Safety and immunogenicity of two heterologous HIV vaccine regimens in healthy, HIV-uninfected adults (TRAVERSE): a randomised, parallel-group, placebo-controlled, double-blind, phase 1/2a study

Lindsey R Baden, Daniel J Stieh, Michal Sarnecki, Stephen R Walsh, Georgia D Tomaras, James G Kublin, M Juliana McElrath, Galit Alter, Guido Ferrari, David Montefiori, Philipp Mann, Steven Nijs, Katleen Callewaert, Paul Goepfert, Srilatha Edupuganti, Etienne Karita, Johannes P Langedijk, Frank Wegmann, Lawrence Corey, Maria G Pau, Dan H Barouch, Hanneke Schuitemaker, Frank Tomaka, Traverse/HVTN 117/HPX2004 Study Team, Julie A Ake, Susan Buchbinder, Karen Buleza, Kristen W Cohen, Trevor A Crowell, Zelda Euler, Ian Frank, Dimitri Goedhart, Michael Keefer, Colleen Kelly, Ken Mayer, Joseph Nkolola, Lauren Peter, Merlin L Robb, Nadine Rouphael, Lorenz Scheppler, Magda Sobieszczyk, Hong Van Tieu, Lindsey R Baden, Daniel J Stieh, Michal Sarnecki, Stephen R Walsh, Georgia D Tomaras, James G Kublin, M Juliana McElrath, Galit Alter, Guido Ferrari, David Montefiori, Philipp Mann, Steven Nijs, Katleen Callewaert, Paul Goepfert, Srilatha Edupuganti, Etienne Karita, Johannes P Langedijk, Frank Wegmann, Lawrence Corey, Maria G Pau, Dan H Barouch, Hanneke Schuitemaker, Frank Tomaka, Traverse/HVTN 117/HPX2004 Study Team, Julie A Ake, Susan Buchbinder, Karen Buleza, Kristen W Cohen, Trevor A Crowell, Zelda Euler, Ian Frank, Dimitri Goedhart, Michael Keefer, Colleen Kelly, Ken Mayer, Joseph Nkolola, Lauren Peter, Merlin L Robb, Nadine Rouphael, Lorenz Scheppler, Magda Sobieszczyk, Hong Van Tieu

Abstract

Background: Bioinformatically designed mosaic antigens increase the breadth of HIV vaccine-elicited immunity. This study compared the safety, tolerability, and immunogenicity of a newly developed, tetravalent Ad26 vaccine with the previously tested trivalent formulation.

Methods: This randomised, parallel-group, placebo-controlled, double-blind, phase 1/2a study (TRAVERSE) was done at 11 centres in the USA and one centre in Rwanda. Eligible participants were adults aged 18 to 50 years, who were HIV-uninfected, healthy at screening based on their medical history and a physical examination including laboratory assessment and vital sign measurements, and at low risk of HIV infection in the opinion of study staff, who applied a uniform definition of low-risk guidelines that was aligned across sites. Enrolled participants were randomly assigned at a 2:1 ratio to tetravalent and trivalent groups. Participants in tetravalent and trivalent groups were then further randomly assigned at a 5:1 ratio to adenovirus 26 (Ad26)-vectored vaccine and placebo subgroups. Randomisation was stratified by region (USA and Rwanda) and based on a computer-generated schedule using randomly permuted blocks prepared under the sponsor's supervision. We masked participants and investigators to treatment allocation throughout the study. On day 0, participants received a first injection of tetravalent vaccine (Ad26.Mos4.HIV or placebo) or trivalent vaccine (Ad26.Mos.HIV or placebo), and those injections were repeated 12 weeks later. At week 24, vaccine groups received a third dose of tetravalent or trivalent together with clade C gp140, and this was repeated at week 48, with placebos again administered to the placebo group. All study vaccines and placebo were administered by intramuscular injection in the deltoid muscle. We assessed adverse events in all participants who received at least one study injection (full analysis set) and Env-specific binding antibodies in all participants who received at least the first three vaccinations according to the protocol-specified vaccination schedule, had at least one measured post-dose blood sample collected, and were not diagnosed with HIV during the study (per-protocol set). This study is registered with Clinicaltrials.gov, NCT02788045.

Findings: Of 201 participants who were enrolled and randomly assigned, 198 received the first vaccination: 110 were in the tetravalent group, 55 in the trivalent group, and 33 in the placebo group. Overall, 185 (93%) completed two scheduled vaccinations per protocol, 180 (91%) completed three, and 164 (83%) completed four. Solicited, self-limiting local, systemic reactogenicity and unsolicited adverse events were similar in vaccine groups and higher than in placebo groups. All participants in the per-protocol set developed clade C Env binding antibodies after the second vaccination, with higher total IgG titres after the tetravalent vaccine than after the trivalent vaccine (10 413 EU/mL, 95% CI 7284-14 886 in the tetravalent group compared with 5494 EU/mL, 3759-8029 in the trivalent group). Titres further increased after the third and fourth vaccinations, persisting at least through week 72. Other immune responses were also higher with the tetravalent vaccine, including the magnitude and breadth of binding antibodies against a cross-clade panel of Env antigens, and the magnitude of IFNγ ELISPOT responses (median 521 SFU/106 peripheral blood mononuclear cells [PBMCs] in the tetravalent group and median 282 SFU/106 PBMCs in the trivalent group after the fourth vaccination) and Env-specific CD4+ T-cell response rates after the third and fourth vaccinations. No interference by pre-existing Ad26 immunity was identified.

Interpretation: The tetravalent vaccine regimen was generally safe, well-tolerated, and found to elicit higher immune responses than the trivalent regimen. Regimens that use this tetravalent vaccine component are being advanced into field trials to assess efficacy against HIV-1 infection.

Funding: National Institutes of Health, Henry M Jackson Foundation for Advancement of Military Medicine and the US Department of Defense, Ragon Institute of MGH, MIT, & Harvard, Bill & Melinda Gates Foundation, and Janssen Vaccines & Prevention.

Copyright © 2020 The Author(s). Published by Elsevier Ltd. This is a Gold Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Study flowchart
Figure 2
Figure 2
Solicited local and systemic adverse events Solicited local (A) and systemic (B) adverse events, as proportions of participants in the three study groups after each vaccination dose in the full analysis set. The full analysis set included all participants who received at least one study injection. Numbers in the columns are the total numbers of participants per group at each timepoint.
Figure 3
Figure 3
Humoral binding antibody immune responses Horizontal bars indicate matched timepoints where the response magnitude is significantly different at p

References

    1. UNAIDS Global HIV & AIDS statistics — 2020 fact sheet.
    1. Fauci AS. An HIV vaccine is essential for ending the HIV/AIDS pandemic. JAMA. 2017;318:1535–1536.
    1. Hammer SM, Sobieszczyk ME, Janes H. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N Engl J Med. 2013;369:2083–2092.
    1. Gray GE, Allen M, Moodie Z. 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.
    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;191:654–665.
    1. Buchbinder SP, Mehrotra DV, Duerr A. 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.
    1. Pitisuttithum P, Gilbert P, Gurwith M. 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.
    1. Haynes BF, Gilbert PB, McElrath MJ. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med. 2012;366:1275–1286.
    1. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 2009;361:2209–2220.
    1. National Institute of Allergy and Infectious Diseases Experimental HIV vaccine regimen ineffective in preventing HIV. Feb 3, 2020.
    1. Taylor BS, Sobieszczyk ME, McCutchan FE, Hammer SM. The challenge of HIV-1 subtype diversity. N Engl J Med. 2008;358:1590–1602.
    1. Fischer W, Perkins S, Theiler J. Polyvalent vaccines for optimal coverage of potential T-cell epitopes in global HIV-1 variants. Nat Med. 2007;13:100–106.
    1. Barouch DH, O'Brien KL, Simmons NL. Mosaic HIV-1 vaccines expand the breadth and depth of cellular immune responses in rhesus monkeys. Nat Med. 2010;16:319–323.
    1. Santra S, Liao HX, Zhang R. Mosaic vaccines elicit CD8+ T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys. Nat Med. 2010;16:324–328.
    1. Barouch DH, Stephenson KE, Borducchi EN. Protective efficacy of a global HIV-1 mosaic vaccine against heterologous SHIV challenges in rhesus monkeys. Cell. 2013;155:531–539.
    1. Baden LR, Walsh SR, Seaman MS. First-in-human randomised, controlled trial of mosaic HIV-1 immunogens delivered via a modified vaccinia Ankara vector. J Infect Dis. 2018;218:633–644.
    1. Barouch DH, Tomaka FL, Wegmann F. Evaluation of a mosaic HIV-1 vaccine in a multicentre, randomised, double-blind, placebo-controlled, phase 1/2a clinical trial (APPROACH) and in rhesus monkeys (NHP 13-19) Lancet. 2018;392:232–243.
    1. Stephenson KE, Wegmann F, Tomaka F. Comparison of shortened mosaic HIV-1 vaccine schedules: a randomised, double-blind, placebo-controlled phase 1 trial (IPCAVD010/HPX1002) and a preclinical study in rhesus monkeys (NHP 17–22) Lancet HIV. 2020;7:e410–e421.
    1. Barouch DH, Liu J, Peter L. 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.
    1. Baden LR, Karita E, Mutua G. Assessment of the safety and immunogenicity of 2 novel vaccine platforms for HIV-1 prevention: a randomized trial. Ann Intern Med. 2016;164:313–322.
    1. Baden LR, Walsh SR, Seaman MS. First-in-human evaluation of the safety and immunogenicity of a recombinant adenovirus serotype 26 HIV-1 Env vaccine (IPCAVD 001) J Infect Dis. 2013;207:240–247.
    1. Baden LR, Liu J, Li H. Induction of HIV-1-specific mucosal immune responses following intramuscular recombinant adenovirus serotype 26 HIV-1 vaccination of humans. J Infect Dis. 2015;211:518–528.
    1. Excler JL, Kim JH. Novel prime-boost vaccine strategies against HIV-1. Expert Rev Vaccines. 2019;18:765–779.
    1. National Institute of Allergy and Infectious Diseases Division of AIDS (DAIDS) table for grading the severity of adult and pediatric adverse events: version 2.0. November, 2014.
    1. Yates NL, deCamp AC, Korber BT. HIV-1 envelope glycoproteins from diverse clades differentiate antibody responses and durability among vaccines. J Virol. 2018;92:e01843–e01917.

Source: PubMed

3
Subscribe