1
|
Nel C, Frater J. Enhancing broadly neutralising antibody suppression of HIV by immune modulation and vaccination. Front Immunol 2024; 15:1478703. [PMID: 39575236 PMCID: PMC11578998 DOI: 10.3389/fimmu.2024.1478703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Although HIV infection can be managed with antiretroviral drugs, there is no cure and therapy has to be taken for life. Recent successes in animal models with HIV-specific broadly neutralising antibodies (bNAbs) have led to long-term virological remission and even possible cures in some cases. This has resulted in substantial investment in human studies to explore bNAbs as a curative intervention for HIV infection. Emerging data are encouraging, but suggest that combinations of bNAbs with other immunomodulatory agents may be needed to induce and sustain long-term viral control. As a result, a number of clinical trials are currently underway exploring these combinations. If successful, the impact for the millions of people living with HIV could be substantial. Here, we review the background to the use of bNAbs in the search for an HIV cure and how different adjunctive agents might be used together to enhance their efficacy.
Collapse
Affiliation(s)
- Carla Nel
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- The National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
2
|
Mothe B, Rosás-Umbert M, Coll P, Manzardo C, Puertas MC, Morón-López S, Llano A, Miranda C, Cedeño S, López M, Alarcón-Soto Y, Melis GG, Langohr K, Barriocanal AM, Toro J, Ruiz I, Rovira C, Carrillo A, Meulbroek M, Crook A, Wee EG, Miró JM, Clotet B, Valle M, Martinez-Picado J, Hanke T, Brander C, Moltó J. HIVconsv Vaccines and Romidepsin in Early-Treated HIV-1-Infected Individuals: Safety, Immunogenicity and Effect on the Viral Reservoir (Study BCN02). Front Immunol 2020; 11:823. [PMID: 32435247 PMCID: PMC7218169 DOI: 10.3389/fimmu.2020.00823] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/09/2020] [Indexed: 12/30/2022] Open
Abstract
Kick&kill strategies combining drugs aiming to reactivate the viral reservoir with therapeutic vaccines to induce effective cytotoxic immune responses hold potential to achieve a functional cure for HIV-1 infection. Here, we report on an open-label, single-arm, phase I clinical trial, enrolling 15 early-treated HIV-1-infected individuals, testing the combination of the histone deacetylase inhibitor romidepsin as a latency-reversing agent and the MVA.HIVconsv vaccine. Romidepsin treatment resulted in increased histone acetylation, cell-associated HIV-1 RNA, and T-cell activation, which were associated with a marginally significant reduction of the viral reservoir. Vaccinations boosted robust and broad HIVconsv-specific T cells, which were strongly refocused toward conserved regions of the HIV-1 proteome. During a monitored ART interruption phase using plasma viral load over 2,000 copies/ml as a criterium for ART resumption, 23% of individuals showed sustained suppression of viremia up to 32 weeks without evidence for reseeding the viral reservoir. Results from this pilot study show that the combined kick&kill intervention was safe and suggest a role for this strategy in achieving an immune-driven durable viremic control.
Collapse
Affiliation(s)
- Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain
| | - Miriam Rosás-Umbert
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
| | - Pep Coll
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
| | | | | | | | - Anuska Llano
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
| | - Cristina Miranda
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Miriam López
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Yovaninna Alarcón-Soto
- Departament d'Estadística i Investigació Operativa, Universitat Politècnica de Catalunya/BARCELONATECH, Barcelona, Spain
| | - Guadalupe Gómez Melis
- Departament d'Estadística i Investigació Operativa, Universitat Politècnica de Catalunya/BARCELONATECH, Barcelona, Spain
| | - Klaus Langohr
- Departament d'Estadística i Investigació Operativa, Universitat Politècnica de Catalunya/BARCELONATECH, Barcelona, Spain
| | - Ana M Barriocanal
- Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain.,Department of Infectious Diseases, Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Jessica Toro
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Irene Ruiz
- Hospital Clinic- IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Cristina Rovira
- Hospital Clinic- IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Antonio Carrillo
- Department of Infectious Diseases, Germans Trias i Pujol Research Institute, Badalona, Spain
| | | | - Alison Crook
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Edmund G Wee
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jose M Miró
- Hospital Clinic- IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
| | - Marta Valle
- Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain.,Pharmacokinetic/Pharmacodynamic Modeling and Simultation, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,ICREA, Barcelona, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, United Kingdom.,Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,ICREA, Barcelona, Spain
| | - José Moltó
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Infectious Diseases, Germans Trias i Pujol Research Institute, Badalona, Spain
| | | |
Collapse
|
3
|
Lacaille-Dubois MA. Updated insights into the mechanism of action and clinical profile of the immunoadjuvant QS-21: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152905. [PMID: 31182297 PMCID: PMC7127804 DOI: 10.1016/j.phymed.2019.152905] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/26/2019] [Accepted: 03/30/2019] [Indexed: 05/06/2023]
Abstract
BACKGROUND Vaccine adjuvants are compounds that significantly enhance/prolong the immune response to a co-administered antigen. The limitations of the use of aluminium salts that are unable to elicite cell responses against intracellular pathogens such as those causing malaria, tuberculosis, or AIDS, have driven the development of new alternative adjuvants such as QS-21, a triterpene saponin purified from Quillaja saponaria. PURPOSE The aim of this review is to attempt to clarify the mechanism of action of QS-21 through either receptors or signaling pathways in vitro and in vivo with special emphasis on the co-administration with other immunostimulants in new adjuvant formulations, called adjuvant systems (AS). Furthermore, the most relevant clinical applications will be presented. METHODS A literature search covering the period 2014-2018 was performed using electronic databases from Sci finder, Science direct, Medline/Pubmed, Scopus, Google scholar. RESULTS Insights into the mechanism of action of QS-21 can be summarized as follows: 1) in vivo stimulation of Th2 humoral and Th1 cell-mediated immune responses through action on antigen presenting cells (APCs) and T cells, leading to release of Th1 cytokines participating in the elimination of intracellular pathogens. 2) activation of the NLRP3 inflammasome in mouse APCs with subsequent release of caspase-1 dependent cytokines, Il-1β and Il-18, important for Th1 responses. 3) synthesis of nearly 50 QS-21 analogs, allowing structure/activity relationships and mechanistic studies. 4) unique synergy mechanism between monophosphoryl lipid A (MPL A) and QS-21, formulated in a liposome (AS01) in the early IFN-γ response, promoting vaccine immunogenicity. The second part of the review is related to phase I-III clinical trials of QS-21, mostly formulated in ASs, to evaluate efficacy, immunogenicity and safety of adjuvanted prophylactic vaccines against infectious diseases, e.g. malaria, herpes zoster, tuberculosis, AIDS and therapeutic vaccines against cancer and Alzheimer's disease. CONCLUSION The most advanced phase III clinical applications led to the development of two vaccines containing QS-21 as part of the AS, the Herpes Zoster vaccine (HZ/su) (Shingrix™) which received a license in 2017 from the FDA and a marketing authorization in the EU in 2018 and the RTS,S/AS01 vaccine (Mosquirix™) against malaria, which was approved by the EMA in 2015 for further implementation in Sub-Saharan countries for routine use.
Collapse
Affiliation(s)
- Marie-Aleth Lacaille-Dubois
- PEPITE EA 4267, Université de Bourgogne Franche-Comté, Laboratoire de Pharmacognosie, UFR des Sciences de Santé, 7, Bd Jeanne d'Arc, 21079 Dijon Cedex, France.
| |
Collapse
|
4
|
Mothe B, Manzardo C, Sanchez-Bernabeu A, Coll P, Morón-López S, Puertas MC, Rosas-Umbert M, Cobarsi P, Escrig R, Perez-Alvarez N, Ruiz I, Rovira C, Meulbroek M, Crook A, Borthwick N, Wee EG, Yang H, Miró JM, Dorrell L, Clotet B, Martinez-Picado J, Brander C, Hanke T. Therapeutic Vaccination Refocuses T-cell Responses Towards Conserved Regions of HIV-1 in Early Treated Individuals (BCN 01 study). EClinicalMedicine 2019; 11:65-80. [PMID: 31312806 PMCID: PMC6610778 DOI: 10.1016/j.eclinm.2019.05.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Strong and broad antiviral T-cell responses targeting vulnerable sites of HIV-1 will likely be a critical component for any effective cure strategy. METHODS BCN01 trial was a phase I, open-label, non-randomized, multicenter study in HIV-1-positive individuals diagnosed and treated during early HIV-1 infection to evaluate two vaccination regimen arms, which differed in the time (8 versus 24 week) between the ChAdV63.HIVconsv prime and MVA.HIVconsv boost vaccinations. The primary outcome was safety. Secondary endpoints included frequencies of vaccine-induced IFN-γ+ CD8+ T cells, in vitro virus-inhibitory capacity, plasma HIV-1 RNA and total CD4+ T-cells associated HIV-1 DNA. (NCT01712425). FINDINGS No differences in safety, peak magnitude or durability of vaccine-induced responses were observed between long and short interval vaccination arms. Grade 1/2 local and systemic post-vaccination events occurred in 22/24 individuals and resolved within 3 days. Weak responses to conserved HIV-1 regions were detected in 50% of the individuals before cART initiation, representing median of less than 10% of their total HIV-1-specific T cells. All participants significantly elevated these subdominant T-cell responses, which after MVA.HIVconsv peaked at median (range) of 938 (73-6,805) IFN-γ SFU/106 PBMC, representing on average 58% of their total anti-HIV-1 T cells. The decay in the size of the HIV-1 reservoir was consistent with the first year of early cART initiation in both arms. INTERPRETATION Heterologous prime-boost vaccination with ChAdV63-MVA/HIVconsv was well-tolerated and refocused pre-cART T-cell responses towards more protective epitopes, in which immune escape is frequently associated with reduced HIV-1 replicative fitness and which are common to most global HIV-1 variants. FUNDING HIVACAT Catalan research program for an HIV vaccine and Fundació Gloria Soler. Vaccine manufacture was jointly funded by the Medical Research Council (MRC) UK and the UK Department for International Development (DFID) under the MRC/DFID Concordat agreements (G0701669. RESEARCH IN CONTEXT Evidence Before this Study: T cells play an important role in the control of HIV infection and may be particularly useful for HIV-1 cure by killing cells with reactivated HIV-1. Evidence is emerging that not all T-cell responses are protective and mainly only those targeting conserved regions of HIV-1 proteins are effective, but typically immunologically subdominant, while those recognizing hypervariable, easy-to-escape immunodominant 'decoys' do not control viremia and do not protect from a loss of CD4 T cells. We pioneered a vaccine strategy focusing T-cell responses on the most conserved regions of the HIV-1 proteome using an immunogen designated HIVconsv. T cells elicited by the HIVconsv vaccines in HIV-uninfected UK and Kenyan adults inhibited in vitro replication of HIV-1 isolates from 4 major global clades A, B, C and D.Added Value of this Study: The present study demonstrated the concept that epitopes subdominant in natural infection, when taken out of the context of the whole HIV-1 proteome and presented to the immune system by a potent simian adenovirus prime-poxvirus MVA boost regimen, can induce strong responses in patients on antiretroviral treatment and efficiently refocus HIV-1-specific T-cells to the protective epitopes delivered by the vaccine.Implications of all the Available Evidence: Nearly all HIV-1 vaccine strategies currently emphasize induction of broadly neutralizing Abs. The HIVconsv vaccine is one of a very few approaches focussing exclusively on elicitation of T cells and, therefore, can complement antibody induction for better prevention and cure. Given the cross-clade reach on the HIVconsv immunogen design, if efficient, the HIVconsv vaccines could be deployed globally. Effective vaccines will likely be a necessary component in combination with other available preventive measures for halting the HIV-1/AIDS epidemic.
Collapse
Affiliation(s)
- Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- Corresponding author at: IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Crta Canyet s/n, 08916, Badalona, Barcelona, Spain.
| | | | | | - Pep Coll
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | | | - Miriam Rosas-Umbert
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Patricia Cobarsi
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Roser Escrig
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Núria Perez-Alvarez
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Technical University of Catalonia, Barcelona, Spain
| | - Irene Ruiz
- Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Cristina Rovira
- Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Alison Crook
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | - Edmund G. Wee
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Hongbing Yang
- Nuffield Department of Medicine and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jose M. Miró
- Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Lucy Dorrell
- Nuffield Department of Medicine and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- ICREA, Pg. Luis Companys 23, Barcelona, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- ICREA, Pg. Luis Companys 23, Barcelona, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, UK
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
5
|
Harrer T, Dinges W, Roman F. Long-term follow-up of HIV-1-infected adults who received the F4/AS01 B HIV-1 vaccine candidate in two randomised controlled trials. Vaccine 2018; 36:2683-2686. [PMID: 29606517 DOI: 10.1016/j.vaccine.2018.03.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/22/2018] [Accepted: 03/15/2018] [Indexed: 11/18/2022]
Abstract
This Phase I/II, open, long-term follow-up study was conducted in antiretroviral therapy (ART)-naïve (N = 212) and ART-treated (N = 19) human immunodeficiency virus 1 (HIV-1)-infected adults, who received an HIV-1 investigational vaccine (F4/AS01B) or placebo in two previous studies (NCT00814762 and NCT01218113). After a minimum of two years and a maximum of four years of follow-up post-vaccination per patient, no significant differences were observed between F4/AS01B and placebo groups in terms of viral load, CD4+ T-cell count and incidence of specific clinical events. Vaccine-induced polyfunctional CD4+ T-cells persisted up to study end and no relevant vaccine-related safety events were reported in F4/AS01B groups. This study has been registered at ClinicalTrials.gov (NCT01092611).
Collapse
Affiliation(s)
- Thomas Harrer
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany.
| | - Warren Dinges
- Seattle Infectious Disease Clinic, Seattle, WA, USA.
| | | |
Collapse
|
6
|
Burny W, Callegaro A, Bechtold V, Clement F, Delhaye S, Fissette L, Janssens M, Leroux-Roels G, Marchant A, van den Berg RA, Garçon N, van der Most R, Didierlaurent AM. Different Adjuvants Induce Common Innate Pathways That Are Associated with Enhanced Adaptive Responses against a Model Antigen in Humans. Front Immunol 2017; 8:943. [PMID: 28855902 PMCID: PMC5557780 DOI: 10.3389/fimmu.2017.00943] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/24/2017] [Indexed: 12/20/2022] Open
Abstract
To elucidate the role of innate responses in vaccine immunogenicity, we compared early responses to hepatitis B virus (HBV) surface antigen (HBsAg) combined with different Adjuvant Systems (AS) in healthy HBV-naïve adults, and included these parameters in multi-parametric models of adaptive responses. A total of 291 participants aged 18–45 years were randomized 1:1:1:1:1 to receive HBsAg with AS01B, AS01E, AS03, AS04, or Alum/Al(OH)3 at days 0 and 30 (ClinicalTrials.gov: NCT00805389). Blood protein, cellular, and mRNA innate responses were assessed at early time-points and up to 7 days after vaccination, and used with reactogenicity symptoms in linear regression analyses evaluating their correlation with HBs-specific CD4+ T-cell and antibody responses at day 44. All AS induced transient innate responses, including interleukin (IL)-6 and C-reactive protein (CRP), mostly peaking at 24 h post-vaccination and subsiding to baseline within 1–3 days. After the second but not the first injection, median interferon (IFN)-γ levels were increased in the AS01B group, and IFN-γ-inducible protein-10 levels and IFN-inducible genes upregulated in the AS01 and AS03 groups. No distinct marker or signature was specific to one particular AS. Innate profiles were comparable between AS01B, AS01E, and AS03 groups, and between AS04 and Alum groups. AS group rankings within adaptive and innate response levels and reactogenicity prevalence were similar (AS01B ≥ AS01E > AS03 > AS04 > Alum), suggesting an association between magnitudes of inflammatory and vaccine responses. Modeling revealed associations between adaptive responses and specific traits of the innate response post-dose 2 (activation of the IFN-signaling pathway, CRP and IL-6 responses). In conclusion, the ability of AS01 and AS03 to enhance adaptive responses to co-administered HBsAg is likely linked to their capacity to activate innate immunity, particularly the IFN-signaling pathway.
Collapse
Affiliation(s)
| | | | | | - Frédéric Clement
- Center for Vaccinology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | | | | | | | - Geert Leroux-Roels
- Center for Vaccinology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | | | | | | | | | | |
Collapse
|
7
|
Tung FY, Tung JK, Pallikkuth S, Pahwa S, Fischl MA. A therapeutic HIV-1 vaccine enhances anti-HIV-1 immune responses in patients under highly active antiretroviral therapy. Vaccine 2016; 34:2225-32. [PMID: 27002500 DOI: 10.1016/j.vaccine.2016.03.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND HIV-1 specific cellular immunity plays an important role in controlling viral replication. In this first-in-human therapeutic vaccination study, a replication-defective HIV-1 vaccine (HIVAX) was tested in HIV-1 infected participants undergoing highly active antiretroviral therapy (HAART) to enhance anti-HIV immunity (Clinicaltrials.gov, identifier NCT01428596). METHODS A010 was a randomized, placebo-controlled trial to evaluate the safety and the immunogenicity of a replication defective HIV-1 vaccine (HIVAX) given as a subcutaneous injection to HIV-1 infected participants who were receiving HAART with HIV-1 viral load <50 copies/ml and CD4 cell count >500 cells/mm(3). HIV-1 specific immune responses were monitored by INF-γ enzyme linked immunospot (Elispot) and intracellular cytokine staining (ICS) assay after vaccination. Following the randomized placebo-controlled vaccination phase, subjects who received HIVAX vaccine and who met eligibility underwent a 12-week analytical antiretroviral treatment interruption (ATI). Viral load was monitored throughout the study. RESULTS HIVAX was well tolerated in trial participants. Transient grade 1 to 2 (mild to moderate) injection site reactions occurred in 8 of 10 vaccinated participants. HIVAX was immunogenic in all vaccinated participants. The functionality of T cells was significantly enhanced after vaccination. Median viral load (3.45 log10 copies/ml, range of 96-12,830 copies/ml) at the end of the 12-week treatment interruption in HIVAX vaccinated group was significantly lower than the pre-treatment levels. Three vaccinated participants extended ATI for up to 2 years with stable CD4 cells and low viral loads. CONCLUSIONS HIVAX vaccine is generally safe, elicits strong anti-HIV-1 immune responses, and may play an important role in controlling viral load during treatment interruption in HIV-1 infected participants.
Collapse
Affiliation(s)
- Frank Y Tung
- GeneCure Biotechnologies, Peachtree Corners, GA, USA.
| | - Jack K Tung
- GeneCure Biotechnologies, Peachtree Corners, GA, USA
| | | | - Savita Pahwa
- University of Miami, School of Medicine, Miami, FL, USA
| | | |
Collapse
|
8
|
Dinges W, Girard PM, Podzamczer D, Brockmeyer NH, García F, Harrer T, Lelievre JD, Frank I, Colin De Verdière N, Yeni GP, Ortega Gonzalez E, Rubio R, Clotet Sala B, DeJesus E, Pérez-Elias MJ, Launay O, Pialoux G, Slim J, Weiss L, Bouchaud O, Felizarta F, Meurer A, Raffi F, Esser S, Katlama C, Koletar SL, Mounzer K, Swindells S, Baxter JD, Schneider S, Chas J, Molina JM, Koutsoukos M, Collard A, Bourguignon P, Roman F. The F4/AS01B HIV-1 Vaccine Candidate Is Safe and Immunogenic, But Does Not Show Viral Efficacy in Antiretroviral Therapy-Naive, HIV-1-Infected Adults: A Randomized Controlled Trial. Medicine (Baltimore) 2016; 95:e2673. [PMID: 26871794 PMCID: PMC4753889 DOI: 10.1097/md.0000000000002673] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The impact of the investigational human immunodeficiency virus type 1 (HIV-1) F4/AS01B vaccine on HIV-1 viral load (VL) was evaluated in antiretroviral therapy (ART)-naive HIV-1 infected adults.This phase IIb, observer-blind study (NCT01218113), included ART-naive HIV-1 infected adults aged 18 to 55 years. Participants were randomized to receive 2 (F4/AS01B_2 group, N = 64) or 3 (F4/AS01B_3 group, N = 62) doses of F4/AS01B or placebo (control group, N = 64) at weeks 0, 4, and 28. Efficacy (HIV-1 VL, CD4 T-cell count, ART initiation, and HIV-related clinical events), safety, and immunogenicity (antibody and T-cell responses) were evaluated during 48 weeks.At week 48, based on a mixed model, no statistically significant difference in HIV-1 VL change from baseline was demonstrated between F4/AS01B_2 and control group (0.073 log10 copies/mL [97.5% confidence interval (CI): -0.088; 0.235]), or F4/AS01B_3 and control group (-0.096 log10 copies/mL [97.5% CI: -0.257; 0.065]). No differences between groups were observed in HIV-1 VL change, CD4 T-cell count, ART initiation, or HIV-related clinical events at intermediate timepoints. Among F4/AS01B recipients, the most frequent solicited symptoms were pain at injection site (252/300 doses), fatigue (137/300 doses), myalgia (105/300 doses), and headache (90/300 doses). Twelve serious adverse events were reported in 6 participants; 1 was considered vaccine-related (F4/AS01B_2 group: angioedema). F4/AS01B induced polyfunctional F4-specific CD4 T-cells, but had no significant impact on F4-specific CD8 T-cell and anti-F4 antibody levels.F4/AS01B had a clinically acceptable safety profile, induced F4-specific CD4 T-cell responses, but did not reduce HIV-1 VL, impact CD4 T-cells count, delay ART initiation, or prevent HIV-1 related clinical events.
Collapse
Affiliation(s)
- Warren Dinges
- From the Seattle Travel and Preventive Medicine, Seattle Infectious Disease Clinic, Seattle, WA, USA (WD); Service des Maladies Infectieuses, Hôpital Saint Antoine, Assistance Publique Hôpitaux de Paris; and INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (P-MG); HIV Unit, Infectious Disease Service, Hospital Universitari de Bellvitge, L'Hospitalet, 08907 Barcelona, Spain (DP); Department of Dermatology, Venerology, and Allergology, St. Josef-Hospital, Ruhr-Universität Bochum, Bochum, Germany (NHB); Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain (FG); Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (TH); Service d'Immunologie Clinique, Hôpital Henri Mondor, Créteil, France (J-DL); University of Pennsylvania, Philadelphia, PA, USA (IF); Service des Maladies Infectieuses et Tropicales, Hôpital Saint Louis, University of Paris Diderot Paris 7, Sorbonne Paris Cité and INSERM U941 (NCDV, J-MM); Hôpital Bichat Claude Bernard, Service des Maladies Infectieuses et Tropicales A, Paris, France (G-PY); Servicio de Enfermedades Infecciosas, Hospital General Universitario de Valencia, Valencia (EOG); Servicio de Enfermedades Infecciosas, Hospital 12 De Octubre, Madrid, Spain (RR); IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Uvic-UCC, Barcelona, Spain (BCS); Orlando Immunology Center, Orlando, FL, USA (EDS); Servicio de Enfermedades Infecciosas, Hospital Ramón Y Cajal, IRYCIS Madrid, Spain (MJPE); Université Paris Descartes, Sorbonne Paris Cité, Inserm, CIC 1417 and F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC); and Assistance Publique Hôpitaux de Paris, Hôpital Cochin (OL); Maladies Infectieuses et Tropicales Co-infections, Hôpital Tenon, Paris, France (GP, JC); Saint Michael's Medical Center, Newark, NJ, USA (JS); Service d'immunologie Clinique, Hôpital Européen Georges Pompidou, Paris, France (LW); Service des Maladie Infectieuses et Tropicales, Hôpital Avicenne, Bobigny, France (OB); Private practice, Bakersfield, CA, USA (FF); Zentrum für Innere Medizin und Infektiologie, Praxis, München, Germany (AM); CMIT, 46 Rue Henri Huchard, Paris, France (FR); HIV Ambulanz, Klinik für Dermatologie, Uniklinikum Essen, Essen, Germany (SE); Service des Maladies Infectieuses et Tropicales, Hôpital de la Pitié-Salpêtrière, Paris, France (CK); The Ohio State University, Division of Infectious Diseases, Columbus, OH (SLK); Philadelphia FIGHT, Philadelphia, PA (KM); University of Nebraska Medical Center, Omaha, NE (SS); Cooper University Hospital, Cooper Medical School of Rowan University, Camden, NJ (JDB); Living Hope Clinical Foundation, Long Beach, CA, USA (SS); and GSK Vaccines, Wavre/Rixensart, Belgium (MK, AC, PB, FR)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Blanco-Heredia J, Lecanda A, Valenzuela-Ponce H, Brander C, Ávila-Ríos S, Reyes-Terán G. Identification of Immunogenic Cytotoxic T Lymphocyte Epitopes Containing Drug Resistance Mutations in Antiretroviral Treatment-Naïve HIV-Infected Individuals. PLoS One 2016; 11:e0147571. [PMID: 26808823 PMCID: PMC4725752 DOI: 10.1371/journal.pone.0147571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/05/2016] [Indexed: 11/18/2022] Open
Abstract
Background Therapeutic HIV vaccines may prove helpful to intensify antiretroviral treatment (ART) efficacy and may be an integral part of future cure strategies. Methods We examined IFN-gamma ELISpot responses to a panel of 218 HIV clade B consensus-based HIV protease-reverse transcriptase peptides, designed to mimic previously described and predicted cytotoxic T lymphocyte epitopes overlapping drug resistance (DR) positions, that either included the consensus sequence or the DR variant sequence, in 49 ART-naïve HIV-infected individuals. Next generation sequencing was used to assess the presence of minority DR variants in circulating viral populations. Results Although a wide spectrum of differential magnitudes of response to DR vs. WT peptide pairs was observed, responses to DR peptides were frequent and strong in the study cohort. No difference between the median magnitudes of response to DR vs. WT peptides was observed. Interestingly, of the 22 peptides that were recognized by >15% of the participants, two-thirds (64%) corresponded to DR peptides. When analysing responses per peptide pair per individual, responses to only WT (median 4 pairs/individual) or DR (median 6 pairs/individual) were more common than responses to both WT and DR (median 2 pairs/individual; p<0.001). While the presence of ELISpot responses to WT peptides was frequently associated with the presence of the corresponding peptide sequence in the patient’s virus (mean 68% of cases), responses to DR peptides were generally not associated with the presence of DR mutations in the viral population, even at low frequencies (mean 1.4% of cases; p = 0.0002). Conclusions Our data suggests that DR peptides are frequently immunogenic and raises the potential benefit of broadening the antigens included in a therapeutic vaccine approach to immunogenic epitopes containing common DR sequences. Further studies are needed to assess the quality of responses elicited by DR peptides.
Collapse
Affiliation(s)
- Juan Blanco-Heredia
- Centre for Infectious Diseases Research, National Institute of Respiratory Diseases, Mexico City, Mexico
| | - Aarón Lecanda
- Centre for Infectious Diseases Research, National Institute of Respiratory Diseases, Mexico City, Mexico
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Humberto Valenzuela-Ponce
- Centre for Infectious Diseases Research, National Institute of Respiratory Diseases, Mexico City, Mexico
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)-UAB, Barcelona, Spain
- University of Vic and Central Catalonia, Vic, Spain
| | - Santiago Ávila-Ríos
- Centre for Infectious Diseases Research, National Institute of Respiratory Diseases, Mexico City, Mexico
- * E-mail: (GRT); (SAR)
| | - Gustavo Reyes-Terán
- Centre for Infectious Diseases Research, National Institute of Respiratory Diseases, Mexico City, Mexico
- * E-mail: (GRT); (SAR)
| |
Collapse
|
10
|
Abstract
Vaccination is the most effective means of preventing and controlling viral infections. The eradication of smallpox and the significant progress made toward polio eradication are clear examples of the great impact of antiviral vaccines. However, viral infections remain a major public health threat and a significant cause of death. Most of the antiviral vaccines introduced over the past century were empirically developed. Poliomyelitis, measles, mumps, and rubella are examples of diseases that are now largely controlled thanks to these empirically developed vaccines. However, there is a growing list of viral pathogens against which effective vaccines are yet to be developed. Recent technological advances will potentially provide us with new platforms that could be harnessed to develop vaccines against emerging and reemerging viral pathogens.
Collapse
|
11
|
Kumarasamy N, Poongulali S, Bollaerts A, Moris P, Beulah FE, Ayuk LN, Demoitié MA, Jongert E, Ofori-Anyinam O. A Randomized, Controlled Safety, and Immunogenicity Trial of the M72/AS01 Candidate Tuberculosis Vaccine in HIV-Positive Indian Adults. Medicine (Baltimore) 2016; 95:e2459. [PMID: 26817879 PMCID: PMC4998253 DOI: 10.1097/md.0000000000002459] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Human immunodeficiency virus (HIV)-associated tuberculosis is a major public health threat. We evaluated the safety and immunogenicity of the candidate tuberculosis vaccine M72/AS01 in HIV-positive and HIV-negative Indian adults.Randomized, controlled observer-blind trial (NCT01262976).We assigned 240 adults (1:1:1) to antiretroviral therapy (ART)-stable, ART-naive, or HIV-negative cohorts. Cohorts were randomized 1:1 to receive M72/AS01 or placebo following a 0, 1-month schedule and followed for 12 months (time-point M13). HIV-specific and laboratory safety parameters, adverse events (AEs), and M72-specific T-cell-mediated and humoral responses were evaluated.Subjects were predominantly QuantiFERON-negative (60%) and Bacille Calmette-Guérin-vaccinated (73%). Seventy ART-stable, 73 ART-naive, and 60 HIV-negative subjects completed year 1. No vaccine-related serious AEs or ART-regimen adjustments, or clinically relevant effects on laboratory parameters, HIV-1 viral loads or CD4 counts were recorded. Two ART-naive vaccinees died of vaccine-unrelated diseases. M72/AS01 induced polyfunctional M72-specific CD4 T-cell responses (median [interquartile range] at 7 days postdose 2: ART-stable, 0.9% [0.7-1.5]; ART-naive, 0.5% [0.2-1.0]; and HIV-negative, 0.6% [0.4-1.1]), persisting at M13 (0.4% [0.2-0.5], 0.09% [0.04-0.2], and 0.1% [0.09-0.2], respectively). Median responses were higher in the ART-stable cohort versus ART-naive cohort from day 30 onwards (P ≤ 0.015). Among HIV-positive subjects (irrespective of ART-status), median responses were higher in QuantiFERON-positive versus QuantiFERON-negative subjects up to day 30 (P ≤ 0.040), but comparable thereafter. Cytokine-expression profiles were comparable between cohorts after dose 2. At M13, M72-specific IgG responses were higher in ART-stable and HIV-negative vaccinees versus ART-naive vaccinees (P ≤ 0.001).M72/AS01 was well-tolerated and immunogenic in this population of ART-stable and ART-naive HIV-positive adults and HIV-negative adults, supporting further clinical evaluation.
Collapse
Affiliation(s)
- Nagalingeswaran Kumarasamy
- From the YRG CARE Medical Centre, VHS (NK, SP, FEB); and GSK Vaccines, Rixensart/Wavre, Belgium (AB, PM, LNA, M-AD, EJ, OO-A)
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
A Phase I Double Blind, Placebo-Controlled, Randomized Study of the Safety and Immunogenicity of an Adjuvanted HIV-1 Gag-Pol-Nef Fusion Protein and Adenovirus 35 Gag-RT-Int-Nef Vaccine in Healthy HIV-Uninfected African Adults. PLoS One 2015; 10:e0125954. [PMID: 25961283 PMCID: PMC4427332 DOI: 10.1371/journal.pone.0125954] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/22/2015] [Indexed: 11/19/2022] Open
Abstract
Background Sequential prime-boost or co-administration of HIV vaccine candidates based on an adjuvanted clade B p24, RT, Nef, p17 fusion protein (F4/AS01) plus a non-replicating adenovirus 35 expressing clade A Gag, RT, Int and Nef (Ad35-GRIN) may lead to a unique immune profile, inducing both strong T-cell and antibody responses. Methods In a phase 1, double-blind, placebo-controlled trial, 146 healthy adult volunteers were randomized to one of four regimens: heterologous prime-boost with two doses of F4/AS01E or F4/AS01B followed by Ad35-GRIN; Ad35-GRIN followed by two doses of F4/AS01B; or three co-administrations of Ad35-GRIN and F4/AS01B. T cell and antibody responses were measured. Results The vaccines were generally well-tolerated, and did not cause serious adverse events. The response rate, by IFN-γ ELISPOT, was greater when Ad35-GRIN was the priming vaccine and in the co-administration groups. F4/AS01 induced CD4+ T-cells expressing primarily CD40L and IL2 +/- TNF-α, while Ad35-GRIN induced predominantly CD8+ T-cells expressing IFN-γ +/- IL2 or TNF-α. Viral inhibition was induced after Ad35-GRIN vaccination, regardless of the regimen. Strong F4-specific antibody responses were induced. Immune responses persisted at least a year after the last vaccination. The complementary response profiles, characteristic of each vaccine, were both expressed after co-administration. Conclusion Co-administration of an adjuvanted protein and an adenovirus vector showed an acceptable safety and reactogenicity profile and resulted in strong, multifunctional and complementary HIV-specific immune responses. Trial Registration ClinicalTrials.gov NCT01264445
Collapse
|
13
|
Lorin C, Vanloubbeeck Y, Baudart S, Ska M, Bayat B, Brauers G, Clarinval G, Donner MN, Marchand M, Koutsoukos M, Mettens P, Cohen J, Voss G. Heterologous prime-boost regimens with a recombinant chimpanzee adenoviral vector and adjuvanted F4 protein elicit polyfunctional HIV-1-specific T-Cell responses in macaques. PLoS One 2015; 10:e0122835. [PMID: 25856308 PMCID: PMC4391709 DOI: 10.1371/journal.pone.0122835] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/15/2015] [Indexed: 01/04/2023] Open
Abstract
HIV-1-specific CD4+ and CD8+ T lymphocytes are important for HIV-1 replication control. F4/AS01 consists of F4 recombinant fusion protein (containing clade B Gag/p24, Pol/RT, Nef and Gag/p17) formulated in AS01 Adjuvant System, and was shown to induce F4-specific polyfunctional CD4+ T-cell responses in humans. While replication-incompetent recombinant HIV-1/SIV antigen-expressing human adenoviral vectors can elicit high-frequency antigen-specific CD8+ T-cell responses, their use is hampered by widespread pre-existing immunity to human serotypes. Non-human adenovirus serotypes associated with lower prevalence may offer an alternative strategy. We evaluated the immunogenicity of AdC7-GRN ('A'), a recombinant chimpanzee adenovirus type 7 vector expressing clade B Gag, RT and Nef, and F4/AS01 ('P'), when delivered intramuscularly in homologous (PP or AA) and heterologous (AAPP or PPAA) prime-boost regimens, in macaques and mice. Vaccine-induced HIV-1-antigen-specific T cells in peripheral blood (macaques), liver, spleen, and intestinal and genital mucosa (mice) were characterized by intracellular cytokine staining. Vaccine-specific IgG antibodies (macaques) were detected using ELISA. In macaques, only the heterologous prime-boost regimens induced polyfunctional, persistent and balanced CD4+ and CD8+ T-cell responses specific to each HIV-1 vaccine antigen. AdC7-GRN priming increased the polyfunctionality of F4/AS01-induced CD4+ T cells. Approximately 50% of AdC7-GRN-induced memory CD8+ T cells exhibited an effector-memory phenotype. HIV-1-specific antibodies were detected with each regimen. In mice, antigen-specific CD4+ and CD8+ T-cell responses were detected in the mucosal and systemic anatomical compartments assessed. When administered in heterologous prime-boost regimens, AdC7-GRN and F4/AS01 candidate vaccines acted complementarily in inducing potent and persistent peripheral blood HIV-1-specific CD4+ and CD8+ T-cell responses and antibodies in macaques. Besides, adenoviral vector priming modulated the cytokine-expression profile of the protein-induced CD4+ T cells. Each regimen induced HIV-1-specific T-cell responses in systemic/local tissues in mice. This suggests that prime-boost regimens combining adjuvanted protein and low-seroprevalent chimpanzee adenoviral vectors represent an attractive vaccination strategy for clinical evaluation.
Collapse
|
14
|
Vaccine Adjuvant Systems containing monophosphoryl lipid A and QS-21 induce strong humoral and cellular immune responses against hepatitis B surface antigen which persist for at least 4 years after vaccination. Vaccine 2014; 33:1084-91. [PMID: 25444781 DOI: 10.1016/j.vaccine.2014.10.078] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/27/2014] [Accepted: 10/28/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Recombinant hepatitis B surface antigen (HBsAg) was used as a model antigen to evaluate persistence of cellular and humoral immune responses when formulated with three different Adjuvant Systems containing 3-O-desacyl-4'-monophosphoryl lipid A (MPL) and QS-21, in an oil-in-water emulsion (AS02B and AS02V), or with liposomes (AS01B). METHODS This is an open, 4-year follow-up of a previous randomised, double-blind study. Healthy subjects aged 18-40 years received three vaccine doses on a month 0, 1, 10 schedule and were initially followed for 18 months. A total of 93 subjects (AS02B: n=30; AS02V: n=28; AS01B: n=35) were enrolled in this follow-up and had an additional blood sample taken at Year 4 (NCT02153320). The primary endpoint was the frequency of HBsAg-specific CD4(+) and CD8(+) T-cells expressing cytokines upon short-term in vitro stimulation of peripheral blood mononuclear cells with HBsAg-derived peptides. Secondary endpoints were anti-HBs antibody titres and frequency of HBsAg-specific memory B-cells. RESULTS A strong and persistent specific CD4(+) T-cell response was observed at Year 4 in all groups. HBsAg-specific CD4(+) T-cells expressed mainly CD40L and IL-2, and to a lesser extent TNF-α and IFN-γ. HBsAg-specific CD8(+) T-cells were not detected in any group. A high, persistent HBsAg-specific humoral immune response was observed in all groups, with all subjects seroprotected (antibody titre ≥10mIU/mL) at Year 4. The geometric mean antibody titre at Year 4 was above 100,000mIU/mL in all groups. A strong memory B-cell response was observed post-dose 2, which tended to increase post-dose 3 and persisted at Year 4 in all groups. CONCLUSION The MPL/QS-21/HBsAg vaccine formulations induced persistent immune responses up to 4 years after first vaccination. These Adjuvant Systems offer potential for combination with recombinant, synthetic or highly purified subunit vaccines, particularly for vaccination against challenging diseases, or in specific populations, although additional studies are needed.
Collapse
|
15
|
Bourguignon P, Clément F, Renaud F, Le Bras V, Koutsoukos M, Burny W, Moris P, Lorin C, Collard A, Leroux-Roels G, Roman F, Janssens M, Vandekerckhove L. Processing of blood samples influences PBMC viability and outcome of cell-mediated immune responses in antiretroviral therapy-naïve HIV-1-infected patients. J Immunol Methods 2014; 414:1-10. [PMID: 25224748 DOI: 10.1016/j.jim.2014.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 09/01/2014] [Accepted: 09/05/2014] [Indexed: 12/22/2022]
Abstract
Intracellular cytokine staining (ICS) assay is increasingly used in vaccine clinical trials to measure antigen-specific T-cell mediated immune (CMI) responses in cryopreserved peripheral blood mononuclear cells (PBMCs) and whole blood. However, recent observations indicate that several parameters involved in blood processing can impact PBMC viability and CMI responses, especially in antiretroviral therapy (ART)-naïve HIV-1-infected individuals. In this phase I study (NCT01610427), we collected blood samples from 22 ART-naïve HIV-1-infected adults. PBMCs were isolated and processed for ICS assay. The individual and combined effects of the following parameters were investigated: time between blood collection and PBMC processing (time-to-process: 2, 7 or 24 h); time between PBMC thawing and initiation of in vitro stimulation with HIV-1 antigens (resting-time: 0, 2, 6 and 18 h); and duration of antigen-stimulation in PBMC cultures (stimulation-time: 6h or overnight). The cell recovery after thawing, cell viability after ICS and magnitude of HIV-specific CD8(+) T-cell responses were considered to determine the optimal combination of process conditions. The impact of time-to-process (2 or 4 h) on HIV-specific CD8(+) T-cell responses was also assessed in a whole blood ICS assay. A higher quality of cells in terms of recovery and viability (up to 81% and >80% respectively) was obtained with shorter time-to-process (less than 7 h) and resting-time (less than 2 h) intervals. Longer (overnight) rather than shorter (6 h) stimulation-time intervals increased the frequency of CD8(+)-specific T-cell responses using ICS in PBMCs without change of the functionality. The CD8(+) specific T-cell responses detected using fresh whole blood showed a good correlation with the responses detected using frozen PBMCs. Our results support the need of standardized procedures for the evaluation of CMI responses, especially in HIV-1-infected, ART-naïve patients.
Collapse
Affiliation(s)
| | - Frédéric Clément
- Center for Vaccinology, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | - Frédéric Renaud
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Vivien Le Bras
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | | | - Wivine Burny
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Philippe Moris
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Clarisse Lorin
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Alix Collard
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Geert Leroux-Roels
- Center for Vaccinology, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| | - François Roman
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Michel Janssens
- GlaxoSmithKline Vaccines, Rue de l'institut 89, Rixensart 1330, Belgium.
| | - Linos Vandekerckhove
- ARC (AIDS Reference Center), Department of Internal Medicine, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
16
|
Safety and immunogenicity of the M72/AS01 candidate tuberculosis vaccine in HIV-infected adults on combination antiretroviral therapy: a phase I/II, randomized trial. AIDS 2014; 28:1769-81. [PMID: 24911353 DOI: 10.1097/qad.0000000000000343] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Tuberculosis (TB) is highly prevalent among HIV-infected people, including those receiving combination antiretroviral therapy (cART), necessitating a well tolerated and efficacious TB vaccine for these populations. We evaluated the safety and immunogenicity of the candidate TB vaccine M72/AS01 in adults with well controlled HIV infection on cART. DESIGN A randomized, observer-blind, controlled trial (NCT00707967). METHODS HIV-infected adults on cART in Switzerland were randomized 3 : 1 : 1 to receive two doses, 1 month apart, of M72/AS01, AS01 or 0.9% physiological saline (N = 22, N = 8 and N = 7, respectively) and were followed up to 6 months postdose 2 (D210). Individuals with CD4⁺ cell counts below 200 cells/μl were excluded. Adverse events (AEs) including HIV-specific and laboratory safety parameters were recorded. Cell-mediated (ICS) and humoral (ELISA) responses were evaluated before vaccination, 1 month after each dose (D30, D60) and D210. RESULTS Thirty-seven individuals [interquartile range (IQR) CD4⁺ cell counts at screening: 438-872 cells/μl; undetectable HIV-1 viremia] were enrolled; 73% of individuals reported previous BCG vaccination, 97.3% tested negative for the QuantiFERON-TB assay. For M72/AS01 recipients, no vaccine-related serious AEs or cART-regimen adjustments were recorded, and there were no clinically relevant effects on laboratory safety parameters, HIV-1 viral loads or CD4⁺ cell counts. M72/AS01 was immunogenic, inducing persistent and polyfunctional M72-specific CD4⁺ T-cell responses [medians 0.70% (IQR 0.37-1.07) at D60] and 0.42% (0.24-0.61) at D210, predominantly CD40L⁺IL-2⁺TNF-α⁺, CD40L⁺IL-2⁺ and CD40L⁺IL-2⁺TNF-α⁺IFN-γ⁺]. All M72/AS01 vaccines were seropositive for anti-M72 IgG after second vaccination until study end. CONCLUSION M72/AS01 was clinically well tolerated and immunogenic in this population, supporting further clinical evaluation in HIV-infected individuals in TB-endemic settings.
Collapse
|
17
|
Leroux-Roels G, Bourguignon P, Willekens J, Janssens M, Clement F, Didierlaurent AM, Fissette L, Roman F, Boutriau D. Immunogenicity and safety of a booster dose of an investigational adjuvanted polyprotein HIV-1 vaccine in healthy adults and effect of administration of chloroquine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:302-11. [PMID: 24391139 PMCID: PMC3957681 DOI: 10.1128/cvi.00617-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/21/2013] [Indexed: 11/20/2022]
Abstract
This phase II study evaluated the effect of chloroquine on the specific CD8(+) T-cell responses to and the safety of a booster dose of investigational human immunodeficiency virus type 1 (HIV-1) F4/AS01(B) vaccine containing 10 μg of recombinant fusion protein (F4) adjuvanted with the AS01(B) adjuvant system. Healthy adults aged 21 to 41 years, primed 3 years before with two F4/AS01(B) doses containing 10 or 30 μg of F4 (ClinicalTrials.gov registration number NCT00434512), were randomized (1:1) to receive the F4/AS01(B) booster administered alone or 2 days after chloroquine (300 mg). F4-specific CD8(+)/CD4(+) T-cell responses were characterized by intracellular cytokine staining and lymphoproliferation assays and anti-F4 antibodies by enzyme-linked immunosorbent assays (ELISAs). No effect of chloroquine on CD4(+)/CD8(+) T-cell and antibody responses and no vaccine effect on CD8(+) T-cell responses (cytokine secretion or proliferation) were detected following F4/AS01(B) booster administration. In vitro, chloroquine had a direct inhibitory effect on AS01(B) adjuvant properties; AS01-induced cytokine production decreased upon coincubation of cells with chloroquine. In the pooled group of participants primed with F4/AS01(B) containing 10 μg of F4, CD4(+) T-cell and antibody responses induced by primary vaccination persisted for at least 3 years. The F4/AS01(B) booster induced strong F4-specific CD4(+) T-cell responses, which persisted for at least 6 months with similar frequencies and polyfunctional phenotypes as following primary vaccination, and high anti-F4 antibody concentrations, reaching higher levels than those following primary vaccination. The F4/AS01(B) booster had a clinically acceptable safety and reactogenicity profile. An F4/AS01(B) booster dose, administered alone or after chloroquine, induced robust antibody and F4-specific CD4(+) T-cell responses but no significant CD8(+) T-cell responses (cytokine secretion or proliferation) in healthy adults. (This study has been registered at ClinicalTrials.gov under registration number NCT00972725).
Collapse
|