Abstract
The immune correlates of risk analysis and recent non-human primate (NHP) challenge studies have generated hypotheses that suggest HIV-1 envelope may be essential and, perhaps, sufficient to induce protective antibody responses against HIV-1 acquisition at the mucosal entry. New prime-boost mosaic and conserved-sequence, together with replicating vector immunisation strategies aiming at inducing immune responses or greater breadth, as well as the development of immunogens inducing broadly neutralising antibodies and mucosal responses, should be actively pursued and tested in humans. Whether the immune correlates of risk identified in RV144 can be extended to other vaccines, other populations, or different modes and intensity of transmission, and against increasing HIV-1 genetic diversity, remains to be demonstrated. Although NHP challenge studies may guide vaccine development, human efficacy trials remain key for answering the critical questions leading to the development of a global HIV-1 vaccine for licensure.
Keywords: HIV vaccine, HIV prevention, clinical trials, efficacy, immune correlates
Introduction
At the end of 2013, 35 million people were living with HIV worldwide, sub-Saharan Africa bearing the heaviest burden. While 70% of HIV cases are attributed to heterosexual transmission, the remaining 30% are attributed to a combination of men who have sex with men (MSM), mother-to child transmission and people who inject drugs (PWID) [1]. Worldwide, the number of people, including children, with new HIV-1 infections has fallen by 38% since 2001. This result can be attributed to aggressive HIV prevention measures including behavioural interventions, wider access to antiretroviral treatment (ART), male circumcision, treatment of sexually transmitted infections, harm reduction and prevention of mother-to-child transmission [2–4]. New prevention strategies are being developed including topical microbicides, pre-exposure prophylaxis (PrEP), and ART for prevention (TasP) [5–11]. While current strategies have led to a decrease in HIV prevalence and HIV-related mortality, they have not stopped HIV transmission. Several factors justify the development of a globally effective HIV-1 vaccine: the challenges associated with human behaviour and acting responsibly in sexual settings, legal barriers in some countries and limited access to ART. Only 25% of US citizens living with HIV have achieved an undetectable viral load [12]. Vaccines represent the most cost-effective public health intervention to counter infectious diseases. Vaccines against diseases such as smallpox, poliomyelitis and diphtheria have reduced the prevalence and incidence of associated illness to eradication and negligible numbers; therefore, providing irrefutable support for the effectiveness of a vaccine strategy to counter HIV transmission [13].
Several challenges hamper the development of a prophylactic HIV-1 vaccine. The virus has extraordinary diversity, even within an infected host. It integrates quickly into host DNA, establishing latent reservoirs inaccessible to treatment and the immune system [14]. The envelope spike is heavily glycosylated and the virus makes use of non-critical, decoy immunodominant epitopes [15]. The substantial replicative capacity of HIV-1 facilitates rapid and adaptive sequence evolution driven by point mutations and viral recombination creating an unprecedented capacity to evade immune responses [16,17]. Broadly neutralising antibodies (bNAb) developed in 20% of HIV-infected subjects are insufficient to limit HIV disease progression within these subjects [18]. Animal models, while helpful in guiding clinical development, remain imperfect in predicting the outcome of human trials. Our understanding of immune correlates of protection has improved but remains limited.
Multiple HIV-1 vaccine Phase I and II trials have been conducted worldwide (reviewed in [19]). Only one of six HIV-1 vaccine efficacy trials was effective in preventing HIV-1 acquisition. The Thai Phase II trial, RV144, was the first trial that showed that a vaccine against HIV is possible, with efficacy of 31% [20]. We review the findings of key clinical efficacy trials and animal studies and develop new perspectives that may lead an HIV-1 vaccine to licensure.
Lessons learned from human efficacy trials and animal challenge studies
Both antibody and cell-mediated immune responses are now thought to be important to counter HIV-1 [21] in both the systemic and mucosal compartments, the entry point for sexual transmission [22]. Among the promising vaccine approaches to achieve this objective, the prime-boost concept was developed by priming the immune system with a vaccine consisting either of a recombinant vector or DNA plasmids expressing HIV-1 proteins, and boosting with soluble HIV-1 proteins or with another vector expressing HIV-1 proteins [23,24]. Table 1 outlines the HIV-1 vaccine efficacy trials conducted so far.
Table 1.
Trial | Details | Population | Area | Outcome |
---|---|---|---|---|
Vax004 | AIDSVAX B/B gp120 (MN and GNE8 subtype B) gp120 in alum | MSM* and women who engage in high-risk behaviour | USA and Europe | No efficacy |
Vax003 | AIDSVAX B/E gp120 (subtype B MN and CRF01_AE CM244) gp120 in alum | PWID† | Thailand | No efficacy |
HVTN 502/Merck 023/Step trial | MRKAd5‡ HIV-1 Gag/Pol/Nef subtype B | High-risk population, MSM, heterosexual men and women | USA | No efficacy |
HVTN 503/ Phambili trial | MRKAd5 HIV-1 Gag/Pol/Nef subtype B | Heterosexual men and women | Republic of South Africa | No efficacy; increased HIV infection observed in vaccinees |
RV144 | ALVAC-HIV§ (vCP1521) and AIDSVAX B/E (subtype B MN and CRF01_AE CM244) rgp120 in alum | Community risk | Thailand | 31.2% efficacy against HIV acquisition at 42 months, 60% at 12 months. No effect on plasma viral load and CD4 count |
HVTN 505 | DNA Gag, Pol, and Nef from HIV-1 subtype B and Env from subtypes A, B, and C and rAd5 subtype B Gag-Pol and Env A, B, and C | Circumcised MSM and transgender individuals lacking infection with Ad5 | USA | No efficacy |
Men who have sex with men;
people who inject drugs;
MRKAd5: recombinant replication-incompetent Ad5 vector;
ALVAC-HIV: recombinant canarypox vector
Vax004
The first HIV-1 vaccine efficacy trial, Vax004, enrolled women at high risk for heterosexual transmission and MSM in the Netherlands and North America. The vaccine was composed of monomeric gp120 envelope subunits derived from GNE8 and MN HIV-1 subtype B isolates, formulated in alum, and aimed at inducing neutralising antibody (NAb) to block HIV-1 transmission. HIV incidence did not differ significantly between vaccine and placebo recipients nor did it affect HIV-1 disease progression [25,26]. High NAb levels against the easy-to-neutralise MN strain were, however, significantly inversely correlated with HIV-1 incidence while low levels against more-difficult-to-neutralise viruses were not, suggesting that level and breadth of elicited NAb were not sufficient for protection. Stratification of HIV-1 acquisition by gender showed that NAb responses were higher in women than men, while behaviour and race had no significant effect [27]. Vaccination was associated with gp120-binding IgG in cervico-vaginal lavage, gingival secretions and gp120 plasma IgA [28]. An inverse correlation was found between antibody-dependent cell-mediated virus inhibition (ADCVI) activity and the rate of HIV acquisition among vaccinated individuals. Fcγ receptor IIIa genotype was associated with an increased rate of HIV-1 infection in low-risk, but not in high-risk vaccinees [29].
Vax003
Vax003, the only HIV-1 vaccine efficacy trial to target PWID, was conducted in Bangkok, Thailand. The vaccine consisted of bivalent gp120 envelope subunits, HIV-1 subtype B MN and CRF01_AE A244, in alum. The antibody levels prior to infection for gp120, A244 V2, A244 V3, blocking of A244 binding to CD4, and MN neutralisation were not significantly different between the HIV-infected and uninfected vaccine recipients and did not correlate with the rate of HIV infection [30].
The failure of these two trials redirected the focus of HIV-1 vaccine development into looking at the efficacy of vaccine-induced cell-mediated responses [19]. The rationale for T cell-based vaccines has been recently reviewed [31–33].
The Step trial
The Step trial (HVTN 502/Merck 023) tested a mixture of replication-defective recombinant adenovirus 5 vectors, MRKAd5, expressing HIV-1 subtype B gag, pol and nef genes, in MSM and women and men at high risk for HIV infection in the US [34]. The trial was halted after an interim analysis showing the vaccine did not confer protection against HIV-1 acquisition, or reduce post-infection plasma viral load despite detection of HIV-specific IFN-γ ELISPOT and intracellular cytokine-staining CD4+ and CD8+ T cell responses in a majority of vaccine recipients [35]. A post hoc multivariate analysis showed a greater risk for HIV-1 infection in uncircumcised men with pre-existing Ad5-neutralising antibodies [36]. The risk waned over time [37]. An additional study did not support sexual behaviour as a risk factor for increased HIV acquisition of uncircumcised individuals in the study who became HIV infected [38]. Ad5 antibodies were not correlated with increased risk for HIV acquisition in unvaccinated individuals [39]. Results from another study suggested that subjects infected during the Step trial seemed to have qualitative immune differences that increased their risk of HIV-1 infection independent of vaccination [40]. A sieve analysis showed evidence of vaccine-elicited immune pressure on the founder virus although no specific CD8+ cytotoxic T lymphocytes (CTL) recognising that epitope could be identified [41]. Despite evidence of anamnestic responses, the sieve effect was not well explained by available measures of T cell immunogenicity. Sequence divergence from the vaccine was not significantly associated with acute viral load [42]. Vaccinees with HLA alleles associated with HIV-1 control had a significantly lower mean viral load over time [43,44]. Intriguingly, non-HIV-specific IFN-γ ELISPOT magnitude was a significant direct correlate of risk (CoR) for HIV-1 infection in the vaccine group [45]. Interestingly, the most highly conserved epitopes were detected at a lower frequency, suggesting that stronger responses to conserved sequences may be as important as breadth for protection [46]. The outcome of the Step trial was recapitulated in an Indian rhesus macaque study where animals vaccinated with a regimen similar to that employed in the Step trial were not protected against an SIVsmE660 challenge [47]. Rhesus macaques chronically infected with a host-range mutant Ad5 (Ad5hr) and immunised with an rAd5 SIVmac239 gag/pol/nef vaccine were challenged with a series of escalating dose penile exposures to SIVmac251. Despite inducing CD8+ T cell responses in 70% of the monkeys, the vaccine did not protect vaccinated animals from penile SIV challenge [48].
The Phambili study
The Phambili study (HVTN 503), a parallel trial conducted in South Africa where the major circulating clade is HIV-1 subtype C, tested the MRKAd5 HIV-1 vaccine. After the results of the Step trial, the Phambili study was halted and treatment allocations unblinded. Only 801 of the 3000 participants had been enrolled in the study. Owing to this setback, statistical power of 80% to evaluate vaccine efficacy was not achieved. There was no evidence of vaccine efficacy, which did not differ by Ad5 antibody titre, gender, age, herpes simplex virus type 2 status or circumcision. There was no significant difference in viral load set-point although there was a trend for a lower viral load in women. HSV-2 infection increased the risk of HIV-1 in men by five times, but not in women [49]. Variables such as early unblinding, untimely interruption of the study, and cessation of vaccinations may have introduced bias to the interpretation of results. An additional factor noted is that the demographics of the Step and Phambili studies were different; moreover, the men in the Phambili study were mostly heterosexual, while men in the Step study were MSM, which suggests that risk factors may also be different [50].
HVTN 505
HVTN 505 was an efficacy trial conducted in 21 sites in the United States. MSM or transgender women were enrolled to receive a vaccine regimen consisting of priming with a mixture of six DNA plasmids containing HIV-1 subtype B gag, pol and nef, and subtypes A, B and C env genes, and boosting with a replication-incompetent recombinant Ad5 vector expressing HIV-1 subtype B gag-pol and subtype A, B and C env genes.
In previous studies, the vaccine regimen was shown to be safe, well tolerated and induced polyfunctional CD4+ and CD8+ T cells, multi-clade anti-Env binding antibodies, and NAb against easy-to-neutralise Tier 1 viruses [51–54]. HVTN 505 was halted for futility, showing no efficacy and no statistically significant effect on viral load and a non-significant excess of HIV infection in the vaccinated group [55].
The vaccine regimen failed to protect NHP against SIVmac251 infection but conferred protection against an intrarectal challenge with SIVsmE660 in half the vaccinated monkeys. Additionally, a reduction in peak plasma virus RNA was observed in Mamu-A*01-positive monkeys, suggesting a role of cytotoxic T lymphocytes in the control of SIV replication. Low levels of neutralising antibodies as well as envelope-specific CD4+ T cell responses were also associated with protection [56].
Comment on recombinant adenovirus type 5-vectored trials
There has been much discussion about the results of these three efficacy trials [22,57]. The nature of futility analyses will hopefully prevent vaccine trials from progressing to the point where enhancement of infection is seen, and once unblinding is undertaken (and for ethical reasons this should be the default) it is difficult with post hoc, unblinded analyses to cleanly study questions related to HIV-1 infection. Adenovirus vectors differ in their use of surface receptors, patterns of induced RNA expression and immunogenicity [58], but responses to the major hexon protein may cross types and species [59,60].
A comparative study evaluated HIV susceptibility and phenotypes of human CD4+ T cells and found that Ad5-specific CD4+ T cells, naturally exposed or introduced via vaccination, are more susceptible to HIV in vitro than CMV-specific CD4+ T cells. Preferential losses or strong reductions of Ad5-specific T cell responses were also observed in HIV-infected individuals. Further analyses showed a prevalence of pro-inflammatory Th17-like, gut-mucosa homing phenotypes of Ad5-specific CD4+ T cells. Flow cytometry analysis of in vitro samples also showed increased preference for infecting both IL-17- and IL-2-producing Ad5-specific CD4+ T cells. This data suggests a possible mechanism for increased HIV infection rate in the vaccine recipients from the Step study. Additionally, the data suggest that HIV susceptibility testing of vector-specific CD4+ T cells from HIV vaccine candidates may be needed [61]. A recent analysis suggests that the proportion of cells, probably CD4+ T cells, producing IFN-© without stimulation by exogenous antigen appears to carry information beyond T cell activation and baseline characteristics that predict risk of HIV-1 infection [45].
Although no increase in activated total or vector-specific mucosal CD4+ T lymphocytes was detected following Ad26 vaccination in humans [62], the assessment of gut CD4+ T cell activation should deserve particular attention in humans vaccinated with any vector, in particular adenovirus-based vectors. Taken together, the studies of recombinant Ad5 vectors raise an important, and more general question about the impact of vaccine vectors on HIV acquisition: the properties of viral vectors that confer adjuvant-like qualities may induce activation or display of homing markers and may render activated CD4+ T cells, and the host, more susceptible to HIV infection [63,64].
RV144
The vector prime and protein-boost concept was applied to RV144, a community-based Phase III efficacy trial that enrolled mostly heterosexual HIV-uninfected 18–30-year-old men and women at ‘community risk’ in Rayong and Chon Buri provinces in Thailand. The prime, ALVAC-HIV (vCP1521), was a canarypox vector expressing HIV-1 env (CRF01_AE 92TH023), gag, protease and the gp41 transmembrane anchoring region subtype B (LAI) genes. The boost, AIDSVAX B/E, was the bivalent monomeric gp120 soluble protein in alum, previously tested in Vax003. The trial participants received ALVAC-HIV or placebo at day 0, and then at 4, 12 and 24 weeks, and AIDSVAX B/E or placebo at weeks 12 and 24. The vaccine regimen was safe, well tolerated and judged adequate for public use [65].
HIV-1 incidence in the RV144 placebo group was 0.28 infections per 100 person-years, more than 10-fold lower than that observed in Vax003, Vax004, Step, Phambili or HVTN 505 [66]. In a pre-specified modified intention-to-treat analysis of 16,395 participants, which excluded seven individuals who were diagnosed with HIV at baseline, the vaccine efficacy was 31.2% at 42 months of follow-up after first vaccination. There were statistically non-significant differences in efficacy between men and women; however, the study was not adequately powered to analyse subgroups. No significant difference was seen in the mean plasma viral load and CD4+ T cell counts between the HIV-infected individuals of the vaccine and placebo groups [20,67]. Post hoc analytical data indicated vaccine efficacy was 44% at 18 months and 60% at 12 months after vaccination, indicating that vaccine efficacy was non-durable [68]. Post-infection CD4+ T cell count and HIV-1 RNA plasma set-point viral load were not different between recipients of vaccine or placebo [69]. A significant reduction in seminal fluid viral load was observed in vaccine recipients who became infected. As observed in previous Phase I and II trials, the vaccine regimen induced binding antibody against HIV-1 Env immunogens and p24 Gag, CD4+ proliferative responses (63%), some CD8+ Tcell responses (24%), NAb against Tcell line-adapted viruses of subtype B and CRF01_AE (96% and 71%, respectively), and antibody-dependent cell-mediated cytotoxicity (ADCC) [20,70,71]. Predominantly CD4+ T cell mediated, IFN-© ELISPOT-positive responses were detected in 41% of the vaccinees and targeted the V2 region, which includes the α4β7 integrin-binding motif [72–74]. A majority of vaccine subjects (97%) had antibody responses against cyclic V2 peptide at 2 weeks post immunisation, declining to 19% at 28 weeks after last injection. Antibody responses targeted the mid-region of the V2 loop that contains conserved epitopes and has the amino acid sequence KQKVHALFYKLDIVPI (HXB2 numbering sequence 169–184) [75]. Intracellular cytokine staining confirmed that Env responses predominated (19 of 30; 63% of vaccine recipients) and were mediated by polyfunctional effector memory CD4+ T cells, with the majority of responders producing both IL-2 and IFN-© (63%). HIV Env antibody titres were higher in subjects with IL-2 compared with those without IL-2-secreting HIV Env-specific effector memory T cells. Proliferation assays revealed that HIV antigen-specific T cells were CD4+, with the majority (80%) expressing CD107a, a functional marker of natural killer cell activity [76]. Although the detection of neutralising activity against Tier 1 viruses was detected in both Vax003 and RV144, the Tier 1 NAb titres were higher after the RV144 regimen compared to two gp120 protein administrations alone, confirming a priming effect for ALVAC-HIV [77].
RV144 and immune correlates of risk
The definition of correlates of protection (CoP) was recently redefined to include mechanistic and non-mechanistic CoP. Correlates of risk (CoR) are the statistically relevant responses that may be a CoP, or indicative of susceptibility to a pathogen or genetic susceptibility to infection [78–81]. Immune CoP in HIV-1 and NHP vaccine studies, and more specifically in RV144, were recently reviewed [22,66].
The efficacy observed in RV144 led to a case–control study being conducted to determine the antibody and cellular immune CoR for HIV acquisition [82]. After a careful stepwise selection process, six primary assay variables were selected: CD4+ T cell responses, V1V2 binding antibodies, neutralising antibodies, ADCC, avidity of IgG antibodies for Env, and IgA antibodies binding to Env. Cryopreserved blood samples were analysed from individuals who became infected after vaccination (case) and on non-infected vaccinees (controls).
Plasma IgG antibodies to scaffolded V1V2 caseA2 (HIV-1 subtype B) envelope proteins correlated inversely with risk of infection, while plasma IgA-envelope binding correlated with risk. Neither low levels of V1V2 antibodies nor high levels of Env-specific IgA antibodies in vaccinees or those in the placebo group correlated with increased rates of infection, ruling out the possibility of vaccine-induced enhancement of risk of infection [82]. In vaccinated subjects with low levels of plasma Env-specific IgA, an inverse correlation with infection was observed between IgG avidity, ADCC, neutralising antibodies, and Env-specific CD4+ T cells. RV144 antibodies to subtype A, C and CRF01_AE gp70 V1V2 scaffold proteins also correlated inversely with risk [83], suggesting that the RV144 regimen might protect against heterosexual transmission of HIV strains heterologous (A and C) to the vaccine components. Two weeks after last vaccination, 97% of RV144 studied plasma samples from vaccine recipients contained antibodies to V2 region synthetic peptides, falling to 19% at 48 weeks, suggesting that waning vaccine efficacy may be correlated with waning V2 antibody response. Interestingly, gp70 V1V2 antibodies were lower in HVTN 505 compared to RV144 [55].
The antibody response to V3 CRF01_AE and neutralising antibodies also inversely correlated with the risk of HIV infection in vaccine recipients with lower levels of Env-specific plasma IgA. In Vax003 and Vax004 (no protection), serum IgG responses targeted the same epitopes as in RV144 with the exception of an additional C1 reactivity in Vax003 and infrequent V2 reactivity in Vax004. Moreover, IgG to linear epitopes in the V2 and V3 regions of gp120 correlate with reduced risk in RV144 [84].
A sieve analysis identified two vaccine-associated genetic signatures in V2 corresponding to sites 169 and 181, further supporting the hypothesis that vaccination-induced immune responses directed against the V2 loop were associated with protection [85]. Monoclonal antibodies from RV144 vaccine recipients bind the V2 K169 residue, providing additional evidence that vaccine-induced antibodies correspond to the observed sieve effect. These V2-specific antibodies can mediate ADCC, neutralisation and low-level virus capture [86,87]. These findings generate the hypothesis that V2 IgG plays a role in protection against HIV-1 acquisition but do not distinguish between mechanistic or non-mechanistic mechanisms of protection [80].
In previous clinical studies, monomeric gp120 induced high levels of Env-specific IgG4 antibodies [88] while ALVAC (vCP1452) prime and gp120 MN in alum boost elicited lower IgG4 relative to IgG1 and IgG3 antibodies [89]. Antigen-specific IgG3 antibodies are associated with a beneficial effect against several pathogens [90–92]. Conversely, IgG4 has been associated with progression to AIDS [93]. IgG3 can fix the complement and has a high affinity for FcγR [94]. In RV144, Env IgG3 was correlated with decreased risk of HIV infection, a response that declined rapidly compared to overall IgG responses [95]. A recent comparison of RV144 and Vax003 showed that Env-specific IgG3 and V1/V2 IgG3 response rates were higher in recipients of the RV144 vaccine compared to Vax003 vaccinees and conversely that IgG4 were considerably lower in RV144. V1/V2 IgG3 responses and IgG3 responses specific for V1/V2 169K correlated with decreased risk of HIV-1 infection after IgA adjustment [95]. Chung et al. recently showed that the RV144 regimen elicited highly coordinated Fc-mediated effector responses, with the selective induction of highly functional IgG3 antibodies. By contrast, Vax003 elicited monofunctional antibody responses influenced by IgG4 selection. Moreover, only RV144 induced IgG1 and IgG3 antibodies that targeted the crown of the HIV envelope V2 loop, although with low coverage of breakthrough viral sequences [96]. ALVAC priming, due to its unique pro-inflammatory cytokine and chemokine response following vaccination in rhesus monkeys and infection in human PBMC, might shift the IgG subclass response to IgG3 in humans after vector prime and envelope protein boost compared with envelope vaccination alone [97]. The contribution of Fc–Fc©R interaction-mediated functions through mechanisms such as ADCC, ADCVI, and antibody-dependent cell phagocytosis (ADCP) antibodies remains to be explored [98,99]. A recent post hoc analysis of RV144 showed a positive association between the FcγRIIC polymorphism and vaccine efficacy, emphasising the role of host genetics in predicting vaccine efficacy [100].
The RV144 direct correlation of plasma IgA with risk of infection is puzzling [101]. It has been suggested that plasma IgA may block IgG activity involving ADCC and phagocytosis [102–104]. In RV144, IgA antibodies elicited by RV144 block C1 region-specific IgG-mediated ADCC [105]. Monoclonal antibodies from RV144 vaccine recipients appeared to bind to a region of V2 that partially overlaps the binding of the bNAb CH01 and PG9. Although not broadly neutralising, these V2-specific antibodies mediate ADCC. Further mapping of ADCC responses from RV144 volunteers shows that a significant portion of the immune responses is directed at the first constant region of gp120 Env (C1) [106]. These C1-directed antibodies are able to block the binding of the A32mAb, which recognises a conformational epitope that includes the C1 domain. The C1-specific antibodies appear to act synergistically with anti-V2 antibodies, possibly by inducing conformational changes that improve the exposure of V2 to binding [107]. IgA purified from serum of HIV-uninfected RV144 vaccinees was able to efficiently opsonise viral particles in the absence of significant aggregation, reflective of monomeric IgA. In contrast, dimeric IgA monoclonal antibodies (mAbs) formed stable viral aggregates, suggesting aggregation as a potential protection mechanism at the mucosal portals of viral entry [108]. Table 2 outlines the immune CoR identified in clinical efficacy trials.
Table 2.
Trial | Details | |
---|---|---|
RV144 | Plasma IgG binding antibody to gp70V1V2 scaffold proteins (subtypes B, A, C and CRF01_AE) inversely correlated with risk of infection | |
Plasma IgA-envelope binding antibodies correlated with risk | ||
In vaccine recipients with low plasma IgA antibodies, an inverse correlation was observed between rate of infection and Env-specific CD4+ T cells, ADCC, neutralising antibodies, and Env IgG avidity | ||
Sieve analysis showed two positions in V2 (169 and 181), which substantiates the hypothesis that protection resulted from vaccine-induced responses against V2 loop | ||
Positive association between the Fc©RIIC polymorphism and vaccine efficacy | ||
Env IgG3 correlated with decreased risk of HIV infection | ||
Vax004 | ADCVI inverse correlated with rate of HIV acquisition | |
High levels of neutralising antibodies to MN inversely correlated with HIV incidence | ||
Fc© receptor IIIa genotype was associated with an increased rate of HIV-1 infection in low-risk, but not in high-risk vaccinees | ||
Vax003 Step trial (HVTN 502) | No correlates identified | |
Presence of HLA alleles and overall T-cell breadth and magnitude of the immune response significantly correlated with lower mean viral load in infected vaccinees suggesting the implication of CD8+ cytotoxic T lymphocytes | ||
Non-HIV-specific ELISPOT magnitude was a significant direct CoR for HIV-1 infection in vaccinees | ||
Vax003 | Analysis ongoing |
CoR: correlate of risk
Innovative approaches to HIV-1 vaccine development
Antibody-based strategies
Capitalising on the RV144 results and correlates of risk analysis
Advanced development of the pox vector prime and protein-boost strategy is proceeding towards efficacy trials in Southern Africa (heterosexual transmission), Thailand (MSM), and China (MSM) [57]. This strategy aims at inducing both non-neutralising (nNAbs) and neutralising (NAbs) antibodies. Designing vaccines to elicit production and concentration of antibodies at mucosal frontlines could aid in the development of an effective vaccine to protect women and MSM against HIV-1 [109]. The role and plausible mechanisms of action of nNAbs in protection against HIV acquisition have recently been reviewed [110,111]. Data from three NHP studies [112–114] suggest that Env is a necessary component for successful protection from SIV acquisition. Mechanisms of protection against acquisition may include inhibition of transcytosis [99,115–120], possible hindrance of HIV mobility by trapping of viruses linked to IgG and IgA within mucin layers outside the cervico-vaginal epithelium [121,122], viral capture by sIgA and IgG, viral aggregation [108], ADCC and ADCVI [117,118,123–125], and ADCP [126]. In Vax004, ADCVI activity was associated with lower infection rates [127]. The inhibition of virus replication in mucosal tissues ex vivo along with HIV-specific nNAb in mucosal secretions is currently being explored in RV144 follow-up studies [128–130].
Antigen design aiming at better-presenting conformational epitopes that induce nNAbs is explored. The antigenicity of the A244 gp120(used on RV144) C1 region and the V2 conformational epitopes could be enhanced by the deletion of 11 N terminus amino acids of gp120 (Δ11). Conformational V1/V2 mAbs gave significantly higher levels of blocking of plasma IgG from A244 Δ11 gp120 immunised animals than IgG from animals immunised with unmodified A244 gp120 [131].
Inducing broadly neutralising antibodies
The challenge for epitope-based vaccine design is that only broadly conserved and exposed epitopes are suitable for vaccine targeting, but these epitopes, in their natural context, tend to elicit poor antibody responses. Perhaps the most difficult step is to design, engineer and produce a stable envelope immunogen that mimics the antigenic profile of the functional envelope spike [132].
Few engineered trimeric envelopes have been able to induce bNAb in animals [133]. Removal of individual glycans proximal to CD4-binding region impairs viral infectivity and results in enhanced capability to induce neutralising activity in mice [134]. The elimination of the glycosylation site near the gp41 loop resulted in enhanced immunogenicity, but immunisation of monkeys with this protein and two others derived from patients with bNAb was not more immunogenic than with one [135]. A stable gp140 trimeric envelope induced bNAb against Tier 1 and Tier 2 viruses with titres substantially higher than those elicited by the corresponding gp120 monomers [136]. The use of multivalent mixtures of natural HIV-1 subtype C envelope immunogens elicited a greater magnitude of NAbs against a panel of Tier 1 viruses than any single clade C trimer alone, but not against Tier 2 viruses [137]. A combination of mosaic envelopes tested in macaques increased the magnitude of NAbs but not the breadth of the response [138]. So far, no trimeric envelope induces bNAb in humans [139].
Another approach, called B cell lineage vaccine design, aims to engage the naïve B cell repertoire residing in bone marrow and secondary lymphoid tissue. Specifically, one or more clonally related bNAb must be isolated and, using next generation sequencing, an antibody lineage constructed through inference that links the mutated bNAb-producing cell to its naïve, germline ancestor. Recombinant antibody technology would express members of that bNAb lineage in order to select HIV-1 envelope constructs that optimally bind them. Those envelope constructs would be used as immunogens in a prime-boost to engage the naïve B cells in vivo and iteratively stimulate B cell ‘evolution’ until bNAb-producing cells are elicited [140]. A recent study determines the viral and antibody evolution leading to induction of a lineage of HIV-1 broadly neutralising antibodies, and provides insights into strategies to elicit similar antibodies by vaccination [141].
Vector-based delivery of broadly neutralising antibodies
Whether bNAb will effectively confer protection against HIV acquisition in humans remains a key question. An alternative to inducing bNAb by vaccination with immunogens is to deliver these bnMAb with viral vectors administered intramuscularly such as an adeno-associated virus (AAV) gene transfer vector expressing antibodies or antibody-like immunoadhesins with predetermined SIV specificity. SIV-specific molecules are endogenously synthesised in myofibres and passively distributed systemically. Long-lasting neutralising activity in serum was generated in monkeys and conferred complete protection against intravenous challenge with virulent SIVmac316 [142,143]. Similarly, full protection against intravenous HIV-1 challenge was obtained in humanised mice which received an AAV vector carrying full-length b12 antibody administered intramuscularly, while those receiving AAV expressing 2G12, 4E10 and 2F5 were partially protected [144]. Moreover, humanised mice receiving AAV vector carrying VRC07 were protected against repeated vaginal challenge with diverse HIV-1 strains [145]. An AAV vector carrying PG9 is now tested in a Phase I trial in Europe ( www.iavi.org).
Cell-mediated-based strategies
Other avenues being explored aim to increase breadth of the CD4+ and CD8+ Tcell immune responses and tackle HIV-1 genetic diversity [32,146,147]. Breadth and magnitude of Tcell responses correlated with control of set-point viral loads in macaques vaccinated with vectors expressing core SIV proteins (no envelope) and challenged with SIV [148]. This increased breadth and depth of epitope recognition may contribute both to protection against infection and to the control of variant viruses that emerge as they mutate away from recognition by cytotoxic T lymphocytes [149].
Conserved sequences
A recent study of the contribution of the lower replication capacity of the transmitted/founder virus and an associated induction of a broad primary HIV-specific T cell response, which was not undermined by rapid epitope escape, to long-term viral control in HIV-1 infection underscores the importance of the earliest CD8+ T cell response that targets regions of the virus proteome that cannot mutate without a high fitness cost. This further emphasises the need for vaccines to elicit a breadth of T cell responses to conserved viral epitopes [150].
Immunogens, including HIV-1 conserved sequences, provide an effective strategy to broaden responses by causing reaction to critical viral elements for which few escape pathways exist [151–154]. Priming with the conserved element vaccine followed by boost with the complete immunogen induces broad cellular and humoral immunity focused on the conserved regions of the virus [155]. Conserved sequences and immune responses have been characterised in animals [156,157] and have conferred partial protection against SIVmac251in macaques [158]. In humans, a combination of DNA, ChAd63 and MVA vectors that was found safe [159] and immunogenic, induced high levels of effector T cells that recognised virus-infected autologous CD4+ cells. In vitro inhibition of HIV-1 replication was mediated by both Gag- and Pol-specific effector CD8+ T cells that targeted epitopes that were subdominant in natural infection [160].
Mosaic immunogens
Polyvalent mosaic immunogens derived by recombination of natural strains of HIV-1 are designed to induce cellular immune responses that recognise genetically diverse circulating virus isolates. Mosaic HIV-1 antigens expressed by recombinant Ad26 vectors markedly augmented both the breadth and depth of similar magnitude of antigen-specific T lymphocyte responses as compared with consensus or natural sequence HIV-1 antigens in rhesus monkeys [161]. Ad26/MVA and Ad26/Ad35 vector-based vaccines expressing HIV-1 mosaic Env, Gag and Pol significantly decreased risk of acquisition following repetitive, intrarectal SHIV-SF162P3 challenges. Protection correlated with vaccine-elicited binding, neutralising and functional non-neutralising antibodies, suggesting that the coordinated activity of multiple antibody functions may contribute to protection against difficult-to-neutralise viruses. However, the vaccine regimens had only a modest effect on viral set-point after challenge [113]. In contrast, similar vector regimens expressing SIVsmE543 antigens afforded >2log10 reductions of set-point viral loads following heterologous SIVmac251 challenges [112]. Ad26 prime and MVA boost mosaic vectors should soon enter human clinical trials.
Replicating vectors
Mimicking the benefits of live-attenuated vaccines, the replicating vector approach was developed from the concept that persistent antigen exposure could confer immune control over viral exposure [162,163]. Live-attenuated SIV vaccine-mediated protection against SIVmac239 challenge strongly correlated with the magnitude and function of SIV-specific effector T cells in the lymph node but not in the blood or with other immune parameters. The maintenance of lymphoid tissue-based, effector-differentiated, SIV-specific T cells that intercept and suppress early wild-type SIV amplification and can control and perhaps clear infection, provides a rationale for the development of persistent vectors such as cytomegalovirus that can elicit and maintain such response [164]. A rhesus cytomegalovirus (RhCMV)-based vaccine expressing SIV proteins induced and maintained a high frequency of SIV-specific CD4+ and CD8+ T cell effector memory (TEM) responses at extra-lymphoid sites without measurable antibody responses to SIV. While the vaccine did not protect against infection, half of the vaccinated monkeys showed a stringent control of intrarectally administered SIVmac239 for more than a year. The outcome of challenge was predicted by peak SIV-specific CD8+ TEM frequencies in peripheral blood before the challenge [165,166]. However, it remains unclear why only 50% of the vaccinated animals were protected. RhCMV-SIV vectors elicit SIV-specific CD8+ T cells that recognise unusual, diverse and highly promiscuous epitopes, including dominant responses to epitopes restricted by MHC-II molecules and modulated by specific genes, suggesting that CMV vectors can be genetically programmed to achieve distinct patterns of CD8+ T cell epitope recognition [167].
Out-of-the-box approach
Prior infection of rhesus macaques with an attenuated SHIV conferred protection against vaginal SIV challenge associated with SIV-specific CTL in cervical vaginal tissues [168], suggesting that a modest vaccine-induced CD8+ T cell response in the context of immune suppression of T cell activation may protect against vaginal HIV-1 transmission [169]. Intriguingly, oral immunisation of macaques with inactivated SIVmac239 induced immune tolerance and elicited CD8+ regulatory T cells (Tregs), completely protecting a majority of animals without inducing SIV-specific antibodies or CTL [170].
Frequency, magnitude and duration of responses
The persistence and boosting of HIV vaccine-induced effector and central memory T cell differentiation as well as of humoral immune responses in the mucosal compartments after a long interval after primary vaccination has not been studied systematically. Long-term maintenance of the memory T cell response is the hallmark of immune protection and, hence, constitutes one of the most important objectives of vaccine-development strategies [171]. Clinical trials show that HIV-1 vaccine-induced immune responses are of modest magnitude and short duration [19]. Several strategies are proposed to remedy these issues. Recently, the RV305 trial vaccinated RV144 vaccine recipients 6–8 years after the original series. The ALVAC-HIV/AIDSVAX B/E combination induced antibody titres higher than the 2-weeks post fourth RV144 vaccination, suggesting an anamnestic response and long-term memory despite the interval between vaccinations [172].
The use of potent adjuvants may also augment and shape antigen-specific antibody responses and contribute to antigen dose sparing. Several adjuvants have been tested in NHP and humans [173] showing a significant benefit of HIV envelope proteins formulated with either MF59 [174] or AS01 [175,176]. In macaques vaccinated with ALVAC-SIV and SIV gp120 in alum or MF59, alum protected macaques from SIVmac251 acquisition while MF59 did not, despite its ability to elicit higher systemic T cell and antibody responses. MF59 altered homing of antibody-producing cells and increased frequency of CXCR3 plasmablasts in blood that positively correlated with anti-envelope IgA serum levels and phagocytosis. Alum, in contrast, increased the frequency of plasmablasts expressing the mucosal integrin a4b7, which positively correlated with IgA responses to cyclic V2 in rectal mucosa. In the alum group, mucosal IgG to cyclic V2 correlated with lower risk of SIVmac251 acquisition. However, mucosal IgG to linear and cyclic V2 correlated with an increased risk of SIVmac251 acquisition in the MF59 group [177]. The formulation of HIV-1 gp120 with L(MPLA) and alum induced significantly higher levels of neutralising antibodies and T cell lymphoproliferation compared to alum, MF59 or MPLA alone [178]. High titres of gp70 V1V2 caseA2 antibodies as early as post second protein administration were elicited. The frequency of antibody response to gp70 V1V2 caseA2 was 100% at 2 weeks post second vaccination and 10 months after the fourth vaccination in the L(MPLA) adjuvant + alum group, while 85% and 100%, respectively, in the alum group. These antibody titres were five- to 10-fold higher than those observed in the group that received the antigen formulated with alum, and were detected at high levels 40 weeks post fourth vaccination and were higher than those observed in RV144. Moreover, antibodies were cross-reactive with gp70 scaffolds for CRF01_AE and subtype C [179]. Formulation of antigens with solid nanoparticles may prolong the duration of antibody responses by increasing antigen deposition/retention locally in the tissue that drives B cell responses. Dendritic cell antigen presentation [180] and development of CD4+ Tfh cells [181] would therefore be enhanced, which would provide critical cytokines and signals required to initiate somatic hypermutation and affinity maturation for effective B cell memory [182].
Modes and routes of administration
The vast majority of HIV-1 vaccine administrations are made by intramuscular injections. DNA constructs induce a better immune response via the intradermal rather than intramuscular or subcutaneous route [183,184]. Electroporation has also been tested with success [185,186]. Immunisation that targets iliac lymph node in macaques with SIV proteins elicited the most consistent mucosal antibody responses in the rectum, vagina, urine, seminal fluid, and blood [187]. Subcutaneous inguinal administration of ALVAC-HIV (vCP205) in humans induced qualitative or quantitative compartmentalisation of immune responses between blood and gut mucosa [188]. Other routes that aim to induce gut and cervico-vaginal mucosal responses remain poorly explored with mixed results including rectal [189–191], vaginal [192,193], oral [194,195], intra-ileal [196], nasal [197,198], aerosol [199], and combined systemic and mucosal [200,201] administrations. Co-delivery of DNA and protein HIV-1 vaccines has been tested in mice and NHP and appears to have an overlap of the prime-boost effect, achieving better immune responses than each component alone [202–204].
Conclusion
Put in perspective, the RV144 CoR analysis and recent NHP challenge studies have generated hypotheses suggesting that Env is essential and perhaps sufficient to induce protective antibody responses against HIV-1 acquisition at the mucosal entry. New prime-boost mosaic and conserved sequences, replicating vector immunisation strategies aimed at inducing immune responses of greater breadth and depth, as well as the development of immunogens inducing broadly neutralising antibodies and mucosal responses are exciting new approaches that will soon enter early-phase testing in humans. Whether the immune CoR identified in RV144 can be extended to other populations with different modes and intensity of transmission and against increasing HIV-1 genetic diversity remains to be demonstrated. Although NHP challenge studies may guide vaccine development, human efficacy trials remain key to answer the critical questions leading to the development of a global HIV-1 vaccine for licensure [205].
Acknowledgements
The preparation of this manuscript was supported in part by an Interagency Agreement Y1-AI-2642-12 between US Army Medical Research and Materiel Command (USAMRMC) and the National Institutes of Allergy and Infectious Diseases. In addition this work was supported by a cooperative agreement (W81XWH-07-2-0067) between the Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., and the US Department of Defense.
Disclaimer
The opinions herein are those of the authors and should not be construed as official or representing the views of the US Department of Defense or Department of the Army.
All authors equally contributed to the preparation of this manuscript.
<>References
- 1. UNAIDS The Gap Report. Geneva, Switzerland: 2014. Available at: http://www.unaids.org/en/resources/documents/2014/20140716_UNAIDS_gap_report ( accessed March 2015).
- 2. Dutta A, Wirtz AL, Baral S et al. Key harm reduction interventions and their impact on the reduction of risky behavior and HIV incidence among people who inject drugs in low-income and middle-income countries. Curr Opin HIV AIDS 2012; 7: 362– 368. [DOI] [PubMed] [Google Scholar]
- 3. Wamai RG, Morris BJ, Bailis SA et al. Male circumcision for HIV prevention: current evidence and implementation in sub-Saharan Africa. J Int AIDS Soc 2011; 14: 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Chi BH, Adler MR, Bolu O et al. Progress, challenges, and new opportunities for the prevention of mother-to-child transmission of HIV under the US President's Emergency Plan for AIDS Relief. J Acquir Immune Defic Syndr 2012; 60 Suppl 3: S78– 87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Kim SC, Becker S, Dieffenbach C et al. Planning for pre-exposure prophylaxis to prevent HIV transmission: challenges and opportunities. J Int AIDS Soc 2010; 13: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Grant RM, Lama JR, Anderson PL et al. Preexposure chemoprophylaxis for HIV prevention in men who have sex with men. N Engl J Med 2010; 363: 2587– 2599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Choopanya K, Martin M, Suntharasamai P et al. Antiretroviral prophylaxis for HIV infection in injecting drug users in Bangkok, Thailand (the Bangkok Tenofovir Study): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2013; 381: 2083– 2090. [DOI] [PubMed] [Google Scholar]
- 8. Mastro TD, Sista N, Abdool-Karim Q.. ARV-based HIV prevention for women – where we are in 2014. J Int AIDS Soc 2014; 17: 19154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Cohen MS, Holmes C, Padian N et al. HIV treatment as prevention: how scientific discovery occurred and translated rapidly into policy for the global response. Health Aff (Millwood) 2012; 31: 1439– 1449. [DOI] [PubMed] [Google Scholar]
- 10. Krakower D, Mayer KH.. Promising prevention approaches: tenofovir gel and prophylactic use of antiretroviral medications. Curr HIV/AIDS Rep 2011; 8: 241– 248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Abdool Karim Q, Abdool Karim SS, Frohlich JA et al. Effectiveness and safety of tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in women. Science 2010; 329: 1168– 1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Fauci AS, Marston HD.. Ending AIDS: is an HIV vaccine necessary? N Engl J Med 2014; 370: 495– 498. [DOI] [PubMed] [Google Scholar]
- 13. Nabel GJ. Designing tomorrow's vaccines. N Engl J Med 2013; 368: 551– 560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Deeks SG, Autran B, Berkhout B et al. Towards an HIV cure: a global scientific strategy. Nat Rev Immunol 2012; 12: 607– 614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Pantophlet R, Wang M, Aguilar-Sino RO, Burton DR.. The human immunodeficiency virus type 1 envelope spike of primary viruses can suppress antibody access to variable regions. J Virol 2009; 83: 1649– 1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Hemelaar J, Gouws E, Ghys PD, Osmanov S.. Global trends in molecular epidemiology of HIV-1 during 2000-2007. AIDS 2011; 25: 679– 689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. McBurney SP, Ross TM.. Viral sequence diversity: challenges for AIDS vaccine designs. Expert Rev Vaccines 2008; 7: 1405– 1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Stamatatos L, Morris L, Burton DR, Mascola JR.. Neutralizing antibodies generated during natural HIV-1 infection: good news for an HIV-1 vaccine? Nat Med 2009; 15: 866– 870. [DOI] [PubMed] [Google Scholar]
- 19. Excler JL, Tomaras GD, Russell ND.. Novel directions in HIV-1 vaccines revealed from clinical trials. Curr Opin HIV AIDS 2013; 8: 421– 431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 2009; 361: 2209– 2220. [DOI] [PubMed] [Google Scholar]
- 21. Walker BD, Ahmed R, Plotkin S.. Moving ahead an HIV vaccine: use both arms to beat HIV. Nat Med 2011; 17: 1194– 1195. [DOI] [PubMed] [Google Scholar]
- 22. O’Connell RJ, Excler JL.. HIV vaccine efficacy and immune correlates of risk. Curr HIV Res 2013; 11: 450– 463. [DOI] [PubMed] [Google Scholar]
- 23. Excler JL, Plotkin S.. The prime-boost concept applied to HIV preventive vaccines. AIDS 1997; 11 Suppl A: S127– 137. [PubMed] [Google Scholar]
- 24. Tartaglia J, Excler JL, El Habib R et al. Canarypox virus-based vaccines: prime-boost strategies to induce cell-mediated and humoral immunity against HIV. AIDS research and human retroviruses 1998; 14 Suppl 3: S291– 298. [PubMed] [Google Scholar]
- 25. Gilbert PB, Ackers ML, Berman PW et al. HIV-1 virologic and immunologic progression and initiation of antiretroviral therapy among HIV-1-infected subjects in a trial of the efficacy of recombinant glycoprotein 120 vaccine. J Infect Dis 2005; 192: 974– 983. [DOI] [PubMed] [Google Scholar]
- 26. Flynn NM, Forthal DN, Harro CD et al. Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection. J Infect Dis 2005; 191: 654– 665. [DOI] [PubMed] [Google Scholar]
- 27. Gilbert PB, Peterson ML, Follmann D et al. Correlation between immunologic responses to a recombinant glycoprotein 120 vaccine and incidence of HIV-1 infection in a phase 3 HIV-1 preventive vaccine trial. J Infect Dis 2005; 191: 666– 677. [DOI] [PubMed] [Google Scholar]
- 28. Schneider JA, Alam SA, Ackers M et al. Mucosal HIV-binding antibody and neutralizing activity in high-risk HIV-uninfected female participants in a trial of HIV-vaccine efficacy. J Infect Dis 2007; 196: 1637– 1644. [DOI] [PubMed] [Google Scholar]
- 29. Forthal DN, Gabriel EE, Wang A et al. Association of Fcgamma receptor IIIa genotype with the rate of HIV infection after gp120 vaccination. Blood 2012; 120: 2836– 2842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Pitisuttithum P, Gilbert P, Gurwith M et al. Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injection drug users in Bangkok, Thailand. J Infect Dis 2006; 194: 1661– 1671. [DOI] [PubMed] [Google Scholar]
- 31. Tongo M, Burgers WA.. Challenges in the design of a T cell vaccine in the context of HIV-1 diversity. Viruses 2014; 6: 3968– 3990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. McDermott AB, Koup RA.. CD8(+) T cells in preventing HIV infection and disease. AIDS 2012; 26: 1281– 1292. [DOI] [PubMed] [Google Scholar]
- 33. McMichael AJ, Haynes BF.. Lessons learned from HIV-1 vaccine trials: new priorities and directions. Nat Immunol 2012; 13: 423– 427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Buchbinder SP, Mehrotra DV, Duerr A et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 2008; 372: 1881– 1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. McElrath MJ, De Rosa SC, Moodie Z et al. HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case-cohort analysis. Lancet 2008; 372: 1894– 1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. D’Souza MP, Frahm N.. Adenovirus 5 serotype vector-specific immunity and HIV-1 infection: a tale of T cells and antibodies. AIDS 2010; 24: 803– 809. [DOI] [PubMed] [Google Scholar]
- 37. Duerr A, Huang Y, Buchbinder S et al. Extended follow-up confirms early vaccine-enhanced risk of HIV acquisition and demonstrates waning effect over time among participants in a randomized trial of recombinant adenovirus HIV vaccine (Step Study). J Infect Dis 2012; 206: 258– 266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Koblin BA, Mayer KH, Noonan E et al. Sexual risk behaviors, circumcision status, and preexisting immunity to adenovirus type 5 among men who have sex with men participating in a randomized HIV-1 vaccine efficacy trial: step study. J Acquir Immune Defic Syndr 2012; 60: 405– 413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Curlin ME, Cassis-Ghavami F, Magaret AS et al. Serological immunity to adenovirus serotype 5 is not associated with risk of HIV infection: a case-control study. AIDS 2011; 25: 153– 158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Cheng C, Wang L, Gall JG et al. Decreased pre-existing Ad5 capsid and Ad35 neutralizing antibodies increase HIV-1 infection risk in the Step trial independent of vaccination. PLoS One 2012; 7: e33969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Rolland M, Tovanabutra S, deCamp AC et al. Genetic impact of vaccination on breakthrough HIV-1 sequences from the STEP trial. Nat Med 2011; 17: 366– 371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Janes H, Frahm N, DeCamp A et al. MRKAd5 HIV-1 Gag/Pol/Nef vaccine-induced T-cell responses inadequately predict distance of breakthrough HIV-1 sequences to the vaccine or viral load. PLoS One 2012; 7: e43396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Fitzgerald DW, Janes H, Robertson M et al. An Ad5-vectored HIV-1 vaccine elicits cell-mediated immunity but does not affect disease progression in HIV-1-infected male subjects: results from a randomized placebo-controlled trial (the Step study). J Infect Dis 2011; 203: 765– 772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Janes H, Friedrich DP, Krambrink A et al. Vaccine-induced gag-specific T cells are associated with reduced viremia after HIV-1 infection. J Infect Dis 2013; 208: 1231– 1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Huang Y, Duerr A, Frahm N et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial reveals an association of nonspecific interferon-gamma secretion with increased HIV-1 infection risk: a cohort-based modeling study. PLoS One 2014; 9: e108631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Li F, Finnefrock AC, Dubey SA et al. Mapping HIV-1 vaccine induced T-cell responses: bias towards less-conserved regions and potential impact on vaccine efficacy in the Step study. PLoS One 2011; 6: e20479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Reynolds MR, Weiler AM, Piaskowski SM et al. A trivalent recombinant Ad5 gag/pol/nef vaccine fails to protect rhesus macaques from infection or control virus replication after a limiting-dose heterologous SIV challenge. Vaccine 2012; 30: 4465– 4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Qureshi H, Ma ZM, Huang Y et al. Low-dose penile SIVmac251 exposure of rhesus macaques infected with adenovirus type 5 (Ad5) and then immunized with a replication-defective Ad5-based SIV gag/pol/nef vaccine recapitulates the results of the phase IIb step trial of a similar HIV-1 vaccine. J Virol 2012; 86: 2239– 2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Dhesi Z, Stebbing J.. The HVTN 503/Phambili study: efficacy is always the issue. Lancet Infect Dis 2011; 11: 490– 491. [DOI] [PubMed] [Google Scholar]
- 50. Gray GE, Allen M, Moodie Z et al. Safety and efficacy of the HVTN 503/Phambili study of a clade-B-based HIV-1 vaccine in South Africa: a double-blind, randomised, placebo-controlled test-of-concept phase 2b study. Lancet Infect Dis 2011; 11: 507– 515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Koup RA, Roederer M, Lamoreaux L et al. Priming immunization with DNA augments immunogenicity of recombinant adenoviral vectors for both HIV-1 specific antibody and T-cell responses. PLoS One 2010; 5: e9015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Kibuuka H, Kimutai R, Maboko L et al. A phase 1/2 study of a multiclade HIV-1 DNA plasmid prime and recombinant adenovirus serotype 5 boost vaccine in HIV-Uninfected East Africans (RV 172). J Infect Dis 2010; 201: 600– 607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Jaoko W, Karita E, Kayitenkore K et al. Safety and immunogenicity study of Multiclade HIV-1 adenoviral vector vaccine alone or as boost following a multiclade HIV-1 DNA vaccine in Africa. PLoS One 2010; 5: e12873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Churchyard GJ, Morgan C, Adams E et al. A phase IIA randomized clinical trial of a multiclade HIV-1 DNA prime followed by a multiclade rAd5 HIV-1 vaccine boost in healthy adults (HVTN204). PLoS One 2011; 6: e21225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Hammer SM, Sobieszczyk ME, Janes H et al. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N Engl J Med 2013; 369: 2083– 2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Letvin NL, Rao SS, Montefiori DC et al. Immune and genetic correlates of vaccine protection against mucosal infection by SIV in monkeys. Sci Transl Med 2011; 3: 81ra36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Excler JL, Robb ML, Kim JH.. HIV-1 vaccines: challenges and new perspectives. Hum Vaccin Immunother 2014; 10: 1734– 1746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Wohlfart C. Neutralization of adenoviruses: kinetics, stoichiometry, and mechanisms. J Virol 1988; 62: 2321– 2328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Farina SF, Gao GP, Xiang ZQ et al. Replication-defective vector based on a chimpanzee adenovirus. J Virol 2001; 75: 11603– 11613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Reyes-Sandoval A, Fitzgerald JC, Grant R et al. Human immunodeficiency virus type 1-specific immune responses in primates upon sequential immunization with adenoviral vaccine carriers of human and simian serotypes. J Virol 2004; 78: 7392– 7399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Hu H, Eller MA, Zafar S et al. Preferential infection of human Ad5-specific CD4 T cells by HIV in Ad5 naturally exposed and recombinant Ad5-HIV vaccinated individuals. Proc Natl Acad Sci U S A 2014; 111: 13439– 13444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Baden LR, Liu J, Li H et al. Induction of HIV-1-Specific Mucosal Immune Responses Following Intramuscular Recombinant Adenovirus Serotype 26 HIV-1 Vaccination of Humans. J Infect Dis 2015; 211: 518– 528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Fauci AS, Marovich MA, Dieffenbach CW et al. Immunology. Immune activation with HIV vaccines. Science 2014; 344: 49– 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Carnathan DG, Wetzel KS, Yu J et al. Activated CD4+CCR5+ T cells in the rectum predict increased SIV acquisition in SIVGag/Tat-vaccinated rhesus macaques. Proc Natl Acad Sci U S A 2015; 112: 518– 523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Pitisuttithum P, Rerks-Ngarm S, Bussaratid V et al. Safety and reactogenicity of canarypox ALVAC-HIV (vCP1521) and HIV-1 gp120 AIDSVAX B/E vaccination in an efficacy trial in Thailand. PLoS One 2011; 6: e27837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Kim JH, Excler JL, Michael NL.. Lessons from the RV144 Thai Phase III HIV-1 Vaccine Trial and the search for correlates of protection. Ann Rev Med 2015; 66: 423– 437. [DOI] [PubMed] [Google Scholar]
- 67. Gilbert PB, Berger JO, Stablein D et al. Statistical interpretation of the RV144 HIV vaccine efficacy trial in Thailand: a case study for statistical issues in efficacy trials. J Infect Dis 2011; 203: 969– 975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Robb ML, Rerks-Ngarm S, Nitayaphan S et al. Risk behaviour and time as covariates for efficacy of the HIV vaccine regimen ALVAC-HIV (vCP1521) and AIDSVAX B/E: a post-hoc analysis of the Thai phase 3 efficacy trial RV 144. Lancet Infect Dis 2012; 12: 531– 537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Rerks-Ngarm S, Paris RM, Chunsutthiwat S et al. Extended evaluation of the virologic, immunologic, and clinical course of volunteers who acquired HIV-1 infection in a phase III vaccine trial of ALVAC-HIV and AIDSVAX B/E. J Infect Dis 2013; 207: 1195– 1205. [DOI] [PubMed] [Google Scholar]
- 70. Karnasuta C, Paris RM, Cox JH et al. Antibody-dependent cell-mediated cytotoxic responses in participants enrolled in a phase I/II ALVAC-HIV/AIDSVAX B/E prime-boost HIV-1 vaccine trial in Thailand. Vaccine 2005; 23: 2522– 2529. [DOI] [PubMed] [Google Scholar]
- 71. Nitayaphan S, Pitisuttithum P, Karnasuta C et al. Safety and immunogenicity of an HIV subtype B and E prime-boost vaccine combination in HIV-negative Thai adults. J Infect Dis 2004; 190: 702– 706. [DOI] [PubMed] [Google Scholar]
- 72. Arthos J, Cicala C, Martinelli E et al. HIV-1 envelope protein binds to and signals through integrin alpha4beta7, the gut mucosal homing receptor for peripheral T cells. Nat Immunol 2008; 9: 301– 309. [DOI] [PubMed] [Google Scholar]
- 73. Nawaz F, Cicala C, Van Ryk D et al. The genotype of early-transmitting HIV gp120s promotes alpha (4) beta(7)-reactivity, revealing alpha (4) beta(7) +/CD4+ T cells as key targets in mucosal transmission. PLoS Pathog 2011; 7: e1001301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Cicala C, Martinelli E, McNally JP et al. The integrin alpha4beta7 forms a complex with cell-surface CD4 and defines a T-cell subset that is highly susceptible to infection by HIV-1. Proc Natl Acad Sci U S A 2009; 106: 20877– 20882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Karasavvas N, Billings E, Rao M et al. The Thai Phase III HIV Type 1 Vaccine trial (RV144) regimen induces antibodies that target conserved regions within the V2 loop of gp120. AIDS research and human retroviruses 2012; 28: 1444– 1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Souza MS, Ratto-Kim S, Chuenarom W et al. The Thai phase III trial (RV144) vaccine regimen induces T cell responses that preferentially target epitopes within the V2 region of HIV-1 envelope. J Immunol 2012; 188: 5166– 5176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Montefiori DC, Karnasuta C, Huang Y et al. Magnitude and breadth of the neutralizing antibody response in the RV144 and Vax003 HIV-1 vaccine efficacy trials. J Infect Dis 2012; 206: 431– 441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Plotkin SA. Vaccines: correlates of vaccine-induced immunity. Clin Infect Dis 2008; 47: 401– 409. [DOI] [PubMed] [Google Scholar]
- 79. Plotkin SA. Complex correlates of protection after vaccination. Clin Infect Dis 2013; 56: 1458– 1465. [DOI] [PubMed] [Google Scholar]
- 80. Plotkin SA, Gilbert PB.. Nomenclature for immune correlates of protection after vaccination. Clin Infect Dis 2012; 54: 1615– 1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Qin L, Gilbert PB, Corey L et al. A framework for assessing immunological correlates of protection in vaccine trials. J Infect Dis 2007; 196: 1304– 1312. [DOI] [PubMed] [Google Scholar]
- 82. Haynes BF, Gilbert PB, McElrath MJ et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 2012; 366: 1275– 1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Zolla-Pazner S, Decamp A, Gilbert PB et al. Vaccine-induced IgG antibodies to V1V2 regions of multiple HIV-1 subtypes correlate with decreased risk of HIV-1 infection. PLoS One 2014; 9: e87572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Gottardo R, Bailer RT, Korber BT et al. Plasma IgG to linear epitopes in the V2 and V3 regions of HIV-1 gp120 correlate with a reduced risk of infection in the RV144 Vaccine Efficacy Trial. PLoS One 2013; 8: e75665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Rolland M, Edlefsen PT, Larsen BB et al. Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2. Nature 2012; 490: 417– 420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Liao HX, Bonsignori M, Alam SM et al. Vaccine induction of antibodies against a structurally heterogeneous site of immune pressure within HIV-1 envelope protein variable regions 1 and 2. Immunity 2013; 38: 176– 186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Liu P, Yates NL, Shen X et al. Infectious virion capture by HIV-1 gp120-specific IgG from RV144 vaccinees. J Virol 2013; 87: 7828– 7836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Gorse GJ, Patel GB, Mandava M et al. MN and IIIB recombinant glycoprotein 120 vaccine-induced binding antibodies to native envelope glycoprotein of human immunodeficiency virus type 1 primary isolates. National Institute of Allergy and Infectious Disease Aids Vaccine Evaluation Group. AIDS Res Hum Retrovirus 1999; 15: 921– 930. [DOI] [PubMed] [Google Scholar]
- 89. Banerjee K, Klasse PJ, Sanders RW et al. IgG subclass profiles in infected HIV type 1 controllers and chronic progressors and in uninfected recipients of Env vaccines. AIDS Res Hum Retrovirus 2010; 26: 445– 458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Roussilhon C, Oeuvray C, Muller-Graf C et al. Long-term clinical protection from falciparum malaria is strongly associated with IgG3 antibodies to merozoite surface protein 3. PLoS Med 2007; 4: e320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Tebo AE, Kremsner PG, Luty AJ.. Plasmodium falciparum: a major role for IgG3 in antibody-dependent monocyte-mediated cellular inhibition of parasite growth in vitro. Exp Parasitol 2001; 98: 20– 28. [DOI] [PubMed] [Google Scholar]
- 92. Kam YW, Simarmata D, Chow A et al. Early appearance of neutralizing immunoglobulin G3 antibodies is associated with chikungunya virus clearance and long-term clinical protection. J Infect Dis 2012; 205: 1147– 1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Ljunggren K, Broliden PA, Morfeldt-Manson L et al. IgG subclass response to HIV in relation to antibody-dependent cellular cytotoxicity at different clinical stages. Clin Exp Immunol 1988; 73: 343– 347. [PMC free article] [PubMed] [Google Scholar]
- 94. Cavacini LA, Kuhrt D, Duval M et al. Binding and neutralization activity of human IgG1 and IgG3 from serum of HIV-infected individuals. AIDS Res Hum Retrovirus 2003; 19: 785– 792. [DOI] [PubMed] [Google Scholar]
- 95. Yates NL, Liao HX, Fong Y et al. Vaccine-induced Env V1-V2 IgG3 correlates withlower HIV-1 infection risk and declines soon after vaccination. Sci Transl Med 2014; 6: 228ra239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Chung AW, Ghebremichael M, Robinson H et al. Polyfunctional Fc-effector profiles mediated by IgG Subclass selection distinguish RV144 and VAX003 vaccines. Sci Transl Med 2014; 6: 228ra238. [DOI] [PubMed] [Google Scholar]
- 97. Teigler JE, Phogat S, Franchini G et al. The canarypox virus vector ALVAC induces distinct cytokine responses compared to the Vaccinia virus-based vectors MVA and NYVAC in Rhesus monkeys. J Virol 2014; 88: 1809- 1814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Forthal D, Hope TJ, Alter G.. New paradigms for functional HIV-specific nonneutralizing antibodies. Curr Opin HIV AIDS 2013; 8: 393– 401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Vargas-Inchaustegui DA, Robert-Guroff M.. Fc receptor-mediated immune responses: new tools but increased complexity in HIV prevention. Curr HIV Res 2013; 11: 407– 420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Li SS, Gilbert PB, Tomaras GD et al. FCGR2C polymorphisms associate with HIV-1 vaccine protection in RV144 trial. J Clin Invest 2014; 124: 3879– 3890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Zhou M, Ruprecht RM.. Are anti-HIV IgAs good guys or bad guys? Retrovirology 2014; 11: 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Griffiss JM, Goroff DK.. IgA blocks IgM and IgG-initiated immune lysis by separate molecular mechanisms. J Immunol 1983; 130: 2882– 2885. [PubMed] [Google Scholar]
- 103. Nikolova EB, Russell MW.. Dual function of human IgA antibodies: inhibition of phagocytosis in circulating neutrophils and enhancement of responses in IL-8-stimulated cells. J Leukoc Biol 1995; 57: 875– 882. [DOI] [PubMed] [Google Scholar]
- 104. Mathew GD, Qualtiere LF, Neel HB 3rd, Pearson GR.. IgA antibody, antibody-dependent cellular cytotoxicity and prognosis in patients with nasopharyngeal carcinoma. Int J Cancer 1981; 27: 175– 180. [DOI] [PubMed] [Google Scholar]
- 105. Tomaras GD, Ferrari G, Shen X et al. Vaccine-induced plasma IgA specific for the C1 region of the HIV-1 envelope blocks binding and effector function of IgG. Proc Natl Acad Sci U S A 2013; 110: 9019– 9024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Bonsignori M, Pollara J, Moody MA et al. Antibody-dependent cellular cytotoxicity-mediating antibodies from an HIV-1 vaccine efficacy trial target multiple epitopes and preferentially use the VH1 gene family. J Virol 2012; 86: 11521– 11532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Pollara J, Bonsignori M, Moody MA et al. HIV-1 vaccine-induced C1 and V2 Env-specific antibodies synergize for increased antiviral activities. J Virol 2014; 88: 7715– 7726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Stieh DJ, King DF, Klein K et al. Aggregate complexes of HIV-1 induced by multimeric antibodies. Retrovirology 2014; 11: 78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Li Q, Zeng M, Duan L et al. Live simian immunodeficiency virus vaccine correlate of protection: local antibody production and concentration on the path of virus entry. J Immunol 2014; 193: 3113– 3125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Su B, Moog C.. Which antibody functions are important for an HIV vaccine? Front Immunol 2014; 5: 289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Excler JL, Ake J, Robb ML et al. Nonneutralizing functional antibodies: a new “old” paradigm for HIV vaccines. Clin Vaccine Immunol 2014; 21: 1023– 1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Barouch DH, Liu J, Li H et al. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys. Nature 2012; 482: 89– 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Barouch DH, Stephenson KE, Borducchi EN et al. Protective efficacy of a global HIV-1 mosaic vaccine against heterologous SHIV challenges in Rhesus monkeys. Cell 2013; 155: 531– 539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Roederer M, Keele BF, Schmidt SD et al. Immunological and virological mechanisms of vaccine-mediated protection against SIV and HIV. Nature 2014; 505: 502– 508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Bomsel M, Tudor D, Drillet AS et al. Immunization with HIV-1 gp41 subunit virosomes induces mucosal antibodies protecting nonhuman primates against vaginal SHIV challenges. Immunity 2011; 34: 269– 280. [DOI] [PubMed] [Google Scholar]
- 116. Watkins JD, Sholukh AM, Mukhtar MM et al. Anti-HIV IgA isotypes: differential virion capture and inhibition of transcytosis are linked to prevention of mucosal R5 SHIV transmission. AIDS 2013; 27: F13– 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Hidajat R, Xiao P, Zhou Q et al. Correlation of vaccine-elicited systemic and mucosal nonneutralizing antibody activities with reduced acute viremia following intrarectal simian immunodeficiency virus SIVmac251 challenge of rhesus macaques. J Virol 2009; 83: 791– 801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Xiao P, Zhao J, Patterson LJ et al. Multiple vaccine-elicited nonneutralizing antienvelope antibody activities contribute to protective efficacy by reducing both acute and chronic viremia following simian/human immunodeficiency virus SHIV89.6P challenge in rhesus macaques. J Virol 2010; 84: 7161– 7173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Leroux-Roels G, Maes C, Clement F et al. Randomized Phase I: safety, immunogenicity and mucosal antiviral activity in young healthy women vaccinated with HIV-1 Gp41 P1 Peptide on Virosomes. PLoS One 2013; 8: e55438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Devito C, Broliden K, Kaul R et al. Mucosal and plasma IgA from HIV-1-exposed uninfected individuals inhibit HIV-1 transcytosis across human epithelial cells. J Immunol 2000; 165: 5170– 5176. [DOI] [PubMed] [Google Scholar]
- 121. Shukair SA, Allen SA, Cianci GC et al. Human cervicovaginal mucus contains an activity that hinders HIV-1 movement. Mucosal Immunol 2013; 6: 427– 434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Fahrbach KM, Malykhina O, Stieh DJ, Hope TJ.. Differential binding of IgG and IgA to mucus of the female reproductive tract. PLoS One 2013; 8: e76176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Shaw GM, Hunter E.. HIV transmission. Cold Spring Harb Perspect Med 2012; 2: ii. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Florese RH, Demberg T, Xiao P et al. Contribution of nonneutralizing vaccine-elicited antibody activities to improved protective efficacy in rhesus macaques immunized with Tat/Env compared with multigenic vaccines. J Immunol 2009; 182: 3718– 3727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Hessell AJ, Hangartner L, Hunter M et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature 2007; 449: 101– 104. [DOI] [PubMed] [Google Scholar]
- 126. Holl V, Peressin M, Moog C.. Antibody-mediated Fcgamma receptor-based mechanisms of HIV inhibition: recent findings and new vaccination strategies. Viruses 2009; 1: 1265– 1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Forthal DN, Gilbert PB, Landucci G, Phan T.. Recombinant gp120 vaccine-induced antibodies inhibit clinical strains of HIV-1 in the presence of Fc receptor-bearing effector cells and correlate inversely with HIV infection rate. J Immunol 2007; 178: 6596– 6603. [DOI] [PubMed] [Google Scholar]
- 128. Hu Q, Frank I, Williams V et al. Blockade of attachment and fusion receptors inhibits HIV-1 infection of human cervical tissue. J Exp Med 2004; 199: 1065– 1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Fletcher PS, Elliott J, Grivel JC et al. Ex vivo culture of human colorectal tissue for the evaluation of candidate microbicides. AIDS 2006; 20: 1237– 1245. [DOI] [PubMed] [Google Scholar]
- 130. Herrera C, Schuetz A, Olejniczak N et al. Preliminary evaluation of mucosal immune responses with mucosal tissue explants in humans vaccinated with ALVAC/AIDSVAX B/E during the ongoing RV305 trial. AIDS Vaccine 2013. October 2013. Barcelona, Spain. Abstract P08.26 B.
- 131. Alam SM, Liao HX, Tomaras GD et al. Antigenicity and immunogenicity of RV144 vaccine AIDSVAX clade E envelope immunogen is enhanced by a gp120 N-terminal deletion. J Virol 2013; 87: 1554– 1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Burton DR, Ahmed R, Barouch DH et al. A blueprint for HIV vaccine discovery. Cell Host Microbe 2012; 12: 396– 407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. McCoy LE, Weiss RA.. Neutralizing antibodies to HIV-1 induced by immunization. J Exp Med 2013; 210: 209– 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Huang X, Jin W, Hu K et al. Highly conserved HIV-1 gp120 glycans proximal to CD4-binding region affect viral infectivity and neutralizing antibody induction. Virology 2012; 423: 97– 106. [DOI] [PubMed] [Google Scholar]
- 135. Quinnan GV Jr., Zhang P, Dong M et al. Neutralizing antibody responses in macaques induced by human immunodeficiency virus type 1 monovalent or trivalent envelope glycoproteins. PLoS One 2013; 8: e59803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Kovacs JM, Nkolola JP, Peng H et al. HIV-1 envelope trimer elicits more potent neutralizing antibody responses than monomeric gp120. Proc Natl Acad Sci U S A 2012; 109: 12111– 12116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Bricault CA, Kovacs JM, Nkolola JP et al. A multivalent clade C HIV-1 Env trimer cocktail elicits a higher magnitude of neutralizing antibodies than any individual component. J Virol 2015; 89: 2507– 2519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Santra S, Muldoon M, Watson S et al. Breadth of cellular and humoral immune responses elicited in rhesus monkeys by multi-valent mosaic and consensus immunogens. Virology 2012; 428: 121– 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Mascola JR, Haynes BF.. HIV-1 neutralizing antibodies: understanding nature's pathways. Immunol Rev 2013; 254: 225– 244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Bonsignori M, Alam SM, Liao HX et al. HIV-1 antibodies from infection and vaccination: insights for guiding vaccine design. Trends Microbiol 2012; 20: 532– 539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Liao HX, Lynch R, Zhou T et al. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 2013; 496: 469– 476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Johnson PR, Schnepp BC, Zhang J et al. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat Med 2009; 15: 901– 906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Schnepp BC, Johnson PR.. Adeno-associated virus delivery of broadly neutralizing antibodies. Curr Opin HIV AIDS 2014; 9: 250– 256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Balazs AB, Chen J, Hong CM et al. Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature 2012; 481: 81– 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Balazs AB, Ouyang Y, Hong CM et al. Vectored immunoprophylaxis protects humanized mice from mucosal HIV transmission. Nat Med 2014; 20: 296– 300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. McMichael AJ, Borrow P, Tomaras GD et al. The immune response during acute HIV-1 infection: clues for vaccine development. Nat Rev Immunol 2010; 10: 11– 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Koup RA, Graham BS, Douek DC.. The quest for a T cell-based immune correlate of protection against HIV: a story of trials and errors. Nat Rev Immunol 2011; 11: 65– 70. [DOI] [PubMed] [Google Scholar]
- 148. Liu J, O’Brien KL, Lynch DM et al. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 2009; 457: 87– 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Santra S, Liao HX, Zhang R et al. Mosaic vaccines elicit CD8+ T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys. Nat Med 2010; 16: 324– 328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Yue L, Pfafferott KJ, Baalwa J et al. Transmitted virus fitness and host T cell responses collectively define divergent infection outcomes in two HIV-1 recipients. PLoS Pathog 2015; 11: e1004565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Letourneau S, Im EJ, Mashishi T et al. Design and pre-clinical evaluation of a universal HIV-1 vaccine. PLoS One 2007; 2: e984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Rolland M, Nickle DC, Mullins JI.. HIV-1 group M conserved elements vaccine. PLoS Pathog 2007; 3: e157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Korber BT, Letvin NL, Haynes BF.. T-cell vaccine strategies for human immunodeficiency virus, the virus with a thousand faces. J Virol 2009; 83: 8300– 8314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Liu Y, McNevin J, Rolland M et al. Conserved HIV-1 epitopes continuously elicit subdominant cytotoxic T-lymphocyte responses. J Infect Dis 2009; 200: 1825– 1833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Kulkarni V, Valentin A, Rosati M et al. HIV-1 conserved elements p24CE DNA vaccine induces humoral immune responses with broad epitope recognition in macaques. PLoS One 2014; 9: e111085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Clutton G, Carpov A, Parks CL et al. Optimizing parallel induction of HIV type 1-specific antibody and T-cell responses by multicomponent subunit vaccines. AIDS 2014; 28: 2495– 2504. [DOI] [PubMed] [Google Scholar]
- 157. Ondondo B, Abdul-Jawad S, Bridgeman A, Hanke T.. Characterization of T-cell responses to conserved regions of the HIV-1 proteome in BALB/c mice. Clin Vaccine Immunol 2014; 21: 1565– 1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Koopman G, Beenhakker N, Nieuwenhuis I et al. DNA/long peptide vaccination against conserved regions of SIV induces partial protection against SIVmac251 challenge. AIDS 2013; 27: 2841– 2851. [DOI] [PubMed] [Google Scholar]
- 159. Hayton EJ, Rose A, Ibrahimsa U et al. Safety and tolerability of conserved region vaccines vectored by plasmid DNA, simian adenovirus and modified vaccinia virus ankara administered to human immunodeficiency virus type 1-uninfected adults in a randomized, single-blind phase I trial. PLoS One 2014; 9: e101591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Borthwick N, Ahmed T, Ondondo B et al. Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther 2014; 22: 464– 475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Barouch DH, O’Brien KL, Simmons NL et al. Mosaic HIV-1 vaccines expand the breadth and depth of cellular immune responses in rhesus monkeys. Nat Med 2010; 16: 319– 323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Parks CL, Picker LJ, King CR.. Development of replication-competent viral vectors for HIV vaccine delivery. Curr Opin HIV AIDS 2013; 8: 402– 411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Excler JL, Parks CL, Ackland J et al. Replicating viral vectors as HIV vaccines: summary report from the IAVI-sponsored satellite symposium at the AIDS vaccine 2009 conference. Biologicals 2010; 38: 511– 521. [DOI] [PubMed] [Google Scholar]
- 164. Hansen SG, Powers CJ, Richards R et al. Evasion of CD8+ T cells is critical for superinfection by cytomegalovirus. Science 2010; 328: 102– 106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Hansen SG, Ford JC, Lewis MS et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 2011; 473: 523– 527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Hansen SG, Piatak M Jr., Ventura AB et al. Immune clearance of highly pathogenic SIV infection. Nature 2013; 502: 100– 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Hansen SG, Sacha JB, Hughes CM et al. Cytomegalovirus vectors violate CD8+ T cell epitope recognition paradigms. Science 2013; 340: 1237874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Genesca M, Skinner PJ, Bost KM et al. Protective attenuated lentivirus immunization induces SIV-specific T cells in the genital tract of rhesus monkeys. Mucosal Immunol 2008; 1: 219– 228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Genesca M, Ma ZM, Wang Y et al. Live-attenuated lentivirus immunization modulates innate immunity and inflammation while protecting rhesus macaques from vaginal simian immunodeficiency virus challenge. J Virol 2012; 86: 9188– 9200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Lu W, Chen S, Lai C et al. Induction of CD8+ regulatory T cells protects macaques against SIV challenge. Cell Rep 2012; 2: 1736– 1746. [DOI] [PubMed] [Google Scholar]
- 171. Tanel A, Fonseca SG, Yassine-Diab B et al. Cellular and molecular mechanisms of memory T-cell survival. Expert Rev Vaccines 2009; 8: 299– 312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Karasavvas N. Investigation of antibody responses induced in RV305 a late boost vaccination of HIV-1 uninfected volunteers that participated in RV144, a Thai trial. AIDS Vaccine 2013. Barcelona, Spain, 2013. Abstract P03.68 LB
- 173. Mastelic B, Garcon N, Del Giudice G et al. Predictive markers of safety and immunogenicity of adjuvanted vaccines. Biologicals 2013; 41: 458– 468. [DOI] [PubMed] [Google Scholar]
- 174. O’Hagan DT, Ott GS, De Gregorio E, Seubert A.. The mechanism of action of MF59 – an innately attractive adjuvant formulation. Vaccine 2012; 30: 4341– 4348. [DOI] [PubMed] [Google Scholar]
- 175. Leroux-Roels I, Koutsoukos M, Clement F et al. Strong and persistent CD4+ T-cell response in healthy adults immunized with a candidate HIV-1 vaccine containing gp120, Nef and Tat antigens formulated in three adjuvant systems. Vaccine 2010; 28: 7016– 7024. [DOI] [PubMed] [Google Scholar]
- 176. Garcon N, Van Mechelen M.. Recent clinical experience with vaccines using MPL- and QS-21-containing adjuvant systems. Expert Rev Vaccines 2011; 10: 471– 486. [DOI] [PubMed] [Google Scholar]
- 177. Vaccari M, Gordon SN, Fourati S et al. Adjuvant dependent mucosal V2 responses and RAS activation in vaccine induced protection from SIVmac251 acquisition. HIV R4P. Cape Town, South Africa, 2014. Abstract OA25.01.
- 178. McElrath MJ. Selection of potent immunological adjuvants for vaccine construction. Semin Cancer Biol 1995; 6: 375– 385. [DOI] [PubMed] [Google Scholar]
- 179. Rao M, Onkar S, Peachman K et al. Potent V2-specific antibodies induced in humans using liposome-encapsulated HIV-1 gp120 recognize a well-exposed V2 epitope on envelope trimer. Keystone Conferences HIV Vaccines: Adaptive Immunity and Beyond. March 2014. Banff, Alberta, Canada. Abstract 2049.
- 180. Moon JJ, Suh H, Bershteyn A et al. Interbilayer-crosslinked multilamellar vesicles as synthetic vaccines for potent humoral and cellular immune responses. Nat Mater 2011; 10: 243– 251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Zhao L, Seth A, Wibowo N et al. Nanoparticle vaccines. Vaccine 2014; 32: 327– 337. [DOI] [PubMed] [Google Scholar]
- 182. Nutt SL, Tarlinton DM.. Germinal center B and follicular helper T cells: siblings, cousins or just good friends? Nat Immunol 2011; 12: 472– 477. [DOI] [PubMed] [Google Scholar]
- 183. Bakari M, Aboud S, Nilsson C et al. Broad and potent immune responses to a low dose intradermal HIV-1 DNA boosted with HIV-1 recombinant MVA among healthy adults in Tanzania. Vaccine 2011; 29: 8417– 8428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Ledgerwood JE, Graham BS.. DNA vaccines: a safe and efficient platform technology for responding to emerging infectious diseases. Hum Vaccin 2009; 5: 623– 626. [DOI] [PubMed] [Google Scholar]
- 185. Vasan S, Hurley A, Schlesinger SJ et al. In vivo electroporation enhances the immunogenicity of an HIV-1 DNA vaccine candidate in healthy volunteers. PLoS One 2011; 6: e19252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Villarreal DO, Talbott KT, Choo DK et al. Synthetic DNA vaccine strategies against persistent viral infections. Expert Rev Vaccines 2013; 12: 537– 554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Lehner T, Wang Y, Ping L et al. The effect of route of immunization on mucosal immunity and protection. J Infect Dis 1999; 179 Suppl 3: S489– s492. [DOI] [PubMed] [Google Scholar]
- 188. Yang OO, Ibarrondo FJ, Price C et al. Differential blood and mucosal immune responses against an HIV-1 vaccine administered via inguinal or deltoid injection. PLoS One 2014; 9: e88621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Belyakov IM, Wyatt LS, Ahlers JD et al. Induction of a mucosal cytotoxic T-lymphocyte response by intrarectal immunization with a replication-deficient recombinant vaccinia virus expressing human immunodeficiency virus 89.6 envelope protein. J Virol 1998; 72: 8264– 8272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Wang SW, Bertley FM, Kozlowski PA et al. An SHIV DNA/MVA rectal vaccination in macaques provides systemic and mucosal virus-specific responses and protection against AIDS. AIDS Res Hum Retrovirus 2004; 20: 846– 859. [DOI] [PubMed] [Google Scholar]
- 191. Yu M, Vajdy M. Mucosal HIV transmission and vaccination strategies through oral compared with vaginal and rectal routes. Expert Opin Biol Ther 2010; 10: 1181– 1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Donnelly L, Curran RM, Tregoning JS et al. Intravaginal immunization using the recombinant HIV-1 clade-C trimeric envelope glycoprotein CN54gp140 formulated within lyophilized solid dosage forms. Vaccine 2011; 29: 4512– 4520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Cranage MP, Fraser CA, Cope A et al. Antibody responses after intravaginal immunisation with trimeric HIV-1 CN54 clade C gp140 in Carbopol gel are augmented by systemic priming or boosting with an adjuvanted formulation. Vaccine 2011; 29: 1421– 1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Ko SY, Cheng C, Kong WP et al. Enhanced induction of intestinal cellular immunity by oral priming with enteric adenovirus 41 vectors. J Virol 2009; 83: 748– 756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Appledorn DM, Aldhamen YA, Godbehere S et al. Sublingual administration of an adenovirus serotype 5 (Ad5)-based vaccine confirms Toll-like receptor agonist activity in the oral cavity and elicits improved mucosal and systemic cell-mediated responses against HIV antigens despite preexisting Ad5 immunity. Clin Vaccine Immunol 2011; 18: 150– 160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Wang L, Cheng C, Ko SY et al. Delivery of human immunodeficiency virus vaccine vectors to the intestine induces enhanced mucosal cellular immunity. J Virol 2009; 83: 7166– 7175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Barnett SW, Srivastava IK, Kan E et al. Protection of macaques against vaginal SHIV challenge by systemic or mucosal and systemic vaccinations with HIV-envelope. AIDS 2008; 22: 339– 348. [DOI] [PubMed] [Google Scholar]
- 198. Kaufman DR, Bivas-Benita M, Simmons NL et al. Route of adenovirus-based HIV-1 vaccine delivery impacts the phenotype and trafficking of vaccine-elicited CD8+ T lymphocytes. J Virol 2010; 84: 5986– 5996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Bolton DL, Song K, Wilson RL et al. Comparison of systemic and mucosal vaccination: impact on intravenous and rectal SIV challenge. Mucosal Immunol 2012; 5: 41– 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Srivastava I, Goodsell A, Zhou F et al. Dynamics of acute and memory mucosal and systemic immune responses against HIV-1 envelope following immunizations through single or combinations of mucosal and systemic routes. Vaccine 2008; 26: 2796– 2806. [DOI] [PubMed] [Google Scholar]
- 201. Karita E, Anzala O, Gazzard B et al. Clinical safety and immunogenicity of two HIV vaccines SeV-G (NP) and Ad35-GRIN in HIV-uninfected, healthy adult volunteers. HIV R4P 2014. Cape Town, South Africa, 2014. Abstract PD03.04 LB.
- 202. Pissani F, Malherbe DC, Schuman JT et al. Improvement of antibody responses by HIV envelope DNA and protein co-immunization. Vaccine 2014; 32: 507– 513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Jalah R, Kulkarni V, Patel V et al. DNA and protein co-immunization improves the magnitude and longevity of humoral immune responses in macaques. PLoS One 2014; 9: e91550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Patel V, Jalah R, Kulkarni V et al. DNA and virus particle vaccination protects against acquisition and confers control of viremia upon heterologous simian immunodeficiency virus challenge. Proc Natl Acad Sci U S A 2013; 110: 2975– 2980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Barouch DH, Michael NL.. Accelerating HIV-1 vaccine efficacy trials. Cell 2014; 159: 969– 972. [DOI] [PMC free article] [PubMed] [Google Scholar]