1
|
He L, Hertel L, James CD, Morgan IM, Klingelhutz AJ, Fu TM, Kauvar LM, McVoy MA. Inhibition of human cytomegalovirus entry into mucosal epithelial cells. Antiviral Res 2024; 230:105971. [PMID: 39074588 PMCID: PMC11408113 DOI: 10.1016/j.antiviral.2024.105971] [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: 04/01/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Human cytomegalovirus (CMV) causes serious developmental disabilities in newborns infected in utero following oral acquisition by the mother. Thus, neutralizing antibodies in maternal saliva have potential to prevent maternal infection and, consequently, fetal transmission and disease. Based on standard cell culture models, CMV entry mediators (and hence neutralizing targets) are cell type-dependent: entry into fibroblasts requires glycoprotein B (gB) and a trimeric complex (TC) of glycoproteins H, L, and O, whereas endothelial and epithelial cell entry additionally requires a pentameric complex (PC) of glycoproteins H and L with UL128, UL130, and UL131A. However, as the mediators of mucosal cell entry and the potential impact of cellular differentiation remained unclear, the present studies utilized mutant viruses, neutralizing antibodies, and soluble TC-receptor to determine the entry mediators required for infection of mucocutaneus cell lines and primary tonsil epithelial cells. Entry into undifferentiated cells was largely PC-dependent, but PC-independent entry could be induced by differentiation. TC-independent entry was also observed and varied by cell line and differentiation. Infection of primary tonsil cells from some donors was entirely TC-independent. In contrast, an antibody to gB or disruption of virion attachment using heparin blocked entry into all cells. These findings indicate that CMV entry into the spectrum of cell types encountered in vivo is likely to be more complex than has been suggested by standard cell culture models and may be influenced by the relative abundance of virion envelope glycoprotein complexes as well as by cell type, tissue of origin, and state of differentiation.
Collapse
Affiliation(s)
- Li He
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Laura Hertel
- Department of Pediatrics, School of Medicine, University of California San Francisco, Oakland, CA, 94609, USA
| | - Claire D James
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Iain M Morgan
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, 52242, USA
| | - Tong-Ming Fu
- Texas Therapeutics Institute, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | | | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
2
|
Pesch MH, Mowers J, Huynh A, Schleiss MR. Intrauterine Fetal Demise, Spontaneous Abortion and Congenital Cytomegalovirus: A Systematic Review of the Incidence and Histopathologic Features. Viruses 2024; 16:1552. [PMID: 39459885 PMCID: PMC11512218 DOI: 10.3390/v16101552] [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: 06/30/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The objective was to review the existing literature reporting on spontaneous abortion (SA) and intrauterine fetal demise (IUFD) associated with cytomegalovirus (CMV) infection. A review using standardized terminology such as 'intrauterine fetal death', 'congenital cytomegalovirus' and 'CMV' was performed using PubMed and Embase (Medline) using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) methodology. Twenty-one studies met inclusion criteria. CMV was identified as a potential or likely factor in a median of 7.1% of SA or IUFD in study cohorts. Of the studies, 11 used fetal remains, 18 used placenta, 6 used serum, and 1 used post-mortem dried blood spot as specimens for testing for CMV. Features commonly observed were fetal thrombotic vasculopathy, hydrops fetalis and chronic villitis. CMV is frequently noted in studies evaluating viral etiologies of SA or IUFD. Large population-based studies are needed to estimate the incidence of CMV-associated SA or IUFD. CMV and congenital CMV should be included on the differential diagnosis in all cases of SA or IUFD of unknown etiology.
Collapse
Affiliation(s)
- Megan H. Pesch
- Division of Developmental and Behavioral Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan Mowers
- Division of Pathology, Ascension Hospital Providence, Southfield, MI 48075, USA;
| | - Anh Huynh
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Mark R. Schleiss
- Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
3
|
Rollman TB, Berkebile ZW, Okae H, Bardwell VJ, Gearhart MD, Bierle CJ. Human trophoblast stem cells restrict human cytomegalovirus replication. J Virol 2024; 98:e0193523. [PMID: 38451085 PMCID: PMC11019952 DOI: 10.1128/jvi.01935-23] [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: 12/11/2023] [Accepted: 02/18/2024] [Indexed: 03/08/2024] Open
Abstract
Placental infection plays a central role in the pathogenesis of congenital human cytomegalovirus (HCMV) infections and is a cause of fetal growth restriction and pregnancy loss. HCMV can replicate in some trophoblast cell types, but it remains unclear how the virus evades antiviral immunity in the placenta and how infection compromises placental development and function. Human trophoblast stem cells (TSCs) can be differentiated into extravillous trophoblasts (EVTs), syncytiotrophoblasts (STBs), and organoids, and this study assessed the utility of TSCs as a model of HCMV infection in the first-trimester placenta. HCMV was found to non-productively infect TSCs, EVTs, and STBs. Immunofluorescence assays and flow cytometry experiments further revealed that infected TSCs frequently only express immediate early viral gene products. Similarly, RNA sequencing found that viral gene expression in TSCs does not follow the kinetic patterns observed during lytic infection in fibroblasts. Canonical antiviral responses were largely not observed in HCMV-infected TSCs and TSC-derived trophoblasts. Rather, infection dysregulated factors involved in cell identity, differentiation, and Wingless/Integrated signaling. Thus, while HCMV does not replicate in TSCs, infection may perturb trophoblast differentiation in ways that could interfere with placental function. IMPORTANCE Placental infection plays a central role in human cytomegalovirus (HCMV) pathogenesis during pregnancy, but the species specificity of HCMV and the limited availability and lifespan of primary trophoblasts have been persistent barriers to understanding how infection impacts this vital organ. Human trophoblast stem cells (TSCs) represent a new approach to modeling viral infection early in placental development. This study reveals that TSCs, like other stem cell types, restrict HCMV replication. However, infection perturbs the expression of genes involved in differentiation and cell fate determination, pointing to a mechanism by which HCMV could cause placental injury.
Collapse
Affiliation(s)
- Tyler B. Rollman
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zachary W. Berkebile
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hiroaki Okae
- Department of Informative Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Vivian J. Bardwell
- Developmental Biology Center, Department of Genetics, Cell Biology and Development and the Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Micah D. Gearhart
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Craig J. Bierle
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
4
|
Langley JM, Gantt S, Halperin SA, Ward B, McNeil S, Ye L, Cai Y, Smith B, Anderson DE, Mitoma FD. An enveloped virus-like particle alum-adjuvanted cytomegalovirus vaccine is safe and immunogenic: A first-in-humans Canadian Immunization Research Network (CIRN) study. Vaccine 2024; 42:713-722. [PMID: 38142214 DOI: 10.1016/j.vaccine.2023.12.019] [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: 08/14/2023] [Revised: 11/06/2023] [Accepted: 12/03/2023] [Indexed: 12/25/2023]
Abstract
INTRODUCTION Cytomegalovirus (CMV) is the most common cause of congenital infection and affected children often have permanent neurodevelopmental sequelae, including hearing loss and intellectual disability. Vaccines to prevent transmission of CMV during pregnancy are a public health priority. This first-in-humans dose-ranging, randomized, placebo-controlled, observer-blinded study evaluated the safety and immunogenicity of an enveloped virus-like particle (eVLP) vaccine expressing a modified form of the CMV glycoprotein B (gB). METHODS Healthy CMV-seronegative 18 to 40-year-olds at 3 Canadian study sites were randomized to one of 4 dose formulations (0.5 µg, 1 µg, or 2 µg gB content with alum) or 1 µg gB without alum, or placebo, given intramuscularly on days 0, 56 and 168. Outcome measures were solicited and unsolicited adverse events (AE), severe AE, gB and AD-2 epitope binding antibody titers and avidity, and neutralizing antibody (nAb) titers to CMV measured in fibroblast and epithelial cell infection assays. RESULTS Among 125 participants, the most common solicited local and general AEs were pain and headache, respectively. A dose-dependent increase in gB binding, avidity and nAb titers was observed after doses 2 and 3, with the highest titers in the alum-adjuvanted 2.0 µg dose recipients after the third dose; in the latter 24 % had responses to the broadly neutralizing AD-2 epitope. Neutralizing activity to CMV infection of fibroblasts was seen in 100 % of 2.0 µg alum-adjuvanted dose recipients, and to epithelial cell infection in 31 %. Epithelial cell nAb titers were positively correlated with higher geometric mean CMV gB binding titers. CONCLUSIONS An eVLP CMV vaccine was immunogenic in healthy CMV-seronegative adults and no safety signals were seen. Alum adjuvantation increased immunogenicity as did higher antigen content and a three dose schedule. This phase 1 trial supports further development of this eVLP CMV vaccine candidate.
Collapse
Affiliation(s)
- Joanne M Langley
- Canadian Center for Vaccinology, (Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority), Canada; Departments of Pediatrics, Dalhousie University, Nova Scotia, Canada; Community Health and Epidemiology, Dalhousie University, Nova Scotia, Canada.
| | - Soren Gantt
- CHU Sainte-Justine Research Centre and the Departments of Microbiology and Pediatrics, University of Montreal (formerly at the Vaccine Evaluation Center, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver BC at the time of the study), Canada
| | - Scott A Halperin
- Canadian Center for Vaccinology, (Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority), Canada; Departments of Pediatrics, Dalhousie University, Nova Scotia, Canada; Microbiology and Immunology, Dalhousie University, Nova Scotia, Canada
| | - Brian Ward
- McGill University Health Centre Vaccine Study Centre, Montreal, PQ, Canada
| | - Shelly McNeil
- Canadian Center for Vaccinology, (Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority), Canada; Departments of Pediatrics, Dalhousie University, Nova Scotia, Canada; Community Health and Epidemiology, Dalhousie University, Nova Scotia, Canada; Department of Medicine, Dalhousie University, Nova Scotia, Canada
| | - Lingyun Ye
- Canadian Center for Vaccinology, (Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority), Canada
| | - Yun Cai
- Canadian Center for Vaccinology, (Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority), Canada
| | - Bruce Smith
- Canadian Center for Vaccinology, (Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority), Canada
| | | | | |
Collapse
|
5
|
Rollman TB, Berkebile ZW, Okae H, Bardwell VJ, Gearhart MD, Bierle CJ. Human Trophoblast Stem Cells Restrict Human Cytomegalovirus Replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571456. [PMID: 38168202 PMCID: PMC10760179 DOI: 10.1101/2023.12.13.571456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Placental infection plays a central role in the pathogenesis of congenital human cytomegalovirus (HCMV) infections and is a cause of fetal growth restriction and pregnancy loss. HCMV can replicate in some trophoblast cell types, but it remains unclear how the virus evades antiviral immunity in the placenta and how infection compromises placental development and function. Human trophoblast stem cells (TSCs) can be differentiated into extravillous trophoblasts (EVTs), syncytiotrophoblasts (STBs), and organoids, and this study assessed the utility of TSCs as a model of HCMV infection in the first trimester placenta. HCMV was found to non-productively infect TSCs, EVTs, and STBs. Immunofluorescence assays and flow cytometry experiments further revealed that infected TSCs frequently only express immediate early viral gene products. Similarly, RNA-sequencing found that viral gene expression in TSCs does not follow the kinetic patterns observed during lytic infection in fibroblasts. Canonical antiviral responses were largely not observed in HCMV-infected TSCs and TSC-derived trophoblasts. Rather, infection dysregulated factors involved in cell identity, differentiation, and WNT signaling. Thus, while HCMV does not replicate in TSCs, infection may perturb trophoblast differentiation in ways that could interfere with placental function. Importance Placental infection plays a central role in HCMV pathogenesis during pregnancy, but the species-specificity of HCMV and the limited availability and lifespan of primary trophoblasts have been persistent barriers to understanding how infection impacts this vital organ. Human TSCs represent a new approach to modeling viral infection early in placental development. This study reveals that TSCs, like other stem cell types, restrict HCMV replication. However, infection perturbs the expression of genes involved in differentiation and cell fate determination, pointing to a mechanism by which HCMV could cause placental injury.
Collapse
|
6
|
Okamoto M, Kurino R, Miura R, Takada K. A fully human neutralizing monoclonal antibody targeting a highly conserved epitope of the human cytomegalovirus glycoprotein B. PLoS One 2023; 18:e0285672. [PMID: 37192198 DOI: 10.1371/journal.pone.0285672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/27/2023] [Indexed: 05/18/2023] Open
Abstract
Human cytomegalovirus causes severe diseases in children (by congenital infection) and immunocompromised patients. Treatment with antiviral agents, such as ganciclovir, is limited by their toxicity. In this study, we investigated the effectiveness of a fully human neutralizing monoclonal antibody to inhibit human cytomegalovirus infection and viral cell-to-cell spread. We isolated a potent neutralizing antibody, EV2038 (IgG1 lambda), targeting human cytomegalovirus glycoprotein B using Epstein-Barr virus transformation. This antibody inhibited human cytomegalovirus infection by all four laboratory strains and 42 Japanese clinical isolates, including ganciclovir-resistant isolates, with a 50% inhibitory concentration (IC50) ranging from 0.013 to 0.105 μg/mL, and 90% inhibitory concentration (IC90) ranging from 0.208 to 1.026 μg/mL, in both human embryonic lung fibroblasts (MRC-5) and human retinal pigment epithelial (ARPE-19) cells. Additionally, EV2038 prevented cell-to-cell spread of eight clinical viral isolates, with IC50 values ranging from 1.0 to 3.1 μg/mL, and IC90 values ranging from 13 to 19 μg/mL, in ARPE-19 cells. EV2038 recognized three discontinuous sequences on antigenic domain 1 of glycoprotein B (amino acids 549-560, 569-576, and 625-632), which were highly conserved among 71 clinical isolates from Japan and the United States. Pharmacokinetics study in cynomolgus monkeys suggested the potential efficacy of EV2038 in vivo, the concentration of which in serum remained higher than the IC90 values of cell-to-cell spread until 28 days after intravenous injection of 10 mg/kg EV2038. Our data strongly support EV2038 as a promising candidate and novel alternative for the treatment of human cytomegalovirus infection.
Collapse
Affiliation(s)
- Miwa Okamoto
- Sapporo Laboratory, EVEC, Inc., Sapporo, Hokkaido, Japan
| | - Rika Kurino
- Sapporo Laboratory, EVEC, Inc., Sapporo, Hokkaido, Japan
| | - Ryu Miura
- Sapporo Laboratory, EVEC, Inc., Sapporo, Hokkaido, Japan
| | - Kenzo Takada
- Sapporo Laboratory, EVEC, Inc., Sapporo, Hokkaido, Japan
| |
Collapse
|
7
|
Jenks JA, Amin S, Sponholtz MR, Kumar A, Wrapp D, Venkatayogi S, Tu JJ, Karthigeyan K, Valencia SM, Connors M, Harnois MJ, Hora B, Rochat E, McLellan JS, Wiehe K, Permar SR. A single, improbable B cell receptor mutation confers potent neutralization against cytomegalovirus. PLoS Pathog 2023; 19:e1011107. [PMID: 36662906 PMCID: PMC9891502 DOI: 10.1371/journal.ppat.1011107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/01/2023] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
Cytomegalovirus (CMV) is a leading cause of infant hearing loss and neurodevelopmental delay, but there are no clinically licensed vaccines to prevent infection, in part due to challenges eliciting neutralizing antibodies. One of the most well-studied targets for CMV vaccines is the viral fusogen glycoprotein B (gB), which is required for viral entry into host cells. Within gB, antigenic domain 2 site 1 (AD-2S1) is a target of potently neutralizing antibodies, but gB-based candidate vaccines have yet to elicit robust responses against this region. We mapped the genealogy of B cells encoding potently neutralizing anti-gB AD-2S1 antibodies from their inferred unmutated common ancestor (UCA) and characterized the binding and function of early lineage ancestors. Surprisingly, we found that a single amino acid heavy chain mutation A33N, which was an improbable mutation rarely generated by somatic hypermutation machinery, conferred broad CMV neutralization to the non-neutralizing UCA antibody. Structural studies revealed that this mutation mediated key contacts with the gB AD-2S1 epitope. Collectively, these results provide insight into potently neutralizing gB-directed antibody evolution in a single donor and lay a foundation for using this B cell-lineage directed approach for the design of next-generation CMV vaccines.
Collapse
Affiliation(s)
- Jennifer A. Jenks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Sharmi Amin
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Madeline R. Sponholtz
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Amit Kumar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Sravani Venkatayogi
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Joshua J. Tu
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Krithika Karthigeyan
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, United States of America
| | - Sarah M. Valencia
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Megan Connors
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, United States of America
| | - Melissa J. Harnois
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Bhavna Hora
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Eric Rochat
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, United States of America
| |
Collapse
|
8
|
Neutralizing Antibodies to Human Cytomegalovirus Recombinant Proteins Reduce Infection in an Ex Vivo Model of Developing Human Placentas. Vaccines (Basel) 2022; 10:vaccines10071074. [PMID: 35891239 PMCID: PMC9315547 DOI: 10.3390/vaccines10071074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 12/03/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the leading viral cause of congenital disease and permanent birth defects worldwide. Although the development of an effective vaccine is a public health priority, no vaccines are approved. Among the major antigenic targets are glycoproteins in the virion envelope, including gB, which facilitates cellular entry, and the pentameric complex (gH/gL/pUL128-131), required for the infection of specialized cell types. In this study, sera from rabbits immunized with the recombinant pentameric complex were tested for their ability to neutralize infection of epithelial cells, fibroblasts, and primary placental cell types. Sera from rhesus macaques immunized with recombinant gB or gB plus pentameric complex were tested for HCMV neutralizing activity on both cultured cells and cell column cytotrophoblasts in first-trimester chorionic villus explants. Sera from rabbits immunized with the pentameric complex potently blocked infection by pathogenic viral strains in amniotic epithelial cells and cytotrophoblasts but were less effective in fibroblasts and trophoblast progenitor cells. Sera from rhesus macaques immunized with the pentameric complex and gB more strongly reduced infection in fibroblasts, epithelial cells, and chorionic villus explants than sera from immunization with gB alone. These results suggest that the pentameric complex and gB together elicit antibodies that could have potential as prophylactic vaccine antigens.
Collapse
|
9
|
Neutralizing Antibodies Limit Cell-Associated Spread of Human Cytomegalovirus in Epithelial Cells and Fibroblasts. Viruses 2022; 14:v14020284. [PMID: 35215877 PMCID: PMC8875165 DOI: 10.3390/v14020284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
Human cytomegalovirus (HCMV) can cause severe clinical disease in immunocompromised individuals, such as allograft recipients and infants infected in utero. Neutralizing activity of antibodies, measured as the ability to prevent the entry of cell-free virus, has been correlated with the reduction in HCMV transmission and the severity of HCMV-associated disease. However, in vivo HCMV amplification may occur mainly via cell-to-cell spread. Thus, quantifying the inhibition of cell-to-cell transmission could be important in the evaluation of therapeutic antibodies and/or humoral responses to infection or immunization. Here, we established a quantitative plaque reduction assay, which allowed for the measurement of the capacity of antibodies to limit HCMV spread in vitro. Using an automated fluorescence spot reader, infection progression was assayed by the expansion of viral plaques during the course of infection with various GFP-expressing viruses. We found that in contrast to non-neutralizing monoclonal antibodies (mAbs), neutralizing mAbs against both glycoprotein B and H (gB and gH) could significantly inhibit viral plaque expansion of different HCMV strains and was equally efficient in fibroblasts as in epithelial cells. In contrast, an anti-pentamer mAb was active only in epithelial cells. Taken together, our data demonstrate that specific anti-HCMV mAbs can significantly limit cell-associated virus spread in vitro.
Collapse
|
10
|
Hyde K, Sultana N, Tran AC, Bileckaja N, Donald CL, Kohl A, Stanton RJ, Strang BL. Limited replication of human cytomegalovirus in a trophoblast cell line. J Gen Virol 2021; 102. [PMID: 34816792 PMCID: PMC8742992 DOI: 10.1099/jgv.0.001683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Several viruses, including human cytomegalovirus (HCMV), are thought to replicate in the placenta. However, there is little understanding of the molecular mechanisms involved in HCMV replication in this tissue. We investigated replication of HCMV in the extravillous trophoblast cell line SGHPL-4, a commonly used model of HCMV replication in the placenta. We found limited HCMV protein expression and virus replication in SGHPL-4 cells. This was associated with a lack of trophoblast progenitor cell protein markers in SGHPL-4 cells, suggesting a relationship between trophoblast differentiation and limited HCMV replication. We proposed that limited HCMV replication in trophoblast cells is advantageous to vertical transmission of HCMV, as there is a greater opportunity for vertical transmission when the placenta is intact and functional. Furthermore, when we investigated the replication of other vertically transmitted viruses in SGHPL-4 cells we found some limitation to replication of Zika virus, but not herpes simplex virus. Thus, limited replication of some, but not all, vertically transmitted viruses may be a feature of trophoblast cells.
Collapse
Affiliation(s)
- Kadeem Hyde
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Nowshin Sultana
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Andy C Tran
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Narina Bileckaja
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Claire L Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Blair L Strang
- Institute for Infection and Immunity, St George's, University of London, London, UK
| |
Collapse
|
11
|
Berkebile ZW, Putri DS, Abrahante JE, Seelig DM, Schleiss MR, Bierle CJ. The Placental Response to Guinea Pig Cytomegalovirus Depends Upon the Timing of Maternal Infection. Front Immunol 2021; 12:686415. [PMID: 34211475 PMCID: PMC8239309 DOI: 10.3389/fimmu.2021.686415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022] Open
Abstract
Human cytomegalovirus (HCMV) infects the placenta, and these placental infections can cause fetal injury and/or demise. The timing of maternal HCMV infection during pregnancy is a determinant of fetal outcomes, but how development affects the placenta's susceptibility to infection, the likelihood of placental injury post-infection, and the frequency of transplacental HCMV transmission remains unclear. In this study, guinea pig cytomegalovirus (GPCMV) was used to model primary maternal infection and compare the effects of infection at two different times on the placenta. When guinea pigs were infected with GPCMV at either 21- or 35-days gestation (dGA), maternal and placental viral loads, as determined by droplet digital PCR, were not significantly affected by the timing of maternal infection. However, when the transcriptomes of gestational age-matched GPCMV-infected and control placentas were compared, significant infection-associated changes in gene expression were only observed after maternal infection at 35 dGA. Notably, transcripts associated with immune activation (e.g. Cxcl10, Ido1, Tgtp1, and Tlr8) were upregulated in the infected placenta. A GPCMV-specific in situ hybridization assay detected rare infected cells in the main placenta after maternal infection at either time, and maternal infection at 35 dGA also caused large areas of GPCMV-infected cells in the junctional zone. As GPCMV infection after mid-gestation is known to cause high rates of stillbirth and/or fetal growth restriction, our results suggest that the placenta becomes sensitized to infection-associated injury late in gestation, conferring an increased risk of adverse pregnancy outcomes after cytomegalovirus infection.
Collapse
Affiliation(s)
- Zachary W. Berkebile
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| | - Dira S. Putri
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| | - Juan E. Abrahante
- Informatics Institute, University of Minnesota, Minneapolis, MN, United States
| | - Davis M. Seelig
- Department of Veterinary Clinical Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Mark R. Schleiss
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| | - Craig J. Bierle
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
12
|
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
|
13
|
Choi KY, El-Hamdi NS, McGregor A. A trimeric capable gB CMV vaccine provides limited protection against a highly cell associated and epithelial tropic strain of cytomegalovirus in guinea pigs. J Gen Virol 2021; 102. [PMID: 33729125 DOI: 10.1099/jgv.0.001579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Multiple strains of human cytomegalovirus (HCMV) can cause congenital cytomegalovirus (cCMV) by primary or secondary infection. The viral gB glycoprotein is a leading vaccine candidate, essential for infection of all cell-types, and immunodominant antibody target. Guinea pig cytomegalovirus (GPCMV) is the only small animal model for cCMV. Various gB vaccines have shown efficacy but studies have utilized truncated gB and protection against prototype strain 22122 with preferential tropism to fibroblasts despite encoding a gH-based pentamer complex for non-fibroblast infection. A highly cell-associated novel strain of GPCMV (TAMYC) with 99 % identity in gB sequence to 22122 exhibited preferred tropism to epithelial cells. An adenovirus vaccine encoding full-length gB (AdgB) was highly immunogenic and partially protected against 22122 strain challenge in vaccinated animals but not when challenged with TAMYC strain. GPCMV studies with AdgB vaccine sera on numerous cell-types demonstrated impaired neutralization (NA50) compared to fibroblasts. GPCMV-convalescent sera including pentamer complex antibodies increased virus neutralization on non-fibroblasts and anti-gB depletion from GPCMV-convalescent sera had minimal impact on epithelial cell neutralization. GPCMV(PC+) 22122-convalescent animals challenged with TAMYC exhibited higher protection compared to AdgB vaccine. Overall, results suggest that antibody response to both gB and PC are important components of a GPCMV vaccine.
Collapse
Affiliation(s)
- K Yeon Choi
- Dept. Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Nadia S El-Hamdi
- Dept. Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Alistair McGregor
- Dept. Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA
| |
Collapse
|
14
|
Singh K, Hamilton ST, Shand AW, Hannan NJ, Rawlinson WD. Receptors in host pathogen interactions between human cytomegalovirus and the placenta during congenital infection. Rev Med Virol 2021; 31:e2233. [PMID: 33709529 DOI: 10.1002/rmv.2233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 11/09/2022]
Abstract
Cellular receptors in human cytomegalovirus (HCMV) mother to child transmission play an important role in congenital infection. Placental trophoblast cells are a significant cell type in placental development, placental functional processes, and in HCMV transmission. Different cells within the placental floating and chorionic villi present alternate receptors for HCMV cell entry. Syncytiotrophoblasts present neonatal Fc receptors that bind and transport circulating maternal immunoglobulin G across the placental interface which can also be bound to HCMV virions, facilitating viral entry into the placenta and foetal circulation. Cytotrophoblast express HCMV receptors including integrin-α1β1, integrin-αVβ3, epidermal growth factor receptor and platelet-derived growth factor receptor alpha. The latter interacts with HCMV glycoprotein-H, glycoprotein-L and glycoprotein-O (gH/gL/gO) trimers (predominantly in placental fibroblasts) and the gH/gL/pUL128, UL130-UL131A pentameric complex in other placental cell types. The pentameric complex allows viral tropism of placental trophoblasts, endothelial cells, epithelial cells, leukocytes and monocytes. This review outlines HCMV ligands and target receptor proteins in congenital HCMV infection.
Collapse
Affiliation(s)
- Krishneel Singh
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Stuart T Hamilton
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, New South Wales, Australia.,School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Antonia W Shand
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Natalie J Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia.,Mercy Perinatal, Mercy Hospital for Women Heidelberg, Victoria, Australia
| | - William D Rawlinson
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, New South Wales, Australia.,School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Njue A, Coyne C, Margulis AV, Wang D, Marks MA, Russell K, Das R, Sinha A. The Role of Congenital Cytomegalovirus Infection in Adverse Birth Outcomes: A Review of the Potential Mechanisms. Viruses 2020; 13:v13010020. [PMID: 33374185 PMCID: PMC7823935 DOI: 10.3390/v13010020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022] Open
Abstract
Human cytomegalovirus (CMV) is a major cause of nonhereditary adverse birth outcomes, including hearing and visual loss, neurologic deficits, and intrauterine growth retardation (IUGR), and may contribute to outcomes such as stillbirth and preterm delivery. However, the mechanisms by which CMV could cause adverse birth outcomes are not fully understood. This study reviewed proposed mechanisms underlying the role of CMV in stillbirth, preterm birth, and IUGR. Targeted literature searches were performed in PubMed and Embase to identify relevant articles. Several potential mechanisms were identified from in vitro studies in which laboratory-adapted and low-passage strains of CMV and various human placental models were used. Potential mechanisms identified included impairment of trophoblast progenitor stem cell differentiation and function, impairment of extravillous trophoblast invasiveness, dysregulation of Wnt signaling pathways in cytotrophoblasts, tumor necrosis factor-α mediated apoptosis of trophoblasts, CMV-induced cytokine changes in the placenta, inhibition of indoleamine 2,3-dioxygenase activity, and downregulation of trophoblast class I major histocompatibility complex molecules. Inherent challenges for the field remain in the identification of suitable in vivo animal models. Nonetheless, we believe that our review provides useful insights into the mechanisms by which CMV impairs placental development and function and how these changes could result in adverse birth outcomes.
Collapse
Affiliation(s)
- Annete Njue
- RTI Health Solutions, Manchester M20 2LS, UK
- Correspondence:
| | - Carolyn Coyne
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | | | - Dai Wang
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.W.); (M.A.M.); (K.R.); (R.D.); (A.S.)
| | - Morgan A. Marks
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.W.); (M.A.M.); (K.R.); (R.D.); (A.S.)
| | - Kevin Russell
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.W.); (M.A.M.); (K.R.); (R.D.); (A.S.)
| | - Rituparna Das
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.W.); (M.A.M.); (K.R.); (R.D.); (A.S.)
| | - Anushua Sinha
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (D.W.); (M.A.M.); (K.R.); (R.D.); (A.S.)
| |
Collapse
|
16
|
Reuter N, Kropff B, Schneiderbanger JK, Alt M, Krawczyk A, Sinzger C, Winkler TH, Britt WJ, Mach M, Thomas M. Cell Fusion Induced by a Fusion-Active Form of Human Cytomegalovirus Glycoprotein B (gB) Is Inhibited by Antibodies Directed at Antigenic Domain 5 in the Ectodomain of gB. J Virol 2020; 94:e01276-20. [PMID: 32641474 PMCID: PMC7459561 DOI: 10.1128/jvi.01276-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 01/22/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen that can cause severe clinical disease in allograft recipients and infants infected in utero Virus-neutralizing antibodies defined in vitro have been proposed to confer protection against HCMV infection, and the virion envelope glycoprotein B (gB) serves as a major target of neutralizing antibodies. The viral fusion protein gB is nonfusogenic on its own and requires glycoproteins H (gH) and L (gL) for membrane fusion, which is in contrast to requirements of related class III fusion proteins, including vesicular stomatitis virus glycoprotein G (VSV-G) or baculovirus gp64. To explore requirements for gB's fusion activity, we generated a set of chimeras composed of gB and VSV-G or gp64, respectively. These gB chimeras were intrinsically fusion active and led to the formation of multinucleated cell syncytia when expressed in the absence of other viral proteins. Utilizing a panel of virus-neutralizing gB-specific monoclonal antibodies (MAbs), we could demonstrate that syncytium formation of the fusogenic gB/VSV-G chimera can be significantly inhibited by only a subset of neutralizing MAbs which target antigenic domain 5 (AD-5) of gB. This observation argues for differential modes of action of neutralizing anti-gB MAbs and suggests that blocking the membrane fusion function of gB could be one mechanism of antibody-mediated virus neutralization. In addition, our data have important implications for the further understanding of the conformation of gB that promotes membrane fusion as well as the identification of structures in AD-5 that could be targeted by antibodies to block this early step in HCMV infection.IMPORTANCE HCMV is a major global health concern, and antiviral chemotherapy remains problematic due to toxicity of available compounds and the emergence of drug-resistant viruses. Thus, an HCMV vaccine represents a priority for both governmental and pharmaceutical research programs. A major obstacle for the development of a vaccine is a lack of knowledge of the nature and specificities of protective immune responses that should be induced by such a vaccine. Glycoprotein B of HCMV is an important target for neutralizing antibodies and, hence, is often included as a component of intervention strategies. By generation of fusion-active gB chimeras, we were able to identify target structures of neutralizing antibodies that potently block gB-induced membrane fusion. This experimental system provides an approach to screen for antibodies that interfere with gB's fusogenic activity. In summary, our data will likely contribute to both rational vaccine design and the development of antibody-based therapies against HCMV.
Collapse
Affiliation(s)
- Nina Reuter
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Kropff
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Karin Schneiderbanger
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mira Alt
- Institut für Virologie, Universitätsklinikum Duisburg-Essen, Essen, Germany
| | - Adalbert Krawczyk
- Institut für Virologie, Universitätsklinikum Duisburg-Essen, Essen, Germany
- Klinik für Infektiologie, Medizinisches Forschungszentrum, Universitätsklinikum Duisburg-Essen, Essen, Germany
| | | | - Thomas H Winkler
- Nikolaus-Fiebiger-Zentrum für Molekulare Medizin, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - William J Britt
- Departments of Pediatrics, Microbiology and Neurobiology, Children's Hospital of Alabama, University of Alabama School of Medicine, Birmingham, Alabama, USA
| | - Michael Mach
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marco Thomas
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
17
|
Charostad J, Mokhtari-Azad D V M T, Mph, Ph D, Yavarian J, Ghavami N, Mahmood Seyed Khorrami S, Behboudi E, Jalilvand S, Shatizadeh Malekshahi S, Zahra Shafiei-Jandaghi N. Detection of human herpes viruses 1-5 in miscarriage: A case-control study. Int J Reprod Biomed 2020; 18:501-508. [PMID: 32803115 PMCID: PMC7385915 DOI: 10.18502/ijrm.v13i7.7367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/23/2019] [Accepted: 01/04/2020] [Indexed: 12/16/2022] Open
Abstract
Background Miscarriage is the spontaneous pregnancy loss before 24 wk of gestation. The incidence rate of miscarriage over the past few decades has shown steady or even growing trends. Viral intrauterine infections are one of the probable etiological causes of miscarriage. Previous evidence have shown that human herpes viruses (HHVs) could be considered as the potential reasons for intrauterine infections and adverse pregnancy outcomes. Objective This case-control study aimed to detect HHV1-5 DNAs in placental tissues and assess their association with miscarriage during the first 24 wk of pregnancy in spontaneous and therapeutic abortions. Materials and Methods Placental tissues from 83 women with spontaneous abortions during the first and the second trimesters of pregnancy and 81 women with therapeutic abortion during the same gestational age were collected. The DNA extraction was performed by the phenol/chloroform method. A part of the DNA polymerase gene of HHVs was amplified with multiplex nested-polymerase chain reaction. The polymerase chain reaction products were subjected to sequencing. Results The results showed the presence of human cytomegalovirus genome in the placenta of both spontaneous (8.4%) and therapeutic (4.9%) abortions. No statistically significant differences were found between these two groups. The other investigated viruses were not detected here. Conclusion In conclusion, like some other studies, no correlation was detected between the HHVs placental infections and the increased risk of spontaneous abortions. In order to find the actual role of HHVs infections in miscarriage, further investigations should be performed on a larger sample size in different areas.
Collapse
Affiliation(s)
- Javad Charostad
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mph
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ph D
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Jila Yavarian
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Ghavami
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Emad Behboudi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
18
|
Sandonís V, García-Ríos E, McConnell MJ, Pérez-Romero P. Role of Neutralizing Antibodies in CMV Infection: Implications for New Therapeutic Approaches. Trends Microbiol 2020; 28:900-912. [PMID: 32448762 DOI: 10.1016/j.tim.2020.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023]
Abstract
Cytomegalovirus (CMV) infection elicits a potent immune response that includes the stimulation of antibodies with neutralizing activity. Recent studies have focused on elucidating the role of neutralizing antibodies in protecting against CMV infection and disease and characterizing viral antigens against which neutralizing antibodies are directed. Here, we provide a synthesis of recent data regarding the role of neutralizing antibodies in protection against CMV infection/disease. We consider the role of humoral immunity in the context of the global CMV-specific immune response, and the implications that recent findings have for vaccine and antibody-based therapy design.
Collapse
Affiliation(s)
- Virginia Sandonís
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), Madrid, Spain
| | - Estéfani García-Ríos
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Michael J McConnell
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Pilar Pérez-Romero
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
| |
Collapse
|
19
|
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
|
20
|
Cui X, Cao Z, Wang S, Adler SP, McVoy MA, Snapper CM. Immunization with Human Cytomegalovirus Core Fusion Machinery and Accessory Envelope Proteins Elicit Strong Synergistic Neutralizing Activities. Vaccines (Basel) 2020; 8:vaccines8020179. [PMID: 32294946 PMCID: PMC7348949 DOI: 10.3390/vaccines8020179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/04/2020] [Accepted: 04/10/2020] [Indexed: 12/15/2022] Open
Abstract
Human cytomegalovirus (HCMV) core fusion machinery proteins gB and gH/gL, and accessory proteins UL128/UL130/UL131A, are the key envelope proteins that mediate HCMV entry into and infection of host cells. To determine whether these HCMV envelope proteins could elicit neutralizing activities synergistically, we immunized rabbits with individual or various combinations of these proteins adsorbed to aluminum hydroxide mixed with CpG-ODN. We then analyzed serum neutralizing activities with multiple HCMV laboratory strains and clinical isolates. HCMV trimeric gB and gH/gL elicited high and moderate titers of HCMV neutralizing activity, respectively. HCMV gB in combination with gH/gL elicited up to 17-fold higher HCMV neutralizing activities compared to the sum of neutralizing activity elicited by the individual proteins analyzed with both fibroblasts and epithelial cells. HCMV gB+gH/gL+UL128/UL130/UL131A in combination increased the neutralizing activity up to 32-fold compared to the sum of neutralizing activities elicited by the individual proteins analyzed with epithelial cells. Adding UL128/UL130/UL131A to gB and gH/gL combination did not increase further the HCMV neutralizing activity analyzed with fibroblasts. These data suggest that the combination of HCMV core fusion machinery envelope proteins gB+gH/gL or the combination of gB and pentameric complex could be ideal vaccine candidates that would induce optimal immune responses against HCMV infection.
Collapse
Affiliation(s)
- Xinle Cui
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +1-301-295-3498
| | - Zhouhong Cao
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Shuishu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Michael A. McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Clifford M. Snapper
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
21
|
Qi Y, He L, Cui X, Hertel L, Freed DC, Fu TM, Kauvar LM, McVoy MA, Ruan Q. Comparative neutralizing potencies of antibodies suggest conservation as well as mechanistic differences in human cytomegalovirus entry into epithelial and endothelial cells. Virol J 2020; 17:50. [PMID: 32268919 PMCID: PMC7144056 DOI: 10.1186/s12985-020-01320-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/20/2020] [Indexed: 11/20/2022] Open
Abstract
Antibody neutralization of cytomegalovirus (CMV) entry into diverse cell types is a key consideration for development of vaccines and immunotherapeutics. CMV entry into fibroblasts differs significantly from entry into epithelial or endothelial cells: fibroblast entry is mediated by gB and gH/gL/gO, whereas both epithelial and endothelial cell entry require an additional pentameric complex (PC) comprised of gH/gL/UL128/UL130/UL131A. Because PC-specific antibodies in CMV-seropositive human sera do not affect fibroblast entry but potently block entry into epithelial or endothelial cells, substantially higher neutralizing potencies for CMV-positive sera are observed when assayed using epithelial cells as targets than when using fibroblasts. That certain sera exhibit similar discordances between neutralizing potencies measured using epithelial vs. endothelial cells (Gerna G. et al.J Gen Virol, 89:853–865, 2008) suggested that additional mechanistic differences may also exist between epithelial and endothelial cell entry. To further explore this issue, neutralizing potencies using epithelial and endothelial cells were simultaneously determined for eight CMV-positive human sera, CMV-hyperimmune globulin, and a panel of monoclonal or anti-peptide antibodies targeting specific epitopes in gB, gH, gH/gL, or the PC. No significant differences were observed between epithelial and endothelial neutralizing potencies of epitope-specific antibodies, CMV-hyperimmune globulin, or seven of the eight human sera. However, one human serum exhibited a six-fold higher potency for neutralizing entry into epithelial cells vs. endothelial cells. These results suggest that epitopes exist that are important for epithelial entry but are less critical, or perhaps dispensable, for endothelial cell entry. Their existence should be considered when developing monoclonal antibody therapies or subunit vaccines representing limited epitopes.
Collapse
Affiliation(s)
- Ying Qi
- Virology laboratory, Shengjing Hospital, China Medical University, Shenyang, People's Republic of China
| | - Li He
- Virginia Commonwealth University, Richmond, VA, USA
| | - Xiaohong Cui
- Virginia Commonwealth University, Richmond, VA, USA
| | - Laura Hertel
- Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | | | - Tong-Ming Fu
- University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | | | - Qiang Ruan
- Virology laboratory, Shengjing Hospital, China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
22
|
Modeling Human Cytomegalovirus-Induced Microcephaly in Human iPSC-Derived Brain Organoids. CELL REPORTS MEDICINE 2020; 1:100002. [PMID: 33205055 PMCID: PMC7659592 DOI: 10.1016/j.xcrm.2020.100002] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 01/16/2020] [Accepted: 02/28/2020] [Indexed: 12/13/2022]
Abstract
Although congenital infection by human cytomegalovirus (HCMV) is well recognized as a leading cause of neurodevelopmental defects, HCMV neuropathogenesis remains poorly understood. A major challenge for investigating HCMV-induced abnormal brain development is the strict CMV species specificity, which prevents the use of animal models to directly study brain defects caused by HCMV. We show that infection of human-induced pluripotent-stem-cell-derived brain organoids by a “clinical-like” HCMV strain results in reduced brain organoid growth, impaired formation of cortical layers, and abnormal calcium signaling and neural network activity. Moreover, we show that the impeded brain organoid development caused by HCMV can be prevented by neutralizing antibodies (NAbs) that recognize the HCMV pentamer complex. These results demonstrate in a three-dimensional cellular biosystem that HCMV can impair the development and function of the human brain and provide insights into the potential capacity of NAbs to mitigate brain defects resulted from HCMV infection. Human iPSC-derived brain organoids to model HCMV-induced brain malformation “Clinical-like” HCMV strain impairs human brain organoid growth and structure HCMV-infected brain organoids exhibit abnormal calcium signaling and neural network HCMV-induced brain organoid abnormality can be prevented by neutralizing antibodies
Collapse
|
23
|
Differential Expression of PDGF Receptor-α in Human Placental Trophoblasts Leads to Different Entry Pathways by Human Cytomegalovirus Strains. Sci Rep 2020; 10:1082. [PMID: 31974453 PMCID: PMC6978357 DOI: 10.1038/s41598-020-57471-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Human cytomegalovirus (CMV) is the leading non-genetic cause of fetal malformation in developed countries. CMV placental infection is a pre-requisite for materno-fetal transmission of virus, and fetal infection. We investigated the roles of the viral pentameric complex gH/gL/pUL128-pUL131A, and cellular platelet-derived growth factor receptor-α (PDGFRα) for CMV infection in first trimester extravillous-derived (SGHPL-4) and villous-derived (HTR-8/SVneo) trophoblast cells. Infection with four CMV clinical and laboratory strains (Merlin, TB40E, Towne, AD169), and Merlin deletion mutants of UL128-, UL130-, and UL131A-genes, showed a cell type-dependent requirement of the viral pentameric complex for infection of trophoblast cells. The viral pentameric complex was essential for infection of villous trophoblasts, but non-essential for extravillous trophoblasts. Blocking of PDGFRα in extravillous trophoblasts, which naturally express PDGFRα, inhibited entry of pentameric complex-deficient CMV strains, but not the entry of pentameric positive CMV strains. Transient expression of PDGFRα in villous trophoblasts, which are naturally deficient in PDGFRα, promoted the entry of CMV strains lacking gH/gL/pUL128-pUL131A, but had no effect on entry of pentameric positive CMV strains. These results suggest PDGFRα is an important cell receptor for entry of CMV mutant strains lacking gH/gL/pUL128-pUL131A complexes in some placental cells, suggesting these entry pathways could be potential antiviral targets.
Collapse
|
24
|
Virus-Like Particles and Nanoparticles for Vaccine Development against HCMV. Viruses 2019; 12:v12010035. [PMID: 31905677 PMCID: PMC7019358 DOI: 10.3390/v12010035] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/21/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) infects more than 70% of the human population worldwide. HCMV is responsible for high morbidity and mortality in immunocompromised patients and remains the leading viral cause of congenital birth defects. Despite considerable efforts in vaccine and therapeutic development, HCMV infection still represents an unmet clinical need and a life-threatening disease in immunocompromised individuals and newborns. Immune repertoire interrogation of HCMV seropositive patients allowed the identification of several potential antigens for vaccine design. However, recent HCMV vaccine clinical trials did not lead to a satisfactory outcome in term of efficacy. Therefore, combining antigens with orthogonal technologies to further increase the induction of neutralizing antibodies could improve the likelihood of a vaccine to reach protective efficacy in humans. Indeed, presentation of multiple copies of an antigen in a repetitive array is known to drive a more robust humoral immune response than its soluble counterpart. Virus-like particles (VLPs) and nanoparticles (NPs) are powerful platforms for multivalent antigen presentation. Several self-assembling proteins have been successfully used as scaffolds to present complex glycoprotein antigens on their surface. In this review, we describe some key aspects of the immune response to HCMV and discuss the scaffolds that were successfully used to increase vaccine efficacy against viruses with unmet medical need.
Collapse
|
25
|
Paradowska E, Jabłońska A, Studzińska M, Kasztelewicz B, Wiśniewska-Ligier M, Dzierżanowska-Fangrat K, Woźniakowska-Gęsicka T, Czech-Kowalska J. Distribution of the CMV glycoprotein gH/gL/gO and gH/gL/pUL128/pUL130/pUL131A complex variants and associated clinical manifestations in infants infected congenitally or postnatally. Sci Rep 2019; 9:16352. [PMID: 31705022 PMCID: PMC6841705 DOI: 10.1038/s41598-019-52906-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 10/22/2019] [Indexed: 11/09/2022] Open
Abstract
Human cytomegalovirus (CMV) is a major cause of morbidity in fetuses following intrauterine infection. The glycoprotein (g) envelope trimeric gH/gL/gO and pentameric gH/gL/pUL128/pUL130/pUL131A complexes are required for CMV entry into fibroblasts and endothelial/epithelial cells, respectively, and both are targets for neutralizing antibodies. The role of sequence variability among viral strains in the outcome of congenital CMV infection is controversial. Variation in the CMV UL75 gene encoding glycoprotein H (gH), the UL115 (gL), the UL74 (gO), and the UL128 locus (UL128L) encoding three structural proteins (pUL128, pUL130, and pUL131A) was determined in 82 newborns with congenital CMV infection and 113 infants with postnatal or unproven congenital CMV infection. Genotyping was performed by sequencing analysis of PCR-amplified fragments and the PCR-restriction fragment length polymorphism (RFLP) method, and the viral load was measured by quantitative real-time PCR. The obtained results demonstrated that (1) different CMV variants and mixed CMV infections can be detected in newborns infected congenitally; (2) the gH1 genotype, UL130 variant 6, and UL131A variant 1 were associated with some signs/symptoms within cohort of pediatric patients, mainly consisting of infants with symptomatic CMV infection. The results revealed that pUL130, pUL131A, and gH polymorphisms seemed to be associated with the outcome of CMV infection in infants.
Collapse
Affiliation(s)
- Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland.
| | - Agnieszka Jabłońska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Mirosława Studzińska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Beata Kasztelewicz
- Department of Clinical Microbiology and Immunology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Małgorzata Wiśniewska-Ligier
- Department of Pediatrics, Immunology, and Nephrology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
- 3rd Department of Pediatrics, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | | | | | - Justyna Czech-Kowalska
- Department of Neonatology and Neonatal Intensive Care, The Children's Memorial Health Institute, Warsaw, Poland
| |
Collapse
|
26
|
Dibo M, Battocchio EC, dos Santos Souza LM, da Silva MDV, Banin-Hirata BK, Sapla MM, Marinello P, Rocha SP, Faccin-Galhardi LC. Antibody Therapy for the Control of Viral Diseases: An Update. Curr Pharm Biotechnol 2019; 20:1108-1121. [DOI: 10.2174/1389201020666190809112704] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/22/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
The epidemiological impact of viral diseases, combined with the emergence and reemergence of some viruses, and the difficulties in identifying effective therapies, have encouraged several studies to develop new therapeutic strategies for viral infections. In this context, the use of immunotherapy for the treatment of viral diseases is increasing. One of the strategies of immunotherapy is the use of antibodies, particularly the monoclonal antibodies (mAbs) and multi-specific antibodies, which bind directly to the viral antigen and bring about activation of the immune system. With current advancements in science and technology, several such antibodies are being tested, and some are already approved and are undergoing clinical trials. The present work aims to review the status of mAb development for the treatment of viral diseases.
Collapse
Affiliation(s)
- Miriam Dibo
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Eduardo C. Battocchio
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lucas M. dos Santos Souza
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | | | - Bruna K. Banin-Hirata
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Milena M.M. Sapla
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Poliana Marinello
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Sérgio P.D. Rocha
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lígia C. Faccin-Galhardi
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| |
Collapse
|
27
|
Tabata T, Petitt M, Fang-Hoover J, Freed DC, Li F, An Z, Wang D, Fu TM, Pereira L. Neutralizing Monoclonal Antibodies Reduce Human Cytomegalovirus Infection and Spread in Developing Placentas. Vaccines (Basel) 2019; 7:vaccines7040135. [PMID: 31569508 PMCID: PMC6963214 DOI: 10.3390/vaccines7040135] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/16/2019] [Accepted: 09/22/2019] [Indexed: 12/18/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects worldwide, yet the most effective strategies for preventing virus transmission during pregnancy are unknown. We measured the efficacy of human monoclonal antibodies (mAbs) to HCMV attachment/entry factors glycoprotein B (gB) and the pentameric complex, gH/gL-pUL128–131, in preventing infection and spread of a clinical strain in primary placental cells and explants of developing anchoring villi. A total of 109 explants from five first-trimester placentas were cultured, and infection was analyzed in over 400 cell columns containing ~120,000 cytotrophoblasts (CTBs). mAbs to gB and gH/gL, 3-25 and 3-16, respectively, neutralized infection in stromal fibroblasts and trophoblast progenitor cells. mAbs to pUL128-131 of the pentameric complex, 1-103 and 2-18, neutralized infection of amniotic epithelial cells better than mAbs 3-25 and 3-16 and hyperimmune globulin. Select mAbs neutralized infection of cell column CTBs, with mAb 2-18 most effective, followed by mAb 3-25. Treatment of anchoring villi with mAbs postinfection reduced spread in CTBs and impaired formation of virion assembly compartments, with mAb 2-18 achieving better suppression at lower concentrations. These results predict that antibodies generated by HCMV vaccines or used for passive immunization have the potential to reduce transplacental transmission and congenital disease.
Collapse
Affiliation(s)
- Takako Tabata
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| | - Matthew Petitt
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| | | | | | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Dai Wang
- Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Tong-Ming Fu
- Merck & Co., Inc., Kenilworth, NJ 07033, USA.
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Lenore Pereira
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
28
|
Cui X, Cao Z, Wang S, Flora M, Adler SP, McVoy MA, Snapper CM. Immunization of Rabbits with Recombinant Human Cytomegalovirus Trimeric versus Monomeric gH/gL Protein Elicits Markedly Higher Titers of Antibody and Neutralization Activity. Int J Mol Sci 2019; 20:ijms20133158. [PMID: 31261659 PMCID: PMC6651862 DOI: 10.3390/ijms20133158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 11/30/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection and HCMV infection of immunosuppressed patients cause significant morbidity and mortality, and vaccine development against HCMV is a major public health priority. HCMV envelope glycoproteins gB, gH, and gL, which constitute the core fusion machinery, play critical roles in HCMV fusion and entry into host cells. HCMV gB and gH/gL have been reported to elicit potent neutralizing antibodies. Recently, the gB/gH/gL complex was identified in the envelope of HCMV virions, and 16–50% of the total gH/gL bound to gB, forming the gB/gH/gL complex. These findings make the gB/gH/gL a unique HCMV vaccine candidate. We previously reported the production of HCMV trimeric gB and gH/gL heterodimers, and immunization with a combination of trimeric gB and gH/gL heterodimers elicited strong synergistic HCMV-neutralizing activity. To further improve the immunogenicity of gH/gL, we produced trimeric gH/gL. Rabbits immunized with HCMV trimeric gH/gL induced up to 38-fold higher serum titers of gH/gL-specific IgG relative to HCMV monomeric gH/gL, and elicited ~10-fold higher titers of complement-dependent and complement-independent HCMV-neutralizing activity for both epithelial cells and fibroblasts. HCMV trimeric gH/gL in combination with HCMV trimeric gB would be a novel promising HCMV vaccine candidate that could induce highly potent neutralizing activities.
Collapse
Affiliation(s)
- Xinle Cui
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Zhouhong Cao
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Shuishu Wang
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Michael Flora
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Clifford M Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
29
|
Abstract
Congenital human cytomegalovirus (HCMV) infection and HCMV infection of the immunosuppressed patients cause significant morbidity and mortality, and vaccine development against HCMV is a major public health priority. Efforts to develop HCMV vaccines have been ongoing for 50 y, though no HCMV vaccine has been licensed; encouraging and promising results have obtained from both preclinical and clinical trials. HCMV infection induces a wide range of humoral and T cell-mediated immune responses, and both branches of immunity are correlated with protection. In recent years, there have been novel approaches toward the development of HCMV vaccines and demonstrated that vaccine candidates could potentially provide superior protection over natural immunity acquired following HCMV infection. Further, rationally designed HCMV protein antigens that express native conformational epitopes could elicit optimal immune response. HCMV vaccine candidates, using a multi-antigen approach, to maximize the elicited protective immunity will most likely be successful in development of HCMV vaccine.
Collapse
Affiliation(s)
- Xinle Cui
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Clifford M Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
30
|
Schampera MS, Arellano-Galindo J, Kagan KO, Adler SP, Jahn G, Hamprecht K. Role of pentamer complex-specific and IgG subclass 3 antibodies in HCMV hyperimmunoglobulin and standard intravenous IgG preparations. Med Microbiol Immunol 2019; 208:69-80. [PMID: 30203132 DOI: 10.1007/s00430-018-0558-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/24/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND HCMV hyperimmunoglobulin-preparations (HIG) contain high concentrations of HCMV-specific IgG. The reduced maternofetal-HCMV-transmission rate of IgG may be due to HCMV-specific neutralizing antibodies against the HCMV pentameric complex (PC). In contrast to HIG, standard intravenous immunoglobulin (IVIG) may have more neutralization (NT) capacity than HIG due to higher IgG subclass 3 levels (Planitzer et al., 2011). METHODS We investigated the HCMV-specific NT-capacity of HIG Cytotect®, using a recombinant pentameric complex (gHgLUL128-131A) for specific antibody-depletion. We used a modified UL130-peptide (TANQNPSPPWSKLTYSKPH) based on original-sequence of Saccoccio et al. (Vaccine 29(15):2705-2711, 2011) (SWSTLTANQNPSPPWSKLTY) as neutralization target. Both UL130-peptides and the PC were bound via sixfold HisTag and anti-HisTag mAbs to magnetic beads to deplete HCMV-specific IgGs from HIG (Cytotect®). Modifying this depletion strategy, we analyzed the role of IgG subclass 3 in both HIG and IVIG. RESULTS After CMV IgG-normalization of HIG and IVIG, we found a significant trend towards a decrease (16%) of neutralization-capacity for the UL130 TAN-peptide, but not for the original UL130 SWS-peptide. However, highly significant loss of NT-capacity could be only observed by PC depletion (42%). The IgG subclass 3 depletion revealed no significant reduction of NT-capacity in both HIG and IVIG. CONCLUSION Via specific antibody depletion, we could demonstrate that pentameric complex-specific antibodies are present in HIG and bind to the recombinant PC resulting in a highly significant reduction of NT-capacity compared to the UL130 TAN-and SWS-peptides. We could not confirm the functional role of IgG subclass 3 neutralizing antibodies in IgG-preparations.
Collapse
Affiliation(s)
- Matthias Stefan Schampera
- Institute of Medical Virology, University Hospital of Tuebingen, Elfriede-Aulhorn-Str. 6, 72076, Tübingen, Germany
| | - Jose Arellano-Galindo
- Institute of Medical Virology, University Hospital of Tuebingen, Elfriede-Aulhorn-Str. 6, 72076, Tübingen, Germany
- Infectious Diseases Laboratory (Virology), Children's Hospital Federico Gómez, México City, Mexico
| | - Karl Oliver Kagan
- Department of Obstetrics and Gynaecology, University Hospital of Tuebingen, Tübingen, Germany
| | | | - Gerhard Jahn
- Institute of Medical Virology, University Hospital of Tuebingen, Elfriede-Aulhorn-Str. 6, 72076, Tübingen, Germany
| | - Klaus Hamprecht
- Institute of Medical Virology, University Hospital of Tuebingen, Elfriede-Aulhorn-Str. 6, 72076, Tübingen, Germany.
| |
Collapse
|
31
|
Nelson CS, Herold BC, Permar SR. A new era in cytomegalovirus vaccinology: considerations for rational design of next-generation vaccines to prevent congenital cytomegalovirus infection. NPJ Vaccines 2018; 3:38. [PMID: 30275984 PMCID: PMC6148244 DOI: 10.1038/s41541-018-0074-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/07/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (HCMV), a member of the beta-herpesvirus family, is the most common cause of congenital infection worldwide as well as an important cause of morbidity in transplant recipients and immunosuppressed individuals. An estimated 1 in 150 infants are infected with HCMV at birth, which can result in lifelong, debilitating neurologic sequelae including microcephaly, sensorineural hearing loss, and cognitive impairment. Natural maternal immunity to HCMV decreases the frequency of reinfection and reduces risk of congenital transmission but does not completely protect against neonatal disease. Thus, a vaccine to reduce the incidence and severity of infant infection is a public health priority. A variety of candidate HCMV vaccine approaches have been tried previously, including live-attenuated viruses, glycoprotein subunit formulations, viral vectors, and single/bivalent DNA plasmids, but all have failed to reach target endpoints in clinical trials. Nevertheless, there is a great deal to be learned from the successes and failures of the HCMV vaccine field (both congenital and transplant-associated), as well as from vaccine development efforts for other herpesvirus pathogens including herpes simplex virus 1 and 2, varicella zoster virus, and Epstein-Barr virus. Here, we review those successes and failures, evaluating recent cutting-edge discoveries that have shaped the HCMV vaccine field and identifying topics of critical importance for future investigation. These considerations will inform rational design and evaluation of next-generation vaccines to prevent HCMV-associated congenital infection and disease.
Collapse
Affiliation(s)
- Cody S. Nelson
- Human Vaccine Institute, Duke University Medical Center, Durham, NC USA
| | - Betsy C. Herold
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY USA
| | - Sallie R. Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, NC USA
| |
Collapse
|
32
|
Cui X, Cao Z, Wang S, Lee RB, Wang X, Murata H, Adler SP, McVoy MA, Snapper CM. Novel trimeric human cytomegalovirus glycoprotein B elicits a high-titer neutralizing antibody response. Vaccine 2018; 36:5580-5590. [PMID: 30082162 PMCID: PMC6556890 DOI: 10.1016/j.vaccine.2018.07.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/03/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Human cytomegalovirus (HCMV) is a major cause of disability in congenitally infected infants and in the immunosuppressed. There is currently no licensed prophylactic HCMV vaccine. The HCMV envelope glycoprotein B (gB) is considered a major vaccine target antigen based on its critical role in mediating viral-host cell fusion and thus viral entry. The natural conformation of HCMV gB within the viral envelope is a trimer, but there has been no reported success in producing a recombinant trimeric gB suitable for vaccine use. Phase II clinical trials of a monomeric recombinant gB protein demonstrated 50% efficacy in preventing HCMV infection in seronegative women of reproductive age, and in reducing viremia in solid organ transplantation recipients. We now report the production of a uniformly trimeric recombinant HCMV gB protein in Chinese ovary cells, as demonstrated by Western blot analysis under modified non-reducing conditions and size exclusion chromatography with multi-angle scattering. Immunization of mice with trimeric HCMV gB induced up to 11-fold higher serum titers of total gB-specific IgG relative to monomeric HCMV gB using Alum + CpG as adjuvants. Further, trimeric HCMV gB elicited 50-fold higher complement-independent and 20-fold higher complement-dependent HCMV neutralizing titers compared to monomeric HCMV gB using the fibroblast cell line, MRC-5, and up to 6-fold higher complement-independent and -dependent HCMV neutralizing titers using the epithelial cell line, ARPE-19. The markedly enhanced HCMV neutralizing activity in response to trimeric HCMV gB was also observed using an additional four distinct clinical HCMV isolates. These data support a role for trimeric HCMV gB as an important component for clinical testing of a prophylactic HCMV vaccine.
Collapse
Affiliation(s)
- Xinle Cui
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States.
| | - Zhouhong Cao
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Shuishu Wang
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Ronzo B Lee
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Xiao Wang
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Haruhiko Murata
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Stuart P Adler
- CMV Research Foundation, Richmond VA 23229, United States
| | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Clifford M Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| |
Collapse
|
33
|
Kobayashi R, Abe M, Oguri K, Torikai M, Nishimura T, Mori H, Koshizuka T, Inoue N. Analysis of relationships between polymorphisms in the genes encoding the pentameric complex and neutralization of clinical cytomegalovirus isolates. Vaccine 2018; 36:5983-5989. [DOI: 10.1016/j.vaccine.2018.08.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/16/2018] [Accepted: 08/18/2018] [Indexed: 10/28/2022]
|
34
|
Zika virus infection of first-trimester human placentas: utility of an explant model of replication to evaluate correlates of immune protection ex vivo. Curr Opin Virol 2018; 27:48-56. [PMID: 29172071 DOI: 10.1016/j.coviro.2017.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/09/2017] [Indexed: 01/08/2023]
Abstract
The emergence of congenital Zika virus (ZIKV) disease, with its devastating effects on the fetus, has prompted development of vaccines and examination of how ZIKV breaches the maternal-fetal barrier. Infection of placental and decidual tissue explants has demonstrated cell types at the uterine-placental interface susceptible to infection and suggests routes for transmission across the placenta and amniochorionic membrane. ZIKV replicates in proliferating Hofbauer cells within chorionic villi in placentas from severe congenital infection. Explants of anchoring villi recapitulate placental architecture and early-stage development and suggest infected Hofbauer cells disseminate virus to fetal blood vessels. ZIKV infection of explants represents a surrogate human model for evaluating protection against transmission by antibodies in vaccine recipients and passive immune formulations and novel therapeutics.
Collapse
|
35
|
Abstract
Why certain viruses cross the physical barrier of the human placenta but others do not is incompletely understood. Over the past 20 years, we have gained deeper knowledge of intrauterine infection and routes of viral transmission. This review focuses on human viruses that replicate in the placenta, infect the fetus, and cause birth defects, including rubella virus, varicella-zoster virus, parvovirus B19, human cytomegalovirus (CMV), Zika virus (ZIKV), and hepatitis E virus type 1. Detailed discussions include ( a) the architecture of the uterine-placental interface, ( b) studies of placental explants ex vivo that provide insights into the infection and spread of CMV and ZIKV to the fetal compartment and how these viruses undermine early development, and ( c) novel treatments and vaccines that limit viral replication and have the potential to reduce dissemination, vertical transmission and the occurrence of congenital disease.
Collapse
Affiliation(s)
- Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, California 94143, USA;
| |
Collapse
|
36
|
Falk JJ, Winkelmann M, Stöhr D, Alt M, Schrezenmeier H, Krawczyk A, Lotfi R, Sinzger C. Identification of Elite Neutralizers With Broad and Potent Neutralizing Activity Against Human Cytomegalovirus (HCMV) in a Population of HCMV-Seropositive Blood Donors. J Infect Dis 2018; 218:876-885. [DOI: 10.1093/infdis/jiy229] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/16/2018] [Indexed: 11/14/2022] Open
Affiliation(s)
| | - Martina Winkelmann
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood-Transfusion Service Baden-Württemberg–Hessen and University Hospital, Ulm, Germany
| | - Dagmar Stöhr
- Institute for Virology, Ulm University Medical Center, Ulm, Germany
| | - Mira Alt
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood-Transfusion Service Baden-Württemberg–Hessen and University Hospital, Ulm, Germany
- Institute for Transfusion Medicine, Ulm University, Ulm, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ramin Lotfi
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood-Transfusion Service Baden-Württemberg–Hessen and University Hospital, Ulm, Germany
- Institute for Transfusion Medicine, Ulm University, Ulm, Germany
| | | |
Collapse
|
37
|
Advancing Our Understanding of Protective Maternal Immunity as a Guide for Development of Vaccines To Reduce Congenital Cytomegalovirus Infections. J Virol 2018; 92:JVI.00030-18. [PMID: 29343580 DOI: 10.1128/jvi.00030-18] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the most common congenitally transmitted pathogen worldwide, impacting an estimated 1 million newborns annually. Congenital HCMV (cCMV) infection is a major global contributor to long-term neurologic deficits, including deafness, microcephaly, and neurodevelopmental delay, as well as to fetal loss and occasional infant mortality. Accordingly, design of a maternal vaccine to prevent cCMV continues to be a top public health priority. Nevertheless, we remain without a licensed vaccine. Maternal immunity provides partial protection, as the risk of vertical HCMV transmission from chronically infected mothers is reduced compared to settings in which the mother is newly infected during pregnancy. Therefore, an understanding of the maternal immune correlates of protection against cCMV is critical to informing design of an efficacious maternal vaccine. Although vaccine development is being assiduously pursued by a large number of pharmaceutical manufacturers, biotechnology organizations, and academic researchers, some pessimism has been expressed regarding the issue of whether a vaccine to protect against cCMV is possible. This pessimism is based on observations that natural immunity is not completely protective against maternal reinfection and congenital transmission. However, we assert that optimism regarding vaccine development is indeed justified, on the basis of accruing evidence of immune correlates of protection-readily achievable by vaccination-that are associated with reduced transmission of HCMV to the fetus in seronegative women. In light of the substantial burden on society conferred by cCMV infection, even a modest reduction in the occurrence of this fetal disease is an important public health goal and justifies aggressive clinical evaluation of vaccines currently in the pipeline.
Collapse
|
38
|
Itell HL, Nelson CS, Martinez DR, Permar SR. Maternal immune correlates of protection against placental transmission of cytomegalovirus. Placenta 2017; 60 Suppl 1:S73-S79. [PMID: 28456432 PMCID: PMC5650553 DOI: 10.1016/j.placenta.2017.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/07/2017] [Accepted: 04/13/2017] [Indexed: 02/02/2023]
Abstract
Human cytomegalovirus (HCMV) is the most common congenitally transmitted pathogen worldwide, impacting an estimated 1 million newborns annually. In a subset of infected infants, congenital HCMV causes severe, long-lasting sequelae, including deafness, microcephaly, neurodevelopmental delay, and even death. Accordingly, a maternal vaccine to prevent congenital HCMV infection continues to be a top public health priority. Nevertheless, all vaccines tested to date have failed to meet clinical trial endpoints. Maternal immunity provides partial protection against congenital HCMV transmission, as vertical transmission from seropositive mothers is relatively rare. Therefore, an understanding of the maternal immune correlates of protection against HCMV congenital infection will be critical to inform design of an efficacious maternal vaccine. This review summarizes our understanding of the innate and adaptive immune correlates of protection against congenital transmission of HCMV, and discusses the advantages and applications of a novel nonhuman primate model of congenital CMV transmission to aid in rational vaccine design and evaluation.
Collapse
Affiliation(s)
- Hannah L Itell
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Cody S Nelson
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - David R Martinez
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sallie R Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
39
|
Itell HL, Kaur A, Deere JD, Barry PA, Permar SR. Rhesus monkeys for a nonhuman primate model of cytomegalovirus infections. Curr Opin Virol 2017; 25:126-133. [PMID: 28888133 DOI: 10.1016/j.coviro.2017.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022]
Abstract
Human cytomegalovirus (HCMV) is the leading opportunistic viral infection in solid organ transplant patients and is the most common congenitally transmitted pathogen worldwide. Despite the significant burden of disease HCMV causes in immunosuppressed patients and infected newborns, there are no licensed preventative vaccines or effective immunotherapeutic treatments for HCMV, largely due to our incomplete understanding of the immune correlates of protection against HCMV infection and disease. Though CMV species-specificity imposes an additional challenge in defining a suitable animal model for HCMV, nonhuman primate (NHP) CMVs are the most genetically related to HCMV. In this review, we discuss the advantages and applicability of rhesus monkey models for studying HCMV infections and pathogenesis and ultimately informing vaccine development.
Collapse
Affiliation(s)
- Hannah L Itell
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Jesse D Deere
- Center for Comparative Medicine, Department of Pathology and Laboratory Medicine, University of California, Davis, CA, USA
| | - Peter A Barry
- Center for Comparative Medicine, Department of Pathology and Laboratory Medicine, University of California, Davis, CA, USA
| | - Sallie R Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
40
|
Bootz A, Karbach A, Spindler J, Kropff B, Reuter N, Sticht H, Winkler TH, Britt WJ, Mach M. Protective capacity of neutralizing and non-neutralizing antibodies against glycoprotein B of cytomegalovirus. PLoS Pathog 2017; 13:e1006601. [PMID: 28854233 PMCID: PMC5595347 DOI: 10.1371/journal.ppat.1006601] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 09/12/2017] [Accepted: 08/22/2017] [Indexed: 01/19/2023] Open
Abstract
Human cytomegalovirus (HCMV) is an important, ubiquitous pathogen that causes severe clinical disease in immunocompromised individuals, such as organ transplant recipients and infants infected in utero. Antiviral chemotherapy remains problematic due to toxicity of the available compounds and the emergence of viruses resistant to available antiviral therapies. Antiviral antibodies could represent a valuable alternative strategy to limit the clinical consequences of viral disease in patients. The envelope glycoprotein B (gB) of HCMV is a major antigen for the induction of virus neutralizing antibodies. However, the role of anti-gB antibodies in the course of the infection in-vivo remains unknown. We have used a murine CMV (MCMV) model to generate and study a number of anti-gB monoclonal antibodies (mAbs) with differing virus-neutralizing capacities. The mAbs were found to bind to similar antigenic structures on MCMV gB that are represented in HCMV gB. When mAbs were used in immunodeficient RAG-/- hosts to limit an ongoing infection we observed a reduction in viral load both with mAbs having potent neutralizing capacity in-vitro as well as mAbs classified as non-neutralizing. In a therapeutic setting, neutralizing mAbs showed a greater capacity to reduce the viral burden compared to non-neutralizing antibodies. Efficacy was correlated with sustained concentration of virus neutralizing mAbs in-vivo rather than their in-vitro neutralizing capacity. Combinations of neutralizing mAbs further augmented the antiviral effect and were found to be as potent in protection as polyvalent serum from immune animals. Prophylactic administration of mAbs before infection was also protective and both neutralizing and non-neutralizing mAbs were equally effective in preventing lethal infection of immunodeficient mice. In summary, our data argue that therapeutic application of potently neutralizing mAbs against gB represent a strategy to modify the outcome of CMV infection in immunodeficient hosts. When present before infection, both neutralizing and non-neutralizing anti-gB exhibited protective capacity.
Collapse
Affiliation(s)
- Anna Bootz
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Astrid Karbach
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Spindler
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Kropff
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nina Reuter
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- Institut für Biochemie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas H. Winkler
- Nikolaus-Fiebiger-Zentrum für Molekulare Medizin, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - William J. Britt
- Departments of Pediatrics, Microbiology and Neurobiology, Children's Hospital of Alabama, University of Alabama, School of Medicine, Birmingham, Alabama, United States of America
| | - Michael Mach
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
41
|
Impact of Antibodies and Strain Polymorphisms on Cytomegalovirus Entry and Spread in Fibroblasts and Epithelial Cells. J Virol 2017; 91:JVI.01650-16. [PMID: 28381568 DOI: 10.1128/jvi.01650-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 03/27/2017] [Indexed: 12/30/2022] Open
Abstract
Cytomegalovirus (CMV) entry into fibroblasts differs from entry into epithelial cells. CMV also spreads cell to cell and can induce syncytia. To gain insights into these processes, 27 antibodies targeting epitopes in CMV virion glycoprotein complexes, including glycoprotein B (gB), gH/gL, and the pentamer, were evaluated for their effects on viral entry and spread. No antibodies inhibited CMV spread in fibroblasts, including those with potent neutralizing activity against fibroblast entry, while all antibodies that neutralized epithelial cell entry also inhibited spread in epithelial cells and a correlation existed between the potencies of these two activities. This suggests that exposure of virions to the cell culture medium is obligatory during spread in epithelial cells but not in fibroblasts. In fibroblasts, the formation of syncytiumlike structures was impaired not only by antibodies to gB or gH/gL but also by antibodies to the pentamer, suggesting a potential role for the pentamer in promoting fibroblast fusion. Four antibodies reacted with linear epitopes near the N terminus of gH, exhibited strain specificity, and neutralized both epithelial cell and fibroblast entry. Five other antibodies recognized conformational epitopes in gH/gL and neutralized both fibroblast and epithelial cell entry. That these antibodies were strain specific for neutralizing fibroblast but not epithelial cell entry suggests that polymorphisms external to certain gH/gL epitopes may influence antibody neutralization during fibroblast but not epithelial cell entry. These findings may have implications for elucidating the mechanisms of CMV entry, spread, and antibody evasion and may assist in determining which antibodies may be most efficacious following active immunization or passive administration.IMPORTANCE Cytomegalovirus (CMV) is a significant cause of birth defects among newborns infected in utero and morbidity and mortality in transplant and AIDS patients. Monoclonal antibodies and vaccines targeting humoral responses are under development for prophylactic or therapeutic use. The findings reported here (i) confirm that cell-to-cell spread of CMV is sensitive to antibody inhibition in epithelial cells but not fibroblasts, (ii) demonstrate that antibodies can restrict the formation in vitro of syncytiumlike structures that resemble syncytial cytomegalic cells that are associated with CMV disease in vivo, and (iii) reveal that neutralization of CMV by antibodies to certain epitopes in gH or gH/gL is both strain and cell type dependent and can be governed by polymorphisms in sequences external to the epitopes. These findings serve to elucidate the mechanisms of CMV entry, spread, and antibody evasion and may have important implications for the development of CMV vaccines and immunotherapeutics.
Collapse
|
42
|
Cytomegalovirus Virions Shed in Urine Have a Reversible Block to Epithelial Cell Entry and Are Highly Resistant to Antibody Neutralization. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00024-17. [PMID: 28404573 DOI: 10.1128/cvi.00024-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/04/2017] [Indexed: 12/31/2022]
Abstract
Cytomegalovirus (CMV) causes sensorineural hearing loss and developmental disabilities in newborns when infections are acquired in utero Pregnant women may acquire CMV from oral exposure to CMV in urine or saliva from young children. Neutralizing antibodies in maternal saliva have the potential to prevent maternal infection and, in turn, fetal infection. As CMV uses different viral glycoprotein complexes to enter different cell types, the first cells to be infected in the oral cavity could determine the type of antibodies needed to disrupt oral transmission. Antibodies targeting the pentameric complex (PC) should block CMV entry into epithelial cells but not into fibroblasts or Langerhans cells (which do not require the PC for entry), while antibodies targeting glycoprotein complexes gB or gH/gL would be needed to block entry into fibroblasts, Langerhans cells, or other cell types. To assess the potential for antibodies to disrupt oral acquisition, CMV from culture-positive urine samples (uCMV) was used to study cell tropisms and sensitivity to antibody neutralization. uCMV entered epithelial cells poorly compared with the entry into fibroblasts. CMV-hyperimmune globulin or monoclonal antibodies targeting gB, gH/gL, or the PC were incapable of blocking the entry of uCMV into either fibroblasts or epithelial cells. Both phenotypes were lost after one passage in cultured fibroblasts, suggestive of a nongenetic mechanism. These results suggest that uCMV virions have a reversible block to epithelial cell entry. Antibodies may be ineffective in preventing maternal oral CMV acquisition but may limit viral spread in blood or tissues, thereby reducing or preventing fetal infection and disease.
Collapse
|
43
|
Plasmablast Response to Primary Rhesus Cytomegalovirus (CMV) Infection in a Monkey Model of Congenital CMV Transmission. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00510-16. [PMID: 28298291 DOI: 10.1128/cvi.00510-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/06/2017] [Indexed: 11/20/2022]
Abstract
Human cytomegalovirus (HCMV) is the most common congenital infection worldwide and the leading infectious cause of neurologic deficits and hearing loss in newborns. Development of a maternal HCMV vaccine to prevent vertical virus transmission is a high priority, yet protective maternal immune responses following acute infection are poorly understood. To characterize the maternal humoral immune response to primary CMV infection, we investigated the plasmablast and early antibody repertoire using a nonhuman primate model with two acutely rhesus CMV (RhCMV)-infected animals-a CD4+ T cell-depleted dam that experienced fetal loss shortly after vertical RhCMV transmission and an immunocompetent dam that did not transmit RhCMV to her infant. Compared to the CD4+ T cell-depleted dam that experienced fetal loss, the immunocompetent, nontransmitting dam had a more rapid and robust plasmablast response that produced a high proportion of RhCMV-reactive antibodies, including the first identified monoclonal antibody specific for soluble and membrane-associated RhCMV envelope glycoprotein B (gB). Additionally, we noted that plasmablast RhCMV-specific antibodies had variable gene usage and maturation similar to those observed in a monkey chronically coinfected with simian immunodeficiency virus (SIV) and RhCMV. This study reveals characteristics of the early maternal RhCMV-specific humoral immune responses to primary RhCMV infection in rhesus monkeys and may contribute to a future understanding of what antibody responses should be targeted by a vaccine to eliminate congenital HCMV transmission. Furthermore, the identification of an RhCMV gB-specific monoclonal antibody underscores the possibility of modeling future HCMV vaccine strategies in this nonhuman primate model.
Collapse
|
44
|
Pereira L, Tabata T, Petitt M, Fang-Hoover J. Congenital cytomegalovirus infection undermines early development and functions of the human placenta. Placenta 2017; 59 Suppl 1:S8-S16. [PMID: 28477968 DOI: 10.1016/j.placenta.2017.04.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 04/19/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
Congenital human cytomegalovirus (HCMV) infection is a major viral cause of birth defects, including microcephaly, neurological deficits, loss of hearing and vision, and intrauterine growth restriction. Despite its public health significance, there is no approved treatment for congenital infection during pregnancy; existing antivirals have unacceptable toxicities. The mechanisms of HCMV-induced placental injury, reduced capacity for compensatory development and transmission to the fetus are poorly understood, limiting the development of alternative strategies for clinical management of the disease. Recently, self-renewing, multipotent trophoblast progenitor cells (TBPCs) were reported to reside in the chorion of the human placenta and differentiate into the mature trophoblast subtypes - transport syncytiotrophoblasts and invasive cytotrophoblasts - forming chorionic villi, the functional units of the placenta. HCMV infects TBPCs, reducing the population of progenitor cells and their functional capacity to self-renew, migrate and differentiate. Human TBPCs and chorionic villus explants from first trimester represent relevant models for evaluating efficacies of new antiviral agents in protecting and restoring growth of the developing placenta in response to adverse conditions. Correlating pathology from complications of congenital HCMV infection with impaired development in the tissue environment of anchoring villus explants and defects in TBPC differentiation may enable identification of molecular pathways that could serve as targets for intervention. Here we summarize studies that could open up novel avenues of research on potential therapeutics to sustain placental development, promote differentiation and improve function and pregnancy outcomes.
Collapse
Affiliation(s)
- Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States.
| | - Takako Tabata
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States
| | - Matthew Petitt
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States
| |
Collapse
|
45
|
Schleiss MR. Cytomegalovirus vaccines under clinical development. J Virus Erad 2016; 2:198-207. [PMID: 27781101 PMCID: PMC5075346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Congenital cytomegalovirus (CMV) infection is the most common infectious cause of disability in newborn infants. CMV also causes serious disease in solid organ (SOT) and haematopoietic stem cell transplant (HSCT) recipients. In otherwise healthy children and adults, primary CMV infection rarely causes illness. However, even asymptomatic CMV infections may predispose an individual towards an increased risk of atherosclerosis, cancer and immune senescence over the life course, although such associations remain controversial. Thus, although a vaccine against congenital CMV infection would have the greatest public health impact and cost-effectiveness, arguably all populations could benefit from an effective immunisation against this virus. Currently there are no licensed CMV vaccines, but there is increased interest in developing and testing potential candidates, driven by the demonstration that a recombinant CMV glycoprotein B (gB) vaccine has some efficacy in prevention of infection in young women and adolescents, and in CMV-seronegative SOT recipients. In this review, the recent and current status of candidate CMV vaccines is discussed. Evolving concepts about proposed correlates of protective immunity in different target populations for CMV vaccination, and how these differences impact current clinical trials, are also reviewed.
Collapse
Affiliation(s)
- Mark R Schleiss
- Department of Pediatrics,
University of Minnesota Medical School,
Minneapolis,
MN,
USA,Corresponding author: Mark R Schleiss,
Department of Pediatrics,
University of Minnesota Medical School,
Division of Pediatric Infectious Diseases and Immunology,
Center for Infectious Diseases and Microbiology Translational Research,
2001 6th Street SE,
Minneapolis,
MN55455,
USA
| |
Collapse
|
46
|
|
47
|
Tabata T, Petitt M, Fang-Hoover J, Zydek M, Pereira L. Persistent Cytomegalovirus Infection in Amniotic Membranes of the Human Placenta. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2970-2986. [PMID: 27638253 DOI: 10.1016/j.ajpath.2016.07.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/25/2016] [Accepted: 07/14/2016] [Indexed: 12/24/2022]
Abstract
Human cytomegalovirus (HCMV) is the leading viral cause of birth defects, including microcephaly, neurological deficits, hearing impairment, and vision loss. We previously reported that epithelial cells in amniotic membranes of placentas from newborns with intrauterine growth restriction and underlying congenital HCMV infection contain viral proteins in cytoplasmic vesicles. Herein, we immunostained amniotic membranes from 51 placentas from symptomatic and asymptomatic congenital infection with HCMV DNA in amniotic fluid and/or newborn saliva, intrauterine growth restriction, preterm deliveries, and controls. We consistently observed HCMV proteins in amniotic epithelial cells (AmEpCs) from infected placentas, sometimes with aberrant morphology. Primary AmEpCs isolated from mid-gestation placentas infected with pathogenic VR1814 proliferated and released infectious progeny for weeks, producing higher virus titers than late-gestation cells that varied by donor. In contrast to intact virion assembly compartments in differentiated retinal pigment epithelial cells, infected AmEpCs made dispersed multivesicular bodies. Primary AmEpCs and explants of amniochorionic membranes from mid-gestation placentas formed foci of infection, and interferon-β production was prolonged. Infected AmEpCs up-regulated anti-apoptotic proteins survivin and Bcl-xL by mechanisms dependent and independent of the activated STAT3. Amniotic membranes naturally expressed both survivin and Bcl-xL, indicating that fetal membranes could foster persistent viral infection. Our results suggest strengthening innate immune responses and reducing viral functions could suppress HCMV infection in the fetal compartment.
Collapse
Affiliation(s)
- Takako Tabata
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California
| | - Matthew Petitt
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California
| | - Martin Zydek
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California
| | - Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
48
|
Tabata T, Petitt M, Puerta-Guardo H, Michlmayr D, Wang C, Fang-Hoover J, Harris E, Pereira L. Zika Virus Targets Different Primary Human Placental Cells, Suggesting Two Routes for Vertical Transmission. Cell Host Microbe 2016; 20:155-66. [PMID: 27443522 PMCID: PMC5257282 DOI: 10.1016/j.chom.2016.07.002] [Citation(s) in RCA: 361] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022]
Abstract
Zika virus (ZIKV) infection during pregnancy is linked to severe birth defects, but mother-to-fetus transmission routes are unknown. We infected different primary cell types from mid- and late-gestation placentas and explants from first-trimester chorionic villi with the prototype Ugandan and a recently isolated Nicaraguan ZIKV strain. ZIKV infects primary human placental cells and explants-cytotrophoblasts, endothelial cells, fibroblasts, and Hofbauer cells in chorionic villi and amniotic epithelial cells and trophoblast progenitors in amniochorionic membranes-that express Axl, Tyro3, and/or TIM1 viral entry cofactors. ZIKV produced NS3 and E proteins and generated higher viral titers in amniotic epithelial cells from mid-gestation compared to late-gestation placentas. Duramycin, a peptide that binds phosphatidylethanolamine in enveloped virions and precludes TIM1 binding, reduced ZIKV infection in placental cells and explants. Our results suggest that ZIKV spreads from basal and parietal decidua to chorionic villi and amniochorionic membranes and that targeting TIM1 could suppress infection at the uterine-placental interface.
Collapse
Affiliation(s)
- Takako Tabata
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew Petitt
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Daniela Michlmayr
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Chunling Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA.
| | - Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
49
|
Manghera A, McLean GR. Human cytomegalovirus vaccination: progress and perspectives of recombinant gB. Future Virol 2016. [DOI: 10.2217/fvl-2016-0039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A vaccine for Human cytomegalovirus (HCMV) remains a high priority as complications following infection are observed in immunocompromised individuals and in congenitally infected neonates. Numerous preclinical and clinical studies have investigated vaccine strategies ranging from live attenuated preparations, nucleic acid-based approaches and recombinant delivery systems to subunit vaccines. These have defined the importance of both cell-mediated and humoral immunity to viral gB in the control of HCMV infection. This review will cover clinical trials investigating vaccine approaches that have incorporated gB and discuss the future perspectives of the recombinant gB subunit vaccine for HCMV.
Collapse
Affiliation(s)
- Avneet Manghera
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Gary R McLean
- National Heart & Lung Institute, Imperial College London, London, UK
- Cellular & Molecular Immunology Research Centre, London Metropolitan University, London, UK
| |
Collapse
|
50
|
An Ex vivo culture model for placental cytomegalovirus infection using slices of Guinea pig placental tissue. Placenta 2015; 37:85-8. [PMID: 26625961 DOI: 10.1016/j.placenta.2015.10.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 01/01/2023]
Abstract
Congenital infection with human cytomegalovirus (CMV) through the placenta is one of the major causes of birth and developmental abnormalities. Guinea pig CMV (GPCMV) causes in utero infection, which makes its animal models useful for studies on congenital diseases. Here, we established an ex vivo culture method for tissue slices prepared from guinea pig placentas and demonstrated that viral spread in the model resembles those in the placenta of GPCMV-infected animals and that the infection is independent of the pentameric glycoprotein complex for endothelial/epithelial cell tropism. Thus, this model affords a useful tool for pathobiological studies on CMV placental infection.
Collapse
|