Subtype C gp140 Vaccine Boosts Immune Responses Primed by the South African AIDS Vaccine Initiative DNA-C2 and MVA-C HIV Vaccines after More than a 2-Year Gap

Glenda E Gray, Kenneth H Mayer, Marnie L Elizaga, Linda-Gail Bekker, Mary Allen, Lynn Morris, David Montefiori, Stephen C De Rosa, Alicia Sato, Niya Gu, Georgia D Tomaras, Timothy Tucker, Susan W Barnett, Nonhlanhla N Mkhize, Xiaoying Shen, Katrina Downing, Carolyn Williamson, Michael Pensiero, Lawrence Corey, Anna-Lise Williamson, Glenda E Gray, Kenneth H Mayer, Marnie L Elizaga, Linda-Gail Bekker, Mary Allen, Lynn Morris, David Montefiori, Stephen C De Rosa, Alicia Sato, Niya Gu, Georgia D Tomaras, Timothy Tucker, Susan W Barnett, Nonhlanhla N Mkhize, Xiaoying Shen, Katrina Downing, Carolyn Williamson, Michael Pensiero, Lawrence Corey, Anna-Lise Williamson

Abstract

A phase I safety and immunogenicity study investigated South African AIDS Vaccine Initiative (SAAVI) HIV-1 subtype C (HIV-1C) DNA vaccine encoding Gag-RT-Tat-Nef and gp150, boosted with modified vaccinia Ankara (MVA) expressing matched antigens. Following the finding of partial protective efficacy in the RV144 HIV vaccine efficacy trial, a protein boost with HIV-1 subtype C V2-deleted gp140 with MF59 was added to the regimen. A total of 48 participants (12 U.S. participants and 36 Republic of South Africa [RSA] participants) were randomized to receive 3 intramuscular (i.m.) doses of SAAVI DNA-C2 of 4 mg (months 0, 1, and 2) and 2 i.m. doses of SAAVI MVA-C of 1.45 × 10(9) PFU (months 4 and 5) (n = 40) or of a placebo (n = 8). Approximately 2 years after vaccination, 27 participants were rerandomized to receive gp140/MF59 at 100 μg or placebo, as 2 i.m. injections, 3 months apart. The vaccine regimen was safe and well tolerated. After the DNA-MVA regimen, CD4(+) T-cell and CD8(+) T-cell responses occurred in 74% and 32% of the participants, respectively. The protein boost increased CD4(+) T-cell responses to 87% of the subjects. All participants developed tier 1 HIV-1C neutralizing antibody responses as well as durable Env binding antibodies that recognized linear V3 and C5 peptides. The HIV-1 subtype C DNA-MVA vaccine regimen showed promising cellular immunogenicity. Boosting with gp140/MF59 enhanced levels of binding and neutralizing antibodies as well as CD4(+) T-cell responses to HIV-1 envelope. (This study has been registered at ClinicalTrials.gov under registration no. NCT00574600 and NCT01423825.).

Copyright © 2016 Gray et al.

Figures

FIG 1
FIG 1
Injection site and systemic reactogenicity. The severity of reactogenicity symptoms is shown for the DNA/MVA prime-boost study (panels A and B) and for the study extension with the protein vaccine or placebo control (panels C and D). Panels A and C indicate local injection site symptoms; panels B and D indicate systemic symptoms. The percentage of participants who experienced no, mild, moderate, or severe symptoms following each administration of placebo (labeled C), DNA (labeled D), MVA (labeled M), or protein (labeled P) is shown by treatment group for each part of the study.
FIG 2
FIG 2
Intracellular cytokine staining assay for vaccine-induced T-cell responses after MVA or placebo vaccinations. The percentages of CD4+ T cells (upper panel) and CD8+ T cells (lower panel) expressing IL-2 or IFN-γ in response to global PTE peptide pools representing the HIV antigens specifically indicated on the x axis or showing a response to any one of these antigens (ANY) are shown. HIV-1-specific CD4+ and CD8+ T-cell responses were measured using a validated ICS assay. Results are from 2 weeks after the first MVA vaccination (M1) and 2 weeks after the second MVA vaccination (M2). Results from the placebo control group are combined from time points 2 weeks after the fourth and fifth placebo injections (far left). Responder data are shown as red dots, and nonresponder data are shown as blue triangles. Box plots show the distribution of the magnitude of response in positive responders only. The box indicates the median and interquartile range (IQR); whiskers extend to the furthest point within 1.5 times the IQR from the upper or lower quartile. Numbers at the top of each panel show the percentage and number of responders from each group among the evaluable participants.
FIG 3
FIG 3
Intracellular cytokine staining assay in study extension participants. The percentages of CD4+ T cells (upper panel) and CD8+ T cells (lower panel) expressing IL-2 or IFN-γ in response to any antigen represented in the global PTE peptide pools (PTEG) as measured using a validated ICS assay are indicated. Results from study extension participants who had received the DNA/MVA regimen are shown over time, from 2 weeks after the first MVA vaccination (M1), 2 weeks after the second MVA vaccination (M2), at extension baseline (B) prior to injection with gp140 or placebo, 2 weeks after each study extension injection, and 6 months after the final injection. Participants received either placebo (DDDMMCC group) or protein (DDDMMPP). Responder data are shown as red dots, and nonresponder data are shown as blue triangles. Box plots show the distribution of the magnitude of response in positive responders only. The box indicates the median and interquartile range (IQR); whiskers extend to the furthest point within 1.5 times the IQR from the upper or lower quartile. Numbers at the top of each panel show the percentage and number of responders from each group among the evaluable participants.
FIG 4
FIG 4
The frequency and magnitude of IgG antibody binding by HIV-1 binding antibody multiplex assay (BAMA). Responses to ConS gp140 CFI (upper panel) and gp140ΔV2.TV1 (lower panel) are shown. The MFI-minus-blank response data summarize the magnitudes at a given time point. Results from study extension participants who had received the DNA/MVA regimen are shown over time, from samples obtained 2 weeks after the first MVA vaccination (M1), 2 weeks after the second MVA vaccination (M2), at extension baseline (B) prior to injection with gp140 or placebo, 2 weeks after each study extension injection, and 6 months after the final injection. Participants received either placebo (DDDMMCC group) or protein (DDDMMPP). Numbers at the top of each panel show the percentage and number of responders from each group among the evaluable participants. Plots include data from responders in red and nonresponders in blue. Box plots show the distribution of the magnitude of response in positive responders only; the midline denotes the median, and the ends of the box denote the 25th and 75th percentiles. Whiskers extend to the most extreme data points within 1.5 times the interquartile range.
FIG 5
FIG 5
Linear epitope-specific response measured by peptide microarray. (A) Proportions of linear binding responses to each epitope region. The peptide region for each epitope is listed as the peptide numbers in the array library in parentheses next to the name of the epitope. Sequences of all peptides in the microarray library have been published previously (15). For each subject, the percentage values for each epitope are calculated as follows: maximum binding to the epitope/sum of maximum binding to all epitopes. Each pie slice represents the average value for all subjects (n = 12) mapped in the study. (B) Clade/strain preferences of the two dominant specificities (V3 and C5). Magnitudes of binding (maximum binding intensities) to V3 and C5 peptides of each clade/strain are plotted. Each bar represents the average value of the results for all 12 subjects mapped.
FIG 6
FIG 6
Neutralization 50% tissue culture infective dose (ID50) titers in the TZM-bl cell assay against the MW965.26 HIV-1C virus. The titer distributions for each treatment group are shown by time point, from samples obtained 2 weeks after the first MVA vaccination (M1), 2 weeks after the second MVA vaccination (M2), at extension baseline (B) prior to the first injection with gp140 or placebo, 2 weeks after each study extension injection, and 6 months after the final injection. Participants received either placebo (DDDMMCC group) or protein (DDDMMPP). Numbers at the top of each panel show the percentage and number of responders from each group among the evaluable participants. Plots include data from responders in red and nonresponders in blue. Box plots show the distribution of the magnitude of response in positive responders only; the midline denotes the median, and the ends of the box denote the 25th and 75th percentiles. Whiskers extend to the most extreme data points within 1.5 times the interquartile range.

References

    1. Williamson AL, Rybicki E, Shephard E, Gray G, Bekker LG, Downing K, Williamson C. 2012. South African HIV-1 vaccine candidates—the journey from the bench to clinical trials. South African Med J 102:452–455.
    1. Williamson C, Morris L, Maughan MF, Ping LH, Dryga SA, Thomas R, Reap EA, Cilliers T, van Harmelen J, Pascual A, Ramjee G, Gray G, Johnston R, Karim SA, Swanstrom R. 2003. Characterization and selection of HIV-1 subtype C isolates for use in vaccine development. AIDS Res Hum Retroviruses 19:133–144. doi:10.1089/088922203762688649.
    1. Burgers WA, van Harmelen JH, Shephard E, Adams C, Mgwebi T, Bourn W, Hanke T, Williamson AL, Williamson C. 2006. Design and preclinical evaluation of a multigene human immunodeficiency virus type 1 subtype C DNA vaccine for clinical trial. J Gen Virol 87:399–410. doi:10.1099/vir.0.81379-0.
    1. Burgers WA, Shephard E, Monroe JE, Greenhalgh T, Binder A, Hurter E, Van Harmelen JH, Williamson C, Williamson AL. 2008. Construction, characterization, and immunogenicity of a multigene modified vaccinia Ankara (MVA) vaccine based on HIV type 1 subtype C. AIDS Res Hum Retroviruses 24:195–206. doi:10.1089/aid.2007.0205.
    1. Burgers WA, Chege GK, Muller TL, van Harmelen JH, Khoury G, Shephard EG, Gray CM, Williamson C, Williamson AL. 2009. 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 90:468–480. doi:10.1099/vir.0.004614-0.
    1. Lian Y, Srivastava I, Gomez-Roman VR, Zur Megede J, Sun Y, Kan E, Hilt S, Engelbrecht S, Himathongkham S, Luciw PA, Otten G, Ulmer JB, Donnelly JJ, Rabussay D, Montefiori D, van Rensburg EJ, Barnett SW. 2005. Evaluation of envelope vaccines derived from the South African subtype C human immunodeficiency virus type 1 TV1 strain. J Virol 79:13338–13349. doi:10.1128/JVI.79.21.13338-13349.2005.
    1. De Rosa SC, Carter DK, McElrath MJ. 2012. OMIP-014: validated multifunctional characterization of antigen-specific human T cells by intracellular cytokine staining. Cytometry A 81:1019–1021.
    1. Horton H, Thomas EP, Stucky JA, Frank I, Moodie Z, Huang Y, Chiu YL, McElrath MJ, De Rosa SC. 2007. Optimization and validation of an 8-color intracellular cytokine staining (ICS) assay to quantify antigen-specific T cells induced by vaccination. J Immunol Methods 323:39–54. doi:10.1016/j.jim.2007.03.002.
    1. Bull M, Lee D, Stucky J, Chiu YL, Rubin A, Horton H, McElrath MJ. 2007. Defining blood processing parameters for optimal detection of cryopreserved antigen-specific responses for HIV vaccine trials. J Immunol Methods 322:57–69. doi:10.1016/j.jim.2007.02.003.
    1. Li F, Malhotra U, Gilbert PB, Hawkins NR, Duerr AC, McElrath JM, Corey L, Self SG. 2006. Peptide selection for human immunodeficiency virus type 1 CTL-based vaccine evaluation. Vaccine 24:6893–6904. doi:10.1016/j.vaccine.2006.06.009.
    1. Tomaras GD, Yates NL, Liu P, Qin L, Fouda GG, Chavez LL, Decamp AC, Parks RJ, Ashley VC, Lucas JT, Cohen M, Eron J, Hicks CB, Liao HX, Self SG, Landucci G, Forthal DN, Weinhold KJ, Keele BF, Hahn BH, Greenberg ML, Morris L, Karim SS, Blattner WA, Montefiori DC, Shaw GM, Perelson AS, Haynes BF. 2008. 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 82:12449–12463. doi:10.1128/JVI.01708-08.
    1. Yates NL, Liao HX, Fong Y, deCamp A, Vandergrift NA, Williams WT, Alam SM, Ferrari G, Yang ZY, Seaton KE, Berman PW, Alpert MD, Evans DT, O'Connell RJ, Francis D, Sinangil F, Lee C, Nitayaphan S, Rerks-Ngarm S, Kaewkungwal J, Pitisuttithum P, Tartaglia J, Pinter A, Zolla-Pazner S, Gilbert PB, Nabel GJ, Michael NL, Kim JH, Montefiori DC, Haynes BF, Tomaras GD. 2014. Vaccine-induced Env V1-V2 IgG3 correlates with lower HIV-1 infection risk and declines soon after vaccination. Sci Transl Med 6:228ra39. doi:10.1126/scitranslmed.3007730.
    1. Tomaras GD, Binley JM, Gray ES, Crooks ET, Osawa K, Moore PL, Tumba N, Tong T, Shen X, Yates NL, Decker J, Wibmer CK, Gao F, Alam SM, Easterbrook P, Abdool Karim S, Kamanga G, Crump JA, Cohen M, Shaw GM, Mascola JR, Haynes BF, Montefiori DC, Morris L. 2011. Polyclonal B cell responses to conserved neutralization epitopes in a subset of HIV-1-infected individuals. J Virol 85:11502–11519. doi:10.1128/JVI.05363-11.
    1. Shen X, Duffy R, Howington R, Cope A, Sadagopal S, Park H, Pal R, Kwa S, Ding S, Yang OO, Fouda GG, Le Grand R, Bolton D, Esteban M, Phogat S, Roederer M, Amara RR, Picker LJ, Seder RA, McElrath MJ, Barnett S, Permar SR, Shattock R, DeVico AL, Felber BK, Pavlakis GN, Pantaleo G, Korber BT, Montefiori DC, Tomaras GD. 27 May 2015. Vaccine-induced epitope-specific antibodies to simian immunodeficiency virus SIVmac239 envelope are distinct from those induced to the human immunodeficiency virus type 1 envelope in nonhuman primates. J Virol doi:10.1128/JVI.03635-14.
    1. Gottardo R, Bailer RT, Korber BT, Gnanakaran S, Phillips J, Shen X, Tomaras GD, Turk E, Imholte G, Eckler L, Wenschuh H, Zerweck J, Greene K, Gao H, Berman PW, Francis D, Sinangil F, Lee C, Nitayaphan S, Rerks-Ngarm S, Kaewkungwal J, Pitisuttithum P, Tartaglia J, Robb ML, Michael NL, Kim JH, Zolla-Pazner S, Haynes BF, Mascola JR, Self S, Gilbert P, Montefiori DC. 2013. 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 8:e75665. doi:10.1371/journal.pone.0075665.
    1. Sarzotti-Kelsoe M, Bailer RT, Turk E, Lin CL, Bilska M, Greene KM, Gao H, Todd CA, Ozaki DA, Seaman MS, Mascola JR, Montefiori DC. 2014. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. J Immunol Methods 409:131–146. doi:10.1016/j.jim.2013.11.022.
    1. Holm S. 1979. A simple sequentially rejective multiple test procedure. Scandinavian J Statistics 6:65–70.
    1. Agresti A, Coull BA. 1998. Approximate is better than “exact” for interval estimation of binomial proportions. Am Statistician 52:119–126.
    1. Elizaga ML, Vasan S, Marovich MA, Sato AH, Lawrence DN, Chaitman BR, Frey SE, Keefer MC; MVA Cardiac Safety Working Group. 2013. Prospective surveillance for cardiac adverse events in healthy adults receiving modified vaccinia Ankara vaccines: a systematic review. PLoS One 8:e54407. doi:10.1371/journal.pone.0054407.
    1. Goepfert PA, Elizaga ML, Seaton K, Tomaras GD, Montefiori DC, Sato A, Hural J, DeRosa SC, Kalams SA, McElrath MJ, Keefer MC, Baden LR, Lama JR, Sanchez J, Mulligan MJ, Buchbinder SP, Hammer SM, Koblin BA, Pensiero M, Butler C, Moss B, Robinson HL; HVTN 205 Study Group; National Institutes of Allergy and Infectious Diseases HIV Vaccines Trials Network . 2014. 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 210:99–110. doi:10.1093/infdis/jiu003.
    1. Spearman P, Lally MA, Elizaga M, Montefiori D, Tomaras GD, McElrath MJ, Hural J, De Rosa SC, Sato A, Huang Y, Frey SE, Sato P, Donnelly J, Barnett S, Corey LJ; HIV Vaccine Trials Network of NIAID. 2011. A trimeric, V2-deleted HIV-1 envelope glycoprotein vaccine elicits potent neutralizing antibodies but limited breadth of neutralization in human volunteers. J Infect Dis 203:1165–1173. doi:10.1093/infdis/jiq175.
    1. Palma P, Romiti ML, Bernardi S, Pontrelli G, Mora N, Santilli V, Tchidjou HK, Aquilani A, Cotugno N, Alghisi F, Lucidi V, Rossi P, Douagi I. 2012. Safety and immunogenicity of a monovalent MF59(R)-adjuvanted A/H1N1 vaccine in HIV-infected children and young adults. Biologicals 40:134–139. doi:10.1016/j.biologicals.2011.12.001.
    1. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, Liao HX, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. 2012. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 366:1275–1286. doi:10.1056/NEJMoa1113425.
    1. Jin X, Morgan C, Yu X, DeRosa S, Tomaras GD, Montefiori DC, Kublin J, Corey L, Keefer MC; NIAID HIV Vaccine Trials Network. 2015. Multiple factors affect immunogenicity of DNA plasmid HIV vaccines in human clinical trials. Vaccine 33:2347–2353. doi:10.1016/j.vaccine.2015.03.036.
    1. 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, Robb ML, Michael NL, Kunasol P, Kim JH; MOPH-TAVEG Investigators. 2009. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 361:2209–2220. doi:10.1056/NEJMoa0908492.
    1. Pereyra F, Addo MM, Kaufmann DE, Liu Y, Miura T, Rathod A, Baker B, Trocha A, Rosenberg R, Mackey E, Ueda P, Lu Z, Cohen D, Wrin T, Petropoulos CJ, Rosenberg ES, Walker BD. 2008. Genetic and immunologic heterogeneity among persons who control HIV infection in the absence of therapy. J Infect Dis 197:563–571. doi:10.1086/526786.

Source: PubMed

3
구독하다