1
|
Permar SR, Schleiss MR, Plotkin SA. A vaccine against cytomegalovirus: how close are we? J Clin Invest 2025; 135:e182317. [PMID: 39744948 DOI: 10.1172/jci182317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
The pursuit of a vaccine against the human cytomegalovirus (HCMV) has been ongoing for more than 50 years. HCMV is the leading infectious cause of birth defects, including damage to the brain, and is a common cause of complications in organ transplantation. The complex biology of HCMV has made vaccine development difficult, but a recent meeting sponsored by the National Institute of Allergy and Infectious Diseases in September of 2023 brought together experts from academia, industry, and federal agencies to discuss progress in the field. The meeting reviewed the status of candidate HCMV vaccines under study and the challenges in clinical trial design in demonstrating efficacy against congenital CMV infection or the reduction of HCMV disease following solid organ transplantation or hematopoietic stem cell transplantation. Discussion in the meeting revealed that, with the numerous candidate vaccines that are under study, it is clear that a safe and effective HCMV vaccine is within reach. Meeting attendees achieved a consensus opinion that even a partially effective vaccine would have a major effect on the global health consequences of HCMV infection.
Collapse
Affiliation(s)
- Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical Center, New York, New York, USA
| | - Mark R Schleiss
- Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Vaxconsult, Doylestown, Pennsylvania, USA
| |
Collapse
|
2
|
Khalil A, Heath PT, Jones CE, Soe A, Ville YG. Congenital Cytomegalovirus Infection: Update on Screening, Diagnosis and Treatment: Scientific Impact Paper No. 56. BJOG 2025; 132:e42-e52. [PMID: 39434207 DOI: 10.1111/1471-0528.17966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 10/23/2024]
Abstract
Cytomegalovirus (CMV) is the most common cause of viral infection in newborn babies, and affects 1 in 200 of all live born infants in high-income countries; and 1 in 71 in low- and middle-income countries. It is a major cause of hearing loss and brain damage. Women may get CMV infection for the first time during pregnancy (primary infection) or may experience 'non-primary' infection, either by reactivation of previous CMV infection or by a new infection with a different strain of the virus. The most common source of infection to pregnant women is the saliva and urine of young children. Therefore, all pregnant women, especially those in regular contact with young children, should be informed about hygiene-based measures to reduce the risks, e.g. handwashing. The UK National Screening Committee recommends against universal antenatal or newborn screening for CMV. Testing for CMV is usually offered only to women who develop symptoms of influenza, glandular fever or hepatitis (liver inflammation) during pregnancy, or for those whom a routine ultrasound scan detects fetal anomalies that suggests possible CMV infection. The risk of harm to the fetus is greatest following primary CMV infection of the woman in early pregnancy, and appears to be very low following infection after 12 weeks of pregnancy. Babies with CMV infection at birth may have jaundice, a rash, enlarged liver or spleen, a small brain, or be small for their gestational age. Around 1 in 8 babies born with CMV infection will have clinically detectable signs at birth. The rest will not have any features detectable by clinical examination alone. Therefore, all infants with CMV infection at birth should be followed up at a minimum of up to 2 years of age or later, depending upon the disease status, to check hearing and brain development. Following primary CMV infection in the first 12 weeks of pregnancy, if the woman starts taking the antiviral medicine valaciclovir (valacyclovir) it reduces the risk of the baby becoming infected. Where CMV infection of the fetus in the womb has been confirmed (by amniocentesis, for example), regular ultrasound scans should be offered every 2-3 weeks until birth. Detailed assessment of the fetal brain is an essential part of these scans. Where maternal CMV infection occurs, but fetal infection is not confirmed, repeated ultrasound scans of the fetus should be offered every 2-3 weeks until birth. In infected fetuses, as well as ultrasound scans, an MRI scan of the brain should be offered at 28-32 weeks of gestation (and sometimes repeated 3-4 weeks later) to assess for any signs of harm to the fetal brain. All babies born to women with confirmed or suspected CMV infection should be tested for CMV with a urine or saliva sample within the first 21 days of life. In newborns with symptomatic CMV infection at birth, treatment with antiviral medicine (valganciclovir or ganciclovir) can reduce hearing loss in 5 out of 6 babies, and improve long-term brain development outcomes in some. There is no licensed vaccine for CMV.
Collapse
|
3
|
Luna E, Ruiz S, Garinot M, Chavagnac C, Agrawal P, Escobar J, Revet L, Asensio MJ, Piras F, Fang FG, Drake DR, Rokbi B, Larocque D, Haensler J. SPA14 liposomes combining saponin with fully synthetic TLR4 agonist provide adjuvanticity to hCMV vaccine candidate. NPJ Vaccines 2024; 9:253. [PMID: 39702373 DOI: 10.1038/s41541-024-01046-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
In the aim of designing and developing a novel saponin-based adjuvant system, we combined the QS21 saponin with low microgram amounts of the fully synthetic TLR4 agonist, E6020, in cholesterol-containing liposomes. The resulting adjuvant system, termed SPA14, appeared as a long-term stable and homogeneous suspension of mostly unilamellar and a few multilamellar vesicles, with an average hydrodynamic diameter of 93 nm, when formulated in citrate buffer at pH 6.0-6.5. When compared in an in vitro human innate immunity construct to AS01B, the QS21/MPL® liposomal adjuvant system of GSK, and with QS21-Liposomes used as benchmarks, SPA14 displayed the strongest immunostimulatory potential based on antigen-presenting cell (APC) activation and cytokine secretion, which was essentially driven by the highly active E6020 agonist in this assay. When tested as an adjuvant in vivo with human cytomegalovirus glycoprotein B (gB) and pentamer complex (PC) as test antigens, SPA14 was generally well tolerated and as active as AS01B for the induction of long-lasting CMV-neutralizing antibodies in mice and non-human primates (NHPs). Both adjuvants promoted the induction of Th-1 responses based on IgG2c production in mice and IFN-γ production in mice and NHPs, but in mice, a higher level of Th-2 cytokines (IL-5) and higher IgG1 over IgG2c secreting cells ratios were obtained with SPA14 indicating that the adjuvant profile of SPA14 could be less Th-1 biased than that of AS01B. From a developability standpoint, SPA14 could be manufactured by a simple and scalable ethanol injection method, owing to the high solubility in ethanol of all its lipidic components, including E6020. Furthermore, E6020 is a single molecule, well-characterized fully synthetic TLR4 agonist constructed in eight synthetic steps from entirely crystalline starting materials and intermediates via an optimized high-yield synthetic route. Overall, our data suggest that SPA14 is a viable, easy-to-manufacture, potent novel adjuvant system that could be broadly applicable as a ready-to-mix adjuvant in the form of a long-term stable liquid formulation.
Collapse
Affiliation(s)
- Ernesto Luna
- Sanofi Vaccines business unit, R&D, Orlando, FL, USA
| | - Sophie Ruiz
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | - Marie Garinot
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | | | - John Escobar
- Sanofi Vaccines business unit, R&D, Orlando, FL, USA
| | - Laurent Revet
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | - Fabienne Piras
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | | | - Bachra Rokbi
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | - Jean Haensler
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France.
| |
Collapse
|
4
|
Billette de Villemeur A, Hoen B, Billaud E, Deruelle P, Goueslard K, Halley des Fontaines V, Minodier P, Parent B, Pozzetto B, Revest M, Salmi LR. Current evidence gaps to support systematic cytomegalovirus screening in pregnancy. EClinicalMedicine 2024; 78:102941. [PMID: 39640941 PMCID: PMC11617987 DOI: 10.1016/j.eclinm.2024.102941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024] Open
Abstract
The benefits of screening for cytomegalovirus (CMV) infection during pregnancy remain a topic of debate. To date, no randomized trial has compared the impact of screening versus routine management on the prevention of severe sequelae in newborns. Furthermore, it is unclear what actions can be taken in case of a positive screening given that there is limited evidence of effective interventions as no treatments showed significant effect on the frequency of congenital cytomegalovirus infections and, as additional challenge, the window for effective treatment initiation after maternal infection is narrow, estimated to be as short as five weeks. Universal screening of all pregnant women could lead to a high number of false positives. There are also concerns regarding the cost-effectiveness of universal screening and the capacity of healthcare professionals that may struggle to manage the increased workload, and we argue that the conditions for implementing such a programme are not yet met. In this Viewpoint we aim at highlighting these challenges and stimulating the forthcoming discussion on how to fill the gaps before CMV screening in pregnancy could be adopted as a standard practice.
Collapse
Affiliation(s)
| | - Bruno Hoen
- Haut Conseil de Santé Publique, Paris F-75000, France
- École de santé publique—UMR 1319 INSPIIRE, Université de Lorraine, Nancy, France
| | - Eric Billaud
- Services de Maladies Infectieuses, CHU de Nantes, Nantes 44000, France
| | - Philippe Deruelle
- Service de Gynécologie-obstétrique, CHU de Montpellier, Montpellier 34000, France
| | - Karine Goueslard
- Department of Clinical Research and Innovation (DRCI), Clinical Research Unit Methodological Support Network (USMR), University Hospital, Dijon, France
| | | | - Philippe Minodier
- Urgences Enfants, CHU Nord, Chemin des Bourrelly, Marseille 13015, France
| | - Bertrand Parent
- EHESP (Ecole des Hautes Etudes en Santé Publique), Rennes 35000, France
- Univ Rennes, EHESP, CNRS, Inserm, Arènes-UMR 6051, RSMS-U 1309, Rennes, France
| | - Bruno Pozzetto
- Service des agents Infectieux et d’hygiène – Hôpital Nord - Centre hospitalier Universitaire (CHU) de Saint-Étienne, Saint-Etienne, France
- Groupe sur l’immunité des muqueuses et agents pathogènes (Gimap) – Centre International de Recherche en Infectiologie (Ciri) – Université Claude-Bernard-Lyon-1 – Institut National de la santé et de la Recherche médicale (Inserm) U1111 – Unité mixte de Recherche 5308 – Centre National de la Recherche scientifique (CNRS) – École Normale supérieure de Lyon – Université Jean-Monnet de Saint-Étienne, Saint-Étienne, France
| | - Matthieu Revest
- Service des Maladies Infectieuses et Réanimation Médicale, CHU Rennes, Rennes Cedex 35033, France
- UMR_1230 BRM (Bacterial RNAs and Medicine), Inserm, Université de Rennes, France
| | - Louis-Rachid Salmi
- Univ. Bordeaux, ISPED, Centre INSERM U1219, Bordeaux Population Health, Bordeaux F-33000, France
- INSERM, ISPED, Centre INSERM U1219, Bordeaux Population Health, Bordeaux F-33000, France
| |
Collapse
|
5
|
Schleiss MR, Crooks CM, Karthigeyan KP, Kruc RM, Otero CE, Wang HY(S, Permar SR, Plotkin SA, Gautam R. Proceedings of the Conference "CMV Vaccine Development-How Close Are We?" (27-28 September 2023). Vaccines (Basel) 2024; 12:1231. [PMID: 39591134 PMCID: PMC11598149 DOI: 10.3390/vaccines12111231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 11/28/2024] Open
Abstract
Congenital cytomegalovirus (cCMV) is the most common infectious cause of disability in children, including sensorineural hearing loss. There is interest in developing a pre-conception vaccine that could confer protective immunity on a woman of child-bearing age, hence resulting in a reduced cCMV disease burden. Other populations, including solid organ transplant (SOT) and hematopoietic stem cell transplant (HSCT) patients, could also benefit from CMV vaccination. To review and discuss vaccines that are in clinical development, a workshop, sponsored by the National Institutes of Health (NIH) and the National Institute of Allergy and Infectious Diseases (NIAID), was empaneled. At this workshop, correlates of protective immunity against CMV, epidemiologic features of CMV transmission, and vaccine platforms in development were reviewed. Representatives from academia, pharma, and the NIH engaged in discussion on the current state-of-the-art in CMV vaccinology. A summary of the presentations from this is provided in this report.
Collapse
Affiliation(s)
- Mark R. Schleiss
- Division of Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Chelsea M. Crooks
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Krithika P. Karthigeyan
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Rebecca M. Kruc
- Department of Pediatrics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Claire E. Otero
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Hsuan-Yuan (Sherry) Wang
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, 1300 York Ave Box 65, New York, NY 10065, USA;
| | - Stanley A. Plotkin
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Rajeev Gautam
- Program Officer at Virology Branch, Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fisher’s Lane, Rockville, MD 20892, USA;
| |
Collapse
|
6
|
Pesch MH, Schleiss MR, Plotkin SA, Permar SR, Gautam R. Beyond Awareness: Hope for a CMV Vaccine! An Introduction to the Conference, "CMV Vaccine Development-How Close Are We?" (27-28 September 2023). Vaccines (Basel) 2024; 12:1226. [PMID: 39591129 PMCID: PMC11598857 DOI: 10.3390/vaccines12111226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/25/2024] [Accepted: 10/14/2024] [Indexed: 11/28/2024] Open
Abstract
Congenital cytomegalovirus (cCMV) is the most common infectious cause of disability in children. The major theme of this National Institute of Allergy and Infectious Diseases (NIAID) workshop, "CMV Vaccine Development-How Close Are We?", was to report progress on the development of a pre-conception vaccine that could confer protective immunity for women of child-bearing age. Such a vaccine could result in a reduced cCMV disease burden, although other populations, including solid organ transplant and hematopoietic stem cell transplant patients, could benefit as well. To frame the compelling need for a cCMV vaccine, a keynote lecture by Dr. Megan Pesch, immediate past-president of the National CMV Foundation and a leading cCMV researcher from the University of Michigan, was given. This manuscript provides a summary of Dr. Pesch's presentation from this workshop, which was written as the introductory conference report for the meeting.
Collapse
Affiliation(s)
- Megan H. Pesch
- Division of Developmental and Behavioral Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Mark R. Schleiss
- Division of Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Stanley A. Plotkin
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, 1300 York Ave Box 65, New York, NY 10065, USA;
| | - Rajeev Gautam
- Program Officer at Virology Branch, Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fisher’s Lane, Rockville, MD 20892, USA;
| |
Collapse
|
7
|
Lembo A, Molinaro A, De Castro C, Berti F, Biagini M. Impact of glycosylation on viral vaccines. Carbohydr Polym 2024; 342:122402. [PMID: 39048237 DOI: 10.1016/j.carbpol.2024.122402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/24/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024]
Abstract
Glycosylation is the most prominent modification important for vaccines and its specific pattern depends on several factors that need to be considered when developing a new biopharmaceutical. Tailor-made glycosylation can be exploited to develop more effective and safer vaccines; for this reason, a deep understanding of both glycoengineering strategies and glycans structures and functions is required. In this review we discuss the recent advances concerning glycoprotein expression systems and the explanation of glycans immunomodulation mechanisms. Furthermore, we highlight how glycans tune the immunological properties among different vaccines platforms (whole virus, recombinant protein, nucleic acid), also comparing commercially available formulations and describing the state-of-the-art analytical technologies for glycosylation analysis. The whole review stresses the aspect of glycoprotein glycans as a potential tool to overcome nowadays medical needs in vaccine field.
Collapse
Affiliation(s)
- Antonio Lembo
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy; GSK, Siena, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Cristina De Castro
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
8
|
Spencer Clinton JL, Hoornweg TE, Tan J, Peng R, Schaftenaar W, Rutten VPMG, de Haan CAM, Ling PD. The EEHV1A gH/gL complex elicits humoral and cell-mediated immune responses in mice. Vaccine 2024; 42:126227. [PMID: 39180978 DOI: 10.1016/j.vaccine.2024.126227] [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: 04/24/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Elephant endotheliotropic herpesvirus (EEHV) causes lethal hemorrhagic disease (HD) in Asian and African elephants. Although rapid detection of viremia and supportive treatments may improve survival rates, an effective vaccine would mitigate the devastating effects of this virus. In elephants, chronic infection with EEHV leads to adaptive immunity against glycoproteins gB and gH/gL, the core entry machinery for most herpesviruses. We previously evaluated two EEHV gB vaccines in mice but not a gH/gL vaccine. Here, we found that inoculation of mice with an adjuvanted EEHV gH/gL subunit vaccine induced a significant antibody response that was similar to the response observed in elephants chronically infected with EEHV. Moreover, the gH/gL heterodimer elicited polyfunctional T cells with a Th1 phenotype but no detectable Th2 response. These results suggest that gH/gL, possibly in combination with gB, may be suitable immunogens for a vaccine comprising herpesvirus glycoproteins that are known to mediate cell entry and infection.
Collapse
Affiliation(s)
- Jennifer L Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Tabitha E Hoornweg
- Department of Biomolecular Health Sciences, Div of Infectious Diseases and Immunology, Fac of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584, CL, Utrecht, Netherlands.
| | - Jie Tan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Rongsheng Peng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Willem Schaftenaar
- Veterinary Advisor EAZA Elephant TAG, Rotterdam Zoo, Blijdorplaan 8, 3041, JG, Rotterdam, Netherlands.
| | - Victor P M G Rutten
- Department of Biomolecular Health Sciences, Div of Infectious Diseases and Immunology, Fac of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584, CL, Utrecht, Netherlands; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa.
| | - Cornelis A M de Haan
- Department of Biomolecular Health Sciences, Div of Infectious Diseases and Immunology, Fac of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584, CL, Utrecht, Netherlands.
| | - Paul D Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Karner D, Kvestak D, Kucan Brlic P, Cokaric Brdovcak M, Lisnic B, Brizic I, Juranic Lisnic V, Golemac M, Tomac J, Krmpotic A, Karkeni E, Libri V, Mella S, Legname G, Altmeppen HC, Hasan M, Jonjic S, Lenac Rovis T. Prion protein alters viral control and enhances pathology after perinatal cytomegalovirus infection. Nat Commun 2024; 15:7754. [PMID: 39237588 PMCID: PMC11377837 DOI: 10.1038/s41467-024-51931-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
Cytomegalovirus (CMV) infection poses risks to newborns, necessitating effective therapies. Given that the damage includes both viral infection of brain cells and immune system-related damage, here we investigate the involvement of cellular prion protein (PrP), which plays vital roles in neuroprotection and immune regulation. Using a murine model, we show the role of PrP in tempering neonatal T cell immunity during CMV infection. PrP-null mice exhibit enhanced viral control through elevated virus-specific CD8 T cell responses, leading to reduced viral titers and pathology. We further unravel the molecular mechanisms by showing CMV-induced upregulation followed by release of PrP via the metalloproteinase ADAM10, impairing CD8 T cell response specifically in neonates. Additionally, we confirm PrP downregulation in human CMV (HCMV)-infected fibroblasts, underscoring the broader relevance of our observations beyond the murine model. Furthermore, our study highlights how PrP, under the stress of viral pathogenesis, reveals its impact on neonatal immune modulation.
Collapse
Affiliation(s)
- Dubravka Karner
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Daria Kvestak
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Paola Kucan Brlic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | | | - Berislav Lisnic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Ilija Brizic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Vanda Juranic Lisnic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Mijo Golemac
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Tomac
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Astrid Krmpotic
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Esma Karkeni
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Valentina Libri
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Sebastien Mella
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Giuseppe Legname
- Department of Neuroscience, Prion Biology Laboratory, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Milena Hasan
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Stipan Jonjic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Tihana Lenac Rovis
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
10
|
Liberati C, Sturniolo G, Brigadoi G, Cavinato S, Visentin S, Cosmi E, Donà D, Rampon O. Burden of Congenital CMV Infection: A Narrative Review and Implications for Public Health Interventions. Viruses 2024; 16:1311. [PMID: 39205285 PMCID: PMC11360585 DOI: 10.3390/v16081311] [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: 07/19/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Cytomegalovirus causes the most common congenital infection worldwide. With most infants asymptomatic at birth, the few affected may present with variable clinical scenarios, from isolated hearing loss to severe neurologic impairment. Public health interventions include all actions at the health system, community, and individual levels that aim at reducing the burden of congenital Cytomegalovirus. This review examines the literature on maternal and neonatal screening programs in light of current evidence for treatment and the development of vaccines against Cytomegalovirus. Potential biases and benefits of these interventions are outlined, with the objective of increasing awareness about the problem and providing readers with data and critical tools to participate in this ongoing debate.
Collapse
Affiliation(s)
- Cecilia Liberati
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Giulia Sturniolo
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Giulia Brigadoi
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Silvia Cavinato
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35126 Padua, Italy;
| | - Silvia Visentin
- Department of Women’s and Children’s Health, Gynecological and Obstetric Clinic, Padua University Hospital, 35126 Padua, Italy; (S.V.); (E.C.)
| | - Erich Cosmi
- Department of Women’s and Children’s Health, Gynecological and Obstetric Clinic, Padua University Hospital, 35126 Padua, Italy; (S.V.); (E.C.)
| | - Daniele Donà
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Osvalda Rampon
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| |
Collapse
|
11
|
d’Angelo P, Zavaglio F, Gabanti E, Zelini P, Fornara C, Bernuzzi S, Spinillo A, Lilleri D, Baldanti F. Evaluation of the In Vitro Capacity of Anti-Human Cytomegalovirus Antibodies to Initiate Antibody-Dependent Cell Cytotoxicity. Microorganisms 2024; 12:1355. [PMID: 39065122 PMCID: PMC11278886 DOI: 10.3390/microorganisms12071355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
In the setting of infectious diseases, antibodies show different functions beyond neutralizing activity. In this study, we investigated the activation of NK cells in vitro in the presence of human cytomegalovirus (HCMV)-specific antibodies and their potential role in the control of HCMV infection through antibody-dependent cell cytotoxicity (ADCC). Retinal pigmented epithelial cells (ARPE-19) infected with the HCMV strain VR1814 were co-cultured with cytokine-activated peripheral blood mononuclear cells (PBMCs) in the presence of sera collected from 23 HCMV-seropositive and 9 HCMV-seronegative donors. Moreover, 13 pregnant women sampled 3 and 6 months after HCMV primary infection and 13 pregnant women with pre-conception immunity were tested and compared. We determined the percentage of activated NK cells via the analysis of CD107a expression as a marker of degranulation. Significantly higher levels of NK-cell activation were observed using 1/100 and 1/10 dilutions of sera from HCMV-seropositive individuals, and when cells were infected for 96 and 120 h, suggesting that NK cells are activated by antibodies directed against late antigens. In the absence of serum NK cells, activation was negligible. In seropositive subjects, the median percentages of CD107a-positive NK cells in the presence of autologous serum and pooled HCMV-positive serum were similar (14.03% [range 0.00-33.56] and 12.42% [range 1.01-46.00], respectively), while NK-cell activation was negligible using an HCMV-negative serum pool. In HCMV-seronegative subjects, the median percentage of activated NK cells was 0.90% [range 0.00-3.92] with autologous serum and 2.07% [0.00-5.76] in the presence of the HCMV-negative serum pool, while it was 8.97% [0.00-26.49] with the pool of HCMV-positive sera. NK-cell activation using hyperimmune globulin is comparable to what is obtained using autologous serum. Sera from subjects at 3 and 6 months post primary infection showed a lower capacity of NK-cell activation than sera from subjects with past infection (p < 0.001). NK activation against HCMV-infected epithelial cells is dependent on the presence of HCMV-specific antibodies. This serum activity increases with time after the onset of HCMV infection. The protective role of NK-cell activation by HCMV-specific serum antibodies should be verified in clinical settings.
Collapse
Affiliation(s)
- Piera d’Angelo
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Federica Zavaglio
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Elisa Gabanti
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- M.A.D. Analisi, 27058 Voghera, Italy
| | - Paola Zelini
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chiara Fornara
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Laboratory Medicine Service, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Stefano Bernuzzi
- Service of Immunohematology and Transfusion Medicine, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Arsenio Spinillo
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation, University of Pavia, 27100 Pavia, Italy
| | - Daniele Lilleri
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Fausto Baldanti
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
12
|
Creisher PS, Klein SL. Pathogenesis of viral infections during pregnancy. Clin Microbiol Rev 2024; 37:e0007323. [PMID: 38421182 PMCID: PMC11237665 DOI: 10.1128/cmr.00073-23] [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] [Indexed: 03/02/2024] Open
Abstract
SUMMARYViral infections during pregnancy are associated with significant adverse perinatal and fetal outcomes. Pregnancy is a unique immunologic and physiologic state, which can influence control of virus replication, severity of disease, and vertical transmission. The placenta is the organ of the maternal-fetal interface and provides defense against microbial infection while supporting the semi-allogeneic fetus via tolerogenic immune responses. Some viruses, such as cytomegalovirus, Zika virus, and rubella virus, can breach these defenses, directly infecting the fetus and having long-lasting consequences. Even without direct placental infection, other viruses, including respiratory viruses like influenza viruses and severe acute respiratory syndrome coronavirus 2, still cause placental damage and inflammation. Concentrations of progesterone and estrogens rise during pregnancy and contribute to immunological adaptations, placentation, and placental development and play a pivotal role in creating a tolerogenic environment at the maternal-fetal interface. Animal models, including mice, nonhuman primates, rabbits, and guinea pigs, are instrumental for mechanistic insights into the pathogenesis of viral infections during pregnancy and identification of targetable treatments to improve health outcomes of pregnant individuals and offspring.
Collapse
Affiliation(s)
- Patrick S Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Payne H, Barnabas S. "Congenital cytomegalovirus in Sub-Saharan Africa-a narrative review with practice recommendations". Front Public Health 2024; 12:1359663. [PMID: 38813410 PMCID: PMC11134569 DOI: 10.3389/fpubh.2024.1359663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/24/2024] [Indexed: 05/31/2024] Open
Abstract
Cytomegalovirus (CMV) is the most common cause of congenital infection internationally, occurring in 0.67% of births, and increasingly recognised as a major public health burden due to the potential for long-term neurodevelopmental and hearing impairment. This burden includes estimates of 10% of childhood cerebral palsy and up to 25% of childhood deafness. In Sub-Saharan Africa, where CMV-seroprevalence is almost ubiquitous, prevalence of congenital CMV (cCMV) is higher than the global average, and yet there is a dearth of research and initiatives to improve recognition, diagnosis and treatment. This narrative review outlines the epidemiology and clinical presentation of cCMV, discusses issues of case identification and treatment in Sub-Saharan Africa, and recommends a framework of strategies to address these challenges. Considering the significant burden of cCMV disease in this setting, it is undoubtably time we embark upon improving diagnosis and care for these infants.
Collapse
Affiliation(s)
- Helen Payne
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, University of Stellenbosch, Cape Town, South Africa
- Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - Shaun Barnabas
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, University of Stellenbosch, Cape Town, South Africa
| |
Collapse
|
14
|
Wu K, Hou YJ, Makrinos D, Liu R, Zhu A, Koch M, Yu WH, Paila YD, Chandramouli S, Panther L, Henry C, DiPiazza A, Carfi A. Characterization of humoral and cellular immunologic responses to an mRNA-based human cytomegalovirus vaccine from a phase 1 trial of healthy adults. J Virol 2024; 98:e0160323. [PMID: 38526054 PMCID: PMC11019844 DOI: 10.1128/jvi.01603-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/25/2024] [Indexed: 03/26/2024] Open
Abstract
mRNA-1647 is an investigational mRNA-based vaccine against cytomegalovirus (CMV) that contains sequences encoding the CMV proteins glycoprotein B and pentamer. Humoral and cellular immune responses were evaluated in blood samples collected from healthy CMV-seropositive and CMV-seronegative adults who participated in a phase 1 trial of a three-dose series of mRNA-1647 (NCT03382405). Neutralizing antibody (nAb) titers against fibroblast and epithelial cell infection in sera from CMV-seronegative mRNA-1647 recipients were higher than those in sera from control CMV-seropositive samples and remained elevated up to 12 months after dose 3. nAb responses elicited by mRNA-1647 were comparable across 14 human CMV (HCMV) strains. Frequencies of antigen-specific memory B cells increased in CMV-seropositive and CMV-seronegative participants after each mRNA-1647 dose and remained elevated for up to 6 months after dose 3. mRNA-1647 elicited robust increases in frequencies and polyfunctionality of CD4+ T helper type 1 and effector CD8+ T cells in samples from CMV-seronegative and CMV-seropositive participants after stimulation with HCMV-specific peptides. The administration of three doses of mRNA-1647 to healthy adults elicited high nAb titers with wide-breadth, long-lasting memory B cells, and strong polyfunctional T-cell responses. These findings support further clinical development of the mRNA-1647 vaccine against CMV.IMPORTANCECytomegalovirus (CMV), a common virus that can infect people of all ages, may lead to serious health problems in unborn babies and those with a weakened immune system. Currently, there is no approved vaccine available to prevent CMV infection; however, the investigational messenger RNA (mRNA)-based CMV vaccine, mRNA-1647, is undergoing evaluation in clinical trials. The current analysis examined samples from a phase 1 trial of mRNA-1647 in healthy adults to better understand how the immune system reacts to vaccination. Three doses of mRNA-1647 produced a long-lasting immune response, thus supporting further investigation of the vaccine in the prevention of CMV infection.CLINICAL TRIALSRegistered at ClinicalTrials.gov (NCT03382405).
Collapse
Affiliation(s)
- Kai Wu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Yixuan Jacob Hou
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Dan Makrinos
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Runxia Liu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Alex Zhu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Matthew Koch
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Wen-Han Yu
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Yamuna D. Paila
- Infectious Disease Development, Moderna, Inc., Cambridge, Massachusetts, USA
| | | | - Lori Panther
- Infectious Disease Development, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Carole Henry
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Anthony DiPiazza
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Andrea Carfi
- Infectious Disease Research, Moderna, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
15
|
Ye X, Shih DJH, Ku Z, Hong J, Barrett DF, Rupp RE, Zhang N, Fu TM, Zheng WJ, An Z. Transcriptional signature of durable effector T cells elicited by a replication defective HCMV vaccine. NPJ Vaccines 2024; 9:70. [PMID: 38561339 PMCID: PMC10984989 DOI: 10.1038/s41541-024-00860-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Human cytomegalovirus (HCMV) is a leading infectious cause of birth defects and the most common opportunistic infection that causes life-threatening diseases post-transplantation; however, an effective vaccine remains elusive. V160 is a live-attenuated replication defective HCMV vaccine that showed a 42.4% efficacy against primary HCMV infection among seronegative women in a phase 2b clinical trial. Here, we integrated the multicolor flow cytometry, longitudinal T cell receptor (TCR) sequencing, and single-cell RNA/TCR sequencing approaches to characterize the magnitude, phenotype, and functional quality of human T cell responses to V160. We demonstrated that V160 de novo induces IE-1 and pp65 specific durable polyfunctional effector CD8 T cells that are comparable to those induced by natural HCMV infection. We identified a variety of V160-responsive T cell clones which exhibit distinctive "transient" and "durable" expansion kinetics, and revealed a transcriptional signature that marks durable CD8 T cells post-vaccination. Our study enhances the understanding of human T-cell immune responses to V160 vaccination.
Collapse
Affiliation(s)
- Xiaohua Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Infectious Disease Research, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - David J H Shih
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Junping Hong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Diane F Barrett
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Richard E Rupp
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tong-Ming Fu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
16
|
Kotton CN, Torre-Cisneros J, Yakoub-Agha I. Slaying the "Troll of Transplantation"-new frontiers in cytomegalovirus management: A report from the CMV International Symposium 2023. Transpl Infect Dis 2024; 26:e14183. [PMID: 37942955 DOI: 10.1111/tid.14183] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 11/10/2023]
Abstract
The 2023 International CMV Symposium took place in Barcelona in May 2023. During the 2-day meeting, delegates and faculty discussed the ongoing challenge of managing the risk of cytomegalovirus infection (the Troll of Transplantation) after solid organ or hematopoietic cell transplantation. Opportunities to improve outcomes of transplant recipients by applying advances in antiviral prophylaxis or pre-emptive therapy, immunotherapy, and monitoring of cell-mediated immunity to routine clinical practice were debated and relevant educational clinical cases presented. This review summarizes the presentations, cases, and discussions from the meeting and describes how further advances are needed before the Troll of Transplantation is slain.
Collapse
Affiliation(s)
- Camille N Kotton
- Transplant and Immunocompromised Host Infectious Diseases, Infectious Diseases Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julian Torre-Cisneros
- Maimónides Institute for Biomedical Research of Cordoba (IMIBIC)/Reina Sofía University Hospital/University of Cordoba (UCO), Cordoba, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
17
|
Rimmer L, Mann DA, Sayer AA, Amarnath S, Granic A. A silver bullet for ageing medicine?: clinical relevance of T-cell checkpoint receptors in normal human ageing. Front Immunol 2024; 15:1360141. [PMID: 38361938 PMCID: PMC10867193 DOI: 10.3389/fimmu.2024.1360141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Immunosenescence describes dysregulation of the immune system with ageing manifested in both the innate and adaptive immunity, including changes in T-cell checkpoint signaling. Through complex and nuanced process, T-cells lose excitatory signaling pathways and upregulate their inhibitory signaling, leading to ineffective immune responses that contribute to the formation of the ageing phenotype. Here we expand on the expression, function, and clinical potential of targeting the T-cell checkpoint signaling in age and highlight interventions offering the most benefits to older adults' health. Notably, modifications in vaccination such as with mTOR inhibitors show immediate clinical relevance and good tolerability. Other proposed treatments, including therapies with monoclonal antibodies fail to show clinical efficacy or tolerability needed for implementation at present. Although T-cell co-signaling fits a valuable niche for translational scientists to manage immunosenescence, future study would benefit from the inclusion of older adults with multiple long-term conditions and polypharmacy, ensuring better applicability to actual patients seen in clinical settings.
Collapse
Affiliation(s)
- Lucy Rimmer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Derek A. Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Avan A. Sayer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Institute for Health and Care Research (NIHR) Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust and Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Shoba Amarnath
- Newcastle University Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Antoneta Granic
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
18
|
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
|
19
|
Šudomová M, Hassan STS. Herpesvirus Diseases in Humans and Animals: Recent Developments, Challenges, and Charting Future Paths. Pathogens 2023; 12:1422. [PMID: 38133305 PMCID: PMC10745940 DOI: 10.3390/pathogens12121422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 11/25/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Herpesviruses, a family of enveloped DNA viruses, pose significant threats to both humans and animals [...].
Collapse
Affiliation(s)
- Miroslava Šudomová
- Museum of Literature in Moravia, Klášter 1, 664 61 Rajhrad, Czech Republic
| | - Sherif T. S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Czech Republic;
| |
Collapse
|
20
|
Doss KM, Heldman MR, Limaye AP. Updates in Cytomegalovirus Prevention and Treatment in Solid Organ Transplantation. Infect Dis Clin North Am 2023:S0891-5520(23)00083-1. [PMID: 37989636 PMCID: PMC11102935 DOI: 10.1016/j.idc.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
The authors summarize recent updates in the prevention and management of cytomegalovirus (CMV) in solid organ transplant (SOT) recipients with a focus on CMV seronegative recipients of organs from seropositive donors (CMV D+/R-) who are at highest risk of CMV infection and disease. They discuss advantages of preemptive therapy for CMV disease prevention in CMV D+/R- liver transplant recipients, letermovir for CMV prophylaxis, and updates in the development of monoclonal antibodies and vaccines as immune-based preventative strategies. They review the roles of maribavir and virus-specific T cells for management of resistant or refractory CMV infection in SOT recipients.
Collapse
Affiliation(s)
- Kathleen M Doss
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Madeleine R Heldman
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, NC, USA
| | - Ajit P Limaye
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
21
|
Swanepoel J, van Zyl G, Hesseling AC, Johnson SM, Moore DAJ, Seddon JA. Human Cytomegalovirus Immunoglobulin G Response and Pulmonary Tuberculosis in Adolescents: A Case-Control Study. Open Forum Infect Dis 2023; 10:ofad487. [PMID: 37937044 PMCID: PMC10627337 DOI: 10.1093/ofid/ofad487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023] Open
Abstract
Background Emerging evidence suggests a link between infection with herpes viruses, particularly human cytomegalovirus (HCMV) and Epstein-Barr virus (EBV), and progression to tuberculosis disease. Methods An unmatched case-control study was conducted among adolescents aged 10-19 years enrolled in an observational study (Teen TB) between November 2020 and November 2021, in Cape Town, South Africa. Fifty individuals with pulmonary tuberculosis and 51 healthy tuberculosis-exposed individuals without tuberculosis were included. Demographics and clinical data were obtained, and serum samples collected at enrolment were tested for HCMV immunoglobulin G (IgG) and EBV nuclear antigen (EBNA) IgG using 2 automated enzyme immunoassays. Odds ratios were estimated using unconditional logistic regression. Results The median age of 101 participants was 15 years (interquartile range, 13-17 years); 55 (54%) were female. All participants were HCMV IgG seropositive, and 95% were EBNA IgG seropositive. Individuals with tuberculosis had higher HCMV IgG titers than healthy controls (P = .04). Individuals with upper-tertile HCMV IgG titers had 3.67 times greater odds of pulmonary tuberculosis than those with IgG titers in the lower tertile (95% confidence interval, 1.05-12.84; P = .04). There was a trend for increasing odds of pulmonary tuberculosis with increasing titers of HCMV IgG (P = .04). In contrast, there was no association between tuberculosis and higher EBNA IgG values. Conclusions There is a high prevalence of sensitization to HCMV and EBV among adolescents in this high-tuberculosis-burden setting. Higher HCMV IgG titers were associated with pulmonary tuberculosis in adolescents.
Collapse
Affiliation(s)
- Jeremi Swanepoel
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gert van Zyl
- Division of Medical Virology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University and National Health Laboratory Service, Tygerberg Academic Hospital, Cape Town, South Africa
| | - Anneke C Hesseling
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Sarah M Johnson
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | - David A J Moore
- TB Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - James A Seddon
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| |
Collapse
|
22
|
Barry PA, Iyer SS, Gibson L. Re-Evaluating Human Cytomegalovirus Vaccine Design: Prediction of T Cell Epitopes. Vaccines (Basel) 2023; 11:1629. [PMID: 38005961 PMCID: PMC10674879 DOI: 10.3390/vaccines11111629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/26/2023] Open
Abstract
HCMV vaccine development has traditionally focused on viral antigens identified as key targets of neutralizing antibody (NAb) and/or T cell responses in healthy adults with chronic HCMV infection, such as glycoprotein B (gB), the glycoprotein H-anchored pentamer complex (PC), and the unique long 83 (UL83)-encoded phosphoprotein 65 (pp65). However, the protracted absence of a licensed HCMV vaccine that reduces the risk of infection in pregnancy regardless of serostatus warrants a systematic reassessment of assumptions informing vaccine design. To illustrate this imperative, we considered the hypothesis that HCMV proteins infrequently detected as targets of T cell responses may contain important vaccine antigens. Using an extant dataset from a T cell profiling study, we tested whether HCMV proteins recognized by only a small minority of participants encompass any T cell epitopes. Our analyses demonstrate a prominent skewing of T cell responses away from most viral proteins-although they contain robust predicted CD8 T cell epitopes-in favor of a more restricted set of proteins. Our findings raise the possibility that HCMV may benefit from evading the T cell recognition of certain key proteins and that, contrary to current vaccine design approaches, including them as vaccine antigens could effectively take advantage of this vulnerability.
Collapse
Affiliation(s)
- Peter A. Barry
- Department of Pathology and Laboratory Medicine, Center for Immunology and Infectious Diseases, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
- California National Primate Research Center, University of California, Davis, CA 95616, USA
| | - Smita S. Iyer
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Laura Gibson
- Departments of Medicine and of Pediatrics, Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Congenital infections are a major cause of childhood multidomain neurodevelopmental disabilities. They contribute to a range of structural brain abnormalities that can cause severe neurodevelopmental impairment, cerebral palsy, epilepsy, and neurosensory impairments. New congenital infections and global viral pandemics have emerged, with some affecting the developing brain and causing neurodevelopmental concerns. This review aims to provide current understanding of fetal infections and their impact on neurodevelopment. RECENT FINDINGS There are a growing list of congenital infections causing neurodevelopmental issues, including cytomegalovirus, Zika virus, syphilis, rubella, lymphocytic choriomeningitis virus, and toxoplasmosis. Fetal exposure to maternal SARS-CoV-2 may also pose risk to the developing brain and impact neurodevelopmental outcomes, although studies have conflicting results. As Zika virus was a recently identified congenital infection, there are several new reports on child neurodevelopment in the Caribbean and Central and South America. For many congenital infections, children with in-utero exposure, even if asymptomatic at birth, may have neurodevelopmental concerns manifest over time. SUMMARY Congenital infections should be considered in the differential diagnosis of a child with neurodevelopmental impairments. Detailed pregnancy history, exposure risk, and testing should guide diagnosis and multidisciplinary evaluation. Children with congenital infections should have long-term follow-up to assess for neurodevelopmental delays and other neurosensory impairments. Children with confirmed delays or high-risk should be referred for rehabilitation therapies.
Collapse
Affiliation(s)
- Olivier Fortin
- Prenatal Pediatrics Institute, Children’s National Hospital, Washington, DC
| | - Sarah B. Mulkey
- Prenatal Pediatrics Institute, Children’s National Hospital, Washington, DC
- Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
24
|
Zhang W, Jiang Y, He Y, Boucetta H, Wu J, Chen Z, He W. Lipid carriers for mRNA delivery. Acta Pharm Sin B 2023; 13:4105-4126. [PMID: 37799378 PMCID: PMC10547918 DOI: 10.1016/j.apsb.2022.11.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 12/05/2022] Open
Abstract
Messenger RNA (mRNA) is the template for protein biosynthesis and is emerging as an essential active molecule to combat various diseases, including viral infection and cancer. Especially, mRNA-based vaccines, as a new type of vaccine, have played a leading role in fighting against the current global pandemic of COVID-19. However, the inherent drawbacks, including large size, negative charge, and instability, hinder its use as a therapeutic agent. Lipid carriers are distinguishable and promising vehicles for mRNA delivery, owning the capacity to encapsulate and deliver negatively charged drugs to the targeted tissues and release cargoes at the desired time. Here, we first summarized the structure and properties of different lipid carriers, such as liposomes, liposome-like nanoparticles, solid lipid nanoparticles, lipid-polymer hybrid nanoparticles, nanoemulsions, exosomes and lipoprotein particles, and their applications in delivering mRNA. Then, the development of lipid-based formulations as vaccine delivery systems was discussed and highlighted. Recent advancements in the mRNA vaccine of COVID-19 were emphasized. Finally, we described our future vision and perspectives in this field.
Collapse
Affiliation(s)
- Wanting Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yuxin Jiang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yonglong He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hamza Boucetta
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jun Wu
- Department of Geriatric Cardiology, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
25
|
Bottino P, Pastrone L, Curtoni A, Bondi A, Sidoti F, Zanotto E, Cavallo R, Solidoro P, Costa C. Antiviral Approach to Cytomegalovirus Infection: An Overview of Conventional and Novel Strategies. Microorganisms 2023; 11:2372. [PMID: 37894030 PMCID: PMC10608897 DOI: 10.3390/microorganisms11102372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a herpesvirus capable of establishing a lifelong persistence in the host through a chronic state of infection and remains an essential global concern due to its distinct life cycle, mutations, and latency. It represents a life-threatening pathogen for immunocompromised patients, such as solid organ transplanted patients, HIV-positive individuals, and hematopoietic stem cell recipients. Multiple antiviral approaches are currently available and administered in order to prevent or manage viral infections in the early stages. However, limitations due to side effects and the onset of antidrug resistance are a hurdle to their efficacy, especially for long-term therapies. Novel antiviral molecules, together with innovative approaches (e.g., genetic editing and RNA interference) are currently in study, with promising results performed in vitro and in vivo. Since HCMV is a virus able to establish latent infection, with a consequential risk of reactivation, infection management could benefit from preventive treatment for critical patients, such as immunocompromised individuals and seronegative pregnant women. This review will provide an overview of conventional antiviral clinical approaches and their mechanisms of action. Additionally, an overview of proposed and developing new molecules is provided, including nucleic-acid-based therapies and immune-mediated approaches.
Collapse
Affiliation(s)
- Paolo Bottino
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Lisa Pastrone
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Antonio Curtoni
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Alessandro Bondi
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Francesca Sidoti
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Elisa Zanotto
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Rossana Cavallo
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Paolo Solidoro
- Pneumology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy;
| | - Cristina Costa
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| |
Collapse
|
26
|
Yu MZ, Wang NN, Zhu JQ, Lin YX. The clinical progress and challenges of mRNA vaccines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1894. [PMID: 37096256 DOI: 10.1002/wnan.1894] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
Owing to the breakthroughs in the prevention and control of the COVID-19 pandemic, messenger RNA (mRNA)-based vaccines have emerged as promising alternatives to conventional vaccine approaches for infectious disease prevention and anticancer treatments. Advantages of mRNA vaccines include flexibility in designing and manipulating antigens of interest, scalability in rapid response to new variants, ability to induce both humoral and cell-mediated immune responses, and ease of industrialization. This review article presents the latest advances and innovations in mRNA-based vaccines and their clinical translations in the prevention and treatment of infectious diseases or cancers. We also highlight various nanoparticle delivery platforms that contribute to their success in clinical translation. Current challenges related to mRNA immunogenicity, stability, and in vivo delivery and the strategies for addressing them are also discussed. Finally, we provide our perspectives on future considerations and opportunities for applying mRNA vaccines to fight against major infectious diseases and cancers. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Meng-Zhen Yu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, People's Republic of China
- University of Chinese Academy of Sciences (UCAS), Beijing, People's Republic of China
| | - Nan-Nan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, People's Republic of China
- University of Chinese Academy of Sciences (UCAS), Beijing, People's Republic of China
| | - Jia-Qing Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, People's Republic of China
| | - Yao-Xin Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, People's Republic of China
- University of Chinese Academy of Sciences (UCAS), Beijing, People's Republic of China
| |
Collapse
|
27
|
McMahon‑Cole H, Johnson A, Sadat Aghamiri S, Helikar T, Crawford LB. Modeling and Remodeling the Cell: How Digital Twins and HCMV Can Elucidate the Complex Interactions of Viral Latency, Epigenetic Regulation, and Immune Responses. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023; 10:141-151. [PMID: 37901689 PMCID: PMC10601359 DOI: 10.1007/s40588-023-00201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 10/31/2023]
Abstract
Purpose of Review Human cytomegalovirus (HCMV), while asymptomatic in most, causes significant complications during fetal development, following transplant or in immunosuppressed individuals. The host-virus interactions regulating viral latency and reactivation and viral control of the cellular environment (immune regulation, differentiation, epigenetics) are highly complex. Understanding these processes is essential to controlling infection and can be leveraged as a novel approach for understanding basic cell biology. Recent Findings Immune digital twins (IDTs) are digital simulations integrating knowledge of human immunology, physiology, and patient-specific clinical data to predict individualized immune responses and targeted treatments. Recent studies used IDTs to elucidate mechanisms of T cells, dendritic cells, and epigenetic control-all key to HCMV biology. Summary Here, we discuss how leveraging the unique biology of HCMV and IDTs will clarify immune response dynamics, host-virus interactions, and viral latency and reactivation and serve as a powerful IDT-validation platform for individualized and holistic health management.
Collapse
Affiliation(s)
- Hana McMahon‑Cole
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Alicia Johnson
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Sara Sadat Aghamiri
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Lindsey B. Crawford
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Center for Virology, Lincoln, NE, USA
- Nebraska Center for Integrated Biomolecular Communication, Lincoln, NE, USA
| |
Collapse
|
28
|
Crooks CM, Chan C, Permar SR. Leveraging preclinical study designs to close gaps in vaccine development for perinatal pathogens. J Exp Med 2023; 220:e20230184. [PMID: 37289272 PMCID: PMC10250551 DOI: 10.1084/jem.20230184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Vaccines to perinatal pathogens are critical for both reducing the burden of endemic pathogens and preparing for the next pandemic. Although they are often at greater risk of severe disease from infection, pregnant people and children are routinely marginalized in the vaccine development process. We highlight several challenges in the vaccine development process and how three tools-translational animal models, human cohort studies of natural infection, and innovative data-use strategies-can speed vaccine development and ensure equity for pregnant people and children in the next pandemic.
Collapse
Affiliation(s)
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
29
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
30
|
Wang HY, Taher H, Kreklywich CN, Schmidt KA, Scheef EA, Barfield R, Otero CE, Valencia SM, Crooks CM, Mirza A, Woods K, Burgt NV, Kowalik TF, Barry PA, Hansen SG, Tarantal AF, Chan C, Streblow DN, Picker LJ, Kaur A, Früh K, Permar SR, Malouli D. The pentameric complex is not required for vertical transmission of cytomegalovirus in seronegative pregnant rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545169. [PMID: 37398229 PMCID: PMC10312687 DOI: 10.1101/2023.06.15.545169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Congenital cytomegalovirus (cCMV) infection is the leading infectious cause of neonatal neurological impairment but essential virological determinants of transplacental CMV transmission remain unclear. The pentameric complex (PC), composed of five subunits, glycoproteins H (gH), gL, UL128, UL130, and UL131A, is essential for efficient entry into non-fibroblast cells in vitro . Based on this role in cell tropism, the PC is considered a possible target for CMV vaccines and immunotherapies to prevent cCMV. To determine the role of the PC in transplacental CMV transmission in a non-human primate model of cCMV, we constructed a PC-deficient rhesus CMV (RhCMV) by deleting the homologues of the HCMV PC subunits UL128 and UL130 and compared congenital transmission to PC-intact RhCMV in CD4+ T cell-depleted or immunocompetent RhCMV-seronegative, pregnant rhesus macaques (RM). Surprisingly, we found that the transplacental transmission rate was similar for PC-intact and PC-deleted RhCMV based on viral genomic DNA detection in amniotic fluid. Moreover, PC-deleted and PC-intact RhCMV acute infection led to similar peak maternal plasma viremia. However, there was less viral shedding in maternal urine and saliva and less viral dissemination in fetal tissues in the PC-deleted group. As expected, dams inoculated with PC-deleted RhCMV demonstrated lower plasma IgG binding to PC-intact RhCMV virions and soluble PC, as well as reduced neutralization of PC-dependent entry of the PC-intact RhCMV isolate UCD52 into epithelial cells. In contrast, binding to gH expressed on the cell surface and neutralization of entry into fibroblasts by the PC-intact RhCMV was higher for dams infected with PC-deleted RhCMV compared to those infected with PC-intact RhCMV. Our data demonstrates that the PC is dispensable for transplacental CMV infection in our non-human primate model. One Sentence Summary Congenital CMV transmission frequency in seronegative rhesus macaques is not affected by the deletion of the viral pentameric complex.
Collapse
|
31
|
Nigro G, Muselli M. Prevention of Congenital Cytomegalovirus Infection: Review and Case Series of Valaciclovir versus Hyperimmune Globulin Therapy. Viruses 2023; 15:1376. [PMID: 37376675 PMCID: PMC10302477 DOI: 10.3390/v15061376] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Cytomegalovirus (CMV) is the most common cause of congenital infections in developed countries because is capable of infecting the fetus after both primary and recurrent maternal infection, and because the virus may be spread for years through infected children. Moreover, CMV is the most serious congenital infection associated with severe neurological and sensorineural sequelae, which can occur at birth or develop later on. Hygienic measures can prevent CMV transmission, which mainly involve contact with children under 3 years of age and attending a nursery or daycare. In animal and human pregnancies, many observational and controlled studies have shown that CMV-specific hyperimmune globulin (HIG) is safe and can significantly decrease maternal-fetal transmission of CMV infection and, mostly, the occurrence of CMV disease. Recently, valaciclovir at the dosage of 8 g/day was also reported to be capable of decreasing the rates of congenital infection and disease. However, comparing the results of our two recent case series, the infants born to women treated with HIG showed significantly lower rates of CMV DNA positivity in urine (9.7% vs. 75.0%; p < 0.0001) and abnormalities after follow-up (0.0% vs. 41.7%; p < 0.0001). The implementation of CMV screening would enable primary prevention via hygiene counseling, improve the understanding and awareness of congenital CMV infection, and increase the knowledge of the potential efficacy of preventive or therapeutic HIG or antiviral administration.
Collapse
Affiliation(s)
- Giovanni Nigro
- Non-Profit Association Mother-Infant Cytomegalovirus Infection (AMICI), 00198 Rome, Italy
| | - Mario Muselli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | | |
Collapse
|
32
|
Canfield D, Gabby L, Vaziri Fard E, Gyamfi-Bannerman C. Cytomegalovirus in Pregnancy. Obstet Gynecol Clin North Am 2023; 50:263-277. [PMID: 37149309 DOI: 10.1016/j.ogc.2023.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cytomegalovirus is a pervasive DNA herpesvirus that, while clinically insignificant to an immunocompetent adult host, can cause significant morbidity to a congenitally infected fetus. Although detection is often possible with several common ultrasonographic markers and good diagnostic accuracy using polymerase chain reaction testing of amniotic fluid, there are no proven prenatal prevention or antenatal treatment options. Therefore, universal screening is not currently recommended in pregnancy. Strategies that have been studied in the past include immunoglobulins, antivirals, and the development of a vaccine. In this review, we will further discuss the themes above, along with future direction for prevention and treatment.
Collapse
Affiliation(s)
- Dana Canfield
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, CA, USA
| | - Lauryn Gabby
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, CA, USA
| | - Elmira Vaziri Fard
- Department of Pathology and Lab Medicine, University of California, San Diego, CA, USA
| | - Cynthia Gyamfi-Bannerman
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, CA, USA.
| |
Collapse
|
33
|
Spatafore D, Warakomski D, Hofmann C, Christanti S, Wagner JM. Investigation into the use of gamma irradiated Cytodex-1 microcarriers to produce a human cytomegalovirus (HCMV) vaccine candidate in epithelial cells. J Biotechnol 2023; 365:62-71. [PMID: 36804577 DOI: 10.1016/j.jbiotec.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
V160 is a viral vaccine candidate against human cytomegalovirus (HCMV) that is manufactured using Adult Retinal Pigment Epithelial cells (ARPE-19) grown on Cytodex-1 microcarriers. The microcarriers are generally hydrated, washed, and autoclaved prior to use, which can be limiting at large production scales. To minimize microcarrier preparation and sterilization, the use of gamma irradiated Cytodex-1 was investigated. Similar ARPE-19 cell growth was observed on heat-sterilized and gamma irradiated Cytodex-1; however, significantly reduced virus production was observed in cultures exposed to gamma irradiated Cytodex-1. Additional experiments suggest that infection inhibition is not exclusive to ARPE-19 but is most directly linked to HCMV V160, as evidenced by similar inhibition of V160 with Vero cells and no inhibition of Measles virus with either cell type. These observations suggest a putative impact on HCMV infection from the presence of extractable(s)/leachable(s) in the gamma irradiated microcarriers. Thorough aseptic rinsing of gamma irradiated Cytodex-1 prior to use can mitigate this impact and enable comparable process performance to heat-sterilized Cytodex-1. Though not fully a "ready-to-use" product for the HCMV V160 production process, utilization of Cytodex-1 microcarriers was possible without requiring heat sterilization, suggesting a potential path forward for large scale production of V160.
Collapse
Affiliation(s)
- Daniel Spatafore
- Process Research & Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Donald Warakomski
- Vaccine Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Carl Hofmann
- Vaccine Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Sianny Christanti
- West Point Technical Operations Labs, Merck & Co., Inc., Rahway, NJ, USA
| | - James M Wagner
- Process Research & Development, Merck & Co., Inc., Rahway, NJ, USA.
| |
Collapse
|
34
|
Abeywickrema M, Kelly D, Kadambari S. Management of neonatal central nervous system viral infections: Knowledge gaps and research priorities. Rev Med Virol 2023; 33:e2421. [PMID: 36639694 DOI: 10.1002/rmv.2421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023]
Abstract
Congenital CMV, enteroviruses, human parechovirus and herpes simplex virus are all common causes of severe central nervous system (CNS) infection in neonates. The introduction of screening (i.e. newborn hearing screening programme), integration of molecular syndromic testing (i.e. multiplex polymerase chain reaction assays) and increase in sexually transmitted infections (i.e. anogenital herpes) have contributed to increases in each of these infections over the last decade. However, therapeutic options are highly limited in part due to the lack of epidemiological data informing trials. This review will describe our current understanding of the clinical burden and epidemiology of these severe neonatal CNS infections, outline the novel antiviral and vaccines in the pipeline and suggest future research studies which could help develop new therapeutics.
Collapse
Affiliation(s)
- Movin Abeywickrema
- Department of Infection, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Dominic Kelly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Seilesh Kadambari
- Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,University College London, Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
35
|
Sehrawat S, Osterrieder N, Schmid DS, Rouse BT. Can the triumph of mRNA vaccines against COVID-19 be extended to other viral infections of humans and domesticated animals? Microbes Infect 2023; 25:105078. [PMID: 36435367 PMCID: PMC9682868 DOI: 10.1016/j.micinf.2022.105078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
The unprecedented success of mRNA vaccines in managing the COVID-19 pandemic raises the prospect of applying the mRNA platform to other viral diseases of humans and domesticated animals, which may lead to more efficacious vaccines for some agents. We briefly discuss reasons why mRNA vaccines achieved such success against COVID-19 and indicate what other virus infections and disease conditions might also be ripe for control using mRNA vaccines. We also evaluate situations where mRNA could prove valuable to rebalance the status of immune responsiveness and achieve success as a therapeutic vaccine approach against infections that induce immunoinflammatory lesions.
Collapse
Affiliation(s)
- Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City, PO Manauli, Mohali 140306, Punjab, India.
| | - Nikolaus Osterrieder
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163 Berlin, Germany; Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 5F, Block 1B, To Yuen Building, 31 To Yuen Street, Kowloon Tong, Hong Kong.
| | - D Scott Schmid
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA.
| | - Barry T Rouse
- College of Veterinary Medicine, University of Tennessee Knoxville, TN 37996-0845, USA.
| |
Collapse
|
36
|
Balani SS, Sadiq S, Jensen CJ, Kizilbash SJ. Prevention and management of CMV infection in pediatric solid organ transplant recipients. Front Pediatr 2023; 11:1098434. [PMID: 36891229 PMCID: PMC9986459 DOI: 10.3389/fped.2023.1098434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023] Open
Abstract
Human cytomegalovirus (CMV) remains one of the most common opportunistic infections following solid organ transplantation in children. CMV causes morbidity and mortality through direct tissue-invasive disease and indirect immunomodulatory effects. In recent years, several new agents have emerged for the prevention and treatment of CMV disease in solid organ transplant recipients. However, pediatric data remain scarce, and many of the treatments are extrapolated from the adult literature. Controversies exist about the type and duration of prophylactic therapies and the optimal dosing of antiviral agents. This review provides an up-to-date overview of treatment modalities used to prevent and treat CMV disease in solid organ transplant (SOT) recipients.
Collapse
Affiliation(s)
- Shanthi S Balani
- Division of Nephrology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Sanober Sadiq
- Division of Nephrology, Department of Pediatrics, University of California, San Francisco, CA, United States
| | - Chelsey J Jensen
- Department of Solid Organ Transplant, University of Minnesota, Minneapolis, MN, United States
| | - Sarah J Kizilbash
- Division of Nephrology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
37
|
Wang S, Zhou X, He X, Ma S, Sun C, Zhang J, Xu X, Jin W, Yan J, Lin P, Mao G. Suppressive effects of pterostilbene on human cytomegalovirus (HCMV) infection and HCMV-induced cellular senescence. Virol J 2022; 19:224. [PMID: 36564838 PMCID: PMC9782289 DOI: 10.1186/s12985-022-01954-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Human cytomegalovirus (HCMV), a member of the β-herpesvirus family, causes the establishment of a latent infection that persists throughout the life of the host and can be reactivated when immunity is weakened. To date, there is no vaccine to prevent HCMV infection, and clinically approved drugs target the stage of viral replication and have obvious adverse reactions. Thus, development of novel therapeutics is urgently needed. METHODS In the current study, we identified a naturally occurring pterostilbene that inhibits HCMV Towne strain replication in human diploid fibroblast WI-38 cells through Western blotting, qPCR, indirect immunofluorescence assay, tissue culture infective dose assays. The time-of-addition experiment was carried out to identify the stage at which pterostilbene acted. Finally, the changes of cellular senescence biomarkers and reactive oxygen species production brought by pterostilbene supplementation were used to partly elucidate the mechanism of anti-HCMV activity. RESULTS Our findings revealed that pterostilbene prevented lytic cytopathic changes, inhibited the expression of viral proteins, suppressed the replication of HCMV DNA, and significantly reduced the viral titre in WI-38 cells. Furthermore, our data showed that pterostilbene predominantly acted after virus cell entry and membrane fusion. The half-maximal inhibitory concentration was determined to be 1.315 μM and the selectivity index of pterostilbene was calculated as 26.73. Moreover, cell senescence induced by HCMV infection was suppressed by pterostilbene supplementation, as shown by a decline in senescence-associated β-galactosidase activity, decreased production of reactive oxygen species and reduced expression of p16, p21 and p53, which are considered biomarkers of cellular senescence. CONCLUSION Together, our findings identify pterostilbene as a novel anti-HCMV agent that may prove useful in the treatment of HCMV replication.
Collapse
Affiliation(s)
- Sanying Wang
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China
| | - Xuqiang Zhou
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China ,grid.268505.c0000 0000 8744 8924College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053 People’s Republic of China
| | - Xinyue He
- grid.469325.f0000 0004 1761 325XCollege of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014 People’s Republic of China
| | - Shushu Ma
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China
| | - Chuan Sun
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China
| | - Jing Zhang
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China
| | - Xiaogang Xu
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China
| | - Weihua Jin
- grid.469325.f0000 0004 1761 325XCollege of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014 People’s Republic of China
| | - Jin Yan
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China
| | - Ping Lin
- Geriatric Department of the 3rd Hospital of Hangzhou, 310009 Hangzhou, People’s Republic of China
| | - Genxiang Mao
- grid.417400.60000 0004 1799 0055Zhejiang Provincial Key Lab of Geriatrics and Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310030 People’s Republic of China ,grid.268505.c0000 0000 8744 8924College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053 People’s Republic of China
| |
Collapse
|
38
|
Neutralization Epitopes in Trimer and Pentamer Complexes Recognized by Potent Cytomegalovirus-Neutralizing Human Monoclonal Antibodies. Microbiol Spectr 2022; 10:e0139322. [PMID: 36342276 PMCID: PMC9784774 DOI: 10.1128/spectrum.01393-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Human cytomegalovirus (HCMV) infects 36% to almost 100% of adults and causes severe complications only in immunocompromised individuals. HCMV viral surface trimeric (gH/gL/gO) and pentameric (gH/gL/UL128/UL130/UL131A) complexes play important roles in HCMV infection and tropism. Here, we isolated and identified a total of four neutralizing monoclonal antibodies (MAbs) derived from HCMV-seropositive blood donors. Based on their reactivity to HCMV trimer and pentamer, these MAbs can be divided into two groups. MAbs PC0012, PC0014, and PC0035 in group 1 bind both trimer and pentamer and neutralize CMV by interfering with the postattachment steps of CMV entering into cells. These three antibodies recognize antigenic epitopes clustered in a similar area, which are overlapped by the epitope recognized by the known neutralizing antibody MSL-109. MAb PC0034 in group 2 binds only to pentamer and neutralizes CMV by blocking the binding of pentamer to cells. Epitope mapping using pentamer mutants showed that amino acid T94 of the subunit UL128 and K27 of UL131A on the pentamer are key epitope-associated residues recognized by PC0034. This study provides new evidence and insight information on the importance of the development of the CMV pentamer as a CMV vaccine. In addition, these newly identified potent CMV MAbs can be attractive candidates for development as antibody therapeutics for the prevention and treatment of HCMV infection. IMPORTANCE The majority of the global population is infected with HCMV, but severe complications occur only in immunocompromised individuals. In addition, CMV infection is a major cause of birth defects in newborns. Currently, there are still no approved prophylactic vaccines or therapeutic monoclonal antibodies (MAbs) for clinical use against HCMV infection. This study identified and characterized a panel of four neutralizing MAbs targeting the HCMV pentamer complex with specific aims to identify a key protein(s) and antigenic epitopes in the HCMV pentamer complex. The study also explored the mechanism by which these newly identified antibodies neutralize HCMV in order to design better HCMV vaccines focusing on the pentamer and to provide attractive candidates for the development of effective cocktail therapeutics for the prevention and treatment of HCMV infection.
Collapse
|
39
|
Turner DL, Mathias RA. The human cytomegalovirus decathlon: Ten critical replication events provide opportunities for restriction. Front Cell Dev Biol 2022; 10:1053139. [PMID: 36506089 PMCID: PMC9732275 DOI: 10.3389/fcell.2022.1053139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous human pathogen that can cause severe disease in immunocompromised individuals, transplant recipients, and to the developing foetus during pregnancy. There is no protective vaccine currently available, and with only a limited number of antiviral drug options, resistant strains are constantly emerging. Successful completion of HCMV replication is an elegant feat from a molecular perspective, with both host and viral processes required at various stages. Remarkably, HCMV and other herpesviruses have protracted replication cycles, large genomes, complex virion structure and complicated nuclear and cytoplasmic replication events. In this review, we outline the 10 essential stages the virus must navigate to successfully complete replication. As each individual event along the replication continuum poses as a potential barrier for restriction, these essential checkpoints represent potential targets for antiviral development.
Collapse
Affiliation(s)
- Declan L. Turner
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Rommel A. Mathias
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
40
|
Carmona AS, Kakkar F, Gantt S. Perinatal Cytomegalovirus Infection. CURRENT TREATMENT OPTIONS IN PEDIATRICS 2022; 8:395-411. [PMID: 36465883 PMCID: PMC9684878 DOI: 10.1007/s40746-022-00261-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/29/2022] [Indexed: 06/17/2023]
Abstract
PURPOSE OF REVIEW There have been recent advances in the field of congenital CMV infection (cCMV) related to antiviral treatment of pregnant women and infants, the implementation of newborn CMV screening programs, and the frequency and diagnosis of complications among infected children. In addition, postnatal CMV infection (pCMV) is increasingly recognized as a potential cause of long-term sequelae in addition to acute complications among preterm infants, raising important questions related to treatment, and prevention. RECENT FINDINGS High-dose valacyclovir appears to be safe and effective for the prevention of cCMV among women with first-trimester primary CMV infection. New studies reveal high rates of vestibular dysfunction and neuropsychiatric manifestations among children with cCMV. Some studies report associations between pCMV and long-term consequences, including neurodevelopmental delay and bronchopulmonary dysplasia, among very low birth weight infants, in addition to high risk of sepsis and death acutely, which has motivated efforts to eliminate the virus from breast milk by different methods. SUMMARY More long-term complications of cCMV are increasingly recognized among children previously thought to be asymptomatic. Although a preventive CMV vaccine may be achievable, strategies to reduce the burden of cCMV disease include maternal education about risk-reduction behaviors, antiviral treatment of pregnant women with primary infection, and newborn screening to allow timely, appropriate care. Similarly, although it remains unclear if pCMV causes long-term problems, there is growing interest in identifying and preventing disease from CMV infections among preterm infants.
Collapse
Affiliation(s)
- Alejandra Sandoval Carmona
- Department of Pediatrics, 3175 Ch. de La Côte-Sainte-Catherine, Université de Montréal, Montréal, Canada
| | - Fatima Kakkar
- Department of Pediatrics, 3175 Ch. de La Côte-Sainte-Catherine, Université de Montréal, Montréal, Canada
- Centre de Recherche du CHU Sainte-Justine, 3175 Ch. de La Côte-Sainte-Catherine, Montréal, Canada
| | - Soren Gantt
- Department of Pediatrics, 3175 Ch. de La Côte-Sainte-Catherine, Université de Montréal, Montréal, Canada
- Centre de Recherche du CHU Sainte-Justine, 3175 Ch. de La Côte-Sainte-Catherine, Montréal, Canada
| |
Collapse
|
41
|
Ralph KMI, Bull K, Trotter C, Wood S, Jones CE. Paediatric health-related quality of life in congenital cytomegalovirus. Arch Dis Child 2022; 107:1004-1011. [PMID: 35768174 DOI: 10.1136/archdischild-2022-324007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/10/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Congenital cytomegalovirus (cCMV) is the most common congenital infection globally. This cross-sectional study aimed to describe the health-related quality of life (HRQoL) of children with cCMV and that of their parents. METHODS Families of children with cCMV in the UK were approached through the charity CMV Action. Parents completed questionnaires about their own HRQoL and that of their child. Children who were able to self-report completed questionnaires about their own HRQoL. Demographic characteristics of the sample were described using descriptive statistics. Questionnaire responses were scored, and domain and summary scores were calculated and compared with UK population norms, where available. RESULTS Seventy families participated, with children aged between 5 months and 18 years. Children with cCMV had poorer HRQoL compared with children from UK population data. HRQoL of children whose health was more severely affected by cCMV was poorer than that of children whose health was less severely affected. Parents of children whose health was moderately or severely affected had greater psychological morbidity and poorer HRQoL in physical, emotional, social, and cognitive functioning domains than parents of less severely affected children. CONCLUSIONS cCMV has a significant effect on the HRQoL of children with cCMV and their parents, with the children with the most significant health needs having the lowest HRQoL compared with those children with little or no effects on their health. This data could contribute to health economic analyses, informing resource allocation to potential interventions for the prevention and treatment of cCMV.
Collapse
Affiliation(s)
| | - Kim Bull
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Caroline Trotter
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | | - Christine E Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
42
|
Plotogea M, Isam AJ, Frincu F, Zgura A, Bacinschi X, Sandru F, Duta S, Petca RC, Edu A. An Overview of Cytomegalovirus Infection in Pregnancy. Diagnostics (Basel) 2022; 12:diagnostics12102429. [PMID: 36292118 PMCID: PMC9600407 DOI: 10.3390/diagnostics12102429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/22/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
The objective of this review was to bring to attention cytomegalovirus (CMV) infection during pregnancy, taking into consideration all relevant aspects, such as maternal diagnosis, fetal infection and prevention, prenatal diagnosis, and postnatal prognosis. A literature review was performed regarding adult and congenital infection. General information regarding this viral infection and potential related medical conditions was provided, considering the issues of maternal infection during pregnancy, transmission to the fetus, and associated congenital infection management. Prenatal diagnosis includes maternal serum testing and the confirmation of the infection in amniotic fluid or fetal blood. Additionally, prenatal diagnosis requires imaging techniques, ultrasound, and complementary magnetic resonance to assess cortical and extracortical anomalies. Imaging findings can predict both fetal involvement and the postnatal prognosis of the newborn, but they are difficult to assess, even for highly trained physicians. In regard to fetal sequelae, the early diagnosis of a potential fetal infection is crucial, and methods to decrease fetal involvement should be considered. Postnatal evaluation is also important, because many newborns may be asymptomatic and clinical anomalies can be diagnosed when sequelae are permanent.
Collapse
Affiliation(s)
- Mihaela Plotogea
- Department of Obstetrics and Gynecology, “Nicolae Malaxa” Clinical Hospital, 022441 Bucharest, Romania
| | - Al Jashi Isam
- Faculty of Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
- Correspondence: (A.J.I.); (F.F.)
| | - Francesca Frincu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Correspondence: (A.J.I.); (F.F.)
| | - Anca Zgura
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Xenia Bacinschi
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Florica Sandru
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Simona Duta
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Razvan Cosmin Petca
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Antoine Edu
- Department of Obstetrics and Gynecology, “Nicolae Malaxa” Clinical Hospital, 022441 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
43
|
Hu X, Wang HY, Otero CE, Jenks JA, Permar SR. Lessons from Acquired Natural Immunity and Clinical Trials to Inform Next-Generation Human Cytomegalovirus Vaccine Development. Annu Rev Virol 2022; 9:491-520. [PMID: 35704747 PMCID: PMC10154983 DOI: 10.1146/annurev-virology-100220-010653] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human cytomegalovirus (HCMV) infection, the most common cause of congenital disease globally, affecting an estimated 1 million newborns annually, can result in lifelong sequelae in infants, such as sensorineural hearing loss and brain damage. HCMV infection also leads to a significant disease burden in immunocompromised individuals. Hence, an effective HCMV vaccine is urgently needed to prevent infection and HCMV-associated diseases. Unfortunately, despite more than five decades of vaccine development, no successful HCMV vaccine is available. This review summarizes what we have learned from acquired natural immunity, including innate and adaptive immunity; the successes and failures of HCMV vaccine human clinical trials; the progress in related animal models; and the analysis of protective immune responses during natural infection and vaccination settings. Finally, we propose novel vaccine strategies that will harness the knowledge of protective immunity and employ new technology and vaccine concepts to inform next-generation HCMV vaccine development.
Collapse
Affiliation(s)
- Xintao Hu
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Claire E Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer A Jenks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA;
| |
Collapse
|
44
|
Spencer Clinton JL, Hoornweg TE, Tan J, Peng R, Schaftenaar W, Rutten VPMG, de Haan CAM, Ling PD. EEHV1A glycoprotein B subunit vaccine elicits humoral and cell-mediated immune responses in mice. Vaccine 2022; 40:5131-5140. [PMID: 35879117 DOI: 10.1016/j.vaccine.2022.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 10/16/2022]
Abstract
Asian elephants are an endangered species facing many threats, including severe hemorrhagic disease (HD) caused by the elephant endotheliotropic herpesvirus (EEHV). EEHV-HD is the leading cause of death in captive juvenile Asian elephants in North America and Europe, and also affects elephants in their natural range countries. Significant challenges exist for successful treatment of EEHV-HD, which include timely recognition of disease onset and limited availability of highly effective treatment options. To address this problem, our goal is to prevent lethal disease in young elephants by developing a vaccine that elicits robust and durable humoral and cell-mediated immunity against EEHV. EEHV glycoprotein B (gB) is a major target for cellular and humoral immunity in elephants previously exposed to EEHV. Therefore, we generated a vaccine containing recombinant EEHV1A gB together with a liposome formulated TLR-4 and saponin combination adjuvant (SLA-LSQ). CD-1 mice that received one or two vaccinations with the vaccine elicited significant anti-gB antibody and polyfunctional CD4+ and CD8+ T cell responses, while no adverse effects of vaccination were observed. Overall, our findings demonstrate that an adjuvanted gB protein subunit vaccine stimulates robust humoral and cell-mediated immune responses and supports its potential use in elephants.
Collapse
Affiliation(s)
- Jennifer L Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Tabitha E Hoornweg
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands.
| | - Jie Tan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Rongsheng Peng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| | - Willem Schaftenaar
- Veterinary Advisor EAZA Elephant TAG, Rotterdam Zoo, Blijdorplaan 8, 3041 JG Rotterdam, The Netherlands.
| | - Victor P M G Rutten
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa.
| | - Cornelis A M de Haan
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands.
| | - Paul D Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA.
| |
Collapse
|
45
|
Krauter S, Büscher N, Bräuchle E, Ortega Iannazzo S, Penner I, Krämer N, Gogesch P, Thomas S, Kreutz M, Dejung M, Freiwald A, Butter F, Waibler Z, Plachter B. An Attenuated Strain of Human Cytomegalovirus for the Establishment of a Subviral Particle Vaccine. Vaccines (Basel) 2022; 10:vaccines10081326. [PMID: 36016214 PMCID: PMC9413975 DOI: 10.3390/vaccines10081326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection is associated with severe disease conditions either following congenital transmission of the virus or viral reactivation in immunosuppressed individuals. Consequently, the establishment of a protective vaccine is of high medical need. Several candidates have been tested in preclinical and clinical studies, yet no vaccine has been licensed. Subviral dense bodies (DB) are a promising vaccine candidate. We have recently provided a GMP-compliant protocol for the production of DB, based on a genetically modified version of the HCMV laboratory strain Towne, expressing the pentameric complex of envelope protein gH-gL-pUL128-131 (Towne-UL130rep). In this work, we genetically attenuated Towne-UL130rep by abrogating the expression of the tegument protein pUL25 and by fusing the destabilizing domain ddFKBP to the N-terminus of the IE1- and IE2-proteins of HCMV. The resulting strain, termed TR-VAC, produced high amounts of DB under IE1/IE2 repressive conditions and concomitant supplementation of the viral terminase inhibitor letermovir to the producer cell culture. TR-VAC DB retained the capacity to induce neutralizing antibodies. A complex pattern of host protein induction was observed by mass spectrometry following exposure of primary human monocytes with TR-VAC DB. Human monocyte-derived dendritic cells (DC) moderately increased the expression of activation markers and MHC molecules upon stimulation with TR-VAC DB. In a co-culture with autologous T cells, the TR-VAC DB-stimulated DC induced a robust HCMV-specific T cell-activation and –proliferation. Exposure of donor-derived monocytic cells to DB led to the activation of a rapid innate immune response. This comprehensive data set thus shows that TR-VAC is an optimal attenuated seed virus strain for the production of a DB vaccine to be tested in clinical studies.
Collapse
Affiliation(s)
- Steffi Krauter
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Nicole Büscher
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Eric Bräuchle
- Division of Immunology, Section 3/1 “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, D-63225 Langen, Germany
| | - Samira Ortega Iannazzo
- Division of Immunology, Section 3/1 “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, D-63225 Langen, Germany
| | - Inessa Penner
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Nadine Krämer
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Patricia Gogesch
- Division of Immunology, Section 3/1 “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, D-63225 Langen, Germany
| | - Simone Thomas
- Leibniz Institute for Immunotherapy, Regensburg and Klinik und Poliklinik für Innere Medizin III, Hämatologie und Internistische Onkologie, University Hospital Regensburg, D-93053 Regensburg, Germany
| | - Marina Kreutz
- Leibniz Institute for Immunotherapy, Regensburg and Klinik und Poliklinik für Innere Medizin III, Hämatologie und Internistische Onkologie, University Hospital Regensburg, D-93053 Regensburg, Germany
| | - Mario Dejung
- Proteomics Core Facility, Institute of Molecular Biology, D-55128 Mainz, Germany
| | - Anja Freiwald
- Proteomics Core Facility, Institute of Molecular Biology, D-55128 Mainz, Germany
| | - Falk Butter
- Proteomics Core Facility, Institute of Molecular Biology, D-55128 Mainz, Germany
| | - Zoe Waibler
- Division of Immunology, Section 3/1 “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, D-63225 Langen, Germany
| | - Bodo Plachter
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
- Correspondence: ; Tel.: +49-6131-179232
| |
Collapse
|
46
|
Abstract
Over a century of research has focused on improving our understanding of congenital cytomegalovirus (cCMV), yet it remains the most common congenital infection in the United States, affecting 3 to 6 per 1000 live born infants each year. Pregnancies affected by cCMV are at a heightened risk of spontaneous abortion and intrauterine fetal demise. Neonates born with cCMV are also at substantial risk for long-term neurodevelopmental sequelae and disability, including sensorineural hearing loss, even those born without clinically apparent disease. Considerable progress has been made in recent years in study of the epidemiology and transmission of cCMV, developing better diagnostic strategies, implementing newborn screening programs, improving therapeutics, and launching vaccine trials. In this article, we review recent developments in the understanding of the virology and immunobiology of cytomegalovirus. We further discuss how this knowledge informs our understanding of the pathophysiology of cCMV and directs strategies aimed at improving outcomes and quality of life for congenitally infected children. We also provide an update on the epidemiology of cCMV in the United States, evolving scientific understanding of maternal-fetal transmission, enhanced screening approaches, and recognition of neonatal and long-term sequelae. Finally, we review the current landscape of pediatric cCMV research and provide recommendations for novel and high-priority areas for future investigation.
Collapse
Affiliation(s)
- Megan H. Pesch
- University of Michigan and CS Mott Children’s Hospital, Division of Developmental and Behavioral Pediatrics, Department of Pediatrics, Ann Arbor, Michigan
| | - Mark R. Schleiss
- Department of Pediatrics, University of Minnesota Medical School, Division of Pediatric Infectious Diseases
| |
Collapse
|
47
|
Liu R, Wu N, Gao H, Liang S, Yue K, -Dong T, Dong X, Xu LP, Wang Y, Zhang XH, Liu J, Huang XJ. Distinct activities of Vδ1 + T cells upon different cytomegalovirus reactivation status after hematopoietic transplantation. Immunology 2022; 167:368-383. [PMID: 35795896 DOI: 10.1111/imm.13542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cytomegalovirus (CMV) reactivation is the most frequent viral infectious complication correlating to non-relapse mortality after allogeneic hematopoietic cell transplantation (alloHCT). The intrinsic anti-CMV immunity has not been completely elucidated. γδ T cells have drawn increasing attentions due to their distinct biological features and potential ability against viral infections. Previous studies reported a general association of γδ T cells or Vδ2-negative γδ T cells with CMV reactivation. Whereas researches for the direct responses and specific functions of γδ T subsets remain limited, especially in the scenario of alloHCT. Herein, we initially demonstrated that Vδ1+ T cells directly and independently recognized cell-free CMV and CMV-infected target cells, and inhibited CMV replication in vitro. The anti-CMV effect of Vδ1+ T cells was partially through TCRγδ, TLR2, and NKG2D receptor pathways. Further investigation about the anti-CMV characteristics of Vδ1+ T cells was performed in a clinical cohort with different CMV reactivation status after alloHCT. We found that occasional CMV reactivation remarkably increased the recovery levels and stimulated the functional activity of Vδ1+ T cells. Whereas disability of Vδ1+ T cells was observed upon refractory CMV reactivation, indicating the differential responses of Vδ1+ T cells under different CMV reactivation status. CXCL10 and IFN-β that were dramatically induced by occasional CMV reactivation could re-activate the deficient Vδ1+ T cells from recipients with refractory CMV reactivation. These findings unveiled the distinct activities of Vδ1+ T cells in anti-CMV immunity after alloHCT and may help develop novel strategies for the treatment of CMV infectious diseases.
Collapse
Affiliation(s)
- Ruoyang Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ning Wu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Gao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Shuang Liang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Keli Yue
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Tianhui -Dong
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xinyu Dong
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Jiangying Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
48
|
Dasari V, Beckett K, Horsefield S, Ambalathingal G, Khanna R. A bivalent CMV vaccine formulated with human compatible TLR9 agonist CpG1018 elicits potent cellular and humoral immunity in HLA expressing mice. PLoS Pathog 2022; 18:e1010403. [PMID: 35737741 PMCID: PMC9223316 DOI: 10.1371/journal.ppat.1010403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022] Open
Abstract
There is now convincing evidence that the successful development of an effective CMV vaccine will require improved formulation and adjuvant selection that is capable of inducing both humoral and cellular immune responses. Here, we have designed a novel bivalent subunit vaccine formulation based on CMV-encoded oligomeric glycoprotein B (gB) and polyepitope protein in combination with human compatible TLR9 agonist CpG1018. The polyepitope protein includes multiple minimal HLA class I-restricted CD8+ T cell epitopes from different antigens of CMV. This subunit vaccine generated durable anti-viral antibodies, CMV-specific CD4+ and CD8+ T cell responses in multiple HLA expressing mice. Antibody responses included broad TH1 isotypes (IgG2a, IgG2b and IgG3) and potently neutralized CMV infection in fibroblasts and epithelial cells. Furthermore, polyfunctional antigen-specific T cell immunity and antiviral antibody responses showed long-term memory maintenance. These observations argue that this novel vaccine strategy, if applied to humans, could facilitate the generation of robust humoral and cellular immune responses which may be more effective in preventing CMV-associated complications in various clinical settings. Human Cytomegalovirus (CMV) is a significant human pathogen. Generally, in healthy people CMV causes mild symptomatic disease, but during pregnancy CMV can transmit from mother to foetus (1 out of every 200 live births worldwide) and lead to sensorineural hearing loss, vision impairment and central nervous system damage. In transplant patients, CMV can cause serious complications leading to organ rejection and even death. Currently, there is no licensed vaccine available to prevent CMV-associated complications in pregnant women and transplant patients. Here, we have developed a novel bivalent CMV vaccine formulation consisting of recombinant CMVpoly and gB proteins in combination with human compatible adjuvant CpG1018. Preclinical immunogenicity evaluation in multiple HLA expressing mice demonstrated that bivalent CMV vaccine formulation consistently generated robust CMV-specific neutralising antibodies, CD4+ and CD8+ T cell responses. More importantly, long-term follow-up analysis showed that the CMV vaccine can induce durable CMV-specific humoral and cellular immune responses. Our results support further development of this bivalent subunit CMV vaccine to test safety, immunogenicity and efficacy in humans.
Collapse
Affiliation(s)
- Vijayendra Dasari
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- * E-mail: (VD); (RK)
| | - Kirrilee Beckett
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Shane Horsefield
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - George Ambalathingal
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- * E-mail: (VD); (RK)
| |
Collapse
|
49
|
Lilleri D, Tassis B, Pugni L, Ronchi A, Pietrasanta C, Spinillo A, Arossa A, Achille C, Vergani P, Ornaghi S, Riboni S, Cavoretto P, Candiani M, Gaeta G, Prefumo F, Fratelli N, Fichera A, Vignali M, Barbasetti Di Prun A, Fabbri E, Cetin I, Locatelli A, Consonni S, Rutolo S, Miotto E, Savasi V, Di Giminiani M, Cromi A, Binda S, Fiorina L, Furione M, Cassinelli G, Klersy C. Prevalence, Outcome, and Prevention of Congenital Cytomegalovirus Infection in Neonates Born to Women With Preconception Immunity (CHILd Study). Clin Infect Dis 2022; 76:513-520. [PMID: 35717635 PMCID: PMC9907511 DOI: 10.1093/cid/ciac482] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/09/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Human cytomegalovirus (HCMV) is the leading infectious cause of congenital disabilities. We designed a prospective study to investigate the rate, outcome, and risk factors of congenital CMV (cCMV) infection in neonates born to immune women, and the potential need and effectiveness of hygiene recommendations in this population. METHODS The study was composed of 2 sequential parts: an epidemiology (part 1) and a prevention (part 2) study. Performance of part 2 depended upon a cCMV rate >0.4%. Women enrolled in part 1 did not receive hygiene recommendations. Newborns were screened by HCMV DNA testing in saliva and cCMV was confirmed by urine testing. RESULTS Saliva swabs were positive for HCMV DNA in 45/9661 newborns and cCMV was confirmed in 18 cases. The rate of cCMV was .19% (95% confidence interval [CI]: .11-.29%), and 3 out of 18 infants with cCMV had symptoms of CMV at birth. Age, nationality, occupation, and contact with children were similar between mothers of infected and noninfected newborns. Twin pregnancy (odds ratio [OR]: 7.2; 95% CI: 1.7-32.2; P = .037) and maternal medical conditions (OR: 3.9; 95% CI: 1.5-10.1; P = .003) appeared associated with cCMV. Given the rate of cCMV was lower than expected, the prevention part of the study was cancelled. CONCLUSIONS Newborns from women with preconception immunity have a low rate of cCMV, which appears to be mostly due to reactivation of the latent virus. Therefore, serological screening in childbearing age would be pivotal to identify HCMV-seropositive women, whose newborns have a low risk of cCMV. CLINICAL TRIALS REGISTRATION www.clinicaltrials.gov (NCT03973359).
Collapse
Affiliation(s)
- Daniele Lilleri
- Correspondence: D. Lilleri, Microbiologia e Virologia, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy ()
| | - Beatrice Tassis
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenza Pugni
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Ronchi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Carlo Pietrasanta
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Arsenio Spinillo
- Ostetricia e Ginecologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessia Arossa
- Ostetricia e Ginecologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Cristian Achille
- Neonatologia e Terapia intensiva neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Patrizia Vergani
- Fondazione Monza Brianza per il Bambino e la sua Mamma Onlus c/o Ospedale San Gerardo, Università Milano-Bicocca Scuola di Medicina e Chirurgia, Monza, Italy
| | - Sara Ornaghi
- Fondazione Monza Brianza per il Bambino e la sua Mamma Onlus c/o Ospedale San Gerardo, Università Milano-Bicocca Scuola di Medicina e Chirurgia, Monza, Italy
| | - Silvia Riboni
- Fondazione Monza Brianza per il Bambino e la sua Mamma Onlus c/o Ospedale San Gerardo, Università Milano-Bicocca Scuola di Medicina e Chirurgia, Monza, Italy
| | - Paolo Cavoretto
- Gynecology and Obstetrics Department, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - Massimo Candiani
- Gynecology and Obstetrics Department, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - Gerarda Gaeta
- Gynecology and Obstetrics Department, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - Federico Prefumo
- ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Nicola Fratelli
- ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Anna Fichera
- ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Michele Vignali
- ASST Fatebenefratelli-Sacco, Ospedale Macedonio Melloni, Milan, Italy,Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Allegra Barbasetti Di Prun
- ASST Fatebenefratelli-Sacco, Ospedale Macedonio Melloni, Milan, Italy,Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Elisa Fabbri
- Dipartimento di Ostetricia e Ginecologia, Ospedale dei Bambini Vittore Buzzi, Università di Milano, Milan, Italy
| | - Irene Cetin
- Dipartimento di Ostetricia e Ginecologia, Ospedale dei Bambini Vittore Buzzi, Università di Milano, Milan, Italy
| | - Anna Locatelli
- ASST Brianza (Ospedali di Carate e Vimercate), Vimercate, Italy
| | - Sara Consonni
- ASST Brianza (Ospedali di Carate e Vimercate), Vimercate, Italy
| | | | | | - Valeria Savasi
- Unit of Obstetrics and Gynecology, ASST Fatebenefratelli-Sacco, Milan, Italy,Department of Biological and Clinical Sciences, University of Milan, Milan, Italy
| | - Maria Di Giminiani
- Unit of Obstetrics and Gynecology, ASST Fatebenefratelli-Sacco, Milan, Italy,Department of Biological and Clinical Sciences, University of Milan, Milan, Italy
| | | | - Sandro Binda
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Loretta Fiorina
- Microbiologia e Virologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Milena Furione
- Microbiologia e Virologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gabriela Cassinelli
- Microbiologia e Virologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Catherine Klersy
- Epidemiologia clinica e Biostatistica, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | |
Collapse
|
50
|
Hong J, Wei D, Zhong L, Wu Q, Chen K, Zhang W, Yang Y, Chen J, Xia N, Zhang X, Chen Y. Glycoprotein B Antibodies Completely Neutralize EBV Infection of B Cells. Front Immunol 2022; 13:920467. [PMID: 35711430 PMCID: PMC9197244 DOI: 10.3389/fimmu.2022.920467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 04/29/2022] [Indexed: 11/26/2022] Open
Abstract
The Epstein-Barr virus (EBV) is the first reported oncogenic herpesvirus that establishes persistent infection in B lymphocytes in 95% of adults worldwide. Glycoprotein B (gB) plays a predominant role in the fusion of the viral envelope with the host cell membrane. Hence, it is of great significance to isolate gB-specific fusion-inhibiting neutralizing antibodies (NAbs). AMMO5 is the only gB NAb but fails to antagonize B-cell infection. It is essential to isolate potent NAbs that can completely block EBV infection of B cells. Using hybridoma technology and neutralization assay, we isolate two gB NAbs 8A9 and 8C12 that are capable of completely neutralizing B-cell infection in vitro. In addition, 8A9 shows cross-reactivity with rhesus lymphocryptovirus (rhLCV) gB. Competitive binding experiments demonstrate that 8A9 and 8C12 recognize novel epitopes that are different from the AMMO5 epitope. The epitopes of 8A9 and 8C12 are mapped to gB D-II, and the AMMO5 epitope is located precisely at gB aa 410-419. We find that 8A9 and 8C12 significantly inhibit gB-derived membrane fusion using a virus-free fusion assay. In summary, this study identifies two gB-specific NAbs that potently block EBV infection of B cells. Our work highlights the importance of gB D-II as a predominant neutralizing epitope, and aids in the rational design of therapeutics or vaccines based on gB.
Collapse
Affiliation(s)
- Junping Hong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Dongmei Wei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Ling Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qian Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Kaiyun Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Wanlin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yanbo Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Junyu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Xiao Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yixin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|