Immunogenicity of a novel Clade B HIV-1 vaccine combination: Results of phase 1 randomized placebo controlled trial of an HIV-1 GM-CSF-expressing DNA prime with a modified vaccinia Ankara vaccine boost in healthy HIV-1 uninfected adults

Susan P Buchbinder, Nicole A Grunenberg, Brittany J Sanchez, Kelly E Seaton, Guido Ferrari, M Anthony Moody, Nicole Frahm, David C Montefiori, Christine M Hay, Paul A Goepfert, Lindsey R Baden, Harriet L Robinson, Xuesong Yu, Peter B Gilbert, M Juliana McElrath, Yunda Huang, Georgia D Tomaras, HIV Vaccine Trials Network (HVTN) 094 Study Group, Susan P Buchbinder, Nicole A Grunenberg, Brittany J Sanchez, Kelly E Seaton, Guido Ferrari, M Anthony Moody, Nicole Frahm, David C Montefiori, Christine M Hay, Paul A Goepfert, Lindsey R Baden, Harriet L Robinson, Xuesong Yu, Peter B Gilbert, M Juliana McElrath, Yunda Huang, Georgia D Tomaras, HIV Vaccine Trials Network (HVTN) 094 Study Group

Abstract

Background: A phase 1 trial of a clade B HIV vaccine in HIV-uninfected adults evaluated the safety and immunogenicity of a DNA prime co-expressing GM-CSF (Dg) followed by different numbers and intervals of modified vaccinia Ankara Boosts (M). Both vaccines produce virus-like particles presenting membrane-bound Env.

Methods: Four US sites randomized 48 participants to receiving 1/10th the DNA dose as DgDgMMM given at 0, 2, 4, 6 and 8 months, or full dose DgDgM_M or DgDgMM_M regimens, given at 0, 2, 4, and 8 months, and 0, 2, 4, 6, and 10 months, respectively. Peak immunogenicity was measured 2 weeks post-last vaccination.

Results: All regimens were well tolerated and safe. Full dose DgDgM_M and DgDgMM_M regimens generated Env-specific IgG to HIV-1 Env in >90%, IgG3 in >80%, and IgA in <20% of participants. Responses to gp140 and gp41 targets were more common and of higher magnitude than to gp120 and V1V2. The gp41 antibody included reactivity to the conserved immunodominant region with specificities known to mediate virus capture and phagocytosis and did not cross-react with a panel of intestinal flora antigens. The 3rd dose of MVA increased the avidity of elicited antibody (7.5% to 39%), the ADCC response to Bal gp120 (14% to 64%), and the one-year durability of the IgG3 responses to gp41 by 4-fold (13% vs. 3.5% retention of peak response). The co-expressed GM-CSF did not enhance responses over those in trials testing this vaccine without GM-CSF.

Conclusion: This DNA/MVA prime-boost regimen induced durable, functional humoral responses that included ADCC, high antibody avidity, and Env IgG1 and IgG3 binding responses to the immunodominant region of gp41. The third, spaced MVA boost improved the overall quality of the antibody response. These products without co-expressed GM-CSF but combined with protein boosts will be considered for efficacy evaluation.

Trial registration: ClinicalTrials.gov NCT01571960.

Conflict of interest statement

Competing Interests: The authors have read the journal's policy and the authors of this manuscript have the following competing interests: SPB, NAG, BJS, KES, GF, MAM, NF, DCM, CMH, PAG, LRB, XY, MJM, YH and GDT are recipients of NIAID funding, and this publication is a result of activities funded by NIAID. This study was also partly funded by GeoVax, Inc. HLR is an employee of GeoVax Labs, Inc. HLR is co-founder and owns stock in GeoVax Labs, Inc. and is an inventor on U.S. Patents 7,795,017 entitled DNA Expression Vectors and Methods of Use; 8,623,379 entitled Compositions and Methods for Generating an Immune Response; 7,867,982 entitled MVA expressing Modified HIV Envelope, gag and pol genes and 9,453,239, entitled recombinant MVA viruses expressing clade A/G, clade B and clade C modified HIV env, gag, and pol genes. This does not alter her adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. CONSORT flow diagram.
Fig 1. CONSORT flow diagram.
Allocation, follow-up, and analysis for HIV Vaccine Trials Network (HVTN) study 094. Abbreviations: 1/10 DgDgMMM, treatment arm receiving two 1/10 doses of DNA followed by three doses of MVA at 0, 2, 4, 6 and 8 months; DgDgMM_M, treatment arm receiving two full doses of DNA followed by three doses of MVA given at 0, 2, 4, 6, and 10 months; DgDgM_M, treatment arm receiving two full doses of DNA followed by two doses of MVA at 0, 2, 4, and 8 months; ADCC, antibody dependent cellular cytotoxicity.
Fig 2. Summary immune responses.
Fig 2. Summary immune responses.
Overall peak binding antibody and ICS response rates among vaccine recipients at two weeks after the 2nd and 3rd MVA in the DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. A positive response to IgG Env or IgA Env indicates positive responses to at least one ENV antigens measured by the BAMA binding antibody assay; a positive response to ANY CD4+ or ANY CD8+ indicates positive responses to any least one peptide pool and/or cytokine measured by the ICS assay.
Fig 3. Peak IgG and IgG3 response…
Fig 3. Peak IgG and IgG3 response rates and magnitudes.
(A) Peak IgG binding antibody response rates. (B) Peak IgG binding response magnitudes. (C) Peak IgG3 binding antibody response rates. (D) Peak IgG3 binding antibody response magnitudes. Responses are to individual antigens at two weeks after the 2nd and 3rd MVA or placebo in the Placebo, DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. Shown are total IgG binding antibody response rates to HIV-1 Env gp140 (ConS gp140), HIV-1 Env gp120 (Con 6 gp120 B), the V1V2 loop, gp41 and the gp41 immunodominant region (IDR).
Fig 4. IgG and IgG3 response rates…
Fig 4. IgG and IgG3 response rates and magnitudes over time.
(A) Total IgG and IgG3 binding antibody response rates. (B) Total IgG and IgG3 binding antibody response median magnitudes and inter-quartile range (IQR) responses. Responses among vaccine recipients over time at two weeks, 6 months, 9 months, and 12 months after the last vaccination in the DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. For the comparison of two visits within an arm, McNemar’s and Wilcoxon signed rank tests were used to compare the response rates (A) and magnitudes (B) among responders, respectively.
Fig 5. Antibody avidity.
Fig 5. Antibody avidity.
Distribution of binding antibody avidity response magnitude among vaccine recipients at two weeks and 6 months after the 2nd and 3rd MVA in the DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. Avidity Index (AI) was calculated for the ID epitope tetramer for those with a positive binding response to the ID epitope tetramer and only for samples not saturated (MFI

Fig 6. A. ADCC response rate (GTL).…

Fig 6. A. ADCC response rate (GTL). B. ADCC response magnitude.

(A) Peak ADCC response…

Fig 6. A. ADCC response rate (GTL). B. ADCC response magnitude.
(A) Peak ADCC response rates. (B) Peak ADCC response magnitudes. Responses are to individual antigens at two weeks after the 2nd and 3rd MVA or placebo in the Placebo, DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. For the DgDgM_M arm, only peak percent Granzyme B activity for BaL gp120 was measured. Positive responses are shown as filled circles and negative responses are shown as open circles (B). Box-plots represent the distribution for the positive responders only. Response rates were compared using Fisher’s exact test (A); response magnitudes among responders across treatment arms were compared using Wilcoxon Rank Sum test (B).

Fig 7. Cellular immune responses measured by…

Fig 7. Cellular immune responses measured by intracellular cytokine staining (ICS).

(A) CD4+ response rates.…

Fig 7. Cellular immune responses measured by intracellular cytokine staining (ICS).
(A) CD4+ response rates. (B) CD4+ response magnitude. (C) CD8+ response rate. (D) CD8+ response magnitudes. Peak ICS response rates and response magnitude to Gag, Env and Pol in the CD4+ (Panels A & B) and CD8+ (Panels C & D) T-cell subsets at two weeks after the 2nd and 3rd MVA or placebo in the Placebo, DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. Shown are response rates (A, C) and response magnitudes (B, D) for the sum of individual antigen responses, reported as % of CD4+ or CD8+ T cells producing IFN-gamma and/or IL-2. Positive responses are shown as filled circles and negative responses are shown as open circles (B, D). Box-plots represent the distribution for the positive responders only. Response rates were compared using Fisher’s exact test (A, C); response magnitudes among responders across treatment arms were compared using Wilcoxon Rank Sum test (B, D).

Fig 8. Radar plot of peak antibody-mediated…

Fig 8. Radar plot of peak antibody-mediated and cellular immune responses by vaccine regimen.

(A)…

Fig 8. Radar plot of peak antibody-mediated and cellular immune responses by vaccine regimen.
(A) Response rates of each assay readout. (B) Response magnitudes of each assay readout. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively.
All figures (8)
Fig 6. A. ADCC response rate (GTL).…
Fig 6. A. ADCC response rate (GTL). B. ADCC response magnitude.
(A) Peak ADCC response rates. (B) Peak ADCC response magnitudes. Responses are to individual antigens at two weeks after the 2nd and 3rd MVA or placebo in the Placebo, DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. For the DgDgM_M arm, only peak percent Granzyme B activity for BaL gp120 was measured. Positive responses are shown as filled circles and negative responses are shown as open circles (B). Box-plots represent the distribution for the positive responders only. Response rates were compared using Fisher’s exact test (A); response magnitudes among responders across treatment arms were compared using Wilcoxon Rank Sum test (B).
Fig 7. Cellular immune responses measured by…
Fig 7. Cellular immune responses measured by intracellular cytokine staining (ICS).
(A) CD4+ response rates. (B) CD4+ response magnitude. (C) CD8+ response rate. (D) CD8+ response magnitudes. Peak ICS response rates and response magnitude to Gag, Env and Pol in the CD4+ (Panels A & B) and CD8+ (Panels C & D) T-cell subsets at two weeks after the 2nd and 3rd MVA or placebo in the Placebo, DgDgM_M and DgDgMM_M groups. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively. Shown are response rates (A, C) and response magnitudes (B, D) for the sum of individual antigen responses, reported as % of CD4+ or CD8+ T cells producing IFN-gamma and/or IL-2. Positive responses are shown as filled circles and negative responses are shown as open circles (B, D). Box-plots represent the distribution for the positive responders only. Response rates were compared using Fisher’s exact test (A, C); response magnitudes among responders across treatment arms were compared using Wilcoxon Rank Sum test (B, D).
Fig 8. Radar plot of peak antibody-mediated…
Fig 8. Radar plot of peak antibody-mediated and cellular immune responses by vaccine regimen.
(A) Response rates of each assay readout. (B) Response magnitudes of each assay readout. DgDgM_M refers to immune responses after the last MVA in the DgDgM_M group; DgDgMM and DgDgMM_M refer to immune responses after the 2nd and 3rd MVA in DgDgMM_M group, respectively.

References

    1. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, Paris R, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 2009;361(23):2209–20. doi:
    1. 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(5):654–65. doi:
    1. Pitisuttithum P, Gilbert P, Gurwith M, Heyward W, Martin M, van Griensven F, 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(12):1661–71. doi:
    1. Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, Li D, 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(9653):1881–93. doi:
    1. Gray GE, Allen M, Moodie Z, Churchyard G, Bekker LG, Nchabeleng M, 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. The Lancet infectious diseases. 2011;11(7):507–15. doi:
    1. Hammer SM, Sobieszczyk ME, Janes H, Karuna ST, Mulligan MJ, Grove D, et al. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N Engl J Med. 2013;369(22):2083–92. doi:
    1. Robb ML, Rerks-Ngarm S, Nitayaphan S, Pitisuttithum P, Kaewkungwal J, Kunasol P, 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. The Lancet infectious diseases. 2012;12(7):531–7. doi:
    1. Tomaras GD, Haynes BF. Advancing Toward HIV-1 Vaccine Efficacy through the Intersections of Immune Correlates. Vaccines. 2014;2(1):15–35. doi:
    1. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med. 2012;366(14):1275–86. doi:
    1. Gottardo R, Bailer RT, Korber BT, Gnanakaran S, Phillips J, Shen X, 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(9):e75665 doi:
    1. Rolland M, Edlefsen PT, Larsen BB, Tovanabutra S, Sanders-Buell E, Hertz T, et al. Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2. Nature. 2012;490(7420):417–20. doi:
    1. Liao HX, Bonsignori M, Alam SM, McLellan JS, Tomaras GD, Moody MA, 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(1):176–86. doi:
    1. Pollara J, Bonsignori M, Moody MA, Liu P, Alam SM, Hwang KK, et al. HIV-1 vaccine-induced C1 and V2 Env-specific antibodies synergize for increased antiviral activities. J Virol. 2014;88(14):7715–26.
    1. Yates NL, Liao H-X, Fong Y, deCamp A, Vandergrift NA, Williams WT, et al. Vaccine-Induced Env V1-V2 IgG3 Correlates with Lower HIV-1 Infection Risk and Declines Soon After Vaccination. Science Translational Medicine. 2014;6(228):228ra39–ra39. doi:
    1. Tomaras GD, Ferrari G, Shen X, Alam SM, Liao HX, Pollara J, 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(22):9019–24. doi:
    1. Goepfert PA, Elizaga ML, Seaton K, Tomaras GD, Montefiori DC, Sato A, et al. Specificity and 6-month durability of immune responses induced by DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J Infect Dis. 2014;210(1):99–110. doi:
    1. Chamcha V, Kannanganat S, Gangadhara S, Nabi R, Kozlowski PA, Montefiori DC, et al. Strong, but Age-Dependent, Protection Elicited by a Deoxyribonucleic Acid/Modified Vaccinia Ankara Simian Immunodeficiency Virus Vaccine. Open forum infectious diseases. 2016;3(1):ofw034 doi:
    1. Ledgerwood JE, Zephir K, Hu Z, Wei CJ, Chang L, Enama ME, et al. Prime-boost interval matters: a randomized phase 1 study to identify the minimum interval necessary to observe the H5 DNA influenza vaccine priming effect. J Infect Dis. 2013;208(3):418–22. doi:
    1. Lai L, Kwa S, Kozlowski PA, Montefiori DC, Ferrari G, Johnson WE, et al. Prevention of infection by a granulocyte-macrophage colony-stimulating factor co-expressing DNA/modified vaccinia Ankara simian immunodeficiency virus vaccine. J Infect Dis. 2011;204(1):164–73. doi:
    1. Smith JM, Amara RR, Campbell D, Xu Y, Patel M, Sharma S, et al. DNA/MVA vaccine for HIV type 1: effects of codon-optimization and the expression of aggregates or virus-like particles on the immunogenicity of the DNA prime. AIDS Res Hum Retroviruses. 2004;20(12):1335–47. doi:
    1. Smith JM, Amara RR, McClure HM, Patel M, Sharma S, Yi H, et al. Multiprotein HIV type 1 clade B DNA/MVA vaccine: construction, safety, and immunogenicity in Macaques. AIDS Res Hum Retroviruses. 2004;20(6):654–65. doi:
    1. Hellerstein M, Xu Y, Marino T, Lu S, Yi H, Wright ER, et al. Co-expression of HIV-1 virus-like particles and granulocyte-macrophage colony stimulating factor by GEO-D03 DNA vaccine. Hum Vaccin Immunother. 2012;8(11):1654–8. doi:
    1. Wyatt LS, Earl PL, Liu JY, Smith JM, Montefiori DC, Robinson HL, et al. Multiprotein HIV type 1 clade B DNA and MVA vaccines: construction, expression, and immunogenicity in rodents of the MVA component. AIDS Res Hum Retroviruses. 2004;20(6):645–53. doi:
    1. Tomaras GD, Yates NL, Liu P, Qin L, Fouda GG, Chavez LL, et al. Initial B-cell responses to transmitted human immunodeficiency virus type 1: virion-binding immunoglobulin M (IgM) and IgG antibodies followed by plasma anti-gp41 antibodies with ineffective control of initial viremia. J Virol. 2008;82(24):12449–63.
    1. Yates NL, Lucas JT, Nolen TL, Vandergrift NA, Soderberg KA, Seaton KE, et al. Multiple HIV-1-specific IgG3 responses decline during acute HIV-1: implications for detection of incident HIV infection. AIDS. 2011;25(17):2089–97. doi:
    1. Archary D, Seaton KE, Passmore JS, Werner L, Deal A, Dunphy LJ, et al. Distinct genital tract HIV-specific antibody profiles associated with tenofovir gel. Mucosal Immunol. 2016.
    1. Gao F, Weaver EA, Lu Z, Li Y, Liao HX, Ma B, et al. Antigenicity and immunogenicity of a synthetic human immunodeficiency virus type 1 group m consensus envelope glycoprotein. J Virol. 2005;79(2):1154–63.
    1. Gaschen B, Taylor J, Yusim K, Foley B, Gao F, Lang D, et al. Diversity considerations in HIV-1 vaccine selection. Science. 2002;296(5577):2354–60. doi:
    1. Liao HX, Sutherland LL, Xia SM, Brock ME, Scearce RM, Vanleeuwen S, et al. A group M consensus envelope glycoprotein induces antibodies that neutralize subsets of subtype B and C HIV-1 primary viruses. Virology. 2006;353(2):268–82. doi:
    1. Morris L, Chen X, Alam M, Tomaras G, Zhang R, Marshall DJ, et al. Isolation of a human anti-HIV gp41 membrane proximal region neutralizing antibody by antigen-specific single B cell sorting. PLoS ONE. 2011;6(9):e23532 doi:
    1. Williams WB, Liao HX, Moody MA, Kepler TB, Alam SM, Gao F, et al. HIV-1 VACCINES. Diversion of HIV-1 vaccine-induced immunity by gp41-microbiota cross-reactive antibodies. Science. 2015;349(6249):aab1253 doi:
    1. Seaton KE, Lan A, Goepfert P, Alam SM, Parekh B, Robinson HL, et al. Increased Avidity Index to Immunodominant Epitope of HIV-1 gp41 Env with MVA prime/boost Vs. DNA Prime MVA/boost In Human HIV-1 Vaccine Trial (HVTN 065). Keystone Symposia, HIV Vaccines 2012; Keystone, CO.
    1. Wei X, Liu X, Dobbs T, Kuehl D, Nkengasong JN, Hu DJ, et al. Development of two avidity-based assays to detect recent HIV type 1 seroconversion using a multisubtype gp41 recombinant protein. AIDS Res Hum Retroviruses. 2010;26(1):61–71. doi:
    1. Pollara J, Hart L, Brewer F, Pickeral J, Packard BZ, Hoxie JA, et al. High-throughput quantitative analysis of HIV-1 and SIV-specific ADCC-mediating antibody responses. Cytometry Part A: the journal of the International Society for Analytical Cytology. 2011;79(8):603–12.
    1. Horton H, Thomas EP, Stucky JA, Frank I, Moodie Z, Huang Y, et al. Optimization and validation of an 8-color intracellular cytokine staining (ICS) assay to quantify antigen-specific T cells induced by vaccination. Journal of immunological methods. 2007;323(1):39–54. doi:
    1. De Rosa SC. Vaccine applications of flow cytometry. Methods. 2012;57(3):383–91. doi:
    1. Montefiori DC. Measuring HIV neutralization in a luciferase reporter gene assay. Methods Mol Biol. 2009;485:395–405. doi:
    1. Sarzotti-Kelsoe M, Bailer RT, Turk E, Lin CL, Bilska M, Greene KM, et al. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. Journal of immunological methods. 2014;409:131–46. doi:
    1. Todd CA, Greene KM, Yu X, Ozaki DA, Gao H, Huang Y, et al. Development and implementation of an international proficiency testing program for a neutralizing antibody assay for HIV-1 in TZM-bl cells. Journal of immunological methods. 2012;375(1–2):57–67. doi:
    1. Agresti A, Coull B. Approximate is Better than" Exact" for Interval Estimation of Binomial Proportions. The American Statistician. 1998;52, No. 2:119–26.
    1. Horton H, Havenar-Daughton C, Lee D, Moore E, Cao J, McNevin J, et al. Induction of human immunodeficiency virus type 1 (HIV-1)-specific T-cell responses in HIV vaccine trial participants who subsequently acquire HIV-1 infection. J Virol. 2006;80(19):9779–88.
    1. Suzuki T, Trapnell BC. Pulmonary Alveolar Proteinosis Syndrome. Clin Chest Med. 2016;37(3):431–40. doi:
    1. Robinson HL. Non-neutralizing antibodies in prevention of HIV infection. Expert opinion on biological therapy. 2013;13(2):197–207. doi:
    1. Burton DR, Hessell AJ, Keele BF, Klasse PJ, Ketas TA, Moldt B, et al. Limited or no protection by weakly or nonneutralizing antibodies against vaginal SHIV challenge of macaques compared with a strongly neutralizing antibody. Proc Natl Acad Sci U S A. 2011;108(27):11181–6. doi:
    1. Moog C, Dereuddre-Bosquet N, Teillaud JL, Biedma ME, Holl V, Van Ham G, et al. Protective effect of vaginal application of neutralizing and nonneutralizing inhibitory antibodies against vaginal SHIV challenge in macaques. Mucosal Immunol. 2014;7(1):46–56. doi:
    1. Tay MZ, Liu P, Williams LD, McRaven MD, Sawant S, Gurley TC, et al. Antibody-Mediated Internalization of Infectious HIV-1 Virions Differs among Antibody Isotypes and Subclasses. PLoS pathogens. 2016;12(8):e1005817 doi:
    1. Santra S, Tomaras GD, Warrier R, Nicely NI, Liao HX, Pollara J, et al. Human Non-neutralizing HIV-1 Envelope Monoclonal Antibodies Limit the Number of Founder Viruses during SHIV Mucosal Infection in Rhesus Macaques. PLoS pathogens. 2015;11(8):e1005042 doi:
    1. Bomsel M, Tudor D, Drillet AS, Alfsen A, Ganor Y, Roger MG, et al. Immunization with HIV-1 gp41 subunit virosomes induces mucosal antibodies protecting nonhuman primates against vaginal SHIV challenges. Immunity. 2011;34(2):269–80. doi:
    1. Li Q, Zeng M, Duan L, Voss JE, Smith AJ, Pambuccian S, 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(6):3113–25. doi:
    1. Voss JE, Macauley MS, Rogers KA, Villinger F, Duan L, Shang L, et al. Reproducing SIVDeltanef vaccine correlates of protection: trimeric gp41 antibody concentrated at mucosal front lines. AIDS. 2016;30(16):2427–38. doi:
    1. Golding H, D'Souza MP, Bradac J, Mathieson B, Fast P. Neutralization of HIV-1. AIDS Res Hum Retroviruses. 1994;10(6):633–43. doi:
    1. Carias AM, Hope TJ. Phagocytosis: cell biology view of antiviral function. Current opinion in HIV and AIDS. 2014;9(3):271–7. doi:
    1. Chung AW, Ghebremichael M, Robinson H, Brown E, Choi I, Lane S, et al. Polyfunctional Fc-Effector Profiles Mediated by IgG Subclass Selection Distinguish RV144 and VAX003 Vaccines. Science Translational Medicine. 2014;6(228):228ra38–ra38. doi:
    1. Goepfert PA, Elizaga ML, Sato A, Qin L, Cardinali M, Hay CM, et al. Phase 1 safety and immunogenicity testing of DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J Infect Dis. 2011;203(5):610–9. doi:
    1. Prentice HA, Tomaras GD, Geraghty DE, Apps R, Fong Y, Ehrenberg PK, et al. HLA class II genes modulate vaccine-induced antibody responses to affect HIV-1 acquisition. Sci Transl Med. 2015;7(296):296ra112 doi:
    1. Gartland AJ, Li S, McNevin J, Tomaras GD, Gottardo R, Janes H, et al. Analysis of HLA A*02 association with vaccine efficacy in the RV144 HIV-1 vaccine trial. J Virol. 2014;88(15):8242–55.
    1. Li SS, Gilbert PB, Tomaras GD, Kijak G, Ferrari G, Thomas R, et al. FCGR2C polymorphisms associate with HIV-1 vaccine protection in RV144 trial. J Clin Invest. 2014;124(9):3879–90. doi:
    1. Manrique A, Adams E, Barouch DH, Fast P, Graham BS, Kim JH, et al. The immune space: a concept and template for rationalizing vaccine development. AIDS Res Hum Retroviruses. 2014;30(11):1017–22. doi:
    1. Centers for Disease Control and Prevention. Annual Report. Division of HIV/AIDS Prevention; 2015.
    1. Beyrer C, Baral SD, van Griensven F, Goodreau SM, Chariyalertsak S, Wirtz AL, et al. Global epidemiology of HIV infection in men who have sex with men. Lancet. 2012;380(9839):367–77. doi:
    1. Desai S, Croxford S, Brown AE, Mitchell H, Hughes G, Delpech V. An overview of the HIV epidemic among men who have sex with men in the United Kingdom, 1999–2013. Euro Surveill. 2015;20(14).
    1. Diez M, Bleda MJ, Varela JR, Ordonana J, Azpiri MA, Vall M, et al. Trends in HIV testing, prevalence among first-time testers, and incidence in most-at-risk populations in Spain: the EPI-VIH Study, 2000 to 2009. Euro Surveill. 2014;19(47):20971

Source: PubMed

3
S'abonner