1
|
Karthigeyan KP, Connors M, Binuya CR, Gross M, Fuller AS, Crooks CM, Wang HY, Sponholtz MR, Byrne PO, Herbek S, Andy C, Gerber LM, Campbell JD, Williams CA, Mitchell E, van der Maas L, Miller I, Yu D, Bottomley MJ, McLellan JS, Permar SR. A human cytomegalovirus prefusion-like glycoprotein B subunit vaccine elicits humoral immunity similar to that of postfusion gB in mice. J Virol 2025:e0217824. [PMID: 40338082 DOI: 10.1128/jvi.02178-24] [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: 12/09/2024] [Accepted: 04/06/2025] [Indexed: 05/09/2025] Open
Abstract
Human cytomegalovirus (HCMV) is the leading infectious cause of birth defects. Despite the global disease burden, there is no Food and Drug Administration (FDA)-approved HCMV vaccine. The most efficacious HCMV vaccine candidates to date have used glycoprotein B (gB), a class III viral fusion protein, in its postfusion form. While some viral fusion proteins have been shown to elicit stronger neutralizing responses in their prefusion conformation, HCMV prefusion-like and postfusion gB were recently shown to elicit antibodies with similar fibroblast neutralization titers in mice. We aimed to define and compare the specificity and functionality of plasma IgG elicited by distinct prefusion-like and postfusion gB constructs. Prefusion-like and postfusion gB elicited comparable IgG responses that predominantly mapped to the AD-5 antigenic domain known to elicit neutralizing antibodies. Interestingly, postfusion gB elicited significantly higher plasma IgG binding to cell-associated gB and antibody-dependent cellular phagocytosis than that of prefusion-like gB. The vaccines elicited comparable neutralization titers of heterologous HCMV strain AD169r in fibroblasts; however, neither elicited neutralizing titers against the vaccine-matched strain Towne in fibroblasts. Our data indicate that gB in this prefusion-like conformation elicits similar specificity and functional humoral immunity to that of postfusion gB, unlike certain class I viral fusion proteins that have been used as vaccine antigens. These findings deepen our understanding of the immune response elicited by class III fusion proteins and may inform further design and testing of conformationally dependent herpesvirus glycoprotein vaccine candidates.IMPORTANCEVaccines against human cytomegalovirus (HCMV) still remain elusive in spite of the high disease burden of the virus, especially in pre-term infants and immunocompromised individuals. While vaccine efforts have focused on vaccine-induced antibodies to neutralize the virus, studies have increasingly shown the importance of other antibody functions in protection against cytomegalovirus (CMV) transmission. In this study, we comprehensively evaluated immune responses elicited by the prefusion state of an important HCMV protein called glycoprotein B (gB) in mice. Our results indicate that prefusion gB elicits immune responses similar to that of postfusion gB in mice and reveals areas for further redesign and testing for prefusion vaccine antigens against CMV and other herpesviruses, which could help in furthering vaccine development against HCMV.
Collapse
Affiliation(s)
| | - Megan Connors
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Christian R Binuya
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Mackensie Gross
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Adelaide S Fuller
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Chelsea M Crooks
- 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
| | - Madeline R Sponholtz
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Patrick O Byrne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - Linda M Gerber
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - John D Campbell
- Dynavax Technologies Corporation, Emeryville, California, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Elizabeth Mitchell
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Lara van der Maas
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Itzayana Miller
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Dong Yu
- Dynavax Technologies Corporation, Emeryville, California, USA
| | | | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
2
|
Nielsen-Saines K, Kalbasi-Romero T, Duarte ACM, Almeida da Silva S, Adachi K, Damasceno L, Kerin T, Fuller T, Deville JG, Moreira ME, Vasconcelos Z, Zin A, Shin-Sim M, Barbosa de Lima SM, Brasil P. Development of Maternal Antibodies Post ZIKV in Pregnancy is Associated with Lower Risk of Microcephaly and Structural Brain Abnormalities in Exposed Infants. J Infect Dis 2025:jiaf146. [PMID: 40257773 DOI: 10.1093/infdis/jiaf146] [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: 09/23/2024] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND We investigated the association between maternal neutralizing antibodies (nAb) to Zika virus (ZIKV) in pregnancy and neonatal outcomes. METHODS In pregnant participants with confirmed ZIKV infection, we determined trimester of infection, collected sera longitudinally, and measured nAbs via plaque reduction. In neonates, adverse outcomes included microcephaly (MC), structural brain abnormalities (SBA), hearing, and eye abnormalities. Associations between trimester of infection, nAbs, and neonatal outcomes were analyzed with Cox regression. RESULTS In total, 137 ZIKV-positive pregnant participants had neutralization assays performed during pregnancy and postdelivery. Infection rates were 29% in the first, 50% in the second, and 21% in the third trimester. Mean ZIKV nAb titer >2 weeks postinfection was 64 258 (SD 213 288). Ten percent of 90% plaque reduction neutralization assays (PRNT90) titers were <500, 10% 500-1000, 73% > 1000, and 7% did not have serologic follow-up; 15%. of infants had adverse findings. Protective factors against MC in 88 mothers with nAb titers available during pregnancy included infection later in gestation (adjusted hazard ratio [aHR], 0.06; P = .036) and adequate nAb titers (aHR, 0.17; P = .014). No SBA was associated with later infection in pregnancy (aHR, 0.16; P = .017) and adequate nAb titers (aHR, 0.34; P = .012). Adjusting for trimester, higher maternal nAb titers were associated with lower risk of MC and SBA. Seven of 137 participants (5.1%) had positive serum ZIKV polymerase chain reaction (PCR) results beyond 14 days (range, 35-269 days). Participants with ZIKV PCR positivity >60 days (n = 2) had infants with MC/SBA. CONCLUSIONS MC and SBA were less frequent in infants of mothers with higher ZIKV nAb titers during pregnancy.
Collapse
Affiliation(s)
- Karin Nielsen-Saines
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Tahmineh Kalbasi-Romero
- Division of General Internal Medicine and Health Services Research, University of California Los Angeles, Los Angeles, California, USA
| | | | | | - Kristina Adachi
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Luana Damasceno
- Acute Febrile Illness Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Tara Kerin
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Trevon Fuller
- Acute Febrile Illness Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Institute of the Environment and Sustainability, University of California Los Angeles, Los Angeles, California, USA
| | - Jaime G Deville
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Maria Elisabeth Moreira
- Fernandes Figueira National Institute of Women's, Children's, and Adolescents' Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Zilton Vasconcelos
- Fernandes Figueira National Institute of Women's, Children's, and Adolescents' Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Andrea Zin
- Fernandes Figueira National Institute of Women's, Children's, and Adolescents' Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Myung Shin-Sim
- Division of General Internal Medicine and Health Services Research, University of California Los Angeles, Los Angeles, California, USA
| | | | - Patricia Brasil
- Acute Febrile Illness Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Wu Q, Chen K, Xue W, Wang G, Yang Y, Li S, Xia N, Chen Y. An insect cell-derived extracellular vesicle-based gB vaccine elicits robust adaptive immune responses against Epstein-Barr virus. SCIENCE CHINA. LIFE SCIENCES 2025; 68:734-745. [PMID: 39499444 DOI: 10.1007/s11427-023-2599-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/19/2024] [Indexed: 11/07/2024]
Abstract
Epstein-Barr virus (EBV), the first identified human tumor virus, is implicated in various human malignancies, infectious mononucleosis, and more recently, multiple sclerosis. Prophylactic vaccines have the potential to effectively prevent EBV infection. Glycoprotein B (gB) serves as the fusogen and plays a pivotal role in the virus entry process, making it a critical target for EBV vaccine development. Surface membrane proteins of enveloped viruses serve as native conformational antigens, making them susceptible to immune recognition. Utilizing lipid membrane-bound viral antigens is a promising strategy for effective vaccine presentation in this context. In this study, we employed a truncated design for gB proteins, observing that these truncated gB proteins prompted a substantial release of extracellular vesicles (EVs) in insect cells. We verified that EVs exhibited abundant gB proteins, displaying the typical virus particle morphology and extracellular vesicle characteristics. gB EVs demonstrated a more efficient humoral and cellular immune response compared with the gB ectodomain trimer vaccine in mice. Moreover, the antisera induced by the gB EVs vaccine exhibited robust antibody-dependent cytotoxicity. Consequently, gB EVs-based vaccines hold significant potential for preventing EBV infection and offer valuable insights for vaccine design.
Collapse
Affiliation(s)
- Qian Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Kaiyun Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Wenhui Xue
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Guosong Wang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Yanbo Yang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| | - Yixin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
4
|
Miller IG, Mahant AM, Jenks JA, Semmes EC, Rochat E, Herbek SL, Andy C, Rodgers NS, Pollara J, Gerber LM, Herold BC, Permar SR. Influence of Distinct Maternal Cytomegalovirus-Specific Neutralizing and Fc Receptor-Binding Responses on Congenital Cytomegalovirus Transmission in HIV-Exposed Neonates. Viruses 2025; 17:325. [PMID: 40143253 PMCID: PMC11946089 DOI: 10.3390/v17030325] [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: 01/22/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Congenital cytomegalovirus (cCMV) is the most common infectious cause of birth defects worldwide, affecting approximately 1 in every 200 live-born infants globally. Recent work has identified potential immune correlates of protection against cCMV transmission including maternal and placentally transferred antibody levels and their function, which may inform the development of maternal active (vaccine) and passive (mono/polyclonal antibody) immunizations. However, these correlates need to also be assessed in diverse cohorts, including women living with HIV who have increased risk of cCMV transmission. Using a case-control design, we investigated whether the magnitude, specificity, function and placental transfer of maternal IgG responses are associated with protection against and/or risk of cCMV transmission in HIV/HCMV co-infection. Within 3 historical cohorts of pregnant women with HIV/HCMV co-infection, we identified 16 cCMV transmitting cases that were matched to 29 cCMV non-transmitting controls. Using a systems serology approach, we found that normalized HCMV-specific IgG binding to FcγR1α was higher in non-transmitting dyads, whereas HCMV-neutralizing antibody responses were higher in transmitting dyads. These findings suggest that engagement of FcγR1α by HCMV-specific IgG may help confer protection against cCMV transmission. Building upon previous research, our study reinforces the critical role of validating maternal humoral immune correlates of cCMV transmission risk across diverse seropositive cohorts, providing essential insights to inform and accelerate the development of effective HCMV vaccines.
Collapse
Affiliation(s)
- Itzayana G. Miller
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA; (I.G.M.); (S.L.H.)
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Aakash Mahant Mahant
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.M.M.); (B.C.H.)
| | - Jennifer A. Jenks
- Human Vaccine Institute, Duke University, Durham, NC 27710, USA; (J.A.J.); (E.C.S.); (E.R.); (J.P.)
- Medical Scientist Training Program, Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Eleanor C. Semmes
- Human Vaccine Institute, Duke University, Durham, NC 27710, USA; (J.A.J.); (E.C.S.); (E.R.); (J.P.)
- Medical Scientist Training Program, Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Eric Rochat
- Human Vaccine Institute, Duke University, Durham, NC 27710, USA; (J.A.J.); (E.C.S.); (E.R.); (J.P.)
| | - Savannah L. Herbek
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA; (I.G.M.); (S.L.H.)
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA; (C.A.); (L.M.G.)
| | - Nicole S. Rodgers
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Justin Pollara
- Human Vaccine Institute, Duke University, Durham, NC 27710, USA; (J.A.J.); (E.C.S.); (E.R.); (J.P.)
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Linda M. Gerber
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA; (C.A.); (L.M.G.)
| | - Betsy C. Herold
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (A.M.M.); (B.C.H.)
- Department of Pediatrics, Children’s Hospital at Montefiore, Bronx, NY 10461, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA; (I.G.M.); (S.L.H.)
| |
Collapse
|
5
|
Otero CE, Petkova S, Ebermann M, Taher H, John N, Hoffmann K, Davalos A, Moström MJ, Gilbride RM, Papen CR, Barber-Axthelm A, Scheef EA, Barfield R, Sprehe LM, Kendall S, Manuel TD, Beechwood T, Nguyen LK, Vande Burgt NH, Chan C, Denton M, Streblow ZJ, Streblow DN, Tarantal AF, Hansen SG, Kaur A, Permar S, Früh K, Hengel H, Malouli D, Kolb P. Rhesus Cytomegalovirus-encoded Fcγ-binding glycoproteins facilitate viral evasion from IgG-mediated humoral immunity. Nat Commun 2025; 16:1200. [PMID: 39885150 PMCID: PMC11782611 DOI: 10.1038/s41467-025-56419-3] [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: 06/06/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Human cytomegalovirus (HCMV) encodes four viral Fc-gamma receptors (vFcγRs) that counteract antibody-mediated activation in vitro, but their role in infection and pathogenesis is unknown. To examine their in vivo function in an animal model evolutionarily closely related to humans, we identified and characterized Rh05, Rh152/151 and Rh173 as the complete set of vFcγRs encoded by rhesus CMV (RhCMV). Each one of these proteins displays functional similarities to their prospective HCMV orthologs with respect to antagonizing host FcγR activation in vitro. When RhCMV-naïve male rhesus macaques were infected with vFcγR-deleted RhCMV, peak plasma DNAemia levels and anti-RhCMV antibody responses were comparable to wildtype infections of both male and female animals. However, the duration of plasma DNAemia was significantly shortened in immunocompetent, but not in CD4 + T cell-depleted animals. Since vFcγRs were not required for superinfection of rhesus macaques, we conclude that these proteins can prolong lytic replication during primary infection by evading virus-specific adaptive immune responses, particularly antibodies.
Collapse
Affiliation(s)
- Claire E Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Sophia Petkova
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Ebermann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nessy John
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Katja Hoffmann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angel Davalos
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Matilda J Moström
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Courtney R Papen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Elizabeth A Scheef
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Lesli M Sprehe
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Savannah Kendall
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Tabitha D Manuel
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Teresa Beechwood
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Linh Khanh Nguyen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nathan H Vande Burgt
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Zachary J Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, 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, CA, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA.
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
6
|
Permar SR, Schleiss MR, Plotkin SA. A vaccine against cytomegalovirus: how close are we? J Clin Invest 2025; 135:e182317. [PMID: 39744948 DOI: 10.1172/jci182317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
The pursuit of a vaccine against the human cytomegalovirus (HCMV) has been ongoing for more than 50 years. HCMV is the leading infectious cause of birth defects, including damage to the brain, and is a common cause of complications in organ transplantation. The complex biology of HCMV has made vaccine development difficult, but a recent meeting sponsored by the National Institute of Allergy and Infectious Diseases in September of 2023 brought together experts from academia, industry, and federal agencies to discuss progress in the field. The meeting reviewed the status of candidate HCMV vaccines under study and the challenges in clinical trial design in demonstrating efficacy against congenital CMV infection or the reduction of HCMV disease following solid organ transplantation or hematopoietic stem cell transplantation. Discussion in the meeting revealed that, with the numerous candidate vaccines that are under study, it is clear that a safe and effective HCMV vaccine is within reach. Meeting attendees achieved a consensus opinion that even a partially effective vaccine would have a major effect on the global health consequences of HCMV infection.
Collapse
Affiliation(s)
- Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical Center, New York, New York, USA
| | - Mark R Schleiss
- Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Vaxconsult, Doylestown, Pennsylvania, USA
| |
Collapse
|
7
|
Shang Z, Li X. Human cytomegalovirus: pathogenesis, prevention, and treatment. MOLECULAR BIOMEDICINE 2024; 5:61. [PMID: 39585514 PMCID: PMC11589059 DOI: 10.1186/s43556-024-00226-7] [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: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Human cytomegalovirus (HCMV) infection remains a significant global health challenge, particularly for immunocompromised individuals and newborns. This comprehensive review synthesizes current knowledge on HCMV pathogenesis, prevention, and treatment strategies. We examine the molecular mechanisms of HCMV entry, focusing on the structure and function of key envelope glycoproteins (gB, gH/gL/gO, gH/gL/pUL128-131) and their interactions with cellular receptors such as PDGFRα, NRP2, and THBD. The review explores HCMV's sophisticated immune evasion strategies, including interference with pattern recognition receptor signaling, modulation of antigen presentation, and regulation of NK and T cell responses. We highlight recent advancements in developing neutralizing antibodies, various vaccine strategies (live-attenuated, subunit, vector-based, DNA, and mRNA), antiviral compounds (both virus-targeted and host-targeted), and emerging cellular therapies such as TCR-T cell approaches. By integrating insights from structural biology, immunology, and clinical research, we identify critical knowledge gaps and propose future research directions. This analysis aims to stimulate cross-disciplinary collaborations and accelerate the development of more effective prevention and treatment strategies for HCMV infections, addressing a significant unmet medical need.
Collapse
Affiliation(s)
- Zifang Shang
- Research Experiment Center, Meizhou Academy of Medical Sciences, Meizhou People's Hospital, Meizhou, 514031, Guangdong, China.
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, 514031, Guangdong, China.
| | - Xin Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| |
Collapse
|
8
|
Slein MD, Backes IM, Kelkar NS, Garland CR, Khanwalkar US, Sholukh AM, Johnston CM, Leib DA, Ackerman ME. Improving antibody-mediated protection against HSV infection by eliminating interactions with the viral Fc receptor gE/gI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624598. [PMID: 39605495 PMCID: PMC11601663 DOI: 10.1101/2024.11.20.624598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Herpes simplex virus (HSV) encodes surface glycoproteins that are host defense evasion molecules, allowing the virus to escape immune clearance. In addition to their role in neuropathogenesis and cell-cell spread, glycoproteins E and I (gE/gI) form a viral Fc receptor (vFcR) for most subclasses and allotypes of human IgG and promote evasion of humoral immune responses. While monoclonal antibodies (mAbs) protect mice from neonatal HSV (nHSV) infections, the impact of the vFcR on mAb-mediated protection by binding to IgG is unknown. Using HSV-1 with intact and ablated gE-mediated IgG Fc binding, and Fc-engineered antibodies with modified ability to interact with gE/gI, we investigated the role of the vFcR in viral pathogenesis and mAb-mediated protection from nHSV. The gD-specific human mAb HSV8 modified to lack binding to gE exhibited enhanced neutralization and in vivo protection compared to its native IgG1 form. This improved protection by the engineered mAbs was dependent on the presence of the vFcR. Human IgG3 allotypes lacking vFcR binding also exhibited enhanced antiviral activity in vivo, suggesting that vaccines that robustly induce IgG3 responses could show enhanced protection. suggesting the value of vaccination strategies that robustly induce this subclass. Lastly, analysis of longitudinal responses to acute primary genital infection in humans raised the possibility that unlike most viruses, HSV may exhibited slow induction of IgG3. In summary, this study demonstrates that mAbs lacking the ability to interact with the vFcR can exhibit improved protection from HSV-offering new prospects for antibody-based interventions.
Collapse
Affiliation(s)
- Matthew D. Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Iara M. Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | | | - Christine M. Johnston
- Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Departments of Medicine and Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104, USA
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
9
|
Semmes EC, Nettere DR, Nelson AN, Hurst JH, Cain DW, Burt TD, Kurtzberg J, Reeves RK, Coyne CB, Fouda GG, Pollara J, Permar SR, Walsh KM. In utero human cytomegalovirus infection expands NK-like FcγRIII+CD8+ T cells that mediate Fc antibody functions. J Clin Invest 2024; 135:e181342. [PMID: 39531313 PMCID: PMC11684805 DOI: 10.1172/jci181342] [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: 03/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Human cytomegalovirus (HCMV) profoundly impacts host T and NK cells across the lifespan, yet how this common congenital infection modulates developing fetal immune cell compartments remains underexplored. Using cord blood from neonates with and without congenital HCMV (cCMV) infection, we identify an expansion of Fcγ receptor III-expressing (FcγRIII-expressing) CD8+ T cells following HCMV exposure in utero. Most FcγRIII+CD8+ T cells express the canonical αβ T cell receptor (TCR), but a proportion express noncanonical γδ TCR. FcγRIII+CD8+ T cells are highly differentiated and have increased expression of NK cell markers and cytolytic molecules. Transcriptional analysis reveals FcγRIII+CD8+ T cells upregulate T-bet and downregulate BCL11B, known transcription factors that govern T/NK cell fate. We show that FcγRIII+CD8+ T cells mediate antibody-dependent IFN-γ production and degranulation against IgG-opsonized target cells, similar to NK cell antibody-dependent cellular cytotoxicity (ADCC). FcγRIII+CD8+ T cell Fc effector functions were further enhanced by IL-15, as has been observed in neonatal NK cells. Our study reveals that FcγRIII+CD8+ T cells elicited in utero by HCMV infection can execute Fc-mediated effector functions bridging cellular and humoral immunity and may be a promising target for antibody-based therapeutics and vaccination in early life.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Boston Children’s Hospital/Boston Medical Center, Boston, Massachusetts, USA
- Medical Scientist Training Program, and
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Danielle R. Nettere
- Medical Scientist Training Program, and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ashley N. Nelson
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Jillian H. Hurst
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
| | - Derek W. Cain
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Trevor D. Burt
- Children’s Health and Discovery Initiative
- Division of Neonatology, Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Children’s Health and Discovery Initiative
- Carolinas Cord Blood Bank, Marcus Center for Cellular Cures, Durham, North Carolina, USA
| | - R. Keith Reeves
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Human Systems Immunology, and
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Carolyn B. Coyne
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Children’s Health and Discovery Initiative
- Division of Infectious Diseases, and
- Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Kyle M. Walsh
- Children’s Health and Discovery Initiative
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| |
Collapse
|
10
|
Mackin SR, Sariol A, Diamond MS. Antibody-mediated control mechanisms of viral infections. Immunol Rev 2024; 328:205-220. [PMID: 39162394 PMCID: PMC11661935 DOI: 10.1111/imr.13383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Antibodies generated after vaccination or natural pathogen exposure are essential mediators of protection against many infections. Most studies with viruses have focused on antibody neutralization, in which protection is conferred by the fragment antigen binding region (Fab) through targeting of different steps in the viral lifecycle including attachment, internalization, fusion, and egress. Beyond neutralization, the fragment crystallizable (Fc) region of antibodies can integrate innate and adaptive immune responses by engaging complement components and distinct Fc gamma receptors (FcγR) on different host immune cells. In this review, we discuss recent advances in our understanding of antibody neutralization and Fc effector functions, and the assays used to measure them. Additionally, we describe the contexts in which these mechanisms are associated with protection against viruses and highlight how Fc-FcγR interactions can improve the potency of antibody-based therapies.
Collapse
Affiliation(s)
- Samantha R. Mackin
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Andrew M. and Jane M. Bursky the Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
11
|
Schleiss MR, Crooks CM, Karthigeyan KP, Kruc RM, Otero CE, Wang HY(S, Permar SR, Plotkin SA, Gautam R. Proceedings of the Conference "CMV Vaccine Development-How Close Are We?" (27-28 September 2023). Vaccines (Basel) 2024; 12:1231. [PMID: 39591134 PMCID: PMC11598149 DOI: 10.3390/vaccines12111231] [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: 07/23/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 11/28/2024] Open
Abstract
Congenital cytomegalovirus (cCMV) is the most common infectious cause of disability in children, including sensorineural hearing loss. There is interest in developing a pre-conception vaccine that could confer protective immunity on a woman of child-bearing age, hence resulting in a reduced cCMV disease burden. Other populations, including solid organ transplant (SOT) and hematopoietic stem cell transplant (HSCT) patients, could also benefit from CMV vaccination. To review and discuss vaccines that are in clinical development, a workshop, sponsored by the National Institutes of Health (NIH) and the National Institute of Allergy and Infectious Diseases (NIAID), was empaneled. At this workshop, correlates of protective immunity against CMV, epidemiologic features of CMV transmission, and vaccine platforms in development were reviewed. Representatives from academia, pharma, and the NIH engaged in discussion on the current state-of-the-art in CMV vaccinology. A summary of the presentations from this is provided in this report.
Collapse
Affiliation(s)
- Mark R. Schleiss
- Division of Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Chelsea M. Crooks
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Krithika P. Karthigeyan
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Rebecca M. Kruc
- Department of Pediatrics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Claire E. Otero
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Hsuan-Yuan (Sherry) Wang
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, 1300 York Ave Box 65, New York, NY 10065, USA;
| | - Stanley A. Plotkin
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Rajeev Gautam
- Program Officer at Virology Branch, Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fisher’s Lane, Rockville, MD 20892, USA;
| |
Collapse
|
12
|
Hederman AP, Remmel CA, Sharma S, Natarajan H, Weiner JA, Wrapp D, Donner C, Delforge ML, d’Angelo P, Furione M, Fornara C, McLellan JS, Lilleri D, Marchant A, Ackerman ME. Discrimination of primary and chronic cytomegalovirus infection based on humoral immune profiles in pregnancy. J Clin Invest 2024; 134:e180560. [PMID: 39207860 PMCID: PMC11473158 DOI: 10.1172/jci180560] [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: 02/23/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUNDMost humans have been infected with cytomegalovirus (CMV) by midlife without clinical signs of disease. However, in settings in which the immune system is undeveloped or compromised, the virus is not adequately controlled and consequently presents a major infectious cause of both congenital disease during pregnancy as well as opportunistic infection in children and adults. With clear evidence that risk to the fetus varies with gestational age at the time of primary maternal infection, further research on humoral responses to primary CMV infection during pregnancy is needed.METHODSHere, systems serology tools were applied to characterize antibody responses to CMV infection in pregnant and nonpregnant women experiencing either primary or chronic infection.RESULTSWhereas strikingly different antibody profiles were observed depending on infection status, limited differences were associated with pregnancy status. Beyond known differences in IgM responses used clinically for identification of primary infection, distinctions observed in IgA and FcγR-binding antibodies and among antigen specificities accurately predicted infection status. Machine learning was used to define the transition from primary to chronic states and predict time since infection with high accuracy. Humoral responses diverged over time in an antigen-specific manner, with IgG3 responses toward tegument decreasing over time as typical of viral infections, while those directed to pentamer and glycoprotein B were lower during acute and greatest during chronic infection.CONCLUSIONIn sum, this work provides insights into the antibody response associated with CMV infection status in the context of pregnancy, revealing aspects of humoral immunity that have the potential to improve CMV diagnostics.FUNDINGCYMAF consortium and NIH NIAID.
Collapse
Affiliation(s)
- Andrew P. Hederman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | | | - Shilpee Sharma
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire, USA
| | - Joshua A. Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas, Austin, Texas, USA
| | - Catherine Donner
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), CUB Hôpital Erasme, Department of Obstetrics and Gynecology, Brussels, Belgium
| | - Marie-Luce Delforge
- ULB, H.U.B., CUB Hôpital Erasme, National Reference Center for Congenital Infections, Brussels, Belgium
| | - Piera d’Angelo
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Milena Furione
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Fornara
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas, Austin, Texas, USA
| | - Daniele Lilleri
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire, USA
| |
Collapse
|
13
|
Sponholtz MR, Byrne PO, Lee AG, Ramamohan AR, Goldsmith JA, McCool RS, Zhou L, Johnson NV, Hsieh CL, Connors M, Karthigeyan KP, Crooks CM, Fuller AS, Campbell JD, Permar SR, Maynard JA, Yu D, Bottomley MJ, McLellan JS. Structure-based design of a soluble human cytomegalovirus glycoprotein B antigen stabilized in a prefusion-like conformation. Proc Natl Acad Sci U S A 2024; 121:e2404250121. [PMID: 39231203 PMCID: PMC11406251 DOI: 10.1073/pnas.2404250121] [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: 02/28/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Human cytomegalovirus (HCMV) glycoprotein B (gB) is a class III membrane fusion protein required for viral entry. HCMV vaccine candidates containing gB have demonstrated moderate clinical efficacy, but no HCMV vaccine has been approved. Here, we used structure-based design to identify and characterize amino acid substitutions that stabilize gB in its metastable prefusion conformation. One variant containing two engineered interprotomer disulfide bonds and two cavity-filling substitutions (gB-C7), displayed increased expression and thermostability. A 2.8 Å resolution cryoelectron microscopy structure shows that gB-C7 adopts a prefusion-like conformation, revealing additional structural elements at the membrane-distal apex. Unlike previous observations for several class I viral fusion proteins, mice immunized with postfusion or prefusion-stabilized forms of soluble gB protein displayed similar neutralizing antibody titers, here specifically against an HCMV laboratory strain on fibroblasts. Collectively, these results identify initial strategies to stabilize class III viral fusion proteins and provide tools to probe gB-directed antibody responses.
Collapse
Affiliation(s)
- Madeline R. Sponholtz
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Patrick O. Byrne
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Alison G. Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ajit R. Ramamohan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Jory A. Goldsmith
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ryan S. McCool
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ling Zhou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Nicole V. Johnson
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ching-Lin Hsieh
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Megan Connors
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Krithika P. Karthigeyan
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Chelsea M. Crooks
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Adelaide S. Fuller
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | | | - Sallie R. Permar
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Jennifer A. Maynard
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX78712
| | - Dong Yu
- Dynavax Technologies Corporation, Emeryville, CA94608
| | | | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
14
|
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
|
15
|
d’Angelo P, Zavaglio F, Gabanti E, Zelini P, Fornara C, Bernuzzi S, Spinillo A, Lilleri D, Baldanti F. Evaluation of the In Vitro Capacity of Anti-Human Cytomegalovirus Antibodies to Initiate Antibody-Dependent Cell Cytotoxicity. Microorganisms 2024; 12:1355. [PMID: 39065122 PMCID: PMC11278886 DOI: 10.3390/microorganisms12071355] [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: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
In the setting of infectious diseases, antibodies show different functions beyond neutralizing activity. In this study, we investigated the activation of NK cells in vitro in the presence of human cytomegalovirus (HCMV)-specific antibodies and their potential role in the control of HCMV infection through antibody-dependent cell cytotoxicity (ADCC). Retinal pigmented epithelial cells (ARPE-19) infected with the HCMV strain VR1814 were co-cultured with cytokine-activated peripheral blood mononuclear cells (PBMCs) in the presence of sera collected from 23 HCMV-seropositive and 9 HCMV-seronegative donors. Moreover, 13 pregnant women sampled 3 and 6 months after HCMV primary infection and 13 pregnant women with pre-conception immunity were tested and compared. We determined the percentage of activated NK cells via the analysis of CD107a expression as a marker of degranulation. Significantly higher levels of NK-cell activation were observed using 1/100 and 1/10 dilutions of sera from HCMV-seropositive individuals, and when cells were infected for 96 and 120 h, suggesting that NK cells are activated by antibodies directed against late antigens. In the absence of serum NK cells, activation was negligible. In seropositive subjects, the median percentages of CD107a-positive NK cells in the presence of autologous serum and pooled HCMV-positive serum were similar (14.03% [range 0.00-33.56] and 12.42% [range 1.01-46.00], respectively), while NK-cell activation was negligible using an HCMV-negative serum pool. In HCMV-seronegative subjects, the median percentage of activated NK cells was 0.90% [range 0.00-3.92] with autologous serum and 2.07% [0.00-5.76] in the presence of the HCMV-negative serum pool, while it was 8.97% [0.00-26.49] with the pool of HCMV-positive sera. NK-cell activation using hyperimmune globulin is comparable to what is obtained using autologous serum. Sera from subjects at 3 and 6 months post primary infection showed a lower capacity of NK-cell activation than sera from subjects with past infection (p < 0.001). NK activation against HCMV-infected epithelial cells is dependent on the presence of HCMV-specific antibodies. This serum activity increases with time after the onset of HCMV infection. The protective role of NK-cell activation by HCMV-specific serum antibodies should be verified in clinical settings.
Collapse
Affiliation(s)
- Piera d’Angelo
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Federica Zavaglio
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Elisa Gabanti
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- M.A.D. Analisi, 27058 Voghera, Italy
| | - Paola Zelini
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chiara Fornara
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Laboratory Medicine Service, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Stefano Bernuzzi
- Service of Immunohematology and Transfusion Medicine, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Arsenio Spinillo
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation, 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, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
16
|
Pighi C, Rotili A, De Luca M, Chiurchiù S, Calò Carducci FI, Rossetti C, Cifaldi L, Bei R, Caforio L, Bernardi S, Palma P, Amodio D. Characterization of Natural Killer Cell Profile in a Cohort of Infected Pregnant Women and Their Babies and Its Relation to CMV Transmission. Viruses 2024; 16:780. [PMID: 38793661 PMCID: PMC11125694 DOI: 10.3390/v16050780] [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: 03/26/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Human cytomegalovirus (CMV) is a common herpesvirus causing lifelong latent infection in most people and is a primary cause of congenital infection worldwide. Given the role of NK cells in the materno-fetal barrier, we investigated peripheral blood NK cell behavior in the context of CMV infection acquired during pregnancy. We analyzed the NK phenotype and CD107a surface mobilization on PBMCs from CMV-transmitting and non-transmitting mothers and newborns with or without congenital infection. NK cells from non-transmitting mothers showed the typical phenotype of CMV-adaptive NK cells, characterized by higher levels of NKG2C, CD57, and KIRs, with reduced NKG2A, compared to transmitting ones. A significantly higher percentage of DNAM-1+, PD-1+, and KIR+NKG2A-CD57+PD-1+ CD56dim cells was found in the non-transmitting group. Accordingly, NK cells from congenital-CMV (cCMV)-infected newborns expressed higher levels of NKG2C and CD57, with reduced NKG2A, compared to non-congenital ones. Furthermore, they showed a significant expansion of CD56dim cells co-expressing NKG2C and CD57 or with a memory-like (KIR+NKG2A-CD57+NKG2C+) phenotype, as well as a significant reduction of the CD57-NKG2C- population. Degranulation assays showed a slightly higher CD107a geomean ratio in NK cells of mothers who were non-transmitting compared to those transmitting the virus. Our findings demonstrate that both CMV-transmitting mothers and cCMV newborns show a specific NK profile. These data can guide studies on predicting virus transmission from mothers and congenital infection in infants.
Collapse
Affiliation(s)
- Chiara Pighi
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.P.); (A.R.); (C.R.); (P.P.)
| | - Arianna Rotili
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.P.); (A.R.); (C.R.); (P.P.)
- PhD Program in “Immunology, Molecular Medicine and Applied Biotechnologies”, Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Maia De Luca
- Infectious Disease Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.D.L.); (S.C.); (F.I.C.C.); (S.B.)
| | - Sara Chiurchiù
- Infectious Disease Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.D.L.); (S.C.); (F.I.C.C.); (S.B.)
| | | | - Chiara Rossetti
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.P.); (A.R.); (C.R.); (P.P.)
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (L.C.); (R.B.)
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (L.C.); (R.B.)
| | - Leonardo Caforio
- Fetal Medicine and Surgery Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Stefania Bernardi
- Infectious Disease Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.D.L.); (S.C.); (F.I.C.C.); (S.B.)
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.P.); (A.R.); (C.R.); (P.P.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Donato Amodio
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (C.P.); (A.R.); (C.R.); (P.P.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| |
Collapse
|
17
|
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
|
18
|
Mader K, Dustin LB. Beyond bNAbs: Uses, Risks, and Opportunities for Therapeutic Application of Non-Neutralising Antibodies in Viral Infection. Antibodies (Basel) 2024; 13:28. [PMID: 38651408 PMCID: PMC11036282 DOI: 10.3390/antib13020028] [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: 03/07/2024] [Revised: 03/27/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024] Open
Abstract
The vast majority of antibodies generated against a virus will be non-neutralising. However, this does not denote an absence of protective capacity. Yet, within the field, there is typically a large focus on antibodies capable of directly blocking infection (neutralising antibodies, NAbs) of either specific viral strains or multiple viral strains (broadly-neutralising antibodies, bNAbs). More recently, a focus on non-neutralising antibodies (nNAbs), or neutralisation-independent effects of NAbs, has emerged. These can have additive effects on protection or, in some cases, be a major correlate of protection. As their name suggests, nNAbs do not directly neutralise infection but instead, through their Fc domains, may mediate interaction with other immune effectors to induce clearance of viral particles or virally infected cells. nNAbs may also interrupt viral replication within infected cells. Developing technologies of antibody modification and functionalisation may lead to innovative biologics that harness the activities of nNAbs for antiviral prophylaxis and therapeutics. In this review, we discuss specific examples of nNAb actions in viral infections where they have known importance. We also discuss the potential detrimental effects of such responses. Finally, we explore new technologies for nNAb functionalisation to increase efficacy or introduce favourable characteristics for their therapeutic applications.
Collapse
Affiliation(s)
| | - Lynn B. Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, UK;
| |
Collapse
|
19
|
Gao F, Mora MC, Constantinides M, Coenon L, Multrier C, Vaillant L, Zhang T, Villalba M. g-NK cells from umbilical cord blood are phenotypically and functionally different than g-NK cells from peripheral blood. Oncoimmunology 2023; 12:2283353. [PMID: 38126036 PMCID: PMC10732642 DOI: 10.1080/2162402x.2023.2283353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023] Open
Abstract
FcRγ-deficient natural killer (NK) cells, designated as g-NK cells, exhibit enhanced antibody-dependent cellular cytotoxicity (ADCC) capacity and increased IFN-γ and TNF-α production, rendering them promising for antiviral and antitumor responses. g-NK cells from peripheral blood (PB) are often associated with prior human cytomegalovirus (HCMV) infection. However, the prevalence, phenotype, and function of g-NK cells in umbilical cord blood (UCB-g-NK) remain unclear. Here, we demonstrate significant phenotypical differences between UCB-g-NK and PB-g-NK cells. Unlike PB-g-NK cells, UCB-g-NK cells did not show heightened cytokine production upon CD16 engagement, in contrast to the conventional NK (c-NK) cell counterparts. Interestingly, following in vitro activation, UCB-g-NK cells also exhibited elevated levels of IFN-γ production, particularly when co-cultured with HCMV and plasma from g-NK+ adults. Furthermore, g-NK+ plasma from PB even facilitated the in vitro expansion of UCB-g-NK cells. These findings underscore the phenotypic and functional heterogeneity of g-NK cells based on their origin and demonstrate that components within g-NK+ plasma may directly contribute to the acquisition of an adult phenotype by the "immature" UCB-g-NK cells.
Collapse
Affiliation(s)
- Fei Gao
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | | | | | - Loïs Coenon
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | - Caroline Multrier
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | - Loïc Vaillant
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | - Tianxiang Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Martin Villalba
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
- Institut du Cancer Avignon-Provence Sainte Catherine, Avignon, France
- IRMB, INSERM, CHRU de Montpellier, CNRS, Univ Montpellier, Montpellier, France
| |
Collapse
|
20
|
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
|