Sequential Immunization with gp140 Boosts Immune Responses Primed by Modified Vaccinia Ankara or DNA in HIV-Uninfected South African Participants

Gavin Churchyard, Koleka Mlisana, Shelly Karuna, Anna-Lise Williamson, Carolyn Williamson, Lynn Morris, Georgia D Tomaras, Stephen C De Rosa, Peter B Gilbert, Niya Gu, Chenchen Yu, Nonhlanhla N Mkhize, Tandile Hermanus, Mary Allen, Michael Pensiero, Susan W Barnett, Glenda Gray, Linda-Gail Bekker, David C Montefiori, James Kublin, Lawrence Corey, Gavin Churchyard, Koleka Mlisana, Shelly Karuna, Anna-Lise Williamson, Carolyn Williamson, Lynn Morris, Georgia D Tomaras, Stephen C De Rosa, Peter B Gilbert, Niya Gu, Chenchen Yu, Nonhlanhla N Mkhize, Tandile Hermanus, Mary Allen, Michael Pensiero, Susan W Barnett, Glenda Gray, Linda-Gail Bekker, David C Montefiori, James Kublin, Lawrence Corey

Abstract

Background: The safety and immunogenicity of SAAVI DNA-C2 (4 mg IM), SAAVI MVA-C (2.9 x 109 pfu IM) and Novartis V2-deleted subtype C gp140 (100 mcg) with MF59 adjuvant in various vaccination regimens was evaluated in HIV-uninfected adults in South Africa.

Methods: Participants at three South African sites were randomized (1:1:1:1) to one of four vaccine regimens: MVA prime, sequential gp140 protein boost (M/M/P/P); concurrent MVA/gp140 (MP/MP); DNA prime, sequential MVA boost (D/D/M/M); DNA prime, concurrent MVA/gp140 boost (D/D/MP/MP) or placebo. Peak HIV specific humoral and cellular responses were measured.

Results: 184 participants were enrolled: 52% were female, all were Black/African, median age was 23 years (range, 18-42 years) and 79% completed all vaccinations. 159 participants reported at least one adverse event, 92.5% were mild or moderate. Five, unrelated, serious adverse events were reported. The M/M/P/P and D/D/MP/MP regimens induced the strongest peak neutralizing and binding antibody responses and the greatest CD4+ T-cell responses to Env. All peak neutralizing and binding antibody responses decayed with time. The MVA, but not DNA, prime contributed to the humoral and cellular immune responses. The D/D/M/M regimen was poorly immunogenic overall but did induce modest CD4+ T-cell responses to Gag and Pol. CD8+ T-cell responses to any antigen were low for all regimens.

Conclusions: The SAAVI DNA-C2, SAAVI MVA-C and Novartis gp140 with MF59 adjuvant in various combinations were safe and induced neutralizing and binding antibodies and cellular immune responses. Sequential immunization with gp140 boosted immune responses primed by MVA or DNA. The best overall immune responses were seen with the M/M/P/P regimen.

Trial registration: ClinicalTrials.gov NCT01418235.

Conflict of interest statement

The authors have read the journal's policy and the authors of this manuscript have the following competing interests: MA and MP are employed by the National Institute of Allergy and Infectious Diseases (NIAID), the study sponsor. GC, KM, SK, CW, A-LW, LM, GDT, SCD, PG, NG, CY, GG, LGB, DCM, JK, LC were recipients of NIAID funding, and this publication is a result of activities funded by NIAID. MA and MP were not involved with the process of funding these awards, nor in their administration or scientific aspects, and, in accordance with NIAID policies, are deferred from decisions regarding funding of coauthors for a requisite period. This study was also partly funded by Novartis Vaccines. At the time the study was conducted, Susan W. Barnett was an employee and stakeholder of Novartis Vaccines and Diagnostics. This does not alter our adherence to PLOS ONE policies on sharing data and materials (as detailed online in the guide for authors, http://www.PLOSone.org/static/editorial.action#competing).

Figures

Fig 1. CONSORT flow diagram: number of…
Fig 1. CONSORT flow diagram: number of individuals assessed for eligibility, enrolled and randomized to vaccine or placebo, followed-up and analysed.
T1: MVA prime, sequential gp140 boost (M/M/P/P); T2 (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4 (D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo. P1-P4 = placebo.
Fig 2. Local and systemic reactinogenicity by…
Fig 2. Local and systemic reactinogenicity by treatment group.
Local and systemic reactinogenicity for the pooled placebo and each vaccine regimen (T1-T4) are shown in the top and bottom panel respectively. None, mild, moderate and severe reactions are colour coded blue, yellow, orange and red respectively. T1: MVA prime, sequential gp140 boost (M/M/P/P); T2 (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4 (D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo. Two-sided unadjusted p-values from Fisher’s exact tests were used to compare proportions of participants that had local or systemic reactions between each pair of vaccine arms.
Fig 3. Peak neutralizing antibody titers.
Fig 3. Peak neutralizing antibody titers.
TZM-bl neutralizing antibody titers by treatment group at peak immunogenicity (2 weeks post last vaccination) over the panel of 5 virus isolates (Tier 1: Clade B [MN.3, SF162], Clade C [MW925]; Tier 2: Clade C [Du151.2, TV1.21]). Each dot represents an individual, with data from responders in red and non-responders in blue. Box plots based on data from responders only are shown. The mid-line of the box denotes the median and the ends of the box denote the 25th and 75th percentiles. Whiskers extend to the extreme data points that are no more than 1.5 times the interquartile range or if no value meets this criterion, to the data extremes. The number and percent positive responders in each group are shown above the graphs. T1: MVA prime, sequential gp140 boost (M/M/P/P); T2: (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4 (D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo.
Fig 4. Peak neutralizing antibody–magnitude breadth curves.
Fig 4. Peak neutralizing antibody–magnitude breadth curves.
Average magnitude-breadth (MB) curves in the TZM-bl assay at peak immunogenicity (2 weeks post last vaccination) by treatment group. The area under the curve (AUC)-MB across all subjects in each group was calculated as the average of the log10 neutralizing antibody titers over the panel of 5 virus isolates (Tier 1: Clade B [MN.3, SF162], Clade C [MW965.26]; Tier 2: Clade C [Du151.2, TV1.21]). T1: MVA prime, sequential gp140 boost (M/M/P/P); T2 (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4(D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo. Holm-Bonferroni adjusted p-values from unequal-variance t-tests were used for testing different mean AUC-MBs between vaccine arms accounting for the six pairs of vaccine arm comparisons.
Fig 5. Kinetics of neutralizing antibody responses.
Fig 5. Kinetics of neutralizing antibody responses.
TZM-bl neutralizing antibody titers by treatment group at all time-points, including the decay time point (visit 14). Neutralizing antibody titers against the viruses (A). MW965.26 (Clade C; Tier 1) and (B) MN.3 (Clade B; Tier 1), that had the highest responses at the peak immunogenicity time point are shown. Each dot represents an individual, with data from responders in red and non-responders in blue. Box plots based on data from responders only are shown. The number and percent positive responders in each group are shown above the graphs. T1: MVA prime, sequential gp140 boost (M/M/P/P), peak responses at visit 12; T2: concurrent MVA/gp140 (MP/MP), peak responses at visit 9; T3: DNA prime, sequential MVA boost (D/D/M/M), peak responses at visit 12; T4: DNA prime, concurrent MVA/gp140 boost (D/D/MP/MP), peak responses at visit 12. The decay kinetics time point was measured at visit 14, 6 months after last vaccination of M/M/P/P, D/D/P/P and D/D/MP/MP and 9 months after last vaccination of MP/MP.
Fig 6. Peak IgG binding antibody response.
Fig 6. Peak IgG binding antibody response.
Binding magnitude of IgG responses to gp120 and gp140 antigens are shown as mean fluorescent intensity (MFI) in top, middle and lower panels, respectively. Positive responders are indicated in red and negative responders in blue. The mid-line of the box denotes the median and the ends of the box denote the 25th and 75th percentiles. The whiskers that extend from the top and bottom of the box extend to the most extreme data points that are no more than 1.5 times the interquartile range (i.e., height of the box) or if no value meets this criterion, to the data extremes. The number and percent positive responders in each group are shown above the graphs. Placebo recipients from all treatment groups are shown together. T1: MVA prime, sequential gp140 boost (M/M/P/P); T2 (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4 (D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo.
Fig 7. Kinetics of IgA binding antibody…
Fig 7. Kinetics of IgA binding antibody responses to Con S gp140 CFI.
Binding magnitude of IgA responses to Con S gp140 CFI is shown as mean fluorescent intensity (MFI) in top, middle and lower panels, respectively. Positive responders are indicated in red and negative responders in blue. The mid-line of the box denotes the median and the ends of the box denote the 25th and 75th percentiles. The whiskers that extend from the top and bottom of the box extend to the most extreme data points that are no more than 1.5 times the interquartile range (i.e., height of the box) or if no value meets this criterion, to the data extremes. The number and percent positive responders in each group are shown above the graphs. Placebo recipients from all treatment groups are shown together. T1: MVA prime, sequential gp140 boost (M/M/P/P); T2 (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4 (D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo.
Fig 8. Kinetics of IgG binding antibody…
Fig 8. Kinetics of IgG binding antibody responses.
The geometric mean (and 95% error bars) of net responders are shown for visit 9, 12 and 14. T1: MVA prime, sequential gp140 boost (M/M/P/P), peak responses at visit 12; T2: concurrent MVA/gp140 (MP/MP), peak responses at visit 9; T3: DNA prime, sequential MVA boost (D/D/M/M), peak responses at visit 12; T4: DNA prime, concurrent MVA/gp140 boost (D/D/MP/MP), peak responses at visit 12. The decay kinetics time point was measured at visit 14, 6 months after last vaccination of M/M/P/P, D/D/P/P and D/D/MP/MP and 9 months after last vaccination of MP/MP.
Fig 9. Peak T-cell responses to vaccine…
Fig 9. Peak T-cell responses to vaccine antigens.
CD4+ and CD8+ background-adjusted T-cell responses as determined by intracellular cytokine staining 2 weeks following the fourth vaccination in groups 1, 3 and 4 (month 6.5) and following the second vaccination in group 2 (month 3.5). Because of overlap of peptides between PTEg peptide pools for the same HIV protein, the magnitude for the protein is calculated as the maximum among the peptide pools for the protein. The overall magnitude is calculated as the sum of the individual HIV protein magnitudes. Plots include data from responders in red and non-responders in blue. Box plots based upon data from responders only are superimposed on the distributions. The mid-line of the box denotes the median and the ends of the box denote the 25th and 75th percentiles. The whiskers that extend from the top and bottom of the box extend to the most extreme data points that are no more than 1.5 times the interquartile range (i.e., height of the box) or if no value meets this criterion, to the data extremes. The number and percent positive responders in each group are shown above the graphs. Placebo recipients from all treatment groups are shown together. T1 (M/M/P/P): MVA prime, sequential gp140 boost; T2 (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4 (D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo.
Fig 10. Polyfunctionality analysis of peak CD4+…
Fig 10. Polyfunctionality analysis of peak CD4+ T-cell Env-specific responses.
Only responses positive for IFN-γ and/or IL-2 are shown; data are for the Env pool with the largest response within each individual at the peak timepoint for each treatment group. The upper left graph shows the proportion of Env-specific cells expressing the indicated number of functions. Total Env-specific cells are determined as cells producing any of the 5 functions alone or in combination, except that granzyme B must be co-expressed with another function since it is constitutively expressed. The next graphs show the proportion of cells within each degree of functionality expressing the indication combinations of functions. T1 (M/M/P/P): MVA prime, sequential gp140 boost; T2 (MP/MP): concurrent MVA/gp140; T3 (D/D/M/M): DNA prime, sequential MVA boost; T4 (D/D/MP/MP): DNA prime, concurrent MVA/gp140 boost) or placebo.

References

    1. Joint United Nations Programme on HIV/AIDS (UNAIDS). GLOBAL REPORT: UNAIDS report on the global AIDS epidemic 2013. 2013. Report No.: UNAIDS / JC2502/1/E.
    1. Shisana OR, Rehele T, Simbayi LC, Zuma K, Jooste S, Zungu, et al. South African National HIV Prevalence, Incidence and Behaviour Survey, 2012 Human Sciences Research Council Press, Cape Town; 2014.
    1. Bor J, Herbst AJ, Newell ML, Barnighausen T. Increases in adult life expectancy in rural South Africa: valuing the scale-up of HIV treatment. Science 2013;339:961–5. 10.1126/science.1230413
    1. Tanser F, Barnighausen T, Grapsa E, Zaidi J, Newell ML. High coverage of ART associated with decline in risk of HIV acquisition in rural KwaZulu-Natal, South Africa. Science 2013;339:966–71. 10.1126/science.1228160
    1. Williamson AL, Rybiki E, Shephard E, Gray G, Bekker LG, Downing K, et al. South African HIV-1 vaccine candidates—the journey from the bench to clinical trials. S Afr Med J 2012;102:452–5.
    1. Williamson C, Morris L, Maughan MF, Ping LH, Dryga SA, Thomas R, et al. Characterization and selection of HIV-1 subtype C isolates for use in vaccine development. AIDS Res Hum Retroviruses 2003;19:133–44.
    1. Burgers WA, van Harmelen JH, Shephard E, Adams C, Mgwebi T, Bourn W, et al. Design and preclinical evaluation of a multigene human immunodeficiency virus type 1 subtype C DNA vaccine for clinical trial. J Gen Virol 2006;87:399–410.
    1. Burgers WA, Shephard E, Monroe JE, Greenhalgh T, Binder A, Hurter E, et al. Construction, characterization, and immunogenicity of a multigene modified vaccinia Ankara (MVA) vaccine based on HIV type 1 subtype C. AIDS Res Hum Retroviruses 2008;24:195–206. 10.1089/aid.2007.0205
    1. Shephard E, Burgers WA, van Harmelen JH, Monroe JE, Greenhalgh T, Williamson C, et al. A multigene HIV type 1 subtype C modified vaccinia Ankara (MVA) vaccine efficiently boosts immune responses to a DNA vaccine in mice. AIDS Res Hum Retroviruses 2008;24:207–17. 10.1089/aid.2007.0206
    1. Burgers WA, Chege GK, Muller TL, van Harmelen JH, Khoury G, Shephard EG, et al. Broad, high-magnitude and multifunctional CD4+ and CD8+ T-cell responses elicited by a DNA and modified vaccinia Ankara vaccine containing human immunodeficiency virus type 1 subtype C genes in baboons. J Gen Virol 2009;90:468–80. 10.1099/vir.0.004614-0
    1. Gray G, Elizaga M, Bekker LG. Immunogenicity of a subtype C HIV vaccine regimen, the SAAVI DNA-C2 vaccine boosted by SAAVI MVA-C vaccine: Results of a Phase I study conducted in South Africa and USA amongst HIV uninfected adults (HVTN 073/SAAVI 102). Conference on Retroviruses and Opportunistic Infections (CROI). 2011.
    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;361:2209–20. 10.1056/NEJMoa0908492
    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;366:1275–86. 10.1056/NEJMoa1113425
    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;372:1881–93. 10.1016/S0140-6736(08)61591-3
    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;369:2083–92. 10.1056/NEJMoa1310566
    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. Lancet Infect Dis 2011;11:507–15. 10.1016/S1473-3099(11)70098-6
    1. Lin L, Finak G, Ushey K, Seshadri C, Hawn TR, Frahm N, et al. COMPASS identifies T-cell subsets correlated with clinical outcomes. Nat Biotechnol 2015;33:610–6. 10.1038/nbt.3187
    1. Cassimatis DC, Atwood JE, Engler RM, Linz PE, Grabenstein JD, Vernalis MN. Smallpox vaccination and myopericarditis: a clinical review. J Am Coll Cardiol 2004;43:1503–10.
    1. Eckart RE, Love SS, Atwood JE, Arness MK, Cassimatis DC, Campbell CL, et al. Incidence and follow-up of inflammatory cardiac complications after smallpox vaccination. J Am Coll Cardiol 2004;44:201–5.
    1. Halsell JS, Riddle JR, Atwood JE, Gardner P, Shope R, Poland GA, et al. Myopericarditis following smallpox vaccination among vaccinia-naive US military personnel. JAMA 2003;289:3283–9.
    1. Elizaga M, Vasan S, Marovich M, Sato A, Lawrence D, Chaitman B, et al. Prospective surveillance for cardiac adverse events in healthy adults receiving modified vaccinia Ankara vaccines: a systematic review. PLoS One 2013;8: e54407 10.1371/journal.pone.0054407
    1. Lian Y, Srivastava I, Gomez-Roman VR, Zur MJ, Sun Y, Kan E, et al. Evaluation of envelope vaccines derived from the South African subtype C human immunodeficiency virus type 1 TV1 strain. J Virol 2005;79:13338–49.
    1. Srivastava IK, Kan E, Sun Y, Sharma VA, Cisto J, Burke B, et al. Comparative evaluation of trimeric envelope glycoproteins derived from subtype C and B HIV-1 R5 isolates. Virology 2008;372:273–90.
    1. O'Hagan DT, Ott GS, Nest GV, Rappuoli R, Giudice GD. The history of MF59((R)) adjuvant: a phoenix that arose from the ashes. Expert Rev Vaccines 2013;12:13–30. 10.1586/erv.12.140
    1. De Rosa SC, Carter DK, McElrath MJ. OMIP-014: validated multifunctional characterization of antigen-specific human T cells by intracellular cytokine staining. Cytometry A 2012;81:1019–21. 10.1002/cyto.a.22218
    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;82:12449–63. 10.1128/JVI.01708-08
    1. Zolla-Pazner S, deCamp A, Gilbert PB, Williams C, Yates NL, Williams WT, 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 10.1371/journal.pone.0087572
    1. Yates NL, Liao H, Fong Y, Camp A, Vandergrift NA, Williams WA, et al. Vaccine-Induced Env V1-V2 IgG3 Correlate with Lower HIV-1 Infection Risk and Decline Soon After Vaccination. Science Translational Medicine 2014;6:228ra39 10.1126/scitranslmed.3007730
    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;38:176–86. 10.1016/j.immuni.2012.11.011
    1. Gaschen B, Taylor J, Yusim K, Foley B, Gao F, Lang D, et al. Diversity considerations in HIV-1 vaccine selection. Science 2002;296:2354–60.
    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;353:268–82.
    1. Montefiori DC. Measuring HIV neutralization in a luciferase reporter gene assay In: Prasad V.R, Kalpana G.V., editors. HIV protocols: Methods in Molecular Biology. Second Edition ed. Humana Press; 2009. p. 395–405.
    1. Seaman MS, Janes H, Hawkins N, Grandpre LE, Devoy C, Giri A, et al. Tiered categorization of a diverse panel of HIV-1 Env pseudoviruses for assessment of neutralizing antibodies. J Virol 2010. February;84(3):1439–52. 10.1128/JVI.02108-09
    1. Liu W. On sample size determination of Dunnett's procedure for comparing several treatments with a control. Journal of Statistical Planning and Inference 62[2], 255–261. 1997.
    1. Huang Y, Gilbert PB, Montefiori DC, Self SG. Simultaneous Evaluation of the Magnitude and Breadth of a Left and Right Censored Multivariate Response, with Application to HIV Vaccine Development. Stat Biopharm Res 2009. February 1;1(1):81–91.
    1. Gorse GJ, Newman MJ, deCamp A, Hay CM, De Rosa SC, Noonan E, et al. DNA and modified vaccinia virus Ankara vaccines encoding multiple cytotoxic and helper T-lymphocyte epitopes of human immunodeficiency virus type 1 (HIV-1) are safe but weakly immunogenic in HIV-1-uninfected, vaccinia virus-naive adults. Clin Vaccine Immunol 2012. May;19(5):649–58. 10.1128/CVI.00038-12
    1. Karasavvas N, Karnasutra C, Ngauy V, Vasan s, Trichavaroj R, de Souza M, et al. 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 P03.68 LB. 2013.
    1. Morris L, Mkize NN, Hermanus T, Chung E, Sato A, Grant S, et al. Boosting antibody responses with gp140 protein two years after DNA/MVA priming: Results from the HVTN 073E Phase I vaccine trial. AID Vaccine 2013 P04.36 LB. 2013.
    1. Richmond JF, Lu S, Santoro JC, Weng J, Hu SL, Montefiori DC, et al. Studies of the neutralizing activity and avidity of anti-human immunodeficiency virus type 1 Env antibody elicited by DNA priming and protein boosting. J Virol 1998. November;72(11):9092–100.
    1. Vaine M, Wang S, Hackett A, Arthos J, Lu S. Antibody responses elicited through homologous or heterologous prime-boost DNA and protein vaccinations differ in functional activity and avidity. Vaccine 2010. April 9;28(17):2999–3007. 10.1016/j.vaccine.2010.02.006
    1. Letvin NL, Montefiori DC, Yasutomi Y, Perry HC, Davies ME, Lekutis C, et al. Potent, protective anti-HIV immune responses generated by bimodal HIV envelope DNA plus protein vaccination. Proc Natl Acad Sci U S A 1997. August 19;94(17):9378–83.
    1. Pal R, Kalyanaraman VS, Nair BC, Whitney S, Keen T, Hocker L, et al. Immunization of rhesus macaques with a polyvalent DNA prime/protein boost human immunodeficiency virus type 1 vaccine elicits protective antibody response against simian human immunodeficiency virus of R5 phenotype. Virology 2006. May 10;348(2):341–53.
    1. Wang S, Kennedy JS, West K, Montefiori DC, Coley S, Lawrence J, et al. Cross-subtype antibody and cellular immune responses induced by a polyvalent DNA prime-protein boost HIV-1 vaccine in healthy human volunteers. Vaccine 2008. July 23;26(31):3947–57.
    1. Frahm N, Friedrich D, Walsh P, DeRosa S, Spearman P, Barnett S, et al. A DNA Prime/Protein Boost Vaccine Leads to Higher B-cell Responses than a Vector Prime/Protein Boost or DNA Prime/Vector Boost Regimens. AIDS Vaccine Bangkok, Thailand, (OA10.06). 2011.
    1. Harari A, Bart PA, Stohr W, Tapia G, Garcia M, Medjitna-Rais E, et al. An HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responses. J Exp Med 2008. January 21;205(1):63–77. 10.1084/jem.20071331
    1. Sandstrom E, Nilsson C, Hejdeman B, Brave A, Bratt G, Robb M, et al. Broad immunogenicity of a multigene, multiclade HIV-1 DNA vaccine boosted with heterologous HIV-1 recombinant modified vaccinia virus Ankara. J Infect Dis 2008. November 15;198(10):1482–90. 10.1086/592507
    1. Bakari M, Aboud S, Nilsson C, Francis J, Buma D, Moshiro 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. October 26;29(46):8417–28. 10.1016/j.vaccine.2011.08.001
    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. July 1;210(1):99–110. 10.1093/infdis/jiu003
    1. McCormack S, Stohr W, Barber T, Bart PA, Harari A, Moog C, et al. EV02: a Phase I trial to compare the safety and immunogenicity of HIV DNA-C prime-NYVAC-C boost to NYVAC-C alone. Vaccine 2008. June 13;26(25):3162–74. 10.1016/j.vaccine.2008.02.072
    1. Gomez CE, Perdiguero B, Garcia-Arriaza J, Esteban M. Poxvirus vectors as HIV/AIDS vaccines in humans. Hum Vaccin Immunother 2012. September;8(9):1192–207. 10.4161/hv.20778
    1. Garcia F, Bernaldo de Quiros JC, Gomez CE, Perdiguero B, Najera JL, Jimenez V, et al. Safety and immunogenicity of a modified pox vector-based HIV/AIDS vaccine candidate expressing Env, Gag, Pol and Nef proteins of HIV-1 subtype B (MVA-B) in healthy HIV-1-uninfected volunteers: A phase I clinical trial (RISVAC02). Vaccine 2011. October 26;29(46):8309–16. 10.1016/j.vaccine.2011.08.098
    1. Gomez CE, Najera JL, Perdiguero B, Garcia-Arriaza J, Sorzano CO, Jimenez V, et al. The HIV/AIDS vaccine candidate MVA-B administered as a single immunogen in humans triggers robust, polyfunctional, and selective effector memory T cell responses to HIV-1 antigens. J Virol 2011. November;85(21):11468–78. 10.1128/JVI.05165-11
    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. March 1;203(5):610–9. 10.1093/infdis/jiq105
    1. Keefer MC, Frey SE, Elizaga M, Metch B, De Rosa SC, Barroso PF, et al. A phase I trial of preventive HIV vaccination with heterologous poxviral-vectors containing matching HIV-1 inserts in healthy HIV-uninfected subjects. Vaccine 2011. February 24;29(10):1948–58. 10.1016/j.vaccine.2010.12.104
    1. Yates NL, Liao HX, 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. Sci Transl Med 2014. March 19;6(228):228ra39 10.1126/scitranslmed.3007730
    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. May 28;110(22):9019–24. 10.1073/pnas.1301456110

Source: PubMed

3
S'abonner