2,372
Views
51
CrossRef citations to date
0
Altmetric
Review

Therapeutic vaccines against HIV infection

, , , &
Pages 569-581 | Published online: 01 May 2012

Abstract

Resistance to medication, adverse effects in the medium-to-long-term and cost all place important limitations on lifelong adherence to combined antiretroviral therapy (cART). In this context, new therapeutic alternatives to cART for life in HIV-infected patients merit investigation. Some data suggest that strong T cell-mediated immunity to HIV can indeed limit virus replication and protect against CD4 depletion and disease progression. The combination of cART with immune therapy to restore and/or boost immune-specific responses to HIV has been proposed, the ultimate aim being to achieve a functional cure. In this scenario, new, induced, HIV-specific immune responses would be able to control viral replication to undetectable levels, mimicking the situation of the minority of patients who control viral replication without treatment and do not progress to AIDS. Classical approaches such as whole inactivated virus or recombinant protein initially proved useful as therapeutic vaccines. Overall, however, the ability of these early vaccines to increase HIV-specific responses was very limited and study results were discouraging, as no consistent immunogenicity was demonstrated and there was no clear impact on viral load. Recent years have seen the development of new approaches based on more innovative vectors such as DNA, recombinant virus or dendritic cells. Most clinical trials of these new vectors have demonstrated their ability to induce HIV-specific immune responses, although they show very limited efficacy in terms of controlling viral replication. However, some preliminary results suggest that dendritic cell-based vaccines are the most promising candidates. To improve the effectiveness of these vaccines, a better understanding of the mechanisms of protection, virological control and immune deterioration is required; without this knowledge, an efficacious therapeutic vaccine will remain elusive.

Introduction

Combined antiretroviral therapy (cART) has become one the best tools to control HIV infection at both the individualCitation1 and social level.Citation2,Citation3 Indeed, cART has achieved impressive reductions in morbidity and mortality among patients with advanced human immunodeficiency virus (HIV).Citation1 However, despite these advances in infection control, the cost of medication, the mandatory high adherence to medication for life, side effectsCitation4 and the risk of development of resistance remain unsolved problems. Furthermore, the HIV epidemic has severely hampered economic growth in developing countries. Although the number of HIV-infected patients under treatment in these countries has increased steadily in recent years, and although the number of new HIV infections has fallen, the demand for treatment continues to rise.Citation5 In fact, the number of new infections is still higher than the number of new patients on treatment. For economic, social, cultural and political reasons, widespread treatment is difficult to implement in these countries. Given this situation, there is a need for new therapeutic alternatives to “cART for life” in HIV-infected patients.Citation6

Two such alternatives which have been proposed are eradication and functional cure.Citation7 There is one report of an HIV-infected patient in whom viral replication remained absent, despite discontinuation of antiretroviral therapy, after transplantation with CCR5Δ32/Δ32 stem cells.Citation8 Recently published follow-ups of this patient strongly suggest that cure of HIV has been achieved.Citation9 Intensification of cART,Citation10 stem cell therapy,Citation11 gene therapyCitation12 and the elimination of latently infected cells are other alternatives to eradication which are currently being assessed.Citation7 However, it is unlikely that therapeutic strategies leading to eradication will emerge as a valid alternative to cART, and in fact, there are no biological models to support this approach. Conversely, the concept of “functional cure,” defined as control of HIV replication without cART, is based on the observation that a small proportion of HIV-infected patients (referred to as long-term non-progressors or elite controllers) are able to control HIV replication to below detectable levels, and without abnormalities in CD4 T-cell counts, for more than 25 y. It has been proposed that the strong HIV-specific immune responses observed in these patients could account for this natural “functional cure.” Specifically, control of viral replication has been associated with HLA types B*57:01, B*27:05 and B*14, and it has been proposed that the viral peptides presented by these HLA molecules induce strong HIV-specific CD8 T-cell responses which are responsible for viral control.Citation13,Citation14 These findings prompted the development of new strategies based on the use of anti-HIV vaccines to induce high frequencies and breadth of response of HIV-specific CD8 cytotoxic T lymphocytes (CTLs), together with strong CD4 T-cell help, in HIV-infected patients.Citation15,Citation16 Recent reports suggest that a strong T cell-mediated immunity to HIV can indeed limit virus replication and protect against CD4 depletion and disease progression.Citation17 Here we will review the basis of the therapeutic vaccine strategy and the different candidates tested in human clinical trials.

Pathogenic Basis for a Therapeutic Vaccine Design

HIV-specific responses against HIV infection in antiretroviral naive patients

The main question to be answered here is whether the immune system can contain viral replication without cART, even if only for limited periods. One major problem is that the types of immunological responses which are able to control viral replication are not known, although it does seem that the relevance of the different response types changes with the stage of HIV infection. Barouch et al. recently published the results of a preventive vaccine which suggest that protection against infection is mediated mainly by antibodies against env, whereas post-infection control of viral replication is associated with cellular responses against gag.Citation18 Recent data reported by Picker's group in relation to SIV-infected rhesus macaques suggest that the induction of effector memory T-cell responses (and not central memory T-cell responses) with a CMV-vectored vaccine is essential for control of viral replication.Citation19

Anti-HIV-1 CTL responses have been detected in all studied cases of acute HIV infection, and it is believed that they reduce the peak plasma viral load (PVL) to the stabilization level (set-point) of PVL which is reached at the end of the acute phase.Citation20,Citation21 It has therefore been hypothesized that CTL play an essential role in controlling viral replication during acute infection. However, it is not clear that neutralizing antibodies contribute to the control of viral replication during acute infection, since a delay in the development of these antibodies is observed which is not associated with changes in viral replication.Citation22 Finally, and in contrast to CTL and neutralizing antibodies, a clonal depletion of HIV-specific CD4+ T cells occurs during acute HIV infection. It has been suggested that the lack of response of these cells is key to explaining the failure to control viral replication during this phase.Citation20,Citation21 In fact, massive infection and loss of memory CD4+ T cells occur in multiple tissues during acute infection,Citation23 and HIV mainly infects HIV-specific CD4+ T cells.Citation24 These findings may provide a basis for explaining why a therapeutic vaccine-induced transient activation of HIV-specific CD4—but not CD8—T cells might have a detrimental effect on HIV replication,Citation25 and could offer important clues for the future design of therapeutic vaccines.

Some data suggest that strong T cell-mediated immunity to HIV can indeed limit virus replication and protect against CD4 depletion and disease progression in chronic HIV-infected patients. Although in most infected patients replication leads to the progressive destruction of the immune system and evolves inevitably toward AIDS, a small number of immunologically “privileged” individuals, or long-term non-progressors (LTNP), have a potent and sustained response of anti-HIV-1 CTL, Th cells and neutralizing HIV-1 antibodies. This is associated with control of viral replication and the presence of very low or undetectable viral concentrations in plasma in the absence of cART.Citation20 Direct data on the critical role of the CTL response in controlling viral replication have been obtained in both the infection model with macaques devoid of CD8+ T lymphocytes,Citation26,Citation27 and in the immunodeficient murine model.Citation28 In addition, there is clear evidence from both humanCitation17,Citation29 and animal modelsCitation30 that a specific helper T response against HIV is crucial in obtaining an optimal, specific CTL response which can control viral replication. This is consistent with other reported data on chronic viral infections in murine models.Citation31 Finally, studies in animal models have shown that high levels of neutralizing antibodies can block infection regardless of the route of exposure to the virus.Citation32 It has also been reported that plasma virus continually and rapidly evolves to escape neutralization, indicating that neutralizing antibodies exert a level of selective pressure. It is probable that neutralizing antibody responses account for the extensive variation in the envelope gene observed in the early months after primary HIV infection.Citation33

Despite the relevance of HIV-specific immune responses in both the acute and chronic stages of infection these responses are not, in most patients, able to control viral replication to undetectable levels and prevent the infection from progressing to AIDS. Many authors hypothesize that selective escape from CD8+ T-cell and other immune responses is the main reason for the failure to control HIV replication.Citation34-Citation42 Some studies suggest that selective escape from CD8+ T-cell responses represents a major driving force of HIV-1 sequence diversity. Allen et al.Citation35 assessed the relationship between viral evolution and adaptive CD8+ T-cell responses in four HIV-infected patients who were studied for five years after acute infection. Nearly two-thirds of the amino acid mutations identified in non-envelope antigens were attributable to CD8+ T-cell selective pressures. The preferential selection of individual residues for mutation and the repeated selection for identical mutations observed in this study suggest that there are significant biochemical constraints on viral evolution. In addition, some mutations induced by immune-mediated selective pressures have an impact on viral replication capacity.Citation39,Citation43 These findings help to decide which epitopes should be used in a therapeutic vaccine in order to avoid or limit the viral escape to HIV CTL response.

In addition to viral escape, the lack of control of viral replication in HIV infection could be secondary to functional defects of immune responses, which may be explained in terms of alterations to the immune system. Although CD4+ and CD8+ cells capable of secreting interferon gamma (IFN-gamma) can be found in most HIV-infected patients, proliferative CD4 responses are normally absentCitation29,Citation44,Citation45 and CD8+ cells have defective cytolytic activity.Citation46,Citation47,Citation48 One explanation for these functional deficits of CD4 and CD8 responses could be that the antigen-presenting functions of the dendritic cells have deteriorated in these patients, which may contribute to the functional defects observed in the Th1 and CTL cellular responses.Citation49,Citation50 The absence of a correct proliferation and expansion of CD4+ responses may, in turn, influence the lack of cytolytic activity of CD8+ cells.Citation44,Citation51 In animal models there is a clear deficit in the secretion of cytokines by CD4+ cells, which starts when PVL peaks in primary infection.Citation52 Lastly, the selective infection of HIV-specific CD4+ cells in infected patients may explain why these responses are quickly lost in HIV infection.Citation24

In summary, it seems clear that both cellular and humoral immune responses are critical for correct control of HIV infection. Viral escape and functional defects in these immune responses are likely to be the main causes of sustained viral replication in HIV infection, and constitute the two main problems that a therapeutic vaccine should solve.

Influence of cART on HIV-specific immune responses

The benefit of cART on the immune system is obtained through an increase in the absolute number of circulating naive CD4 T lymphocytes, a concomitant reduction in the number of T lymphocytes with activation markers, and restoration of the response to memory antigens.Citation53 However, despite the clinical efficacy of cARTCitation54,Citation55 this treatment by itself is unable to eradicate the infection, even if it were administered for more than 60 y.Citation56,Citation57 This is mainly due to the fact that therapy cannot eliminate latent HIV-1 in the form of integrated proviral DNA, as well as to the existence of low levels of viral replication, which makes even cell-to-cell infection possible.Citation58,Citation59 Furthermore, cART is incapable of restoring the immune-specific response to HIV,Citation17,Citation45 and, in fact, it leads to a fall in the specific CTL response due to the lack of antigenic exposure.Citation60 Some reports have shown that the helper proliferative response to HIV p24 Ag presented by some cART patients does not reflect an improvement in the immune phenotype or function of CD4 or CD8 cells, but in fact is secondary to the small increases in viremia typically observed in patients taking cART.Citation61 This would explain the rapid ‘rebound’ of viral load that occurs within days or weeks of suspending cART, even after several years of effective therapy.Citation62,Citation63 This rebound occurs even if cART is initiated in very early-stage HIV-infected patients, in whom the immune system is theoretically still well preserved (circulating CD4+ T lymphocytes above 500 cells/mL; PVL: 5,000–10,000 copies/ml). Similarly, these viral dynamics occur even when immune restoration is practically complete in terms of the homeostasis of T lymphocytes and their subpopulations, as well as in terms of the capacity for response to polyclonal stimuli and memory antigens with cART.Citation62,Citation64 Taken together these data suggest that the first challenge for a therapeutic vaccine is to demonstrate that it is possible to induce de novo HIV-specific responses that are able to control viral replication, albeit only partially. If this is the case, it would be a proof of concept that would justify further investigation of different immunotherapies.

Therapeutic Vaccines

Objectives of a therapeutic vaccine

The main objective of a therapeutic vaccine is to induce or increase immune responses against HIV infection using a planned exposure to HIV viral antigens. In the ideal scenario, these responses would control viral replication to an undetectable level, mimicking the situation of the minority of patients who control viral replication without treatment and do not progress to AIDS. This has been termed “functional cure” and it is observed in most other infections (e.g., herpes or tuberculosis). Previous research has suggested that partial control of viral replication could be enough to delay the initiation of cART in antiretroviral naive patients or to offer “drug holidays” in already-treated patients. However, the current knowledge of HIV pathogenesis precludes this “partial remission” of infection from being a conceivable clinical objective. Even antiretroviral naive patients with a normal CD4 T-cell count could be at higher risk of suffering non-AIDS events (cancer, cardiovascular or neurological events) than the general population.Citation65-Citation67 In addition, it seems that the early initiation of antiretroviral therapy significantly improves survival, as compared with deferred therapy.Citation68 Furthermore, the discontinuation of antiretroviral therapy in order to reduce drug-related adverse effects and to improve the patient’s quality of life has been shown to be associated with increased morbidity and mortality.Citation69 It has been suggested that immune activation due to viral load rebound after ART interruption would explain this increase in the rates of AIDS-defining and non-AIDS-defining malignancies or cardiovascular events among patients who discontinued cART. Untreated HIV infection is characterized by a profound and continuous state of immune activation manifested by an increased turnover of B and T lymphocytes, natural killer cells and a high level of pro-inflammatory cytokines such as IL-7 and TNF-α.Citation70 Another relevant feature is the elevation of activated CD8+ T-cells expressing the DR+/CD38+ phenotype, which is considered as a surrogate marker of disease progression.Citation71,Citation72 Systemic immune activation could lead, first, to exhaustion of the immune system, including lymph node fibrosis,Citation73,Citation74 retention of effector T cells in lymph nodes,Citation75,Citation76 clonal exhaustion and thymic dysfunction. In addition, it could contribute to the spreading of HIV infection by generating more targets for HIV, thereby allowing ongoing HIV replication.Citation24 In summary, partial control of viral replication remains deleterious for HIV-infected patients, and the objective of a therapeutic vaccine should be complete remission of infection. Nonetheless, if a therapeutic vaccine were shown to be capable of partially controlling viral load this would be a proof of concept justifying further investigation of new candidates.

A second objective has recently been proposed for therapeutic vaccines.Citation7 Based on data related to the eradication of HIV in the “Berlin patient”Citation8,Citation9 it has been suggested that the combination of cART intensification with a vaccine able to induce effective HIV-specific responses could purge the HIV reservoir. This strategy, together with others using different drugs or gene therapy, is already being assessed in clinical trials.Citation7

Types of therapeutic vaccines

Most of the therapeutic vaccines tested have previously been used as preventive candidates. Classical approaches such as whole inactivated virus (REMUNE) or recombinant protein (gp120) were initially shown to be useful. In general, however, the capacity of these early vaccines to increase HIV-specific responses was very limited and study results were discouraging, as no consistent immunogenicity was demonstrated and there was no clear impact on viral load.Citation77,Citation78-Citation81,Citation82-Citation89 Recent years have seen the development of new approaches based on more innovative vectors such as DNA, recombinant virus or dendritic cells. Most of the clinical trials involving these new vectors have been exploratory and with a low number of patients. In fact, in randomized studies no therapeutic vaccine has yet to demonstrate lasting and potent effectiveness in terms of controlling viral load, improving CD4 T-cell count or delaying clinical progression. These weak results have precluded further development. Although a great number of candidates have been used in clinical trials, this review will focus on those summarized in .

Table 1. Therapeutic vaccine clinical trials

Remune

The Remune vaccine initially aroused considerable expectation. This is a vaccine of an inactivated whole virus in which the envelope protein has been removed during the process of inactivation, which is performed by synthesis and formulated with incomplete Freund adjuvant. The vaccine stems from a virus originally obtained in Zaire and contains a type-A envelope and type-G gag. It has been administered to more than 3,000 people with an antiviral-controlled virus. The results showed that it was able to induce gag-specific helper responses which were sometimes very potent, but did not suggest a capacity for immunological control of viral replication.Citation87-Citation90 The best and most extensive study aimed to determine whether the addition of Remune would confer added clinical efficacy to that achievable by cART. This was a multicenter, double-blind, placebo-controlled, randomized trial conducted at 77 centers in the United States, and involved 2527 cART-treated, HIV-infected patients with a CD4 T-cell level between 300 × 106/L and 549 × 106/L. There were no differences in HIV progression-free survival, changes in HIV RNA or CD4 cell counts when comparing Remune and placebo recipients.Citation89

Subunit vaccines

Multiple strategies have been used to design therapeutic HIV vaccines based on peptides or recombinant proteins, including vaccination with a recombinant envelope protein,Citation91-Citation100 HIV p24-like peptides (Vacc-4x),Citation101-Citation103 a Tat proteinCitation104-Citation106 and a combination of Nef-Tat fusion protein and envelope glycoprotein gp120 formulated with AS02A adjuvant.Citation107 The basis for the use of these candidates is that a therapeutic HIV vaccine that induces HIV-1-specific CD4+ T-cell responses could improve antiviral immunity, resulting in delayed disease progression.

A number of clinical trials using recombinant gp120 or gp160 as immunogens have been performed both in combination and without antiretrovirals.Citation91-Citation100 Overall, these trials have not demonstrated clinical benefit, although an improvement in specific lymphocyte proliferation is observed in some of them.

Vacc-4x consists of four synthetic peptides corresponding to conserved domains of the HIV-1 protein p24 that preferentially include HLA-A2 restricted elements. To ensure optimal exposure of the immunogen to antigen-presenting cells (APCs) the vaccine was given intradermally together with granulocyte-macrophage colony-stimulating factor (GM-CSF).Citation101 Forty HIV-infected patients on cART were vaccinated with either low-dose or high-dose Vacc-4x over 26 weeks. HIV-associated specific responses were safely induced in 90% of patients in a dose-dependent manner and were influenced by the HLA haplotype.Citation103 Thereafter, cART was discontinued in patients for 14 weeks. Patients with the strongest delayed-type hypersensitivity (DTH) responses before treatment interruption tended to achieve lower actual HIV RNA levels.Citation101 Finally, long-term HIV-specific immune responses and clinical outcomes were evaluated. Vacc-4x-induced cellular immune responses were unchanged 1.5 y after completing immunization, and 62% of patients were still off cART.Citation102

Based on efficacy studies in animal modelsCitation108 and those suggesting that the presence of an anti-Tat immune response correlates with a slower progression to disease,Citation109 some authors have proposed that a preventive and therapeutic Tat protein-based vaccine deserves to be investigated. The first randomized, double blind, placebo-controlled phase I vaccine trial based on the native Tat protein was conducted in HIV-infected asymptomatic individuals.Citation106 The vaccine was well tolerated and induced and/or maintained Tat-specific T helper cell responses in all subjects, with a wide spectrum of functional anti-Tat antibodies. In addition, 2.5 y after the last immunization Tat-specific humoral and cellular immune responses were still detectable in vaccinees.Citation105 These data provided the basis for a phase II study in 87 successfully-treated HIV-infected patients, the results of which have recently been reported.Citation104 It was confirmed that immunization with Tat was safe and immunogenic. In addition, there was a consistent decrease in cellular and biochemical activation markers and persistent increases in the number of regulatory T cells. This was associated with stable increases in the percentage and absolute numbers of both CD4 T cells and B lymphocytes, as well as with a reduction in the percentage of CD8 T cells and NK lymphocytes. The authors concluded that Tat immunization in patients on cART could help to restore immune homeostasis. The improvement in immune system function (as opposed to functional cure) is a benefit that some other investigators have claimed as an alternative primary end-point for therapeutic vaccines.Citation90,Citation110 However, the clinical relevance of the recovery of these parameters is not clear. Until this issue is clarified, functional cure remains the best objective for a therapeutic vaccine.

A clinical trial of the gp120/NefTat/AS02A therapeutic vaccine in chronically HIV-1-infected volunteers on suppressive ART found the vaccine to be safe, well tolerated and able to induce strong gp120-specific CD4+ T-cell responses, and a higher number of vaccinees developed both HIV-1-specific CD4+ T-cell responses and CD8+ T-cell proliferation.Citation107 Therapy interruption to determine whether these immunological responses might lead to improved control of HIV-1viral load was not performed.

Vaccines using DNA as a vector

DNA vaccines are based on bacterial plasmids that express the desired antigen in situ, which leads to induction of an immune response of the Th1 type. The first trial, reported in 1998, included nine asymptomatic HIV-1-infected patients who were immunized with DNA constructs encoding the nef, rev or tat regulatory genes of HIV-1.Citation111 The vaccination induced HIV-1-specific cellular responses, but was not able to control viral replication or change CD4+ T-cell counts. These results were confirmed with the same candidate in patients on cART.Citation112 A further two candidates have been tested in HIV-infected patients on cART, the first encoding env/rev and gag/pol genesCitation113 and the second encoding HIVA, an immunogen comprising HIV-1 clade A p24/p17 fused to a string of cytotoxic T-cell epitopes.Citation114 These candidates were safe but their capacity to boost virus-specific CD4 and CD8 T-cell responses was poor. Given these results, novel means of vaccine delivery have been proposed for further improvement of DNA vaccine potency. DermaVir, a single plasmid DNA (pDNA) immunogen expressing 15 HIV antigens, has been formulated in a nanoparticle with polyethylenimine-mannose and glucose, and has been developed for topical application to deliver DNA-encoded antigens into Langerhans cells.Citation115 This candidate has recently been tested in the context of structured therapy interruption in a pilot clinical trial.Citation116 Although HIV DNA immunization induces broader and higher magnitudes of HIV-specific immune responses compared with structured therapy interruptions alone, this candidate did not have any effect on the dynamics of viral rebound, set-point viral load or CD4 T-cell counts.

Viral vector vaccines

Given the poor results obtained with Remune, subunit vaccines and vaccines using DNA as the vector it has been proposed that outcomes could be improved by using viral vectors as immunogens. Live recombinant vaccines are made of a live viral or bacterial vector that is engineered to carry the genes that encode HIV antigens. The antigens are presented on the surface of the cell as externalized peptides in the context of MHC class I molecules, thus priming for CD8+ T-cell responses. The best studied vaccine vectors in humans are the pox viruses.Citation117 Vaccinia virus engineered with HIV-1 genes has been shown to induce virus-specific cellular and humoral immune responses in immunized macaques, as well as protection against simian immunodeficiency virus (SIV) infection when immunization with such constructs has been followed with boosting by recombinant proteins. However, due to the development of life-threatening disseminated vaccinia infections in immunosuppressed individuals there is a reluctance to use replication-competent vaccinia virus as a vector system in large human trials. Particular attention has, however, been focused on pox viruses with limited in vivo replicative capacity and which, therefore, are non-pathogenic in animal models and humansCitation118,Citation119; examples include MVA and NYVAC, which carry deletions of the genes associated with pathogenicity, or avian pox viruses (canary pox and fowl pox). These vectors are restricted to the early stages of virus replication in human cells but initiate protein synthesis and thus elicit specific immune responses to the recombinant antigen.Citation118,Citation120-Citation123

The best studied vector has been canary pox (ALVAC).Citation25,Citation77,Citation124-Citation128,Citation129-Citation131 The international QUEST studyCitation124 was performed on patients who started treatment during the acute phase. After a mean of two years of virological control, 79 patients were randomized to receive either immunization with ALVAC-HIV vCP1452, with ALVAC-HIV vCP1452 plus Remune, or placebo. ALVAC-HIV vCP1452 expresses HIV-1 env and gag gene products and nef and pol gene products encompassing known human HLA-A2–restricted cytotoxic T-cell (CTL) epitopes from these genes. The choice of vaccine candidates for this study was dictated by their safety records in HIV-1-infected subjects and by immunogenicity. The combination of vaccines in a single treatment arm was selected for its potential to enhance CD4 T helper cells (Remune) and CTLs (ALVAC-HIV vCP1452). After 24 weeks’ immunization, cART was interrupted. There was no difference between the groups in terms of viral rebound dynamics or viral load figures. These results confirm the findings of a previous small randomized pilot study of HIV-infected patients treated with cART during acute infection and who received ALVAC-HIV vCP1452 and recombinant gp160.Citation77,Citation125

Similar results with ALVAC-HIV vCP1452 have been obtained in chronic HIV-infected patients. The ACTG 5068 included 99 subjects receiving successful cART who were randomized to undergo continued cART, structured therapy interruptions (STIs), ALVAC-HIV vCP1452 immunization, or STIs and ALVAC-HIV vCP1452 immunization. Subjects in the two STI arms had significantly enhanced immunological control of HIV replication compared with subjects in the two arms without STIs. ALVAC-HIV vCP1452 did not affect viral load measures.Citation129 A separate placebo-controlled study conducted in chronically-infected patients suggested that the immunogenicity conferred by the ALVAC vCP1452 candidate vaccine alone could be deleterious, since it was associated with higher virus production following treatment interruption.Citation130 Further research suggested that ALVAC vCP1452 was only weakly immunogenic, it failing to elicit significant HIV-specific CD8 T-cell responses and inducing activated CD4 T-cell responses that could favor greater viral rebound after treatment interruption.Citation25 These results appear to conflict with those of a clinical trial reported recently by Angel et al.Citation131 This was a randomized, placebo-controlled, double-blind study in which effectively-treated HIV-infected individuals with high CD4 T-cell counts were randomized to receive ALVAC vCP1452 with Remune, ALVAC vCP1452 alone or placebo over 20 weeks. At week 24, participants interrupted cART. ALVAC with or without Remune did not lower the viral load set-point, although it tended to delay viral load rebound and was associated with a greater time to meet pre-set criteria to restart ART. In contrast to the findings reported by Autran et al.Citation130 and Papagno et al.Citation25 the induction of helper CD4 T-cell responses was not associated with a worse virological outcome. At all events, further research is required to determine whether the best strategy to induce viral control in chronic HIV-infected patients should be based on inducing robust and broad CD8 T-cell responses but only modestly activating CD4 T-cell responses.

The best results for a viral vector have been reported with ALVAC vCP1433 in combination with Lipo-6T vaccines, followed by three cycles of subcutaneous interleukin-2.Citation126,Citation132 ALVAC-HIV vCP1433 expresses several HIV genes for gp120, gp41, 55 polyprotein, for a portion of pol encoding the protease and for genes expressing cytotoxic T-lymphocyte peptides from pol and nef. The Lipo-6T vaccine is a mixture of the tetanus toxoid TT-830–843 class II-restricted universal CD4 epitope combined with certain peptides of Gag, Nef and Pol. Chronic HIV-infected patients were randomized to receive either cART alone (n = 37) or four doses of ALVAC-HIV vCP1433 and Lipo-6T followed by three cycles of IL-2 (n = 34). Vaccination improved the proliferative response against p24 Ag, as well as CD8 T-cell responses. After cART interruption, 24% of the Vac-IL-2 group lowered their viral set-point, compared with 5% of controls. Vaccine-elicited immunological responses were predictive of virological control. Vaccination with the same product (ALVAC-HIV vCP1433) in a French clinical trial was safe, immunogenic and associated with prolongation of treatment discontinuation.Citation128

A pilot clinical trial of a therapeutic vaccination based on a fowl pox vector and involving patients with primary infection has been reported by Emery et al.Citation133 After a mean of four years of cART, 35 patients with controlled viral replication were randomized to be vaccinated with a fowl pox vector free of HIV sequences, a vector containing gag/pol sequences, or a vector containing gag/pol sequences and a gene which encodes human interferon gamma. Surprisingly, there were few differences between the groups in terms of the presence of CTL cells, as measured by ELISPOT, or in cytolytic responses after vaccination and before interruption of treatment. Treatment was not interrupted in ten patients. There were no differences in the control of viral replication between the placebo group and the group vaccinated with gag/pol. However, patients immunized with gag/pol and interferon gamma had better control of viral replication, with a mean viral load of 0.8 log10 less than the other two groups, although the differences between groups were not significant. The absence of immune responses in the two vaccinated groups is disappointing, while the response in the interferon group is surprising and merits further investigation.

The modified vaccinia virus Ankara (MVA) was developed toward the end of the smallpox eradication campaign in the 1970s, the aim being to obtain a vaccine with a better safety profile than the ones in use at that time. MVA was dereived from vaccinia by over 570 passages in chicken embryo fibroblast cells (CEF).Citation134 The complete genomic sequence has been sequenced and has a length of 178 kb. It consists of 193 open reading frames (ORFs), corresponding to 177 genes, of which 25 are split and/or have suffered mutations resulting in truncated proteins. These numerous mutations, affecting host interactive proteins and some structural proteins, explain the attenuated phenotype of MVA.Citation119 There is clinical experience with MVA as a vaccine against smallpox in over 120,000 people.Citation118,Citation120 During extensive field studies, including with high-risk patients, no side effects were associated with the use of the MVA vaccine.Citation118,Citation120 The combination of a very good safety profile and the ability to deliver antigens in a highly immunogenic way makes MVA suitable as a vaccine vector. It can stimulate antibody and T-cell responses even in the presence of pre-existing antibodies,Citation135 and it has been shown to be effective in primates and humans for several viruses, including SIV and SHIV.Citation121,Citation136-Citation139 Harrer et al.Citation140 investigated the safety and immunogenicity of an HIV-1 nef-expressing MVA in 14 cART-treated HIV-infected patients. Vaccination with MVA-nef was safe, was associated with recognition of new CTL epitopes, and after interruption of cART viremia remained below the pre-cART viral load in 9/14 patients. In another pilot study, vaccination of 16 HIV-1-infected cART-treated individuals with MVA expressing HIV-1 gag p24, p17 and a CD8+ T-cell epitope string was not only safe but also significantly increased the frequencies of CD8+ and CD4+ T cells secreting IFN-gamma and enhanced proliferative responses to gag peptides.Citation141-Citation143

Finally, in ACTG A5197, HIV-1-infected individuals who received a recombinant adenovirus serotype 5 HIV-1 gag therapeutic vaccine had a 0.5 log10 lower PVL after a 16-week ATI, compared with those who received placebo.Citation144,Citation145

In summary, although most viral vector vaccines have been able to induce HIV-specific immune responses in clinical trials they have shown very limited efficacy in terms of controlling viral replication. New therapeutic strategies are therefore warranted.

Dendritic cell-based vaccines

Dendritic cells (DC) are the most potent professional antigen-processing and presenting cells (APC), with the unique ability to induce both primary and secondary immune responses of CD4+ and CD8+ T lymphocytes.Citation146,Citation147 A wide variety of data in experimental in vivo and in vitro systems have shown that DC are able to engulf exogenous soluble proteins, tumor cell lysates, and inactivated virus and virus-infected apoptotic cells, as well as presenting antigens not only in the MHC-class II pathway to helper CD4+ T cells (Th)Citation146 but also in the MHC-class I pathway to cytotoxic CD8+ T lymphocytes (CTL), a phenomenon also known as “cross-presentation” or “cross-priming”Citation148-Citation150.

In vitro culture of human blood monocytes (MO) with GM-CSF and IL-4 gives rise to myeloid DC.Citation151,Citation152 Some data suggest that these human myeloid DC not only activate naive CD4+ T cells but also promote their differentiation into Th1 cells, preventing the differentiation into Th2.Citation153 In addition, antigen-pulsed DC secrete antigen-presenting particles (termed exosomes) which express functional MHC-class I and II and T-cell co-stimulatory molecules, and they are able to prime CTL in vivo and eradicate or suppress the growth of established murine tumors.Citation154 Autologous DC pulsed in vitro with a variety of inactivated pathogens have also been shown to induce a potent protective immunity in murine models of human infections.Citation154-Citation158 All these data indicate that in vitro-generated DC may be the most potent cellular adjuvant for a vaccine preparation designed to induce effective Th1 and CTL responses to tumor antigens and intracellular pathogens.

It has been reported that an inactivated SIV-pulsed therapeutic dendritic-cell vaccine was able to elicit effective and lasting SIV-specific cellular and humoral immunity. This study found that after three immunizations administered at two-week intervals the SIV cellular DNA and the SIV plasma RNA levels decreased by 50-fold and 1,000-fold, respectively. This decrease in viral load was negatively correlated with SIV-specific T-cell responses. Neutralizing antibody responses also increased significantly and remained increased throughout the follow-up period.Citation159 Finally, two studies in a hu-PBL-SCID mouse model suggest that mice immunized with inactivated virus-pulsed dendritic cells elicit a potent immune response against HIV, which protects the animals from virus challenge.Citation160,Citation161

There are at least ten published clinical trials of DC immunotherapy for HIV infection in humans.Citation162-Citation169,Citation170,Citation171 Most of them found that DC immunotherapy elicited immunological responses, even though the design of the trials and the HIV antigens used to pulse DC were very different. It should be noted, however, that virological responses were only observed in four of these trials.Citation163,Citation164,Citation170,Citation171

Two of these four trials were performed in untreated patients. The first was a preliminary, non-controlled, non-randomized study that showed striking results for a MD-DC-based vaccine loaded with high doses of chemically-inactivated (with aldrithiol-2) autologous virus in untreated HIV patients.Citation163 Plasma viral load (PVL) decreased by 90% for at least one year in eight of 18 patients. This decrease in PVL was associated with strong and sustained HIV-1-specific cellular responses. The second trial was a double-blind, placebo-controlled study using the same schedule and immunizing the same type of patients (HIV-1-infected patients off cART).Citation170 Only a modest virological response, which was maintained for at least 48 weeks in the vaccinated group, was observed.Citation170 This change in VL in the immunized patients was small (ranging from 0.30 to 0.60 log10) but clinically significant when compared with the placebo group, and it was maintained up to week 48. However, this small decrease in VL in immunized patients was inversely correlated with a weak increase in HIV-1-specific CD8+ T-cell responses.

The other two trials involved antiretroviral-treated patients. The first was an open, randomized (2:1) clinical trial using heat-inactivated autologous virus in cART-treated HIV-1-infected patients, and reported partial and transient control of viral replication after interruption of antiretroviral therapy.Citation164 In this study, autologous DC vaccine pulsed ex vivo with heat-inactivated autologous HIV-1 elicited only weak Th1- and HIV-1-specific CD8+ T-cell responses. Recently, Routy et al.,Citation171 in an uncontrolled study, reported the final results of an immunotherapy regimen consisting of MD-DC electroporated with mRNA encoding autologous HIV-1 antigens (Gag, Nef, Rev, Vpr); this was administered monthly in 33 subjects in four intradermal doses in combination with cART, followed by two more doses during a 12-week cART interruption (STI). They found that eight out of 24 subjects (33%) met the criteria for the primary endpoint of three instances of viral load < 1,000 copies/mL, while 11 out of 24 subjects (46%) met the criteria for the secondary endpoint of three instances of viral load < 10,000 copies/mL. At week 12 of the STI, 16/24 subjects responded with a mean reduction in viral load of –1.2 log compared with their pre-cART viral load value. These data are impressive and are similar to those reported by Lu et al.Citation163 with HIV-1-infected patients who were vaccinated when treated with cART. These data need to be confirmed in a randomized, placebo-controlled study. A review of DC-based vaccine clinical trials has recently been published.Citation172

Conclusions

Our knowledge of the immunological control of HIV viral replication and the causes of cellular and humoral immune deterioration is limited, and we lack immunological methods able to demonstrate an efficacious immune control of HIV in vivo. The efficacy of immune therapy and therapeutic vaccines has been modest in the best of cases, although some preliminary results with dendritic cell-based vaccines seem promising. Efforts must now be redoubled in order to improve our understanding of the mechanisms of protection, virological control and immune deterioration, as without this knowledge an efficacious therapeutic vaccine remains a long way off. Given the toxicity and long-term efficacy problems associated with current drugs the search for therapeutic vaccines must be seen as a priority line of investigation.

Acknowledgments

This study was partially supported by the following grants: FIS PS09/01297, TRA-094, EC10–153, FIS PI10/02984, SAF2006–26667-E, RIS*, HIVACAT**, ORVACS***. F.G. was recipient of a research grant from IDIBAPS****, Barcelona, Spain. M.P. was supported by contract FIS 03/0072 from the Fundació Privada Clínic per a la Recerca Biomèdica, in collaboration with the Spanish Department of Health. *Red Temática Cooperativa de Grupos de Investigación en Sida del Fondo de Investigación Sanitaria (FIS). **HIVACAT: HIV development program in Catalonia. ***ORVACS: Objectif recherche vaccin sida. ****IDIBAPS: Institut d’Investigacions Biomèdiques August Pi I Sunyer.

References

  • Palella FJ, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. In: 1998. pp. 853-860.
  • Montaner JS. Treatment as prevention--a double hat-trick. Lancet 2011; 378:208 - 9; http://dx.doi.org/10.1016/S0140-6736(11)60821-0; PMID: 21763923
  • Montaner JS, Lima VD, Barrios R, Yip B, Wood E, Kerr T, et al. Association of highly active antiretroviral therapy coverage, population viral load, and yearly new HIV diagnoses in British Columbia, Canada: a population-based study. Lancet 2010; 376:532 - 9; http://dx.doi.org/10.1016/S0140-6736(10)60936-1; PMID: 20638713
  • Martinez E, Mocroft A, García-Viejo M, Pérez-Cuevas J, Blanco J, Mallolas J, et al. A prospective cohort study on the risk for lipodystrophy in HIV-1 infected patients treated with protease inhibitor-containing regimens. Lancet 2001; 357:592 - 8; http://dx.doi.org/10.1016/S0140-6736(00)04056-3; PMID: 11558485
  • Sidibé M. Newsmaker interview: Michel Sidibé. New HIV infections drop, but treatment demands rise. Interview by Jon Cohen. [Abstract] Science 2010; 330:1301; PMID: 21127221
  • García F, Ruiz L, Lopez-Bernaldo de Quirós J, Moreno S, Domingo P. INMUNOTERAPIA Y VACUNAS TERAPEUTICAS EN LA INFECCIÓN POR VIH. Enf Inf Microb Clin 2005; 23:Suppl 2 84 - 94; http://dx.doi.org/10.1016/S0213-005X(05)75164-8
  • Cohen J. The emerging race to cure HIV infections. Science 2011; 332:784 - 5, 787-9; http://dx.doi.org/10.1126/science.332.6031.784; PMID: 21566173
  • Hütter G, Nowak D, Mossner M, Ganepola S, Müssig A, Allers K, et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med 2009; 360:692 - 8; http://dx.doi.org/10.1056/NEJMoa0802905; PMID: 19213682
  • Allers K, Hütter G, Hofmann J, Loddenkemper C, Rieger K, Thiel E, et al. Evidence for the cure of HIV infection by CCR5Δ32/Δ32 stem cell transplantation. Blood 2011; 117:2791 - 9; http://dx.doi.org/10.1182/blood-2010-09-309591; PMID: 21148083
  • Buzón MJ, Massanella M, Llibre JM, Esteve A, Dahl V, Puertas MC, et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med 2010; 16:460 - 5; http://dx.doi.org/10.1038/nm.2111; PMID: 20228817
  • Kambal A, Mitchell G, Cary W, Gruenloh W, Jung Y, Kalomoiris S, et al. Generation of HIV-1 resistant and functional macrophages from hematopoietic stem cell-derived induced pluripotent stem cells. Mol Ther 2011; 19:584 - 93; http://dx.doi.org/10.1038/mt.2010.269; PMID: 21119622
  • Johnson PR, Schnepp BC, Zhang J, Connell MJ, Greene SM, Yuste E, et al. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat Med 2009; 15:901 - 6; http://dx.doi.org/10.1038/nm.1967; PMID: 19448633
  • McMichael AJ, Jones EY. Genetics. First-class control of HIV-1. Science 2010; 330:1488 - 90; http://dx.doi.org/10.1126/science.1200035; PMID: 21148380
  • Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI, Walker BD, et al, International HIV Controllers Study. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 2010; 330:1551 - 7; http://dx.doi.org/10.1126/science.1195271; PMID: 21051598
  • Autran B, Carcelain G. AIDS. Boosting immunity to HIV--can the virus help?. Science 2000; 290:946 - 9; http://dx.doi.org/10.1126/science.290.5493.946; PMID: 11184735
  • Autran B, Carcelain G, Combadiere B, Debre P. Therapeutic vaccines for chronic infections. Science 2004; 305:205 - 8; http://dx.doi.org/10.1126/science.1100600; PMID: 15247470
  • Letvin NL, Walker BD. Immunopathogenesis and immunotherapy in AIDS virus infections. Nat Med 2003; 9:861 - 6; http://dx.doi.org/10.1038/nm0703-861; PMID: 12835706
  • Barouch DH, Liu J, Li H, Maxfield LF, Abbink P, Lynch DM, et al. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys. Nature 2012; 482:89 - 93; http://dx.doi.org/10.1038/nature10766; PMID: 22217938
  • Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 2011; 473:523 - 7; http://dx.doi.org/10.1038/nature10003; PMID: 21562493
  • Rosenberg ES, Billingsley JM, Caliendo AM, Boswell SL, Sax PE, Kalams SA, et al. Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia. [see comments] Science 1997; 278:1447 - 50; http://dx.doi.org/10.1126/science.278.5342.1447; PMID: 9367954
  • Rosenberg ES, Altfeld M, Poon SH, Phillips MN, Wilkes BM, Eldridge RL, et al. Immune control of HIV-1 after early treatment of acute infection. Nature 2000; 407:523 - 6; http://dx.doi.org/10.1038/35035103; PMID: 11029005
  • Moore JP, Cao Y, Ho DD, Koup RA. Development of the anti-gp120 antibody response during seroconversion to human immunodeficiency virus type 1. J Virol 1994; 68:5142 - 55; PMID: 8035514
  • Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer M. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature 2005; 434:1093 - 7; http://dx.doi.org/10.1038/nature03501; PMID: 15793563
  • Douek DC, Brenchley JM, Betts MR, Ambrozak DR, Hill BJ, Okamoto Y, et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature 2002; 417:95 - 8; http://dx.doi.org/10.1038/417095a; PMID: 11986671
  • Papagno L, Alter G, Assoumou L, Murphy RL, Garcia F, Clotet B, et al, ORVACS Study Group. Comprehensive analysis of virus-specific T-cells provides clues for the failure of therapeutic immunization with ALVAC-HIV vaccine. AIDS 2011; 25:27 - 36; http://dx.doi.org/10.1097/QAD.0b013e328340fe55; PMID: 21076273
  • Schmitz JE, Kuroda MJ, Santra S. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. In: 1999. pp. 857-860.
  • Metzner KJ, Jin X, Lee FV, Gettie A, Bauer DE, Di Mascio M, et al. Effects of in vivo CD8(+) T cell depletion on virus replication in rhesus macaques immunized with a live, attenuated simian immunodeficiency virus vaccine. J Exp Med 2000; 191:1921 - 31; http://dx.doi.org/10.1084/jem.191.11.1921; PMID: 10839807
  • de Quiros JC, Shupert WL, McNeil AC, Gea-Banacloche JC, Flanigan M, Savage A, et al. Resistance to replication of human immunodeficiency virus challenge in SCID-Hu mice engrafted with peripheral blood mononuclear cells of nonprogressors is mediated by CD8(+) T cells and associated with a proliferative response to p24 antigen. J Virol 2000; 74:2023 - 8; http://dx.doi.org/10.1128/JVI.74.4.2023-2028.2000; PMID: 10644376
  • Day CL, Walker BD. Progress in defining CD4 helper cell responses in chronic viral infections. J Exp Med 2003; 198:1773 - 7; http://dx.doi.org/10.1084/jem.20031947; PMID: 14676292
  • Hel Z, Nacsa J, Tryniszewska E, Tsai WP, Parks RW, Montefiori DC, et al. Containment of simian immunodeficiency virus infection in vaccinated macaques: correlation with the magnitude of virus-specific pre- and postchallenge CD4+ and CD8+ T cell responses. J Immunol 2002; 169:4778 - 87; PMID: 12391187
  • Kaech SM, Ahmed R. Immunology. CD8 T cells remember with a little help. Science 2003; 300:263 - 5; http://dx.doi.org/10.1126/science.1084511; PMID: 12690179
  • Nishimura Y, Igarashi T, Haigwood NL, Sadjadpour R, Donau OK, Buckler C, et al. Transfer of neutralizing IgG to macaques 6 h but not 24 h after SHIV infection confers sterilizing protection: implications for HIV-1 vaccine development. Proc Natl Acad Sci U S A 2003; 100:15131 - 6; http://dx.doi.org/10.1073/pnas.2436476100; PMID: 14627745
  • Richman DD, Wrin T, Little SJ, Petropoulos CJ. Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proc Natl Acad Sci U S A 2003; 100:4144 - 9; http://dx.doi.org/10.1073/pnas.0630530100; PMID: 12644702
  • Gao X, Bashirova A, Iversen AK, Phair J, Goedert JJ, Buchbinder S, et al. AIDS restriction HLA allotypes target distinct intervals of HIV-1 pathogenesis. Nat Med 2005; 11:1290 - 2; http://dx.doi.org/10.1038/nm1333; PMID: 16288280
  • Allen TM, Altfeld M, Geer SC, Kalife ET, Moore C, O’sullivan KM, et al. Selective escape from CD8+ T-cell responses represents a major driving force of human immunodeficiency virus type 1 (HIV-1) sequence diversity and reveals constraints on HIV-1 evolution. J Virol 2005; 79:13239 - 49; http://dx.doi.org/10.1128/JVI.79.21.13239-13249.2005; PMID: 16227247
  • Allen TM, Yu XG, Kalife ET, Reyor LL, Lichterfeld M, John M, et al. De novo generation of escape variant-specific CD8+ T-cell responses following cytotoxic T-lymphocyte escape in chronic human immunodeficiency virus type 1 infection. J Virol 2005; 79:12952 - 60; http://dx.doi.org/10.1128/JVI.79.20.12952-12960.2005; PMID: 16188997
  • Feeney ME, Tang Y, Pfafferott K, Roosevelt KA, Draenert R, Trocha A, et al. HIV-1 viral escape in infancy followed by emergence of a variant-specific CTL response. J Immunol 2005; 174:7524 - 30; PMID: 15944251
  • Altfeld M, Allen TM, Kalife ET, Frahm N, Addo MM, Mothe BR, et al. The majority of currently circulating human immunodeficiency virus type 1 clade B viruses fail to prime cytotoxic T-lymphocyte responses against an otherwise immunodominant HLA-A2-restricted epitope: implications for vaccine design. J Virol 2005; 79:5000 - 5; http://dx.doi.org/10.1128/JVI.79.8.5000-5005.2005; PMID: 15795285
  • Allen TM, Altfeld M, Yu XG, O’Sullivan KM, Lichterfeld M, Le Gall S, et al. Selection, transmission, and reversion of an antigen-processing cytotoxic T-lymphocyte escape mutation in human immunodeficiency virus type 1 infection. J Virol 2004; 78:7069 - 78; http://dx.doi.org/10.1128/JVI.78.13.7069-7078.2004; PMID: 15194783
  • Draenert R, Le Gall S, Pfafferott KJ, Leslie AJ, Chetty P, Brander C, et al. Immune selection for altered antigen processing leads to cytotoxic T lymphocyte escape in chronic HIV-1 infection. J Exp Med 2004; 199:905 - 15; http://dx.doi.org/10.1084/jem.20031982; PMID: 15067030
  • Leslie AJ, Pfafferott KJ, Chetty P, Draenert R, Addo MM, Feeney M, et al. HIV evolution: CTL escape mutation and reversion after transmission. Nat Med 2004; 10:282 - 9; http://dx.doi.org/10.1038/nm992; PMID: 14770175
  • Goulder PJ, Brander C, Tang Y, Tremblay C, Colbert RA, Addo MM, et al. Evolution and transmission of stable CTL escape mutations in HIV infection. Nature 2001; 412:334 - 8; http://dx.doi.org/10.1038/35085576; PMID: 11460164
  • Leslie AJ, Pfafferott KJ, Chetty P, Draenert R, Addo MM, Feeney M, et al. HIV evolution: CTL escape mutation and reversion after transmission. Nat Med 2004; 10:282 - 9; http://dx.doi.org/10.1038/nm992; PMID: 14770175
  • Pitcher CJ, Quittner C, Peterson DM, Connors M, Koup RA, Maino VC, et al. HIV-1-specific CD4+ T cells are detectable in most individuals with active HIV-1 infection, but decline with prolonged viral suppression. Nat Med 1999; 5:518 - 25; http://dx.doi.org/10.1038/8400; PMID: 10229228
  • Plana M, Garcia F, Gallart MT, Miro JM, Gatell JM. Lack of T-cell proliferative response to HIV-1 antigens after one year of HAART in very early HIV-1 disease. Lancet 1998; 352:1194 - 5; http://dx.doi.org/10.1016/S0140-6736(05)60532-6; PMID: 9777842
  • Yokomaku Y, Miura H, Tomiyama H, Kawana-Tachikawa A, Takiguchi M, Kojima A, et al. Impaired processing and presentation of CTL epitopes are major escape mechanisms from CTL immune pressure in Human Immunodeficiency Virus type 1 infection. J Virol 2004; 78:1324 - 32; http://dx.doi.org/10.1128/JVI.78.3.1324-1332.2004; PMID: 14722287
  • Seder RA, Ahmed R. Similarities and differences in CD4+ and CD8+ effector and memory T cell generation. Nat Immunol 2003; 4:835 - 42; http://dx.doi.org/10.1038/ni969; PMID: 12942084
  • Yang OO, Sarkis PT, Trocha A, Kalams SA, Johnson RP, Walker BD. Impacts of avidity and specificity on the antiviral efficiency of HIV-1-specific CTL. J Immunol 2003; 171:3718 - 24; PMID: 14500671
  • Donaghy H, Gazzard B, Gotch F, Patterson S. Dysfunction and infection of freshly isolated blood myeloid and plasmacytoid dendritic cells in patients infected with HIV-1. Blood 2003; 101:4505 - 11; http://dx.doi.org/10.1182/blood-2002-10-3189; PMID: 12576311
  • Chehimi J, Campbell DE, Azzoni L, Bacheller D, Papasavvas E, Jerandi G, et al. Persistent decreases in blood plasmacytoid dendritic cell number and function despite effective highly active antiretroviral therapy and increased blood myeloid dendritic cells in HIV-infected individuals. J Immunol 2002; 168:4796 - 801; PMID: 11971031
  • McNeil AC, Shupert WL, Iyasere CA, Hallahan CW, Mican JA, Davey RT Jr., et al. High-level HIV-1 viremia suppresses viral antigen-specific CD4(+) T cell proliferation. Proc Natl Acad Sci U S A 2001; 98:13878 - 83; http://dx.doi.org/10.1073/pnas.251539598; PMID: 11717444
  • McKay PF, Barouch DH, Schmitz JE, Veazey RS, Gorgone DA, Lifton MA, et al. Global dysfunction of CD4 T-lymphocyte cytokine expression in simian-human immunodeficiency virus/SIV-infected monkeys is prevented by vaccination. J Virol 2003; 77:4695 - 702; http://dx.doi.org/10.1128/JVI.77.8.4695-4702.2003; PMID: 12663776
  • Autran B, Carcelain G, Li TS, Blanc C, Mathez D, Tubiana R, et al. Positive effects of combined antiretroviral therapy on CD4+ T cell homeostasis and function in advanced HIV disease. [see comments] Science 1997; 277:112 - 6; http://dx.doi.org/10.1126/science.277.5322.112; PMID: 9204894
  • Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD. HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and viral generation time. Science 1996; 271:1582 - 6; http://dx.doi.org/10.1126/science.271.5255.1582; PMID: 8599114
  • Perelson AS, Essunger P, Cao Y. Decay characteristics of HIV-1 infected compartments during combination therapy. In: 1997. pp. 188-191.
  • Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 1999; 5:512 - 7; http://dx.doi.org/10.1038/8394; PMID: 10229227
  • Blankson JN, Persaud D, Siliciano RF. The challenge of viral reservoirs in HIV-1 infection. Annu Rev Med 2002; 53:557 - 93; http://dx.doi.org/10.1146/annurev.med.53.082901.104024; PMID: 11818490
  • Zhang L, Ramratnam B, Tenner-Racz K, He Y, Vesanen M, Lewin S, et al. Quantifying residual HIV-1 replication in patients receiving combination antiretroviral therapy. N Engl J Med 1999; 340:1605 - 13; http://dx.doi.org/10.1056/NEJM199905273402101; PMID: 10341272
  • Furtado MR, Callaway DS, Phair JP, Kunstman KJ, Stanton JL, Macken CA, et al. Persistence of HIV-1 transcription in peripheral-blood mononuclear cells in patients receiving potent antiretroviral therapy. [see comments] N Engl J Med 1999; 340:1614 - 22; http://dx.doi.org/10.1056/NEJM199905273402102; PMID: 10341273
  • Gamberg J, Barrett L, Bowmer MI, Howley C, Grant M. Factors related to loss of HIV-specific cytotoxic T lymphocyte activity. AIDS 2004; 18:597 - 604; http://dx.doi.org/10.1097/00002030-200403050-00003; PMID: 15090764
  • Lange CG, Xu Z, Patterson BK, Medvik K, Harnisch B, Asaad R, et al. Proliferation responses to HIVp24 during antiretroviral therapy do not reflect improved immune phenotype or function. AIDS 2004; 18:605 - 13; http://dx.doi.org/10.1097/00002030-200403050-00004; PMID: 15090765
  • García F, Plana M, Vidal C, Cruceta A, O’Brien WA, Pantaleo G, et al. Dynamics of viral load rebound and immunological changes after stopping effective antiretroviral therapy. AIDS 1999; 13:F79 - 86; http://dx.doi.org/10.1097/00002030-199907300-00002; PMID: 10449278
  • Ruiz L, Martinez-Picado J, Romeu J, Paredes R, Zayat MK, Marfil S, et al. Structured treatment interruption in chronically HIV-1 infected patients after long-term viral suppression. [Abstract 354 ] [Abstract] AIDS 2000; 14:397 - 403; http://dx.doi.org/10.1097/00002030-200003100-00013; PMID: 10770542
  • Plana M, García F, Gallart T, Tortajada C, Soriano A, Palou E, et al. Immunological benefits of antiretroviral therapy in very early stages of asymptomatic chronic HIV-1 infection. AIDS 2000; 14:1921 - 33; http://dx.doi.org/10.1097/00002030-200009080-00007; PMID: 10997396
  • Reekie J, Gatell JM, Yust I, Bakowska E, Rakhmanova A, Losso M, et al, EuroSIDA in EuroCoord. Fatal and nonfatal AIDS and non-AIDS events in HIV-1-positive individuals with high CD4 cell counts according to viral load strata. AIDS 2011; 25:2259 - 68; http://dx.doi.org/10.1097/QAD.0b013e32834cdb4b; PMID: 21918422
  • Mocroft A, Reiss P, Gasiorowski J, Ledergerber B, Kowalska J, Chiesi A, et al, EuroSIDA Study Group. Serious fatal and nonfatal non-AIDS-defining illnesses in Europe. J Acquir Immune Defic Syndr 2010; 55:262 - 70; http://dx.doi.org/10.1097/QAI.0b013e3181e9be6b; PMID: 20700060
  • Lodwick RK, Sabin CA, Porter K, Ledergerber B, van Sighem A, Cozzi-Lepri A, et al, Study Group on Death Rates at High CD4 Count in Antiretroviral Naive Patients. Death rates in HIV-positive antiretroviral-naive patients with CD4 count greater than 350 cells per microL in Europe and North America: a pooled cohort observational study. Lancet 2010; 376:340 - 5; http://dx.doi.org/10.1016/S0140-6736(10)60932-4; PMID: 20638118
  • Kitahata MM, Gange SJ, Abraham AG, Merriman B, Saag MS, Justice AC, et al, NA-ACCORD Investigators. Effect of early versus deferred antiretroviral therapy for HIV on survival. N Engl J Med 2009; 360:1815 - 26; http://dx.doi.org/10.1056/NEJMoa0807252; PMID: 19339714
  • El-Sadr WM, Lundgren JD, Neaton JD, Gordin F, Abrams D, Arduino RC, et al, Strategies for Management of Antiretroviral Therapy (SMART) Study Group. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med 2006; 355:2283 - 96; http://dx.doi.org/10.1056/NEJMoa062360; PMID: 17135583
  • Kahn JO, Walker BD. Current concepts: acute human immunodeficiency virus type 1 infection. In: 1998. pp. 33-39.
  • Mocroft A, Bofill M, Lipman M, Medina E, Borthwick N, Timms A, et al. CD8+,CD38+ lymphocyte percent: a useful immunological marker for monitoring HIV-1-infected patients. J Acquir Immune Defic Syndr Hum Retrovirol 1997; 14:158 - 62; http://dx.doi.org/10.1097/00042560-199702010-00009; PMID: 9052725
  • Bofill M, Mocroft A, Lipman M, Medina E, Borthwick NJ, Sabin CA, et al. Increased numbers of primed activated CD8+CD38+CD45RO+ T cells predict the decline of CD4+ T cells in HIV-1-infected patients. AIDS 1996; 10:827 - 34; http://dx.doi.org/10.1097/00002030-199607000-00005; PMID: 8828739
  • Diaz A, García F, Mozos A, Caballero M, León A, Martinez A, et al. Lymphoid tissue collagen deposition in HIV-infected patients correlates with the imbalance between matrix metalloproteinases and their inhibitors. J Infect Dis 2011; 203:810 - 3; http://dx.doi.org/10.1093/infdis/jiq129; PMID: 21343147
  • Diaz A, Alós L, León A, Mozos A, Caballero M, Martinez A, et al, Study Group of Lymphoid Tissue immunopathogenesis in HIV infection. Factors associated with collagen deposition in lymphoid tissue in long-term treated HIV-infected patients. AIDS 2010; 24:2029 - 39; http://dx.doi.org/10.1097/QAD.0b013e32833c3268; PMID: 20588162
  • Garrido M, Mozos A, Martínez A, García F, Serafín A, Morente V, et al. HIV-1 upregulates intercellular adhesion molecule-1 gene expression in lymphoid tissue of patients with chronic HIV-1 infection. J Acquir Immune Defic Syndr 2007; 46:268 - 74; http://dx.doi.org/10.1097/QAI.0b013e318142c74c; PMID: 17786132
  • Mozos A, Garrido M, Carreras J, Plana M, Diaz A, Alos L, et al. Redistribution of FOXP3-positive regulatory T cells from lymphoid tissues to peripheral blood in HIV-infected patients. J Acquir Immune Defic Syndr 2007; 46:529 - 37; http://dx.doi.org/10.1097/QAI.0b013e31815b69ae; PMID: 18193494
  • Markowitz M, Jin X, Hurley A, Simon V, Ramratnam B, Louie M, et al. Discontinuation of antiretroviral therapy commenced early during the course of human immunodeficiency virus type 1 infection, with or without adjunctive vaccination. J Infect Dis 2002; 186:634 - 43; http://dx.doi.org/10.1086/342559; PMID: 12195350
  • MacGregor RR, Ginsberg R, Ugen KE, Baine Y, Kang CU, Tu XM, et al. T-cell responses induced in normal volunteers immunized with a DNA-based vaccine containing HIV-1 env and rev. AIDS 2002; 16:2137 - 43; http://dx.doi.org/10.1097/00002030-200211080-00005; PMID: 12409734
  • MacGregor RR, Boyer JD, Ciccarelli RB, Ginsberg RS, Weiner DB. Safety and immune responses to a DNA-based human immunodeficiency virus (HIV) type I env/rev vaccine in HIV-infected recipients: follow-up data. J Infect Dis 2000; 181:406; http://dx.doi.org/10.1086/315199; PMID: 10608800
  • Schooley RT, Spino C, Kuritzkes D, Walker BD, Valentine FA, Hirsch MS, et al. Two 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 and 214. J Infect Dis 2000; 182:1357 - 64; http://dx.doi.org/10.1086/315860; PMID: 11023459
  • Smith D, Gow I, Colebunders R, Weller I, Tchamouroff S, Weber J, et al, 006 Study Group. Therapeutic vaccination (p24-VLP) of patients with advanced HIV-1 infection in the pre-HAART era does not alter CD4 cell decline. HIV Med 2001; 2:272 - 5; http://dx.doi.org/10.1046/j.1468-1293.2001.00080.x; PMID: 11737409
  • Trauger RJ, Daigle AE, Giermakowska W, Moss RB, Jensen F, Carlo DJ. Safety and immunogenicity of a gp120-depleted, inactivated HIV-1 immunogen: results of a double-blind, adjuvant controlled trial. J Acquir Immune Defic Syndr Hum Retrovirol 1995; 10:Suppl 2 S74 - 82; PMID: 7552517
  • Eron JJ Jr., Ashby MA, Giordano MF, Chernow M, Reiter WM, Deeks SG, et al. Randomised trial of MNrgp120 HIV-1 vaccine in symptomless HIV-1 infection. Lancet 1996; 348:1547 - 51; http://dx.doi.org/10.1016/S0140-6736(96)05283-X; PMID: 8950881
  • Sandström E, Wahren B. Therapeutic immunisation with recombinant gp160 in HIV-1 infection: a randomised double-blind placebo-controlled trial. Nordic VAC-04 Study Group. Lancet 1999; 353:1735 - 42; http://dx.doi.org/10.1016/S0140-6736(98)06493-9; PMID: 10347985
  • Peters BS, Cheingsong-Popov R, Callow D, Foxall R, Patou G, Hodgkin K, et al. A pilot phase II study of the safety and immunogenicity of HIV p17/p24:VLP (p24-VLP) in asymptomatic HIV seropositive subjects. J Infect 1997; 35:231 - 5; http://dx.doi.org/10.1016/S0163-4453(97)92814-0; PMID: 9459393
  • MacGregor RR, Boyer JD, Ugen KE, Lacy KE, Gluckman SJ, Bagarazzi ML, et al. First human trial of a DNA-based vaccine for treatment of human immunodeficiency virus type 1 infection: safety and host response. J Infect Dis 1998; 178:92 - 100; PMID: 9652427
  • Robbins GK, Addo MM, Troung H, Rathod A, Habeeb K, Davis B, et al. Augmentation of HIV-1-specific T helper cell responses in chronic HIV-1 infection by therapeutic immunization. AIDS 2003; 17:1121 - 6; http://dx.doi.org/10.1097/00002030-200305230-00002; PMID: 12819512
  • Lederman MM, Douek DC. Sometimes help may not be enough. AIDS 2003; 17:1249 - 51; http://dx.doi.org/10.1097/00002030-200305230-00018; PMID: 12819528
  • Kahn JO, Cherng DW, Mayer K, Murray H, Lagakos S. Evaluation of HIV-1 immunogen, an immunologic modifier, administered to patients infected with HIV having 300 to 549 x 10(6)/L CD4 cell counts: A randomized controlled trial. JAMA 2000; 284:2193 - 202; http://dx.doi.org/10.1001/jama.284.17.2193; PMID: 11056590
  • Lichterfeld M, Kaufmann DE, Yu XG, Mui SK, Addo MM, Johnston MN, et al. Loss of HIV-1-specific CD8+ T cell proliferation after acute HIV-1 infection and restoration by vaccine-induced HIV-1-specific CD4+ T cells. J Exp Med 2004; 200:701 - 12; http://dx.doi.org/10.1084/jem.20041270; PMID: 15381726
  • Gorse GJ, Simionescu RE, Patel GB. Cellular immune responses in asymptomatic human immunodeficiency virus type 1 (HIV-1) infection and effects of vaccination with recombinant envelope glycoprotein of HIV-1. Clin Vaccine Immunol 2006; 13:26 - 32; http://dx.doi.org/10.1128/CVI.13.1.26-32.2006; PMID: 16425996
  • Birx DL, Loomis-Price LD, Aronson N, Brundage J, Davis C, Deyton L, et al. Efficacy testing of recombinant human immunodeficiency virus (HIV) gp160 as a therapeutic vaccine in early-stage HIV-1-infected volunteers. rgp160 Phase II Vaccine Investigators. J Infect Dis 2000; 181:881 - 9; http://dx.doi.org/10.1086/315308; PMID: 10720508
  • Eron JJ Jr., Ashby MA, Giordano MF, Chernow M, Reiter WM, Deeks SG, et al. Randomised trial of MNrgp120 HIV-1 vaccine in symptomless HIV-1 infection. Lancet 1996; 348:1547 - 51; http://dx.doi.org/10.1016/S0140-6736(96)05283-X; PMID: 8950881
  • Pontesilli O, Guerra EC, Ammassari A, Tomino C, Carlesimo M, Antinori A, et al. Phase II controlled trial of post-exposure immunization with recombinant gp160 versus antiretroviral therapy in asymptomatic HIV-1-infected adults. VaxSyn Protocol Team. AIDS 1998; 12:473 - 80; http://dx.doi.org/10.1097/00002030-199805000-00008; PMID: 9543445
  • Sitz KV, Ratto-Kim S, Hodgkins AS, Robb ML, Birx DL. Proliferative responses to human immunodeficiency virus type 1 (HIV-1) gp120 peptides in HIV-1-infected individuals immunized with HIV-1 rgp120 or rgp160 compared with nonimmunized and uninfected controls. J Infect Dis 1999; 179:817 - 24; http://dx.doi.org/10.1086/314685; PMID: 10068576
  • Redfield RR, Birx DL, Ketter N, Tramont E, Polonis V, Davis C, et al, Military Medical Consortium for Applied Retroviral Research. A phase I evaluation of the safety and immunogenicity of vaccination with recombinant gp160 in patients with early human immunodeficiency virus infection. N Engl J Med 1991; 324:1677 - 84; http://dx.doi.org/10.1056/NEJM199106133242401; PMID: 1674589
  • Schooley RT, Spino C, Kuritzkes D, Walker BD, Valentine FA, Hirsch MS, et al. Two 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 and 214. J Infect Dis 2000; 182:1357 - 64; http://dx.doi.org/10.1086/315860; PMID: 11023459
  • Valentine FT, Kundu S, Haslett PA, Katzenstein D, Beckett L, Spino C, et al. A 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). J Infect Dis 1996; 173:1336 - 46; http://dx.doi.org/10.1093/infdis/173.6.1336; PMID: 8648205
  • Wahren B, Bratt G, Persson C, Levén B, Hinkula J, Gilljam G, et al. Improved cell-mediated immune responses in HIV-1-infected asymptomatic individuals after immunization with envelope glycoprotein gp160. J Acquir Immune Defic Syndr 1994; 7:220 - 9; PMID: 7906300
  • Wright PF, Lambert JS, Gorse GJ, Hsieh RH, McElrath MJ, Weinhold K, et al. Immunization with envelope MN rgp120 vaccine in human immunodeficiency virus-infected pregnant women. J Infect Dis 1999; 180:1080 - 8; http://dx.doi.org/10.1086/314985; PMID: 10479134
  • Kran AM, Sommerfelt MA, Sørensen B, Nyhus J, Baksaas I, Bruun JN, et al. Reduced viral burden amongst high responder patients following HIV-1 p24 peptide-based therapeutic immunization. Vaccine 2005; 23:4011 - 5; http://dx.doi.org/10.1016/j.vaccine.2005.03.010; PMID: 15963359
  • Kran AM, Sørensen B, Sommerfelt MA, Nyhus J, Baksaas I, Kvale D. Long-term HIV-specific responses and delayed resumption of antiretroviral therapy after peptide immunization targeting dendritic cells. AIDS 2006; 20:627 - 30; http://dx.doi.org/10.1097/01.aids.0000210620.75707.ac; PMID: 16470131
  • Kran AM, Sørensen B, Nyhus J, Sommerfelt MA, Baksaas I, Bruun JN, et al. HLA- and dose-dependent immunogenicity of a peptide-based HIV-1 immunotherapy candidate (Vacc-4x). AIDS 2004; 18:1875 - 83; http://dx.doi.org/10.1097/00002030-200409240-00003; PMID: 15353973
  • Ensoli B, Bellino S, Tripiciano A, Longo O, Francavilla V, Marcotullio S, et al. Therapeutic immunization with HIV-1 Tat reduces immune activation and loss of regulatory T-cells and improves immune function in subjects on HAART. PLoS One 2010; 5:e13540; http://dx.doi.org/10.1371/journal.pone.0013540; PMID: 21085635
  • Longo O, Tripiciano A, Fiorelli V, Bellino S, Scoglio A, Collacchi B, et al. Phase I therapeutic trial of the HIV-1 Tat protein and long term follow-up. Vaccine 2009; 27:3306 - 12; http://dx.doi.org/10.1016/j.vaccine.2009.01.090; PMID: 19208456
  • Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R, Narciso P, et al. The therapeutic phase I trial of the recombinant native HIV-1 Tat protein. AIDS 2008; 22:2207 - 9; http://dx.doi.org/10.1097/QAD.0b013e32831392d4; PMID: 18832884
  • Lichterfeld M, Gandhi RT, Simmons RP, Flynn T, Sbrolla A, Yu XG, et al. Induction of Strong HIV-1-specific CD4+ T Cell Responses using an HIV-1 gp120/NefTat Vaccine adjuvanted with AS02A in ARV Treated HIV-1-Infected Individuals. J Acquir Immune Defic Syndr 2011;
  • Cafaro A, Titti F, Fracasso C, Maggiorella MT, Baroncelli S, Caputo A, et al. Vaccination with DNA containing tat coding sequences and unmethylated CpG motifs protects cynomolgus monkeys upon infection with simian/human immunodeficiency virus (SHIV89.6P). Vaccine 2001; 19:2862 - 77; http://dx.doi.org/10.1016/S0264-410X(01)00002-0; PMID: 11282197
  • Allen TM, O’Connor DH, Jing P, Dzuris JL, Mothé BR, Vogel TU, et al. Tat-specific cytotoxic T lymphocytes select for SIV escape variants during resolution of primary viraemia. Nature 2000; 407:386 - 90; http://dx.doi.org/10.1038/35036559; PMID: 11014195
  • Fernandez-Cruz E, Moreno S, Navarro J, Clotet B, Bouza E, Carbone J, et al, STIR-2102 TEAM. Therapeutic immunization with an inactivated HIV-1 Immunogen plus antiretrovirals versus antiretroviral therapy alone in asymptomatic HIV-infected subjects. Vaccine 2004; 22:2966 - 73; http://dx.doi.org/10.1016/j.vaccine.2004.03.040; PMID: 15297045
  • Calarota S, Bratt G, Nordlund S, Hinkula J, Leandersson AC, Sandström E, et al. Cellular cytotoxic response induced by DNA vaccination in HIV-1-infected patients. Lancet 1998; 351:1320 - 5; http://dx.doi.org/10.1016/S0140-6736(97)09440-3; PMID: 9643795
  • Hejdeman B, Boström AC, Matsuda R, Calarota S, Lenkei R, Fredriksson EL, et al. DNA immunization with HIV early genes in HIV type 1-infected patients on highly active antiretroviral therapy. AIDS Res Hum Retroviruses 2004; 20:860 - 70; http://dx.doi.org/10.1089/0889222041725190; PMID: 15320990
  • MacGregor RR, Boyer JD, Ugen KE, Tebas P, Higgins TJ, Baine Y, et al. Plasmid vaccination of stable HIV-positive subjects on antiviral treatment results in enhanced CD8 T-cell immunity and increased control of viral “blips”. Vaccine 2005; 23:2066 - 73; http://dx.doi.org/10.1016/j.vaccine.2005.01.010; PMID: 15755572
  • Dorrell L, Yang H, Iversen AK, Conlon C, Suttill A, Lancaster M, et al. Therapeutic immunization of highly active antiretroviral therapy-treated HIV-1-infected patients: safety and immunogenicity of an HIV-1 gag/poly-epitope DNA vaccine. AIDS 2005; 19:1321 - 3; http://dx.doi.org/10.1097/01.aids.0000180104.65640.16; PMID: 16052088
  • Lisziewicz J, Trocio J, Xu J, Whitman L, Ryder A, Bakare N, et al. Control of viral rebound through therapeutic immunization with DermaVir. AIDS 2005; 19:35 - 43; http://dx.doi.org/10.1097/00002030-200501030-00004; PMID: 15627031
  • Gudmundsdotter L, Wahren B, Haller BK, Boberg A, Edbäck U, Bernasconi D, et al. Amplified antigen-specific immune responses in HIV-1 infected individuals in a double blind DNA immunization and therapy interruption trial. Vaccine 2011; 29:5558 - 66; http://dx.doi.org/10.1016/j.vaccine.2011.01.064; PMID: 21300092
  • Pantaleo G, Esteban M, Jacobs B, Tartaglia J. Poxvirus vector-based HIV vaccines. Curr Opin HIV AIDS 2010; 5:391 - 6; http://dx.doi.org/10.1097/COH.0b013e32833d1e87; PMID: 20978379
  • Stickl H, Hochstein-Mintzel V, Mayr A, Huber HC, Schäfer H, Holzner A. [MVA vaccination against smallpox: clinical tests with an attenuated live vaccinia virus strain (MVA) (author’s transl)]. Dtsch Med Wochenschr 1974; 99:2386 - 92; http://dx.doi.org/10.1055/s-0028-1108143; PMID: 4426258
  • Antoine G, Scheiflinger F, Dorner F, Falkner FG. The complete genomic sequence of the modified vaccinia Ankara strain: comparison with other orthopoxviruses. Virology 1998; 244:365 - 96; http://dx.doi.org/10.1006/viro.1998.9123; PMID: 9601507
  • Stickl HA. Smallpox vaccination and its consequences: first experiences with the highly attenuated smallpox vaccine “MVA”. Prev Med 1974; 3:97 - 101; http://dx.doi.org/10.1016/0091-7435(74)90066-8; PMID: 4205586
  • Mwau M, Cebere I, Sutton J, Chikoti P, Winstone N, Wee EG, et al. A human immunodeficiency virus 1 (HIV-1) clade A vaccine in clinical trials: stimulation of HIV-specific T-cell responses by DNA and recombinant modified vaccinia virus Ankara (MVA) vaccines in humans. J Gen Virol 2004; 85:911 - 9; http://dx.doi.org/10.1099/vir.0.19701-0; PMID: 15039533
  • Cebere I, Dorrell L, McShane H, Simmons A, McCormack S, Schmidt C, et al. Phase I clinical trial safety of DNA- and modified virus Ankara-vectored human immunodeficiency virus type 1 (HIV-1) vaccines administered alone and in a prime-boost regime to healthy HIV-1-uninfected volunteers. Vaccine 2006; 24:417 - 25; http://dx.doi.org/10.1016/j.vaccine.2005.08.041; PMID: 16176847
  • Goonetilleke N, Moore S, Dally L, Winstone N, Cebere I, Mahmoud A, et al. Induction of multifunctional human immunodeficiency virus type 1 (HIV-1)-specific T cells capable of proliferation in healthy subjects by using a prime-boost regimen of DNA- and modified vaccinia virus Ankara-vectored vaccines expressing HIV-1 Gag coupled to CD8+ T-cell epitopes. J Virol 2006; 80:4717 - 28; http://dx.doi.org/10.1128/JVI.80.10.4717-4728.2006; PMID: 16641265
  • Kinloch-de Loes S, Hoen B, Smith DE, Autran B, Lampe FC, Phillips AN, et al, QUEST Study Group. Impact of therapeutic immunization on HIV-1 viremia after discontinuation of antiretroviral therapy initiated during acute infection. J Infect Dis 2005; 192:607 - 17; http://dx.doi.org/10.1086/432002; PMID: 16028129
  • Jin X, Ramanathan M Jr., Barsoum S, Deschenes GR, Ba L, Binley J, et al. Safety and immunogenicity of ALVAC vCP1452 and recombinant gp160 in newly human immunodeficiency virus type 1-infected patients treated with prolonged highly active antiretroviral therapy. J Virol 2002; 76:2206 - 16; http://dx.doi.org/10.1128/jvi.76.5.2206-2216.2002; PMID: 11836398
  • Lévy Y, Gahéry-Ségard H, Durier C, Lascaux AS, Goujard C, Meiffrédy V, et al, ANRS 093 Study Group. Immunological and virological efficacy of a therapeutic immunization combined with interleukin-2 in chronically HIV-1 infected patients. AIDS 2005; 19:279 - 86; PMID: 15718838
  • Johnson DC, McFarland EJ, Muresan P, Fenton T, McNamara J, Read JS, et al. Safety and immunogenicity of an HIV-1 recombinant canarypox vaccine in newborns and infants of HIV-1-infected women. J Infect Dis 2005; 192:2129 - 33; http://dx.doi.org/10.1086/498163; PMID: 16288378
  • Tubiana R, Carcelain G, Vray M, Gourlain K, Dalban C, Chermak A, et al, for the Vacciter Study group. Therapeutic immunization with a human immunodeficiency virus (HIV) type 1-recombinant canarypox vaccine in chronically HIV-infected patients: The Vacciter Study (ANRS 094). Vaccine 2005; 23:4292 - 301; http://dx.doi.org/10.1016/j.vaccine.2005.04.013; PMID: 15927325
  • Jacobson JM, Pat Bucy R, Spritzler J, Saag MS, Eron JJ Jr., Coombs RW, et al, National Institute of Allergy and Infectious Diseases-AIDS Clinical Trials Group 5068 Protocol Team. Evidence that intermittent structured treatment interruption, but not immunization with ALVAC-HIV vCP1452, promotes host control of HIV replication: the results of AIDS Clinical Trials Group 5068. J Infect Dis 2006; 194:623 - 32; http://dx.doi.org/10.1086/506364; PMID: 16897661
  • Autran B, Murphy RL, Costagliola D, Tubiana R, Clotet B, Gatell J, et al, ORVACS Study Group. Greater viral rebound and reduced time to resume antiretroviral therapy after therapeutic immunization with the ALVAC-HIV vaccine (vCP1452). AIDS 2008; 22:1313 - 22; http://dx.doi.org/10.1097/QAD.0b013e3282fdce94; PMID: 18580611
  • Angel JB, Routy JP, Tremblay C, Ayers D, Woods R, Singer J, et al. A randomized controlled trial of HIV therapeutic vaccination using ALVAC with or without Remune. AIDS 2011; 25:731 - 9; http://dx.doi.org/10.1097/QAD.0b013e328344cea5; PMID: 21330911
  • Lévy Y, Durier C, Lascaux AS, Meiffrédy V, Gahéry-Ségard H, Goujard C, et al, ANRS 093 Study Group. Sustained control of viremia following therapeutic immunization in chronically HIV-1-infected individuals. AIDS 2006; 20:405 - 13; http://dx.doi.org/10.1097/01.aids.0000206504.09159.d3; PMID: 16439874
  • Emery S, Kelleher AD, Workman C, Puls RL, Bloch M, Baker D, et al. Influence of IFNgamma co-expression on the safety and antiviral efficacy of recombinant fowlpox virus HIV therapeutic vaccines following interruption of antiretroviral therapy. Hum Vaccin 2007; 3:260 - 7; PMID: 18340117
  • Stickl H, Hochstein-Mintzel V, Mayr A, Huber HC, Schäfer H, Holzner A. [MVA vaccination against smallpox: clinical tests with an attenuated live vaccinia virus strain (MVA) (author’s transl)]. Dtsch Med Wochenschr 1974; 99:2386 - 92; http://dx.doi.org/10.1055/s-0028-1108143; PMID: 4426258
  • Ramírez JC, Gherardi MM, Esteban M. Biology of attenuated modified vaccinia virus Ankara recombinant vector in mice: virus fate and activation of B- and T-cell immune responses in comparison with the Western Reserve strain and advantages as a vaccine. J Virol 2000; 74:923 - 33; http://dx.doi.org/10.1128/JVI.74.2.923-933.2000; PMID: 10623755
  • Barouch DH, Santra S, Kuroda MJ, Schmitz JE, Plishka R, Buckler-White A, et al. Reduction of simian-human immunodeficiency virus 89.6P viremia in rhesus monkeys by recombinant modified vaccinia virus Ankara vaccination. J Virol 2001; 75:5151 - 8; http://dx.doi.org/10.1128/JVI.75.11.5151-5158.2001; PMID: 11333896
  • Ourmanov I, Brown CR, Moss B, Carroll M, Wyatt L, Pletneva L, et al. Comparative efficacy of recombinant modified vaccinia virus Ankara expressing simian immunodeficiency virus (SIV) Gag-Pol and/or Env in macaques challenged with pathogenic SIV. J Virol 2000; 74:2740 - 51; http://dx.doi.org/10.1128/JVI.74.6.2740-2751.2000; PMID: 10684290
  • Amara RR, Villinger F, Staprans SI, Altman JD, Montefiori DC, Kozyr NL, et al. Different patterns of immune responses but similar control of a simian-human immunodeficiency virus 89.6P mucosal challenge by modified vaccinia virus Ankara (MVA) and DNA/MVA vaccines. J Virol 2002; 76:7625 - 31; http://dx.doi.org/10.1128/JVI.76.15.7625-7631.2002; PMID: 12097576
  • Cosma A, Nagaraj R, Bühler S, Hinkula J, Busch DH, Sutter G, et al. Therapeutic vaccination with MVA-HIV-1 nef elicits Nef-specific T-helper cell responses in chronically HIV-1 infected individuals. Vaccine 2003; 22:21 - 9; http://dx.doi.org/10.1016/S0264-410X(03)00538-3; PMID: 14604567
  • Harrer E, Bäuerle M, Ferstl B, Chaplin P, Petzold B, Mateo L, et al. Therapeutic vaccination of HIV-1-infected patients on HAART with a recombinant HIV-1 nef-expressing MVA: safety, immunogenicity and influence on viral load during treatment interruption. Antivir Ther 2005; 10:285 - 300; PMID: 15865223
  • Dorrell L, Yang H, Ondondo B, Dong T, di Gleria K, Suttill A, et al. Expansion and diversification of virus-specific T cells following immunization of human immunodeficiency virus type 1 (HIV-1)-infected individuals with a recombinant modified vaccinia virus Ankara/HIV-1 Gag vaccine. J Virol 2006; 80:4705 - 16; http://dx.doi.org/10.1128/JVI.80.10.4705-4716.2006; PMID: 16641264
  • Ondondo BO, Yang H, Dong T, di Gleria K, Suttill A, Conlon C, et al. Immunisation with recombinant modified vaccinia virus Ankara expressing HIV-1 gag in HIV-1-infected subjects stimulates broad functional CD4+ T cell responses. Eur J Immunol 2006; 36:2585 - 94; http://dx.doi.org/10.1002/eji.200636508; PMID: 17013989
  • Dorrell L, Williams P, Suttill A, Brown D, Roberts J, Conlon C, et al. Safety and tolerability of recombinant modified vaccinia virus Ankara expressing an HIV-1 gag/multiepitope immunogen (MVA.HIVA) in HIV-1-infected persons receiving combination antiretroviral therapy. Vaccine 2007; 25:3277 - 83; http://dx.doi.org/10.1016/j.vaccine.2007.01.005; PMID: 17257714
  • Schooley RT, Spritzler J, Wang H, Lederman MM, Havlir D, Kuritzkes DR, et al, AIDS Clinical Trials Group 5197 Study Team. AIDS clinical trials group 5197: a placebo-controlled trial of immunization of HIV-1-infected persons with a replication-deficient adenovirus type 5 vaccine expressing the HIV-1 core protein. J Infect Dis 2010; 202:705 - 16; http://dx.doi.org/10.1086/655468; PMID: 20662716
  • Li JZ, Brumme ZL, Brumme CJ, Wang H, Spritzler J, Robertson MN, et al, AIDS Clinical Trials Group A5197 Study Team. Factors associated with viral rebound in HIV-1-infected individuals enrolled in a therapeutic HIV-1 gag vaccine trial. J Infect Dis 2011; 203:976 - 83; http://dx.doi.org/10.1093/infdis/jiq143; PMID: 21402549
  • Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature 1998; 392:245 - 52; http://dx.doi.org/10.1038/32588; PMID: 9521319
  • Bottomly K. T cells and dendritic cells get intimate. Science 1999; 283:1124 - 5; http://dx.doi.org/10.1126/science.283.5405.1124; PMID: 10075571
  • Kovacsovics-Bankowski M, Rock KL. A phagosome-to-cytosol pathway for exogenous antigens presented on MHC class I molecules. Science 1995; 267:243 - 6; http://dx.doi.org/10.1126/science.7809629; PMID: 7809629
  • Rock KL. A new foreign policy: MHC class I molecules monitor the outside world. Immunol Today 1996; 17:131 - 7; http://dx.doi.org/10.1016/0167-5699(96)80605-0; PMID: 8820271
  • Rock KL, Clark K. Analysis of the role of MHC class II presentation in the stimulation of cytotoxic T lymphocytes by antigens targeted into the exogenous antigen-MHC class I presentation pathway. J Immunol 1996; 156:3721 - 6; PMID: 8621907
  • Peters JH, Gieseler R, Thiele B, Steinbach F. Dendritic cells: from ontogenetic orphans to myelomonocytic descendants. Immunol Today 1996; 17:273 - 8; http://dx.doi.org/10.1016/0167-5699(96)80544-5; PMID: 8962630
  • Fan Z, Huang XL, Zheng L, Wilson C, Borowski L, Liebmann J, et al. Cultured blood dendritic cells retain HIV-1 antigen-presenting capacity for memory CTL during progressive HIV-1 infection. J Immunol 1997; 159:4973 - 82; PMID: 9366424
  • Rissoan MC, Soumelis V, Kadowaki N, Grouard G, Briere F, de Waal Malefyt R, et al. Reciprocal control of T helper cell and dendritic cell differentiation. Science 1999; 283:1183 - 6; http://dx.doi.org/10.1126/science.283.5405.1183; PMID: 10024247
  • Filgueira L, Nestlé FO, Rittig M, Joller HI, Groscurth P. Human dendritic cells phagocytose and process Borrelia burgdorferi. J Immunol 1996; 157:2998 - 3005; PMID: 8816408
  • Mbow ML, Zeidner N, Panella N, Titus RG, Piesman J. Borrelia burgdorferi-pulsed dendritic cells induce a protective immune response against tick-transmitted spirochetes. Infect Immun 1997; 65:3386 - 90; PMID: 9234802
  • Su H, Messer R, Whitmire W, Fischer E, Portis JC, Caldwell HD. Vaccination against chlamydial genital tract infection after immunization with dendritic cells pulsed ex vivo with nonviable Chlamydiae. J Exp Med 1998; 188:809 - 18; http://dx.doi.org/10.1084/jem.188.5.809; PMID: 9730883
  • Brossart P, Goldrath AW, Butz EA, Martin S, Bevan MJ. Virus-mediated delivery of antigenic epitopes into dendritic cells as a means to induce CTL. J Immunol 1997; 158:3270 - 6; PMID: 9120283
  • Svensson M, Stockinger B, Wick MJ. Bone marrow-derived dendritic cells can process bacteria for MHC-I and MHC-II presentation to T cells. J Immunol 1997; 158:4229 - 36; PMID: 9126984
  • Lu W, Wu X, Lu Y, Guo W, Andrieu JM. Therapeutic dendritic-cell vaccine for simian AIDS. Nat Med 2003; 9:27 - 32; http://dx.doi.org/10.1038/nm806; PMID: 12496959
  • Lapenta C, Santini SM, Logozzi M, Spada M, Andreotti M, Di Pucchio T, et al. Potent immune response against HIV-1 and protection from virus challenge in hu-PBL-SCID mice immunized with inactivated virus-pulsed dendritic cells generated in the presence of IFN-alpha. J Exp Med 2003; 198:361 - 7; http://dx.doi.org/10.1084/jem.20021924; PMID: 12874266
  • Yoshida A, Tanaka R, Murakami T, Takahashi Y, Koyanagi Y, Nakamura M, et al. Induction of protective immune responses against R5 human immunodeficiency virus type 1 (HIV-1) infection in hu-PBL-SCID mice by intrasplenic immunization with HIV-1-pulsed dendritic cells: possible involvement of a novel factor of human CD4(+) T-cell origin. J Virol 2003; 77:8719 - 28; http://dx.doi.org/10.1128/JVI.77.16.8719-8728.2003; PMID: 12885891
  • Kundu SK, Engleman E, Benike C, Shapero MH, Dupuis M, van Schooten WC, et al. A pilot clinical trial of HIV antigen-pulsed allogeneic and autologous dendritic cell therapy in HIV-infected patients. AIDS Res Hum Retroviruses 1998; 14:551 - 60; http://dx.doi.org/10.1089/aid.1998.14.551; PMID: 9591709
  • Lu W, Arraes LC, Ferreira WT, Andrieu JM. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nat Med 2004; 10:1359 - 65; http://dx.doi.org/10.1038/nm1147; PMID: 15568033
  • García F, Lejeune M, Climent N, Gil C, Alcami J, Morente V, et al. Therapeutic immunization with dendritic cells loaded with inactivated autologous HIV-1 in chronic HIV-1 infected patients. J Infect Dis 2005; 195:1680 - 5; http://dx.doi.org/10.1086/429340
  • Ide F, Nakamura T, Tomizawa M, Kawana-Tachikawa A, Odawara T, Hosoya N, et al. Peptide-loaded dendritic-cell vaccination followed by treatment interruption for chronic HIV-1 infection: a phase 1 trial. J Med Virol 2006; 78:711 - 8; http://dx.doi.org/10.1002/jmv.20612; PMID: 16628588
  • Connolly NC, Whiteside TL, Wilson C, Kondragunta V, Rinaldo CR, Riddler SA. Therapeutic immunization with human immunodeficiency virus type 1 (HIV-1) peptide-loaded dendritic cells is safe and induces immunogenicity in HIV-1-infected individuals. Clin Vaccine Immunol 2008; 15:284 - 92; http://dx.doi.org/10.1128/CVI.00221-07; PMID: 17942609
  • Gandhi RT, O’Neill D, Bosch RJ, Chan ES, Bucy RP, Shopis J, et al, AIDS Clinical Trials Group A5130 team. A randomized therapeutic vaccine trial of canarypox-HIV-pulsed dendritic cells vs. canarypox-HIV alone in HIV-1-infected patients on antiretroviral therapy. Vaccine 2009; 27:6088 - 94; http://dx.doi.org/10.1016/j.vaccine.2009.05.016; PMID: 19450647
  • Kloverpris H, Karlsson I, Bonde J, Thorn M, Vinner L, Pedersen AE, et al. Induction of novel CD8+ T-cell responses during chronic untreated HIV-1 infection by immunization with subdominant cytotoxic T-lymphocyte epitopes. AIDS 2009; 23:1329 - 40; http://dx.doi.org/10.1097/QAD.0b013e32832d9b00; PMID: 19528789
  • Routy JP, Boulassel MR, Yassine-Diab B, Nicolette C, Healey D, Jain R, et al. Immunologic activity and safety of autologous HIV RNA-electroporated dendritic cells in HIV-1 infected patients receiving antiretroviral therapy. Clin Immunol 2010; 134:140 - 7; http://dx.doi.org/10.1016/j.clim.2009.09.009; PMID: 19889582
  • García F, Climent N, Assoumou L, Gil C, González N, Alcamí J, et al, DCV2/MANON07- AIDS Vaccine Research Objective Study Group. A therapeutic dendritic cell-based vaccine for HIV-1 infection. J Infect Dis 2011; 203:473 - 8; http://dx.doi.org/10.1093/infdis/jiq077; PMID: 21233310
  • Routy JP, Angel J, Vezina S, Tremblay C, Loutfy M, Gill J, et al. Final Analysis of a Phase 2 Study of an Autologous DC Immunotherapy (AGS-004) Showed Positive Outcomes in Primary Endpoint of Viral Load Control, and Favorable Safety and Immunogenicity Profile, in Subjects Undergoing Structured Treatment Interruption of ART (Abstract 385) [Abstract]. 18th Conference on Retroviruses and Opportunistic Infections Boston February 27-March 2 2011.
  • García F, Routy JP. Challenges in dendritic cells-based therapeutic vaccination in HIV-1 infection Workshop in dendritic cell-based vaccine clinical trials in HIV-1. Vaccine 2011; 29:6454 - 63; PMID: 21791232

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.