Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Aug 1.
Published in final edited form as: Curr Opin Immunol. 2016 Jun 3;41:39–46. doi: 10.1016/j.coi.2016.05.011

New Concepts in HIV-1 Vaccine Development

Kathryn E Stephenson a,b, Helen T D’Couto a, Dan H Barouch a,b,#
PMCID: PMC4992607  NIHMSID: NIHMS791144  PMID: 27268856

Abstract

With 2 million people newly infected with HIV-1 in 2014, an effective HIV-1 vaccine remains a major public health priority. HIV-1 vaccine efficacy trials in humans, complemented by active and passive immunization studies in non-human primates, have identified several key vaccine-induced immunological responses that may correlate with protection against HIV-1 infection. Potential correlates of protection in these studies include V2-specific, polyfunctional, and broadly neutralizing antibody responses, as well as effector memory T cell responses. Here we review how these correlates of protection are guiding current approaches to HIV-1 vaccine development. These approaches include improvements on the ALVAC-HIV/AIDSVAX B/E vaccine regimen used in the RV144 clinical trial in Thailand, adenovirus serotype 26 vectors with gp140 boosting, intravenous infusions of bNAbs, and replicating viral vectors.

Keywords: HIV-1, vaccine, immune correlates, clinical trial

Introduction

Over three decades after the discovery of human immunodeficiency virus type 1 (HIV-1), a vaccine remains elusive. Variable adherence to combination antiretroviral therapy (cART), limited drug availability, and poor infrastructure are some of the public health roadblocks that show that an HIV-1 vaccine is still needed [13]. While cART has resulted in dramatic progress in the treatment of HIV-1 [4], the virus persists in a viral reservoir that is established early in acute infection [58], suggesting that there is only a short timeframe within which a vaccine has to be effective. Moreover, enormous HIV-1 diversity makes the design of a successful global vaccine challenging [911]. Nevertheless, recent preclinical and clinical vaccine studies have identified several vaccine-induced immune responses that appear to correlate with protection against infection [1215]. Although these correlates of protection are statistical associations at the present time and may or may not be causal mechanisms of protection [16], they serve as essential guideposts on the pathway of HIV-1 vaccine development and provide scientific hypotheses that the next generation of HIV-1 vaccines will test.

In this review article, we provide an overview of the current HIV-1 vaccine clinical trial landscape, focusing on immune correlates of protection and trials that are underway or in the pipeline. We begin by providing a brief summary of HIV-1 vaccine clinical development from the first efficacy trial, launched in 1998, to the present (Table 1). We then describe several current approaches in HIV-1 vaccine development, each of which is focused on eliciting a different immune response based on immune correlates of protection (Table 2, Figure 1). The first of these approaches is aimed at generating antibody responses specific to the V2 region of the HIV-1 Env protein, with a canarypox viral vector and the Env protein gp120 [17]. The second approach is aimed at generating polyfunctional Env-specific antibody responses, with adenovirus serotype 26 (Ad26) and/or modified vaccinia Ankara (MVA) viral vectors, mosaic HIV-1 sequences, and the Env protein gp140 [1820]. The third approach is aimed at generating or delivering broadly neutralizing antibodies (bNAbs), exemplified by passive infusion of the CD4-binding site monoclonal antibody (mAb) VRC01 [21,22]. The fourth approach is aimed at generating effector memory T cell responses with the use of live, replicating viral vectors, exemplified by cytomegalovirus vectors.

Table 1.

Completed HIV-1 Vaccine Efficacy Trials

Trial Vaccine Description Immune Responses Observed Efficacy Outcome
Vax004 AIDSVAX B/B gp120 (MN and
GNE8 subtype B) gp120 in alum
Non-neutralizing antibody response;
ADCVI
No efficacy
Vax003 AIDSVAX B/E gp120 (subtype B
MN and CRF01_AE CM244) gp120
in alum
Non-neutralizing antibody response No efficacy
HVTN 502/STEP
Trial
Adenovirus type 5 Clade B
gag/pol/nef
HIV-1 specific CD4+ and CD8+
responses
No efficacy, increased
infection risk
HVTN 503
(Phambili trial)
Adenovirus type 5 Clade B
gag/pol/nef
HIV-1 specific CD4+ and CD8+
responses
No efficacy, increased
infection risk
RV144 ALVAC-HIV (recombinant
canarypox vector)/vCP1521 and
AIDSVAX B/E rgp120 in alum
Humoral and cellular immune responses;
Non-neutralizing Ab to V1V2*, high
ADCC, HIV-1 specific IgG3, FcγRIIC
receptor
* Correlate of protection
31.2 % efficacy at 42
months, 60% efficacy at 12 months
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
T-cell responses to HIV-1 potential T-
cell epitopes; CD4+ HIV Gag responses
No efficacy

HVTN, HIV Vaccine Trials Network; ADCVI, antibody-dependent cell-mediated virus inhibition; ADCC, antibody-dependent cellular cytotoxicity

Table 2.

Ongoing HIV-1 Vaccine Clinical Trials Based on Known Immune Correlates of Protection

Vaccines that Elicit V2-Specific Antibody Responses

Extended Boosting of RV144 Regimen ALVAC-HIV/AIDSVAX B/E in Thailand NCT01435135 U.S. Army
Testing of RV144 Regimen in New
Region
ALVAC-HIV, AIDSVAX B/E in South Africa NCT02109354 HVTN
Adapting RV144 Regimen to New
Region
ALVAC-HIV (vCP2438), bivalent Subtype
C/MF59 in South Africa
NCT02404311 NIAID
Changing RV144 Adjuvants ALVAC/gp120 with Alum vs. MF59 Planned HVTN
ALVAC/gp120 Planned HVTN

Vaccines that Elicit Polyfunctional Antibody Responses

Novel Vaccine Regimens based on
Ad26 Vector, Mosaic Immunogens, and
gp140 Protein
Ad26.Mos.HIV/MVA-Mosaic/ gp140 NCT02315703 Crucell Holland BV
Ad26.Mos.HIV/Clade C gp140 NCT02685020 Crucell Holland BV
MVA-Mosaic NCT02218125 Crucell Holland BV
Clade C gp140 NCT02304185 Crucell Holland BV

Efforts that Elicit (or Deliver) Broadly Neutralizing Antibodies

Passive Infusion of bNAbs VRC01 NCT02256631 NIAID
VRC01 NCT02568215 NIAID
VRC01LS NCT022599896 NIAID
VRC01 efficacy Planned
10-1074 NCT02511990 Rockefeller Univ.
bNAb Epitope Scaffolds None yet
B Cell Lineage Design None yet
Gene Delivery rAAV1-PG9DP NCT01937455 IAVI

Vaccines that Elicit Effector Memory T Cells at Mucosal Sites of Infection

Replicating Vectors VSV-Indiana HIV Gag NCT01438606 NIAID
rVSV HIV Gag NCT01578889 NIAID
Ad4-mgag, Ad4-EnvC150 NCT01989533 NIAID
rcAd26.MOS1.HIV-Env NCT02366013 IAVI
HCMV/HIV Planned

HVTN, HIV Vaccine Trials Network; NIAID, National Institute of Allergy and Infectious Diseases; IAVI, International AIDS Vaccine Institute

Figure 1. Potential correlates of protection for ongoing HIV vaccine studies.

Figure 1

(1) V1V2-specific and/or polyfunctional non-neutralizing antibodies (NnAb, left panel). The cartoon represents the mechanism by which non-neutralizing antibodies may bind to infected CD4+ T cells, triggering complement activation, antibody-dependent cellular phagocytosis (ADCP), and/or antibody-dependent cellular cytotoxicity (ADCC). (2) Broadly neutralizing antibodies (nAb, middle panel). The cartoon represents neutralization of the infecting virus via binding to the Envelope glycoprotein. (C) Effector memory T-cells at mucosal sites. The cartoon depicts continuous antigen stimulation via an effector memory T-cell pool.

HIV-1 Vaccine Clinical Development: 1998 – Present

To date there have been only 6 HIV-1 vaccine candidates that have reached efficacy trials (Table 1). The early VAX003 and VAX004 trials used the Env subunit protein gp120 to induce immune responses, but the vaccines did not elicit neutralizing antibodies and failed to show efficacy [2326]. The focus subsequently shifted towards vaccine strategies aimed at inducing cellular immune responses, owing to the prominent role of CD8+ T cell responses in control of HIV-1 replication [27,28]. The Step study (HVTN 502) was the first efficacy trial that focused on cell-mediated immunity using an adenovirus serotype 5 (Ad5) viral vector to express HIV-1 Gag, Pol, and Nef internal proteins. The Step trial did not confer protection against infection or induce virologic control, and increased rates of infection were observed in a subset of vaccinees [29,30]. The Phambili trial (HVTN 503) tested the same vaccine used in the Step trial and also did not reduce infection rates [31]. The only signal of efficacy of an HIV-1 vaccine candidate was shown in the RV144 trial in Thailand, which showed 31% efficacy with a prime-boost regimen of a canarypox viral vector (ALVAC-HIV) and the Env subunit protein gp120 (AIDSVAX B/E) [17]. The HVTN 505 trial tested a multi-clade DNA prime with a recombinant Ad5 boost designed to generate both humoral and cellular responses, and this vaccine also did not show efficacy [3234]. The lessons from these trials suggest that the global diversity of HIV-1 strains and the challenge of inducing functional immune responses are major obstacles to a successful HIV-1 vaccine.

Current Landscape of HIV-1 Vaccine Clinical Research

Vaccines that Elicit V2-Specific Antibody Responses

RV144 used an ALVAC-HIV/AIDSVAX B/E regimen based on clade B and CRF01_AE sequences that matched the prevailing HIV-1 strains in Thailand [17,35]. Subsequent analyses showed that non-neutralizing antibodies to variable loops 1 and 2 (V1V2) regions of HIV-1 Env were associated with a reduced risk of HIV-1 acquisition [12,15], and a genetic sieve analysis demonstrated that the ALVAC-HIV/AIDSVAX B/E regimen had improved efficacy against viruses that matched the immunogen sequence in the V2 location [15]. Moreover, high levels of antibody-dependent cellular cytotoxicity (ADCC) were also shown to correlate with a reduced risk of HIV-1 acquisition in RV144 [36] and V2-specific monoclonal antibodies isolated from RV144 vaccinees were shown to mediate ADCC against HIV-1-infected CD4+ T cells [37]. HIV-1 specific IgG3 responses were also noted to correlate with protection in RV144 and distinguish this trial from the VAX003 trial [13].

These data suggested a path forward for HIV-1 vaccine development by raising the hypothesis that non-neutralizing V2-specific antibodies, particularly if well-matched to local circulating strains, might have the potential to block HIV-1 acquisition (Table 2, Figure 1). As a result, multiple strategies are being pursued to build on the RV144 findings, including:

  • Changing the RV144 vaccine schedule to improve the magnitude and durability of V2specific antibody responses. For example, the RV305 trial is studying extended boosting of the ALVAC-HIV/AIDSVAX B/E regimen in Thailand (NCT01435135).

  • Transferring the RV144 regimen to a new region to test whether the V2-specific responses generated in Thailand might also provide protection in a different region. The HVTN 097 trial is testing this strategy by studying the ALVAC-HIV/AIDSVAX B/E regimen in South Africa (NCT02109354); early data indicates similar cellular responses as RV144 [38].

  • Adapting the RV144 regimen to a different region to test whether changing the vaccine sequences to match local prevailing strains will generate regional V2-specific responses. The HVTN 100 trial is pursuing this strategy with testing of an ALVAC-HIV (vCP2438)/bivalent subtype C gp120/MF59 regimen in South Africa (NCT02404311), with plans to move to efficacy testing in the HVTN 702 trial [39].

  • Using different adjuvants to the RV144 regimen to enhance immune responses [40,41][42]. For example, HVTN 107 is a planned phase1/2a study in Zimbabwe comparing ALVAC-HIV (vCP2438)/bivalent subtype C gp120 with either MF59 or aluminum hydroxide [43].

Vaccines that Elicit Polyfunctional Antibody Responses

The modest and transient vaccine efficacy observed in the RV144 trial has also inspired efforts to push forward with additional vaccine concepts. Polyfunctional Env-specific antibody responses were associated with a significantly reduced risk of simian-human immunodeficiency virus (SHIV) and simian immunodeficiency virus (SIV) acquisition following repetitive rectal challenges in nonhuman primates [44]. Specifically, we showed that rhesus monkeys primed with Ad26 vectors expressing SIVsmE543 Env, Gag, and Pol and boosted with adjuvanted SIVmac32H Env gp140 demonstrated complete protection in 50% of vaccinated animals against a series of repeated, heterologous, intrarectal SIVmac251 challenges that infected all controls. Analysis of antibody Fc functions, including ADCC, antibody-dependent cellular phagocytosis (ADCP), antibody-dependent natural killer (NK) cell activities (ADNKA), antibody-dependent complement deposition (ADCD), and glycosylation profiles, revealed that the protein boost resulted in a more polyfunctional antibody response that also correlated with protective efficacy. These data supported the results of earlier vaccine studies in non-human primates that also demonstrated that non-neutralizing antibody effector functions correlated with protective efficacy [45,46].

Novel Adenovirus serotype 26 vectors are thus being evaluated in clinical studies in combination with the Env trimeric protein gp140. In initial studies, an Ad26 vector expressing a clade A HIV-1 Env immunogen (Ad26.EnvA.01) was shown to be safe, well-tolerated, and immunogenic in three completed clinical trials (NCT00618605, NCT01103687, and NCT01215149). Ad26 vectors have been shown in multiple models to be biologically distinct from Ad5 [47], the vector used in the Step study. Ad26 has also been shown to elicit polyfunctional antibody response in humans using systems serology [48]. This platform characterized Fc-effector functions, ADCC, ADCP, NK cell IFN-γ secretion, NK MIP-1β secretion, and NK CD107a activation as well as multiple other biophysical measurements [48]. Using this platform, Chung et al. demonstrated that Ad26.EnvA.01 elicited a network of antibody functions and Fc-receptor binding activity that was clearly distinguished from responses elicited by the RV144, VAX003/004, and Step regimens.

Multivalent Ad26 vectors have recently been developed that express mosaic immunogens (Ad26.Mos.HIV) with the goal of generating polyfunctional antibodies and diverse cellular immune responses that recognize HIV-1 epitopes from global viruses. Mosaic HIV-1 Env, Gag, and Pol sequences are bioinformatically designed to optimize coverage of global HIV-1 diversity [4951][52], and have been shown to expand cellular and humoral immune responses in non-human primates when expressed by adenoviral vectors [44,5355]. The clinical trial HIV-V-A004/IPCAVD009 (NCT02315703) launched in 2015, is evaluating Ad26.Mos.HIV, MVA vectors expressing the same mosaic antigens (MVA-mosaic) and a clade C gp140 boost. This study is sponsored by Crucell Holland B.V. (Janssen Pharmaceuticals), and has completed enrollment of 400 subjects in Rwanda, South Africa, Thailand, Uganda, and the USA. The MVA-mosaic vaccine was also evaluated in a phase 1 clinical trial, HIV-V-A002/IPCAVD006, that launched in 2014; this study is examining two injections of MVA-mosaic in unvaccinated healthy individuals and individuals who previously received the Ad26.EnvA.01 vaccine (NCT02218125) [18,19]. A third vaccine trial of mosaic immunogens is launching in 2016 and is testing Ad26.Mos.HIV and clade C gp140 using shorter vaccine schedules (HPX1002/IPCAVD 010; NCT02685020).

Finally, Env gp140 protein subunit immunogens are also undergoing clinical testing as part of multi-component vaccine regimens to elicit polyfunctional antibodies. For example, the South African AIDS Vaccine Initiative (SAAVI) has begun testing of a Novartis subtype C gp140 boost in the context of a previous DNA/MVA/truncated gp160 vaccine (HVTN 073, NCT01423825 and HVTN 073E, NCT01423825). As mentioned above, a clade C gp140 trimeric protein is also being tested in the HIV-V-A003/IPCAVD008 (NCT02304185), HIV-V-A004/IPCAVD009, and HPX1002/IPCAVD010 studies.

Efforts to Elicit (or Deliver) Broadly Neutralizing Antibodies

One of the unsolved goals in HIV-1 vaccine development is the generation of immunogens that can elicit broadly neutralizing antibodies (bNAbs) [5658]. Such bNAbs are found in about 10–30% of infected people and develop several years after infection [59]. The sole target of these antibodies is the Env glycoprotein, a highly diverse structure with glycosylation patterns that hide many of the potential neutralizing antibody targets [60,61]. Several broad and potent mAbs have shown promise in passive protection studies in nonhuman primates. PGT121, which targets a glycan-dependent site on the V3 loop of Env, elicited sterilizing immunity in macaques against SHIV infection at low doses of 1–5 mg/kg [62]. Other antibodies, including VRC01, 2F5, 2G12, b12, 4E10, 3BNC117, 10–1074, and others also elicited protection against SHIV challenge in non-human primates in combination or at higher doses [6366][67].

Multiple approaches are being pursued to elicit bNAbs by immunization. One approach consists of designing immunogens that better mimic bNAb epitopes, e.g., novel native-like Env trimeric proteins such as BG505 SOSIP.664 or bNAb epitope mimics on scaffolds [6871]. Another approach is to use immunogens that bind germline precursors of bNAbs and thus stimulate different stages of the bNAb clonal lineage and allow for evolution of B-cells to production of bNAb, known as B-cell lineage vaccines [72].

Another option for the delivery of bNAbs is gene transfer, which is sometimes referred to as vectored immunoprophylaxis. Adeno-associated virus (AAV) vectors are being used for this task to enable the direct expression of bNAbs. SIV modified immunoadhesins expressed via AAVs showed protection and long term neutralization in a non-human primate study [73]. AAVs have also been used to induce lifelong mAb production in humanized mice, affording protection against HIV [74]. Another immunoadhesin, CD4-Ig was combined with a modified CCR5 binding peptide without inhibiting hydrophobic residues to enhance Env binding. This combination was expressed for up to 10 months in non-human primates and showed protection against SHIV challenge [75]. One AAV delivering the neutralizing antibody PG9 is currently being tested in a phase I study (NCT01937455).

Vaccine strategies to induce bNAbs are in their early stages in terms of clinical development and a full review of strategies to induce bNAbs is beyond the scope of this review. Efforts to administer bNAbs passively are therefore being pursued in proof-of-concept studies to establish whether bNAbs protect against HIV-1 infection in humans. The first bNAb to begin human clinical trials is VCR-HIVMAB060-00-AB (VRC01), which targets the CD4 binding site. VRC01 neutralizes an extensive global panel of Env-pseudotyped viral strains in vitro [22,76,77], and passive transfer of VRC01 along with two other bNAbs 10E8 and PG9 showed complete protection against SHIV challenge in rhesus macaques [67]. Thus far, VRC01 has been shown to be safe and well-tolerated, and to suppress viral replication transiently in HIV-1-infected individuals in the Phase 1 clinical trials VRC601 and VRC602 [78,79]. A phase 2b safety and efficacy study of VRC01 has recently begun and early phase clinical trials of other bNAbs are underway (NCT02256631; NCT02568215 (HVTN 703); NCT02599896 (VRC606)).

Vaccines that Elicit Effector Memory T Cells at Mucosal Sites of Infection

The HIV-1 vaccine strategies discussed above aim to elicit humoral immune responses – V2-specific, polyfunctional, and broadly neutralizing antibodies – as these responses may correlate with reduced risk of HIV-1 acquisition. Data from non-human primates also suggests that replicating viral vectors may elicit cellular immune responses that can prevent systemic HIV-1 infection by limiting the spread of mucosally acquired infection. For example, a rhesus cytomegalovirus (RhCMV) vector expressing SIV antigens elicited SIV-specific effectormemory T cells responses at mucosal sites of infection [80], prevented systemic, progressive dissemination of infection [81], and led to control of viremia and pathogenesis against SIVmac239 in rhesus macaques [82]. Such an approach suggests that the RhCMV vector established a persistent HIV-1 effector T-cell pool that could rapidly control viral replication at the time of infection.

Several replicating vectors are being explored for their ability to induce persistent immune responses via continuous antigen stimulation, especially at mucosal sites of potential infection. The hypothesis is that these immune responses may inhibit viral infection prior to establishment of a viral reservoir and diversification of the infecting virus, thus containing or aborting early infection. In addition to modified attenuated human CMV vectors, which are not yet in clinical trials [82], replicating adenoviruses (Ad) are also being explored, as they persist in mucosal sites [83]. For example, mucosal IgA titers following immunization with a replicating Ad-SIV vaccine in macaques correlated with improved control of viremia [84]. Clinical advancement of replicating Ad vectors is underway with replication-competent recombinant adenovirus serotype 4 (Ad4) HIV-1 vaccine regimens (Ad4-mgag and Ad4-EnvC150); a phase 1 clinical trial testing the oral and intranasal routes of administration is currently underway (NCT01989533). In addition, an oral replicating adenovirus serotype 26 (Ad26) vector expressing HIV-1 Env is also in Phase 1 testing (NCT02366013) [85]. Additionally, the vesicular stomatitis virus (VSV) vector has entered phase 1 clinical testing (NCT01438606) [86], and plans are in place to conduct a follow-up trial using a DNA prime-VSV vector boost (NCT01578889).

Conclusions

An HIV-1 vaccine has been a daunting challenge due to global HIV-1 diversity and the difficulties in inducing protective antibody responses and cellular immune responses. Here we discuss four current approaches to the development of a successful HIV-1 vaccine, each based on different immune correlates of protection. The first approach is to develop vaccines that elicit V2-specific antibody responses and that are well matched to locally circulating strains. Examples of this approach include efforts to build on the ALVAC/AIDSVAX B/E vaccine regimens tested in RV144 with ongoing clinical trials in Thailand and Africa. The second approach is to develop vaccines to elicit diverse, polyfunctional antibody responses. Examples of this approach include efforts to test Ad26 vectors, mosaic immunogens, and trimeric gp140 protein regimens. The third approach is to elicit (or deliver) broadly neutralizing antibodies through immunization, gene delivery, or passive administration. The fourth strategy is to elicit effector memory T cells through the use of replicating vectors. As all four of these approaches move towards efficacy trials, the field will have an unprecedented opportunity to test prospectively which immune correlates are confirmed and afford protection against HIV-1 in humans.

Highlights.

  • Immune correlates of protection are guiding HIV-1 vaccine development.

  • RV144 follow-on trials aim to generate antibody responses to the V1V2 region of Env.

  • Immunogens are being tested to generate polyfunctional Env antibody responses.

  • Passive transfer of broadly neutralizing antibodies is being tested in clinical trials.

  • Replicating viral vectors may establish a pool of effector memory T-cells.

Acknowledgments

We acknowledge support from the National Institutes of Health (AI095985, AI096040, AI100663, AI124377, OD011170), the Bill and Melinda Gates Foundation, and the Ragon Institute of MGH, MIT, and Harvard.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Fox MP, Rosen S. Retention of Adult Patients on Antiretroviral Therapy in Low- and Middle-Income Countries. JAIDS J. Acquir. Immune Defic. Syndr. 2015;69:98–108. doi: 10.1097/QAI.0000000000000553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Schaecher KL. The importance of treatment adherence in HIV. Am. J. Manag. Care. 2013;19:s231–s237. [PubMed] [Google Scholar]
  • 3.Obiako OR, Muktar HM. Challenges of HIV treatment in resource-poor countries: a review. Niger. J. Med. 19:361–368. doi: 10.4314/njm.v19i4.69785. [DOI] [PubMed] [Google Scholar]
  • 4.AIDS by the numbers 2015 | UNAIDS. 2015 [Google Scholar]
  • 5.Barouch DH, Deeks SG. Immunologic strategies for HIV-1 remission and eradication. Science. 2014;345:169–174. doi: 10.1126/science.1255512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chun T-W, Justement JS, Murray D, Hallahan CW, Maenza J, Collier AC, Sheth PM, Kaul R, Ostrowski M, Moir S, et al. Rebound of plasma viremia following cessation of antiretroviral therapy despite profoundly low levels of HIV reservoir: implications for eradication. AIDS. 2010;24:2803–2808. doi: 10.1097/QAD.0b013e328340a239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chun T-W, Moir S, Fauci AS. HIV reservoirs as obstacles and opportunities for an HIV cure. Nat. Immunol. 2015;16:584–589. doi: 10.1038/ni.3152. [DOI] [PubMed] [Google Scholar]
  • 8.Sigal A, Kim JT, Balazs AB, Dekel E, Mayo A, Milo R, Baltimore D. Cell-to-cell spread of HIV permits ongoing replication despite antiretroviral therapy. Nature. 2011;477:95–98. doi: 10.1038/nature10347. [DOI] [PubMed] [Google Scholar]
  • 9.Hemelaar J. The origin and diversity of the HIV-1 pandemic. Trends Mol. Med. 2012;18:182–192. doi: 10.1016/j.molmed.2011.12.001. [DOI] [PubMed] [Google Scholar]
  • 10.Hemelaar J, Gouws E, Ghys PD, Osmanov S. Global and regional distribution of HIV-1 genetic subtypes and recombinants in 2004. AIDS. 2006;20:W13–W23. doi: 10.1097/01.aids.0000247564.73009.bc. [DOI] [PubMed] [Google Scholar]
  • 11.Ndung’u T, Weiss RA. On HIV diversity. AIDS. 2012;26:1255–1260. doi: 10.1097/QAD.0b013e32835461b5. [DOI] [PubMed] [Google Scholar]
  • 12. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N. Engl. J. Med. 2012;366:1275–1286. doi: 10.1056/NEJMoa1113425. This paper showed an inverse correlation between V1V2 Abs against different HIV-1 subtypes and risk of HIV-1 infection in the RV144 trial, establishing a method to test V1V2 antibodies as a correlate of protection in future vaccine trials.
  • 13.Yates NL, Liao H-X, Fong Y, deCamp A, Vandergrift NA, Williams WT, Alam SM, Ferrari G, Yang Z, Seaton KE, et al. Vaccine-induced Env V1–V2 IgG3 correlates with lower HIV-1 infection risk and declines soon after vaccination. Sci. Transl. Med. 2014;6:228ra39. doi: 10.1126/scitranslmed.3007730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zolla-Pazner S, deCamp A, Gilbert PB, Williams C, Yates NL, Williams WT, Howington R, Fong Y, Morris DE, Soderberg KA, 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: 10.1371/journal.pone.0087572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rolland M, Edlefsen PT, Larsen BB, Tovanabutra S, Sanders-Buell E, Hertz T, deCamp AC, Carrico C, Menis S, Magaret CA, et al. Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2. Nature. 2012;490:417–420. doi: 10.1038/nature11519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Plotkin SA, Gilbert PB. Nomenclature for immune correlates of protection after vaccination. Clin. Infect. Dis. 2012;54:1615–1617. doi: 10.1093/cid/cis238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, Paris R, Premsri N, Namwat C, de Souza M, Adams E, Benenson M, Gurunathan S, Tartaglia J, McNeil JG, Francis DP, Stablein D, Birx DL, Chunsuttiwat S, Khamboonruang C, Thongcharoen P, Rob M-TI. Vaccination with ALVAC and AIDSVAX to Prevent HIV-1 Infection in Thailand. N Engl J Med. 2009;361:2209–2220. doi: 10.1056/NEJMoa0908492. [DOI] [PubMed] [Google Scholar]
  • 18.Baden LR, Walsh SR, Seaman MS, Johnson JA, Tucker RP, Kleinjan JA, Gothing JA, Engelson BA, Carey BR, Oza A, et al. First-in-human evaluation of a hexon chimeric adenovirus vector expressing HIV-1 Env (IPCAVD 002) J. Infect. Dis. 2014;210:1052–1061. doi: 10.1093/infdis/jiu217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Barouch DH, Liu J, Peter L, Abbink P, Iampietro MJ, Cheung A, Alter G, Chung A, Dugast A-S, Frahm N, et al. Characterization of humoral and cellular immune responses elicited by a recombinant adenovirus serotype 26 HIV-1 Env vaccine in healthy adults (IPCAVD 001) J. Infect. Dis. 2013;207:248–256. doi: 10.1093/infdis/jis671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nkolola JP, Bricault CA, Cheung A, Shields J, Perry J, Kovacs JM, Giorgi E, van Winsen M, Apetri A, Brinkman-van der Linden ECM, et al. Characterization and immunogenicity of a novel mosaic M HIV-1 gp140 trimer. J. Virol. 2014;88:9538–9552. doi: 10.1128/JVI.01739-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Caskey M, Klein F, Lorenzi JCC, Seaman MS, West AP, Buckley N, Kremer G, Nogueira L, Braunschweig M, Scheid JF, et al. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature. 2015;522:487–491. doi: 10.1038/nature14411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wu X, Yang Z-Y, Li Y, Hogerkorp C-M, Schief WR, Seaman MS, Zhou T, Schmidt SD, Wu L, Xu L, et al. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science. 2010;329:856–861. doi: 10.1126/science.1187659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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: 10.4049/jimmunol.178.10.6596. [DOI] [PubMed] [Google Scholar]
  • 24.Gilbert PB, Peterson ML, Follmann D, Hudgens MG, Francis DP, Gurwith M, Heyward WL, Jobes DV, Popovic V, Self SG, 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: 10.1086/428405. [DOI] [PubMed] [Google Scholar]
  • 25.Flynn NM, Forthal DN, Harro CD, Judson FN, Mayer KH, Para MF. Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection. J. Infect. Dis. 2005;191:654–665. doi: 10.1086/428404. [DOI] [PubMed] [Google Scholar]
  • 26.Pitisuttithum P, Gilbert P, Gurwith M, Heyward W, Martin M, van Griensven F, Hu D, Tappero JW, Choopanya K. 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: 10.1086/508748. [DOI] [PubMed] [Google Scholar]
  • 27.Emu B, Sinclair E, Hatano H, Ferre A, Shacklett B, Martin JN, McCune JM, Deeks SG. HLA class I-restricted T-cell responses may contribute to the control of human immunodeficiency virus infection, but such responses are not always necessary for long-term virus control. J. Virol. 2008;82:5398–5407. doi: 10.1128/JVI.02176-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cao Y, Qin L, Zhang L, Safrit J, Ho DD. Virologic and immunologic characterization of long-term survivors of human immunodeficiency virus type 1 infection. N. Engl. J. Med. 1995;332:201–208. doi: 10.1056/NEJM199501263320401. [DOI] [PubMed] [Google Scholar]
  • 29.Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, Li D, Gilbert PB, Lama JR, Marmor M, Del Rio C, 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: 10.1016/S0140-6736(08)61591-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Duerr A, Huang Y, Buchbinder S, Coombs RW, Sanchez J, del Rio C, Casapia M, Santiago S, Gilbert P, Corey L, 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: 10.1093/infdis/jis342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Gray GE, Allen M, Moodie Z, Churchyard G, Bekker L-G, Nchabeleng M, Mlisana K, Metch B, de Bruyn G, Latka MH, 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: 10.1016/S1473-3099(11)70098-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Letvin NL, Rao SS, Montefiori DC, Seaman MS, Sun Y, Lim S-Y, Yeh WW, Asmal M, Gelman RS, Shen L, et al. Immune and Genetic Correlates of Vaccine Protection Against Mucosal Infection by SIV in Monkeys. Sci. Transl. Med. 2011;3:81ra36. doi: 10.1126/scitranslmed.3002351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Churchyard GJ, Morgan C, Adams E, Hural J, Graham BS, Moodie Z, Grove D, Gray G, Bekker L-G, McElrath MJ, 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: 10.1371/journal.pone.0021225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hammer SM, Sobieszczyk ME, Janes H, Karuna ST, Mulligan MJ, Grove D, Koblin BA, Buchbinder SP, Keefer MC, Tomaras GD, et al. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N. Engl. J. Med. 2013;369:2083–2092. doi: 10.1056/NEJMoa1310566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Excler JL, Plotkin S. The prime-boost concept applied to HIV preventive vaccines. AIDS. 1997;11(Suppl A):S127–S137. [PubMed] [Google Scholar]
  • 36.Bonsignori M, Pollara J, Moody MA, Alpert MD, Chen X, Hwang K-K, Gilbert PB, Huang Y, Gurley TC, Kozink DM, 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: 10.1128/JVI.01023-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Liao H-X, Bonsignori M, Alam SM, McLellan JS, Tomaras GD, Moody MA, Kozink DM, Hwang K-K, Chen X, Tsao C-Y, 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: 10.1016/j.immuni.2012.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gray GE, Andersen-Nissen E, Grunenberg N, Huang Y, Roux S, Laher F, Innes C, Gu N, DiazGranados C, Phogat S, et al. HVTN 097: Evaluation of the RV144 Vaccine Regimen in HIV Uninfected South African Adults. AIDS Res. Hum. Retroviruses. 2014;30:A33–A34. [Google Scholar]
  • 39.O’Connell RJ, Kim JH, Corey L, Michael NL. Human immunodeficiency virus vaccine trials. Cold Spring Harb. Perspect. Med. 2012;2:a007351. doi: 10.1101/cshperspect.a007351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.McElrath MJ. Selection of potent immunological adjuvants for vaccine construction. Semin. Cancer Biol. 1995;6:375–385. doi: 10.1016/1044-579x(95)90007-1. [DOI] [PubMed] [Google Scholar]
  • 41.Rhee EG, Barouch DH. Translational Mini-Review Series on Vaccines for HIV: Harnessing innate immunity for HIV vaccine development. Clin. Exp. Immunol. 2009;157:174–180. doi: 10.1111/j.1365-2249.2009.03928.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Vaccari M, Gordon SN, Fourati S, Schifanella L, Cameron M, Keele BF, Shen X, Tomaras G, Billings E, Rao M, et al. Adjuvant Dependent Mucosal V2 Responses and RAS Activation in Vaccine Induced Protection from SIV mac251 Acquisitiona. AIDS Res. Hum. Retroviruses. 2014;30:A64–A65. [Google Scholar]
  • 43.HVTN Studies. HVTN Ongoing Protoc [Google Scholar]
  • 44. Barouch DH, Alter G, Broge T, Linde C, Ackerman ME, Brown EP, Borducchi EN, Smith KM, Nkolola JP, Liu J, et al. Protective efficacy of adenovirus/protein vaccines against SIV challenges in rhesus monkeys. Science. 2015;349:320–324. doi: 10.1126/science.aab3886. In this study, protective efficacy of Ad26 expressing SIVsmE453 Env/Gag/Pol and Env gp140 correlated with polyfunctionality of Env-specific antibody responses. The protein boost was critical to eliciting these polyfunctional antibody responses.
  • 45.Barouch DH, Stephenson KE, Borducchi EN, Smith K, Stanley K, McNally AG, Liu J, Abbink P, Maxfield LF, Seaman MS, et al. Protective efficacy of a global HIV-1 mosaic vaccine against heterologous SHIV challenges in rhesus monkeys. Cell. 2013;155:531–539. doi: 10.1016/j.cell.2013.09.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ackerman ME, Moldt B, Wyatt RT, Dugast A-S, McAndrew E, Tsoukas S, Jost S, Berger CT, Sciaranghella G, Liu Q, et al. A robust, high-throughput assay to determine the phagocytic activity of clinical antibody samples. J. Immunol. Methods. 2011;366:8–19. doi: 10.1016/j.jim.2010.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Barouch DH, Picker LJ. Novel vaccine vectors for HIV-1. Nat. Rev. Microbiol. 2014;12:765–771. doi: 10.1038/nrmicro3360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Chung AW, Kumar MP, Arnold KB, Yu WH, Schoen MK, Dunphy LJ, Suscovich TJ, Frahm N, Linde C, Mahan AE, et al. Dissecting Polyclonal Vaccine-Induced Humoral Immunity against HIV Using Systems Serology. Cell. 2015;163:988–998. doi: 10.1016/j.cell.2015.10.027. Systems serology is an approach to broadly characterize the non-neutralizing Fc-mediated effector functions of antibodies that may correlate with protection against infection and is a platform that could be used to compare HIV-1 vaccines.
  • 49.Fischer W, Perkins S, Theiler J, Bhattacharya T, Yusim K, Funkhouser R, Kuiken C, Haynes B, Letvin NL, Walker BD, et al. Polyvalent vaccines for optimal coverage of potential T-cell epitopes in global HIV-1 variants. Nat. Med. 2007;13:100–106. doi: 10.1038/nm1461. [DOI] [PubMed] [Google Scholar]
  • 50.Ndhlovu ZM, Piechocka-Trocha A, Vine S, McMullen A, Koofhethile KC, Goulder PJR, Ndung’u T, Barouch DH, Walker BD. Mosaic HIV-1 Gag antigens can be processed and presented to human HIV-specific CD8+ T cells. J. Immunol. 2011;186:6914–6924. doi: 10.4049/jimmunol.1004231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Thurmond J, Yoon H, Kuiken C, Yusim K, Perkins S, Theiler J, Bhattacharya T, Korber B, Fischer W. Web-based design and evaluation of T-cell vaccine candidates. Bioinformatics. 2008;24:1639–1640. doi: 10.1093/bioinformatics/btn251. [DOI] [PubMed] [Google Scholar]
  • 52.Kothe DL, Li Y, Decker JM, Bibollet-Ruche F, Zammit KP, Salazar MG, Chen Y, Weng Z, Weaver EA, Gao F, et al. Ancestral and consensus envelope immunogens for HIV-1 subtype C. Virology. 2006;352:438–449. doi: 10.1016/j.virol.2006.05.011. [DOI] [PubMed] [Google Scholar]
  • 53.Barouch DH, O’Brien KL, Simmons NL, King SL, Abbink P, Maxfield LF, Sun Y-H, La Porte A, Riggs AM, Lynch DM, 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: 10.1038/nm.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Stephenson KE, SanMiguel A, Simmons NL, Smith K, Lewis MG, Szinger JJ, Korber B, Barouch DH. Full-length HIV-1 immunogens induce greater magnitude and comparable breadth of T lymphocyte responses to conserved HIV-1 regions compared with conserved-region-only HIV-1 immunogens in rhesus monkeys. J. Virol. 2012;86:11434–11440. doi: 10.1128/JVI.01779-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Santra S, Muldoon M, Watson S, Buzby A, Balachandran H, Carlson KR, Mach L, Kong W-P, McKee K, Yang Z-Y, 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: 10.1016/j.virol.2012.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Stamatatos L. HIV vaccine design: the neutralizing antibody conundrum. Curr. Opin. Immunol. 2012;24:316–323. doi: 10.1016/j.coi.2012.04.006. [DOI] [PubMed] [Google Scholar]
  • 57.Hraber P, Korber BT, Lapedes AS, Bailer RT, Seaman MS, Gao H, Greene KM, McCutchan F, Williamson C, Kim JH, et al. Impact of clade, geography, and age of the epidemic on HIV-1 neutralization by antibodies. J. Virol. 2014;88:12623–12643. doi: 10.1128/JVI.01705-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Klein F, Mouquet H, Dosenovic P, Scheid JF, Scharf L, Nussenzweig MC. Antibodies in HIV-1 vaccine development and therapy. Science. 2013;341:1199–1204. doi: 10.1126/science.1241144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.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: 10.1038/nm.1949. [DOI] [PubMed] [Google Scholar]
  • 60.Moore PL, Gray ES, Wibmer CK, Bhiman JN, Nonyane M, Sheward DJ, Hermanus T, Bajimaya S, Tumba NL, Abrahams M-R, et al. Evolution of an HIV glycan-dependent broadly neutralizing antibody epitope through immune escape. Nat. Med. 2012;18:1688–1692. doi: 10.1038/nm.2985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Klein JS, Bjorkman PJ. Few and far between: how HIV may be evading antibody avidity. PLoS Pathog. 2010;6:e1000908. doi: 10.1371/journal.ppat.1000908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Moldt B, Rakasz EG, Schultz N, Chan-Hui P-Y, Swiderek K, Weisgrau KL, Piaskowski SM, Bergman Z, Watkins DI, Poignard P, et al. Highly potent HIV-specific antibody neutralization in vitro translates into effective protection against mucosal SHIV challenge in vivo. Proc. Natl. Acad. Sci. U. S. A. 2012;109:18921–18925. doi: 10.1073/pnas.1214785109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mascola JR, Lewis MG, Stiegler G, Harris D, VanCott TC, Hayes D, Louder MK, Brown CR, Sapan CV, Frankel SS, et al. Protection of Macaques against pathogenic simian/human immunodeficiency virus 89.6PD by passive transfer of neutralizing antibodies. J. Virol. 1999;73:4009–4018. doi: 10.1128/jvi.73.5.4009-4018.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Parren PWHI, Marx PA, Hessell AJ, Luckay A, Harouse J, Cheng-Mayer C, Moore JP, Burton DR. Antibody Protects Macaques against Vaginal Challenge with a Pathogenic R5 Simian/Human Immunodeficiency Virus at Serum Levels Giving Complete Neutralization In Vitro. J. Virol. 2001;75:8340–8347. doi: 10.1128/JVI.75.17.8340-8347.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hessell AJ, Rakasz EG, Tehrani DM, Huber M, Weisgrau KL, Landucci G, Forthal DN, Koff WC, Poignard P, Watkins DI, et al. Broadly neutralizing monoclonal antibodies 2F5 and 4E10 directed against the human immunodeficiency virus type 1 gp41 membrane-proximal external region protect against mucosal challenge by simian-human immunodeficiency virus SHIVBa-L. J. Virol. 2010;84:1302–1313. doi: 10.1128/JVI.01272-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hessell AJ, Rakasz EG, Poignard P, Hangartner L, Landucci G, Forthal DN, Koff WC, Watkins DI, Burton DR. Broadly neutralizing human anti-HIV antibody 2G12 is effective in protection against mucosal SHIV challenge even at low serum neutralizing titers. PLoS Pathog. 2009;5:e1000433. doi: 10.1371/journal.ppat.1000433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pegu A, Yang Z, Boyington JC, Wu L, Ko S-Y, Schmidt SD, McKee K, Kong W-P, Shi W, Chen X, et al. Neutralizing antibodies to HIV-1 envelope protect more effectively in vivo than those to the CD4 receptor. Sci. Transl. Med. 2014;6:243ra88. doi: 10.1126/scitranslmed.3008992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mann JK, Ndung’u T. HIV-1 vaccine immunogen design strategies. Virol. J. 2015;12:3. doi: 10.1186/s12985-014-0221-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sanders RW, Derking R, Cupo A, Julien J-P, Yasmeen A, de Val N, Kim HJ, Blattner C, de la Peña AT, Korzun J, et al. A next-generation cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS Pathog. 2013;9:e1003618. doi: 10.1371/journal.ppat.1003618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sanders RW, Schiffner L, Master A, Kajumo F, Guo Y, Dragic T, Moore JP, Binley JM. Variable-loop-deleted variants of the human immunodeficiency virus type 1 envelope glycoprotein can be stabilized by an intermolecular disulfide bond between the gp120 and gp41 subunits. J. Virol. 2000;74:5091–5100. doi: 10.1128/jvi.74.11.5091-5100.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Georgiev IS, Joyce MG, Yang Y, Sastry M, Zhang B, Baxa U, Chen RE, Druz A, Lees CR, Narpala S, et al. Single-Chain Soluble BG505.SOSIP gp140 Trimers as Structural and Antigenic Mimics of Mature Closed HIV-1 Env. J. Virol. 2015;89:5318–5329. doi: 10.1128/JVI.03451-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Haynes BF, Kelsoe G, Harrison SC, Kepler TB. B-cell-lineage immunogen design in vaccine development with HIV-1 as a case study. Nat. Biotechnol. 2012;30:423–433. doi: 10.1038/nbt.2197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Johnson PR, Schnepp BC, Zhang J, Connell MJ, Greene SM, Yuste E, Desrosiers RC, Clark KR. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat. Med. 2009;15:901–906. doi: 10.1038/nm.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Balazs AB, Chen J, Hong CM, Rao DS, Yang L, Baltimore D. Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature. 2012;481:81–84. doi: 10.1038/nature10660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Gardner MR, Kattenhorn LM, Kondur HR, von Schaewen M, Dorfman T, Chiang JJ, Haworth KG, Decker JM, Alpert MD, Bailey CC, et al. AAV-expressed eCD4-Ig provides durable protection from multiple SHIV challenges. Nature. 2015;519:87–91. doi: 10.1038/nature14264. This study demonstrated that eCD4-Ig, a fusion of CD4I-Ig and a CCR5-mimetic sulfopeptide expressed via an AAV vector neutralized a larger panel of viruses than bNAbs and protected macaques against multiple SHIV-AD8 challenges.
  • 76.Nakamura KJ, Cerini C, Sobrera ER, Heath L, Sinkala M, Kankasa C, Thea DM, Mullins JI, Kuhn L, Aldrovandi GM. Coverage of primary mother-to-child HIV transmission isolates by second-generation broadly neutralizing antibodies. AIDS. 2013;27:337–346. doi: 10.1097/QAD.0b013e32835cadd6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Russell ES, Ojeda S, Fouda GG, Meshnick SR, Montefiori D, Permar SR, Swanstrom R. Short communication: HIV type 1 subtype C variants transmitted through the bottleneck of breastfeeding are sensitive to new generation broadly neutralizing antibodies directed against quaternary and CD4-binding site epitopes. AIDS Res. Hum. Retroviruses. 2013;29:511–515. doi: 10.1089/aid.2012.0197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Ledgerwood JE, Coates EE, Yamshchikov G, Saunders JG, Holman L, Enama ME, DeZure A, Lynch RM, Gordon I, Plummer S, et al. Safety, pharmacokinetics and neutralization of the broadly neutralizing HIV-1 human monoclonal antibody VRC01 in healthy adults. Clin. Exp. Immunol. 2015;182:289–301. doi: 10.1111/cei.12692. This phase I trial evaluated the bNAB, VRC01, as part of efforts to understand whether passive transfer of bNAbs is a viable path for HIV-1 vaccine development.
  • 79.Lynch RM, Boritz E, Coates EE, DeZure A, Madden P, Costner P, Enama ME, Plummer S, Holman L, Hendel CS, et al. Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection. Sci. Transl. Med. 2015;7:319ra206. doi: 10.1126/scitranslmed.aad5752. [DOI] [PubMed] [Google Scholar]
  • 80.Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, Whizin N, Oswald K, Shoemaker R, Swanson T, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011;473:523–527. doi: 10.1038/nature10003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Hansen SG, Vieville C, Whizin N, Coyne-Johnson L, Siess DC, Drummond DD, Legasse AW, Axthelm MK, Oswald K, Trubey CM, et al. Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nat. Med. 2009;15:293–299. doi: 10.1038/nm.1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Hansen SG, Piatak M, Ventura AB, Hughes CM, Gilbride RM, Ford JC, Oswald K, Shoemaker R, Li Y, Lewis MS, et al. Immune clearance of highly pathogenic SIV infection. Nature. 2013;502:100–104. doi: 10.1038/nature12519. A RhCMV/SIV vector in rhesus macaqus led to control and clearance of a pathogenic lentiviral infection. The continuous presence of an effector memory T-cell population maintained by CMV vectors might be a potential correlate of protection and mechanism of controlling potential HIV-1 infections.
  • 83.Patterson LJ, Kuate S, Daltabuit-Test M, Li Q, Xiao P, McKinnon K, DiPasquale J, Cristillo A, Venzon D, Haase A, et al. Replicating adenovirus-simian immunodeficiency virus (SIV) vectors efficiently prime SIV-specific systemic and mucosal immune responses by targeting myeloid dendritic cells and persisting in rectal macrophages, regardless of immunization route. Clin. Vaccine Immunol. 2012;19:629–637. doi: 10.1128/CVI.00010-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Xiao P, Patterson LJ, Kuate S, Brocca-Cofano E, Thomas MA, Venzon D, Zhao J, DiPasquale J, Fenizia C, Lee EM, et al. Replicating adenovirus-simian immunodeficiency virus (SIV) recombinant priming and envelope protein boosting elicits localized, mucosal IgA immunity in rhesus macaques correlated with delayed acquisition following a repeated low-dose rectal SIV(mac251) ch. J. Virol. 2012;86:4644–4657. doi: 10.1128/JVI.06812-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Maxfield LF, Abbink P, Stephenson KE, Borducchi EN, Ng’ang'a D, Kirilova MM, Paulino N, Boyd M, Shabram P, Ruan Q, et al. Attenuation of Replication-Competent Adenovirus Serotype 26 Vaccines by Vectorization. Clin. Vaccine Immunol. 2015;22:1166–1175. doi: 10.1128/CVI.00510-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Rose NF, Marx PA, Luckay A, Nixon DF, Moretto WJ, Donahoe SM, Montefiori D, Roberts A, Buonocore L, Rose JK. An Effective AIDS Vaccine Based on Live Attenuated Vesicular Stomatitis Virus Recombinants. Cell. 2001;106:539–549. doi: 10.1016/s0092-8674(01)00482-2. [DOI] [PubMed] [Google Scholar]

RESOURCES