1
|
Shim GH. Treatment of congenital cytomegalovirus infection. Clin Exp Pediatr 2023; 66:384-394. [PMID: 36596746 PMCID: PMC10475861 DOI: 10.3345/cep.2022.01032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/26/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022] Open
Abstract
Congenital cytomegalovirus (CMV) is the most common cause of congenital infection worldwide, the most common nongenetic cause of sensorineural hearing loss in children, and a cause of neurodevelopmental disorders in the brain. Infants with symptomatic congenital CMV infection may benefit from hearing and neurodevelopmental outcomes, particularly if antiviral treatment is initiated within the first month of life. Infants with life-threatening symptoms are recommended to receive 2-6 weeks of intravenous ganciclovir and then switch to oral valganciclovir, and those without life-threatening symptoms are recommended to use oral valganciclovir during the entire 6-month period. During antiviral drug treatment, absolute neutrophil count, platelet count, blood urea nitrogen, creatinine, and liver function tests were performed to identify neutropenia, thrombocytopenia, renal failure, and liver failure. This review investigated the evidence to date of treating congenital CMV infection.
Collapse
Affiliation(s)
- Gyu Hong Shim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| |
Collapse
|
2
|
Piccirilli G, Gabrielli L, Bonasoni MP, Chiereghin A, Turello G, Borgatti EC, Simonazzi G, Felici S, Leone M, Salfi NCM, Santini D, Lazzarotto T. Fetal Brain Damage in Human Fetuses with Congenital Cytomegalovirus Infection: Histological Features and Viral Tropism. Cell Mol Neurobiol 2023; 43:1385-1399. [PMID: 35933637 PMCID: PMC10006254 DOI: 10.1007/s10571-022-01258-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 07/08/2022] [Indexed: 11/24/2022]
Abstract
Human cytomegalovirus (HCMV) causes congenital neurological lifelong disabilities. To date, the neuropathogenesis of brain injury related to congenital HCMV (cCMV) infection is poorly understood. This study evaluates the characteristics and pathogenetic mechanisms of encephalic damage in cCMV infection. Ten HCMV-infected human fetuses at 21 weeks of gestation were examined. Specifically, tissues from different brain areas were analyzed by: (i) immunohistochemistry (IHC) to detect HCMV-infected cell distribution, (ii) hematoxylin-eosin staining to evaluate histological damage and (iii) real-time PCR to quantify tissue viral load (HCMV-DNA). The differentiation stage of HCMV-infected neural/neuronal cells was assessed by double IHC to detect simultaneously HCMV-antigens and neural/neuronal markers: nestin (a marker of neural stem/progenitor cells), doublecortin (DCX, marker of cells committed to the neuronal lineage) and neuronal nuclei (NeuN, identifying mature neurons). HCMV-positive cells and viral DNA were found in the brain of 8/10 (80%) fetuses. For these cases, brain damage was classified as mild (n = 4, 50%), moderate (n = 3, 37.5%) and severe (n = 1, 12.5%) based on presence and frequency of pathological findings (necrosis, microglial nodules, microglial activation, astrocytosis, and vascular changes). The highest median HCMV-DNA level was found in the hippocampus (212 copies/5 ng of human DNA [hDNA], range: 10-7,505) as well as the highest mean HCMV-infected cell value (2.9 cells, range: 0-23), followed by that detected in subventricular zone (1.7 cells, range: 0-19). These findings suggested a preferential viral tropism for both neural stem/progenitor cells and neuronal committed cells, residing in these regions, confirmed by the expression of DCX and nestin in 94% and 63.3% of HCMV-positive cells, respectively. NeuN was not found among HCMV-positive cells and was nearly absent in the brain with severe damage, suggesting HCMV does not infect mature neurons and immature neural/neuronal cells do not differentiate into neurons. This could lead to known structural and functional brain defects from cCMV infection.
Collapse
Affiliation(s)
- Giulia Piccirilli
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Liliana Gabrielli
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | | | - Angela Chiereghin
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Gabriele Turello
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Eva Caterina Borgatti
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Giuliana Simonazzi
- Department of Obstetrics and Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Silvia Felici
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Marta Leone
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | | | - Donatella Santini
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Tiziana Lazzarotto
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
3
|
Congenital Cytomegalovirus in Algiers, Algeria, a Descriptive Case Series Study. JOURNAL OF MEDICAL MICROBIOLOGY AND INFECTIOUS DISEASES 2022. [DOI: 10.52547/jommid.10.2.87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
4
|
Geris JM, Spector LG, Roesler M, Hernandez-Alvarado N, Blackstad M, Nelson HH, Schleiss MR. High prevalence of asymptomatic CMV shedding in healthy children attending the minnesota state fair. J Clin Virol 2022; 148:105102. [PMID: 35158280 PMCID: PMC8918022 DOI: 10.1016/j.jcv.2022.105102] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Young children in the household are a known risk factor for maternal CMV infection and consequently, congenital infection in infants. However, little is known about viral shedding in pre-school aged children. OBJECTIVES To estimate the prevalence of CMV DNA shedding and CMV antibodies among healthy children and their mothers. STUDY DESIGN A study of children ages 0 through 5 years was undertaken at the 2019 Minnesota State Fair. Children and their mothers were assessed for CMV shedding by procurement of a saliva swab for CMV PCR testing. An optional finger-stick for capillary blood was used to assess CMV antibodies. RESULTS A total of 109 children and 85 mothers were enrolled. The prevalence of CMV saliva shedding among children (mean age 3.1 years, SE=0.16) and their mothers was 12/109 (11.0%) and 1/85 (1.2%), respectively. The prevalence of CMV DNA among children peaked at 3 years of age (26%) while the mean viral load was greatest at one year of age (236,693 IU/mL). CMV IgG antibodies among those who agreed to a finger-stick were detected in 16/35 mothers (45.7%) and 0/7 children (0%). Mothers of children aged 5 years or greater had the highest seroprevalence (61.5%). CONCLUSIONS The prevalence of CMV salivary shedding in this unselected sample of young children was approximately 11.0%. The overall maternal seroprevalence in our sample was <50%, suggesting these women are at risk for acquisition of a primary CMV infection in subsequent pregnancies.
Collapse
Affiliation(s)
- Jennifer M Geris
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA; Institute for Molecular Virology, University of Minnesota, Minneapolis MN, USA.
| | - Logan G Spector
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Michelle Roesler
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Nelmary Hernandez-Alvarado
- Division of Pediatric Infectious Diseases and Immunology, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Mark Blackstad
- Division of Pediatric Infectious Diseases and Immunology, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Heather H Nelson
- Division of Epidemiology & Community Health, University of Minnesota School of Public Health, Minneapolis MN, USA
| | - Mark R Schleiss
- Institute for Molecular Virology, University of Minnesota, Minneapolis MN, USA; Division of Pediatric Infectious Diseases and Immunology, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| |
Collapse
|
5
|
Kim JH, Roh KJ, Nam GS, Son EJ. Audiologic Status of Children with Confirmed Cytomegalovirus Infection: a Case Series. J Korean Med Sci 2020; 35:e244. [PMID: 32743988 PMCID: PMC7402926 DOI: 10.3346/jkms.2020.35.e244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/04/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) infection is the most common non-genetic cause of sensorineural hearing loss (SHNL) in children. Only about 10% to 15% of children with congenital CMV are symptomatic, and most are not diagnosed at birth. About 7% to 15% of clinically asymptomatic patients may develop later complications, including SNHL, which is the most common sequela in clinically asymptomatic patients. In this study, hearing status was investigated in children with confirmed CMV infection and neonatal hearing screening (NHS) histories were reviewed to explore hearing loss caused by CMV. METHODS The medical records of 58 children who were diagnosed with confirmed CMV infection were reviewed for clinical symptoms and signs of CMV infection. Hearing status was evaluated with age-appropriate audiological test batteries. RESULTS A total of 58 children (M:F = 32:26 patients; age at study: mean, 5.62 years, range, 1-10 years) were diagnosed serologically with CMV infection (14 patients, 21.1%), or diagnosed via PCR of serum (5, 7.9%) and/or PCR from urine (19, 26.8%). Hearing loss was confirmed in 11 children (19.0%), being bilateral in 6 (54.5%), and unilateral in 5 (45.5%). Note that 7 of 17 ears with hearing loss passed NHS and were diagnosed only after re-evaluation when CMV infection was identified. CONCLUSION Hearing loss is a serious complication of CMV infection in children. Our results highlight the importance of timely audiological evaluation in children with clinically symptomatic CMV infection even if they pass NHS.
Collapse
Affiliation(s)
- Ji Hyung Kim
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Jin Roh
- Department of Otorhinolaryngology, Inje University Seoul Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Gi Sung Nam
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jin Son
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Department of Otorhinolaryngology, Yongin Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Turriziani Colonna A, Buonsenso D, Pata D, Salerno G, Chieffo DPR, Romeo DM, Faccia V, Conti G, Molle F, Baldascino A, De Waure C, Acampora A, Luciano R, Santangelo R, Valentini P. Long-Term Clinical, Audiological, Visual, Neurocognitive and Behavioral Outcome in Children With Symptomatic and Asymptomatic Congenital Cytomegalovirus Infection Treated With Valganciclovir. Front Med (Lausanne) 2020; 7:268. [PMID: 32793607 PMCID: PMC7393008 DOI: 10.3389/fmed.2020.00268] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/15/2020] [Indexed: 11/25/2022] Open
Abstract
Cytomegalovirus (CMV) is the most common cause of congenital infection in humans. There are no enough data on long-term outcome of newborns with congenital CMV (cCMV) infection, particularly for those asymptomatic at birth. For this reason, we performed this study to evaluate long-term audiological, visual, neurocognitive, and behavioral outcome in patients with symptomatic and asymptomatic cCMV infection treated with oral Valganciclovir (VGC). Thirty-six newborns with confirmed cCMV infection were evaluated: 12 (33.3%) symptomatic at birth and 24 asymptomatic (66.7%). No one had cognitive impairment. Cognitive assessment scales resulted abnormal in 4/35 patients (11.4%). 11/21 patients (52.4%) achieved abnormal scores in neuropsychological tests. The language evaluation gave pathological results in 6/21 (28.5%) patients. 6/35 patients (17.1%) developed SNHL, all symptomatic at birth except one. None of the 34 patients evaluated developed CMV retinopathy. Our study shows that both symptomatic and asymptomatic newborns with cCMV infection develop long-term sequelae, particularly in the behavioral and communicative areas, independently from the trimester of maternal infection.
Collapse
Affiliation(s)
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Pata
- Istituto di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gilda Salerno
- Istituto di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Daniela P R Chieffo
- Istituto di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy.,Istituto di Neuropsichiatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico M Romeo
- Istituto di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy.,Istituto di Neuropsichiatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valerio Faccia
- Unit of Pediatrics, Department of Gynecologic, Pediatric and Neonatologic Sciences, "Sant'Andrea" University Hospital, University "Sapienza" of Rome, Rome, Italy
| | - Guido Conti
- Department of Aging, Neurologic, Orthopedic and Head and Neck Science, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.,Istituto di Otorinolaringoiatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fernando Molle
- Department of Aging, Neurologic, Orthopedic and Head and Neck Science, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.,Istituto di Oculistica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Baldascino
- Department of Aging, Neurologic, Orthopedic and Head and Neck Science, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.,Istituto di Oculistica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara De Waure
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Anna Acampora
- Istituto di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy.,Istituto di Igiene, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rita Luciano
- Istituto di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rosaria Santangelo
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Microbiology and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Piero Valentini
- Istituto di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
7
|
Sohal K, Moshy J, Owibingire S, Shuaibu I. Hearing loss in children: A review of literature. JOURNAL OF MEDICAL SCIENCES 2020. [DOI: 10.4103/jmedsci.jmedsci_166_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
8
|
Chen SJ, Wang SC, Chen YC. Antiviral Agents as Therapeutic Strategies Against Cytomegalovirus Infections. Viruses 2019; 12:v12010021. [PMID: 31878068 PMCID: PMC7019738 DOI: 10.3390/v12010021] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/17/2022] Open
Abstract
Cytomegalovirus (CMV) is a threat to human health in the world, particularly for immunologically weak patients. CMV may cause opportunistic infections, congenital infections and central nervous system infections. CMV infections are difficult to treat due to their specific life cycles, mutation, and latency characteristic. Despite recent advances, current drugs used for treating active CMV infections are limited in their efficacy, and the eradication of latent infections is impossible. Current antiviral agents which target the UL54 DNA polymerase are restricted because of nephrotoxicity and viral resistance. CMV also cannot be prevented or eliminated with a vaccine. Fortunately, letermovir which targets the human CMV (HCMV) terminase complex has been recently approved to treat CMV infections in humans. The growing point is developing antiviral agents against both lytically and latently infected cells. The nucleic acid-based therapeutic approaches including the external guide sequences (EGSs)-RNase, the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system and transcription activator-like effector nucleases (TALENs) are being explored to remove acute and/or latent CMV infections. HCMV vaccine is being developed for prophylaxis. Additionally, adoptive T cell therapy (ACT) has been experimentally used to combate drug-resistant and recurrent CMV in patients after cell and/or organ transplantation. Developing antiviral agents is promising in this area to obtain fruitful outcomes and to have a great impact on humans for the therapy of CMV infections.
Collapse
Affiliation(s)
- Shiu-Jau Chen
- Department of Neurosurgery, Mackay Memorial Hospital, Taipei 10491, Taiwan;
- Department of Medicine, Mackay Medicine College, Taipei 25245, Taiwan
| | - Shao-Cheng Wang
- Jianan Psychiatric Center, Ministry of Health and Welfare, Tainan 71742, Taiwan;
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Yuan-Chuan Chen
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
- Correspondence:
| |
Collapse
|
9
|
McCrary H, Sheng X, Greene T, Park A. Long-term hearing outcomes of children with symptomatic congenital CMV treated with valganciclovir. Int J Pediatr Otorhinolaryngol 2019; 118:124-127. [PMID: 30611098 PMCID: PMC6363845 DOI: 10.1016/j.ijporl.2018.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Congenital human cytomegalovirus (cCMV) is a leading cause of pediatric hearing loss. Recent literature has suggested that valganciclovir (VGCV) therapy can improve hearing outcomes. The objective of this study was to evaluate the long-term hearing outcomes among symptomatic CMV patients treated with VGCV. METHODS A retrospective chart review of symptomatic CMV patients treated with VGCV was completed. The primary endpoint was the change in best ear hearing scores prior to treatment and after follow-up audiograms. A paired-sample t-test was used to evaluate the data. RESULTS A total of 16 children were included in the study and participants were followed for an average of 3.2 years. There was a measurable worsening, but not a statistically significant change in the best ear hearing scores, where the mean change was 11.9 dB (p-value = 0.070). However, 14/16 patients (87.5%, p-value<0.001) were found to have clinically significant worsening of hearing. The mean change in hearing scores for the left and right ear was 14.2 dB (p-value = 0.023) and 15.5 dB (p-value = 0.032), respectively. Mean elapsed time for progressive loss was 2.6 ± 0.2 years. When comparing the better or worse ear, there was no pattern for which ear deteriorated earlier or more frequently. CONCLUSIONS Our data did show a measurable, but not a statistically significant worsening outcome in best ear hearing. There was a significant change in both left and right ear hearing. Our results suggest that VGCV may provide only a short-term improvement in hearing outcomes; however, these preliminary post-hoc findings suggest the need for a more rigorous evaluation.
Collapse
Affiliation(s)
- Hilary McCrary
- University of Utah School of Medicine, Division of Otolaryngology - Head and Neck Surgery, Utah, United States
| | - Xiaoming Sheng
- University of Utah School of Nursing, Utah, United States
| | - Tom Greene
- University of Utah School of Medicine, Department of Population Health Sciences, Division of Biostatistics, Utah, United States
| | - Albert Park
- University of Utah School of Medicine, Division of Otolaryngology - Head and Neck Surgery, Utah, United States.
| |
Collapse
|
10
|
Pilot Study of Model-Based Dosage Individualization of Ganciclovir in Neonates and Young Infants with Congenital Cytomegalovirus Infection. Antimicrob Agents Chemother 2018; 62:AAC.00075-18. [PMID: 29507070 DOI: 10.1128/aac.00075-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/28/2018] [Indexed: 01/20/2023] Open
Abstract
Newborns with congenital cytomegalovirus (CMV) infection are at high risk for developing permanent sequelae. Intravenous ganciclovir therapy is frequently used for the treatment of congenital CMV infection. A target area under the concentration-time curve from 0 to 24 h (AUC0-24) of 40 to 50 μg · h/ml is recommended. The standard dose has resulted in a large variability in ganciclovir exposure in newborns, indicating the unmet need of dosage individualization for this vulnerable population, but the implementation of this strategy remains challenging in clinical practice. We aim to evaluate the clinical utility of model-based dosage individualization of ganciclovir in newborns using an opportunistic sampling approach. The predictive performance of a published ganciclovir population pharmacokinetic model was evaluated using an independent patient cohort. The individual dose was adjusted based on the target AUC0-24 to ensure its efficacy. A total of 26 newborns with congenital CMV infection were included in the present study. Only 11 (42.3%) patients achieved the target AUC0-24 after being given the standard dose. For all the subtherapeutic patients (achieving <80% of the target AUC) (n = 5), a model-based dosage adjustment was performed using the Bayesian estimation method combined with the opportunistic sampling strategy. The adjusted doses were increased by 28.6% to 60.0% in these five patients, and all adapted AUC0-24 values achieved the target (range, 48.6 to 66.1 μg · h/ml). The clinical utility of model-based dosing individualization of ganciclovir was demonstrated in newborns with congenital CMV infection. The population pharmacokinetic model combined with the opportunistic sampling strategy provides a clinically feasible method to adapt the ganciclovir dose in neonatal clinical practice. (This study has been registered at ClinicalTrials.gov under registration no. NCT03113344.).
Collapse
|
11
|
Humanes Cytomegalievirus (HCMV). Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2018; 61:116-128. [DOI: 10.1007/s00103-017-2661-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
12
|
Thackeray R, Magnusson BM, Christensen EM. Effectiveness of message framing on women's intention to perform cytomegalovirus prevention behaviors: a cross-sectional study. BMC WOMENS HEALTH 2017; 17:134. [PMID: 29262815 PMCID: PMC5738799 DOI: 10.1186/s12905-017-0492-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/05/2017] [Indexed: 11/10/2022]
Abstract
Background The purpose of this study was to evaluate the effect of message framing on women’s intention to perform cytomegalovirus (CMV) prevention behaviors involving handwashing, not sharing food and eating utensils, not kissing a child on the lips and not placing a pacifier in the mouth after it was in a child’s mouth. Methods An online panel of women 18–40 years, who were pregnant or planning a pregnancy were randomized in a 2 × 2 factorial design to receive 1 of 4 CMV fact sheets. The fact sheets were framed as either what could be gained or be lost by following (or not) the recommendations and the likelihood of being affected by CMV (i.e., small chance or one of the most common infections in infants). The questionnaire measured CMV knowledge, participation in CMV risk or prevention behaviors, perceived severity of and susceptibly to CMV, and the perceived control over and the efficacy of recommended prevention behaviors. The dependent variable, intention to modify behavior, was an index score that ranged from 0 to 16 with higher values indicating greater intention. Linear regression was used to evaluate the association between all independent variables and overall behavioral intention. Results The sample included 840 women; 15.5% were familiar with CMV. Behavioral intention was high (M = 10.43; SD = 5.13) but did not differ across the message frames (p = 0.23). Overall, behavioral intention was predicted by CMV knowledge, message credibility, perceived severity of CMV, perceived behavioral control and response efficacy. Significant interactions with gain vs. loss frame were observed for perceived behavioral control (p = 0.03) and response efficacy (p = .003). Conclusions Framing CMV messages by what women stand to gain or lose interacts with perceived behavioral control and response efficacy to influence behavioral intention. Perceived behavioral control and response efficacy were most predictive of behavioral intention overall regardless of frame. Messaging that focuses on these two variables, particularly for avoiding kissing a child on the lips and sharing food, cups and utensils, may result in greater gains in intention to participate in CMV prevention behaviors.
Collapse
Affiliation(s)
- Rosemary Thackeray
- Department of Health Science, Brigham Young University, 4103 LSB, Provo, UT, 84602, USA.
| | - Brianna M Magnusson
- Department of Health Science, Brigham Young University, 4103 LSB, Provo, UT, 84602, USA
| | - Emily M Christensen
- Department of Health Science, Brigham Young University, 4103 LSB, Provo, UT, 84602, USA
| |
Collapse
|
13
|
Thackeray R, Magnusson BM. Women's attitudes toward practicing cytomegalovirus prevention behaviors. Prev Med Rep 2016; 4:517-524. [PMID: 27747148 PMCID: PMC5061468 DOI: 10.1016/j.pmedr.2016.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/19/2016] [Accepted: 09/25/2016] [Indexed: 11/28/2022] Open
Abstract
Congenital cytomegalovirus (CMV) infection causes severe disabilities and developmental delays. Women's awareness of CMV is low. Only about half of healthcare providers report counseling women about behaviors to reduce CMV risk and public health education is limited. Routine CMV counseling is not recommend. Providers may lack time to counsel women; other conditions may take priority for counseling; there may be a perception that women are reluctant to follow advice. This cross-sectional descriptive study examined women's attitudes toward CMV prevention behaviors. Data were collected from an online panel of 840 U.S. women 18–40 years of age, who had a child < 5 years of age, and were pregnant or planning a pregnancy in the next 12 months. Questions assessed CMV awareness, frequency of past behaviors that transmit CMV, and attitudes toward eight CMV prevention behaviors. Only 15.5% of women were somewhat or very familiar with CMV. Very few women (6.1%) reported hearing from their provider about CMV. Women held positive attitudes toward the CMV prevention behaviors and perceived them as feasible. Least positive attitudes were toward not kissing a child on the lips and not sharing foods. Predictors of positive attitudes were CMV awareness, past behavior, talking to a healthcare provider, and perceived risk reduction. Healthcare providers and public health practitioners should collaborate to increase CMV awareness. Encouraging behaviors to reduce saliva sharing may result in greater gains in reducing CMV infection. Overall, women hold favorable attitudes toward CMV prevention behaviors. Women hold less favorable attitudes toward not kissing a child on the lips. Women also hold less favorable attitudes toward not sharing food with a child. Positive attitudes predicted by belief that their risk of infection would be reduced. Providers and public health practitioners need to work together to increase awareness.
Collapse
Affiliation(s)
- Rosemary Thackeray
- Brigham Young University, Department of Health Science, 4103 LSB, Provo, UT 84602, United States
| | - Brianna M Magnusson
- Brigham Young University, Department of Health Science, 4103 LSB, Provo, UT 84602, United States
| |
Collapse
|
14
|
Reuschel E, Barabas S, Zeman F, Bendfeldt H, Rascle A, Deml L, Seelbach-Goebel B. Functional impairment of CMV-reactive cellular immunity during pregnancy. J Med Virol 2016; 89:324-331. [PMID: 27447923 DOI: 10.1002/jmv.24639] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2016] [Indexed: 01/14/2023]
Abstract
Cytomegalovirus (CMV) is the most common congenital viral infection. Mother-to-child transmission can cause severe child disability. Intact CMV-specific cell-mediated immunity (CMI) was shown to prevent uncontrolled replication in healthy individuals. This study aimed to determine whether CMV-specific CMI is impaired in pregnant women, thus potentially increasing the overall risk for active CMV replication and transmission. CMV-specific CMI in peripheral blood of 60 pregnant women was determined using T-Track® CMV for detection of IE-1 and pp65-reactive effector cells by IFN-γ ELISpot, and compared to the CMV-IgG and -IgM serostatus. CMV-specific CMI was detected in 65% of CMV-seropositive pregnant women. Five percent of CMV-IgG seronegative women showed IE-1- but not pp65-reactive cells. The overall number of CMV-reactive cells in pregnant women was significantly lower compared to a matched non-pregnant control group (P < 0.001). No significant difference in CMV-specific CMI was detected in the course of the three trimesters of pregnancy of CMV-IgG seropositive women. Postpartum (median days postnatal = 123), IE-1- and pp65-specific CMI remained significantly lower than in the non-pregnant control group (P < 0.001 and 0.0032, respectively). Functional analysis of CMV-reactive immune cells using T-Track® CMV therefore suggests a systemic down-regulation of CMV-specific CMI in pregnant women. Further studies are needed to investigate whether this may be indicative of a higher susceptibility to CMV reactivation or transmission. J. Med. Virol. 89:324-331, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Edith Reuschel
- Department of Obstetrics and Gynecology, University of Regensburg, Hospital of the Barmherzige Brueder, Regensburg, Germany
| | | | - Florian Zeman
- Center for Clinical Studies, University of Regensburg, Regensburg, Germany
| | | | - Anne Rascle
- Lophius Biosciences GmbH, Regensburg, Germany
| | - Ludwig Deml
- Lophius Biosciences GmbH, Regensburg, Germany
| | - Birgit Seelbach-Goebel
- Department of Obstetrics and Gynecology, University of Regensburg, Hospital of the Barmherzige Brueder, Regensburg, Germany
| |
Collapse
|
15
|
Abstract
Throughout pregnancy, the placenta acts as a physical and immunological barrier against the hematogenous transmission of viruses from mother to fetus. Despite this, very little is known regarding the specific mechanisms by which the placenta shields the developing fetus from viral infections or about the strategies utilized by select viruses to bypass and/or weaken the placental barrier. In this review, we summarize studies regarding virus-host interactions at the placental interface and explore key areas for future investigation. We focus our review on placental trophoblasts, which form the barrier between maternal and fetal circulations and thus govern the cross talk between the maternal and fetal microenvironments.
Collapse
Affiliation(s)
- Elizabeth Delorme-Axford
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania 15219;
| | - Yoel Sadovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania 15219; .,Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Science, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Carolyn B Coyne
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania 15219; .,Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Science, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| |
Collapse
|
16
|
Reitter A, Buxmann H, Haberl AE, Schlösser R, Kreibich M, Keppler OT, Berger A. Incidence of CMV co-infection in HIV-positive women and their neonates in a tertiary referral centre: a cohort study. Med Microbiol Immunol 2015; 205:63-71. [PMID: 26155982 DOI: 10.1007/s00430-015-0427-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/29/2015] [Indexed: 01/21/2023]
Abstract
Co-infection with CMV in HIV-positive pregnant women is associated with perinatal mother-to-child transmission (MTCT) of both viruses. This retrospective study reports on the incidence of maternal and neonatal CMV (presence of anti-CMV IgG and IgM, CMV DNA PCR and/or CMV virus isolation) in high-risk pregnancies due to maternal HIV infection, MTCT of HIV and/or CMV. One hundred and eleven maternal samples and 75 matched neonatal samples were available for HIV and subsequent CMV testing. In this cohort of HIV-positive pregnant women, 96 (86.5 %) serum samples were anti-CMV IgG positive. In nine (9.4 %) of these, anti-CMV IgM was detected, and in none of them a maternal primary CMV infection was suspected. Fifty-seven (51.8 %) maternal serum samples were tested retrospectively by CMV DNA PCR; one sample was positive (0.9 %). All matched neonates were tested for HIV by PCR in the first month of life; HIV transmission was detected in one case. In 74 (67.2 %) of neonates, CMV testing was performed. Sixty-six of these serum samples were tested retrospectively by CMV DNA PCR. Two newborns (2.7 %) showed laboratory markers for CMV infection (one by detection of CMV DNA in plasma, and one by isolation of CMV from a urine sample). In the follow-up, neither of these two showed clinical signs for active CMV disease. We discussed these findings in the light of the national official guidelines. All CMV transmissions occurred due to maternal reinfection or endogenous reactivation. This suggests the success of highly active antiretroviral therapy in preventing MTCT of HIV and CMV disease and highlights the importance of adequate care and follow-up.
Collapse
Affiliation(s)
- A Reitter
- Department of Obstetrics and Gynaecology, University Hospital Frankfurt, Goethe-University, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany.
| | - H Buxmann
- Division of Neonatology, Department of Pediatrics, University Hospital Frankfurt, Goethe-University, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany
| | - A E Haberl
- Department of Infectious Diseases, University Hospital Frankfurt, Goethe-University, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany
| | - R Schlösser
- Division of Neonatology, Department of Pediatrics, University Hospital Frankfurt, Goethe-University, Theodor Stern Kai 7, 60590, Frankfurt am Main, Germany
| | - M Kreibich
- Institute of Medical Virology, National Reference Center for Retroviruses, University Hospital Frankfurt, Goethe-University, Paul Ehrlich Strasse 40, 60596, Frankfurt am Main, Germany
| | - O T Keppler
- Institute of Medical Virology, National Reference Center for Retroviruses, University Hospital Frankfurt, Goethe-University, Paul Ehrlich Strasse 40, 60596, Frankfurt am Main, Germany
| | - A Berger
- Institute of Medical Virology, National Reference Center for Retroviruses, University Hospital Frankfurt, Goethe-University, Paul Ehrlich Strasse 40, 60596, Frankfurt am Main, Germany
| |
Collapse
|
17
|
RNA interference-mediated targeting of human cytomegalovirus immediate-early or early gene products inhibits viral replication with differential effects on cellular functions. J Virol 2012; 86:5660-73. [PMID: 22438545 DOI: 10.1128/jvi.06338-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Viral drug toxicity, resistance, and an increasing immunosuppressed population warrant continued research into new avenues for limiting diseases associated with human cytomegalovirus (HCMV). In this study, a small interfering RNA (siRNA), siX3, was designed to target coding sequences within shared exon 3 of UL123 and UL122 transcripts encoding IE1 and IE2 immediate-early proteins of HCMV. Pretreatment of cells with siX3 reduced the levels of viral protein expression, DNA replication, and progeny virus production compared to control siRNA. Two siRNAs against UL54 and overlapping transcripts (UL55-57) were compared to siX3 in HCMV infection and were also found to be effective at inhibiting HCMV replication. Further investigation into the effects of the siRNAs on viral replication showed that pretreatment with each of the siRNAs resulted in an inhibition in the formation of mature replication compartments. The ability of these siRNAs to prevent or reduce certain cytopathic effects associated with HCMV infection was also examined. Infected cells pretreated with siX3, but not siUL54, retained promyelocytic leukemia (PML) protein in cellular PML bodies, an essential component of this host intrinsic antiviral defense. DNA damage response proteins, which are localized in nuclear viral replication compartments, were reduced in the siX3- and siUL54-treated cells. siX3, but not siUL54, prevented DNA damage response signaling early after infection. Therapeutic efficacy was demonstrated by treating cells with siRNAs after HCMV replication had commenced. Together, these findings suggest that siRNAs targeting exon 3 of the major IE genes or the UL54-57 transcripts be further studied for their potential development into anti-HCMV therapeutics.
Collapse
|
18
|
de Vries JJC, Barbi M, Binda S, Claas ECJ. Extraction of DNA from dried blood in the diagnosis of congenital CMV infection. Methods Mol Biol 2012; 903:169-75. [PMID: 22782817 DOI: 10.1007/978-1-61779-937-2_10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Viral DNA detection in dried blood spotted on filter paper, dried blood spots (DBS), is valuable in the diagnosis of viral infections, with at the moment congenital cytomegalovirus (CMV) being the most common application. CMV detection in clinical samples taken within the first 2-3 weeks after birth differentiates congenital CMV infection from the in general harmless postnatal acquired cytomegalovirus infection. DBS render the possibility to diagnose congenital CMV infection retrospectively, e.g., when late-onset hearing loss, the most frequently encountered symptom of congenital CMV infection, becomes manifest. Additionally, CMV DNA detection in DBS can be of usage in recently advocated newborn screening on congenital CMV infection. The procedure of CMV DNA detection in DBS consists of two separate steps: (1) DNA extraction from the DBS, followed by (2) CMV DNA amplification. Here, we describe two efficient methods for the extraction of DNA from DBS. Sensitivity, specificity, and applicability of the methods for high-throughput usage are discussed.
Collapse
Affiliation(s)
- Jutte J C de Vries
- Department of Medical Microbiology, Clinical Microbiological Laboratory, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| | | | | | | |
Collapse
|
19
|
Valganciclovir treatment in a 6-month-old infant with asymptomatic congenital cytomegalovirus infection and late hearing loss. Pediatr Infect Dis J 2011; 30:1124-5. [PMID: 22105427 DOI: 10.1097/inf.0b013e3182293439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Paradiž KR, Seme K, Puklavec E, Paro-Panjan D, Poljak M. Prevalence of congenital cytomegalovirus infection in Slovenia: a study on 2,841 newborns. J Med Virol 2011; 84:109-15. [PMID: 22028094 DOI: 10.1002/jmv.22230] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2011] [Indexed: 12/23/2022]
Abstract
Human cytomegalovirus (CMV) is the most frequent cause of congenital infection in humans. In the first prevalence study of congenital CMV infection in Eastern and Central Europe, all neonates born in a 22-month period in two Slovenian maternity units (total of 2,841 newborns) were screened prospectively for congenital CMV infection by a real-time polymerase chain reaction (PCR) in urine. In all newborns with positive screening results, plasma and dried blood spots (DBS) collected at birth were tested additionally for CMV DNA. Congenital CMV infection was confirmed by virus isolation from a urine sample collected within the first 2 weeks of life. Congenital CMV infection was identified in four out of 2,841 newborns tested (incidence 0.14%; 95% CI, 0.05-0.39%). In four newborns with confirmed congenital infection, the concentration of CMV DNA in urine ranged from 4.68 to 8.18 log(10) copies/ml, all four newborns had detectable CMV DNA in plasma taken at birth (1.26-3.34 log(10) copies/ml) and two out of four had detectable CMV DNA in DBS collected during newborn metabolic screening. None of the four newborns with confirmed congenital CMV infection was symptomatic. The study showed that the prevalence of congenital CMV infection at birth in Slovenia is among the lowest in the world and that CMV DNA PCR testing of urine is a suitable and affordable real-time screening strategy for congenital CMV infection. If it is performed in 24 mini-pools, the cost of screening is 1.4 €/newborn and the cost of detecting a single newborn with congenital CMV infection 1,000 €.
Collapse
|
21
|
Lombardi G, Garofoli F, Stronati M. Congenital cytomegalovirus infection: treatment, sequelae and follow-up. J Matern Fetal Neonatal Med 2011; 23 Suppl 3:45-8. [PMID: 20807160 DOI: 10.3109/14767058.2010.506753] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cytomegalovirus (CMV) is the most common cause of congenital infection affecting about 1% of all the live births worldwide. Its prevalence in the developed world seems to be slightly lower, ranging between 0.6 and 0.7%. Symptoms can be detected at birth in 10-15% of the congenitally infected of which 50-90% will develop sequelae, the most frequent being sensorineural hearing loss (SNHL), visual defect, psychomotor impairment, mental retardation, cerebral palsy and seizures. Eighty-five to 90% of the infected newborns are asymptomatic but 10-15% of them are equally at risk for sensorineural sequelae, like 20-30% of all the infected children. Therefore it is important a time prolonged and closer follow-up of infected children that we propose should be until 6 years of age. This should lead to an early intervention, better management and eventually even control the long-term sequelae. Infants born with symptomatic congenital infection have a worse prognosis than those with no evidence of clinical disease, and ganciclovir (GCV) intravenous 6 mg/kg every 12 h for 6 weeks is the most used therapy for symptomatic newborns. Valganciclovir (V-GCV) syrup is a pro-drug of GCV and presents high oral bioavailability. To date, it is possible to administer this drug at home, and the tolerability profile may allow for wider indications and longer treatments.
Collapse
Affiliation(s)
- Giuseppina Lombardi
- Neonatology and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | | |
Collapse
|
22
|
Schottstedt V, Blümel J, Burger R, Drosten C, Gröner A, Gürtler L, Heiden M, Hildebrandt M, Jansen B, Montag-Lessing T, Offergeld R, Pauli G, Seitz R, Schlenkrich U, Strobel J, Willkommen H, von König CHW. Human Cytomegalovirus (HCMV) - Revised. ACTA ACUST UNITED AC 2010; 37:365-375. [PMID: 21483467 DOI: 10.1159/000322141] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 07/13/2010] [Indexed: 02/05/2023]
Affiliation(s)
- Volkmar Schottstedt
- Arbeitskreis Blut, Untergruppe «Bewertung Blutassoziierter Krankheitserreger»
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Coll O, Benoist G, Ville Y, Weisman LE, Botet F, Anceschi MM, Greenough A, Gibbs RS, Carbonell-Estrany X. Guidelines on CMV congenital infection. J Perinat Med 2010; 37:433-45. [PMID: 19673682 DOI: 10.1515/jpm.2009.127] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Congenital cytomegalovirus (CMV) infection occurs in 0.6-0.7% of all newborns and is the most prevalent infection-related cause of congenital neurological handicap. Vertical transmission occurs in around 30% of cases, but the fetus is not always affected. Symptomatic newborns at birth have a much higher risk of suffering severe neurological sequelae. Detection of specific IgG and IgM and IgG avidity seem to be the most reliable tests to identify a primary infection but interpretation in a clinical context may be difficult. If a seroconversion is documented or a fetal infection is suspected by ultrasound markers, an amniocentesis should be performed to confirm a vertical transmission. In the absence of a confirmed fetal infection with fetal structural anomalies, a pregnancy termination should be discouraged. Fetal prognosis is mainly correlated to the presence of brain damage. Despite promising results with the use of antiviral drugs and CMV hyperimmune globulin (HIG), results have to be interpreted with caution. Pregnant women should not be systematically tested for CMV during pregnancy. Managing CMV screening should be restricted to pregnancies where a primary infection is suspected or among women at high risk. The magnitude of congenital CMV disease and the value of interventions to prevent its transmission or to decrease the sequelae need to be established before implementing public health interventions. In this paper, aspects of CMV infection in the pregnant woman and her infant are reviewed.
Collapse
Affiliation(s)
- Oriol Coll
- Department of Maternal-Fetal Medicine, Institut Clínic de Ginecologia, Obstetrícia i Neonatologia, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Despite advances in peripartum care, maternal morbidity and mortality associated with infections in pregnancy are increasing even in developed countries. Recently published data from the Center for Disease Control's Pregnancy Mortality Surveillance System indicates that although maternal mortality from hemorrhage, embolism, and anesthesia has declined in the United States, the proportion of maternal deaths due to infections has increased. During 1991–7 infection accounted for 13.2% of pregnancy-related deaths overall and 36.3% of abortion-related deaths. The greatest infection risk is found in blacks, older women, women without prenatal care, and women with multiple pregnancy. In the United States pregnancy rates are stable or increasing in these groups. Infection is also a major cause of morbidity and mortality for the fetus and newborn. Many perinatal infections are associated with intra-uterine growth retardation and low birthweight, or cause fetal and neonatal brain injury. Infections, particularly bacterial vaginosis and chorioamnionitis, can result in preterm delivery of live-born infants (delivery before 37 weeks gestation), or stillbirth. A multitude of immunologic, endocrinologic, metabolic, physiologic, and anatomic changes influence the likelihood and course of many infections during pregnancy. Some of these changes are intrinsic, and occur in all normal pregnancies, while others occur to varying degrees in normal and abnormal pregnancies.
Collapse
|
25
|
Kim CS. Congenital and perinatal cytomegalovirus infection. KOREAN JOURNAL OF PEDIATRICS 2010. [DOI: 10.3345/kjp.2010.53.1.14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Chun Soo Kim
- Departments of Pediatrics, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
26
|
Oral valganciclovir treatment in newborns with symptomatic congenital cytomegalovirus infection. Eur J Clin Microbiol Infect Dis 2009; 28:1465-70. [DOI: 10.1007/s10096-009-0806-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Accepted: 08/10/2009] [Indexed: 10/20/2022]
|
27
|
Schreiber A, Härter G, Schubert A, Bunjes D, Mertens T, Michel D. Antiviral treatment of cytomegalovirus infection and resistant strains. Expert Opin Pharmacother 2009; 10:191-209. [PMID: 19236193 DOI: 10.1517/14656560802678138] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This review discusses the management of resistant cytomegalovirus and prevention strategies for fatal therapy failures. Five drugs, ganciclovir/valganciclovir, cidofovir, foscarnet and fomivirsen, have been approved so far for the treatment of human cytomegalovirus (HCMV) diseases. Except for fomivirsen, all of the approved drugs share the same target molecule, the viral DNA polymerase. The emergence of drug-resistant HCMV has also been reported for all of them. For optimal care of patients, the clinical virologist has to provide the most meaningful assays for monitoring of therapy and early detection of emerging drug-resistant HCMV. Additionally, a quantitative drug monitoring would be helpful. New antiviral agents are urgently needed with less adverse effects, good oral bioavailability and possibly novel targets or mechanisms of action to avoid cross-resistance and to improve the ability to suppress the selection of resistant virus strains by combination therapy. Compounds like maribavir, leflunomide and artesunate, which exhibit anti-HCMV activity in vitro and in patients need to be evaluated in clinical studies. Besides these, new therapy approaches like immunotherapy or new diagnostic techniques like pyrosequencing have to be considered in the future.
Collapse
Affiliation(s)
- Andreas Schreiber
- Universitätsklinikum Ulm, Institut für Virologie, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | | | | | | | | | | |
Collapse
|