1
|
Portet Sulla V, Rafek R, Bertin-Jung I, Siest JP, Bouthry E, Rogier O, Jadoui A, Vauloup-Fellous C, Perillaud-Dubois C. Reliability of CMV-IgG kinetics in the diagnosis of CMV primary infection: sensitivity, specificity, and clinical implications. Microbiol Spectr 2025:e0045525. [PMID: 40525841 DOI: 10.1128/spectrum.00455-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 05/05/2025] [Indexed: 06/19/2025] Open
Abstract
Diagnosis of cytomegalovirus (CMV) primary infection (PI) during pregnancy relies on serology (CMV-IgG, IgM, and IgG avidity). However, as for toxoplasmosis, subsequent serology testing 3-5 weeks later is often performed to confirm the diagnosis. In this study, we aimed to show that testing CMV-IgG with different assays may lead to misinterpretation of CMV-IgG kinetics and to determine the sensitivity and specificity of CMV-IgG stability and significant increase to exclude or confirm recent CMV PI. We conducted a retrospective study on (i) a CMV-IgG external quality control program (2015-2022) and (ii) on CMV serology results obtained in our virology laboratory (2013-2023) in pregnant women with positive CMV-IgM and a subsequent serum sample collected 3-5 weeks later. Analysis of 21 CMV-IgG external quality control serum samples highlighted significant differences in CMV-IgG values, with variations up to a factor of 185 between different immunoassays for the same positive sample. In 434 pregnant women, the sensitivity of a significant CMV-IgG increase to predict recent PI was 32.9% (95% CI = 26.5-39.2), while CMV-IgG stability specificity to exclude PI <3 months was 32.9% (95% CI = 26.5-39.2). Our observations highlight the discrepancies in CMV-IgG values with different assays and the major importance of CMV-IgG avidity in the diagnosis of recent CMV PI in case of positive CMV-IgM. We also demonstrate that retesting IgG on a sample collected 3-5 weeks later is not helpful and can be confusing.IMPORTANCEThis article is the first to address cytomegalovirus (CMV)-IgG kinetics and their reliability in the serological diagnosis of CMV. In our experience, many clinical virologists and laboratory practitioners still rely on kinetics for diagnosis. However, our study clearly demonstrates that this approach is misleading and that avidity testing should always be performed. Additionally, we conducted a robust study highlighting discrepancies between CMV serology techniques, emphasizing the importance for practitioners, particularly gynecologists, to avoid monitoring serology results using different testing methods.
Collapse
Affiliation(s)
- Vincent Portet Sulla
- WHO Rubella National Reference Laboratory, Division of Virology, Department of Biology Genetics, Paul Brousse Hospital, Paris Saclay University Hospital, APHP, Villejuif, France
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Paris Saclay University, INSERM U1184, CEA, Fontenay-aux-Roses, France
| | - Rana Rafek
- WHO Rubella National Reference Laboratory, Division of Virology, Department of Biology Genetics, Paul Brousse Hospital, Paris Saclay University Hospital, APHP, Villejuif, France
| | | | | | - Elise Bouthry
- Department of Virology, Angers University Hospital, Angers, France
| | - Olivier Rogier
- WHO Rubella National Reference Laboratory, Division of Virology, Department of Biology Genetics, Paul Brousse Hospital, Paris Saclay University Hospital, APHP, Villejuif, France
| | - Abir Jadoui
- WHO Rubella National Reference Laboratory, Division of Virology, Department of Biology Genetics, Paul Brousse Hospital, Paris Saclay University Hospital, APHP, Villejuif, France
| | - Christelle Vauloup-Fellous
- WHO Rubella National Reference Laboratory, Division of Virology, Department of Biology Genetics, Paul Brousse Hospital, Paris Saclay University Hospital, APHP, Villejuif, France
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Paris Saclay University, INSERM U1184, CEA, Fontenay-aux-Roses, France
| | - Claire Perillaud-Dubois
- Virology Department, Sorbonne University, Saint-Antoine Hospital, AP-HP, Pierre Louis Epidemiology and Public Health Institute (iPLESP), INSERM 1136, Paris, France
| |
Collapse
|
2
|
Hadjiiona A, Michaelides I, Kummer P, Kappelmeyer M, Koeninger A, Reuschel E. Frequency of CMV testing during pregnancy-a retrospective study. Arch Gynecol Obstet 2025; 311:1297-1304. [PMID: 39890645 PMCID: PMC12033204 DOI: 10.1007/s00404-025-07962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025]
Abstract
PURPOSE The cytomegalovirus (CMV) belongs to the family of human Herpesviridae and is distributed worldwide. It is the most common cause of viral congenital infections and can have serious consequences for the health of the fetus in the event of a vertical transmission. This study, taking place for the first time in Upper Palatinate, Bavaria, aims to evaluate the frequency of CMV testing among pregnant women in our region in Germany, which for some individuals can be an expensive individual health service. METHODS Retrospectively, 1000 pregnant patients aged 17-45 years who were treated in the University Clinic St. Hedwig, Regensburg, Germany, were included in the study. It was investigated whether a CMV test was carried out during pregnancy and which results were obtained. RESULTS 597 patients (59.7%) had not received a CMV test during pregnancy. Among the 403 (40.3%) patients who had undergone CMV testing, seropositivity was detected in 143 (35.5%). 257 patients (63.8%) were seronegative, while 3 (0.74%) had a primary infection. CONCLUSION Although CMV is the most common pathogen of viral congenital infections and can severely impair the health of affected newborns, CMV diagnostics during pregnancy is still not an integral part of the maternity guidelines in Germany, but rather an individual healthcare service, meaning that the patients undergoing the test must bear the full cost. An antiviral treatment with valacyclovir has shown good preventive and therapeutic success, but unfortunately, there is currently no vaccination available to prevent vertical transmission, which is why early diagnosis and hygiene measures are the most important means of preventing seroconversion of the mother and possible infection of the fetus.
Collapse
Affiliation(s)
- A Hadjiiona
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstrasse 1-3, 93049, Regensburg, Germany.
| | - I Michaelides
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Regensburg, Regensburg, Germany
| | - P Kummer
- Department of Otorhinolaryngology, Department of Phoniatrics and Pediatric Audiology, University Hospital Regensburg, Regensburg, Germany
| | - M Kappelmeyer
- Chair of Obstetrics and Gynecology, Focus: Obstetrics, University of Regensburg, Biopark 1, Regensburg, Germany
| | - A Koeninger
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstrasse 1-3, 93049, Regensburg, Germany
| | - E Reuschel
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstrasse 1-3, 93049, Regensburg, Germany
| |
Collapse
|
3
|
Kang H, Wang L, Chen Y, Dong T, Pan Y, Liu Q. Screening and prenatal diagnosis of fetal cytomegalovirus infection: experience in a western Chinese city. BMC Infect Dis 2025; 25:542. [PMID: 40240971 PMCID: PMC12004672 DOI: 10.1186/s12879-025-10910-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVE To explore the application value of serology testing and fetal ultrasound examination in screening for fetal cytomegalovirus(CMV) infection. To explore the application value of CMV polymerase chain reaction (PCR) in amnio fluids in prenatal diagnosis of fetal CMV infection. METHODS This is a retrospective study performed at Chengdu Women's and Children's Central Hospital between 2021 and 2024. The baseline of preconception CMV infections and recent infections were investigated via serology testing. Pregnant women with suspicious serology for recent infection and/or ultrasound abnormalities suggestive of fetal infection underwent amniocentesis to obtain amniotic fluid for CMV PCR. The pregnancy outcomes of women with suspicious serology for recent infection and those who underwent amniocentesis were analyzed. RESULTS The seroprevalence was 95.65%(55262/57778) and 0.99%(571/57778) for immunoglobulin G(IgG) and immunoglobulin M (IgM) antibodies respectively. Among the 315 pregnant women with positive IgM results who performed CMV IgG avidity index and/or dynamic IgM/ IgG tests, only 2.22%(7/315) confirmed recent infection and 2.54%(8/315) suspected recent infection: three terminated pregnancies including one with positive CMV PCR results and two with other reasons, and 12 gave birth to asymptomatic babies. 576 pregnant women with suspicious serology for recent infection and/or ultrasound abnormalities suggestive of fetal infection performed CMV PCR in amniotic fluid. Among them, we got 438 pregnancy outcomes, including 25 terminated pregnancies, eight stillbirths, and 405 live births. None of the 405 live births had symptoms related to CMV infection at birth, except two with mild hearing loss without known cause. Four fetal infections were diagnosed, including three who performed amniocentesis and CMV PCR and one accidentally diagnosed by copy number variation sequencing(CNV-seq). Three of them were negative for IgM in first-trimester screening. CONCLUSION Ultrasound screening plays a more important role than serology screening in areas with a high seroprevalence of CMV antibodies. Assays for both chromosome disorders and CMV PCR in amniotic fluids should be suggested to pregnant women with ultrasound abnormalities suggestive of fetal infection.
Collapse
Affiliation(s)
- Han Kang
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Lingxi Wang
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Yifei Chen
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Ting Dong
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Yuchun Pan
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Qingsong Liu
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China.
| |
Collapse
|
4
|
Habel JR, Nguyen THO, Allen LF, Hagen RR, Kedzierski L, Allen EK, Jia X, Li S, Tarasova I, Minervina AA, Pogorelyy MV, Saunders PM, Clatch A, Evrard M, Xu C, Koay HF, Khan MAAK, de Alwis N, Mackay LK, Barrow AD, Douros C, Karapanagiotidis T, Nicholson S, Bond K, Williamson DA, Lappas M, Walker S, Hannan NJ, Brooks AG, Schroeder J, Crawford JC, Thomas PG, Rowntree LC, Kedzierska K. Single-cell immune profiling of third trimester pregnancies defines importance of chemokine receptors and prevalence of CMV-induced NK cells in the periphery and decidua. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.24.25324489. [PMID: 40196247 PMCID: PMC11974773 DOI: 10.1101/2025.03.24.25324489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Human pregnancy presents a unique physiological state that allows for growth of an antigenically dissimilar foetus and requires specific adaptations of the immune system. The immune system plays an important role in establishing and maintaining successful pregnancy, yet deep understanding of immunological responses in pregnancy is lacking. To provide in-depth understanding of the immunological landscape of pregnancy, with a focus on NK cells, we used high-throughput cell surface proteome screening of >350 markers and scRNAseq with 130 CITE-seq antibodies to identify key differentially expressed molecules and investigated their potential roles in altered immunity. We identified skewing in NK cell subsets towards higher frequencies of CD56bright cells caused by a reduced number of CD56dim cells in the peripheral blood during pregnancy and provide evidence for a role of chemokine receptor, CX3CR1, in NK cell activation. In addition, we defined a new cytomegalovirus (CMV)-induced decidual NK cell population in CMV+ pregnancies with tissue-residency markers. Overall, our data provide fundamental knowledge into how NK cell immunity is altered in pregnancy, and key knowledge needed to inform vaccine and therapeutic strategies to manage infections or pregnancy complications.
Collapse
Affiliation(s)
- Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ruth R Hagen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - E Kaitlynn Allen
- Computational Sciences Initiative, Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Shihan Li
- Computational Sciences Initiative, Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Ilariya Tarasova
- Computational Sciences Initiative, Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Anastasia A Minervina
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mikhail V Pogorelyy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Philippa M Saunders
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Allison Clatch
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Maximilien Evrard
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Calvin Xu
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Md Abdullah-Al-Kamran Khan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Natasha de Alwis
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Alexander D Barrow
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Celia Douros
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Theo Karapanagiotidis
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katherine Bond
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Microbiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Deborah A Williamson
- Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Martha Lappas
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
| | - Susan Walker
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
| | - Natalie J Hannan
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jan Schroeder
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| |
Collapse
|
5
|
Smith MD, Seleme MC, Marquez-Lago T, Chen JW, Mach M, Britt WJ. Early control of cochlear viral load limits cochlear inflammation and prevents virus-induced sensorineural hearing loss. J Neuroinflammation 2025; 22:92. [PMID: 40122833 PMCID: PMC11931849 DOI: 10.1186/s12974-025-03416-4] [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: 12/02/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Human cytomegalovirus (HCMV) is the most common viral infection acquired in utero and a leading cause of neurodevelopmental abnormalities, including sensorineural hearing loss (SNHL). In previous studies using a murine model of HCMV induced SNHL, hearing loss was correlated with virus-induced cochlear inflammation but not cochlear viral load. However, these previous findings were determined at the time of auditory testing, a time poiont well past critical periods of auditory development. In the current study, cochlear virus load early in auditory development could be correlated with the magnitude of virus-induced cochlear inflammation, cochlear histopathology and the development of hearing loss. Transcriptional profiling at early times after infection revealed dysregulation of multiple well described deafness-related genes (DRG). Treatment with antiviral antibodies early after infection decreased cochlear virus load, cochlear inflammation, cochlear histopathology, and normalized DRG expression arguing that virus-induced cochlear inflammation can result in pleiotropic effects on the developing auditory system. Finally, this model also demonstrated that sterilizing immunity was unnecessary for prevention of SNHL, thus providing a rationale for inteventions that could limit, but not completely prevent HCMV infection of the developing auditory system.
Collapse
Affiliation(s)
- Matthew D Smith
- Department of Microbiology, Heersink School of Medicince, UAB, Birmingham, Ala, USA
| | - Maria C Seleme
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Jiung-Wen Chen
- Department of Genetics, Heersink School of Medicince, UAB, Birmingham, Ala, USA
| | - Michael Mach
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - William J Britt
- Department of Pediatrics, Microbiology, and Neurobiology, Heersink School of Medicince, UAB, Birmingham, Ala, USA.
| |
Collapse
|
6
|
Payne H, Aaltoranta M, Veikkolainen V, Kent N, Gkouleli T, Lennon A, Ramgoolam T, Adams SP. A high-sensitivity, high-throughput newborn screening assay for congenital cytomegalovirus-is it time for universal screening in the United Kingdom? Front Pediatr 2025; 13:1543132. [PMID: 40171173 PMCID: PMC11959010 DOI: 10.3389/fped.2025.1543132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/20/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction Congenital cytomegalovirus (cCMV) is the leading cause of neurodevelopmental and hearing impairment resulting from in utero infection, affecting over a million infants globally each year. Early antiviral treatment can limit sequelae; however, most newborns are diagnosed late-or not at all-due to the lack of universal screening. Ensuring the availability of appropriate screening tools is critical to facilitate accurate and timely cCMV diagnosis. Methods A high-sensitivity, high-throughput commercial CMV PCR kit targeting the RRP30 control gene and a conserved region of CMV DNA was provided by Revvity and tested in three population groups: (1) leftover dried blood spot (DBS) samples from the UK newborn screening programme, (2) DBS samples from children with CMV viraemia unrelated to cCMV, and (3) DBS and dried saliva samples from infants with and without cCMV. Results Of 3,345 anonymised newborn DBS samples analysed, CMV was detected in 22 cases (0.66%), with a mean cycle threshold value of 36.70 (range 31.87-41.68). Assay development demonstrated a sensitivity of 2.04 CMV IU per reaction. This level of sensitivity was replicated using DBS samples prepared from infant/child blood samples with known levels of CMV, suggesting that the sensitivity reflects 2,000-3,000 CMV IU/mL blood. Discussion We demonstrated high analytical sensitivity of the qPCR assay with an optimal extraction protocol, making it an effective strategy for cCMV screening using DBS samples. These data suggest a potential cCMV incidence rate of up to 0.66% in the United Kingdom, equivalent to 3,960 infants per year, 25% of whom may develop long-term sequelae, which could be improved through early diagnosis and treatment.
Collapse
Affiliation(s)
- H. Payne
- Section of Paediatric Infectious Disease, School of Medicine, Imperial College London, London, United Kingdom
| | - M. Aaltoranta
- Research and Development, Revvity Inc., Turku, Finland
| | | | - N. Kent
- SIHMDS-Haematology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - T. Gkouleli
- SIHMDS-Haematology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - A. Lennon
- Department of Virology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - T. Ramgoolam
- Department of Infection, Immunity and Inflammation, Institute of Child Health, London, United Kingdom
- Newborn Screening Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - S. P. Adams
- SIHMDS-Haematology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
7
|
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: 2] [Impact Index Per Article: 2.0] [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
|
8
|
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
|
9
|
Pinninti SG, Britt WJ, Boppana SB. Auditory and Vestibular Involvement in Congenital Cytomegalovirus Infection. Pathogens 2024; 13:1019. [PMID: 39599572 PMCID: PMC11597862 DOI: 10.3390/pathogens13111019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Congenital cytomegalovirus infection (cCMV) is a frequent cause of non-hereditary sensorineural hearing loss (SNHL) and developmental disabilities. The contribution of cCMV to childhood hearing loss has been estimated to be about 25% of all hearing loss in children at 4 years of age. Although the vestibular insufficiency (VI) in cCMV has not been well-characterized and therefore, underestimated, recent studies suggest that VI is also frequent in children with cCMV and can lead to adverse neurodevelopmental outcomes. The pathogenesis of SNHL and VI in children with cCMV has been thought to be from direct viral cytopathic effects as well as local inflammatory responses playing a role. Hearing loss in cCMV can be of varying degrees of severity, unilateral or bilateral, present at birth or develop later (late-onset), and can progress or fluctuate in early childhood. Therefore, newborn hearing screening fails to identify a significant number of children with CMV-related SNHL. Although the natural history of cCMV-associated VI has not been well characterized, recent data suggests that it is likely that VI also varies considerably with respect to the laterality, timing of onset, degree of the deficit, and continued deterioration during early childhood. This article summarizes the current understanding of the natural history and pathogenesis of auditory and vestibular disorders in children with cCMV.
Collapse
Affiliation(s)
- Swetha G. Pinninti
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.G.P.); (W.J.B.)
| | - William J. Britt
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.G.P.); (W.J.B.)
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Suresh B. Boppana
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.G.P.); (W.J.B.)
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
10
|
Szulc W, Szydłowska N, Smyk JM, Majewska A. Progress and Challenges in the Management of Congenital Cytomegalovirus Infection. Clin Pract 2024; 14:2445-2462. [PMID: 39585019 PMCID: PMC11587044 DOI: 10.3390/clinpract14060191] [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: 07/17/2024] [Revised: 10/22/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024] Open
Abstract
Congenital cytomegalovirus (CMV) infection is the most common intrauterine viral infection with a significant impact on the foetus and newborn. Current diagnostic practice includes serological testing for specific antibodies, but there are no global screening protocols. Maternal CMV screening is often performed in conjunction with antenatal ultrasound. While most infections are asymptomatic, severe cases can lead to long-term disability or death. Antiviral therapies, mainly ganciclovir and valganciclovir, are reserved for symptomatic patients, especially those with central nervous system involvement. Although effective, these treatments are associated with significant side effects such as neutropenia and hepatotoxicity. Foscarnet and cidofovir are used as alternatives, but their efficacy and safety require further study in paediatric patient populations. The effectiveness of passive prophylaxis is still uncertain. The lack of universally accepted guidelines for diagnosis, treatment, and prevention and the risk of serious side effects highlight the need for continued research. This review evaluates current therapeutic strategies, discusses their efficacy and associated risks, and highlights the need for innovative approaches to improve outcomes for affected neonates.
Collapse
Affiliation(s)
| | | | | | - Anna Majewska
- Department of Medical Microbiology, Medical University of Warsaw, Chalubinskiego 5 Str., 02-004 Warsaw, Poland; (W.S.); (J.M.S.)
| |
Collapse
|
11
|
Al Beloushi M, Saleh H, Ahmed B, Konje JC. Congenital and Perinatal Viral Infections: Consequences for the Mother and Fetus. Viruses 2024; 16:1698. [PMID: 39599813 PMCID: PMC11599085 DOI: 10.3390/v16111698] [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: 06/05/2024] [Revised: 10/13/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Viruses are the most common congenital infections in humans and an important cause of foetal malformations, neonatal morbidity, and mortality. The effects of these infections, which are transmitted in utero (transplacentally), during childbirth or in the puerperium depend on the timing of the infections. These vary from miscarriages (usually with infections in very early pregnancy), congenital malformations (when the infections occur during organogenesis) and morbidity (with infections occurring late in pregnancy, during childbirth or after delivery). The most common of these viruses are cytomegalovirus, hepatitis, herpes simplex type-2, parvovirus B19, rubella, varicella zoster and zika viruses. There are currently very few efficacious antiviral agents licensed for use in pregnancy. For most of these infections, therefore, prevention is mainly by vaccination (where there is a vaccine). The administration of immunoglobulins to those exposed to the virus to offer passive immunity or appropriate measures to avoid being infected would be options to minimise the infections and their consequences. In this review, we discuss some of the congenital and perinatal infections and their consequences on both the mother and fetus and their management focusing mainly on prevention.
Collapse
Affiliation(s)
- Mariam Al Beloushi
- Women’s Wellness and Research Centre Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (M.A.B.); (H.S.)
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Huda Saleh
- Women’s Wellness and Research Centre Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (M.A.B.); (H.S.)
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Badreldeen Ahmed
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
- Feto Maternal Centre, Al Markhiya Doha, Doha P.O. Box 34181, Qatar
- Department of Obstetrics and Gynaecology Weill Cornell Medicine, Doha P.O. Box 24144, Qatar
| | - Justin C. Konje
- Feto Maternal Centre, Al Markhiya Doha, Doha P.O. Box 34181, Qatar
- Department of Obstetrics and Gynaecology Weill Cornell Medicine, Doha P.O. Box 24144, Qatar
- Department of Health Sciences, University of Leicester, P.O. Box 7717, Leicester LE2 7LX, UK
| |
Collapse
|
12
|
Forli F, Capobianco S, Berrettini S, Bruschini L, Lorenzoni F, Fiori S, Lazzerini F. Long-term outcomes of congenital cytomegalovirus infection in children early identified by extended hearing-targeted screening. Int J Pediatr Otorhinolaryngol 2024; 184:112070. [PMID: 39191004 DOI: 10.1016/j.ijporl.2024.112070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVES Congenital Cytomegalovirus (cCMV) has been associated with hearing, vision, and neurodevelopmental long-term sequelae. Despite the social burden associated with the disease, a universally accepted consensus on screening, diagnostic, therapeutic and follow-up approaches has not been reached. The present observational retrospective study aims at describing long-term sequelae and radiological abnormalities associated with cCMV in children early identified by extended hearing-targeted screening and evaluated by audiological follow-up in a single III Level Audiological Referral Center for at least 2 years. METHODS Audiological neonatal and follow-up data were available for all subjects. Data collection included clinical neonatal and virological assessment at birth. Ophthalmological, neurodevelopmental and neuroradiological follow-up abnormalities compatible with cCMV sequelae were collected by clinical reports. Spearman's rank correlation coefficient (rho-ρ) was used to evaluate possible correlations among the considered parameters. RESULTS 61 newborns were identified by extended hearing-targeted cCMV screening and diagnosed mostly (83.6 %) by PCR viral DNA extraction in urine collected within the 15° day of life. Seventeen babies were born preterm, with a mean gestational age of 33.5 weeks. Sixteen patients (26.2 %) were admitted to an Intensive or sub-Intensive Neonatal Care Unit. At birth, 35 newborns were symptomatic (57.3 %), and 19 of them received antiviral treatment by valganciclovir or ganciclovir. Overall, 20 children (32.7 %) were diagnosed with sensorineural hearing loss (SNHL), among them 17 (85 %) were refer at the newborn hearing screening while 3 (15 %) were Pass. 5/20 children (25 %) presented isolated SNHL, while in 15/20 (75 %) children SNHL was associated to other long-term sequelae. In 5 patients (25 %) a progression of the hearing threshold was observed, with a mean age of progression of 26 months of age. Risk factors for progression were a worse final hearing threshold (Spearman's ρ = 0.434; p = 0.0001) and a worse hearing threshold at birth (Spearman's ρ = 0.298; p = 0.020). Thirteen children were fitted with hearing aids, 8 of whom subsequently underwent cochlear implantation. Concerning long term impairments, 10/61 children (17 %) presented a variety of ophthalmological sequelae, while 16/40 cCMV patients (40 %) were diagnosed with neurodevelopmental abnormalities. Language delays were significantly associated with a worse hearing threshold (ρ = 0.582; p = 0.0001) and with other neurocognitive abnormalities (ρ = 0.677, p = 0.0001). 30 children underwent radiological brain evaluation by Magnetic Resonance Imaging, and 63.3 % of them presented abnormalities compatible with cCMV. Mean viral load at birth did not show significant associations with long-term sequelae. CONCLUSIONS The study highlights the diverse and significant long-term sequelae of cCMV infection detected through early screening. With a significant proportion of cCMV children developing sensorineural hearing loss, ophthalmological and neurodevelopmental issues, the results emphasize the importance of continuous, multidisciplinary follow-up. Early identification and tailored interventions are crucial for improving the long-term health and quality of life of children affected by cCMV.
Collapse
Affiliation(s)
- Francesca Forli
- Otolaryngology, Audiology, and Phoniatrics Unit, University of Pisa, Pisa, Italy.
| | - Silvia Capobianco
- Otolaryngology, Audiology, and Phoniatrics Unit, University of Pisa, Pisa, Italy
| | - Stefano Berrettini
- Otolaryngology, Audiology, and Phoniatrics Unit, University of Pisa, Pisa, Italy
| | - Luca Bruschini
- Otolaryngology, Audiology, and Phoniatrics Unit, University of Pisa, Pisa, Italy
| | - Francesca Lorenzoni
- Neonatal Unit and Neonatal Intensive Care Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Simona Fiori
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy; Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Lazzerini
- Otolaryngology, Audiology, and Phoniatrics Unit, University of Pisa, Pisa, Italy
| |
Collapse
|
13
|
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] [Download PDF] [Figures] [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
|
14
|
Gana N, Huluță I, Cătănescu MȘ, Apostol LM, Nedelea FM, Sima RM, Botezatu R, Panaitescu AM, Gică N. Congenital Cytomegalovirus-Related Hearing Loss. Audiol Res 2024; 14:507-517. [PMID: 38920964 PMCID: PMC11200402 DOI: 10.3390/audiolres14030043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
Congenital hearing loss is a significant global health concern that affects millions of newborns and infants worldwide, posing substantial challenges for affected individuals, their families, and healthcare systems. This condition, present at birth, can stem from genetic factors, in utero exposures, infections, or complications during pregnancy or childbirth. The spectrum of congenital hearing loss ranges from mild to profound, impacting the development of speech, language, and cognitive skills, thereby influencing educational achievements, social integration, and future employment opportunities. Early detection and intervention strategies, such as newborn hearing screenings, genetic counseling, and the use of hearing aids or cochlear implants, are crucial for mitigating these impacts. This review article aims to explore the diagnostic approaches and management strategies for congenital cytomegalovirus-related hearing loss, emphasizing the importance of interdisciplinary care and the potential for technological advances to improve outcomes for affected individuals.
Collapse
Affiliation(s)
- Nicoleta Gana
- Filantropia Clinical Hospital, 011171 Bucharest, Romania; (N.G.); (I.H.); (M.-Ș.C.); (R.-M.S.); (R.B.); (A.M.P.); (N.G.)
| | - Iulia Huluță
- Filantropia Clinical Hospital, 011171 Bucharest, Romania; (N.G.); (I.H.); (M.-Ș.C.); (R.-M.S.); (R.B.); (A.M.P.); (N.G.)
- Gynecology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Mihai-Ștefan Cătănescu
- Filantropia Clinical Hospital, 011171 Bucharest, Romania; (N.G.); (I.H.); (M.-Ș.C.); (R.-M.S.); (R.B.); (A.M.P.); (N.G.)
| | - Livia-Mihaela Apostol
- Gynecology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Florina Mihaela Nedelea
- Gynecology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Romina-Marina Sima
- Filantropia Clinical Hospital, 011171 Bucharest, Romania; (N.G.); (I.H.); (M.-Ș.C.); (R.-M.S.); (R.B.); (A.M.P.); (N.G.)
| | - Radu Botezatu
- Filantropia Clinical Hospital, 011171 Bucharest, Romania; (N.G.); (I.H.); (M.-Ș.C.); (R.-M.S.); (R.B.); (A.M.P.); (N.G.)
- Gynecology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Anca Maria Panaitescu
- Filantropia Clinical Hospital, 011171 Bucharest, Romania; (N.G.); (I.H.); (M.-Ș.C.); (R.-M.S.); (R.B.); (A.M.P.); (N.G.)
- Gynecology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Nicolae Gică
- Filantropia Clinical Hospital, 011171 Bucharest, Romania; (N.G.); (I.H.); (M.-Ș.C.); (R.-M.S.); (R.B.); (A.M.P.); (N.G.)
- Gynecology Department, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| |
Collapse
|
15
|
Fourgeaud J, Magny JF, Couderc S, Garcia P, Maillotte AM, Benard M, Pinquier D, Minodier P, Astruc D, Patural H, Parat S, Guillois B, Garenne A, Guilleminot T, Parodi M, Bussières L, Ghout I, Ville Y, Leruez-Ville M. Predictors of the Outcome at 2 Years in Neonates With Congenital Cytomegalovirus Infection. Pediatrics 2024; 153:e2023063531. [PMID: 38487823 DOI: 10.1542/peds.2023-063531] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Approximately 20% of neonates with congenital cytomegalovirus (cCMV) develop long-term sequelae. The ability to accurately predict long-term outcomes as early as the neonatal period would help to provide for appropriate parental counseling and treatment indications. With this study, we aimed to identify neonatal predictive markers of cCMV long-term outcomes. METHODS As this study's subjects, we chose neonates diagnosed with cCMV in 13 hospitals throughout France recruited from 2013 to 2017 and evaluated for at least 2 years with thorough clinical, audiology, and imaging evaluations and psychomotor development tests. RESULTS A total of 253 neonates were included, and 3 were later excluded because of the identification of a genetic disorder. A total of 227 were followed up for 2 years: 187/227 (82%) and 34/227 (15%) were infected after a maternal primary or nonprimary infection, respectively, 91/227 (40%) were symptomatic at birth, and 44/227 (19%) had cCMV sequelae. Maternal primary infection in the first trimester was the strongest prognosis factor (odds ratio = 38.34 [95% confidence interval, 5.02-293], P < .001). A predictive model of no risk of sequelae at 2 years of age according to normal hearing loss at birth, normal cerebral ultrasound, and normal platelet count had 98% specificity, 69% sensitivity, and 0.89 area under the curve (95% confidence interval, 0.83-0.96). CONCLUSIONS In the studied population, children with normal hearing at birth, normal platelet count at birth, and a normal cranial ultrasound had no risk of neurologic sequelae and a low risk of delayed unilateral sensorineural hearing loss. The use of this model based on readily available neonatal markers should help clinicians establish a personalized care pathway for each cCMV neonate.
Collapse
Affiliation(s)
- Jacques Fourgeaud
- URP 7328 FETUS, Université Paris Cité, Paris, France
- Virology Laboratory, Reference Laboratory for Cytomegalovirus Infections
| | | | - Sophie Couderc
- Maternity, Hospital Intercommunal Poissy-Saint Germain, Poissy, France
| | - Patricia Garcia
- Neonatology and Intensive Care Department, AP-HM, Hospital La Conception, Marseille, France
| | | | - Melinda Benard
- Department of Neonatology, Toulouse University Hospital, Infinity, Université Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Didier Pinquier
- Department of Neonatology, Rouen University Hospital, Rouen, France
| | | | - Dominique Astruc
- Department of Neonatology, Strasbourg University Hospital, Strasbourg, France
| | - Hugues Patural
- Department of Neonatology, Saint-Etienne, University Hospital, Saint-Etienne, France
| | - Sophie Parat
- Maternity, AP-HP, Hospital Cochin, Paris, France
| | - Bernard Guillois
- Department of Neonatology, CHU de Caen, Caen, France
- Université Caen Normandie Medical School, Caen, France
| | | | - Tiffany Guilleminot
- URP 7328 FETUS, Université Paris Cité, Paris, France
- Virology Laboratory, Reference Laboratory for Cytomegalovirus Infections
| | | | - Laurence Bussières
- URP 7328 FETUS, Université Paris Cité, Paris, France
- Clinical Research Unit, P-HP
| | - Idir Ghout
- Cegedim Health Data, Boulogne-Billancourt, France
| | - Yves Ville
- URP 7328 FETUS, Université Paris Cité, Paris, France
- Maternity, AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - Marianne Leruez-Ville
- URP 7328 FETUS, Université Paris Cité, Paris, France
- Virology Laboratory, Reference Laboratory for Cytomegalovirus Infections
| |
Collapse
|
16
|
Hillam K, Suarez D, Nielson C, Traxler A, Sommer E, Winslow A, Holley A, Huang E, Hughes M, Firpo MA, Rower J, Park AH. Hearing Following Prolonged and Delayed Ganciclovir Treatment in a Murine Cytomegalovirus Model. Laryngoscope 2024; 134:433-438. [PMID: 37421238 DOI: 10.1002/lary.30860] [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: 04/03/2023] [Revised: 05/20/2023] [Accepted: 06/14/2023] [Indexed: 07/10/2023]
Abstract
OBJECTIVE Compare hearing outcomes utilizing standard, prolonged and delayed ganciclovir (GCV) therapy in a murine model of cytomegalovirus (CMV). METHODS BALB/c mice were inoculated with mouse cytomegalovirus (mCMV) or saline via intracerebral injection on postnatal day 3 (p3). Intraperitoneal GCV or saline was administered at 12 h intervals for the duration of the standard (p3-p17), delayed (p30-p44), or prolonged treatment windows (p3-p31). Auditory thresholds were assessed using distortion product otoacoustic emission (DPOAE) and auditory brainstem response (ABR) testing at 4, 6, and 8 weeks of age. Blood and tissue samples were harvested from mice on p17 and p37 one hour after GCV administration, and their concentrations were assessed via liquid chromatography-mass spectrometry. RESULTS A delayed course of GCV improved ABR but not DPOAE thresholds in mCMV-infected mice. A prolonged course of GCV did not provide better hearing thresholds than those administered standard treatment. The average GCV concentration in all 17-day-old mice tissue was significantly higher than those in older 37-day-old mice. CONCLUSION Delayed GCV treatment provided a hearing benefit on ABR over untreated mCMV infected mice. Prolonged CGV administration showed no benefit compared to a shorter duration GCV treatment. GCV drug concentrations both systemically and in the cochlea are much lower in older mice. These results have potential implications for the clinical management of cCMV infected children. LEVEL OF EVIDENCE NA Laryngoscope, 134:433-438, 2024.
Collapse
Affiliation(s)
- Katrina Hillam
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Daniel Suarez
- Department of Otolaryngology, State University of New York Downstate Medical Center, Stony Brook, New York, U.S.A
| | - Christopher Nielson
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Abigail Traxler
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Elizabeth Sommer
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Anna Winslow
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Anna Holley
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Emily Huang
- Johns Hopkins University School of Medicine, Baltimore, Maryland, U.S.A
| | - Maura Hughes
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Matthew A Firpo
- Department of Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| | - Joseph Rower
- University of Utah Center for Human Toxicology and Department of Pharmacology and Toxicology, Salt Lake City, Utah, U.S.A
| | - Albert H Park
- Department of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, Utah, U.S.A
| |
Collapse
|
17
|
STOKES CALEB, J. MELVIN ANN. Viral Infections of the Fetus and Newborn. AVERY'S DISEASES OF THE NEWBORN 2024:450-486.e24. [DOI: 10.1016/b978-0-323-82823-9.00034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
18
|
Boppana SB, van Boven M, Britt WJ, Gantt S, Griffiths PD, Grosse SD, Hyde TB, Lanzieri TM, Mussi-Pinhata MM, Pallas SE, Pinninti SG, Rawlinson WD, Ross SA, Vossen ACTM, Fowler KB. Vaccine value profile for cytomegalovirus. Vaccine 2023; 41 Suppl 2:S53-S75. [PMID: 37806805 DOI: 10.1016/j.vaccine.2023.06.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/28/2023] [Accepted: 06/02/2023] [Indexed: 10/10/2023]
Abstract
Cytomegalovirus (CMV) is the most common infectious cause of congenital malformation and a leading cause of developmental disabilities such as sensorineural hearing loss (SNHL), motor and cognitive deficits. The significant disease burden from congenital CMV infection (cCMV) led the US National Institute of Medicine to rank CMV vaccine development as the highest priority. An average of 6.7/1000 live births are affected by cCMV, but the prevalence varies across and within countries. In contrast to other congenital infections such as rubella and toxoplasmosis, the prevalence of cCMV increases with CMV seroprevalence rates in the population. The true global burden of cCMV disease is likely underestimated because most infected infants (85-90 %) have asymptomatic infection and are not identified. However, about 7-11 % of those with asymptomatic infection will develop SNHL throughout early childhood. Although no licensed CMV vaccine exists, several candidate vaccines are in development, including one currently in phase 3 trials. Licensure of one or more vaccine candidates is feasible within the next five years. Various models of CMV vaccine strategies employing different target populations have shown to provide substantial benefit in reducing cCMV. Although CMV can cause end-organ disease with significant morbidity and mortality in immunocompromised individuals, the focus of this vaccine value profile (VVP) is on preventing or reducing the cCMV disease burden. This CMV VVP provides a high-level, comprehensive assessment of the currently available data to inform the potential public health, economic, and societal value of CMV vaccines. The CMV VVP was developed by a working group of subject matter experts from academia, public health groups, policy organizations, and non-profit organizations. All contributors have extensive expertise on various elements of the CMV VVP and have described the state of knowledge and identified the current gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Suresh B Boppana
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Michiel van Boven
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, and Julius Center for Health Sciences and Primary Care, Department of Epidemiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - William J Britt
- Departments of Pediatrics, Microbiology, and Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, USA
| | - Soren Gantt
- Centre de recherche du CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Paul D Griffiths
- Emeritus Professor of Virology, University College London, United Kingdom
| | - Scott D Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Terri B Hyde
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Tatiana M Lanzieri
- Measles, Rubella, and Cytomegalovirus Epidemiology Team, Viral Vaccine Preventable Diseases Branch / Division of Viral Diseases. National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Marisa M Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Sarah E Pallas
- Global Immunization Division, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Swetha G Pinninti
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William D Rawlinson
- Serology and Virology Division, NSW Health Pathology Randwick, Prince of Wales Hospital, Sydney, Australia; School of Biotechnology and Biomolecular Sciences, and School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Shannon A Ross
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ann C T M Vossen
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karen B Fowler
- Departments of Pediatrics and Epidemiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
19
|
Choodinatha HK, Jeon MR, Choi BY, Lee KN, Kim HJ, Park JY. Cytomegalovirus infection during pregnancy. Obstet Gynecol Sci 2023; 66:463-476. [PMID: 37537975 PMCID: PMC10663402 DOI: 10.5468/ogs.23117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/06/2023] [Accepted: 07/04/2023] [Indexed: 08/05/2023] Open
Abstract
Cytomegalovirus (CMV) infection during pregnancy is a global silent problem. Additionally, it is the leading cause of congenital infections, non-genetic sensorineural hearing loss, and neurodevelopmental delays in infants. However, this has barely been recognized globally. This condition lacks adequate attention, which is further emphasized by the lack of awareness among healthcare workers and the general population. The impact of CMV infection is often overlooked because of the asymptomatic nature of its presentation in infected pregnant women and newborns, difficulty in diagnosis, and the perception that infants born to women with pre-existing antibodies against CMV have normal neonatal outcomes. This article highlights the latest information on the epidemiology, transmission, clinical manifestations, and development of CMV infection and its management. We reviewed the pathophysiology and clinical manifestations of CMV infection in pregnant women, diagnostic methods, including screening and prognostic markers, and updates in treatment modalities. Current advancements in research on vaccination and hyperimmunoglobulins with worldwide treatment protocols are highlighted.
Collapse
Affiliation(s)
- Harshitha Kallubhavi Choodinatha
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Korea
| | - Min Ryeong Jeon
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Korea
| | - Bo Young Choi
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Korea
| | - Kyong-No Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Korea
| | - Hyeon Ji Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Korea
| | - Jee Yoon Park
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Korea
| |
Collapse
|
20
|
Schirwani-Hartl N, Palmrich P, Haberl C, Perkmann-Nagele N, Kiss H, Berger A, Rittenschober-Böhm J, Kasprian G, Kienast P, Khalil A, Binder J. Biweekly Versus Monthly Hyperimmune Globulin Therapy for Primary Cytomegalovirus Infection in Pregnancy. J Clin Med 2023; 12:6776. [PMID: 37959240 PMCID: PMC10649935 DOI: 10.3390/jcm12216776] [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: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Primary cytomegalovirus (CMV) infection during pregnancy is associated with an increased risk of congenital CMV (cCMV). Hyperimmune globulin (HIG) therapy has been proposed as a potential prophylaxis to reduce maternal-fetal transmission. Data on whether the administration of HIG every 2 weeks offers benefits over HIG administration every 4 weeks are lacking. This was a retrospective analysis including pregnant women with primary CMV infection diagnosed in the first or early second trimester between 2010 and 2022 treated with HIG every 4 weeks (300 IE HIG per kg) or every 2 weeks (200 IE HIG per kg), respectively. In total, 36 women (4 weeks: n = 26; 2 weeks: n = 10) and 39 newborns (4 weeks: n = 29; 2 weeks: n = 10) were included. The median gestational age at the first HIG administration was 13.1 weeks. There was no significant difference in the cCMV rates between the women who received HIG every 4 versus every 2 weeks (n = 8/24 [33.3%] vs. 3/10 [30.0%]; p = 0.850). An abnormal fetal ultrasound was present in three fetuses and fetal magnetic resonance imaging (MRI) anomalies in four fetuses were related to cCMV infection, with no significant difference in the frequency between the two groups. A larger study will be needed to determine whether HIG administration every 2 instead of every 4 weeks improves the maternal-fetal transmission rates.
Collapse
Affiliation(s)
- Nawa Schirwani-Hartl
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (N.S.-H.); (P.P.); (C.H.); (H.K.)
| | - Pilar Palmrich
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (N.S.-H.); (P.P.); (C.H.); (H.K.)
| | - Christina Haberl
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (N.S.-H.); (P.P.); (C.H.); (H.K.)
| | | | - Herbert Kiss
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (N.S.-H.); (P.P.); (C.H.); (H.K.)
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (A.B.); (J.R.-B.)
| | - Judith Rittenschober-Böhm
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (A.B.); (J.R.-B.)
| | - Gregor Kasprian
- Department of Radiology, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, 1090 Vienna, Austria; (G.K.); (P.K.)
| | - Patric Kienast
- Department of Radiology, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, 1090 Vienna, Austria; (G.K.); (P.K.)
| | - Asma Khalil
- Fetal Medicine Unit, St George’s University Hospitals NHS Foundation Trust, University of London, London WC1E 6BT, UK;
| | - Julia Binder
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (N.S.-H.); (P.P.); (C.H.); (H.K.)
| |
Collapse
|
21
|
Yin MZ, Gu YY, Shu JT, Zhang B, Su M, Zhang LP, Jiang YH, Qin G. Cost-effectiveness of cytomegalovirus vaccination for females in China: A decision-analytical Markov study. Vaccine 2023; 41:5825-5833. [PMID: 37580210 DOI: 10.1016/j.vaccine.2023.08.011] [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: 04/23/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND The global burden of disease caused by congenital cytomegalovirus (CMV) infection is high. Previous modeling studies have suggested that CMV vaccination may be cost-effective in developed countries. Congenital CMV infection is more likely driven by maternal non-primary infection in China. We aimed to measure the effectiveness and cost-effectiveness of population-level CMV vaccination in Chinese females. METHODS A decision tree Markov model was developed to simulate potential CMV vaccination strategies in a multi-cohort setting, with a population size of 1,000,000 each for the infant, adolescent (10-year-old) and young adult (20-year-old) cohorts. The hypothetical vaccines were assumed to have 50% efficacy, 20 years of protection, 70% coverage, at a price of US$120/dose for base-case analysis. Costs and disability-adjusted life years (DALYs) were discounted by 3% per year and the vaccination would be considered cost-effective if an incremental cost-effectiveness ratio (ICER) was lower than 2021 Chinese per capita GDP (US$12,500). FINDINGS For the pre-infection (PRI) vaccine efficacy type, the adolescent strategy was the most cost-effective, with an ICER of US$12,213 (12,134 to 12,291) pre DALY averted, compared with the next best strategy (young adult strategy). For pre- and post-infection (P&PI) efficacy type, the young adult strategy was the most cost-effective as it was cost-saving. In one-way analysis varying the PRI vaccine price, the infant strategy, adolescent strategy and the young adult strategy would be a dominant strategy over others if the vaccine cost ≤US$60, US$61-121 and US$122-251 per dose respectively. In contrast, the young adult strategy continued to be the preferred strategy until the P&PI vaccine price exceeded US$226/dose. Our main results were robust under a wide variety of sensitivity analyses and scenario analyses. INTERPRETATION CMV vaccination for females would be cost-effective and even cost-saving in China. Our findings had public health implications for control of CMV diseases.
Collapse
Affiliation(s)
- Meng-Zhao Yin
- Department of Infectious Diseases, Affiliated Hospital of Nantong University, Nantong, JS, China
| | - Yuan-Yuan Gu
- Centre for the Health Economy, Macquarie University, Sydney, NSW, Australia
| | - Jun-Tao Shu
- Department of Epidemiology and Biostatistics, School of Public Health, Nantong University, Nantong, JS, China
| | - Bin Zhang
- Department of Infectious Diseases, Affiliated Hospital of Nantong University, Nantong, JS, China
| | - Min Su
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, JS, China
| | - Lu-Ping Zhang
- Department of Otolaryngology, Affiliated Hospital of Nantong University, Nantong, JS, China.
| | - Yin-Hua Jiang
- Clinical Medicine Research Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, JS, China.
| | - Gang Qin
- Department of Infectious Diseases, Affiliated Hospital of Nantong University, Nantong, JS, China; Department of Epidemiology and Biostatistics, School of Public Health, Nantong University, Nantong, JS, China.
| |
Collapse
|
22
|
Pellkofer Y, Hammerl M, Griesmaier E, Sappler M, Gizewski ER, Kiechl-Kohlendorfer U, Neubauer V. The Effect of Postnatal Cytomegalovirus Infection on (Micro)structural Cerebral Development in Very Preterm Infants at Term-Equivalent Age. Neonatology 2023; 120:727-735. [PMID: 37634498 DOI: 10.1159/000532084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION There are some data indicating a negative impact of postnatal cytomegalovirus (CMV) infection on long-term neurodevelopmental outcome of preterm infants. So far, there is only little knowledge about a cerebral imaging correlate of these neurodevelopmental alterations induced by postnatal CMV infection in preterm infants. The aim of the current study was to investigate the effect of postnatal CMV infection on the incidence of brain injury and on microstructural brain maturation in very preterm infants at term-equivalent age. METHODS Infants <32 gestational weeks (02/2011-11/2018) received cerebral MRI including axial diffusion-weighted images at term-equivalent age. All infants were screened for CMV infection using urine/saliva samples, and infection was regarded as acquired postnatal if a sample became positive >5 postnatal days. We compared brain injury as well as fractional anisotropy and apparent diffusion coefficient in 14 defined cerebral regions between infants with and without postnatal CMV infection. RESULTS 401 infants were eligible, of whom 18 (4.5%) infants had a postnatal CMV infection. There were no significant differences in rates of brain injury or in microstructural brain development between both groups. This applied equally to the subgroup of infants <28 gestational weeks. CONCLUSION Although infants with postnatal CMV infection were born more immature and more frequently suffered from complications related to immaturity, we neither observed a higher rate of preterm brain injury nor disadvantageous alterations in microstructural brain maturation at term-equivalent age.
Collapse
Affiliation(s)
- Yasmin Pellkofer
- Department of Pediatrics II, Neonatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marlene Hammerl
- Department of Pediatrics II, Neonatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke Griesmaier
- Department of Pediatrics II, Neonatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Maria Sappler
- Department of Pediatrics II, Neonatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke Ruth Gizewski
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
- Neuroimaging Research Core Facility, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Vera Neubauer
- Department of Pediatrics II, Neonatology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
23
|
Sanchez-Durán MA, Maiz N, Liutsko L, Bielsa-Pascual J, García-Sierra R, Zientalska AM, Velasco I, Vazquez E, Gracia O, Ribas A, Sitja N, Nadales M, Martinez C, Gonce A, Frick MA, Guerrero-Martínez M, Violán C, Torán P, Falguera-Puig G, Gol R. Universal screening programme for cytomegalovirus infection in the first trimester of pregnancy: study protocol for an observational multicentre study in the area of Barcelona (CITEMB study). BMJ Open 2023; 13:e071997. [PMID: 37474185 PMCID: PMC10357649 DOI: 10.1136/bmjopen-2023-071997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
INTRODUCTION Congenital cytomegalovirus (cCMV) is the leading cause of non-genetic sensorineural hearing loss and one of the main causes of neurological disability. Despite this, no universal screening programme for cCMV has been implemented in Spain. A recent study has shown that early treatment with valaciclovir, initiated in the first trimester and before the onset of signs in the fetus, reduces the risk of fetal infection. This finding favours the implementation of a universal screening programme for cCMV.The aim of this study is to evaluate the performance of a universal screening programme for cCMV during the first trimester of pregnancy in a primary care setting. METHODS AND ANALYSIS This is an observational multicentre cohort study. The study will be conducted in four primary care settings from the Northern Metropolitan Barcelona area and three related hospitals and will last 3 years and will consist of a recruitment period of 18 months.In their first pregnancy visit, pregnant women will be offered to add a CMV serology test to the first trimester screening tests. Pregnant women with primary infection will be referred to the reference hospital, where they will continue treatment and follow-up according to the clinical protocol of the referral hospital, which includes treatment with valacyclovir. A CMV-PCR will be performed at birth on newborns of mothers with primary infection, and those who are infected will undergo neonatal follow-up for at least 12 months of life.For the analysis, the acceptance rate, the prevalence of primary CMV infections and the CMV seroprevalence in the first trimester of pregnancy will be studied. ETHICS AND DISSEMINATION Ethical approval was obtained from the University Institute Foundation for Primary Health Care Research Jordi Gol i Gurina Ethics Committee 22/097-P dated 27 April 2022.
Collapse
Affiliation(s)
- Maria Angeles Sanchez-Durán
- Maternal Fetal Medicine Unit, Department of Obstetrics; Universitat Autònoma de Barcelona, Hospital Vall d'Hebron, Barcelona, Spain
| | - Nerea Maiz
- Maternal Fetal Medicine Unit, Department of Obstetrics; Universitat Autònoma de Barcelona, Hospital Vall d'Hebron, Barcelona, Spain
| | - Liudmila Liutsko
- Unitat de Suport a la Recerca Metropolitana Nord, IDIAP Jordi Gol, Mataró, Spain
- Area Metropolitana Nord, Institut Català de la Salut, Barcelona, Spain
| | - Jofre Bielsa-Pascual
- Unitat de Suport a la Recerca Metropolitana Nord, IDIAP Jordi Gol, Mataró, Spain
| | - Rosa García-Sierra
- Unitat de Suport a la Recerca Metropolitana Nord, IDIAP Jordi Gol, Mataró, Spain
- Multidisciplinary Research Group in Health and Society (GREMSAS) (2017-SGR-917), Barcelona, Spain
| | - Aneta Monika Zientalska
- Service of Obstetrics and Gynecology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Inés Velasco
- Service of Obstetrics and Gynecology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Eva Vazquez
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Esquerra, Institut Català de la Salut, Barcelona, Spain
| | - Olga Gracia
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Muntanya, Institut Català de la Salut, Barcelona, Spain
| | - Aleida Ribas
- Atencio a la Salut Sexual i Reproductiva (ASSIR) St. Adrià de Besòs, Servei d'Atenció Primària Barcelonès Nord i Maresme, Institut Català de la Salut, Barcelona, Spain
| | - Nuria Sitja
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Santa Coloma de Gramenet, Atenció Primària Metropolitana Nord, Institut Català de la Salut, Santa Coloma de Gramenet, Spain
| | - Maria Nadales
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Badalona, Institut Català de la Salut, Badalona, Barcelona, Spain
| | - Cristina Martinez
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Catalunya, Institut Català de la Salut, Barcelona, Spain
- Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Anna Gonce
- Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- BCNatal: Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Hospital Clinic de Barcelona, Barcelona, Spain
| | - Marie Antoinette Frick
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Vall d'Hebron, Barcelona, Spain
| | - Mercedes Guerrero-Martínez
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Santa Coloma de Gramenet, Atenció Primària Metropolitana Nord, Institut Català de la Salut, Santa Coloma de Gramenet, Spain
- Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, IDIAP Jordi Gol, Barcelona, Spain
| | - Concepción Violán
- Unitat de Suport a la Recerca Metropolitana Nord, IDIAP Jordi Gol, Mataró, Spain
- Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain
| | - Pere Torán
- Unitat de Suport a la Recerca Metropolitana Nord, IDIAP Jordi Gol, Mataró, Spain
- Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain
| | - Gemma Falguera-Puig
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Metropolitana Nord, Institut Català de la Salut, Sabadell, Spain
- GRASSIR, Grup preconsolidat en Recerca en Atenció a la Salut Sexual i Reproductiva. IDIAP i AGAUR, IDIAP Jordi Gol, Barcelona, Spain
| | - Roser Gol
- Atencio a la Salut Sexual i Reproductiva (ASSIR) Badalona, Institut Català de la Salut, Badalona, Barcelona, Spain
- Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, IDIAP Jordi Gol, Barcelona, Spain
| |
Collapse
|
24
|
Nigro G, Muselli M, on behalf of the Congenital Cytomegalic Disease Collaborating Group. 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: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [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
|
25
|
Ben Shoham A, Schlesinger Y, Miskin I, Kalderon Z, Michaelson-Cohen R, Wiener-Well Y. Cytomegalovirus (CMV) seroprevalence among women at childbearing age, maternal and congenital CMV infection: policy implications of a descriptive, retrospective, community-based study. Isr J Health Policy Res 2023; 12:16. [PMID: 37098565 PMCID: PMC10131385 DOI: 10.1186/s13584-023-00566-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/20/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Maternal CMV infection during pregnancy, either primary or non-primary, may be associated with fetal infection and long-term sequelae. While guidelines recommend against it, screening for CMV in pregnant women is a prevalent clinical practice in Israel. Our aim is to provide updated, local, clinically relevant, epidemiological information about CMV seroprevalence among women at childbearing age, the incidence of maternal CMV infection during pregnancy and the prevalence of congenital CMV (cCMV), as well as to provide information about the yield of CMV serology testing. METHODS We performed a descriptive, retrospective study of women at childbearing age who were members of Clalit Health Services in the district of Jerusalem and had at least one gestation during the study period (2013-2019). We utilized serial serology tests to determine CMV serostatus at baseline and at pre/periconception and identified temporal changes in CMV serostatus. We then conducted a sub-sample analysis integrating inpatient data on newborns of women who gave birth in a single large medical center. cCMV was defined as either positive urine CMV-PCR test in a sample collected during the first 3 weeks of life, neonatal diagnosis of cCMV in the medical records, or prescription of valganciclovir during the neonatal period. RESULTS The study population Included 45,634 women with 84,110 associated gestational events. Initial CMV serostatus was positive in 89% women, with variation across different ethno-socioeconomic subgroups. Based on consecutive serology tests, the detected incidence rate of CMV infection was 2/1000 women follow-up years, among initially seropositive women, and 80/1000 women follow-up years, among initially seronegative women. CMV infection in pregnancy was identified among 0.2% of women who were seropositive at pre/periconception and among 10% of women who were seronegative. In a subsample, which included 31,191 associated gestational events, we identified 54 newborns with cCMV (1.9/1000 live births). The prevalence of cCMV among newborns of women who were seropositive at pre/periconception was lower than among newborns of women who were seronegative (2.1 vs. 7.1/1000). Frequent serology tests among women who were seronegative at pre/periconception detected most primary CMV infections in pregnancy that resulted in cCMV (21/24). However, among women who were seropositive, serology tests prior to birth detected none of the non-primary infections that resulted in cCMV (0/30). CONCLUSIONS In this retrospective community-based study among women of childbearing age characterized by multiparity and high seroprevalence of CMV, we find that consecutive CMV serology testing enabled to detect most primary CMV infections in pregnancy that led to cCMV in newborns but failed to detect non-primary CMV infections in pregnancy. Conducting CMV serology tests among seropositive women, despite guidelines' recommendations, has no clinical value, while it is costly and introduces further uncertainties and distress. We thus recommend against routine CMV serology testing among women who were seropositive in a prior serology test. We recommend CMV serology testing prior to pregnancy only among women known to be seronegative or women whose serology status is unknown.
Collapse
Affiliation(s)
- Assaf Ben Shoham
- Clalit Health Services, Yehuda Burla 26/28, 9371426, Jerusalem, Israel.
| | - Yechiel Schlesinger
- Wilf Children's Hospital, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ian Miskin
- Clalit Health Services, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ziva Kalderon
- Clalit Health Services, Yehuda Burla 26/28, 9371426, Jerusalem, Israel
| | - Rachel Michaelson-Cohen
- Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yonit Wiener-Well
- Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
26
|
Capra D, DosSantos MF, Sanz CK, Acosta Filha LG, Nunes P, Heringer M, Ximenes-da-Silva A, Pessoa L, de Mattos Coelho-Aguiar J, da Fonseca ACC, Mendes CB, da Rocha LS, Devalle S, Niemeyer Soares Filho P, Moura-Neto V. Pathophysiology and mechanisms of hearing impairment related to neonatal infection diseases. Front Microbiol 2023; 14:1162554. [PMID: 37125179 PMCID: PMC10140533 DOI: 10.3389/fmicb.2023.1162554] [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: 02/09/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
The inner ear, the organ of equilibrium and hearing, has an extraordinarily complex and intricate arrangement. It contains highly specialized structures meticulously tailored to permit auditory processing. However, hearing also relies on both peripheral and central pathways responsible for the neuronal transmission of auditory information from the cochlea to the corresponding cortical regions. Understanding the anatomy and physiology of all components forming the auditory system is key to better comprehending the pathophysiology of each disease that causes hearing impairment. In this narrative review, the authors focus on the pathophysiology as well as on cellular and molecular mechanisms that lead to hearing loss in different neonatal infectious diseases. To accomplish this objective, the morphology and function of the main structures responsible for auditory processing and the immune response leading to hearing loss were explored. Altogether, this information permits the proper understanding of each infectious disease discussed.
Collapse
Affiliation(s)
- Daniela Capra
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos F. DosSantos
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Odontologia (PPGO), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Propriedades Mecânicas e Biologia Celular (PropBio), Departamento de Prótese e Materiais Dentários, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Carolina K. Sanz
- Laboratório de Propriedades Mecânicas e Biologia Celular (PropBio), Departamento de Prótese e Materiais Dentários, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lionete Gall Acosta Filha
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Propriedades Mecânicas e Biologia Celular (PropBio), Departamento de Prótese e Materiais Dentários, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Priscila Nunes
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Manoela Heringer
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Luciana Pessoa
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Juliana de Mattos Coelho-Aguiar
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho (HUCFF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Carolina Carvalho da Fonseca
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | | | - Sylvie Devalle
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Paulo Niemeyer Soares Filho
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura-Neto
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho (HUCFF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Sartori P, Egloff C, Hcini N, Vauloup Fellous C, Périllaud-Dubois C, Picone O, Pomar L. Primary, Secondary, and Tertiary Prevention of Congenital Cytomegalovirus Infection. Viruses 2023; 15:v15040819. [PMID: 37112800 PMCID: PMC10146889 DOI: 10.3390/v15040819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/11/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Cytomegalovirus infection is the most common congenital infection, affecting about 1% of births worldwide. Several primary, secondary, and tertiary prevention strategies are already available during the prenatal period to help mitigate the immediate and long-term consequences of this infection. In this review, we aim to present and assess the efficacy of these strategies, including educating pregnant women and women of childbearing age on their knowledge of hygiene measures, development of vaccines, screening for cytomegalovirus infection during pregnancy (systematic versus targeted), prenatal diagnosis and prognostic assessments, and preventive and curative treatments in utero.
Collapse
Affiliation(s)
- Pauline Sartori
- School of Health Sciences (HESAV), University of Applied Sciences and Arts Western Switzerland, 1011 Lausanne, Switzerland
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Charles Egloff
- Assistance Publique-Hôpitaux de Paris APHP, Nord, Service de Gynécologie Obstétrique, Hôpital Louis Mourier, 92700 Colombes, France
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
| | - Najeh Hcini
- Department of Obstetrics and Gynaecology, West French Guiana Hospital Center, French 97320, Guyana
- CIC Inserm 1424 et DFR Santé Université Guyane, 97320 ST Laurent du Maroni, France
| | - Christelle Vauloup Fellous
- Université Paris-Saclay, INSERM U1193, 94804 Villejuif, France
- Laboratoire de Virologie, AP-HP, Hôpital Paul-Brousse, 94804 Villejuif, France
- Groupe de Recherche sur les Infections Pendant la Grossesse (GRIG), 75000 Paris, France
| | - Claire Périllaud-Dubois
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
- Virology Laboratory, AP-HP, Sorbonne Université, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Olivier Picone
- Assistance Publique-Hôpitaux de Paris APHP, Nord, Service de Gynécologie Obstétrique, Hôpital Louis Mourier, 92700 Colombes, France
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
- Groupe de Recherche sur les Infections Pendant la Grossesse (GRIG), 75000 Paris, France
| | - Léo Pomar
- School of Health Sciences (HESAV), University of Applied Sciences and Arts Western Switzerland, 1011 Lausanne, Switzerland
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
28
|
Management of pregnancy in left ventricular assist device and heart transplant recipients: a concise review. Curr Opin Cardiol 2023; 38:257-265. [PMID: 36927986 DOI: 10.1097/hco.0000000000001036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
PURPOSE OF REVIEW Women of reproductive age are increasingly undergoing heart transplantation (HT) or left ventricular assist device (LVAD) implantation for advanced heart failure. This review is intended to give an overview of the current state of the art management of pregnancy in patients with LVAD or HT recipients. RECENT FINDINGS Heart transplant recipients are at increased risk for graft rejection, renal dysfunction, preeclampsia and worsening of comorbidities (hypertension and diabetes). Patients with LVAD are at higher risk of thromboembolic events, infections, right ventricular failure and require close surveillance during pregnancy. Preconception counseling must be offered to all women of reproductive age group with HT or LVAD to avoid unplanned pregnancies. SUMMARY A multidisciplinary approach with close antepartum and postpartum surveillance is recommended.
Collapse
|
29
|
Cytomegalovirus Anterior Uveitis in an Immunocompetent Patient. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2023. [DOI: 10.1097/ipc.0000000000001200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
30
|
Soriano-Ramos M, Esquivel-De la Fuente E, Albert Vicent E, de la Calle M, Baquero-Artigao F, Domínguez-Rodríguez S, Cabanes M, Gómez-Montes E, Goncé A, Valdés-Bango M, Viñuela-Benéitez MC, Muñoz-Chápuli Gutiérrez M, Saavedra-Lozano J, Cuadrado Pérez I, Encinas B, Castells Vilella L, de la Serna Martínez M, Tagarro A, Rodríguez-Molino P, Giménez Quiles E, García Alcázar D, García Burguillo A, Folgueira MD, Navarro D, Blázquez-Gamero D, the CYTRIC Study Group. The role of the T-cell mediated immune response to Cytomegalovirus infection in intrauterine transmission. PLoS One 2023; 18:e0281341. [PMID: 36745589 PMCID: PMC9901742 DOI: 10.1371/journal.pone.0281341] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Prognostic markers for fetal transmission of Cytomegalovirus (CMV) infection during pregnancy are poorly understood. Maternal CMV-specific T-cell responses may help prevent fetal transmission and thus, we set out to assess whether this may be the case in pregnant women who develop a primary CMV infection. METHODS A multicenter prospective study was carried out at 8 hospitals in Spain, from January 2017 to April 2020. Blood samples were collected from pregnant women at the time the primary CMV infection was diagnosed to assess the T-cell response. Quantitative analysis of interferon producing specific CMV-CD8+/CD4+ cells was performed by intracellular cytokine flow cytometry. RESULTS In this study, 135 pregnant women with a suspected CMV infection were evaluated, 60 of whom had a primary CMV infection and samples available. Of these, 24 mothers transmitted the infection to the fetus and 36 did not. No association was found between the presence of specific CD4 or CD8 responses against CMV at the time maternal infection was diagnosed and the risk of fetal transmission. There was no transmission among women with an undetectable CMV viral load in blood at diagnosis. CONCLUSIONS In this cohort of pregnant women with a primary CMV infection, no association was found between the presence of a CMV T-cell response at the time of maternal infection and the risk of intrauterine transmission. A detectable CMV viral load in the maternal blood at diagnosis of the primary maternal infection may represent a relevant biomarker associated with fetal transmission.
Collapse
Affiliation(s)
- María Soriano-Ramos
- Department of Neonatology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (imas12), Fundación Biomédica del Hospital Universitario 12 de Octubre (FBHU12O), Madrid, Spain
- * E-mail:
| | - Estrella Esquivel-De la Fuente
- Instituto de Investigación Hospital 12 de Octubre (imas12), Fundación Biomédica del Hospital Universitario 12 de Octubre (FBHU12O), Madrid, Spain
| | - Eliseo Albert Vicent
- Microbiology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | - Fernando Baquero-Artigao
- Department of Infectious Diseases and Tropical Pediatrics, Hospital Universitario Infantil La Paz, Madrid, Spain
| | - Sara Domínguez-Rodríguez
- Instituto de Investigación Hospital 12 de Octubre (imas12), Fundación Biomédica del Hospital Universitario 12 de Octubre (FBHU12O), Madrid, Spain
| | - María Cabanes
- Obstetrics Department, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Enery Gómez-Montes
- Obstetrics Department, Fetal Medicine Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Anna Goncé
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetricia i Neonatologia, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Marta Valdés-Bango
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut Clínic de Ginecología, Obstetricia i Neonatologia, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Mª Carmen Viñuela-Benéitez
- Obstetrics Department, Hospital Gregorio Marañón, Complutense University, Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Mar Muñoz-Chápuli Gutiérrez
- Obstetrics Department, Hospital Gregorio Marañón, Complutense University, Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Jesús Saavedra-Lozano
- Hospital General Universitario Gregorio Marañón, Pediatric Infectious Diseases Unit, Universidad Complutense, Madrid, Spain
| | | | - Begoña Encinas
- Obstetrics Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Laura Castells Vilella
- Department of Neonatology, Hospital Universitari General de Catalunya, Grupo Quiron Salud, Sant Cugat del Vallès, Barcelona, Spain
| | | | - Alfredo Tagarro
- Paediatrics Department, Paediatrics Research Group, Hospital Universitario Infanta Sofía, Universidad Europea de Madrid, Madrid, Spain
| | - Paula Rodríguez-Molino
- Department of Infectious Diseases and Tropical Pediatrics, Hospital Universitario Infantil La Paz, Madrid, Spain
| | - Estela Giménez Quiles
- Microbiology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Diana García Alcázar
- Obstetrics Department, Fetal Medicine Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | - David Navarro
- Microbiology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Daniel Blázquez-Gamero
- Instituto de Investigación Hospital 12 de Octubre (imas12), Fundación Biomédica del Hospital Universitario 12 de Octubre (FBHU12O), Madrid, Spain
- Pediatric Infectious Diseases Unit, Hospital Universitario 12 de Octubre, Universidad Complutense, RITIP, Madrid, Spain
| | - the CYTRIC Study Group
- Pediatric Infectious Diseases Unit, Hospital Universitario 12 de Octubre, Universidad Complutense, RITIP, Madrid, Spain
| |
Collapse
|
31
|
Liu X, He G, Lan Y, Guo W, Liu X, Li J, Liu A, He M, Liu X, Fan Z, Zhang Y. Virome and metagenomic analysis reveal the distinct distribution of microbiota in human fetal gut during gestation. Front Immunol 2023; 13:1079294. [PMID: 36685560 PMCID: PMC9850102 DOI: 10.3389/fimmu.2022.1079294] [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: 10/25/2022] [Accepted: 12/14/2022] [Indexed: 01/07/2023] Open
Abstract
Studies have shown that fetal immune cell activation may result from potential exposure to microbes, although the presence of microbes in fetus has been a controversial topic. Here, we combined metagenomic and virome techniques to investigate the presence of bacteria and viruses in fetal tissues (small intestine, cecum, and rectum). We found that the fetal gut is not a sterile environment and has a low abundance but metabolically rich microbiome. Specifically, Proteobacteria and Actinobacteria were the dominant bacteria phyla of fetal gut. In total, 700 species viruses were detected, and Human betaherpesvirus 5 was the most abundant eukaryotic viruses. Especially, we first identified Methanobrevibacter smithii in fetal gut. Through the comparison with adults' gut microbiota we found that Firmicutes and Bacteroidetes gradually became the main force of gut microbiota during the process of growth and development. Interestingly, 6 antibiotic resistance genes were shared by the fetus and adults. Our results indicate the presence of microbes in the fetal gut and demonstrate the diversity of bacteria, archaea and viruses, which provide support for the studies related to early fetal immunity. This study further explores the specific composition of viruses in the fetal gut and the similarities between fetal and adults' gut microbiota, which is valuable for understanding human fetal immunity development during gestation.
Collapse
Affiliation(s)
- Xu Liu
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, China
| | - Guolin He
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yue Lan
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Weijie Guo
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xuyuan Liu
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jing Li
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Anqing Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Miao He
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Xinhui Liu
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, China,*Correspondence: Yaoyao Zhang, ; Zhenxin Fan,
| | - Yaoyao Zhang
- Key Laboratory of Bioresources and Ecoenvironment, Ministry of Education, College of Life Sciences, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,*Correspondence: Yaoyao Zhang, ; Zhenxin Fan,
| |
Collapse
|
32
|
Uchôa LRA, Lucato LT. Newer Updates in Pediatric Intracranial Infection. Semin Roentgenol 2023; 58:88-109. [PMID: 36732014 DOI: 10.1053/j.ro.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/24/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Luiz Ricardo Araújo Uchôa
- Neuroradiology Section, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, São Paulo, Brazil.
| | - Leandro Tavares Lucato
- Neuroradiology Section, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, São Paulo, Brazil; Grupo Fleury, São Paulo, Brazil
| |
Collapse
|
33
|
Kennedy KM, de Goffau MC, Perez-Muñoz ME, Arrieta MC, Bäckhed F, Bork P, Braun T, Bushman FD, Dore J, de Vos WM, Earl AM, Eisen JA, Elovitz MA, Ganal-Vonarburg SC, Gänzle MG, Garrett WS, Hall LJ, Hornef MW, Huttenhower C, Konnikova L, Lebeer S, Macpherson AJ, Massey RC, McHardy AC, Koren O, Lawley TD, Ley RE, O'Mahony L, O'Toole PW, Pamer EG, Parkhill J, Raes J, Rattei T, Salonen A, Segal E, Segata N, Shanahan F, Sloboda DM, Smith GCS, Sokol H, Spector TD, Surette MG, Tannock GW, Walker AW, Yassour M, Walter J. Questioning the fetal microbiome illustrates pitfalls of low-biomass microbial studies. Nature 2023; 613:639-649. [PMID: 36697862 PMCID: PMC11333990 DOI: 10.1038/s41586-022-05546-8] [Citation(s) in RCA: 191] [Impact Index Per Article: 95.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/09/2022] [Indexed: 01/26/2023]
Abstract
Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.
Collapse
Affiliation(s)
- Katherine M Kennedy
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Marcus C de Goffau
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Wellcome Sanger Institute, Cambridge, UK
| | - Maria Elisa Perez-Muñoz
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marie-Claire Arrieta
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, South Korea
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Thorsten Braun
- Department of Obstetrics and Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederic D Bushman
- Department of Microbiology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel Dore
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jonathan A Eisen
- Department of Evolution and Ecology, University of California, Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephanie C Ganal-Vonarburg
- Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michael G Gänzle
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
- Department of Medicine and Division of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Chair of Intestinal Microbiome, ZIEL-Institute for Food and Health, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Curtis Huttenhower
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liza Konnikova
- Departments of Pediatrics and Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Andrew J Macpherson
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Ruth C Massey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Alice Carolyn McHardy
- Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), Hannover Braunschweig site, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Trevor D Lawley
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jeroen Raes
- VIB Center for Microbiology, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thomas Rattei
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eran Segal
- Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Harry Sokol
- Gastroenterology Department, AP-HP, Saint Antoine Hospital, Centre de Recherche Saint-Antoine, CRSA, INSERM and Sorbonne Université, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gerald W Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alan W Walker
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Moran Yassour
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
- Department of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
34
|
Barlinn R, Dudman SG, Rollag H, Trogstad L, Lindstrøm JC, Magnus P. Maternal cytomegalovirus infection and delayed language development in children at 3 years of age-a nested case-control study in a large population-based pregnancy cohort. PLoS One 2022; 17:e0278623. [PMID: 36455052 PMCID: PMC9714838 DOI: 10.1371/journal.pone.0278623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION Maternal cytomegalovirus (CMV) infection in pregnancy may result in vertical transmission of CMV to the child. Long-term effects of congenital CMV infection include visual, cognitive as well as neurological impairment. The aim of this study was to estimate the odds ratios for CMV seropositivity and seroconversion in mothers, with and without delayed language development in 3 year old children, nested within a large cohort. MATERIAL AND METHODS The Norwegian Mother, Father and Child Cohort Study (MoBa) is a prospective population-based pregnancy cohort that includes 95 200 mothers and 114 500 children. Blood samples were obtained from mothers during pregnancy weeks 17 or 18 in pregnancy and after birth. We included 300 women from MoBa with children suffering from delayed language development at three years of age, based on validated questionnaires. Within the cohort, 1350 randomly selected women were included as controls to perform a nested case-control study. The cases and controls were tested for CMV IgG antibodies by an enzyme-linked immunosorbent assay. RESULTS Among mothers of cases, 63.2% were CMV-IgG positive in the sample at birth, as compared to 55.9% among controls; OR 1.36, (95% CI; 1.05 to 1.76). Also, among case mothers, 8/118 (6.8%) initially seronegative cases, seroconverted. Among initially seronegative controls, seroconversion occurred in 23/618 (3.7%) mothers. The OR for seroconversion in cases as compared to control mothers was 1.88 (CI; 0.82 to 4.31), thus not statistically significant different. CONCLUSION This study shows a higher risk of delayed language development at three years of age in children born by mothers seropositive for CMV, compared to children born from seronegative mothers.
Collapse
Affiliation(s)
- Regine Barlinn
- Division for Infection Control and Environmental Health, Department of Microbiology, Oslo University Hospital, Norwegian Institute of Public Health, Oslo, Norway
- * E-mail:
| | - Susanne G. Dudman
- Department of Microbiology, Oslo University Hospital, and University of Oslo, Oslo, Norway
| | - Halvor Rollag
- Department of Microbiology, Oslo University Hospital, and University of Oslo, Oslo, Norway
| | - Lill Trogstad
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Jonas C. Lindstrøm
- Division for Infection Control and Environmental Health, Department of Methods Development and Analytics, Norwegian Institute of Public Health, Oslo, Norway
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
Raynor EM, Martin HL, Poehlein E, Lee H, Lantos P. Impact of maternal cytomegalovirus seroconversion on newborn and childhood hearing loss. Laryngoscope Investig Otolaryngol 2022; 7:1626-1633. [PMID: 36258861 PMCID: PMC9575047 DOI: 10.1002/lio2.904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Objectives/hypothesis The objective of this study is to describe long-term hearing outcomes in infants born to mothers with a known cytomegalovirus (CMV) positivity who were not tested for congenital CMV. Study type Clinical research study. Design Retrospective cohort study. Methods Retrospective chart review was performed for mothers seropositive to CMV. Mother-infant dyads (130) were identified between January 1, 2013 and January 1, 2017. Outcomes data was collected through June 1, 2020. Demographics, risk factors for hearing loss, evidence of CMV infection, other causes of hearing loss, need for speech therapy services, and results of all hearing tests were collected. Results All 130 infants were asymptomatic and 5 were tested for congenital CMV. Five were negative for CMV and excluded from analyses. Of the remaining 125, only 1 had low-viral avidity IgG antibodies. None had IgM antibodies. Four children (3.2%) had hearing loss at last audiogram and one child had delayed onset SNHL due to an enlarged vestibular aqueduct. Speech therapy for communication was required for 33 children (26.4%). Conclusions Knowledge of maternal perinatal CMV status can allow for education about possible sequelae of cCMV, as well as trigger an alert for testing babies born to mothers with low-viral avidity IgG during the first trimester, when the risk of vertical transmission is highest. Also, babies born to CMV positive mothers may be more at risk for communication delays necessitating intervention. Studies focusing on the impact of maternal CMV related to childhood communication deficits could elucidate any direct relationships.
Collapse
Affiliation(s)
- Eileen M. Raynor
- Department of Head and Neck Surgery and Communication SciencesDuke UniversityDurhamNorth CarolinaUSA
| | - Hannah L. Martin
- Department of Head and Neck Surgery and Communication SciencesDuke UniversityDurhamNorth CarolinaUSA
| | - Emily Poehlein
- Department of Biostatistics and BioinformaticsDuke UniversityDurhamNorth CarolinaUSA
| | - Hui‐Jie Lee
- Department of Biostatistics and BioinformaticsDuke UniversityDurhamNorth CarolinaUSA
| | - Paul Lantos
- Department of Pediatric Infectious DiseaseDuke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
36
|
Neutralizing Antibodies to Human Cytomegalovirus Recombinant Proteins Reduce Infection in an Ex Vivo Model of Developing Human Placentas. Vaccines (Basel) 2022; 10:vaccines10071074. [PMID: 35891239 PMCID: PMC9315547 DOI: 10.3390/vaccines10071074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 12/03/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the leading viral cause of congenital disease and permanent birth defects worldwide. Although the development of an effective vaccine is a public health priority, no vaccines are approved. Among the major antigenic targets are glycoproteins in the virion envelope, including gB, which facilitates cellular entry, and the pentameric complex (gH/gL/pUL128-131), required for the infection of specialized cell types. In this study, sera from rabbits immunized with the recombinant pentameric complex were tested for their ability to neutralize infection of epithelial cells, fibroblasts, and primary placental cell types. Sera from rhesus macaques immunized with recombinant gB or gB plus pentameric complex were tested for HCMV neutralizing activity on both cultured cells and cell column cytotrophoblasts in first-trimester chorionic villus explants. Sera from rabbits immunized with the pentameric complex potently blocked infection by pathogenic viral strains in amniotic epithelial cells and cytotrophoblasts but were less effective in fibroblasts and trophoblast progenitor cells. Sera from rhesus macaques immunized with the pentameric complex and gB more strongly reduced infection in fibroblasts, epithelial cells, and chorionic villus explants than sera from immunization with gB alone. These results suggest that the pentameric complex and gB together elicit antibodies that could have potential as prophylactic vaccine antigens.
Collapse
|
37
|
Gabrani C, Mitsikas D, Giannakou K, Lamnisos D. Congenital Cytomegalovirus Infection and Ophthalmological Disorders: A Systematic Review. J Pediatr Ophthalmol Strabismus 2022; 60:86-94. [PMID: 35611826 DOI: 10.3928/01913913-20220426-01] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Congenital cytomegalovirus (CMV) infection is the most common congenital viral infection and can be a major cause of neurodevelopmental disabilities, including various ocular disorders in infants and young children. This review summarizes the evidence on the association between congenital CMV infection and the type and frequency of ocular disorders. A systematic search was conducted across PubMed and Cochrane Library from inception through December 2021 to identify studies examining the association between congenital CMV infection and the occurrence of ocular disorders in children born with this infection. Seventeen articles were identified. A total of 306 ocular disorders were identified in 977 children, with 45 of them (4.6%) being related to cases of chorioretinitis. A total of 259 of the 306 (84.6%) ocular disorders occurred in symptomatic children. Four studies did not report the number of children with ophthalmological disorders. The remaining 13 studies reporting this parameter included 666 children, of whom 85 had ophthalmological disorders (12.7%). Vision was tested in 556 children and visual impairement was detected in 91 of them (16.3%). Given the high prevalence of congenital CMV infection and the frequent occurrence of ocular disorders, the question that arises is whether there should be worldwide screening for congenital CMV infection so that both symptomatic and asymptomatic children can be detected in time to reduce the burden of the disease and its complications. [J Pediatr Ophthalmol Strabismus. 20XX;X(X):XX-XX.].
Collapse
|
38
|
Karamchandani U, Ahmed U, Rufai SR, Tan N, Tan W, Petrushkin H, Solebo AL. Long-term ocular and visual outcomes following symptomatic and asymptomatic congenital CMV infection: a systematic review protocol. BMJ Open 2022; 12:e059038. [PMID: 35584878 PMCID: PMC9119163 DOI: 10.1136/bmjopen-2021-059038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Cytomegalovirus (CMV) is one of the most common congenitally acquired infections worldwide. Visual impairment is a common outcome for symptomatic infants, with long-term ophthalmic surveillance often recommended. However, there are no clear guidelines for ophthalmic surveillance in infants with asymptomatic disease. We aim to conduct a systematic review to establish the overall prevalence and incidence of eye and vision related disorders following congenital CMV infection (cCMV). METHODS AND ANALYSIS A systematic review and meta-analysis (pending appropriate data for analysis) of cross-sectional and longitudinal studies will be conducted. The PubMed, Embase and CINAHL databases will be searched up to 29 March 2022 without date or language restrictions. Studies will be screened by at least two independent reviewers. Methodological quality of included studies will be assessed using the Joanna Briggs Institute tool. The primary outcome measures will be incidence and/or prevalence of vision impairment or ophthalmic disorders in patients with symptomatic and asymptomatic cCMV infection. A narrative synthesis will be conducted for all included studies. The overall prevalence will be estimated by pooling data using a random-effects model. Heterogeneity between studies will be estimated using Cochran's Q and the I2 statistics. Egger's test will be used to assess for publication bias. ETHICS AND DISSEMINATION Ethical approval is not required as there is no primary data collection. Study findings will be disseminated at scientific meetings and through publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER This is not a clinical trial, but the protocol has been registered: CRD42021284678 (PROSPERO).
Collapse
Affiliation(s)
| | - Umar Ahmed
- Imperial College London, London, UK
- Department of Ophthalmology, Royal Free London NHS Foundation Trust, London, UK
| | - Sohaib R Rufai
- Ulverscroft Eye Unit, University of Leicester, Leicester, Leicestershire, UK
- Department of Ophthalmology, Great Ormond Street Hospital, London, UK
| | - Naomi Tan
- Department of Ophthalmology, Royal Free London NHS Foundation Trust, London, UK
| | - Weijen Tan
- Department of Ophthalmology, Great Ormond Street Hospital, London, UK
| | - Harry Petrushkin
- Department of Uveitis, Moorfields Eye Hospital NHS Trust, London, UK
- Department of Rheumatology, Great Ormond Street Hospital, London, UK
| | - Ameenat Lola Solebo
- Department of Ophthalmology, Great Ormond Street Hospital, London, UK
- Department of Rheumatology, Great Ormond Street Hospital, London, UK
- UCL GOS Institute of Child Health, University College London, London, UK
| |
Collapse
|
39
|
Choi KY, El-Hamdi NS, McGregor A. Cross Strain Protection against Cytomegalovirus Reduces DISC Vaccine Efficacy against CMV in the Guinea Pig Model. Viruses 2022; 14:760. [PMID: 35458490 PMCID: PMC9031936 DOI: 10.3390/v14040760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023] Open
Abstract
Congenital cytomegalovirus (CMV) is a leading cause of disease in newborns and a vaccine is a high priority. The guinea pig is the only small animal model for congenital CMV but requires guinea pig cytomegalovirus (GPCMV). Previously, a disabled infectious single cycle (DISC) vaccine strategy demonstrated complete protection against congenital GPCMV (22122 strain) and required neutralizing antibodies to various viral glycoprotein complexes. This included gB, essential for all cell types, and the pentamer complex (PC) for infection of non-fibroblast cells. All GPCMV research has utilized prototype strain 22122 limiting the translational impact, as numerous human CMV strains exist allowing re-infection and congenital CMV despite convalescent immunity. A novel GPCMV strain isolate (designated TAMYC) enabled vaccine cross strain protection studies. A GPCMV DISC (PC+) vaccine (22122 strain) induced a comprehensive immune response in animals, but vaccinated animals challenged with the TAMYC strain virus resulted in sustained viremia and the virus spread to target organs (liver, lung and spleen) with a significant viral load in the salivary glands. Protection was better than natural convalescent immunity, but the results fell short of previous DISC vaccine sterilizing immunity against the homologous 22122 virus challenge, despite a similarity in viral glycoprotein sequences between strains. The outcome suggests a limitation of the current DISC vaccine design against heterologous infection.
Collapse
Affiliation(s)
| | | | - Alistair McGregor
- Department Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (K.Y.C.); (N.S.E.-H.)
| |
Collapse
|
40
|
Zhao X, Bai X, Xi Y. Intrauterine Infection and Mother-to-Child Transmission of Hepatitis B Virus: Route and Molecular Mechanism. Infect Drug Resist 2022; 15:1743-1751. [PMID: 35437345 PMCID: PMC9013253 DOI: 10.2147/idr.s359113] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/26/2022] [Indexed: 01/01/2023] Open
Affiliation(s)
- Xianlei Zhao
- Division of Human Reproduction and Developmental Genetics, the Women’s Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, People’s Republic of China
- Institute of Genetics and Department of Human Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310058, People’s Republic of China
| | - Xiaoxia Bai
- Division of Human Reproduction and Developmental Genetics, the Women’s Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, People’s Republic of China
| | - Yongmei Xi
- Division of Human Reproduction and Developmental Genetics, the Women’s Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, People’s Republic of China
- Institute of Genetics and Department of Human Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310058, People’s Republic of China
- Correspondence: Yongmei Xi, Email
| |
Collapse
|
41
|
Karageorgou I, Kossyvakis A, Jiménez J, Garcia I, Mentis AFA. Cytomegalovirus DNA detection in pregnant women with a high IgG avidity index: a valuable tool for diagnosing non-primary infections? J Matern Fetal Neonatal Med 2022; 35:9399-9405. [PMID: 35139746 DOI: 10.1080/14767058.2022.2038130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND/AIM Congenital human cytomegalovirus infection (cCMV) is the commonest congenital infection, and it can result in hearing loss and neurodevelopmental delay. Even if primary infections are more frequent and cause more severe congenital cCMV manifestations compared to NPIs, and despite partial protection from maternal immunity, the highest birth prevalence of cCMV is observed in seropositive women with non-primary CMV infection (NPI). Given that NPI contribute significantly to the overall burden of cCMV, their accurate diagnosis of NPI remains clinically important. Considering that the serological testing for CMV infection is not always reliable, we sought to determine whether detection of CMV DNA in pregnant women with a high IgG avidity index (AI) can help diagnose NPI. MATERIALS AND METHODS Human CMV serology screening (IgG, IgM, and IgG AI) was performed for confirmation of CMV infection in serum samples from mainly pregnant women with indications of CMV infection due to IgG+ and IgM+-positive samples in other laboratories. Pregnant women (or those with termination of pregnancy during the last period) with adequate IgG levels to perform IgG AI were included. Demographic data and mean gestation week at the time of screening were recorded. Serological testing was performed using CE-IVD commercial kits. CMV DNAemia detection by real time PCR (RT-PCR) was applied to confirm suspected CMV infection. RESULTS Nine-hundred and thirty-four pregnant women CMV IgG positive with adequate IgG titers for AI testing were included in the study. The percentage of women with a high AI was 71.8% (671/934); among them, nearly 2.4% (16/671) had presence of CMV DNA. Also, 12.4% of women (116/934) had intermediate IgG AI and 15.7% of women (147/934) had low IgG AI. The presence of CMV DNA was observed in 13.8% (16/116) and 39.5% (58/147) of the groups with intermediate and low IgG AI, respectively. A high CMV IgG AI was associated with a negative CMV PCR status (p-value <.00001). CONCLUSIONS CMV DNA was present in 2.4% of seropositive women with high IgG AI, indicating active NPI and thus, harboring the risk of cCMV sequelae to the fetus. Moreover, the incidence of NPI may have been underestimated due to single timepoint testing. In order to detect CMV NPI in a seropositive woman, regular and frequent serology testing as well as detection of CMV DNAemia are required which render the whole diagnostic process impractical and not cost-effective.
Collapse
Affiliation(s)
- Ioulia Karageorgou
- Diagnostic Services Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | | | - Juan Jiménez
- Department of Mathematical Sciences and Informatics and Health Research Institute (IdISBa), University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Irene Garcia
- Department of Mathematical Sciences and Informatics and Health Research Institute (IdISBa), University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Alexios-Fotios A Mentis
- Diagnostic Services Laboratory, Hellenic Pasteur Institute, Athens, Greece.,University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece.,UNESCO Chair on Adolescent Health Care, Center for Adolescent Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
42
|
Megli CJ, Coyne CB. Infections at the maternal-fetal interface: an overview of pathogenesis and defence. Nat Rev Microbiol 2022; 20:67-82. [PMID: 34433930 PMCID: PMC8386341 DOI: 10.1038/s41579-021-00610-y] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2021] [Indexed: 02/08/2023]
Abstract
Infections are a major threat to human reproductive health, and infections in pregnancy can cause prematurity or stillbirth, or can be vertically transmitted to the fetus leading to congenital infection and severe disease. The acronym 'TORCH' (Toxoplasma gondii, other, rubella virus, cytomegalovirus, herpes simplex virus) refers to pathogens directly associated with the development of congenital disease and includes diverse bacteria, viruses and parasites. The placenta restricts vertical transmission during pregnancy and has evolved robust mechanisms of microbial defence. However, microorganisms that cause congenital disease have likely evolved diverse mechanisms to bypass these defences. In this Review, we discuss how TORCH pathogens access the intra-amniotic space and overcome the placental defences that protect against microbial vertical transmission.
Collapse
Affiliation(s)
- Christina J Megli
- Division of Maternal-Fetal Medicine, Division of Reproductive Infectious Disease, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine and the Magee Womens Research Institute, Pittsburgh, PA, USA.
| | - Carolyn B Coyne
- Department of Molecular Genetics and Microbiology and the Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
43
|
Talavera-Barber M, Flint K, Graber B, Dhital R, Kaptsan I, Medoro AK, Sánchez PJ, Shimamura M. Antibody Titers Against Human Cytomegalovirus gM/gN and gB Among Pregnant Women and Their Infants. Front Pediatr 2022; 10:846254. [PMID: 35813379 PMCID: PMC9259787 DOI: 10.3389/fped.2022.846254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
Congenital CMV (cCMV) infection can affect infants born to mothers with preconceptional seroimmunity. To prevent cCMV due to nonprimary maternal infection, vaccines eliciting responses exceeding natural immunity may be required. Anti-gM/gN antibodies have neutralizing capacity in-vitro and in animal models, but anti-gM/gN antibodies have not been characterized among seroimmune pregnant women. Paired maternal and infant cord sera from 92 CMV seropositive mothers and their full-term or preterm infants were tested for anti-gM/gN antibody titers in comparison with anti-gB titers and neutralizing activity. Anti-gM/gN titers were significantly lower than anti-gB titers for all groups and did not correlate with serum neutralizing capacity. Further study is needed to determine if higher anti-gM/gN antibody titers might enhance serum neutralizing capacity among seropositive adults.
Collapse
Affiliation(s)
- Maria Talavera-Barber
- Avera McKennan Hospital and University Medical Center, Avera Research Institute, Sioux Falls, SD, United States
| | - Kaitlyn Flint
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Brianna Graber
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Ravi Dhital
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Irina Kaptsan
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Alexandra K Medoro
- Division of Neonatology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, United States.,Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Pablo J Sánchez
- Division of Neonatology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, United States.,Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, United States.,Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Masako Shimamura
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
44
|
Périllaud-Dubois C, Belhadi D, Laouénan C, Mandelbrot L, Picone O, Vauloup-Fellous C. Current practices of management of maternal and congenital Cytomegalovirus infection during pregnancy after a maternal primary infection occurring in first trimester of pregnancy: Systematic review. PLoS One 2021; 16:e0261011. [PMID: 34860861 PMCID: PMC8641894 DOI: 10.1371/journal.pone.0261011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/22/2021] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Congenital CMV infection is the first worldwide cause of congenital viral infection but systematic screening of pregnant women and newborns for CMV is still debated in many countries. OBJECTIVES This systematic review aims to provide the state of the art on current practices concerning management of maternal and congenital CMV infection during pregnancy, after maternal primary infection (PI) in first trimester of pregnancy. DATA SOURCES Electronically searches on databases and hand searches in grey literature. STUDY ELIGIBILITY CRITERIA AND PARTICIPANTS Primary outcome was listing biological, imaging, and therapeutic management interventions in two distinct populations: population 1 are pregnant women with PI, before or without amniocentesis; population 2 are pregnant women with congenitally infected fetuses (after positive amniocentesis). Secondary outcome was pregnancy outcome in population 2. RESULTS Out of 4,134 studies identified, a total of 31 studies were analyzed, with 3,325 pregnant women in population 1 and 1,021 pregnant women in population 2, from 7 countries (Belgium, France, Germany, Israel, Italy, Spain and USA). In population 1, ultrasound (US) examination frequency was 0.75/month, amniocentesis in 82% cases, maternal viremia in 14% and preventive treatment with hyperimmune globulins (HIG) or valaciclovir in respectively 14% and 4% women. In population 2, US examination frequency was 1.5/month, magnetic resonance imaging (MRI) in 44% cases at 32 weeks gestation (WG), fetal blood sampling (FBS) in 24% at 28 WG, and curative treatment with HIG or valaciclovir in respectively 9% and 8% patients. CONCLUSIONS This systematic review illustrates management of maternal and congenital CMV during pregnancy in published and non-published literature, in absence of international consensus. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42019124342.
Collapse
Affiliation(s)
- Claire Périllaud-Dubois
- Département de Virologie, AP-HP.Sorbonne Université, Hôpital Saint-Antoine, Paris, France
- Université de Paris, INSERM UMR1137, IAME, Paris, France
- GRIG, Groupe de Recherche sur les Infections pendant la Grossesse, Paris, France
- * E-mail:
| | - Drifa Belhadi
- Université de Paris, INSERM UMR1137, IAME, Paris, France
- Department of Epidemiology, Biostatistic and Clinical Research, AP-HP, Hôpital Bichat, Paris, France
| | - Cédric Laouénan
- Université de Paris, INSERM UMR1137, IAME, Paris, France
- Department of Epidemiology, Biostatistic and Clinical Research, AP-HP, Hôpital Bichat, Paris, France
| | - Laurent Mandelbrot
- Université de Paris, INSERM UMR1137, IAME, Paris, France
- GRIG, Groupe de Recherche sur les Infections pendant la Grossesse, Paris, France
- AP-HP, Hôpital Louis Mourier, Service de Gynécologie-Obstétrique, Colombes, France
| | - Olivier Picone
- Université de Paris, INSERM UMR1137, IAME, Paris, France
- GRIG, Groupe de Recherche sur les Infections pendant la Grossesse, Paris, France
- AP-HP, Hôpital Louis Mourier, Service de Gynécologie-Obstétrique, Colombes, France
| | - Christelle Vauloup-Fellous
- GRIG, Groupe de Recherche sur les Infections pendant la Grossesse, Paris, France
- Laboratoire de Virologie, AP-HP.Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Saclay, INSERM U1193, Villejuif, France
| |
Collapse
|
45
|
Hyde K, Sultana N, Tran AC, Bileckaja N, Donald CL, Kohl A, Stanton RJ, Strang BL. Limited replication of human cytomegalovirus in a trophoblast cell line. J Gen Virol 2021; 102. [PMID: 34816792 PMCID: PMC8742992 DOI: 10.1099/jgv.0.001683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Several viruses, including human cytomegalovirus (HCMV), are thought to replicate in the placenta. However, there is little understanding of the molecular mechanisms involved in HCMV replication in this tissue. We investigated replication of HCMV in the extravillous trophoblast cell line SGHPL-4, a commonly used model of HCMV replication in the placenta. We found limited HCMV protein expression and virus replication in SGHPL-4 cells. This was associated with a lack of trophoblast progenitor cell protein markers in SGHPL-4 cells, suggesting a relationship between trophoblast differentiation and limited HCMV replication. We proposed that limited HCMV replication in trophoblast cells is advantageous to vertical transmission of HCMV, as there is a greater opportunity for vertical transmission when the placenta is intact and functional. Furthermore, when we investigated the replication of other vertically transmitted viruses in SGHPL-4 cells we found some limitation to replication of Zika virus, but not herpes simplex virus. Thus, limited replication of some, but not all, vertically transmitted viruses may be a feature of trophoblast cells.
Collapse
Affiliation(s)
- Kadeem Hyde
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Nowshin Sultana
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Andy C Tran
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Narina Bileckaja
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Claire L Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Blair L Strang
- Institute for Infection and Immunity, St George's, University of London, London, UK
| |
Collapse
|
46
|
Devlieger R, Buxmann H, Nigro G, Enders M, Jückstock J, Siklós P, Wartenberg-Demand A, Schüttrumpf J, Schütze J, Rippel N, Herbold M, Niemann G, Friese K. Serial Monitoring and Hyperimmunoglobulin versus Standard of Care to Prevent Congenital Cytomegalovirus Infection: A Phase III Randomized Trial. Fetal Diagn Ther 2021; 48:611-623. [PMID: 34569538 DOI: 10.1159/000518508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/12/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Nonrandomized studies support the potential of cytomegalovirus hyperimmunoglobulin (CMV-HyperIg) in preventing maternofetal CMV transmission, but prospective interventional studies show equivocal results. We pre-sent a prospective phase-III international randomized open-label trial on the potential effect of CMV-HyperIg following serial monitoring of CMV serostatus. METHODS CMV-seronegative pregnant women (gestational age [GA] <14 weeks) were 1:1 randomized to monthly CMV-serostatus monitoring and CMV-HyperIg upon seroconversion (treatment), or routine prenatal care with CMV-serostatus testing at end of pregnancy (control). Ethical considerations required that control subjects with confirmed seroconversion be offered Cytotect®. The primary endpoint was the proportion of fetuses/newborns with congenital CMV infection. Secondary endpoints included neonatal CMV disease and safety during the 24-month follow-up. RESULTS The treatment arm counted 4,800 randomized subjects: 52 seroconverted (median GA 24 [11-35] weeks), of which 45 completed follow-up. The control arm counted 4,735 randomized subjects: 42 seroconverted, of which 34 completed follow-up (evaluable data for 28 newborns) and 8 subjects chose off-label Cytotect®. Congenital CMV rates were 13/28 newborns (46.4% [CI 27.51; 66.13]) vs. 16/45 newborns (35.6% [CI 21.87; 51.22]) in control and treated arms, respectively (p = 0.46). Newborn CMV disease was mostly mild and spontaneously resolving. There were no major safety concerns. The target sample was not reached within an acceptable time frame. CONCLUSIONS Serial monitoring of CMV serostatus with CMV-HyperIg treatment was associated with a mild nonsignificant reduction in the vertical CMV transmission rate. Studies on the optimal preventive strategy are hampered by epidemiological and ethical challenges and should focus on GA-dependent transmission rates and accurate dating of infection.
Collapse
Affiliation(s)
- Roland Devlieger
- Maternal and Fetal Medicine at University Hospital Leuven, Leuven, Belgium.,Department of Obstetrics at GZA campus Sint-Augustinus, Wilrijk, Belgium
| | - Horst Buxmann
- Department of Pediatric and Adolescent Medicine, Division for Neonatology University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | | | - Julia Jückstock
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Pal Siklós
- Szent István Kórház (St. Stephan Hospital), Budapest, Hungary
| | | | | | | | | | | | | | - Klaus Friese
- Director of Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
47
|
Zhou Q, Wang Q, Shen H, Zhang Y, Zhang S, Li X, Acharya G. Seroprevalence of Cytomegalovirus and Associated Factors Among Preconception Women: A Cross-Sectional Nationwide Study in China. Front Public Health 2021; 9:631411. [PMID: 34513776 PMCID: PMC8425481 DOI: 10.3389/fpubh.2021.631411] [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: 11/24/2020] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Cytomegalovirus seroconversion during pregnancy is common and has a substantial risk of congenital infection with longterm sequale. Screening during pregnancy or vaccination have not been shown to be effective for eliminating congenital infections. Preconception screening policy has not been evaluated adequately in a large scale. This nationwide study aimed to investigate epidemiological features of cytomegalovirus seropositivity and its geographic variation among Chinese women planning a pregnancy to gather epidemiological evidence as an essential for developing novel prevention strategies. Method: This cross-sectional sero-epidemiological survey enrolled women intending to become pregnant within 6 months in mainland China during 2010-2012. The primary outcomes in this study were cytomegalovirus Immunoglobulin G and M seropositivity. Secondary outcomes were the associations between Immunoglobulin G and Immunoglobulin M, with socio-demographic characteristics, including age, occupation, education level, place of residence, and ethnicity. The overall seropositivity and regional disparity was analyzed on the individual and regional level, respectively. Results: This study included data from 1,564,649 women from 31 provinces in mainland China. Among participants, 38.6% (n = 603,511) were cytomegalovirus immunoglobulin G+, 0.4% (n = 6,747) were immunoglobulin M+, and 0.2% (n = 2,879) were immunoglobulin M+ and immunoglobulin G+. On individual level, participant's age, ethnicity, and residing region were significantly associated with IgG+, IgM+, and IgM+IgG+ (P < 0.001), while occupation, education level, and place of residence were not statistically significant (P > 0.05). On regional level, cytomegalovirus immunoglobulin G and immunoglobulin M seropositivity was highest in the eastern region (49.5 and 0.5%, respectively), and lowest in the western region (26.9 and 0.4%, respectively). This geographic variation was also noted at the provincial level, characterized by higher provincial immunoglobulin M+ and immunoglobulin G+ rates associated with higher immunoglobulin G seropositivity. In the subgroup analysis of immunoglobulin G seropositivity, areas of higher immunoglobulin G positivity had a higher rate of immunoglobulin M+, indicating an expected increased risk of reinfection and primary infection. Conclusions: A substantial proportion of women (>60%) were susceptible to cytomegalovirus in preconception period in China, and immunoglobulin G seropositivity was seen at a low-medium level with substantial geographic variation. Integration of cytomegalovirus antibody testing in preconception screening program based on regional immunoglobulin G seropositivity, should be considered to promote strategies directed toward preventing sero-conversion during pregnancy to reduce the risk of this congenital infection.
Collapse
Affiliation(s)
- Qiongjie Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.,Women's Health and Perinatology Research Group, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Qiaomei Wang
- Department of Maternal and Child Health, National Health and Family Planning Commission of the People's Republic of China, Beijing, China
| | - Haiping Shen
- Department of Maternal and Child Health, National Health and Family Planning Commission of the People's Republic of China, Beijing, China
| | - Yiping Zhang
- Department of Maternal and Child Health, National Health and Family Planning Commission of the People's Republic of China, Beijing, China
| | - Shikun Zhang
- Department of Maternal and Child Health, National Health and Family Planning Commission of the People's Republic of China, Beijing, China
| | - Xiaotian Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Ganesh Acharya
- Women's Health and Perinatology Research Group, Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway.,Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Obstetrics and Gynecology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
48
|
Moulden J, Sung CYW, Brizic I, Jonjic S, Britt W. Murine Models of Central Nervous System Disease following Congenital Human Cytomegalovirus Infections. Pathogens 2021; 10:1062. [PMID: 34451526 PMCID: PMC8400215 DOI: 10.3390/pathogens10081062] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022] Open
Abstract
Human cytomegalovirus infection of the developing fetus is a leading cause of neurodevelopmental disorders in infants and children, leading to long-term neurological sequela in a significant number of infected children. Current understanding of the neuropathogenesis of this intrauterine infection is limited because of the complexity of this infection, which includes maternal immunological responses that are overlaid on virus replication in the CNS during neurodevelopment. Furthermore, available data from human cases are observational, and tissues from autopsy studies have been derived from only the most severe infections. Animal models of this human infection are also limited by the strict species specificity of cytomegaloviruses. However, informative models including non-human primates and small animal models have been developed. These include several different murine models of congenital HCMV infection for the study of CMV neuropathogenesis. Although individual murine models do not completely recapitulate all aspects of the human infection, each model has provided significant information that has extended current understanding of the neuropathogenesis of this human infection. This review will compare and contrast different murine models in the context of available information from human studies of CNS disease following congenital HCMV infections.
Collapse
Affiliation(s)
- Jerome Moulden
- Department of Microbiology, UAB School of Medicine, Birmingham, Al 35294, USA;
| | - Cathy Yea Won Sung
- Laboratory of Hearing Biology and Therapeutics, NIDCD, NIH, Bethesda, MD 20892, USA;
| | - Ilija Brizic
- Center for Proteomics and Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (I.B.); (S.J.)
| | - Stipan Jonjic
- Center for Proteomics and Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (I.B.); (S.J.)
| | - William Britt
- Department of Microbiology, UAB School of Medicine, Birmingham, Al 35294, USA;
- Department of Pediatrics and Neurobiology, UAB School of Medicine, Birmingham, Al 35294, USA
| |
Collapse
|
49
|
Congenital Human Cytomegalovirus Infection: A Narrative Review of Maternal Immune Response and Diagnosis in View of the Development of a Vaccine and Prevention of Primary and Non-Primary Infections in Pregnancy. Microorganisms 2021; 9:microorganisms9081749. [PMID: 34442828 PMCID: PMC8398868 DOI: 10.3390/microorganisms9081749] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 12/19/2022] Open
Abstract
Congenital cytomegalovirus infection (cCMV) may affect about 1% of all newborns all over the world as a result of either a primary or recurrent human cytomegalovirus (HCMV) infection. While about 90% of infants affected by cCMV are asymptomatic at birth, the remaining 10% are symptomatic often with neurodevelopmental impairment and sensorineural hearing loss. In view of identifying the best approach to vaccine prevention of cCMV, this review will examine the most important steps made in the study of the immune response to, and diagnosis of, HCMV infection. The maternal immune response and immune correlates of protection are being partially identified with a partial contribution given by our laboratory. The diagnosis of primary infection is often difficult to achieve in the first three months of pregnancy, which is the time primarily involved in virus transmission to the fetus in association with the most severe symptoms and sequelae. Prevention of cCMV is anticipated by prevention of primary infection in early pregnancy by means of different measures, such as (i) behavioral-educational measures, (ii) immunoglobulin administration, (iii) antiviral treatment with valaciclovir. However, the most promising approach to cCMV prevention appears to be the development of a non-living vaccine, including at least three viral antigens: gB, pentamer complex gHgLpUL128L, and pp65, which have been shown to be able to stimulate both the humoral and the cellular arms of the maternal immune response. Primary HCMV infection may be managed in pregnancy by counseling of the couples involved by a team of specialists that includes virologists, obstetricians, infectivologists and neonatologists.
Collapse
|
50
|
New evidence on prognostic features, prevention and treatment of congenital Cytomegalovirus infection. Curr Opin Obstet Gynecol 2021; 32:342-350. [PMID: 32739974 DOI: 10.1097/gco.0000000000000651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Congenital Cytomegalovirus (CMV) infection remains a major cause of lifelong disability, with no systematic screening implemented in pregnancy or the postnatal period. In this review article, we outline the preventive strategies, antenatal prognostic features and experimental therapies as well as evidence of efficacy from recent trials. RECENT FINDINGS A recent randomized, double blinded, placebo-controlled study investigated the efficacy of Valaciclovir in women contracting primary CMV in the periconception period or first trimester. They concluded that Valaciclovir at a dose of 8 g/day is effective in reducing the rate of foetal CMV infection following early maternal primary infection. Administration of CMV hyperimmune globulin (HIG) was investigated in a recent randomized double-masked controlled trial. This study concluded that CMV HIG was ineffective at reducing the risk of congenital CMV among women with primary CMV in early pregnancy. SUMMARY Congenital CMV infection remains a significant cause of disability. There is currently no vaccine available, with the best preventive strategy being patient education on transmission as well as hygiene measures to reduce risk of exposure. Experimental therapies have been investigated in recent years and there is evidence supporting the use of Valaciclovir. Data for the efficacy of CMV HIG remains inconsistent and administration is currently limited to clinical trial settings.
Collapse
|