1
|
Borthwick N, Fernandez N, Hayes PJ, Wee EGT, Akis Yildirim BM, Baines A, Baker M, Byard N, Conway O, Glaze M, Jenkin D, Larkworthy C, Luciw M, Platt A, Poulton I, Thomas M, Quaddy J, Watson M, Crook A, Cicconi P, Hanke T. Safety and immunogenicity of the ChAdOx1-MVA-vectored conserved mosaic HIVconsvX candidate T-cell vaccines in HIV-CORE 005.2, an open-label, dose-escalation, first-in-human, phase 1 trial in adults living without HIV-1 in the UK. THE LANCET. MICROBE 2025; 6:100956. [PMID: 39612921 DOI: 10.1016/j.lanmic.2024.100956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 05/29/2024] [Accepted: 07/18/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND An HIV-1 vaccine is long overdue. Although vaccine research focuses on the induction of broadly neutralising antibodies, challenging infections such as HIV-1 could require parallel induction of protective T cells. It is important to recognise that not all T cells contribute to protection equally. Previously, we developed a T-cell immunogen-based bivalent mosaic vaccine, HIVconsvX, delivered by vaccine vectors ChAdOx1 and modified vaccinia Ankara. In this study, we tested the HIVconsvX vaccine regimen for the first time in humans. Other ongoing trials will assess the contribution of the vaccine-induced killer T cells to the control of HIV-1. METHODS HIV-CORE 005.2 was an open-label, dose-escalation, first-in-human, phase 1 trial done at the Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Oxford, UK. Eligible participants were healthy volunteers aged 18-65 years living without HIV-1 and at a low likelihood of acquiring it. Because it was the first administration of ChAdOx1.tHIVconsv1 (C1) to humans, participants were assigned stepwise to two groups. Volunteer group 1 received a low dose of C1 on enrolment. Following a satisfactory safety review 7 days after vaccination, volunteer group 2 received a full dose of C1 boosted by vaccines MVA.tHIVconsv3 (M3) and MVA.tHIVconsv4 (M4) 4 weeks later in regimen C1-M3M4 and were followed up until day 140. Focusing on the full vaccine doses in group 2, the primary outcome was the local and systemic safety of the vaccine. The secondary outcome was the frequency and breadth of epitope recognition by vaccine-induced T cells determined by IFN-γ ELISPOT assay using peripheral blood mononuclear cells (PBMC) at peak (1 and 2 weeks after the M3M4 boost) and at the end of the study, assessed against volunteer's pre-vaccination levels. The HIV-CORE 005.2 trial is registered at ClinicalTrials.gov (NCT04586673) and is closed. FINDINGS Between July 3, 2021, and Aug 3, 2022, 13 participants were recruited and assigned to group 1 (n=3) and group 2 (n=10). Low-dose C1 was safe and well tolerated in group 1, and all three vaccine components were well tolerated in volunteer group 2. There were no serious adverse events. Local and systemic reactogenicities were consistent with intramuscular needle administration of immunogenic substances. All volunteers responded, and their vaccine-elicited T-cell frequencies peaked at a median of 4433 (IQR 2750-5820) IFN-γ spot-forming units per 106 PBMC and recognised a median of 9 (IQR 9-10) peptide pools out of 10, indicating that the responses were broadly specific and each vaccine recipient targeted at least nine epitopes on HIV-1. These frequencies were 7·4 times lower by day 140 (ie, 3 months later). T cells proliferated upon antigen re-exposure and displayed multiple effector functions, recognised variant epitopes, and inhibited HIV-1 from the four major global clades A, B, C, and D. INTERPRETATION These results inform and support a programme of clinical evaluations of the HIVconsvX T-cell vaccines together with other cutting-edge tools for HIV-1 cure and prevention such as latency reactivating agents, passively infused combinations of broadly neutralising antibodies, and active Env-based vaccines or immunomodulators. FUNDING EU Horizon 2020 Research and Innovation programme, Medical Research Council and Foreign Commonwealth and Development Office Concordat agreement, European and Developing Countries Clinical Trials Partnership, National Institute for Health Research Oxford Biomedical Research Centre, and IAVI.
Collapse
Affiliation(s)
- Nicola Borthwick
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | | | - Peter J Hayes
- IAVI Human Immunology Laboratory, Imperial College, London, UK
| | - Edmund G-T Wee
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | | | - Andrea Baines
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Megan Baker
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Nicholas Byard
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Oliver Conway
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Molly Glaze
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Daniel Jenkin
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Colin Larkworthy
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Michael Luciw
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Abigail Platt
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Ian Poulton
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Merin Thomas
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Jack Quaddy
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Marion Watson
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Alison Crook
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Paola Cicconi
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, Oxford University, Oxford, UK; Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
2
|
Link EK, Tscherne A, Sutter G, Smith ER, Gurwith M, Chen RT, Volz A. A Brighton collaboration standardized template with key considerations for a benefit/risk assessment for a viral vector vaccine based on a non-replicating modified vaccinia virus Ankara viral vector. Vaccine 2025; 43:126521. [PMID: 39612556 DOI: 10.1016/j.vaccine.2024.126521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) was formed to evaluate the safety and other key features of new platform technology vaccines. This manuscript provides an overview of Modified Vaccinia virus Ankara (MVA)-vectored vaccines and reviews molecular and biological key features of this platform. In particular, this review aims to provide fundamental information about the promising candidate vaccine MVA-MERS-S which has been evaluated successfully in different preclinical animal models and has undergone clinical testing including a phase Ib study involving more than 170 participants. Infectious diseases continue to be a major cause of human death worldwide. In this context, emerging zoonotic infectious diseases pose a particular challenge for public health systems. In the last two decades, three different respiratory coronaviruses, including the Middle East respiratory syndrome Coronavirus (MERS-CoV) have emerged. For many years, safe and efficacious vaccines have been a major tool to combat infectious diseases. Here, we report on a promising candidate vaccine (MVA-MERS-S) against MERS-CoV based on MVA. Upon application, MVA-MERS-S has been well tolerated and immunogenic, inducing both, cellular and humoral immune responses in different animal models and humans. We demonstrate that the MVA vector platform, with the example of MVA-MERS-S, is a viable and effective tool for producing safe, immunogenic, and efficient vaccines against emerging infectious diseases.
Collapse
Affiliation(s)
- Ellen K Link
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Alina Tscherne
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Gerd Sutter
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Emily R Smith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA.
| | - Marc Gurwith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T Chen
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Asisa Volz
- University of Veterinary Medicine Hannover, Institute of Virology, Hannover, Germany
| |
Collapse
|
3
|
Cua S, Tello BA, Farelo MA, Rodriguez E, Escalante GM, Mutsvunguma LZ, Ogembo JG, Reidel IG. A Novel Monoclonal Antibody Against a Modified Vaccinia Ankara (MVA) Envelope Protein as a Tool for MVA Virus Titration by Flow Cytometry. Viruses 2024; 16:1628. [PMID: 39459960 PMCID: PMC11512277 DOI: 10.3390/v16101628] [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: 09/12/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Modified vaccinia Ankara (MVA) virus is a widely used vaccine platform, making accurate titration essential for vaccination studies. However, the current plaque forming unit (PFU) assay, the standard for MVA titration, is prone to observer bias and other limitations that affect accuracy and precision. To address these challenges, we developed a new flow cytometry-based quantification method using a highly specific monoclonal antibody (mAb) for the detection of MVA-infected cells, as a more accurate titration assay. Through previous work, we serendipitously identified three MVA-specific hybridoma antibody clones, which we characterized through ELISA, immunoblot, and flow cytometry, confirming their specificity for MVA. Sequencing confirmed that each antibody was monoclonal, and mass spectrometry results revealed that all mAbs target the MVA cell surface binding protein (CSBP, MVA105L). We next optimized the titration protocol using the most effective mAb, 33C7 by refining culture conditions and staining protocols to enhance sensitivity and minimize background. Our optimized method demonstrated superior sensitivity, reliability, and reduced processing time when compared with the traditional PFU assay, establishing it as a more accurate and efficient approach for MVA titration.
Collapse
Affiliation(s)
- Simeon Cua
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Brenda A Tello
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Mafalda A Farelo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Esther Rodriguez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA
| | - Gabriela M Escalante
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lorraine Z Mutsvunguma
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Ivana G Reidel
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
4
|
Ndiaye A, Suneson K, Njuguna I, Ambler G, Hanke T, John-Stewart G, Jaoko W, Reilly M. Growth patterns and their contributing factors among HIV-exposed uninfected infants. MATERNAL AND CHILD NUTRITION 2020; 17:e13110. [PMID: 33269548 PMCID: PMC7988866 DOI: 10.1111/mcn.13110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 06/11/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023]
Abstract
With expanded HIV treatment and prevention programmes, most infants born to HIV‐positive women are uninfected, but the patterns and determinants of their growth are not well described. This study aimed to assess growth patterns in a cohort of HIV‐exposed uninfected (HEU) infants who participated in an experimental HIV vaccine trial and to test for associations with maternal and infant factors, including in‐utero exposure to antiretroviral therapy (ART), mode of delivery, exclusive breastfeeding, mother's education and receipt of the vaccine. Infants in the trial were seen at regular clinic visits from birth to 48 weeks of age. From the anthropometric measurements at these visits, weight‐for‐age z‐scores (WAZ), weight‐for‐length z‐scores (WLZ) and length‐for‐age z‐scores (LAZ) were computed using World Health Organization (WHO) software and reference tables. Growth patterns were investigated with respect to maternal and infant factors, using linear mixed regression models. From 94 infants included at birth, growth data were available for 75.5% at 48 weeks. The determinants of infant growth in this population are multifactorial: infant LAZ during the first year was significantly lower among infants delivered by caesarean section (p = 0.043); both WAZ and LAZ were depressed among infants with longer exposure to maternal ART (WAZ: p = 0.015; LAZ: p < 0.0001) and among infants of mothers with lower educational level (WAZ: p = 0.038; LAZ: p < 0.0001); the effect of maternal education was modified by breastfeeding practice, with no differences seen in exclusively breastfed infants. These findings inform intervention strategies to preserve growth in this vulnerable infant population.
Collapse
Affiliation(s)
- Aminata Ndiaye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Klara Suneson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.,Faculty of Medicine, Department of Clinical Sciences, Psychiatry, Lund University, Lund, Sweden
| | - Irene Njuguna
- Research and Programs, Kenyatta National Hospital, Nairobi, Kenya.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Gwen Ambler
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Tomas Hanke
- Jenner Institute, University of Oxford, Oxford, UK
| | - Grace John-Stewart
- Department of Epidemiology, University of Washington, Seattle, Washington, USA.,Departments of Global Health, Pediatrics, and Medicine, University of Washington, Seattle, Washington, USA
| | - Walter Jaoko
- KAVI-Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Marie Reilly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
5
|
Harnessing early life immunity to develop a pediatric HIV vaccine that can protect through adolescence. PLoS Pathog 2020; 16:e1008983. [PMID: 33180867 PMCID: PMC7660516 DOI: 10.1371/journal.ppat.1008983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
6
|
Chea LS, Wyatt LS, Gangadhara S, Moss B, Amara RR. Novel Modified Vaccinia Virus Ankara Vector Expressing Anti-apoptotic Gene B13R Delays Apoptosis and Enhances Humoral Responses. J Virol 2019; 93:e01648-18. [PMID: 30541829 PMCID: PMC6384055 DOI: 10.1128/jvi.01648-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/17/2018] [Indexed: 01/18/2023] Open
Abstract
Modified vaccinia virus Ankara (MVA), an attenuated poxvirus, has been developed as a potential vaccine vector for use against cancer and multiple infectious diseases, including human immunodeficiency virus (HIV). MVA is highly immunogenic and elicits strong cellular and humoral responses in preclinical models and humans. However, there is potential to further enhance the immunogenicity of MVA, as MVA-infected cells undergo rapid apoptosis, leading to faster clearance of recombinant antigens and potentially blunting a greater response. Here, we generated MVA-B13R by replacing the fragmented 181R/182R genes of MVA with a functional anti-apoptotic gene, B13R, and confirmed its anti-apoptotic function against chemically induced apoptosis in vitro In addition, MVA-B13R showed a significant delay in induction of apoptosis in muscle cells derived from mice and humans, as well as in plasmacytoid dendritic cells (pDCs) and CD141+ DCs from rhesus macaques, compared to the induction of apoptosis in MVA-infected cells. MVA-B13R expressing simian immunodeficiency virus (SIV) Gag and Pol and HIV envelope (SHIV) (MVA-B13R/SHIV) produced higher levels of envelope in the supernatants than MVA/SHIV-infected DF-1 cells in vitro Immunization of BALB/c mice showed induction of higher levels of envelope-specific antibody-secreting cells and memory B cells, higher IgG antibody titers, and better persistence of antibody titers with MVA-B13R/SHIV than with MVA/SHIV. Gene set enrichment analysis of draining lymph node cells from day 1 after immunization showed negative enrichment for interferon responses in MVA-B13R/SHIV-immunized mice compared to the responses in MVA/SHIV-immunized mice. Taken together, these results demonstrate that restoring B13R functionality in MVA significantly delays MVA-induced apoptosis in muscle and antigen-presenting cells in vitro and augments vaccine-induced humoral immunity in mice.IMPORTANCE MVA is an attractive viral vector for vaccine development due to its safety and immunogenicity in multiple species and humans even under conditions of immunodeficiency. Here, to further improve the immunogenicity of MVA, we developed a novel vector, MVA-B13R, by replacing the fragmented anti-apoptotic genes 181R/182R with a functional version derived from vaccinia virus, B13R Our results show that MVA-B13R significantly delays apoptosis in antigen-presenting cells and muscle cells in vitro and augments vaccine-induced humoral immunity in mice, leading to the development of a novel vector for vaccine development against infectious diseases and cancer.
Collapse
Affiliation(s)
- Lynette S Chea
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Linda S Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sailaja Gangadhara
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Itell HL, McGuire EP, Muresan P, Cunningham CK, McFarland EJ, Borkowsky W, Permar SR, Fouda GG. Development and application of a multiplex assay for the simultaneous measurement of antibody responses elicited by common childhood vaccines. Vaccine 2018; 36:5600-5608. [PMID: 30087048 PMCID: PMC6130906 DOI: 10.1016/j.vaccine.2018.07.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 11/18/2022]
Abstract
Because vaccine co-administration can affect elicited immune responses, it is important to evaluate new vaccines in the context of pre-existing vaccination schedules. This is particularly necessary for new pediatric vaccines, as the World Health Organization's infant immunization program already schedules several vaccines to be administered during the first months of life. To facilitate the assessment of inter-vaccine interference, we developed a pediatric vaccine multiplex assay (PVMA) to simultaneously measure antibodies against vaccines commonly administered to infants, including hepatitis B, Haemophilus influenzae type B, diphtheria, tetanus, pertussis, rubella, and respiratory syncytial virus (RSV). Comparison of antibody concentrations determined by enzyme-linked immunosorbent assays (ELISAs) and the PVMA demonstrated that the PVMA is highly sensitive, specific, reproducible, and accurate. Moreover, the PVMA requires half the time to assess a cohort compared to ELISAs, and only costs marginally more. Demonstrating the utility of the assay, we employed the PVMA to assess vaccine interference in the setting of a candidate vaccine, using the infant HIV vaccines from the completed Pediatric AIDS Clinical Trials Group (PACTG) protocols 230 and 326 as examples. There was no substantial difference in antibody concentrations between vaccine and placebo recipients, which suggests that HIV vaccination did not disrupt antibody responses elicited by routine pediatric vaccines. Thus, the PVMA is a reliable, high-throughput technique that requires minimal sample volume to measure multiple antibody concentrations concurrently, and is an efficient alternative to ELISAs for the measurement of vaccine-elicited antibody responses in large cohorts.
Collapse
Affiliation(s)
| | | | - Petronella Muresan
- Center for Biostatistics in AIDS Research, T.H. Chan Harvard School of Public Health, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
8
|
Nemes E, Hesseling AC, Tameris M, Mauff K, Downing K, Mulenga H, Rose P, van der Zalm M, Mbaba S, Van As D, Hanekom WA, Walzl G, Scriba TJ, McShane H, Hatherill M. Safety and Immunogenicity of Newborn MVA85A Vaccination and Selective, Delayed Bacille Calmette-Guerin for Infants of Human Immunodeficiency Virus-Infected Mothers: A Phase 2 Randomized, Controlled Trial. Clin Infect Dis 2018; 66:554-563. [PMID: 29028973 PMCID: PMC5849090 DOI: 10.1093/cid/cix834] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/23/2017] [Indexed: 12/20/2022] Open
Abstract
Background Vaccination of human immunodeficiency virus (HIV)-infected infants with bacille Calmette-Guérin (BCG) is contraindicated. HIV-exposed newborns need a new tuberculosis vaccination strategy that protects against tuberculosis early in life and avoids the potential risk of BCG disease until after HIV infection has been excluded. Methods This double-blind, randomized, controlled trial compared newborn MVA85A prime vaccination (1 × 108 PFU) vs Candin® control, followed by selective, deferred BCG vaccination at age 8 weeks for HIV-uninfected infants and 12 months follow-up for safety and immunogenicity. Results A total of 248 HIV-exposed infants were enrolled. More frequent mild-moderate reactogenicity events were seen after newborn MVA85A vaccination. However, no significant difference was observed in the rate of severe or serious adverse events, HIV acquisition (n = 1 per arm), or incident tuberculosis disease (n = 5 MVA85A; n = 3 control) compared to the control arm. MVA85A vaccination induced modest but significantly higher Ag85A-specific interferon gamma (IFNγ)+ CD4+ T cells compared to control at weeks 4 and 8 (P < .0001). BCG did not further boost this response in MVA85A vaccinees. The BCG-induced Ag85A-specific IFNγ+ CD4+ T-cell response at weeks 16 and 52 was of similar magnitude in the control arm compared to the MVA85A arm at all time points. Proliferative capacity, functional profiles, and memory phenotype of BCG-specific CD4 responses were similar across study arms. Conclusions MVA85A prime vaccination of HIV-exposed newborns was safe and induced an early modest antigen-specific immune response that did not interfere with, or enhance, immunogenicity of subsequent BCG vaccination. New protein-subunit and viral-vectored tuberculosis vaccine candidates should be tested in HIV-exposed newborns. Clinical Trials Registration NCT01650389.
Collapse
Affiliation(s)
- Elisa Nemes
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | - Anneke C Hesseling
- Desmond Tutu Tuberculosis Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences
| | - Michele Tameris
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | - Katya Mauff
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | - Katrina Downing
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | - Humphrey Mulenga
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | - Penelope Rose
- Desmond Tutu Tuberculosis Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences
| | - Marieke van der Zalm
- Desmond Tutu Tuberculosis Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences
| | - Sharon Mbaba
- Desmond Tutu Tuberculosis Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences
| | - Danelle Van As
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | - Gerhard Walzl
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research/Medical Research Council Centre for Molecular and Cellular Biology, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| | | | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Science & Technology/National Research Foundation, University of Cape Town
| |
Collapse
|
9
|
Milligan C, Slyker JA, Overbaugh J. The Role of Immune Responses in HIV Mother-to-Child Transmission. Adv Virus Res 2017; 100:19-40. [PMID: 29551137 DOI: 10.1016/bs.aivir.2017.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
HIV mother-to-child transmission (MTCT) represents a success story in the HIV/AIDS field given the significant reduction in number of transmission events with the scale-up of antiretroviral treatment and other prevention methods. Nevertheless, MTCT still occurs and better understanding of the basic biology and immunology of transmission will aid in future prevention and treatment efforts. MTCT is a unique setting given that the transmission pair is known and the infant receives passively transferred HIV-specific antibodies from the mother while in utero. Thus, infant exposure to HIV occurs in the face of HIV-specific antibodies, especially during delivery and breastfeeding. This review highlights the immune correlates of protection in HIV MTCT including humoral (neutralizing antibodies, antibody-dependent cellular cytotoxicity, and binding epitopes), cellular, and innate immune factors. We further discuss the future implications of this research as it pertains to opportunities for passive and active vaccination with the ultimate goal of eliminating HIV MTCT.
Collapse
Affiliation(s)
- Caitlin Milligan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, United States.
| | | | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
10
|
Muturi-Kioi V, Lewis D, Launay O, Leroux-Roels G, Anemona A, Loulergue P, Bodinham CL, Aerssens A, Groth N, Saul A, Podda A. Neutropenia as an Adverse Event following Vaccination: Results from Randomized Clinical Trials in Healthy Adults and Systematic Review. PLoS One 2016; 11:e0157385. [PMID: 27490698 PMCID: PMC4974007 DOI: 10.1371/journal.pone.0157385] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/26/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In the context of early vaccine trials aimed at evaluating the safety profile of novel vaccines, abnormal haematological values, such as neutropenia, are often reported. It is therefore important to evaluate how these trials should be planned not to miss potentially important safety signals, but also to understand the implications and the clinical relevance. METHODOLOGY We report and discuss the results from five clinical trials (two with a new Shigella vaccine in the early stage of clinical development and three with licensed vaccines) where the absolute neutrophil counts (ANC) were evaluated before and after vaccination. Additionally, we have performed a systematic review of the literature on cases of neutropenia reported during vaccine trials to discuss our results in a more general context. PRINCIPAL FINDINGS Both in our clinical trials and in the literature review, several cases of neutropenia have been reported, in the first two weeks after vaccination. However, neutropenia was generally transient and had a benign clinical outcome, after vaccination with either multiple novel candidates or well-known licensed vaccines. Additionally, the vaccine recipients with neutropenia frequently had lower baseline ANC than non-neutropenic vaccinees. In many instances neutropenia occurred in subjects of African descent, known to have lower ANC compared to western populations. CONCLUSIONS It is important to include ANC and other haematological tests in early vaccine trials to identify potential safety signals. Post-vaccination neutropenia is not uncommon, generally transient and clinically benign, but many vaccine trials do not have a sampling schedule that allows its detection. Given ethnic variability in the level of circulating neutrophils, normal ranges taking into account ethnicity should be used for determination of trial inclusion/exclusion criteria and classification of neutropenia related adverse events. TRIAL REGISTRATION ClinicalTrials.gov NCT02017899, NCT02034500, NCT01771367, NCT01765413, NCT02523287.
Collapse
Affiliation(s)
| | - David Lewis
- Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| | - Odile Launay
- Université Paris Descartes, Sorbonne Paris cité, and Inserm CIC 1417, F-CRIN I-Reivac, Assistance Publique Hôpitaux de Paris, CIC Cochin-Pasteur, Paris, France
| | | | | | - Pierre Loulergue
- Université Paris Descartes, Sorbonne Paris cité, and Inserm CIC 1417, F-CRIN I-Reivac, Assistance Publique Hôpitaux de Paris, CIC Cochin-Pasteur, Paris, France
| | - Caroline L. Bodinham
- Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| | | | | | - Allan Saul
- Novartis Vaccines Institute for Global Health, Siena, Italy
| | - Audino Podda
- Novartis Vaccines Institute for Global Health, Siena, Italy
| |
Collapse
|
11
|
|
12
|
Meseda CA, Atukorale V, Kuhn J, Schmeisser F, Weir JP. Percutaneous Vaccination as an Effective Method of Delivery of MVA and MVA-Vectored Vaccines. PLoS One 2016; 11:e0149364. [PMID: 26895072 PMCID: PMC4760941 DOI: 10.1371/journal.pone.0149364] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/29/2016] [Indexed: 12/22/2022] Open
Abstract
The robustness of immune responses to an antigen could be dictated by the route of vaccine inoculation. Traditional smallpox vaccines, essentially vaccinia virus strains, that were used in the eradication of smallpox were administered by percutaneous inoculation (skin scarification). The modified vaccinia virus Ankara is licensed as a smallpox vaccine in Europe and Canada and currently undergoing clinical development in the United States. MVA is also being investigated as a vector for the delivery of heterologous genes for prophylactic or therapeutic immunization. Since MVA is replication-deficient, MVA and MVA-vectored vaccines are often inoculated through the intramuscular, intradermal or subcutaneous routes. Vaccine inoculation via the intramuscular, intradermal or subcutaneous routes requires the use of injection needles, and an estimated 10 to 20% of the population of the United States has needle phobia. Following an observation in our laboratory that a replication-deficient recombinant vaccinia virus derived from the New York City Board of Health strain elicited protective immune responses in a mouse model upon inoculation by tail scarification, we investigated whether MVA and MVA recombinants can elicit protective responses following percutaneous administration in mouse models. Our data suggest that MVA administered by percutaneous inoculation, elicited vaccinia-specific antibody responses, and protected mice from lethal vaccinia virus challenge, at levels comparable to or better than subcutaneous or intramuscular inoculation. High titers of specific neutralizing antibodies were elicited in mice inoculated with a recombinant MVA expressing the herpes simplex type 2 glycoprotein D after scarification. Similarly, a recombinant MVA expressing the hemagglutinin of attenuated influenza virus rgA/Viet Nam/1203/2004 (H5N1) elicited protective immune responses when administered at low doses by scarification. Taken together, our data suggest that MVA and MVA-vectored vaccines inoculated by scarification can elicit protective immune responses that are comparable to subcutaneous vaccination, and may allow for antigen sparing when vaccine supply is limited.
Collapse
Affiliation(s)
- Clement A. Meseda
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food & Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, United States of America
| | - Vajini Atukorale
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food & Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, United States of America
| | - Jordan Kuhn
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food & Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, United States of America
| | - Falko Schmeisser
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food & Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, United States of America
| | - Jerry P. Weir
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food & Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, United States of America
| |
Collapse
|
13
|
Klein N, Palma P, Luzuriaga K, Pahwa S, Nastouli E, Gibb DM, Rojo P, Borkowsky W, Bernardi S, Zangari P, Calvez V, Compagnucci A, Wahren B, Foster C, Munoz-Fernández MÁ, De Rossi A, Ananworanich J, Pillay D, Giaquinto C, Rossi P. Early antiretroviral therapy in children perinatally infected with HIV: a unique opportunity to implement immunotherapeutic approaches to prolong viral remission. THE LANCET. INFECTIOUS DISEASES 2015; 15:1108-1114. [PMID: 26187030 DOI: 10.1016/s1473-3099(15)00052-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 05/04/2015] [Accepted: 05/08/2015] [Indexed: 02/02/2023]
Abstract
From the use of antiretroviral therapy to prevent mother-to-child transmission to the possibility of HIV cure hinted at by the Mississippi baby experience, paediatric HIV infection has been pivotal to our understanding of HIV pathogenesis and management. Daily medication and indefinite antiretroviral therapy is recommended for children infected with HIV. Maintenance of life-long adherence is difficult and the incidence of triple-class virological failure after initiation of antiretroviral therapy increases with time. This challenge shows the urgent need to define novel strategies to provide long-term viral suppression that will allow safe interruption of antiretroviral therapy without viral rebound and any associated complications. HIV-infected babies treated within a few days of birth have a unique combination of a very small pool of integrated viruses, a very high proportion of relatively HIV resistant naive T cells, and an unparalleled capacity to regenerate an immune repertoire. These features make this group the optimum model population to investigate the potential efficacy of immune-based therapies. If successful, these investigations could change the way we manage HIV infection.
Collapse
Affiliation(s)
- Nigel Klein
- Institute of Child Health, University College London, London, UK.
| | - Paolo Palma
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Katherine Luzuriaga
- Program in Molecular Medicine, University of Massachusetts Medical School Worcester, MA, USA
| | - Savita Pahwa
- Miami Center for AIDS Research Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Eleni Nastouli
- Department of Virology, University College London, London, UK
| | - Diane M Gibb
- Medical Research Council Clinical Trials Unit, London, UK
| | - Pablo Rojo
- Department of Pediatrics, Hospital 12 de Octubre, Madrid, Spain
| | | | - Stefania Bernardi
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Paola Zangari
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Vincent Calvez
- Pierre et Marie Curie University and Pitié-Salpêtrière Hospital, Paris, France
| | - Alexandra Compagnucci
- Institut National de la Santé et de la Recherche Médicale SC10-US019 Clinical Trials and Infectious Diseases, Villejuif, Paris, France
| | - Britta Wahren
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Foster
- Imperial College Healthcare National Health Service Trust, London, UK
| | | | - Anita De Rossi
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology-DiSCOG, University of Padova, and Istituto Oncologico Veneto, Padova, Italy
| | - Jintanat Ananworanich
- US Military HIV Research Program, Walter Reed Army Institute of Research and Henry M Jackson Foundation for the Advancement of Military Medicine, Silver Spring, MD, USA
| | - Deenan Pillay
- Africa Centre, Mtubatuba, KwaZulu Natal, South Africa
| | - Carlo Giaquinto
- Department of Women's and Children's Health, University of Padova, and Penta Foundation, Padova, Italy
| | - Paolo Rossi
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy.
| |
Collapse
|
14
|
Fouda GG, Cunningham CK, Permar SR. Infant HIV-1 vaccines: supplementing strategies to reduce maternal-child transmission. JAMA 2015; 313:1513-4. [PMID: 25898044 PMCID: PMC4520313 DOI: 10.1001/jama.2015.1382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Genevieve G Fouda
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina2Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| | - Coleen K Cunningham
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Sallie R Permar
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina2Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
15
|
Characterization of T-cell responses to conserved regions of the HIV-1 proteome in BALB/c mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:1565-72. [PMID: 25230940 PMCID: PMC4248756 DOI: 10.1128/cvi.00587-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A likely requirement for a protective vaccine against human immunodeficiency virus type 1 (HIV-1)/AIDS is, in addition to eliciting antibody responses, induction of effective T cells. To tackle HIV-1 diversity by T-cell vaccines, we designed an immunogen, HIVconsv, derived from the most functionally conserved regions of the HIV-1 proteome and demonstrated its high immunogenicity in humans and rhesus macaques when delivered by regimens combining plasmid DNA, nonreplicating simian (chimpanzee) adenovirus ChAdV-63, and nonreplicating modified vaccinia virus Ankara (MVA) as vectors. Here, we aimed to increase the decision power for iterative improvements of this vaccine strategy in the BALB/c mouse model. First, we found that prolonging the period after the ChAdV63.HIVconsv prime up to 6 weeks increased the frequencies of HIV-1-specific, gamma interferon (IFN-γ)-producing T cells induced by the MVA.HIVconsv boost. Induction of strong responses allowed us to map comprehensively the H-2d-restricted T-cell responses to these regions and identified 8 HIVconsv peptides, of which three did not contain a previously described epitope and were therefore considered novel. Induced effector T cells were oligofunctional and lysed sensitized targets in vitro. Our study therefore provides additional tools for studying and optimizing vaccine regimens in this commonly used small animal model, which will in turn guide vaccine improvements in more expensive nonhuman primate and human clinical trials.
Collapse
|