1
|
Akingbola A, Adegbesan A, Adewole O, Adegoke K, Benson AE, Jombo PA, Uchechukwu Eboson S, Oluwasola V, Aiyenuro A. The mRNA-1647 vaccine: A promising step toward the prevention of cytomegalovirus infection (CMV). Hum Vaccin Immunother 2025; 21:2450045. [PMID: 39825496 DOI: 10.1080/21645515.2025.2450045] [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: 10/07/2024] [Revised: 12/14/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Cytomegalovirus (CMV) is a leading cause of congenital infections and significant health complications in immunocompromised individuals. With no licensed CMV vaccine available, the development of the mRNA-1647 offers promising advancements in CMV prevention. We have reviewed results from Phase 1 and 2 clinical trials of the mRNA-1647 vaccine, demonstrating robust immune responses in both seronegative and seropositive participants. Vaccines exhibited significantly elevated neutralizing antibody titers against CMV, particularly in fibroblast and epithelial cells, with sustained responses lasting up to 18 months post-vaccination. The mRNA-1647 vaccine triggered strong T-cell and memory B-cell responses, suggesting its potential for long-term protection against CMV infection. The ongoing Phase 3 CMVictory trial evaluates the safety and immunogenicity of mRNA-1647 in women of childbearing age, with preliminary data showing promise in preventing congenital CMV transmission. This vaccine could significantly reduce CMV-related morbidity and mortality, particularly in newborns and immunocompromised individuals, addressing a critical unmet medical need.
Collapse
Affiliation(s)
| | - Abiodun Adegbesan
- African Cancer Institute, Department of Global Health, Stellenbosch University, Cape Town, South Africa
| | | | - Kolade Adegoke
- Faculty of Clinical Sciences, Obafemi Awolowo University Ile-Ife, Osun State,Nigeria
| | | | - Paul Ayomide Jombo
- Internal Medicine, Basildon and Thurrock University Hospitals NHS Foundation Trust: Basildon SS165NL, England, Essex, England, UK
| | | | - Victor Oluwasola
- Babcock University Teaching Hospital, Ilishan-Remo, Ogun State, Nigeria
| | - Ademola Aiyenuro
- Division of Virology, Department of Pathology, University of Cambridge, England, UK
| |
Collapse
|
2
|
Schultz EP, Ponsness L, Lanchy JM, Zehner M, Klein F, Ryckman BJ. Human cytomegalovirus gH/gL/gO binding to PDGFRα provides a regulatory signal activating the fusion protein gB that can be blocked by neutralizing antibodies. J Virol 2025; 99:e0003525. [PMID: 40202318 PMCID: PMC12090739 DOI: 10.1128/jvi.00035-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Herpesviruses require membrane fusion for entry and spread, a process facilitated by the fusion glycoprotein B (gB) and the regulatory factor gH/gL. The human cytomegalovirus (HCMV) gH/gL can be modified by the accessory protein gO, or the set of proteins UL128, UL130, and UL131. While the binding of the gH/gL/gO and gH/gL/UL128-131 complexes to cellular receptors, including PDGFRα and NRP2, has been well-characterized structurally, the specific role of receptor engagements by the gH/gL/gO and gH/gL/UL128-131 in regulation of fusion has remained unclear. We describe a cell-cell fusion assay that can quantitatively measure fusion on a timescale of minutes and demonstrate that binding of gH/gL/gO to PDGFRα dramatically enhances gB-mediated cell-cell fusion. In contrast, gH/gL/pUL128-131-regulated fusion is significantly slower, and gH/gL alone cannot promote gB fusion activity within this timescale. The genetic diversity of gO influenced the observed cell-cell fusion rates, correlating with previously reported effects on HCMV infectivity. Mutations in gL that had no effect on the formation of gH/gL/gO or binding to PDGFRa dramatically reduced the cell-cell fusion rate, suggesting that gL plays a critical role in linking the gH/gL/gO-PDGFRa receptor binding to activation of gB. Several neutralizing human monoclonal antibodies were found to potently block gH/gL/gO-PDGFRa-regulated cell-cell fusion, suggesting this mechanism as a therapeutic target. IMPORTANCE Development of vaccines and therapeutics targeting the fusion apparatus of human cytomegalovirus (HCMV) has been limited by the lack of an in vitro cell-cell fusion assay that faithfully models the receptor-dependent fusion characteristic of HCMV entry. The cell-cell fusion assay described here demonstrated that the binding of gH/gL/gO to its receptor, PDGFRα, serves to regulate the activity of the fusion protein gB, and this is specifically vulnerable to inhibition by neutralizing antibodies. Moreover, the measurement of fusion kinetics allows for mutational studies of the fusion mechanism, assessing the influence of genetic diversity among the viral glycoproteins and studying the mechanism of neutralizing antibodies.
Collapse
Affiliation(s)
- Eric P. Schultz
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana, USA
| | - Lars Ponsness
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
| | - Jean-Marc Lanchy
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana, USA
| | - Matthias Zehner
- Laboratory for Infection and Immune Biology, University of Cologne, Cologne, Germany
- Institute of Virology, University Cologne, Cologne, Germany
- Faculty of Medicine, University of Cologne, Cologne, Germany
- University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Klein
- Institute of Virology, University Cologne, Cologne, Germany
- Faculty of Medicine, University of Cologne, Cologne, Germany
- University Hospital Cologne, University of Cologne, Cologne, Germany
- Laboratory of Experimental Immunology, University of Cologne, Cologne, Germany
| | - Brent J. Ryckman
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana, USA
| |
Collapse
|
3
|
Lankina A, Raposo M, Hargreaves A, Atkinson C, Griffiths P, Reeves MB. Developing a Vaccine Against Human Cytomegalovirus: Identifying and Targeting HCMV's Immunological Achilles' Heel. Vaccines (Basel) 2025; 13:435. [PMID: 40432047 DOI: 10.3390/vaccines13050435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Human cytomegalovirus (HCMV) is a critical pathogen in immunocompromised populations, such as organ transplant recipients as well as congenitally infected neonates with immature immune systems. Despite decades of research and the growing financial burden associated with the management of HCMV, there is no licensed vaccine to date. In this review, we aim to outline the complexity of HCMV and the antigens it presents and the journey and challenges of developing an effective HCMV vaccine, as well as further highlight the recent analyses of the most successful vaccine candidate so far-gB/MF59.
Collapse
Affiliation(s)
- Anastasia Lankina
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Marta Raposo
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Alexander Hargreaves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Claire Atkinson
- School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Paul Griffiths
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Matthew B Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK
| |
Collapse
|
4
|
Wang HY, Taher H, Kreklywich CN, Schmidt KA, Scheef EA, Barfield R, Otero CE, Valencia SM, Zhang K, Callahan C, Monticolo F, Qiao Y, Gilbride RM, Crooks CM, Mirza A, Knight K, Moström MJ, Manuel TD, Sprehe L, Kendall S, Burgt NV, Kowalik TF, Barry PA, Hansen SG, Shu J, Tarantal AF, Chan C, Streblow DN, Picker LJ, Kaur A, Früh K, Permar SR, Malouli D. The pentameric complex is not required for congenital CMV transmission in seronegative rhesus macaques. Sci Transl Med 2025; 17:eadm8961. [PMID: 40073152 DOI: 10.1126/scitranslmed.adm8961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 08/29/2024] [Accepted: 01/22/2025] [Indexed: 03/14/2025]
Abstract
Congenital cytomegalovirus (cCMV) is the leading infectious cause of neonatal neurological impairment worldwide, but the viral factors enabling vertical spread across the placenta remain undetermined. The pentameric complex (PC), composed of the subunits gH/gL/UL128/UL130/UL131A, has been demonstrated to be important for entry into nonfibroblast cells in vitro. These findings link the PC to broad cell tropism and virus dissemination in vivo, denoting all subunits as potential targets for intervention strategies and vaccine development. To determine the relevance of the PC for congenital transmission in a translational nonhuman primate model, we engineered a rhesus CMV (RhCMV) mutant lacking the orthologs of UL128 and UL130, which demonstrated diminished infection of epithelial cells in vitro. However, intravenous inoculation of either CD4+ T cell-depleted or immunocompetent RhCMV-seronegative pregnant rhesus macaques (RMs) in the early second trimester with the PC-deficient mutant resulted in maternal RhCMV peak plasma viremia similar to inoculations with PC-intact RhCMV, although virus shedding in saliva and urine was limited. Infections with the PC-intact virus induced IgG responses that neutralized RhCMV entry into epithelial cells in tissue culture. These responses were reduced, but not absent, from animals infected with the PC-deficient virus, which also induced IgG responses against gH. Moreover, congenital CMV transmission was confirmed in multiple animals infected with PC-deficient virus by detecting viral DNA in the amniotic fluid, indicating that transplacental transmission in RMs is not contingent on the PC.
Collapse
Affiliation(s)
- Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Duke University Medical Center, Durham, NC 27710, USA
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Craig N Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Kimberli A Schmidt
- Department of Pathology and Laboratory Medicine, Center for Immunology and Infectious Diseases, California National Primate Research Center, University of California Davis, Davis, CA 95616, USA
| | - Elizabeth A Scheef
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics and Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Claire E Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Duke University Medical Center, Durham, NC 27710, USA
| | | | - Ke Zhang
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Boston, MA 02142, USA
| | - Claire Callahan
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Francesco Monticolo
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yueqing Qiao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Chelsea M Crooks
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anne Mirza
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Kelsey Knight
- Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Matilda J Moström
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Tabitha D Manuel
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Lesli Sprehe
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Savannah Kendall
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Nathan Vande Burgt
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Timothy F Kowalik
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Peter A Barry
- Department of Pathology and Laboratory Medicine, Center for Immunology and Infectious Diseases, California National Primate Research Center, University of California Davis, Davis, CA 95616, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jian Shu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Boston, MA 02142, USA
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA 02139, USA
| | - Alice F Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California Davis, Davis, CA 95616, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics and Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| |
Collapse
|
5
|
Schultz EP, Ponsness L, Lanchy JM, Zehner M, Klein F, Ryckman BJ. Human cytomegalovirus gH/gL/gO binding to PDGFRα provides a regulatory signal activating the fusion protein gB that can be blocked by neutralizing antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631902. [PMID: 39829861 PMCID: PMC11741351 DOI: 10.1101/2025.01.08.631902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Herpesviruses require membrane fusion for entry and spread, a process facilitated by the fusion glycoprotein B (gB) and the regulatory factor gH/gL. The human cytomegalovirus (HCMV) gH/gL can be modified by the accessory protein gO, or the set of proteins UL128, UL130 and UL131. While the binding of the gH/gL/gO and gH/gL/UL128-131 complexes to cellular receptors including PDFGRα and NRP2 has been well-characterized structurally, the specific role of receptor engagements by the gH/gL/gO and gH/gL/UL128-131 in regulation of fusion has remained unclear. We describe a cell-cell fusion assay that can quantitatively measure fusion on a timescale of minutes and demonstrate that binding of gH/gL/gO to PDGFRα dramatically enhances gB-mediated cell-cell fusion. In contrast, gH/gL/pUL128-131-regulated fusion is significantly slower and gH/gL alone cannot promote gB fusion activity within this timescale. The genetic diversity of gO influenced the observed cell-cell fusion rates, correlating with previously reported effects on HCMV infectivity. Mutations in gL that had no effect on formation of gH/gL/gO or binding to PDGFRα dramatically reduced the cell-cell fusion rate, suggesting that gL plays a critical role in linking the gH/gL/gO-PDGFRα receptor-binding to activation of gB. Several neutralizing human monoclonal antibodies were found to potently block gH/gL/gO-PDGFRα regulated cell-cell fusion, suggesting this mechanism as a therapeutic target.
Collapse
Affiliation(s)
- Eric P. Schultz
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Lars Ponsness
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jean-Marc Lanchy
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Matthias Zehner
- Laboratory for Infection and Immune Biology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brent J. Ryckman
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
6
|
Fierro C, Brune D, Shaw M, Schwartz H, Knightly C, Lin J, Carfi A, Natenshon A, Kalidindi S, Reuter C, Miller J, Panther L. Safety and Immunogenicity of a Messenger RNA-Based Cytomegalovirus Vaccine in Healthy Adults: Results From a Phase 1 Randomized Clinical Trial. J Infect Dis 2024; 230:e668-e678. [PMID: 38478705 PMCID: PMC11420795 DOI: 10.1093/infdis/jiae114] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/11/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND This phase 1 trial evaluated the safety, reactogenicity, and immunogenicity of mRNA-1647, a messenger RNA (mRNA)-based cytomegalovirus (CMV) vaccine, in CMV-seronegative and -seropositive adults. METHODS Participants were randomly assigned to receive 30, 90, 180, or 300 µg of mRNA-1647 or placebo on a 0-, 2-, and 6-month schedule and followed for 12 months after the last dose. RESULTS A total of 154 (80 CMV-seronegative and 74 CMV-seropositive) participants were enrolled; 118 participants were randomized to mRNA-1647 and 36 to placebo. Mean (standard deviation) age was 32.5 (8.6) and 35.1 (8.9) years in the placebo and mRNA-1647 groups, respectively, in phase B (63% and 64% female) and 42.5 (6.2) and 33.3 (8.7) years, respectively, in phase C (2% and 16% female). No deaths, related serious adverse events, or adverse events of special interest were reported. Most adverse reactions were grade ≤2 severity. Increased neutralizing antibody, binding antibody, and antigen-specific cell-mediated responses were observed across mRNA-1647 treatment groups, regardless of CMV serostatus. CONCLUSIONS This phase 1, first-in-human trial demonstrated that mRNA-1647 has an acceptable safety profile in adults and elicits humoral and cellular immune responses. Clinical Trials Registration. NCT03382405.
Collapse
Affiliation(s)
- Carlos Fierro
- Johnson County Clin-Trials, Department of Clinical Safety & Risk Management, Lenexa, Kansas
| | | | | | | | - Conor Knightly
- Moderna, Inc, Department of Clinical Development Operations, Cambridge, Massachusetts
| | - Jiang Lin
- Moderna, Inc, Department of Biostatistics, Cambridge, Massachusetts
| | - Andrea Carfi
- Moderna, Inc, Department of Research and Development, Cambridge, Massachusetts
| | - Andrew Natenshon
- Moderna, Inc, Department of Infectious Disease Development, Cambridge, Massachusetts
| | - Shiva Kalidindi
- Moderna, Inc, Department of Statistical Programming, Cambridge, Massachusetts
| | - Caroline Reuter
- Johnson County Clin-Trials, Department of Clinical Safety & Risk Management, Lenexa, Kansas
| | - Jacqueline Miller
- Moderna, Inc, Department of Infectious Diseases, Cambridge, Massachusetts
| | - Lori Panther
- Moderna, Inc, Department of Infectious Diseases, Cambridge, Massachusetts
| |
Collapse
|
7
|
Zavaglio F, d'Angelo P, Fornara C, Zelini P, Comolli G, Furione M, Arossa A, Spinillo A, Lilleri D, Baldanti F. Different Antigen-Specific CD4 + and CD8 + T-Cell Response against HCMV Proteins in Pregnant Women with Primary Infection and in Control Subjects with Remote Infection. J Clin Med 2024; 13:5448. [PMID: 39336935 PMCID: PMC11432261 DOI: 10.3390/jcm13185448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Human cytomegalovirus (HCMV) is the most frequent cause of congenital infections. The HCMV-specific T-cell response in primary infection may help define reliable correlates of immune protection in pregnancy. In this study, the antigen-specific T-cell response against different HCMV proteins (IE-1, pp65, gB, gHgLpUL128L) was investigated in pregnant women with primary infection and in control subjects with remote infection to identify possible components of a vaccine. Methods: Blood samples from 35 pregnant women with HCMV primary infection and 30 HCMV-seropositive healthy adult subjects with remote infection were tested. The antigen-specific T-cell response was measured using cytokine intracellular staining after stimulation with IE-1, pp65, gB and gHgLpUL128L peptides pool. Results: The pp65-specific CD4+ T-cell response was higher in pregnant women with HCMV primary infection at the late time point and in control subjects with remote infection, while the pregnant women at the early time point showed a higher gB-specific CD8+ T-cell response. Regarding the CD4+ and CD8+ T-cell phenotypes, we observed that HCMV-specific CD4+ and CD8+ T cells expressing CD45RA+ remained constant in pregnant women with primary infection at the early and late time points and in subjects with remote infection, while HCMV-specific CD4+ and CD8+ T cells expressing IL-7R+ or producing IL-2 were higher in control subjects with remote infection than in pregnant women with HCMV primary infection. Conclusions: The T-cell response was higher against gB in the early phase of infection and against pp65 in the late phase. Therefore, these proteins should be taken into consideration as candidates for a vaccine.
Collapse
Affiliation(s)
- Federica Zavaglio
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Piera d'Angelo
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chiara Fornara
- General Clinical Laboratory, with Specialist Areas of Clinical Pathology, Microbiology and Virology, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Paola Zelini
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giuditta Comolli
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Milena Furione
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Alessia Arossa
- Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Arsenio Spinillo
- Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Daniele Lilleri
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Fausto Baldanti
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
8
|
Hu X, Karthigeyan KP, Herbek S, Valencia SM, Jenks JA, Webster H, Miller IG, Connors M, Pollara J, Andy C, Gerber LM, Walter EB, Edwards KM, Bernstein DI, Hou J, Koch M, Panther L, Carfi A, Wu K, Permar SR. Human Cytomegalovirus mRNA-1647 Vaccine Candidate Elicits Potent and Broad Neutralization and Higher Antibody-Dependent Cellular Cytotoxicity Responses Than the gB/MF59 Vaccine. J Infect Dis 2024; 230:455-466. [PMID: 38324766 PMCID: PMC11326847 DOI: 10.1093/infdis/jiad593] [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: 08/17/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND MF59-adjuvanted gB subunit (gB/MF59) vaccine demonstrated approximately 50% efficacy against human cytomegalovirus (HCMV) acquisition in multiple clinical trials, suggesting that efforts to improve this vaccine design might yield a vaccine suitable for licensure. METHODS A messenger RNA (mRNA)-based vaccine candidate encoding HCMV gB and pentameric complex (PC), mRNA-1647, is currently in late-stage efficacy trials. However, its immunogenicity has not been compared to the partially effective gB/MF59 vaccine. We assessed neutralizing and Fc-mediated immunoglobulin G (IgG) effector antibody responses induced by mRNA-1647 in both HCMV-seropositive and -seronegative vaccinees from a first-in-human clinical trial through 1 year following third vaccination using a systems serology approach. Furthermore, we compared peak anti-gB antibody responses in seronegative mRNA-1647 vaccinees to that of seronegative gB/MF59 vaccine recipients. RESULTS mRNA-1647 vaccination elicited and boosted HCMV-specific IgG responses in seronegative and seropositive vaccinees, respectively, including neutralizing and Fc-mediated effector antibody responses. gB-specific IgG responses were lower than PC-specific IgG responses. gB-specific IgG and antibody-dependent cellular phagocytosis responses were lower than those elicited by gB/MF59. However, mRNA-1647 elicited higher neutralization and antibody-dependent cellular cytotoxicity (ADCC) responses. CONCLUSIONS Overall, mRNA-1647 vaccination induced polyfunctional and durable HCMV-specific antibody responses, with lower gB-specific IgG responses but higher neutralization and ADCC responses compared to the gB/MF59 vaccine. CLINICAL TRIALS REGISTRATION NCT03382405 (mRNA-1647) and NCT00133497 (gB/MF59).
Collapse
Affiliation(s)
- Xintao Hu
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | | | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Sarah M Valencia
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| | - Jennifer A Jenks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| | - Helen Webster
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| | - Itzayana G Miller
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Megan Connors
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York
| | - Linda M Gerber
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina
| | - Kathryn M Edwards
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David I Bernstein
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Jacob Hou
- Moderna, Inc, Cambridge, Massachusetts
| | | | | | | | - Kai Wu
- Moderna, Inc, Cambridge, Massachusetts
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| |
Collapse
|
9
|
Wu K, Hou YJ, Makrinos D, Liu R, Zhu A, Koch M, Yu WH, Paila YD, Chandramouli S, Panther L, Henry C, DiPiazza A, Carfi A. Characterization of humoral and cellular immunologic responses to an mRNA-based human cytomegalovirus vaccine from a phase 1 trial of healthy adults. J Virol 2024; 98:e0160323. [PMID: 38526054 PMCID: PMC11019844 DOI: 10.1128/jvi.01603-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/25/2024] [Indexed: 03/26/2024] Open
Abstract
mRNA-1647 is an investigational mRNA-based vaccine against cytomegalovirus (CMV) that contains sequences encoding the CMV proteins glycoprotein B and pentamer. Humoral and cellular immune responses were evaluated in blood samples collected from healthy CMV-seropositive and CMV-seronegative adults who participated in a phase 1 trial of a three-dose series of mRNA-1647 (NCT03382405). Neutralizing antibody (nAb) titers against fibroblast and epithelial cell infection in sera from CMV-seronegative mRNA-1647 recipients were higher than those in sera from control CMV-seropositive samples and remained elevated up to 12 months after dose 3. nAb responses elicited by mRNA-1647 were comparable across 14 human CMV (HCMV) strains. Frequencies of antigen-specific memory B cells increased in CMV-seropositive and CMV-seronegative participants after each mRNA-1647 dose and remained elevated for up to 6 months after dose 3. mRNA-1647 elicited robust increases in frequencies and polyfunctionality of CD4+ T helper type 1 and effector CD8+ T cells in samples from CMV-seronegative and CMV-seropositive participants after stimulation with HCMV-specific peptides. The administration of three doses of mRNA-1647 to healthy adults elicited high nAb titers with wide-breadth, long-lasting memory B cells, and strong polyfunctional T-cell responses. These findings support further clinical development of the mRNA-1647 vaccine against CMV.IMPORTANCECytomegalovirus (CMV), a common virus that can infect people of all ages, may lead to serious health problems in unborn babies and those with a weakened immune system. Currently, there is no approved vaccine available to prevent CMV infection; however, the investigational messenger RNA (mRNA)-based CMV vaccine, mRNA-1647, is undergoing evaluation in clinical trials. The current analysis examined samples from a phase 1 trial of mRNA-1647 in healthy adults to better understand how the immune system reacts to vaccination. Three doses of mRNA-1647 produced a long-lasting immune response, thus supporting further investigation of the vaccine in the prevention of CMV infection.CLINICAL TRIALSRegistered at ClinicalTrials.gov (NCT03382405).
Collapse
Affiliation(s)
- Kai Wu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Yixuan Jacob Hou
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Dan Makrinos
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Runxia Liu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Alex Zhu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Matthew Koch
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Wen-Han Yu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Yamuna D. Paila
- Infectious Disease Development, Moderna, Inc., Cambridge, Massachusetts, USA
| | | | - Lori Panther
- Infectious Disease Development, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Carole Henry
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Anthony DiPiazza
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Andrea Carfi
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
10
|
Ye X, Shih DJH, Ku Z, Hong J, Barrett DF, Rupp RE, Zhang N, Fu TM, Zheng WJ, An Z. Transcriptional signature of durable effector T cells elicited by a replication defective HCMV vaccine. NPJ Vaccines 2024; 9:70. [PMID: 38561339 PMCID: PMC10984989 DOI: 10.1038/s41541-024-00860-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Human cytomegalovirus (HCMV) is a leading infectious cause of birth defects and the most common opportunistic infection that causes life-threatening diseases post-transplantation; however, an effective vaccine remains elusive. V160 is a live-attenuated replication defective HCMV vaccine that showed a 42.4% efficacy against primary HCMV infection among seronegative women in a phase 2b clinical trial. Here, we integrated the multicolor flow cytometry, longitudinal T cell receptor (TCR) sequencing, and single-cell RNA/TCR sequencing approaches to characterize the magnitude, phenotype, and functional quality of human T cell responses to V160. We demonstrated that V160 de novo induces IE-1 and pp65 specific durable polyfunctional effector CD8 T cells that are comparable to those induced by natural HCMV infection. We identified a variety of V160-responsive T cell clones which exhibit distinctive "transient" and "durable" expansion kinetics, and revealed a transcriptional signature that marks durable CD8 T cells post-vaccination. Our study enhances the understanding of human T-cell immune responses to V160 vaccination.
Collapse
Affiliation(s)
- Xiaohua Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Infectious Disease Research, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - David J H Shih
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Junping Hong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Diane F Barrett
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Richard E Rupp
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tong-Ming Fu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
11
|
Das R, Blázquez-Gamero D, Bernstein DI, Gantt S, Bautista O, Beck K, Conlon A, Rosenbloom DIS, Wang D, Ritter M, Arnold B, Annunziato P, Russell KL. Safety, efficacy, and immunogenicity of a replication-defective human cytomegalovirus vaccine, V160, in cytomegalovirus-seronegative women: a double-blind, randomised, placebo-controlled, phase 2b trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:1383-1394. [PMID: 37660711 DOI: 10.1016/s1473-3099(23)00343-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND A vaccine that prevents cytomegalovirus (CMV) infection in women could reduce the incidence of congenital CMV infection, a major cause of neurodevelopmental disability. We aimed to assess the safety and efficacy of a replication-defective investigational CMV vaccine, V160, in CMV-seronegative women. METHODS This phase 2b, randomised, double-blind, placebo-controlled study was conducted at 90 sites in seven countries (USA, Finland, Canada, Israel, Spain, Russia, and Australia). Eligible participants were generally healthy, CMV-seronegative, non-pregnant, 16-35-year-old women of childbearing potential with exposure to children aged 5 years or younger. Participants were randomly assigned using central randomisation via an interactive response technology system 1:1:1 to one of three groups: V160 three-dose regimen (V160 at day 1, month 2, and month 6), V160 two-dose regimen (V160 on day 1, placebo at month 2, and V160 at month 6), or placebo (saline solution at day 1, month 2, and month 6). The primary outcomes were the efficacy of three doses of V160 in reducing the incidence of primary CMV infection during the follow-up period starting 30 days after the last dose of vaccine using a fixed event rate design, and the safety and tolerability of the two-dose and three-dose V160 regimens. We planned to test the efficacy of a two-dose regimen of V160 in reducing the incidence of primary CMV infection only if the primary efficacy hypothesis was met. Analyses for the primary efficacy endpoint were performed on the per-protocol efficacy population; safety analyses included all randomly assigned participants who received study vaccine. The primary efficacy hypothesis was tested at prespecified interim and final analyses. The study was ongoing and efficacy data continued to accrue at the time of final testing of the primary efficacy hypothesis. Vaccine efficacy was re-estimated after final testing of the primary efficacy hypothesis based on all available efficacy data at end of study. This trial is registered at ClinicalTrials.gov (NCT03486834) and EudraCT (2017-004233-86) and is complete. FINDINGS Between April 30, 2018, and Aug 30, 2019, 7458 participants were screened, of whom 2220 were randomly assigned to the V160 three-dose group (n=733), V160 two-dose group (n=733), or placebo group (n=734). A total of 523 participants in the V160 three-dose group and 519 in the placebo group were included in the final hypothesis testing. Of these, there were 11 cases of CMV infection in the V160 three-dose group and 20 cases in the placebo group. The vaccine efficacy for the V160 three-dose group was 44·6% (95% CI -15·2 to 74·8) at the final testing of the primary efficacy hypothesis, a result corresponding to failure to demonstrate the primary efficacy hypothesis. On the basis of this result, the study was terminated for futility. The re-estimate of vaccine efficacy for the V160 three-dose group based on all available efficacy data at end of study (556 participants in the V160 three-dose group and 543 in the placebo group) was 42·4% (95% CI -13·5 to 71·1). A total of 728 participants in the V160 three-dose group, 729 in the V160 two-dose group, and 732 in the placebo group were included in the safety analyses. The most common solicited injection-site adverse event was injection-site pain (680 [93%] in the V160 three-dose group, 659 [90%] in the V160 two-dose group, and 232 [32%] in the placebo group). The most common solicited systemic adverse event was fatigue (457 [63%] in the V160 three-dose group, 461 [63%] in the V160 two-dose group, and 357 [49%] in the placebo group). No vaccine-related serious adverse events or deaths were reported. INTERPRETATION V160 was generally well tolerated and immunogenic; however, three doses of the vaccine did not reduce the incidence of primary CMV infection in CMV-seronegative women compared with placebo. This study provides insights into the design of future CMV vaccine efficacy trials, particularly for the identification of CMV infection using molecular assays. FUNDING Merck Sharp & Dohme, a subsidiary of Merck & Co, Rahway, NJ, USA (MSD).
Collapse
Affiliation(s)
| | - Daniel Blázquez-Gamero
- Instituto de Investigación Hospital 12 de Octubre (imas12), Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - David I Bernstein
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Soren Gantt
- CHU Sainte-Justine Research Centre, Université de Montréal, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zehner M, Alt M, Ashurov A, Goldsmith JA, Spies R, Weiler N, Lerma J, Gieselmann L, Stöhr D, Gruell H, Schultz EP, Kreer C, Schlachter L, Janicki H, Laib Sampaio K, Stegmann C, Nemetchek MD, Dähling S, Ullrich L, Dittmer U, Witzke O, Koch M, Ryckman BJ, Lotfi R, McLellan JS, Krawczyk A, Sinzger C, Klein F. Single-cell analysis of memory B cells from top neutralizers reveals multiple sites of vulnerability within HCMV Trimer and Pentamer. Immunity 2023; 56:2602-2620.e10. [PMID: 37967532 DOI: 10.1016/j.immuni.2023.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/02/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023]
Abstract
Human cytomegalovirus (HCMV) can cause severe diseases in fetuses, newborns, and immunocompromised individuals. Currently, no vaccines are approved, and treatment options are limited. Here, we analyzed the human B cell response of four HCMV top neutralizers from a cohort of 9,000 individuals. By single-cell analyses of memory B cells targeting the pentameric and trimeric HCMV surface complexes, we identified vulnerable sites on the shared gH/gL subunits as well as complex-specific subunits UL128/130/131A and gO. Using high-resolution cryogenic electron microscopy, we revealed the structural basis of the neutralization mechanisms of antibodies targeting various binding sites. Moreover, we identified highly potent antibodies that neutralized a broad spectrum of HCMV strains, including primary clinical isolates, that outperform known antibodies used in clinical trials. Our study provides a deep understanding of the mechanisms of HCMV neutralization and identifies promising antibody candidates to prevent and treat HCMV infection.
Collapse
Affiliation(s)
- Matthias Zehner
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany.
| | - Mira Alt
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Artem Ashurov
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Jory A Goldsmith
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Rebecca Spies
- Institute for Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Nina Weiler
- Institute for Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Justin Lerma
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lutz Gieselmann
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Dagmar Stöhr
- Institute for Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Henning Gruell
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Eric P Schultz
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Christoph Kreer
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Linda Schlachter
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Hanna Janicki
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | | | - Cora Stegmann
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Michelle D Nemetchek
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Sabrina Dähling
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Leon Ullrich
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brent J Ryckman
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Ramin Lotfi
- Institute for Transfusion Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Adalbert Krawczyk
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Christian Sinzger
- Institute for Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
13
|
Boppana SB, van Boven M, Britt WJ, Gantt S, Griffiths PD, Grosse SD, Hyde TB, Lanzieri TM, Mussi-Pinhata MM, Pallas SE, Pinninti SG, Rawlinson WD, Ross SA, Vossen ACTM, Fowler KB. Vaccine value profile for cytomegalovirus. Vaccine 2023; 41 Suppl 2:S53-S75. [PMID: 37806805 DOI: 10.1016/j.vaccine.2023.06.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/28/2023] [Accepted: 06/02/2023] [Indexed: 10/10/2023]
Abstract
Cytomegalovirus (CMV) is the most common infectious cause of congenital malformation and a leading cause of developmental disabilities such as sensorineural hearing loss (SNHL), motor and cognitive deficits. The significant disease burden from congenital CMV infection (cCMV) led the US National Institute of Medicine to rank CMV vaccine development as the highest priority. An average of 6.7/1000 live births are affected by cCMV, but the prevalence varies across and within countries. In contrast to other congenital infections such as rubella and toxoplasmosis, the prevalence of cCMV increases with CMV seroprevalence rates in the population. The true global burden of cCMV disease is likely underestimated because most infected infants (85-90 %) have asymptomatic infection and are not identified. However, about 7-11 % of those with asymptomatic infection will develop SNHL throughout early childhood. Although no licensed CMV vaccine exists, several candidate vaccines are in development, including one currently in phase 3 trials. Licensure of one or more vaccine candidates is feasible within the next five years. Various models of CMV vaccine strategies employing different target populations have shown to provide substantial benefit in reducing cCMV. Although CMV can cause end-organ disease with significant morbidity and mortality in immunocompromised individuals, the focus of this vaccine value profile (VVP) is on preventing or reducing the cCMV disease burden. This CMV VVP provides a high-level, comprehensive assessment of the currently available data to inform the potential public health, economic, and societal value of CMV vaccines. The CMV VVP was developed by a working group of subject matter experts from academia, public health groups, policy organizations, and non-profit organizations. All contributors have extensive expertise on various elements of the CMV VVP and have described the state of knowledge and identified the current gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Suresh B Boppana
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Michiel van Boven
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, and Julius Center for Health Sciences and Primary Care, Department of Epidemiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - William J Britt
- Departments of Pediatrics, Microbiology, and Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, USA
| | - Soren Gantt
- Centre de recherche du CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Paul D Griffiths
- Emeritus Professor of Virology, University College London, United Kingdom
| | - Scott D Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Terri B Hyde
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Tatiana M Lanzieri
- Measles, Rubella, and Cytomegalovirus Epidemiology Team, Viral Vaccine Preventable Diseases Branch / Division of Viral Diseases. National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Marisa M Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Sarah E Pallas
- Global Immunization Division, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Swetha G Pinninti
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William D Rawlinson
- Serology and Virology Division, NSW Health Pathology Randwick, Prince of Wales Hospital, Sydney, Australia; School of Biotechnology and Biomolecular Sciences, and School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Shannon A Ross
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ann C T M Vossen
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karen B Fowler
- Departments of Pediatrics and Epidemiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
14
|
Hassanzadeh Y, Yaghobi R, Pakzad P, Geramizadeh B. Decreased frequency of Th22 cells and IL-22 cytokine in kidney transplant patients with active cytomegalovirus infection. BMC Immunol 2023; 24:18. [PMID: 37403036 PMCID: PMC10318775 DOI: 10.1186/s12865-023-00555-2] [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: 11/28/2022] [Accepted: 06/29/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND The immunity of CD4+ T cell subsets against human cytomegalovirus (HCMV) is considerable due to their essential role in controlling the infection in transplant individuals. Previously explained CD4+ subsets such as T helper (Th) 1 have been proven to have a protective role against HCMV infection, while the role of the recently identified Th22 subset has not been described yet. Here, the frequency changes of Th22 cells and the IL-22 cytokine production were investigated in kidney transplant recipients with and without HCMV infection. METHODS Twenty kidney transplant patients and ten healthy controls were enrolled in this study. Patients were categorized into HCMV + and HCMV- groups based on the HCMV DNA real-time PCR results. After isolating CD4+ T cells from PBMCs, the phenotype (CCR6+CCR4+CCR10+) and cytokine profile (IFN-γ-IL-17-IL-22+) of Th22 cells were analyzed by flow cytometry. The gene expression of Aryl Hydrocarbon Receptor (AHR) transcription factor was analyzed by real-time PCR. RESULTS The phenotype frequency of these cells was lower in recipients with infection than in those without infection and healthy controls (1.88 ± 0.51 vs. 4.31 ± 1.05; P = 0.03 and 4.22 ± 0.72; P = 0.01, respectively). A lower Th22 cytokine profile was observed in patients with infection than in the two other groups (0.18 ± 0.03 vs. 0.20 ± 0.03; P = 0.96 and 0.33 ± 0.05; P = 0.04, respectively). AHR expression was also lower in patients with active infection. CONCLUSIONS Overall, this study for the first time suggests that the reduced levels of Th22 subset and IL-22 cytokine in patients with active HCMV infection might indicate the protective role of these cells against HCMV.
Collapse
Affiliation(s)
- Yashgin Hassanzadeh
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parviz Pakzad
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Bita Geramizadeh
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
Wang HY, Taher H, Kreklywich CN, Schmidt KA, Scheef EA, Barfield R, Otero CE, Valencia SM, Crooks CM, Mirza A, Woods K, Burgt NV, Kowalik TF, Barry PA, Hansen SG, Tarantal AF, Chan C, Streblow DN, Picker LJ, Kaur A, Früh K, Permar SR, Malouli D. The pentameric complex is not required for vertical transmission of cytomegalovirus in seronegative pregnant rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545169. [PMID: 37398229 PMCID: PMC10312687 DOI: 10.1101/2023.06.15.545169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Congenital cytomegalovirus (cCMV) infection is the leading infectious cause of neonatal neurological impairment but essential virological determinants of transplacental CMV transmission remain unclear. The pentameric complex (PC), composed of five subunits, glycoproteins H (gH), gL, UL128, UL130, and UL131A, is essential for efficient entry into non-fibroblast cells in vitro . Based on this role in cell tropism, the PC is considered a possible target for CMV vaccines and immunotherapies to prevent cCMV. To determine the role of the PC in transplacental CMV transmission in a non-human primate model of cCMV, we constructed a PC-deficient rhesus CMV (RhCMV) by deleting the homologues of the HCMV PC subunits UL128 and UL130 and compared congenital transmission to PC-intact RhCMV in CD4+ T cell-depleted or immunocompetent RhCMV-seronegative, pregnant rhesus macaques (RM). Surprisingly, we found that the transplacental transmission rate was similar for PC-intact and PC-deleted RhCMV based on viral genomic DNA detection in amniotic fluid. Moreover, PC-deleted and PC-intact RhCMV acute infection led to similar peak maternal plasma viremia. However, there was less viral shedding in maternal urine and saliva and less viral dissemination in fetal tissues in the PC-deleted group. As expected, dams inoculated with PC-deleted RhCMV demonstrated lower plasma IgG binding to PC-intact RhCMV virions and soluble PC, as well as reduced neutralization of PC-dependent entry of the PC-intact RhCMV isolate UCD52 into epithelial cells. In contrast, binding to gH expressed on the cell surface and neutralization of entry into fibroblasts by the PC-intact RhCMV was higher for dams infected with PC-deleted RhCMV compared to those infected with PC-intact RhCMV. Our data demonstrates that the PC is dispensable for transplacental CMV infection in our non-human primate model. One Sentence Summary Congenital CMV transmission frequency in seronegative rhesus macaques is not affected by the deletion of the viral pentameric complex.
Collapse
|
16
|
Gomes AC, Baraniak IA, McIntosh MR, Sodi I, Langstone T, Siddiqui S, Atkinson C, McLean GR, Griffiths PD, Reeves MB. A temperature-dependent virus-binding assay reveals the presence of neutralizing antibodies in human cytomegalovirus gB vaccine recipients' sera. J Gen Virol 2023; 104:001860. [PMID: 37310000 PMCID: PMC10661908 DOI: 10.1099/jgv.0.001860] [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: 03/22/2023] [Accepted: 05/15/2023] [Indexed: 06/14/2023] Open
Abstract
Human cytomegalovirus (HCMV) remains an important cause of mortality in immune-compromised transplant patients and following congenital infection. Such is the burden, an effective vaccine strategy is considered to be of the highest priority. The most successful vaccines to date have focused on generating immune responses against glycoprotein B (gB) - a protein essential for HCMV fusion and entry. We have previously reported that an important component of the humoral immune response elicited by gB/MF59 vaccination of patients awaiting transplant is the induction of non-neutralizing antibodies that target cell-associated virus with little evidence of concomitant classical neutralizing antibodies. Here we report that a modified neutralization assay that promotes prolonged binding of HCMV to the cell surface reveals the presence of neutralizing antibodies in sera taken from gB-vaccinated patients that cannot be detected using standard assays. We go on to show that this is not a general feature of gB-neutralizing antibodies, suggesting that specific antibody responses induced by vaccination could be important. Although we can find no evidence that these neutralizing antibody responses are a correlate of protection in vivo in transplant recipients their identification demonstrates the utility of the approach in identifying these responses. We hypothesize that further characterization has the potential to aid the identification of functions within gB that are important during the entry process and could potentially improve future vaccine strategies directed against gB if they prove to be effective against HCMV at higher concentrations.
Collapse
Affiliation(s)
- Ariane C. Gomes
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| | - Ilona A. Baraniak
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| | - Megan R. McIntosh
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| | - Isabella Sodi
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| | - Toby Langstone
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| | - Saima Siddiqui
- London Metropolitan University, School of Human Sciences, London, N7 8DB, UK
| | - Claire Atkinson
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| | - Gary R. McLean
- London Metropolitan University, School of Human Sciences, London, N7 8DB, UK
- Imperial College London, National Heart and Lung Institute, London, W2 1PG, UK
| | - Paul D. Griffiths
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| | - Matthew B. Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Campus, London, NW3 2PP, UK
| |
Collapse
|
17
|
Sartori P, Egloff C, Hcini N, Vauloup Fellous C, Périllaud-Dubois C, Picone O, Pomar L. Primary, Secondary, and Tertiary Prevention of Congenital Cytomegalovirus Infection. Viruses 2023; 15:v15040819. [PMID: 37112800 PMCID: PMC10146889 DOI: 10.3390/v15040819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/11/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Cytomegalovirus infection is the most common congenital infection, affecting about 1% of births worldwide. Several primary, secondary, and tertiary prevention strategies are already available during the prenatal period to help mitigate the immediate and long-term consequences of this infection. In this review, we aim to present and assess the efficacy of these strategies, including educating pregnant women and women of childbearing age on their knowledge of hygiene measures, development of vaccines, screening for cytomegalovirus infection during pregnancy (systematic versus targeted), prenatal diagnosis and prognostic assessments, and preventive and curative treatments in utero.
Collapse
Affiliation(s)
- Pauline Sartori
- School of Health Sciences (HESAV), University of Applied Sciences and Arts Western Switzerland, 1011 Lausanne, Switzerland
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Charles Egloff
- Assistance Publique-Hôpitaux de Paris APHP, Nord, Service de Gynécologie Obstétrique, Hôpital Louis Mourier, 92700 Colombes, France
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
| | - Najeh Hcini
- Department of Obstetrics and Gynaecology, West French Guiana Hospital Center, French 97320, Guyana
- CIC Inserm 1424 et DFR Santé Université Guyane, 97320 ST Laurent du Maroni, France
| | - Christelle Vauloup Fellous
- Université Paris-Saclay, INSERM U1193, 94804 Villejuif, France
- Laboratoire de Virologie, AP-HP, Hôpital Paul-Brousse, 94804 Villejuif, France
- Groupe de Recherche sur les Infections Pendant la Grossesse (GRIG), 75000 Paris, France
| | - Claire Périllaud-Dubois
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
- Virology Laboratory, AP-HP, Sorbonne Université, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Olivier Picone
- Assistance Publique-Hôpitaux de Paris APHP, Nord, Service de Gynécologie Obstétrique, Hôpital Louis Mourier, 92700 Colombes, France
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
- Groupe de Recherche sur les Infections Pendant la Grossesse (GRIG), 75000 Paris, France
| | - Léo Pomar
- School of Health Sciences (HESAV), University of Applied Sciences and Arts Western Switzerland, 1011 Lausanne, Switzerland
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
18
|
Gomes AC, Baraniak IA, Lankina A, Moulder Z, Holenya P, Atkinson C, Tang G, Mahungu T, Kern F, Griffiths PD, Reeves MB. The cytomegalovirus gB/MF59 vaccine candidate induces antibodies against an antigenic domain controlling cell-to-cell spread. Nat Commun 2023; 14:1041. [PMID: 36823200 PMCID: PMC9950427 DOI: 10.1038/s41467-023-36683-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Vaccination against human cytomegalovirus (CMV) infection remains high priority. A recombinant form of a protein essential for CMV entry, glycoprotein B (gB), demonstrated partial protection in a clinical trial (NCT00299260) when delivered with the MF59 adjuvant. Although the antibody titre against gB correlated with protection poor neutralising responses against the 5 known antigenic domains (AD) of gB were evident. Here, we show that vaccination of CMV seronegative patients induces an antibody response against a region of gB we term AD-6. Responses to the polypeptide AD-6 are detected in >70% of vaccine recipients yet in <5% of naturally infected people. An AD-6 antibody binds to gB and to infected cells but not the virion directly. Consistent with this, the AD-6 antibody is non-neutralising but, instead, prevents cell-cell spread of CMV in vitro. The discovery of AD-6 responses has the potential to explain part of the protection mediated by gB vaccines against CMV following transplantation.
Collapse
Affiliation(s)
- A C Gomes
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - I A Baraniak
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - A Lankina
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - Z Moulder
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - P Holenya
- JPT Peptide Technologies GmbH, Berlin, Germany
| | - C Atkinson
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - G Tang
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - T Mahungu
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - F Kern
- JPT Peptide Technologies GmbH, Berlin, Germany
- Experimental Medicine, Brighton and Sussex Medical School, Brighton, United Kingdom
| | - P D Griffiths
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom
| | - M B Reeves
- Institute of Immunity & Transplantation, UCL, London, NW3 2PP, United Kingdom.
| |
Collapse
|
19
|
Human Cytomegalovirus pUL11, a CD45 Ligand, Disrupts CD4 T Cell Control of Viral Spread in Epithelial Cells. mBio 2022; 13:e0294622. [PMID: 36445084 PMCID: PMC9765415 DOI: 10.1128/mbio.02946-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Human cytomegalovirus (HCMV) encodes numerous immunomodulatory genes that facilitate its persistence. Previously described mechanisms by which HCMV avoids T cell control typically involve evasion of detection by infected cells. Here, we show that the virus also inhibits T cells directly via an interaction between the pUL11 glycoprotein on infected cells and the CD45 phosphatase on T cells. The antiviral functions of CD4 T cells are impaired as a result of this interaction, largely via induced interleukin 10 (IL-10) secretion in the CD4 T cell central memory compartment, resulting in enhanced viral spread. This establishes CD45 as an inhibitory receptor that regulates antiviral T cell functions and has parallels with the manipulation of natural killer (NK) cells by HCMV. By coculturing donor T cells with HCMV-infected epithelial cells, we observed that CD4 T cells can respond to epithelial cell antigen presentation and can control HCMV spread via cytolytic and cytokine-dependent mechanisms. pUL11 impairs both mechanisms. We showed that pUL11-induced IL-10 secretion requires IL-2, mTOR, and T cell receptor signaling. This characterization of the effects of the pUL11-CD45 interaction may allow for the development of new antiviral therapies and treatments for inflammatory disorders. IMPORTANCE Human cytomegalovirus (HCMV) is adept at avoiding its host's immune defenses, both by evading detection and by directly inhibiting immune cells. This can lead to a loss of control of the infection, and dangerous disease can result, particularly in cases in which an individual's immune system is immature, weak, or suppressed. T cells form a crucial part of the response to HCMV and are used in cellular HCMV therapies. We show that an interaction between a viral glycoprotein (pUL11) and a T cell surface receptor (CD45) impairs T cell memory functions and allows for increased viral spread. This defines a new immunomodulatory strategy for the virus as well as a new T cell regulatory mechanism. These results are important, as they increase our understanding of how T cells function and how HCMV disrupts them. This will allow for the development of new antiviral therapies that restore T cell functions and indicates a new target for controlling pathological T cell disorders.
Collapse
|
20
|
Davies EL, Noor M, Lim EY, Houldcroft CJ, Okecha G, Atkinson C, Reeves MB, Jackson SE, Wills MR. HCMV carriage in the elderly diminishes anti-viral functionality of the adaptive immune response resulting in virus replication at peripheral sites. Front Immunol 2022; 13:1083230. [PMID: 36591233 PMCID: PMC9797693 DOI: 10.3389/fimmu.2022.1083230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection and periodic reactivation is, generally, well controlled by adaptative immune responses in the healthy. In older people, overt HCMV disease is rarely seen despite the association of HCMV with increased risk of mortality; evidence from studies of unwell aged populations suggest that HCMV seropositivity is an important co-morbidity factor. HCMV genomes have been detected in urine from older donors, suggesting that the immune response prevents systemic disease but possibly immunomodulation due to lifelong viral carriage may alter its efficacy at peripheral tissue sites. Previously we have demonstrated that there were no age-related expansions of T cell responses to HCMV or increase in latent viral carriage with age and these T cells produced anti-viral cytokines and viremia was very rarely detected. To investigate the efficacy of anti-HCMV responses with increasing age, we used an in vitro Viral Dissemination Assay (VDA) using autologous dermal fibroblasts to determine the anti-viral effector capacity of total PBMC, as well as important subsets (T cells, NK cells). In parallel we assessed components of the humoral response (antibody neutralization) and combined this with qPCR detection of HCMV in blood, saliva and urine in a cohort of young and old donors. Consistent with previous studies, we again show HCMV specific cIL-10, IFNγ and TNFα T cell responses to peptides did not show an age-related defect. However, assessment of direct anti-viral cellular and antibody-mediated adaptive immune responses using the VDA shows that older donors are significantly less able to control viral dissemination in an in vitro assay compared to young donors. Corroborating this observation, we detected viral genomes in saliva samples only from older donors, these donors had a defect in cellular control of viral spread in our in vitro assay. Phenotyping of fibroblasts used in this study shows expression of a number of checkpoint inhibitor ligands which may contribute to the defects observed. The potential to therapeutically intervene in checkpoint inhibitor pathways to prevent HCMV reactivation in the unwell aged is an exciting avenue to explore.
Collapse
Affiliation(s)
- Emma L. Davies
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Mahlaqua Noor
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Eleanor Y. Lim
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Charlotte J. Houldcroft
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Georgina Okecha
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Claire Atkinson
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Matthew B. Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Sarah E. Jackson
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Mark R. Wills
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| |
Collapse
|
21
|
Hu X, Wang HY, Otero CE, Jenks JA, Permar SR. Lessons from Acquired Natural Immunity and Clinical Trials to Inform Next-Generation Human Cytomegalovirus Vaccine Development. Annu Rev Virol 2022; 9:491-520. [PMID: 35704747 PMCID: PMC10154983 DOI: 10.1146/annurev-virology-100220-010653] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human cytomegalovirus (HCMV) infection, the most common cause of congenital disease globally, affecting an estimated 1 million newborns annually, can result in lifelong sequelae in infants, such as sensorineural hearing loss and brain damage. HCMV infection also leads to a significant disease burden in immunocompromised individuals. Hence, an effective HCMV vaccine is urgently needed to prevent infection and HCMV-associated diseases. Unfortunately, despite more than five decades of vaccine development, no successful HCMV vaccine is available. This review summarizes what we have learned from acquired natural immunity, including innate and adaptive immunity; the successes and failures of HCMV vaccine human clinical trials; the progress in related animal models; and the analysis of protective immune responses during natural infection and vaccination settings. Finally, we propose novel vaccine strategies that will harness the knowledge of protective immunity and employ new technology and vaccine concepts to inform next-generation HCMV vaccine development.
Collapse
Affiliation(s)
- Xintao Hu
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Claire E Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer A Jenks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
| |
Collapse
|
22
|
Spencer Clinton JL, Hoornweg TE, Tan J, Peng R, Schaftenaar W, Rutten VPMG, de Haan CAM, Ling PD. EEHV1A glycoprotein B subunit vaccine elicits humoral and cell-mediated immune responses in mice. Vaccine 2022; 40:5131-5140. [PMID: 35879117 DOI: 10.1016/j.vaccine.2022.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 10/16/2022]
Abstract
Asian elephants are an endangered species facing many threats, including severe hemorrhagic disease (HD) caused by the elephant endotheliotropic herpesvirus (EEHV). EEHV-HD is the leading cause of death in captive juvenile Asian elephants in North America and Europe, and also affects elephants in their natural range countries. Significant challenges exist for successful treatment of EEHV-HD, which include timely recognition of disease onset and limited availability of highly effective treatment options. To address this problem, our goal is to prevent lethal disease in young elephants by developing a vaccine that elicits robust and durable humoral and cell-mediated immunity against EEHV. EEHV glycoprotein B (gB) is a major target for cellular and humoral immunity in elephants previously exposed to EEHV. Therefore, we generated a vaccine containing recombinant EEHV1A gB together with a liposome formulated TLR-4 and saponin combination adjuvant (SLA-LSQ). CD-1 mice that received one or two vaccinations with the vaccine elicited significant anti-gB antibody and polyfunctional CD4+ and CD8+ T cell responses, while no adverse effects of vaccination were observed. Overall, our findings demonstrate that an adjuvanted gB protein subunit vaccine stimulates robust humoral and cell-mediated immune responses and supports its potential use in elephants.
Collapse
Affiliation(s)
- Jennifer L Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Tabitha E Hoornweg
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands.
| | - Jie Tan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Rongsheng Peng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Willem Schaftenaar
- Veterinary Advisor EAZA Elephant TAG, Rotterdam Zoo, Blijdorplaan 8, 3041 JG Rotterdam, The Netherlands.
| | - Victor P M G Rutten
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa.
| | - Cornelis A M de Haan
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands.
| | - Paul D Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| |
Collapse
|
23
|
Majewska A, Mlynarczyk-Bonikowska B. 40 Years after the Registration of Acyclovir: Do We Need New Anti-Herpetic Drugs? Int J Mol Sci 2022; 23:ijms23073431. [PMID: 35408788 PMCID: PMC8998721 DOI: 10.3390/ijms23073431] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 01/17/2023] Open
Abstract
Herpes simplex virus types 1 and 2 HSV1 and 2, namely varicella-zoster VZV and cytomegalovirus CMV, are among the most common pathogens worldwide. They remain in the host body for life. The course of infection with these viruses is often asymptomatic or mild and self-limiting, but in immunocompromised patients, such as solid organ or bone marrow transplant recipients, the course can be very severe or even life-threatening. Unfortunately, in the latter group, the highest percentage of infections with strains resistant to routinely used drugs is observed. On the other hand, frequent recurrences of genital herpes can be a problem even in people with normal immunity. Genital herpes also increases the risk of acquiring sexually transmitted diseases, including HIV infection and, if present in pregnant women, poses a risk to the fetus and newborn. Even more frequently than herpes simplex, congenital infections can be caused by cytomegalovirus. We present the most important anti-herpesviral agents, the mechanisms of resistance to these drugs, and the associated mutations in the viral genome. Special emphasis was placed on newly introduced drugs such as maribavir and brincidofovir. We also briefly discuss the most promising substances in preclinical testing as well as immunotherapy options and vaccines currently in use and under investigation.
Collapse
Affiliation(s)
- Anna Majewska
- Department of Medical Microbiology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, Poland;
| | - Beata Mlynarczyk-Bonikowska
- Department of Dermatology, Immunodermatology and Venereology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
- Correspondence: ; Tel.: +48-225021313
| |
Collapse
|
24
|
Ataya M, Redondo-Pachón D, Llinàs-Mallol L, Yélamos J, Alari-Pahissa E, Pérez-Sáez MJ, Altadill M, Raïch-Regué D, Vilches C, Pascual J, Crespo M, López-Botet M. Long-Term Evolution of the Adaptive NKG2C + NK Cell Response to Cytomegalovirus Infection in Kidney Transplantation: An Insight on the Diversity of Host-Pathogen Interaction. THE JOURNAL OF IMMUNOLOGY 2021; 207:1882-1890. [PMID: 34470855 DOI: 10.4049/jimmunol.2100055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 08/01/2021] [Indexed: 11/19/2022]
Abstract
Human CMV infection is frequent in kidney transplant recipients (KTR). Pretransplant Ag-specific T cells and adaptive NKG2C+ NK cells associate with reduced incidence of infection in CMV+ KTR. Expansions of adaptive NKG2C+ NK cells were reported in posttransplant CMV-infected KTR. To further explore this issue, NKG2C+ NK, CD8+, and TcRγδ T cells were analyzed pretransplant and at different time points posttransplant for ≥24 mo in a cohort of CMV+ KTR (n = 112), stratified according to CMV viremia detection. In cryopreserved samples from a subgroup (n = 49), adaptive NKG2C+ NK cell markers and T cell subsets were compared after a longer follow-up (median, 56 mo), assessing the frequencies of CMV-specific T cells and viremia at the last time point. Increased proportions of NKG2C+ NK, CD8+, and TcRγδ T cells were detected along posttransplant evolution in viremia(+) KTR. However, the individual magnitude and kinetics of the NKG2C+ NK response was variable and only exceptionally detected among viremia(-) KTR, presumably reflecting subclinical viral replication events. NKG2C+ expansions were independent of KLRC2 zygosity and associated with higher viral loads at diagnosis; no relation with other clinical parameters was perceived. Increased proportions of adaptive NKG2C+ NK cells (CD57+, ILT2+, FcεRIγ-) were observed after resolution of viremia long-term posttransplant, coinciding with increased CD8+ and Vδ2- γδ T cells; at that stage CMV-specific T cells were comparable to viremia(-) cases. These data suggest that adaptive NKG2C+ NK cells participate with T cells to restore CMV replication control, although their relative contribution cannot be discerned.
Collapse
Affiliation(s)
| | - Dolores Redondo-Pachón
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | | | - José Yélamos
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Immunology Laboratory, Department of Pathology, Hospital del Mar, Barcelona, Spain; and
| | | | - María J Pérez-Sáez
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | | | - Dàlia Raïch-Regué
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain
| | - Carlos Vilches
- Immunogenetics-HLA, Instituto de Investigación Sanitaria Puerta de Hierro Segovia de Arana, Majadahonda, Madrid, Spain
| | - Julio Pascual
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Marta Crespo
- Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Miguel López-Botet
- Universitat Pompeu Fabra, Barcelona, Spain; .,Instituto Hospital del Mar de Investigaciones Médicas, Barcelona, Spain.,Immunology Laboratory, Department of Pathology, Hospital del Mar, Barcelona, Spain; and
| |
Collapse
|
25
|
Immune Prophylaxis and Therapy for Human Cytomegalovirus Infection. Int J Mol Sci 2021; 22:ijms22168728. [PMID: 34445434 PMCID: PMC8395925 DOI: 10.3390/ijms22168728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Human Cytomegalovirus (HCMV) infection is widespread and can result in severe sequelae in susceptible populations. Primary HCMV infection of naïve individuals results in life-long latency characterized by frequent and sporadic reactivations. HCMV infection elicits a robust antibody response, including neutralizing antibodies that can block the infection of susceptible cells in vitro and in vivo. Thus, antibody products and vaccines hold great promise for the prevention and treatment of HCMV, but to date, most attempts to demonstrate their safety and efficacy in clinical trials have been unsuccessful. In this review we summarize publicly available data on these products and highlight new developments and approaches that could assist in successful translation of HCMV immunotherapies.
Collapse
|
26
|
A conditionally replication-defective cytomegalovirus vaccine elicits potent and diverse functional monoclonal antibodies in a phase I clinical trial. NPJ Vaccines 2021; 6:79. [PMID: 34078915 PMCID: PMC8172929 DOI: 10.1038/s41541-021-00342-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/24/2021] [Indexed: 02/03/2023] Open
Abstract
A conditionally replication-defective human cytomegalovirus (HCMV) vaccine, V160, was shown to be safe and immunogenic in a two-part, double-blind, randomized, placebo-controlled phase I clinical trial (NCT01986010). However, the specificities and functional properties of V160-elicited antibodies remain undefined. Here, we characterized 272 monoclonal antibodies (mAbs) isolated from single memory B cells of six V160-vaccinated subjects. The mAbs bind to diverse HCMV antigens, including multiple components of the pentamer, gB, and tegument proteins. The most-potent neutralizing antibodies target the pentamer-UL subunits. The binding sites of the antibodies overlap with those of antibodies responding to natural HCMV infection. The majority of the neutralizing antibodies target the gHgL subunit. The non-neutralizing antibodies predominantly target the gB and pp65 proteins. Sequence analysis indicated that V160 induced a class of gHgL antibodies expressing the HV1-18/KV1-5 germline genes in multiple subjects. This study provides valuable insights into primary targets for anti-HCMV antibodies induced by V160 vaccination.
Collapse
|
27
|
Feldmann S, Grimm I, Stöhr D, Antonini C, Lischka P, Sinzger C, Stegmann C. Targeted mutagenesis on PDGFRα-Fc identifies amino acid modifications that allow efficient inhibition of HCMV infection while abolishing PDGF sequestration. PLoS Pathog 2021; 17:e1009471. [PMID: 33780515 PMCID: PMC8031885 DOI: 10.1371/journal.ppat.1009471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 04/08/2021] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
Platelet-derived growth factor receptor alpha (PDGFRα) serves as an entry receptor for the human cytomegalovirus (HCMV), and soluble PDGFRα-Fc can neutralize HCMV at a half-maximal effective concentration (EC50) of about 10 ng/ml. While this indicates a potential for usage as an HCMV entry inhibitor PDGFRα-Fc can also bind the physiological ligands of PDGFRα (PDGFs), which likely interferes with the respective signaling pathways and represents a potential source of side effects. Therefore, we tested the hypothesis that interference with PDGF signaling can be prevented by mutations in PDGFRα-Fc or combinations thereof, without losing the inhibitory potential for HCMV. To this aim, a targeted mutagenesis approach was chosen. The mutations were quantitatively tested in biological assays for interference with PDGF-dependent signaling as well as inhibition of HCMV infection and biochemically for reduced affinity to PDGF-BB, facilitating quantification of PDGFRα-Fc selectivity for HCMV inhibition. Mutation of Ile 139 to Glu and Tyr 206 to Ser strongly reduced the affinity for PDGF-BB and hence interference with PDGF-dependent signaling. Inhibition of HCMV infection was less affected, thus increasing the selectivity by factor 4 and 8, respectively. Surprisingly, the combination of these mutations had an additive effect on binding of PDGF-BB but not on inhibition of HCMV, resulting in a synergistic 260fold increase of selectivity. In addition, a recently reported mutation, Val 242 to Lys, was included in the analysis. PDGFRα-Fc with this mutation was fully effective at blocking HCMV entry and had a drastically reduced affinity for PDGF-BB. Combining Val 242 to Lys with Ile 139 to Glu and/or Tyr 206 to Ser further reduced PDGF ligand binding beyond detection. In conclusion, this targeted mutagenesis approach identified combinations of mutations in PDGFRα-Fc that prevent interference with PDGF-BB but maintain inhibition of HCMV, which qualifies such mutants as candidates for the development of HCMV entry inhibitors.
Collapse
Affiliation(s)
- Svenja Feldmann
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Dagmar Stöhr
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | - Chiara Antonini
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Peter Lischka
- AiCuris Anti-infective Cures GmbH, Wuppertal, Germany
| | | | - Cora Stegmann
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
28
|
Perotti M, Marcandalli J, Demurtas D, Sallusto F, Perez L. Rationally designed Human Cytomegalovirus gB nanoparticle vaccine with improved immunogenicity. PLoS Pathog 2020; 16:e1009169. [PMID: 33370407 PMCID: PMC7794029 DOI: 10.1371/journal.ppat.1009169] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/08/2021] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the primary viral cause of congenital birth defects and causes significant morbidity and mortality in immune-suppressed transplant recipients. Despite considerable efforts in vaccine development, HCMV infection still represents an unmet clinical need. In recent phase II trials, a MF59-adjuvanted gB vaccine showed only modest efficacy in preventing infection. These findings might be attributed to low level of antibodies (Abs) with a neutralizing activity induced by this vaccine. Here, we analyzed the immunogenicity of each gB antigenic domain (AD) and demonstrated that domain I of gB (AD5) is the main target of HCMV neutralizing antibodies. Furthermore, we designed, characterized and evaluated immunogenic responses to two different nanoparticles displaying a trimeric AD5 antigen. We showed that mice immunization with nanoparticles induces sera neutralization titers up to 100-fold higher compared to those obtained with the gB extracellular domain (gBECD). Collectively, these results illustrate with a medically relevant example the advantages of using a general approach combining antigen discovery, protein engineering and scaffold presentation for modern development of subunit vaccines against complex pathogens.
Collapse
Affiliation(s)
- Michela Perotti
- Institute for Research in Biomedicine, Università della Svizzera italiana, faculty of Biomedical Sciences, Bellinzona, Switzerland.,Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Jessica Marcandalli
- Institute for Research in Biomedicine, Università della Svizzera italiana, faculty of Biomedical Sciences, Bellinzona, Switzerland
| | - Davide Demurtas
- BioEM Facility, School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, faculty of Biomedical Sciences, Bellinzona, Switzerland.,Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Laurent Perez
- Institute for Research in Biomedicine, Università della Svizzera italiana, faculty of Biomedical Sciences, Bellinzona, Switzerland.,University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Department of Medicine, Division of Immunology and Allergy, Center for Human Immunology (CHIL), Lausanne, Switzerland
| |
Collapse
|
29
|
Materne EC, Lilleri D, Garofoli F, Lombardi G, Furione M, Zavattoni M, Gibson L. Cytomegalovirus-Specific T Cell Epitope Recognition in Congenital Cytomegalovirus Mother-Infant Pairs. Front Immunol 2020; 11:568217. [PMID: 33329532 PMCID: PMC7732427 DOI: 10.3389/fimmu.2020.568217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/09/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Congenital cytomegalovirus (cCMV) infection is the most common infection acquired before birth and from which about 20% of infants develop permanent neurodevelopmental effects regardless of presence or absence of symptoms at birth. Viral escape from host immune control may be a mechanism of CMV transmission and infant disease severity. We sought to identify and compare CMV epitopes recognized by mother-infant pairs. We also hypothesized that if immune escape were occurring, then one pattern of longitudinal CD8 T cell responses restricted by shared HLA alleles would be maternal loss (by viral escape) and infant gain (by viral reversion to wildtype) of CMV epitope recognition. Methods: The study population consisted of 6 women with primary CMV infection during pregnancy and their infants with cCMV infection. CMV UL83 and UL123 peptides with known or predicted restriction by maternal MHC class I alleles were identified, and a subset was selected for testing based on several criteria. Maternal or infant cells were stimulated with CMV peptides in the IFN-γ ELISpot assay. Results: Overall, 14 of 25 (56%; 8 UL83 and 6 UL123) peptides recognized by mother-infant pairs were not previously reported as CD8 T cell epitopes. Of three pairs with longitudinal samples, one showed maternal loss and infant gain of responses to a CMV epitope restricted by a shared HLA allele. Conclusions: CD8 T cell responses to multiple novel CMV epitopes were identified, particularly in infants. Moreover, the hypothesized pattern of CMV immune escape was observed in one mother-infant pair. These findings emphasize that knowledge of paired CMV epitope recognition allows exploration of viral immune escape that may operate within the maternal-fetal system. Our work provides rationale for future studies of this potential mechanism of CMV transmission during pregnancy or clinical outcomes of infants with cCMV infection.
Collapse
Affiliation(s)
- Emma C Materne
- University of Massachusetts Medical School, Worcester, MA, United States
| | - Daniele Lilleri
- Unità Operativa Complessa (UOC) Laboratorio Genetica - Trapiantologia e Malattie Cardiovascolari, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Francesca Garofoli
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Giuseppina Lombardi
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Milena Furione
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Maurizio Zavattoni
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Laura Gibson
- University of Massachusetts Medical School, Worcester, MA, United States.,Department of Medicine, UMass Memorial Medical Center, Worcester, MA, United States.,Department of Pediatrics, UMass Memorial Medical Center, Worcester, MA, United States
| |
Collapse
|
30
|
Nelson CS, Jenks JA, Pardi N, Goodwin M, Roark H, Edwards W, McLellan JS, Pollara J, Weissman D, Permar SR. Human Cytomegalovirus Glycoprotein B Nucleoside-Modified mRNA Vaccine Elicits Antibody Responses with Greater Durability and Breadth than MF59-Adjuvanted gB Protein Immunization. J Virol 2020; 94:e00186-20. [PMID: 32051265 PMCID: PMC7163130 DOI: 10.1128/jvi.00186-20] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
A vaccine to prevent maternal acquisition of human cytomegalovirus (HCMV) during pregnancy is a primary strategy to reduce the incidence of congenital disease. The MF59-adjuvanted glycoprotein B (gB) protein subunit vaccine (gB/MF59) is the most efficacious vaccine tested to date for this indication. We previously identified that gB/MF59 vaccination elicited poor neutralizing antibody responses and an immunodominant response against gB antigenic domain 3 (AD-3). Thus, we sought to test novel gB vaccines to improve functional antibody responses and reduce AD-3 immunodominance. Groups of juvenile New Zealand White rabbits were administered 3 sequential doses of the full-length gB protein with an MF59-like squalene-based adjuvant, the gB ectodomain protein (lacking AD-3) with squalene adjuvant, or lipid nanoparticle (LNP)-encapsulated nucleoside-modified mRNA encoding full-length gB. All vaccines were highly immunogenic with similar kinetics and comparable peak gB-binding and functional antibody responses. The AD-3-immunodominant IgG response following human gB/MF59 vaccination was closely mimicked in rabbits. Though gB ectodomain subunit vaccination eliminated targeting of epitopes in AD-3, it did not improve vaccine-elicited neutralizing or nonneutralizing antibody functions. gB nucleoside-modified mRNA-LNP-immunized rabbits exhibited an enhanced durability of vaccine-elicited antibody responses. Furthermore, the gB mRNA-LNP vaccine enhanced the breadth of IgG binding responses against discrete gB peptides. Finally, low-magnitude gB-specific T cell activity was observed in the full-length gB protein and mRNA-LNP groups, though not in ectodomain-vaccinated rabbits. Altogether, these data suggest that the use of gB nucleoside-modified mRNA-LNP vaccines is a viable strategy for improving on the partial efficacy of gB/MF59 vaccination and should be further evaluated in preclinical models.IMPORTANCE Human cytomegalovirus (HCMV) is the most common infectious cause of infant birth defects, resulting in permanent neurological disability for one newborn child every hour in the United States. After more than a half century of research and development, we remain without a clinically licensed vaccine or immunotherapeutic to reduce the burden of HCMV-associated disease. In this study, we sought to improve upon the glycoprotein B protein vaccine (gB/MF59), the most efficacious HCMV vaccine evaluated in a clinical trial, via targeted modifications to either the protein structure or vaccine formulation. Utilization of a novel vaccine platform, nucleoside-modified mRNA formulated in lipid nanoparticles, increased the durability and breadth of vaccine-elicited antibody responses. We propose that an mRNA-based gB vaccine may ultimately prove more efficacious than the gB/MF59 vaccine and should be further evaluated for its ability to elicit antiviral immune factors that can prevent HCMV-associated disease.
Collapse
Affiliation(s)
- Cody S Nelson
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer A Jenks
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Norbert Pardi
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Matthew Goodwin
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Hunter Roark
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Whitney Edwards
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Justin Pollara
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sallie R Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|