Your browser doesn't support javascript.
loading
Immune correlates analysis of the Imbokodo (HVTN 705/HPX2008) efficacy trial of a mosaic HIV-1 vaccine regimen evaluated in Southern African people assigned female sex at birth: a two-phase case-control study.
Kenny, Avi; van Duijn, Janine; Dintwe, One; Heptinstall, Jack; Burnham, Randy; Sawant, Sheetal; Zhang, Lu; Mielke, Dieter; Khuzwayo, Sharon; Omar, Faatima Laher; Stanfield-Oakley, Sherry; Keyes, Taylor; Dunn, Brooke; Goodman, Derrick; Fong, Youyi; Benkeser, David; Zou, Rodger; Hural, John; Hyrien, Ollivier; Juraska, Michal; Luedtke, Alex; van der Laan, Lars; Giorgi, Elena E; Magaret, Craig; Carpp, Lindsay N; Pattacini, Laura; van de Kerkhof, Tom; Korber, Bette; Willems, Wouter; Fisher, Leigh H; Schuitemaker, Hanneke; Swann, Edith; Kublin, James G; Pau, Maria G; Buchbinder, Susan; Tomaka, Frank; Nijs, Steven; Lavreys, Ludo; Gelderblom, Huub C; Corey, Lawrence; Mngadi, Kathryn; Gray, Glenda E; Borducchi, Erica; Hendriks, Jenny; Seaton, Kelly E; Zolla-Pazner, Susan; Barouch, Dan H; Ferrari, Guido; De Rosa, Stephen C; McElrath, M Juliana.
Afiliación
  • Kenny A; Department of Biostatistics, University of Washington, Seattle, WA, USA.
  • van Duijn J; Janssen Vaccines & Prevention BV, Leiden, the Netherlands.
  • Dintwe O; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa.
  • Heptinstall J; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA.
  • Burnham R; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Sawant S; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA.
  • Zhang L; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA.
  • Mielke D; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA.
  • Khuzwayo S; Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa.
  • Omar FL; Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa.
  • Stanfield-Oakley S; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA.
  • Keyes T; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA.
  • Dunn B; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA.
  • Goodman D; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA.
  • Fong Y; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Benkeser D; Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
  • Zou R; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Hural J; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Hyrien O; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Juraska M; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Luedtke A; Department of Statistics, University of Washington, Seattle, WA, USA.
  • van der Laan L; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Statistics, University of Washington, Seattle, WA, USA.
  • Giorgi EE; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Magaret C; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Carpp LN; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Pattacini L; Janssen Vaccines & Prevention BV, Leiden, the Netherlands.
  • van de Kerkhof T; Janssen Vaccines & Prevention BV, Leiden, the Netherlands.
  • Korber B; Los Alamos National Laboratory, Los Alamos, NM, USA; New Mexico Consortium, Los Alamos, NM, USA.
  • Willems W; Janssen Research & Development BE, Beerse, Belgium.
  • Fisher LH; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Schuitemaker H; Janssen Vaccines & Prevention BV, Leiden, the Netherlands.
  • Swann E; Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
  • Kublin JG; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Pau MG; Janssen Infectious Diseases BV, Beerse, Belgium.
  • Buchbinder S; San Francisco Department of Public Health, San Francisco, CA, USA.
  • Tomaka F; Janssen Research & Development, LLC, Titusville, NJ, USA.
  • Nijs S; Janssen Infectious Diseases BV, Beerse, Belgium.
  • Lavreys L; Janssen Infectious Diseases BV, Beerse, Belgium.
  • Gelderblom HC; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • Corey L; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, 98195, USA.
  • Mngadi K; The Aurum Institute, Johannesburg, South Africa.
  • Gray GE; South African Medical Research Council, Cape Town, South Africa.
  • Borducchi E; Center for Virology & Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.
  • Hendriks J; Janssen Vaccines & Prevention BV, Leiden, the Netherlands.
  • Seaton KE; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA.
  • Zolla-Pazner S; Icahn School of Medicine at Mount Sinai, New York, NY, USA.
  • Barouch DH; Center for Virology & Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA, USA.
  • Ferrari G; Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA.
  • De Rosa SC; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
  • McElrath MJ; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
EBioMedicine ; 108: 105320, 2024 Sep 04.
Article en En | MEDLINE | ID: mdl-39236556
ABSTRACT

BACKGROUND:

The HVTN 705 Imbokodo trial of 2636 people without HIV and assigned female sex at birth, conducted in southern Africa, evaluated a heterologous HIV-1 vaccine regimen mosaic adenovirus 26-based vaccine (Ad26.Mos4.HIV) at Months 0, 3, 6, 12 and alum-adjuvanted clade C gp140 at Months 6, 12. Per-protocol vaccine efficacy (VE) against HIV-1 diagnosis from seven to 24 months was 14.1% (95% CI -22.0% to 39.5%). Immune correlates analysis was performed for markers selected based on prior evidence in efficacy trials and/or nonhuman primate models.

METHODS:

Humoral and cellular immune response markers at Month 7 were evaluated as immune correlates of risk and of protection in a breakthrough case-control cohort (n = 52 cases, 246 non-cases). Primary markers were IgG binding to vaccine-strain gp140, IgG3 binding to diverse Env antigens (IgG3 Env breadth), IgG3 binding to diverse V1V2 antigens (IgG3 V1V2 breadth), antibody-dependent phagocytosis against the vaccine-strain gp140, Env-specific CD4+ and CD8+ T-cell responses, and multi-epitope functions.

FINDINGS:

No immune markers were statistically significant correlates of risk. IgG3 V1V2 breadth trended toward an inverse association hazard ratio 0.70 (95% CI 0.36 to 1.35; p = 0.29) per 10-fold increase and 0.51 (95% CI 0.21 to 1.24; p = 0.14) in a Cox model with all primary markers. The VE estimate was 11.8% (95% CI -17.9% to 34.0%) at all IgG3 V1V2 breadth values below 667 weighted geometric mean net MFI; just above this value, the VE estimate sharply increased to 62.6% (95% CI -17.9% to 89.6%), and further increased to 80.9% (95% CI -17.9% to 99.5%) at 1471 MFI, the 95th percentile of the marker distribution. Mediation analysis yielded a VE of 35.7% (95% CI 15.0% to 51.3%) attributable to the vaccine's impact on this marker.

INTERPRETATION:

The trend in association of greater IgG3 V1V2 antibody breadth with lower likelihood of HIV acquisition is consistent with the identification of antibodies against V1V2 as immune correlates in three other HIV vaccine efficacy trials and suggests that a greater emphasis should be placed on studying this region in the HIV-1 envelope as a vaccine immunogen.

FUNDING:

National Institute of Allergy and Infectious Diseases and Janssen Vaccines & Prevention BV.
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: EBioMedicine Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: EBioMedicine Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos