1
|
Calzone KA, Badzek L. The Genomic Essential Competencies Expected for all Registered Nurses. Nurs Clin North Am 2025; 60:243-255. [PMID: 40345757 PMCID: PMC12064882 DOI: 10.1016/j.cnur.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Evidence-based genomic applications improve the quality and safety of health care. Nurses irrespective of their role, level of training, clinical specialty, need to achieve genomic competency as defined by competencies internationally. This includes nursing educators who teach nurses in training as well as nursing leadership who guide policy and support infrastructures. Genomic resources are abundant to help achieve competency, which is aimed at further improving the quality, safety, and outcomes of nursing care.
Collapse
Affiliation(s)
- Kathleen A Calzone
- Genetics Branch, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, Building 37, Rm 6002C, Bethesda, MD 20892, USA.
| | - Laurie Badzek
- Penn State University 201 Nursing Sciences Building, University Park, PA 16802, USA
| |
Collapse
|
2
|
Tonkin ET, Dwyer AA, Dewell S, Badzek L, Calzone KA. Global Health and Genomics. Nurs Clin North Am 2025; 60:217-228. [PMID: 40345755 PMCID: PMC12064883 DOI: 10.1016/j.cnur.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Evidence based genomic applications improve the quality and safety of healthcare. Nurses, irrespective of their role, level of training, or clinical specialty, need to achieve genomic competency as defined internationally. This includes nursing educators who teach nurses in training as well as nursing leadership who guide policy and support infrastructure. Genomic resources are abundant to help achieve competency which in turn improves quality, safety, and outcomes of nursing care.
Collapse
Affiliation(s)
- Emma T Tonkin
- Genomics Healthcare, Faculty of Life Sciences and Education [GTAB031, Glyntaff Campus], University of South Wales, Pontypridd, Wales CF37 1DL, UK. https://twitter.com/emmattonkin
| | - Andrew A Dwyer
- Boston College William F. Connell School of Nursing, 140 Commonwealth Avenue, Maloney Hall 364, Chestnut Hill, MA 20476, USA. https://twitter.com/Andrew_A_Dwyer
| | - Sarah Dewell
- School of Nursing, Thompson Rivers University, School of Nursing, 805 TRU Way, Kamloops, BC V2C 0C8, Canada. https://twitter.com/drsarahdewell
| | - Laurie Badzek
- Ross and Carol Nese College of Nursing, Penn State University, 201 Nursing Sciences Building, University Park, PA 16802, USA. https://twitter.com/lbadzek
| | - Kathleen A Calzone
- Genetics Branch, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, Building 37, Room 6002C, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Goldin MR, Ruderfer DM, Bick A, Roden DM, Schuler BA, Robinson JR. Benefits and barriers to broad implementation of genomic sequencing in the NICU. Am J Hum Genet 2025:S0002-9297(25)00148-X. [PMID: 40367948 DOI: 10.1016/j.ajhg.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025] Open
Abstract
Genome (GS) and exome (ES) sequencing as first-tier diagnostic tests have the potential to increase rates of genetic diagnoses and acutely change the management of neonates in the neonatal intensive care unit (NICU). However, the widespread implementation of genomic sequencing has been limited by several barriers. In this systematic review, we analyze the current literature on the utilization of GS and ES in infants in the NICU to identify the benefits, barriers, and components of successful implementation. Across the 42 studies that discussed GS and ES in the NICU setting, six themes were identified: disease detection, timeliness of results, cost, provider attitudes, parental attitudes, and equitable access. Benefits of GS and ES include high disease detection rates, timely results, and possible reduction in healthcare costs by reducing time spent in the NICU. Additionally, clinicians find GS/ES to be important and useful, and parents and caregivers largely perceive GS/ES to be beneficial. Barriers to widespread GS/ES include availability of personnel to facilitate timely diagnosis and coverage of cost. Additionally, clinicians report worries about a lack of genetics knowledge, informed consent, results return, and potential harm. Parents consistently report low levels of anxiety, decisional conflict, harm, or regret. Finally, the lack of availability of translated consent documents limits the participation of families who do not speak English or Spanish. Continued work is essential to optimize these technologies and ensure equitable access.
Collapse
Affiliation(s)
| | - Douglas M Ruderfer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander Bick
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dan M Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bryce A Schuler
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jamie R Robinson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatric Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
4
|
Moore C, Arenchild M, Waldman B, Rego S, Kingsmore SF, Field J, Barnhart J, Nee S, Nofsinger R. Rapid Whole-Genome Sequencing as a First-Line Test Is Likely to Significantly Reduce the Cost of Acute Care in a Private Payer System. J Appl Lab Med 2025:jfaf045. [PMID: 40248916 DOI: 10.1093/jalm/jfaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Genetic disorders are a leading contributor to morbidity and mortality in neonatal and pediatric intensive care units. Rapid whole-genome sequencing (rWGS) has demonstrated improved clinical outcomes and reduced costs of care. The objective of this study was to predict the effect of rWGS on healthcare spending if implemented as a first-line diagnostic test in the Blue Shield of California (BSC) private payer system. METHODS This study applied private payer reimbursement methods and rates to clinical outcomes of rWGS on pediatric inpatient care as determined by a previous study of publicly insured infants in Project Baby Bear. BSC patients who were clinically similar to the Project Baby Bear cohort were identified by matching on diagnosis-related group and severity of illness. Payment data from these BSC patients was used to estimate the financial impact of clinical outcomes resulting from rWGS testing in a commercially insured pediatric population. RESULTS The analysis estimated a reduction of $5.8 million to $7.8 million in inpatient payments due to an estimated 457 to 592 avoided inpatient days due to rWGS results. With an estimated cost of sequencing at $2.7 million for the entire cohort (n = 184), the financial impact of rWGS as a first-tier test in the intensive care unit resulted in estimated net savings to BSC of $16 730 to $28 061 per patient sequenced. CONCLUSIONS Implementation of rWGS using the protocols established in Project Baby Bear is likely to result in significant reductions in healthcare spending among privately insured patients.
Collapse
Affiliation(s)
- Christy Moore
- Health Trend Solutions, Blue Shield of California, Oakland, CA, United States
| | - Madison Arenchild
- Rady Children's Institute for Genomic Medicine, San Diego, CA, United States
| | - Bryce Waldman
- Rady Children's Institute for Genomic Medicine, San Diego, CA, United States
| | - Seema Rego
- Rady Children's Institute for Genomic Medicine, San Diego, CA, United States
| | - Stephen F Kingsmore
- Rady Children's Institute for Genomic Medicine, San Diego, CA, United States
| | - Justin Field
- Health Trend Solutions, Blue Shield of California, Oakland, CA, United States
| | - Jason Barnhart
- Health Trend Solutions, Blue Shield of California, Oakland, CA, United States
| | - Stephanie Nee
- Health Trend Solutions, Blue Shield of California, Oakland, CA, United States
| | - Russell Nofsinger
- Rady Children's Institute for Genomic Medicine, San Diego, CA, United States
| |
Collapse
|
5
|
Abarca-Barriga HH, Vásquez Sotomayor F, Punil-Luciano R, Laso-Salazar MC, Barrón-Pastor HJ. Identification of intragenic variants in pediatric patients with intellectual disability in Peru. BMC Med Genomics 2025; 18:76. [PMID: 40251579 PMCID: PMC12008840 DOI: 10.1186/s12920-025-02141-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/04/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Intellectual disability in Latin America can reach a frequency of 12% of the population, these may include nutritional deficiencies, exposure to toxic or infectious agents, and the lack of universal neonatal screening programs. In 90% of patients with intellectual disability, the etiology can be attributed to variants in the genome. OBJECTIVE to determine intragenic variants in patients with intellectual disability between 5 and 18 years old at Instituto Nacional de Salud del Niño. METHODS It is a descriptive cross-sectional study with convenience sampling. A total of 124 children diagnosed with intellectual disability were selected based on psychological test results and availability for whole exome sequencing. In addition, a chromosomal analysis of 6.55 M was performed on ten patients with a negative result in sequencing. Relative and absolute frequencies and measures of central tendency and dispersion were determined according to their nature. In addition, multiple linear regression and Poisson regression were used to determine the association between some clinical characteristics and the probability of occurrence in patients with positive results. RESULTS The median age of the patients was 6.3 (IQR = 5.95), males accounted for 57.3%, and 91.9% of the cases had mild intellectual disability. Exome sequencing determined the etiology in 30.6% of patients with intellectual disability, of which 52.6% were autosomal dominant inheritance. The most frequent genes found were MECP2, STXBP1 and LAMA2. A broad genotype-phenotype correlation was identified, highlighting the genetic heterogeneity of intellectual disability in this population. The presence of dermatologic lesions, dystonia, peripheral neurological disorders, and fourth finger flexion limitation were observed more frequently in patients with intellectual disability with "positive results". CONCLUSIONS This study shows that one-third of patients with intellectual disability exhibit intragenic variants, highlighting the importance of genetic analysis for accurate diagnosis. The identification of genes such as MECP2, STXBP1, and LAMA2 underscores the genetic heterogeneity of intellectual disability in the studied population. These findings emphasize the need for genetic testing in clinical management and the implementation of early detection programs in Peru.
Collapse
Affiliation(s)
- Hugo Hernán Abarca-Barriga
- Instituto de Investigaciones de Ciencias Biomédicas, Facultad de Medicina Humana, Universidad Ricardo Palma, Av. Benavides 5440. Santiago de Surco, Lima, 1801, Perú.
- Servicio de Genética & Errores Innatos del Metabolismo, Instituto Nacional de Salud del Niño- Breña, Lima, Perú.
- Facultad de Medicina Humana, Universidad Nacional Mayor de San Marcos. Lima, Lima, Perú.
| | - Flor Vásquez Sotomayor
- Instituto de Investigaciones de Ciencias Biomédicas, Facultad de Medicina Humana, Universidad Ricardo Palma, Av. Benavides 5440. Santiago de Surco, Lima, 1801, Perú
- Servicio de Genética & Errores Innatos del Metabolismo, Instituto Nacional de Salud del Niño- Breña, Lima, Perú
| | - Renzo Punil-Luciano
- Servicio de Genética & Errores Innatos del Metabolismo, Instituto Nacional de Salud del Niño- Breña, Lima, Perú
| | - María Cristina Laso-Salazar
- Servicio de Genética & Errores Innatos del Metabolismo, Instituto Nacional de Salud del Niño- Breña, Lima, Perú
| | | |
Collapse
|
6
|
Hong Y, Wang Y, Shu W. Immunocyte phenotypes and childhood disease susceptibility: insights from bidirectional Mendelian randomization and implications for immunomodulatory therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04091-1. [PMID: 40178601 DOI: 10.1007/s00210-025-04091-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Immune cells are essential for maintaining immune homeostasis during childhood and influence both growth and disease susceptibility. However, the causal relationships between immunocyte phenotypes and childhood diseases remain unclear. This study employed a two-sample Mendelian Randomization (MR) analysis to assess causal associations between 731 immunocyte phenotypes and four major childhood diseases: childhood obesity, childhood absence epilepsy, childhood asthma, and childhood allergies. Genome-wide association study (GWAS) data were used, and stringent instrumental variable (IV) selection and multiple sensitivity analyses, including MR-Egger, weighted median, and leave-one-out tests, were applied to validate the robustness of the results. Significant associations were identified between specific T cell, monocyte, and B cell phenotypes and childhood diseases. Notably, CD8bright T cells and CD19 + B cells were positively correlated with childhood obesity, while monocyte subtypes were strongly associated with asthma pathophysiology. Reverse MR analysis indicated no significant causal effects of childhood diseases on immune phenotypes, except for negative associations between childhood asthma and TCRgd AC, and childhood allergy and CD28 + CD45RA + CD4 + cells. These findings highlight the critical role of immune dysregulation in childhood disease etiology and suggest potential targets for immunomodulatory therapies. Understanding these immune-disease interactions may inform novel pharmacological interventions, particularly in immune-mediated disorders such as asthma and obesity. Further research into immune-targeted therapies could enhance treatment strategies for pediatric conditions associated with chronic inflammation and immune dysfunction.
Collapse
Affiliation(s)
- Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China.
| | - Yi Wang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China
| | - Wanyi Shu
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China
| |
Collapse
|
7
|
Cohen JL, Duyzend M, Adelson SM, Yeo J, Fleming M, Ganetzky R, Hale R, Mitchell DM, Morton SU, Reimers R, Roberts A, Strong A, Tan W, Thiagarajah JR, Walker MA, Green RC, Gold NB. Advancing precision care in pregnancy through a treatable fetal findings list. Am J Hum Genet 2025:S0002-9297(25)00110-7. [PMID: 40209713 DOI: 10.1016/j.ajhg.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 04/12/2025] Open
Abstract
The use of genomic sequencing (GS) for prenatal diagnosis of fetuses with sonographic abnormalities has grown tremendously over the past decade. Fetal GS also offers an opportunity to identify incidental genomic variants that are unrelated to the fetal phenotype but may be relevant to fetal and newborn health. There are currently no guidelines for reporting incidental findings from fetal GS. In the United States, GS for adults and children is recommended to include a list of "secondary findings" genes (ACMG SF v.3.2) that are associated with disorders for which surveillance or treatment can reduce morbidity and mortality. The genes on ACMG SF v.3.2 predominantly cause adult-onset disorders. Importantly, many genetic disorders with fetal and infantile onset are treatable as well. A proposed solution is to create a "treatable fetal findings list," which can be offered to pregnant individuals undergoing fetal GS or, eventually, as a standalone cell-free fetal DNA screening test. In this integrative review, we propose criteria for a treatable fetal findings list, then identify genetic disorders with clinically available or emerging fetal interventions and those for which clinical detection and intervention in the first week of life might lead to improved outcomes. Finally, we synthesize the potential benefits, limitations, and risks of a treatable fetal findings list.
Collapse
Affiliation(s)
- Jennifer L Cohen
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Michael Duyzend
- Department of Pediatrics, Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sophia M Adelson
- Brigham and Women's Hospital, Boston, MA, USA; Stanford School of Medicine, Stanford, CA, USA
| | - Julie Yeo
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Mark Fleming
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rebecca Ganetzky
- Center for Computational Genomic Medicine and Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Rebecca Hale
- Department of Pediatrics, Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Deborah M Mitchell
- Division of Pediatric Endocrinology, Massachusetts General Hospital, Boston, MA, USA
| | - Sarah U Morton
- Department of Pediatrics, Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rebecca Reimers
- Rady Children's Institute for Genomic Medicine, Rady Children's Hospital Division of Perinatology, Scripps Research Translational Institute, University of California, San Diego, San Diego, CA, USA
| | - Amy Roberts
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Department of Cardiology, Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Alanna Strong
- Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA, USA; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Weizhen Tan
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Division of Pediatric Nephrology, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Jay R Thiagarajah
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Melissa A Walker
- Department of Neurology, Division of Pediatric Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert C Green
- Mass General Brigham, Boston, MA, USA; Broad Institute, Cambridge, MA, USA; Ariadne Labs, Boston, MA, USA
| | - Nina B Gold
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Gold NB, Omorodion JO, Del Rosario MC, Rivera-Cruz G, Hsu CY, Ziniel SI, Holm IA. Preferences of parents from diverse backgrounds on genomic screening of apparently healthy newborns. J Genet Couns 2025; 34:e1994. [PMID: 39465664 DOI: 10.1002/jgc4.1994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/22/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
Genomic sequencing has been proposed as a strategy to expand newborn screening. Perspectives on genomic newborn screening from parents of diverse racial, ethnic, and socioeconomic backgrounds are needed to shape equitable implementation of this modality. We conducted 20 semi-structured interviews (15 English, 5 Spanish) and seven focus groups (4 English, 3 Spanish) with parents from diverse backgrounds to assess their perspectives regarding which disorders and variants might be screened, data privacy, and barriers to pursuing specialized care. Parents felt that genomic newborn screening would provide them with improved understanding of their children's health and had the potential to yield health and personal benefits. Themes that became evident included: interest in childhood and family health risks, the value of emotional preparation and personal planning, understanding of uncertain and low-risk results, concerns regarding data privacy, and concerns about support following the receipt of a positive newborn screening result. The expected benefits and concerns expressed by parents of diverse backgrounds regarding genomic newborn screening should guide future policy decisions. Their preferences should be considered prior to the implementation of large-scale genomic newborn screening programs.
Collapse
Affiliation(s)
- Nina B Gold
- Division of Medical Genetics and Metabolism, Massachusetts General Hospital for Children, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacklyn O Omorodion
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Maya C Del Rosario
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Graduate Medical Sciences, Boston University, Boston, Massachusetts, USA
| | - Greysha Rivera-Cruz
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Reproductive Endocrinology and Infertility, Stanford University School of Medicine, Stanford School of Medicine, Stanford, California, USA
| | - Celeste Y Hsu
- Harvard Medical School Center for Bioethics, Boston, Massachusetts, USA
- University of California San Diego School of Medicine, La Jolla, California, USA
| | - Sonja I Ziniel
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Section of Pediatric Hospital Medicine, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Ingrid A Holm
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Hao C, Hu X, Guo R, Qi Z, Jin F, Zhang X, Xie L, Liu H, Liu Y, Ni X, Li W. Targeted gene sequencing and hearing follow-up in 7501 newborns reveals an improved strategy for newborn hearing screening. Eur J Hum Genet 2025; 33:468-475. [PMID: 39443691 PMCID: PMC11986168 DOI: 10.1038/s41431-024-01711-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/08/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
Hearing loss is a common congenital condition. Concurrent newborn hearing and limited genetic screening has been implemented in China for the last decade. However, the role of gene sequencing screening has not been evaluated. In this study, we enrolled 7501 newborns (52.7% male, 47.3% female) in our Newborn Screening with Targeted Sequencing (NESTS) program, and 90 common deafness genes were sequenced for them. Hearing status assessments were conducted via telephone from February 2021 to August 2022, for children aged 3 to 48 months. Of the universal newborn hearing screening, 126 (1.7%) newborns did not pass. Targeted sequencing identified 150 genetically positive newborns (2.0%), with 25 exhibiting dual-positive results in both screening. Following diagnostic audiometry revealed 18 hearing loss newborns and half of them had abnormal results in both screening. The positive predictive value for universal newborn hearing screening alone was merely 14.3% (18/126). However, when combined with targeted sequencing, this rate increased to 36.0% (9/25). Furthermore, limited genetic screening identified 316 carriers of hot-spot variants, but none exhibited biallelic variants. All 15 hot-spot carriers who failed physical screening demonstrated normal hearing during follow-up. In this cohort study of 7501 Newborns, Combining targeted sequencing with universal newborn hearing screening demonstrated technical feasibility and clinical utility of identifying individuals with hearing loss, especially when coupled with genetic counseling and closed-loop management. It is suggested to use this integrated method as an improved strategy instead of the current limited genetic screening program in some regions of China.
Collapse
Affiliation(s)
- Chanjuan Hao
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
- Henan Key Laboratory of Inherited Metabolic Diseases, Pediatric Research Institute of Zhengzhou Children's Hospital, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China.
| | - Xuyun Hu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Henan Key Laboratory of Inherited Metabolic Diseases, Pediatric Research Institute of Zhengzhou Children's Hospital, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Ruolan Guo
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Henan Key Laboratory of Inherited Metabolic Diseases, Pediatric Research Institute of Zhengzhou Children's Hospital, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Zhan Qi
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Feng Jin
- Shunyi Women and Children's Healthcare Hospital of Beijing Children's Hospital, Beijing, China
| | - Xiaofen Zhang
- Shunyi Women and Children's Healthcare Hospital of Beijing Children's Hospital, Beijing, China
| | - Limin Xie
- Shunyi Women and Children's Healthcare Hospital of Beijing Children's Hospital, Beijing, China
| | - Haihong Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yuanhu Liu
- Shunyi Women and Children's Healthcare Hospital of Beijing Children's Hospital, Beijing, China
- Department of Otolaryngology Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xin Ni
- Department of Otolaryngology Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Wei Li
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
- Henan Key Laboratory of Inherited Metabolic Diseases, Pediatric Research Institute of Zhengzhou Children's Hospital, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
10
|
Kansal R. Rapid Whole-Genome Sequencing in Critically Ill Infants and Children with Suspected, Undiagnosed Genetic Diseases: Evolution to a First-Tier Clinical Laboratory Test in the Era of Precision Medicine. CHILDREN (BASEL, SWITZERLAND) 2025; 12:429. [PMID: 40310077 PMCID: PMC12025730 DOI: 10.3390/children12040429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
The completion of the Human Genome Project in 2003 has led to significant advances in patient care in medicine, particularly in diagnosing and managing genetic diseases and cancer. In the realm of genetic diseases, approximately 15% of critically ill infants born in the U.S.A. are diagnosed with genetic disorders, which comprise a significant cause of mortality in neonatal and pediatric intensive care units. The introduction of rapid whole-genome sequencing (rWGS) as a first-tier test in critically ill children with suspected, undiagnosed genetic diseases is a breakthrough in the diagnosis and subsequent clinical management of such infants and older children in intensive care units. Rapid genome sequencing is currently being used clinically in the USA, the UK, the Netherlands, Sweden, and Australia, among other countries. This review is intended for students and clinical practitioners, including non-experts in genetics, for whom it provides a historical background and a chronological review of the relevant published literature for the progression of pediatric diagnostic genomic sequencing leading to the development of pediatric rWGS in critically ill infants and older children with suspected but undiagnosed genetic diseases. Factors that will help to develop rWGS as a clinical test in critically ill infants and the limitations are briefly discussed, including an evaluation of the clinical utility and accessibility of genetic testing, education for parents and providers, cost-effectiveness, ethical challenges, consent issues, secondary findings, data privacy concerns, false-positive and false-negative results, challenges in variant interpretation, costs and reimbursement, the limited availability of genetic counselors, and the development of evidence-based guidelines, which would all need to be addressed to facilitate the implementation of pediatric genomic sequencing in an effective widespread manner in the era of precision medicine.
Collapse
Affiliation(s)
- Rina Kansal
- Molecular Oncology and Genetics, Diagnostic Laboratories, Versiti Blood Center of Wisconsin, Milwaukee, WI 53233, USA;
- Department of Pathology and Anatomical Sciences, The University at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
11
|
Wenger TL, Scott A, Kruidenier L, Sikes M, Keefe A, Buckingham KJ, Marvin CT, Shively KM, Bacus T, Sommerland OM, Anderson K, Gildersleeve H, Davis CJ, Love-Nichols J, MacDuffie KE, Miller DE, Yu JH, Snook A, Johnson B, Veenstra DL, Parish-Morris J, McWalter K, Retterer K, Copenheaver D, Friedman B, Juusola J, Ryan E, Varga R, Doherty DA, Dipple K, Chong JX, Kruszka P, Bamshad MJ. SeqFirst: Building equity access to a precise genetic diagnosis in critically ill newborns. Am J Hum Genet 2025; 112:508-522. [PMID: 39999847 PMCID: PMC11947171 DOI: 10.1016/j.ajhg.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Access to a precise genetic diagnosis (PrGD) in critically ill newborns is limited and inequitable because the complex inclusion criteria used to prioritize testing eligibility omit many patients at high risk for a genetic condition. SeqFirst-neo is a program to test whether a genotype-driven workflow using simple, broad exclusion criteria to assess eligibility for rapid genome sequencing (rGS) increases access to a PrGD in critically ill newborns. All 408 newborns admitted to a neonatal intensive care unit between January 2021 and February 2022 were assessed, and of 240 eligible infants, 126 were offered rGS (i.e., intervention group [IG]) and compared to 114 infants who received conventional care in parallel (i.e., conventional care group [CCG]). A PrGD was made in 62/126 (49.2%) IG neonates compared to 11/114 (9.7%) CCG infants. The odds of receiving a PrGD were ∼9 times greater in the IG vs. the CCG, and this difference was maintained at 12 months follow-up. Access to a PrGD in the IG vs. CCG differed significantly between infants identified as non-White (34/74, 45.9% vs. 6/29, 20.7%; p = 0.024) and Black (8/10, 80.0% vs. 0/4; p = 0.015). Neonatologists were significantly less successful at predicting a PrGD in non-White than non-Hispanic White infants. The use of a standard workflow in the IG with a PrGD revealed that a PrGD would have been missed in 26/62 (42%) infants. The use of simple, broad exclusion criteria that increase access to genetic testing significantly increases access to a PrGD, improves access equity, and results in fewer missed diagnoses.
Collapse
Affiliation(s)
- Tara L Wenger
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Abbey Scott
- Seattle Children's Hospital, Seattle, WA 98105, USA
| | | | - Megan Sikes
- Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Alexandra Keefe
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Kati J Buckingham
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Colby T Marvin
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Kathryn M Shively
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Tamara Bacus
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | - Kailyn Anderson
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Heidi Gildersleeve
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Chayna J Davis
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | - Katherine E MacDuffie
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Treuman Katz Center for Pediatric Bioethics and Palliative Care, Seattle Children's Research Institute, Seattle, WA 98121, USA
| | - Danny E Miller
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Hospital, Seattle, WA 98105, USA; Brotman Bay Institute, Seattle, WA 98195, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Joon-Ho Yu
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Treuman Katz Center for Pediatric Bioethics and Palliative Care, Seattle Children's Research Institute, Seattle, WA 98121, USA; Institute for Public Health Genetics, University of Washington, Seattle, WA 98195, USA
| | | | | | - David L Veenstra
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Julia Parish-Morris
- Department of Biomedical and Health Informatics, Perelman School of Medicine, Philadelphia, PA 19146, USA
| | | | - Kyle Retterer
- GeneDx, Gaithersburg, MD 20877, USA; Geisinger, Danville, PA 17822, USA
| | | | | | | | | | | | - Daniel A Doherty
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Hospital, Seattle, WA 98105, USA; Brotman Bay Institute, Seattle, WA 98195, USA
| | - Katrina Dipple
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Jessica X Chong
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Brotman Bay Institute, Seattle, WA 98195, USA
| | | | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Hospital, Seattle, WA 98105, USA; Brotman Bay Institute, Seattle, WA 98195, USA.
| |
Collapse
|
12
|
Couce ML, Cernada M, Boix H, Sánchez-Redondo MD, Espinosa Fernández MG, González-Pacheco N, Martín A, Pérez-Muñuzuri A. Current situation and new steps in newborn screening in Spain. An Pediatr (Barc) 2025; 102:503775. [PMID: 40044559 DOI: 10.1016/j.anpede.2025.503775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/19/2025] [Indexed: 03/23/2025] Open
Abstract
After more than 50 years of experience, newborn screening (NBS) programs represent one of the most significant advancements in public health, particularly in pediatric and neonatal care, benefiting almost 350 000 children annually in Spain. Following the inclusion of congenital hearing loss screening in 2003 and screening for seven congenital diseases by newborn blood spot test in 2014 as part of the population-wide neonatal screening program of the National Health System (NHS), significant advances have been achieved in recent years. This progress is evident in the implementation of screening for critical congenital heart diseases, approved in January 2024 by the National Public Health Commission of the Interterritorial Council of the NHS, as well as screening for congenital diseases through the newborn blood spot test, with the incorporation of new conditions enabled by advances in second-tier testing and emerging scientific evidence. Neonatologists and pediatricians must keep abreast of these developments and where the field is heading, as even more rapid progress may take place with the advent of genomic newborn screening.
Collapse
Affiliation(s)
- María L Couce
- Servicio de Neonatología, Hospital Clínico Universitario de Santiago; IDIS, RICORS-SAMID, Universidad de Santiago de Compostela, Santiago de Compostela (La Coruña), Spain.
| | - María Cernada
- Servicio de Neonatología, Hospital Universitario y Politécnico La Fe; Grupo de Investigación en Perinatología, Instituto de Investigación Sanitaria La Fe, Valencia, España, Spain
| | - Hector Boix
- Dirección Territorial de Pediatría, Hospital Universitari Dexeus, Barcelona, Spain
| | | | | | | | - Ana Martín
- Servicio de Neonatología, Hospital Sant Joan de Dèu, Barcelona, Spain
| | - Alejandro Pérez-Muñuzuri
- Servicio de Neonatología, Hospital Clínico Universitario de Santiago; IDIS, RICORS-SAMID, Universidad de Santiago de Compostela, Santiago de Compostela (La Coruña), Spain
| |
Collapse
|
13
|
Huang L, Lai HJ, Furuya KN, Antos NJ, Asfour F, Boyne KL, Howenstine M, Rock MJ, Sawicki GS, Gaffin JM, Worthey EA, Farrell PM. The Frequency and Potential Implications of HFE Genetic Variants in Children With Cystic Fibrosis. Pediatr Pulmonol 2025; 60:e71042. [PMID: 40071665 PMCID: PMC11898569 DOI: 10.1002/ppul.71042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Genetic modifiers have been identified that increase the risks of lung disease and other complications, such as diabetes in people with cystic fibrosis (CF). Variants in the hemochromatosis gene (HFE) were reported in a study of adults to be associated with worse lung disease. OBJECTIVES To ascertain the frequency of HFE variants, particularly C282Y (c.845G > A) and H63D (c.187C > G) and to determine if they are associated with variations in the onset and early severity of CF lung disease as well as abnormalities in iron status. DESIGN We studied with whole genome sequencing and clinical outcome measures in a cohort of 104 children with CF at 5-6 years old who were previously found to show an association between aggregated genetic modifiers and an earlier onset and a more severe lung disease phenotype. RESULTS In our cohort, 23% have H63D and 11% have C282Y. Lung function at age 6 years and Pseudomonas aeruginosa infections did not differ by HFE variants, but having C282Y was associated with more pulmonary exacerbations in the first 6 years of life. Three patients have H63D/C282Y genotype, and all showed phenotypic expression of hemochromatosis with abnormal iron indices. CONCLUSION Our study revealed that the presence of HFE variant C282Y in people with CF may lead to more severe lung disease manifestations beginning in early childhood. There is a risk of hemochromatosis in CF patients with two HFE variants, and thus they should be followed for evidence of iron overload.
Collapse
Affiliation(s)
- Leslie Huang
- Department of PediatricsUniversity of Wisconsin – MadisonMadisonWisconsinUSA
| | - HuiChuan J. Lai
- Department of PediatricsUniversity of Wisconsin – MadisonMadisonWisconsinUSA
- Department of Nutritional SciencesUniversity of Wisconsin – MadisonMadisonWisconsinUSA
| | - Katryn N. Furuya
- Department of PediatricsUniversity of Wisconsin – MadisonMadisonWisconsinUSA
| | - Nicholas J. Antos
- Department of PediatricsMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Fadi Asfour
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | | | | | - Michael J. Rock
- Department of PediatricsUniversity of Wisconsin – MadisonMadisonWisconsinUSA
| | - Gregory S. Sawicki
- Department of PediatricsBoston Children's HospitalBostonMassachusettsUSA
| | - Jonathan M. Gaffin
- Department of PediatricsBoston Children's HospitalBostonMassachusettsUSA
| | | | - Philip M. Farrell
- Department of PediatricsUniversity of Wisconsin – MadisonMadisonWisconsinUSA
- Department of Population Health SciencesUniversity of Wisconsin – MadisonMadisonWisconsinUSA
| |
Collapse
|
14
|
Uveges MK, Smith HS, Pereira S, Genetti C, McGuire AL, Beggs AH, Green RC, Holm IA. Family genetic risk communication and reverse cascade testing in the BabySeq project. Genet Med 2025; 27:101350. [PMID: 39731470 DOI: 10.1016/j.gim.2024.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
PURPOSE Genomic sequencing of newborns can initiate disease surveillance and therapy for children and may identify at-risk relatives through reverse cascade testing. We explored genetic risk communication and reverse cascade testing among families of newborns who underwent exome sequencing and were identified as having a risk for an autosomal dominant disease. METHODS We conducted semistructured interviews with parents of newborns enrolled in the BabySeq Project who had a pathogenic or likely pathogenic variant associated with an autosomal dominant childhood- and/or adult-onset disease returned. We used directed content analysis to derive themes. RESULTS From 11 families, all first-degree relatives (n = 32, 100%), 29 second-degree relatives (76%), and 26 third-degree relatives (43%) were informed of their risk. All parents (n = 22, 69% of first-degree relatives), 4 (11%) second-degree relatives, and 1 (2%) third-degree relatives underwent cascade testing. Most parents preferred to handle risk communication themselves. Parents with positive cascade testing but no associated symptoms were less inclined to share findings with relatives but highly motivated to share results if the variant's associated disease severity was high, as perceived with adult-onset conditions. One new subtheme, family member traits, was identified and defined as a relative's propensity to anxiety/concern after risk communications but did not diminish risk communication. CONCLUSION Findings can inform more effective notification and testing practices for families of newborns at risk for hereditary genetic conditions.
Collapse
Affiliation(s)
- Melissa K Uveges
- Boston College, William F. Connell School of Nursing, Chestnut Hill, MA; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA.
| | - Hadley Stevens Smith
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX; Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA; Center for Bioethics, Harvard Medical School, Boston, MA
| | - Stacey Pereira
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - Casie Genetti
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA
| | - Amy L McGuire
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - Alan H Beggs
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Robert C Green
- Harvard Medical School, Boston, MA; Mass General Brigham, Boston, MA; The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ingrid A Holm
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Mademont-Soler I, Castells-Sarret N, Cisneros A, Foj L, Benavent-Bofill C, Xunclà M, Viñas-Jornet M, Ros A, Rey N, Blanco I, López-Ortega R, Obón M, Plaja A. Incidental Findings Identified by Prenatal Microarray Analysis and Consensus Reporting Criteria of the Catalan Public Health Network XIGENICS. Prenat Diagn 2025; 45:326-347. [PMID: 39815072 DOI: 10.1002/pd.6746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/25/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
OBJECTIVE The study aimed to evaluate the frequency of pathogenic copy number variants (CNVs) classified as incidental findings (IFs) in prenatal diagnosis and to develop consensus recommendations for standardizing their reporting across six centers within the Catalan public health system (XIGENICS network). METHOD A retrospective review of 4219 consecutive prenatal microarrays performed within the network from 2018 to 2023 was conducted, including all referral reasons. To develop consensus recommendations, several discussion meetings were held along with an extensive review of the existing literature. RESULTS A total of 69 IFs were identified in 68 samples, revealing a detection rate of 1.6%. They included: 5 CNVs associated with neurodevelopmental disorders and/or congenital defects with complete penetrance, 41 CNVs for neurodevelopmental disorders and/or congenital defects with incomplete penetrance, 4 disorders that can potentially be prevented or treated, 5 non-childhood onset neurological disorders, 13 X-linked disorders (mainly STS and DMD deletions), and 1 deletion of the SHOX gene. Long-term follow-up revealed that newborns with high penetrance neurosusceptibility CNVs exhibited clinical manifestations more frequently than those with low penetrance CNVs. At the time of reporting, 52 IFs were disclosed, while 17 were not. According to the new consensus criteria, 43 IFs would now be reported, 17 would not, and 9 would depend on parental decision. CNVs consistent with the referral reason were identified in 4% of cases. CONCLUSION This study represents the largest series rigorously documenting all identified IFs in consecutive pregnancies evaluated by microarray, including both reported and unreported findings. IFs were found at a higher frequency than previously recognized, underscoring the need for specific clinical attention. Comprehensive consensus reporting recommendations were developed to ensure uniformity of criteria, and an ad hoc committee was established to manage complex cases.
Collapse
Affiliation(s)
- Irene Mademont-Soler
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial ICS Girona, Hospital Universitari de Girona Dr. Josep Trueta, Girona, Spain
- Grup de Trastorns del Neurodesenvolupament, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain
| | - Neus Castells-Sarret
- Àrea de Genètica Clínica i Molecular, Hospital Universitari Vall d'Hebron, Institut Català de la Salut, Barcelona, Spain
- Grup de Medicina Genètica, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Adela Cisneros
- Servei Hematologia Laboratori, Laboratori Clínic Territorial ICS Metropolitana Nord, ICO-Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Laura Foj
- Unitat de Citogenètica i Genètica Mèdica, Laboratori Clínic Territorial ICS Lleida, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Clara Benavent-Bofill
- Unitat de Genètica Clínica, Laboratori Clínic Territorial ICS Camp de Tarragona i Terres de l'Ebre, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Mar Xunclà
- Àrea de Genètica Clínica i Molecular, Hospital Universitari Vall d'Hebron, Institut Català de la Salut, Barcelona, Spain
- Grup de Medicina Genètica, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina Viñas-Jornet
- Àrea de Genètica Clínica i Molecular, Hospital Universitari Vall d'Hebron, Institut Català de la Salut, Barcelona, Spain
- Grup de Medicina Genètica, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andrea Ros
- Servei de Genètica Clínica, Laboratori Clínic Territorial ICS Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Unitat de Recerca en Genòmica Clínica, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Natalia Rey
- Àrea de Genètica Clínica i Molecular, Hospital Universitari Vall d'Hebron, Institut Català de la Salut, Barcelona, Spain
- Grup de Medicina Genètica, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ignacio Blanco
- Servei de Genètica Clínica, Laboratori Clínic Territorial ICS Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Unitat de Recerca en Genòmica Clínica, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Ricard López-Ortega
- Unitat de Citogenètica i Genètica Mèdica, Laboratori Clínic Territorial ICS Lleida, Hospital Universitari Arnau de Vilanova, Lleida, Spain
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - María Obón
- Àrea de Genètica Clínica i Consell Genètic, Laboratori Clínic Territorial ICS Girona, Hospital Universitari de Girona Dr. Josep Trueta, Girona, Spain
- Grup de Trastorns del Neurodesenvolupament, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, Girona, Spain
| | - Alberto Plaja
- Àrea de Genètica Clínica i Molecular, Hospital Universitari Vall d'Hebron, Institut Català de la Salut, Barcelona, Spain
- Grup de Medicina Genètica, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Braconi D, Nadwa H, Bernardini G, Santucci A. Omics and rare diseases: challenges, applications, and future perspectives. Expert Rev Proteomics 2025; 22:107-122. [PMID: 39956998 DOI: 10.1080/14789450.2025.2468300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/08/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
INTRODUCTION Rare diseases (RDs) are a heterogeneous group of diseases recognized as a relevant global health priority but posing aspects of complexity, such as geographical scattering of affected individuals, improper/late diagnosis, limited awareness, difficult surveillance and monitoring, limited understanding of natural history, and lack of treatment. Usually, RDs have a pediatric onset and are life-long, multisystemic, and associated with a poor prognosis. AREAS COVERED In this work, we review how high-throughput omics technologies such as genomics, transcriptomics, proteomics, metabolomics, epigenomics, and other well-established omics, which are increasingly more affordable and efficient, can be applied to the study of RDs promoting diagnosis, understanding of pathological mechanisms, biomarker discovery, and identification of treatments. EXPERT OPINION RDs, despite their challenges, offer a niche where collaborative efforts and personalized treatment strategies might be feasible using omics technologies. Specialized consortia fostering multidisciplinary collaboration, data sharing, and the development of biobanks and registries can be built; multi-omics approaches, including so far less exploited omics technologies, along with the implementation of AI tools can be undertaken to deepen our understanding of RDs, driving biomarker discovery and clinical interventions. Nevertheless, technical, ethical, legal, and societal issues must be clearly defined and addressed.
Collapse
Affiliation(s)
- Daniela Braconi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Haidara Nadwa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulia Bernardini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| |
Collapse
|
17
|
Mardinoglu A, Palsson BØ. Genome-scale models in human metabologenomics. Nat Rev Genet 2025; 26:123-140. [PMID: 39300314 DOI: 10.1038/s41576-024-00768-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/22/2024]
Abstract
Metabologenomics integrates metabolomics with other omics data types to comprehensively study the genetic and environmental factors that influence metabolism. These multi-omics data can be incorporated into genome-scale metabolic models (GEMs), which are highly curated knowledge bases that explicitly account for genes, transcripts, proteins and metabolites. By including all known biochemical reactions catalysed by enzymes and transporters encoded in the human genome, GEMs analyse and predict the behaviour of complex metabolic networks. Continued advancements to the scale and scope of GEMs - from cells and tissues to microbiomes and the whole body - have helped to design effective treatments and develop better diagnostic tools for metabolic diseases. Furthermore, increasing amounts of multi-omics data are incorporated into GEMs to better identify the underlying mechanisms, biomarkers and potential drug targets of metabolic diseases.
Collapse
Affiliation(s)
- Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.
| | - Bernhard Ø Palsson
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA.
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
18
|
Ziegler A, Koval-Burt C, Kay DM, Suchy SF, Begtrup A, Langley KG, Hernan R, Amendola LM, Boyd BM, Bradley J, Brandt T, Cohen LL, Coffey AJ, Devaney JM, Dygulska B, Friedman B, Fuleihan RL, Gyimah A, Hahn S, Hofherr S, Hruska KS, Hu Z, Jeanne M, Jin G, Johnson DA, Kavus H, Leibel RL, Lobritto SJ, McGee S, Milner JD, McWalter K, Monaghan KG, Orange JS, Pimentel Soler N, Quevedo Y, Ratner S, Retterer K, Shah A, Shapiro N, Sicko RJ, Silver ES, Strom S, Torene RI, Williams O, Ustach VD, Wynn J, Taft RJ, Kruszka P, Caggana M, Chung WK. Expanded Newborn Screening Using Genome Sequencing for Early Actionable Conditions. JAMA 2025; 333:232-240. [PMID: 39446378 PMCID: PMC11503470 DOI: 10.1001/jama.2024.19662] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/03/2024] [Indexed: 10/27/2024]
Abstract
Importance The feasibility of implementing genome sequencing as an adjunct to traditional newborn screening (NBS) in newborns of different racial and ethnic groups is not well understood. Objective To report interim results of acceptability, feasibility, and outcomes of an ongoing genomic NBS study in a diverse population in New York City within the context of the New York State Department of Health Newborn Screening Program. Design, Setting, and Participants The Genomic Uniform-screening Against Rare Disease in All Newborns (GUARDIAN) study was a multisite, single-group, prospective, observational investigation of supplemental newborn genome screening with a planned enrollment of 100 000 participants. Parent-reported race and ethnicity were recorded at the time of recruitment. Results of the first 4000 newborns enrolled in 6 New York City hospitals between September 2022 and July 2023 are reported here as part of a prespecified interim analysis. Exposure Sequencing of 156 early-onset genetic conditions with established interventions selected by the investigators were screened in all participants and 99 neurodevelopmental disorders associated with seizures were optional. Main Outcomes and Measures The primary outcome was screen-positive rate. Additional outcomes included enrollment rate and successful completion of sequencing. Results Over 11 months, 5555 families were approached and 4000 (72.0%) consented to participate. Enrolled participants reflected a diverse group by parent-reported race (American Indian or Alaska Native, 0.5%; Asian, 16.5%; Black, 25.1%; Native Hawaiian or Other Pacific Islander, 0.1%; White, 44.7%; 2 or more races, 13.0%) and ethnicity (Hispanic, 44.0%; not Hispanic, 56.0%). The majority of families consented to screening of both groups of conditions (both groups, 90.6%; disorders with established interventions only, 9.4%). Testing was successfully completed for 99.6% of cases. The screen-positive rate was 3.7%, including treatable conditions that are not currently included in NBS. Conclusions and Relevance These interim findings demonstrate the feasibility of targeted interpretation of a predefined set of genes from genome sequencing in a population of different racial and ethnic groups. DNA sequencing offers an additional method to improve screening for conditions already included in NBS and to add those that cannot be readily screened because there is no biomarker currently detectable in dried blood spots. Additional studies are required to understand if these findings are generalizable to populations of different racial and ethnic groups and whether introduction of sequencing leads to changes in management and improved health outcomes. Trial Registration ClinicalTrials.gov Identifier: NCT05990179.
Collapse
Affiliation(s)
- Alban Ziegler
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Carrie Koval-Burt
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Denise M. Kay
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany
| | | | | | | | - Rebecca Hernan
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | | | - Brenna M. Boyd
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Jennifer Bradley
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany
| | | | | | | | | | - Beata Dygulska
- NewYork-Presbyterian Brooklyn Methodist Hospital, New York
| | | | - Ramsay L. Fuleihan
- Division of Allergy, Immunology & Rheumatology, Columbia University Irving Medical Center, New York, New York
| | - Awura Gyimah
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Sihoun Hahn
- Department of Pediatrics, Biochemical Genetics, University of Washington, Seattle Children’s Hospital, Seattle
| | | | | | - Zhanzhi Hu
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
| | - Médéric Jeanne
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
| | - Guanjun Jin
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | | | - Haluk Kavus
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Rudolph L. Leibel
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Steven J. Lobritto
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | | | - Joshua D. Milner
- Division of Allergy, Immunology & Rheumatology, Columbia University Irving Medical Center, New York, New York
| | | | | | - Jordan S. Orange
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Nicole Pimentel Soler
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Yeyson Quevedo
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Samantha Ratner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | | | - Ankur Shah
- Division of Pediatric Cardiology, Department of Pediatrics, Weill Cornell Medical College, New York, New York
| | | | - Robert J. Sicko
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany
| | - Eric S. Silver
- Division of Pediatric Cardiology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | | | | | - Olatundun Williams
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | | | - Julia Wynn
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | | | | | - Michele Caggana
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany
| | - Wendy K. Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Maron JL, Terry SF. From the Perspective of the Child: Ethical Considerations for the Implementation of Genomic Sequencing into Neonatal and Pediatric Care. Clin Chem 2025; 71:18-20. [PMID: 39749504 DOI: 10.1093/clinchem/hvae112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 01/04/2025]
Affiliation(s)
- Jill L Maron
- Department of Pediatrics, Pediatrician-in-Chief, Women & Infants Hospital of Rhode Island, Providence, RI, United States
| | - Sharon F Terry
- President and CEO, Genetic Alliance, Inc., Washington, DC, United States
| |
Collapse
|
20
|
Jeanne M, Chung WK. DNA Sequencing in Newborn Screening: Opportunities, Challenges, and Future Directions. Clin Chem 2025; 71:77-86. [PMID: 39749512 DOI: 10.1093/clinchem/hvae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/01/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Newborn screening is a public health system designed to identify infants at risk for conditions early in life to facilitate timely intervention and treatment to prevent or mitigate adverse health outcomes. Newborn screening programs use tandem mass spectrometry as a platform to detect several treatable inborn errors of metabolism, and the T-cell receptor excision circle assay to detect some inborn errors of the immune system. Recent advancements in DNA sequencing have decreased the cost of sequencing and allow us to consider DNA sequencing as an additional platform to complement other newborn screening methods. CONTENT This review provides an overview of DNA-based newborn screening, including its applications, opportunities, challenges, and future directions. We discuss the potential benefits of expanded DNA sequencing in newborn screening, such as expanding conditions screened and improved specificity and sensitivity of currently screened conditions. Additionally, we examine the ethical, legal, and social implications of implementing genomic sequencing in newborn screening programs, including issues related to consent, privacy, equity, data interpretation, scalability, and psychosocial impact on families. Additionally, we explore emerging strategies for addressing current limitations and advancing the field of newborn screening. SUMMARY DNA sequencing in newborn screening has the potential to improve the diagnosis and management of rare diseases but also presents significant challenges that need to be addressed before implementation at the population level.
Collapse
Affiliation(s)
- Médéric Jeanne
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
21
|
Bick SL, Nathan A, Park H, Green RC, Wojcik MH, Gold NB. Estimating the sensitivity of genomic newborn screening for treatable inherited metabolic disorders. Genet Med 2025; 27:101284. [PMID: 39355980 PMCID: PMC11717630 DOI: 10.1016/j.gim.2024.101284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
PURPOSE Over 30 research groups and companies are exploring newborn screening using genomic sequencing (NBSeq), but the sensitivity of this approach is not well understood. METHODS We identified individuals with treatable inherited metabolic disorders (IMDs) and ascertained the proportion whose DNA analysis revealed explanatory deleterious variants (EDVs). We examined variables associated with EDV detection and estimated the sensitivity of DNA-first NBSeq. We further predicted the annual rate of true-positive and false-negative NBSeq results in the United States for several conditions on the Recommended Uniform Screening Panel. RESULTS We identified 635 individuals with 80 unique IMDs. In univariate analyses, Black race (OR = 0.37, 95% CI: 0.16-0.89, P = .02) and public insurance (OR = 0.60, 95% CI: 0.39-0.91, P = .02) were less likely to be associated with finding EDVs. Had all individuals been screened with NBSeq, the sensitivity would have been 80.3%. We estimated that between 0 and 649.9 cases of Recommended Uniform Screening Panel IMDs would be missed annually by NBSeq in the United States. CONCLUSION The overall sensitivity of NBSeq for treatable IMDs is estimated at 80.3%. That sensitivity will likely be lower for Black infants and those who are on public insurance.
Collapse
Affiliation(s)
- Sarah L Bick
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Division of Medical Genetics and Metabolism, Massachusetts General Hospital for Children, Boston, MA; Harvard Medical School, Boston, MA
| | - Aparna Nathan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Hannah Park
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA; Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA
| | - Robert C Green
- Harvard Medical School, Boston, MA; Mass General Brigham, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA; Ariadne Labs, Boston, MA
| | - Monica H Wojcik
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA; Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA
| | - Nina B Gold
- Division of Medical Genetics and Metabolism, Massachusetts General Hospital for Children, Boston, MA; Harvard Medical School, Boston, MA.
| |
Collapse
|
22
|
Aishworiya R, Chin HL, Savulescu J. Should newborn genetic testing for autism be introduced? JOURNAL OF MEDICAL ETHICS 2024:jme-2024-110166. [PMID: 39626956 DOI: 10.1136/jme-2024-110166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025]
Abstract
This manuscript provides a review of the potential role of newborn genetic testing for autism, and whether the state has an inherent responsibility to facilitate and subsidise this. This is situated within the broader construct of benefits and limitations of genetic testing currently. Potential benefits of such presymptomatic genetic testing include facilitating earlier diagnosis and access to appropriate intervention which can improve the treatment outcome for the child and indirectly benefit caregivers and society by reducing the care needs of the child and adult in future. However, there are several limitations to newborn genetic testing including the variable penetrance of 'autism-risk' genes, marked phenotypic heterogeneity of autism, real-world limitations in access to treatment, potential psychological harm to caregivers and financial considerations. We hence argue for facilitation of diagnostic genetic testing instead, especially for parents who seek to have greater understanding of recurrence likelihoods, related to reproductive decision-making. Facilitation of such testing can be in the form of both financial subsidies and infrastructural elements including availability of testing facilities and trained healthcare personnel for individualised pregenetic and postgenetic test counselling.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- Division of Developmental and Behavioural Paediatrics, Department of Paediatrics, Khoo Teck Puat - National University Children's Medical Institute, National University Hospital, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hui-Lin Chin
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Genetics and Metabolism, Department of Paediatrics, Khoo Teck Puat - National University Children's Medical Institute, National University Hospital, Singapore
| | - Julian Savulescu
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Oxford Uehiro Centre for Practical Ethics, Faculty of Philosophy, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Mori M, Chaudhari BP, Ream MA, Kemper AR. Promises and challenges of genomic newborn screening (NBS) - lessons from public health NBS programs. Pediatr Res 2024:10.1038/s41390-024-03689-0. [PMID: 39516573 DOI: 10.1038/s41390-024-03689-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Newborn screening (NBS) in the United States began in the 1960s to detect inborn errors of metabolism that benefited from presymptomatic treatment compared with treatment after the development of symptoms and diagnosis. Over time, it expanded to include endocrinological disorders, hematological disorders, immunodeficiencies, and other treatable diseases such as lysosomal storage diseases (LSD), cystic fibrosis, X-linked adrenoleukodystrophy, and spinal muscular dystrophy. This expansion has been driven by new technologies (e.g., tandem mass spectrometry) and novel treatments (e.g., enzyme replacement therapy and stem cell transplant for LSDs). Advances in next-generation gene sequencing (NGS) enable rapid identification of many additional conditions that might benefit from early presymptomatic intervention. We review the NGS technologies that evolved as diagnostic testing and suggest issues to be resolved before their potential application to screening the asymptomatic population. We illustrate the importance of selecting diseases to be screened and propose recommendations to follow when variants of uncertain significance are found. We address ethical issues around achieving equity in the sensitivity of genomic NBS, access to follow-up and management, especially for people from diverse backgrounds, and other considerations. Finally, we discuss the potential benefits and harms of genomic NBS to the overall health of children with monogenic diseases. IMPACT: Genomic newborn screening programs are ongoing worldwide. Public discussion is needed as to whether genomic newborn screening should be offered as a public health program and, if so, what conditions should be screened for. Providers should understand that the sensitivity of genomic newborn screening is especially low for newborns from non-European populations. Methylation, large structural variants and repeat expansion variants are not amenable to next-generation sequencing-based genomic newborn screening. The article serves as a comprehensive guide to understanding issues that need to be solved before genomic newborn screening is implemented as a public health program.
Collapse
Affiliation(s)
- Mari Mori
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA.
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
| | - Bimal P Chaudhari
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Division of Neonatology, Nationwide Children's Hospital, Columbus, OH, USA
- The Steve and Cindy Rasmussen Institute of Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Margie A Ream
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA
- Division of Division of Child Neurology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Alex R Kemper
- The Ohio State University College of Medicine Department of Pediatrics, Columbus, OH, USA
- Division of Primary Care Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
24
|
Lantos JD. Neonatal bioethics, AI, and genomics. Early Hum Dev 2024; 198:106130. [PMID: 39405800 DOI: 10.1016/j.earlhumdev.2024.106130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/04/2024] [Indexed: 11/12/2024]
Abstract
Artificial intelligence (AI) and synthetic biology will transform civilization. The only question is how. In this paper, I explore some recent developments in medical AI, genomics, and synthetic biology. I speculate about the implications of these technologies for the practice of medicine and conclude that they will fundamentally alter our ideas of health, disease, medicine, and what it means to be human. I have three conclusions. First, AI and synthetic biology will force us to examine whether humanistic skills can be uniquely human and, if so, whether they are skills or natural gifts. AI will offer opportunities to examine what we mean by empathy, how we develop skills in communication, and when the human touch is essential for healing. Second, these technologies will change the ways that we will assess the value of doctors' work. Skills that can be mechanized will be devalued and delegated to machines. Doctors will either need to learn new skills or become irrelevant. Finally, AI and synthetic biology will force us to deeply examine what it means to be human. For humans to remain uniquely valuable, we will need to develop those aspects of our humanity that cannot be mechanized. Doctors will need to carefully attune themselves to the non-physical aspects of disease and suffering. Ultimately, AI and synthetic biology will force us to redesign both or systems of medical education and the systems of health care delivery in ways that meet both the medical and non-medical needs of patients.
Collapse
Affiliation(s)
- John D Lantos
- JDL Bioethics Consulting, 385 Lake Shore Drive, Pleasantville, NY 10570, United States of America.
| |
Collapse
|
25
|
MacDuffie KE, Cohn B, Appelbaum P, Brothers KB, Doherty D, Goldenberg AJ, Reynolds E, Smith HS, Wheeler A, Yu JH. Early Intervention services in the era of genomic medicine: setting a research agenda. Pediatr Res 2024:10.1038/s41390-024-03668-5. [PMID: 39438712 DOI: 10.1038/s41390-024-03668-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
Newborn genomic sequencing (NBSeq) has the potential to substantially improve early detection of rare genetic conditions, allowing for pre-symptomatic treatment to optimize outcomes. Expanding conceptions of the clinical utility of NBSeq include earlier access to behavioral early intervention to support the acquisition of core motor, cognitive, communication, and adaptive skills during critical windows in early development. However, important questions remain about equitable access to early intervention programs for the growing number of infants identified with a genetic condition via NBSeq. We review the current NBSeq public health, clinical, and research landscape, and highlight ongoing international research efforts to collect population-level data on the utility of NBSeq for healthy newborns. We then explore the challenges facing a specific Early Intervention (EI) system-the US federally supported "Part C" system-for meeting the developmental needs of young children with genetic diagnoses, including structural limitations related to funding, variable eligibility criteria, and lack of collaboration with newborn screening programs. We conclude with a set of questions to guide future research at the intersection of NBSeq, newborn screening, and EI, which once answered, can steer future policy to ensure that EI service systems can optimally support the developmental needs of infants impacted by broader implementation of NBSeq. IMPACT: Existing literature on the clinical benefits of genome sequencing in newborns tends to focus on earlier provision of medical interventions, with less attention to the ongoing developmental needs of very young children with genetic conditions. This review outlines the developmental needs of a growing number of children diagnosed with genetic conditions in infancy and describes the strengths and limitations of the United States Early Intervention system (IDEA Part C) for meeting those needs.
Collapse
Affiliation(s)
- Katherine E MacDuffie
- Treuman Katz Center for Pediatric Bioethics and Palliative Care, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.
| | - Betty Cohn
- Institute for Public Health Genetics, University of Washington, Seattle, WA, USA
| | - Paul Appelbaum
- Department of Psychiatry, Columbia University Irving Medical Center and NY State Psychiatric Institute, New York, NY, USA
| | - Kyle B Brothers
- Department of Pediatrics, University of Louisville, Louisville, KY, USA
| | - Dan Doherty
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Aaron J Goldenberg
- Department of Bioethics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Hadley Stevens Smith
- Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Center for Bioethics, Harvard Medical School, Boston, MA, USA
| | - Anne Wheeler
- RTI International, Research Triangle Park, NC, USA
| | - Joon-Ho Yu
- Treuman Katz Center for Pediatric Bioethics and Palliative Care, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Public Health Genetics, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Shubina J, Tolmacheva E, Maslennikov D, Kochetkova T, Mukosey I, Sadelov I, Goltsov A, Barkov I, Ekimov A, Rogacheva M, Stupko O, Pavlova N, Kuznetsova M, Dokshukina A, Vasiliev G, Bolshakova A, Kovalskaia V, Korovko A, Pomerantseva E, Tsabai P, Buyanovskaya O, Zaretskaya N, Karetnikova N, Grebenshchikova E, Degtyareva A, Bokerija E, Kholin A, Rebrikov D, Degtyarev D, Trofimov D, Sukhih G. WES-based screening of 7,000 newborns: A pilot study in Russia. HGG ADVANCES 2024; 5:100334. [PMID: 39033325 PMCID: PMC11350501 DOI: 10.1016/j.xhgg.2024.100334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/17/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024] Open
Abstract
The effective implementation of whole-exome sequencing- and whole-genome sequencing-based diagnostics in the management of children affected with genetic diseases and the rapid decrease in the cost of next-generation sequencing (NGS) enables the expansion of this method to newborn genetic screening programs. Such NGS-based screening greatly increases the number of diseases that can be detected compared to conventional newborn screening, as the latter is aimed at early detection of a limited number of inborn diseases. Moreover, genetic testing provides new possibilities for family members of the proband, as many variants responsible for adult-onset conditions are inherited from the parents. However, the idea of NGS-based screening in healthy children raises issues of medical and ethical integrity as well as technical questions, including interpretation of the observed variants. Pilot studies have shown that both parents and medical professionals have moved forward and are enthused about these new possibilities. However, either the number of participants or the number of genes studied in previous investigations thus far has been limited to a few hundred, restricting the scope of potential findings. Our current study (NCT05325749) includes 7,000 apparently healthy infants born at our center between February 2021 and May 2023, who were screened for pathogenic variants in 2,350 genes. Clinically significant variants associated with early-onset diseases that can be treated, prevented, or where symptoms can be alleviated with timely introduced symptomatic therapy, were observed in 0.9% of phenotypically normal infants, 2.1% of the screened newborns were found to carry variants associated with reduced penetrance or monogenic diseases of adult-onset and/or variable expressivity, and 0.3% had chromosomal abnormalities. Here, we report our results and address questions regarding the interpretation of variants in newborns who were presumed to be healthy.
Collapse
Affiliation(s)
- Jekaterina Shubina
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia.
| | - Ekaterina Tolmacheva
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Dmitry Maslennikov
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Taisiya Kochetkova
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Irina Mukosey
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Igor Sadelov
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Andrey Goltsov
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Ilya Barkov
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Aleksey Ekimov
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Margarita Rogacheva
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Olga Stupko
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Nadezhda Pavlova
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Maria Kuznetsova
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Alina Dokshukina
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Grigory Vasiliev
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Anna Bolshakova
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Valeriia Kovalskaia
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Anastasia Korovko
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Ekaterina Pomerantseva
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Polina Tsabai
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Olga Buyanovskaya
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Nadezhda Zaretskaya
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Natalia Karetnikova
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | | | - Anna Degtyareva
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Ekaterina Bokerija
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Alexey Kholin
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Denis Rebrikov
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia; Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Dmitry Degtyarev
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Dmitriy Trofimov
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| | - Gennady Sukhih
- National Medical Research Center for Obstetrics, Gynecology, and Perinatology of the Ministry of Health of the Russian Federation, 117198 Moscow, Russia
| |
Collapse
|
27
|
Del Rosario MC, Walmsley SA, Harrison BW, Stephens CT, Zettler B, Rivera-Cruz G, Agrawal P, Brower A, Chigbu S, Christensen KD, Genetti CA, Givens R, Gold NB, Reeves IV, Schichter I, Shariat H, Simon S, Smith HS, Uveges M, Green RC, Holm IA, Pereira S. Diverse Participant Recruitment for Infant Sequencing in the BabySeq Project. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.01.24314717. [PMID: 39417101 PMCID: PMC11482871 DOI: 10.1101/2024.10.01.24314717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Purpose It is essential that studies of genomic sequencing (GS) in newborns and children include individuals from under-represented racial and ethnic groups (URG) to ensure future applications are equitably implemented. We conducted interviews with parents from URG to better understand their perspectives on GS research, develop strategies to reduce barriers to enrollment, and facilitate research participation. Methods Semi-structured interviews with 50 parents from URG. Results Nearly all parents (44) said they would be interested in participating in an infant GS study. Parents were interested in participating in GS research for reasons including clinical utility, personal utility, and/or family health benefits. Deterrents to enrollment cited by parents were discomfort with enrollment procedures (e.g., not wanting a heel stick), limited emotional bandwidth, unfavorable perceptions of the study, and concerns about potential results. Most parents (35 of 40) said they would want to receive all types of genetic results, including actionable and non-actionable, as well as childhood- and adult-onset. Conclusion Our findings demonstrate that parents from URG are interested in participating in GS research. Based upon these findings, we provide recommendations for designing GS studies that are responsive to their concerns.
Collapse
|
28
|
Smith HS, Zettler B, Genetti CA, Hickingbotham MR, Coleman TF, Lebo M, Nagy A, Zouk H, Mahanta L, Christensen KD, Pereira S, Shah ND, Gold NB, Walmsley S, Edwards S, Homayouni R, Krasan GP, Hakonarson H, Horowitz CR, Gelb BD, Korf BR, McGuire AL, Holm IA, Green RC. The BabySeq Project: A clinical trial of genome sequencing in a diverse cohort of infants. Am J Hum Genet 2024; 111:2094-2106. [PMID: 39288765 PMCID: PMC11480845 DOI: 10.1016/j.ajhg.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/15/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Efforts to implement and evaluate genome sequencing (GS) as a screening tool for newborns and infants are expanding worldwide. The first iteration of the BabySeq Project (2015-2019), a randomized controlled trial of newborn sequencing, produced novel evidence on medical, behavioral, and economic outcomes. The second iteration of BabySeq, which began participant recruitment in January 2023, examines GS outcomes in a larger, more diverse cohort of more than 500 infants up to one year of age recruited from pediatric clinics at several sites across the United States. The trial aims for families who self-identify as Black/African American or Hispanic/Latino to make up more than 50% of final enrollment, and key aspects of the trial design were co-developed with a community advisory board. All enrolled families receive genetic counseling and a family history report. Half of enrolled infants are randomized to receive GS with comprehensive interpretation of pathogenic and likely pathogenic variants in more than 4,300 genes associated with childhood-onset and actionable adult-onset conditions, as well as larger-scale chromosomal copy number variants classified as pathogenic or likely pathogenic. GS result reports include variants associated with disease (Mendelian disease risks) and carrier status of autosomal-recessive and X-linked disorders. Investigators evaluate the utility and impacts of implementing a GS screening program in a diverse cohort of infants using medical record review and longitudinal parent surveys. In this perspective, we describe the rationale for the second iteration of the BabySeq Project, the outcomes being assessed, and the key decisions collaboratively made by the study team and community advisory board.
Collapse
Affiliation(s)
- Hadley Stevens Smith
- Department of Population Medicine, Precision Medicine Translational Research (PROMoTeR) Center, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA; Center for Bioethics, Harvard Medical School, Boston, MA 02215, USA
| | - Bethany Zettler
- Department of Medicine, Mass General Brigham, Boston, MA 02115, USA; Ariadne Labs, Boston, MA 02215, USA
| | - Casie A Genetti
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA
| | - Madison R Hickingbotham
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA
| | - Tanner F Coleman
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Matthew Lebo
- Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Boston, MA 02139, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Anna Nagy
- Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Boston, MA 02139, USA
| | - Hana Zouk
- Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Boston, MA 02139, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Lisa Mahanta
- Laboratory for Molecular Medicine, Mass General Brigham Personalized Medicine, Boston, MA 02139, USA
| | - Kurt D Christensen
- Department of Population Medicine, Precision Medicine Translational Research (PROMoTeR) Center, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Stacey Pereira
- Center for Medical Ethics & Health Policy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nidhi D Shah
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA; Dartmouth Health Children's, Lebanon, NH 03756, USA
| | - Nina B Gold
- Massachusetts General Hospital for Children, Division of Medical Genetics and Metabolism, Boston, MA 02114, USA; Harvard Medical School, Department of Pediatrics, Boston, MA 02115, USA
| | - Sheyenne Walmsley
- Department of Medicine, Mass General Brigham, Boston, MA 02115, USA; Ariadne Labs, Boston, MA 02215, USA
| | | | - Ramin Homayouni
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Graham P Krasan
- Department of Pediatrics, Corewell Health William Beaumont University Hospital, Royal Oak, MI 48073, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, The Joseph Stokes Jr. Research Institute of Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carol R Horowitz
- Department of Population Health Science and Policy, Institute for Health Equity Research, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and the Departments of Pediatrics and Genetics & Genomic Sciences Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruce R Korf
- Department of Genetics, UAB Heersink School of Medicine, Birmingham, AL 35233, USA
| | - Amy L McGuire
- Center for Medical Ethics & Health Policy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ingrid A Holm
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Department of Pediatrics, Boston, MA 02115, USA.
| | - Robert C Green
- Department of Medicine, Mass General Brigham, Boston, MA 02115, USA; Ariadne Labs, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| |
Collapse
|
29
|
Shaikh H, Lyle ANJ, Oslin E, Gray MM, Weiss EM. Eligible Infants Included in Neonatal Clinical Trials and Reasons for Noninclusion: A Systematic Review. JAMA Netw Open 2024; 7:e2441372. [PMID: 39453652 PMCID: PMC11581680 DOI: 10.1001/jamanetworkopen.2024.41372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/31/2024] [Indexed: 10/26/2024] Open
Abstract
Importance Results of clinical trials can only represent included participants, and many neonatal trials fail due to insufficient participation. Infants not included in research may differ from those included in meaningful ways, biasing the sample and limiting the generalizability of findings. Objective To describe the proportion of eligible infants included in neonatal clinical trials and the reasons for noninclusion. Evidence Review A systematic search of Cochrane CENTRAL was performed by retrieving articles meeting the following inclusion criteria: full-length, peer-reviewed articles describing clinical trial results in at least 20 human infants from US neonatal intensive care units, published in English, and added to Cochrane CENTRAL between 2017 and 2022. Retrieved articles were screened for inclusion by 2 independent researchers. Findings In total 120 articles met inclusion criteria and 91 of these (75.8%) reported the number of infants eligible for participation, which totaled 26 854 in aggregate. Drawing from these, an aggregate of 11 924 eligible infants (44.4%) were included in reported results. Among all eligible infants, most reasons for noninclusion in results were classified as modifiable or potentially modifiable by the research team. Parents declining to participate (8004 infants [29.8%]) or never being approached (2507 infants [9.3%]) were the 2 predominant reasons for noninclusion. Other modifiable reasons included factors related to study logistics, such as failure to appropriately collect data on enrolled infants (859 of 26 854 infants [3.2%]) and other reasons (1907 of 26 854 infants [7.1%]), such as loss to follow-up or eligible participants that were unaccounted for. Nonmodifiable reasons, including clinical change or death, accounted for a small proportion of eligible infants who were not included (858 of 26 854 infants [3.2%]). Conclusions and Relevance This systematic review of reporting on eligible infants included and not included in neonatal clinical trials highlights the need for improved documentation on the flow of eligible infants through neonatal clinical trials and may also inform recruitment expectations for trialists designing future protocols. Improved adherence to standardized reporting may clarify which potential participants are being missed, improving understanding of the generalizability of research findings. Furthermore, these findings suggest that future work to understand why parents decline to participate in neonatal research trials and why some are never approached about research may help increase overall participation.
Collapse
Affiliation(s)
- Henna Shaikh
- Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Allison N J Lyle
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Medical Group-Neonatology, Louisville, Kentucky
| | - Ellie Oslin
- Department of Pediatrics, University of Washington School of Medicine, Seattle
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Medical Group-Neonatology, Louisville, Kentucky
| | - Megan M Gray
- Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Elliott Mark Weiss
- Department of Pediatrics, University of Washington School of Medicine, Seattle
- Treuman Katz Center for Pediatric Bioethics & Palliative Care, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
30
|
Al-Dewik N, Abuarja T, Younes S, Nasrallah G, Alsharshani M, Ibrahim FE, Samara M, Farrell T, Abdulrouf PV, Qoronfleh MW, Al Rifai H. Precision medicine activities and opportunities for shaping maternal and neonatal health in Qatar. Per Med 2024; 21:313-333. [PMID: 39347749 DOI: 10.1080/17410541.2024.2394397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 08/16/2024] [Indexed: 10/01/2024]
Abstract
Precision Medicine (PM) is a transformative clinical medicine strategy that aims to revolutionize healthcare by leveraging biological information and biomarkers. In the context of maternal and neonatal health, PM enables personalized care from preconception through the postnatal period. Qatar has emerged as a key player in PM research, with dedicated programs driving advancements and translating cutting-edge research into clinical applications. This article delves into neonatal and maternal health in Qatar, emphasizing PM programs and initiatives that have been implemented. It also features noteworthy clinical cases that demonstrate the effectiveness of precision interventions. Furthermore, the article highlights the role of pharmacogenomics in addressing various maternal health conditions. The review further explores potential advancements in the application of PM in maternal and neonatal healthcare in Qatar.
Collapse
Affiliation(s)
- Nader Al-Dewik
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
- Department of Neonatology, Neonatal Intensive Care Unit, Newborn Screening Unit, Women's Wellness & Research Center, Hamad Medical Corporation, Doha, 3050, Qatar
- Translational Research Institute (TRI), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
- Genomics & Precision Medicine (GPM), College of Health & Life Science (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Tala Abuarja
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Salma Younes
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University (QU), Doha, 2713, Qatar
| | - Gheyath Nasrallah
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University (QU), Doha, 2713, Qatar
| | - Mohamed Alsharshani
- Diagnostic Genetics Division (DGD), Department of Laboratory Medicine & Pathology (DLMP), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Faisal E Ibrahim
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Muthanna Samara
- Department of Psychology, Kingston University London, Kingston upon Thames, London, KT1 2EE, United Kingdom
| | - Thomas Farrell
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Palli Valapila Abdulrouf
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - M Walid Qoronfleh
- Q3 Research Institute (QRI), Healthcare Research & Policy Division, 7227 Rachel Drive, Ypsilanti, MI 48917, USA
| | - Hilal Al Rifai
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
- Department of Neonatology, Neonatal Intensive Care Unit, Newborn Screening Unit, Women's Wellness & Research Center, Hamad Medical Corporation, Doha, 3050, Qatar
| |
Collapse
|
31
|
Seed L, Scott A, Pichini A, Peter M, Tadros S, Sortica da Costa C, Hill M. Perceptions of genomic newborn screening: a cross-sectional survey conducted with UK medical students. BMJ Open 2024; 14:e089108. [PMID: 39317512 PMCID: PMC11423729 DOI: 10.1136/bmjopen-2024-089108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND With the potential to identify a vast number of rare diseases soon after birth, genomic newborn screening (gNBS) could facilitate earlier interventions and improve health outcomes. Designing a gNBS programme will involve balancing stakeholders' opinions and addressing concerns. The views of medical students-future clinicians who would deliver gNBS-have not yet been explored. METHODS We conducted a nationwide online survey of UK medical students via the REDCap platform. Perceptions of gNBS, including scope of testing and potential benefits and drawbacks, were explored using a mix of multiple-choice questions, Likert scales, visual analogue scales and free-text questions. RESULTS In total, 116 medical students across 16 universities participated. Overall, 45% supported gNBS, with a positively skewed mean support score of 3.24 (SD 1.26, range: 1.0-5.0), and 55% felt it relevant to their future practice. Almost all agreed that infant-onset and childhood-onset diseases and conditions with effective treatments should be included. Most felt that earlier interventions and personalised care would be the most important benefit of gNBS. Other perceived benefits included earlier diagnoses, diagnosing more patients and enabling research for new treatments. However, several perceived challenges were highlighted: risk of genomic discrimination, incidental or uncertain findings, data security and breaching children's future autonomy. Students expressed conflicting opinions on the psychological impact on families, but most were concerned about a lack of support due to current resource limitations in health services. Students frequently reported having insufficient knowledge to form an opinion, which may reflect gaps in genomics education at medical school and the current lack of evidence base for gNBS. CONCLUSION Although some support for gNBS was demonstrated, ethicolegal and social challenges were raised, emphasising a need for ongoing discussions about the implications of gNBS.
Collapse
Affiliation(s)
- Lydia Seed
- Infection, Immunity and Inflammation Department, University College London Great Ormond Street Institute of Child Health, London, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Anna Scott
- Infection, Immunity and Inflammation Department, University College London Great Ormond Street Institute of Child Health, London, UK
- School of Medicine, University of Southampton, Southampton, UK
| | | | - Michelle Peter
- NHS North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Genetics and Genomic Medicine, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Shereen Tadros
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children, London, UK
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Cristine Sortica da Costa
- Infection, Immunity and Inflammation Department, University College London Great Ormond Street Institute of Child Health, London, UK
- Neonatal Intensive Care Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Melissa Hill
- North Thames Regional Genetics Service, Great Ormond Street Hospital For Children NHS Foundation Trust, London, UK
- Genetic and Genomic Medicine, University College London Great Ormond Street Institute of Child Health Library, London, UK
| |
Collapse
|
32
|
Unnikumaran Y, Lietsch M, Brower A. Charting the Ethical Frontier in Newborn Screening Research: Insights from the NBSTRN ELSI Researcher Needs Survey. Int J Neonatal Screen 2024; 10:64. [PMID: 39311366 PMCID: PMC11417897 DOI: 10.3390/ijns10030064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/26/2024] Open
Abstract
From 2008 to 2024, the Newborn Screening Translational Research Network (NBSTRN), part of the National Institute of Child Health and Human Development (NICHD) Hunter Kelly Newborn Screening Program, served as a robust infrastructure to facilitate groundbreaking research in newborn screening (NBS), public health, rare disease, and genomics. Over its sixteen years, NBSTRN developed into a significant international network, supporting innovative research on novel technologies to screen, diagnose, treat, manage, and understand the natural history of more than 280 rare diseases. The NBSTRN tools and resources were used by a variety of stakeholders including researchers, clinicians, state NBS programs, parents, families, and policy makers. Resources and expertise for the newborn screening community in ethical, legal, and social issues (ELSI) has been an important area of focus for the NBSTRN and this includes efforts across the NBS system from pilot studies of candidate conditions to public health implementation of screening for new conditions, and the longitudinal follow-up of NBS-identified individuals to inform health outcomes and disease understanding. In 2023, the NBSTRN conducted a survey to explore ELSI issues in NBS research, specifically those encountered by the NBS community. Since NBS research involves collaboration among researchers, state NBS programs, clinicians, and families, the survey was broadly designed and disseminated to engage all key stakeholders. With responses from 88 members of the NBS community, including researchers and state NBS programs, the survey found that individuals rely most on institutional and collegial resources when they encounter ELSI questions. Most survey responses ranked privacy as extremely or very important in NBS research and identified the need for policies that address informed consent in NBS research. The survey results highlight the need for improved collaborative resources and educational programs focused on ELSI for the NBS community. The survey results inform future efforts in ELSI and NBS research in the United States (U.S.) and the rest of the world, including the development of policies and expanded ELSI initiatives and tools that address the needs of all NBS stakeholders.
Collapse
Affiliation(s)
- Yekaterina Unnikumaran
- American College of Medical Genetics and Genomics (ACMG), Bethesda, MD 20814, USA; (Y.U.); (M.L.)
| | - Mei Lietsch
- American College of Medical Genetics and Genomics (ACMG), Bethesda, MD 20814, USA; (Y.U.); (M.L.)
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Brower
- American College of Medical Genetics and Genomics (ACMG), Bethesda, MD 20814, USA; (Y.U.); (M.L.)
- Genetic Medicine, Munore-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Department of Health Professions., Creighton University School of Medicine, Omaha, NE 68178, USA
| |
Collapse
|
33
|
Taylor-Miller T, Tucker K, Sugo E, Anazodo A, Mowat D. Clues for Early Diagnosis of MEN2B Syndrome Before Medullary Thyroid Carcinoma. Pediatrics 2024; 154:e2022059517. [PMID: 39148481 DOI: 10.1542/peds.2022-059517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 08/17/2024] Open
Abstract
Early onset medullary thyroid carcinoma, later pheochromocytomas, and nonspecific extra-endocrine features (hypermobility and persistent constipation) are part of the clinical phenotype of Multiple Endocrine Neoplasia type 2B (MEN2B). A de novo pathogenic M918T variant in the rearranged during transfection proto-oncogene is usually identified. Affected children are often seen by multiple clinicians over a long period before consideration of a diagnosis of MEN2B, with metastatic medullary thyroid carcinoma often the precipitator. We describe the clinical presentation and course of 5 children ultimately diagnosed with MEN2B in New South Wales and the Australian Capital Territory, Australia between 1989 and 2021. All cases had intestinal ganglioneuromatosis that could have prompted an earlier diagnosis. Population wide newborn genomic screening for rare diseases is on the horizon. We propose that MEN2B genomic screening should be included in newborn screening programs and that careful exclusion of intestinal ganglioneuromatosis would allow earlier identification leading to improved clinical outcomes.
Collapse
Affiliation(s)
- Tashunka Taylor-Miller
- Centre for Clinical Genetics, Sydney Children's Hospital Randwick, New Sout Wales, Australia
| | - Katherine Tucker
- Hereditary Cancer Centre, Prince of Wales Hospital Randwick, New Sout Wales, Australia
- Discipline of Paediatrics, School of Clinical Medicine, University of New South Wales, Medicine and Health, Sydney, Australia
| | - Ella Sugo
- Department of Anatomical Histopathology, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Antoinette Anazodo
- Discipline of Paediatrics, School of Clinical Medicine, University of New South Wales, Medicine and Health, Sydney, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - David Mowat
- Centre for Clinical Genetics, Sydney Children's Hospital Randwick, New Sout Wales, Australia
- Discipline of Paediatrics, School of Clinical Medicine, University of New South Wales, Medicine and Health, Sydney, Australia
| |
Collapse
|
34
|
Bhatia S, Pal S, Kulshrestha S, Gupta D, Soni A, Saxena R, Bijarnia-Mahay S, Verma IC, Puri RD. Role of next generation sequencing in diagnosis and management of critically ill children with suspected monogenic disorder. Eur J Hum Genet 2024; 32:1106-1115. [PMID: 38605122 PMCID: PMC11369102 DOI: 10.1038/s41431-024-01569-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/19/2024] [Accepted: 02/12/2024] [Indexed: 04/13/2024] Open
Abstract
Next generation sequencing based diagnosis has emerged as a promising tool for evaluating critically ill neonates and children. However, there is limited data on its utility in developing countries. We assessed its diagnostic rate and clinical impact on management of pediatric patients with a suspected genetic disorder requiring critical care. The study was conducted at a single tertiary hospital in Northern India. We analyzed 70 children with an illness requiring intensive care and obtained a precise molecular diagnosis in 32 of 70 probands (45.3%) using diverse sequencing techniques such as clinical exome, whole exome, and whole genome. A significant change in clinical outcome was observed in 13 of 32 (40.6%) diagnosed probands with a change in medication in 11 subjects and redirection to palliative care in two subjects. Additional benefits included specific dietary management (three cases), avoidance of a major procedure (one case) and better reproductive counseling. Dramatic therapeutic responses were observed in three cases with SCN1A, SCN2A and KCNQ2-related epileptic encephalopathy. A delayed turn-around for sequencing results was perceived as a major limiting factor in the study, as rapid and ultra-rapid sequencing was not available. Achieving a precise molecular diagnosis has great utility in managing critically ill patients with suspected genetic disorders in developing countries.
Collapse
Affiliation(s)
- Sameer Bhatia
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Swasti Pal
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Samarth Kulshrestha
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Dhiren Gupta
- Department of Paediatrics, Institute of Child Health, Sir Ganga Ram Hospital, New Delhi, India
| | - Arun Soni
- Department of Neonatology, Institute of Child Health, Sir Ganga Ram Hospital, New Delhi, India
| | - Renu Saxena
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Sunita Bijarnia-Mahay
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Ishwar Chander Verma
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Ratna Dua Puri
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India.
| |
Collapse
|
35
|
Betzler IR, Hempel M, Mütze U, Kölker S, Winkler E, Dikow N, Garbade SF, Schaaf CP, Brennenstuhl H. Comparative analysis of gene and disease selection in genomic newborn screening studies. J Inherit Metab Dis 2024; 47:945-970. [PMID: 38757337 DOI: 10.1002/jimd.12750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
Genomic newborn screening (gNBS) is on the horizon given the decreasing costs of sequencing and the advanced understanding of the impact of genetic variants on health and diseases. Key to ongoing gNBS pilot studies is the selection of target diseases and associated genes to be included. In this study, we present a comprehensive analysis of seven published gene-disease lists from gNBS studies, evaluating gene-disease count, composition, group proportions, and ClinGen curations of individual disorders. Despite shared selection criteria, we observe substantial variation in total gene count (median 480, range 237-889) and disease group composition. An intersection was identified for 53 genes, primarily inherited metabolic diseases (83%, 44/53). Each study investigated a subset of exclusive gene-disease pairs, and the total number of exclusive gene-disease pairs was positively correlated with the total number of genes included per study. While most pairs receive "Definitive" or "Strong" ClinGen classifications, some are labeled as "Refuted" (n = 5) or "Disputed" (n = 28), particularly in genetic cardiac diseases. Importantly, 17%-48% of genes lack ClinGen curation. This study underscores the current absence of consensus recommendations for selection criteria for target diseases for gNBS resulting in diversity in proposed gene-disease pairs, their coupling with gene variations and the use of ClinGen curation. Our findings provide crucial insights into the selection of target diseases and accompanying gene variations for future gNBS program, emphasizing the necessity for ongoing collaboration and discussion about criteria harmonization for panel selection to ensure the screening's objectivity, integrity, and broad acceptance.
Collapse
Affiliation(s)
- Isabel R Betzler
- Institute of Human Genetics, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Maja Hempel
- Institute of Human Genetics, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Ulrike Mütze
- Centre for Child and Adolescent Medicine, Division of Child Neurology and Metabolic Medicine, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Kölker
- Centre for Child and Adolescent Medicine, Division of Child Neurology and Metabolic Medicine, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Eva Winkler
- Section of Translational Medical Ethics, National Center for Tumour Diseases, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Nicola Dikow
- Institute of Human Genetics, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Sven F Garbade
- Centre for Child and Adolescent Medicine, Division of Child Neurology and Metabolic Medicine, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Christian P Schaaf
- Institute of Human Genetics, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| | - Heiko Brennenstuhl
- Institute of Human Genetics, Heidelberg University and University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
36
|
del Rosario MC, Swenson KB, Coury S, Schwab J, Green RC, Gold NB. Genetic counselors' perspectives on genomic screening of apparently healthy newborns in the United States. GENETICS IN MEDICINE OPEN 2024; 2:101885. [PMID: 39669612 PMCID: PMC11613815 DOI: 10.1016/j.gimo.2024.101885] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 12/14/2024]
Abstract
Purpose There is growing international interest in using genomic sequencing to screen newborns and children for treatable genomic conditions. Although recent research has demonstrated increasing support for using genomic sequencing to screen newborns and children for treatable genomic conditions among various stakeholders, little is known about the perspectives of genetic counselors (GCs) in the United States, who are frequently engaged in the disclosure of positive newborn screening results and coordination of follow-up testing and management. Methods This study utilized a cross-sectional 3-section survey to explore GCs' perspectives on the benefits, limitations, and ethical and practical considerations of genomic sequencing in newborns as an adjunct screen to standard newborn screening (NBS). Additionally, we evaluated GCs' views on specific genes that could be added to NBS via sequencing. Results Of 176 GCs who participated in the study, most endorsed the addition of NBSeq for conditions that typically manifest in childhood and have a well-defined treatment or management protocol. Some perspectives, such as attitudes toward health inequity, varied by practice region. Most respondents endorsed 13 of 25 specific genetic conditions for inclusion in NBSeq. Conclusion Our findings demonstrate GCs' support for the expansion of NBS using genomic sequencing in the United States and the need for ongoing investigation of ethical and practical concerns related to its implementation.
Collapse
Affiliation(s)
- Maya C. del Rosario
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA
- Master’s Program in Genetic Counseling, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Kathleen B. Swenson
- Department of Medical Sciences & Education, Graduate Medical Sciences, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Stephanie Coury
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Ariadne Labs, Boston, MA
| | | | - Robert C. Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Ariadne Labs, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Broad Institute, Boston, MA
| | - Nina B. Gold
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
37
|
Wai HA, Svobodova E, Herrera NR, Douglas AGL, Holloway JW, Baralle FE, Baralle M, Baralle D. Tailored antisense oligonucleotides designed to correct aberrant splicing reveal actionable groups of mutations for rare genetic disorders. Exp Mol Med 2024; 56:1816-1825. [PMID: 39085356 PMCID: PMC11371919 DOI: 10.1038/s12276-024-01292-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 08/02/2024] Open
Abstract
Effective translation of rare disease diagnosis knowledge into therapeutic applications is achievable within a reasonable timeframe; where mutations are amenable to current antisense oligonucleotide technology. In our study, we identified five distinct types of abnormal splice-causing mutations in patients with rare genetic disorders and developed a tailored antisense oligonucleotide for each mutation type using phosphorodiamidate morpholino oligomers with or without octa-guanidine dendrimers and 2'-O-methoxyethyl phosphorothioate. We observed variations in treatment effects and efficiencies, influenced by both the chosen chemistry and the specific nature of the aberrant splicing patterns targeted for correction. Our study demonstrated the successful correction of all five different types of aberrant splicing. Our findings reveal that effective correction of aberrant splicing can depend on altering the chemical composition of oligonucleotides and suggest a fast, efficient, and feasible approach for developing personalized therapeutic interventions for genetic disorders within short time frames.
Collapse
Affiliation(s)
- Htoo A Wai
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Eliska Svobodova
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Natalia Romero Herrera
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andrew G L Douglas
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Francisco E Baralle
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- Fondazione Fegato, Area Science Park Basovizza, 34149, Trieste, Italy
| | - Marco Baralle
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149, Trieste, Italy
| | - Diana Baralle
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
38
|
Rodriguez KM, Vaught J, Salz L, Foley J, Boulil Z, Van Dongen-Trimmer HM, Whalen D, Oluchukwu O, Liu KC, Burton J, Syngal P, Vargas-Shiraishi O, Kingsmore SF, Kobayashi ES, Coufal NG. Rapid Whole-Genome Sequencing and Clinical Management in the PICU: A Multicenter Cohort, 2016-2023. Pediatr Crit Care Med 2024; 25:699-709. [PMID: 38668387 PMCID: PMC11300160 DOI: 10.1097/pcc.0000000000003522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
OBJECTIVES Analysis of the clinical utility of rapid whole-genome sequencing (rWGS) outside of the neonatal period is lacking. We describe the use of rWGS in PICU and cardiovascular ICU (CICU) patients across four institutions. DESIGN Ambidirectional multisite cohort study. SETTING Four tertiary children's hospitals. PATIENTS Children 0-18 years old in the PICU or CICU who underwent rWGS analysis, from May 2016 to June 2023. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS A total of 133 patients underwent clinical, phenotype-driven rWGS analysis, 36 prospectively. A molecular diagnosis was identified in 79 patients (59%). Median (interquartile range [IQR]) age was 6 months (IQR 1.2 mo-4.6 yr). Median time for return of preliminary results was 3 days (IQR 2-4). In 79 patients with a molecular diagnosis, there was a change in ICU management in 19 patients (24%); and some change in clinical management in 63 patients (80%). Nondiagnosis changed management in 5 of 54 patients (9%). The clinical specialty ordering rWGS did not affect diagnostic rate. Factors associated with greater odds ratio (OR [95% CI]; OR [95% CI]) of diagnosis included dysmorphic features (OR 10.9 [95% CI, 1.8-105]) and congenital heart disease (OR 4.2 [95% CI, 1.3-16.8]). Variables associated with greater odds of changes in management included obtaining a genetic diagnosis (OR 16.6 [95% CI, 5.5-62]) and a shorter time to genetic result (OR 0.8 [95% CI, 0.76-0.9]). Surveys of pediatric intensivists indicated that rWGS-enhanced clinical prognostication ( p < 0.0001) and contributed to a decision to consult palliative care ( p < 0.02). CONCLUSIONS In this 2016-2023 multiple-PICU/CICU cohort, we have shown that timely genetic diagnosis is feasible across institutions. Application of rWGS had a 59% (95% CI, 51-67%) rate of diagnostic yield and was associated with changes in critical care management and long-term patient management.
Collapse
Affiliation(s)
- Katherine M. Rodriguez
- Rady Children’s Hospital, San Diego, CA
- Department of Pediatrics, University of California, San Diego
- Rady Children’s Institute for Genomic Medicine, San Diego, CA
| | - Jordan Vaught
- Rady Children’s Hospital, San Diego, CA
- Department of Pediatrics, University of California, San Diego
| | - Lisa Salz
- Rady Children’s Institute for Genomic Medicine, San Diego, CA
| | | | | | | | | | - Okonkwo Oluchukwu
- OSF Children’s Hospital of Illinois, Peoria, IL
- University of Illinois College of Medicine at Peoria, Peoria, IL
| | - Kuang Chuen Liu
- University of Illinois College of Medicine at Peoria, Peoria, IL
| | - Jennifer Burton
- OSF Children’s Hospital of Illinois, Peoria, IL
- University of Illinois College of Medicine at Peoria, Peoria, IL
| | - Prachi Syngal
- OSF Children’s Hospital of Illinois, Peoria, IL
- University of Illinois College of Medicine at Peoria, Peoria, IL
| | | | | | - Erica Sanford Kobayashi
- Rady Children’s Institute for Genomic Medicine, San Diego, CA
- Cedars-Sinai Medical Center, Department of Pediatrics, Los Angeles, CA
- Children’s Hospital of Orange County, Orange, CA
| | - Nicole G. Coufal
- Rady Children’s Hospital, San Diego, CA
- Department of Pediatrics, University of California, San Diego
- Rady Children’s Institute for Genomic Medicine, San Diego, CA
| |
Collapse
|
39
|
Ashenden AJ, Chowdhury A, Anastasi LT, Lam K, Rozek T, Ranieri E, Siu CWK, King J, Mas E, Kassahn KS. The Multi-Omic Approach to Newborn Screening: Opportunities and Challenges. Int J Neonatal Screen 2024; 10:42. [PMID: 39051398 PMCID: PMC11270328 DOI: 10.3390/ijns10030042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
Newborn screening programs have seen significant evolution since their initial implementation more than 60 years ago, with the primary goal of detecting treatable conditions within the earliest possible timeframe to ensure the optimal treatment and outcomes for the newborn. New technologies have driven the expansion of screening programs to cover additional conditions. In the current era, the breadth of screened conditions could be further expanded by integrating omic technologies such as untargeted metabolomics and genomics. Genomic screening could offer opportunities for lifelong care beyond the newborn period. For genomic newborn screening to be effective and ready for routine adoption, it must overcome barriers such as implementation cost, public acceptability, and scalability. Metabolomics approaches, on the other hand, can offer insight into disease phenotypes and could be used to identify known and novel biomarkers of disease. Given recent advances in metabolomic technologies, alongside advances in genomics including whole-genome sequencing, the combination of complementary multi-omic approaches may provide an exciting opportunity to leverage the best of both approaches and overcome their respective limitations. These techniques are described, along with the current outlook on multi-omic-based NBS research.
Collapse
Affiliation(s)
- Alex J. Ashenden
- Department of Biochemical Genetics, SA Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia (T.R.)
| | - Ayesha Chowdhury
- Department of Molecular Pathology, SA Pathology, Adelaide, SA 5000, Australia; (A.C.); (L.T.A.)
| | - Lucy T. Anastasi
- Department of Molecular Pathology, SA Pathology, Adelaide, SA 5000, Australia; (A.C.); (L.T.A.)
| | - Khoa Lam
- Department of Biochemical Genetics, SA Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia (T.R.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Tomas Rozek
- Department of Biochemical Genetics, SA Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia (T.R.)
| | - Enzo Ranieri
- Department of Biochemical Genetics, SA Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia (T.R.)
| | - Carol Wai-Kwan Siu
- Department of Biochemical Genetics, SA Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia (T.R.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Jovanka King
- Immunology Directorate, SA Pathology, Adelaide, SA 5000, Australia
- Department of Allergy and Clinical Immunology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia
- Discipline of Paediatrics, Women’s and Children’s Hospital, The University of Adelaide, Adelaide, SA 5006, Australia
| | - Emilie Mas
- Department of Biochemical Genetics, SA Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia (T.R.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Karin S. Kassahn
- Department of Molecular Pathology, SA Pathology, Adelaide, SA 5000, Australia; (A.C.); (L.T.A.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
40
|
Linga BG, Mohammed SGAA, Farrell T, Rifai HA, Al-Dewik N, Qoronfleh MW. Genomic Newborn Screening for Pediatric Cancer Predisposition Syndromes: A Holistic Approach. Cancers (Basel) 2024; 16:2017. [PMID: 38893137 PMCID: PMC11171256 DOI: 10.3390/cancers16112017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
As next-generation sequencing (NGS) has become more widely used, germline and rare genetic variations responsible for inherited illnesses, including cancer predisposition syndromes (CPSs) that account for up to 10% of childhood malignancies, have been found. The CPSs are a group of germline genetic disorders that have been identified as risk factors for pediatric cancer development. Excluding a few "classic" CPSs, there is no agreement regarding when and how to conduct germline genetic diagnostic studies in children with cancer due to the constant evolution of knowledge in NGS technologies. Various clinical screening tools have been suggested to aid in the identification of individuals who are at greater risk, using diverse strategies and with varied outcomes. We present here an overview of the primary clinical and molecular characteristics of various CPSs and summarize the existing clinical genomics data on the prevalence of CPSs in pediatric cancer patients. Additionally, we discuss several ethical issues, challenges, limitations, cost-effectiveness, and integration of genomic newborn screening for CPSs into a healthcare system. Furthermore, we assess the effectiveness of commonly utilized decision-support tools in identifying patients who may benefit from genetic counseling and/or direct genetic testing. This investigation highlights a tailored and systematic approach utilizing medical newborn screening tools such as the genome sequencing of high-risk newborns for CPSs, which could be a practical and cost-effective strategy in pediatric cancer care.
Collapse
Affiliation(s)
- BalaSubramani Gattu Linga
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
- Translational and Precision Medicine Research, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
| | | | - Thomas Farrell
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
| | - Hilal Al Rifai
- Neonatal Intensive Care Unit (NICU), Newborn Screening Unit, Department of Pediatrics and Neonatology, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
| | - Nader Al-Dewik
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
- Translational and Precision Medicine Research, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
- Neonatal Intensive Care Unit (NICU), Newborn Screening Unit, Department of Pediatrics and Neonatology, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
- Genomics and Precision Medicine (GPM), College of Health & Life Science (CHLS), Hamad Bin Khalifa University (HBKU), Doha 0974, Qatar
- Faculty of Health and Social Care Sciences, Kingston University and St George’s University of London, Kingston upon Thames, Surrey, London KT1 2EE, UK
| | - M. Walid Qoronfleh
- Healthcare Research & Policy Division, Q3 Research Institute (QRI), Ann Arbor, MI 48197, USA
| |
Collapse
|
41
|
Cornel MC, van der Meij KRM, van El CG, Rigter T, Henneman L. Genetic Screening-Emerging Issues. Genes (Basel) 2024; 15:581. [PMID: 38790210 PMCID: PMC11121342 DOI: 10.3390/genes15050581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
In many countries, some form of genetic screening is offered to all or part of the population, either in the form of well-organized screening programs or in a less formalized way. Screening can be offered at different phases of life, such as preconception, prenatal, neonatal and later in life. Screening should only be offered if the advantages outweigh the disadvantages. Technical innovations in testing and treatment are driving changes in the field of prenatal and neonatal screening, where many jurisdictions have organized population-based screening programs. As a result, a greater number and wider range of conditions are being added to the programs, which can benefit couples' reproductive autonomy (preconception and prenatal screening) and improve early diagnosis to prevent irreversible health damage in children (neonatal screening) and in adults (cancer and cascade screening). While many developments in screening are technology-driven, citizens may also express a demand for innovation in screening, as was the case with non-invasive prenatal testing. Relatively new emerging issues for genetic screening, especially if testing is performed using DNA sequencing, relate to organization, data storage and interpretation, benefit-harm ratio and distributive justice, information provision and follow-up, all connected to acceptability in current healthcare systems.
Collapse
Affiliation(s)
- Martina C. Cornel
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Karuna R. M. van der Meij
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Carla G. van El
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Tessel Rigter
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
| | - Lidewij Henneman
- Section Community Genetics, Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1007 MB Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1100 DD Amsterdam, The Netherlands
| |
Collapse
|
42
|
Lunke S, Bouffler SE, Downie L, Caruana J, Amor DJ, Archibald A, Bombard Y, Christodoulou J, Clausen M, De Fazio P, Greaves RF, Hollizeck S, Kanga-Parabia A, Lang N, Lynch F, Peters R, Sadedin S, Tutty E, Eggers S, Lee C, Wall M, Yeung A, Gaff C, Gyngell C, Vears DF, Best S, Goranitis I, Stark Z. Prospective cohort study of genomic newborn screening: BabyScreen+ pilot study protocol. BMJ Open 2024; 14:e081426. [PMID: 38569677 PMCID: PMC11146401 DOI: 10.1136/bmjopen-2023-081426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
INTRODUCTION Newborn bloodspot screening (NBS) is a highly successful public health programme that uses biochemical and other assays to screen for severe but treatable childhood-onset conditions. Introducing genomic sequencing into NBS programmes increases the range of detectable conditions but raises practical and ethical issues. Evidence from prospectively ascertained cohorts is required to guide policy and future implementation. This study aims to develop, implement and evaluate a genomic NBS (gNBS) pilot programme. METHODS AND ANALYSIS The BabyScreen+ study will pilot gNBS in three phases. In the preimplementation phase, study materials, including education resources, decision support and data collection tools, will be designed. Focus groups and key informant interviews will also be undertaken to inform delivery of the study and future gNBS programmes. During the implementation phase, we will prospectively recruit birth parents in Victoria, Australia, to screen 1000 newborns for over 600 severe, treatable, childhood-onset conditions. Clinically accredited whole genome sequencing will be performed following standard NBS using the same sample. High chance results will be returned by genetic healthcare professionals, with follow-on genetic and other confirmatory testing and referral to specialist services as required. The postimplementation phase will evaluate the feasibility of gNBS as the primary aim, and assess ethical, implementation, psychosocial and health economic factors to inform future service delivery. ETHICS AND DISSEMINATION This project received ethics approval from the Royal Children's Hospital Melbourne Research Ethics Committee: HREC/91500/RCHM-2023, HREC/90929/RCHM-2022 and HREC/91392/RCHM-2022. Findings will be disseminated to policy-makers, and through peer-reviewed journals and conferences.
Collapse
Affiliation(s)
- Sebastian Lunke
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| | - Sophie E Bouffler
- Australian Genomics Health Alliance, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Lilian Downie
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| | - Jade Caruana
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - David J Amor
- University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Alison Archibald
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| | - Yvonne Bombard
- Genomics Health Services Research Program, St Michael's Hospital, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - John Christodoulou
- University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Marc Clausen
- Genomics Health Services Research Program, St Michael's Hospital, Toronto, Ontario, Canada
| | - Paul De Fazio
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Ronda F Greaves
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| | - Sebastian Hollizeck
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Anaita Kanga-Parabia
- University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Nitzan Lang
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Fiona Lynch
- University of Melbourne, Melbourne, Victoria, Australia
| | | | - Simon Sadedin
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Erin Tutty
- University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Stefanie Eggers
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Crystle Lee
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Meaghan Wall
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| | - Alison Yeung
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
| | - Clara Gaff
- University of Melbourne, Melbourne, Victoria, Australia
- Melbourne Genomics Health Alliance, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Christopher Gyngell
- University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Danya F Vears
- University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Stephanie Best
- Australian Genomics Health Alliance, Parkville, Victoria, Australia
- Department of Health Services Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ilias Goranitis
- University of Melbourne, Melbourne, Victoria, Australia
- Australian Genomics Health Alliance, Parkville, Victoria, Australia
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
- Australian Genomics Health Alliance, Parkville, Victoria, Australia
| |
Collapse
|
43
|
Xi BX, Liu SY, Xu YT, Zhang DD, Hu Q, Liu AG. Genetic Analysis of Two Novel GPI Variants Disrupting H Bonds and Localization Characteristics of 55 Gene Variants Associated with Glucose-6-phosphate Isomerase Deficiency. Curr Med Sci 2024; 44:426-434. [PMID: 38561594 DOI: 10.1007/s11596-024-2857-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/04/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Glucose-6-phosphate isomerase (GPI) deficiency is a rare hereditary nonspherocytic hemolytic anemia caused by GPI gene variants. This disorder exhibits wide heterogeneity in its clinical manifestations and molecular characteristics, often posing challenges for precise diagnoses using conventional methods. To this end, this study aimed to identify the novel variants responsible for GPI deficiency in a Chinese family. METHODS The clinical manifestations of the patient were summarized and analyzed for GPI deficiency phenotype diagnosis. Novel compound heterozygous variants of the GPI gene, c.174C>A (p.Asn58Lys) and c.1538G>T (p.Trp513Leu), were identified using whole-exome and Sanger sequencing. The AlphaFold program and Chimera software were used to analyze the effects of compound heterozygous variants on GPI structure. RESULTS By characterizing 53 GPI missense/nonsense variants from previous literature and two novel missense variants identified in this study, we found that most variants were located in exons 3, 4, 12, and 18, with a few localized in exons 8, 9, and 14. This study identified novel compound heterozygous variants associated with GPI deficiency. These pathogenic variants disrupt hydrogen bonds formed by highly conserved GPI amino acids. CONCLUSION Early family-based sequencing analyses, especially for patients with congenital anemia, can help increase diagnostic accuracy for GPI deficiency, improve child healthcare, and enable genetic counseling.
Collapse
Affiliation(s)
- Bi-Xin Xi
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Si-Ying Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Ting Xu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - De-Dong Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qun Hu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ai-Guo Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
44
|
Chan TCH, Mak CM, Yeung MCW, Law ECY, Cheung J, Wong TK, Cheng VWS, Lee JKH, Wong JCL, Fung CW, Belaramani KM, Kwok AMK, Tsang KY. Harnessing Next-Generation Sequencing as a Timely and Accurate Second-Tier Screening Test for Newborn Screening of Inborn Errors of Metabolism. Int J Neonatal Screen 2024; 10:19. [PMID: 38535123 PMCID: PMC10971432 DOI: 10.3390/ijns10010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 11/12/2024] Open
Abstract
In this study, we evaluated the implementation of a second-tier genetic screening test using an amplicon-based next-generation sequencing (NGS) panel in our laboratory during the period of 1 September 2021 to 31 August 2022 for the newborn screening (NBS) of six conditions for inborn errors of metabolism: citrullinemia type II (MIM #605814), systemic primary carnitine deficiency (MIM #212140), glutaric acidemia type I (MIM #231670), beta-ketothiolase deficiency (#203750), holocarboxylase synthetase deficiency (MIM #253270) and 3-hydroxy-3-methylglutaryl-CoA lyase deficiency (MIM # 246450). The custom-designed NGS panel can detect sequence variants in the relevant genes and also specifically screen for the presence of the hotspot variant IVS16ins3kb of SLC25A13 by the copy number variant calling algorithm. Genetic second-tier tests were performed for 1.8% of a total of 22,883 NBS samples. The false positive rate for these six conditions after the NGS second-tier test was only 0.017%, and two cases of citrullinemia type II would have been missed as false negatives if only biochemical first-tier testing was performed. The confirmed true positive cases were citrullinemia type II (n = 2) and systemic primary carnitine deficiency (n = 1). The false positives were later confirmed to be carrier of citrullinemia type II (n = 2), carrier of glutaric acidemia type I (n = 1) and carrier of systemic primary carnitine deficiency (n = 1). There were no false negatives reported. The incorporation of a second-tier genetic screening test by NGS greatly enhanced our program's performance with 5-working days turn-around time maintained as before. In addition, early genetic information is available at the time of recall to facilitate better clinical management and genetic counseling.
Collapse
Affiliation(s)
- Toby Chun Hei Chan
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Chloe Miu Mak
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Matthew Chun Wing Yeung
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Eric Chun-Yiu Law
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Jana Cheung
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Tsz Ki Wong
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Vincent Wing-Sang Cheng
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Jacky Kwan Ho Lee
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Jimmy Chi Lap Wong
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| | - Cheuk Wing Fung
- Metabolic Medicine Unit, Department of Pediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Hong Kong SAR, China; (C.W.F.); (K.M.B.); (A.M.K.K.)
| | - Kiran Moti Belaramani
- Metabolic Medicine Unit, Department of Pediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Hong Kong SAR, China; (C.W.F.); (K.M.B.); (A.M.K.K.)
| | - Anne Mei Kwun Kwok
- Metabolic Medicine Unit, Department of Pediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Hong Kong SAR, China; (C.W.F.); (K.M.B.); (A.M.K.K.)
| | - Kwok Yeung Tsang
- Newborn Screening Laboratory, Department of Pathology, Hong Kong Children’s Hospital, Hong Kong SAR, China; (T.C.H.C.); (M.C.W.Y.); (T.K.W.); (J.K.H.L.); (J.C.L.W.); (K.Y.T.)
| |
Collapse
|
45
|
Horton R, Wright CF, Firth HV, Turnbull C, Lachmann R, Houlston RS, Lucassen A. Challenges of using whole genome sequencing in population newborn screening. BMJ 2024; 384:e077060. [PMID: 38443063 DOI: 10.1136/bmj-2023-077060] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Affiliation(s)
- Rachel Horton
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Caroline F Wright
- Department of Clinical and Biomedical Sciences (Medical School), University of Exeter, Exeter, UK
| | | | | | - Robin Lachmann
- University College London Hospitals NHS Trust National Hospital for Neurology and Neurosurgery, London, UK
| | | | | |
Collapse
|
46
|
Kingsmore SF, Nofsinger R, Ellsworth K. Rapid genomic sequencing for genetic disease diagnosis and therapy in intensive care units: a review. NPJ Genom Med 2024; 9:17. [PMID: 38413639 PMCID: PMC10899612 DOI: 10.1038/s41525-024-00404-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/15/2024] [Indexed: 02/29/2024] Open
Abstract
Single locus (Mendelian) diseases are a leading cause of childhood hospitalization, intensive care unit (ICU) admission, mortality, and healthcare cost. Rapid genome sequencing (RGS), ultra-rapid genome sequencing (URGS), and rapid exome sequencing (RES) are diagnostic tests for genetic diseases for ICU patients. In 44 studies of children in ICUs with diseases of unknown etiology, 37% received a genetic diagnosis, 26% had consequent changes in management, and net healthcare costs were reduced by $14,265 per child tested by URGS, RGS, or RES. URGS outperformed RGS and RES with faster time to diagnosis, and higher rate of diagnosis and clinical utility. Diagnostic and clinical outcomes will improve as methods evolve, costs decrease, and testing is implemented within precision medicine delivery systems attuned to ICU needs. URGS, RGS, and RES are currently performed in <5% of the ~200,000 children likely to benefit annually due to lack of payor coverage, inadequate reimbursement, hospital policies, hospitalist unfamiliarity, under-recognition of possible genetic diseases, and current formatting as tests rather than as a rapid precision medicine delivery system. The gap between actual and optimal outcomes in children in ICUs is currently increasing since expanded use of URGS, RGS, and RES lags growth in those likely to benefit through new therapies. There is sufficient evidence to conclude that URGS, RGS, or RES should be considered in all children with diseases of uncertain etiology at ICU admission. Minimally, diagnostic URGS, RGS, or RES should be ordered early during admissions of critically ill infants and children with suspected genetic diseases.
Collapse
Affiliation(s)
- Stephen F Kingsmore
- Rady Children's Institute for Genomic Medicine, Rady Children's Hospital, San Diego, CA, USA.
| | - Russell Nofsinger
- Rady Children's Institute for Genomic Medicine, Rady Children's Hospital, San Diego, CA, USA
| | - Kasia Ellsworth
- Rady Children's Institute for Genomic Medicine, Rady Children's Hospital, San Diego, CA, USA
| |
Collapse
|
47
|
Wang X, Sun Y, Zhao JY, Guan XW, Wang YY, Hong DY, Zhang ZL, Li YH, Yang PY, Jiang T, Xu ZF. Utility, benefits, and risks of newborn genetic screening carrier reports for families. J Glob Health 2024; 14:04044. [PMID: 38389402 PMCID: PMC10884785 DOI: 10.7189/jogh.14.04044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Background Newborn genetic screening (NBGS) based on next-generation sequencing offers enhanced disease detection and better detection rates than traditional newborn screening. However, challenges remain, especially around reporting the NBGS carrier results. Therefore, we aimed to investigate the NBGS carrier parents' views on NBGS and NBGS reports in China. Methods We distributed a survey querying demographic information, knowledge and perceptions of NBGS, the impact of NBGS on a total of 2930 parents, and their decision-making to parents of newborns reported as carriers in NBGS in Nanjing, China in 2022. Results The average age of the survey respondents was 30.7 years (standard deviation = 3.6). Most (68.38%) felt informed about NBGS, especially women, the highly educated, and high earners. Nearly all (98.74%) saw NBGS as crucial for early disease detection, with 73.18% believing it positively impacts their future. However, 19.16% felt it might cause anxiety, especially among the less educated. Concerns included potential discrimination due to exposed genetic data and strained family ties. Many suggested NBGS coverage by medical insurance to ease financial burdens. Conclusions Through our study, we gained insights into parents' perspectives and concerns regarding the NBGS carrier result reporting, thus providing relevant information for further refinement and clinical promotion of the NBGS project.
Collapse
Affiliation(s)
- Xin Wang
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Yun Sun
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | | | - Xian-Wei Guan
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Yan-Yun Wang
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Dong-Yang Hong
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Zhi-Lei Zhang
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Ya-Hong Li
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Pei-Ying Yang
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Tao Jiang
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| | - Zheng-Feng Xu
- Genetic Medicine Center, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, China
| |
Collapse
|
48
|
Thompson L, Larson A, Salz L, Veith R, Tsai JP, Jayakar A, Chapman R, Gupta A, Kingsmore SF, Dimmock D, Bedrick A, Galindo MK, Casas K, Mohamed M, Straight L, Khan MA, Salyakina D. Multi-center implementation of rapid whole genome sequencing provides additional evidence of its utility in the pediatric inpatient setting. Front Pediatr 2024; 12:1349519. [PMID: 38440187 PMCID: PMC10909823 DOI: 10.3389/fped.2024.1349519] [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: 12/05/2023] [Accepted: 02/01/2024] [Indexed: 03/06/2024] Open
Abstract
Objective Multi-center implementation of rapid whole genome sequencing with assessment of the clinical utility of rapid whole genome sequencing (rWGS), including positive, negative and uncertain results, in admitted infants with a suspected genetic disease. Study design rWGS tests were ordered at eight hospitals between November 2017 and April 2020. Investigators completed a survey of demographic data, Human Phenotype Ontology (HPO) terms, test results and impacts of results on clinical care. Results A total of 188 patients, on general hospital floors and intensive care unit (ICU) settings, underwent rWGS testing. Racial and ethnic characteristics of the tested infants were broadly representative of births in the country at large. 35% of infants received a diagnostic result in a median of 6 days. The most common HPO terms for tested infants indicated an abnormality of the nervous system, followed by the cardiovascular system, the digestive system, the respiratory system and the head and neck. Providers indicated a major change in clinical management because of rWGS for 32% of infants tested overall and 70% of those with a diagnostic result. Also, 7% of infants with a negative rWGS result and 23% with a variant of unknown significance (VUS) had a major change in management due to testing. Conclusions Our study demonstrates that the implementation of rWGS is feasible across diverse institutions, and provides additional evidence to support the clinical utility of rWGS in a demographically representative sample of admitted infants and includes assessment of the clinical impact of uncertain rWGS results in addition to both positive and negative results.
Collapse
Affiliation(s)
- Lauren Thompson
- Division of Genetics and Metabolism, Nicklaus Children’s Hospital, Miami, FL, United States
| | - Austin Larson
- Department of Pediatrics, Children’s Hospital Colorado, Aurora, CO, United States
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lisa Salz
- RCIGM, Rady Children’s Hospital San Diego, San Diego, CA, United States
| | - Regan Veith
- Genomic Medicine, Children’s Minnesota, Minneapolis, MN, United States
| | - John-Paul Tsai
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Anuj Jayakar
- Division of Genetics and Metabolism, Nicklaus Children’s Hospital, Miami, FL, United States
| | - Rachel Chapman
- Fetal & Neonatal Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Department of Pediatrics, USC Keck School of Medicine, Los Angeles, CA, United States
| | - Apeksha Gupta
- Division of Genetics and Metabolism, Nicklaus Children’s Hospital, Miami, FL, United States
| | | | - David Dimmock
- RCIGM, Rady Children’s Hospital San Diego, San Diego, CA, United States
| | - Alan Bedrick
- Department of Pediatrics, Banner Diamond Children’s Medical Center, Tucson, AZ, United States
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Maureen Kelly Galindo
- Department of Pediatrics, Banner Diamond Children’s Medical Center, Tucson, AZ, United States
| | - Kari Casas
- Department of Pediatrics, Sanford Children’s Fargo, Fargo, SD, United States
| | - Mohamed Mohamed
- Department of Pediatrics, Sanford Children’s Fargo, Fargo, SD, United States
| | - Lisa Straight
- Department of Pediatrics, Sanford Children’s Sioux Falls, Sioux Falls, SD, United States
| | - M. Akram Khan
- Department of Pediatrics, Sanford Children’s Sioux Falls, Sioux Falls, SD, United States
| | - Daria Salyakina
- Division of Genetics and Metabolism, Nicklaus Children’s Hospital, Miami, FL, United States
| |
Collapse
|
49
|
Watanabe H, Nagano N, Tsuji Y, Noto N, Ayusawa M, Morioka I. Challenges of pediatric pharmacotherapy: A narrative review of pharmacokinetics, pharmacodynamics, and pharmacogenetics. Eur J Clin Pharmacol 2024; 80:203-221. [PMID: 38078929 DOI: 10.1007/s00228-023-03598-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/20/2023] [Indexed: 02/07/2024]
Abstract
PURPOSE Personalized pharmacotherapy, including for the pediatric population, provides optimal treatment and has emerged as a major trend owing to advanced drug therapeutics and diversified drug selection. However, it is essential to understand the growth and developmental characteristics of this population to provide appropriate drug therapy. In recent years, clinical pharmacogenetics has accumulated knowledge in pediatric pharmacotherapy, and guidelines from professional organizations, such as the Clinical Pharmacogenetics Implementation Consortium, can be consulted to determine the efficacy of specific drugs and the risk of adverse effects. However, the existence of a large knowledge gap hinders the use of these findings in clinical practice. METHODS We provide a narrative review of the knowledge gaps in pharmacokinetics (PK) and pharmacodynamics (PD) in the pediatric population, focusing on the differences from the perspective of growth and developmental characteristics. In addition, we explored PK/PD in relation to pediatric clinical pharmacogenetics. RESULTS The lack of direct and indirect biomarkers for more accurate assessment of the effects of drug administration limits the current knowledge of PD. In addition, incorporating pharmacogenetic insights as pivotal covariates is indispensable in this comprehensive synthesis for precision therapy; therefore, we have provided recommendations regarding the current status and challenges of personalized pediatric pharmacotherapy. The integration of clinical pharmacogenetics with the health care system and institution of educational programs for health care providers is necessary for its safe and effective implementation. A comprehensive understanding of the physiological and genetic complexities of the pediatric population will facilitate the development of effective and personalized pharmacotherapeutic strategies.
Collapse
Affiliation(s)
- Hirofumi Watanabe
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1 Kami-cho Ooyaguchi, Itabashi-ku, Tokyo, 173-8610, Japan.
| | - Nobuhiko Nagano
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1 Kami-cho Ooyaguchi, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Yasuhiro Tsuji
- Laboratory of Clinical Pharmacometrics, School of Pharmacy, Nihon University, Chiba, Japan
| | - Nobutaka Noto
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1 Kami-cho Ooyaguchi, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Mamoru Ayusawa
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1 Kami-cho Ooyaguchi, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Ichiro Morioka
- Department of Pediatrics and Child Health, Nihon University School of Medicine, 30-1 Kami-cho Ooyaguchi, Itabashi-ku, Tokyo, 173-8610, Japan
| |
Collapse
|
50
|
Yu B, Yang Y, Zhou L, Wang Q. Evaluating a Novel Newborn Screening Methodology: Combined Genetic and Biochemical Screenings. Arch Med Res 2024; 55:102959. [PMID: 38295467 DOI: 10.1016/j.arcmed.2024.102959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/20/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE Analysis of four newborn screening modes using newborn genomic sequencing (nGS) and traditional biochemical screening (TBS). METHODS Prospective clinical study with a total of 1,012 newborn samples from retrospective TBS. Three independent groups performed the study under strict double-blind conditions according to the screening modes: independent biochemical (IBS), independent NeoSeq (INS), sequential (SS), and combined (CS) screening. Using targeted sequencing, the NeoSeq panel included 154 pathogenic genes covering 86 diseases. RESULTS Of the 1,012 newborns, 120 were diagnosed were diagnosed with genetic diseases Among them, 52 cases were within the scope of TBS and 68 additional cases were identified through nGS. The number of cases detected per screening mode was 50, 113, 56, and 119 for IBS, INS, SS, and CS, respectively. CS was the most satisfactory screening mode, with the detection rate of 99.17%, the specificity and positive predictive value of 100%, and the negative predictive value of 99.89%. In addition, of the 68 cases identified by nGS (96 variants in 31 pathogenic genes), only four participants (5.9%) had clinical manifestations consistent with the disease. The experimental reporting cycles of CS and INS were the shortest. CONCLUSIONS CS was the most satisfactory method for newborn screening, which combined nGS with TBS to improve early diagnosis.
Collapse
Affiliation(s)
- Bin Yu
- Department of Medical Genetics, Changzhou Maternal and Child Health Hospital, Changzhou Medical Center of Nanjing Medical University, Changzhou, Jiangsu Province, China.
| | - Yuqi Yang
- Department of Medical Genetics, Changzhou Maternal and Child Health Hospital, Changzhou Medical Center of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Lingna Zhou
- Department of Medical Genetics, Changzhou Maternal and Child Health Hospital, Changzhou Medical Center of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Qiuwei Wang
- Department of Neonatology, Changzhou Children's Hospital of Nantong Medical University, Changzhou, Jiangsu Province, China.
| |
Collapse
|