Safety and immunogenicity of an HIV-1 prefusion-stabilized envelope trimer (Trimer 4571) vaccine in healthy adults: A first-in-human open-label, randomized, dose-escalation, phase 1 clinical trial

Katherine V Houser, Martin R Gaudinski, Myra Happe, Sandeep Narpala, Raffaello Verardi, Edward K Sarfo, Angela R Corrigan, Richard Wu, Ro Shauna Rothwell, Laura Novik, Cynthia S Hendel, Ingelise J Gordon, Nina M Berkowitz, Cora Trelles Cartagena, Alicia T Widge, Emily E Coates, Larisa Strom, Somia Hickman, Michelle Conan-Cibotti, Sandra Vazquez, Olga Trofymenko, Sarah Plummer, Judy Stein, Christopher L Case, Martha Nason, Andrea Biju, Danealle K Parchment, Anita Changela, Cheng Cheng, Hongying Duan, Hui Geng, I-Ting Teng, Tongqing Zhou, Sarah O'Connell, Chris Barry, Kevin Carlton, Jason G Gall, Britta Flach, Nicole A Doria-Rose, Barney S Graham, Richard A Koup, Adrian B McDermott, John R Mascola, Peter D Kwong, Julie E Ledgerwood, VRC 018 Study Team, Katherine V Houser, Martin R Gaudinski, Myra Happe, Sandeep Narpala, Raffaello Verardi, Edward K Sarfo, Angela R Corrigan, Richard Wu, Ro Shauna Rothwell, Laura Novik, Cynthia S Hendel, Ingelise J Gordon, Nina M Berkowitz, Cora Trelles Cartagena, Alicia T Widge, Emily E Coates, Larisa Strom, Somia Hickman, Michelle Conan-Cibotti, Sandra Vazquez, Olga Trofymenko, Sarah Plummer, Judy Stein, Christopher L Case, Martha Nason, Andrea Biju, Danealle K Parchment, Anita Changela, Cheng Cheng, Hongying Duan, Hui Geng, I-Ting Teng, Tongqing Zhou, Sarah O'Connell, Chris Barry, Kevin Carlton, Jason G Gall, Britta Flach, Nicole A Doria-Rose, Barney S Graham, Richard A Koup, Adrian B McDermott, John R Mascola, Peter D Kwong, Julie E Ledgerwood, VRC 018 Study Team

Abstract

Background: Advances in therapeutic drugs have increased life-expectancies for HIV-infected individuals, but the need for an effective vaccine remains. We assessed safety and immunogenicity of HIV-1 vaccine, Trimer 4571 (BG505 DS-SOSIP.664) adjuvanted with aluminum hydroxide (alum), in HIV-negative adults.

Methods: We conducted a phase I, randomized, open-label, dose-escalation trial at the National Institutes of Health Clinical Center in Bethesda, MD, USA. Eligible participants were HIV-negative, healthy adults between 18-50 years. Participants were randomized 1:1 to receive Trimer 4571 adjuvanted with 500 mcg alum by either the subcutaneous (SC) or intramuscular (IM) route at weeks 0, 8, and 20 in escalating doses of 100 mcg or 500 mcg. The primary objectives were to evaluate the safety and tolerability of Trimer 4571 with a secondary objective of evaluating vaccine-induced antibody responses. The primary and safety endpoints were evaluated in all participants who received at least one dose of Trimer 4571. Trial results were summarized using descriptive statistics. This trial is registered at ClinicalTrials.gov, NCT03783130.

Findings: Between March 7 and September 11, 2019, 16 HIV-negative participants were enrolled, including six (38%) males and ten (62%) females. All participants received three doses of Trimer 4571. Solicited reactogenicity was mild to moderate in severity, with one isolated instance of severe injection site redness (6%) following a third 500 mcg SC administration. The most commonly reported solicited symptoms included mild injection site tenderness in 14 (88%) and mild myalgia in six (38%) participants. The most frequent unsolicited adverse event attributed to vaccination was mild injection site pruritus in six (38%) participants. Vaccine-induced seropositivity occurred in seven (44%) participants and resolved in all but one (6%). No serious adverse events occurred. Trimer 4571-specific binding antibodies were detected in all groups two weeks after regimen completion, primarily focused on the glycan-free trimer base. Neutralizing antibody activity was limited to the 500 mcg dose groups.

Interpretation: Trimer 4571 was safe, well tolerated, and immunogenic in this first-in-human trial. While this phase 1 trial is limited in size, our results inform and support further evaluation of prefusion-stabilized HIV-1 envelope trimers as a component of vaccine design strategies to generate broadly neutralizing antibodies against HIV-1.

Funding: Intramural Research Program of the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health.

Keywords: BG505 DS-SOSIP.664; HIV-1; NIH; Phase 1 clinical trial; Trimer 4571; Vaccine.

Conflict of interest statement

CC, HG, JRM, and PDK are listed on patent applications involving Trimer 4571. All other authors declare no competing interests.

© 2022 The Authors.

Figures

Figure 1
Figure 1
Trial CONSORT diagram. Participants were enrolled according to dose-escalation protocol and randomized to receive three doses of adjuvanted Trimer 4571 at weeks 0, 8, and 20. All participants completed the protocol. IM denotes intramuscular, and SC subcutaneous route of vaccine administration. Participants who withdrew during enrollment did so because: time commitment for study procedures (10 participants, 19%), concern of potential risks of study procedures (28 participants, 52%), number of procedures/blood draws (2 participants, 4%), lost contact (5 participants, 9%), study closed to enrollment (7 participants, 13%), participants chose another study (1 participants, 2%) and other (29, 54%). Of the twenty-eight (28) participants that withdrew due to concern of potential risks, 23 (82%) had concerns with vaccine induced seropositivity (VISP). Participants could choose more than one withdrawal reason/category. * 500 mcg of alum was field mixed with Trimer 4571 for all study groups.
Figure 2
Figure 2
Maximum local and systemic reactogenicity reported within 7 days after receipt of each Trimer 4571 vaccination according to dose and route of administration. Percent of participants who reported a local or systemic symptom (y-axis) in the seven days following each vaccination. The severity of solicited adverse events was graded mild, moderate, and severe. Data are n (%). IM denotes intramuscular, and SC subcutaneous.
Figure 3
Figure 3
Vaccination with Trimer 4571 induces trimer-specific antibody responses by ELISA. Trimer 4571-specific antibody titers in serum were measured by ELISA at baseline and two weeks after each vaccination. HIV+ samples were used as positive controls. ELISA endpoint titer was defined as the reciprocal of the greatest dilution with an optical density value above 0·1. Geometric mean titers (GMTs) with 95% confidence intervals (CIs) are indicated by error bars. Horizontal dotted line indicates the assay negative cut off. IM denotes intramuscular, and SC subcutaneous. Significant differences from baseline to two weeks after regimen completion were observed for 100 mcg SC (p=0·029), 500 mcg IM (p=0·0052), and 500 mcg SC (p=0·0002) groups.
Figure 4
Figure 4
Vaccination with Trimer 4571 induces antibodies against trimer immunogen but not against other Env regions. Anti-Trimer 4571, V3 peptide, fusion peptide (FP), gp120, and gp41 responses at baseline and two weeks after regimen completion were assessed serologically by ELISA. HIV+ human sera were used as assay controls. Horizontal dotted lines indicate the assay negative cut offs. ELISA endpoint titer was defined as the reciprocal of the greatest dilution with an optical density value above 0·1. Significant increase in Trimer 4571 ELISA Endpoint titer was observed from baseline to two weeks after regimen completion across all study groups combined (p

Figure 5

Vaccination with Trimer 4571 elicits…

Figure 5

Vaccination with Trimer 4571 elicits antibodies directed against glycan-free trimer base. A) ELISA…

Figure 5
Vaccination with Trimer 4571 elicits antibodies directed against glycan-free trimer base. A) ELISA base-binding endpoint antibody titers in serum were measured with or without trimer base-directed antibodies (RM19R) at two weeks after regimen completion. ELISA endpoint titer was defined as the reciprocal of the greatest dilution with an optical density value above 0·1. B) Ratios of responses directed to base epitopes were calculated as 1 - (endpoint titer with blocking/endpoint titer without blocking), so that the value is 1·00 when all responses are specific to the glycan-free base. IM denotes intramuscular, and SC subcutaneous

Figure 6

Electron Microscopy Polyclonal Epitope Mapping…

Figure 6

Electron Microscopy Polyclonal Epitope Mapping (EMPEM) of the antibody responses to Trimer 4571…

Figure 6
Electron Microscopy Polyclonal Epitope Mapping (EMPEM) of the antibody responses to Trimer 4571 distinguished three different antibody classes. A) Representative micrograph of negatively stained Trimer 4571 and Fab complexes (bar size: 100nm). B) Representative reference-free 2D classes of Fab bound to Trimer 4571 (bar size: 10 nm) C) 3D reconstruction of antibody classes detected at two weeks after regimen completion.
Figure 5
Figure 5
Vaccination with Trimer 4571 elicits antibodies directed against glycan-free trimer base. A) ELISA base-binding endpoint antibody titers in serum were measured with or without trimer base-directed antibodies (RM19R) at two weeks after regimen completion. ELISA endpoint titer was defined as the reciprocal of the greatest dilution with an optical density value above 0·1. B) Ratios of responses directed to base epitopes were calculated as 1 - (endpoint titer with blocking/endpoint titer without blocking), so that the value is 1·00 when all responses are specific to the glycan-free base. IM denotes intramuscular, and SC subcutaneous
Figure 6
Figure 6
Electron Microscopy Polyclonal Epitope Mapping (EMPEM) of the antibody responses to Trimer 4571 distinguished three different antibody classes. A) Representative micrograph of negatively stained Trimer 4571 and Fab complexes (bar size: 100nm). B) Representative reference-free 2D classes of Fab bound to Trimer 4571 (bar size: 10 nm) C) 3D reconstruction of antibody classes detected at two weeks after regimen completion.

References

    1. Fauci AS, Lane HC. Four decades of HIV/AIDS - much accomplished, much to do. N Engl J Med. 2020;383(1):1–4.
    1. Barouch DH, Yang ZY, Kong WP, et al. A human T-cell leukemia virus type 1 regulatory element enhances the immunogenicity of human immunodeficiency virus type 1 DNA vaccines in mice and nonhuman primates. J Virol. 2005;79(14):8828–8834.
    1. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 2009;361(23):2209–2220.
    1. Kwong PD, Mascola JR. HIV-1 vaccines based on antibody identification, B cell ontogeny, and epitope structure. Immunity. 2018;48(5):855–871.
    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):654–665.
    1. Pitisuttithum P, Gilbert P, Gurwith M, 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):1661–1671.
    1. Graham BS, Keefer MC, McElrath MJ, et al. Safety and immunogenicity of a candidate HIV-1 vaccine in healthy adults: recombinant glycoprotein (rgp) 120. A randomized, double-blind trial. NIAID AIDS vaccine evaluation group. Ann Intern Med. 1996;125(4):270–279.
    1. Gilbert P, Wang M, Wrin T, et al. Magnitude and breadth of a nonprotective neutralizing antibody response in an efficacy trial of a candidate HIV-1 gp120 vaccine. J Infect Dis. 2010;202(4):595–605.
    1. Overbaugh J, Morris L. The antibody response against HIV-1. Cold Spring Harb Perspect Med. 2012;2(1)
    1. Pancera M, Zhou T, Druz A, et al. Structure and immune recognition of trimeric pre-fusion HIV-1 Env. Nature. 2014;514(7523):455–461.
    1. Munro JB, Gorman J, Ma X, et al. Conformational dynamics of single HIV-1 envelope trimers on the surface of native virions. Science. 2014;346(6210):759–763.
    1. Kwon YD, Pancera M, Acharya P, et al. Crystal structure, conformational fixation and entry-related interactions of mature ligand-free HIV-1 Env. Nat Struct Mol Biol. 2015;22(7):522–531.
    1. Guttman M, Cupo A, Julien JP, et al. Antibody potency relates to the ability to recognize the closed, pre-fusion form of HIV Env. Nat Commun. 2015;6:6144.
    1. Sanders RW, Derking R, Cupo A, 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(9)
    1. Kwong PD, DeKosky BJ, Ulmer JB. Antibody-guided structure-based vaccines. Semin Immunol. 2020;50
    1. Crank MC, Ruckwardt TJ, Chen M, et al. A proof of concept for structure-based vaccine design targeting RSV in humans. Science. 2019;365(6452):505–509.
    1. Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2 - preliminary report. N Engl J Med. 2020;383(20):1920–1931.
    1. Binley JM, Sanders RW, Clas B, et al. A recombinant human immunodeficiency virus type 1 envelope glycoprotein complex stabilized by an intermolecular disulfide bond between the gp120 and gp41 subunits is an antigenic mimic of the trimeric virion-associated structure. J Virol. 2000;74(2):627–643.
    1. Zhou T, Doria-Rose NA, Cheng C, et al. Quantification of the impact of the HIV-1-glycan shield on antibody elicitation. Cell Rep. 2017;19(4):719–732.
    1. Chuang GY, Lai YT, Boyington JC, et al. Development of a 3Mut-apex-stabilized envelope trimer that expands HIV-1 neutralization breadth when used to boost fusion peptide-directed vaccine-elicited responses. J Virol. 2020;94(13)
    1. Chuang GY, Geng H, Pancera M, et al. Structure-based design of a soluble prefusion-closed HIV-1 Env trimer with reduced CD4 affinity and improved immunogenicity. J Virol. 2017;91(10)
    1. Xu K, Acharya P, Kong R, et al. Epitope-based vaccine design yields fusion peptide-directed antibodies that neutralize diverse strains of HIV-1. Nat Med. 2018;24(6):857–867.
    1. Gulla K, Cibelli N, Cooper JW, et al. A non-affinity purification process for GMP production of prefusion-closed HIV-1 envelope trimers from clades A and C for clinical evaluation. Vaccine. 2021;39(25):3379–3387.
    1. Cooper CJ, Metch B, Dragavon J, Coombs RW, Baden LR, Force NHVTNV-IST . Vaccine-induced HIV seropositivity/reactivity in noninfected HIV vaccine recipients. JAMA. 2010;304(3):275–283.
    1. Ivleva VB, Cooper JW, Arnold FJ, Lei QP. Overcoming challenges in structural characterization of HIV-1 envelope glycoprotein by LC-MS/MS. J Am Soc Mass Spectrom. 2019;30(9):1663–1678.
    1. Decker JM, Bibollet-Ruche F, Wei X, et al. Antigenic conservation and immunogenicity of the HIV coreceptor binding site. J Exp Med. 2005;201(9):1407–1419.
    1. Nogal B, Bianchi M, Cottrell CA, et al. Mapping polyclonal antibody responses in non-human primates vaccinated with HIV Env trimer subunit vaccines. Cell Rep. 2020;30(11) 3755-65 e7.
    1. Corrigan AR, Duan H, Cheng C, et al. Fusion peptide priming reduces immune responses to HIV-1 envelope trimer base. Cell Rep. 2021;35(1)
    1. Gilbert PB, Peterson ML, Follmann D, 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(5):666–677.
    1. Ou L, Kong WP, Chuang GY, et al. Preclinical development of a fusion peptide conjugate as an HIV vaccine immunogen. Sci Rep. 2020;10(1):3032.
    1. Cheng C, Duan H, Xu K, et al. Immune monitoring reveals fusion peptide priming to imprint cross-clade HIV-neutralizing responses with a characteristic early B cell signature. Cell Rep. 2020;32(5)
    1. Kong R, Xu K, Zhou T, et al. Fusion peptide of HIV-1 as a site of vulnerability to neutralizing antibody. Science. 2016;352(6287):828–833.
    1. Struwe WB, Chertova E, Allen JD, et al. Site-specific glycosylation of virion-derived HIV-1 Env is mimicked by a soluble trimeric immunogen. Cell Rep. 2018;24(8) 1958-66.e5.
    1. Baylor NW, Egan W, Richman P. Aluminum salts in vaccines–US perspective. Vaccine. 2002;20(Suppl 3):S18–S23.

Source: PubMed

3
Subscribe