Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 450
Filter
1.
Travel Med Infect Dis ; 62: 102760, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39293589

ABSTRACT

Toxoplasma gondii (T. gondii), an obligate intracellular parasite, is considered as an opportunistic infection and causes toxoplasmosis in humans and animals. Congenital toxoplasmosis can influence pregnancy and cause mild to severe consequences for the fetal and neonatal. During early T. gondii infection, neutrophils as the most abundant white blood cells provide a front line of defense mechanism against infection. The activated dendritic cells are then responsible for initiating an inflammatory response via T-helper 1 (Th1) cells. As part of its robust immune response, the infected host cells produce interferon (IFN-γ). IFN-γ inhibits T. gondii replication and promotes its transformation from an active form to tissue cysts. Although anti- T. gondii antibodies play an important role in infection control, T-helper 2 (Th2) immune response, can facilitate the growth and proliferation of T. gondii in the host cell. In pregnant women infected with T. gondii, the expression of cytokines may vary and in response diverse outcomes are expected. Cytokine profiles serve as valuable indicators for estimating the patho-immunological effects of T. gondii infection. This demonstrates the intricate relationship between pro-inflammatory and anti-inflammatory cytokines, as well as their influence on the various pregnancy outcomes in T. gondii infection.

2.
Viruses ; 16(9)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39339911

ABSTRACT

Oropouche virus (OROV) is an emerging arbovirus endemic in Latin America and the Caribbean that causes Oropouche fever, a febrile illness that clinically resembles some other arboviral infections. It is currently spreading through Brazil and surrounding countries, where, from 1 January to 1 August 2024, more than 8000 cases have been identified in Bolivia, Brazil, Columbia, and Peru and for the first time in Cuba. Travelers with Oropouche fever have been identified in the United States and Europe. A significant occurrence during this epidemic has been the report of pregnant women infected with OROV who have had miscarriages and stillborn fetuses with placental, umbilical blood and fetal somatic organ samples that were RT-PCR positive for OROV and negative for other arboviruses. In addition, there have been four cases of newborn infants having microcephaly, in which the cerebrospinal fluid tested positive for IgM antibodies to OROV and negative for other arboviruses. This communication examines the biology, epidemiology, and clinical features of OROV, summarizes the 2023-2024 Oropouche virus epidemic, and describes the reported cases of vertical transmission and congenital infection, fetal death, and microcephaly in pregnant women with Oropouche fever, addresses experimental animal infections and potential placental pathology findings of OROV, and reviews other bunyavirus agents that can cause vertical transmission. Recommendations are made for pregnant women travelling to the regions affected by the epidemic.


Subject(s)
Bunyaviridae Infections , Infectious Disease Transmission, Vertical , Microcephaly , Orthobunyavirus , Placenta , Pregnancy Complications, Infectious , Stillbirth , Pregnancy , Female , Humans , Microcephaly/virology , Microcephaly/epidemiology , Orthobunyavirus/genetics , Orthobunyavirus/isolation & purification , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/epidemiology , Placenta/virology , Bunyaviridae Infections/transmission , Bunyaviridae Infections/epidemiology , Bunyaviridae Infections/virology , Infant, Newborn , Fetus/virology , Animals
3.
J Pediatr Pharmacol Ther ; 29(4): 429-433, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39144389

ABSTRACT

In recent years, rates of syphilis in adults have been on the rise resulting in an increase in the number of neonates born with congenital syphilis. National organizations including the Centers for Disease Control and Prevention as well as The US Preventative Services Task Force recommend routine testing of pregnant persons to identify and provide maternal syphilis treatment prior to delivery. Significant variability exists between states for these screenings, resulting in some pregnant persons not being diagnosed prior to delivery. The Pediatric Pharmacy Association (PPA) believes that pharmacists, along with other health care providers can help by ensuring optimal syphilis testing and treatment pathways for pregnant individuals and newborns are included in their workplaces. PPA also supports pharmacists working to increase treatment compliance by providing medication education and counseling regarding optimal treatment of syphilis infections, as well as work with state and local governments to standardize treatment recommendations.

4.
Neonatology ; : 1-5, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39137732

ABSTRACT

INTRODUCTION: Intrauterine herpes simplex virus (HSV) infection is uncommon and challenging to diagnose, requiring detection of HSV in skin lesions within 48 h post-birth. CASE PRESENTATION: A preterm female infant presented with the typical triad of blisters, microcephaly, and chorioretinitis, but the initial diagnostic approach was elusive due to negative results for TORCH pathogens from vesicles/serum. Referred at 7 months for developmental delay and epilepsy, her brain imaging showed calcification and cortical dysplasia. Polymerase chain reaction (PCR) of her preserved dried umbilical cord detected HSV-2 DNA, diagnosing intrauterine HSV infection. HSV-2 was later found in relapsed blisters at 8 months but not in cerebrospinal fluid or brain tissue. A literature review identified 104 congenital/intrauterine HSV cases; 28.8% presented the typical triad, and 50% were diagnosed using specimens collected 48 h post-birth. CONCLUSION: This case marks the first retrospective diagnosis of intrauterine HSV infection via PCR on preserved umbilical cord, underscoring its diagnostic value.

5.
Curr Res Microb Sci ; 7: 100257, 2024.
Article in English | MEDLINE | ID: mdl-39070527

ABSTRACT

Human cytomegalovirus is an ancient virus that has co-evolved with humans. It establishes a life-long infection in suspectable individuals for which there is no vaccination or cure. The virus can be transmitted to a developing fetus in seropositive pregnant women, and it is the leading cause of congenital infectious disease. While the majority of infected infants remain asymptomatic at birth, congenital cytomegalovirus infection can lead to substantial long-term neurodevelopmental impairments in survivors, resulting in considerable economic and social hardships. Recent discoveries regarding cytomegalovirus pathophysiology and viral replication cycles might enable the development of innovative diagnostics and therapeutics, including an effective vaccine. This Review will detail our understanding of human cytomegalovirus infection, with an in-depth discussion regarding the viral genome and transcriptome that contributes to its pathophysiology. The neonate's clinical course will also be highlighted, including maternal and neonatal testing, treatment recommendations, and long-term outcomes.

6.
Pathogens ; 13(7)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39057769

ABSTRACT

Neurodevelopmental outcomes for preschool-age children in the United States with in utero Zika virus (ZIKV) exposure have not yet been reported. We performed a case-control study to assess whether children exposed in utero to ZIKV have abnormal neurodevelopment at age 4-5 years compared to unexposed controls. Thirteen ZIKV-exposed cases that did not have microcephaly or other specific features of congenital Zika syndrome and 12 controls were evaluated between ages 4-5 years. Child neurodevelopment was assessed using the Pediatric Evaluation of Disability Inventory, Behavior Rating Inventory of Executive Function, Peabody Picture Vocabulary Test, Bracken School Readiness Assessment (BSRA), and Movement Assessment Battery for Children (MABC). Caregivers answered questions on the child's medical history and family demographics. Cases and controls were evaluated at mean (SD) ages 4.9 (0.3) and 4.8 (0.4) years, respectively. Caregivers reported more behavior and mood problems in cases than controls. MABC scores showed more gross and fine motor coordination difficulties among cases than controls. Controls trended towards higher performance on concepts underlying school readiness on BSRA. Three cases had a diagnosis of autism spectrum disorder or global developmental delay. Continued follow-up through school age for children with prenatal ZIKV exposure is needed to understand the impact of in utero ZIKV exposure on motor coordination, cognition, executive function, and academic achievement.

7.
Viruses ; 16(7)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39066285

ABSTRACT

Mpox (monkeypox) is a neglected tropical disease that has received increased attention since the multi-nation outbreak that began in 2022. The virus is endemic in West and Central Africa, where the Democratic Republic of the Congo (DRC) is the most affected country. Clade I monkeypox virus (MPXV) infection is endemic in the DRC and has an overall case fatality rate of 10.6% among children and adults. A study conducted in Sankuru Province, DRC, from 2007 to 2011 demonstrated that 75% of pregnant women with mpox had miscarriages or stillbirth. Further analysis of a stillborn fetus showed that MPXV could infect both the placenta and fetus, causing congenital infection. No additional cases of Clade I MPXV in pregnant women were reported until a new outbreak occurred in South Kivu Province during 2023 and 2024. Eight pregnant women having Clade I MPXV infection were identified, of whom four had either miscarriages or stillbirth, representing a 50% fetal mortality rate. These reports confirm previous data from the DRC that indicate the capability of Clade I MPXV to affect the fetus, causing congenital infection and fetal loss in a high percentage of cases. In this article, we review both past and new data from the DRC on the effects of Clade I MPXV during pregnancy and discuss the association of mpox with fetal loss.


Subject(s)
Abortion, Spontaneous , Disease Outbreaks , Mpox (monkeypox) , Pregnancy Complications, Infectious , Stillbirth , Humans , Female , Pregnancy , Stillbirth/epidemiology , Democratic Republic of the Congo/epidemiology , Abortion, Spontaneous/epidemiology , Adult , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/virology , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/virology , Monkeypox virus/genetics , Young Adult
9.
Diagnostics (Basel) ; 14(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39001326

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) is a neglected rodent-borne arenavirus, primarily spread by common house mouse species. Acquired human infections range from asymptomatic to mild flu-like symptoms and self-resolving neurological diseases. In contrast, intrauterine LCMV infection is associated with high mortality and morbidity. Infection of the fetus often leads to fetal death, and surviving fetuses may develop vision impairment and central nervous system developmental disorders. LCMV is mainly diagnosed by serological methods using in-house indirect immunofluorescence assays. LCMV nucleic acid is detected by the nested RT-PCR method and confirmed by Sanger sequencing. In Hungary, 23 acquired lymphocytic choriomeningitis cases were diagnosed between 2017 and 2023. Ten out of 23 confirmed patients proved to be positive by the PCR method. Two cases of intrauterine LCMV infections were detected in 2019 and 2021, respectively. The IgG antibody titers measured in the infant's serum samples were much higher than the IgG titers of the maternal serum samples. Both IgM and IgA antibodies were detectable in the infants' sera. As the microbiological diagnosis of LCMV is rather challenging and the symptoms are very similar to the clinical picture of other common teratogenic pathogens such as cytomegalovirus or Toxoplasma gondii, intrauterine LCMV infections might still be underdiagnosed.

11.
Annu Rev Virol ; 11(1): 363-379, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38848605

ABSTRACT

Interferon lambda (IFN-λ, type III IFN, IL-28/29) is a family of antiviral cytokines that are especially important at barrier sites, including the maternal-fetal interface. Recent discoveries have identified important roles for IFN-λ during pregnancy, particularly in the context of congenital infections. Here, we provide a comprehensive review of the activity of IFN-λ at the maternal-fetal interface, highlighting cell types that produce and respond to IFN-λ in the placenta, decidua, and endometrium. Further, we discuss the role of IFN-λ during infections with congenital pathogens including Zika virus, human cytomegalovirus, rubella virus, and Listeria monocytogenes. We discuss advances in experimental models that can be used to fill important knowledge gaps about IFN-λ-mediated immunity.


Subject(s)
Interferons , Placenta , Humans , Pregnancy , Female , Interferons/immunology , Placenta/immunology , Placenta/virology , Animals , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Interferon Lambda , Maternal-Fetal Exchange/immunology , Decidua/immunology , Virus Diseases/immunology , Virus Diseases/virology , Endometrium/immunology , Endometrium/virology , Antiviral Agents , Interleukins/immunology , Interleukins/metabolism
12.
J Pediatr ; 272: 114084, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38705230

ABSTRACT

OBJECTIVE: The objective of this study was to assess the role of T-lymphocyte immune responses in newborns with congenital cytomegalovirus (CMV) infection (cCMV) and their potential association with the development of long-term sequelae. STUDY DESIGN: A multicenter, prospective study from 2017 to 2022 was conducted across 8 hospitals in Spain. Blood samples were collected within the first month of life from neonates diagnosed with cCMV. Intracellular cytokine staining was employed to evaluate the presence of CMV-specific interferon-gamma (IFN-γ)-producing CD8+ and CD4+ T lymphocytes (CMV-IFN-γ-CD8+/CD4+) using flow cytometry. The development of sequelae, including hearing loss and neurologic impairment, was assessed during follow-up. RESULTS: In total, 64 newborns were included; 42 infants (65.6%) had symptomatic cCMV. The median age at the last follow-up visit was 25.3 months (IQR 20.1-34.4). Eighteen infants had long-term sequelae (28.1%), predominantly hearing loss (20.3%) and neurologic disorders (15.6%). No relationship was observed between total count or percentage of CMV-specific IFN-γ-CD8+ or CD4+ lymphocytes and long-term sequelae. Multivariable analysis demonstrated an association between lower total lymphocyte count and long-term sequelae (aOR 0.549, 95% CI: 0.323-0.833), which requires further study. CONCLUSIONS: CMV-specific IFN-γ-CD4+ and CD8+ T-lymphocyte responses in neonates with cCMV were not predictive of long-term sequelae.


Subject(s)
Cytomegalovirus Infections , Humans , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/complications , Infant, Newborn , Prospective Studies , Male , Female , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Spain , Interferon-gamma/blood , Infant , Follow-Up Studies , Immunity, Cellular , Cytomegalovirus/immunology , Hearing Loss/immunology
13.
BMC Genomics ; 25(1): 321, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38556880

ABSTRACT

Congenital infection caused by vertical transmission of microsporidia N. bombycis can result in severe economic losses in the silkworm-rearing industry. Whole-transcriptome analyses have revealed non-coding RNAs and their regulatory networks in N. bombycis infected embryos and larvae. However, transcriptomic changes in the microsporidia proliferation and host responses in congenitally infected embryos and larvae remains unclear. Here, we simultaneously compared the transcriptomes of N. bombycis and its host B. mori embryos of 5-day and larvae of 1-, 5- and 10-day during congenital infection. For the transcriptome of N. bombycis, a comparison of parasite expression patterns between congenital-infected embryos and larva showed most genes related to parasite central carbon metabolism were down-regulated in larvae during infection, whereas the majority of genes involved in parasite proliferation and growth were up-regulated. Interestingly, a large number of distinct or shared differentially expressed genes (DEGs) were revealed by the Venn diagram and heat map, many of them were connected to infection related factors such as Ricin B lectin, spore wall protein, polar tube protein, and polysaccharide deacetylase. For the transcriptome of B. mori infected with N. bombycis, beyond numerous DEGs related to DNA replication and repair, mRNA surveillance pathway, RNA transport, protein biosynthesis, and proteolysis, with the progression of infection, a large number of DEGs related to immune and infection pathways, including phagocytosis, apoptosis, TNF, Toll-like receptor, NF-kappa B, Fc epsilon RI, and some diseases, were successively identified. In contrast, most genes associated with the insulin signaling pathway, 2-oxacarboxylic acid metabolism, amino acid biosynthesis, and lipid metabolisms were up-regulated in larvae compared to those in embryos. Furthermore, dozens of distinct and three shared DEGs that were involved in the epigenetic regulations, such as polycomb, histone-lysine-specific demethylases, and histone-lysine-N-methyltransferases, were identified via the Venn diagram and heat maps. Notably, many DEGs of host and parasite associated with lipid-related metabolisms were verified by RT-qPCR. Taken together, simultaneous transcriptomic analyses of both host and parasite genes lead to a better understanding of changes in the microsporidia proliferation and host responses in embryos and larvae in N. bombycis congenital infection.


Subject(s)
Bombyx , Nosema , Animals , Transcriptome , Larva/genetics , Larva/metabolism , Histones/metabolism , Lysine/metabolism , Nosema/physiology , Gene Expression Profiling , Cell Proliferation , Lipids , Bombyx/genetics
14.
Am J Obstet Gynecol ; 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38527603

ABSTRACT

BACKGROUND: Cytomegalovirus is responsible for the most common congenital infection, affecting 0.5% to 1.0% of live births in Europe. Congenital cytomegalovirus infection can be diagnosed during pregnancy by viral DNA amplification in the amniotic fluid, but the prognosis of fetuses without severe brain abnormalities remains difficult to establish on the basis of prenatal imaging alone. OBJECTIVE: To identify predictors of moderate to severe symptomatic cytomegalovirus infection among fetal blood parameters and to propose an algorithm on the basis of these parameters and on prenatal imaging that would provide the best positive and negative predictive values. STUDY DESIGN: Fetal blood sampling at 21-28 weeks gestation was performed in fetuses with congenital cytomegalovirus infection confirmed by amniocentesis after maternal infection in the first-trimester or periconceptional period. We compared the levels of hemoglobin, thrombocytes, γ-glutamyl transpeptidase, aspartate aminotransferase, alanine aminotransferase, ß2-microglobulin, immunoglobulins G and M, and cytomegalovirus DNA viral loads in amniotic fluid and fetal blood between those with moderate to severe symptomatic infection and those with asymptomatic to mild infection (median follow-up of 36 months for live births). RESULTS: Among 58 fetuses included, 25 (43%) had a moderate to severe symptomatic infection: 16 with severe cerebral abnormalities, 5 with multiple signs or symptoms at birth, 2 with bilateral sensorineural hearing loss, and 2 with neurodevelopmental delay. The values of thrombocytes, aspartate aminotransferase, ß2 microglobulin, Immunoglobulin M, and cytomegalovirus viral loads differed significantly between fetuses with moderate to severe symptomatic infection and those with asymptomatic to mild infection. The optimal strategy to predict moderate to severe symptomatic infection was to first perform fetal brain imaging, followed by fetal blood sampling with the following cutoffs: thrombocytes <120,000/mL, viremia ≥5 log10/mL, and ß2 microglobulin ≥12 mg/L). This recursive algorithm had a negative predictive value of 100% for moderately to severely symptomatic infection. CONCLUSION: The combination of thrombocytes, ß2-microglobulin, and cytomegalovirus viral load in fetal blood can be used for prognosis determination, particularly in cytomegalovirus-infected fetuses without severe brain abnormalities at the time of prenatal diagnosis. Future studies should evaluate whether these parameters remain useful in infected fetuses who have been treated with valacyclovir before fetal blood sampling.

15.
Int J Mol Sci ; 25(5)2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38473883

ABSTRACT

Congenital cytomegalovirus (CMV) infection is the main cause of non-hereditary sensorineural hearing loss (SNHL). In order to shed light on SNHL pathophysiology, we examined the auditory pathway in CMV-infected fetuses; the temporal lobe, in particular the auditory cortex, and the inner ear. We investigated both inner ears and temporal lobes of 20 human CMV-infected fetuses at 21 weeks of gestation. As a negative group, five fetuses from spontaneous miscarriages without CMV infection were studied. Inner ears and temporal lobes were histologically examined, immunohistochemistry for CMV and CMV-PCR were performed. On the auditory cortex, we evaluated the local microglial reaction to the infection. CMV-positive cells were found in 14/20 brains and the damage was classified as severe, moderate, or mild, according to histological features. Fetuses with severe brain damage had a statistically higher temporal lobe viral load and a higher number of activated microglial cells in the auditory cortex compared to fetuses with mild brain damage (p: 0.01; p: 0.01). In the inner ears, the marginal cells of the stria vascularis were the most CMV positive. In our study, CMV affected the auditory pathway, suggesting a tropism for this route. In addition, in the auditory cortex, microglial activation may favor further tissue damage contributing to hearing loss.


Subject(s)
Cytomegalovirus Infections , Hearing Loss, Sensorineural , Humans , Cytomegalovirus , Auditory Pathways/pathology , Hearing Loss, Sensorineural/etiology , Fetus/pathology
16.
Virol J ; 21(1): 63, 2024 03 08.
Article in English | MEDLINE | ID: mdl-38459575

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) is one of the most important pathogens associated with congenital infection worldwide. Most congenital CMV-infected infants are asymptomatic at birth; however, some can develop delayed sequelae, especially hearing loss. METHODS: This study aimed to investigate the prevalence of congenital CMV infection in a neonatal intensive care unit in a low-income region of Brazil. The objectives extended to identifying associated factors, assessing the clinical status of infected newborns, and undertaking a two-year follow-up to discern potential long-term consequences in the affected infants. This cross-sectional prospective study enrolled newborns up to three weeks of life requiring intensive medical care. We employed a convenience sampling method to include 498 newborns and 477 mothers in the study. Categorical variables underwent analysis employing Fisher's exact test, whereas the examination of continuous variables involved the Mann‒Whitney test. RESULTS: CMV DNA was detected in saliva/urine samples from 6 newborns (1.21%), confirming congenital infection. We noted a significantly greater incidence (OR: 11.48; 95% CI: 2.519-52.33; p = 0.0094) of congenital infection among twins (7.14%) than among nontwins (0.66%). The twin patients exhibited discordant infection statuses, suggesting that only one of the babies tested positive for CMV. Most of the infected children were born to mothers who initiated sexual activity at a younger age (p = 0.0269). Only three out of the six newborns diagnosed with CMV infection underwent comprehensive clinical assessments and received continuous follow-up until they reached two years of age. Only one of the children had weight and height measurements below the norm for their age, coupled with developmental delays. CONCLUSIONS: The prevalence of congenital CMV infection among newborns admitted to the NICU was low and similar to that in the general population. However, we found a significantly greater incidence of congenital CMV infection in twins than in singletons. Interestingly, the twin-infected patients exhibited discordant infection statuses, suggesting that CMV was present in only one of the babies. We also found that most of the infected children were born to mothers who initiated sexual activity at a younger age. Diagnostic accessibility and comprehensive surveillance programs are imperative for effectively managing and preventing congenital CMV infections.


Subject(s)
Cytomegalovirus Infections , Intensive Care Units, Neonatal , Infant , Child , Female , Humans , Infant, Newborn , Brazil/epidemiology , Prevalence , Cross-Sectional Studies , Prospective Studies , Cytomegalovirus Infections/complications , Cytomegalovirus/genetics
17.
Aust N Z J Obstet Gynaecol ; 64(4): 354-360, 2024 08.
Article in English | MEDLINE | ID: mdl-38380539

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the indications for maternal TORCH (Toxoplasma gondii, rubella, cytomegalovirus (CMV), and herpes simplex virus (HSV)) serology, with a focus on the yield in isolated fetal growth restriction (FGR). MATERIALS AND METHODS: A retrospective review of antenatal TORCH testing between January 2014 and December 2018 was carried out at two hospitals in Melbourne, Australia. TORCH testing ordered for pregnancy losses and stillbirth was excluded. RESULTS: Medical records of 718 pregnancies were reviewed, representing 760 fetuses. Isolated FGR was the indication for TORCH screening in 71.2% of pregnancies. Screens ordered for isolated FGR were positive in 7.4% (95% CI 5.5-10.0%). There were 49 positive maternal immunoglobulin M (CMV = 34, Toxoplasma = 15). Two acute maternal infections during pregnancy were diagnosed (CMV = 1, Toxoplasma = 1), with both screens ordered to assess symptomatic maternal illness. There was one neonatal CMV infection, born to a woman with symptomatic primary CMV. No maternal or neonatal rubella or HSV infections were identified. We found a diagnostic yield of TORCH screening for isolated FGR of 0.0% (95% CI 0.00-0.8%). An estimated AUD$64 269.75 was expended on maternal TORCH screens in this study. CONCLUSION: Maternal TORCH testing for isolated FGR is of no diagnostic yield and should be abandoned.


Subject(s)
Cytomegalovirus Infections , Fetal Growth Retardation , Herpes Simplex , Pregnancy Complications, Infectious , Rubella , Humans , Female , Fetal Growth Retardation/diagnosis , Pregnancy , Retrospective Studies , Adult , Pregnancy Complications, Infectious/diagnosis , Rubella/diagnosis , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/congenital , Herpes Simplex/diagnosis , Toxoplasmosis/diagnosis , Australia , Prenatal Diagnosis
18.
J Clin Microbiol ; 62(4): e0031323, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38391188

ABSTRACT

Human cytomegalovirus (CMV) is the leading cause of congenital infection worldwide and the most common cause of non-genetic sensorineural hearing loss. As there is no vaccine or other specific intervention to prevent congenital CMV infection, there is a need to identify maternal and congenital infections with sensitive and specific testing as early as possible. There is no widely accepted practice for screening during pregnancy or in all newborns for identification of possible cases of congenital CMV. Currently, screening during pregnancy is limited to those identified as at risk followed by fetal and/or neonatal testing when congenital infection is suspected. This review focuses primarily on the current status of laboratory testing for diagnosis of maternal and congenital CMV infections. Primary maternal infection is best diagnosed using serologic testing, including CMV IgM, IgG, and avidity testing, while fetal infection should be assessed by nucleic acid amplification testing (NAAT) of amniotic fluid. Urine and saliva NAATs are the mainstay for diagnosis of congenital CMV in the first 3 weeks of life. Testing of dried blood spots can be useful for diagnosis of congenital CMV outside of the newborn period. The gaps in knowledge such as the prognostic value of viral loads in various sample types are addressed.


Subject(s)
Cytomegalovirus Infections , Fetal Diseases , Pregnancy Complications, Infectious , Pregnancy , Female , Humans , Infant, Newborn , Cytomegalovirus/genetics , Pregnancy Complications, Infectious/diagnosis , Prognosis , Fetal Diseases/diagnosis
19.
J Pediatr ; 268: 113945, 2024 May.
Article in English | MEDLINE | ID: mdl-38336204

ABSTRACT

OBJECTIVE: To assess the efficacy of valganciclovir in infants with hearing loss and clinically inapparent congenital cytomegalovirus infection (cCMV), as there is no consensus on treatment of this group. STUDY DESIGN: A nationwide, nonrandomized controlled trial, comparing 6 weeks of oral valganciclovir to no treatment in infants with cCMV, recruited after newborn hearing screening resulted in referral to an audiologist. The choice whether to treat was left to parents of subjects. Eligible subjects were full term infants aged <13 weeks with sensorineural hearing loss and diagnosed with cCMV through dried blood spot testing. The primary outcome, measured by linear and ordinal logistic regression, was change in best-ear hearing from baseline to follow-up at 18-22 months of age. RESULTS: Thirty-seven participants were included in the final analysis, of whom 25 were in the treatment group and 12 in the control group. The majority of subjects in both groups had neuroimaging abnormalities, which were mostly mild. Hearing deterioration was more likely in the control group compared with the treatment group (common OR 0.10, 95% CI 0.02-0.45, P = .003). Mean best-ear hearing deteriorated by 13.7 dB in the control group, compared with improvement of 3.3 dB in the treatment group (difference 17 dB, 95% CI 2.6 - 31.4, P = .02). CONCLUSIONS: We investigated treatment in children with hearing loss and clinically inapparent cCMV. Although our study was nonrandomized, it is the first prospective and controlled trial in this population. Valganciclovir-treated children with hearing loss and inapparent cCMV had less hearing deterioration at 18 through 22 months of age than control subjects. EUDRACT REGISTRY NUMBER: 2013-003068-30.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Hearing Loss, Sensorineural , Valganciclovir , Humans , Valganciclovir/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/complications , Antiviral Agents/therapeutic use , Male , Female , Infant , Infant, Newborn , Hearing Loss, Sensorineural/drug therapy , Treatment Outcome , Ganciclovir/analogs & derivatives , Ganciclovir/therapeutic use , Neonatal Screening , Prospective Studies , Follow-Up Studies , Administration, Oral
20.
J Pediatr ; 268: 113934, 2024 May.
Article in English | MEDLINE | ID: mdl-38309519

ABSTRACT

OBJECTIVE: The objective of this study was to determine if valganciclovir initiated after 1 month of age improves congenital cytomegalovirus-associated sensorineural hearing loss. STUDY DESIGN: We conducted a randomized, double-blind, placebo-controlled phase 2 trial of 6 weeks of oral valganciclovir at US (n = 12) and UK (n = 9) sites. Patients of ages 1 month through 3 years with baseline sensorineural hearing loss were enrolled. The primary outcome was change in total ear hearing between baseline and study month 6. Secondary outcome measures included change in best ear hearing and reduction in cytomegalovirus viral load in blood, saliva, and urine. RESULTS: Of 54 participants enrolled, 35 were documented to have congenital cytomegalovirus infection and were randomized (active group: 17; placebo group: 18). Mean age at enrollment was 17.8 ± 15.8 months (valganciclovir) vs 19.5 ± 13.1 months (placebo). Twenty (76.9%) of the 26 ears from subjects in the active treatment group did not have worsening of hearing, compared with 27 (96.4%) of 28 ears from subjects in the placebo group (P = .09). All other comparisons of total ear or best ear hearing outcomes were also not statistically significant. Saliva and urine viral loads decreased significantly in the valganciclovir group but did not correlate with change in hearing outcome. CONCLUSIONS: In this randomized controlled trial, initiation of antiviral therapy beyond the first month of age did not improve hearing outcomes in children with congenital cytomegalovirus-associated sensorineural hearing loss. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT01649869.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Ganciclovir , Hearing Loss, Sensorineural , Valganciclovir , Humans , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/complications , Valganciclovir/therapeutic use , Valganciclovir/administration & dosage , Hearing Loss, Sensorineural/drug therapy , Hearing Loss, Sensorineural/virology , Hearing Loss, Sensorineural/etiology , Antiviral Agents/therapeutic use , Antiviral Agents/administration & dosage , Male , Female , Double-Blind Method , Infant , Administration, Oral , Ganciclovir/analogs & derivatives , Ganciclovir/therapeutic use , Ganciclovir/administration & dosage , Child, Preschool , Treatment Outcome , Viral Load , Infant, Newborn
SELECTION OF CITATIONS
SEARCH DETAIL