1
|
Gray GE, Mngadi K, Lavreys L, Nijs S, Gilbert PB, Hural J, Hyrien O, Juraska M, Luedtke A, Mann P, McElrath MJ, Odhiambo JA, Stieh DJ, van Duijn J, Takalani AN, Willems W, Tapley A, Tomaras GD, Van Hoof J, Schuitemaker H, Swann E, Barouch DH, Kublin JG, Corey L, Pau MG, Buchbinder S, Tomaka F. Mosaic HIV-1 vaccine regimen in southern African women (Imbokodo/HVTN 705/HPX2008): a randomised, double-blind, placebo-controlled, phase 2b trial. THE LANCET. INFECTIOUS DISEASES 2024; 24:1201-1212. [PMID: 39038477 PMCID: PMC11512672 DOI: 10.1016/s1473-3099(24)00358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND HIV type 1 (HIV-1) remains a global health concern, with the greatest burden in sub-Saharan Africa. Despite 40 years of research, no vaccine candidate has shown durable and protective efficacy against HIV-1 acquisition. Although pre-exposure prophylaxis in groups with high vulnerability can be very effective, barriers to its use, such as perceived low acquisition risk, fear of stigma, and concerns about side-effects, remain. Thus, a population-based approach, such as an HIV-1 vaccine, is needed. The current study aimed to evaluate the efficacy and safety of a heterologous HIV-1 vaccine regimen, consisting of a tetravalent mosaic adenovirus 26-based vaccine (Ad26.Mos4.HIV) and aluminium phosphate-adjuvanted clade C glycoprotein (gp) 140, in young women at risk of acquiring HIV-1 in southern Africa. METHODS This randomised, double-blind, phase 2b study enrolled sexually active women without HIV-1 or HIV-2 aged 18-35 years at 23 clinical research sites in Malawi, Mozambique, South Africa, Zambia, and Zimbabwe. Participants were centrally randomly assigned (1:1) to receive intramuscular injections of vaccine or saline placebo in stratified permuted blocks via an interactive web response system. Study participants, study site personnel (except those with primary responsibility for study vaccine preparation and dispensing), and investigators were masked to treatment group allocation. The vaccine regimen consisted of Ad26.Mos4.HIV administered at months 0 and 3 followed by Ad26.Mos4.HIV administered concurrently with aluminium phosphate-adjuvanted clade C gp140 at months 6 and 12. The primary efficacy outcome was vaccine efficacy in preventing laboratory-confirmed HIV-1 acquisition diagnosed between visits at month 7 and month 24 after the first vaccination (VE[7-24]) in the per-protocol population, which included participants who had not acquired HIV-1 4 weeks after the third vaccination, received all planned vaccinations at the first three vaccination visits within the protocol-specified windows, and had no major protocol deviations that could affect vaccine efficacy. Primary safety outcomes were assessed in randomly assigned participants who received one study injection or more based on the actual injection received. The primary safety endpoints were the incidences of unsolicited adverse events (AEs), solicited local and systemic AEs, serious AEs, AEs of special interest, and AEs leading to discontinuation of vaccination. This trial is registered with ClinicalTrials.gov, NCT03060629, and is complete. FINDINGS Between Nov 3, 2017, and June 30, 2019, 2654 women were randomly assigned, of whom 2636 women (median age of 23 years [IQR 20-25]) were enrolled and received at least one study injection (1313 assigned vaccine, 1323 placebo; 1317 received vaccine, 1319 placebo). Analysis of the primary efficacy outcome in the per-protocol cohort included 1080 women in the vaccine group and 1108 women in the placebo group; the incidence of HIV-1 acquisition per 100 person-years over months 7-24 after the first vaccination was 3·38 (95% CI 2·54-4·41) in the vaccine group and 3·94 (3·04-5·03) in the placebo group, with an estimated VE(7-24) of 14·10% (95% CI -22·00 to 39·51; p=0·40). There were no serious unsolicited AEs, AEs of special interest, or deaths related to the study vaccine. In the vaccine group, 663 (50·3%) of 1317 participants had grade 1 or 2 solicited local AEs and ten (0·8%) of 1317 participants had grade 3 or 4 solicited local AEs. In the placebo group, 305 (23·1%) of 1319 participants had grade 1 or 2 solicited local AEs and three (0·2%) of 1319 participants had grade 3 or 4 solicited local AEs. 863 (65·5%) of 1317 participants in the vaccine group had grade 1 or 2 solicited systemic AEs and 34 (2·6%) of 1317 participants had grade 3 or 4 solicited systemic AEs. 763 (57·8%) of 1319 participants in the placebo group had grade 1 or 2 solicited systemic AEs and 20 (1·5%) of 1319 participants had grade 3 or 4 solicited systemic AEs. Overall, three (0·2%) of 1317 participants in the vaccine group and three (0·2%) of 1319 participants in the placebo group discontinued vaccination due to an unsolicited AE, and three (0·2%) of 1317 participants in the vaccine group and one (0·1%) of 1319 participants in the placebo group discontinued vaccination due to a solicited AE. INTERPRETATION The heterologous Ad26.Mos4.HIV and clade C gp140 vaccine regimen was safe and well tolerated but did not show efficacy in preventing HIV-1 acquisition in a population of young women in southern Africa at risk of HIV-1. FUNDING Division of AIDS at the National Institute of Allergy and Infectious Diseases through the HIV Vaccine Trials Network, Bill & Melinda Gates Foundation, Janssen Vaccines & Prevention, US Army Medical Materiel Development Activity, and Ragon Institute.
Collapse
Affiliation(s)
- Glenda E Gray
- South African Medical Research Council, Cape Town, South Africa.
| | | | | | - Steven Nijs
- Janssen Infectious Diseases, Beerse, Belgium
| | - Peter B Gilbert
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Statistics, University of Washington, Seattle, WA, USA
| | - John Hural
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | | | - M Juliana McElrath
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jackline A Odhiambo
- Hutchinson Center Research Institute of South Africa, Cape Town, South Africa
| | | | | | | | | | - Asa Tapley
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Duke University, Durham, NC, USA; Department of Surgery, Duke University, Durham, NC, USA; Department of Integrative Immunobiology, Duke University, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| | | | | | - Edith Swann
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Dan H Barouch
- Center for Virology and 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
| | | | - Lawrence Corey
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria G Pau
- Janssen Infectious Diseases, Beerse, Belgium
| | - Susan Buchbinder
- San Francisco Department of Public Health, San Francisco, CA, USA
| | - Frank Tomaka
- Janssen Research & Development, Titusville, NJ, USA
| |
Collapse
|
2
|
Kenny A, van Duijn J, Dintwe O, Heptinstall J, Burnham R, Sawant S, Zhang L, Mielke D, Khuzwayo S, Omar FL, Stanfield-Oakley S, Keyes T, Dunn B, Goodman D, Fong Y, Benkeser D, Zou R, Hural J, Hyrien O, Juraska M, Luedtke A, van der Laan L, Giorgi EE, Magaret C, Carpp LN, Pattacini L, van de Kerkhof T, Korber B, Willems W, Fisher LH, Schuitemaker H, Swann E, Kublin JG, Pau MG, Buchbinder S, Tomaka F, Nijs S, Lavreys L, Gelderblom HC, Corey L, Mngadi K, Gray GE, Borducchi E, Hendriks J, Seaton KE, Zolla-Pazner S, Barouch DH, Ferrari G, De Rosa SC, McElrath MJ, Andersen-Nissen E, Stieh DJ, Tomaras GD, Gilbert PB. 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. EBioMedicine 2024; 108:105320. [PMID: 39236556 PMCID: PMC11404224 DOI: 10.1016/j.ebiom.2024.105320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/09/2024] [Accepted: 08/17/2024] [Indexed: 09/07/2024] Open
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.
Collapse
Affiliation(s)
- Avi Kenny
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | - One Dintwe
- 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
| | - Jack Heptinstall
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA
| | - Randy Burnham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sheetal Sawant
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA
| | - Lu Zhang
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA
| | - Dieter Mielke
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA
| | - Sharon Khuzwayo
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Faatima Laher Omar
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Sherry Stanfield-Oakley
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA
| | - Taylor Keyes
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA
| | - Brooke Dunn
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA
| | - Derrick Goodman
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Rodger Zou
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Lars van der Laan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Statistics, University of Washington, Seattle, WA, USA
| | - Elena E Giorgi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Craig Magaret
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | - Bette Korber
- Los Alamos National Laboratory, Los Alamos, NM, USA; New Mexico Consortium, Los Alamos, NM, USA
| | | | - Leigh H Fisher
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Edith Swann
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Maria G Pau
- Janssen Infectious Diseases BV, Beerse, Belgium
| | - Susan Buchbinder
- San Francisco Department of Public Health, San Francisco, CA, USA
| | - Frank Tomaka
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Steven Nijs
- Janssen Infectious Diseases BV, Beerse, Belgium
| | | | - Huub C Gelderblom
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lawrence Corey
- 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
| | | | - Glenda E Gray
- South African Medical Research Council, Cape Town, South Africa
| | - Erica Borducchi
- Center for Virology & Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jenny Hendriks
- Janssen Vaccines & Prevention BV, Leiden, the Netherlands
| | - Kelly E Seaton
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA
| | | | - Dan H Barouch
- 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
| | - Guido Ferrari
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA
| | - Stephen C De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Erica Andersen-Nissen
- 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
| | - Daniel J Stieh
- Janssen Vaccines & Prevention BV, Leiden, the Netherlands
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Durham, NC, USA; Department of Integrative Immunobiology, Duke University, Durham, NC, USA.
| | - Peter B Gilbert
- Department of Biostatistics, University of Washington, Seattle, WA, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
3
|
Govindan R, Stephenson KE. HIV Vaccine Development at a Crossroads: New B and T Cell Approaches. Vaccines (Basel) 2024; 12:1043. [PMID: 39340073 PMCID: PMC11435826 DOI: 10.3390/vaccines12091043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Despite rigorous scientific efforts over the forty years since the onset of the global HIV pandemic, a safe and effective HIV-1 vaccine remains elusive. The challenges of HIV vaccine development have proven immense, in large part due to the tremendous sequence diversity of HIV and its ability to escape from antiviral adaptive immune responses. In recent years, several phase 3 efficacy trials have been conducted, testing a similar hypothesis, e.g., that non-neutralizing antibodies and classical cellular immune responses could prevent HIV-1 acquisition. These studies were not successful. As a result, the field has now pivoted to bold novel approaches, including sequential immunization strategies to drive the generation of broadly neutralizing antibodies and human CMV-vectored vaccines to elicit MHC-E-restricted CD8+ T cell responses. Many of these vaccine candidates are now in phase 1 trials, with early promising results.
Collapse
Affiliation(s)
- Ramesh Govindan
- Division of Infectious Diseases and Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Kathryn E. Stephenson
- Division of Infectious Diseases and Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
- Harvard Medical School, Boston, MA 02115, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
4
|
Pekarek MJ, Weaver EA. Influenza B Virus Vaccine Innovation through Computational Design. Pathogens 2024; 13:755. [PMID: 39338946 PMCID: PMC11434669 DOI: 10.3390/pathogens13090755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
As respiratory pathogens, influenza B viruses (IBVs) cause a significant socioeconomic burden each year. Vaccine and antiviral development for influenza viruses has historically viewed IBVs as a secondary concern to influenza A viruses (IAVs) due to their lack of animal reservoirs compared to IAVs. However, prior to the global spread of SARS-CoV-2, the seasonal epidemics caused by IBVs were becoming less predictable and inducing more severe disease, especially in high-risk populations. Globally, researchers have begun to recognize the need for improved prevention strategies for IBVs as a primary concern. This review discusses what is known about IBV evolutionary patterns and the effect of the spread of SARS-CoV-2 on these patterns. We also analyze recent advancements in the development of novel vaccines tested against IBVs, highlighting the promise of computational vaccine design strategies when used to target both IBVs and IAVs and explain why these novel strategies can be employed to improve the effectiveness of IBV vaccines.
Collapse
Affiliation(s)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
5
|
Nelson GW, van Duijn J, Yuki Y, Pau MG, Tomaka F, Lavreys L, DeRosa SC, McElrath MJ, Kirk GD, Michael NL, Haas DW, Deeks SG, Wolinsky S, Walker B, Barouch DH, Stieh D, Carrington M. Prediction of differential Gag versus Env responses to a mosaic HIV-1 vaccine regimen by HLA class I alleles. J Virol 2024; 98:e0028124. [PMID: 39046263 PMCID: PMC11338073 DOI: 10.1128/jvi.00281-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/21/2024] [Indexed: 07/25/2024] Open
Abstract
HLA class I variation has the strongest effect genome-wide on outcome after HIV infection, and as such, an understanding of the impact of HLA polymorphism on response to HIV vaccination may inform vaccine design. We sought HLA associations with HIV-directed immunogenicity in the phase 1/2a APPROACH vaccine trial, which tested vaccine regimens containing mosaic inserts in Ad26 and MVA vectors, with or without a trimeric gp140 protein. While there were no HLA allelic associations with the overall cellular immune response to the vaccine assessed by ELISpot (Gag, Pol, and Env combined), significant associations with differential response to Gag compared to Env antigens were observed. Notably, HLA class I alleles known to associate with disease susceptibility in HIV natural history cohorts are associated with stronger Env-directed responses, whereas protective alleles are associated with stronger Gag-directed responses. Mean viral loads determined for each HLA allele in untreated individuals correlated negatively with the strength of the Gag response minus the Env response in Black vaccinees based on both ELISpot and CD8+ T cell ICS responses. As the association of T cell responses to conserved Gag epitopes with lower viral load in untreated individuals is well established, our data raise the possibility that the Ad26.Mos.HIV vaccine may induce more effective cellular responses in those with HLA alleles that confer improved virologic control in untreated HIV infection.IMPORTANCENo vaccine tested to date has shown sufficient efficacy against HIV infection. A vaccine that induces robust responses in one individual may fail to do so in another individual due to variation in HLA class I genes, loci central to the immune response. Extensive data have shown the strong effect of HLA variation on outcome after HIV infection, but very little is known about the effect of such variation on HIV vaccine success. Here, we identify a link between the effect of HLA variation on HIV disease outcome and immune responses to an HIV vaccine. HLA variants associated with better HIV control after infection also induce stronger responses against the HIV Gag protein relative to the Env protein after vaccination. Given the virologic control conferred by responses to Gag in natural history of HIV infection, these data suggest that HLA alleles conferring protection after HIV infection may also support a more effective cellular response to HIV vaccination.
Collapse
Affiliation(s)
- George W. Nelson
- Basic Science Program
Frederick National Laboratory for Cancer Research, National Cancer
Institute and Laboratory of Integrative Cancer Immunology, Center for
Cancer Research, National Cancer
Institute, Bethesda,
Maryland, USA
| | | | - Yuko Yuki
- Basic Science Program
Frederick National Laboratory for Cancer Research, National Cancer
Institute and Laboratory of Integrative Cancer Immunology, Center for
Cancer Research, National Cancer
Institute, Bethesda,
Maryland, USA
| | - Maria G. Pau
- Janssen Vaccines
& Prevention,
Leiden, the Netherlands
| | - Frank Tomaka
- Janssen Research and
Development, Titusville,
New Jersey, USA
| | | | - Steven C. DeRosa
- Department of
Laboratory Medicine and Pathology, University of
Washington, Seattle,
Washington, USA
- Division of Vaccine
and Infectious Disease, Fred Hutchinson Cancer
Center, Seattle,
Washington, USA
| | - M. Juliana McElrath
- Department of
Laboratory Medicine and Pathology, University of
Washington, Seattle,
Washington, USA
- Division of Vaccine
and Infectious Disease, Fred Hutchinson Cancer
Center, Seattle,
Washington, USA
- Department of
Medicine, Division of Allergy and Infectious Diseases, University of
Washington, Seattle,
Washington, USA
| | - Gregory D. Kirk
- Department of
Epidemiology, Bloomberg School of Public Health, Johns Hopkins
University, Baltimore,
Maryland, USA
| | - Nelson L. Michael
- US Military HIV
Research Program, Walter Reed Army Institute of
Research, Silver Spring,
Maryland, USA
| | - David W. Haas
- Department of
Medicine, Vanderbilt University School of
Medicine, Nashville,
Tennessee, USA
| | - Steven G. Deeks
- Department of
Medicine, University of California,
San Francisco, California,
USA
| | - Steven Wolinsky
- Division of
Infectious Diseases, Department of Medicine, The Feinberg School of
Medicine, Northwestern University,
Chicago, Illinois, USA
| | - Bruce Walker
- Ragon Institute of
Massachusetts General Hospital, Massachusetts Institute of Technology
and Harvard University,
Cambridge, Massachusetts,
USA
| | - Dan H. Barouch
- Ragon Institute of
Massachusetts General Hospital, Massachusetts Institute of Technology
and Harvard University,
Cambridge, Massachusetts,
USA
- Center for Virology
and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard
Medical School, Boston,
Massachusetts, USA
- Harvard Medical
School, Boston,
Massachusetts, USA
| | - Daniel Stieh
- Janssen Vaccines
& Prevention,
Leiden, the Netherlands
| | - Mary Carrington
- Ragon Institute of
Massachusetts General Hospital, Massachusetts Institute of Technology
and Harvard University,
Cambridge, Massachusetts,
USA
| |
Collapse
|
6
|
Wolf T, Calisan KK, Stitz J, Barbe S. The effects of high shear rates on the average hydrodynamic diameter measured in biomimetic HIV Gag virus-like particle dispersions. Front Bioeng Biotechnol 2024; 12:1367405. [PMID: 38860137 PMCID: PMC11163053 DOI: 10.3389/fbioe.2024.1367405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/07/2024] [Indexed: 06/12/2024] Open
Abstract
HIV Gag virus-like particles (HIV Gag VLPs) are promising HIV vaccine candidates. In the literature, they are often described as shear-sensitive particles, and authors usually recommend the operation of tangential flow filtration (TFF) gently at shear rates below 4,000 s-1 to 6,000 s-1. This in turn poses a severe limitation to the performance of TFF-mediated concentration of VLPs, which would be substantially enhanced by working at higher shear rates. To our knowledge, studies examining the shear sensitivity of HIV Gag VLPs and providing detailed information and evidence for the fragility of these particles have not been conducted yet. Thus, we investigated the effect of high shear rates on the colloidal stability of mosaic VLPs (Mos-VLPs) as relevant examples for HIV Gag VLPs. For this purpose, Mos-VLPs were exposed to different shear rates ranging from 3,395 s-1 to 22, 365 s-1 for 2 h. The average hydrodynamic diameter (AHD) and the polydispersity index (PDI) of the associated particle size distribution were used as stability indicators and measured after the treatment and during storage through dynamic light scattering. At high shear rates, we observed an increase in both AHD and PDI during the storage of HIV Mos1.Gag VLPs (bVLP-without envelope proteins) and Mos1.Gag + Mos2S.Env VLPs (eVLP-with envelope proteins). eVLPs exhibited higher colloidal stability than bVLPs, and we discuss the potential stabilizing role of envelope proteins. We finally demonstrated that the dispersion medium also has a considerable impact on the stability of Mos-VLPs.
Collapse
Affiliation(s)
- Tobias Wolf
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Leverkusen, Germany
- Institue of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Kerim Kadir Calisan
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Leverkusen, Germany
| | - Jörn Stitz
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Leverkusen, Germany
| | - Stéphan Barbe
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Leverkusen, Germany
| |
Collapse
|
7
|
Bai H, Lewitus E, Li Y, Thomas PV, Zemil M, Merbah M, Peterson CE, Thuraisamy T, Rees PA, Hajduczki A, Dussupt V, Slike B, Mendez-Rivera L, Schmid A, Kavusak E, Rao M, Smith G, Frey J, Sims A, Wieczorek L, Polonis V, Krebs SJ, Ake JA, Vasan S, Bolton DL, Joyce MG, Townsley S, Rolland M. Contemporary HIV-1 consensus Env with AI-assisted redesigned hypervariable loops promote antibody binding. Nat Commun 2024; 15:3924. [PMID: 38724518 PMCID: PMC11082178 DOI: 10.1038/s41467-024-48139-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
An effective HIV-1 vaccine must elicit broadly neutralizing antibodies (bnAbs) against highly diverse Envelope glycoproteins (Env). Since Env with the longest hypervariable (HV) loops is more resistant to the cognate bnAbs than Env with shorter HV loops, we redesigned hypervariable loops for updated Env consensus sequences of subtypes B and C and CRF01_AE. Using modeling with AlphaFold2, we reduced the length of V1, V2, and V5 HV loops while maintaining the integrity of the Env structure and glycan shield, and modified the V4 HV loop. Spacers are designed to limit strain-specific targeting. All updated Env are infectious as pseudoviruses. Preliminary structural characterization suggests that the modified HV loops have a limited impact on Env's conformation. Binding assays show improved binding to modified subtype B and CRF01_AE Env but not to subtype C Env. Neutralization assays show increases in sensitivity to bnAbs, although not always consistently across clades. Strikingly, the HV loop modification renders the resistant CRF01_AE Env sensitive to 10-1074 despite the absence of a glycan at N332.
Collapse
Affiliation(s)
- Hongjun Bai
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Eric Lewitus
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Yifan Li
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Paul V Thomas
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Michelle Zemil
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Mélanie Merbah
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Caroline E Peterson
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Thujitha Thuraisamy
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Phyllis A Rees
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Agnes Hajduczki
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Vincent Dussupt
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Bonnie Slike
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Letzibeth Mendez-Rivera
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Annika Schmid
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Erin Kavusak
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Mekhala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Gabriel Smith
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Jessica Frey
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Alicea Sims
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Lindsay Wieczorek
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Victoria Polonis
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Shelly J Krebs
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Julie A Ake
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Sandhya Vasan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Diane L Bolton
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - M Gordon Joyce
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Samantha Townsley
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Morgane Rolland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA.
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA.
| |
Collapse
|
8
|
Schmidt S, Mengistu M, Daffis S, Ahmadi-Erber S, Deutschmann D, Grigoriev T, Chu R, Leung C, Tomkinson A, Uddin MN, Moshkani S, Robek MD, Perry J, Lauterbach H, Orlinger K, Fletcher SP, Balsitis S. Alternating Arenavirus Vector Immunization Generates Robust Polyfunctional Genotype Cross-Reactive Hepatitis B Virus-Specific CD8 T-Cell Responses and High Anti-Hepatitis B Surface Antigen Titers. J Infect Dis 2024; 229:1077-1087. [PMID: 37602681 DOI: 10.1093/infdis/jiad340] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023] Open
Abstract
Hepatitis B Virus (HBV) is a major driver of infectious disease mortality. Curative therapies are needed and ideally should induce CD8 T cell-mediated clearance of infected hepatocytes plus anti-hepatitis B surface antigen (HBsAg) antibodies (anti-HBs) to neutralize residual virus. We developed a novel therapeutic vaccine using non-replicating arenavirus vectors. Antigens were screened for genotype conservation and magnitude and genotype reactivity of T cell response, then cloned into Pichinde virus (PICV) vectors (recombinant PICV, GS-2829) and lymphocytic choriomeningitis virus (LCMV) vectors (replication-incompetent, GS-6779). Alternating immunizations with GS-2829 and GS-6779 induced high-magnitude HBV T cell responses, and high anti-HBs titers. Dose schedule optimization in macaques achieved strong polyfunctional CD8 T cell responses against core, HBsAg, and polymerase and high titer anti-HBs. In AAV-HBV mice, GS-2829 and GS-6779 were efficacious in animals with low pre-treatment serum HBsAg. Based on these results, GS-2829 and GS-6779 could become a central component of cure regimens.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ruth Chu
- Gilead Sciences, Foster City, California, USA
| | - Cleo Leung
- Gilead Sciences, Foster City, California, USA
| | | | - Mohammad Nizam Uddin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Safiehkhatoon Moshkani
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Michael D Robek
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Jason Perry
- Gilead Sciences, Foster City, California, USA
| | | | | | | | | |
Collapse
|
9
|
Lewitus E, Li Y, Bai H, Pham P, Rolland M. HIV-1 Gag, Pol, and Env diversified with limited adaptation since the 1980s. mBio 2024; 15:e0174923. [PMID: 38329340 PMCID: PMC10936417 DOI: 10.1128/mbio.01749-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/02/2024] [Indexed: 02/09/2024] Open
Abstract
Knowledge of HIV-1 global sequence diversity is critical for developing an effective prophylactic against HIV-1 infection. We developed the Hervé platform to analyze and visualize trends in HIV-1 diversification. Using Hervé, we analyzed 4,830 Env, 4,407 Gag, and 3,002 Pol publicly available independent sequences corresponding to subtypes A1, A6, B, C, D, F1, and G and circulating recombinant forms (CRFs) 01_AE, 02_AG, and 07_BC; sequences were sampled between 1980 and 2020 from 82 countries. HIV-1 diversified with a median of 1.82 amino acid substitutions per year in Env, 0.297 in Gag, and 0.779 in Pol. Yet, Env subtype B diversification plateaued post-2000. Pairwise diversity within subtypes and CRFs increased by 41.82% (range = 24.85%-54.41%) in Env, 56.93% (15.38%-89.16%) in Gag, and 46.12% (11.70%-70.57%) in Pol. Consensus sequences based on sequences sampled in each decade remained relatively stable over time. Similarly, at antibody epitope sites, only 0-8 residues that were minority variants became consensus over time in any subtype/CRF and only one known drug resistance mutation site differed from the reference (subtype G). The apparent contradiction between the fast diversification of HIV-1 and its limited adaptation illustrates that HIV-1 evolution is not directional and its consensus is at the intersection of millions of within-host selective processes occurring in a star-like manner. While a consensus sequence is a better representation of HIV-1 diversity than any individual sequence, consensus sequences have progressively become more distant from the circulating sequences they represent. IMPORTANCE Global surveillance of HIV-1 sequences is critical for designing relevant prophylactic and therapeutic interventions to infection. We designed an open-source platform, Hervé, for analyzing and visualizing the diversification dynamics of HIV-1 protein sequences. We characterized the evolution of over 12,000 HIV-1 Env, Gag, and Pol protein sequences from 1980-2020 and found that, despite a steady increase in intra-subtype and circulating recombinant form diversity, the most frequent residue at each site, i.e., the consensus, has varied only moderately.
Collapse
Affiliation(s)
- Eric Lewitus
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Yifan Li
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Hongjun Bai
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Phuc Pham
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Morgane Rolland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| |
Collapse
|
10
|
Kaur A, Vaccari M. Exploring HIV Vaccine Progress in the Pre-Clinical and Clinical Setting: From History to Future Prospects. Viruses 2024; 16:368. [PMID: 38543734 PMCID: PMC10974975 DOI: 10.3390/v16030368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 04/01/2024] Open
Abstract
The human immunodeficiency virus (HIV) continues to pose a significant global health challenge, with millions of people affected and new cases emerging each year. While various treatment and prevention methods exist, including antiretroviral therapy and non-vaccine approaches, developing an effective vaccine remains the most crucial and cost-effective solution to combating the HIV epidemic. Despite significant advancements in HIV research, the HIV vaccine field has faced numerous challenges, and only one clinical trial has demonstrated a modest level of efficacy. This review delves into the history of HIV vaccines and the current efforts in HIV prevention, emphasizing pre-clinical vaccine development using the non-human primate model (NHP) of HIV infection. NHP models offer valuable insights into potential preventive strategies for combating HIV, and they play a vital role in informing and guiding the development of novel vaccine candidates before they can proceed to human clinical trials.
Collapse
Affiliation(s)
- Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Monica Vaccari
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
11
|
van Duijn J, Stieh D, Fernandez N, King D, Gilmour J, Tolboom J, Callewaert K, Willems W, Pau MG, De Rosa SC, McElrath MJ, Barouch DH, Hayes P. Mosaic HIV-1 vaccination induces anti-viral CD8 + T cell functionality in the phase 1/2a clinical trial APPROACH. J Virol 2023; 97:e0112623. [PMID: 37811993 PMCID: PMC10617392 DOI: 10.1128/jvi.01126-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023] Open
Abstract
IMPORTANCE The functionality of CD8+ T cells against human immunodeficiency virus-1 (HIV-1) antigens is indicative of HIV-progression in both animal models and people living with HIV. It is, therefore, of interest to assess CD8+ T cell responses in a prophylactic vaccination setting, as this may be an important component of the immune system that inhibits HIV-1 replication. T cell responses induced by the adenovirus serotype 26 (Ad26) mosaic vaccine regimen were assessed previously by IFN-γ ELISpot and flow cytometric assays, yet these assays only measure cytokine production but not the capacity of CD8+ T cells to inhibit replication of HIV-1. In this study, we demonstrate direct anti-viral function of the clinical Ad26 mosaic vaccine regimen through ex vivo inhibition of replication of diverse clades of HIV-1 isolates in the participant's own CD4+ T cells.
Collapse
Affiliation(s)
| | - Daniel Stieh
- Janssen Vaccines & Prevention B.V., Leiden, the Netherlands
| | - Natalia Fernandez
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Deborah King
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Jill Gilmour
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| | - Jeroen Tolboom
- Janssen Vaccines & Prevention B.V., Leiden, the Netherlands
| | | | | | - Maria G. Pau
- Janssen Vaccines & Prevention B.V., Leiden, the Netherlands
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Peter Hayes
- IAVI Human Immunology Laboratory, Imperial College, London, United Kingdom
| |
Collapse
|
12
|
Ahmed K, Jha S. Oncoviruses: How do they hijack their host and current treatment regimes. Biochim Biophys Acta Rev Cancer 2023; 1878:188960. [PMID: 37507056 DOI: 10.1016/j.bbcan.2023.188960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Viruses have the ability to modulate the cellular machinery of their host to ensure their survival. While humans encounter numerous viruses daily, only a select few can lead to disease progression. Some of these viruses can amplify cancer-related traits, particularly when coupled with factors like immunosuppression and co-carcinogens. The global burden of cancer development resulting from viral infections is approximately 12%, and it arises as an unfortunate consequence of persistent infections that cause chronic inflammation, genomic instability from viral genome integration, and dysregulation of tumor suppressor genes and host oncogenes involved in normal cell growth. This review provides an in-depth discussion of oncoviruses and their strategies for hijacking the host's cellular machinery to induce cancer. It delves into how viral oncogenes drive tumorigenesis by targeting key cell signaling pathways. Additionally, the review discusses current therapeutic approaches that have been approved or are undergoing clinical trials to combat malignancies induced by oncoviruses. Understanding the intricate interactions between viruses and host cells can lead to the development of more effective treatments for virus-induced cancers.
Collapse
Affiliation(s)
- Kainat Ahmed
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sudhakar Jha
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA.
| |
Collapse
|
13
|
Stieh DJ, Barouch DH, Comeaux C, Sarnecki M, Stephenson KE, Walsh SR, Sawant S, Heptinstall J, Tomaras GD, Kublin JG, McElrath MJ, Cohen KW, De Rosa SC, Alter G, Ferrari G, Montefiori D, Mann P, Nijs S, Callewaert K, Goepfert PA, Edupuganti S, Karita E, Seaman MS, Corey L, Baden LR, Pau MG, Schuitemaker H, Tomaka F. Safety and Immunogenicity of Ad26-Vectored HIV Vaccine With Mosaic Immunogens and a Novel Mosaic Envelope Protein in HIV-Uninfected Adults: A Phase 1/2a Study. J Infect Dis 2023; 227:939-950. [PMID: 36348617 PMCID: PMC10202119 DOI: 10.1093/infdis/jiac445] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Developing a cross-clade, globally effective HIV vaccine remains crucial for eliminating HIV. METHODS This placebo-controlled, double-blind, phase 1/2a study enrolled healthy HIV-uninfected adults at low risk for HIV infection. They were randomized (1:4:1) to receive 4 doses of an adenovirus 26-based HIV-1 vaccine encoding 2 mosaic Gag and Pol, and 2 mosaic Env proteins plus adjuvanted clade C gp140 (referred to here as clade C regimen), bivalent protein regimen (clade C regimen plus mosaic gp140), or placebo. Primary end points were safety and antibody responses. RESULTS In total 152/155 participants (clade C, n = 26; bivalent protein, n = 103; placebo, n = 26) received ≥1 injection. The highest adverse event (AE) severity was grade 3 (local pain/tenderness, 12%, 2%, and 0% of the respective groups; solicited systemic AEs, 19%, 15%, 0%). HIV-1 mosaic gp140-binding antibody titers were 79 595 ELISA units (EU)/mL and 137 520 EU/mL in the clade C and bivalent protein groups (P < .001) after dose 4 and 16 862 EU/mL and 25 162 EU/mL 6 months later. Antibody response breadth against clade C gp140 and clade C/non-clade C gp120 was highest in the bivalent protein group. CONCLUSIONS Adding mosaic gp140 to the clade C regimen increased and broadened the elicited immune response without compromising safety or clade C responses. Clinical Trials Registration. NCT02935686.
Collapse
Affiliation(s)
| | - Dan H Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | | | | | - Kathryn E Stephenson
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen R Walsh
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sheetal Sawant
- Department of Surgery, Center for Human Systems Immunology, and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jack Heptinstall
- Department of Surgery, Center for Human Systems Immunology, and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Georgia D Tomaras
- Department of Surgery, Center for Human Systems Immunology, and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kristen W Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stephen C De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Guido Ferrari
- Department of Surgery, Center for Human Systems Immunology, and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - David Montefiori
- Department of Surgery, Center for Human Systems Immunology, and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Philipp Mann
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Steven Nijs
- Janssen Research and Development, Beerse, Belgium
| | | | - Paul A Goepfert
- Division of Infectious Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Srilatha Edupuganti
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Michael S Seaman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Lindsey R Baden
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria G Pau
- Janssen Vaccines and Prevention Leiden, the Netherlands
| | | | - Frank Tomaka
- Janssen Research and Development, Titusville, New Jersey, USA
| |
Collapse
|
14
|
Cohen KW, Fiore-Gartland A, Walsh SR, Yusim K, Frahm N, Elizaga ML, Maenza J, Scott H, Mayer KH, Goepfert PA, Edupuganti S, Pantaleo G, Hutter J, Morris DE, De Rosa SC, Geraghty DE, Robb ML, Michael NL, Fischer W, Giorgi EE, Malhi H, Pensiero MN, Ferrari G, Tomaras GD, Montefiori DC, Gilbert PB, McElrath MJ, Haynes BF, Korber BT, Baden LR. Trivalent mosaic or consensus HIV immunogens prime humoral and broader cellular immune responses in adults. J Clin Invest 2023; 133:e163338. [PMID: 36787249 PMCID: PMC9927951 DOI: 10.1172/jci163338] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/27/2022] [Indexed: 02/15/2023] Open
Abstract
BACKGROUNDMosaic and consensus HIV-1 immunogens provide two distinct approaches to elicit greater breadth of coverage against globally circulating HIV-1 and have shown improved immunologic breadth in nonhuman primate models.METHODSThis double-blind randomized trial enrolled 105 healthy HIV-uninfected adults who received 3 doses of either a trivalent global mosaic, a group M consensus (CON-S), or a natural clade B (Nat-B) gp160 env DNA vaccine followed by 2 doses of a heterologous modified vaccinia Ankara-vectored HIV-1 vaccine or placebo. We performed prespecified blinded immunogenicity analyses at day 70 and day 238 after the first immunization. T cell responses to vaccine antigens and 5 heterologous Env variants were fully mapped.RESULTSEnv-specific CD4+ T cell responses were induced in 71% of the mosaic vaccine recipients versus 48% of the CON-S recipients and 48% of the natural Env recipients. The mean number of T cell epitopes recognized was 2.5 (95% CI, 1.2-4.2) for mosaic recipients, 1.6 (95% CI, 0.82-2.6) for CON-S recipients, and 1.1 (95% CI, 0.62-1.71) for Nat-B recipients. Mean breadth was significantly greater in the mosaic group than in the Nat-B group using overall (P = 0.014), prime-matched (P = 0.002), heterologous (P = 0.046), and boost-matched (P = 0.009) measures. Overall T cell breadth was largely due to Env-specific CD4+ T cell responses.CONCLUSIONPriming with a mosaic antigen significantly increased the number of epitopes recognized by Env-specific T cells and enabled more, albeit still limited, cross-recognition of heterologous variants. Mosaic and consensus immunogens are promising approaches to address global diversity of HIV-1.TRIAL REGISTRATIONClinicalTrials.gov NCT02296541.FUNDINGUS NIH grants UM1 AI068614, UM1 AI068635, UM1 AI068618, UM1 AI069412, UL1 RR025758, P30 AI064518, UM1 AI100645, and UM1 AI144371, and Bill & Melinda Gates Foundation grant OPP52282.
Collapse
Affiliation(s)
- Kristen W. Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stephen R. Walsh
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Karina Yusim
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Marnie L. Elizaga
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Hyman Scott
- San Francisco Department of Public Health, San Francisco, California, USA
| | - Kenneth H. Mayer
- Harvard Medical School, Boston, Massachusetts, USA
- The Fenway Institute, Fenway Health, Boston, Massachusetts, USA
| | | | | | | | - Julia Hutter
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Daryl E. Morris
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Daniel E. Geraghty
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Merlin L. Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Nelson L. Michael
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Will Fischer
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Elena E. Giorgi
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Harmandeep Malhi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Michael N. Pensiero
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - David C. Montefiori
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Bette T. Korber
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Lindsey R. Baden
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
15
|
Wrapp D, Mu Z, Thakur B, Janowska K, Ajayi O, Barr M, Parks R, Mansouri K, Edwards RJ, Hahn BH, Acharya P, Saunders KO, Haynes BF. Structure-Based Stabilization of SOSIP Env Enhances Recombinant Ectodomain Durability and Yield. J Virol 2023; 97:e0167322. [PMID: 36633409 PMCID: PMC9888283 DOI: 10.1128/jvi.01673-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
The envelope glycoprotein (Env) is the main focus of human immunodeficiency virus type 1 (HIV-1) vaccine development due to its critical role in viral entry. Despite advances in protein engineering, many Env proteins remain recalcitrant to recombinant expression due to their inherent metastability, making biochemical and immunological experiments impractical or impossible. Here, we report a novel proline stabilization strategy to facilitate the production of prefusion Env trimers. This approach, termed "2P," works synergistically with previously described SOSIP mutations and dramatically increases the yield of recombinantly expressed Env ectodomains without altering the antigenic or conformational properties of near-native Env. We determined that the 2P mutations function by enhancing the durability of the prefusion conformation and that this stabilization strategy is broadly applicable to evolutionarily and antigenically diverse Env constructs. These findings provide a new Env stabilization platform to facilitate biochemical research and expand the number of Env variants that can be developed as future HIV-1 vaccine candidates. IMPORTANCE Recent estimates have placed the number of new human immunodeficiency virus type 1 (HIV-1) infections at approximately 1.5 million per year, emphasizing the ongoing and urgent need for an effective vaccine. The envelope (Env) glycoprotein is the main focus of HIV-1 vaccine development, but, due to its inherent metastability, many Env variants are difficult to recombinantly express in the relatively large quantities that are required for biochemical studies and animal trials. Here, we describe a novel structure-based stabilization strategy that works synergistically with previously described SOSIP mutations to increase the yield of prefusion HIV-1 Env.
Collapse
Affiliation(s)
- Daniel Wrapp
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Zekun Mu
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Bhishem Thakur
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Katarzyna Janowska
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Oluwatobi Ajayi
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert J. Edwards
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Beatrice H. Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Kevin O. Saunders
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Department of Microbiology and Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
16
|
Safety and immunogenicity of a mosaic vaccine booster against Omicron and other SARS-CoV-2 variants: a randomized phase 2 trial. Signal Transduct Target Ther 2023; 8:20. [PMID: 36596779 PMCID: PMC9808735 DOI: 10.1038/s41392-022-01295-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 01/04/2023] Open
Abstract
An ongoing randomized, double-blind, controlled phase 2 trial was conducted to evaluate the safety and immunogenicity of a mosaic-type recombinant vaccine candidate, named NVSI-06-09, as a booster dose in subjects aged 18 years and older from the United Arab Emirates (UAE), who had administered two or three doses of inactivated vaccine BBIBP-CorV at least 6 months prior to enrollment. The participants were randomly assigned with 1:1 to receive a booster dose of NVSI-06-09 or BBIBP-CorV. The primary outcomes were immunogenicity and safety against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant, and the exploratory outcome was cross-immunogenicity against other circulating strains. Between May 25 and 30, 2022, 516 adults received booster vaccination with 260 in NVSI-06-09 group and 256 in BBIBP-CorV group. Interim results showed a similar safety profile between two booster groups, with low incidence of adverse reactions of grade 1 or 2. For immunogenicity, by day 14 post-booster, the fold rises in neutralizing antibody geometric mean titers (GMTs) from baseline elicited by NVSI-06-09 were remarkably higher than those by BBIBP-CorV against the prototype strain (19.67 vs 4.47-fold), Omicron BA.1.1 (42.35 vs 3.78-fold), BA.2 (25.09 vs 2.91-fold), BA.4 (22.42 vs 2.69-fold), and BA.5 variants (27.06 vs 4.73-fold). Similarly, the neutralizing GMTs boosted by NVSI-06-09 against Beta and Delta variants were also 6.60-fold and 7.17-fold higher than those by BBIBP-CorV. Our findings indicated that a booster dose of NVSI-06-09 was well-tolerated and elicited broad-spectrum neutralizing responses against divergent SARS-CoV-2 variants, including Omicron and its sub-lineages.
Collapse
|
17
|
Bi J, Zheng Y, Wang C, Ding Y. An Attention Based Bidirectional LSTM Method to Predict the Binding of TCR and Epitope. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3272-3280. [PMID: 34559661 DOI: 10.1109/tcbb.2021.3115353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The T-cell epitope prediction has always been a long-term challenge in immunoinformatics and bioinformatics. Studying the specific recognition between T-cell receptor (TCR) and peptide-major histocompatibility complex (p-MHC) complexes can help us better understand the immune mechanism, it's also make a signification contribution in developing vaccines and targeted drugs. Meanwhile, more advanced methods are needed for distinguishing TCRs binding from different epitopes. In this paper, we introduce a hybrid model composed of bidirectional long short-term memory networks (BiLSTM), attention and convolutional neural networks (CNN) that can identified the binding of TCRs to epitopes. The BiLSTM can more completely extract amino acid forward and backward information in the sequence, and attention mechanism can focus on amino acids at certain positions from complex sequences to capture the most important feature, then CNN was used to further extract salient features to predict the binding of TCR-epitope. In McPAS dataset, the AUC value (the area under ROC curve) of naive TCR-epitope binding is 0.974 and specific TCR-epitope binding is 0.887. The model has achieved better prediction results than other existing models (TCRGP, ERGO, NetTCR), and some experiments are used to analyze the advantages of our model. The algorithm is available at https://github.com/bijingshu/BiAttCNN.git.
Collapse
|
18
|
Optimal sequence-based design for multi-antigen HIV-1 vaccines using minimally distant antigens. PLoS Comput Biol 2022; 18:e1010624. [PMID: 36315492 PMCID: PMC9621458 DOI: 10.1371/journal.pcbi.1010624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
The immense global diversity of HIV-1 is a significant obstacle to developing a safe and effective vaccine. We recently showed that infections established with multiple founder variants are associated with the development of neutralization breadth years later. We propose a novel vaccine design strategy that integrates the variability observed in acute HIV-1 infections with multiple founder variants. We developed a probabilistic model to simulate this variability, yielding a set of sequences that present the minimal diversity seen in an infection with multiple founders. We applied this model to a subtype C consensus sequence for the Envelope (Env) (used as input) and showed that the simulated Env sequences mimic the mutational landscape of an infection with multiple founder variants, including diversity at antibody epitopes. The derived set of multi-founder-variant-like, minimally distant antigens is designed to be used as a vaccine cocktail specific to a HIV-1 subtype or circulating recombinant form and is expected to promote the development of broadly neutralizing antibodies. Diverse HIV-1 populations are generally thought to promote neutralizing responses. Current leading HIV-1 vaccine design strategies maximize the distance between antigens to attempt to cover global HIV-1 diversity or serialize immunizations to recapitulate the temporal evolution of HIV-1 during infection. To date, no vaccine has elicited broadly neutralizing antibodies. As we recently demonstrated that infection with multiple HIV-1 founder variants is predictive of neutralization breadth, we propose a novel strategy that endeavors to promote the development of broadly neutralizing antibodies by replicating the diversity of multi-founder variant acute infections. By training an HIV-1 Env consensus sequence on the diversity from acute infections with multiple founders, we derived in silico a set of minimally distant antigens that is representative of the diversity seen in a multi-founder acute infection. As the model is particular to the input sequence, it can produce antigens specific to any HIV-1 subtype or circulating recombinant form (CRF). We applied this to HIV-1 subtype C and obtained a set of minimally distant antigens that can be used as a vaccine cocktail.
Collapse
|
19
|
Araújo NM, Rubio IGS, Toneto NPA, Morale MG, Tamura RE. The use of adenoviral vectors in gene therapy and vaccine approaches. Genet Mol Biol 2022; 45:e20220079. [PMID: 36206378 PMCID: PMC9543183 DOI: 10.1590/1678-4685-gmb-2022-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022] Open
Abstract
Adenovirus was first identified in the 1950s and since then this pathogenic group
of viruses has been explored and transformed into a genetic transfer vehicle.
Modification or deletion of few genes are necessary to transform it into a
conditionally or non-replicative vector, creating a versatile tool capable of
transducing different tissues and inducing high levels of transgene expression.
In the early years of vector development, the application in monogenic diseases
faced several hurdles, including short-term gene expression and even a fatality.
On the other hand, an adenoviral delivery strategy for treatment of cancer was
the first approved gene therapy product. There is an increasing interest in
expressing transgenes with therapeutic potential targeting the cancer hallmarks,
inhibiting metastasis, inducing cancer cell death or modulating the immune
system to attack the tumor cells. Replicative adenovirus as vaccines may be even
older and date to a few years of its discovery, application of non-replicative
adenovirus for vaccination against different microorganisms has been
investigated, but only recently, it demonstrated its full potential being one of
the leading vaccination tools for COVID-19. This is not a new vector nor a new
technology, but the result of decades of careful and intense work in this
field.
Collapse
Affiliation(s)
- Natália Meneses Araújo
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil.
| | - Ileana Gabriela Sanchez Rubio
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | | | - Mirian Galliote Morale
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | - Rodrigo Esaki Tamura
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil.
| |
Collapse
|
20
|
Liu X, Zhao T, Wang L, Li M, Sun C, Shu Y. Strategies targeting hemagglutinin cocktail as a potential universal influenza vaccine. Front Microbiol 2022; 13:1014122. [PMID: 36246271 PMCID: PMC9558277 DOI: 10.3389/fmicb.2022.1014122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Vaccination is the most effective means of protecting people from influenza virus infection. The effectiveness of existing vaccines is very limited due to antigenic drift of the influenza virus. Therefore, there is a requirement to develop a universal vaccine that provides broad and long-lasting protection against influenza. CD8+ T-cell response played a vital role in controlling influenza virus infection, reducing viral load, and less clinical syndrome. In this study, we optimized the HA sequences of human seasonal influenza viruses (H1N1, H3N2, Victoria, and Yamagata) by designing multivalent vaccine antigen sets using a mosaic vaccine design strategy and genetic algorithms, and designed an HA mosaic cocktail containing the most potential CTL epitopes of seasonal influenza viruses. We then tested the recombinant mosaic antigen, which has a significant number of potential T-cell epitopes. Results from genetic evolutionary analyses and 3D structural simulations demonstrated its potential to be an effective immunogen. In addition, we have modified an existing neutralizing antibody-based seasonal influenza virus vaccine to include a component that activates cross-protective T cells, which would provide an attractive strategy for improving human protection against seasonal influenza virus drift and mutation and provide an idea for the development of a rationally designed influenza vaccine targeting T lymphocyte immunity.
Collapse
Affiliation(s)
- Xuejie Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Tianyi Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Liangliang Wang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Minchao Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yuelong Shu,
| |
Collapse
|
21
|
Zhang J, Han ZB, Liang Y, Zhang XF, Jin YQ, Du LF, Shao S, Wang H, Hou JW, Xu K, Lei W, Lei ZH, Liu ZM, Zhang J, Hou YN, Liu N, Shen FJ, Wu JJ, Zheng X, Li XY, Li X, Huang WJ, Wu GZ, Su JG, Li QM. A mosaic-type trimeric RBD-based COVID-19 vaccine candidate induces potent neutralization against Omicron and other SARS-CoV-2 variants. eLife 2022; 11:e78633. [PMID: 36004719 PMCID: PMC9481243 DOI: 10.7554/elife.78633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Large-scale populations in the world have been vaccinated with COVID-19 vaccines, however, breakthrough infections of SARS-CoV-2 are still growing rapidly due to the emergence of immune-evasive variants, especially Omicron. It is urgent to develop effective broad-spectrum vaccines to better control the pandemic of these variants. Here, we present a mosaic-type trimeric form of spike receptor-binding domain (mos-tri-RBD) as a broad-spectrum vaccine candidate, which carries the key mutations from Omicron and other circulating variants. Tests in rats showed that the designed mos-tri-RBD, whether used alone or as a booster shot, elicited potent cross-neutralizing antibodies against not only Omicron but also other immune-evasive variants. Neutralizing antibody ID50 titers induced by mos-tri-RBD were substantially higher than those elicited by homo-tri-RBD (containing homologous RBDs from prototype strain) or the BIBP inactivated COVID-19 vaccine (BBIBP-CorV). Our study indicates that mos-tri-RBD is highly immunogenic, which may serve as a broad-spectrum vaccine candidate in combating SARS-CoV-2 variants including Omicron.
Collapse
Affiliation(s)
- Jing Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zi Bo Han
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Liang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xue Feng Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Qin Jin
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
| | - Li Fang Du
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Shuai Shao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Hui Wang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Jun Wei Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ke Xu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Wenwen Lei
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ze Hua Lei
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zhao Ming Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Zhang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Ya Nan Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ning Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Fu Jie Shen
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Juan Wu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xiang Zheng
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Yu Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Wei Jin Huang
- National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Gui Zhen Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ji Guo Su
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Qi Ming Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| |
Collapse
|
22
|
Akbari E, Ajdary S, Ardakani EM, Agi E, Milani A, Seyedinkhorasani M, Khalaj V, Bolhassani A. Immunopotentiation by linking Hsp70 T-cell epitopes to Gag-Pol-Env-Nef-Rev multiepitope construct and increased IFN-gamma secretion in infected lymphocytes. Pathog Dis 2022; 80:6608937. [PMID: 35704612 DOI: 10.1093/femspd/ftac021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 11/12/2022] Open
Abstract
Therapeutic human immunodeficiency virus (HIV) vaccines can boost the anti-HIV host immunity to control viral replication and eliminate viral reservoirs in the absence of anti-retroviral therapy. In this study, two computationally designed multiepitope Gag-Pol-Env-Nef-Rev and Hsp70-Gag-Pol-Env-Nef-Rev constructs harboring immunogenic and highly conserved HIV T cell epitopes were generated in E. coli as polypeptide vaccine candidates. Furthermore, the multiepitope gag-pol-env-nef-rev and hsp70-gag-pol-env-nef-rev DNA vaccine constructs were prepared and complexed with MPG cell-penetrating peptide. The immunogenicity of the multiepitope constructs were evaluated using the homologous and heterologous prime/boost strategies in mice. Moreover, the secretion of IFN-γ was assessed in infected lymphocytes in vitro. Our data showed that the homologous polypeptide regimens could significantly induce a mixture of IgG1 and IgG2a antibody responses, activate T cells to secret IFN-γ, IL-5, IL-10, and generate Granzyme B. Moreover, IFN-γ secretion was significantly enhanced in single-cycle replicable (SCR) HIV-1 virions-infected splenocytes in these groups compared to uninfected splenocytes. The linkage of heat shock protein 70 (Hsp70) epitopes to Gag-Pol-Env-Nef-Rev polypeptide in the homologous regimen increased significantly cytokines and Granzyme B levels, and IFN-γ secretion in virions-infected splenocytes. Briefly, both designed constructs in the homologous regimens can be used as a promising vaccine candidate against HIV infection.
Collapse
Affiliation(s)
- Elahe Akbari
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.,Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Elnaz Agi
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Vahid Khalaj
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Sánchez-Martínez A, Acevedo-Sáenz L, Alzate-Ángel JC, Álvarez CM, Guzmán F, Roman T, Urcuqui-Inchima S, Cardona-Maya WD, Velilla PA. Functional Profile of CD8 + T-Cells in Response to HLA-A*02:01-Restricted Mutated Epitopes Derived from the Gag Protein of Circulating HIV-1 Strains from Medellín, Colombia. Front Immunol 2022; 13:793982. [PMID: 35392101 PMCID: PMC8980466 DOI: 10.3389/fimmu.2022.793982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
CD8+ T-cells play a crucial role in the control of HIV replication. HIV-specific CD8+ T-cell responses rapidly expand since the acute phase of the infection, and it has been observed that HIV controllers harbor CD8+ T-cells with potent anti-HIV capacity. The development of CD8+ T-cell-based vaccine against HIV-1 has focused on searching for immunodominant epitopes. However, the strong immune pressure of CD8+ T-cells causes the selection of viral variants with mutations in immunodominant epitopes. Since HIV-1 mutations are selected under the context of a specific HLA-I, the circulation of viral variants with these mutations is highly predictable based on the most prevalent HLA-I within a population. We previously demonstrated the adaptation of circulating strains of HIV-1 to the HLA-A*02 molecule by identifying mutations under positive selection located in GC9 and SL9 epitopes derived from the Gag protein. Also, we used an in silico prediction approach and evaluated whether the mutations found had a higher or lower affinity to the HLA-A*02. Although this strategy allowed predicting the interaction between mutated peptides and HLA-I, the functional response of CD8+ T-cells that these peptides induce is unknown. In the present work, peripheral blood mononuclear cells from 12 HIV-1+ HLA-A*02:01+ individuals were stimulated with the mutated and wild-type peptides derived from the GC9 and SL9 epitopes. The functional profile of CD8+ T-cells was evaluated using flow cytometry, and the frequency of subpopulations was determined according to their number of functions and the polyfunctionality index. The results suggest that the quality of the response (polyfunctionality) could be associated with the binding affinity of the peptide to the HLA molecule, and the functional profile of specific CD8+ T-cells to mutated epitopes in individuals under cART is maintained.
Collapse
Affiliation(s)
- Alexandra Sánchez-Martínez
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Liliana Acevedo-Sáenz
- Grupo Cuidado Enfermería CES, Facultad de Enfermería, Universidad CES, Medellín, Colombia
| | - Juan Carlos Alzate-Ángel
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia.,Unidad de Micología Médica y Experimental, Corporación para Investigaciones Biológicas, Medellín, Universidad de Santander (CIB-UDES), Bucaramanga, Colombia
| | - Cristian M Álvarez
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Fanny Guzmán
- Núcleo de Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Tanya Roman
- Núcleo de Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Walter D Cardona-Maya
- Grupo Reproducción, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Paula Andrea Velilla
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
24
|
Bouza E, Arribas JR, Alejos B, Bernardino JI, Coiras M, Coll P, Del Romero J, Fuster MJ, Górgolas M, Gutiérrez A, Gracia D, Hernando V, Martínez-Picado J, Martínez Sesmero JM, Martínez E, Moreno S, Mothe B, Navarro ML, Podzamczer D, Pulido F, Ramos JT, Ruiz-Mateos E, Suárez García I, Palomo E. Past and future of HIV infection. A document based on expert opinion. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2022; 35:131-156. [PMID: 35018404 PMCID: PMC8972691 DOI: 10.37201/req/083.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/02/2021] [Indexed: 11/15/2022]
Abstract
HIV infection is now almost 40 years old. In this time, along with the catastrophe and tragedy that it has entailed, it has also represented the capacity of modern society to take on a challenge of this magnitude and to transform an almost uniformly lethal disease into a chronic illness, compatible with a practically normal personal and relationship life. This anniversary seemed an ideal moment to pause and reflect on the future of HIV infection, the challenges that remain to be addressed and the prospects for the immediate future. This reflection has to go beyond merely technical approaches, by specialized professionals, to also address social and ethical aspects. For this reason, the Health Sciences Foundation convened a group of experts in different aspects of this disease to discuss a series of questions that seemed pertinent to all those present. Each question was presented by one of the participants and discussed by the group. The document we offer is the result of this reflection.
Collapse
Affiliation(s)
- E Bouza
- Servicio de Microbiología Clínica y Enfermedades Infecciosas del Hospital General Universitario Gregorio Marañón, Universidad Complutense. CIBERES. Ciber de Enfermedades Respiratorias. Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Affiliation(s)
- Paul Munson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
26
|
Lewitus E, Townsley SM, Li Y, Donofrio GC, Dearlove BL, Bai H, Sanders-Buell E, O’Sullivan AM, Bose M, Kibuuka H, Maganga L, Nitayaphan S, Sawe FK, Eller LA, Michael NL, Polonis VR, Ake JA, Vasan S, Robb ML, Tovanabutra S, Krebs SJ, Rolland M. HIV-1 infections with multiple founders associate with the development of neutralization breadth. PLoS Pathog 2022; 18:e1010369. [PMID: 35303045 PMCID: PMC8967031 DOI: 10.1371/journal.ppat.1010369] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/30/2022] [Accepted: 02/16/2022] [Indexed: 12/21/2022] Open
Abstract
Eliciting broadly neutralizing antibodies (bnAbs) is a cornerstone of HIV-1 vaccine strategies. Comparing HIV-1 envelope (env) sequences from the first weeks of infection to the breadth of antibody responses observed several years after infection can help define viral features critical to vaccine design. We investigated the relationship between HIV-1 env genetics and the development of neutralization breadth in 70 individuals enrolled in a prospective acute HIV-1 cohort. Half of the individuals who developed bnAbs were infected with multiple HIV-1 founder variants, whereas all individuals with limited neutralization breadth had been infected with single HIV-1 founders. Accordingly, at HIV-1 diagnosis, env diversity was significantly higher in participants who later developed bnAbs compared to those with limited breadth (p = 0.012). This association between founder multiplicity and the subsequent development of neutralization breadth was also observed in 56 placebo recipients in the RV144 vaccine efficacy trial. In addition, we found no evidence that neutralization breath was heritable when analyzing env sequences from the 126 participants. These results demonstrate that the presence of slightly different HIV-1 variants in acute infection could promote the induction of bnAbs, suggesting a novel vaccine strategy, whereby an initial immunization with a cocktail of minimally distant antigens would be able to initiate bnAb development towards breadth. Vaccines against viral pathogens protect through the induction of broadly neutralizing antibodies (bnAbs). No HIV-1 vaccine has successfully elicited bnAbs, and a successful HIV-1 vaccine will need to accelerate the process of development of a broadly neutralizing response that typically takes a couple of years to develop in natural infection. We studied diversity in the HIV-1 envelope gene from initial infection to several years out in 126 individuals from two cohorts. We showed that the development of bnAbs at 2–3 years was not due to transmissible viral genetics, but rather associated with diversity during the first month of infection. We propose that designing a vaccine that mimics an infection with multiple, minimally distant founder variants may successfully elicit the development of bnAbs and provide effective prophylaxis against HIV-1.
Collapse
Affiliation(s)
- Eric Lewitus
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Samantha M. Townsley
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Yifan Li
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Gina C. Donofrio
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Bethany L. Dearlove
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Hongjun Bai
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Eric Sanders-Buell
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Anne Marie O’Sullivan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Meera Bose
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Hannah Kibuuka
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Lucas Maganga
- National Institute for Medical Research-Mbeya Medical Research Center, Mbeya, Tanzania
| | | | - Fredrick K. Sawe
- Kenya Medical Research Institute/U.S. Army Medical Research Directorate-Africa/Kenya-Henry Jackson Foundation MRI, Kericho, Kenya
| | - Leigh Anne Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Nelson L. Michael
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Victoria R. Polonis
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Julie A. Ake
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Sandhya Vasan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Merlin L. Robb
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Sodsai Tovanabutra
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Shelly J. Krebs
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Morgane Rolland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
27
|
Enhanced Cross-Reactive and Polyfunctional Effector-Memory T Cell Responses by ICVAX-a Human PD1-Based Bivalent HIV-1 Gag-p41 Mosaic DNA Vaccine. J Virol 2022; 96:e0216121. [PMID: 35297660 PMCID: PMC9006887 DOI: 10.1128/jvi.02161-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vaccine-induced protective T cell immunity is necessary for HIV-1 functional cure. We previously reported that rhesus PD1-Gag-based DNA vaccination sustained simian-human immunodeficiency virus (SHIV) suppression by inducing effector-memory CD8+ T cells. Here, we investigated a human PD1-Gag-based DNA vaccine, namely, ICVAX, for clinical translation. PD1-based dendritic cell targeting and mosaic antigenic designs were combined to generate the ICVAX by fusing the human soluble PD1 domain with a bivalent HIV-1 Gag-p41 mosaic antigen. The mosaic antigen was cross-reactive with patients infected with B, CRF07/08_BC, and CRF01_AE variants. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses than mosaic Gag-p41 alone, and suppressed EcoHIV infection more efficiently. In macaques, ICVAX elicited polyfunctional effector-memory T cell responses that targeted multiple nonoverlapping epitopes of the Gag-p41 antigen. Furthermore, ICVAX manufactured following good manufacturing practices proved potent immunogenicity in macaques after biannual homologous vaccination, warranting clinical evaluation of ICVAX as an immunotherapy against HIV-1. IMPORTANCE This study presents that ICVAX, a PD1-based DNA vaccine against HIV-1, could induce broad and polyfunctional T cell responses against different HIV-1 subtypes. ICVAX encodes a recombinant antigen consisting of the human soluble PD1 domain fused with two mosaic Gag-p41 antigens. The mosaic antigens cover more than 500 HIV-1 strains circulating in China including the subtypes B/B’, CRF01_AE, and CRF07/08_BC. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses, with better EcoHIV suppression than the nontargeting mosaic Gag-p41 DNA vaccine. Moreover, both lab-generated and GMP-grade ICVAX also elicited strong polyfunctional effector-memory T cell responses in rhesus macaques with good immunogenicity against multiple nonoverlapping epitopes of the Gag-p41 antigen. This study therefore highlights the great potential to translate the PD1-based DNA vaccine approach into clinical use, and opens up new avenues for alternative HIV-1 vaccine design for HIV-1 preventive and functional cure.
Collapse
|
28
|
van Heuvel Y, Schatz S, Rosengarten JF, Stitz J. Infectious RNA: Human Immunodeficiency Virus (HIV) Biology, Therapeutic Intervention, and the Quest for a Vaccine. Toxins (Basel) 2022; 14:toxins14020138. [PMID: 35202165 PMCID: PMC8876946 DOI: 10.3390/toxins14020138] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022] Open
Abstract
Different mechanisms mediate the toxicity of RNA. Genomic retroviral mRNA hijacks infected host cell factors to enable virus replication. The viral genomic RNA of the human immunodeficiency virus (HIV) encompasses nine genes encoding in less than 10 kb all proteins needed for replication in susceptible host cells. To do so, the genomic RNA undergoes complex alternative splicing to facilitate the synthesis of the structural, accessory, and regulatory proteins. However, HIV strongly relies on the host cell machinery recruiting cellular factors to complete its replication cycle. Antiretroviral therapy (ART) targets different steps in the cycle, preventing disease progression to the acquired immunodeficiency syndrome (AIDS). The comprehension of the host immune system interaction with the virus has fostered the development of a variety of vaccine platforms. Despite encouraging provisional results in vaccine trials, no effective vaccine has been developed, yet. However, novel promising vaccine platforms are currently under investigation.
Collapse
Affiliation(s)
- Yasemin van Heuvel
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Germany
| | - Stefanie Schatz
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Germany
| | - Jamila Franca Rosengarten
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Germany
| | - Jörn Stitz
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Correspondence:
| |
Collapse
|
29
|
Rosengarten JF, Schatz S, Wolf T, Barbe S, Stitz J. Components of a HIV-1 vaccine mediate virus-like particle (VLP)-formation and display of envelope proteins exposing broadly neutralizing epitopes. Virology 2022; 568:41-48. [DOI: 10.1016/j.virol.2022.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
|
30
|
Swanson PA, Padilla M, Hoyland W, McGlinchey K, Fields PA, Bibi S, Faust SN, McDermott AB, Lambe T, Pollard AJ, Durham NM, Kelly EJ. AZD1222/ChAdOx1 nCoV-19 vaccination induces a polyfunctional spike protein-specific T H1 response with a diverse TCR repertoire. Sci Transl Med 2021; 13:eabj7211. [PMID: 34591596 PMCID: PMC9924073 DOI: 10.1126/scitranslmed.abj7211] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/20/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
AZD1222 (ChAdOx1 nCoV-19), a replication-deficient simian adenovirus–vectored vaccine, has demonstrated safety, efficacy, and immunogenicity against coronavirus disease 2019 in clinical trials and real-world studies. We characterized CD4+ and CD8+ T cell responses induced by AZD1222 vaccination in peripheral blood mononuclear cells from 296 unique vaccine recipients aged 18 to 85 years who enrolled in the phase 2/3 COV002 trial. Total spike protein–specific CD4+ T cell helper type 1 (TH1) and CD8+ T cell responses were increased in AZD1222-vaccinated adults of all ages after two doses of AZD1222. CD4+ TH2 responses after AZD1222 vaccination were not detected. Furthermore, AZD1222-specific TH1 and CD8+ T cells both displayed a high degree of polyfunctionality in all adult age groups. T cell receptor β (TCRβ) sequences from vaccinated participants mapped against TCR sequences known to react to SARS-CoV-2 revealed substantial breadth and depth across the SARS-CoV-2 spike protein for both AZD1222-induced CD4+ and CD8+ T cell responses. Overall, AZD1222 vaccination induced a polyfunctional TH1-dominated T cell response, with broad CD4+ and CD8+ T cell coverage across the SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
- Phillip A. Swanson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcelino Padilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wesley Hoyland
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly McGlinchey
- Discovery, Research and Early Development, Oncology R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | | | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and NIHR Oxford Biomedical Research Centre, Oxford OX4 6PG, UK
| | - Saul N. Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Adrian B. McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7FZ, UK
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and NIHR Oxford Biomedical Research Centre, Oxford OX4 6PG, UK
| | - Nicholas M. Durham
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Elizabeth J. Kelly
- Translational Medicine, Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | | |
Collapse
|
31
|
Kim J, Vasan S, Kim JH, Ake JA. Current approaches to HIV vaccine development: a narrative review. J Int AIDS Soc 2021; 24 Suppl 7:e25793. [PMID: 34806296 PMCID: PMC8606871 DOI: 10.1002/jia2.25793] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The development of an effective vaccine to protect against HIV is a longstanding global health need complicated by challenges inherent to HIV biology and to the execution of vaccine efficacy testing in the context of evolving biomedical prevention interventions. This review describes lessons learnt from previous efficacy trials, highlights unanswered questions, and surveys new approaches in vaccine development addressing these gaps. METHODS We conducted a targeted peer-reviewed literature search of articles and conference abstracts from 1989 through 2021 for HIV vaccine studies and clinical trials. The US National Library of Medicine's Clinical Trials database was accessed to further identify clinical trials involving HIV vaccines. The content of the review was also informed by the authors' own experience and engagement with collaborators in HIV vaccine research. DISCUSSION The HIV vaccine field has successfully developed multiple vaccine platforms through advanced clinical studies; however, the modest efficacy signal of the RV144 Thai trial remains the only demonstration of HIV vaccine protection in humans. Current vaccine strategies include prime-boost strategies to improve elicitation of immune correlates derived from RV144, combination mosaic antigens, novel viral vectors, antigens designed to elicit broadly neutralizing antibody, new nucleic acid platforms and potent adjuvants to enhance immunogenicity across multiple classes of emerging vaccine candidates. CONCLUSIONS HIV vaccine developers have applied lessons learnt from previous successes and failures to innovative vaccine design approaches. These strategies have yielded novel mosaic antigen constructs now in efficacy testing, produced a diverse pipeline of early-stage immunogens and novel adjuvants, and advanced the field towards a globally effective HIV vaccine.
Collapse
Affiliation(s)
- Jiae Kim
- US Military HIV Research ProgramWalter Reed Army Institute of ResearchSilver SpringMarylandUSA
- Henry M. Jackson Foundation for the Advancement of Military MedicineBethesdaMarylandUSA
| | - Sandhya Vasan
- US Military HIV Research ProgramWalter Reed Army Institute of ResearchSilver SpringMarylandUSA
- Henry M. Jackson Foundation for the Advancement of Military MedicineBethesdaMarylandUSA
| | | | - Julie A. Ake
- US Military HIV Research ProgramWalter Reed Army Institute of ResearchSilver SpringMarylandUSA
| |
Collapse
|
32
|
Ensoli B, Moretti S, Borsetti A, Maggiorella MT, Buttò S, Picconi O, Tripiciano A, Sgadari C, Monini P, Cafaro A. New insights into pathogenesis point to HIV-1 Tat as a key vaccine target. Arch Virol 2021; 166:2955-2974. [PMID: 34390393 PMCID: PMC8363864 DOI: 10.1007/s00705-021-05158-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
Despite over 30 years of enormous effort and progress in the field, no preventative and/or therapeutic vaccines against human immunodeficiency virus (HIV) are available. Here, we briefly summarize the vaccine strategies and vaccine candidates that in recent years advanced to efficacy trials with mostly unsatisfactory results. Next, we discuss a novel and somewhat contrarian approach based on biological and epidemiological evidence, which led us to choose the HIV protein Tat for the development of preventive and therapeutic HIV vaccines. Toward this goal, we review here the role of Tat in the virus life cycle as well as experimental and epidemiological evidence supporting its key role in the natural history of HIV infection and comorbidities. We then discuss the preclinical and clinical development of a Tat therapeutic vaccine, which, by improving the functionality and homeostasis of the immune system and by reducing the viral reservoir in virologically suppressed vaccinees, helps to establish key determinants for intensification of combination antiretroviral therapy (cART) and a functional cure. Future developments and potential applications of the Tat therapeutic vaccine are also discussed, as well as the rationale for its use in preventative strategies. We hope this contribution will lead to a reconsideration of the current paradigms for the development of HIV/AIDS vaccines, with a focus on targeting of viral proteins with key roles in HIV pathogenesis.
Collapse
Affiliation(s)
- Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Teresa Maggiorella
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Stefano Buttò
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonella Tripiciano
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
33
|
Elangovan R, Jenks M, Yun J, Dickson-Tetteh L, Kirtley S, Hemelaar J. Global and Regional Estimates for Subtype-Specific Therapeutic and Prophylactic HIV-1 Vaccines: A Modeling Study. Front Microbiol 2021; 12:690647. [PMID: 34335516 PMCID: PMC8320730 DOI: 10.3389/fmicb.2021.690647] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/27/2021] [Indexed: 11/18/2022] Open
Abstract
Global HIV-1 genetic diversity forms a major obstacle to the development of an HIV vaccine. It may be necessary to employ subtype-specific HIV-1 vaccines in individual countries according to their HIV-1 subtype distribution. We estimated the global and regional need for subtype-specific HIV-1 vaccines. We took into account the proportions of different HIV-1 variants circulating in each country, the genetic composition of HIV-1 recombinants, and the different genome segments (gag, pol, env) that may be incorporated into vaccines. We modeled different scenarios according to whether countries would employ subtype-specific HIV-1 vaccines against (1) the most common subtype; (2) subtypes contributing more than 5% of HIV infections; or (3) all circulating subtypes. For therapeutic vaccines targeting the most common HIV-1 subtype in each country, 16.5 million doses of subtype C vaccine were estimated globally, followed by subtypes A (14.3 million) and B (4.2 million). A vaccine based on env required 2.6 million subtype E doses, and a vaccine based on pol required 4.8 million subtype G doses. For prophylactic vaccines targeting the most common HIV-1 subtype in each country, 1.9 billion doses of subtype A vaccine were estimated globally, followed by subtype C (1.1 billion) and subtype B (1.0 billion). A vaccine based on env required 1.2 billion subtype E doses, and a vaccine based on pol required 0.3 billion subtype G doses. If subtype-specific HIV-1 vaccines are also directed against less common subtypes in each country, vaccines targeting subtypes D, F, H, and K are also needed and would require up to five times more vaccine doses in total. We conclude that to provide global coverage, subtype-specific HIV-1 vaccines need to be directed against subtypes A, B, and C. Vaccines targeting env also need to include subtype E and those targeting pol need to include subtype G.
Collapse
Affiliation(s)
| | - Michael Jenks
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Jason Yun
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Leslie Dickson-Tetteh
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Shona Kirtley
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Centre for Statistics in Medicine, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Joris Hemelaar
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
34
|
Fries CN, Chen JL, Dennis ML, Votaw NL, Eudailey J, Watts BE, Hainline KM, Cain DW, Barfield R, Chan C, Moody MA, Haynes BF, Saunders KO, Permar SR, Fouda GG, Collier JH. HIV envelope antigen valency on peptide nanofibers modulates antibody magnitude and binding breadth. Sci Rep 2021; 11:14494. [PMID: 34262096 PMCID: PMC8280189 DOI: 10.1038/s41598-021-93702-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/23/2021] [Indexed: 01/02/2023] Open
Abstract
A major challenge in developing an effective vaccine against HIV-1 is the genetic diversity of its viral envelope. Because of the broad range of sequences exhibited by HIV-1 strains, protective antibodies must be able to bind and neutralize a widely mutated viral envelope protein. No vaccine has yet been designed which induces broadly neutralizing or protective immune responses against HIV in humans. Nanomaterial-based vaccines have shown the ability to generate antibody and cellular immune responses of increased breadth and neutralization potency. Thus, we have developed supramolecular nanofiber-based immunogens bearing the HIV gp120 envelope glycoprotein. These immunogens generated antibody responses that had increased magnitude and binding breadth compared to soluble gp120. By varying gp120 density on nanofibers, we determined that increased antigen valency was associated with increased antibody magnitude and germinal center responses. This study presents a proof-of-concept for a nanofiber vaccine platform generating broad, high binding antibody responses against the HIV-1 envelope glycoprotein.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maria L Dennis
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Nicole L Votaw
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Joshua Eudailey
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brian E Watts
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin O Saunders
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, New York-Presbyterian/Weill Cornell Medicine, New York, NY, 10065, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
35
|
Where to Next? Research Directions after the First Hepatitis C Vaccine Efficacy Trial. Viruses 2021; 13:v13071351. [PMID: 34372558 PMCID: PMC8310243 DOI: 10.3390/v13071351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/03/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
Thirty years after its discovery, the hepatitis C virus (HCV) remains a leading cause of liver disease worldwide. Given that many countries continue to experience high rates of transmission despite the availability of potent antiviral therapies, an effective vaccine is seen as critical for the elimination of HCV. The recent failure of the first vaccine efficacy trial for the prevention of chronic HCV confirmed suspicions that this virus will be a challenging vaccine target. Here, we examine the published data from this first efficacy trial along with the earlier clinical and pre-clinical studies of the vaccine candidate and then discuss three key research directions expected to be important in ongoing and future HCV vaccine development. These include the following: 1. design of novel immunogens that generate immune responses to genetically diverse HCV genotypes and subtypes, 2. strategies to elicit broadly neutralizing antibodies against envelope glycoproteins in addition to cytotoxic and helper T cell responses, and 3. consideration of the unique immunological status of individuals most at risk for HCV infection, including those who inject drugs, in vaccine platform development and early immunogenicity trials.
Collapse
|
36
|
Hauser A, Carnell G, Held K, Sulbaran G, Tischbierek N, Rogers L, Pollakis G, Tonks P, Hoelscher M, Ding S, Sanders RW, Geldmacher C, Sattentau Q, Weissenhorn W, Heeney JL, Peterhoff D, Wagner R. Stepwise Conformational Stabilization of a HIV-1 Clade C Consensus Envelope Trimer Immunogen Impacts the Profile of Vaccine-Induced Antibody Responses. Vaccines (Basel) 2021; 9:750. [PMID: 34358165 PMCID: PMC8310183 DOI: 10.3390/vaccines9070750] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 01/16/2023] Open
Abstract
Stabilization of the HIV-1 Envelope glycoprotein trimer (Env) in its native pre-fusion closed conformation is regarded as one of several requirements for the induction of neutralizing antibody (nAb) responses, which, in turn, will most likely be a prerequisite for the development of an efficacious preventive vaccine. Here, we systematically analyzed how the stepwise stabilization of a clade C consensus (ConC) Env immunogen impacts biochemical and biophysical protein traits such as antigenicity, thermal stability, structural integrity, and particle size distribution. The increasing degree of conformational rigidification positively correlates with favorable protein characteristics, leading to optimized homogeneity of the protein preparations, increased thermal stability, and an overall favorable binding profile of structure-dependent broadly neutralizing antibodies (bnAbs) and non-neutralizing antibodies (non-nAbs). We confirmed that increasing the structural integrity and stability of the Env trimers positively correlates with the quality of induced antibody responses by the immunogens. These and other data contribute to the selection of ConCv5 KIKO as novel Env immunogens for use within the European Union's H2020 Research Consortium EHVA (European HIV Alliance) for further preclinical analysis and phase 1 clinical development.
Collapse
Affiliation(s)
- Alexandra Hauser
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (A.H.); (N.T.); (D.P.)
| | - George Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK; (G.C.); (P.T.); (J.L.H.)
| | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (L.R.); (M.H.); (C.G.)
- German Center for Infection Research (DZIF), Partner site Munich, 80802 Munich, Germany
| | - Guidenn Sulbaran
- Institut de Biologie Structurale (IBS), University Grenoble Alpes, CEA, CNRS, 38000 Grenoble, France; (G.S.); (W.W.)
| | - Nadine Tischbierek
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (A.H.); (N.T.); (D.P.)
| | - Lisa Rogers
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (L.R.); (M.H.); (C.G.)
- German Center for Infection Research (DZIF), Partner site Munich, 80802 Munich, Germany
| | - Georgios Pollakis
- Department of Clinical Infection, Microbiology, and Immunology (CIMI), University of Liverpool, Liverpool L69 7BE, UK;
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections (HPRU EZI), Liverpool L69 7BE, UK
| | - Paul Tonks
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK; (G.C.); (P.T.); (J.L.H.)
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (L.R.); (M.H.); (C.G.)
- German Center for Infection Research (DZIF), Partner site Munich, 80802 Munich, Germany
| | - Song Ding
- EuroVacc Foundation, 1105 BP Amsterdam, The Netherlands;
| | - Rogier W. Sanders
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (L.R.); (M.H.); (C.G.)
- German Center for Infection Research (DZIF), Partner site Munich, 80802 Munich, Germany
| | - Quentin Sattentau
- The Sir Willian Dunn School of Pathology, The University of Oxford, Oxford OX1 3RE, UK;
| | - Winfried Weissenhorn
- Institut de Biologie Structurale (IBS), University Grenoble Alpes, CEA, CNRS, 38000 Grenoble, France; (G.S.); (W.W.)
| | - Jonathan L. Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK; (G.C.); (P.T.); (J.L.H.)
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (A.H.); (N.T.); (D.P.)
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, 93053 Regensburg, Germany; (A.H.); (N.T.); (D.P.)
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
37
|
Lee SH, Beck BR, Hwang SH, Song SK. Feeding olive flounder (Paralichthys olivaceus) with Lactococcus lactis BFE920 expressing the fusion antigen of Vibrio OmpK and FlaB provides protection against multiple Vibrio pathogens: A universal vaccine effect. FISH & SHELLFISH IMMUNOLOGY 2021; 114:253-262. [PMID: 33979691 DOI: 10.1016/j.fsi.2021.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 06/12/2023]
Abstract
Vibriosis, an illness caused by the Vibrio bacteria species, results in significant economic loss in olive flounder farms. Here we present a novel anti-Vibrio feed vaccine protecting multiple strains of Vibrio pathogens, a universal vaccine effect. The vaccine was generated by engineering Lactococcus lactis BFE920 to express the fusion antigens of Vibrio outer membrane protein K (OmpK) and flagellin B subunit (FlaB). These antigen genes are highly conserved among Vibrio species. Olive flounder (7.1 ± 0.8 g and 140 ± 10 g) were fed the vaccine adsorbed to a regular feed (1 × 107 CFU/g) for one week with a 1-week interval, repeating three times (a triple boost). The vaccinated fish increased the significant levels of antigen-specific antibodies, T cell numbers (CD4-1, CD4-2, and CD8α), cytokine production (T-bet and IFN-γ), and innate immune responses (TLR5M, IL-1β, and IL-12p40). Also, the survival rates of adult and juvenile fish fed the vaccine were significantly elevated when challenged with V. anguillarum, V. alginolyticus, and V. harveyi. In addition, weight gain rate and feed conversion ratio were improved in vaccinated fish. The feed vaccine protected multiple Vibrio pathogens, a universal vaccine effect, by activating innate and adaptive immune responses. This oral vaccine may be developed as an anti-Vibrio vaccine to protect against a broad spectrum of Vibrio pathogens.
Collapse
Affiliation(s)
- Soon Ho Lee
- School of Life Science, Handong University, 558 Handong-ro, Pohang-city, Gyeongbuk, 37554, South Korea
| | - Bo Ram Beck
- School of Life Science, Handong University, 558 Handong-ro, Pohang-city, Gyeongbuk, 37554, South Korea
| | - Seok-Hong Hwang
- School of Life Science, Handong University, 558 Handong-ro, Pohang-city, Gyeongbuk, 37554, South Korea
| | - Seong Kyu Song
- School of Life Science, Handong University, 558 Handong-ro, Pohang-city, Gyeongbuk, 37554, South Korea.
| |
Collapse
|
38
|
Karch CP, Matyas GR. The current and future role of nanovaccines in HIV-1 vaccine development. Expert Rev Vaccines 2021; 20:935-944. [PMID: 34184607 DOI: 10.1080/14760584.2021.1945448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: An efficacious vaccine for HIV-1 has been sought for over 30 years to eliminate the virus from the human population. Many challenges have occurred in the attempt to produce a successful immunogen, mainly caused by the basic biology of the virus. Immunogens have been developed focusing on inducing one or more of the following types of immune responses; neutralizing antibodies, non-neutralizing antibodies, and T-cell mediated responses. One way to better present and develop an immunogen for HIV-1 is through the use of nanotechnology and nanoparticles.Areas covered: This article gives a basic overview of the HIV-1 vaccine field, as well as nanotechnology, specifically nanovaccines. It then covers the application of nanovaccines made from biological macromolecules to HIV-1 vaccine development for neutralizing antibodies, non-neutralizing antibodies, and T-cell-mediated responses.Expert opinion: Nanovaccines are an area that is ripe for further exploration in HIV-1 vaccine field. Not only are nanovaccines capable of carrying and presenting antigens in native-like conformations, but they have also repeatedly been shown to increase immunogenicity over recombinant antigens alone. Only through further research can the true role of nanovaccines in the development of an efficacious HIV-1 vaccine be established.
Collapse
Affiliation(s)
- Christopher P Karch
- Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Laboratory of Adjuvant and Antigen Research, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
39
|
Adenovirus-vectored vaccine containing multidimensionally conserved parts of the HIV proteome is immunogenic in rhesus macaques. Proc Natl Acad Sci U S A 2021; 118:2022496118. [PMID: 33514660 DOI: 10.1073/pnas.2022496118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
An effective vaccine that can protect against HIV infection does not exist. A major reason why a vaccine is not available is the high mutability of the virus, which enables it to evolve mutations that can evade human immune responses. This challenge is exacerbated by the ability of the virus to evolve compensatory mutations that can partially restore the fitness cost of immune-evading mutations. Based on the fitness landscapes of HIV proteins that account for the effects of coupled mutations, we designed a single long peptide immunogen comprising parts of the HIV proteome wherein mutations are likely to be deleterious regardless of the sequence of the rest of the viral protein. This immunogen was then stably expressed in adenovirus vectors that are currently in clinical development. Macaques immunized with these vaccine constructs exhibited T-cell responses that were comparable in magnitude to animals immunized with adenovirus vectors with whole HIV protein inserts. Moreover, the T-cell responses in immunized macaques strongly targeted regions contained in our immunogen. These results suggest that further studies aimed toward using our vaccine construct for HIV prophylaxis and cure are warranted.
Collapse
|
40
|
Wong YC, Liu W, Yim LY, Li X, Wang H, Yue M, Niu M, Cheng L, Ling L, Du Y, Chen SMY, Cheung KW, Wang H, Tang X, Tang J, Zhang H, Song Y, Chakrabarti LA, Chen Z. Sustained viremia suppression by SHIVSF162P3CN-recalled effector-memory CD8+ T cells after PD1-based vaccination. PLoS Pathog 2021; 17:e1009647. [PMID: 34125864 PMCID: PMC8202916 DOI: 10.1371/journal.ppat.1009647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
HIV-1 functional cure requires sustained viral suppression without antiretroviral therapy. While effector-memory CD8+ T lymphocytes are essential for viremia control, few vaccines elicit such cellular immunity that could be potently recalled upon viral infection. Here, we investigated a program death-1 (PD1)-based vaccine by fusion of simian immunodeficiency virus capsid antigen to soluble PD1. Homologous vaccinations suppressed setpoint viremia to undetectable levels in vaccinated macaques following a high-dose intravenous challenge by the pathogenic SHIVSF162P3CN. Poly-functional effector-memory CD8+ T cells were not only induced after vaccination, but were also recalled upon viral challenge for viremia control as determined by CD8 depletion. Vaccine-induced effector memory CD8+ subsets displayed high cytotoxicity-related genes by single-cell analysis. Vaccinees with sustained viremia suppression for over two years responded to boost vaccination without viral rebound. These results demonstrated that PD1-based vaccine-induced effector-memory CD8+ T cells were recalled by AIDS virus infection, providing a potential immunotherapy for functional cure. HIV-1/AIDS remains a major global pandemic although treatment regimen has improved. Identifying efficacious vaccines and therapeutics to achieve long-term viral control with very low/undetectable plasma viral loads in the absence of antiretroviral therapy, a status known as functional cure, would be highly beneficial. We previously demonstrated that antigens fused to a soluble program death-1 (PD1) domain could effectively bind and be cross-presented by dendritic cells that constitutively expressed PD1 ligands. When applied in the form of DNA vaccination, this antigen-targeting strategy was highly immunogenic in mice. Here, we investigated the efficacy of the PD1-based DNA vaccine approach against pathogenic simian-human immunodeficiency virus challenge in rhesus monkeys. Our results showed that homologous PD1-based DNA vaccinations induced highly functional effector-memory CD8+ T cells carrying a unique cytotoxicity gene expression profile. These T cells actively supressed viremia in monkeys and were re-activated via boost vaccination at 2 years after viral challenge without viral rebound. In summary, our study demonstrates the potential application of PD1-based DNA vaccination to control AIDS virus infection.
Collapse
Affiliation(s)
- Yik Chun Wong
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Wan Liu
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Lok Yan Yim
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Xin Li
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Department of Veterinary Medicine, Foshan University, Foshan, China
| | - Hui Wang
- HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Ming Yue
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Mengyue Niu
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Lin Cheng
- HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Lijun Ling
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Yanhua Du
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Samantha M. Y. Chen
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Ka-Wai Cheung
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Haibo Wang
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Xian Tang
- HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
- Virus and Immunity Unit, Pasteur Institute, Paris, France; INSERM U1108, Paris, France
| | - Jiansong Tang
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Haoji Zhang
- Department of Veterinary Medicine, Foshan University, Foshan, China
| | - Youqiang Song
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Lisa A. Chakrabarti
- Virus and Immunity Unit, Pasteur Institute, Paris, France; INSERM U1108, Paris, France
| | - Zhiwei Chen
- AIDS Institute, Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
- * E-mail:
| |
Collapse
|
41
|
Wee EG, Moyo N, Hannoun Z, Giorgi EE, Korber B, Hanke T. Effect of epitope variant co-delivery on the depth of CD8 T cell responses induced by HIV-1 conserved mosaic vaccines. Mol Ther Methods Clin Dev 2021; 21:741-753. [PMID: 34169114 PMCID: PMC8187930 DOI: 10.1016/j.omtm.2021.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/29/2021] [Indexed: 11/27/2022]
Abstract
To stop the HIV-1 pandemic, vaccines must induce responses capable of controlling vast HIV-1 variants circulating in the population as well as those evolved in each individual following transmission. Numerous strategies have been proposed, of which the most promising include focusing responses on the vulnerable sites of HIV-1 displaying the least entropy among global isolates and using algorithms that maximize vaccine match to circulating HIV-1 variants by vaccine cocktails of optimized complementing sequences. In this study, we investigated CD8 T cell responses induced by a bi-valent mosaic of highly conserved HIVconsvX regions delivered by a combination of simian adenovirus ChAdOx1 and poxvirus MVA. We compared partially and fully mono- and bi-valent prime-boost regimens and their ability to elicit T cells recognizing natural epitope variants using an interferon-γ enzyme-linked immunospot (ELISPOT) assay. We used 11 well-defined CD8 T cell epitopes in two mouse haplotypes and, for each epitope, assessed recognition of the two vaccine forms together with the other most frequent epitope variants in the HIV-1 database. We conclude that for the magnitude and depth of epitope recognition, CD8 T cell responses benefitted in most comparisons from the combined bi-valent mosaic and envisage the main advantage of the bi-valent vaccine during its deployment to diverse populations.
Collapse
Affiliation(s)
- Edmund G. Wee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Nathifa Moyo
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Zara Hannoun
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Bette Korber
- Los Alamos National Laboratory, Los Alamos, NM, USA
- New Mexico Consortium, Los Alamos, NM, USA
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
42
|
Boniolo F, Dorigatti E, Ohnmacht AJ, Saur D, Schubert B, Menden MP. Artificial intelligence in early drug discovery enabling precision medicine. Expert Opin Drug Discov 2021; 16:991-1007. [PMID: 34075855 DOI: 10.1080/17460441.2021.1918096] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Precision medicine is the concept of treating diseases based on environmental factors, lifestyles, and molecular profiles of patients. This approach has been found to increase success rates of clinical trials and accelerate drug approvals. However, current precision medicine applications in early drug discovery use only a handful of molecular biomarkers to make decisions, whilst clinics gear up to capture the full molecular landscape of patients in the near future. This deep multi-omics characterization demands new analysis strategies to identify appropriate treatment regimens, which we envision will be pioneered by artificial intelligence.Areas covered: In this review, the authors discuss the current state of drug discovery in precision medicine and present our vision of how artificial intelligence will impact biomarker discovery and drug design.Expert opinion: Precision medicine is expected to revolutionize modern medicine; however, its traditional form is focusing on a few biomarkers, thus not equipped to leverage the full power of molecular landscapes. For learning how the development of drugs can be tailored to the heterogeneity of patients across their molecular profiles, artificial intelligence algorithms are the next frontier in precision medicine and will enable a fully personalized approach in drug design, and thus ultimately impacting clinical practice.
Collapse
Affiliation(s)
- Fabio Boniolo
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,School of Medicine, Chair of Translational Cancer Research and Institute for Experimental Cancer Therapy, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Emilio Dorigatti
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Statistical Learning and Data Science, Department of Statistics, Ludwig Maximilian Universität München, Munich, Germany
| | - Alexander J Ohnmacht
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Dieter Saur
- School of Medicine, Chair of Translational Cancer Research and Institute for Experimental Cancer Therapy, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Benjamin Schubert
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Mathematics, Technical University of Munich, Garching, Germany
| | - Michael P Menden
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany.,German Centre for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
43
|
Expression of a Novel HIV-1 Gag-Pol-Env-Nef-Rev Multi-Epitope Construct in Escherichia coli. JOURNAL OF MEDICAL MICROBIOLOGY AND INFECTIOUS DISEASES 2021. [DOI: 10.52547/jommid.9.2.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
44
|
Nagashima KA, Mousa JJ. Next-Generation Influenza HA Immunogens and Adjuvants in Pursuit of a Broadly Protective Vaccine. Viruses 2021; 13:v13040546. [PMID: 33805245 PMCID: PMC8064354 DOI: 10.3390/v13040546] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/20/2022] Open
Abstract
Influenza virus, a highly mutable respiratory pathogen, causes significant disease nearly every year. Current vaccines are designed to protect against circulating influenza strains of a given season. However, mismatches between vaccine strains and circulating strains, as well as inferior vaccine effectiveness in immunodeficient populations, represent major obstacles. In an effort to expand the breadth of protection elicited by influenza vaccination, one of the major surface glycoproteins, hemagglutinin (HA), has been modified to develop immunogens that display conserved regions from multiple viruses or elicit a highly polyclonal antibody response to broaden protection. These approaches, which target either the head or the stalk domain of HA, or both domains, have shown promise in recent preclinical and clinical studies. Furthermore, the role of adjuvants in bolstering the robustness of the humoral response has been studied, and their effects on the vaccine-elicited antibody repertoire are currently being investigated. This review will discuss the progress made in the universal influenza vaccine field with respect to influenza A viruses from the perspectives of both antigen and adjuvant, with a focus on the elicitation of broadly neutralizing antibodies.
Collapse
Affiliation(s)
- Kaito A. Nagashima
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Jarrod J. Mousa
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Correspondence:
| |
Collapse
|
45
|
Gebre MS, Brito LA, Tostanoski LH, Edwards DK, Carfi A, Barouch DH. Novel approaches for vaccine development. Cell 2021; 184:1589-1603. [PMID: 33740454 PMCID: PMC8049514 DOI: 10.1016/j.cell.2021.02.030] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023]
Abstract
Vaccines are critical tools for maintaining global health. Traditional vaccine technologies have been used across a wide range of bacterial and viral pathogens, yet there are a number of examples where they have not been successful, such as for persistent infections, rapidly evolving pathogens with high sequence variability, complex viral antigens, and emerging pathogens. Novel technologies such as nucleic acid and viral vector vaccines offer the potential to revolutionize vaccine development as they are well-suited to address existing technology limitations. In this review, we discuss the current state of RNA vaccines, recombinant adenovirus vector-based vaccines, and advances from biomaterials and engineering that address these important public health challenges.
Collapse
Affiliation(s)
- Makda S. Gebre
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- co-first authors
| | | | - Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- co-first authors
| | | | - Andrea Carfi
- Moderna, Inc., Cambridge, MA, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, USA
| |
Collapse
|
46
|
Karch CP, Burkhard P, Matyas GR, Beck Z. The diversity of HIV-1 fights against vaccine efficacy: how self-assembling protein nanoparticle technology may fight back. Nanomedicine (Lond) 2021; 16:673-680. [PMID: 33715403 DOI: 10.2217/nnm-2020-0450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An efficacious HIV-1 vaccine has remained an elusive target for almost 40 years. The sheer diversity of the virus is one of the major roadblocks for vaccine development. HIV-1 frequently mutates and various strains predominate in different geographic regions, making the development of a globally applicable vaccine extremely difficult. Multiple approaches have been taken to overcome the issue of viral diversity, including sequence optimization, development of consensus and mosaic sequences and the use of different prime-boost approaches. To develop an efficacious vaccine, these approaches may need to be combined. One way to potentially synergize these approaches is to use a rationally designed protein nanoparticle that allows for the native-like presentation of antigens, such as the self-assembling protein nanoparticle.
Collapse
Affiliation(s)
- Christopher P Karch
- US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910, USA.,Henry M Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Peter Burkhard
- Alpha-O Peptides, Lörracherstrasse 50, 4125 Riehen, Switzerland
| | - Gary R Matyas
- US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910, USA
| | - Zoltan Beck
- US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910, USA.,Henry M Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA.,Current address: VRD, Pfizer, 401 N Middletown Rd, Pearl River, NY 10965, USA
| |
Collapse
|
47
|
Conti S, Kaczorowski KJ, Song G, Porter K, Andrabi R, Burton DR, Chakraborty AK, Karplus M. Design of immunogens to elicit broadly neutralizing antibodies against HIV targeting the CD4 binding site. Proc Natl Acad Sci U S A 2021; 118:e2018338118. [PMID: 33637649 PMCID: PMC7936365 DOI: 10.1073/pnas.2018338118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
A vaccine which is effective against the HIV virus is considered to be the best solution to the ongoing global HIV/AIDS epidemic. In the past thirty years, numerous attempts to develop an effective vaccine have been made with little or no success, due, in large part, to the high mutability of the virus. More recent studies showed that a vaccine able to elicit broadly neutralizing antibodies (bnAbs), that is, antibodies that can neutralize a high fraction of global virus variants, has promise to protect against HIV. Such a vaccine has been proposed to involve at least three separate stages: First, activate the appropriate precursor B cells; second, shepherd affinity maturation along pathways toward bnAbs; and, third, polish the Ab response to bind with high affinity to diverse HIV envelopes (Env). This final stage may require immunization with a mixture of Envs. In this paper, we set up a framework based on theory and modeling to design optimal panels of antigens to use in such a mixture. The designed antigens are characterized experimentally and are shown to be stable and to be recognized by known HIV antibodies.
Collapse
Affiliation(s)
- Simone Conti
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Kevin J Kaczorowski
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ge Song
- Scripps Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Katelyn Porter
- Scripps Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Raiees Andrabi
- Scripps Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Dennis R Burton
- Scripps Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
| | - Arup K Chakraborty
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139;
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Martin Karplus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138;
- Laboratoire de Chimie Biophysique, Institut de Science et d'Ingénierie Supramoléculaires, Université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
48
|
Adams P, Iserentant G, Servais JY, Vandekerckhove L, Vanham G, Seguin-Devaux C. Cytotoxic CD8+ T Cells Expressing CXCR5 Are Detectable in HIV-1 Elite Controllers After Prolonged In Vitro Peptide Stimulation. Front Immunol 2021; 11:622343. [PMID: 33717056 PMCID: PMC7945035 DOI: 10.3389/fimmu.2020.622343] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/21/2020] [Indexed: 11/23/2022] Open
Abstract
Antiretroviral therapy (ART) is not curative as HIV-1 persists in long-lived viral reservoirs. Consequently, patients are dependent on life-long drug adherence with possible side effects. To overcome these limitations strategies of a functional cure aim at ART free viral remission. In this study, we sought to identify detailed subsets of anti-viral CD8+ T cell immunity linked to natural long-term control of HIV-1 infection. Here, we analyzed HIV controllers and ART suppressed progressors for in vitro viral suppressive capacity (VSC) at baseline and after peptide stimulation. Functional properties and phenotypes of CD8+ T cells were assessed by IFN-γ ELISPOT and 18 color flow cytometry. HIV controllers showed significantly increased suppression at baseline as well as after peptide stimulation. IFN-γ secretion and the proliferation marker Ki67 positively correlated with VSC. Moreover, the detailed phenotype of three distinct multifunctional memory CD8+ T cell subsets were specific traits of HIV controllers of which two correlated convincingly with VSC. Our results underline the importance of multifunctional CD8+ T cell responses during natural control. Especially the role of CXCR5 expressing cytotoxic subsets emphasizes potential surveillance in sites of reservoir persistence and demand further study.
Collapse
Affiliation(s)
- Philipp Adams
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Departments of Biomedical and Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Gilles Iserentant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jean-Yves Servais
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | | | - Guido Vanham
- Departments of Biomedical and Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | | |
Collapse
|
49
|
Eslamizar L, Petrovas C, Leggat DJ, Furr K, Lifton ML, Levine G, Ma S, Fletez-Brant C, Hoyland W, Prabhakaran M, Narpala S, Boswell K, Yamamoto T, Liao HX, Pickup D, Ramsburg E, Sutherland L, McDermott A, Roederer M, Montefiori D, Koup RA, Haynes BF, Letvin NL, Santra S. Recombinant MVA-prime elicits neutralizing antibody responses by inducing antigen-specific B cells in the germinal center. NPJ Vaccines 2021; 6:15. [PMID: 33495459 PMCID: PMC7835239 DOI: 10.1038/s41541-020-00277-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/07/2020] [Indexed: 01/23/2023] Open
Abstract
The RV144 HIV-1 vaccine trial has been the only clinical trial to date that has shown any degree of efficacy and associated with the presence of vaccine-elicited HIV-1 envelope-specific binding antibody and CD4+ T-cell responses. This trial also showed that a vector-prime protein boost combined vaccine strategy was better than when used alone. Here we have studied three different priming vectors-plasmid DNA, recombinant MVA, and recombinant VSV, all encoding clade C transmitted/founder Env 1086 C gp140, for priming three groups of six non-human primates each, followed by a protein boost with adjuvanted 1086 C gp120 protein. Our data showed that MVA-priming favors the development of higher antibody binding titers and neutralizing activity compared with other vectors. Analyses of the draining lymph nodes revealed that MVA-prime induced increased germinal center reactivity characterized by higher frequencies of germinal center (PNAhi) B cells, higher frequencies of antigen-specific B-cell responses as well as an increased frequency of the highly differentiated (ICOShiCD150lo) Tfh-cell subset.
Collapse
Affiliation(s)
- Leila Eslamizar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Integrative Toxicology, Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., 175 Briar Ridge Road, Ridgefield, CT, 06877, USA
| | - Constantinos Petrovas
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA.
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| | | | - Kathryn Furr
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michelle L Lifton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gail Levine
- Foundation for the National Institutes of Health, Bethesda, MD, USA
| | - Steven Ma
- Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | | | | | | | | | | | | | - Hua-Xin Liao
- Foundation for the National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | - Norman L Letvin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sampa Santra
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
50
|
Olusola BA, Olaleye DO, Odaibo GN. Non-synonymous Substitutions in HIV-1 GAG Are Frequent in Epitopes Outside the Functionally Conserved Regions and Associated With Subtype Differences. Front Microbiol 2021; 11:615721. [PMID: 33505382 PMCID: PMC7829476 DOI: 10.3389/fmicb.2020.615721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/15/2020] [Indexed: 12/22/2022] Open
Abstract
In 2019, 38 million people lived with HIV-1 infection resulting in 690,000 deaths. Over 50% of this infection and its associated deaths occurred in Sub-Saharan Africa. The West African region is a known hotspot of the HIV-1 epidemic. There is a need to develop an HIV-1 vaccine if the HIV epidemic would be effectively controlled. Few protective cytotoxic T Lymphocytes (CTL) epitopes within the HIV-1 GAG (HIV_gagconsv) have been previously identified to be functionally conserved among the HIV-1 M group. These epitopes are currently the focus of universal HIV-1 T cell-based vaccine studies. However, these epitopes' phenotypic and genetic properties have not been observed in natural settings for HIV-1 strains circulating in the West African region. This information is critical as the usefulness of universal HIV-1 vaccines in the West African region depends on these epitopes' occurrence in strains circulating in the area. This study describes non-synonymous substitutions within and without HIV_gagconsv genes isolated from 10 infected Nigerians at the early stages of HIV-1 infection. Furthermore, we analyzed these substitutions longitudinally in five infected individuals from the early stages of infection till after seroconversion. We identified three non-synonymous substitutions within HIV_gagconsv genes isolated from early HIV infected individuals. Fourteen and nineteen mutations outside the HIV_gagconsv were observed before and after seroconversion, respectively, while we found four mutations within the HIV_gagconsv. These substitutions include previously mapped CTL epitope immune escape mutants. CTL immune pressure likely leaves different footprints on HIV-1 GAG epitopes within and outside the HIV_gagconsv. This information is crucial for universal HIV-1 vaccine designs for use in the West African region.
Collapse
Affiliation(s)
| | | | - Georgina N. Odaibo
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|