1
|
Lorenzetti L, Dinh N, Whitcomb C, Martinez A, Chatani M, Lievense B, Nhamo D, Slack C, Eley N, MacQueen K. Meta-analysis of Pregnancy Events in Biomedical HIV Prevention Trials in Sub-Saharan Africa: Implications for Gender Transformative Trials. AIDS Behav 2024; 28:3850-3872. [PMID: 39153025 PMCID: PMC11471715 DOI: 10.1007/s10461-024-04459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/19/2024]
Abstract
Historically, pregnant and lactating populations (PLP) have been excluded or disenrolled from biomedical HIV prevention trials, despite being more likely to acquire HIV during pregnancy and the post-partum period. We conducted a meta-analysis of pregnancy events in biomedical HIV prevention trials in sub-Saharan Africa to support trialists moving toward more inclusive clinical and implementation studies. We searched peer-reviewed literature reporting pregnancy events and contraceptive requirements in HIV prevention trials between 2001 and 2022. We hypothesized four variables to explain variation: contraceptive requirements, study start year, study product, and sub-region. We fit a meta-analytic model to estimate individual effect sizes and sampling variances, then conducted sub-group analyses to assess moderating effects. We identified 38 references for inclusion, across which the proportion of pregnancy events was 8% (95% confidence interval [CI]: 6-10%) with high heterogeneity (I2 = 99%). Studies not requiring contraceptives (21%, 95%CI: 7-48%) reported a significantly higher proportion of pregnancy events than studies requiring two methods (5%, 95%CI: 2-10%). Studies launched between 2001 and 2007 (11%, 95%CI: 8-16%), microbicide gel trials (12%, 95%CI: 8-18%), and studies conducted in Western Africa (28%, 95%CI: 13-51%) reported higher proportions of pregnancy events than reference groups. Together, these variables have a moderating effect on pregnancy events (p < 0.0001), explaining 63% of heterogeneity in trials. Results describe how, over time, more stringent contraceptive requirements reduced pregnancy events, which ensured necessary statistical power but limited reproductive choice by participants. With the move toward continuing PLP on experimental products, trialists can utilize estimated pregnancy events reported here to inform strategies that accommodate participants' changing fertility preferences.
Collapse
Affiliation(s)
- Lara Lorenzetti
- Behavioral, Epidemiological and Clinical Sciences Division, Durham, NC, FHI 360, USA.
| | - Nhi Dinh
- Behavioral, Epidemiological and Clinical Sciences Division, Durham, NC, FHI 360, USA
| | - Cason Whitcomb
- Behavioral, Epidemiological and Clinical Sciences Division, Durham, NC, FHI 360, USA
| | - Andres Martinez
- Behavioral, Epidemiological and Clinical Sciences Division, Durham, NC, FHI 360, USA
| | | | | | | | - Catherine Slack
- School of Law, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Natalie Eley
- Behavioral, Epidemiological and Clinical Sciences Division, Durham, NC, FHI 360, USA
| | - Kathleen MacQueen
- Behavioral, Epidemiological and Clinical Sciences Division, Durham, NC, FHI 360, USA
| |
Collapse
|
2
|
Pancras G, Ezekiel M, Mbugi E, Merz JF. Should HIV Vaccines Be Made Available at No or Subsidized Cost? A Qualitative Inquiry of HIV Vaccine Trial Stakeholders in Tanzania. AJOB Empir Bioeth 2024; 15:206-213. [PMID: 37889211 PMCID: PMC11052918 DOI: 10.1080/23294515.2023.2274599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
BACKGROUND The world has come closer than ever to discovering a viable HIV vaccine. However, it remains less certain whether HIV vaccines should be made available to participants and communities in which trials are run no or subsidized cost. Hence the essence of this inquiry. METHODOLOGY This is a case study design using in-depth interviews (IDI) and focus group discussions (FGD) with researchers of HIV vaccine trials, institutional review board (IRB) members, HIV advocates, a policy maker, and members of community advisory board (CAB) in Tanzania. Participants were purposively selected and data thematically analyzed using MAXQDA software. RESULTS Hosting a vaccine trial and the financial incapacity of individuals at increased risk of HIV were among the reasons in favor of free access to HIV vaccines. In contrast, the view that vaccines should be provided at a subsidized cost was related to high costs of vaccine development, financial return expectations by investors, and the fear of labeling the free vaccine as less important. Moreover, apart from governments and international organizations, well-off individuals could share the cost burden. CONCLUSION Stakeholders engaging in active discussion about sharing the viable vaccine ought to take the aforementioned concerns into account and ensure unhindered access to individuals and host communities in Tanzania and beyond.
Collapse
Affiliation(s)
- Godwin Pancras
- Department of Bioethics and Health Professionalism, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Tanzania; P.O. Box 65001, Dar es Salaam, Tanzania
| | - Mangi Ezekiel
- Department of Behavioral Sciences, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Tanzania; P.O. Box 65001, Dar es Salaam, Tanzania
| | - Erasto Mbugi
- Department of Biochemistry, School of Medicine, Muhimbili University of Health and Allied Sciences, Tanzania; P.O. Box 65001, Dar es Salaam, Tanzania
| | - Jon F. Merz
- Department of Medical Ethics & Health Policy, Perelman School of Medicine at the University of Pennsylvania, Blockley Hall Floor 14, 423 Guardian Drive, Philadelphia, Pennsylvania 19104-4884 USA
| |
Collapse
|
3
|
Perdiguero B, Pérez P, Marcos-Villar L, Albericio G, Astorgano D, Álvarez E, Sin L, Elena Gómez C, García-Arriaza J, Esteban M. Highly attenuated poxvirus-based vaccines against emerging viral diseases. J Mol Biol 2023:168173. [PMID: 37301278 DOI: 10.1016/j.jmb.2023.168173] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Although one member of the poxvirus family, variola virus, has caused one of the most devastating human infections worldwide, smallpox, the knowledge gained over the last 30 years on the molecular, virological and immunological mechanisms of these viruses has allowed the use of members of this family as vectors for the generation of recombinant vaccines against numerous pathogens. In this review, we cover different aspects of the history and biology of poxviruses with emphasis on their application as vaccines, from first- to fourth-generation, against smallpox, monkeypox, emerging viral diseases highlighted by the World Health Organization (COVID-19, Crimean-Congo haemorrhagic fever, Ebola and Marburg virus diseases, Lassa fever, Middle East respiratory syndrome and severe acute respiratory syndrome, Nipah and other henipaviral diseases, Rift Valley fever and Zika), as well as against one of the most concerning prevalent virus, the Human Immunodeficiency Virus, the causative agent of AcquiredImmunodeficiency Syndrome. We discuss the implications in human health of the 2022 monkeypox epidemic affecting many countries, and the rapid prophylactic and therapeutic measures adopted to control virus dissemination within the human population. We also describe the preclinical and clinical evaluation of the Modified Vaccinia virus Ankara and New York vaccinia virus poxviral strains expressing heterologous antigens from the viral diseases listed above. Finally, we report different approaches to improve the immunogenicity and efficacy of poxvirus-based vaccine candidates, such as deletion of immunomodulatory genes, insertion of host-range genes and enhanced transcription of foreign genes through modified viral promoters. Some future prospects are also highlighted.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| |
Collapse
|
4
|
The Prevalence, Incidence, and Risk Factors for HIV Among Female Sex Workers-A Cohort Being Prepared for a Phase IIb HIV Vaccine Trial in Dar es Salaam, Tanzania. J Acquir Immune Defic Syndr 2022; 91:439-448. [PMID: 36126184 PMCID: PMC9646411 DOI: 10.1097/qai.0000000000003097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 06/20/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND A cohort of female sex workers (FSWs) was established to determine HIV prevalence and incidence, and associated factors in preparation for a phase IIb HIV vaccine and pre-exposure prophylaxis trial (PrEPVacc). SETTING A cohort of FSWs in Dar es Salaam, Tanzania. METHODS FSWs aged 18-45 years were recruited using a respondent-driven sampling method. Social demographic data, HIV risk behavioral assessments, and blood samples for testing of HIV, syphilis, hepatitis B (HBV), and hepatitis C (HCV) infections were collected at baseline and then at 3, 6, 9, and 12 months. Poisson regressions were used to estimate the prevalence ratios for factors associated with HIV prevalence and to estimate the 12-month HIV incidence rate. RESULTS Between October and December 2018, a total of 773 FSWs were screened for eligibility and 700 were enrolled. The baseline prevalence of HIV, syphilis, HBV, and HCV was 7.6%, 1.2%, 1.7%, and 1.0%, respectively. HIV prevalence was associated with older age, using illicit drugs, and being infected with syphilis, HBV, or HCV. Attendance at 12 months was 80% (562/700). Twenty-one FSWs seroconverted during follow-up, giving a 12-month HIV incidence rate of 3.45 per 100 person-years at risk (95% CI; 2.25-5.28/100 person-years at risk). The HIV incidence rate was higher among FSWs aged 18-24 years, FSWs who used drugs, and those diagnosed with syphilis, HBV, or HCV. CONCLUSION The high HIV incidence rate and retention rate among FSWs enrolled into the cohort demonstrate that this population is suitable for participation in HIV prevention trials.
Collapse
|
5
|
Tarimo EAM, Ambikile J, Munseri P, Bakari M. Personal experiences following acquiring HIV infection while volunteering in Phase I/II HIV vaccine trials: A qualitative study from Tanzania. PLoS One 2022; 17:e0276404. [PMID: 36288332 PMCID: PMC9605023 DOI: 10.1371/journal.pone.0276404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Participation in HIV vaccine trials is an essential step towards development of an effective preventive vaccine. A Phase I/II HIV vaccine trial enrolls volunteers at low risk of acquiring HIV infection, however a few may still become infected. Understanding the experiences of volunteers who acquired HIV infection while participating in such trials is essential for future research. Here, we describe experiences of HIV infected volunteers in Phase I/II HIV vaccine trials conducted in urban Tanzania. MATERIALS AND METHODS We used a case study design. In-depth interviews were conducted with four participants who became HIV infected during long follow-up visits after completion of vaccination schedules in a Phase I/II trial. Between 3 and 8 years after HIV positive diagnosis, each participant was interviewed at three time points within a two-year interval so as to allow for accumulation of experiences and cross-checking the emerging constructs. Data was analyzed using a qualitative data analysis framework. RESULTS Analysis revealed that participation in HIV vaccine trials involves balancing controversies and the spirit of informed decision. The participants declared that they did not acquire HIV from the experimental vaccine. Disclosure of HIV status within the family was gender specific. Men were hesitant to disclose their HIV status to their sexual partners fearing for the consequences. Women's attempt to disclose their HIV status yielded negative reactions from the sexual partners. The acquired knowledge from the HIV vaccine research enabled the participants to cope with the uncertainties and their health status. CONCLUSIONS The knowledge acquired during the Phase I/II HIV vaccine trial appears to be an essential resource to cope with uncertainties post research. The HIV vaccine trial implementers need to understand the challenges the volunteers may confront after the trial while coping with their health status. Longitudinal studies are essential to trace the effects of uncertainties to the individual participants.
Collapse
Affiliation(s)
- Edith A. M. Tarimo
- Department of Nursing Management, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Joel Ambikile
- Department of Clinical Nursing, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Patricia Munseri
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Muhammad Bakari
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| |
Collapse
|
6
|
Msafiri F, Manjate A, Lindroth S, Tembe N, Chissumba RM, Cumbane V, Jani I, Aboud S, Lyamuya E, Andersson S, Nilsson C. Vaccine-Induced Seroreactivity Impacts the Accuracy of HIV Testing Algorithms in Sub-Saharan Africa: An Exploratory Study. Vaccines (Basel) 2022; 10:vaccines10071062. [PMID: 35891226 PMCID: PMC9316099 DOI: 10.3390/vaccines10071062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 01/27/2023] Open
Abstract
The detection of vaccine-induced HIV antibody responses by rapid diagnostic tests (RDTs) may confound the interpretation of HIV testing results. We assessed the impact of vaccine-induced seroreactivity (VISR) on the diagnosis of HIV in sub-Saharan Africa. Samples collected from healthy participants of HIVIS and TaMoVac HIV vaccine trials after the final vaccination were analyzed for VISR using HIV testing algorithms used in Mozambique and Tanzania that employ two sequential RDTs. The samples were also tested for VISR using Enzygnost HIV Integral 4 ELISA and HIV western blot assays. Antibody titers to subtype C gp140 were determined using an in-house enzyme-linked immunosorbent assay (ELISA). The frequency of VISR was 93.4% (128/137) by Enzygnost HIV Integral 4 ELISA, and 66.4% (91/137) by western blot assay (WHO interpretation). The proportion of vaccine recipients that would have been misdiagnosed as HIV-positive in Mozambique was half of that in Tanzania: 26.3% (36/137) and 54.0% (74/137), respectively, p < 0.0001. In conclusion, the HIV RDTs and algorithms assessed here will potentially misclassify a large proportion of the HIV vaccine recipients if no other test is used. Increased efforts are needed to develop differential serological or molecular tools for use at the point of care.
Collapse
Affiliation(s)
- Frank Msafiri
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (S.A.); (E.L.)
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
- Correspondence:
| | - Alice Manjate
- Faculdade de Medicina, Universidade Eduardo Mondlane, Maputo P.O. Box 257, Mozambique;
- School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (S.L.); (S.A.)
| | - Sarah Lindroth
- School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (S.L.); (S.A.)
| | - Nelson Tembe
- Instituto Nacional de Saúde, Maputo P.O. Box 3943, Mozambique; (N.T.); (R.M.C.); (V.C.); (I.J.)
| | | | - Victoria Cumbane
- Instituto Nacional de Saúde, Maputo P.O. Box 3943, Mozambique; (N.T.); (R.M.C.); (V.C.); (I.J.)
| | - Ilesh Jani
- Instituto Nacional de Saúde, Maputo P.O. Box 3943, Mozambique; (N.T.); (R.M.C.); (V.C.); (I.J.)
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (S.A.); (E.L.)
| | - Eligius Lyamuya
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (S.A.); (E.L.)
| | - Sören Andersson
- School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (S.L.); (S.A.)
- Public Health Agency of Sweden, 17182 Solna, Sweden
| | - Charlotta Nilsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
- Public Health Agency of Sweden, 17182 Solna, Sweden
| |
Collapse
|
7
|
Maki Y, Kashiwagi S, Kimizuka Y. Laser vaccine adjuvants: Light-augmented immune responses. Vaccine 2021; 39:6805-6812. [PMID: 34666921 DOI: 10.1016/j.vaccine.2021.09.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 01/10/2023]
Abstract
Adjuvants are essential for ensuring the efficacy of modern vaccines. Considering frequent local and systemic adverse reactions, research into the development of safer and more effective adjuvants is being actively conducted. In recent years, the novel concept of laser vaccine adjuvants, which use the physical energy of light, has been developed. For long, light has been known to affect the physiological functions in living organisms. Since the development of lasers as stable light sources, laser adjuvants have evolved explosively in multiple ways over recent decades. Future laser adjuvants would have the potential not only to enhance the efficacy of conventional vaccine preparations but also to salvage candidate vaccines abandoned during development because of insufficient immunogenicity or owing to their inability to be combined with conventional adjuvants. Furthermore, the safety and efficacy of non-invasive laser adjuvants make them advantageous for vaccine dose sparing, which would be favorable for the timely and equitable global distribution of vaccines. In this review, we first describe the basics of light-tissue interactions, and then summarize the classification of lasers, the history of laser adjuvants, and the mechanisms by which different lasers elicit an immune response.
Collapse
Affiliation(s)
- Yohei Maki
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, USA
| | - Yoshifumi Kimizuka
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| |
Collapse
|
8
|
Eldershaw SA, Pearce H, Inman CF, Piper KP, Abbotts B, Stephens C, Nicol S, Croft W, Powell R, Begum J, Taylor G, Nunnick J, Walsh D, Sirovica M, Saddique S, Nagra S, Ferguson P, Moss P, Malladi R. DNA and modified vaccinia Ankara prime-boost vaccination generates strong CD8 + T cell responses against minor histocompatibility antigen HA-1. Br J Haematol 2021; 195:433-446. [PMID: 34046897 DOI: 10.1111/bjh.17495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/27/2021] [Indexed: 11/29/2022]
Abstract
Allogeneic immune responses underlie the graft-versus-leukaemia effect of stem cell transplantation, but disease relapse occurs in many patients. Minor histocompatibility antigen (mHAg) peptides mediate alloreactive T cell responses and induce graft-versus-leukaemia responses when expressed on patient haematopoietic tissue. We vaccinated nine HA-1-negative donors against HA-1 with a 'prime-boost' protocol of either two or three DNA 'priming' vaccinations prior to 'boost' with modified vaccinia Ankara (MVA). HA-1-specific CD8+ T cell responses were observed in seven donors with magnitude up to 1·5% of total CD8+ T cell repertoire. HA-1-specific responses peaked two weeks post-MVA challenge and were measurable in most donors after 12 months. HA-1-specific T cells demonstrated strong cytotoxic activity and lysed target cells with endogenous HA-1 protein expression. The pattern of T cell receptor (TCR) usage by HA-1-specific T cells revealed strong conservation of T cell receptor beta variable 7-9 (TRBV7-9) usage between donors. These findings describe one of the strongest primary peptide-specific CD8+ T cell responses yet recorded to a DNA-MVA prime-boost regimen and this may reflect the strong immunogenicity of mHAg peptides. Prime-boost vaccination in donors or patients may prove of substantial benefit in boosting graft-versus-leukaemia responses.
Collapse
MESH Headings
- Adult
- Aged
- Allografts
- Antigens, Neoplasm/immunology
- Cytotoxicity, Immunologic
- Epitopes/immunology
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Graft vs Leukemia Effect/immunology
- HLA-A2 Antigen/immunology
- Hematopoietic Stem Cell Transplantation
- Humans
- Immunogenicity, Vaccine
- Immunologic Memory
- Male
- Middle Aged
- Minor Histocompatibility Antigens/immunology
- Oligopeptides/immunology
- Peptides/immunology
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Vaccination
- Vaccines, Attenuated
- Vaccines, DNA/immunology
- Vaccines, DNA/therapeutic use
- Vaccinia virus/immunology
- Viral Vaccines/immunology
- Viral Vaccines/therapeutic use
Collapse
Affiliation(s)
- Suzy A Eldershaw
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Charlotte F Inman
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Karen P Piper
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Ben Abbotts
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Christine Stephens
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Samantha Nicol
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Wayne Croft
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Richard Powell
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Jusnara Begum
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Graham Taylor
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
| | - Jane Nunnick
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Donna Walsh
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Mirjana Sirovica
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Shamyla Saddique
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Sandeep Nagra
- Department of Haematology, Birmingham Health Partners, Queen Elizabeth Hospital, Birmingham, UK
| | - Paul Ferguson
- Department of Haematology, Birmingham Health Partners, Queen Elizabeth Hospital, Birmingham, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
- Department of Haematology, Birmingham Health Partners, Queen Elizabeth Hospital, Birmingham, UK
| | - Ram Malladi
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Birmingham, UK
- Department of Haematology, Birmingham Health Partners, Queen Elizabeth Hospital, Birmingham, UK
| |
Collapse
|
9
|
Reciprocal Inhibition of Immunogenic Performance in Mice of Two Potent DNA Immunogens Targeting HCV-Related Liver Cancer. Microorganisms 2021; 9:microorganisms9051073. [PMID: 34067686 PMCID: PMC8156932 DOI: 10.3390/microorganisms9051073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 11/29/2022] Open
Abstract
Chronic HCV infection and associated liver cancer impose a heavy burden on the healthcare system. Direct acting antivirals eliminate HCV, unless it is drug resistant, and partially reverse liver disease, but they cannot cure HCV-related cancer. A possible remedy could be a multi-component immunotherapeutic vaccine targeting both HCV-infected and malignant cells, but also those not infected with HCV. To meet this need we developed a two-component DNA vaccine based on the highly conserved core protein of HCV to target HCV-infected cells, and a renowned tumor-associated antigen telomerase reverse transcriptase (TERT) based on the rat TERT, to target malignant cells. Their synthetic genes were expression-optimized, and HCV core was truncated after aa 152 (Core152opt) to delete the domain interfering with immunogenicity. Core152opt and TERT DNA were highly immunogenic in BALB/c mice, inducing IFN-γ/IL-2/TNF-α response of CD4+ and CD8+ T cells. Additionally, DNA-immunization with TERT enhanced cellular immune response against luciferase encoded by a co-delivered plasmid (Luc DNA). However, DNA-immunization with Core152opt and TERT mix resulted in abrogation of immune response against both components. A loss of bioluminescence signal after co-delivery of TERT and Luc DNA into mice indicated that TERT affects the in vivo expression of luciferase directed by the immediate early cytomegalovirus and interferon-β promoters. Panel of mutant TERT variants was created and tested for their expression effects. TERT with deleted N-terminal nucleoli localization signal and mutations abrogating telomerase activity still suppressed the IFN-β driven Luc expression, while the inactivated reverse transcriptase domain of TERT and its analogue, enzymatically active HIV-1 reverse transcriptase, exerted only weak suppressive effects, implying that suppression relied on the presence of the full-length/nearly full-length TERT, but not its enzymatic activity. The effect(s) could be due to interference of the ectopically expressed xenogeneic rat TERT with biogenesis of mRNA, ribosomes and protein translation in murine cells, affecting the expression of immunogens. HCV core can aggravate this effect, leading to early apoptosis of co-expressing cells, preventing the induction of immune response.
Collapse
|
10
|
Differential Immune Response Following Intranasal and Intradermal Infection with Francisella tularensis: Implications for Vaccine Development. Microorganisms 2021; 9:microorganisms9050973. [PMID: 33946283 PMCID: PMC8145380 DOI: 10.3390/microorganisms9050973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022] Open
Abstract
Francisella tularensis (Ft) is a Gram-negative, facultative intracellular coccobacillus that is the etiological agent of tularemia. Interestingly, the disease tularemia has variable clinical presentations that are dependent upon the route of infection with Ft. Two of the most likely routes of Ft infection include intranasal and intradermal, which result in pneumonic and ulceroglandular tularemia, respectively. While there are several differences between these two forms of tularemia, the most notable disparity is between mortality rates: the mortality rate following pneumonic tularemia is over ten times that of the ulceroglandular disease. Understanding the differences between intradermal and intranasal Ft infections is important not only for clinical diagnoses and treatment but also for the development of a safe and effective vaccine. However, the immune correlates of protection against Ft, especially within the context of infection by disparate routes, are not yet fully understood. Recent advances in different animal models have revealed new insights in the complex interplay of innate and adaptive immune responses, indicating dissimilar patterns in both responses following infection with Ft via different routes. Further investigation of these differences will be crucial to predicting disease outcomes and inducing protective immunity via vaccination or natural infection.
Collapse
|
11
|
Ulmer JB, Liu MA. Path to Success and Future Impact of Nucleic Acid Vaccines: DNA and mRNA. MOLECULAR FRONTIERS JOURNAL 2021. [DOI: 10.1142/s2529732521400022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The rapid development of mRNA vaccines for COVID-19 has both astonished the world and raised concerns about their safety, perhaps because many people do not realize the decades’ long efforts for nucleic acid vaccines, both mRNA and DNA vaccines, including the licensure of several veterinary DNA vaccines. This manuscript traces the milestones for nucleic acid vaccine research and development (R&D), with a focus on the immune and safety issues they both raised and answered. The characteristics of the two entities are compared, demonstrating the similarities and differences between them, the advantages and disadvantages, which might lead toward using one or the other technology for different indications. In addition, as the SARS-CoV-2 pandemic has once again highlighted the importance of One Health, that is, the interactions between animal and human pathogens, focus will also be given to how DNA vaccine utilization and studies both in large domestic animals and in wildlife pave the way for more integrated approaches for vaccines to respond quickly to, and prevent, the global impacts of emerging diseases.
Collapse
|
12
|
Msafiri F, Joachim A, Held K, Nadai Y, Chissumba RM, Geldmacher C, Aboud S, Stöhr W, Viegas E, Kroidl A, Bakari M, Munseri PJ, Wahren B, Sandström E, Robb ML, McCormack S, Joseph S, Jani I, Ferrari G, Rao M, Biberfeld G, Lyamuya E, Nilsson C. Frequent Anti-V1V2 Responses Induced by HIV-DNA Followed by HIV-MVA with or without CN54rgp140/GLA-AF in Healthy African Volunteers. Microorganisms 2020; 8:microorganisms8111722. [PMID: 33158007 PMCID: PMC7693996 DOI: 10.3390/microorganisms8111722] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
Antibody responses that correlated with reduced risk of HIV acquisition in the RV144 efficacy trial were assessed in healthy African volunteers who had been primed three times with HIV-DNA (subtype A, B, C) and then randomized into two groups; group 1 was boosted twice with HIV-MVA (CRF01_AE) and group 2 with the same HIV-MVA coadministered with subtype C envelope (Env) protein (CN54rgp140/GLA-AF). The fine specificity of plasma Env-specific antibody responses was mapped after the final vaccination using linear peptide microarray technology. Binding IgG antibodies to the V1V2 loop in CRF01_AE and subtype C Env and Env-specific IgA antibodies were determined using enzyme-linked immunosorbent assay. Functional antibody-dependent cellular cytotoxicity (ADCC)-mediating antibody responses were measured using luciferase assay. Mapping of linear epitopes within HIV-1 Env demonstrated strong targeting of the V1V2, V3, and the immunodominant region in gp41 in both groups, with additional recognition of two epitopes located in the C2 and C4 regions in group 2. A high frequency of V1V2-specific binding IgG antibody responses was detected to CRF01_AE (77%) and subtype C antigens (65%). In conclusion, coadministration of CN54rgp140/GLA-AF with HIV-MVA did not increase the frequency, breadth, or magnitude of anti-V1V2 responses or ADCC-mediating antibodies induced by boosting with HIV-MVA alone.
Collapse
Affiliation(s)
- Frank Msafiri
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (A.J.); (S.A.); (E.L.)
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
- Correspondence: or
| | - Agricola Joachim
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (A.J.); (S.A.); (E.L.)
| | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (Y.N.); (C.G.); (A.K.)
- German Center for Infection Research (DZIF), partner site Munich, 80802 Munich, Germany
| | - Yuka Nadai
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (Y.N.); (C.G.); (A.K.)
- German Center for Infection Research (DZIF), partner site Munich, 80802 Munich, Germany
| | | | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (Y.N.); (C.G.); (A.K.)
- German Center for Infection Research (DZIF), partner site Munich, 80802 Munich, Germany
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (A.J.); (S.A.); (E.L.)
| | - Wolfgang Stöhr
- MRC Clinical Trials Unit at UCL, London WC1V 6LJ, UK; (W.S.); (S.M.)
| | - Edna Viegas
- Instituto Nacional de Saúde, Maputo 3943, Mozambique; (R.M.C.); (E.V.); (I.J.)
| | - Arne Kroidl
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, 80802 Munich, Germany; (K.H.); (Y.N.); (C.G.); (A.K.)
- German Center for Infection Research (DZIF), partner site Munich, 80802 Munich, Germany
| | - Muhammad Bakari
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (M.B.); (P.J.M.)
| | - Patricia J. Munseri
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (M.B.); (P.J.M.)
| | - Britta Wahren
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Nobel’s Rd 16, 17177 Stockholm, Sweden;
| | - Eric Sandström
- Karolinska Institutet at Södersjukhuset, Södersjukhuset, 11883 Stockholm, Sweden;
| | - Merlin L. Robb
- The Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA;
| | - Sheena McCormack
- MRC Clinical Trials Unit at UCL, London WC1V 6LJ, UK; (W.S.); (S.M.)
| | | | - Ilesh Jani
- Instituto Nacional de Saúde, Maputo 3943, Mozambique; (R.M.C.); (E.V.); (I.J.)
| | - Guido Ferrari
- Department of Surgery and Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Mangala Rao
- United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA;
| | - Gunnel Biberfeld
- Department of Global Public Health, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Eligius Lyamuya
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam P.O. Box 65001, Tanzania; (A.J.); (S.A.); (E.L.)
| | - Charlotta Nilsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
- Department of Microbiology, Public Health Agency of Sweden, 17182 Solna, Sweden
| |
Collapse
|
13
|
A Zigzag but Upward Way to Develop an HIV-1 Vaccine. Vaccines (Basel) 2020; 8:vaccines8030511. [PMID: 32911701 PMCID: PMC7564621 DOI: 10.3390/vaccines8030511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 01/04/2023] Open
Abstract
After decades of its epidemic, the human immunodeficiency virus type 1 (HIV-1) is still rampant worldwide. An effective vaccine is considered to be the ultimate strategy to control and prevent the spread of HIV-1. To date, hundreds of clinical trials for HIV-1 vaccines have been tested. However, there is no HIV-1 vaccine available yet, mostly because the immune correlates of protection against HIV-1 infection are not fully understood. Currently, a variety of recombinant viruses-vectored HIV-1 vaccine candidates are extensively studied as promising strategies to elicit the appropriate immune response to control HIV-1 infection. In this review, we summarize the current findings on the immunological parameters to predict the protective efficacy of HIV-1 vaccines, and highlight the latest advances on HIV-1 vaccines based on viral vectors.
Collapse
|
14
|
Lévy Y, Lacabaratz C, Ellefsen-Lavoie K, Stöhr W, Lelièvre JD, Bart PA, Launay O, Weber J, Salzberger B, Wiedemann A, Surenaud M, Koelle DM, Wolf H, Wagner R, Rieux V, Montefiori DC, Yates NL, Tomaras GD, Gottardo R, Mayer B, Ding S, Thiébaut R, McCormack S, Chêne G, Pantaleo G. Optimal priming of poxvirus vector (NYVAC)-based HIV vaccine regimens for T cell responses requires three DNA injections. Results of the randomized multicentre EV03/ANRS VAC20 Phase I/II Trial. PLoS Pathog 2020; 16:e1008522. [PMID: 32589686 PMCID: PMC7319597 DOI: 10.1371/journal.ppat.1008522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 04/06/2020] [Indexed: 12/22/2022] Open
Abstract
DNA vectors have been widely used as a priming of poxvirus vaccine in prime/boost regimens. Whether the number of DNA impacts qualitatively or quantitatively the immune response is not fully explored. With the aim to reinforce T-cell responses by optimizing the prime-boost regimen, the multicentric EV03/ANRS VAC20 phase I/II trial, randomized 147 HIV-negative volunteers to either 3xDNA plus 1xNYVAC (weeks 0, 4, 8 plus 24; n = 74) or to 2xDNA plus 2xNYVAC (weeks 0, 4 plus 20, 24; n = 73) groups. T-cell responses (IFN-γ ELISPOT) to at least one peptide pool were higher in the 3xDNA than the 2xDNA groups (91% and 80% of vaccinees) (P = 0.049). In the 3xDNA arm, 26 (37%) recipients developed a broader T-cell response (Env plus at least to one of the Gag, Pol, Nef pools) than in the 2xDNA (15; 22%) arms (primary endpoint; P = 0.047) with a higher magnitude against Env (at week 26) (P<0.001). In both groups, vaccine regimens induced HIV-specific polyfunctional CD4 and CD8 T cells and the production of Th1, Th2 and Th17/IL-21 cytokines. Antibody responses were also elicited in up to 81% of vaccines. A higher percentage of IgG responders was noted in the 2xDNA arm compared to the 3xDNA arm, while the 3xDNA group tended to elicit a higher magnitude of IgG3 response against specific Env antigens. We show here that the modulation of the prime strategy, without modifying the route or the dose of administration, or the combination of vectors, may influence the quality of the responses. Development of a safe and effective HIV-1 vaccine would undoubtedly be the best solution for the ultimate control of the worldwide AIDS pandemic. To date, only one large phase III trial (RV144 Thai study) showed a partial and modest protection against HIV infection. This result raised hope in the field and encouraged the development of vaccines or strategies in order to improve vaccine efficacy. Several vaccine strategies designed to elicit broad HIV-specific T cells and/or neutralizing antibodies to prevent HIV-1 transmission are under evaluation. Among diverse candidate vaccines, the safety and immunogenicity of multi-gene DNA-based and Pox-virus derived vaccines have been evaluated in several clinical studies. The present study was designed to optimize the combination of these two vaccines with the aim of determining the optimal number of DNA primes for a poxvirus-based HIV vaccine regimen. We show here that the prime boost combination is highly immunogenic and that the number of DNA primes induces differentially T cell and antibody responses. A better priming of poxvirus-based vaccine regimens for T cells is obtained with 3 DNA injections. Our results contribute and extend data of several preclinical studies pointing out the potential interest of DNA as a prime capable not only of improving immune responses but also of imprinting the long-term responses to boost vaccines.
Collapse
Affiliation(s)
- Yves Lévy
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, équipe 16, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service d’Immunologie Clinique, Créteil, France
- * E-mail:
| | - Christine Lacabaratz
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, équipe 16, Créteil, France
| | | | | | - Jean-Daniel Lelièvre
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, équipe 16, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service d’Immunologie Clinique, Créteil, France
| | | | - Odile Launay
- Université de Paris, Faculté de médecine Paris Descartes; Inserm, CIC 1417, F-CRIN I-REIVAC; Assistance Publique-Hôpitaux de Paris, CIC Cochin Pasteur, Paris, France
| | | | - Bernd Salzberger
- University Hospital, Institute of Clinical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Aurélie Wiedemann
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, équipe 16, Créteil, France
| | - Mathieu Surenaud
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, équipe 16, Créteil, France
| | - David M. Koelle
- Department of Medicine & Department of Global Health, University of Washington, Fred Hutchinson Cancer Research Center Seattle, Washington, United States of America
| | - Hans Wolf
- University Hospital, Institute of Clinical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- University Hospital, Institute of Clinical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Véronique Rieux
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, équipe 16, Créteil, France
- ANRS, Paris, France
| | - David C. Montefiori
- Department of Surgery, Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Nicole L. Yates
- Department of Surgery, Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Georgia D. Tomaras
- Department of Surgery, Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Bryan Mayer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Song Ding
- EuroVacc Foundation, Lausanne, Switzerland
| | - Rodolphe Thiébaut
- Inserm, Bordeaux Population Health Research Center, UMR 1219, University Bordeaux, ISPED, CIC 1401-EC, Univ Bordeaux, Bordeaux, France
- CHU de Bordeaux, pôle de santé publique, Bordeaux, France
- INRIA SISTM, Talence, France
| | | | - Geneviève Chêne
- Inserm, Bordeaux Population Health Research Center, UMR 1219, University Bordeaux, ISPED, CIC 1401-EC, Univ Bordeaux, Bordeaux, France
- CHU de Bordeaux, pôle de santé publique, Bordeaux, France
| | - Giuseppe Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Joachim A, Ahmed MIM, Pollakis G, Rogers L, Hoffmann VS, Munseri P, Aboud S, Lyamuya EF, Bakari M, Robb ML, Wahren B, Sandstrom E, Nilsson C, Biberfeld G, Geldmacher C, Held K. Induction of Identical IgG HIV-1 Envelope Epitope Recognition Patterns After Initial HIVIS-DNA/MVA-CMDR Immunization and a Late MVA-CMDR Boost. Front Immunol 2020; 11:719. [PMID: 32411138 PMCID: PMC7198863 DOI: 10.3389/fimmu.2020.00719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/30/2020] [Indexed: 01/16/2023] Open
Abstract
In the RV144 trial, to date the only HIV-1 vaccine efficacy trial demonstrating a modestly reduced risk of HIV-1 acquisition, antibody responses toward the HIV Envelope protein (Env) variable (V) 2 and V3 regions were shown to be correlated with a reduced risk of infection. These potentially protective antibody responses, in parallel with the vaccine efficacy, however, waned quickly. Dissecting vaccine-induced IgG recognition of antigenic regions and their variants within the HIV-1 Env from different vaccine trials will aid in designing future HIV-1 immunogens and vaccination schedules. We, therefore, analyzed the IgG response toward linear HIV-1 Env epitopes elicited by a multi-clade, multigene HIVIS-DNA priming, and heterologous recombinant modified vaccinia virus Ankara (MVA-CMDR) boosting regimen (HIVIS03) and assessed whether a late MVA-CMDR boost 3 years after completion of the initial vaccination schedule (HIVIS06) restored antibody responses toward these epitopes. Here we report that vaccination schedule in the HIVIS03 trial elicited IgG responses against linear epitopes within the V2 and V3 tip as well as against the gp41 immunodominant region in a high proportion of vaccinees. Antibodies against the V2 and gp41 Env regions were restricted to variants with close homology to the MVA-CMDR immunogen sequence, while V3 responses were more cross-reactive. Boosting with a late third MVA-CMDR after 3 years effectively restored waned IgG responses to linear Env epitopes and induced targeting of identical antigenic regions and variants comparable to the previous combined HIVIS-DNA/MVA-CMDR regimen. Our findings support the notion that anti-HIV-1 Env responses, associated with a reduced risk of infection in RV144, could be maintained by regular boosting with a single dose of MVA-CMDR.
Collapse
Affiliation(s)
- Agricola Joachim
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam, Tanzania
| | - Mohamed I M Ahmed
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Georgios Pollakis
- Faculty of Health and Life Science, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,NIHR Health Protection Research Unit in Emerging and Zoonotic Infections (HPRU EZI), Liverpool, United Kingdom
| | - Lisa Rogers
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Verena S Hoffmann
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Munich, Germany
| | - Patricia Munseri
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam, Tanzania
| | - Eligius F Lyamuya
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences (MUHAS), Dar es Salaam, Tanzania
| | - Muhammad Bakari
- Tanzania Ministry of Health, Community Development, Gender, Elderly, and Children, Dodoma, Tanzania
| | - Merlin L Robb
- Walter Reed Army Institute of Research (WRAIR), Rockville, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Britta Wahren
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Eric Sandstrom
- Department of Clinical Science and Education, Karolinska Institutet, Sodersjukhuset, Stockholm, Sweden
| | - Charlotta Nilsson
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.,The Public Health Agency of Sweden, Solna, Sweden
| | - Gunnel Biberfeld
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| |
Collapse
|
16
|
Recruitment using respondent driven sampling, risk behaviors assessment and willingness of young female sex workers (18-25 years) in Dar Es Salaam, Tanzania to participate in HIV vaccine trials. BMC Public Health 2019; 19:1537. [PMID: 31796002 PMCID: PMC6889671 DOI: 10.1186/s12889-019-7822-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/21/2019] [Indexed: 01/31/2023] Open
Abstract
Background Despite the present HIV preventive and treatment programs, the prevalence of HIV is still high in eastern and southern Africa, among young women and populations at high. risk for HIV transmission such as sex workers. There is a need to prepare a suitable population that will participate in efficacy HIV vaccine trials to determine the efficacy of HIV vaccines that had proven to be safe and immune potent. Methods It was a cross-sectional study that recruited 600 female sex workers using respondent-driven sampling in Dar es Salaam. The study examined recruitment approaches, risk behaviors and willingness of young female sex workers to participate in an HIV vaccine trial. Descriptive statistics described risk behaviors and willingness of the participants to participate in efficacy HIV vaccine trials. The logistic regression model computed the likelihood of willingness to participate in the trials with selected variables. Results The study demonstrated 53% were less than 20 years old, 96% were single, and 22% lived in brothels. Eighty percent of the participants started selling sex at the age between 15 and 19 years old, 61% used illicit drugs for the first time when they were less than 20 years old, 24% had anal sex ever. Eighty-nine percent had more than 20-lifetime sexual partners, and 56% had unprotected sexual intercourse with sex clients. Ninety-one percent expressed a willingness to participate in the HIV vaccine trial. Sixty-one percent did not need permission from anyone for participating in a trial. Ninety-one percent expressed willingness to participate in the efficacy of HIV vaccine trial. In the logistic regression model, willingness was significantly associated with the need to ask permission for participation in HIV vaccine trial from sex agent. Conclusion Respondent-driven sampling provided a rapid means of reaching young female sex workers who reported high-risk behaviors. The majority expressed a high level of willingness to participate in the HIV vaccine trial which was marginally correlated to the need to seek consent for participation in the trial from the sex brokers. Future HIV vaccine trials involving this population should consider involving the brokers in the trials because they form an essential part of the community for the participants.
Collapse
|
17
|
Tarimo EAM, Ambikile J, Munseri P, Bakari M. Perception of potential harm and benefits of HIV vaccine trial participation: A qualitative study from urban Tanzania. PLoS One 2019; 14:e0224831. [PMID: 31703092 PMCID: PMC6839895 DOI: 10.1371/journal.pone.0224831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The development of an effective preventive HIV vaccine is the best-known option to halt incident HIV infections. Participants in HIV vaccine trials may possess expectations shaped by existing socio-cultural contexts that are important to understand to allow for improved trial design. Here, we describe post-phase I/II HIV vaccine trial perceptions within participating communities in Dar es Salaam, Tanzania. MATERIALS AND METHODS This descriptive qualitative study was conducted in May 2016. We conducted eight focus group discussions, each consisting of 5 to 12 participants. Four groups comprised of the past phase I/II HIV vaccine trial participants and four groups involved those who did not participate. We used a thematic analysis approach. RESULTS Ongoing concerns existed among non-vaccine trial participants who believed that those who participated in HIV vaccine trials were infected with HIV. Limited post-HIV vaccine trial result dissemination, the pre-existing negative beliefs about vaccines, and experiences from other previous medical experiments fueled these concerns. The participants anticipated that broader dissemination of facts regarding HIV vaccine trials using media, former volunteers, and flyers would reduce the reported concerns. In contrast, some participants embraced the benefits gained through participating in HIV vaccine trials. HIV vaccine trial participants appreciated trial interventions, such as health status check-ups, knowledge acquisition, and facilitation of access to medical services. They envisioned mutual benefits in the form of community protection and capacity building among the local scientists. CONCLUSIONS The future conduct of HIV vaccine trials in Tanzania requires wider community dissemination of information and post-trial feedback to alleviate concerns among the participating communities. Interventions such as medical services may represent essential incentives to the HIV vaccine trial volunteers. In future HIV vaccine trials, it is crucial to boost individual and perceived mutual benefits.
Collapse
Affiliation(s)
- Edith A. M. Tarimo
- Department of Nursing Management, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Joel Ambikile
- Department of Clinical Nursing, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Patricia Munseri
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Muhammad Bakari
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| |
Collapse
|
18
|
Affiliation(s)
| | - Jerome H. Kim
- International Vaccine Institute, Seoul, Republic of Korea
| |
Collapse
|
19
|
Nadai Y, Held K, Joseph S, Ahmed MIM, Hoffmann VS, Peterhoff D, Missanga M, Bauer A, Joachim A, Reimer U, Zerweck J, McCormack S, Cope AV, Tatoud R, Shattock RJ, Robb ML, Sandstroem EG, Hoelscher M, Maboko L, Bakari M, Kroidl A, Wagner R, Weber J, Pollakis G, Geldmacher C. Envelope-Specific Recognition Patterns of HIV Vaccine-Induced IgG Antibodies Are Linked to Immunogen Structure and Sequence. Front Immunol 2019; 10:717. [PMID: 31105688 PMCID: PMC6492543 DOI: 10.3389/fimmu.2019.00717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/18/2019] [Indexed: 11/13/2022] Open
Abstract
Background: A better understanding of the parameters influencing vaccine-induced IgG recognition of individual antigenic regions and their variants within the HIV Envelope protein (Env) can help to improve design of preventive HIV vaccines. Methods: Env-specific IgG responses were mapped in samples of the UKHVC003 Standard Group (UK003SG, n = 11 from UK) and TaMoVac01 (TMV01, n = 17 from Tanzania) HIV vaccine trials. Both trials consisted of three immunizations with DNA, followed by two boosts with recombinant Modified Vaccinia Virus Ankara (MVA), either mediating secretion of gp120 (UK003SG) or the presentation of cell membrane bound gp150 envelopes (TMV01) from infected cells, and an additional two boosts with 5 μg of CN54gp140 protein adjuvanted with glucopyranosyl lipid adjuvant (GLA). Env immunogen sequences in UK003SG were solely based on the clade C isolate CN54, whereas in TMV01 these were based on clades A, C, B, and CRF01AE. The peptide microarray included 8 globally representative Env sequences, CN54gp140 and the MVA-encoded Env immunogens from both trials, as well as additional peptide variants for hot spots of immune recognition. Results: After the second MVA boost, UK003SG vaccinees almost exclusively targeted linear, non-glycosylated antigenic regions located in the inter-gp120 interface. In contrast, TMV01 recipients most strongly targeted the V2 region and an immunodominant region in gp41. The V3 region was frequently targeted in both trials, with a higher recognition magnitude for diverse antigenic variants observed in the UK003SG (p < 0.0001). After boosting with CN54gp140/GLA, the overall response magnitude increased with a more comparable recognition pattern of antigenic regions and variants between the two trials. Recognition of most immunodominant regions within gp120 remained significantly stronger in UK003SG, whereas V2-region recognition was not boosted in either group. Conclusions: IgG recognition of linear antigenic Env regions differed between the two trials particularly after the second MVA boost. Structural features of the MVA-encoded immunogens, such as secreted, monomeric gp120 vs. membrane-anchored, functional gp150, and differences in prime-boost immunogen sequence variability most probably contributed to these differences. Prime-boosting with multivalent Env immunogens during TMV01 did not improve variant cross-recognition of immunodominant peptide variants in the V3 region.
Collapse
Affiliation(s)
- Yuka Nadai
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Kathrin Held
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Sarah Joseph
- MRC Clinical Trials Unit at UCL, London, United Kingdom
| | - Mohamed I M Ahmed
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Verena S Hoffmann
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | | | - Asli Bauer
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,NIMR-Mbeya Medical Research Center, Mbeya, Tanzania
| | - Agricola Joachim
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Ulf Reimer
- JPT Peptide Technologies, Berlin, Germany
| | | | | | - Alethea V Cope
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Roger Tatoud
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Robin J Shattock
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Merlin Lee Robb
- US Military HIV Research Program, Silver Spring, MD, United States
| | - Eric G Sandstroem
- Department of Clinical Science and Education, Karolinska Institutet at Södersjukhuset, Stockholm, Sweden
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | | | - Muhammad Bakari
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Arne Kroidl
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany.,Institute of Clinical Microbiology and Hygiene, University Hospital, Regensburg, Germany
| | - Jonathan Weber
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Georgios Pollakis
- Institute of Global Health (CIMI), University of Liverpool, Liverpool, United Kingdom
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| |
Collapse
|
20
|
Criscuolo E, Caputo V, Diotti RA, Sautto GA, Kirchenbaum GA, Clementi N. Alternative Methods of Vaccine Delivery: An Overview of Edible and Intradermal Vaccines. J Immunol Res 2019; 2019:8303648. [PMID: 30949518 PMCID: PMC6425294 DOI: 10.1155/2019/8303648] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 01/26/2023] Open
Abstract
Vaccines are recognized worldwide as one of the most important tools for combating infectious diseases. Despite the tremendous value conferred by currently available vaccines toward public health, the implementation of additional vaccine platforms is also of key importance. In fact, currently available vaccines possess shortcomings, such as inefficient triggering of a cell-mediated immune response and the lack of protective mucosal immunity. In this regard, recent work has been focused on vaccine delivery systems, as an alternative to injectable vaccines, to increase antigen stability and improve overall immunogenicity. In particular, novel strategies based on edible or intradermal vaccine formulations have been demonstrated to trigger both a systemic and mucosal immune response. These novel vaccination delivery systems offer several advantages over the injectable preparations including self-administration, reduced cost, stability, and elimination of a cold chain. In this review, the latest findings and accomplishments regarding edible and intradermal vaccines are described in the context of the system used for immunogen expression, their molecular features and capacity to induce a protective systemic and mucosal response.
Collapse
Affiliation(s)
- E. Criscuolo
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
| | - V. Caputo
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
- Pomona Ricerca S.r.l., Turin, Italy
| | - R. A. Diotti
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
- Pomona Ricerca S.r.l., Turin, Italy
| | - G. A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | | | - N. Clementi
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
| |
Collapse
|
21
|
Kozlowski PA, Aldovini A. Mucosal Vaccine Approaches for Prevention of HIV and SIV Transmission. CURRENT IMMUNOLOGY REVIEWS 2019; 15:102-122. [PMID: 31452652 PMCID: PMC6709706 DOI: 10.2174/1573395514666180605092054] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Optimal protective immunity to HIV will likely require that plasma cells, memory B cells and memory T cells be stationed in mucosal tissues at portals of viral entry. Mucosal vaccine administration is more effective than parenteral vaccine delivery for this purpose. The challenge has been to achieve efficient vaccine uptake at mucosal surfaces, and to identify safe and effective adjuvants, especially for mucosally administered HIV envelope protein immunogens. Here, we discuss strategies used to deliver potential HIV vaccine candidates in the intestine, respiratory tract, and male and female genital tract of humans and nonhuman primates. We also review mucosal adjuvants, including Toll-like receptor agonists, which may adjuvant both mucosal humoral and cellular immune responses to HIV protein immunogens.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Anna Aldovini
- Department of Medicine, and Harvard Medical School, Boston Children’s Hospital, Department of Pediatrics, Boston MA, 02115, USA
| |
Collapse
|
22
|
Optimizing the immunogenicity of HIV prime-boost DNA-MVA-rgp140/GLA vaccines in a phase II randomized factorial trial design. PLoS One 2018; 13:e0206838. [PMID: 30496299 PMCID: PMC6264478 DOI: 10.1371/journal.pone.0206838] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/18/2018] [Indexed: 12/16/2022] Open
Abstract
Background We evaluated the safety and immunogenicity of (i) an intradermal HIV-DNA regimen given with/without intradermal electroporation (EP) as prime and (ii) the impact of boosting with modified vaccinia virus Ankara (HIV-MVA) administered with or without subtype C CN54rgp140 envelope protein adjuvanted with Glucopyranosyl Lipid A (GLA-AF) in volunteers from Tanzania and Mozambique. Methods Healthy HIV-uninfected adults (N = 191) were randomized twice; first to one of three HIV-DNA intradermal priming regimens by needle-free ZetaJet device at weeks 0, 4 and 12 (Group I: 2x0.1mL [3mg/mL], Group II: 2x0.1mL [3mg/mL] plus EP, Group III: 1x0.1mL [6mg/mL] plus EP). Second the same volunteers received 108 pfu HIV-MVA twice, alone or combined with CN54rgp140/GLA-AF, intramuscularly by syringe, 16 weeks apart. Additionally, 20 volunteers received saline placebo. Results Vaccinations and electroporation did not raise safety concerns. After the last vaccination, the overall IFN-γ ELISpot response rate to either Gag or Env was 97%. Intradermal electroporation significantly increased ELISpot response rates to HIV-DNA-specific Gag (66% group I vs. 86% group II, p = 0.026), but not to the HIV-MVA vaccine-specific Gag or Env peptide pools nor the magnitude of responses. Co-administration of rgp140/GLA-AF with HIV-MVA did not impact the frequency of binding antibody responses against subtype B gp160, C gp140 or E gp120 antigens (95%, 99%, 79%, respectively), but significantly enhanced the magnitude against subtype B gp160 (2700 versus 300, p<0.001) and subtype C gp140 (24300 versus 2700, p<0.001) Env protein. At relatively low titers, neutralizing antibody responses using the TZM-bl assay were more frequent in vaccinees given adjuvanted protein boost. Conclusion Intradermal electroporation increased DNA-induced Gag response rates but did not show an impact on Env-specific responses nor on the magnitude of responses. Co-administration of HIV-MVA with rgp140/GLA-AF significantly enhanced antibody responses.
Collapse
|
23
|
Chissumba RM, Luciano A, Namalango E, Bauer A, Bhatt N, Wahren B, Nilsson C, Geldmacher C, Scarlatti G, Jani I, Kestens L. Regulatory T cell abundance and activation status before and after priming with HIVIS-DNA and boosting with MVA-HIV/rgp140/GLA-AF may impact the magnitude of the vaccine-induced immune responses. Immunobiology 2018; 223:792-801. [PMID: 30121146 DOI: 10.1016/j.imbio.2018.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 08/11/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND Little is known about regulatory CD4 T cells (Tregs) in the context of HIV vaccines. Tregs can be differentiated into resting (FoxP3+CD45RA+ - rTregs), activated (FoxP3HighCD45RA- - aTregs) and memory (FoxP3LowCD45RA- - mTregs). Tregs, as CD4 T cells, are also frequent targets for HIV infection. We studied how the abundance and phenotypes of Tregs in terms of activation status and expression of HIV-1 binding molecules would have changed during vaccination in healthy volunteers participating in a phase IIa HIV vaccine clinical trial. Subjects were primed three times with HIVIS-DNA and boosted twice with MVA-CMDR-HIV alone (n = 12) or MVA-CMDR combined with protein CN54rgp140 (n = 13). The proportions of β7 integrin in all CD4 T cells and in the Tregs subset decreased moderately after the final vaccination (p = 0.001 and p = 0.033, respectively) and the rTregs proportion within the total Tregs were also decreased after the final vaccination (p = 0.038). All these proportions returned to normal values within the three months after the final vaccination. The magnitude of HIV-Envelope-specific IFNγ + T cells after vaccination (r = 0.66; p = 0.021) correlated directly with the proportion of Tregs, and correlated inversely correlated with ratios of Th17/Tregs (r = -0.75; p = 0.0057) and Th17/mTregs (r = -0.78; p = 0.0065). Higher titers of IgG gp140 antibodies were observed in subjects with higher mTregs proportions (r = 0.52; p = 0.022). Interestingly, pre-vaccination levels of mTregs correlated with vaccine-induced Env-binding antibodies (r = 0.57; p = 0.01) and presence of neutralizing antibodies (r = 0.61; p = 0.01), while the pre-vaccination Th17/mTregs ratio correlated inversely with the magnitude of cellular IFN-γ ELISpot responses (r = -0.9; p = 0.002). Taken together, these results suggest that pre- and post-vaccination Tregs, their activation status, the Th17/Tregs ratio and other host factors affecting Treg abundance, have an impact on the magnitude of HIV vaccine-induced immune responses. Moreover, the DNA-HIVIS/MVA-HIV regimen, alone or in combination with CN54rgp140 induced moderate and temporary alterations of the Tregs activation status. We also show a decrease in expression of the HIV-1 ligand β7 integrin on Tregs and all CD4 T cells.
Collapse
Affiliation(s)
- Raquel Matavele Chissumba
- Instituto Nacional de Saúde, Ministry of Health, Maputo, Mozambique; Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| | - Abílio Luciano
- Instituto de Ciências de Saúde, Ministry of Health, Maputo, Mozambique
| | | | - Asli Bauer
- National Institute for Medical Research, Mbeya Medical Research Center, Mbeya, Tanzania
| | - Nilesh Bhatt
- Instituto Nacional de Saúde, Ministry of Health, Maputo, Mozambique
| | - Britta Wahren
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Charlotta Nilsson
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden; Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Department of Immunology, Transplant and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Ilesh Jani
- Instituto Nacional de Saúde, Ministry of Health, Maputo, Mozambique
| | - Luc Kestens
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
24
|
Codon optimization and improved delivery/immunization regimen enhance the immune response against wild-type and drug-resistant HIV-1 reverse transcriptase, preserving its Th2-polarity. Sci Rep 2018; 8:8078. [PMID: 29799015 PMCID: PMC5967322 DOI: 10.1038/s41598-018-26281-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
DNA vaccines require a considerable enhancement of immunogenicity. Here, we optimized a prototype DNA vaccine against drug-resistant HIV-1 based on a weak Th2-immunogen, HIV-1 reverse transcriptase (RT). We designed expression-optimized genes encoding inactivated wild-type and drug-resistant RTs (RT-DNAs) and introduced them into mice by intradermal injections followed by electroporation. RT-DNAs were administered as single or double primes with or without cyclic-di-GMP, or as a prime followed by boost with RT-DNA mixed with a luciferase-encoding plasmid (“surrogate challenge”). Repeated primes improved cellular responses and broadened epitope specificity. Addition of cyclic-di-GMP induced a transient increase in IFN-γ production. The strongest anti-RT immune response was achieved in a prime-boost protocol with electroporation by short 100V pulses done using penetrating electrodes. The RT-specific response, dominated by CD4+ T-cells, targeted epitopes at aa 199–220 and aa 528–543. Drug-resistance mutations disrupted the epitope at aa 205–220, while the CTL epitope at aa 202–210 was not affected. Overall, multiparametric optimization of RT strengthened its Th2- performance. A rapid loss of RT/luciferase-expressing cells in the surrogate challenge experiment revealed a lytic potential of anti-RT response. Such lytic CD4+ response would be beneficial for an HIV vaccine due to its comparative insensitivity to immune escape.
Collapse
|
25
|
Viegas EO, Tembe N, Nilsson C, Meggi B, Maueia C, Augusto O, Stout R, Scarlatti G, Ferrari G, Earl PL, Wahren B, Andersson S, Robb ML, Osman N, Biberfeld G, Jani I, Sandström E, the TaMoVac Study Group. Intradermal HIV-1 DNA Immunization Using Needle-Free Zetajet Injection Followed by HIV-Modified Vaccinia Virus Ankara Vaccination Is Safe and Immunogenic in Mozambican Young Adults: A Phase I Randomized Controlled Trial. AIDS Res Hum Retroviruses 2018; 34:193-205. [PMID: 28969431 DOI: 10.1089/aid.2017.0121] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We assessed the safety and immunogenicity of HIV-DNA priming using Zetajet™, a needle-free device intradermally followed by intramuscular HIV-MVA boosts, in 24 healthy Mozambicans. Volunteers were randomized to receive three immunizations of 600 μg (n = 10; 2 × 0.1 ml) or 1,200 μg (n = 10; 2 × 0.2 ml) of HIV-DNA (3 mg/ml), followed by two boosts of 108 pfu HIV-MVA. Four subjects received placebo saline injections. Vaccines and injections were safe and well tolerated with no difference between the two priming groups. After three HIV-DNA immunizations, IFN-γ ELISpot responses to Gag were detected in 9/17 (53%) vaccinees, while none responded to Envelope (Env). After the first HIV-MVA, the overall response rate to Gag and/or Env increased to 14/15 (93%); 14/15 (93%) to Gag and 13/15 (87%) to Env. There were no significant differences between the immunization groups in frequency of response to Gag and Env or magnitude of Gag responses. Env responses were significantly higher in the higher dose group (median 420 vs. 157.5 SFC/million peripheral blood mononuclear cell, p = .014). HIV-specific antibodies to subtype C gp140 and subtype B gp160 were elicited in all vaccinees after the second HIV-MVA, without differences in titers between the groups. Neutralizing antibody responses were not detected. Two (13%) of 16 vaccinees, one in each of the priming groups, exhibited antibodies mediating antibody-dependent cellular cytotoxicity to CRF01_AE. In conclusion, HIV-DNA vaccine delivered intradermally in volumes of 0.1-0.2 ml using Zetajet was safe and well tolerated. Priming with the 1,200 μg dose of HIV-DNA generated higher magnitudes of ELISpot responses to Env.
Collapse
Affiliation(s)
- Edna Omar Viegas
- Instituto Nacional de Saúde, Maputo, Mozambique
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Eduardo Mondlane University, Maputo, Mozambique
| | - Nelson Tembe
- Instituto Nacional de Saúde, Maputo, Mozambique
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Eduardo Mondlane University, Maputo, Mozambique
| | - Charlotta Nilsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Public Health Agency of Sweden, Stockholm, Sweden
| | | | | | | | | | | | - Guido Ferrari
- Department of Surgery and Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina
| | - Patricia L. Earl
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAD)/National Institutes of Health (NIH), Bethesda, Maryland
| | - Britta Wahren
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sören Andersson
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Merlin L. Robb
- The Military HIV Research Program, Walter Reed Army Institute of Research and The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland
| | | | - Gunnel Biberfeld
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ilesh Jani
- Instituto Nacional de Saúde, Maputo, Mozambique
| | - Eric Sandström
- Department of Education and Clinical Research, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
26
|
Ake JA, Schuetz A, Pegu P, Wieczorek L, Eller MA, Kibuuka H, Sawe F, Maboko L, Polonis V, Karasavva N, Weiner D, Sekiziyivu A, Kosgei J, Missanga M, Kroidl A, Mann P, Ratto-Kim S, Anne Eller L, Earl P, Moss B, Dorsey-Spitz J, Milazzo M, Laissa Ouedraogo G, Rizvi F, Yan J, Khan AS, Peel S, Sardesai NY, Michael NL, Ngauy V, Marovich M, Robb ML. Safety and Immunogenicity of PENNVAX-G DNA Prime Administered by Biojector 2000 or CELLECTRA Electroporation Device With Modified Vaccinia Ankara-CMDR Boost. J Infect Dis 2017; 216:1080-1090. [PMID: 28968759 DOI: 10.1093/infdis/jix456] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/01/2017] [Indexed: 01/24/2023] Open
Abstract
Background We report the first-in-human safety and immunogenicity evaluation of PENNVAX-G DNA/modified vaccinia Ankara-Chiang Mai double recombinant (MVA-CMDR) prime-boost human immuonodeficiency virus (HIV) vaccine, with intramuscular DNA delivery by either Biojector 2000 needle-free injection system (Biojector) or CELLECTRA electroporation device. Methods Healthy, HIV-uninfected adults were randomized to receive 4 mg of PENNVAX-G DNA delivered intramuscularly by Biojector or electroporation at baseline and week 4 followed by intramuscular injection of 108 plaque forming units of MVA-CMDR at weeks 12 and 24. The open-label part A was conducted in the United States, followed by a double-blind, placebo-controlled part B in East Africa. Solicited and unsolicited adverse events were recorded, and immune responses were measured. Results Eighty-eight of 100 enrolled participants completed all study injections, which were generally safe and well tolerated, with more immediate, but transient, pain in the electroporation group. Cellular responses were observed in 57% of vaccine recipients tested and were CD4 predominant. High rates of binding antibody responses to CRF01_AE antigens, including gp70 V1V2 scaffold, were observed. Neutralizing antibodies were detected in a peripheral blood mononuclear cell assay, and moderate antibody-dependent, cell-mediated cytotoxicity activity was demonstrated. Discussion The PVG/MVA-CMDR HIV-1 vaccine regimen is safe and immunogenic. Substantial differences in safety or immunogenicity between modes of DNA delivery were not observed. Clinical Trials Registration NCT01260727.
Collapse
Affiliation(s)
- Julie A Ake
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring
| | - Alexandra Schuetz
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda.,Armed Forces Research Institute of Medical Sciences, Department of Retrovirology, Bangkok, Thailand
| | - Poonam Pegu
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Lindsay Wieczorek
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Michael A Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Hannah Kibuuka
- Makerere University/Walter Reed Project, Kampala, Uganda
| | | | - Leonard Maboko
- National Institute of Medical Research, Mbeya Medical Research Centre, Mbeya, United Republic of Tanzania
| | - Victoria Polonis
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring
| | - Nicos Karasavva
- Armed Forces Research Institute of Medical Sciences, Department of Retrovirology, Bangkok, Thailand
| | | | | | | | - Marco Missanga
- National Institute of Medical Research, Mbeya Medical Research Centre, Mbeya, United Republic of Tanzania
| | - Arne Kroidl
- National Institute of Medical Research, Mbeya Medical Research Centre, Mbeya, United Republic of Tanzania.,Division of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich, Germany
| | - Philipp Mann
- National Institute of Medical Research, Mbeya Medical Research Centre, Mbeya, United Republic of Tanzania.,Division of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich, Germany
| | - Silvia Ratto-Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Leigh Anne Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | | | | | - Julie Dorsey-Spitz
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - Mark Milazzo
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| | - G Laissa Ouedraogo
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda
| | - Farrukh Rizvi
- Military Infectious Diseases Research Program, Ft. Detrick, Maryland
| | - Jian Yan
- Inovio Pharmaceuticals, Inc, Plymouth Meeting, Pennsylvania
| | - Amir S Khan
- Inovio Pharmaceuticals, Inc, Plymouth Meeting, Pennsylvania
| | - Sheila Peel
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring
| | | | - Nelson L Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring
| | - Viseth Ngauy
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Armed Forces Research Institute of Medical Sciences, Department of Retrovirology, Bangkok, Thailand
| | - Mary Marovich
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring
| | - Merlin L Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda
| |
Collapse
|
27
|
Okeke MI, Okoli AS, Diaz D, Offor C, Oludotun TG, Tryland M, Bøhn T, Moens U. Hazard Characterization of Modified Vaccinia Virus Ankara Vector: What Are the Knowledge Gaps? Viruses 2017; 9:v9110318. [PMID: 29109380 PMCID: PMC5707525 DOI: 10.3390/v9110318] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/21/2017] [Accepted: 10/26/2017] [Indexed: 12/17/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is the vector of choice for human and veterinary applications due to its strong safety profile and immunogenicity in vivo. The use of MVA and MVA-vectored vaccines against human and animal diseases must comply with regulatory requirements as they pertain to environmental risk assessment, particularly the characterization of potential adverse effects to humans, animals and the environment. MVA and recombinant MVA are widely believed to pose low or negligible risk to ecosystem health. However, key aspects of MVA biology require further research in order to provide data needed to evaluate the potential risks that may occur due to the use of MVA and MVA-vectored vaccines. The purpose of this paper is to identify knowledge gaps in the biology of MVA and recombinant MVA that are of relevance to its hazard characterization and discuss ongoing and future experiments aimed at providing data necessary to fill in the knowledge gaps. In addition, we presented arguments for the inclusion of uncertainty analysis and experimental investigation of verifiable worst-case scenarios in the environmental risk assessment of MVA and recombinant MVA. These will contribute to improved risk assessment of MVA and recombinant MVA vaccines.
Collapse
Affiliation(s)
- Malachy I Okeke
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
| | - Arinze S Okoli
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
| | - Diana Diaz
- Molecular Inflammation Research Group, Institute of Medical Biology, University i Tromsø (UiT)-The Arctic University of Norway, N-9037 Tromso, Norway.
| | - Collins Offor
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences Piaristengasse 1, A-3500 Krems, Austria.
| | - Taiwo G Oludotun
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences Piaristengasse 1, A-3500 Krems, Austria.
| | - Morten Tryland
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
- Artic Infection Biology, Department of Artic and Marine Biology, UIT-The Artic University of Norway, N-9037 Tromso, Norway.
| | - Thomas Bøhn
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
| | - Ugo Moens
- Molecular Inflammation Research Group, Institute of Medical Biology, University i Tromsø (UiT)-The Arctic University of Norway, N-9037 Tromso, Norway.
| |
Collapse
|
28
|
Chea LS, Amara RR. Immunogenicity and efficacy of DNA/MVA HIV vaccines in rhesus macaque models. Expert Rev Vaccines 2017; 16:973-985. [PMID: 28838267 DOI: 10.1080/14760584.2017.1371594] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Despite 30 years of research on HIV, a vaccine to prevent infection and limit disease progression remains elusive. The RV144 trial showed moderate, but significant protection in humans and highlighted the contribution of antibody responses directed against HIV envelope as an important immune correlate for protection. Efforts to further build upon the progress include the use of a heterologous prime-boost regimen using DNA as the priming agent and the attenuated vaccinia virus, Modified Vaccinia Ankara (MVA), as a boosting vector for generating protective HIV-specific immunity. Areas covered: In this review, we summarize the immunogenicity of DNA/MVA vaccines in non-human primate models and describe the efficacy seen in SIV infection models. We discuss immunological correlates of protection determined by these studies and potential approaches for improving the protective immunity. Additionally, we describe the current progress of DNA/MVA vaccines in human trials. Expert commentary: Efforts over the past decade have provided the opportunity to better understand the dynamics of vaccine-induced immune responses and immune correlates of protection against HIV. Based on what we have learned, we outline multiple areas where the field will likely focus on in the next five years.
Collapse
Affiliation(s)
- Lynette Siv Chea
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| | - Rama Rao Amara
- a Emory Vaccine Center, Department of Microbiology and Immunology , Yerkes National Primate Research Center, Emory University , Atlanta , GA , USA
| |
Collapse
|
29
|
Preferential Targeting of Conserved Gag Regions after Vaccination with a Heterologous DNA Prime-Modified Vaccinia Virus Ankara Boost HIV-1 Vaccine Regimen. J Virol 2017; 91:JVI.00730-17. [PMID: 28701395 DOI: 10.1128/jvi.00730-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/14/2017] [Indexed: 11/20/2022] Open
Abstract
Prime-boost vaccination strategies against HIV-1 often include multiple variants for a given immunogen for better coverage of the extensive viral diversity. To study the immunologic effects of this approach, we characterized breadth, phenotype, function, and specificity of Gag-specific T cells induced by a DNA-prime modified vaccinia virus Ankara (MVA)-boost vaccination strategy, which uses mismatched Gag immunogens in the TamoVac 01 phase IIa trial. Healthy Tanzanian volunteers received three injections of the DNA-SMI vaccine encoding a subtype B and AB-recombinant Gagp37 and two vaccinations with MVA-CMDR encoding subtype A Gagp55 Gag-specific T-cell responses were studied in 42 vaccinees using fresh peripheral blood mononuclear cells. After the first MVA-CMDR boost, vaccine-induced gamma interferon-positive (IFN-γ+) Gag-specific T-cell responses were dominated by CD4+ T cells (P < 0.001 compared to CD8+ T cells) that coexpressed interleukin-2 (IL-2) (66.4%) and/or tumor necrosis factor alpha (TNF-α) (63.7%). A median of 3 antigenic regions were targeted with a higher-magnitude median response to Gagp24 regions, more conserved between prime and boost, compared to those of regions within Gagp15 (not primed) and Gagp17 (less conserved; P < 0.0001 for both). Four regions within Gagp24 each were targeted by 45% to 74% of vaccinees upon restimulation with DNA-SMI-Gag matched peptides. The response rate to individual antigenic regions correlated with the sequence homology between the MVA- and DNA Gag-encoded immunogens (P = 0.04, r2 = 0.47). In summary, after the first MVA-CMDR boost, the sequence-mismatched DNA-prime MVA-boost vaccine strategy induced a Gag-specific T-cell response that was dominated by polyfunctional CD4+ T cells and that targeted multiple antigenic regions within the conserved Gagp24 protein.IMPORTANCE Genetic diversity is a major challenge for the design of vaccines against variable viruses. While including multiple variants for a given immunogen in prime-boost vaccination strategies is one approach that aims to improve coverage for global virus variants, the immunologic consequences of this strategy have been poorly defined so far. It is unclear whether inclusion of multiple variants in prime-boost vaccination strategies improves recognition of variant viruses by T cells and by which mechanisms this would be achieved, either by improved cross-recognition of multiple variants for a given antigenic region or through preferential targeting of antigenic regions more conserved between prime and boost. Engineering vaccines to induce adaptive immune responses that preferentially target conserved antigenic regions of viral vulnerability might facilitate better immune control after preventive and therapeutic vaccination for HIV and for other variable viruses.
Collapse
|
30
|
Hinkula J, Petkov S, Ljungberg K, Hallengärd D, Bråve A, Isaguliants M, Falkeborn T, Sharma S, Liakina V, Robb M, Eller M, Moss B, Biberfeld G, Sandström E, Nilsson C, Markland K, Blomberg P, Wahren B. HIVIS-DNA or HIVISopt-DNA priming followed by CMDR vaccinia-based boosts induce both humoral and cellular murine immune responses to HIV. Heliyon 2017; 3:e00339. [PMID: 28721397 PMCID: PMC5496381 DOI: 10.1016/j.heliyon.2017.e00339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023] Open
Abstract
Background In order to develop a more effective prophylactic HIV-1 vaccine it is important optimize the components, improve Envelope glycoprotein immunogenicity as well as to explore prime-boost immunization schedules. It is also valuable to include several HIV-1 subtype antigens representing the world-wide epidemic. Methods HIVIS-DNA plasmids which include Env genes of subtypes A, B and C together with Gag subtypes A and B and RTmut/Rev of subtype B were modified as follows: the Envelope sequences were shortened, codon optimized, provided with an FT4 sequence and an immunodominant region mutated. The reverse transcriptase (RT) gene was shortened to contain the most immunogenic N-terminal fragment and fused with an inactivated viral protease vPR gene. HIVISopt-DNA thus contains fewer plasmids but additional PR epitopes compared to the native HIVIS-DNA. DNA components were delivered intradermally to young Balb/c mice once, using a needle-free Biojector® immediately followed by dermal electroporation. Vaccinia-based MVA-CMDR boosts including Env gene E and Gag-RT genes A were delivered intramuscularly by needle, once or twice. Results Both HIVIS-DNA and HIVISopt-DNA primed humoral and cell mediated responses well. When boosted with heterologous MVA-CMDR (subtypes A and E) virus inhibitory neutralizing antibodies were obtained to HIV-1 subtypes A, B, C and AE. Both plasmid compositions boosted with MVA-CMDR generated HIV-1 specific cellular responses directed against HIV-1 Env, Gag and Pol, as measured by IFNγ ELISpot. It was shown that DNA priming augmented the vector MVA immunological boosting effects, the HIVISopt-DNA with a trend to improved (Env) neutralization, the HIVIS-DNA with a trend to better (Gag) cell mediated immune reponses. Conclusions HIVIS-DNA was modified to obtain HIVISopt-DNA that had fewer plasmids, and additional epitopes. Even with one DNA prime followed by two MVA-CMDR boosts, humoral and cell-mediated immune responses were readily induced by priming with either DNA construct composition. Priming by HIV-DNA augmented neutralizing antibody responses revealed by boosting with the vaccinia-based heterologous sequences. Cellular and antibody responses covered selected strains representing HIV-1 subtypes A, B, C and CRF01_AE. We assume this is related to the inclusion of heterologous full genes in the vaccine schedule.
Collapse
Affiliation(s)
- J Hinkula
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden.,Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - S Petkov
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - K Ljungberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - D Hallengärd
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - A Bråve
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - M Isaguliants
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - T Falkeborn
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - S Sharma
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - V Liakina
- Faculty of Medicine, Vilnius University 2, 08661 Vilnius, Lithuania
| | - M Robb
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, 20892 MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, 20892 MD, USA
| | - M Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, 20892 MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, 20892 MD, USA
| | - B Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, 20892 MD, USA
| | - G Biberfeld
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - E Sandström
- Department of South Hospital, Karolinska Institutet, 11883 Stockholm, Sweden
| | - C Nilsson
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - K Markland
- Clinical Research Center and Vecura, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - P Blomberg
- Clinical Research Center and Vecura, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - B Wahren
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
31
|
Fusion to Flaviviral Leader Peptide Targets HIV-1 Reverse Transcriptase for Secretion and Reduces Its Enzymatic Activity and Ability to Induce Oxidative Stress but Has No Major Effects on Its Immunogenic Performance in DNA-Immunized Mice. J Immunol Res 2017; 2017:7407136. [PMID: 28717654 PMCID: PMC5498913 DOI: 10.1155/2017/7407136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/13/2017] [Indexed: 01/10/2023] Open
Abstract
Reverse transcriptase (RT) is a key enzyme in viral replication and susceptibility to ART and a crucial target of immunotherapy against drug-resistant HIV-1. RT induces oxidative stress which undermines the attempts to make it immunogenic. We hypothesized that artificial secretion may reduce the stress and make RT more immunogenic. Inactivated multidrug-resistant RT (RT1.14opt-in) was N-terminally fused to the signal providing secretion of NS1 protein of TBEV (Ld) generating optimized inactivated Ld-carrying enzyme RT1.14oil. Promotion of secretion prohibited proteasomal degradation increasing the half-life and content of RT1.14oil in cells and cell culture medium, drastically reduced the residual polymerase activity, and downmodulated oxidative stress. BALB/c mice were DNA-immunized with RT1.14opt-in or parental RT1.14oil by intradermal injections with electroporation. Fluorospot and ELISA tests revealed that RT1.14opt-in and RT1.14oil induced IFN-γ/IL-2, RT1.14opt-in induced granzyme B, and RT1.14oil induced perforin production. Perforin secretion correlated with coproduction of IFN-γ and IL-2 (R = 0,97). Both DNA immunogens induced strong anti-RT antibody response. Ld peptide was not immunogenic. Thus, Ld-driven secretion inferred little change to RT performance in DNA immunization. Positive outcome was the abrogation of polymerase activity increasing safety of RT-based DNA vaccines. Identification of the molecular determinants of low cellular immunogenicity of RT requires further studies.
Collapse
|
32
|
Billings E, Sanders-Buell E, Bose M, Kijak GH, Bradfield A, Crossler J, Arroyo MA, Maboko L, Hoffmann O, Geis S, Birx DL, Kim JH, Michael NL, Robb ML, Hoelscher M, Tovanabutra S. HIV-1 Genetic Diversity Among Incident Infections in Mbeya, Tanzania. AIDS Res Hum Retroviruses 2017; 33:373-381. [PMID: 27841669 PMCID: PMC5372774 DOI: 10.1089/aid.2016.0111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In preparation for vaccine trials, HIV-1 genetic diversity was surveyed between 2002 and 2006 through the Cohort Development study in the form of a retrospective and prospective observational study in and around the town of Mbeya in Tanzania's Southwest Highlands. This study describes the molecular epidemiology of HIV-1 strains obtained from 97 out of 106 incident HIV-1 infections identified in three subpopulations of participants (one rural, two urban) from the Mbeya area. Near full-genome or half-genome sequencing showed a subtype distribution of 40% C, 17% A1, 1% D, and 42% inter-subtype recombinants. Compared to viral subtyping results previously obtained from the retrospective phase of this study, the overall proportion of incident viral strains did not change greatly during the study course, suggesting maturity of the epidemic. A comparison to a current Phase I-II vaccine being tested in Africa shows ∼17% amino acid sequence difference between the gp120 of the vaccine and subtype C incident strains. Phylogenetic and recombinant breakpoint analysis of the incident strains revealed the emergence of CRF41_CD and many unique recombinants, as well as the presence of six local transmission networks most of which were confined to the rural subpopulation. In the context of vaccine cohort selection, these results suggest distinct infection transmission dynamics within these three geographically close subpopulations. The diversity and genetic sequences of the HIV-1 strains obtained during this study will greatly contribute to the planning, immunogen selection, and analysis of vaccine-induced immune responses observed during HIV-1 vaccine trials in Tanzania and neighboring countries.
Collapse
Affiliation(s)
- Erik Billings
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| | - Eric Sanders-Buell
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| | - Meera Bose
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| | - Gustavo H. Kijak
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| | - Andrea Bradfield
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| | - Jacqueline Crossler
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| | - Miguel A. Arroyo
- United States Military HIV Research Program/Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | - Oliver Hoffmann
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
- NIMR-Mbeya Medical Research Center, Mbeya, Tanzania
| | - Steffen Geis
- NIMR-Mbeya Medical Research Center, Mbeya, Tanzania
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
| | - Deborah L. Birx
- United States Military HIV Research Program/Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Jerome H. Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Nelson L. Michael
- United States Military HIV Research Program/Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Merlin L. Robb
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
- German Centre for Infection Research (DZIF), Munich partner site, Munich, Germany
| | - Sodsai Tovanabutra
- United States Military HIV Research Program/Henry M. Jackson Foundation, Silver Spring, Maryland
| |
Collapse
|
33
|
Joseph S, Quinn K, Greenwood A, Cope AV, McKay PF, Hayes PJ, Kopycinski JT, Gilmour J, Miller AN, Geldmacher C, Nadai Y, Ahmed MIM, Montefiori DC, Dally L, Bouliotis G, Lewis DJM, Tatoud R, Wagner R, Esteban M, Shattock RJ, McCormack S, Weber J. A Comparative Phase I Study of Combination, Homologous Subtype-C DNA, MVA, and Env gp140 Protein/Adjuvant HIV Vaccines in Two Immunization Regimes. Front Immunol 2017; 8:149. [PMID: 28275375 PMCID: PMC5319954 DOI: 10.3389/fimmu.2017.00149] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 01/30/2017] [Indexed: 01/11/2023] Open
Abstract
There remains an urgent need for a prophylactic HIV vaccine. We compared combined MVA and adjuvanted gp140 to sequential MVA/gp140 after DNA priming. We expected Env-specific CD4+ T-cells after DNA and MVA priming, and Env-binding antibodies in 100% individuals after boosting with gp140 and that combined vaccines would not compromise safety and might augment immunogenicity. Forty volunteers were primed three times with DNA plasmids encoding (CN54) env and (ZM96) gag-pol-nef at 0, 4 and 8 weeks then boosted with MVA-C (CN54 env and gag-pol-nef) and glucopyranosyl lipid adjuvant—aqueous formulation (GLA-AF) adjuvanted CN54gp140. They were randomised to receive them in combination at the same visit at 16 and 20 weeks (accelerated) or sequentially with MVA-C at 16, 20, and GLA-AF/gp140 at 24 and 28 weeks (standard). All vaccinations were intramuscular. Primary outcomes included ≥grade 3 safety events and the titer of CN54gp140-specific binding IgG. Other outcomes included neutralization, binding antibody specificity and T-cell responses. Two participants experienced asymptomatic ≥grade 3 transaminitis leading to discontinuation of vaccinations, and three had grade 3 solicited local or systemic reactions. A total of 100% made anti-CN54gp140 IgG and combining vaccines did not significantly alter the response; geometric mean titer 6424 (accelerated) and 6578 (standard); neutralization of MW965.2 Tier 1 pseudovirus was superior in the standard group (82 versus 45% responders, p = 0.04). T-cell ELISpot responses were CD4+ and Env-dominant; 85 and 82% responding in the accelerated and standard groups, respectively. Vaccine-induced IgG responses targeted multiple regions within gp120 with the V3 region most immunodominant and no differences between groups detected. Combining MVA and gp140 vaccines did not result in increased adverse events and did not significantly impact upon the titer of Env-specific binding antibodies, which were seen in 100% individuals. The approach did however affect other immune responses; neutralizing antibody responses, seen only to Tier 1 pseudoviruses, were poorer when the vaccines were combined and while T-cell responses were seen in >80% individuals in both groups and similarly CD4 and Env dominant, their breadth/polyfunctionality tended to be lower when the vaccines were combined, suggesting attenuation of immunogenicity and cautioning against this accelerated regimen.
Collapse
Affiliation(s)
- Sarah Joseph
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London , London , UK
| | - Killian Quinn
- Department of Medicine, Imperial College London , London , UK
| | | | - Alethea V Cope
- Department of Medicine, Imperial College London , London , UK
| | - Paul F McKay
- Department of Medicine, Imperial College London , London , UK
| | - Peter J Hayes
- IAVI Human Immunology Laboratory, Imperial College London , London , UK
| | | | - Jill Gilmour
- IAVI Human Immunology Laboratory, Imperial College London , London , UK
| | - Aleisha N Miller
- ICTU, Department of Public Health, Imperial College London , London , UK
| | - Christof Geldmacher
- Department of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich, Munich, Germany; German Centre for Infection Research (DZIF), Munich, Germany
| | - Yuka Nadai
- Department of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich, Munich, Germany; German Centre for Infection Research (DZIF), Munich, Germany
| | - Mohamed I M Ahmed
- Department of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich, Munich, Germany; German Centre for Infection Research (DZIF), Munich, Germany
| | | | - Len Dally
- The EMMES Corporation , Rockville, MD , USA
| | - George Bouliotis
- ICTU, Department of Public Health, Imperial College London , London , UK
| | - David J M Lewis
- Clinical Research Centre, University of Surrey, Guildford, UK; Clinical Research Facility, Imperial College Healthcare NHS Trust, London, UK
| | - Roger Tatoud
- Department of Medicine, Imperial College London , London , UK
| | - Ralf Wagner
- University of Regensburg and University Hospital Regensburg , Regensburg , Germany
| | | | | | - Sheena McCormack
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London , London , UK
| | - Jonathan Weber
- Department of Medicine, Imperial College London , London , UK
| |
Collapse
|
34
|
The Influence of Community Members on Participation by Youth in an HIV Vaccine Trial in Tanzania. PLoS One 2016; 11:e0168660. [PMID: 27997617 PMCID: PMC5173279 DOI: 10.1371/journal.pone.0168660] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 12/05/2016] [Indexed: 11/19/2022] Open
Abstract
In sub-Saharan Africa, the burden of HIV is high among young people and it is of the utmost importance that they be recruited into vaccination trials. Since community members influence the willingness of young people to participate in the vaccination trials, ascertaining their opinions is essential to overcoming barriers to such participation. Here, in seven focus group discussions we explored the views of 44 community members identified as someone they felt close by youth in Tanzania. The transcripts of these discussions were examined using content analysis. Our participants expressed that community members would be directly involved in the decisions of young people about whether or not to participate in an HIV vaccine trial. In general, they felt that community members would provide social support for youth during the trial and perceived that youth might have misconceptions concerning the vaccine and trial process. The participants pointed out structural factors such as substance use, poverty, stigma and unemployment that are barriers to participation. In conclusion, involvement of community members could be an integral part of the recruitment and retention of young people in HIV vaccine trials in Tanzania.
Collapse
|
35
|
Volz A, Sutter G. Modified Vaccinia Virus Ankara: History, Value in Basic Research, and Current Perspectives for Vaccine Development. Adv Virus Res 2016; 97:187-243. [PMID: 28057259 PMCID: PMC7112317 DOI: 10.1016/bs.aivir.2016.07.001] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Safety tested Modified Vaccinia virus Ankara (MVA) is licensed as third-generation vaccine against smallpox and serves as a potent vector system for development of new candidate vaccines against infectious diseases and cancer. Historically, MVA was developed by serial tissue culture passage in primary chicken cells of vaccinia virus strain Ankara, and clinically used to avoid the undesirable side effects of conventional smallpox vaccination. Adapted to growth in avian cells MVA lost the ability to replicate in mammalian hosts and lacks many of the genes orthopoxviruses use to conquer their host (cell) environment. As a biologically well-characterized mutant virus, MVA facilitates fundamental research to elucidate the functions of poxvirus host-interaction factors. As extremely safe viral vectors MVA vaccines have been found immunogenic and protective in various preclinical infection models. Multiple recombinant MVA currently undergo clinical testing for vaccination against human immunodeficiency viruses, Mycobacterium tuberculosis or Plasmodium falciparum. The versatility of the MVA vector vaccine platform is readily demonstrated by the swift development of experimental vaccines for immunization against emerging infections such as the Middle East Respiratory Syndrome. Recent advances include promising results from the clinical testing of recombinant MVA-producing antigens of highly pathogenic avian influenza virus H5N1 or Ebola virus. This review summarizes our current knowledge about MVA as a unique strain of vaccinia virus, and discusses the prospects of exploiting this virus as research tool in poxvirus biology or as safe viral vector vaccine to challenge existing and future bottlenecks in vaccinology.
Collapse
Affiliation(s)
- A Volz
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany
| | - G Sutter
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany.
| |
Collapse
|
36
|
Joachim A, Bauer A, Joseph S, Geldmacher C, Munseri PJ, Aboud S, Missanga M, Mann P, Wahren B, Ferrari G, Polonis VR, Robb ML, Weber J, Tatoud R, Maboko L, Hoelscher M, Lyamuya EF, Biberfeld G, Sandström E, Kroidl A, Bakari M, Nilsson C, McCormack S. Boosting with Subtype C CN54rgp140 Protein Adjuvanted with Glucopyranosyl Lipid Adjuvant after Priming with HIV-DNA and HIV-MVA Is Safe and Enhances Immune Responses: A Phase I Trial. PLoS One 2016; 11:e0155702. [PMID: 27192151 PMCID: PMC4871571 DOI: 10.1371/journal.pone.0155702] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/02/2016] [Indexed: 12/18/2022] Open
Abstract
Background A vaccine against HIV is widely considered the most effective and sustainable way of reducing new infections. We evaluated the safety and impact of boosting with subtype C CN54rgp140 envelope protein adjuvanted in glucopyranosyl lipid adjuvant (GLA-AF) in Tanzanian volunteers previously given three immunizations with HIV-DNA followed by two immunizations with recombinant modified vaccinia virus Ankara (HIV-MVA). Methods Forty volunteers (35 vaccinees and five placebo recipients) were given two CN54rgp140/GLA-AF immunizations 30–71 weeks after the last HIV-MVA vaccination. These immunizations were delivered intramuscularly four weeks apart. Results The vaccine was safe and well tolerated except for one episode of asymptomatic hypoglycaemia that was classified as severe adverse event. Two weeks after the second HIV-MVA vaccination 34 (97%) of the 35 previously vaccinated developed Env-specific binding antibodies, and 79% and 84% displayed IFN-γ ELISpot responses to Gag and Env, respectively. Binding antibodies to subtype C Env (included in HIV-DNA and protein boost), subtype B Env (included only in HIV-DNA) and CRF01_AE Env (included only in HIV-MVA) were significantly boosted by the CN54rgp140/GLA-AF immunizations. Functional antibodies detected using an infectious molecular clone virus/peripheral blood mononuclear cell neutralization assay, a pseudovirus/TZM-bl neutralization assay or by assays for antibody-dependent cellular cytotoxicity (ADCC) were not significantly boosted. In contrast, T-cell proliferative responses to subtype B MN antigen and IFN-γ ELISpot responses to Env peptides were significantly enhanced. Four volunteers not primed with HIV-DNA and HIV-MVA before the CN54rgp140/GLA-AF immunizations mounted an antibody response, while cell-mediated responses were rare. After the two Env subtype C protein immunizations, a trend towards higher median subtype C Env binding antibody titers was found in vaccinees who had received HIV-DNA and HIV-MVA prior to the two Env protein immunizations as compared to unprimed vaccinees (p = 0.07). Conclusion We report excellent tolerability, enhanced binding antibody responses and Env-specific cell-mediated immune responses but no ADCC antibody increase after two immunizations with a subtype C rgp140 protein adjuvanted in GLA-AF in healthy volunteers previously immunized with HIV-DNA and HIV-MVA. Trial Registration International Clinical Trials Registry PACTR2010050002122368
Collapse
Affiliation(s)
- Agricola Joachim
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: ;
| | - Asli Bauer
- National Institute for Medical Research-Mbeya, Medical Research Center, Mbeya, Tanzania
- Department of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich (LMU), Munich, Germany
| | - Sarah Joseph
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Christof Geldmacher
- Department of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich (LMU), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Patricia J. Munseri
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Marco Missanga
- National Institute for Medical Research-Mbeya, Medical Research Center, Mbeya, Tanzania
| | - Philipp Mann
- National Institute for Medical Research-Mbeya, Medical Research Center, Mbeya, Tanzania
| | - Britta Wahren
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Guido Ferrari
- Department of Surgery and Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Victoria R. Polonis
- The Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Merlin L. Robb
- The Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- The Military HIV Research Program, The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | | | | | - Leonard Maboko
- National Institute for Medical Research-Mbeya, Medical Research Center, Mbeya, Tanzania
| | - Michael Hoelscher
- National Institute for Medical Research-Mbeya, Medical Research Center, Mbeya, Tanzania
- Department of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich (LMU), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Eligius F. Lyamuya
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Gunnel Biberfeld
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Eric Sandström
- Venhälsan, Karolinska Insitutet at Södersjukhuset, Stockholm, Sweden
| | - Arne Kroidl
- National Institute for Medical Research-Mbeya, Medical Research Center, Mbeya, Tanzania
- Department of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich (LMU), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Muhammad Bakari
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Charlotta Nilsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- The Public Health Agency of Sweden, Solna, Sweden
- Department of Laboratory Medicine, Karolinska Institutet Huddinge, Stockholm, Sweden
| | - Sheena McCormack
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| |
Collapse
|
37
|
Prime-boost vaccine strategy against viral infections: Mechanisms and benefits. Vaccine 2016; 34:413-423. [DOI: 10.1016/j.vaccine.2015.11.062] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/01/2023]
|
38
|
Sheets RL, Zhou T, Knezevic I. Scientific and regulatory challenges in evaluating clinical trial protocols for HIV-1/AIDS vaccines - A review from a regulatory perspective. Biologicals 2015; 44:90-110. [PMID: 26732973 DOI: 10.1016/j.biologicals.2015.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 11/20/2015] [Accepted: 11/21/2015] [Indexed: 11/29/2022] Open
Abstract
Clinical development of prophylactic HIV/AIDS vaccines presents many scientific challenges that result in challenges for regulators reviewing clinical trial applications (CTAs). The World Health Organization (WHO) has the responsibility to provide technical support to these regulators. The search for an HIV/AIDS vaccine will only succeed through well-designed, -conducted and -controlled human efficacy studies reviewed and approved by regulators in countries worldwide, particularly in countries where the epidemic has hit hardest, such as in sub-Saharan Africa and Asia. This review summarizes the current candidates in development and focuses on challenges regulators face when reviewing CTAs, such as the evolving landscape of "standard of prevention," trials in adolescents, adaptive trial designs, correlates of protection and their analysis, and access to successful vaccines. There are many unknowns in the field of HIV/AIDS vaccine development and often, there is not a clear right or wrong approach because of the scientific challenges described in this review. Consequently, regulators should not feel that decisions need be made in isolation, when there are many available international collaborative efforts and opportunities to seek expert advice. The WHO provides many such opportunities and support to regulators across the globe.
Collapse
Affiliation(s)
| | - TieQun Zhou
- Technologies Standards and Norms Team, Regulation of Medicines and Other Health Technologies, Department of Essential Medicines and Health Products, Health Systems and Innovation, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Ivana Knezevic
- Technologies Standards and Norms Team, Regulation of Medicines and Other Health Technologies, Department of Essential Medicines and Health Products, Health Systems and Innovation, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| |
Collapse
|
39
|
Chamcha V, Jones A, Quigley BR, Scott JR, Amara RR. Oral Immunization with a Recombinant Lactococcus lactis-Expressing HIV-1 Antigen on Group A Streptococcus Pilus Induces Strong Mucosal Immunity in the Gut. THE JOURNAL OF IMMUNOLOGY 2015; 195:5025-34. [PMID: 26482408 DOI: 10.4049/jimmunol.1501243] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/16/2015] [Indexed: 01/25/2023]
Abstract
The induction of a potent humoral and cellular immune response in mucosal tissue is important for the development of an effective HIV vaccine. Most of the current HIV vaccines under development use the i.m. route for immunization, which is relatively poor in generating potent and long-lived mucosal immune responses. In this article, we explore the ability of an oral vaccination with a probiotic organism, Lactococcus lactis, to elicit HIV-specific immune responses in the mucosal and systemic compartments of BALB/c mice. We expressed the HIV-1 Gag-p24 on the tip of the T3 pilus of Streptococcus pyogenes as a fusion to the Cpa protein (LL-Gag). After four monthly LL-Gag oral immunizations, we observed strong Gag-specific IgG and IgA responses in serum, feces, and vaginal secretions. However, the Gag-specific CD8 T cell responses in the blood were at or below our detection limit. After an i.m. modified vaccinia Ankara/Gag boost, we observed robust Gag-specific CD8 T cell responses both in systemic and in mucosal tissues, including intraepithelial and lamina propria lymphocytes of the small intestine, Peyer's patches, and mesenteric lymph nodes. Consistent with strong immunogenicity, the LL-Gag induced activation of CD11c(+) CD11b(+) dendritic cells in the Peyer's patches after oral immunization. Our results demonstrate that oral immunization with L. lactis expressing an Ag on the tip of the group A Streptococcus pilus serves as an excellent vaccine platform to induce strong mucosal humoral and cellular immunity against HIV.
Collapse
Affiliation(s)
- Venkateswarlu Chamcha
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Andrew Jones
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and
| | - Bernard R Quigley
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30329
| | - June R Scott
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30329
| | - Rama Rao Amara
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329; and Department of Microbiology and Immunology, Emory University, Atlanta, GA 30329
| |
Collapse
|