Congenital By Elena Grebenciucova MD (Dr. Grebenciucova of the University of Chicago has no relevant financial relationships to disclose.) Raymond P Roos MD (Dr. Roos of the University of Chicago owns stock in Amgen, Express Scripts, Isis, and Merck.) Originally released November 1, 1993; last updated February 20, 2017; expires February 20, 2020

Introduction

This article includes discussion of congenital cytomegalovirus, intrauterine cytomegalovirus , prenatal cytomegalovirus, and cytomegalic inclusion disease. The foregoing terms may include synonyms, similar disorders, variations in usage, and abbreviations.

Overview

Cytomegalovirus (CMV) is a ubiquitous agent responsible for most intrauterine . Besides well-known symptoms and findings such as hearing loss and microcephaly, congenital cytomegalovirus infection can also underlie certain cerebral anomalies and static leukodystrophies. In this article, the authors discuss uncommon presentations of asymptomatic congenital cytomegalovirus, predicted outcomes of congenital multi-strain cytomegalovirus infection, and updates on potential cytomegalovirus vaccines.

Key points • Cytomegalovirus is the most common infection of the developing brain. • Congenital cytomegalovirus infection can cause a host of cerebral abnormalities, including calcifications, ventriculomegaly, white matter lesions, cortical atrophy, and cortical migration abnormalities. • Affected children can have neurologic impairments that range from sensorineural hearing loss to profound mental and motor deficits.

Historical note and terminology

In 1904, Ribbert described a stillborn with congenital who had large inclusion-bearing cells in the kidney (Ribbert 1904). A similar report followed (Jesionek and Kiolemenoglou 1904). These are probably the first cases of congenital cytomegalovirus described in literature. By 1921, it was postulated that the inclusions were due to an infectious agent, most likely a virus (Goodpasture and Talbot 1921; Lipschutz 1921). The agent was subsequently called "salivary gland virus," and the infection was referred to as "cytomegalic inclusion disease.” Murine cytomegalovirus was successfully grown in tissue culture in 1954 (Smith 1954). Two years later, human cytomegalovirus was isolated from congenitally infected children using similar techniques (Smith 1956; Weller et al 1957). The name "cytomegalovirus" was accepted in 1960 (Weller et al 1960).

Clinical manifestations

Presentation and course

Only 5% to 12% of infected are symptomatic at birth. However, more symptoms appear on longer follow-up because congenital cytomegalovirus is a chronic persistent infection.

Symptomatic . The most common abnormalities at birth are , thrombocytopenia, petechiae, purpura, hepato- or , intrauterine growth retardation, microcephaly, and sensorineural hearing loss (5% to 10% at birth and up to 40% during childhood). Less common are hemolytic , ascites, hydrops, prematurity, inguinal hernia, pneumonitis, and ocular problems (chorioretinitis, microphthalmos). The presence or absence of microcephaly predicts nervous system involvement (Gandhi et al 2010). Various neurologic problems can occur: encephalitis, mental retardation, , abnormal tone or movements, hydrocephalus, leukodystrophy, or migration abnormalities such as lissencephaly or schizencephaly (Tatli et al 2005).{embed="pagecomponents/media_embed" entry_id="8690"} CT scan is abnormal in 70% of symptomatic children, most commonly showing parenchymal calcifications, and correlates with neurologic outcome. As an alternative to CT, ultrasonography can be combined with magnetic resonance imaging in the newborn period.

Asymptomatic neonatal infection. Approximately 85% of infected infants will fall into this group (Pass 2010). Some will be diagnosed as a result of neonatal screening for hearing or with late sequelae, such as hearing loss (6% to 25%) or permanent neurologic impairment (13.5%). Of interest, a unique case of leukemoid reaction (>100,000 WBCs) as the only presenting feature of congenital cytomegalovirus infection in an otherwise asymptomatic preterm infant has been described (Isik et al 2014).

Hearing should be assessed periodically during childhood because hearing loss is the most common sign of congenital cytomegalovirus infection, especially first trimester infections, and can progress. Of all affected symptomatic and asymptomatic infants, 5% have hearing loss at birth and 15.4%, at 6 years (Gandhi et al 2010).

Prognosis and complications

Prognosis relates to the timing of in utero infection as well as the presence or absence of symptoms at birth and the type of maternal infection. Primary maternal cytomegalovirus infection and symptomatic congenital infection increase the likelihood of significant problems. Children symptomatic at birth have a mortality rate of 10% to 20% and are more likely to develop permanent sequelae than asymptomatic ones (50% vs. 13%) (Dollard et al 2007). Ascites and are associated with an increased death rate. Ventriculomegaly, microcephaly, and calcification are associated with neurologic damage (Maruyama et al 2007). Cortical migration disorders such as lissencephaly or polymicrogyria can occur, but largely depend on the timing of the in utero infection. Lissencephaly is observed if the infection occurs prior to 16 to 18 weeks of gestation (Barkovich and Lindan 1994). Polymicrogyria may occur if the fetus is infected between 18 and 24 weeks of gestation. Frontal and temporal polymicrogyria are the most common patterns of migrational disorder (White et al 2014). Death within the first month results from severe hepatic disease, disseminated intravascular coagulation, or secondary bacterial infections; death in the first year results from progressive liver disease or severe failure to thrive. Death after 1 year of age is seen in those with severe neurologic damage and usually results from complications of infection, aspiration, or malnutrition. Subsequent and significant additional deficits occur in up to 95% of symptomatic infections. These include hearing loss (bilateral in up to 70%), neurologic impairment (mental retardation, motor deficits, seizures, and behavioral or cognitive deficits), dental defects (abnormal enamel, severe caries, crumbling primary teeth), and vision loss due to chorioretinitis or optic atrophy. Hearing loss occurs in one fifth of asymptomatic and one third of symptomatic congenitally infected infants. It can improve, worsen, or fluctuate during childhood. It must be noted that congenital co-infection with multiple cytomegalovirus strains is not currently shown to be predictive of either severity of symptoms or sensorineural hearing loss at birth (Pati et al 2013).

Asymptomatic infection does not carry a risk of death, but it can lead to morbidity. Hearing loss and (according to some studies) neurodevelopmental defects develop in 5% to 17% of these patients.

A combination of imaging characteristics, cerebrospinal fluid profile and head circumference adjusted for weight, can be helpful in predicting neurologic outcomes after congenital infection. Imaging-based predictive scales based on the presence of destructive lesions (dilated ventricles, germinolysis, calcifications, and atrophy), teratogenic abnormalities (cerebellar hypoplasia, dysgenesis of the corpus callosum), and white matter signal abnormality on MRI can be useful (Alarcon et al 2016).

Clinical vignette

A 3-month-old girl was admitted for and anemia. She was born at term as the first child of healthy parents with a birthweight of 2200 g. The pregnancy and delivery were uneventful. On physical examination she had microcephaly, jaundice, respiratory difficulty, and hepatosplenomegaly. Laboratory tests revealed elevated liver enzymes, hemolytic anemia, and thrombocytopenia. Chest x-ray showed interstitial pneumonia. Congenital cytomegalovirus infection was diagnosed on the basis of specific IgM in the serum, the presence of inclusion bodies in urinary epithelial cells, and a positive polymerase chain reaction with high viral load in the urine.

Biological basis

Etiology and pathogenesis

Human cytomegalovirus is an enveloped double-stranded DNA member of the herpes virus group. It is ubiquitous and infects only humans. Like other herpes agents, cytomegalovirus becomes latent in the host after primary infection and may reactivate to cause secondary infection. Different cytomegalovirus strains exist, and reinfections also occur. Congenital cytomegalovirus is due to fetal infection by maternal virus. Both primary and secondary maternal infections can involve the fetus at any time during pregnancy. Congenital cytomegalovirus produces a chronic infection: virus is recoverable months to years after birth.

Congenital cytomegalovirus is an intrauterine infection, distinct from perinatal cytomegalovirus infection. In perinatal infections, virus is transmitted to the newborn at the time of birth, or shortly after, by way of infected birth canal, breast milk, or blood transfusion. Congenital infection probably results from virus acquired by susceptible women immediately before or during early pregnancy followed by viremia, placental infection, and hematogenous and transplacental spread to the fetus. Alternative postulated, but unproven, mechanisms include: reactivation of latent virus within uterine tissue, transovarian infection, or spermatozoan infection. Preexisting maternal immunity protects considerably from virus transmission. Only 1.5% of secondary maternal infections involve the fetus, compared to 30% to 40% of primary infections. Still, more than 60% of all the infants infected in utero are born to mothers with preconception immunity and who have a secondary infection by new strains during pregnancy. However, severe symptoms, and specifically severe and progressive hearing loss, are more frequent during primary infection (Ross et al 2006).

The extent of damage due to cytomegalovirus varies widely and is correlated with viral load (Boppana et al 2005) as well as immune-mediated damage for cytotoxic T lymphocytes and consequent hypoxic cerebral damage (Gabrielli et al 2012). Tumor necrosis factor receptor UL144 polymorphisms have been suggested as a predisposing factor affecting virus load (Arav-Boger et al 2006). Gap junction protein beta-2 (GJB2) mutations are more frequent in patients with cytomegalovirus and hearing loss than those without (21% vs. 3%) (Ross et al 2007).

Cytomegalovirus can infect many different cell types and all major organs. The cochlea is frequently involved, as is the central nervous system. There is a predilection for periependymal neurons and glia, with focal encephalitis and periependymitis. Necrotic periependymal tissue subsequently calcifies. Calcifications are typically periventricular but may also be scattered throughout the brain. Cytomegalovirus produces cytolysis, with focal necrosis and a localized mononuclear inflammatory response. Tissue damage results from direct effects of the inflammatory response as well as an associated vasculopathy resulting in ischemia and encephalomalacia, immune-mediated reactions, and apoptosis (DeBiasi et al 2002).

Typical pathology involves cytomegalic brain cells with intranuclear inclusions. When cytomegalovirus infects developing CNS tissue, it can produce microcephaly with neuronal migration defects. Severe destructive changes lead to more severe brain abnormalities such as porencephalic cysts, cerebellar hypoplasia, aqueductal stenosis, and hydrocephalus. Postnatally, MRI can detect white matter abnormalities that occur in several patterns: predominantly parietal lobe lesions, multifocal white matter lesions with polymicrogyria, and diffuse white matter lesions with polymicrogyria (van der Knaap et al 2004).

An in vitro model using neural stem cells showed that CMV activates the peroxisome proliferator-activated receptor γ (PPARγ), which inhibits neural stem cell differentiation into neurons and, thus, may underlie one of the key pathogenic mechanisms behind CMV-associated neurologic sequelae (Chavanas 2016). Cytomegalovirus can induce specific chromosomal damage that requires viral entry into the cell, but not de novo viral protein expression, a probable mechanism for damage in the developing fetal brain (Fortunato and Spector 2003). Another mechanism in the pathogenesis might be related to down regulation of the EGF receptor on the cell surface by cytomegalovirus (Beutler et al 2003).

Congenital cytomegalovirus is a persistent chronic infection. Half of infected children show viremia for months and viruria for 6 years or more. Cytomegalovirus may be excreted in saliva for 2 to 4 years. Late sequelae reflect this chronic infection of developing tissue. In addition, some abnormalities that are present at birth may not be detectable until the infant is older.

Epidemiology"

The prevalence of congenital cytomegalovirus infection varies from 0.15% to 2.0% in different countries. Approximately 40,000 congenitally infected infants are born in the United States each year. Polymerase chain reaction of dried blood samples demonstrated that 0.7% of newborns in California were positive for cytomegalovirus; however, not all newborns with congenital infection can be identified, even by this sensitive method (Pass 2010). Ten to twenty percent show signs of neurologic damage; approximately half will manifest problems within the first 6 years of life. Congenital cytomegalovirus is the most common cause of nonhereditary sensorineural hearing loss in children.

Seropositivity varies with age, geographic location, cultural and socioeconomic background, and childrearing practices. In the United States, seroprevalence is 40% to 60% among middle and upper income people, compared with 80% in lower income groups. Seroconversion rates during pregnancy range from 0.7% to 4.1%. Primary maternal infection carries a higher risk for fetal damage, but recurrent maternal infection may also be associated with symptomatic infection in the infant. The rate of transmission to infants born to mothers who had a primary infection or a recurrent infection during pregnancy was 32% and 1.4%, respectively. Neurologic symptoms have occasionally been observed in infants born to mothers with preconceptional immunity. Nonwhite race, low-socioeconomic status, premature birth, and neonatal intensive care unit admittance were risk factors for congenital cytomegalovirus infection (Kenneson and Cannon 2007). Significant epidemiological associations were found between congenital cytomegalovirus infection and the following: caring for school children in the year before delivery; onset of sexual activity less than 2 years before delivery; sexually transmitted diseases during pregnancy; household size of more than 3 people; and maternal age of less than 25 years. Women who carried both of the first 2 factors were at greatest risk for delivering an infected baby (Fowler and Pass 2006).

Prevention

The goal of preventing congenital cytomegalovirus infection with a vaccine is of the highest priority. The presence of cytomegalovirus genes that encode proteins that lead to evasion of the immune system complicates vaccine design. In addition, the immunological basis for prevention is not sufficiently identified.

Currently there are no effective vaccines available, although several potential candidates are emerging (Swanson and Schleiss 2013). A phase II trial studying recombinant genetically modified gB, which is an immunodominant envelope protein, in an adjuvant (MF59) versus placebo in cytomegalovirus seronegative women proved to be immunogenic and showed a 50% reduction in the rate of maternal infection (Pass et al 2009). It must, however, be noted that most of the efficacy was observed in the first 12 to 15 months. Despite being moderately efficacious, this vaccine ultimately failed to induce a durable and robust cellular and humoral immunity (Wang and Fu 2014). Another potential vaccine that underwent phase I trials combined gB protein and pp65/IE1 fusion protein was successful at neutralizing antibody response (Bernstein et al 2009). In a phase 2 trial, this vaccine (TransVax) reduced posttransplant cytomegalovirus viremia in patients undergoing hematopoietic cell transplant. TransVax is currently undergoing a phase 3 trial in hematopoietic cell transplant patients. The cytomegalovirus vaccine CymVectin combines plasmids that express pp65/IE1 fusion protein with gB formulated with Vaxfectin, an adjuvant that helps induce a robust humoral and cellular response. It is currently in late preclinical development. Many other candidate vaccines are currently in preclinical development as well. These combine peptides, subunits, or all multisubunit complexes of the pentameric complex necessary for the endothelial and epithelial cell entry by the virus (McVoy 2013).

At this time, neither primary prevention, nor standardized, high-throughput screening tests and follow-up protocols for secondary and tertiary prevention are available. Therefore, current preventive measures include counseling young women, assessing their serologic status to identify those at risk for primary infection, and recommending appropriate hygiene measures in situations with risk of cytomegalovirus exposure, such as daycare centers. Recommendations for pregnant women include avoidance of iatrogenic transmission through blood transfusion and adherence to hygiene measures. Infants actively excrete virus. Awareness of this fact, with appropriate contact isolation and proper hygienic measures, minimizes risk of cytomegalovirus transmission. For pregnant women who have acquired a primary cytomegalovirus infection, maternal-to-fetal transmission is probably preventable using cytomegalovirus hyperimmune globulin (Adler et al 2007).

Although a large percentage of pregnant women shed the virus during pregnancy, their infants do not develop congenital infection (Cheeran et al 2009). Therefore, prenatal diagnosis of intrauterine infection is accomplished by serologic studies (ie, detection of virus-specific IgG in a previously seronegative pregnant woman or of specific IgM with low IgG avidity) and most reliably by PCR of the amniotic fluid collected at least 7 weeks after the presumed time of maternal infection and after 21 weeks of gestation (Yinon et al 2010). A low avidity index indicates recent infection and may help to distinguish pregnancies in which invasive prenatal diagnosis is necessary. Ebina and colleagues showed that a cut-off value of less than 40% IgG avidity index carried a specificity of 96.1% and a sensitivity of 64.3% for prediction of congenital infection (Ebina et al 2014).

IgM-positivity, seroconversion, or anti-CMV antibodies of low/moderate avidity are observed for approximately 18 to 20 weeks after primary infection.

Yinon and colleagues assessed the quality of evidence for diagnosis of intrauterine infection (Yinon et al 2010). Routine screening of pregnant women for cytomegalovirus by serologic testing is currently not recommended – a practice that is not without some controversy (Din et al 2011). Screening is advised when an influenza-like illness is experienced during pregnancy; when ultrasonographic findings suggesting infection are detected; and for seronegative pregnant women who work in child care or health care. In the case of primary maternal infection, there is a 30% to 40% risk for fetal infection and 20% to 25% risk for sequelae. In cases of proven secondary infection (high IgG avidity), counselors should estimate a lower transmission rate. Ultrasonography findings that show an abnormal pattern of periventricular echogenicity, ventriculomegaly, and various parenchymal abnormalities may support the diagnosis of suspected cytomegalovirus infection in mid or late pregnancy and may indicate the need for serological tests. However, ultrasound abnormalities predict symptomatic congenital infection in only a third of cases (Guerra et al 2008).

Risk factors for asymptomatic infants developing late hearing loss include an intense and prolonged maternal antibody response, which suggests infection early in pregnancy and continuing antigenic stimulation. Genetic mutations predisposing the child to hearing loss due to cytomegalovirus, if confirmed in larger populations, may be screened to identify infants at risk. Universal screening of hearing in neonates and preschoolers is likely to reduce the most frequent effect of this congenital infection.

Differential diagnosis

The differential diagnosis of congenital cytomegalovirus includes other congenital infections (rubella, , syphilis, herpes simplex, hepatitis, and varicella zoster). Both cytomegalovirus and intrauterine parvovirus B19 infection can cause fetal hydrops. Rubella produces heart defects, , a salt-and-pepper retinopathy (rather than chorioretinitis), and, rarely, produces cerebral calcifications. Toxoplasmosis typically has scattered, rather than periventricular, calcifications, and its associated skin is maculopapular. produces long-bone changes (osteochondritis, epiphysitis), , and mucous membrane lesions; brain calcifications are unusual. Herpes simplex may produce skin and mucous membrane vesicles. Neonatal herpes is generally a perinatal infection, and presents acutely 1 to 3 weeks after birth.

Bacterial sepsis is also in the differential, as are noninfectious disorders such as hemolytic diseases (due to blood type or factor incompatibilities or primary red cell defects), metabolic disorders (galactosemia, tyrosinemia), immune thrombocytopenia, reticuloendotheliosis, and congenital leukemia. The differential diagnosis widens dramatically when congenital cytomegalovirus produces only mild or single organ disease.

Metabolic disorders, particularly respiratory chain defects, can cause cortical migration abnormalities and cerebral calcifications similar to congenital cytomegalovirus infection. Severe lactic acidosis in the neonate suggests a metabolic disturbance that can be proven by measurement of mitochondrial enzyme activities.

Attention has been drawn to the resemblance between congenital cytomegalovirus and autosomal recessive cystic leukoencephalopathy without megalencephaly. In the latter, children are asymptomatic at birth but show developmental delay within the first year of life, with a normal or small head circumference. Bilateral cystic lesions in the anterior temporal lobes, enlarged inferior horns, and multifocal white matter alterations can be seen on brain MRI. Brain computed tomography demonstrates intracranial calcifications that can mimic congenital cytomegalovirus infection (Henneke et al 2009).

Diagnostic workup

Congenital cytomegalovirus should be suspected in any newborn with signs suggesting this diagnosis or when there has been maternal seroconversion during pregnancy. In pregnant women, the diagnosis is essentially based on the detection of IgG and IgM antibodies. The presence of the cytomegalovirus DNA by PCR in the maternal uterine cervical secretions may be predictive of congenital cytomegalovirus infection (Tanimura et al 2017). The detection of viral DNA in the amniotic fluid confirms congenital infection. However, amniocentesis must be performed after 21 weeks' gestation and at least 6 weeks after seroconversion in order to reliably detect cytomegalovirus. A high cytomegalovirus load in the amniotic fluid on quantitative PCR correlates with symptomatic infection.

The gold standard for diagnosing congenital cytomegalovirus infection is isolation of the virus from newborn babies' urine, saliva, conjunctival or rectal swabs, or leukocytes through conventional or rapid cell culture techniques. All samples should be obtained within the first 2 weeks of life because later samples do not allow differentiation between intrauterine and intra- or postpartum infection. Detection of viral DNA by PCR is the most widely used diagnostic test and can be performed even retrospectively on blood, saliva, or urine samples obtained around birth. Quantitative PCR methods can be useful to distinguish between cytomegalovirus infection and cytomegalovirus disease; a significantly higher viral load is seen in symptomatic infants. A urine-based screening program can identify asymptomatic cases with low viral loads in the blood (Inoue and Koyano 2008). Real-time PCR in neonatal blood samples showed a lower sensitivity than the rapid culture of saliva, limiting its value as a screening test (Boppana et al 2010). According to the data derived from the CMV and Hearing Multicenter Screening (CHIMES) study, real-time PCR of saliva and urine are as reliable as virus culture, and provide a good diagnostic tool for high-volume screening (Ross et al 2014).

Less-established virus detection techniques include electron microscopy, enzyme-linked immunosorbent assay detection of antigen, cytologic examination on sloughed cells in urine, or immunohistochemistry or in situ hybridization in tissue samples, especially in the case of or abortion.

Serologic tests are not a substitute for virus isolation or histopathology, but are useful adjuncts for diagnosis. The absence of IgG against cytomegalovirus in cord or infant blood rules out congenital infection, and the presence of IgM supports this diagnosis; however, false-positive and false-negative reactions do occur. One study suggested that the neonatal serum IgM has a poor sensitivity (40.7%) for the diagnosis of congenital cytomegalovirus infection, being positive in only 48.8% of those with a symptomatic infection and 22.1% of those asymptomatic (Bilavsky et al 2017).

Other common laboratory findings are increased cord blood IgM level, atypical lymphocytosis, thrombocytopenia, anemia, direct hyperbilirubinemia, and increased aspartate transaminase. Cerebrospinal fluid may show increased protein in cases with neurologic damage or hearing loss and mild mononuclear pleocytosis. A variety of imaging approaches have been used to detect and confirm congenital cytomegalovirus infection (Lanari et al 2012). Neuroimaging may show intracranial (particularly periventricular) calcifications in 25% to 50%, multicentric encephalomalacia, leukodystrophy, ventriculomegaly, or migration abnormalities. Although pachygyria and lissencephaly can also be seen, the most typical findings are polymicrogyria, cerebellar hypoplasia, hippocampal malformations, and temporal lobe lesions (Doneda et al 2010). The latter typically consist of dilatation of temporal horns, white matter hyperintensity, and cysts in the temporal poles. For most abnormalities, including migrational defects, MRI is preferred. In suspected cases, fetal or postnatal MRI may be helpful in understanding the extent of cerebral damage. In this way the MRI may help decide the need for prenatal or postnatal antiviral therapy, or in some severely affected cases, the need for discussion about the possible termination of pregnancy as an option (Averill et al 2015).{embed="pagecomponents/media_embed" entry_id="8691"}{embed="pagecomponents/media_embed" entry_id="8692"}{embed="pagecomponents/media_embed" entry_id="8693"}{embed="pagecomponents/media_embed" entry_id="8694"}

Management

Prenatal management. Pregnant women with documented primary cytomegalovirus infection are offered termination, but the possibility of asymptomatic fetal infection can cause major difficulties in the decision process. At present, there is no recommended treatment for pregnant women with cytomegalovirus infection. Although hyperimmune IgG against cytomegalovirus was initially thought to lower the risk of congenital infection (Schleiss 2008), a randomized trial of hyperimmune globulin administered to 128 women showed that it did not significantly modify the course of primary infection. However, the power of the study to detect the difference was small, at 33%. It must also be noted that the rate of obstetrical adverse events was higher in the hyperimmune globulin group than in the placebo group (13% vs. 2%) (Revello et al 2014). A large phase 3, randomized, placebo-controlled, double-blind trial that will evaluate hyperimmune globulin prophylaxis is currently recruiting patients, NCT01376778.

A controlled trial evaluating oral valacyclovir administered to pregnant women to evaluate in utero efficacy in modifying primary congenital infection, NCT01037712, was terminated early due to not having enough enrollment. Fetal growth disturbance, brain calcifications, and microcephaly can be monitored by ultrasonography.

The most recent best practice guideline from the American Society for Maternal-Fetal Medicine does not recommend antenatal treatment with ganciclovir or valacyclovir (Society for Maternal-Fetal Medicine et al 2016). The guideline specifies that these options or cytomegalovirus hyperimmune immunoglobulin should be offered only as a part of a research protocol. Postnatal treatment. Infants with severe symptomatic infections require supportive care for systemic disease, eye problems, or seizures. Ganciclovir appears to preserve or improve hearing and neurodevelopmental status (Oliver et al 2009). Although intravenous ganciclovir and oral valganciclovir are used in the treatment of congenital cytomegalovirus infection, their use is limited by the potential for toxicity, especially neutropenia (Nassetta et al 2009). Oral treatment with valganciclovir appears preferable because of easier administration, high bioavailability, and efficacy on clinical and virological parameters (Gandhi et al 2010). The combined use of antiviral agents and anticytomegalovirus immunoglobulin in severe disease may be more efficient than either agent alone. Cytomegalovirus-induced infantile spasms are not treated with adrenocorticotropic hormone to avoid a disseminated infection. Infected infants require serial medical, audiologic, psychometric, and eye examinations to detect late sequelae. Older children may require specific therapeutic interventions and educational assistance. Cochlear implants are effective for hearing loss. Gandhi and colleagues published an evidence-based review and algorithm for the management of cases of suspected of congenital cytomegalovirus infection (Gandhi et al 2010).

Special considerations

Pregnancy

Congenital cytomegalovirus involves maternal infection during pregnancy. Primary maternal infections during the first half of pregnancy carry the greatest risk of fetal damage. Pregnant women should avoid contact with suspected cases.

References cited

Adler SP, Nigro G, Pereira L. Recent advances in the prevention and treatment of congenital cytomegalovirus infections. Semin Perinatol 2007;31:10-8. PMID 17317422

Alarcon A, Martinez-Biarge M, Cabañas F, Quero J, García-Alix A. A prognostic neonatal neuroimaging scale for symptomatic congenital cytomegalovirus infection. Neonatology 2016;110(4):277-85. PMID 27344149

Arav-Boger R, Battaglia CA, Lazzarotto T, et al. Cytomegalovirus (CMV)-encoded UL144 (truncated tumor necrosis factor receptor) and outcome of congenital CMV infection. J Infect Dis 2006;194:464-73. PMID 16845629

Averill LW, Kandula VV, Akyol Y, Epelman M. Fetal brain magnetic resonance imaging findings in congenital cytomegalovirus infection with postnatal imaging correlation. Semin Ultrasound CT MR 2015;36(6):476-86. PMID 26614131

Barkovich AJ, Lindan CE. Congenital cytomegalovirus infection of the brain: imaging analysis and embryologic considerations. Am J Neuroradiol 1994;15(4):703-15. PMID 8010273

Bernstein DI, Reap EA, Katen K, et al. Randomized, double-blind, phase I trial of an alphavirus replicon vaccine for cytomegalovirus in CMV seronegative adult volunteers. Vaccine 2009;28(2):484-93. PMID 19857446

Beutler T, Hoflich C, Stevens PA, Kruger DH, Prosch S. Downregulation of the epidermal growth factor receptor by human cytomegalovirus infection in human fetal lung fibroblasts. Am J Respir Cell Mol Biol 2003;28(1):86-94. PMID 12495936

Bilavsky E, Watad S, Levy I, et al. Positive IgM in congenital CMV infection. Clin Pediatr (Phila) 2017;56(4):371-5. PMID 28006975

Boppana SB, Fowler KB, Pass RF, et al. Congenital cytomegalovirus infection: association between virus burden in infancy and hearing loss. J Pediatr 2005;146:817-23. PMID 15973325

Boppana SB, Ross SA, Novak Z, et al. Dried blood spot real-time polymerase chain reaction assays to screen newborns for congenital cytomegalovirus infection. JAMA 2010;303(14):1375-82. PMID 20388893

Chavanas S. Peroxisome proliferator-activated receptor γ (PPARγ) activation: a key determinant of neuropathogeny during congenital infection by cytomegalovirus. Neurogenesis (Austin) 2016;3(1):e1231654. PMID 27844024

Cheeran MC, Lokensgard JR, Schleiss MR. Neuropathogenesis of congenital cytomegalovirus infection: disease mechanisms and prospects for intervention. Clin Microbiol Rev 2009;22(1):99-126. PMID 19136436 DeBiasi RL, Kleinschmidt-DeMasters BK, Richardson-Burns S, Tyler KL. Central nervous system apoptosis in human herpes simplex virus and cytomegalovirus encephalitis. J Infect Dis 2002;186(11):1547-57. PMID 12447729

Din ES, Brown CJ, Grosse SD, et al. Attitudes toward newborn screening for cytomegalovirus infection. Pediatrics 2011;128(6):e1434-42. PMID 22084323

Dollard SC, Grosse SD, Ross DS. New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection. Rev Med Virol 2007;17(5):355-63. PMID 17542052

Doneda C, Parazzini C, Righini A, et al. Early cerebral lesions in cytomegalovirus infection: prenatal MR imaging. Radiology 2010;255(2):613-21. PMID 20413771

Ebina Y, Minematsu T, Sonoyama A, et al. The IgG avidity for the prediction of congenital cytomegalovirus infection in a prospective cohort study. J Perinat Med 2014;42(6):755-9. PMID 24663226

Fortunato EA, Spector DH. Viral induction of site-specific chromosome damage. Rev Med Virol 2003;13(1):21-37. PMID 12516060

Fowler KB, Pass RF. Risk factors for congenital cytomegalovirus infection in the offspring of young women: exposure to young children and recent onset of sexual activity. Pediatrics 2006;118(2):e286-92. PMID 16847076

Gabrielli L, Bonasoni MP, Dantini D, et al. Congential cytomegalovirus infection: patterns of fetal brain damage. Clin Microbiol Infect 2012;18(10):E419-E427. PMID 22882294

Gandhi RS, Fernandez-Alvarez JR, Rabe H. Management of congenital cytomegalovirus infection: an evidence-based approach. Acta Paediatr 2010;99(4):509-15. PMID 20050828

Goodpasture E, Talbot FB. Concerning the nature of "protozoanlike" cells in certain lesions of infancy. Am J Dis Child 1921;21:415-25.

Guerra B, Simonazzi G, Puccetti C, et al. Ultrasound prediction of symptomatic congenital cytomegalovirus infection. Am J Obstet Gynecol 2008;198(4):380. PMID 18191802

Henneke M, Diekmann S, Ohlenbusch A, et al. RNASET2-deficient cystic leukoencephalopathy resembles congenital cytomegalovirus brain infection. Nat Genet 2009;41(7):773-5. PMID 19525954

Inoue N, Koyano S. Evaluation of screening tests for congenital cytomegalovirus infection. Pediatr Infect Dis J 2008;27(2):182-4. PMID 18174856

Isik DU, Aydemir O, Kale Y, et al. Severe leukemoid reaction in a preterm infant with congenital congenital cytomegalovirus infection. J Pediatr Hematol Oncol 2014;36(5):e310-2. PMID 24072252

Jesionek A, Kiolemenoglou B. Uber einen Befund von protozoenartigen Gebilden in den Organen eines hereditarleustiochen Fotus. Muenchen Medizinische Wochenschrift 1904;51:1905-7.

Kenneson A, Cannon MJ. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol 2007;17:253-76. PMID 17579921

Lanari M, Capretti MG, Lazzarotto T, et al. Neuroimaging in CMV congenital infected neonates: how and when. Early Hum Dev 2012;88 Suppl 2:S3-5. PMID 22633509

Lipschutz B. Untersuchungen uber die Aetiologic der Krankheiten d. Herpes genitalis, usw. Arch F Derm Syph 1921;136:428-82.

Maruyama Y, Sameshima H, Kamitomo M, et al. Fetal manifestations and poor outcomes of congenital cytomegalovirus infections: possible candidates for intrauterine antiviral treatments. J Obstet Gynaecol Res 2007;33(5):619-23. PMID 17845318

McVoy MA. Cytomegalovirus vaccines. Clin Infect Dis 2013;57 Suppl 4:S196-9. PMID 24257427 Nassetta L, Kimberlin D, Whitley R. Treatment of congenital cytomegalovirus infection: implications for future therapeutic strategies. J Antimicrob Chemother 2009;63(5):862-7. PMID 19287011

Oliver SE, Cloud GA, Sánchez PJ, et al. Neurodevelopmental outcomes following ganciclovir therapy in symptomatic congenital cytomegalovirus infections involving the central nervous system. J Clin Virol 2009;46 Suppl 4:S22-6. PMID 19766534

Pass RF. Congenital cytomegalovirus infection: screening and treatment. J Pediatr 2010;157(2):179-80. PMID 20542286

Pass RF, Zhang C, Evans A, et al. Vaccine prevention of maternal cytomegalovirus infection. N Engl J Med 2009;360:1191-9. PMID 19297572

Pati SK, Pinninti S, Novak Z, et al. Genotypic diversity and mixed infection in newborn disease and hearing loss in congenital cytomegalovirus infection. Pediatr Infect Dis J 2013;32(10):1050-4. PMID 23694837

Revello MG, Lazzarotto T, Guerra B, et al. A randomized trial of hyperimmune globulin to prevent congenital cytomegalovirus. N Eng J Med 2014;370(14):1316-26. PMID 24693891

Ribbert H. Uber protozoenartigen Zellen in der nireeines syphilitischen Neugeboren und in der Parotis von Kindren. Zentralbl Allg Pathol 1904;15:945-76.

Ross SA, Ahmed A, Palmer AL, et al. Detection of congenital cytomegalovirus infection by real-time polymerase chain reaction analysis of saliva or urine specimens. National Institute on Deafness and Other Communication Disorders CHIMES Study. J Infect Dis 2014;210(9):1415-8. PMID 24799600

Ross SA, Fowler KB, Ashrith G, et al. Hearing loss in children with congenital cytomegalovirus infection born to mothers with preexisting immunity. J Pediatr 2006;148(3):332-6. PMID 16615962

Ross SA, Novak Z, Kumbla RA, Zhang K, Fowler KB, Boppana S. GJB2 and GJB6 mutations in children with congenital cytomegalovirus infection. Pediatr Res 2007;61(6):687-91. PMID 17426645

Schleiss MR. Congenital cytomegalovirus infection: update on management strategies. Curr Treat Options Neurol 2008;10(3):186-92. PMID 18579022

Smith MG. Propagation of salivary gland virus of the mouse in tissue culture. Proc Soc Exp Biol Med 1954;86(3):435-40. PMID 13194679

Smith MG. Propagation in tissue cultures of a cytopathogenic virus from the human salivary gland virus (SGV) disease. Proc Soc Exp Biol Med 1956;92(2):424-30. PMID 13350368

Society for Maternal-Fetal Medicine, Hughes BL, Gyamfi-Bannerman C. Diagnosis and antenatal management of congenital cytomegalovirus infection. Am J Obstet Gynecol 2016;214(6):B5-B11. PMID 26902990

Swanson EC, Schleiss MR. Congenital cytomegalovirus infection: new prospects for prevention and therapy. Pediatr Clin North Am 2013;60(2)335-49. PMID 23481104

Tanimura K, Tairaku S, Ebina Y, et al. Prediction of congenital cytomegalovirus infection in high-risk pregnant women. Clin Infect Dis 2017;64(2):159-65. PMID 27986675

Tatli B, Ozmen M, Aydinli N, Caliskan M. Not a new leukodystrophy but congenital cytomegalovirus infection. J Child Neurol 2005;20:525-7. PMID 15996404 van der Knaap MS, Vermeulen G, Barkhof F, Hart AA, Loeber JG, Weel JF. Pattern of white matter abnormalities at MR imaging: use of polymerase chain reaction testing of Guthrie cards to link pattern with congenital cytomegalovirus infection. Radiology 2004;230(2):529-36. PMID 14752192

Wang D, Fu TM. Progress on human cytomegalovirus vaccine for prevention of congenital infection and disease. Curr Opin Virol 2014;6:13-23. PMID 24632198

Weller TH, Hanshaw JB, Scott DE. Serologic differentiation of viruses responsible for cytomegalic inclusion disease. Virology 1960;12:130-2. PMID 13843861

Weller TH, Macauley JC, Craig JM, Wirth P. Isolation of intranuclear inclusion-producing agents from infants with illnesses resembling cytomegalic inclusion disease. Proc Soc Exp Biol Med 1957;94:4-12. PMID 13400856

White AL, Hedlund GL, Bale JF Jr. Congenital cytomegalovirus infection and brain clefting. Pediatr Neurol 2014;50(3):218-23. PMID 24373410

Yinon Y, Farine D, Yudin MH, et al. Cytomegalovirus infection in pregnancy. J Obstet Gynaecol Can 2010;32(4):348-54. PMID 20500943

**References especially recommended by the author or editor for general reading.

Former authors

Patricia K Coyle MD (original author) and Banu Anlar MD

ICD and OMIM codes

ICD codes

ICD-9: Neonatal candida infection: 771.7

ICD-10: Neonatal : P37.5

Profile

Age range of presentation

0-01 month 01-23 months

Sex preponderance male=female

Family history none

Heredity none

Population groups selectively affected none selectively affected

Occupation groups selectively affected none selectively affected

Differential diagnosis list other congenital infections rubella toxoplasmosis syphilis Herpes simplex neonatal Herpes hepatitis varicella zoster bacterial sepsis hemolytic diseases metabolic disorders: galactosemia, tyrosinemia immune thrombocytopenia reticuloendotheliosis congenital leukemia

Associated disorders

Aqueductal stenosis Cerebellar hypoplasia Congenital infection syndrome Cytolysis Mental retardation Periventricular calcification Porencephalic cysts Sensorineural hearing loss

Other topics to consider

Acquired cytomegalovirus Autism spectrum disorder Hearing loss in infants and children Microcephaly Neuropathies associated with cytomegalovirus infection

Copyright© 2001-2017 MedLink Corporation. All rights reserved.