Next Article in Journal
Transcription of Endogenous Retroviruses: Broad and Precise Mechanisms of Control
Next Article in Special Issue
The Effects of Viral Infections on the Molecular and Signaling Pathways Involved in the Development of the PAOs
Previous Article in Journal
SARS-CoV-2 Modulation of HIV Latency Reversal in a Myeloid Cell Line: Direct and Bystander Effects
Previous Article in Special Issue
Polyploid Giant Cancer Cells: A Distinctive Feature in the Transformation of Epithelial Cells by High-Risk Oncogenic HCMV Strains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Burden of Congenital CMV Infection: A Narrative Review and Implications for Public Health Interventions

1
Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy
2
Infectious and Tropical Diseases Unit, Padua University Hospital, 35126 Padua, Italy
3
Department of Women’s and Children’s Health, Gynecological and Obstetric Clinic, Padua University Hospital, 35126 Padua, Italy
*
Author to whom correspondence should be addressed.
Co-last authors.
Viruses 2024, 16(8), 1311; https://doi.org/10.3390/v16081311
Submission received: 19 July 2024 / Revised: 14 August 2024 / Accepted: 15 August 2024 / Published: 17 August 2024
(This article belongs to the Special Issue 65-Year Anniversary of the Discovery of Cytomegalovirus)

Abstract

:
Cytomegalovirus causes the most common congenital infection worldwide. With most infants asymptomatic at birth, the few affected may present with variable clinical scenarios, from isolated hearing loss to severe neurologic impairment. Public health interventions include all actions at the health system, community, and individual levels that aim at reducing the burden of congenital Cytomegalovirus. This review examines the literature on maternal and neonatal screening programs in light of current evidence for treatment and the development of vaccines against Cytomegalovirus. Potential biases and benefits of these interventions are outlined, with the objective of increasing awareness about the problem and providing readers with data and critical tools to participate in this ongoing debate.

1. Introduction

The burden of congenital CMV infection (cCMV) is related to the sequelae of fetal invasion, possibly leading to irreversible complications. In screened populations, 40 up to 58% of symptomatic and 13.5% of asymptomatic infants at birth develop permanent sequelae [1]. Neurologic sequelae, including sensorineural hearing loss (SNHL), are mostly limited to the acquisition of primary infection during the first trimester of pregnancy [2,3,4], and symptomatic newborns seem at higher risk for developing delayed-onset SNHL [5]. Although treatment for cCMV has been used for more than 20 years, evidence is still weak, with data coming from a few clinical trials (from the Collaborative Antiviral Study Group—CASG) and observational studies [6,7,8,9]. Based on current evidence, treatment is considered for symptomatic infected children with severe disease or with at least one organ involved [10]. In Europe, no treatment is indicated in asymptomatic children or mild disease cases [11]. In fact, all treatment studies excluded asymptomatic children, so no recommendation is available for this category. Given the lack of evidence, some European experts suggest treating isolated SNHL cases. In the same way, weak data denote antiviral utilization for delayed-onset SNHL [12,13], with some ongoing trials [14,15]. Regarding the treatment of pregnant mothers, no evidence strongly indicates the need for antiviral therapy. However, valacyclovir therapy of periconceptional or first-trimester primary CMV infection seems to reduce vertical transmission and neonatal infections [16]. Hyperimmune CMV globulin seem to have a role in decreasing the inflammatory response to CMV and subsequent tissue damage [17].
Being the most widespread congenital infection, several public health interventions aiming at reducing the burden of disease have been proposed (or rejected) by health systems and public authorities, leaving space for wide debates still without a clear answer. Public intervention should include the epidemiology and surveillance of the disease, outreach to the population of interest (pregnant women), screening programs, health teaching, vaccination plans, and policy development (Figure 1). The aim of this review is to critically appraise public health interventions reported in the existing literature to reduce the burden of cCMV, with a special focus on neonatal screening studies and vaccine development.

2. Materials and Methods

To explore cCMV public awareness strategies, a literature search on PubMed was conducted between January 2000 and December 2023 and on national and international society websites in 2024. This search was restricted to original English-language studies and websites.
Concerning neonatal screening, studies focusing solely on epidemiological aspects or on the performance of different diagnostic methods were excluded. Additionally, studies where the targeted population was identified based only on maternal history or neonatal clinical suspicion of infection were excluded. It is reasonable to assume that these infants would be tested regardless and thus would not benefit from a screening program.
Vaccine studies for CMV were included if they involved the population of interest (adolescents, children, and women of childbearing age). Vaccine studies on transplant recipients and phase 1 trials were excluded.

3. Results

3.1. Awareness and Education in Pregnancy

The greatest risk to pregnant women is exposure to the urine or saliva of young children. Information on CMV disease in pregnancy would properly engage the women in prevention strategies, strengthening hygienic precautions that will reduce the possibility of CMV infections among others [19,20,21,22]. Physicians and obstetricians should raise awareness and provide education, these actions still being largely unpracticed [23]. A systematic review by Barber et al. on the pregnancy prevention of CMV acquisition through hygiene-based behavioral interventions showed that preventive and hygienic measures are acceptable to pregnant women and have the potential to reduce the risk of infection during pregnancy [24]. In the latest update of the National Institute for Health and Care Excellence (NICE) guidelines for antenatal care, it is recommended that at the first antenatal appointment, women are provided with information on infections that can impact pregnancy, including CMV, and that their personal risk factors and concerns (such as occupation and living with other children) are discussed [25].
The Center for Disease Control and Prevention (CDC) offers a dedicated website providing information to parents about the disease and its prevention, with fact sheets and an infographic [26]. CDC since 2020 is promoting a CMV Awareness Month to increase awareness.
In the UK, the “CMV Action” organization promotes knowledge, supports families, and promotes scientific advances in the field of congenital CMV [27]. Such great initiatives are still rare and are of paramount importance in terms of general awareness.

3.2. Maternal Screening

The identification of CMV infection during pregnancy can cause significant maternal anxiety because of the uncertainty of what the significance of the disease will be and leads to more testing and the possible (unnecessary) termination of pregnancy [28]. Practices about maternal screening are heterogeneous in the absence of an international consensus and recommendations [29]. Several considerations need to be carried out when balancing the feasibility of maternal serologic screening. First of all, almost half of congenital infections occur as non-primary maternal infection with CMV rather than primary infection, even if the risk of vertical transmission is lower in this case (<3.5% in CMV-seropositive mothers versus 32% during primary infection [30]). Reinfection is, however, the main modality of acquiring cCMV in high-seroprevalence populations, which are mostly low–middle income countries [31]. In the United States of America, only 25% of infants with cCMV were attributable to a primary maternal infection, with three quarters of cases born to immune mothers [32]. Secondly, invasive testing procedures (amniotic fluid PCR and viral culture combined) yield almost 100% positive and negative predictive values for congenital infection [20], but an association between viral loads and symptomatic infection at birth is debated, with no clear evidence [33]. If reinfection is at a lower risk for mother-to-child transmission, infants born to immune mothers are still at risk of symptomatic infection and SNHL. This was shown in a Brazilian prospective study (with a 96.7% CMV-immune population rate), where 60% of SNHL affected infants were born to a mother with non-primary CMV infection [34]. Maternal serologic screening alone therefore misses a considerable part of vertical infections and affected children. With these uncertainties, guidelines until now have suggested against the universal screening of CMV during pregnancy [29]. However, the recently published consensus from the European Congenital Cytomegalovirus Initiative made important recommendations on treatment options for pregnant women with primary CMV infection, suggesting valacyclovir therapy and, in some instances, considering CMV hyperimmune globulin. These recommendations are based on available studies. Two randomized controlled trials on the use of CMV hyperimmune globulin during pregnancy did not show a reduction in the vertical transmission of the infection or clinical outcomes at birth [35,36]. However, in a trial carried out by Revello, a reduction in vertical transmission from 40% to 30% was observed, even if not reaching significance because of the limited sample size. On the other hand, the utilization of hyperimmune globulin was associated with an increased incidence of obstetrical adverse effects [35]. Some earlier data suggest the potential role of CMV hyperimmune globulin in reducing the risk of poor clinical outcomes during infant follow-up [37,38,39]. In the same way, a retrospective database study showed that high-dose hyperimmune globulin administration was associated with a lower rate of fetal infection [40]. In recent years, studies on valacyclovir have shown promising results in reducing the rate of vertical transmission [16,41,42,43]. Other antivirals have shown ex vivo efficacy in first-trimester placental trophoblast cell cultures and third-trimester placental explant histocultures [44]. With these treatment options, maternal screening in the first trimester becomes crucial to identify women who would most likely benefit from treatments and to enhance hygiene measures and education for seronegative women [17].

3.3. Neonatal Screening

The screening of neonates would identify all children suffering from this condition. Available methods (PCR on saliva and urine) are, in fact, highly sensitive and specific. Universal screening for CMV at birth is not currently included in legislation in most countries. A debate among experts was raised in recent decades due to the potential benefit of this action (diagnosis, treatment, and follow-up), which was counter-balanced by the fact that most infections are asymptomatic and evidence for treatment does not demonstrate a strong benefit [9].
Many countries, regarding law indications or internal local practice, are adopting targeted CMV screening for infants failing the neonatal hearing screening (NHS). Currently, many countries adopt NHS [45] through evoked otoacoustic emissions (OAEs) or an automated measurement of auditory brainstem responses (ABRs), recommended for all infants before the age of three months in the United States, the United Kingdom, and Europe [46].

3.3.1. Universal Screening

The studies included are summarized in Table 1.
Universal screening would diagnose asymptomatic babies, including isolated SNHL or asymptomatic abnormal brain findings. In the most numerous series from Fowler (USA), 8% of cCMV cases were found with isolated SNHL [47]. Other smaller studies obtained variable percentages, from 0 to 10%. In the study by Lorenzoni et al., universal screening was performed on premature and small-for-gestational-age newborns, revealing an increased incidence of cCMV and isolated SNHL (17%) [48]. The treatment and clinical outcomes of these children are largely unreported in the series analyzed: only four studies reported treatment and short-term outcomes. For this reason, coupled with the uncertainty of valganciclovir efficacy in asymptomatic infants, the long-term clinical significance of universal screening is hard to estimate. Larger, prospective, and longer studies of coupled universal screening with therapeutic trials would further clarify the benefit.
Table 1. Summary of studies included on universal screening for congenital CMV (cCMV). SNHL: sensorineural hearing loss; PCR: polymerase chain reaction; LO-SNHL: late-onset SNHL; SGA: small gestational age; OAEs: otoacoustic emissions; ABRs: automated measurement of auditory brain-stem responses; FP: false positive; DBS: dried blood spot; CNS: central nervous system; HL: hearing loss; US: ultrasound; gw: gestational week; n.a.: not applicable.
Table 1. Summary of studies included on universal screening for congenital CMV (cCMV). SNHL: sensorineural hearing loss; PCR: polymerase chain reaction; LO-SNHL: late-onset SNHL; SGA: small gestational age; OAEs: otoacoustic emissions; ABRs: automated measurement of auditory brain-stem responses; FP: false positive; DBS: dried blood spot; CNS: central nervous system; HL: hearing loss; US: ultrasound; gw: gestational week; n.a.: not applicable.
Author, Year, Ref, CountryStudy Design and Time of EnrollmentPopulationMethod of Universal ScreeningResultsSymptomatic Newborns at Birth (Other than Isolated HL)Isolated HL Confirmed CasesTreatmentOutcomeComments
Schlesinger, 2005 [49]
Israel
Multicenter prospective study
From May 98 to August 99
Live-born infantsPCR on urine14 diagnosed/2000 screened for CMV2 symptomatic (microcephaly, hepatitis)no HL foundn.a.n.a.This study did not identify any isolated SNHL, no information about follow-up for LO-SNHL was provided.
Lorenzoni, 2013 [48]
Italy
Monocenter prospective
From January 2012 to July 2013
Premature newborns (<37 gw) and SGA term infants (weight <3rd percentile)PCR on urine12 (10 preterms, 2 SGA)/383 screened/504 premature or SGA1 preterm (lissencephaly)2n.a.n.a.Increased incidence of cCMV and isolated SHL (17%) in this populations.
Barkai, 2014 [50]
Israel
Single-center prospective study
From May 2011 to May 2012
Live-born infantsPCR on saliva confirmed by urine48 cCMV/9845 screened for CMV/10,137 live-born infants014 infants1 LO-SNHL at 3 months of ageIncidence of neonatal hearing loss: 2%. The infant diagnosed with HL passed the OAE screening and was confirmed on ABR.
Fowler, 2017 [47]
USA
Multicenter prospective
From March 2007 to March 2012
Live-born infantsPCR on saliva or DBS443 cCMV identified out of 100,332 testedn.a.35 confirmedn.a.n.a.Incidence of neonatal hearing loss: 8%. The lack of CMV confirmation on urine may give some FP patients. 15 cCMV cases with confirmed SNHL passed the OAE screening.
Dar, 2017 [51]
India
Multicenter, prospective study
From December 2010 to May 2012
Live-born infantsPCR on saliva20 diagnosed/1720 screened12n.a.n.a.Incidence of neonatal hearing loss: 10%.
1 out of 2 neonates with cCMV and SNHL passed the initial HS. The lack of CMV confirmation on urine may give some FP patients.
Yamamoto, 2020 [52]
Brazil
Multicenter, prospective study
From September 2013 to April 2017
Live-born infantsPCR on saliva, confirmed on urine68 diagnosed/11,900 tested447Neonatal SNHL: between the 4 isolated SNHL, 1 progressed and 1 was stable at 18–48 months at follow-up For the 4 symptomatic babies, all had SNHL, one progressive and 3 stable at follow-up. For the other 49 cases, no late-onset HL was detected at a median 36-month follow-upIncidence of neonatal hearing loss: 5.8%.
1 neonate with cCMV and SNHL passed the initial HS. Targeted screening would have missed 12.5% of infants with SNHL.
Yamada, 2020 [53]
Japan
Multicenter prospective study
From November 2009 to March 2018
Live-born infantsPCR on urine56 diagnosed/11,736 tested for CMV194n.a.Between the 4 isolated SNHL, 2 normal development and 2 mild sequele.The incidence of isolated SNHL in this population is 7.1%.
Blazquez-Gamero, 2020 [54]
Spain
Prospective, monocenter
From February 2017 to February 2018
Live-born infantsPCR on saliva, confirmed by urine15 positive out of 3190 tested20n.a.No infants (13 available at follow-up) developed SNH at 25 monthsThe incidence of isolated SNHL in this population is 0%.
Letamendia-Richard, 2022 [55]
France
Monocenter, retrospective
From single unit, 2016–2020
Live-born infantsPCR on saliva at birth, confirmed by urine63 confirmed infections/15,341 tested/15,649 live-born infants8 infants small for gestational age, no one with HL1n.a.n.a.The child with isolated SNHL had hepatomegaly at prenatal US and his mother had known seroconversion, so it would have been diagnosed without intervention.
Chiereghin, 2022 [56]
Italy
Multicenter prospective study
From February 2019 to July 2020
Live-born infantsPCR on saliva confirmed by urine21 confirmed cCMV/3151 screened for CMV1 case with severe CNS disease and HL12 (6 months)1 asymptomatic infant developed LO-SNHL at 5 months of ageIncidence of neonatal hearing loss: 4.7%.
No information regarding hearing screening test.

3.3.2. Targeted Screening

The studies included are reported in Table 2.
Regarding targeted screening studies, they were mainly retrospective experiences from centers locally performing cCMV screening for infants failing NHS. The primary limitation in understanding the potential of screening is that most studies did not exclude symptomatic infants or those with known or suspected maternal infection during pregnancy. Even when considering isolated cases of sensorineural hearing loss (SNHL), these could still originate from mothers with known infections and be diagnosed without screening. Clinical outcomes are available from only two studies [57,58]. Furthermore, concerns exist regarding the sensitivity of hearing tests performed at birth. False negatives could result in missing cases of neonatal hearing loss, thereby depriving these children of the benefits of both pharmacological and non-pharmacological therapies and precluding early intervention for those with delayed-onset SNHL.
A prospective study by Fowler enrolled 443 children with cCMV identified through universal neonatal screening over 5 years: 35 newborns were diagnosed with SNHL by a confirmatory hearing test [47]. Of these 35 newborns, 15 (43%) passed their hearing screening. In this study, therefore, targeted screening missed 43% of cCMV with a neonatal-onset hearing deficit. One neonate out of two and one neonates out of four with confirmed SNHL passed the HS in the series from Dar (India) and Yamamoto (Brazil), respectively [51,52].
Another aspect that emerged during the search is that some infants classified as symptomatic within screening programs are, in practice, asymptomatic at birth. They are deemed symptomatic only due to laboratory abnormalities or abnormal cerebral findings that would otherwise remain undiagnosed [49,53,59].
In this context, it is worth questioning whether an asymptomatic newborn is truly ‘asymptomatic’. The significance of nonspecific ultrasound or MRI findings, such as isolated lenticulostriatal vasculopathy, remains uncertain, and the long-term outcomes associated with these findings are still unknown [11]. The follow-up of diagnosed cases would lead to the development of a scoring system aimed at more accurately quantifying the risk of adverse outcomes. This would expand the knowledge base to facilitate studies on treatment.
From the scant clinical data and follow-up availability in the search, it is hard to extrapolate a long-term clinical outcome of targeted and universal screening for cCMV. This would be clarified in the occurrence of clear treatment indications and evidence of efficacy. However, mainly due to the paucity of data and the fact that most cCMV cases are mild, large trials of treatment are missing, and recommendations are weak.
Table 2. Summary of studies included on targeted screening for congenital CMV. SNHL: sensorineural hearing loss; PCR: polymerase chain reaction; NHS: neonatal hearing screening; LO-SNHL: late-onset SNHL; OAE: otoacoustic emission; ABR: auditory brain-stem response; HIV: human immunodeficiency virus; HL: hearing loss; DBS: dried blood spot; CNS: central nervous system; n.a.: not applicable.
Table 2. Summary of studies included on targeted screening for congenital CMV. SNHL: sensorineural hearing loss; PCR: polymerase chain reaction; NHS: neonatal hearing screening; LO-SNHL: late-onset SNHL; OAE: otoacoustic emission; ABR: auditory brain-stem response; HIV: human immunodeficiency virus; HL: hearing loss; DBS: dried blood spot; CNS: central nervous system; n.a.: not applicable.
Author, Year, Ref, CountryStudy Design and Time of EnrollmentPatientMethod of Targeted ScreeningResultsSymptomatic Newborns at Birth (Other than Isolated HL)Isolated HL Confirmed CasesTreatmentOutcomes of cCMV CasesComments
Stehel, 2008 [60]
Texas (USA)
Monocenter, retrospective
From September 1999 to August 2004
Patients failing HS, mother infected with HIV, clinical or lab signs suggestivePCR on urine24 confirmed/483 screened/572 failing HS. 98 n.a.n.a.The inclusion criteria for screening were not stringent. It was hard to predict if the screening would be different to normal clinical practice.
Williams, 2014 [61]
UK
Multicenter prospective
From August 2010 to October 2012
Infants < 22 days old failing NHS. Known cCMV excludedPCR on urine or saliva6 diagnosed/407 screened/411 recruited after failing NHSn.a.3n.a.n.a.Clinical data and outcome missing.
Kawada, 2015 [62]
Japan
Prospective study
From January 2011 to December 2013
Infants failing NHSPCR on saliva or urine6 confirmed out fo 127 failing NHS06valgancyclovir for 6 weeksonly 1 out of 6 improved at 1-year follow-upValganciclovir did not show to significantly improved hearing function.
Roth, 2017 [63]
Israel
Single-center retrospective study
From 2014 to 2015
Infants failing NHSPCR on saliva confirmed by urine4 confirmed cCMV/180 tested for CMV/200 failing NHS23n.a.n.a.Targeted screening identified 1 child (out of 200 failing NHS) who needed treatment. Outcomes missing.
Diener, 2017 [58]
Utah (USA)
Retrospective multicenter
From 2013 to 2015
Live-born infants failing NHS. Infants with suggestive symptoms were excludedPCR on saliva, confirmed on urine14 diagnosed/314 screened for CMV/509 failing HS06n.a.n.a.No information on follow-up and outcome.
Rawlinson, 2018 [59]
Australia
Monocenter, retrospective study
From October 2009 to Oct 2016
Infants failing HS and formal audiological testing (ABR)PCR on saliva up to 2011, after 2011, positivity on saliva was confirmed on urine19 diagnosed/323 screened/502 infants with confirmed HL 4156 out of 19 (only 4 started within the first month of life)n.a. No clinical outcome, no follow-up. Symptomatic infants were not excluded from the study (4 out of 19 confirmed) and were reasonably diagnosed without this intervention.
Beswick, 2019 [64]
Australia
Multicenter, retrospective
From August 2014 to April 2016
Neonates failing NHS (twice OAE)PCR on saliva, confirmed by urine and blood3 diagnosed out of 234 screened/347 failing NHS021, valganciclovirn.a. Intervention allowed diagnosis and treatment of one otherwise asymptomatic infant. No clinical outcome provided.
Pellegrinelli, 2019 [65]
Italy
Observational single-center study
From 2014 to 2018
Infants failing NHS (AOE)PCR on DBS5 DBS tested positive/82 DBS screened/89 failing NHSn.a.5n.a.n.a.DBS method may have missed some CMV diagnoses.
Ronner, 2021 [57]
Massachusetts (USA)
Monocenter, retrospective chart review,
From 2013 to 2020 (screening from 2015). Targeted screening was implemented in 2015 for 2 nurseries, from 2016 to all nurseries
Infants failing NHSPrimary PCR on saliva8 confirmed/528 tested for CMV/891 failing NHSn.a.6valganciclovirhearing stable in 3, progressed in 2, improved in 1.Hearing function improved in 1 patient out of 6 diagnosed and treated for isolated SNHL. Not specified if symptomatic infants were excluded from the study.
Khi Chung, 2022 [66]
Netherlands
National, prospective observational
From 2012 to 2016
Infants failing NHS (three rounds: two OAEs, one ABR)PCR on DBS54 confirmed/1374 DBS screened/1381 infants failing NHSn.a.n.a. (48 infants had confirmed HL, but other concurrent symptoms were not excluded or specified in the study)n.an.a.Symptomatic children were not excluded and granular data about clinical scenario were not provided.
Fourgeaud, 2022 [67]
France
Multicenter, prospective study
From 2014 to 2017
Newborns failing NHS (twice OAE in 3 centers, twice ABR in 2 centers)PCR on saliva
Confirmatory test on saliva and blood
2 confirmed/231 screened for CMV/236 failing NHS n.a.n.a. (2 cases of HL but no information on other symptoms)valganciclovirn.a.No granular data about clinical scenario of confirmed cases. Not specified if symptomatic infants were excluded from the study.
Webb, 2022 [68]
Australia
Prospective, multicenter
From June 2019 to March 2020
Infants failing NHSPCR on saliva, confirmed on urine and plasma1 positive out of 96 tested01valganciclovir started at 32 days of life for 6 monthsn.a.Good feasibility and acceptability.
Zhang, 2023 [69]
Japan
Single-center observational prospective study
From October 2018 to October 2021
Newborns with suggestive perinatal conditions, including failing NHS (twice ABR)PCR on urine1 positive out of 12 failing NHS, 1 positive screened because of abnormal CNS findings, 1 positive screened for suspected maternal infection during pregnancyn.a.12 treated with valgancyclovirn.a.No clinical outcome.

3.4. Vaccinations

Key factors considered in the vaccine prioritization strategies include disease burden, vaccine effectiveness and safety, the feasibility of additional recommendations in the context of the existing vaccination schedule, equity of access, and whether vaccination is a good use of public funds [70].
Cytomegalovirus is not highly contagious, and it is believed that protecting 50–60% of the population could lead to viral elimination, making vaccination cost effective [71]. Several efforts are currently focused on vaccine development, with some aimed at preventing infection in transplant patients and others targeting the prevention of congenital infection [72].
Regarding cCMV disease, vaccination efforts should focus on protecting women of childbearing age from acquiring the infection. To achieve this goal, the target population should primarily include young women, adolescents, and young children as they are the main sources of infection for pregnant mothers [73].
Here is a summary of vaccines studied in the population of interest, which includes adolescents, children, and women of childbearing age (Table 3). Studies on transplant recipients and phase 1 trials are excluded from this summary.
Three CMV vaccines have reached phases 2 and 3 in healthy individuals. The MF59-adjuvated recombinant CMV glycoprotein B vaccine was studied in two phase 2 trials in post-partum seronegative women and adolescents [74,75]. While in the first study, the vaccine’s effectiveness in preventing the infection in post-partum women seemed good [74], in the second study on adolescents, it was not significant. No phase 3 trial is ongoing. V160 is a whole-virus vaccine derived from the live-attenuated AD169 strain. A recently published phase 2 trial by Das was terminated early because of the futility in preventing the primary CMV infection in vaccinated women compared to the placebo [76]. The mRNA-1647 vaccine seem to induce higher neutralization and antibody-dependent cellular cytotoxicity responses compared to the gB/MF59 vaccine [77]. A phase 2 trial of the CMV mRNA-1647 vaccine was terminated in 2023 [78]. The results are not available, but in 2020, the company declared a positive seven-month interim safety and immunogenicity analysis. The following phase 3 trial is expected to evaluate the efficacy of the vaccine in a larger population of healthy individuals at risk of having early post-vaccination contact with young children (NCT05085366) [79]. Moreover, 7545 patients were enrolled, but the results have still not been reported.
The immunogenicity of CMV vaccines has not been studied in young children. This is due to the lack of positive effectiveness data on adults, unknown duration of immunity, and mildness of the disease in infancy [73].
Table 3. Summary of studies (published or ongoing) included on vaccination for CMV. Nab: neutralizing antibody.
Table 3. Summary of studies (published or ongoing) included on vaccination for CMV. Nab: neutralizing antibody.
VaccineAuthor, Year or Trial ID NumberStudy DesignPopulationOutcomeEnrolment TimeResults
bB-MF59: MF59 adjuvated recombinant CMV envelope glicoproteinB subunitPass, 2009 [74]Phase 2, placebo-controlled, randomized, double-blind trial.Post-partum, seronegative women, aged 14–40 years and healthy.Effectiveness in preventing CMV infection during a 42-month periodAugust 1999 to April 2006464 subject enrolled. Vaccine recipients were more likely to remain uninfected than placebo recipients (p = 0.02).
bB-MF59: MF59 adjuvated recombinant CMV envelope glicoproteinB subunitBernstein, 2017 [75]Phase 2, multicenter, randomized, double-blind, controlled study.Healthy adolescent females.Effectiveness in preventing CMV infection, immunogenicity, safety.June 2006–June 2013402 subjects enrolled. CMV infection occurred without significant differences between vaccinated and control individuals.
V160: whole-virus vaccine that is derived from the live-attenuated AD169 strainDas, 2023 [76]Phase 2b, multicenter, randomized, double-blind, placebo-controlled study.Healthy, CMV-seronegative, non-pregnant, 16–35-year-old women of childbearing potential with exposure to children aged 5 years or younger.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; vaccine safety.April 2018–August 20192220 enrolled. 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. The study was terminated due to futility.
mRNA-1647NCT04232280 [78]Phase 2, randomized, observer-blind, placebo-controlled, dose-finding trial. Part 1: to inform the selection of the middle dose level for further development. Part 2: to further evaluate the safety and immunogenicity of the middle dose level of mRNA-1647 vaccine or placebo.Healthy participants seropositive or seronegative, males or females, 18 to 40 years of age.Safety, immunogenicity (NAb titers)September 2020–April 2023315 subjects enrolled. No results reported.
mRNA-1647Ongoing trial NCT05085366 [79]Phase 3, randomized, observer-blind, placebo-controlled study.Participants aged ≥20 years, has or anticipates having direct exposure within 7 months after the planned first dose (in the home, socially, or occupationally) to at least 1 child ≤5 years of age. Enrollment estimated 6900 subjects.Efficacy (seroconversion from a negative to a positive result) in females and in all participants. Safety.October 2021–April 2024 Enrolled 7454 patients, no results reported.

4. Final Considerations and Future Prospectives

Despite being a widespread disease with a substantial body of literature addressing congenital CMV, evidence regarding the effectiveness of various interventions remains weak. The absence of effective treatment options for asymptomatic children or LO-SNHL complicates the implementation of screening by health systems. This underscores the need, at higher levels, to weigh the allocation of public funds against numerous competing health priorities. The promotion of trials during pregnancy is primarily hindered by ethical concerns, which limits the advancement of therapeutic strategies to prevent fetus damages by CMV infection. It is crucial to enhance education among pregnant women and increase awareness among healthcare providers about congenital CMV at the community, local, and national organizational levels. Early education remains, in fact, the main useful strategy to prevent primary maternal infection during pregnancy, and awareness initiatives should be supported by health systems.
To date, vaccine research has predominantly focused on immunosuppressed patients, and data regarding healthy women do not demonstrate significant effectiveness. The network between gynecologists, obstetricians, infectious disease specialists and pediatric infectious disease teams for managing mother–infant pairs should be strengthened and established as the standard of care for cCMV by public health systems. In this view, maternal and neonatal universal screening emerges as a possible way forward, along with continued research on vaccines. It would ease the understanding of risk factors for infection and sequelae (such as prematurity, low birth weight, and brain imaging anomalies), the development of scoring systems, and the optimization of follow-up modalities. This would allow researchers to construct a basis for new therapeutic trials.

Author Contributions

Conceptualization, C.L. and D.D.; methodology, C.L., D.D. and G.B.; research, C.L. and G.S.; data curation, C.L. and G.S.; writing—original draft preparation, C.L. and G.S.; writing—review and editing, G.B., D.D. and O.R.; supervision, S.C., S.V., E.C., D.D. and O.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

All data are included in the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dollard, S.C.; Grosse, S.D.; Ross, D.S. New Estimates of the Prevalence of Neurological and Sensory Sequelae and Mortality Associated with Congenital Cytomegalovirus Infection. Rev. Med. Virol. 2007, 17, 355–363. [Google Scholar] [CrossRef] [PubMed]
  2. Faure-bardon, V.; Magny, J.; Parodi, M.; Couderc, S.; Garcia, P.; Maillotte, A.; Benard, M. Sequelae of Congenital Cytomegalovirus Following Maternal Primary Infections Are Limited to Those Acquired in the First Trimester of Pregnancy. Clin. Infect. Dis. 2019, 69, 1526–1532. [Google Scholar] [CrossRef] [PubMed]
  3. Gindes, L.; Teperberg-Oikawa, M.; Sherman, D.; Pardo, J.; Rahav, G. Congenital Cytomegalovirus Infection Following Primary Maternal Infection in the Third Trimester. BJOG Int. J. Obstet. Gynaecol. 2008, 115, 830–835. [Google Scholar] [CrossRef] [PubMed]
  4. Enders, G.; Daiminger, A.; Bäder, U.; Exler, S.; Enders, M. Intrauterine Transmission and Clinical Outcome of 248 Pregnancies with Primary Cytomegalovirus Infection in Relation to Gestational Age. J. Clin. Virol. 2011, 52, 244–246. [Google Scholar] [CrossRef] [PubMed]
  5. Riga, M.; Korres, G.; Chouridis, P.; Naxakis, S.; Danielides, V. Congenital Cytomegalovirus Infection Inducing Non-Congenital Sensorineural Hearing Loss during Childhood; a Systematic Review. Int. J. Pediatr. Otorhinolaryngol. 2018, 115, 156–164. [Google Scholar] [CrossRef]
  6. Nigro, G.; Scholz, H.; Bartmann, U. Ganciclovir Therapy for Symptomatic Congenital Cytomegalovirus Infection in Infants: A Two-Regimen Experience. J. Pediatr. 1994, 124, 318–322. [Google Scholar] [CrossRef]
  7. Kimberlin, D.W.; Jester, P.M.; Sánchez, P.J.; Ahmed, A.; Arav-Boger, R.; Michaels, M.G.; Ashouri, N.; Englund, J.A.; Estrada, B.; Jacobs, R.F.; et al. Valganciclovir for Symptomatic Congenital Cytomegalovirus Disease. N. Engl. J. Med. 2015, 372, 933–943. [Google Scholar] [CrossRef]
  8. Whitley, R.J.; Cloud, G.; Gruber, W.; Storch, G.A.; Demmler, G.J.; Jacobs, R.F.; Dankner, W.; Spector, S.A.; Starr, S.; Pass, R.F.; et al. Ganciclovir Treatment of Symptomatic Congenital Cytomegalovirus Infection: Results of a Phase II Study. National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group. J. Infect. Dis. 1997, 175, 1080–1086. [Google Scholar] [CrossRef]
  9. Oliver, S.E.; Cloud, G.A.; Sánchez, P.J.; Demmler, G.J.; Dankner, W.; Shelton, M.; Jacobs, R.F.; Vaudry, W.; Pass, R.F.; Soong, S.J.; et al. Neurodevelopmental Outcomes Following Ganciclovir Therapy in Symptomatic Congenital Cytomegalovirus Infections Involving the Central Nervous System. J. Clin. Virol. 2009, 46, 22–26. [Google Scholar] [CrossRef]
  10. Rawlinson, W.D.; Boppana, S.B.; Fowler, K.B.; Kimberlin, D.W.; Lazzarotto, T.; Alain, S.; Daly, K.; Doutré, S.; Gibson, L.; Giles, M.L.; et al. Congenital Cytomegalovirus Infection in Pregnancy and the Neonate: Consensus Recommendations for Prevention, Diagnosis, and Therapy. Lancet Infect. Dis. 2017, 17, e177–e188. [Google Scholar] [CrossRef]
  11. Luck, S.E.; Wieringa, J.W.; Blázquez-Gamero, D.; Henneke, P.; Schuster, K.; Butler, K.; Capretti, M.G.; Cilleruelo, M.J.; Curtis, N.; Garofoli, F.; et al. Congenital Cytomegalovirus a European Expert Consensus Statement on Diagnosis and Management. Pediatr. Infect. Dis. J. 2017, 36, 1205–1213. [Google Scholar] [CrossRef] [PubMed]
  12. Dorfman, L.; Amir, J.; Attias, J.; Bilavsky, E. Treatment of Congenital Cytomegalovirus beyond the Neonatal Period: An Observational Study. Eur. J. Pediatr. 2020, 179, 807–812. [Google Scholar] [CrossRef]
  13. Amir, J.; Attias, J.; Pardo, J. Treatment of Late-Onset Hearing Loss in Infants with Congenital Cytomegalovirus Infection. Clin. Pediatr. 2014, 53, 444–448. [Google Scholar] [CrossRef]
  14. Randomized Controlled Trial of Valganciclovir for Cytomegalovirus Infected Hearing Impaired Infants—Full Text View—ClinicalTrials.Gov. Available online: https://classic.clinicaltrials.gov/ct2/show/study/NCT03107871 (accessed on 27 November 2023).
  15. Study Details|Valganciclovir Therapy in Infants and Children With Congenital CMV Infection and Hearing Loss|ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/study/NCT01649869 (accessed on 16 November 2023).
  16. Chatzakis, C.; Shahar-Nissan, K.; Faure-Bardon, V.; Picone, O.; Hadar, E.; Amir, J.; Egloff, C.; Vivanti, A.; Sotiriadis, A.; Leruez-Ville, M.; et al. The Effect of Valacyclovir on Secondary Prevention of Congenital Cytomegalovirus Infection, Following Primary Maternal Infection Acquired Periconceptionally or in the First Trimester of Pregnancy. An Individual Patient Data Meta-Analysis. Am. J. Obstet. Gynecol. 2023, 230, 109–117.e2. [Google Scholar] [CrossRef] [PubMed]
  17. Nigro, G.; Muselli, M. Prevention of Congenital Cytomegalovirus Infection: Review and Case Series of Valaciclovir versus Hyperimmune Globulin Therapy. Viruses 2023, 15, 1376. [Google Scholar] [CrossRef]
  18. MNDOH Public Health Interventions (Population-Based). Minn. Dep. Health 2019, 2, 453–468.
  19. Demmler-Harrison, G.J. Congenital Cytomegalovirus: Public Health Action towards Awareness, Prevention, and Treatment. J. Clin. Virol. 2009, 46 (Suppl. S4), S1–S5. [Google Scholar] [CrossRef] [PubMed]
  20. Revello, M.G.; Tibaldi, C.; Masuelli, G.; Frisina, V.; Sacchi, A.; Furione, M.; Arossa, A.; Spinillo, A.; Klersy, C.; Ceccarelli, M.; et al. Prevention of Primary Cytomegalovirus Infection in Pregnancy. eBioMedicine 2015, 2, 1205–1210. [Google Scholar] [CrossRef]
  21. Adler, S.P.; Finney, J.W.; Manganello, A.M.; Best, A.M. Prevention of Child-to-Mother Transmission of Cytomegalovirus among Pregnant Women. J. Pediatr. 2004, 145, 485–491. [Google Scholar] [CrossRef] [PubMed]
  22. Vauloup-Fellous, C.; Picone, O.; Cordier, A.G.; Parent-du-Châtelet, I.; Senat, M.V.; Frydman, R.; Grangeot-Keros, L. Does Hygiene Counseling Have an Impact on the Rate of CMV Primary Infection during Pregnancy? Results of a 3-Year Prospective Study in a French Hospital. J. Clin. Virol. 2009, 46 (Suppl. S4), S49–S53. [Google Scholar] [CrossRef]
  23. Knowledge and Practices of Obstetricians and Gynecologists Regarding Cytomegalovirus Infection During Pregnancy—United States. 2007. Available online: https://www.cdc.gov/mmwr/preview/mmwrhtml/mm5703a2.htm (accessed on 25 November 2023).
  24. Barber, V.; Calvert, A.; Vandrevala, T.; Star, C.; Khalil, A.; Griffiths, P.; Heath, P.T.; Jones, C.E. Prevention of Acquisition of Cytomegalovirus Infection in Pregnancy through Hygiene-Based Behavioral Interventions: A Systematic Review and Gap Analysis. Pediatr. Infect. Dis. J. 2020, 39, 949–954. [Google Scholar] [CrossRef] [PubMed]
  25. NICE Guideline, Antenatal Care. 2021. Available online: www.nice.org.uk/guidance/ng201 (accessed on 27 November 2023).
  26. Resources for Pregnant Women and Parents|CDC. Available online: https://www.cdc.gov/cmv/resources/pregnant-women-parents.html (accessed on 27 November 2023).
  27. CMV Action—What Is CMV? Available online: https://cmvaction.org.uk/ (accessed on 27 November 2023).
  28. Périllaud-Dubois, C.; Belhadi, D.; Laouénan, C.; Mandelbrot, L.; Picone, O.; Vauloup-Fellous, C. Current Practices of Management of Maternal and Congenital Cytomegalovirus Infection during Pregnancy after a Maternal Primary Infection Occurring in First Trimester of Pregnancy: Systematic Review. PLOS ONE 2021, 16, e0261011. [Google Scholar] [CrossRef] [PubMed]
  29. Xie, M.; Tripathi, T.; Holmes, N.E.; Hui, L. Serological Screening for Cytomegalovirus during Pregnancy: A Systematic Review of Clinical Practice Guidelines and Consensus Statements. Prenat. Diagn. 2023, 43, 959–967. [Google Scholar] [CrossRef] [PubMed]
  30. Leruez-Ville, M.; Chatzakis, C.; Lilleri, D.; Blazquez-Gamero, D.; Alarcon, A.; Bourgon, N.; Foulon, I.; Fourgeaud, J.; Gonce, A.; Jones, C.E.; et al. Consensus Recommendation for Prenatal, Neonatal and Postnatal Management of Congenital Cytomegalovirus Infection from the European Congenital Infection Initiative (ECCI). Lancet Reg. Health-Eur. 2024, 40, 100892. [Google Scholar] [CrossRef] [PubMed]
  31. Stagno, S.; Pass, R.F.; Dworsky, M.E.; Henderson, R.E.; Moore, E.G.; Walton, P.D.; Alford, C.A. Congenital Cytomegalovirus Infection. N. Engl. J. Med. 1982, 306, 945–949. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, C.; Zhang, X.; Bialek, S.; Cannon, M.J. Attribution of Congenital Cytomegalovirus Infection to Primary versus Non-Primary Maternal Infection. Clin. Infect. Dis. 2011, 52, 11–14. [Google Scholar] [CrossRef]
  33. Davis, N.L.; King, C.C.; Kourtis, A.P. Cytomegalovirus Infection in Pregnancy. Birth Defects Res. 2017, 109, 336–346. [Google Scholar] [CrossRef]
  34. Yamamoto, A.Y.; Mussi-Pinhata, M.M.; Isaac, M.D.L.; Amaral, F.R.; Carvalheiro, C.G.; Aragon, D.C.; Da Silva Manfredi, A.K.; Boppana, S.B.; Britt, W.J. Congenital Cytomegalovirus Infection as a Cause of Sensorineural Hearing Loss in a Highly Immune Population. Pediatr. Infect. Dis. J. 2011, 30, 1043–1046. [Google Scholar] [CrossRef]
  35. Revello, M.G.; Lazzarotto, T.; Guerra, B.; Spinillo, A.; Ferrazzi, E.; Kustermann, A.; Guaschino, S.; Vergani, P.; Todros, T.; Frusca, T.; et al. A Randomized Trial of Hyperimmune Globulin to Prevent Congenital Cytomegalovirus. N. Engl. J. Med. 2014, 370, 1316–1326. [Google Scholar] [CrossRef]
  36. Hughes, B.L.; Clifton, R.G.; Rouse, D.J.; Saade, G.R.; Dinsmoor, M.J.; Reddy, U.M.; Pass, R.; Allard, D.; Mallett, G.; Fette, L.M.; et al. A Trial of Hyperimmune Globulin to Prevent Congenital Cytomegalovirus Infection. N. Engl. J. Med. 2021, 385, 436–444. [Google Scholar] [CrossRef]
  37. Visentin, S.; Manara, R.; Milanese, L.; Da Roit, A.; Forner, G.; Salviato, E.; Citton, V.; Magno, F.M.; Orzan, E.; Morando, C.; et al. Early Primary Cytomegalovirus Infection in Pregnancy: Maternal Hyperimmunoglobulin Therapy Improves Outcomes among Infants at 1 Year of Age. Clin. Infect. Dis. 2012, 55, 497–503. [Google Scholar] [CrossRef]
  38. Adler, S.P. Primary Maternal Cytomegalovirus Infection during Pregnancy: Do We Have a Treatment Option? Clin. Infect. Dis. 2012, 55, 504–506. [Google Scholar] [CrossRef]
  39. Nigro, G.; Adler, S.P.; La Torre, R.; Best, A.M. Passive Immunization during Pregnancy for Congenital Cytomegalovirus Infection. N. Engl. J. Med. 2005, 353, 1350–1362. [Google Scholar] [CrossRef]
  40. Nigro, G.; Adler, S.P.; Lasorella, S.; Iapadre, G.; Maresca, M.; Mareri, A.; Di Paolantonio, C.; Catenaro, M.; Tambucci, R.; Mattei, I.; et al. High-Dose Cytomegalovirus (CMV) Hyperimmune Globulin and Maternal CMV DNAemia Independently Predict Infant Outcome in Pregnant Women with a Primary CMV Infection. Clin. Infect. Dis. 2020, 71, 1491–1498. [Google Scholar] [CrossRef]
  41. Shahar-Nissan, K.; Pardo, J.; Peled, O.; Krause, I.; Bilavsky, E.; Wiznitzer, A.; Hadar, E.; Amir, J. Valaciclovir to Prevent Vertical Transmission of Cytomegalovirus after Maternal Primary Infection during Pregnancy: A Randomised, Double-Blind, Placebo-Controlled Trial. Lancet 2020, 396, 779–785. [Google Scholar] [CrossRef] [PubMed]
  42. Faure-Bardon, V.; Fourgeaud, J.; Stirnemann, J.; Leruez-Ville, M.; Ville, Y. Secondary Prevention of Congenital Cytomegalovirus Infection with Valacyclovir Following Maternal Primary Infection in Early Pregnancy. Ultrasound Obstet. Gynecol. 2021, 58, 576–581. [Google Scholar] [CrossRef]
  43. Egloff, C.; Sibiude, J.; Vauloup-Fellous, C.; Benachi, A.; Bouthry, E.; Biquard, F.; Hawkins-Villarreal, A.; Houhou-Fidouh, N.; Mandelbrot, L.; Vivanti, A.J.; et al. New Data on Efficacy of Valacyclovir in Secondary Prevention of Maternal–Fetal Transmission of Cytomegalovirus. Ultrasound Obstet. Gynecol. 2023, 61, 59–66. [Google Scholar] [CrossRef] [PubMed]
  44. Hamilton, S.T.; Marschall, M.; Rawlinson, W.D. Investigational Antiviral Therapy Models for the Prevention and Treatment of Congenital Cytomegalovirus Infection during Pregnancy. Antimicrob. Agents Chemother. 2021, 65, e01627-20. [Google Scholar] [CrossRef]
  45. Wroblewska-Seniuk, K.E.; Dabrowski, P.; Szyfter, W.; Mazela, J. Universal Newborn Hearing Screening: Methods and Results, Obstacles, and Benefits. Pediatr. Res. 2017, 81, 415–422. [Google Scholar] [CrossRef]
  46. Kennedy, C.R.; McCann, D.C.; Campbell, M.J.; Law, C.M.; Mullee, M.; Petrou, S.; Watkin, P.; Worsfold, S.; Yuen, H.M.; Stevenson, J. Language Ability after Early Detection of Permanent Childhood Hearing Impairment. N. Engl. J. Med. 2006, 354, 2131–2141. [Google Scholar] [CrossRef] [PubMed]
  47. Fowler, K.B.; Mccollister, F.P.; Sabo, D.L.; Shoup, A.G.; Owen, K.E.; Woodruff, J.L.; Cox, E.; Mohamed, L.S.; Choo, D.I.; Boppana, S.B. A Targeted Approach for Congenital Cytomegalovirus Screening Within Newborn Hearing Screening. Pediatrics 2017, 139. [Google Scholar] [CrossRef]
  48. Lorenzoni, F.; Lunardi, S.; Liumbruno, A.; Ferri, G.; Madrigali, V.; Fiorentini, E.; Forli, F.; Berrettini, S.; Boldrini, A.; Ghirri, P. Neonatal Screening for Congenital Cytomegalovirus Infection in Preterm and Small for Gestational Age Infants. J. Matern. Neonatal Med. 2014, 27, 1589–1593. [Google Scholar] [CrossRef] [PubMed]
  49. Schlesinger, Y.; Reich, D.; Eidelman, A.I.; Schimmel, M.S.; Hassanin, J.; Miron, D. Congenital Cytomegalovirus Infection in Israel: Screening in Different Subpopulations. Isr. Med. Assoc. J. 2005, 7, 237–240. [Google Scholar] [PubMed]
  50. Barkai, G.; Ari-Even Roth, D.; Barzilai, A.; Tepperberg-Oikawa, M.; Mendelson, E.; Hildesheimer, M.; Kuint, J. Universal Neonatal Cytomegalovirus Screening Using Saliva—Report of Clinical Experience. J. Clin. Virol. 2014, 60, 361–366. [Google Scholar] [CrossRef]
  51. Dar, L.; Namdeo, D.; Kumar, P.; Thakar, A.; Kant, S.; Rai, S.; Singh, P.K.; Kabra, M.; Fowler, K.B.; Boppana, S.B. Congenital Cytomegalovirus Infection and Permanent Hearing Loss in Rural North Indian Children. Pediatr. Infect. Dis. J. 2017, 36, 670. [Google Scholar] [CrossRef]
  52. Yamamoto, A.Y.; Anastasio, A.R.T.; Massuda, E.T.; Isaac, M.L.; Manfredi, A.K.S.; Cavalcante, J.M.S.; Carnevale-Silva, A.; Fowler, K.B.; Boppana, S.B.; Britt, W.J.; et al. Contribution of Congenital Cytomegalovirus Infection to Permanent Hearing Loss in a Highly Seropositive Population: The Brazilian Cytomegalovirus Hearing and Maternal Secondary Infection Study. Clin. Infect. Dis. 2020, 70, 1379–1384. [Google Scholar] [CrossRef]
  53. Yamada, H.; Tanimura, K.; Fukushima, S.; Fujioka, K.; Deguchi, M.; Sasagawa, Y.; Tairaku, S.; Funakoshi, T.; Morioka, I. A Cohort Study of the Universal Neonatal Urine Screening for Congenital Cytomegalovirus Infection. J. Infect. Chemother. 2020, 26, 790–794. [Google Scholar] [CrossRef] [PubMed]
  54. Blázquez-Gamero, D.; Soriano-Ramos, M.; Vicente, M.; Pallás-Alonso, C.R.; Pérez-Rivilla, A.; García-Álvarez, M.; Pinilla Martín, M.T.; Freire, X.; De Vergas, J.; De Aragón, A.M.; et al. Prevalence and Clinical Manifestations of Congenital Cytomegalovirus Infection in a Screening Program in Madrid (PICCSA Study). Pediatr. Infect. Dis. J. 2020, 39, 1050–1056. [Google Scholar] [CrossRef]
  55. Letamendia-Richard, E.; Périllaud-Dubois, C.; de La Guillonnière, L.; Thouard, I.; Cordier, A.G.; Roque-Afonso, A.M.; de Luca, D.; Benachi, A.; Vauloup-Fellous, C. Universal Newborn Screening for Congenital Cytomegalovirus Infection: Feasibility and Relevance in a French Type-III Maternity Cohort. BJOG 2022, 129, 291–299. [Google Scholar] [CrossRef]
  56. Chiereghin, A.; Pavia, C.; Turello, G.; Borgatti, E.C.; Baiesi Pillastrini, F.; Gabrielli, L.; Gibertoni, D.; Marsico, C.; De Paschale, M.; Manco, M.T.; et al. Universal Newborn Screening for Congenital Cytomegalovirus Infection – From Infant to Maternal Infection: A Prospective Multicenter Study. Front. Pediatr. 2022, 10, 1–11. [Google Scholar] [CrossRef]
  57. Ronner, E.A.; Glovsky, C.K.; Herrmann, B.S.; Woythaler, M.A.; Pasternack, M.S.; Cohen, M.S. Congenital Cytomegalovirus Targeted Screening Implementation and Outcomes: A Retrospective Chart Review. Otolaryngol. Neck Surg. 2022, 167, 178–182. [Google Scholar] [CrossRef] [PubMed]
  58. Diener, M.L.; Zick, C.D.; McVicar, S.B.; Boettger, J.; Park, A.H. Outcomes from a Hearing-Targeted Cytomegalovirus Screening Program. Pediatrics 2017, 139. [Google Scholar] [CrossRef] [PubMed]
  59. Rawlinson, W.D.; Palasanthiran, P.; Hall, B.; Al Yazidi, L.; Cannon, M.J.; Cottier, C.; van Zuylen, W.J.; Wilkinson, M. Neonates with Congenital Cytomegalovirus and Hearing Loss Identified via the Universal Newborn Hearing Screening Program. J. Clin. Virol. 2018, 102, 110–115. [Google Scholar] [CrossRef] [PubMed]
  60. Stehel, E.K.; Shoup, A.G.; Owen, K.E.; Jackson, G.L.; Sendelbach, D.M.; Boney, L.F.; Sánchez, P.J. Newborn Hearing Screening and Detection of Congenital Cytomegalovirus Infection. Pediatrics 2008, 121, 970–975. [Google Scholar] [CrossRef] [PubMed]
  61. Williams, E.J.; Kadambari, S.; Berrington, J.E.; Luck, S.; Atkinson, C.; Walter, S.; Embleton, N.D.; James, P.; Griffiths, P.; Davis, A.; et al. Feasibility and Acceptability of Targeted Screening for Congenital CMV-Related Hearing Loss. Arch. Dis. Child. Fetal Neonatal Ed. 2014, 99, 230–236. [Google Scholar] [CrossRef] [PubMed]
  62. Kawada, J.; Torii, Y.; Kawano, Y.; Suzuki, M.; Kamiya, Y.; Kotani, T.; Kikkawa, F.; Kimura, H.; Ito, Y. Viral Load in Children with Congenital Cytomegalovirus Infection Identified on Newborn Hearing Screening. J. Clin. Virol. 2015, 65, 41–45. [Google Scholar] [CrossRef] [PubMed]
  63. Roth, D.A.E.; Lubin, D.; Kuint, J.; Teperberg-Oikawa, M.; Mendelson, E.; Strauss, T.; Barkai, G. Contribution of Targeted Saliva Screening for Congenital CMV-Related Hearing Loss in Newborns Who Fail Hearing Screening. Arch. Dis. Child. Fetal Neonatal Ed. 2017, 102, F519–F524. [Google Scholar] [CrossRef] [PubMed]
  64. Beswick, R.; David, M.; Higashi, H.; Thomas, D.; Nourse, C.; Koh, G.; Koorts, P.; Jardine, L.A.; Clark, J.E. Integration of Congenital Cytomegalovirus Screening within a Newborn Hearing Screening Programme. J. Paediatr. Child Health 2019, 55, 1381–1388. [Google Scholar] [CrossRef] [PubMed]
  65. Pellegrinelli, L.; Galli, C.; Primache, V.; Alde, M.; Fagnani, E.; Di Berardino, F.; Zanetti, D.; Pariani, E.; Ambrosetti, U.; Binda, S. Diagnosis of Congenital CMV Infection via DBS Samples Testing and Neonatal Hearing Screening: An Observational Study in Italy. BMC Infect. Dis. 2019, 19, 652. [Google Scholar] [CrossRef] [PubMed]
  66. Chung, P.K.; Schornagel, F.; Oudesluys-Murphy, A.M.; De Vries, L.S.; Soede, W.; Van Zwet, E.; Vossen, A. Targeted Screening for Congenital Cytomegalovirus Infection: Clinical, Audiological and Neuroimaging Findings. Arch. Dis. Child. Fetal Neonatal Ed. 2023, 108, F302–F308. [Google Scholar] [CrossRef]
  67. Fourgeaud, J.; Boithias, C.; Walter-Nicolet, E.; Kermorvant, E.; Couderc, S.; Parat, S.; Pol, C.; Mousset, C.; Bussières, L.; Guilleminot, T.; et al. Performance of Targeted Congenital Cytomegalovirus Screening in Newborns Failing Universal Hearing Screening: A Multicenter Study. J. Bone Jt. Surg. 2022, 41, 478–481. [Google Scholar] [CrossRef]
  68. Webb, E.; Gillespie, A.N.; Poulakis, Z.; Gartland, T.; Buttery, J.; Casalaz, D.; Daley, A.J.; Donath, S.; Gwee, A.; Jacobs, S.E.; et al. Feasibility and Acceptability of Targeted Salivary Cytomegalovirus Screening through Universal Newborn Hearing Screening. J. Paediatr. Child Health 2022, 58, 288–294. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, Y.; Egashira, T.; Egashira, M.; Ogiwara, S.; Tomino, H.; Shichijo, A.; Mizukami, T.; Ogata, T.; Moriuchi, H.; Takayanagi, T.; et al. Expanded Targeted Screening for Congenital Cytomegalovirus Infection. Congenit. Anom. 2023, 63, 79–82. [Google Scholar] [CrossRef] [PubMed]
  70. Lee, G.; Carr, W.; Reingold, A.; Hunter, P.; Lee, G.; Temte, J.; Campos-Outcalt, D.; Rubin, L.; O’Leary, S.; Savoy, M.; et al. Updated Framework for Development of Evidence-Based Recommendations by the Advisory Committee on Immunization Practices. MMWR. Morb. Mortal. Wkly. Rep. 2018, 67, 1271–1272. [Google Scholar] [CrossRef] [PubMed]
  71. Griffiths, P.D. Burden of Disease Associated with Human Cytomegalovirus and Prospects for Elimination by Universal Immunisation. Lancet Infect. Dis. 2012, 12, 790–798. [Google Scholar] [CrossRef] [PubMed]
  72. Plotkin, S.A.; Wang, D.; Oualim, A.; Diamond, D.J.; Kotton, C.N.; Mossman, S.; Carfi, A.; Anderson, D.; Dormitzer, P.R. The Status of Vaccine Development Against the Human Cytomegalovirus. J. Infect. Dis. 2020, 221, S113–S122. [Google Scholar] [CrossRef] [PubMed]
  73. Krause, P.R.; Bialek, S.R.; Boppana, S.B.; Griffiths, P.D.; Laughlin, C.A.; Ljungman, P.; Mocarski, E.S.; Pass, R.F.; Read, J.S.; Schleiss, M.R.; et al. Priorities for CMV Vaccine Development. Vaccine 2013, 32, 4–10. [Google Scholar] [CrossRef] [PubMed]
  74. Pass, R.F.; Zhang, C.; Evans, A.; Simpson, T.; Andrews, W.; Huang, M.-L.; Corey, L.; Hill, J.; Davis, E.; Flanigan, C.; et al. Vaccine Prevention of Maternal Cytomegalovirus Infection. N. Engl. J. Med. 2009, 360, 1191–1199. [Google Scholar] [CrossRef] [PubMed]
  75. Bernstein, D.I.; Munoz, F.M.; Callahan, S.T.; Rupp, R.; Wootton, S.H.; Edwards, K.M.; Turley, C.B.; Stanberry, L.R.; Patel, S.M.; Mcneal, M.M.; et al. Safety and Efficacy of a Cytomegalovirus Glycoprotein B (GB) Vaccine in Adolescent Girls: A Randomized Clinical Trial. Vaccine 2016, 34, 313. [Google Scholar] [CrossRef]
  76. Das, R.; Blázquez-Gamero, D.; Bernstein, D.I.; Gantt, S.; Bautista, O.; Beck, K.; Conlon, A.; Rosenbloom, D.I.S.; Wang, D.; Ritter, M.; et al. 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. Lancet Infect. Dis. 2023, 23, 1383–1394. [Google Scholar] [CrossRef] [PubMed]
  77. Hu, X.; Karthigeyan, K.P.; Herbek, S.; Valencia, S.M.; Jenks, J.A.; Webster, H.; Miller, I.G.; Connors, M.; Pollara, J.; Andy, C.; et al. Human Cytomegalovirus MRNA-1647 Vaccine Candidate Elicits Potent and Broad Neutralization and Higher Antibody-Dependent Cellular Cytotoxicity Responses Than the GB/MF59 Vaccine. J. Infect. Dis. 2024, 230, 455–466. [Google Scholar] [CrossRef] [PubMed]
  78. Study Details|Dose-Finding Trial to Evaluate the Safety and Immunogenicity of Cytomegalovirus (CMV) Vaccine MRNA-1647 in Healthy Adults |ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/study/NCT04232280?term=NCT04232280&rank=1 (accessed on 27 November 2023).
  79. Study Details|A Study to Evaluate the Efficacy, Safety, and Immunogenicity of MRNA-1647 Cytomegalovirus (CMV) Vaccine in Healthy Participants 16 to 40 Years of Age |ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/study/NCT05085366?term=NCT05085366&rank=1 (accessed on 27 November 2023).
Figure 1. Preventive public measures proposed to reduce the cCMV burden. Adapted from Public Health Interventions (population-based), Minnesota Department of Health [18].
Figure 1. Preventive public measures proposed to reduce the cCMV burden. Adapted from Public Health Interventions (population-based), Minnesota Department of Health [18].
Viruses 16 01311 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liberati, C.; Sturniolo, G.; Brigadoi, G.; Cavinato, S.; Visentin, S.; Cosmi, E.; Donà, D.; Rampon, O. Burden of Congenital CMV Infection: A Narrative Review and Implications for Public Health Interventions. Viruses 2024, 16, 1311. https://doi.org/10.3390/v16081311

AMA Style

Liberati C, Sturniolo G, Brigadoi G, Cavinato S, Visentin S, Cosmi E, Donà D, Rampon O. Burden of Congenital CMV Infection: A Narrative Review and Implications for Public Health Interventions. Viruses. 2024; 16(8):1311. https://doi.org/10.3390/v16081311

Chicago/Turabian Style

Liberati, Cecilia, Giulia Sturniolo, Giulia Brigadoi, Silvia Cavinato, Silvia Visentin, Erich Cosmi, Daniele Donà, and Osvalda Rampon. 2024. "Burden of Congenital CMV Infection: A Narrative Review and Implications for Public Health Interventions" Viruses 16, no. 8: 1311. https://doi.org/10.3390/v16081311

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop