1
|
Oliveira ADS, Pereira JG, Nunes GT, Sousa Junior IPD, Sarmento DJDS, Lopes JIF, Amorim Filho L, Paula VSD. Prevalence and investigation of Cytomegalovirus (HCMV) in blood donors from the main blood establishment in Rio de Janeiro/Brazil. Braz J Infect Dis 2025; 29:104508. [PMID: 39922051 PMCID: PMC11848753 DOI: 10.1016/j.bjid.2025.104508] [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: 06/27/2024] [Revised: 12/17/2024] [Accepted: 01/17/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Human Cytomegalovirus (HCMV) remains a significant cause of morbidity and mortality among pregnant women and immunocompromised patients. HCMV transmission can occur through blood transfusions and typically results in asymptomatic infections in newborns and young individuals or causes symptoms like infectious mononucleosis when symptomatic infections arise. HCMV infection poses a notable risk to transfusion recipients, particularly in vulnerable groups such as premature newborns and immunosuppressed patients. The risk persists even after prophylaxis ends, especially in patients who undergo organ transplantation and receive blood or blood products from a seropositive donor while being seronegative themselves (D+/R-). MATERIALS AND METHODS Here, we investigated the serological and molecular prevalence of HCMV among 980 blood donors from the main blood bank in Rio de Janeiro, Brazil, using chemiluminescence and real-time PCR (TaqMan). The data underwent univariate, bivariate, and multivariate statistical analyses using the SPSS program, version 20.0. RESULTS The average age of donors was 38.53 years, with a majority being male (53.9 %). The prevalence of cytomegalovirus was 88.5 %, and HCMV DNA was detected in 1.2 % of the samples. DISCUSSION Given that there are approximately 100,000 blood donations per year, this prevalence rate is considerably high compared to that in developed countries. These findings underscore the critical need for ongoing surveillance and molecular testing to ensure the safety of blood supplies.
Collapse
Affiliation(s)
- Agildo da Silva Oliveira
- Instituto Estadual de Hematologia Arthur de Siqueira Cavalcanti/HEMORIO, Rio de Janeiro, RJ, Brazil; Instituto Oswaldo Cruz (Fiocruz), Laboratório de Virologia Molecular e Parasitologia, Rio de Janeiro, RJ, Brazil
| | - Jéssica Gonçalves Pereira
- Instituto Oswaldo Cruz (Fiocruz), Laboratório de Virologia Molecular e Parasitologia, Rio de Janeiro, RJ, Brazil
| | - Gabrielle Tantos Nunes
- Instituto Estadual de Hematologia Arthur de Siqueira Cavalcanti/HEMORIO, Rio de Janeiro, RJ, Brazil; Instituto Oswaldo Cruz (Fiocruz), Laboratório de Virologia Molecular e Parasitologia, Rio de Janeiro, RJ, Brazil
| | | | - Dmitry José de Santana Sarmento
- Universidade Estadual da Paraíba, Faculdade de Odontologia, Departamento de Diagnóstico Oral, Araruna, PB, Brazil; Centro Universitário Facisa (UNIFACISA), Departamento de Medicina, Campina Grande, PB Brazil
| | | | - Luiz Amorim Filho
- Instituto Estadual de Hematologia Arthur de Siqueira Cavalcanti/HEMORIO, Rio de Janeiro, RJ, Brazil
| | - Vanessa Salete de Paula
- Instituto Oswaldo Cruz (Fiocruz), Laboratório de Virologia Molecular e Parasitologia, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
2
|
Katungye RV, Musooko M, Sekikubo M, Mutabazi T, Kyohere M, Tusubira V, Sendagala JN, Peacock J, Le Doare K, Nakimuli A. Seroprevalence of Cytomegalovirus Among Pregnant Women at Kawempe National Referral Hospital, Uganda: A Cross-sectional Study. Open Forum Infect Dis 2024; 11:S200-S205. [PMID: 40070698 PMCID: PMC11891130 DOI: 10.1093/ofid/ofae604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Background Maternal primary cytomegalovirus (CMV) infection is associated with abortion and congenital anomalies. In Uganda, the burden of maternal CMV infection is not well studied. This study thus assessed the seroprevalence and factors associated with CMV infection among pregnant women at Kawempe National Referral Hospital in Kampala. This work forms a part of the PROGRESS study, an observational cohort study undertaken in Kampala, Uganda, between November 2018 and April 2021. Methods We conducted a cross-sectional study between September 2020 and January 2021 among the 639 pregnant women admitted to the labor ward at a government hospital. Sociodemographic, medical, obstetric, and socioeconomic data were collected. Blood samples from study participants were drawn and analyzed for the presence of CMV immunoglobulin G (IgG) and IgM using enzyme-linked immunosorbent assay-based quantitative assays. Further analysis of all IgM-positive samples was conducted using CMV IgG avidity assays. All infants had a nasal polymerase chain reaction (PCR) on the first day of life to investigate CMV positivity. Logistic regression was performed to determine the factors associated with CMV infection. Results Seroprevalence of CMV IgG among the 637 women was universal (100%), and that of CMV (IgM) was 5.8% (37/637). CMV (IgM) was associated with being low socioeconomic status (odds ratio, 3.44; 95% CI, 1.05-11.32; P = .04). Transmission risk was low, and no infant had a positive PCR for CMV at birth. Conclusions Universally, by the time women in Kampala conceive, they will have been exposed to CMV. Women of lower socioeconomic status were more likely to have recent CMV infection than their more affluent counterparts, highlighting the need for screening guidelines in this setting.
Collapse
Affiliation(s)
- Richard V Katungye
- Department of Obstetrics and Gynaecology, Makerere University College of Health Sciences, Kampala, Uganda
- Directorate of Health Services, Special Forces Command, Uganda People's Defense Forces, Entebbe, Uganda
| | - Moses Musooko
- Department of Obstetrics and Gynaecology, Makerere University College of Health Sciences, Kampala, Uganda
| | - Musa Sekikubo
- Department of Obstetrics and Gynaecology, Makerere University College of Health Sciences, Kampala, Uganda
| | - Tobius Mutabazi
- Directorate of Health Services, Special Forces Command, Uganda People's Defense Forces, Entebbe, Uganda
- School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Mary Kyohere
- Makerere University—Johns Hopkins University Research Collaboration, Kampala, Uganda
- Institute for Infection and Immunity, St. George's, University of London, London, UK
| | - Valerie Tusubira
- Makerere University—Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - Juliet Nsimire Sendagala
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Joseph Peacock
- Institute for Infection and Immunity, St. George's, University of London, London, UK
| | - Kirsty Le Doare
- Makerere University—Johns Hopkins University Research Collaboration, Kampala, Uganda
- Institute for Infection and Immunity, St. George's, University of London, London, UK
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Annettee Nakimuli
- Department of Obstetrics and Gynaecology, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
3
|
Balegamire SJ, Mâsse B, Audibert F, Lamarre V, Giguere Y, Forest JC, Boucoiran I. Association Between Maternal Cytomegalovirus Seropositivity, Preterm Birth, and Preeclampsia in Two Cohorts From Quebec, Canada: A Mediation Analysis. Am J Reprod Immunol 2024; 92:e13941. [PMID: 39436114 DOI: 10.1111/aji.13941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
PROBLEM Preterm birth and preeclampsia significantly contribute to infant morbidity and mortality, posing critical public health concerns. Viral infections, particularly Cytomegalovirus (CMV), associated with chronic inflammation, may play a role in these adverse pregnancy outcomes. The contribution of CMV to preterm birth and preeclampsia requires further investigation. METHOD OF STUDY Data from 6048 pregnant women from two prospective Quebec cohorts, recruited between May 2005 and August 2012, were analyzed. First-trimester CMV serology was the exposure variable. Associations were assessed using multivariable logistic regression adjusted by inverse probability treatment weighting (IPTW) of propensity scores. Mediation analyses estimated the direct effect of maternal CMV serostatus on preterm birth, excluding mediation by preeclampsia. RESULTS Preterm birth and preeclampsia proportions were 5.1% (95% CI: 4.6-5.7) and 1.9% (95% CI: 1.6-2.3), respectively. Multivariable logistic regression adjusted by IPTW showed associations between CMV seropositivity and preterm birth (OR 1.20, 95% CI: 1.02-1.41) and CMV seropositivity and preeclampsia (OR 1.41, 95% CI: 1.08-1.84). Mediation analysis indicated that 97% of the total effect of CMV seropositivity on preterm birth is direct, with the remaining 3% mediated by preeclampsia. CONCLUSIONS CMV seropositivity appears to be a risk factor for both preterm birth and preeclampsia. The effect of maternal CMV seropositivity on preterm birth is primarily direct, not mediated by preeclampsia. Future studies should explore the impact of preventive measures against CMV infection on the incidence of preterm delivery and preeclampsia.
Collapse
Affiliation(s)
- Safari Joseph Balegamire
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
| | - Benoît Mâsse
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Applied Clinical Research Unit, CHU Sainte Justine Research Center, Montreal, Canada
| | - François Audibert
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Valerie Lamarre
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Yves Giguere
- CHU de Québec-Université Laval Research Center, Quebec City, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Jean-Claude Forest
- CHU de Québec-Université Laval Research Center, Quebec City, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Isabelle Boucoiran
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Leruez-Ville M, Chatzakis C, Lilleri D, Blazquez-Gamero D, Alarcon A, Bourgon N, Foulon I, Fourgeaud J, Gonce A, Jones CE, Klapper P, Krom A, Lazzarotto T, Lyall H, Paixao P, Papaevangelou V, Puchhammer E, Sourvinos G, Vallely P, Ville Y, Vossen A. Consensus recommendation for prenatal, neonatal and postnatal management of congenital cytomegalovirus infection from the European congenital infection initiative (ECCI). THE LANCET REGIONAL HEALTH. EUROPE 2024; 40:100892. [PMID: 38590940 PMCID: PMC10999471 DOI: 10.1016/j.lanepe.2024.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/10/2024]
Abstract
Congenital cytomegalovirus (cCMV) infection carries a significant burden with a 0.64% global prevalence and a 17-20% chance of serious long-term effects in children. Since the last guidelines, our understanding, particularly regarding primary maternal infections, has improved. A cCMV guidelines group was convened under the patronage of the European Society of Clinical Virology in April 2023 to refine these insights. The quality and validity of selected studies were assessed for potential biases and the GRADE framework was employed to evaluate quality of evidence across key domains. The resulting recommendations address managing cCMV, spanning prevention to postnatal care. Emphasizing early and accurate maternal diagnosis through serological tests enhances risk management and prevention strategies, including using valaciclovir to prevent vertical transmission. The guidelines also strive to refine personalized postnatal care based on risk assessments, ensuring targeted interventions for affected families.
Collapse
Affiliation(s)
- Marianne Leruez-Ville
- Université Paris Cité, URP 7328 FETUS, F-75015, Paris, France
- Virology Laboratory, Reference Laboratory for Cytomegalovirus Infections, Hôpital Necker Enfants Malades, GHU Paris Centre, AP-HP, Paris, France
| | - Christos Chatzakis
- Obstetrics, Fetal Medicine Surgery and Imaging Unit, Hôpital Necker Enfants Malades, GHU Paris Centre, AP-HP, Paris, France
- Second Department of Obstetrics and Gynecology of Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Daniele Lilleri
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Daniel Blazquez-Gamero
- Pediatric Infectious Diseases Unit, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (imas12), Universidad Complutense, Madrid, Spain
| | - Ana Alarcon
- Department of Neonatology, Hospital Hospital Sant Joan de Déu, BCNatal (Barcelona Center for Maternal, Fetal and Neonatal Medicine), Institut de Recerca Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Nicolas Bourgon
- Obstetrics, Fetal Medicine Surgery and Imaging Unit, Hôpital Necker Enfants Malades, GHU Paris Centre, AP-HP, Paris, France
| | - Ina Foulon
- Department of Otorhinolaryngology and Head & Neck Surgery, Vrije Universiteit Brussels, University Hospital UZ Brussel, Brussels Health Campus. De Poolster, Rehabilitation Centre, Brussels, Belgium
| | - Jacques Fourgeaud
- Université Paris Cité, URP 7328 FETUS, F-75015, Paris, France
- Virology Laboratory, Reference Laboratory for Cytomegalovirus Infections, Hôpital Necker Enfants Malades, GHU Paris Centre, AP-HP, Paris, France
| | - Anna Gonce
- BCNatal: Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain
| | - Christine E. Jones
- Faculty of Medicine and Institute for Life Sciences, University of Southampton and NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, United Kingdom
| | - Paul Klapper
- Microbiology and Virology Unit (EIGen), School of Biological Sciences, University of Manchester, Manchester, M139PT, UK
| | - André Krom
- Department of Medical Ethics and Health Law, Leiden University Medical Center, Leiden, the Netherlands
| | - Tiziana Lazzarotto
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | | - Paulo Paixao
- CHRC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056, Lisbon, Portugal
| | - Vassiliki Papaevangelou
- Third Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Greece
| | | | - George Sourvinos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, 71003, Greece
| | - Pamela Vallely
- Microbiology and Virology Unit (EIGen), School of Biological Sciences, University of Manchester, Manchester, M139PT, UK
| | - Yves Ville
- Université Paris Cité, URP 7328 FETUS, F-75015, Paris, France
- Obstetrics, Fetal Medicine Surgery and Imaging Unit, Hôpital Necker Enfants Malades, GHU Paris Centre, AP-HP, Paris, France
| | - Ann Vossen
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
5
|
Mocanu AG, Stoian DL, Daescu AMC, Motofelea AC, Ciohat IM, Navolan DB, Vilibic-Cavlek T, Bogdanic M, Nemescu D, Tomescu L, Carabineanu A. The Impact of Latent Cytomegalovirus Infection on Spontaneous Abortion History and Pregnancy Outcomes in Romanian Pregnant Women. Microorganisms 2024; 12:731. [PMID: 38674675 PMCID: PMC11052013 DOI: 10.3390/microorganisms12040731] [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: 02/29/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Cytomegalovirus (CMV), a DNA virus that belongs to the Orthoherpesviridae family, infects 40-100% of people. Primary/non-primary CMV infection during pregnancy could cause fetal disabilities. After primary infection, CMV causes a latent infection and resides in cells of the myeloid compartment (CD34+, monocytes). Few studies have analyzed the impact of latent CMV infections on miscarriage history, pregnancy complications, and neonatal outcomes. METHODS Serum samples from 806 pregnant women (28.29 ± 4.50 years old) who came for a consultation at the Timisoara Clinical Emergency City Hospital between 2008 and 2010 were tested for anti-CMV IgM/IgG antibodies, and data about demography, obstetrical history, pregnancy complications, birth, and neonate were collected. The data were compared between the groups with and without latent CMV infection, and statistical significance was calculated. RESULTS We did not find a difference regarding cesarean section (OR = 0.916, p = 0.856), placental abruption (OR = 1.004, p = 1.00), pregnancy-induced hypertension rate (OR = 1.078, p = 1.00), secondary sex ratio (0.882, p = 0.857), APGAR score (p = 0.225), gestational age at birth (p = 0.434), or birth weight (p = 0.365). A borderline significant difference was found regarding the presence of miscarriage history: OR = 8.467, p = 0.051. CONCLUSIONS The presence of latent CMV infection does not affect the likelihood of complications in healthy women. A borderline significantly higher prevalence of miscarriage history was found in women with latent CMV infection.
Collapse
Affiliation(s)
- Adelina Geanina Mocanu
- Department of Obstetrics-Gynecology, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania; (A.G.M.); (L.T.)
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania
| | - Dana Liana Stoian
- Department of Endocrinology, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania;
| | - Ana-Maria Cristina Daescu
- Department of Psychiatry, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania;
| | - Alexandru Catalin Motofelea
- Department of Internal Medicine, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania;
| | - Ioana Mihaela Ciohat
- Laboratory of Antenatal Medicine, Timisoara City Emergency Hospital, 300202 Timisoara, Romania;
| | - Dan Bogdan Navolan
- Department of Obstetrics-Gynecology, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania; (A.G.M.); (L.T.)
| | - Tatjana Vilibic-Cavlek
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia;
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Maja Bogdanic
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia;
| | - Dragos Nemescu
- Department of Obstetrics-Gynecology, Gr. T. Popa University of Medicine and Pharmacy Iasi, Universitatiistr. nr. 16, 700115 Iasi, Romania;
| | - Larisa Tomescu
- Department of Obstetrics-Gynecology, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania; (A.G.M.); (L.T.)
| | - Adrian Carabineanu
- Department of Surgery, Victor Babes University of Medicine and Pharmacy Timisoara, P-ta Eftimie Murgu nr. 2, 300041 Timisoara, Romania;
| |
Collapse
|
6
|
Das R, Blázquez-Gamero D, Bernstein DI, Gantt S, Bautista O, Beck K, Conlon A, Rosenbloom DIS, Wang D, Ritter M, Arnold B, Annunziato P, Russell KL. Safety, efficacy, and immunogenicity of a replication-defective human cytomegalovirus vaccine, V160, in cytomegalovirus-seronegative women: a double-blind, randomised, placebo-controlled, phase 2b trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:1383-1394. [PMID: 37660711 DOI: 10.1016/s1473-3099(23)00343-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND A vaccine that prevents cytomegalovirus (CMV) infection in women could reduce the incidence of congenital CMV infection, a major cause of neurodevelopmental disability. We aimed to assess the safety and efficacy of a replication-defective investigational CMV vaccine, V160, in CMV-seronegative women. METHODS This phase 2b, randomised, double-blind, placebo-controlled study was conducted at 90 sites in seven countries (USA, Finland, Canada, Israel, Spain, Russia, and Australia). Eligible participants were generally healthy, CMV-seronegative, non-pregnant, 16-35-year-old women of childbearing potential with exposure to children aged 5 years or younger. Participants were randomly assigned using central randomisation via an interactive response technology system 1:1:1 to one of three groups: V160 three-dose regimen (V160 at day 1, month 2, and month 6), V160 two-dose regimen (V160 on day 1, placebo at month 2, and V160 at month 6), or placebo (saline solution at day 1, month 2, and month 6). The primary outcomes were the efficacy of three doses of V160 in reducing the incidence of primary CMV infection during the follow-up period starting 30 days after the last dose of vaccine using a fixed event rate design, and the safety and tolerability of the two-dose and three-dose V160 regimens. We planned to test the efficacy of a two-dose regimen of V160 in reducing the incidence of primary CMV infection only if the primary efficacy hypothesis was met. Analyses for the primary efficacy endpoint were performed on the per-protocol efficacy population; safety analyses included all randomly assigned participants who received study vaccine. The primary efficacy hypothesis was tested at prespecified interim and final analyses. The study was ongoing and efficacy data continued to accrue at the time of final testing of the primary efficacy hypothesis. Vaccine efficacy was re-estimated after final testing of the primary efficacy hypothesis based on all available efficacy data at end of study. This trial is registered at ClinicalTrials.gov (NCT03486834) and EudraCT (2017-004233-86) and is complete. FINDINGS Between April 30, 2018, and Aug 30, 2019, 7458 participants were screened, of whom 2220 were randomly assigned to the V160 three-dose group (n=733), V160 two-dose group (n=733), or placebo group (n=734). A total of 523 participants in the V160 three-dose group and 519 in the placebo group were included in the final hypothesis testing. Of these, there were 11 cases of CMV infection in the V160 three-dose group and 20 cases in the placebo group. The vaccine efficacy for the V160 three-dose group was 44·6% (95% CI -15·2 to 74·8) at the final testing of the primary efficacy hypothesis, a result corresponding to failure to demonstrate the primary efficacy hypothesis. On the basis of this result, the study was terminated for futility. The re-estimate of vaccine efficacy for the V160 three-dose group based on all available efficacy data at end of study (556 participants in the V160 three-dose group and 543 in the placebo group) was 42·4% (95% CI -13·5 to 71·1). A total of 728 participants in the V160 three-dose group, 729 in the V160 two-dose group, and 732 in the placebo group were included in the safety analyses. The most common solicited injection-site adverse event was injection-site pain (680 [93%] in the V160 three-dose group, 659 [90%] in the V160 two-dose group, and 232 [32%] in the placebo group). The most common solicited systemic adverse event was fatigue (457 [63%] in the V160 three-dose group, 461 [63%] in the V160 two-dose group, and 357 [49%] in the placebo group). No vaccine-related serious adverse events or deaths were reported. INTERPRETATION V160 was generally well tolerated and immunogenic; however, three doses of the vaccine did not reduce the incidence of primary CMV infection in CMV-seronegative women compared with placebo. This study provides insights into the design of future CMV vaccine efficacy trials, particularly for the identification of CMV infection using molecular assays. FUNDING Merck Sharp & Dohme, a subsidiary of Merck & Co, Rahway, NJ, USA (MSD).
Collapse
Affiliation(s)
| | - Daniel Blázquez-Gamero
- Instituto de Investigación Hospital 12 de Octubre (imas12), Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - David I Bernstein
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Soren Gantt
- CHU Sainte-Justine Research Centre, Université de Montréal, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Tan NK, Pope CF, Carrington D. Performance evaluation of fully automated cobas® 6800 CMV PCR for the detection and quantification of cytomegalovirus DNA in neonatal urine and saliva, and adult urine, saliva, and vaginal secretion. J Med Virol 2023; 95:e29223. [PMID: 37966419 DOI: 10.1002/jmv.29223] [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: 06/23/2023] [Revised: 10/22/2023] [Accepted: 10/28/2023] [Indexed: 11/16/2023]
Abstract
Laboratory testing for cytomegalovirus (CMV) in bodily fluids is essential to manage congenital and prenatal CMV infection. The rapid and fully automated cobas® CMV PCR is approved only for the testing of plasma in transplant patients. To evaluate the performance of the cobas® CMV to detect and quantify CMV DNA in neonatal and adult female urine, saliva, and vaginal secretion, the limit of detection (LoD), limit of quantification (LoQ), imprecision, linearity, PCR efficiency, bias, analytical specificity, cross-reactivity, and cross-contamination of the cobas® CMV for urine, saliva, and vaginal secretion was determined. The performance of the assay was evaluated prospectively with two laboratory-developed PCR assays using neonatal and adult urine, saliva swabs, and vaginal swabs. The LoD and LoQ were 31 and 100 IU/mL, respectively, for urine, and 81 and 100 IU/mL, respectively, for vaginal secretion. The LoD and LoQ for saliva were the same (200 IU/mL). The cobas® CMV was precise (coefficient of variation ≤10%), linear (R2 ≥ 0.995), and efficient (1.07 and 1.09) between 100 and 250,000 IU/mL for the sample types. The bias and analytical specificity was <±0.30 log10 IU/mL and 100%, respectively. Cross-reactivity with non-CMV pathogens was not detected. Cross-contamination rate was 0.28%. The diagnostic accuracy, sensitivity, and specificity of the cobas® CMV for neonatal urine and saliva were ≥95.0%, ≥93.3%, and ≥90.4%, respectively. The overall percent agreement for adult urine, saliva, and vaginal secretion was 86.6%, 94.5%, and 89.4%, respectively. Taken together, the cobas® CMV demonstrated acceptable analytical and diagnostic performance, and is suitable for routine diagnostic laboratory investigation of CMV infection in neonates and adults.
Collapse
Affiliation(s)
- Ngee Keong Tan
- Department of Medical Microbiology, Infection and Immunity, South West London Pathology, St George's University Hospitals NHS Foundation Trust, London, UK
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cassie F Pope
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK
- Institute for Infection and Immunity, St George's, University of London, London, UK
| | - David Carrington
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
8
|
Barry PA, Iyer SS, Gibson L. Re-Evaluating Human Cytomegalovirus Vaccine Design: Prediction of T Cell Epitopes. Vaccines (Basel) 2023; 11:1629. [PMID: 38005961 PMCID: PMC10674879 DOI: 10.3390/vaccines11111629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/26/2023] Open
Abstract
HCMV vaccine development has traditionally focused on viral antigens identified as key targets of neutralizing antibody (NAb) and/or T cell responses in healthy adults with chronic HCMV infection, such as glycoprotein B (gB), the glycoprotein H-anchored pentamer complex (PC), and the unique long 83 (UL83)-encoded phosphoprotein 65 (pp65). However, the protracted absence of a licensed HCMV vaccine that reduces the risk of infection in pregnancy regardless of serostatus warrants a systematic reassessment of assumptions informing vaccine design. To illustrate this imperative, we considered the hypothesis that HCMV proteins infrequently detected as targets of T cell responses may contain important vaccine antigens. Using an extant dataset from a T cell profiling study, we tested whether HCMV proteins recognized by only a small minority of participants encompass any T cell epitopes. Our analyses demonstrate a prominent skewing of T cell responses away from most viral proteins-although they contain robust predicted CD8 T cell epitopes-in favor of a more restricted set of proteins. Our findings raise the possibility that HCMV may benefit from evading the T cell recognition of certain key proteins and that, contrary to current vaccine design approaches, including them as vaccine antigens could effectively take advantage of this vulnerability.
Collapse
Affiliation(s)
- Peter A. Barry
- Department of Pathology and Laboratory Medicine, Center for Immunology and Infectious Diseases, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
- California National Primate Research Center, University of California, Davis, CA 95616, USA
| | - Smita S. Iyer
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Laura Gibson
- Departments of Medicine and of Pediatrics, Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
9
|
Moström MJ, Yu S, Tran D, Saccoccio FM, Versoza CJ, Malouli D, Mirza A, Valencia S, Gilbert M, Blair RV, Hansen S, Barry P, Früh K, Jensen JD, Pfeifer SP, Kowalik TF, Permar SR, Kaur A. Protective effect of pre-existing natural immunity in a nonhuman primate reinfection model of congenital cytomegalovirus infection. PLoS Pathog 2023; 19:e1011646. [PMID: 37796819 PMCID: PMC10553354 DOI: 10.1371/journal.ppat.1011646] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
Congenital cytomegalovirus (cCMV) is the leading infectious cause of neurologic defects in newborns with particularly severe sequelae in the setting of primary CMV infection in the first trimester of pregnancy. The majority of cCMV cases worldwide occur after non-primary infection in CMV-seropositive women; yet the extent to which pre-existing natural CMV-specific immunity protects against CMV reinfection or reactivation during pregnancy remains ill-defined. We previously reported on a novel nonhuman primate model of cCMV in rhesus macaques where 100% placental transmission and 83% fetal loss were seen in CD4+ T lymphocyte-depleted rhesus CMV (RhCMV)-seronegative dams after primary RhCMV infection. To investigate the protective effect of preconception maternal immunity, we performed reinfection studies in CD4+ T lymphocyte-depleted RhCMV-seropositive dams inoculated in late first / early second trimester gestation with RhCMV strains 180.92 (n = 2), or RhCMV UCD52 and FL-RhCMVΔRh13.1/SIVgag, a wild-type-like RhCMV clone with SIVgag inserted as an immunological marker, administered separately (n = 3). An early transient increase in circulating monocytes followed by boosting of the pre-existing RhCMV-specific CD8+ T lymphocyte and antibody response was observed in the reinfected dams but not in control CD4+ T lymphocyte-depleted dams. Emergence of SIV Gag-specific CD8+ T lymphocyte responses in macaques inoculated with the FL-RhCMVΔRh13.1/SIVgag virus confirmed reinfection. Placental transmission was detected in only one of five reinfected dams and there were no adverse fetal sequelae. Viral whole genome, short-read, deep sequencing analysis confirmed transmission of both reinfection RhCMV strains across the placenta with ~30% corresponding to FL-RhCMVΔRh13.1/SIVgag and ~70% to RhCMV UCD52, consistent with the mixed human CMV infections reported in infants with cCMV. Our data showing reduced placental transmission and absence of fetal loss after non-primary as opposed to primary infection in CD4+ T lymphocyte-depleted dams indicates that preconception maternal CMV-specific CD8+ T lymphocyte and/or humoral immunity can protect against cCMV infection.
Collapse
Affiliation(s)
- Matilda J. Moström
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Shan Yu
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Dollnovan Tran
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Frances M. Saccoccio
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
| | - Cyril J. Versoza
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Daniel Malouli
- Oregon Health and Sciences University, Beaverton, Oregon, United States of America
| | - Anne Mirza
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sarah Valencia
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
| | - Margaret Gilbert
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Robert V. Blair
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Scott Hansen
- Oregon Health and Sciences University, Beaverton, Oregon, United States of America
| | - Peter Barry
- University of California, Davis, California, United States of America
| | - Klaus Früh
- Oregon Health and Sciences University, Beaverton, Oregon, United States of America
| | - Jeffrey D. Jensen
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Susanne P. Pfeifer
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Timothy F. Kowalik
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
- Weill Cornell Medicine, New York, New York State, United States of America
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| |
Collapse
|
10
|
Tan NK, Carrington D, Pope CF. Detecting human cytomegalovirus in urine, vagina and saliva: Impact of biological fluids and storage durations and temperatures on CMV DNA recovery. J Med Virol 2023; 95:e29081. [PMID: 37675875 DOI: 10.1002/jmv.29081] [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: 06/23/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
Sample collection, transport and storage conditions vary in the human cytomegalovirus (CMV) shedding literature. Currently, limited data exist on the impact of biological fluids and pre-analytical sample handling on the detection of CMV DNA. To evaluate CMV DNA recovery from urine, vaginal fluid and saliva stored in different conditions, adult urine, vaginal and saliva fluids and swabs, stored with or without selected nucleic acid preservation media at various durations and temperatures, was compared by polymerase chain reaction (PCR) quantitation of spiked samples and self-collected urine (n = 45) and vaginal swabs (n = 58) from CMV seropositive pregnant women. There was a time-dependent reduction in CMV DNA recovery from urine, urine diluted in phosphate-buffered saline, and saliva stored at 2-8°C, but not from urine preserved in cobas® PCR transport media (CPM) (urine/CPM). For vaginal fluid, a reduction in recovery was evident after 7 days storage at 2-8°C. CMV DNA recovery over 91 days was similar between -80°C and -20°C storage for urine and vaginal swabs preserved in CPM, and saliva swabs preserved in eNAT® PCR transport media. A statistically significant change in CMV DNA recovery after 25 months storage (median) at -80°C was not observed for self-collected urine/CPM and vaginal swab/CPM from pregnant women. Taken together, recovery of CMV DNA is dependent on fluid type and storage conditions. To improve the validity and reliability of detection at different storage durations and temperatures, the use of nucleic acid preserving transport media at the point of collection for urine, vaginal fluid and saliva may be essential.
Collapse
Affiliation(s)
- Ngee Keong Tan
- Department of Medical Microbiology, Infection and Immunity, South West London Pathology, St George's University Hospitals NHS Foundation Trust, London, UK
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David Carrington
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Cassie F Pope
- Infection Care Group, St George's University Hospitals NHS Foundation Trust, London, UK
- Institute for Infection and Immunity, St George's, University of London, London, UK
| |
Collapse
|
11
|
d'Angelo P, Zelini P, Zavaglio F, Piccini S, Cirasola D, Arossa A, Spinillo A, Lilleri D, Baldanti F. Correlates of postnatal human cytomegalovirus transmission in term babies in the first year. J Med Virol 2023; 95:e29105. [PMID: 37728300 DOI: 10.1002/jmv.29105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
Postnatal human cytomegalovirus (HCMV) infection in newborns is well characterized for preterm infants but less so for term infants. We sought to analyze the rates and routes of HCMV transmission in full-term infants during the first year of life. A cohort of 120 HCMV seropositive mothers and their 122 newborns were tested after delivery for HCMV-DNA shedding in different bodily fluids. Postnatal HCMV infection was defined as the detection of >2.5 × 102 HCMV-DNA copies/mL in infants' saliva swabs. Maternal neutralizing antibody serum titer, HCMV-specific T-cell response, and HCMV glycoprotein B immunoglobulin G on breastmilk were analyzed. HCMV shedding was detected in 67 of 120 mothers (55.8%), and 20 of 122 infants (16.4%) developed HCMV infection within the first 3 months of life. Six additional infants were infected during the first year, for a postnatal infection rate of 21.3%. Viral shedding was more frequent in breastmilk than saliva, urine, and vaginal secretions, and the mothers of infected infants showed higher levels of HCMV-DNA in milk. No association was found between the antibody levels in serum or milk and maternal viral shedding, whereas a slightly lower frequency of HCMV-specific CD4+ T-cells with long-term memory phenotype was observed in women with HCM-DNA-positive milk. About one out of five infants develop HCMV infection within the first year of life. Breastmilk appears the major route of transmission of the infection, maternal saliva has a minor role whereas the role of vaginal secretions is negligible.
Collapse
Affiliation(s)
- Piera d'Angelo
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Paola Zelini
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Federica Zavaglio
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Piccini
- Department of Obstetric and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Daniela Cirasola
- Microbiology and Virology Analysis Laboratory, Humanitas Research Hospital, Milano, Italy
| | - Alessia Arossa
- Department of Obstetric and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arsenio Spinillo
- Department of Obstetric and Gynecology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Daniele Lilleri
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fausto Baldanti
- Molecular Virology Unit, Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
12
|
Crooks CM, Chan C, Permar SR. Leveraging preclinical study designs to close gaps in vaccine development for perinatal pathogens. J Exp Med 2023; 220:e20230184. [PMID: 37289272 PMCID: PMC10250551 DOI: 10.1084/jem.20230184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Vaccines to perinatal pathogens are critical for both reducing the burden of endemic pathogens and preparing for the next pandemic. Although they are often at greater risk of severe disease from infection, pregnant people and children are routinely marginalized in the vaccine development process. We highlight several challenges in the vaccine development process and how three tools-translational animal models, human cohort studies of natural infection, and innovative data-use strategies-can speed vaccine development and ensure equity for pregnant people and children in the next pandemic.
Collapse
Affiliation(s)
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
13
|
Moström M, Yu S, Tran D, Saccoccio F, Versoza CJ, Malouli D, Mirza A, Valencia S, Gilbert M, Blair R, Hansen S, Barry P, Früh K, Jensen JD, Pfeifer SP, Kowalik TF, Permar SR, Kaur A. Protective effect of pre-existing natural immunity in a nonhuman primate reinfection model of congenital cytomegalovirus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536057. [PMID: 37090643 PMCID: PMC10120644 DOI: 10.1101/2023.04.10.536057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Congenital cytomegalovirus (cCMV) is the leading infectious cause of neurologic defects in newborns with particularly severe sequelae in the setting of primary CMV infection in the first trimester of pregnancy. The majority of cCMV cases worldwide occur after non-primary infection in CMV-seropositive women; yet the extent to which pre-existing natural CMV-specific immunity protects against CMV reinfection or reactivation during pregnancy remains ill-defined. We previously reported on a novel nonhuman primate model of cCMV in rhesus macaques where 100% placental transmission and 83% fetal loss were seen in CD4 + T lymphocyte-depleted rhesus CMV (RhCMV)-seronegative dams after primary RhCMV infection. To investigate the protective effect of preconception maternal immunity, we performed reinfection studies in CD4+ T lymphocyte-depleted RhCMV-seropositive dams inoculated in late first / early second trimester gestation with RhCMV strains 180.92 ( n =2), or RhCMV UCD52 and FL-RhCMVΔRh13.1/SIV gag , a wild-type-like RhCMV clone with SIV gag inserted as an immunological marker ( n =3). An early transient increase in circulating monocytes followed by boosting of the pre-existing RhCMV-specific CD8+ T lymphocyte and antibody response was observed in the reinfected dams but not in control CD4+ T lymphocyte-depleted dams. Emergence of SIV Gag-specific CD8+ T lymphocyte responses in macaques inoculated with the FL-RhCMVΔRh13.1/SIV gag virus confirmed reinfection. Placental transmission was detected in only one of five reinfected dams and there were no adverse fetal sequelae. Viral whole genome, short-read, deep sequencing analysis confirmed transmission of both reinfection RhCMV strains across the placenta with ∼30% corresponding to FL-RhCMVΔRh13.1/SIV gag and ∼70% to RhCMV UCD52, consistent with the mixed human CMV infections reported in infants with cCMV. Our data showing reduced placental transmission and absence of fetal loss after non-primary as opposed to primary infection in CD4+ T lymphocyte-depleted dams indicates that preconception maternal CMV-specific CD8+ T lymphocyte and/or humoral immunity can protect against cCMV infection. Author Summary Globally, pregnancies in CMV-seropositive women account for the majority of cases of congenital CMV infection but the immune responses needed for protection against placental transmission in mothers with non-primary infection remains unknown. Recently, we developed a nonhuman primate model of primary rhesus CMV (RhCMV) infection in which placental transmission and fetal loss occurred in RhCMV-seronegative CD4+ T lymphocyte-depleted macaques. By conducting similar studies in RhCMV-seropositive dams, we demonstrated the protective effect of pre-existing natural CMV-specific CD8+ T lymphocytes and humoral immunity against congenital CMV after reinfection. A 5-fold reduction in congenital transmission and complete protection against fetal loss was observed in dams with pre-existing immunity compared to primary CMV in this model. Our study is the first formal demonstration in a relevant model of human congenital CMV that natural pre-existing CMV-specific maternal immunity can limit congenital CMV transmission and its sequelae. The nonhuman primate model of non-primary congenital CMV will be especially relevant to studying immune requirements of a maternal vaccine for women in high CMV seroprevalence areas at risk of repeated CMV reinfections during pregnancy.
Collapse
Affiliation(s)
- Matilda Moström
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Shan Yu
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Dollnovan Tran
- Tulane National Primate Research Center, Tulane University, Covington LA
| | | | - Cyril J. Versoza
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ
| | | | - Anne Mirza
- University of Massachusetts Chan Medical School, Worcester, MA
| | - Sarah Valencia
- Duke Human Vaccine Institute, Duke University, Durham, NC
| | - Margaret Gilbert
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Robert Blair
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Scott Hansen
- Oregon Health and Sciences University, Beaverton, OR
| | | | - Klaus Früh
- Oregon Health and Sciences University, Beaverton, OR
| | - Jeffrey D. Jensen
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ
| | - Susanne P. Pfeifer
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ
| | | | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, NC
- Weill Cornell Medicine, New York, NY
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington LA
| |
Collapse
|
14
|
Rahimzadeh G, Safar MJ, Rezai S, Rezai MS, Movahedi FS. Seroepidemiology of HBV, HCV, HIV, HTLV, and CMV in Pregnant Women Referring to Sari Birth Cohort. Adv Biomed Res 2022; 11:97. [PMID: 36660762 PMCID: PMC9843599 DOI: 10.4103/abr.abr_334_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background Congenital infections are among the most important conditions threatening human fetal health, the majority of which are caused by viral agents. Screening pregnant women for viral infections is essential because such infections can cause serious consequences for both the mother and the infant. So, this study aimed to serologically investigate sexually transmitted viral infections in pregnant women and also find the association between the prevalence of viral infections and epidemiological parameters in pregnant women of Sari, Iran. Materials and Methods This descriptive, observational study was performed in pregnant women referring to Sari Birth Cohort Center between 2018 and 2020. A total of 1092 blood samples were investigated for hepatitis B (HBV), hepatitis C (HCV), human immunodeficiency virus (HIV), human T-lymphotropic virus (HTLV), and cytomegalovirus (CMV) serological markers by enzyme-linked immunosorbent assay (ELISA). Results The prevalence of HBsAg positivity, HCV, HIV, and HTLV was 0.2%, 0.09%, 0.09%, and 0.2%, respectively. The percentage of participants with CMV-IgM and -IgG antibody titers above normal was 0.2% and 91.8%, respectively. Pregnant women whose educational level was bachelor's degree or lower, those who did not use a male condom before pregnancy, or those with a family history of infectious disease were found to be more likely to have HBV, HCV, HIV, HTLV, and CMV infections. Conclusion Family history, maternal age, pregnancy stage, and not using a male condom are among the risk factors for sexually transmitted viral infections in pregnant women in Sari.
Collapse
Affiliation(s)
- Golnar Rahimzadeh
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Jafar Safar
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shaghayegh Rezai
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Sadegh Rezai
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran,Address for correspondence: Prof. Mohammad Sadegh Rezai, Professor, Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran. E-mail:
| | | |
Collapse
|
15
|
Sapuan S, Theodosiou AA, Strang BL, Heath PT, Jones CE. A systematic review and meta-analysis of the prevalence of human cytomegalovirus shedding in seropositive pregnant women. Rev Med Virol 2022; 32:e2399. [PMID: 36196755 PMCID: PMC9786761 DOI: 10.1002/rmv.2399] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/25/2022] [Accepted: 09/12/2022] [Indexed: 12/30/2022]
Abstract
The detection of human cytomegalovirus (HCMV) in an individual's bodily fluid by culture techniques or through HCMV DNA detection by polymerase chain reaction, is known as HCMV shedding. Human cytomegalovirus shedding has the potential to transmit HCMV infection, where an individual can become infected with HCMV through contact with the bodily fluid of another individual containing HCMV. Human cytomegalovirus shedding can occur in primary infection and in non-primary infection for individuals with prior infection (HCMV seropositive). Human cytomegalovirus infection causes few or no symptoms in a pregnant woman, but can cause significant harm to her foetus if congenital CMV (cCMV) infection occurs. The association between HCMV shedding in HCMV seropositive pregnant women and the vertical transmission of HCMV to result in cCMV infection is poorly investigated, challenged by a limited understanding of the distribution of HCMV shedding in HCMV seropositive pregnant women. We systematically reviewed the published literature to describe the prevalence of HCMV shedding in HCMV seropositive women during pregnancy up to delivery. This analysis identified nine studies that met our eligibility criteria. In these studies, the prevalence of HCMV shedding in any bodily fluid of HCMV seropositive women during pregnancy and at delivery ranged from 0% to 42.5%. A meta-analysis, performed on six of the nine studies with suitable sample sizes, estimated a pooled prevalence of 21.5% [95% CI 12.7%,30.3%]. To our knowledge, this is the first review to systematically search the literature to summarise the prevalence of HCMV shedding in HCMV seropositive pregnant women. These estimates can help in the development of disease burden models and therapeutic or preventative strategies against cCMV infection in the context of non-primary maternal HCMV infection.
Collapse
Affiliation(s)
- Shari Sapuan
- St George's, University of LondonCentre for Neonatal and Paediatric InfectionLondonUK
| | | | - Blair L. Strang
- St George's, University of LondonInstitute for Infection and ImmunityLondonUK
| | - Paul T. Heath
- St George's, University of LondonCentre for Neonatal and Paediatric InfectionLondonUK
| | - Christine E. Jones
- Faculty of Medicine and Institute for Life SciencesUniversity of Southampton and NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research CentreUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| |
Collapse
|
16
|
Human cytomegalovirus (HCMV) long-term shedding and HCMV-specific immune response in pregnant women with primary HCMV infection. Med Microbiol Immunol 2022; 211:249-260. [PMID: 35960328 DOI: 10.1007/s00430-022-00747-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/21/2022] [Indexed: 10/15/2022]
Abstract
Human cytomegalovirus (HCMV) shedding has been extensively investigated in newborns and in young children, however, much less is known about it in immunocompetent adults. Shedding of HCMV was investigated in saliva, vaginal secretions and urine of pregnant women experiencing primary infection along with the development of the HCMV-specific immune response. Thirty-three pregnant women shed HCMV DNA in peripheral biological fluids at least until one year after onset of infection, while in blood HCMV DNA was cleared earlier. Significantly higher levels of viral load were found in vaginal secretions compared to saliva and urine. All subjects examined two years after the onset of infection showed a high avidity index, with IgM persisting in 36% of women. Viral load in blood was directly correlated with levels of HCMV-specific IgM and inversely correlated with levels of IgG specific for the pentameric complex gH/gL/pUL128L; in addition, viral load in blood was inversely correlated with percentage of HCMV-specific CD4+ and CD8+ expressing IL-7R (long-term memory, LTM) while viral load in biological fluids was inversely correlated with percentage of HCMV-specific CD4+ and CD8+ effector memory RA+(TEMRA). In conclusion, viral shedding during primary infection in pregnancy persists in peripheral biological fluids for at least one year and the development of both antibodies (including those directed toward the pentameric complex) and memory T cells are associated with viral clearance.
Collapse
|
17
|
Hehnly C, Ssentongo P, Bebell LM, Burgoine K, Bazira J, Fronterre C, Kumbakumba E, Mulondo R, Mbabazi-Kabachelor E, Morton SU, Ngonzi J, Ochora M, Olupot-Olupot P, Mugamba J, Onen J, Roberts DJ, Sheldon K, Sinnar SA, Smith J, Ssenyonga P, Kiwanuka J, Paulson JN, Meier FA, Ericson JE, Broach JR, Schiff SJ. Cytomegalovirus infections in infants in Uganda: Newborn-mother pairs, neonates with sepsis, and infants with hydrocephalus. Int J Infect Dis 2022; 118:24-33. [PMID: 35150915 PMCID: PMC9058984 DOI: 10.1016/j.ijid.2022.02.005] [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: 09/21/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES To estimate the prevalence of cytomegalovirus (CMV) infections among newborn-mother pairs, neonates with sepsis, and infants with hydrocephalus in Uganda. DESIGN AND METHODS Three populations-newborn-mother pairs, neonates with sepsis, and infants (≤3 months) with nonpostinfectious (NPIH) or postinfectious (PIH) hydrocephalus-were evaluated for CMV infection at 3 medical centers in Uganda. Quantitative PCR (qPCR) was used to characterize the prevalence of CMV. RESULTS The overall CMV prevalence in 2498 samples across all groups was 9%. In newborn-mother pairs, there was a 3% prevalence of cord blood CMV positivity and 33% prevalence of maternal vaginal shedding. In neonates with clinical sepsis, there was a 2% CMV prevalence. Maternal HIV seropositivity (adjusted odds ratio [aOR] 25.20; 95% confidence interval [CI] 4.43-134.26; p = 0.0001), residence in eastern Uganda (aOR 11.06; 95% CI 2.30-76.18; p = 0.003), maternal age <25 years (aOR 4.54; 95% CI 1.40-19.29; p = 0.02), and increasing neonatal age (aOR 1.08 for each day older; 95% CI 1.00-1.16; p = 0.05), were associated risk factors for CMV in neonates with clinical sepsis. We found a 2-fold higher maternal vaginal shedding in eastern (45%) vs western (22%) Uganda during parturition (n = 22/49 vs 11/50, the Fisher exact test; p = 0.02). In infants with PIH, the prevalence in blood was 24% and in infants with NPIH, it was 20%. CMV was present in the cerebrospinal fluid (CSF) of 13% of infants with PIH compared with 0.5% of infants with NPIH (n = 26/205 vs 1/194, p < 0.0001). CONCLUSIONS Our findings highlight that congenital and postnatal CMV prevalence is substantial in this African setting, and the long-term consequences are uncharacterized.
Collapse
Affiliation(s)
- Christine Hehnly
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Paddy Ssentongo
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, USA; Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Lisa M Bebell
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathy Burgoine
- Neonatal Unit, Department of Paediatrics and Child Health, Mbale Regional Referral Hospital, Mbale, Uganda; Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; Mbale Clinical Research Institute, Mbale Regional Referral Hospital, Mbale, Uganda
| | - Joel Bazira
- Department of Microbiology, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Claudio Fronterre
- Centre for Health Informatics, Computing, and Statistics, Lancaster University, Lancaster, United Kingdom
| | - Elias Kumbakumba
- Department of Pediatrics, Mbarara University of Science and Technology, Mbarara, Uganda
| | | | | | - Sarah U Morton
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Joseph Ngonzi
- Department of Obstetrics and Gynaecology, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Moses Ochora
- Centre for Health Informatics, Computing, and Statistics, Lancaster University, Lancaster, United Kingdom
| | - Peter Olupot-Olupot
- Mbale Clinical Research Institute, Mbale Regional Referral Hospital, Mbale, Uganda; Busitema University, Busitema, Uganda
| | - John Mugamba
- CURE Children's Hospital of Uganda, Mbale, Uganda
| | - Justin Onen
- Mulago National Referral Hospital, Kampala, Uganda
| | - Drucilla J Roberts
- Department of Pathology, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston, MA, USA
| | - Kathryn Sheldon
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Shamim A Sinnar
- Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jasmine Smith
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | | | - Julius Kiwanuka
- Department of Pediatrics, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Joseph N Paulson
- Department of Biostatistics, Product Development, Genentech Inc., San Francisco, CA, USA
| | | | - Jessica E Ericson
- Division of Pediatric Infectious Disease, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - James R Broach
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Steven J Schiff
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, USA; Center for Infectious Disease Dynamics, and Department of Physics, The Pennsylvania State University, University Park, PA, USA; Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
18
|
Jarduli-Maciel LR, de Azevedo JTC, Clave E, Costa TCDM, Arruda LCM, Fournier I, Palma PVB, Lima KC, Elias JB, Stracieri ABP, Pieroni F, Cunha R, Darrigo-Júnior LG, Grecco CES, Covas DT, Silva-Pinto AC, De Santis GC, Simões BP, Oliveira MC, Toubert A, Malmegrim KCR. Allogeneic haematopoietic stem cell transplantation resets T- and B-cell compartments in sickle cell disease patients. Clin Transl Immunology 2022; 11:e1389. [PMID: 35474905 PMCID: PMC9035210 DOI: 10.1002/cti2.1389] [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: 05/23/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/12/2022] Open
Abstract
Objectives Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is the only currently available curative treatment for sickle cell disease (SCD). Here, we comprehensively evaluated the reconstitution of T- and B-cell compartments in 29 SCD patients treated with allo-HSCT and how it correlated with the development of acute graft-versus-host disease (aGvHD). Methods T-cell neogenesis was assessed by quantification of signal-joint and β-chain TCR excision circles. B-cell neogenesis was evaluated by quantification of signal-joint and coding-joint K-chain recombination excision circles. T- and B-cell peripheral subset numbers were assessed by flow cytometry. Results Before allo-HSCT (baseline), T-cell neogenesis was normal in SCD patients compared with age-, gender- and ethnicity-matched healthy controls. Following allo-HSCT, T-cell neogenesis declined but was fully restored to healthy control levels at one year post-transplantation. Peripheral T-cell subset counts were fully restored only at 24 months post-transplantation. Occurrence of acute graft-versus-host disease (aGvHD) transiently affected T- and B-cell neogenesis and overall reconstitution of T- and B-cell peripheral subsets. B-cell neogenesis was significantly higher in SCD patients at baseline than in healthy controls, remaining high throughout the follow-up after allo-HSCT. Notably, after transplantation SCD patients showed increased frequencies of IL-10-producing B-regulatory cells and IgM+ memory B-cell subsets compared with baseline levels and with healthy controls. Conclusion Our findings revealed that the T- and B-cell compartments were normally reconstituted in SCD patients after allo-HSCT. In addition, the increase of IL-10-producing B-regulatory cells may contribute to improve immune regulation and homeostasis after transplantation.
Collapse
Affiliation(s)
- Luciana Ribeiro Jarduli-Maciel
- Graduate Program in Biosciences Applied to Pharmacy School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | - Júlia Teixeira Cottas de Azevedo
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Graduate Program in Basic and Applied Immunology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | | | - Thalita Cristina de Mello Costa
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | | | - Isabelle Fournier
- Laboratoire d'Immunologie et d'Histocompatibilité Hôpital Saint-Louis AP-HP Paris France
| | - Patrícia Vianna Bonini Palma
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | - Keli Cristina Lima
- Graduate Program in Biosciences Applied to Pharmacy School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | | | | | - Fabiano Pieroni
- Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Renato Cunha
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | | | | | - Dimas Tadeu Covas
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Ana Cristina Silva-Pinto
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Gil Cunha De Santis
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Belinda Pinto Simões
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Antoine Toubert
- Université de Paris INSERM UMR 1160 IRSL Paris France.,Laboratoire d'Immunologie et d'Histocompatibilité Hôpital Saint-Louis AP-HP Paris France
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Department of Clinical Analysis, Toxicology and Food Sciences School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| |
Collapse
|
19
|
Gatta LA, Rochat E, Weber JM, Valencia S, Erkanli A, Dotters-Katz SK, Permar S, Hughes BL. Clinical factors associated with cytomegalovirus shedding among seropositive pregnant women. Am J Obstet Gynecol MFM 2022; 4:100560. [PMID: 34990874 PMCID: PMC9942897 DOI: 10.1016/j.ajogmf.2021.100560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/17/2021] [Accepted: 12/29/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Both neighborhood disadvantage and close contact with children have been associated with seroprevalence of cytomegalovirus in pregnancy. However, it is unknown which individual factors influence whether seropositive women are likely to have ongoing viral shedding. OBJECTIVE This study aimed to define the frequency of and risk factors for ongoing maternal cytomegalovirus shedding across gestation among seropositive pregnant women. STUDY DESIGN This was a prospective cohort study of women who were cytomegalovirus seropositive at a single tertiary care hospital between September 1, 2018, and September 1, 2020. The participants were eligible if positive for cytomegalovirus immunoglobulin G during the first trimester of pregnancy. Urine samples were planned to be collected from each trimester. DNA was isolated in urine samples to detect and quantitate cytomegalovirus immediate-early 1 gene. Participants were classified as "ever shedder" if cytomegalovirus was detected in any urine sample and "never shedder" if cytomegalovirus was never detected. Patient demographics and characteristics were compared between groups. Stochastic search variable selection (with a posterior probability of inclusion of >0.5) was used to identify predictors of cytomegalovirus shedding at any time point. Forward selection modeling was used as a sensitivity check for independent risks. RESULTS A total of 240 participants who were cytomegalovirus immunoglobulin G seropositive were enrolled, with 567 urine samples analyzed across gestation. Fifty-eight participants (24.2%) were "never shedders", and 182 participants (75.8%) were "ever shedders." The characteristics and demographics were similar between cohorts. With stochastic search variable selection, nulliparity was the only variable selected (odds ratio, 1.82; 95% credible interval, 1.00-4.09; Bayes factor, 2.22). Furthermore, nulliparity was selected with standard logistic regression, with an odds ratio and 95% confidence interval of 1.89 (1.00-3.58). Sociodemographic characteristics, such as age, race, education level, occupation, children at home, children in daycare, housing type, insurance type, income, and concurrent infections, were not associated with shedding. The only positive neonatal sample (0.42%) was detected from a participant who had cytomegalovirus detected in all 3 time points. CONCLUSION Approximately 75% of women who were positive for cytomegalovirus immunoglobulin G shed virus at some point during gestation. Nulliparity was the only variable selected that was associated with shedding.
Collapse
Affiliation(s)
- Luke A Gatta
- Department of Obstetrics and Gynecology, Duke University Hospital, Durham, NC (Drs Gatta, Dotters-Katz, and Hughes); Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Duke University Hospital, Durham, NC (Drs Gatta, Dotters-Katz, and Hughes).
| | - Eric Rochat
- Duke Human Vaccine Institute, Duke University Hospital, Durham, NC (Mr Rochat and Drs Valencia and Permar)
| | - Jeremy M Weber
- Department of Biostatistics and Bioinformatics, Duke University Hospital, Durham, NC (Mr Weber and Dr Erkanli)
| | - Sarah Valencia
- Duke Human Vaccine Institute, Duke University Hospital, Durham, NC (Mr Rochat and Drs Valencia and Permar)
| | - Alaattin Erkanli
- Department of Biostatistics and Bioinformatics, Duke University Hospital, Durham, NC (Mr Weber and Dr Erkanli)
| | - Sarah K Dotters-Katz
- Department of Obstetrics and Gynecology, Duke University Hospital, Durham, NC (Drs Gatta, Dotters-Katz, and Hughes); Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Duke University Hospital, Durham, NC (Drs Gatta, Dotters-Katz, and Hughes)
| | - Sallie Permar
- Duke Human Vaccine Institute, Duke University Hospital, Durham, NC (Mr Rochat and Drs Valencia and Permar); Department of Pediatrics, Weill Cornell Medical Center, New York, NY (Dr Permar)
| | - Brenna L Hughes
- Department of Obstetrics and Gynecology, Duke University Hospital, Durham, NC (Drs Gatta, Dotters-Katz, and Hughes); Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Duke University Hospital, Durham, NC (Drs Gatta, Dotters-Katz, and Hughes)
| |
Collapse
|
20
|
Tan NK, Pope CF, Carrington D. Screening for cytomegalovirus shedding in vagina and saliva: Significant differences between biological fluids, swab types and storage durations in DNA recovery. J Clin Virol 2022; 146:105055. [DOI: 10.1016/j.jcv.2021.105055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/06/2021] [Accepted: 12/11/2021] [Indexed: 10/19/2022]
|
21
|
Congenital Cytomegalovirus Infections Mother-Newborn Pair Study in Southern Ethiopia. CANADIAN JOURNAL OF INFECTIOUS DISEASES AND MEDICAL MICROBIOLOGY 2021; 2021:4646743. [PMID: 35003406 PMCID: PMC8739911 DOI: 10.1155/2021/4646743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/08/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022]
Abstract
Introduction. Congenital cytomegalovirus (cCMV) is a common cause of neurodevelopmental delays and sensorineural hearing loss of infants, yet the prevalence of cCMV and the associated factors in Ethiopia are not studied. Hence, this study was to assess the prevalence and associated factors of cCMV in Southern Ethiopia. Methodology. A mother-newborn pair cross-sectional study was conducted at Hawassa University Comprehensive and Specialized Hospital, Ethiopia. Newborn’s saliva sample was tested for cCMV using Alethia CMV molecular assay. Mothers’ serum was tested serologically for anti-CMV IgM and IgG by EUROIMMUN ELISA. Pregnant women responded to a questionnaire about their previous and current obstetric history and sociodemographic characteristics. The chi-square (χ2) test and independent-sample t-test were used to determine the associations between infections and possible risk factors; then, potential variables were screened for multivariable analysis. Results. A total of 593 mother-newborn pairs were assessed. CMV was detected in 14 of 593 newborn saliva swabs (2.4%; 95% CI 1.2–3.7). As assessed by CMV IgM-positive results, maternal CMV seropositivity was 8.3% (49/593); thus, the rate of mother-to-child transmission of CMV was 28% (14/49) among CMV IgM-positive women. Congenital CMV infection was significantly associated with maternal exposure through nursery school children in the household, women sharing a feeding cup with children, and any of the detected curable STIs during pregnancy. Birth weight was negatively associated with CMV infection. Maternal age, gravidity, level of education, and sharing of children feeding utensils were not associated with cCMV infection. Conclusion. A high rate of cCMV infection in the absence of awareness demands further in-depth investigation in Ethiopia. Thus, policymakers must take appropriate action through the antenatal care system for prevention strategies and put in place a constant health education and awareness creation of pregnant women about the causes of infection and hygienic measures.
Collapse
|
22
|
Zelini P, Piera d'Angelo, De Cicco M, Achille C, Sarasini A, Fiorina L, Cirasola D, Marazzi V, Piccini S, Furione M, Arossa A, Muscettola G, Spinillo A, Lilleri D. Human cytomegalovirus non-primary infection during pregnancy: antibody response, risk factors and newborn outcome. Clin Microbiol Infect 2021; 28:1375-1381. [PMID: 34555536 DOI: 10.1016/j.cmi.2021.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Human cytomegalovirus (HCMV) non-primary infections can occur in pregnant women and may result in congenital infection. Comprehensive studies investigating the frequency, characteristics, risk factors and immune response of non-primary infection in pregnancy are missing, while rate of vertical transmission is not known. METHODS HCMV non-primary infection was investigated prospectively in 250 pregnant women. Blood and urine samples as well as saliva and vaginal swabs were collected at 13, 21 and 31 weeks of gestation and at delivery. HCMV-DNA and specific IgG and IgM levels were determined. RESULTS Overall, 105/250 pregnant women (42.0%) developed non-primary infection. HCMV-DNA was detected more frequently in the vaginal secretions (84/250 of the women, 33.6%) than in the urine (35/250, 14.0%), in the saliva (26/250, 10.4%), and in the blood (7/250, 3.0%). The rate of HCMV non-primary infection increased significantly with the progression of pregnancy (from 12.9% in the first trimesters of gestation to 21.9% at delivery, p<0.01). IgM was detected in 25/250 of the women (10.0%), with no association with non-primary infection, while anti-gB IgG was significantly higher (p<0.01) in women with non-primary infection. Age and close contact with children were not associated with non-primary infection. No woman with non-primary infection transmitted the infection to the fetus (95% confidence interval of transmission rate: 0-3.5%). CONCLUSION Although HCMV non-primary infection is frequent during pregnancy, the rate of congenital infection as a consequence of non-primary infection is likely to be ≤3.5%.
Collapse
Affiliation(s)
- Paola Zelini
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy.
| | - Piera d'Angelo
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Marica De Cicco
- Genetics, Transplantology and Cardiovascular Diseases Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Cristian Achille
- Neonatology and Neonatal Intensive Care Unit, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Antonella Sarasini
- Molecular Virology Unit, Department of Microbiology and Virology, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Loretta Fiorina
- Genetics, Transplantology and Cardiovascular Diseases Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Daniela Cirasola
- Genetics, Transplantology and Cardiovascular Diseases Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Valentina Marazzi
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Stefania Piccini
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Milena Furione
- Molecular Virology Unit, Department of Microbiology and Virology, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessia Arossa
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Giulia Muscettola
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Arsenio Spinillo
- Department of Obstetrics and Gynecology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Daniele Lilleri
- Genetics, Transplantology and Cardiovascular Diseases Laboratories, IRCCS Policlinico San Matteo Foundation, Pavia, Italy; Molecular Virology Unit, Department of Microbiology and Virology, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| |
Collapse
|
23
|
Dorfman JR, Balla SR, Pathirana J, Groome MJ, Madhi SA, Moore PL. In utero human cytomegalovirus infection is associated with increased levels of putatively protective maternal antibodies in nonprimary infection: evidence for boosting but not protection. Clin Infect Dis 2021; 73:e981-e987. [PMID: 33560335 DOI: 10.1093/cid/ciab099] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/02/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Although primary maternal cytomegalovirus infections are associated with higher risk of in utero transmission, most fetal infections worldwide result from nonprimary maternal infections. Antibodies directed at glycoprotein B and the gH/gL/pUL128-130-131 pentamer can neutralize virus, and higher levels of antibody directed at several particular pentamer epitopes defined by monoclonal antibodies (mAbs) are associated with reduced risk of fetal cytomegalovirus transmission during primary maternal infection. This had not been explored in maternal nonprimary infection. METHODS In a setting where most maternal cytomegalovirus infections are nonprimary, 42 mothers of infants with congenital CMV infections (transmitters) were compared to 75 cytomegalovirus-seropositive mothers whose infants were cytomegalovirus-uninfected (nontransmitters). Control infants were matched by sex, maternal HIV status and gestational age. We measured the ability of maternal antibodies to block three key pentameric epitopes: one in the gH subunit, another straddling UL130/UL131 and the third straddling gH/gL/UL128/UL130. We tested if levels of antibodies directed at these epitopes were higher in nontransmitters compared to transmitters. RESULTS Levels of all three putatively protective pentamer-directed antibodies were significantly higher in transmitters compared to nontransmitters. In contrast, antibodies targeting an epitope on glycoprotein B were not different. Total antibody specific for pentamer and for gB were also higher in transmitters. CONCLUSIONS We found no evidence that higher levels of any CMV-specific antibodies were associated with reduced risk of congenital CMV infection in nonprimary maternal infection. Instead, we found higher maternal antibody targeting epitopes on CMV pentamer in transmitters than nontransmitters, providing evidence for antibody boosting but not protection.
Collapse
Affiliation(s)
- Jeffrey R Dorfman
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Division of Medical Virology, Department of Pathology, Stellenbosch University, Cape Town, South Africa
| | - Sashkia R Balla
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jayani Pathirana
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Michelle J Groome
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Penny L Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
24
|
Mussi-Pinhata MM, Yamamoto AY. Natural History of Congenital Cytomegalovirus Infection in Highly Seropositive Populations. J Infect Dis 2021; 221:S15-S22. [PMID: 32134482 DOI: 10.1093/infdis/jiz443] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Maternal preconceptional cytomegalovirus (CMV) immunity does not protect the fetus from acquiring congenital CMV infection (cCMV). Nonprimary infections due to recurrence of latent infections or reinfection with new virus strains during pregnancy can result in fetal infection. Because the prevalence of cCMV increases with increasing maternal CMV seroprevalence, the vast majority of the cases of cCMV throughout the world follow nonprimary maternal infections and is more common in individuals of lower socioeconomic background. Horizontal exposures to persons shedding virus in bodily secretions (young children, sexual activity, household crowding, low income) probably increase the risk of acquisition of an exogenous nonprimary CMV infection and fetal transmission. In addition, more frequent acquisition of new antibody reactivities in transmitter mothers suggest that maternal reinfection by new viral strains could be a major source of congenital infection in such populations. However, the exact frequency of CMV nonprimary infection in seroimmune women during pregnancy and the rate of intrauterine transmission in these women are yet to be defined. Usually, the birth prevalence of cCMV is high (≥7:1000) in highly seropositive populations. There is increasing evidence that the frequency and severity of the clinical and laboratory abnormalities in infants with congenital CMV infection born to mothers with nonprimary CMV infection are similar to infants born after a primary maternal infection. This is particularly true for sensorineural hearing loss, which contributes to one third of all early-onset hearing loss in seropositive populations. This brief overview will discuss the need for more research to better clarify the natural history of cCMV in highly seropositive populations, which, in almost all populations, remains incompletely defined.
Collapse
|
25
|
Boppana SB, Britt WJ. Recent Approaches and Strategies in the Generation of Anti-human Cytomegalovirus Vaccines. Methods Mol Biol 2021; 2244:403-463. [PMID: 33555597 DOI: 10.1007/978-1-0716-1111-1_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human cytomegalovirus is the largest human herpesvirus and shares many core features of other herpesviruses such as tightly regulated gene expression during genome replication and latency as well as the establishment of lifelong persistence following infection. In contrast to stereotypic clinical syndromes associated with alpha-herpesvirus infections, almost all primary HCMV infections are asymptomatic and acquired early in life in most populations in the world. Although asymptomatic in most individuals, HCMV is a major cause of disease in hosts with deficits in adaptive and innate immunity such as infants who are infected in utero and allograft recipients following transplantation. Congenital HCMV is a commonly acquired infection in the developing fetus that can result in a number of neurodevelopmental abnormalities. Similarly, HCMV is a major cause of disease in allograft recipients in the immediate and late posttransplant period and is thought to be a major contributor to chronic allograft rejection. Even though HCMV induces robust innate and adaptive immune responses, it also encodes a vast array of immune evasion functions that are thought aid in its persistence. Immune correlates of protective immunity that prevent or modify intrauterine HCMV infection remain incompletely defined but are thought to consist primarily of adaptive responses in the pregnant mother, thus making congenital HCMV a potentially vaccine modifiable disease. Similarly, HCMV infection in allograft recipients is often more severe in recipients without preexisting adaptive immunity to HCMV. Thus, there has been a considerable effort to modify HCMV specific immunity in transplant recipient either through active immunization or passive transfer of adaptive effector functions. Although efforts to develop an efficacious vaccine and/or passive immunotherapy to limit HCMV disease have been underway for nearly six decades, most have met with limited success at best. In contrast to previous efforts, current HCMV vaccine development has relied on observations of unique properties of HCMV in hopes of reproducing immune responses that at a minimum will be similar to that following natural infection. However, more recent findings have suggested that immunity following naturally acquired HCMV infection may have limited protective activity and almost certainly, is not sterilizing. Such observations suggest that either the induction of natural immunity must be specifically tailored to generate protective activity or alternatively, that providing targeted passive immunity to susceptible populations could be prove to be more efficacious.
Collapse
Affiliation(s)
- Suresh B Boppana
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA.,Departments of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - William J Britt
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA. .,Departments of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA. .,Departments of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
26
|
Alain S, Garnier-Geoffroy F, Labrunie A, Montané A, Marin B, Gatet M, Grosjean J, Dufour V, Saugeras M, Postil D, Hantz S. Cytomegalovirus (CMV) Shedding in French Day-Care Centers: A Nationwide Study of Epidemiology, Risk Factors, Centers' Practices, and Parents' Awareness of CMV. J Pediatric Infect Dis Soc 2020; 9:686-694. [PMID: 32068854 DOI: 10.1093/jpids/piz097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/12/2020] [Indexed: 11/12/2022]
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) remains an important healthcare burden, resulting from primary or secondary infection in pregnant women. Exposure to young children's saliva is a major risk factor, as prevalence of CMV shedding can reach 34%. METHODS This cross-sectional, multicenter, nationwide study was conducted in randomly selected day care centers (DCCs), and complemented with a survey among parents and DCCs. All children aged >3 months were eligible. The study measured the CMV shedding prevalence in children's saliva and described CMV genotypes epidemiology. The risk factors for CMV shedding and high viral load were evaluated using multivariable models. RESULTS A total of 93 DCCs participated. Among the 1770 enrolled children with evaluable samples, the CMV shedding prevalence was 40% (713/1770, 95% confidence interval, 34.6-46.1), independently associated with children aged between 12 and 18 months, history of CMV infection in ≥1 parents, a mid-level income. Prevalence increased with DCC staff workload and attending children number. Viral load was ≥5 log-copies CMV/mL in 48% (342/713). Risk factors for higher viral load included children aged between 12 and 18 months, and still being breastfed. The most frequent genotype combinations were gB1-gN4c-gH2 (6.9%), gB1-gN2-gH2 (6.3%), gB4a-gN3a-gH1 (6.3%), and gB1-gN3b-gH2 (5,7%). CMV awareness was low in parents: their serological status was unknown by 72% of mothers and 82% of fathers. Only 41% knew something about CMV. CONCLUSIONS CMV shedding was independently associated with risk factors related to the children, family and DCC. Some of these risk factors may influence prevention strategies, including through an improved information provided to parents. CLINICAL TRIALS REGISTRATION NCT01704222.
Collapse
Affiliation(s)
- Sophie Alain
- National Reference Center for Herpesviruses, Limoges, France.,Inserm U1092 University Limoges, Limoges, France.,Bacteriology, Virology, Hygiene Department, CHU Limoges, Limoges, France
| | - Françoise Garnier-Geoffroy
- National Reference Center for Herpesviruses, Limoges, France.,Inserm U1092 University Limoges, Limoges, France.,Bacteriology, Virology, Hygiene Department, CHU Limoges, Limoges, France
| | - Anaïs Labrunie
- Centre for Methodology and Data Management (Cebimer Department), CHU Limoges, Limoges, France
| | | | - Benoit Marin
- Centre for Methodology and Data Management (Cebimer Department), CHU Limoges, Limoges, France.,General Directorate for Health, Paris, France
| | | | - Jérôme Grosjean
- National Reference Center for Herpesviruses, Limoges, France.,Bacteriology, Virology, Hygiene Department, CHU Limoges, Limoges, France.,Biology Department, Chambery Hospital, Chambery, France
| | - Véronique Dufour
- Families and Early Childhood Directorate, Infantile Maternal Protection Department, Paris, France
| | - Mathilde Saugeras
- Center of Clinical Investigation Department, CHU Limoges, Limoges, France
| | - Deborah Postil
- Centre for Methodology and Data Management (Cebimer Department), CHU Limoges, Limoges, France
| | - Sébastien Hantz
- National Reference Center for Herpesviruses, Limoges, France.,Inserm U1092 University Limoges, Limoges, France.,Bacteriology, Virology, Hygiene Department, CHU Limoges, Limoges, France
| |
Collapse
|
27
|
Pang J, Slyker JA, Roy S, Bryant J, Atkinson C, Cudini J, Farquhar C, Griffiths P, Kiarie J, Morfopoulou S, Roxby AC, Tutil H, Williams R, Gantt S, Goldstein RA, Breuer J. Mixed cytomegalovirus genotypes in HIV-positive mothers show compartmentalization and distinct patterns of transmission to infants. eLife 2020; 9:e63199. [PMID: 33382036 PMCID: PMC7806273 DOI: 10.7554/elife.63199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022] Open
Abstract
Cytomegalovirus (CMV) is the commonest cause of congenital infection and particularly so among infants born to HIV-infected women. Studies of congenital CMV infection (cCMVi) pathogenesis are complicated by the presence of multiple infecting maternal CMV strains, especially in HIV-positive women, and the large, recombinant CMV genome. Using newly developed tools to reconstruct CMV haplotypes, we demonstrate anatomic CMV compartmentalization in five HIV-infected mothers and identify the possibility of congenitally transmitted genotypes in three of their infants. A single CMV strain was transmitted in each congenitally infected case, and all were closely related to those that predominate in the cognate maternal cervix. Compared to non-transmitted strains, these congenitally transmitted CMV strains showed statistically significant similarities in 19 genes associated with tissue tropism and immunomodulation. In all infants, incident superinfections with distinct strains from breast milk were captured during follow-up. The results represent potentially important new insights into the virologic determinants of early CMV infection.
Collapse
Affiliation(s)
- Juanita Pang
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Jennifer A Slyker
- Departments of Global Health and Epidemiology, University of WashingtonSeattleUnited States
| | - Sunando Roy
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Josephine Bryant
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Claire Atkinson
- Institute of Immunology and Transplantation, Division of Infection and Immunity, University College LondonLondonUnited Kingdom
| | - Juliana Cudini
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Carey Farquhar
- Departments of Global Health, Epidemiology, Medicine (Div. Allergy and Infectious Diseases), University of WashingtonSeattleUnited States
| | - Paul Griffiths
- Institute of Immunology and Transplantation, Division of Infection and Immunity, University College LondonLondonUnited Kingdom
| | - James Kiarie
- University of Nairobi, Department of Obstetrics and Gynaecology, World Health OrganizationNairobiKenya
| | - Sofia Morfopoulou
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Alison C Roxby
- Departments of Global Health, Epidemiology, Medicine (Div. Allergy and Infectious Diseases), University of WashingtonSeattleUnited States
| | - Helena Tutil
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Rachel Williams
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Soren Gantt
- Research Centre of the Sainte-Justine University Hospital, Department of Microbiology, Infectious Diseases and Immunology, University of Montréal QCMontréalCanada
| | - Richard A Goldstein
- Division of Infection and Immunity, University College London, Cruciform BuildingLondonUnited Kingdom
| | - Judith Breuer
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, University College LondonLondonUnited Kingdom
| |
Collapse
|
28
|
Osterholm EA, Schleiss MR. Impact of breast milk-acquired cytomegalovirus infection in premature infants: Pathogenesis, prevention, and clinical consequences? Rev Med Virol 2020; 30:1-11. [PMID: 32662174 PMCID: PMC8173712 DOI: 10.1002/rmv.2117] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023]
Abstract
Maternal-fetal transmission of cytomegalovirus (CMV) represents the most common infectious cause of long-term neurodevelopmental disability in children. Congenital CMV (cCMV) infection is associated with microcephaly, seizure disorders, cognitive disability, developmental delay, and sensorineural hearing loss (SNHL). Of these disabilities, SNHL is the most common, affecting approximately 10% of infants with cCMV. Although the sequelae of cCMV are well recognized, it is much less clear what long-term morbidities may occur in neonates that acquire post-natal CMV infection. Post-natal CMV (pCMV) infection is most commonly transmitted by breast-feeding, and in full-term infants is of little consequence. However, in preterm, very-low birthweight (VLBW) infants (<1500 g), pCMV can result in a severe sepsis-like syndrome, with wide-ranging end-organ disease manifestations. Although such short-term complications are well recognized among clinicians caring for premature infants, the long-term risks with respect to adverse neurodevelopmental outcomes remain controversial. In this review, we provide an overview of the clinical manifestations of breast milk-acquired pCMV infection. In particular, we summarize studies that have examined-sometimes with conflicting conclusions-the risks of long-term adverse neurodevelopmental outcome in VLBW infants that acquire pCMV from breast milk. We highlight proposed preventive strategies and antiviral interventions, and offer recommendations for high-priority areas for future basic science and clinical research.
Collapse
Affiliation(s)
- Erin A. Osterholm
- Department of Pediatrics, Division of Neonatology, University of Minnesota, Minneapolis, Minnesota
| | - Mark R. Schleiss
- Department of Pediatrics, Division of Infectious Diseases and Immunology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
29
|
Khodabandehloo M, Sharifi P. Seroprevalence of Cytomegalovirus Antibodies by Electrochemiluminescence Method in Young Women Referred to the Clinical Laboratory, Sanandaj, Iran. INTERNATIONAL JOURNAL OF EPIDEMIOLOGIC RESEARCH 2020. [DOI: 10.34172/ijer.2020.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background and aims: Maternal primary and recurrent infection of cytomegalovirus (CMV) may be transmitted to the fetus during pregnancy and may have complications such as death or growth, along with the development retardation of the fetus and infant. The aim of this study was to determine the prevalence of immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies against CMV in young women, Sanandaj, Iran. Methods: To this end, 90 women (15-40 years old) referring to a clinical laboratory were randomly selected and announced their informed consent to participate in this cross-sectional study. Demographic information and women’s data were collected, including pregnancy, history of abortion, and history of blood transfusion. Then, women’s sera were measured for CMV IgG and IgM antibodies using the electrochemiluminescence technique. Finally, the data were analyzed by SPSS statistical software. Results: The prevalence of IgG and IgM antibodies against CMV in women was 92.2% (95% CI = 86.5-97.8) and 0%, respectively. In addition, the average CMV IgG antibody level was about 137.52 ± 85.215 SD IU/mL. The results revealed a significant statistical association between IgG antibody and pregnancy (P value = 0.012) while there was no association between CMV IgG antibody and other demographic data. Conclusions: In general, high percentages of women had CMV IgG antibody whereas 7.8% of them were susceptible. They are expected to acquire CMV primary infection, and therefore, the screening of antibodies to CMV is suggested for prenatal care.
Collapse
Affiliation(s)
- Mazaher Khodabandehloo
- Ph.D, Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Pezhman Sharifi
- M.Sc, Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
30
|
Park JH, Kim N, Shin S, Roh EY, Yoon JH, Park H. Prevalence and correlated factors of sexually transmitted infections among women attending a Korean sexual assault center. J Forensic Leg Med 2020; 71:101935. [PMID: 32342905 DOI: 10.1016/j.jflm.2020.101935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/05/2020] [Indexed: 11/26/2022]
Abstract
Long-term observation of the prevalence of sexually transmitted infections (STIs) is unclear in sexual assault victims (SAVs).We aimed to identify the trends in the prevalence of STIs in SAVs and factors correlated with STIs. Between January 2010 and May 2019, a retrospective observational study was conducted in a regional sexual assault center in Seoul. Data were collected from sexual assault questionnaires and laboratory results. PCR for STIs, including Chlamydia trachomatis (CT), Neisseria gonorrhoeae (NG), Mycoplasma genitalium (MG), Mycoplasma hominis (MH), Ureaplasma urealyticum (UU), Trichomonas vaginalis (TV) and cytomegalovirus (CMV), was performed via vaginal swab. Associations between STIs and covariates were examined by logistic regression. A total of 1843 SAVs were included, and 60.2% were positive for at least one STI. UU was the dominant agent (32.6%), followed by MH (27.7%) and CT (17.4%). The annual positivity rate ofMH showed a significant decreasing trend (tau = -0.600, P value = 0.020). The highest positivity rate of STI by age group was 68.0% (282/415) in the 15-19 years of age group. Age (OR = 0.951, 95% confidence interval [CI] = 0.934-0.968) and pyuria (OR = 1.898, 95% CI = 1.543-2.329) were associated with clinically important STIs (NG, CT and TV). The prevalence of STIs in SAVs was consistently high over 10 years, and most STIs were more prevalent in SAVs than in the general population. Presumptive treatment is still needed, especially for young SAVs with pyuria.
Collapse
Affiliation(s)
- Jae Hyeon Park
- Department of Laboratory Medicine, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| | - Namhee Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Department of Laboratory Medicine, Seoul National University Boramae Medical Center, 20, Borama-ro 5-gil, Dongjak-gu, Seoul, 07061, South Korea.
| | - Sue Shin
- Department of Laboratory Medicine, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Department of Laboratory Medicine, Seoul National University Boramae Medical Center, 20, Borama-ro 5-gil, Dongjak-gu, Seoul, 07061, South Korea.
| | - Eun Youn Roh
- Department of Laboratory Medicine, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Department of Laboratory Medicine, Seoul National University Boramae Medical Center, 20, Borama-ro 5-gil, Dongjak-gu, Seoul, 07061, South Korea.
| | - Jong Hyun Yoon
- Department of Laboratory Medicine, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Department of Laboratory Medicine, Seoul National University Boramae Medical Center, 20, Borama-ro 5-gil, Dongjak-gu, Seoul, 07061, South Korea.
| | - Hyunwoong Park
- Department of Laboratory Medicine, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea; Department of Laboratory Medicine, Seoul National University Boramae Medical Center, 20, Borama-ro 5-gil, Dongjak-gu, Seoul, 07061, South Korea.
| |
Collapse
|
31
|
Abstract
: The use of cytomegalovirus (CMV) as a vaccine vector to express antigens against multiple infectious diseases, including simian immunodeficiency virus, Ebola virus, plasmodium, and mycobacterium tuberculosis, in rhesus macaques has generated extraordinary levels of protective immunity against subsequent pathogenic challenge. Moreover, the mechanisms of immune protection have altered paradigms about viral vector-mediated immunity against ectopically expressed vaccine antigens. Further optimization of CMV-vectored vaccines, particularly as this approach moves to human clinical trials will be augmented by a more complete understanding of how CMV engenders mechanisms of immune protection. This review summarizes the particulars of the specific CMV vaccine vector that has been used to date (rhesus CMV strain 68-1) in relation to CMV natural history.
Collapse
|
32
|
Abstract
Cytomegalovirus (CMV) enters latency after primary infection and can reactivate periodically with virus excreted in body fluids which can be called shedding. CMV shedding during the early stage of pregnancy is associated with adverse pregnancy outcome. The shedding pattern in healthy seropositive women who plan to have babies has not been well characterised. Vaginal swabs, urine and blood were collected from 1262 CMV IgG-positive women who intended to have babies and tested for CMV DNA by fluorogenic quantitative PCR method. Serum IgM was also detected. The association between sociodemographic characteristics and CMV shedding prevalence was analysed. Among 1262 seropositive women, 12.8% (161/1262) were detected CMV DNA positive in at least one body fluid. CMV DNA was more frequently detected in vaginal secretion (10.5%) than in urine (3.2%) and blood (0.6%) also with higher viral loads (P < 0.00). CMV shedding was more likely detected in IgM-positive women than IgM-negative women (29.5% (13/44) vs. 12.2% (148/1218); OR 3.03, 95% CI 1.55–5.93; P = 0.001). CMV shedding in vaginal secretion was highly correlated with shedding in urine, the immune state of IgM, the adverse pregnant history and younger age. CMV shedding was more commonly detected in vaginal secretion than in urine or blood with higher viral loads among healthy seropositive women of reproductive age. Further studies are needed to figure out whether the shedding is occasional or continuous and whether it is associated with adverse pregnancy outcomes.
Collapse
|
33
|
Diogo MC, Glatter S, Binder J, Kiss H, Prayer D. The MRI spectrum of congenital cytomegalovirus infection. Prenat Diagn 2020; 40:110-124. [PMID: 31802515 PMCID: PMC7027449 DOI: 10.1002/pd.5591] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/16/2019] [Accepted: 10/07/2019] [Indexed: 01/01/2023]
Abstract
Human cytomegalovirus (CMV) is an ubiquitous pathogen, with a high worldwide seroprevalence. When acquired in the prenatal period, congenital CMV (cCMV) is a major cause of neurodevelopmental sequelae and hearing loss. cCMV remains an underdiagnosed condition, with no systematic screening implemented in pregnancy or in the postnatal period. Therefore, imaging takes a prominent role in prenatal diagnosis of cCMV. With the prospect of new viable therapies, accurate and timely diagnosis becomes paramount, as well as identification of fetuses at risk for neurodevelopmental sequelae. Fetal magnetic resonance imaging (MRI) provides a complementary method to ultrasound (US) in fetal brain and body imaging. Anterior temporal lobe lesions are the most specific finding, and MRI is superior to US in their detection. Other findings such as ventriculomegaly, cortical malformations and calcifications, as well as hepatosplenomegaly, liver signal changes and abnormal effusions are unspecific. However, when seen in combination these should raise the suspicion of fetal infection, highlighting the need for a full fetal assessment. Still, some fetuses deemed normal on prenatal imaging are symptomatic at birth or develop delayed cCMV-associated symptoms, leaving room for improvement of diagnostic tools. Advanced MR sequences may help in this field and in determining prognosis, but further studies are needed.
Collapse
Affiliation(s)
- Mariana C. Diogo
- Department of Image Guided TherapyUniversity Clinic for Neuroradiology and Musculoskeletal Radiology, Medical University of ViennaViennaAustria
| | - Sarah Glatter
- Department of Image Guided TherapyUniversity Clinic for Neuroradiology and Musculoskeletal Radiology, Medical University of ViennaViennaAustria
- Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Julia Binder
- Department of Obstetrics and GynecologyMedical University of ViennaViennaAustria
| | - Herbert Kiss
- Department of Obstetrics and GynecologyMedical University of ViennaViennaAustria
| | - Daniela Prayer
- Department of Image Guided TherapyUniversity Clinic for Neuroradiology and Musculoskeletal Radiology, Medical University of ViennaViennaAustria
| |
Collapse
|
34
|
Dons’koi BV, Tutchenko TM, Chernyshov VP, Stepaniuk KS. HCMV seropositivity is associated with specific proinflammatory immune phenotype in women with implantation failure. Immunol Lett 2020; 217:84-90. [DOI: 10.1016/j.imlet.2019.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/07/2019] [Accepted: 11/17/2019] [Indexed: 01/06/2023]
|
35
|
Plotkin SA. Seroconversion for Cytomegalovirus Infection During Pregnancy and Fetal Infection in a Highly Seropositive Population: "The BraCHS Study," by Mussi-Pinhata et al. J Infect Dis 2019; 218:1188-1190. [PMID: 29868893 DOI: 10.1093/infdis/jiy322] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/01/2018] [Indexed: 11/14/2022] Open
Affiliation(s)
- Stanley A Plotkin
- Emeritus Professor of Pediatrics, University of Pennsylvania, Philadelphia
| |
Collapse
|
36
|
Parolini F, Hadzic N, Davenport M. Adjuvant therapy of cytomegalovirus IgM + ve associated biliary atresia: Prima facie evidence of effect. J Pediatr Surg 2019; 54:1941-1945. [PMID: 30772005 DOI: 10.1016/j.jpedsurg.2018.12.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/14/2018] [Accepted: 12/16/2018] [Indexed: 12/19/2022]
Abstract
AIM OF STUDY CMV-IgM + ve associated biliary atresia (CMV-BA) is a distinct etiological subgroup characterized by older age at presentation and a greater degree of inflammation and hepatic fibrosis, leading to a worse outcome. We report our experience with adjuvant antiviral therapy after Kasai portoenterostomy (KPE). METHODS Single-center prospective database identification of CMV-IgM + ve associated BA managed between 2003 and 2017. Since 2011, IV ganciclovir (5 mg/kg b.d.) and/or oral valganciclovir (520 mg/m2 b.d.) were started in the early postoperative period in selected cases and continued until negativity of CMV DNA load [Anti-Viral Therapy (AVT) Group 1]. Clearance of jaundice was defined as achieving a total bilirubin ≤20 μmol/L in post-KPE period and tested with a Fisher test; native liver survival (NLS) and overall actuarial survival (OS) were compared with untreated BA CMV IgM + ve patients (Group 2) using a Log-Rank test. A P value of <0.05 was regarded as significant. Data are quoted as median (IQ range). RESULTS During the 14-year period, 376 infants with histologically confirmed BA were treated; of those 38(10%) were CMV IgM + ve at presentation. One child was considered too late at presentation for KPE and underwent primary liver transplantation while another only started AVT one month after KPE. Both were excluded from survival analysis. Therefore 36 underwent KPE [AVT Group 1 (n = 8) and Control Group 2 (n = 28)]. Overall age at surgery was 67(53-77) days. There was no difference in age at surgery (P = 0.26); bilirubin (P = 0.12); or AST (P = 0.15) between Group 1 and Group 2. Viral load data were available in 16 with a trend towards higher counts in the AVT group 1 [4935 (2668-18,817) vs. 1296 (253-10,471) c/ml; P = 0.06]. Clearance of jaundice was higher in AVT Group 1 (75% vs 21%, P = 0.009). There was no difference in OS (P = 0.24) but NLS was improved in the AVT Group 1 (75% vs. 25% at 2 years; P = 0.04). CONCLUSIONS Although this finding may be regarded as preliminary, adjuvant antiviral therapy appeared to improve outcome in infants with CMV IgM + ve BA. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
- Filippo Parolini
- Department of Paediatric Surgery, Denmark Hill, London, UK SE5 9RS; Department of Hepatology Kings College Hospital, Denmark Hill, London, UK SE5 9RS
| | - Nedim Hadzic
- Department of Paediatric Surgery, Denmark Hill, London, UK SE5 9RS; Department of Hepatology Kings College Hospital, Denmark Hill, London, UK SE5 9RS
| | - Mark Davenport
- Department of Paediatric Surgery, Denmark Hill, London, UK SE5 9RS; Department of Hepatology Kings College Hospital, Denmark Hill, London, UK SE5 9RS.
| |
Collapse
|
37
|
Gomes AC, Griffiths PD, Reeves MB. The Humoral Immune Response Against the gB Vaccine: Lessons Learnt from Protection in Solid Organ Transplantation. Vaccines (Basel) 2019; 7:E67. [PMID: 31319553 PMCID: PMC6789498 DOI: 10.3390/vaccines7030067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022] Open
Abstract
Human cytomegalovirus (hCMV) is considered to be the highest priority for vaccine development. This view is underscored by the significant morbidity associated with congenital hCMV infection and viraemia in transplant patients. Although a number of vaccines have been trialed, none have been licensed. The hCMV vaccine candidate that has performed best in clinical trials to date is the recombinant glycoprotein B (gB) vaccine that has demonstrated protection, ranging from a 43% to 50% efficacy in three independent phase II trials. In this review, we focus on data from the phase II trial performed in solid organ transplant patients and the outcomes of follow-up studies attempting to identify immunological and mechanistic correlates of protection associated with this vaccine strategy. We relate this to other vaccine studies of gB as well as other vaccine strategies to determine areas of commonality and divergence. Finally, through the review, we discuss the unique challenges and opportunities presented with vaccine studies in transplant populations with recommendations that could empower subsequent trials.
Collapse
Affiliation(s)
- Ariane C Gomes
- Institute for Immunity and Transplantation, University College London, London NW3 2PF, UK
| | - Paul D Griffiths
- Institute for Immunity and Transplantation, University College London, London NW3 2PF, UK
| | - Matthew B Reeves
- Institute for Immunity and Transplantation, University College London, London NW3 2PF, UK.
| |
Collapse
|
38
|
Abstract
Background: Congenital cytomegalovirus (cCMV) is the leading cause of nongenetic congenital hearing loss in much of the world and a leading cause of neurodevelopmental disabilities. Infected babies can be born to women who are seropositive and seronegative prior to pregnancy, and the incidence is approximately 0.6%-0.7% in the United States. Symptoms vary from mild to severe, and hearing loss can be delayed in onset and progressive. Methods: We reviewed the literature to summarize the epidemiology, clinical manifestations, diagnosis, treatment, and future directions of cCMV. Results: The best way to diagnose the infection is with polymerase chain reaction of urine or saliva within 3 weeks after birth, followed by a repeat confirmatory test if positive. Moderately to severely symptomatic neonates should be treated for 6 months with valganciclovir, and some practitioners also choose to treat infants who have isolated hearing loss only. Treatment is not recommended for asymptomatic infants. All infected infants should be screened for hearing loss and neurodevelopmental sequelae. Universal and targeted screening may be cost effective. Currently, no vaccine is commercially available, although multiple candidates are under study. Conclusion: Congenitally acquired cytomegalovirus is found in all communities around the world with a disease burden that is greater than many other well-known diseases. Advances are being made in prevention and treatment; however, improved awareness of the disease among clinicians and patients is needed.
Collapse
|
39
|
Tran C, Bennett MV, Gould JB, Lee HC, Lanzieri TM. Cytomegalovirus Infection among Infants in Neonatal Intensive Care Units, California, 2005 to 2016. Am J Perinatol 2019; 37:146-150. [PMID: 30895580 PMCID: PMC6754307 DOI: 10.1055/s-0039-1683958] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
AIM The main purpose of this article is to assess trends in cytomegalovirus (CMV) infection reported among infants in California neonatal intensive care units (NICUs) during 2005 to 2016. STUDY DESIGN The California Perinatal Quality Care Collaborative collects data on all very low birth weight (VLBW, birth weight ≤ 1,500 g) and acutely ill infants > 1,500 g, representing 92% of NICUs in California. We compared clinical characteristics and length of hospital stay among infants with and without reported CMV infection (CMV-positive viral culture or polymerase chain reaction). RESULTS During 2005 to 2016, CMV infection was reported in 174 VLBW infants and 145 infants > 1,500 g, or 2.7 (range: 1.5-4.7) and 1.2 (range: 0.8-1.7) per 1,000 infants, respectively (no significant annual trend). Among infants > 1,500 g, 12 (8%) versus 4,928 (4%) of those reported with versus without CMV infection died (p < 0.05). The median hospital stay was significantly longer among infants reported with versus without CMV infection for both VLBW infants (98 vs. 46 days) and infants > 1,500 g (61 vs. 14 days) (p < 0.001). CONCLUSION Reports of CMV infection remained stable over a 12-year period. Although we were not able to assess whether infection was congenital or postnatal, CMV infection among infants > 1,500 g was associated with increased mortality.
Collapse
Affiliation(s)
- Chinh Tran
- University of California Irvine, School of Medicine, Irvine, CA,California Perinatal Quality Care Collaborative (CPQCC), Stanford, CA
| | - Mihoko V. Bennett
- California Perinatal Quality Care Collaborative (CPQCC), Stanford, CA,Stanford University, School of Medicine, Stanford, CA
| | - Jeffrey B. Gould
- California Perinatal Quality Care Collaborative (CPQCC), Stanford, CA,Stanford University, School of Medicine, Stanford, CA
| | - Henry C. Lee
- California Perinatal Quality Care Collaborative (CPQCC), Stanford, CA,Stanford University, School of Medicine, Stanford, CA
| | - Tatiana M. Lanzieri
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA
| |
Collapse
|
40
|
Mussi-Pinhata MM, Yamamoto AY, Aragon DC, Duarte G, Fowler KB, Boppana S, Britt WJ. Seroconversion for Cytomegalovirus Infection During Pregnancy and Fetal Infection in a Highly Seropositive Population: "The BraCHS Study". J Infect Dis 2018; 218:1200-1204. [PMID: 29868783 PMCID: PMC6129109 DOI: 10.1093/infdis/jiy321] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022] Open
Abstract
We determined the risk of seroconversion in seronegative pregnant women living in a high seroprevalence population. Cytomegalovirus (CMV)-immunoglobulin G reactivity was determined at the 1st trimester in all women and sequentially for seronegative women. A total of 1915 of 1952 (98.1%; 95% confidence interval [CI], 97.4%-98.7%) women were seropositive, and 36 (1.8%; 95% CI, 1.3%-2.6%) were seronegative. Five of the 36-seronegative women seroconverted for a cumulative rate of 13.9% (95% CI, 4.8%-30.6%). Congenital CMV infection was diagnosed in 1 of 36 infants (2.8%; 95% CI, 0.5%-63.9%) born to seronegative women compared with 8 of 1685 (0.5%; 95% CI, 0.2%-1.0%) infants born to seropositive mothers. Even with a high risk of primary infection in seronegative women, most CMV-infected infants were born to women with pre-existing seroimmunity.
Collapse
Affiliation(s)
| | | | - Davi C Aragon
- Departments of Pediatrics, University of São Paulo, Brazil
| | - Geraldo Duarte
- Departments of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Karen B Fowler
- Department of Pediatrics, The University of Alabama at Birmingham
| | - Suresh Boppana
- Department of Pediatrics, The University of Alabama at Birmingham
| | - William J Britt
- Department of Pediatrics, The University of Alabama at Birmingham
| |
Collapse
|
41
|
Britt WJ. Maternal Immunity and the Natural History of Congenital Human Cytomegalovirus Infection. Viruses 2018; 10:v10080405. [PMID: 30081449 PMCID: PMC6116058 DOI: 10.3390/v10080405] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) is the most common viral infection of the developing fetus, and a significant cause of neurodevelopmental abnormalities in infants and children. Congenital HCMV infections account for an estimated 25% of all cases of hearing loss in the US. It has long been argued that maternal adaptive immune responses to HCMV can modify both the likelihood of intrauterine transmission of HCMV, and the severity of fetal infection and risk of long term sequelae in infected infants. Over the last two decades, multiple studies have challenged this paradigm, including findings that have demonstrated that the vast majority of infants with congenital HCMV infections in most populations are born to women with established immunity prior to conception. Furthermore, the incidence of clinically apparent congenital HCMV infection in infants born to immune and non-immune pregnant women appears to be similar. These findings from natural history studies have important implications for the design, development, and testing of prophylactic vaccines and biologics for this perinatal infection. This brief overview will provide a discussion of existing data from human natural history studies and animal models of congenital HCMV infections that have described the role of maternal immunity in the natural history of this perinatal infection.
Collapse
Affiliation(s)
- William J Britt
- Departments of Pediatrics, Microbiology, and Neurobiology, University of Alabama School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|