma, Kaposi’s sarcoma, etc) by patients treated with HAART, there are several reasons why development of an HIV-1 vaccine is still warranted. Five of these reasons are as follows: 1) nearly two-thirds of the patients who contract HIV-1 live in underdeveloped countries and cannot afford the expensive HAART regimen,Citation1 2) both the ART and HAART regimen are complex and are disruptive to patients’ lives and diets, making long-term compliance an issue,Citation2 3) the potential side effects of ART/HAART treatments negatively affect the long-term health of patients and include diabetes, cardiovascular disease, fractures, etc,Citation3Citation5 4) development of HAART drug resistance, and 5) the presence of latent HIV-1 reservoirs harboring viral strains that were produced through mutation throughout the duration of the infection of the host also play a role in the failure of HAART.Citation6 These reasons, as well as many others, underscore the need for a prophylactic HIV-1 vaccine.

Possibly, the strongest argument for development of a prophylactic vaccine may be the need for control of the virus spread worldwide. Every day, 7500 patients worldwide are infected with HIV-1.Citation1 Production of a vaccine that could inhibit infection, reduce spread, or both would aid in the reduction of the burden of AIDS and AIDS-related diseases. The expenses incurred by the AIDS epidemic can hardly be calculated. They range from tens of thousands of dollars per patient for the HAART regimen, to millions of dollars required for building of orphanages by governments for children whose parents have succumbed to the disease, to the unknown cost of educational materials and condoms in the effort to prevent further spread of the disease. This public health challenge has not gone unnoticed and has been addressed by scientists’ ongoing efforts to develop a safe and effective HIV-1 vaccine.

Prophylactic vs therapeutic vaccines

A prophylactic HIV-1 vaccine would offer sterilizing immunity to patients, preventing infection upon presentation of the virus. A prophylactic vaccine must also be effective at all possible portals of HIV-1 entry, especially the mucosa.Citation7 For this to occur, the vaccine must offer broad and durable immunity. Several consortia have worked diligently to produce a vaccine that will induce broadly reactive neutralizing antibodies (Nabs). These consortia include major international efforts as well as efforts of individual countries, regions, and institutions including, but not limited to: the International AIDS Vaccine Initiative Neutralizing Antibody Consortium,Citation8 the Center for HIV-AIDS Vaccine Immunology, the HIV Vaccine Trials Network, US Military HIV Research Program, the Collaboration for AIDS Vaccine Discovery, and the Vaccine Research Center at the National Institutes of Allergy and Infectious Diseases of the National Institutes of Health. To date, however, no HIV-1 candidate vaccine has induced broadly reactive Nabs.Citation8

In the absence of a vaccine that can prevent infection of HIV-1, there are still many benefits to be realized from production of a therapeutic vaccine. A therapeutic vaccine would be supremely valuable if it were able to increase the titer of virus necessary for infection, increase the time to clinical manifestation of virus, control viral load after infection, and reduce secondary transmission.Citation9Citation13 A vaccine that could induce this type of response would invariably decrease contagiousness, decrease the need for costly and potentially dangerous ART/HAART, and decrease the number of opportunistic infections of patients.

While the effect of controlling the normal HIV-1 pathology with therapeutic vaccines will be favorable for the individual patient as well as society, the effect of preventing HIV-1 infections in humans with a prophylactic vaccine is also broadly appealing. This potential for eradicating the HIV-1 virus from human hosts drives scientists to continue to find ways to circumvent the challenges presented by this unique virus in order to induce production of the Nabs that are critical for sterilizing immunity. This review, therefore, will focus on the specific challenges presented by HIV-1 and strides that have been made toward creating a prophylactic vaccine, including past efforts that have failed and lessons that have been learned from those failures. We will also discuss novel vaccine options and some of the promising trials that are currently underway.

Current challenges to creating an HIV-1 vaccine

While several problems face scientists who are attempting to create an HIV-1 vaccine, three problems in particular have posed extremely daunting challenges. These three problems are 1) degree of diversity of the virus, 2) ability of the virus to evade the hosts’ immunity, and 3) lack of appropriate animal models in which to test vaccine candidates. These three major problems will be discussed in more detail below.

Degree of diversity

Traditionally, prophylactic vaccines have been made by exposing some part of a pathogen’s structure as an antigen to the host’s immune system, and eliciting an immune response, resulting in the production of long-term memory lymphocytes that are capable of mounting a strong immune response upon later infection with the pathogen. The premise upon which this manipulation of the immune system is based is the ability of the immune system to make long-lasting antibodies to conserved structures on exposed proteins that are native to the pathogen. Ideally, both humoral and cell-mediated immunity would be induced creating long-lasting immunity. Traditional attempts to recreate this process using live attenuated simian immunodeficiency virus-1 (SIV-1) viruses in an effort to vaccinate macaques against SIV-1 have been proven safe and effective in macaques that were subsequently challenged with SIV-1.Citation14,Citation15 However, an incidental study of the effect of live-attenuated HIV-1 (containing deletions of the nef gene and the long terminal repeat) was proven pathogenic in humans when three out of six treated patients developed late-onset immunosuppression.Citation16Citation18 Killed viruses have also been tested as a potential vaccine approach, but safety concerns have halted their use. These safety concerns include incomplete inactivation of the virus leading to potential residual infectivity during the vaccine preparation.Citation19 Due to the ineffectiveness of traditional vaccine approaches to date, scientists have attempted to use recombinant HIV-1 proteins to stimulate the production of Nabs. These attempts failed due to their inability to induce a lasting, broad range of Nabs that would inhibit infection in humans.Citation20Citation23 Perhaps these failures are a result of the inherent diversity of HIV-1. This diversity has presented a major roadblock to development of a prophylactic vaccine. There are three main groups of HIV-1 (M, O, and N)Citation24 as well as a recently discovered group, P.Citation25 Each group consists of several subtypes, clades. The various clades display biological differences with respect to transmission,Citation26 replication,Citation27 and disease progression.Citation28,Citation29 These differences result in an inability to produce a generalizable vaccine that would induce the breadth of Nabs necessary to counter an infection by a wide range of HIV-1 clades that may be encountered in a natural setting.Citation30 The degree of diversity seen in HIV-1 is greater than that of any other virus observed.Citation31,Citation32 This problem is being addressed by development of multiclade (multiple env and/or subtype B gag, pol, nef)Citation33,Citation34 and mosaic vaccines which incorporate sets of 10 immunogenic proteins from 4 different clades or bivalent proteins from clades B and C.Citation35Citation37 There are proof of principle studies that illustrate immunological protection against HIV-1 in nonhuman primates that were passively treated with broadly reactive Nabs.Citation38Citation40 These studies show that protection against infection with HIV-1 can be conferred by the presence of broadly reactive Nabs. The next step toward production of a prophylactic vaccine would involve induction of production of these or similar broadly reactive Nabs by the host’s immune system.

Immune evasion

The rate at which the HIV-1 virus mutates, due to the nature of the reverse transcriptase enzyme responsible for transcribing its RNA, ensures that nearly every daughter virion will have a different genome than its parent.Citation41 When these changes occur in the HIV-1 Env protein that is needed for antibody recognition, they inhibit the immune system’s ability to mount a sufficient response. One attempt to circumvent this problem has been to induce the production of Nabs to the conserved regions of HIV-1 proteins. A major problem with this approach is that the conserved regions of HIV-1 proteins are often shielded from exposure to Nabs within the HIV-1 envelope. The native structure of the envelope protein, reportedly the only HIV-1 protein susceptible to Nabs,Citation31 shields it from the immune system as a glycosylated trimer of heterodimers. The glycosylation of the envelope protein allows for the carbohydrates to masquerade as ‘self ’ thereby forming an immunologically silent face and protects neighboring epitopes via an ‘evolving glycan shield’.Citation42Citation44 Additionally, the gp41 coreceptor binding site, another conserved site, is not presented until primary binding to CD4+ has occurred.Citation45 An attempt to create antibodies to the CD4-binding region of the gp120 protein was made in rhesus macaques in 2007 and results indicated that vaccinated hosts were able to withstand challenge with SHIV.Citation46 Other attempts to create an HIV-1 vaccine have focused on overcoming the ability of HIV-1 to escape immune surveillance through use of antibodies that are able to neutralize diverse isolates of HIV-1. These antibodies include PG9, PG16,Citation47 2F5, 2G12, 4E10, b12,Citation48Citation51 and most recently sCD4-17bCitation52 and others.Citation53 Identification of these antibodies gives hope that their induction or the induction of other such broadly reactive Nabs may provide the basis for a prophylactic vaccine in the future.

Lack of appropriate animal models

The use of animal models for development of therapeutics offers the benefit of thorough testing and validation prior to introduction of a vaccine in humans. In the past, vaccines were made by observing and then mimicking the immune response mounted by individuals who had recovered from a particular disease. To date, however, there are no known cases of individuals who have recovered from HIV-1 infection. However, data can be gathered from long-term nonprogressors – patients who have been infected with HIV-1 for at least 7 years and do not display any HIV-1-related symptoms.Citation54,Citation55 Another option that may be critical to the development of a prophylactic vaccine is the use of relevant animal models. Such models will allow for analysis of the effect of a potential vaccine on an intact host prior to use in humans.

One particular challenge with the use of animal models for development of a prophylactic HIV-1 vaccine is that there are very few naturally occurring disease models of HIV-1. Only a few nonhuman primates are susceptible to infection with HIV-1 and infected animals do not progress to AIDS.Citation56 Therefore, it is important to use other disease models that mimic the HIV-AIDS pathologic progression.Citation57 One such potential model is feline immunodeficiency virus (FIV). FIV was discovered in 1986 and is known to cause an AIDS-like disease in domestic cats and mimics HIV-related dementia in humans.Citation58 A vaccine for FIV was approved by the FDA in 2002.Citation59 While the FIV model is potentially informative, its use is not sufficient as a basis for development of a prophylactic HIV-1 vaccine.

An ideal animal model would display a pathological response to infection with HIV-1 that is very similar to the one that occurs in humans. Unfortunately, HIV-1 does not cause pathology leading to the development of AIDS in any host other than humans.Citation60Citation63 However, animal models have been developed and used that allow partial understanding of the pathology of HIV-1, the natural immunological response to infection, and the response of the host to novel therapeutics. One of these models involves the simian immunodeficiency virusMAC (SIVMAC) that replicates and causes an AIDS-like disease in baboons, cynomolgus, and pigtailed macaques. While the similarities of SIVMAC to HIV-1 have allowed for insight into pathology, transmission, and immunological response of the infected host to the virus, the differences between SIVMAC and HIV-1 are still too great to be able to draw conclusions regarding potential human responses to an HIV-1 prophylactic vaccine.Citation63 Therefore, to broaden the scope of animal model usage, a chimeric SHIV virus was engineered to incorporate both SIV and HIV-1 proteins or genes.Citation64 While macaques infected with SHIV do go on to develop AIDS, the time to progression is much different from the time to progression to AIDS of HIV-1-infected humans. Infection of macaques with SIVmac251 strain mimics HIV-1 infection in humans by leading to chronic, slow disease progression. Route and dose required for infection, viral tropism, replicative capacity of the viruses, and pathology of SIV/SHIV-infected monkeys are all very different than these parameters in humans.Citation65,Citation66 This distinction has been well characterized by the recent Phase IIb STEP trial, which involved 3000 healthy, uninfected volunteers. The result of this trial was termination at its first scheduled efficacy assessment due to its failure to suppress viral load in subsequently infected individuals and then-suspected increased HIV-1 infection due to interaction of the immune system with vaccine components.Citation67 The vaccine, a recombinant adenovirus serotype 5 (Ad5) virus incorporating the gag, pol, and nef genes from HIV-1, had been previously tested in an SHIV model in macaques and the results of that experiment were not suggestive of the results of the human trial.Citation68

This disparity underscores the need for animal models that more closely reflect the pathology seen in human infection with HIV-1 as well as identification of immunological correlates of protection that reflect control of HIV-1 viral load in human subjects. Therefore, the search for an appropriate animal model or the appropriate use of current animal models in the search for a prophylactic HIV-1 vaccine continues. Until a model can be derived that will allow for observation of each stage of infection, progression of disease, and response of the immune system in a way that is comparable to this process in humans, we will not be able to logically predict which vaccine candidates should be moved forward to clinical trials.

Several attempts to stimulate the immune system to provide protection against HIV infection have been attempted so far (). Hope for creating a prophylactic vaccine lies in the ability of the scientific community to identify and induce a broad neutralizing antibody response that would offer sterilizing immunity to vaccinated patients. To this end, several novel approaches are being studied.

Table 1 Historical vaccine attempts to inhibit HIV-1 infection

Novel vaccine options

As mentioned in the previous section, there are several daunting problems facing scientists who are attempting to create an HIV-1 vaccine. In hopes of creating a vaccine which elicits sterilizing immunity to HIV-1, researchers have focused their efforts on (1) the use of plasmid DNA vaccines, (2) live recombinant vectors for vaccine development (expressing or presenting HIV antigens), and (3) mucosal immunity. These critical topics will be discussed in more detail below.

Plasmid DNA vaccines

Vaccines should elicit a robust immune response that is long lasting and is able to provide protection against various strains of a pathogen. Plasmid DNA vaccinations can induce a strong humoral and T-cell response. DNA-based vaccination has been used as a powerful tool to fight against parasitic, fungal, bacterial, and viral infections.Citation115Citation119 There are multiple advantages for using plasmid DNA for vaccination: they are generally safe, nontoxic, and through the delivery of a gene encoding important immunogenic epitopes, the DNAbased vaccine exploits biosynthetic machinery of the host cell. One such example was in 1990, whereby Wolff and colleagues illustrated protein expression after intramuscular (IM) injection of plasmid DNA into myocytes.Citation120 Despite these promising results, there had been speculation regarding DNA vaccination strategies. For example, it was shown that protein production in response to DNA plasmids that contained HIV inserts elicited substantial cellular response in mice and nonhuman primates. However, these products were poorly immunogenic in humans.

One strategy to improve immune response of the plasmid DNA vaccine strategy is by coadministration of DNA plasmids coding for cytokines (eg, INF-g, IL-2, IL-12, IL-18, and IL-15).Citation121Citation124 A second strategy which has been utilized to improve plasmid DNA vaccination has been the administration of plasmid DNA with adjuvants (eg, CpG oligodeoxynucleotides), or the use of DNA-delivery systems (eg, microparticles, cochleates, and linear polyenimines).Citation125Citation128 A third strategy to improve vaccine efficacy involves the coadministration of plasmid DNA in combination with viral vectors. For instance, research performed by Harari and colleagues in 2008 demonstrated that vaccination by means of an HIV-1 clade C DNA prime in combination with a pox vector (NYVAC) boost induces a reliable polyfunctional and long-lasting anti-HIV T-cell response in human participants.Citation129 Along these same lines, work recently published by Jaoko and group demonstrated safety and immunogenicity of a multiclade HIV-1 Ad-based vaccine alone or in combination with a multiclade HIV-1 DNA vaccine in Africa. These results also demonstrated that DNA priming increased the frequency and magnitude of cellular and humoral responses; however, there was no effect of recombinant Ad5 dosage on immunogenicity endpoints.Citation130

The previously mentioned DNA-delivery strategies have been used in combination with viral vectors or alone by means of a variety of immunization routes (eg, IM, intravenous [IV], intradermal [ID], intranasal [IN], oral, rectal, or vaginal). In the majority of reported studies, DNA vaccines have been administered by the IM and/or ID routes. However, as it relates to HIV vaccination, mucosal immunity could potentially be an important factor to consider, with mucosal immunity being achieved optimally by IN or oral routes of administration. The topic of mucosal immunity will be discussed in more detail in a later section within this review.

After immunization, it is assumed that the DNA vaccination immunogen is produced in the skeletal muscles, dendritic cells, and macrophages at the site of immunization. However, in adults, the skeletal muscles are not involved in a high level of protein synthesis as compared to the liver. Therefore, the delivery of DNA to cells, which are capable of high protein synthesis, such as hepatocytes, epithelia cells of the intestines, or salivary pancreas, may result in high levels of protein expression. The hepatocytes express enzymes involved in the formation of intrachain and interchain disulfide bonds required for proper folding and assembly of proteins. In addition, the liver expresses glycosyltranferases, which are essential for synthesis of both N- and O-linked glycan side chains; this may not be the case for other cell types,Citation131,Citation132 the significance of this point being the fact that broadly crossclade Nabs such as 2G12 recognize glycan moieties on the heavily glycosylated HIV-1 envelope antigens.Citation44,Citation133,Citation134 Another advantage of protein expression within the liver is that significantly lower amounts of DNA are needed for protein expression of a particular antigen in the hepatocytes vs another cell type. For the immunization of humans, milligram quantities of DNA are necessary to achieve adequate levels of immune response.Citation119 Any method whereby there would be a reduction in DNA quantity needed to vaccinate humans would provide significant economic advantages. Based on the previously mentioned reasons, it is not a surprise that the liver has been exploited extensively as a site for gene delivery due to its ability to produce proteins and glycoproteins.Citation135Citation138

Hydrodynamic delivery is the application of controlled hydrodynamic pressure in capillaries to enhance endothelial and parenchymal cell permeability; this methodology had its inception in the late 1990s with investigations into intravascular injection of plasmid DNA solution for gene delivery in whole animals.Citation139Citation142 Hydrodynamic plasmid DNA delivery is well tolerated in mice. In 2008, Raska and colleagues demonstrated in mice that IV hydrodynamic vaccination with HIV-1 envelope DNA injections resulted in high levels of expression of HIV antigen in the liver. In mice, immunological data illustrated that hydrodynamic administration of HIV-1 plasmid DNA was superior to vaccination with DNA by IN, ID, IM, and intrasplenic routes. Further results illustrated that after boosting, hydrodynamic vaccination yielded levels of HIV-1-specific antibodies that were 40-fold higher than those elicited by other routes tested.Citation132

However, this delivery scheme is not feasible in large animals and humans. As an alternative, receptor-mediated DNA binding to hepatocytes could be a viable approach. Molecules with terminal galactose residues covalently linked to DNA are recognized by the hepatocyte-expressed galacto-sespecific asialoglycoproteinCitation143 receptor for internalization.Citation144 This alternative would avoid delivery through the hepatic system and the need for expansion of the blood volume. In addition, galactose-linked DNA packaged in delivery vehicles such as liposomes, choleates, or microspheres can be given by oral administration, which would be absorbed by the intestine and ultimately delivered to the hepatic vein. As an additional alternative to hydrodynamic delivery in humans, it might be possible to express HIV antigens in the liver by means of plasmid DNA delivery via viral vectors such as the Ad. Ads have been shown to transduce the liver efficiently in vivo by means of the hexon proteins.Citation145,Citation146 In this regard, production of translation of HIV-1 proteins primarily in the liver might allow for the production of heavily glycosylated HIV-1 envelope antigens and thus the production of Nabs.

Live recombinant vectors for vaccine development

Viral vectors are potent inducers of cellular and humoral response. Viral vectors can express proteins from bacteria or viral pathogens to vaccinate against infectious diseases. There are several viral vaccine vectors that have been used successfully in models for vaccination. These vectors include alphaviruses, human rhinoviruses (HRVs), Ads, picornaviruses, poxviruses, measles viruses, influenza, and vaccinia viruses.Citation30,Citation129,Citation147Citation156 Each of these vectors has its respective disadvantages and advantages with respect to vaccine development. Some advantages of a few of these vectors include their ability to naturally infect a wide variety of cell types and tissues of interest.Citation157Citation162 Each respective vector has its own set of disadvantages. For instance, one disadvantage of using the poliovirus or the HRV as a vaccine vector is the insert size limit restriction of these vectors as compared to the large insert size (~8 kb) accommodation of Ad vectors.

The most common disadvantage of the majority of viral vaccine vectors is reduced vaccine efficacy due to vector preexisting immunity (PEI).Citation163Citation167 Various strategies have been employed to circumvent the problems associated with vector PEI. Specifically, as it relates to Ad vectors, PEI is a tremendous problem. Of the identified serotypes of Ad vectors, human serotypes 5 (Ad5) and 2 (Ad2) have been the most extensively used for gene therapy protocols. Ad5 has been used for HIV-1 vaccination protocols, most recently in the STEP study. As it relates to Ad2 and Ad5, PEI to these vectors may be found in up to 50% of the American population and up to 95% of the population of other countries. This Ad PEI can limit the effectiveness of Ad-based vaccinations.Citation168Citation170 To circumvent Ad2 or Ad5 PEI, researchers have employed the use of vector chimeras,Citation166,Citation171 use of alternative serotypes,Citation172Citation178 and the use of nonhuman Ads,Citation151 such as chimpanzee Ad. The chimpanzee Ad virus was demonstrated to not be significantly neutralized by human sera, which gives chimpanzee Ad an advantage for human vaccine development.Citation179Citation181

Other strategies have been used to reduce the immune response against Ad vectors such as the use of helperdependent Ad (HD-Ads) vectors,Citation182Citation187 the use of Ad delivery in combination with biochemical modifications such as PEGylation,Citation188Citation194 and the use of vector delivery by means of cell vehicles.Citation195,Citation196 With respect to the HD-Ads, these vectors were produced to further increase the safety and cloning capacity of first-generation Ad vectors. HD-Ads lack Ad genes and contain only the packaging signals and end terminal repeats. These vectors were designed to avoid cellular immunity and diminish liver toxicity, thus promoting long-term transgene expression.Citation197Citation200 The reduced immune response against HD-Ads has allowed for transgene expression in mice and baboons for years.Citation182,Citation183,Citation185,Citation200 This long-term transgene expression could be helpful for antigen production for an HIV vaccine, thus producing an opportunity to have increased protection against HIV, with reduced frequency of vaccinations.

Although Nabs to Ad5 may reduce the immunogenicity of Ad5-based vectors in animal model systems, their effect on the immunity in subjects with previous Ad5 exposure is still largely unknown. As previously mentioned, the STEP trial, which tested a Merck recombinant Ad5 (rAd5) vaccine (encoding HIV-1 gag, pol, and ne1 genes), failed to yield protection, either by lowering viral load or by decreasing acquisition of infection.Citation13 Analysis of data from this study aroused speculation that subjects with pre-existing Nabs from wild-type Ad5 infection had an increased risk of HIV infection after vaccination. One recent study has shown that there was no causative role for Ad5-specific CD4+ T cells in increasing HIV-1 susceptibility in the Merck trial.Citation201 In this regard, there are multiple studies ongoing to elucidate a concrete finding with respect to the role of Ad5 PEI and increased activation of CD4+ T cells in the mucosal milieu.Citation202,Citation203

Recently, there was a report by Cheng and colleagues that attempted to characterize the specificity of rAd5 Nabs in Ad5- immune subjects and determine the impact of Ad exposure on immune responses elicited by Ad5-based vaccinations. Cheng and colleagues reported that rAd5 Nabs were directed toward different components of the Ad virion, depending on whether the Ad5 infection was natural or from Ad-based HIV vaccine trials. For example, Ad Nabs generated by natural infection are directed primarily to fiber components, while vector exposure elicits responses primarily to capsid proteins other than fiber. Nabs elicited by natural infection significantly reduced the CD8+ and CD4+ cell responses to HIV Gag after DNA/rAd5 vaccination. This report concluded that Ad5 Nabs differ based on the route of exposure and that previous Ad5 exposure compromises Ad5 vaccine-induced immunity to weak immunogens, such as HIV-1 Gag.Citation204 These results have a tremendous impact on HIV-1 vaccine trials and the design of next generation viral vaccine vectors.

Viral vectors such as Ad, influenza, and polio have been used as vaccine vectors for many reasons. One important advantage of these vectors, which makes them attractive, is that they can provide mucosal immunity because they can easily infect the mucosal surfaces as well as act to induce cytokine and chemokine production at the mucosal entry sites. Ad, influenza, and polio also have the advantage of being able to be delivered orally, without the use of needles. This is an important fact in developing countries where needle cost is prohibitive to vaccine administration. As it relates to HIV vaccine development, mucosal immunity is a debatable factor to consider.

Mucosal HIV immunity

When deciding upon a vaccine agent, the importance of considering if the ultimate goal is to induce systemic immunity, mucosal immunity, or both is worth careful consideration.Citation205Citation207 It is believed that 80% of HIV-1 infection will occur from heterosexual viral transmission and most of the rest will occur from homosexual or perinatal transmission.Citation152 Although the biology of sexual transmission is poorly understood, it is clear that the essential first step in the infection pathway is the transfer of infectious virus or HIV-infected cells through the mucosal surfaces. After HIV has entered a new host, the HIV or HIV-infected cells will soon encounter susceptible host target cells at the mucosal point of entry where the virus replicates and then invades local lymphatic tissues, initiating systemic HIV infection. On this basis, strong immunity is required to provide a protective immunological barrier at the most common point of entry, the mucosal surfaces of the reproductive tract. Due to the compartmentalization of the secretory and systemic immune systems, parenterally administered antigens do not consistently stimulate mucosal immunity.Citation152 Therefore, it is important to consider a vaccine regime that induces mucosal immunity.

Since CD4+ CCR5+ memory T cells are the primary target of HIV infection in the gut and mucosa and rapid depletion of this subset occurs early after infection,Citation208,Citation209 several studies have investigated the role of HIV mucosal immunity. Previous studies have demonstrated the importance of a mucosal SIV/HIV vaccine producing both strong mucosal antibody and CD8+ response capable of blocking the escape of virus from the intestinal mucosa into systemic lymphoid organs.Citation207,Citation210Citation214 However, in other instances, the necessity of exclusive mucosal HIV immunity will be further debated based on the promising results found in a heterologous prime/boost regimen using DNA/89.6-expressing SIV and HIV-1 transcriptsCitation215,Citation216 and modified vaccinia virus Ankara (MVA/89.6)-expressing SIV and HIV-1 transcripts under the control of vaccinia virus early/late promoter. In this case, either ID or IM DNA/MVA vaccination was able to provide protection against a intrarectal SHIV-89.6 challenge. Citation153 Along these same lines, recently, promising results were found by Hessell and colleagues in 2010. Hessell and colleagues demonstrated that after an IV administration of monoclonal antibodies 2F5 or 4E10 to six monkeys followed by a SHIVba-L challenge, five out of six monkeys from either group showed complete protection and sterilizing immunity. A low level of viral replication could not be ruled out for the six monkeys in either group.Citation217

Replicative Ad yields a robust immune response at the mucosal sites partly because Ad is known to infect and replicate in epithelial cells.Citation218Citation221 Various strategies have been used to achieve mucosal immunity via the oral route. One such strategy embodies the development of replication-defective recombinant Ad serotype 41 (Ad41) vector.Citation222 Serotype 41 vectors are being currently used because Ad41 has a natural tropism for the gut and causes no pathological disease outside of the gastrointestinal tract.Citation223 Ad41 vectors are likely to have a preferential tropism for the gut because Ad41 appears to have a resistance to acidic pHCitation224 and the capsid configuration of long and short fibers allows the Ad41 virus to preferentially infect the gut.Citation177,Citation225

Live recombinant vectors for vaccine development engineered to express/present HIV-1 antigens

As previously mentioned, viral vectors are potent inducers of cellular and humoral responses. Of note, viral vectors have been practically used for human applications and have progressed treating a variety of disease contexts such as cancer and infectious diseases.Citation226Citation229 Traditional viral vector immunization embodies the concept that the vector uses the host cell machinery to express antigens, which are encoded as transgenes within the viral vector. Cellular and humoral immune responses are generated against these antigens. Over the last 20 years, several viral vectors have been derived to express HIV-1 antigens for vaccine purposes.

Some researchers have taken an alternative approach to conventional transgene expression of antigens by means of viral vectors; this alternative approach embodies the capsid incorporation of antigens. This innovative paradigm is based upon the vector presenting the antigen as a component of the capsid rather than an encoded transgene. Incorporation of immunogenic peptides into the vector capsid offers potential advantages. In this regard, the processing of the capsid-incorporated antigen via the exogenous pathway should result in a strong humoral response similar to the response provoked by native Ad capsid proteins. In this arrangement, potentially, HIV peptide antigens accrue the potent immunostimulatory effects of the native Ad vector capsid proteins, which effectively perform an adjuvant function. On this basis, the immune response directed against vector capsid proteins with repetitive vector administration should achieve a booster effect against the incorporated antigen.Citation230 Most importantly, as it relates to HIV infection, this strategy yields the potential of generating antibodies to HIV proteins. Recent crystallographic, cryo-electron tomography, and molecular modeling studies have provided valuable insight to molecular surfaces recognized by antibodies as well as assisted in rationale vaccine design of immunogens.Citation231Citation235 These structural technologies can also potentially improve the abilities of scientists to advance the antigen capsidincorporation strategy. If the antigen capsid incorporation is effective, it can provide a way forward with respect to inducing sterilizing immunity.Citation68,Citation236,Citation237

The antigen capsid-incorporation strategy has been used for Ad-based vaccines in the context of many diseases.Citation230,Citation238Citation242 One of the first examples where the antigen capsid-incorporation strategy was used was with research performed by Crompton in 1994.Citation242 Crompton and colleagues inserted an eight-amino acid sequence of the VP1 capsid protein of poliovirus type 3 into two regions of the Ad2 serotype hexon. One of the chimeric vectors produced grew well in tissue culture. In addition, antiserum raised against the Ad with the polio insert specifically recognized the VP1 capsid of polio type 3. As it relates to Ad5 serotype, Wu and group demonstrated that HisCitation6 epitopes could be incorporated into Ad hexon hypervariable regions (HVRs) 1–7 (now reclassified as 1–9) without perturbing viral viability and any major biological characteristics such as replication, thermostability, or native infectivity. This study by Wu and colleagues demonstrated that HisCitation6 appeared to be surface exposed at these regions.Citation243 With respect to peptide incorporation within Ad5 hexon, HVR2 and HVR5 appear to be the most promising locales for peptide/antigen incorporation based on X-ray and peptide analyses along with molecular studies.Citation244 Our laboratory and others have focused on incorporations at HVR5 or single-site incorporations (such as fiber and pIX).Citation230,Citation238Citation241–,Citation243,Citation245,Citation246 However, we recognized that the ability to place antigen within multiple sites of the Ad capsid protein would hold important potential for presenting multiple epitopes/antigens or several copies of the same epitope within a single Ad vector-based vaccine.

In an effort to create multivalent HIV vaccine vectors, our 2008 study explored the use of Ad5 HVR2 and HVR5 in hopes of creating vectors which contained HIV antigenic epitopes at both locales. To compare the flexibility and capacities of Ad5 HVR2 and HVR5, we genetically incorporated identical epitopes of incrementally increasing size within HVR2 or HVR5 of Ad5 hexon. We incorporated identical epitopes ranging from 33 to 83 amino acids within the Ad5 hexon HVR2 or HVR5 region. Viable viruses were produced with incorporations of 33 amino acids plus a 12-amino acid linker at HVR2 or HVR5. In addition, viable viruses were produced with incorporations of up to 53 amino acids plus a 12-amino acid linker at HVR5. With respect to identical antigen incorporations at Ad5 HVR2 or HVR5, HVR5 was more permissive allowing an epitope incorporation of 65 amino acids in total. These model antigens were surface exposed via ELISA analysis. In vivo immunization with these vectors illustrated an antigen-specific immune response.Citation240

Along these same lines, Abe and colleagues evaluated the ability of Ad5-based vectors expressing an HIV transgene to induce antigen-specific immune responses under Ad5 preimmune conditions. To overcome limitations that are generally experienced as a result of PEI to Ad5, they constructed vectors that have a modification in the HVR5. Their study characterized various immunological parameters generated by these vectors such as vector neutralization, acquisition of adaptive immune response, and comparison of protective immunity. First, in order to evaluate the utility of the modified Ad vector, they measured the neutralizing activity of sera by a modified Ad vector. They administered Ad-Luc (luciferase protein expressed as a transgene in the Ad E1 region), Ad-HisLuc (HisCitation6 epitope presented in HVR5 region and luciferase protein expressed as a transgene), or Ad-END/AAALuc vector (containing three amino acid mutations in HVR5 and expressing luciferase protein) to mice IM. After administration of these vectors, neutralizing activity against Ad5 was observed for 0–8 weeks. The hexon-modified vector (Ad-HisLuc) generated the lowest Ad5-specific neutralizing activity, which was significantly lower than what was generated by Ad-Luc at weeks 6 and 8, and by Ad-End/AAALuc vector at week 8. The individual neutralizing activity of Ad-HisLuc immunization was significantly lower than that of Ad-Luc immunization. Additional studies performed by Abe and colleagues support the concept that modified hexon thwarts Ad5 Nabs and promotes cellular immune responses. Citation247 Studies performed by this research group indicate that a change in the immunogenic epitope is necessary to avoid neutralization by pre-existing Nabs.

Our recently published work exploits the antigen capsidincorporation strategy for HIV vaccination. Our novel vectors were constructed in hopes of moving toward the goal of creating vectors that will provide cellular and humoral HIV immunity. Our study is the first of its kind to genetically incorporate an HIV antigen within the Ad5 hexon HVR2 alone or in combination with genomic incorporation of a Gag transgene (Ad5/HVR2-MPER-L15(Gag)). In this study, we successfully incorporated a 24-amino acid epitope of HIV-1 within HVR2. The HIV-1 region selected for HVR2 incorporation was the membrane proximal ectodomain region (MPER) derived from HIV-1 glycoprotein 41 (gp41). Our rationale for choosing a portion of the MPER (EKNEKELLELDKWASLWNWFDITN) derived from gp41 was based on the fact that the gp41 envelope protein ectodomain is a target of three broadly neutralizing anti-HIV-1 antibodies.Citation248 When the MPER was incorporated into HVR2 in combination with transgene expression, we observed growth kinetics and thermostability changes similar to those of other capsid-incorporated vectors generated in other studies,Citation249,Citation250 indicating that incorporation of the MPER epitope within HVR2 was not dramatically detrimental to virological characteristics.Citation250,Citation251 In addition, we demonstrated that the MPER epitope is surface exposed within HVR2. Most importantly, we observed a humoral anti-HIV response in mice vaccinated with the hexon-modified vector. The MPER-modified vector allows boosting compared to AdCMVGag, possibly because the Ad5/HVR2-MPER-L15(Gag) Ad elicits less anti-Ad5 immune response. It is possible that the MPER epitope reduced the immunogenicity of the Ad5 vector. This finding is noteworthy because HVR2 has not been fully explored for antigen capsid-incorporation strategies.Citation252 These vectors are currently being analyzed by cryo-electron microscopic analysis to determine the critical correlates related to antigen placement/configuration and immune response.

In addition, with respect to HIV-1 vaccination, the antigen capsid-incorporation strategy has been evaluated within the context of HRV. Research groups have constructed human rhinovirus:HIV-1 chimeras in an effort to stimulate immunity against HIV-1.Citation148,Citation253 In an effort to develop HIV-1 vaccines, researchers within this same group generated combinatorial libraries of HRV capsid-incorporated HIV-1 gp41 epitope. Their results indicated that they were successful in eliciting antibodies whose activity can mimic the Nab effect.Citation149

Commercial and clinical Ad development of HIV-1 vaccines have progressed preferentially more than vector systems such as HRV because the flexibility of Ad generally exceeds current rhinovirus systems. For example, because HRV is a relatively small RNA virus, the HRV platform can display an array limited to 60 copies of a single HIV-1 epitope.Citation148,Citation253 In contrast, the Ad vector platform allows incorporation of the HIV-1 MPER epitope into three structurally distinct locales, including HVR2, HVR5,Citation247 and protein IX (our unpublished data). In comparison, the Ad MPER antigen capsid-incorporation display platform could present an array of 720 HIV-1 epitope copies within Ad hexon and 240 HIV-1 epitope copies within pIX. If a multivalent Ad vector is generated with HIV-1 epitopes within the hexon and the pIX locales, this would represent 960 HIV epitopes within one Ad vector. Another significant difference between the Ad and HRV platforms is in the number of locales that have been successfully used for heterologous epitope insertion. Finally, in contrast to the rhinovirus that lacks this capacity, the Ad platform has sufficient coding capacity allowing for HIV-1 transgene expression in combination with presenting the same or a different antigen on the viral capsid surface. This latter finding is important because it provides the basis for constructing vectors that will provide cellular and humoral HIV-1 immunity. Vectors which provide both cellular and humoral immunity may be the way forward with respect to prophylactic HIV vaccine development.

Promising results in an effort to produce an HIV vaccine

Recently, there have been encouraging developments regarding HIV vaccination. In the 1980s, in Thailand, there was a substantial increase in the prevalence of infection with HIV-1.Citation254Citation256 By first observation, these groups consisted of intravenous-drug users and commercial sex workers; this infected group then expanded to the general population.Citation101 By the mid 1990s, the overall seroprevalence of HIV-1 reached a peak of 3.7% among members of the Royal Thai Army and of 12.5% among people from Northern Thailand.Citation255,Citation257 The Thai Ministry of Public Health acted by starting an effective HIV-prevention campaign. With this effort, the number of new HIV-1 infections per year decreased from an estimated 143,000 in 1990 to 14,000 in 2007.Citation255,Citation258Citation260 Although this decrease was promising, there was still a desire to do more to prevent HIV infection. To achieve this goal, an HIV Phase III study was begun.

The Thai Phase III HIV vaccine study, also known as RV144, opened in the fall of 2003. The placebo-controlled trial tested the safety and effectiveness of a prime-boost regimen of two vaccines: ALVAC-HIV vaccine (the prime), a modified canarypox vaccine, and AIDSVAX B/E vaccine (booster), a gp 120 vaccine. The vaccines were based on the subtype E and B HIV-1 strains that commonly circulate in Thailand. The subtype B HIV-1 strain is the most commonly found strain in the United States. The trial, conducted in the Chonburi and Rayong provinces of Thailand, enrolled 16,402 women and men aged 18–30 years at various levels of risk for HIV infection. Study participants received the placebo or ALVAC HIV vaccine at enrollment and again after 1, 3, and 6 months. The placebo or AIDSVAX B/E vaccine was given to participants at 3 and 6 months. Participants were tested for HIV-1 infection every 6 months for 3 years. During each clinic visit, study participants were counseled on how to prevent HIV-1 infection.

The results showed that 74 of 8198 placebo recipients became infected with HIV-1 compared with 51 of 8197 participants who received the vaccine. This level of effectiveness in preventing HIV-1 infection was found to be statistically significant. The vaccine strategy had no effect, however, on the amount of virus in the blood of volunteers who acquired HIV-1 infection during the study. Based on the final analysis of the study, the surgeon general of the US Army, the trial sponsor, announced that the prime-boost investigational vaccine regimen was safe and 31% effective in preventing HIV-1 infection. With respect to an HIV-1 vaccine that can provide sterilizing HIV immunity, this is the best result in humans to date. However, the modest protection effect appeared limited to low-risk individuals, and there were data which suggest that this effect was confined to the first year following administration of the vaccine. Efforts must continue to focus on evaluating the immune response induced by the vaccine to establish potential correlates of protection.

Conclusion

Over the last three decades, the world has been faced with the emergence and subsequent epidemic of HIV/AIDS. There has been much progress with respect to diagnosis and prevention. On the treatment front, there have been several significant advances with respect to drug development (ie, ART/HAART). However, there is a desperate need for an effective and safe vaccine. There has been tremendous difficulty with regard to developing a vaccine that provides sterilizing immunity. This has been the case due to some of the factors mentioned in this review such as HIV diversity, immune evasion, and lack of appropriate animal models. Due to these obstacles, many researchers assumed that the control of HIV-1 viremia by vaccination would be a more realistic goal than the development of sterilizing immunity.

The road to a safe and effective HIV-1 vaccine received a serious setback in the fall of 2007 with the premature termination of the Merck-HIV-1 Vaccine STEP trial due to the lack of efficacy and early speculation that the vaccine might have increased the risk of HIV infection in some populations of vaccinees. In late 2009, promising results came in from Thailand in response to their efforts to create a safe and effective vaccine against HIV-1. A community-based, randomized, multicenter, double-blinded, placebo-controlled efficacy trial using a prime-boost combination showed 31% effectiveness in preventing HIV-1 infection. These results lend promise to the hope of producing an HIV-1 vaccine vector that yields sterilizing HIV-1 immunity.

In the future, research scientists must work together to increase HIV-1 vaccine effectiveness beyond 31%. Realization of this goal may be accomplished by some of the techniques mentioned in this review, such as acquisition of HIV mucosal immunity, development of effective prime-boost strategies, development of better animal models, better molecular antigen modeling and presentation, avoidance of PEI (by the means of using novel vector serotypes in combination with PEGylation), and/or induction of Nabs (by means of capsid incorporation of HIV antigens within viral vectors). These are just a few considerations that scientists and clinicians must consider with respect to the development of an effective and safe HIV-1 vaccine. Scientists and clinicians must also consider that one vector or scheme may not be sufficient with respect to providing effective HIV-1 immunity and some combination of the above-mentioned potential strategies may offer the most promising method of producing an effective HIV-1 prophylactic vaccine.

Acknowledgments

Grant support was provided by National Institutes of Health grants 1R33AI076096-01, 2P30AI027767-21A1, and 3P30CA013148-38S9. We thank Erin E Thacker for her thoughtful insight.

Disclosure

The authors report no conflicts of interest in this work.

References

  • UNAIDS2008 Report on the Global AIDS Epidemic2010 Available from: http://www.unaids.org/en/KnowledgeCentre/HIVData/GlobalReport/2008/2008_Global_report.aspAccessed 2010 Oct 3
  • DugganJMLocherAFinkBOkontaCChakrabortyJAdherence to antiretroviral therapy: a survey of factors associated with medication usageAIDS Care20092191141114720024773
  • ThiebautRDaucourtVMerciePLipodystrophy, metabolic disorders, and human immunodeficiency virus infection: Aquitaine Cohort, France, 1999. Groupe d’Epidemiologie Clinique du Syndrome d’Immunodeficience Acquise en AquitaineClin Infect Dis20003161482148711096016
  • CarrAMillerJLawMCooperDAA syndrome of lipoatrophy, lactic acidaemia and liver dysfunction associated with HIV nucleoside analogue therapy: contribution to protease inhibitor-related lipodystrophy syndromeAIDS2000143F253210716495
  • MondyKYarasheskiKPowderlyWGLongitudinal evolution of bone mineral density and bone markers in human immunodeficiency virus-infected individualsClin Infect Dis200336448249012567307
  • NoeAPlumJVerhofstedeCThe latent HIV-1 reservoir in patients undergoing HAART: an archive of pre-HAART drug resistanceJ Antimicrob Chemother200555441041215728140
  • ShacklettBLMucosal immunity to HIV: a review of recent literatureCurr Opin HIV AIDS20083554154719373019
  • BurtonDRDesrosiersRCDomsRWHIV vaccine design and the neutralizing antibody problemNat Immunol20045323323614985706
  • McMichaelAJHIV vaccinesAnnu Rev Immunol20062422725516551249
  • EminiEAKoffWCAIDS/HIV. Developing an AIDS vaccine: need, uncertainty, hopeScience200430456791913191415218131
  • ThornerARBarouchDHHIV-1 vaccine development: progress and prospectsCurr Infect Dis Rep200791717517254507
  • RobinsonHLAmaraRRT cell vaccines for microbial infectionsNat Med200511Suppl 4S253215812486
  • SekalyRPThe failed HIV Merck vaccine study: a step back or a launching point for future vaccine development?J Exp Med2008205171218195078
  • AlmondNKentKCranageMRudEClarkeBStottEJProtection by attenuated simian immunodeficiency virus in macaques against challenge with virus-infected cellsLancet19953458961134213447752758
  • Stahl-HennigCDittmerUNissleinTRapid development of vaccine protection in macaques by live-attenuated simian immuno-deficiency virusJ Gen Virol199677Pt 12296929819000087
  • LearmontJTindallBEvansLLong-term symptomless HIV-1 infection in recipients of blood products from a single donorLancet199234088248638671357294
  • BerkhoutBVerhoefKvan WamelJLBackNKGenetic instability of live, attenuated human immunodeficiency virus type 1 vaccine strainsJ Virol1999732113811459882315
  • LearmontJCookLDunckleyHSullivanJSUpdate on long-term symptomless HIV type 1 infection in recipients of blood products from a single donorAIDS Res Hum Retroviruses199511117734182
  • DuerrAWasserheitJNCoreyLHIV vaccines: new frontiers in vaccine developmentClin Infect Dis200643450051116838241
  • FlynnNMForthalDNHarroCDJudsonFNMayerKHParaMFPlacebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infectionJ Infect Dis2005191565466515688278
  • GilbertPBPetersonMLFollmannDCorrelation between immunologic responses to a recombinant glycoprotein 120 vaccine and incidence of HIV-1 infection in a phase 3 HIV-1 preventive vaccine trialJ Infect Dis2005191566667715688279
  • GrahamBSMascolaJRLessons from failure – preparing for future HIV-1 vaccine efficacy trialsJ Infect Dis2005191564764915688276
  • PitisuttithumPGilbertPGurwithMRandomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injection drug users in Bangkok, ThailandJ Infect Dis2006194121661167117109337
  • HemelaarJGouwsEGhysPDOsmanovSGlobal and regional distribution of HIV-1 genetic subtypes and recombinants in 2004AIDS20062016W132317053344
  • PlantierJCLeozMDickersonJEA new human immunodeficiency virus derived from gorillasNat Med200915887187219648927
  • RenjifoBGilbertPChaplinBPreferential in-utero transmission of HIV-1 subtype C as compared to HIV-1 subtype A or DAIDS200418121629163615280773
  • BhoopatLRithapornTSKhunamornpongSBhoopatTTaylorCRThornerPSCell reservoirs in lymph nodes infected with HIV-1 subtype E differ from subtype B: identification by combined in situ polymerase chain reaction and immunohistochemistryMod Pathol200619225526316341147
  • VasanARenjifoBHertzmarkEDifferent rates of disease progression of HIV type 1 infection in Tanzania based on infecting subtypeClin Infect Dis200642684385216477563
  • KaleebuPNankyaILYirrellDLRelation between chemokine receptor use, disease stage, and HIV-1 subtypes A and D: results from a rural Ugandan cohortJ Acquir Immune Defic Syndr2007451283317310935
  • CohenJHIV. Escape artist par excellenceScience200329956121505150812624245
  • MooreJPParrenPWBurtonDRGenetic subtypes, humoral immunity, and human immunodeficiency virus type 1 vaccine developmentJ Virol200175135721572911390574
  • McBurneySPRossTMViral sequence diversity: challenges for AIDS vaccine designsExpert Rev Vaccines2008791405141718980542
  • SeamanMSLeblancDFGrandpreLEStandardized assessment of NAb responses elicited in rhesus monkeys immunized with single- or multi-clade HIV-1 envelope immunogensVirology2007367117518617599382
  • KibuukaHKimutaiRMabokoLA phase 1/2 study of a multiclade HIV-1 DNA plasmid prime and recombinant adenovirus serotype 5 boost vaccine in HIV-Uninfected East Africans (RV 172)J Infect Dis2010201460060720078213
  • SantraSLiaoHXZhangRMosaic vaccines elicit CD8(+) T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeysNat Med201016332432820173754
  • CoreyLMcElrathMJHIV vaccines: mosaic approach to virus diversityNat Med201016326827020208511
  • BarouchDHO’BrienKLSimmonsNLMosaic HIV-1 vaccines expand the breadth and depth of cellular immune responses in rhesus monkeysNat Med201016331932320173752
  • ParrenPWMarxPAHessellAJAntibody protects macaques against vaginal challenge with a pathogenic R5 simian/human immunodeficiency virus at serum levels giving complete neutralization in vitroJ Virol200175178340834711483779
  • BabaTWLiskaVHofmann-LehmannRHuman neutralizing monoclonal antibodies of the IgG1 subtype protect against mucosal simian-human immunodeficiency virus infectionNat Med20006220020610655110
  • MascolaJRStieglerGVanCottTCProtection of macaques against vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive infusion of neutralizing antibodiesNat Med20006220721010655111
  • KorberBGaschenBYusimKThakallapallyRKesmirCDetoursVEvolutionary and immunological implications of contemporary HIV-1 variationBr Med Bull200158194211714622
  • WyattRKwongPDDesjardinsEThe antigenic structure of the HIV gp120 envelope glycoproteinNature199839366867057119641684
  • KwongPDWyattRRobinsonJSweetRWSodroskiJHendricksonWAStructure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibodyNature199839366866486599641677
  • WeiXDeckerJMWangSAntibody neutralization and escape by HIV-1Nature2003422692930731212646921
  • FenouilletEBarboucheRJonesIMCell entry by enveloped viruses: redox considerations for HIV and SARS-coronavirusAntioxid Redox Signal2007981009103417567241
  • DeVicoAFoutsTLewisGKAntibodies to CD4-induced sites in HIV gp120 correlate with the control of SHIV challenge in macaques vaccinated with subunit immunogensProc Natl Acad Sci U S A200710444174771748217956985
  • WalkerLMPhogatSKChan-HuiPYBroad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine targetScience2009326595028528919729618
  • BurtonDRPyatiJKoduriREfficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibodyScience19942665187102410277973652
  • MusterTSteindlFPurtscherMA conserved neutralizing epitope on gp41 of human immunodeficiency virus type 1J Virol19936711664266477692082
  • StieglerGKunertRPurtscherMA potent cross-clade neutralizing human monoclonal antibody against a novel epitope on gp41 of human immunodeficiency virus type 1AIDS Res Hum Retroviruses200117181757176511788027
  • TrkolaAPomalesABYuanHCross-clade neutralization of primary isolates of human immunodeficiency virus type 1 by human monoclonal antibodies and tetrameric CD4-IgGJ Virol19956911660966177474069
  • LagenaurLAVillarroelVABundocVDeyBBergerEAsCD4-17b bifunctional protein: extremely broad and potent neutralization of HIV-1 Env pseudotyped viruses from genetically diverse primary isolatesRetrovirology201071120158904
  • CortiDLangedijkJPHinzAAnalysis of memory B cell responses and isolation of novel monoclonal antibodies with neutralizing breadth from HIV-1-infected individualsPLoS One201051e880520098712
  • EasterbrookPJSchragerLKLong-term nonprogression in HIV infection: methodological issues and scientific prioritiesReport of an International European Community-National Institutes of Health Workshop, The Royal SocietyLondon, England1995 Nov 27–29Scientific Coordinating CommitteeAIDS Res Hum Retroviruses19981414121112289764904
  • ParoliMPropatoAAccapezzatoDFrancavillaVSchiaffellaEBarnabaVThe immunology of HIV-infected long-term nonprogressors– a current viewImmunol Lett20017912127129
  • JohnsonBKStoneGALong-term observations of human immunodeficiency virus-infected chimpanzeesAIDS Res Hum Retroviruses1993943753788352831
  • GardnerMBSimian and feline immunodeficiency viruses: animal lentivirus models for evaluation of AIDS vaccines and antiviral agentsAntiviral Res19911542672861659310
  • PodellMMarchPABuckWRMathesLEThe feline model of neuroAIDS: understanding the progression towards AIDS dementiaJ Psychopharmacol200014320521311106298
  • YamamotoJKSanouMPAbbottJRColemanJKFeline immunodeficiency virus model for designing HIV/AIDS vaccinesCurr HIV Res201081142520210778
  • MosierDEGuliziaRJBairdSMWilsonDBSpectorDHSpectorSAHuman immunodeficiency virus infection of human-PBL-SCID miceScience199125149957917941990441
  • McCuneJMNamikawaRShihCCRabinLKaneshimaHSuppression of HIV infection in AZT-treated SCID-hu miceScience199024749425645662300816
  • BaenzigerSTussiwandRSchlaepferEDisseminated and sustained HIV infection in CD34+ cord blood cell-transplanted Rag2−/−gamma c−/− miceProc Natl Acad Sci U S A200610343159511595617038503
  • GardnerMBLuciwPAAnimal models of AIDSFASEB J1989314259326062556312
  • LiJLordCIHaseltineWLetvinNLSodroskiJInfection of cynomolgus monkeys with a chimeric HIV-1/SIVmac virus that expresses the HIV-1 envelope glycoproteinsJ Acquir Immune Defic Syndr1992576396461613662
  • ReimannKALiJTVeazeyRA chimeric simian/human immunodeficiency virus expressing a primary patient human immunodeficiency virus type 1 isolate env causes an AIDS-like disease after in vivo passage in rhesus monkeysJ Virol19967010692269288794335
  • NishimuraYIgarashiTDonauOKHighly pathogenic SHIVs and SIVs target different CD4+ T cell subsets in rhesus monkeys, explaining their divergent clinical coursesProc Natl Acad Sci U S A200410133123241232915297611
  • BuchbinderSPMehrotraDVDuerrAEfficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trialLancet200837296531881189319012954
  • ShiverJWFuTMChenLReplication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunityNature2002415686933133511797011
  • KestlerHWIIIRinglerDJMoriKImportance of the nef gene for maintenance of high virus loads and for development of AIDSCell19916546516622032289
  • DanielMDKirchhoffFCzajakSCSehgalPKDesrosiersRCProtective effects of a live attenuated SIV vaccine with a deletion in the nef geneScience19922585090193819411470917
  • RudEWCranageMYonJMolecular and biological characterization of simian immunodeficiency virus macaque strain 32H proviral clones containing nef size variantsJ Gen Virol199475Pt 35295438126450
  • CranageMPWhatmoreAMSharpeSAMacaques infected with live attenuated SIVmac are protected against superinfection via the rectal mucosaVirology199722911431549123856
  • MarthasMLSutjiptoSHigginsJImmunization with a live, attenuated simian immunodeficiency virus (SIV) prevents early disease but not infection in rhesus macaques challenged with pathogenic SIVJ Virol1990648369437002164591
  • ShacklettBLShawKEAdamsonLALive, attenuated simian immunodeficiency virus SIVmac-M4, with point mutations in the Env transmembrane protein intracytoplasmic domain, provides partial protection from mucosal challenge with pathogenic SIVmac251J Virol20027622113651137812388697
  • Protection of macaques against simian immunodeficiency virus infection with inactivated vaccines: comparison of adjuvants, doses and challenge viruses. The European Concerted Action on ‘Macaque Models for AIDS Research’Vaccine19951332953007631516
  • PutkonenPNilssonCHildKWhole inactivated SIV vaccine grown on human cells fails to protect against homologous SIV grown on simian cellsJ Med Primatol19932223100103
  • Stahl-HennigCVossGDittmerUProtection of monkeys by a split vaccine against SIVmac depends upon biological properties of the challenge virusAIDS1993767877958363756
  • KusuharaHHohdatsuTOkumuraMDual-subtype vaccine (Fel-O-Vax FIV) protects cats against contact challenge with heterologous subtype B FIV infected catsVet Microbiol200510834155165
  • PuRColemanJOmoriMDual-subtype FIV vaccine protects cats against in vivo swarms of both homologous and heterologous subtype FIV isolatesAIDS200115101225123711426067
  • HohdatsuTOkadaSMotokawaKAizawaCYamamotoJKKoyamaHEffect of dual-subtype vaccine against feline immunodeficiency virus infectionVet Microbiol19975824155165
  • PuRColemanJCoismanJDual-subtype FIV vaccine (Fel-O-Vax FIV) protection against a heterologous subtype B FIV isolateJ Feline Med Surg200571657015686976
  • BirxDLLoomis-PriceLDAronsonNEfficacy testing of recombinant human immunodeficiency virus (HIV) gp160 as a therapeutic vaccine in early-stage HIV-1-infected volunteers. rgp160 hase II Vaccine InvestigatorsJ Infect Dis2000181388188910720508
  • EronJJJrAshbyMAGiordanoMFRandomised trial of MNrgp120 HIV-1 vaccine in symptomless HIV-1 infectionLancet19963489041154715518950881
  • PontesilliOGuerraECAmmassariAPhase II controlled trial of post-exposure immunization with recombinant gp160 versus antiretroviral therapy in asymptomatic HIV-1-infected adults. VaxSyn Protocol TeamAIDS19981254734809543445
  • Ratto-KimSSitzKVGarnerRPRepeated immunization with recombinant gp160 human immunodeficiency virus (HIV) envelope protein in early HIV-1 infection: evaluation of the T cell proliferative responseJ Infect Dis199917923373449878016
  • RedfieldRRBirxDLKetterNA phase I evaluation of the safety and immunogenicity of vaccination with recombinant gp160 in patients with early human immunodeficiency virus infection. Military Medical Consortium for Applied Retroviral ResearchN Engl J Med199132424167716841674589
  • SchooleyRTSpinoCKuritzkesDTwo double-blinded, randomized, comparative trials of 4 human immunodeficiency virus type 1 (HIV-1) envelope vaccines in HIV-1-infected individuals across a spectrum of disease severity: AIDS Clinical Trials Groups 209 a nd 214J Infect Dis200018251357136411023459
  • ValentineFTKunduSHaslettPAA randomized, placebo-controlled study of the immunogenicity of human immunodeficiency virus (HIV) rgp160 vaccine in HIV-infected subjects with > or = 400/mm3 CD4 T lymphocytes (AIDS Clinical Trials Group Protocol 137)
  • WahrenBBrattGPerssonCImproved cell-mediated immune responses in HIV-1-infected asymptomatic individuals after immunization with envelope glycoprotein gp160J Acquir Immune Defic Syndr1994732202297906300
  • WrightPFLambertJSGorseGJImmunization with envelope MN rgp120 vaccine in human immunodeficiency virus-infected pregnant womenJ Infect Dis199918041080108810479134
  • WeberJCheinsong-PopovRCallowDImmunogenicity of the yeast recombinant p17/p24:Ty virus-like particles (p24-VLP) in healthy volunteersVaccine19951398318347483805
  • VeenstraJWilliamsIGColebundersRImmunization with recombinant p17/p24:Ty virus-like particles in human immunodeficiency virus-infected personsJ Infect Dis199617448628668843231
  • KelleherADRoggensackMJaramilloABSafety and immunogenicity of a candidate therapeutic vaccine, p24 virus-like particle, combined with zidovudine, in asymptomatic subjects. Community HIV Research Network InvestigatorsAIDS19981221751829468366
  • BensonEMClarksonJLawMTherapeutic vaccination with p24-VLP and zidovudine augments HIV-specific cytotoxic T lymphocyte activity in asymptomatic HIV-infected individualsAIDS Res Hum Retroviruses199915210511310029243
  • OkudaKBukawaHHamajimaKInduction of potent humoral and cell-mediated immune responses following direct injection of DNA encoding the HIV type 1 env and rev gene productsAIDS Res Hum Retroviruses19951189339437492440
  • NeumannJStitzJKonigRRetroviral vectors for vaccine development: induction of HIV-1-specific humoral and cellular immune responses in rhesus macaques using a novel MLV(HIV-1) pseudotype vectorJ Biotechnol2006124361562516563543
  • BuffaVNegriDRLeonePA single administration of lentiviral vectors expressing either full-length human immunodeficiency virus 1 (HIV-1)(HXB2) Rev/Env or codon-optimized HIV-1(JR-FL) gp120 generates durable immune responses in miceJ Gen Virol200687Pt 61625163416690927
  • FaulEJAyePPPapaneriABRabies virus-based vaccines elicit neutralizing antibodies, poly-functional CD8+ T cell, and protect rhesus macaques from AIDS-like disease after SIV(mac251) challengeVaccine200928229930819879223
  • McKennaPMKoserMLCarlsonKRHighly attenuated rabies virus-based vaccine vectors expressing simian-human immunodeficiency virus89.6P Env and simian immunodeficiency virusmac239 Gag are safe in rhesus macaques and protect from an AIDS-like diseaseJ Infect Dis2007195798098817330788
  • QuinnanGVJrYuXFLewisMGProtection of rhesus monkeys against infection with minimally pathogenic simian-human immunodeficiency virus: correlations with neutralizing antibodies and cytotoxic T cellsJ Virol20057963358336915731230
  • Rerks-NgarmSPitisuttithumPNitayaphanSVaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in ThailandN Engl J Med2009361232209222019843557
  • JohnsonPRSchneppBCZhangJVector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeysNat Med200915890190619448633
  • ChencinerNRandrianarison-JewtoukoffVDelpeyrouxFEnhancement of humoral immunity to SIVenv following simultaneous inoculation of mice by three recombinant adenoviruses encoding SIVenv/poliovirus chimeras, Tat and RevAIDS Res Hum Retroviruses19971398018069171225
  • SantraSSeamanMSXuLReplication-defective adenovirus serotype 5 vectors elicit durable cellular and humoral immune responses in nonhuman primatesJ Virol200579106516652215858035
  • NatukRJDavisARChandaPKAdenovirus vectored vaccinesDev Biol Stand19948271777958485
  • BruceCBAkriggASharpeSAHankeTWilkinsonGWCranageMPReplication-deficient recombinant adenoviruses expressing the human immunodeficiency virus Env antigen can induce both humoral and CTL immune responses in miceJ Gen Virol199980Pt 102621262810573155
  • ZhanXMartinLNSlobodKSMulti-envelope HIV-1 vaccine devoid of SIV components controls disease in macaques challenged with heterologous pathogenic SHIVVaccine200523464753065320
  • LaubeLSBurrascanoMDejesusCECytotoxic T lymphocyte and antibody responses generated in rhesus monkeys immunized with retroviral vector-transduced fibroblasts expressing human immunodeficiency virus type-1 IIIB ENV/REV proteinsHum Gene Ther1994578538627981310
  • BuffaVNegriDRLeonePEvaluation of a self-inactivating lentiviral vector expressing simian immunodeficiency virus gag for induction of specific immune responses in vitro and in vivoViral Immunol200619469070117201664
  • HalseyRJTanzerFLMeyersAChimaeric HIV-1 subtype C Gag molecules with large in-frame C-terminal polypeptide fusions form virus-like particlesVirus Res2008133225926818329748
  • PillaySMeyersAWilliamsonALRybickiEPOptimization of chimeric HIV-1 virus-like particle production in a baculovirus-insect cell expression systemBiotechnol Prog20092541153116019572400
  • DemlLKratochwilGOsterriederNKnuchelRWolfHWagnerRIncreased incorporation of chimeric human immunodeficiency virus type 1 gp120 proteins into Pr55 gag virus-like particles by an Epstein-Barr virus gp220/350-derived transmembrane domainVirology1997235110259300033
  • DemlLSchirmbeckRReimannJWolfHWagnerRRecombinant human immunodeficiency Pr55 gag virus-like particles presenting chimeric envelope glycoproteins induce cytotoxic T-cells and neutralizing antibodiesVirology1997235126399300034
  • KangCYLuoLWainbergMALiYDevelopment of HIV/AIDS vaccine using chimeric gag-env virus-like particlesBiol Chem1999380335336410223338
  • DonnellyJJUlmerJBShiverJWLiuMADNA vaccinesAnnu Rev Immunol1997156176489143702
  • GarzonMRBerraondoPCrettazJInduction of gp120-specific protective immune responses by genetic vaccination with linear polyethylenimine-plasmid complexVaccine200523111384139215661387
  • LiuMADNA vaccines: a reviewJ Intern Med2003253440241012653868
  • RaskaMBelakovaJWudattuNKComparison of protective effect of protein and DNA vaccines hsp90 in murine model of systemic candidiasisFolia Microbiol (Praha)2005501778215954537
  • GiriMUgenKEWeinerDBDNA vaccines against human immunodeficiency virus type 1 in the past decadeClin Microbiol Rev200417237038915084506
  • WolffJAMaloneRWWilliamsPDirect gene transfer into mouse muscle in vivoScience19902474949 Pt 1146514681690918
  • CalarotaSAWeinerDBEnhancement of human immunodeficiency virus type 1-DNA vaccine potency through incorporation of T-helper 1 molecular adjuvantsImmunol Rev2004199849915233728
  • Billaut-MulotOIdziorekTLoyensMCapronABahrGMModulation of cellular and humoral immune responses to a multiepitopic HIV-1 DNA vaccine by interleukin-18 DNA immunization/viral protein boostVaccine200119202228032811
  • DaleCJde RoseRWilsonKMEvaluation in macaques of HIV-1 DNA vaccines containing primate CpG motifs and fowlpoxvirus vaccines co-expressing IFNgamma or IL-12Vaccine200423218819715531036
  • BarouchDHSantraSSteenbekeTDAugmentation and suppression of immune responses to an HIV-1 DNA vaccine by plasmid cytokine/Ig administrationJ Immunol19981614187518829712056
  • HashidaMNishikawaMYamashitaFTakakuraYCell-specific delivery of genes with glycosylated carriersAdv Drug Deliv Rev200152318719611718943
  • ZhangYSchlachetzkiFLiJYBoadoRJPardridgeWMOrgan-specific gene expression in the rhesus monkey eye following intravenous non-viral gene transferMol Vis2003946547214551536
  • ZouSMErbacherPRemyJSBehrJPSystemic linear polyethylenimine (L-PEI)-mediated gene delivery in the mouseJ Gene Med20002212813410809146
  • ZuberGZammut-ItalianoLDautyEBehrJPTargeted gene delivery to cancer cells: directed assembly of nanometric DNA particles coated with folic acidAngew Chem Int Ed Engl200342232666266912813749
  • HarariABartPAStohrWAn HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responsesJ Exp Med20082051637718195071
  • JaokoWKaritaEKayitenkoreKSafety and immunogenicity study of multiclade HIV-1 adenoviral vector vaccine alone or as boost following a multiclade HIV-1 DNA vaccine in AfricaPLoS One201059e1287320877623
  • CombeEPirmanTStekarJHoulierMLMirandPPDifferential effect of lentil feeding on proteosynthesis rates in the large intestine, liver and muscle of ratsJ Nutr Biochem2004151121714711455
  • RaskaMMoldoveanuZNovakJDelivery of DNA HIV-1 vaccine to the liver induces high and long-lasting humoral immune responsesVaccine200826121541155118304708
  • LetvinNLWalkerBDImmunopathogenesis and immunotherapy in AIDS virus infectionsNat Med20039786186612835706
  • ReitterJNMeansREDesrosiersRCA role for carbohydrates in immune evasion in AIDSNat Med1998466796849623976
  • HellerLJaroszeskiMJCoppolaDPottingerCGilbertRHellerRElectrically mediated plasmid DNA delivery to hepatocellular carcinomas in vivoGene Ther200071082682910845719
  • Vorup-JensenTJensenUBLiuHTail-vein injection of mannan-binding lectin DNA leads to high expression levels of multimeric protein in liverMol Ther20013686787411407900
  • BudkerVGSubbotinVMBudkerTSebestyenMGZhangGWolffJAMechanism of plasmid delivery by hydrodynamic tail vein injection. II. Morphological studiesJ Gene Med20068787488816718734
  • SebestyenMGBudkerVGBudkerTMechanism of plasmid delivery by hydrodynamic tail vein injection. I. Hepatocyte uptake of various moleculesJ Gene Med20068785287316724360
  • BudkerVZhangGDankoIWilliamsPWolffJThe efficient expression of intravascularly delivered DNA in rat muscleGene Ther1998522722769578848
  • LiuFSongYLiuDHydrodynamics-based transfection in animals by systemic administration of plasmid DNAGene Ther1999671258126610455434
  • ZhangGBudkerVWolffJAHigh levels of foreign gene expression in hepatocytes after tail vein injections of naked plasmid DNAHum Gene Ther199910101735173710428218
  • SudaTLiuDHydrodynamic gene delivery: its principles and applicationsMol Ther200715122063206917912237
  • StockertRJThe asialoglycoprotein receptor: relationships between structure, function, and expressionPhysiol Rev19957535916097624395
  • MaierMAYannopoulosCGMohamedNSynthesis of antisense oligonucleotides conjugated to a multivalent carbohydrate cluster for cellular targetingBioconjug Chem2003141182912526688
  • WaddingtonSNMcVeyJHBhellaDAdenovirus serotype 5 hexon mediates liver gene transferCell2008132339740918267072
  • KalyuzhniyOdi PaoloNCSilvestryMAdenovirus serotype 5 hexon is critical for virus infection of hepatocytes in vivoProc Natl Acad Sci U S A2008105145483548818391209
  • LiSLockeEBruderJViral vectors for malaria vaccine developmentVaccine200725142567257416914237
  • SmithADGeislerSCChenAAHuman rhinovirus type 14:human immunodeficiency virus type 1 (HIV-1) V3 loop chimeras from a combinatorial library induce potent neutralizing antibody responses against HIV-1J Virol19987216516599420270
  • ArnoldGFVelascoPKHolmesAKBroad neutralization of human immunodeficiency virus type 1 (HIV-1) elicited from human rhinoviruses that display the HIV-1 gp41 ELDKWA epitopeJ Virol200983105087510019279101
  • PattersonSPapagatsiasTBenlahrechAUse of adenovirus in vaccines for HIVHandb Exp Pharmacol200918827529319031031
  • BangariDSMittalSKDevelopment of nonhuman adenoviruses as vaccine vectorsVaccine200624784986216297508
  • CrottySAndinoRPoliovirus vaccine strains as mucosal vaccine vectors and their potential use to develop an AIDS vaccineAdv Drug Deliv Rev200456683585215063593
  • AmaraRRVillingerFAltmanJDControl of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccineScience20012925514697411393868
  • GherardiMMEstebanMRecombinant poxviruses as mucosal vaccine vectorsJ Gen Virol200586Pt 112925293616227213
  • LangleyWABradleyKCLiZNSmithMESchnellMJSteinhauerDAInduction of neutralizing antibody responses to anthrax protective antigen by using influenza virus vectors: implications for disparate immune system priming pathwaysJ Virol201084168300830720504926
  • SlobodKSLockeyTDHowlettNSubcutaneous administration of a recombinant vaccinia virus vaccine expressing multiple envelopes of HIV-1Eur J Clin Microbiol Infect Dis200423210611014735404
  • DmitrievIKrasnykhVMillerCRAn adenovirus vector with genetically modified fibers demonstrates expanded tropism via utilization of a coxsackievirus and adenovirus receptor-independent cell entry mechanismJ Virol19987212970697139811704
  • BarnettBGCrewsCJDouglasJTTargeted adenoviral vectorsBiochim Biophys Acta200215751311412020813
  • KayMAGloriosoJCNaldiniLViral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeuticsNat Med200171334011135613
  • TangyFNaimHYLive attenuated measles vaccine as a potential multivalent pediatric vaccination vectorViral Immunol200518231732616035943
  • EsolenLMWardBJMoenchTRGriffinDEInfection of monocytes during measlesJ Infect Dis1993168147528515132
  • HillemanMRVaccinology, immunology, and comparative pathogenesis of measles in the quest for a preventative against AIDSAIDS Res Hum Retroviruses19941013128179962
  • MandlSHixLAndinoRPreexisting immunity to poliovirus does not impair the efficacy of recombinant poliovirus vaccine vectorsJ Virol200175262262711134275
  • ThackerETimaresLMatthewsQLStrategies to overcome host immunity to adenovirus vectors in vaccine developmentExpert Rev Vaccines20098676177719485756
  • SumidaSMTruittDMLemckertAANeutralizing antibodies to adenovirus serotype 5 vaccine vectors are directed primarily against the adenovirus hexon proteinJ Immunol2005174117179718515905562
  • WuHDmitrievIKashentsevaESekiTWangMCurielDTConstruction and characterization of adenovirus serotype 5 packaged by serotype 3 hexonJ Virol20027624127751278212438602
  • RobertsDMNandaAHavengaMJHexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunityNature2006441709023924316625206
  • ChirmuleNPropertKMagosinSQianYQianRWilsonJImmune responses to adenovirus and adeno-associated virus in humansGene Ther1999691574158310490767
  • Kass-EislerAFalck-PedersenEElfenbeinDHAlviraMButtrickPMLeinwandLAThe impact of developmental stage, route of administration and the immune system on adenovirus-mediated gene transferGene Ther1994163954027584105
  • Kass-EislerALeinwandLGallJBloomBFalck-PedersenECircumventing the immune response to adenovirus-mediated gene therapyGene Ther1996321541628867863
  • YouilRTonerTJSuQHexon gene switch strategy for the generation of chimeric recombinant adenovirusHum Gene Ther200213231132011812286
  • MastrangeliAHarveyBGYaoJ‘Sero-switch’ adenovirus-mediated in vivo gene transfer: circumvention of anti-adenovirus humoral immune defenses against repeat adenovirus vector administration by changing the adenovirus serotypeHum Gene Ther19967179878825871
  • MoffattSHaysJHogenEschHMittalSKCircumvention of vector-specific neutralizing antibody response by alternating use of human and non-human adenoviruses: implications in gene therapyVirology2000272115916710873758
  • LemckertAAGrimbergenJSmitsSGeneration of a novel replication-incompetent adenoviral vector derived from human adenovirus type 49: manufacture on PER.C6 cells, tropism and immunogenicityJ Gen Virol200687Pt 102891289916963747
  • ShayakhmetovDMLieberADependence of adenovirus infectivity on length of the fiber shaft domainJ Virol20007422102741028611044071
  • KiddAHChroboczekJCusackSRuigrokRWAdenovirus type 40 virions contain two distinct fibersVirology1993192173848517033
  • YehHYPieniazekNPieniazekDGelderblomHLuftigRBHuman adenovirus type 41 contains two fibersVirus Res19943321791987975882
  • Pichla-GollonSLDrinkerMZhouXStructure-based identification of a major neutralizing site in an adenovirus hexonJ Virol20078141680168917108028
  • RoySGaoGLuYCharacterization of a family of chimpanzee adenoviruses and development of molecular clones for gene transfer vectorsHum Gene Ther200415551953015144581
  • FarinaSFGaoGPXiangZQReplication-defective vector based on a chimpanzee adenovirusJ Virol20017523116031161311689642
  • KobingerGPFeldmannHZhiYChimpanzee adenovirus vaccine protects against Zaire Ebola virusVirology2006346239440116356525
  • UmanaPGerdesCAStoneDEfficient FLPe recombinase enables scalable production of helper-dependent adenoviral vectors with negligible helper-virus contaminationNat Biotechnol200119658258511385466
  • LowensteinPRThomasCEUmanaPHigh-capacity, helperdependent, ‘gutless’ adenoviral vectors for gene transfer into brainMethods Enzymol200234629231111883074
  • XiongWGoverdhanaSSciasciaSARegulatable gutless adenovirus vectors sustain inducible transgene expression in the brain in the presence of an immune response against adenovirusesJ Virol2006801273716352528
  • XiongWCandolfiMKroegerKMImmunization against the transgene but not the TetON switch reduces expression from gutless adenoviral vectors in the brainMol Ther200816234335118180781
  • WeaverEANehetePNBuchlSSComparison of replication-competent, first generation, and helper-dependent adenoviral vaccinesPLoS One200943e505919333387
  • WeaverEANehetePNNeheteBPProtection against mucosal SHIV challenge by peptide and helper-dependent adenovirus vaccinesViruses20091392093820107521
  • WeaverEABarryMAEffects of shielding adenoviral vectors with polyethylene glycol (PEG) on vector-specific and vaccine-mediated immune responsesHum Gene Ther200819121369138218778197
  • O’RiordanCRSongAPEGylated adenovirus for targeted gene therapyMethods Mol Biol200843413316018470643
  • EtoYYoshiokaYMukaiYOkadaNNakagawaSDevelopment of PEGylated adenovirus vector with targeting ligandInt J Pharm20083541238
  • EtoYGaoJQSekiguchiFNeutralizing antibody evasion ability of adenovirus vector induced by the bioconjugation of methoxypolyethylene glycol succinimidyl propionate (MPEG-SPA)Biol Pharm Bull200427693693815187452
  • EtoYGaoJQSekiguchiFPEGylated adenovirus vectors containing RGD peptides on the tip of PEG show high transduction efficiency and antibody evasion abilityJ Gene Med20057560461215543536
  • MokHPalmerDJNgPBarryMAEvaluation of polyethylene glycol modification of first-generation and helper-dependent adenoviral vectors to reduce innate immune responsesMol Ther2005111667915585407
  • CroyleMALeHTLinseKDPEGylated helper-dependent adenoviral vectors: highly efficient vectors with an enhanced safety profileGene Ther200512757958715647765
  • ZabaletaALlopizDArribillagaLVaccination against hepatitis C virus with dendritic cells transduced with an adenovirus encoding NS3 proteinMol Ther200816121021717923840
  • RothJCCurielDTPereboevaLCell vehicle targeting strategiesGene Ther2008151071672918369326
  • BangariDSMittalSKCurrent strategies and future directions for eluding adenoviral vector immunityCurr Gene Ther20066221522616611043
  • BrownBDShiCXRawleFEFactors influencing therapeutic efficacy and the host immune response to helper-dependent adenoviral gene therapy in hemophilia A miceJ Thromb Haemost20042111111814717974
  • de GeestBSnoeysJvan LinthoutSLievensJCollenDElimination of innate immune responses and liver inflammation by PEGylation of adenoviral vectors and methylprednisoloneHum Gene Ther200516121439145116390275
  • MorralNO’NealWRiceKAdministration of helper-dependent adenoviral vectors and sequential delivery of different vector serotype for long-term liver-directed gene transfer in baboonsProc Natl Acad Sci U S A19999622128161282110536005
  • HutnickNACarnathanDGDubeySABaseline Ad5 serostatus does not predict Ad5 HIV vaccine-induced expansion of adenovirus-specific CD4+ T cellsNat Med200915887687819620962
  • KaufmanDRBarouchDHTranslational mini-review series on vaccines for HIV: T lymphocyte trafficking and vaccine-elicited mucosal immunityClin Exp Immunol2009157216517319604255
  • KaufmanDRBivas-BenitaMSimmonsNLMillerDBarouchDHRoute of adenovirus-based HIV-1 vaccine delivery impacts the phenotype and trafficking of vaccine-elicited CD8+ T lymphocytesJ Virol201084125986599620357087
  • ChengCGallJGNasonMDifferential specificity and immunogenicity of adenovirus type 5 neutralizing antibodies elicited by natural infection or immunizationJ Virol201084163063819846512
  • OgraPLKarzonDTDistribution of poliovirus antibody in serum, nasopharynx and alimentary tract following segmental immunization of lower alimentary tract with poliovaccineJ Immunol19691026142314304977607
  • OgraPLOgraSSal-NakeebSCoppolaPRLocal antibody response to experimental poliovirus infection in the central nervous system of rhesus monkeysInfect Immun1973869319374361727
  • AhlersJDBelyakovIMStrategies for optimizing targeting and delivery of mucosal HIV vaccinesEur J Immunol200939102657266919609978
  • MehandruSPolesMATenner-RaczKPrimary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tractJ Exp Med2004200676177015365095
  • BrenchleyJMSchackerTWRuffLECD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tractJ Exp Med2004200674975915365096
  • BelyakovIMHelZKelsallBMucosal AIDS vaccine reduces disease and viral load in gut reservoir and blood after mucosal infection of macaquesNat Med20017121320132611726972
  • BelyakovIMKuznetsovVAKelsallBImpact of vaccine-induced mucosal high-avidity CD8+ CTLs in delay of AIDS viral dissemination from mucosaBlood200610783258326416373659
  • BelyakovIMIsakovDZhuQDzutsevABerzofskyJAA novel functional CTL avidity/activity compartmentalization to the site of mucosal immunization contributes to protection of macaques against simian/human immunodeficiency viral depletion of mucosal CD4+ T cellsJ Immunol2007178117211722117513770
  • StaatsHFMontgomerySPPalkerTJIntranasal immunization is superior to vaginal, gastric, or rectal immuni - zation for the induction of systemic and mucosal anti-HIV antibody responsesAIDS Res Hum Retroviruses199713119459529223410
  • KozlowskiPACu-UvinSNeutraMRFlaniganTPComparison of the oral, rectal, and vaginal immunization routes for induction of antibodies in rectal and genital tract secretions of womenInfect Immun1997654138713949119478
  • GorelickRJBenvenisteREGagliardiTDNucleocapsid protein zinc-finger mutants of simian immunodeficiency virus strain mne produce virions that are replication defective in vitro and in vivoVirology199925322592709918884
  • SauterMMPelchen-MatthewsABronRAn internalization signal in the simian immunodeficiency virus transmembrane protein cytoplasmic domain modulates expression of envelope glycoproteins on the cell surfaceJ Cell Biol199613257958118603913
  • HessellAJRakaszEGTehraniDMBroadly neutralizing monoclonal antibodies 2F5 and 4E10 directed against the human immunodeficiency virus type 1 gp41 membrane-proximal external region protect against mucosal challenge by simian-human immunodeficiency virus SHIVBa-LJ Virol20108431302131319906907
  • NatukRJLubeckMDChandaPKImmunogenicity of recombinant human adenovirus-human immunodeficiency virus vaccines in chimpanzeesAIDS Res Hum Retroviruses1993953954048318268
  • LubeckMDNatukRMyagkikhMLong-term protection of chimpanzees against high-dose HIV-1 challenge induced by immunizationNat Med1997366516589176492
  • PattersonLJMalkevitchNVenzonDProtection against mucosal simian immunodeficiency virus SIV(mac251) challenge by using replicating adenovirus-SIV multigene vaccine priming and subunit boostingJ Virol20047852212222114963117
  • MalkevitchNRohneDPinczewskiJEvaluation of combination DNA/replication-competent Ad-SIV recombinant immunization regimens in rhesus macaquesAIDS Res Hum Retroviruses200420223524415018712
  • LemialeFHaddadaHNabelGJBroughDEKingCRGallJGNovel adenovirus vaccine vectors based on the enteric-tropic serotype 41Vaccine200725112074208417250935
  • ChristensenMLHuman viral gastroenteritisClin Microbiol Rev19892151892644024
  • FavierALBurmeisterWPChroboczekJUnique physicochemical properties of human enteric Ad41 responsible for its survival and replication in the gastrointestinal tractVirology200432219310415063120
  • PieniazekNJSlemendaSBPieniazekDVelardeJJrLuftigRBHuman enteric adenovirus type 41 (Tak) contains a second fiber protein geneNucleic Acids Res19901871901
  • McConnellMJHannaPCImperialeMJAdenovirus-based prime-boost immunization for rapid vaccination against anthraxMol Ther200715120321017164792
  • SullivanNJSanchezARollinPEYangZYNabelGJDevelopment of a preventive vaccine for Ebola virus infection in primatesNature2000408681260560911117750
  • LiuRYWuLZHuangBJAdenoviral expression of a truncated S1 subunit of SARS-CoV spike protein results in specific humoral immune responses against SARS-CoV in ratsVirus Res200511212243116022896
  • Gomez-RomanVRRobert-GuroffMAdenoviruses as vectors for HIV vaccinesAIDS Rev20035317818514598567
  • McConnellMJDanthinneXImperialeMJCharacterization of a permissive epitope insertion site in adenovirus hexonJ Virol200680115361537016699016
  • SchiefWRBanYEStamatatosLChallenges for structure-based HIV vaccine designCurr Opin HIV AIDS20094543144020048708
  • LinGNaraPLDesigning immunogens to elicit broadly neutralizing antibodies to the HIV-1 envelope glycoproteinCurr HIV Res20075651454118045109
  • WalkerLMBurtonDRRational antibody-based HIV-1 vaccine design: current approaches and future directionsCurr Opin Immunol201022335836620299194
  • LiuJBartesaghiABorgniaMJSapiroGSubramaniamSMolecular architecture of native HIV-1 gp120 trimersNature2008455720910911318668044
  • ZhouTXuLDeyBStructural definition of a conserved neutralization epitope on HIV-1 gp120Nature2007445712973273717301785
  • LifsonJDMartinMAOne step forwards, one step backNature2002415686927227311796990
  • LetvinNLBarouchDHMontefioriDCProspects for vaccine protection against HIV-1 infection and AIDSAnnu Rev Immunol200220739911861598
  • WorgallSBuschARivaraMModification to the capsid of the adenovirus vector that enhances dendritic cell infection and transgene-specific cellular immune responsesJ Virol20047852572258014963160
  • WorgallSKrauseARivaraMProtection against P. aeruginosa with an adenovirus vector containing an OprF epitope in the capsidJ Clin Invest200511551281128915841217
  • MatthewsQLYangPWuQOptimization of capsid-incorporated antigens for a novel adenovirus vaccine approachJ Virol2008598
  • KrauseAJohJHHackettNREpitopes expressed in different adenovirus capsid proteins induce different levels of epitope-specific immunityJ Virol200680115523553016699033
  • CromptonJToogoodCIWallisNHayRTExpression of a foreign epitope on the surface of the adenovirus hexonJ Gen Virol199475Pt 11331397509367
  • WuHHanTBelousovaNIdentification of sites in adenovirus hexon for foreign peptide incorporationJ Virol20057963382339015731232
  • RuxJJKuserPRBurnettRMStructural and phylogenetic analysis of adenovirus hexons by use of high-resolution X-ray crystallographic, molecular modeling, and sequence-based methodsJ Virol200377179553956612915569
  • WorgallSKrauseAQiuJJohJHackettNRCrystalRGProtective immunity to pseudomonas aeruginosa induced with a capsid-modified adenovirus expressing P. aeruginosa OprFJ Virol20078124138011380817942539
  • VigneEMahfouzIDedieuJFBrieAPerricaudetMYehPRGD inclusion in the hexon monomer provides adenovirus type 5-based vectors with a fiber knob-independent pathway for infectionJ Virol19997365156516110233980
  • AbeSOkudaKUraTAdenovirus type 5 with modified hexons induces robust transgene-specific immune responses in mice with pre-existing immunity against adenovirus type 5J Gene Med200911757057919391169
  • OfekGTangMSamborAStructure and mechanistic analysis of the anti-human immunodeficiency virus type 1 antibody 2F5 in complex with its gp41 epitopeJ Virol20047819107241073715367639
  • MatthewsQLSibleyDAWuHGenetic incorporation of a herpes simplex virus type 1 thymidine kinase and firefly luciferase fusion into the adenovirus protein IX for functional display on the virionMol Imaging20065451051917150163
  • TangYLeLPMatthewsQLHanTWuHCurielDTDerivation of a triple mosaic adenovirus based on modification of the minor capsid protein IXVirology2008377239140018570971
  • LiJLeLSibleyDAMathisJMCurielDTGenetic incorporation of HSV-1 thymidine kinase into the adenovirus protein IX for functional display on the virionVirology2005338224725815996701
  • MatthewsQLFatimaATangYHIV antigen incorporation within adenovirus hexon hypervariable 2 for a novel HIV vaccine approachPLoS One201057e1181520676400
  • SmithADResnickDAZhangAGeislerSCArnoldEArnoldGFUse of random systematic mutagenesis to generate viable human rhinovirus 14 chimeras displaying human immunodeficiency virus type 1 V3 loop sequencesJ Virol19946815755798254775
  • BrownTPeerapatanapokinWThe Asian Epidemic Model: a process model for exploring HIV policy and programme alternatives in AsiaSex Transm Infect200480Suppl 1i19i2415249695
  • PhoolcharoenWHIV/AIDS prevention in Thailand: success and challengesScience19982805371187318749669947
  • WenigerBGLimpakarnjanaratKUngchusakKThe epidemiology of HIV infection and AIDS in ThailandAIDS19915Suppl 2S71851845063
  • NitayaphanSBrownAEPreventive HIV vaccine development in ThailandAIDS199812Suppl BS1551619679641
  • HanenbergRSRojanapithayakornWKunasolPSokalDCImpact of Thailand’s HIV-control programme as indicated by the decline of sexually transmitted diseasesLancet199434489172432457913163
  • NelsonKECelentanoDDSuprasertSRisk factors for HIV infection among young adult men in northern ThailandJAMA199327089559608345647
  • Rerks-NgarmSBrownAEKhamboonruangCThongcharoenPKunasolPHIV/AIDS preventive vaccine ‘prime-boost’ phase III trial: foundations and initial lessons learned from ThailandAIDS200620111471147916847401