1
|
Lorenzo MM, Marín-López A, Chiem K, Jimenez-Cabello L, Ullah I, Utrilla-Trigo S, Calvo-Pinilla E, Lorenzo G, Moreno S, Ye C, Park JG, Matía A, Brun A, Sánchez-Puig JM, Nogales A, Mothes W, Uchil PD, Kumar P, Ortego J, Fikrig E, Martinez-Sobrido L, Blasco R. Vaccinia Virus Strain MVA Expressing a Prefusion-Stabilized SARS-CoV-2 Spike Glycoprotein Induces Robust Protection and Prevents Brain Infection in Mouse and Hamster Models. Vaccines (Basel) 2023; 11:1006. [PMID: 37243110 PMCID: PMC10220993 DOI: 10.3390/vaccines11051006] [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: 04/14/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The COVID-19 pandemic has underscored the importance of swift responses and the necessity of dependable technologies for vaccine development. Our team previously developed a fast cloning system for the modified vaccinia virus Ankara (MVA) vaccine platform. In this study, we reported on the construction and preclinical testing of a recombinant MVA vaccine obtained using this system. We obtained recombinant MVA expressing the unmodified full-length SARS-CoV-2 spike (S) protein containing the D614G amino-acid substitution (MVA-Sdg) and a version expressing a modified S protein containing amino-acid substitutions designed to stabilize the protein a in a pre-fusion conformation (MVA-Spf). S protein expressed by MVA-Sdg was found to be expressed and was correctly processed and transported to the cell surface, where it efficiently produced cell-cell fusion. Version Spf, however, was not proteolytically processed, and despite being transported to the plasma membrane, it failed to induce cell-cell fusion. We assessed both vaccine candidates in prime-boost regimens in the susceptible transgenic K18-human angiotensin-converting enzyme 2 (K18-hACE2) in mice and in golden Syrian hamsters. Robust immunity and protection from disease was induced with either vaccine in both animal models. Remarkably, the MVA-Spf vaccine candidate produced higher levels of antibodies, a stronger T cell response, and a higher degree of protection from challenge. In addition, the level of SARS-CoV-2 in the brain of MVA-Spf inoculated mice was decreased to undetectable levels. Those results add to our current experience and range of vaccine vectors and technologies for developing a safe and effective COVID-19 vaccine.
Collapse
Affiliation(s)
- María M. Lorenzo
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Alejandro Marín-López
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Kevin Chiem
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Luis Jimenez-Cabello
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Irfan Ullah
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Sergio Utrilla-Trigo
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Sandra Moreno
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Alejandro Matía
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Alejandro Brun
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Juana M. Sánchez-Puig
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Aitor Nogales
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA; (W.M.); (P.D.U.)
| | - Pradeep D. Uchil
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA; (W.M.); (P.D.U.)
| | - Priti Kumar
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Erol Fikrig
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Luis Martinez-Sobrido
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Rafael Blasco
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| |
Collapse
|
2
|
Gómez CE, Perdiguero B, García-Arriaza J, Cepeda V, Sánchez-Sorzano CÓ, Mothe B, Jiménez JL, Muñoz-Fernández MÁ, Gatell JM, López Bernaldo de Quirós JC, Brander C, García F, Esteban M. A Phase I Randomized Therapeutic MVA-B Vaccination Improves the Magnitude and Quality of the T Cell Immune Responses in HIV-1-Infected Subjects on HAART. PLoS One 2015; 10:e0141456. [PMID: 26544853 PMCID: PMC4636254 DOI: 10.1371/journal.pone.0141456] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/08/2015] [Indexed: 11/29/2022] Open
Abstract
Trial Design Previous studies suggested that poxvirus-based vaccines might be instrumental in the therapeutic HIV field. A phase I clinical trial was conducted in HIV-1-infected patients on highly active antiretroviral therapy (HAART), with CD4 T cell counts above 450 cells/mm3 and undetectable viremia. Thirty participants were randomized (2:1) to receive either 3 intramuscular injections of MVA-B vaccine (coding for clade B HIV-1 Env, Gag, Pol and Nef antigens) or placebo, followed by interruption of HAART. Methods The magnitude, breadth, quality and phenotype of the HIV-1-specific T cell response were assayed by intracellular cytokine staining (ICS) in 22 volunteers pre- and post-vaccination. Results MVA-B vaccine induced newly detected HIV-1-specific CD4 T cell responses and expanded pre-existing responses (mostly against Gag, Pol and Nef antigens) that were high in magnitude, broadly directed and showed an enhanced polyfunctionality with a T effector memory (TEM) phenotype, while maintaining the magnitude and quality of the pre-existing HIV-1-specific CD8 T cell responses. In addition, vaccination also triggered preferential CD8+ T cell polyfunctional responses to the MVA vector antigens that increase in magnitude after two and three booster doses. Conclusion MVA-B vaccination represents a feasible strategy to improve T cell responses in individuals with pre-existing HIV-1-specific immunity. Trial Registration ClinicalTrials.gov NCT01571466
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Victoria Cepeda
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carlos Óscar Sánchez-Sorzano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Beatriz Mothe
- IrsiCaixa-HIVACAT, Hospital Universitari Germans Trias i Pujol, Autonomous University of Barcelona, Badalona, Spain
| | | | | | | | | | - Christian Brander
- IrsiCaixa-HIVACAT, Hospital Universitari Germans Trias i Pujol, Autonomous University of Barcelona, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, and University of Vic and Central Catalonia, Vic, Spain
| | | | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
3
|
Mothe B, Climent N, Plana M, Rosàs M, Jiménez JL, Muñoz-Fernández MÁ, Puertas MC, Carrillo J, Gonzalez N, León A, Pich J, Arnaiz JA, Gatell JM, Clotet B, Blanco J, Alcamí J, Martinez-Picado J, Alvarez-Fernández C, Sánchez-Palomino S, Guardo AC, Peña J, Benito JM, Rallón N, Gómez CE, Perdiguero B, García-Arriaza J, Esteban M, López Bernaldo de Quirós JC, Brander C, García F. Safety and immunogenicity of a modified vaccinia Ankara-based HIV-1 vaccine (MVA-B) in HIV-1-infected patients alone or in combination with a drug to reactivate latent HIV-1. J Antimicrob Chemother 2015; 70:1833-42. [PMID: 25724985 DOI: 10.1093/jac/dkv046] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/03/2015] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES The safety, immunogenicity, impact on the latent reservoir and rebound of viral load after therapeutic HIV-1 vaccination with recombinant modified vaccinia Ankara-based (MVA-B) HIV-1 vaccine expressing monomeric gp120 and the fused Gag-Pol-Nef polyprotein of clade B with or without a drug to reactivate latent HIV-1 (disulfiram) were assessed. METHODS HIV-1-infected patients were randomized to receive three injections of MVA-B (n = 20) or placebo (n = 10). Twelve patients (eight who received vaccine and four who were given placebo) received a fourth dose of MVA-B followed by 3 months of disulfiram. Combined ART (cART) was discontinued 8 weeks after the last dose of MVA-B. Clinical Trials.gov identifier: NCT01571466. RESULTS MVA-B was safe and well tolerated. A minor, but significant, increase in the T cell responses targeting vaccine inserts of Gag was observed [a median of 290, 403 and 435 spot-forming-cells/10(6) PBMCs at baseline, after two vaccinations and after three vaccinations, respectively; P = 0.02 and P = 0.04]. After interruption of cART, a modest delay in the rebound of the plasma viral load in participants receiving vaccine but not disulfiram was observed compared with placebo recipients (P = 0.01). The dynamics of the viral load rebound did not change in patients receiving MVA-B/disulfiram. No changes in the proviral reservoir were observed after disulfiram treatment. CONCLUSIONS MVA-B vaccination was a safe strategy to increase Gag-specific T cell responses in chronically HIV-1-infected individuals, but it did not have a major impact on the latent reservoir or the rebound of plasma viral load after interruption of cART when given alone or in combination with disulfiram.
Collapse
Affiliation(s)
- Beatriz Mothe
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain 'Lluita contra la Sida' Foundation, Hospital Germans Trias i Pujol, Badalona, Spain Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain
| | - Nuria Climent
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Montserrat Plana
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Miriam Rosàs
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain
| | | | | | - María C Puertas
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Jorge Carrillo
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Nuria Gonzalez
- AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III, Madrid, Spain
| | - Agathe León
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Judit Pich
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Joan Albert Arnaiz
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Jose M Gatell
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Bonaventura Clotet
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain 'Lluita contra la Sida' Foundation, Hospital Germans Trias i Pujol, Badalona, Spain Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain
| | - Julià Blanco
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain
| | - José Alcamí
- AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Martinez-Picado
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain Universitat Autònoma de Barcelona, Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | | | | | - Alberto C Guardo
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - José Peña
- Hospital Reina Sofía, Córdoba, Spain
| | - José M Benito
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Barcelona, Madrid, Spain
| | - Norma Rallón
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Barcelona, Madrid, Spain
| | | | | | | | | | | | - Christian Brander
- Irsicaixa AIDS Research Institute-HIVACAT, Hospital Germans Trias i Pujol, Badalona, Spain Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Spain Universitat Autònoma de Barcelona, Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Felipe García
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
4
|
Characterization of T-cell responses to conserved regions of the HIV-1 proteome in BALB/c mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:1565-72. [PMID: 25230940 PMCID: PMC4248756 DOI: 10.1128/cvi.00587-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A likely requirement for a protective vaccine against human immunodeficiency virus type 1 (HIV-1)/AIDS is, in addition to eliciting antibody responses, induction of effective T cells. To tackle HIV-1 diversity by T-cell vaccines, we designed an immunogen, HIVconsv, derived from the most functionally conserved regions of the HIV-1 proteome and demonstrated its high immunogenicity in humans and rhesus macaques when delivered by regimens combining plasmid DNA, nonreplicating simian (chimpanzee) adenovirus ChAdV-63, and nonreplicating modified vaccinia virus Ankara (MVA) as vectors. Here, we aimed to increase the decision power for iterative improvements of this vaccine strategy in the BALB/c mouse model. First, we found that prolonging the period after the ChAdV63.HIVconsv prime up to 6 weeks increased the frequencies of HIV-1-specific, gamma interferon (IFN-γ)-producing T cells induced by the MVA.HIVconsv boost. Induction of strong responses allowed us to map comprehensively the H-2d-restricted T-cell responses to these regions and identified 8 HIVconsv peptides, of which three did not contain a previously described epitope and were therefore considered novel. Induced effector T cells were oligofunctional and lysed sensitized targets in vitro. Our study therefore provides additional tools for studying and optimizing vaccine regimens in this commonly used small animal model, which will in turn guide vaccine improvements in more expensive nonhuman primate and human clinical trials.
Collapse
|
5
|
Ondondo BO. The influence of delivery vectors on HIV vaccine efficacy. Front Microbiol 2014; 5:439. [PMID: 25202303 PMCID: PMC4141443 DOI: 10.3389/fmicb.2014.00439] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/03/2014] [Indexed: 12/31/2022] Open
Abstract
Development of an effective HIV/AIDS vaccine remains a big challenge, largely due to the enormous HIV diversity which propels immune escape. Thus novel vaccine strategies are targeting multiple variants of conserved antibody and T cell epitopic regions which would incur a huge fitness cost to the virus in the event of mutational escape. Besides immunogen design, the delivery modality is critical for vaccine potency and efficacy, and should be carefully selected in order to not only maximize transgene expression, but to also enhance the immuno-stimulatory potential to activate innate and adaptive immune systems. To date, five HIV vaccine candidates have been evaluated for efficacy and protection from acquisition was only achieved in a small proportion of vaccinees in the RV144 study which used a canarypox vector for delivery. Conversely, in the STEP study (HVTN 502) where human adenovirus serotype 5 (Ad5) was used, strong immune responses were induced but vaccination was more associated with increased risk of HIV acquisition than protection in vaccinees with pre-existing Ad5 immunity. The possibility that pre-existing immunity to a highly promising delivery vector may alter the natural course of HIV to increase acquisition risk is quite worrisome and a huge setback for HIV vaccine development. Thus, HIV vaccine development efforts are now geared toward delivery platforms which attain superior immunogenicity while concurrently limiting potential catastrophic effects likely to arise from pre-existing immunity or vector-related immuno-modulation. However, it still remains unclear whether it is poor immunogenicity of HIV antigens or substandard immunological potency of the safer delivery vectors that has limited the success of HIV vaccines. This article discusses some of the promising delivery vectors to be harnessed for improved HIV vaccine efficacy.
Collapse
Affiliation(s)
- Beatrice O Ondondo
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford Oxford, UK
| |
Collapse
|
6
|
Iyer SS, Amara RR. DNA/MVA Vaccines for HIV/AIDS. Vaccines (Basel) 2014; 2:160-78. [PMID: 26344473 PMCID: PMC4494194 DOI: 10.3390/vaccines2010160] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 01/31/2014] [Accepted: 02/06/2014] [Indexed: 11/16/2022] Open
Abstract
Since the initial proof-of-concept studies examining the ability of antigen-encoded plasmid DNA to serve as an immunogen, DNA vaccines have evolved as a clinically safe and effective platform for priming HIV-specific cellular and humoral responses in heterologous "prime-boost" vaccination regimens. Direct injection of plasmid DNA into the muscle induces T- and B-cell responses against foreign antigens. However, the insufficient magnitude of this response has led to the development of approaches for enhancing the immunogenicity of DNA vaccines. The last two decades have seen significant progress in the DNA-based vaccine platform with optimized plasmid constructs, improved delivery methods, such as electroporation, the use of molecular adjuvants and novel strategies combining DNA with viral vectors and subunit proteins. These innovations are paving the way for the clinical application of DNA-based HIV vaccines. Here, we review preclinical studies on the DNA-prime/modified vaccinia Ankara (MVA)-boost vaccine modality for HIV. There is a great deal of interest in enhancing the immunogenicity of DNA by engineering DNA vaccines to co-express immune modulatory adjuvants. Some of these adjuvants have demonstrated encouraging results in preclinical and clinical studies, and these data will be examined, as well.
Collapse
Affiliation(s)
- Smita S Iyer
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA.
| | - Rama R Amara
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA.
| |
Collapse
|
7
|
Afolabi MO, Ndure J, Drammeh A, Darboe F, Mehedi SR, Rowland-Jones SL, Borthwick N, Black A, Ambler G, John-Stewart GC, Reilly M, Hanke T, Flanagan KL. A phase I randomized clinical trial of candidate human immunodeficiency virus type 1 vaccine MVA.HIVA administered to Gambian infants. PLoS One 2013; 8:e78289. [PMID: 24205185 PMCID: PMC3813444 DOI: 10.1371/journal.pone.0078289] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 09/07/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND A vaccine to decrease transmission of human immunodeficiency virus type 1 (HIV-1) during breast-feeding would complement efforts to eliminate infant HIV-1 infection by antiretroviral therapy. Relative to adults, infants have distinct immune development, potentially high-risk of transmission when exposed to HIV-1 and rapid progression to AIDS when infected. To date, there have been only three published HIV-1 vaccine trials in infants. TRIAL DESIGN We conducted a randomized phase I clinical trial PedVacc 001 assessing the feasibility, safety and immunogenicity of a single dose of candidate vaccine MVA.HIVA administered intramuscularly to 20-week-old infants born to HIV-1-negative mothers in The Gambia. METHODS Infants were followed to 9 months of age with assessment of safety, immunogenicity and interference with Expanded Program on Immunization (EPI) vaccines. The trial is the first stage of developing more complex prime-boost vaccination strategies against breast milk transmission of HIV-1. RESULTS From March to October 2010, 48 infants (24 vaccine and 24 no-treatment) were enrolled with 100% retention. The MVA.HIVA vaccine was safe with no difference in adverse events between vaccinees and untreated infants. Two vaccine recipients (9%) and no controls had positive ex vivo interferon-γ ELISPOT assay responses. Antibody levels elicited to the EPI vaccines, which included diphtheria, tetanus, whole-cell pertussis, hepatitis B virus, Haemophilus influenzae type b and oral poliovirus, reached protective levels for the vast majority and were similar between the two arms. CONCLUSIONS A single low-dose of MVA.HIVA administered to 20-week-old infants in The Gambia was found to be safe and without interference with the induction of protective antibody levels by EPI vaccines, but did not alone induce sufficient HIV-1-specific responses. These data support the use of MVA carrying other transgenes as a boosting vector within more complex prime-boost vaccine strategies against transmission of HIV-1 and/or other infections in this age group. TRIAL REGISTRATION ClinicalTrials.gov NCT00982579. The Pan African Clinical Trials Registry PACTR2008120000904116.
Collapse
Affiliation(s)
| | - Jorjoh Ndure
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Abdoulie Drammeh
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Fatoumatta Darboe
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Shams-Rony Mehedi
- Statistics and Data Management Department, Medical Research Council Unit, Fajara, The Gambia
| | | | - Nicola Borthwick
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Antony Black
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Gwen Ambler
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Grace C. John-Stewart
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Marie Reilly
- Department of Medical Epidemiology and Biostatistics, Stockholm, Sweden
| | - Tomáš Hanke
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Katie L. Flanagan
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| |
Collapse
|
8
|
Greenberg RN, Overton ET, Haas DW, Frank I, Goldman M, von Krempelhuber A, Virgin G, Bädeker N, Vollmar J, Chaplin P. Safety, immunogenicity, and surrogate markers of clinical efficacy for modified vaccinia Ankara as a smallpox vaccine in HIV-infected subjects. J Infect Dis 2012; 207:749-58. [PMID: 23225902 DOI: 10.1093/infdis/jis753] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-infected persons are at higher risk for serious complications associated with traditional smallpox vaccines. Alternative smallpox vaccines with an improved safety profile would address this unmet medical need. METHODS The safety and immunogenicity of modified vaccinia Ankara (MVA) was assessed in 91 HIV-infected adult subjects (CD4(+) T-cell counts, ≥350 cells/mm(3)) and 60 uninfected volunteers. The primary objectives were to evaluate the safety of MVA and immunogenicity in HIV-infected and uninfected subjects. As a measure of the potential efficacy of MVA, the ability to boost the memory response in people previously vaccinated against smallpox was evaluated by the inclusion of vaccinia-experienced HIV-infected and HIV-uninfected subjects. RESULTS MVA was well tolerated and immunogenic in all subjects. Antibody responses were comparable between uninfected and HIV-infected populations, with only 1 significantly lower total antibody titer at 2 weeks after the second vaccination, while no significant differences were observed for neutralizing antibodies. MVA rapidly boosted the antibody responses in vaccinia-experienced subjects, supporting the efficacy of MVA against variola. CONCLUSIONS MVA is a promising candidate as a safer smallpox vaccine, even for immunocompromised individuals, a group for whom current smallpox vaccines have an unacceptable safety profile.
Collapse
Affiliation(s)
- Richard N Greenberg
- Infectious Diseases Division, University of Kentucky School of Medicine, Lexington, KY 40536-0093, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Sanou MP, De Groot AS, Murphey-Corb M, Levy JA, Yamamoto JK. HIV-1 Vaccine Trials: Evolving Concepts and Designs. Open AIDS J 2012; 6:274-88. [PMID: 23289052 PMCID: PMC3534440 DOI: 10.2174/1874613601206010274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/18/2012] [Accepted: 09/20/2012] [Indexed: 12/24/2022] Open
Abstract
An effective prophylactic HIV-1 vaccine is needed to eradicate the HIV/AIDS pandemic but designing such a vaccine is a challenge. Despite many advances in vaccine technology and approaches to generate both humoral and cellular immune responses, major phase-II and -III vaccine trials against HIV/AIDS have resulted in only moderate successes. The modest achievement of the phase-III RV144 prime-boost trial in Thailand re-emphasized the importance of generating robust humoral and cellular responses against HIV. While antibody-directed approaches are being pursued by some groups, others are attempting to develop vaccines targeting cell-mediated immunity, since evidence show CTLs to be important for the control of HIV replication. Phase-I and -IIa multi-epitope vaccine trials have already been conducted with vaccine immunogens consisting of known CTL epitopes conserved across HIV subtypes, but have so far fallen short of inducing robust and consistent anti-HIV CTL responses. The concepts leading to the development of T-cell epitope-based vaccines, the outcomes of related clinical vaccine trials and efforts to enhance the immunogenicity of cell-mediated approaches are summarized in this review. Moreover, we describe a novel approach based on the identification of SIV and FIV antigens which contain conserved HIV-specific T-cell epitopes and represent an alternative method for developing an effective HIV vaccine against global HIV isolates.
Collapse
Affiliation(s)
- Missa P Sanou
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611, USA
| | - Anne S De Groot
- EpiVax Inc., University of Rhode Island, Providence, RI 02903, USA
| | - Michael Murphey-Corb
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, E1252 Biomedical Science Tower 200, Lothrop Street, Pittsburgh, PA 15261, USA
| | - Jay A Levy
- Department of Medicine, University of California San Francisco, S-1280, 513 Parnassus Ave, San Francisco, CA 94143, USA
| | - Janet K Yamamoto
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611, USA
| |
Collapse
|
10
|
García-Arriaza J, Arnáez P, Jiménez JL, Gómez CE, Muñoz-Fernández MÁ, Esteban M. Vector replication and expression of HIV-1 antigens by the HIV/AIDS vaccine candidate MVA-B is not affected by HIV-1 protease inhibitors. Virus Res 2012; 167:391-6. [PMID: 22659488 DOI: 10.1016/j.virusres.2012.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 05/22/2012] [Accepted: 05/23/2012] [Indexed: 02/07/2023]
Abstract
MVA-B is an attenuated poxvirus vector expressing human immunodeficiency virus type 1 Env, Gag, Pol, and Nef antigens from clade B, and is considered a promising HIV/AIDS vaccine candidate. Recently, a phase I clinical trial in human healthy volunteers has shown that MVA-B is safe and highly immunogenic, inducing broad, polyfunctional, and long-lasting CD4(+) and CD8(+) T cell responses to HIV-1 antigens, with preference for effector memory T cells; and it also triggers the induction of specific antibodies to Env in most of the vaccines. While MVA recombinants expressing HIV-1 antigens are being used or plan to use in therapeutic clinical trials, little is known on the effect of HIV-1 highly active antiretroviral therapy in MVA life cycle. To define this role, here we have evaluated in established cell cultures and human dendritic cells to what extent different HIV-1 protease inhibitors affect virus replication and expression of HIV-1 antigens during MVA-B infection. The results obtained revealed that the most commonly used HIV-1 protease inhibitors (atazanavir, ritonavir, and lopinavir) had no effect on MVA-B virus growth kinetics, even at higher concentrations than those normally used on HAART. Furthermore, expression of gp120 and the fused Gag-Pol-Nef polyprotein in permissive and non-permissive cells infected with MVA-B were also not affected. These findings are relevant information for the therapeutic use of MVA-B as an HIV-1/AIDS vaccine.
Collapse
Affiliation(s)
- Juan García-Arriaza
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
11
|
Lisziewicz J, Bakare N, Calarota SA, Bánhegyi D, Szlávik J, Ujhelyi E, Tőke ER, Molnár L, Lisziewicz Z, Autran B, Lori F. Single DermaVir immunization: dose-dependent expansion of precursor/memory T cells against all HIV antigens in HIV-1 infected individuals. PLoS One 2012; 7:e35416. [PMID: 22590502 PMCID: PMC3348904 DOI: 10.1371/journal.pone.0035416] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 03/14/2012] [Indexed: 11/25/2022] Open
Abstract
Background The GIHU004 study was designed to evaluate the safety and immunogenicity of three doses of DermaVir immunization in HIV-infected subjects on fully suppressive combination antiretroviral therapy (cART). Methodology/Principal Findings This first-in-human dose escalation study was conducted with three topical DermaVir doses targeted to epidermal Langerhans cells to express fifteen HIV antigens in draining lymph nodes: 0.1 mg DNA targeted to two, 0.4 mg and 0.8 mg DNA targeted to four lymph nodes. Particularly, in the medium dose cohort 0.1 mg DNA was targeted per draining lymph node via ∼8 million Langerhans cells located in 80 cm2 epidermis area. The 28-days study with 48-week safety follow-up evaluated HIV-specific T cell responses against Gag p17, Gag p24 and Gag p15, Tat and Rev antigens. DermaVir-associated side effects were mild, transient and not dose-dependent. Boosting of HIV-specific effector CD4+ and CD8+ T cells expressing IFN-gamma and IL-2 was detected against several antigens in every subject of the medium dose cohort. The striking result was the dose-dependent expansion of HIV-specific precursor/memory T cells with high proliferation capacity. In low, medium and high dose cohorts this HIV-specific T cell population increased by 325-, 136,202 and 50,759 counts after 4 weeks, and by 3,899, 9,878 and 18,382 counts after one year, respectively, compared to baseline. Conclusions/Significance Single immunization with the DermaVir candidate therapeutic vaccine was safe and immunogenic in HIV-infected individuals. Based on the potent induction of Gag, Tat and Rev-specific memory T cells, especially in the medium dose cohort, we speculate that DermaVir boost T cell responses specific to all the 15 HIV antigens expressed from the single DNA. For durable immune reactivity repeated DermaVir immunization might be required since the frequency of DermaVir-boosted HIV-specific memory T cells decreased during the 48-week follow up. Trial Registration ClinicalTrial.gov NCT00712530.
Collapse
Affiliation(s)
- Julianna Lisziewicz
- Genetic Immunity, Budapest, Hungary and Genetic Immunity Inc, Mclean, Virginia, United States of America.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Guzman E, Cubillos-Zapata C, Cottingham MG, Gilbert SC, Prentice H, Charleston B, Hope JC. Modified vaccinia virus Ankara-based vaccine vectors induce apoptosis in dendritic cells draining from the skin via both the extrinsic and intrinsic caspase pathways, preventing efficient antigen presentation. J Virol 2012; 86:5452-66. [PMID: 22419811 PMCID: PMC3347273 DOI: 10.1128/jvi.00264-12] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/05/2012] [Indexed: 02/03/2023] Open
Abstract
Dendritic cells (DC) are potent antigen-presenting cells and central to the induction of immune responses following infection or vaccination. The collection of DC migrating from peripheral tissues by cannulation of the afferent lymphatic vessels provides DC which can be used directly ex vivo without extensive in vitro manipulations. We have previously used bovine migrating DC to show that recombinant human adenovirus 5 vectors efficiently transduce afferent lymph migrating DEC-205(+) CD11c(+) CD8(-) DC (ALDC). We have also shown that recombinant modified vaccinia virus Ankara (MVA) infects ALDC in vitro, causing downregulation of costimulatory molecules, apoptosis, and cell death. We now show that in the bovine system, modified vaccinia virus Ankara-induced apoptosis in DC draining from the skin occurs soon after virus binding via the caspase 8 pathway and is not associated with viral gene expression. We also show that after virus entry, the caspase 9 pathway cascade is initiated. The magnitude of T cell responses to mycobacterial antigen 85A (Ag85A) expressed by recombinant MVA-infected ALDC is increased by blocking caspase-induced apoptosis. Apoptotic bodies generated by recombinant MVA (rMVA)-Ag85A-infected ALDC and containing Ag85A were phagocytosed by noninfected migrating ALDC expressing SIRPα via actin-dependent phagocytosis, and these ALDC in turn presented antigen. However, the addition of fresh ALDC to MVA-infected cultures did not improve on the magnitude of the T cell responses; in contrast, these noninfected DC showed downregulation of major histocompatibility complex class II (MHC-II), CD40, CD80, and CD86. We also observed that MVA-infected ALDC promoted migration of DEC-205(+) SIRPα(+) CD21(+) DC as well as CD4(+) and CD8(+) T cells independently of caspase activation. These in vitro studies show that induction of apoptosis in DC by MVA vectors is detrimental to the subsequent induction of T cell responses.
Collapse
Affiliation(s)
- E Guzman
- Institute for Animal Health, Compton, Newbury, Berkshire, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
13
|
García F, León A, Gatell JM, Plana M, Gallart T. Therapeutic vaccines against HIV infection. Hum Vaccin Immunother 2012; 8:569-81. [PMID: 22634436 DOI: 10.4161/hv.19555] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Resistance to medication, adverse effects in the medium-to-long-term and cost all place important limitations on lifelong adherence to combined antiretroviral therapy (cART). In this context, new therapeutic alternatives to 'cART for life' in HIV-infected patients merit investigation. Some data suggest that strong T cell-mediated immunity to HIV can indeed limit virus replication and protect against CD4 depletion and disease progression. The combination of cART with immune therapy to restore and/or boost immune-specific responses to HIV has been proposed, the ultimate aim being to achieve a 'functional cure'. In this scenario, new, induced, HIV-specific immune responses would be able to control viral replication to undetectable levels, mimicking the situation of the minority of patients who control viral replication without treatment and do not progress to AIDS. Classical approaches such as whole inactivated virus or recombinant protein initially proved useful as therapeutic vaccines. Overall, however, the ability of these early vaccines to increase HIV-specific responses was very limited and study results were discouraging, as no consistent immunogenicity was demonstrated and there was no clear impact on viral load. Recent years have seen the development of new approaches based on more innovative vectors such as DNA, recombinant virus or dendritic cells. Most clinical trials of these new vectors have demonstrated their ability to induce HIV-specific immune responses, although they show very limited efficacy in terms of controlling viral replication. However, some preliminary results suggest that dendritic cell-based vaccines are the most promising candidates. To improve the effectiveness of these vaccines, a better understanding of the mechanisms of protection, virological control and immune deterioration is required; without this knowledge, an efficacious therapeutic vaccine will remain elusive.
Collapse
Affiliation(s)
- Felipe García
- Hospital Clinic-HIVACAT, IDIBAPS, University of Barcelona, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
14
|
Chung J, Rossi JJ, Jung U. Current progress and challenges in HIV gene therapy. Future Virol 2011; 6:1319-1328. [PMID: 22754586 DOI: 10.2217/fvl.11.113] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
HIV-1 causes AIDS, a syndrome that affects millions of people globally. Existing HAART is efficient in slowing down disease progression but cannot eradicate the virus. Furthermore the severity of the side effects and the emergence of drug-resistant mutants call for better therapy. Gene therapy serves as an attractive alternative as it reconstitutes the immune system with HIV-resistant cells and could thereby provide a potential cure. The feasibility of this approach was first demonstrated with the 'Berlin patient', who was functionally cured from HIV/AIDS with undetectable HIV-1 viral load after transplantation of bone marrow harboring a naturally occurring CCR5 mutation that blocks viral entry. Here, we give an overview of the current status of HIV gene therapy and remaining challenges and obstacles.
Collapse
Affiliation(s)
- Janet Chung
- Division of Molecular & Cell Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, CA 91010, USA
| | | | | |
Collapse
|
15
|
Comprehensive analysis of virus-specific T-cells provides clues for the failure of therapeutic immunization with ALVAC-HIV vaccine. AIDS 2011; 25:27-36. [PMID: 21076273 DOI: 10.1097/qad.0b013e328340fe55] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND HIV-specific T-cell-based vaccines have been extensively studied in both prevention and therapeutic settings, with most studies failing to show benefit, and some suggesting harm. We previously performed a multicenter, double-blind, placebo-controlled phase II clinical trial in which 65 antiretroviral-treated patients were randomized to receive an HIV-1 recombinant canarypox vaccine (vCP1452) or placebo, followed by analytical treatment interruption. Patients exposed to vaccine had higher levels of viral replication and more rapid time to treatment resumption. OBJECTIVE In the present study we report the results from extensive immunological investigations to test whether the preferential expansion of HIV-specific CD4(+), rather than CD8(+) T cells, could account for these unexpected results. METHODS Polychromatic flow cytometry was used to characterize the functional and phenotypic profile of antigen-specific CD8(+) and CD4(+) T cells induced by the immunization. RESULTS We found a significant increase in HIV-specific CD4(+) T cells producing IFN-γ and IL-2 in the 4 injections arm compared to the placebo arm following vaccination. In contrast, no difference was observed following vaccination in the phenotype and functional capacity within the CD8(+) T-cell compartment. Neither HLA biases, nor immune hyper-activation, or Env-specific facilitating antibodies were associated with the enhanced virus rebound observed in vaccinees. CONCLUSION Our data suggest that a vaccine-induced transient activation of HIV-specific CD4(+) but not CD8(+) T cells may have a detrimental effect on HIV outcomes. These findings may provide a mechanistic basis for higher rates of HIV acquisition or replication that have been associated with some T-cell vaccines.
Collapse
|
16
|
Cafaro A, Macchia I, Maggiorella MT, Titti F, Ensoli B. Innovative approaches to develop prophylactic and therapeutic vaccines against HIV/AIDS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 655:189-242. [PMID: 20047043 DOI: 10.1007/978-1-4419-1132-2_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The acquired immunodeficiency syndrome (AIDS) emerged in the human population in the summer of 1981. According to the latest United Nations estimates, worldwide over 33 million people are infected with human immunodeficiency virus (HIV) and the prevalence rates continue to rise globally. To control the alarming spread of HIV, an urgent need exists for developing a safe and effective vaccine that prevents individuals from becoming infected or progressing to disease. To be effective, an HIV/AIDS vaccine should induce broad and long-lasting humoral and cellular immune responses, at both mucosal and systemic level. However, the nature of protective immune responses remains largely elusive and this represents one of the major roadblocks preventing the development of an effective vaccine. Here we summarize our present understanding of the factors responsible for resistance to infection or control of progression to disease in human and monkey that may be relevant to vaccine development and briefly review recent approaches which are currently being tested in clinical trials. Finally, the rationale and the current status of novel strategies based on nonstructural HIV-1 proteins, such as Tat, Nef and Rev, used alone or in combination with modified structural HIV-1 Env proteins are discussed.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, V.le Regina Elena 299, 00161, Rome, Italy
| | | | | | | | | |
Collapse
|
17
|
Kutscher S, Allgayer S, Dembek CJ, Bogner JR, Protzer U, Goebel FD, Erfle V, Cosma A. MVA-nef induces HIV-1-specific polyfunctional and proliferative T-cell responses revealed by the combination of short- and long-term immune assays. Gene Ther 2010; 17:1372-83. [DOI: 10.1038/gt.2010.90] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
18
|
Ondondo BO, Rowland-Jones SL, Dorrell L, Peterson K, Cotten M, Whittle H, Jaye A. Comprehensive analysis of HIV Gag-specific IFN-gamma response in HIV-1- and HIV-2-infected asymptomatic patients from a clinical cohort in The Gambia. Eur J Immunol 2009; 38:3549-60. [PMID: 19016530 DOI: 10.1002/eji.200838759] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Majority of HIV-2-infected individuals meet the criteria of long-term non-progressors. This has been linked to superior qualitative HIV-2-specific cellular immune responses that correlate with viral control. However, it is unknown whether this is due to frequent targeting of immunodominant Gag epitopes in HIV-2 than HIV-1 infection. We describe a comprehensive comparison of the magnitude, breadth and frequency of Gag responses and the degree of cross-recognition of frequently targeted, immunodominant Gag peptides in a cross-sectional study of asymptomatic HIV-1- and HIV-2-infected individuals. Fresh PBMC from 20 HIV-1- and 20 HIV-2-infected patients with similar CD4(+) T-cell counts (p=0.36) were stimulated with pools of HIV-1 and/or HIV-2 Gag peptides in an IFN-gamma ELISPOT assay. We found no difference in the cumulative magnitude of IFN-gamma responses (p=0.75) despite significantly lower plasma viral loads in HIV-2-infected people (p<0.0001). However, Gag211-290 was targeted with significantly higher magnitude in HIV-2-infected subjects (p=0.03) although this did not correlate with viral control. There was no difference in frequently targeted Gag peptides, the breadth, immunodominance or cross-recognition of Gag peptide pools between the two infections. This suggests that other factors may control viral replication in HIV-2 infection.
Collapse
Affiliation(s)
- Beatrice O Ondondo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK.
| | | | | | | | | | | | | |
Collapse
|
19
|
Greenough TC, Cunningham CK, Muresan P, McManus M, Persaud D, Fenton T, Barker P, Gaur A, Panicali D, Sullivan JL, Luzuriaga K. Safety and immunogenicity of recombinant poxvirus HIV-1 vaccines in young adults on highly active antiretroviral therapy. Vaccine 2008; 26:6883-93. [PMID: 18940219 PMCID: PMC2845914 DOI: 10.1016/j.vaccine.2008.09.084] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 09/17/2008] [Accepted: 09/25/2008] [Indexed: 02/05/2023]
Abstract
A trial to evaluate the safety and immunogenicity of recombinant modified vaccinia Ankara (MVA) and fowlpox (FP) vectors expressing multiple HIV-1 proteins was conducted in twenty HIV-1 infected youth with suppressed viral replication on HAART. The MVA and FP-based multigene HIV-1 vaccines were safe and well tolerated. Increased frequencies of HIV-1 specific CD4+ proliferative responses and cytokine secreting cells were detected following immunization. Increased frequencies and breadth of HIV-1 specific CD8 T-cell responses were also detected. Plasma HIV-1-specific antibody levels and neutralizing activity were unchanged following vaccination. Poxvirus-based vaccines may merit further study in therapeutic vaccine protocols.
Collapse
Affiliation(s)
- Thomas C Greenough
- Pediatrics and Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Feng Y, Wang S, Luo F, Ruan Y, Kang L, Xiang X, Chao T, Peng G, Zhu C, Mu Y, Zhu Y, Zhang X, Wu J. A novel recombinant bacterial vaccine strain expressing dual viral antigens induces multiple immune responses to the Gag and gp120 proteins of HIV-1 in immunized mice. Antiviral Res 2008; 80:272-9. [PMID: 18639586 PMCID: PMC7114238 DOI: 10.1016/j.antiviral.2008.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 06/15/2008] [Accepted: 06/16/2008] [Indexed: 11/27/2022]
Abstract
Recombinant Salmonella enterica serovar Typhi can function as a live vector to deliver foreign antigens to the mammalian immune system and induce both mucosal and systemic immunity. In this study, we generated a recombinant Salmonella Typhi strain pilS−pilT−Gag+(pVAX1-gp120) harboring the human immunodeficiency virus (HIV) gag gene integrated into the bacterial chromosome and gp120 gene carried by a plasmid. Mice inoculated with this recombinant bacterium through intranasal route produced high titers of IgG to gp120 in sera and IgA to gp120 in fecal washes. In addition, Gag-specific and gp120-specific cytotoxic T lymphocyte (CTL) responses were observed in sorted spleen lymphocytes of immunized mice. These results demonstrated that this recombinant Salmonella Typhi strain elicits multiple immune responses against both Gag and gp120 antigens of HIV, and thus would be a potential vaccine candidate to the prevention of HIV/AIDS.
Collapse
Affiliation(s)
- Yong Feng
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wychang, Wuhan 430072, Hubei, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Moran E, Simmons C, Chau NV, Luhn K, Wills B, Phuong Dung N, Thao LTT, Hien TT, Farrar J, Rowland-Jones S, Dong T. Preservation of a critical epitope core region is associated with the high degree of flaviviral cross-reactivity exhibited by a dengue-specific CD4+ T cell clone. Eur J Immunol 2008; 38:1050-1057. [PMID: 18383038 PMCID: PMC4333208 DOI: 10.1002/eji.200737699] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Dengue is a member of the Flaviviridae, a large group of related viruses some of which co-circulate in certain regions (e.g. dengue and Yellow fever in South America). Immune responses cross-reactive between different dengue serotypes are important in the pathogenesis of dengue disease but it is not known whether previous infection with one flavivirus might affect the clinical course of subsequent infections with other members of the family. CD4+ T cells have been shown to be important in the production of cytokines in response to dengue infection and can demonstrate significant epitope cross-reactivity. Here, we describe the generation and characterisation of CD4+ T cell clones from a patient experiencing acute dengue infection. These clones were DRB1*15+ and recognised epitope variants not only within other dengue viruses but certain other flaviviruses. This cross-reactivity was dependent upon the presence of a five-amino acid core region, consistent with structural observations of class II MHC binding to TCR demonstrating that only a subset of residues within an epitope bound to a class II molecule are "read out" by the TCR. This capacity of CD4+ T cell clones to recognise a given epitope despite considerable variation between viruses may be of pathological significance, particularly in regions where related viruses co-circulate.
Collapse
Affiliation(s)
- Edward Moran
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Cameron Simmons
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | | | - Kerstin Luhn
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Bridget Wills
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | - Nguyen Phuong Dung
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | | | - Tran Tinh Hien
- Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | - Jeremy Farrar
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | - Sarah Rowland-Jones
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Tao Dong
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
22
|
Stahl-Hennig C, Kuate S, Franz M, Suh YS, Stoiber H, Sauermann U, Tenner-Racz K, Norley S, Park KS, Sung YC, Steinman R, Racz P, Uberla K. Atraumatic oral spray immunization with replication-deficient viral vector vaccines. J Virol 2007; 81:13180-90. [PMID: 17898066 PMCID: PMC2169087 DOI: 10.1128/jvi.01400-07] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of needle-free vaccines is one of the recently defined "grand challenges in global health" (H. Varmus, R. Klausner, R. Klausner, R. Zerhouni, T. Acharya, A. S. Daar, and P. A. Singer, Science 302:398-399, 2003). To explore whether a natural pathway to the inductive site of the mucosa-associated lymphatic tissue could be exploited for atraumatic immunization purposes, replication-deficient viral vector vaccines were sprayed directly onto the tonsils of rhesus macaques. Tonsillar immunization with viral vector vaccines encoding simian immunodeficiency virus (SIV) antigens induced cellular and humoral immune responses. Viral RNA levels after a stringent SIV challenge were reduced, providing a level of protection similar to that observed after systemic immunization with the same vaccines. Thus, atraumatic oral spray immunization with replication-deficient vectors can overcome the epithelial barrier, deliver the vaccine antigen to the mucosa-associated lymphatic tissue, and avoid induction of tolerance, providing a novel approach to circumvent acceptability problems of syringe and needle vaccines for children and in developing countries.
Collapse
|
23
|
Duke CM, Maguire CA, Keefer MC, Federoff HJ, Bowers WJ, Dewhurst S. HSV-1 amplicon vectors elicit polyfunctional T cell responses to HIV-1 Env, and strongly boost responses to an adenovirus prime. Vaccine 2007; 25:7410-21. [PMID: 17868958 PMCID: PMC2092414 DOI: 10.1016/j.vaccine.2007.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2007] [Revised: 08/03/2007] [Accepted: 08/07/2007] [Indexed: 12/11/2022]
Abstract
HSV-1 amplicon vectors elicit strong T-cell responses to encoded antigens but the qualitative nature of these responses is poorly understood. Antigen-specific CD4(+) and CD8(+) T-cell responses to amplicon and adenovirus (rAd5) vectors encoding HIV-1 gp120 were assessed following immunization of mice, by performing intracellular cytokine staining for IFNgamma, IL2 and TNFalpha, following stimulation of splenocytes with a HIV-1 Env peptide pool. The quality of the primary T-cell response to amplicon and rAd5 vectors was strikingly similar, but there were qualitative differences in responses to amplicon vectors that incorporated different promoters upstream of gp120 - suggesting that promoters can significantly influence immune response quality. When prime-boost combinations were studied, a rAd5 prime and amplicon boost elicited the highest T-cell response. Furthermore, protocols that incorporated a rAd5 prime consistently elicited a greater proportion of polyfunctional CD4(+) T-cells-regardless of boost. This suggests that initial priming can shape immune response quality after a boost. Overall, these findings provide insight into effective vector combinations for HIV-1 vaccine development.
Collapse
Affiliation(s)
- Cindy M.P. Duke
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Casey A. Maguire
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Michael C. Keefer
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Howard J. Federoff
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
- Department of Center for Aging and Development, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - William J. Bowers
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
- Department of Center for Aging and Development, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| | - Stephen Dewhurst
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY. USA
| |
Collapse
|
24
|
Im EJ, Saubi N, Virgili G, Sander C, Teoh D, Gatell JM, McShane H, Joseph J, Hanke T. Vaccine platform for prevention of tuberculosis and mother-to-child transmission of human immunodeficiency virus type 1 through breastfeeding. J Virol 2007; 81:9408-18. [PMID: 17596303 PMCID: PMC1951420 DOI: 10.1128/jvi.00707-07] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Most children in Africa receive their vaccine against tuberculosis at birth. Those infants born to human immunodeficiency virus type 1 (HIV-1)-positive mothers are at high risk of acquiring HIV-1 infection through breastfeeding in the first weeks of their lives. Thus, the development of a vaccine which would protect newborns against both of these major global killers is a logical yet highly scientifically, ethically, and practically challenging aim. Here, a recombinant lysine auxotroph of Mycobacterium bovis bacillus Calmette-Guérin (BCG), a BCG strain that is safer than those currently used and expresses an African HIV-1 clade-derived immunogen, was generated and shown to be stable and to induce durable, high-quality HIV-1-specific CD4(+)- and CD8(+)-T-cell responses. Furthermore, when the recombinant BCG vaccine was used in a priming-boosting regimen with heterologous components, the HIV-1-specific responses provided protection against surrogate virus challenge, and the recombinant BCG vaccine alone protected against aerosol challenge with M. tuberculosis. Thus, inserting an HIV-1-derived immunogen into the scheduled BCG vaccine delivered at or soon after birth may prime HIV-1-specific responses, which can be boosted by natural exposure to HIV-1 in the breast milk and/or by a heterologous vaccine such as recombinant modified vaccinia virus Ankara delivering the same immunogen, and decrease mother-to-child transmission of HIV-1 during breastfeeding.
Collapse
Affiliation(s)
- Eung-Jun Im
- Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe, Oxford OX3 9DS, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hanke T, McMichael AJ, Dorrell L. Clinical experience with plasmid DNA- and modified vaccinia virus Ankara-vectored human immunodeficiency virus type 1 clade A vaccine focusing on T-cell induction. J Gen Virol 2007; 88:1-12. [PMID: 17170430 DOI: 10.1099/vir.0.82493-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Candidate human immunodeficiency virus type 1 (HIV-1) vaccines focusing on T-cell induction, constructed as pTHr.HIVA DNA and modified vaccinia virus Ankara (MVA).HIVA, were delivered in a heterologous prime-boost regimen. The vaccines were tested in several hundred healthy or HIV-1-infected volunteers in Europe and Africa. Whilst larger trials of hundreds of volunteers suggested induction of HIV-1-specific T-cell responses in <15 % of healthy vaccinees, a series of small, rapid trials in 12-24 volunteers at a time with a more in-depth analysis of vaccine-elicited T-cell responses proved to be highly informative and provided more encouraging results. These trials demonstrated that the pTHr.HIVA vaccine alone primed consistently weak and mainly CD4(+), but also CD8(+) T-cell responses, and the MVA.HIVA vaccine delivered a consistent boost to both CD4(+) and CD8(+) T cells, which was particularly strong in HIV-1-infected patients. Thus, whilst the search is on for ways to enhance T-cell priming, MVA is a useful boosting vector for human subunit genetic vaccines.
Collapse
Affiliation(s)
- Tomáš Hanke
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| | - Andrew J McMichael
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| | - Lucy Dorrell
- Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, The John Radcliffe, Oxford OX3 9DS, UK
| |
Collapse
|
26
|
Dorrell L, Williams P, Suttill A, Brown D, Roberts J, Conlon C, Hanke T, McMichael A. Safety and tolerability of recombinant modified vaccinia virus Ankara expressing an HIV-1 gag/multiepitope immunogen (MVA.HIVA) in HIV-1-infected persons receiving combination antiretroviral therapy. Vaccine 2007; 25:3277-83. [PMID: 17257714 DOI: 10.1016/j.vaccine.2007.01.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Revised: 12/14/2006] [Accepted: 01/04/2007] [Indexed: 12/23/2022]
Abstract
The safety of attenuated poxviruses in HIV-1-infected individuals is an important consideration in their application as vaccine vectors, first, because new HIV-1 infections may occur in vaccine trials involving persons at high risk of infection and secondly, therapeutic vaccinations are a potential means to enhance virus-specific immune responses once infection has occurred. We administered a candidate modified vaccinia virus Ankara-vectored HIV-1 vaccine, MVA.HIVA, by intradermal injection to 16 chronically infected adults during highly active antiretroviral therapy. Vaccinations were well tolerated and there were no serious adverse events. No breakthrough viraemia occurred after immunisations or throughout follow-up. These data confirm the safety of MVA.HIVA in HIV-1-infected individuals and provide support for further evaluation of MVA-vectored vaccines in prophylactic and therapeutic immunisation strategies.
Collapse
Affiliation(s)
- Lucy Dorrell
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, UK.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Rodríguez JW, Pagan NO, Ocasio MC, Ríos Z, Cubano LA, Boukli NM, Otero M, Hunter R, Nair MP, Rios-Olivares E. Induction of a Soluble Anti-HIV-1 factor (s) with IFN-γ, IL-10, and β-Chemokine Modulating Activity by an Influenza-Bacterial Polyantigenic Mixture. ACTA ACUST UNITED AC 2007; 3. [PMID: 24327810 DOI: 10.3844/ajidsp.2007.267.275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Partial immune restoration may be obtained with highly active antiretroviral therapy (HAART), but specific anti-HIV-1 immune responses do not appear to improve substantially. We have demonstrated that a soluble factor(s) induced by a mixture of inactivated influenza and bacterial vaccines called polyantigenic immunomodulator (PAI), possesses strong immunoregulatory and anti-HIV-1 activities. In the present study, we show that culture fluids from both PAI-stimulated peripheral blood mononuclear cells (PBMC) and CD8+ T-cells of HIV-1 infected patients were able to suppress HIV-1 replication in an MHC-unrestricted fashion. The PAI-induced antiviral activity was eliminated when culture fluids were pre-heated at 100°C for 10 min. and it is associated with induction of IFN-γ, MIP-1α, MIP-1β, and RANTES production, but inhibition of IL-10. Furthermore, this induction is dependent on the immunological status (CD4:CD8 ratio) of the HIV-1 infected patient. Taken together, our results suggest that the MHC-unrestricted HIV-1 suppression that is induced by culture fluids from PAI-stimulated PBMC may result from the stimulation of immune cell subpopulations to produce a heat-labile antiviral soluble factor(s), which in turn modulate cytokine and β-chemokine production. The identification of this PAI-induced soluble factor(s) may have major therapeutic potential.
Collapse
Affiliation(s)
- José W Rodríguez
- Department of Microbiology and Immunology, Universidad Central del Caribe School of Medicine, Bayamón, Puerto Rico
| | | | | | | | | | | | | | | | | | | |
Collapse
|