1
|
Montanari G, Candela E, Baronio F, Ferrari V, Biasucci G, Lanari M, Ortolano R. Early-Onset Inherited Metabolic Diseases: When Clinical Symptoms Precede Newborn Screening-Insights from Emilia-Romagna (Italy). CHILDREN (BASEL, SWITZERLAND) 2025; 12:464. [PMID: 40310096 PMCID: PMC12025479 DOI: 10.3390/children12040464] [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: 02/27/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Expanded Newborn Screening (ENS) allows the early identification of many inherited metabolic diseases (IMDs) for which timely treatment can modify the natural history. For most IMDs, diagnosis by ENS is pre-clinical. However, clinical symptoms may emerge for certain conditions before screening results become available. METHODS We describe six cases of patients with early-onset IMDs born between 2013 and 2023, who were admitted or transferred to Sant'Orsola University Hospital in Bologna (Italy). RESULTS Over the study period, 379,013 newborns underwent ENS in the Italian region of Emilia-Romagna. Excluding cases of congenital hypothyroidism, pre-clinical diagnoses from ENS were 410. In addition, six cases of IMD presented with early-onset clinical symptomatology, an antecedent to the outcome of newborn screening (incidence over 11 years of 1.58 cases per 100,000 infants). Among these patients, three were diagnosed with Urea Cycle Disorders (UCDs)-two with Citrullinemia type I (CIT1) and one with Argininosuccinic Acidemia (ASA); two were diagnosed with Methylmalonic Acidemia (MMA); and one was found to have Medium-Chain Acyl-CoA Dehydrogenase Deficiency (MCADD). CONCLUSIONS Our 11-year experience with ENS has shown that clinical onset can occur between the second and fourth day of life, though rare. Even if dried blood spot (DBS) collection was performed 24-48 h after birth, the time required for sample transportation and processing would still delay result availability, making early intervention unlikely. Therefore, our experience supports performing ENS at 48-72 h, as currently implemented in Italy, while also highlighting the advantages and limitations of earlier screening.
Collapse
Affiliation(s)
- Giulia Montanari
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (G.M.); (V.F.)
| | - Egidio Candela
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.L.); (R.O.)
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Massarenti 11, 40126 Bologna, Italy
| | - Federico Baronio
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.L.); (R.O.)
| | - Vittorio Ferrari
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (G.M.); (V.F.)
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Marcello Lanari
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.L.); (R.O.)
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Massarenti 11, 40126 Bologna, Italy
| | - Rita Ortolano
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.L.); (R.O.)
| |
Collapse
|
2
|
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
|
3
|
Boemer F, Hovhannesyan K, Piazzon F, Minner F, Mni M, Jacquemin V, Mashhadizadeh D, Benmhammed N, Bours V, Jacquinet A, Harvengt J, Bulk S, Dideberg V, Helou L, Palmeira L, Dangouloff T, Servais L. Population-based, first-tier genomic newborn screening in the maternity ward. Nat Med 2025; 31:1339-1350. [PMID: 39875687 PMCID: PMC12003153 DOI: 10.1038/s41591-024-03465-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: 07/22/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025]
Abstract
The rapid development of therapies for severe and rare genetic conditions underlines the need to incorporate first-tier genetic testing into newborn screening (NBS) programs. A workflow was developed to screen newborns for 165 treatable pediatric disorders by deep sequencing of regions of interest in 405 genes. The prospective observational BabyDetect pilot project was launched in September 2022 in a maternity ward of a public hospital in the Liege area, Belgium. In this ongoing observational study, 4,260 families have been informed of the project, and 3,847 consented to participate. To date, 71 disease cases have been identified, 30 of which were not detected by conventional NBS. Glucose-6-phosphate dehydrogenase deficiency was the most frequent disorder detected, with 44 positive individuals. Of the remaining 27 cases, 17 were recessive disorders. We also identified one false-positive case in a newborn in whom two variants in the AGXT gene were identified, which were subsequently shown to be located on the maternal allele. Nine heterozygous variants were identified in genes associated with dominant conditions. Results from the BabyDetect project demonstrate the importance of integrating biochemical and genomic methods in NBS programs. Challenges must be addressed in variant interpretation within a presymptomatic population and in result reporting and diagnostic confirmation.
Collapse
Affiliation(s)
- François Boemer
- Biochemical Genetics Lab, Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium.
| | | | - Flavia Piazzon
- Human Genetics Laboratory, GIGA-R Institute, University of Liege, Liege, Belgium
| | - Frédéric Minner
- Human Genetics Laboratory, GIGA-R Institute, University of Liege, Liege, Belgium
| | - Myriam Mni
- Human Genetics Laboratory, GIGA-R Institute, University of Liege, Liege, Belgium
| | - Valérie Jacquemin
- Human Genetics Laboratory, GIGA-R Institute, University of Liege, Liege, Belgium
| | | | - Noor Benmhammed
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, CHU Liege, University of Liege, Liege, Belgium
| | - Vincent Bours
- Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium
| | - Adeline Jacquinet
- Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium
| | - Julie Harvengt
- Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium
| | - Saskia Bulk
- Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium
| | - Vinciane Dideberg
- Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium
| | - Laura Helou
- Bioinformatics Unit, Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium
| | - Leonor Palmeira
- Bioinformatics Unit, Department of Human Genetics, CHU Liege, University of Liege, Liege, Belgium
| | - Tamara Dangouloff
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, CHU Liege, University of Liege, Liege, Belgium
| | - Laurent Servais
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, CHU Liege, University of Liege, Liege, Belgium
- MDUK Neuromuscular Center, Department of Pediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Minten T, Bick S, Adelson S, Gehlenborg N, Amendola LM, Boemer F, Coffey AJ, Encina N, Ferlini A, Kirschner J, Russell BE, Servais L, Sund KL, Taft RJ, Tsipouras P, Zouk H, Bick D, Green RC, Gold NB. Data-driven consideration of genetic disorders for global genomic newborn screening programs. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.03.24.24304797. [PMID: 38585998 PMCID: PMC10996735 DOI: 10.1101/2024.03.24.24304797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Purpose Over 30 international studies are exploring newborn sequencing (NBSeq) to expand the range of genetic disorders included in newborn screening. Substantial variability in gene selection across programs exists, highlighting the need for a systematic approach to prioritize genes. Methods We assembled a dataset comprising 25 characteristics about each of the 4,390 genes included in 27 NBSeq programs. We used regression analysis to identify several predictors of inclusion, and developed a machine learning model to rank genes for public health consideration. Results Among 27 NBSeq programs, the number of genes analyzed ranged from 134 to 4,299, with only 74 (1.7%) genes included by over 80% of programs. The most significant associations with gene inclusion across programs were presence on the US Recommended Uniform Screening Panel (inclusion increase of 74.7%, CI: 71.0%-78.4%), robust evidence on the natural history (29.5%, CI: 24.6%-34.4%) and treatment efficacy (17.0%, CI: 12.3%- 21.7%) of the associated genetic disease. A boosted trees machine learning model using 13 predictors achieved high accuracy in predicting gene inclusion across programs (AUC = 0.915, R² = 84%). Conclusion The machine learning model developed here provides a ranked list of genes that can adapt to emerging evidence and regional needs, enabling more consistent and informed gene selection in NBSeq initiatives.
Collapse
|
5
|
Blair DR, Risch N. Reduced Penetrance is Common Among Predicted Loss-of-Function Variants and is Likely Driven by Residual Allelic Activity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.09.23.24314008. [PMID: 39399029 PMCID: PMC11469360 DOI: 10.1101/2024.09.23.24314008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Loss-of-function genetic variants (LoFs) often result in severe phenotypes, including autosomal dominant diseases driven by haploinsufficiency. Due to low carrier frequencies, their penetrance is generally unknown but typically variable. Here, we investigate the penetrance of >6,000 predicted LoFs (pLoFs) linked to 91 haploinsufficient diseases using a cohort of ≈24,000 carriers with linked electronic health record data. We find evidence for widespread reduced penetrance, which persisted after accounting for variant annotation artifacts, missed diagnoses, and incomplete clinical data. We thus hypothesized that many pLoFs have incomplete penetrance, which may be driven by residual allelic activity. To test this, we trained machine learning models to predict pLoF penetrance using variant-specific genomic features that may correlate with incomplete loss-of-function. The models were predictive of pLoF penetrance across a range of diseases and variant types, including those with prior clinical evidence for pathogenicity. This suggests that many pLoFs have incomplete penetrance due to residual allelic activity, complicating disease prognostication in asymptomatic carriers.
Collapse
|
6
|
Knoppers BM, Bonilha AE, Laberge AM, Ahmed A, Newson AJ. Genomic sequencing in newborn screening: balancing consent with the right of the asymptomatic at-risk child to be found. Eur J Hum Genet 2025; 33:182-188. [PMID: 39134767 PMCID: PMC11840138 DOI: 10.1038/s41431-024-01677-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/13/2024] [Accepted: 07/25/2024] [Indexed: 02/21/2025] Open
Abstract
In this paper, we explore key aspects of the complex ethical and legal landscape surrounding consent in the context of incorporating genomic sequencing into existing newborn bloodspot screening programs. In particular, we consider the potential impact of genomic sequencing on the health rights of the child in relation to existing consent practices in newborn screening. We begin with an introduction to newborn screening programs and their population health goals. We then discuss public health ethics as a rationale underpinning newborn screening before turning to consent. We go on to describe seven current research projects on genomic sequencing in newborn screening and then introduce the 'right of the asymptomatic at-risk child to be found' as a useful concept to draw on when considering consent to newborn screening. We draw on this novel right to argue for the adoption of "appropriate consent" when it comes to certain uses of genomics in newborn screening. We contend that, for 'virtual panels' at least, appropriate consent proportionately balances the ongoing universality of newborn screening for important health conditions with an acknowledgement of the complex outcomes that bringing a complicated diagnostic technology into the screening domain will generate.
Collapse
Affiliation(s)
- Bartha Maria Knoppers
- Centre of Genomics and Policy, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Ana Eliza Bonilha
- McGill University Health Centre, Department of Human Genetics, Montreal, QC, Canada
| | - Anne-Marie Laberge
- Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Montreal, QC, Canada
| | | | - Ainsley J Newson
- Sydney Health Ethics, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
7
|
Kamolvisit W, Cheawsamoot C, Chetruengchai W, Kor-Anantakul P, Thangpong R, Srichomthong C, Assawapitaksakul A, Syananondh K, Kontun S, Buasong A, Od-Ek P, Suphapeetiporn K, Shotelersuk V. Singleton rapid long-read genome sequencing as first tier genetic test for critically Ill children with suspected genetic diseases. Eur J Hum Genet 2025:10.1038/s41431-025-01818-9. [PMID: 40011756 DOI: 10.1038/s41431-025-01818-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025] Open
Abstract
Timely genetic testing is crucial for diagnosing pediatric patients in the intensive care units (ICUs) without known etiology. We aim to explore the benefits of singleton rapid long-read genome sequencing (rLR-GS) in critically ill children admitted to ICU with a suspected genetic etiology. Children younger than 18 years of age admitted to the ICU with a suspected genetic etiology at two tertiary hospitals in Thailand from August 2023 to May 2024 were included. rLR-GS was performed. The value of a molecular diagnosis for changing patient management outcomes was assessed. Eighteen patients were recruited. Singleton rLR-GS identified seventeen likely pathogenic (LP) or pathogenic (P) variants, resulting in the diagnosis of eleven distinct genetic disorders with autosomal recessive, autosomal dominant, and mitochondrial inheritance patterns. This yielded a diagnostic rate of 61% (11/18) with a median turnaround time of nine days. Specifically, rLR-GS identified three pathogenic structural variants (SVs), including large deletions of 19 kb, 2.4 kb, and 10.1 kb. Additionally, it provided phasing information for the two variants in each of the six patients with autosomal recessive disorders. Furthermore, the identification of two SVs and the phasing information led to the reclassification of three single nucleotide variants (SNV), one in each patient, from variants of unknown significance (VUS) to LP. The application of rLR-GS resulted in significant changes in the management of all eleven patients. This proof-of-concept study demonstrated the utility of singleton rLR-GS as a first-tier diagnostic approach for critically ill patients with unknown causes.
Collapse
Affiliation(s)
- Wuttichart Kamolvisit
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Chanatjit Cheawsamoot
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Wanna Chetruengchai
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Phawin Kor-Anantakul
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Rungroj Thangpong
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Chalurmpon Srichomthong
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Adjima Assawapitaksakul
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Kusuma Syananondh
- Respiratory Unit, Pediatric Department, Buddhachinaraj Phitsanulok Hospital, Phitsanulok, Thailand
| | - Sineenat Kontun
- Neurology Unit, Pediatric Department, Buddhachinaraj Phitsanulok Hospital, Phitsanulok, Thailand
| | - Aayalida Buasong
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Phichittra Od-Ek
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Kanya Suphapeetiporn
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Hoppe B, Martin-Higueras C, Borghese L, Kaspar S, Reusch B, Beck BB, Walli A, Janzen E, Hegert S, Janzen N, Hohenfellner K. Effective Newborn Screening for Type 1 and 3 Primary Hyperoxaluria. Kidney Int Rep 2025; 10:177-183. [PMID: 39810772 PMCID: PMC11725795 DOI: 10.1016/j.ekir.2024.10.006] [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: 09/02/2024] [Accepted: 10/07/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Newborn screening (NBS) programs for a defined set of eligible diseases have been enormously successful, but genomic NBS allowing for detection of additional treatable disorders has not been broadly implemented. All 3 types of primary hyperoxaluria (PH1-3) are rare autosomal recessive diseases caused by distinct defects of glyoxylate metabolism that are diagnosed genetically with certainty. Early diagnosis and treatment are mandatory to avoid renal failure or sequalae associated with persistent hyperoxaluria. Methods This prospective pilot study was undertaken within the framework of the German NBS. DNA samples extracted from dried blood spot cards were screened by multiplex polymerase chain reaction (PCR) for the 2 most prevalent variants: AGXT c.508G>A (PH1) and HOGA1 c.700 + 5G>T (PH3). Heterozygous AGXT/HOGA1 carriers received repeated spot urine analyses and, in case of persistent hyperoxaluria, complete Sanger sequencing of AGXT and HOGA1 genes, respectively. Results Between March 15, 2022 and June 30, 2023, additional screening for PH1 and PH3 was performed in 77,199 out of 222,638 newborns included in the regular NBS program. No homozygous individuals, but 274 potential carriers for the AGXT mistargeting and 287 potential carriers for the HOGA1 splice variant were identified. Further workup revealed 2 already symptomatic compound heterozygous infants, 1 with PH1 (genotype c.508G>A; c.33delC) and 1 with PH3 (genotype: c.700 + 5G>T; c.134C>G). A second symptomatic patient with PH1 (father of an identified carrier; genotype: c.508G>A; c.508G>A) was uncovered via family history. Conclusion This pilot study demonstrates the efficacy of a genomic neonatal screening program for PH even in relatively small cohorts.
Collapse
Affiliation(s)
| | | | | | - Sophie Kaspar
- Institute of Human Genetics, Center for Molecular Medicine Cologne, and Center for Rare and Hereditary Kidney Disease, Cologne, University Hospital of Cologne, Cologne, Germany
| | - Björn Reusch
- Institute of Human Genetics, Center for Molecular Medicine Cologne, and Center for Rare and Hereditary Kidney Disease, Cologne, University Hospital of Cologne, Cologne, Germany
| | - Bodo B. Beck
- Institute of Human Genetics, Center for Molecular Medicine Cologne, and Center for Rare and Hereditary Kidney Disease, Cologne, University Hospital of Cologne, Cologne, Germany
| | - Adam Walli
- Wisplinghoff Laboratory, Cologne, Germany
| | | | | | - Nils Janzen
- Screening Laboratory Hannover, Germany
- Department of Clinical Chemistry, Hannover Medical School, Hannover, Germany
- Division of Laboratory Medicine, Centre for Children and Adolescents, Hannover, Germany
| | | |
Collapse
|
12
|
Kojima D, Shibata M, Shikano H, Maruo Y, Fujii H. Identification of a novel missense variant in the AVP gene in a Japanese pedigree with familial neurohypophyseal diabetes insipidus. Clin Pediatr Endocrinol 2025; 34:77-82. [PMID: 39777129 PMCID: PMC11701011 DOI: 10.1297/cpe.2024-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
Familial neurohypophyseal diabetes insipidus is a rare genetic disease caused by AVP gene variants and is characterized by progressive polyuria and polydipsia in early childhood. Herein, we have reported the clinical symptoms and genetic test results of a Japanese patient with a family history of polyuria and polydipsia for over five generations. The proband was a 6-yr-old boy who was referred for the evaluation of polyuria and polydipsia. A hypertonic saline infusion test showed no increase in AVP levels and a water deprivation test followed by vasopressin administration confirmed the diagnosis of central diabetes insipidus. Genetic analyses of the patient and his affected mother revealed a novel heterozygous missense variant (c.308T>A, p.V103D). This variant was located in the region encoding the neurophysin II moiety. Computational analysis predicted that p.V103D is pathogenic, and a structural change was detected by viewing the three-dimensional structure of the protein model. To our knowledge, this is the first study to identify a novel missense variant, p.V103D, in a Japanese family with central diabetes insipidus. These findings expand the panel of AVP variants and facilitate the genetic diagnosis of familial neurohypophyseal diabetes insipidus.
Collapse
Affiliation(s)
- Daiei Kojima
- Department of Pediatrics, Ogaki Municipal Hospital, Ogaki, Japan
| | - Masami Shibata
- Department of Pediatrics, Ogaki Municipal Hospital, Ogaki, Japan
| | - Hiroaki Shikano
- Department of Pediatrics, Ogaki Municipal Hospital, Ogaki, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Hidehiko Fujii
- Department of Pediatrics, Ogaki Municipal Hospital, Ogaki, Japan
| |
Collapse
|
13
|
Pauly K, Woontner M, Abdenur JE, Chaudhari BP, Gosselin R, Kripps KA, Thomas JA, Wempe MF, Gospe SM, Coughlin CR. Feasibility of newborn screening for pyridoxine-dependent epilepsy. Mol Genet Metab 2025; 144:109002. [PMID: 39729892 DOI: 10.1016/j.ymgme.2024.109002] [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: 10/19/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Pyridoxine-dependent epilepsy (PDE-ALDH7A1) is a developmental epileptic encephalopathy historically characterized by seizures that are resistant to antiseizure medications. Treatment with pyridoxine and lysine reduction therapies are associated with seizure control and improved developmental outcomes. In rare circumstances, patients have died prior to diagnosis and treatment with pyridoxine, and many patients are diagnosed after six months of age when lysine reduction therapies have limited efficacy. Recently two new metabolites were identified (2S,6S-/2S,6R-oxopropylpiperidine-2-carboxylic acid, 2-OPP and 6-oxo-pipecolate, 6-oxo-pip), and we evaluated these metabolites as potential newborn screening biomarkers. METHODS We recruited participants with a confirmed diagnosis of PDE-ALDH7A1 and retrieved their residual dried blood spots from state-sponsored newborn screening programs. We evaluated the dried blood spots for 2-OPP using commercially available newborn screening kits and equipment, and developed a second-tier test for 6-oxo-pip using LC-MS/MS. RESULTS We received eight residual dried blood spots collected before the onset of seizures and the diagnosis of PDE-ALDH7A1. In our newborn screening experiments, 2-OPP was elevated in 7 of 8 samples from affected participants with a mean of 3.08 μmol/L (95 % CI 2.17-3.99) compared to a mean of 0.09 μmol/L (95 % CI 0.09-0.10) in controls (p < 0.001). Second tier testing demonstrated elevated 6-oxo-pip in all samples from affected participants with a mean of 5.66 μmol/L (95 % CI 1.51-9.81) and was undetectable in controls (p < 0.001). CONCLUSION Patients with PDE-ALDH7A1 can be identified using neonatal dried blood spots prior to the onset of symptoms. The use of commercially available newborn screening approaches demonstrates the feasibility of newborn screening for this treatable condition.
Collapse
Affiliation(s)
- Kristine Pauly
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael Woontner
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jose E Abdenur
- Division of Metabolic Disorders, CHOC Children's Hospital, Orange, CA, USA
| | - Bimal P Chaudhari
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, Division of Neonatology, Nationwide Children's Hospital, Columbus, OH, USA; Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Rachel Gosselin
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kimberly A Kripps
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Janet A Thomas
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael F Wempe
- School of Pharmacy, Department of Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA; Univeristy of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sidney M Gospe
- Departments of Neurology and Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Duke University, Durham, NC, USA
| | - Curtis R Coughlin
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
14
|
Schnabel‐Besson E, Garbade SF, Gleich F, Grünert SC, Krämer J, Thimm E, Hennermann JB, Freisinger P, Burgard P, Gramer G, Morath MA, Tuncel AT, Keßler S, Hoffmann GF, Kölker S, Mütze U. Parental and child's psychosocial and financial burden living with an inherited metabolic disease identified by newborn screening. J Inherit Metab Dis 2025; 48:e12784. [PMID: 39189622 PMCID: PMC11670445 DOI: 10.1002/jimd.12784] [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: 04/25/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/28/2024]
Abstract
Newborn screening (NBS) is one of the most effective measures of secondary prevention. While the benefit of NBS on the clinical long-term outcomes of children with inherited metabolic diseases (IMD) has been demonstrated, the potential burden of families living with an early diagnosed and treated child with an IMD has not been thoroughly investigated. The aim of this longitudinal questionnaire-based study on 369 families living with a child with an IMD was to investigate the psychosocial and financial burden following a true-positive NBS. The reported psychosocial burden differed between children and their parents, and was associated with the child's age, diagnosis, and treatment. At younger ages, parent-reported burden was higher for the parents than for the individual child, while it increased for children and decreased for parents as the child grew older. Furthermore, psychosocial burden increased if the child required a strict dietary treatment and was at risk of metabolic decompensation. Regardless of diagnosis and treatment, the developmental delay of their child independently increased the parental psychosocial burden. Financial burden was reported by 24% of all families, and was higher in low-income families and in families whose children required dietary treatment. In conclusion, a substantial psychosocial and financial burden was revealed for children and their families after true-positive NBS. Since this burden is likely to have a negative impact on the long-term individual health benefits of NBS, this study underlines the importance of regularly assessing the psychosocial and financial needs of these families.
Collapse
Affiliation(s)
- Elena Schnabel‐Besson
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Sven F. Garbade
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Florian Gleich
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Sarah C. Grünert
- Department of General Pediatrics, Adolescent Medicine and NeonatologyMedical Center ‐ University of Freiburg, Faculty of MedicineFreiburgGermany
| | - Johannes Krämer
- Department of Pediatric and Adolescent MedicineUlm University Medical SchoolUlmGermany
| | - Eva Thimm
- Department of General Pediatrics, Neonatology, and Pediatric CardiologyUniversity Children's Hospital, Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Julia B. Hennermann
- Villa MetabolicaCenter for Pediatric and Adolescent Medicine, Mainz University Medical CenterMainzGermany
| | - Peter Freisinger
- Children's Hospital Reutlingen, Klinikum am SteinenbergReutlingenGermany
| | - Peter Burgard
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Gwendolyn Gramer
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
- Department for Inborn Metabolic DiseasesUniversity Children's Hospital, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Marina A. Morath
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - A. Tunç Tuncel
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Svenja Keßler
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Georg F. Hoffmann
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Stefan Kölker
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| | - Ulrike Mütze
- Department of Pediatrics I, Division of Pediatric Neurology and Metabolic MedicineHeidelberg University, Medical Faculty HeidelbergHeidelbergGermany
| |
Collapse
|
15
|
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
|
16
|
Veldman A, Sikkema-Raddatz B, Derks TGJ, van Karnebeek CDM, Kiewiet MBG, Mulder MF, Nelen MR, Rubio-Gozalbo ME, Sinke RJ, de Sain-van der Velden MG, Visser G, de Vries MC, Westra D, Williams M, Wevers RA, Heiner-Fokkema MR, van Spronsen FJ. Newborn Screening by DNA-First: Systematic Evaluation of the Eligibility of Inherited Metabolic Disorders Based on Treatability. Int J Neonatal Screen 2024; 11:1. [PMID: 39846587 PMCID: PMC11755635 DOI: 10.3390/ijns11010001] [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: 10/14/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
The biomarker-based Dutch Newborn Screening (NBS) panel (as of 2024) comprises 19 inherited metabolic disorders (IMDs). With the use of next-generation sequencing (NGS) as a first-tier screen, NBS could expand to include IMDs that lack a reliable biochemical footprint in dried blood spots, while also reducing secondary findings. To be eligible for inclusion in NBS, an IMD needs to fulfill the Wilson and Jungner criteria, with treatability being one of the most important criteria. In this study, we aimed to identify IMDs eligible for DNA-first NBS when considering only treatability in the context of NBS as a prerequisite. First, three independent reviewers performed a systematic literature review of the 1459 genotypic IMDs and their causative gene(s), as described in the International Classification of Inherited Metabolic Disorders (dated 1 February 2021), applying 16 criteria to exclude non-treatable disorders. Eligible disorders were then discussed in three online meetings with a project group of clinical laboratory geneticists, medical laboratory specialists specialized in IMD, and pediatricians with expertise in IMDs. Based on treatability, we identified 100 genes, causing 95 IMDs, as eligible for NBS, including 42 causal genes for the IMDs in the current biomarker-based NBS. The other 58 genes are primarily associated with treatable defects in amino acid metabolism and fatty acid oxidation. Other IMDs were excluded, most often because of insufficient literature. As the evaluation of treatability was not straightforward, we recommend the development of standardized treatability scores for the inclusion of IMDs in NBS.
Collapse
Affiliation(s)
- Abigail Veldman
- Division of Metabolic Diseases, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9718 GZ Groningen, The Netherlands
| | - Birgit Sikkema-Raddatz
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Terry G. J. Derks
- Division of Metabolic Diseases, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9718 GZ Groningen, The Netherlands
| | - Clara D. M. van Karnebeek
- Departments of Pediatrics and Human Genetics, Emma Center for Personalized Medicine, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - M. B. Gea Kiewiet
- Division of Metabolic Diseases, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9718 GZ Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Margaretha F. Mulder
- Departments of Pediatrics and Human Genetics, Emma Center for Personalized Medicine, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Marcel R. Nelen
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - M. Estela Rubio-Gozalbo
- Division of Metabolic Diseases, Department of Pediatrics, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Richard J. Sinke
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | | | - Gepke Visser
- Departments of Pediatrics and Human Genetics, Emma Center for Personalized Medicine, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Maaike C. de Vries
- Division of Metabolic Disorders, Department of Pediatrics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Dineke Westra
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Monique Williams
- Center for Lysosomal and Metabolic Diseases, Department of Pediatrics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Ron A. Wevers
- Department of Human Genetics, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - M. Rebecca Heiner-Fokkema
- Laboratory of Metabolic Diseases, Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9718 GZ Groningen, The Netherlands
| | - Francjan J. van Spronsen
- Division of Metabolic Diseases, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9718 GZ Groningen, The Netherlands
| |
Collapse
|
17
|
Kingsmore SF, Wright M, Smith LD, Liang Y, Mowrey WR, Protopsaltis L, Bainbridge M, Baker M, Batalov S, Blincow E, Cao B, Caylor S, Chambers C, Ellsworth K, Feigenbaum A, Frise E, Guidugli L, Hall KP, Hansen C, Kiel M, Van Der Kraan L, Krilow C, Kwon H, Madhavrao L, Lefebvre S, Leipzig J, Mardach R, Moore B, Oh D, Olsen L, Ontiveros E, Owen MJ, Reimers R, Scharer G, Schleit J, Shelnutt S, Mehtalia SS, Oriol A, Sanford E, Schwartz S, Wigby K, Willis MJ, Yandell M, Kunard CM, Defay T. Prequalification of genome-based newborn screening for severe childhood genetic diseases through federated training based on purifying hyperselection. Am J Hum Genet 2024; 111:2618-2642. [PMID: 39642867 PMCID: PMC11639087 DOI: 10.1016/j.ajhg.2024.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 12/09/2024] Open
Abstract
Genome-sequence-based newborn screening (gNBS) has substantial potential to improve outcomes in hundreds of severe childhood genetic disorders (SCGDs). However, a major impediment to gNBS is imprecision due to variants classified as pathogenic (P) or likely pathogenic (LP) that are not SCGD causal. gNBS with 53,855 P/LP variants, 342 genes, 412 SCGDs, and 1,603 therapies was positive in 74% of UK Biobank (UKB470K) adults, suggesting 97% false positives. We used the phenomenon of purifying hyperselection, which acts to decrease the frequency of SCGD causal diplotypes, to reduce false positives. Training of gene-disease-inheritance mode-diplotype tetrads in 618,290 control and affected subjects identified 293 variants or haplotypes and seven genes with variable inheritance contributing higher positive diplotype counts than consistent with purifying hyperselection and with little or no evidence of SCGD causality. With these changes, 2.0% of UKB470K adults were positive. In contrast, gNBS was positive in 7.2% of 3,118 critically ill children with suspected SCGDs and 7.9% of 705 infant deaths. When compared with rapid diagnostic genome sequencing (RDGS), gNBS had 99.1% recall. In eight true-positive children, gNBS was projected to decrease time to diagnosis by a median of 121 days and avoid life-threatening disease presentations in four children, organ damage in six children, ∼$1.25 million in healthcare cost, and ten (1.4%) infant deaths. Federated training predicated on purifying hyperselection provides a general framework to attain high precision in population screening. Federated training across many biobanks and clinical trials can provide a privacy-preserving mechanism for qualification of gNBS in diverse genetic ancestries.
Collapse
Affiliation(s)
- Stephen F Kingsmore
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA.
| | - Meredith Wright
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Laurie D Smith
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Yupu Liang
- Alexion, AstraZeneca Rare Disease, Boston, MA 02210, USA
| | | | - Liana Protopsaltis
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Matthew Bainbridge
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Mei Baker
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | - Sergey Batalov
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Eric Blincow
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Bryant Cao
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Sara Caylor
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Christina Chambers
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Katarzyna Ellsworth
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Annette Feigenbaum
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA; Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Erwin Frise
- Fabric Genomics, Inc., Oakland, CA 94612, USA
| | - Lucia Guidugli
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | | | - Christian Hansen
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Mark Kiel
- Genomenon Inc., Ann Arbor, MI 48108, USA
| | - Lucita Van Der Kraan
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | | | - Hugh Kwon
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Lakshminarasimha Madhavrao
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | | | | | - Rebecca Mardach
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA; Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Barry Moore
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84132, USA
| | - Danny Oh
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Lauren Olsen
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Eric Ontiveros
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Mallory J Owen
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Rebecca Reimers
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Scripps Research Translational Institute, La Jolla, CA 92037, USA
| | - Gunter Scharer
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Jennifer Schleit
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | | | | | - Albert Oriol
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Erica Sanford
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | | | - Kristen Wigby
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Mary J Willis
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Mark Yandell
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84132, USA
| | | | - Thomas Defay
- Alexion, AstraZeneca Rare Disease, Boston, MA 02210, USA
| |
Collapse
|
18
|
Kingsmore SF, Wright M, Olsen L, Schultz B, Protopsaltis L, Averbuj D, Blincow E, Carroll J, Caylor S, Defay T, Ellsworth K, Feigenbaum A, Gover M, Guidugli L, Hansen C, Van Der Kraan L, Kunard CM, Kwon H, Madhavrao L, Leipzig J, Liang Y, Mardach R, Mowrey WR, Nguyen H, Niemi AK, Oh D, Saad M, Scharer G, Schleit J, Mehtalia SS, Sanford E, Smith LD, Willis MJ, Wigby K, Reimers R. Genome-based newborn screening for severe childhood genetic diseases has high positive predictive value and sensitivity in a NICU pilot trial. Am J Hum Genet 2024; 111:2643-2667. [PMID: 39642868 PMCID: PMC11639094 DOI: 10.1016/j.ajhg.2024.10.020] [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: 08/07/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 12/09/2024] Open
Abstract
Large prospective clinical trials are underway or planned that examine the clinical utility and cost effectiveness of genome-based newborn screening (gNBS). One gNBS platform, BeginNGS, currently screens 53,575 variants for 412 severe childhood genetic diseases with 1,603 efficacious therapies. Retrospective evaluation of BeginNGS in 618,290 subjects suggests adequate sensitivity and positive predictive value (PPV) to proceed to prospective studies. To inform pivotal clinical trial design, we undertook a pilot clinical trial. We enrolled 120 infants in a regional neonatal intensive care unit (NICU) who were not under consideration for rapid diagnostic genome sequencing (RDGS). Each enrollee received BeginNGS and two index tests (California state NBS and RDGS). California NBS identified 4 of 4 true positive (TP) findings (TP rate 3.6%, sensitivity 100%) and 11 false positive (FP) findings (PPV 27%). RDGS identified 41 diagnostic findings in 36 neonates (diagnostic rate 30%). BeginNGS identified 5 of 6 on-target TP disorders (TP rate 4.2%, 95% confidence interval 1%-8%, sensitivity 83%) and no FPs (PPV 100%). Changes in management were anticipated following the return of 27 RDGS results in 25 enrollees (clinical utility [CU] 21%), 3 of 4 NBS TPs (CU 2.7%), and all BeginNGS TPs (CU 4.2%). The incidence of actionable genetic diseases in NICU infants not being considered for RDGS suggests (1) performance of RDGS in ∼20% of admissions misses many genetic diagnoses, (2) NICU enrollment in gNBS trials will greatly increase power to test endpoints, and (3) NICUs may be attractive for early implementation of consented BeginNGS screening.
Collapse
Affiliation(s)
- Stephen F Kingsmore
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA.
| | - Meredith Wright
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Lauren Olsen
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Brandan Schultz
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Liana Protopsaltis
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Dan Averbuj
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Eric Blincow
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Jeanne Carroll
- Rady Children's Hospital, San Diego, CA 92123, USA; Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Sara Caylor
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Thomas Defay
- Alexion, Astra Zeneca Rare Disease, Boston, MA 02210, USA
| | - Katarzyna Ellsworth
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Annette Feigenbaum
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA; Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Mia Gover
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Lucia Guidugli
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Christian Hansen
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Lucita Van Der Kraan
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | | | - Hugh Kwon
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Lakshminarasimha Madhavrao
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | | | - Yupu Liang
- Alexion, Astra Zeneca Rare Disease, Boston, MA 02210, USA
| | - Rebecca Mardach
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA; Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | | | - Hung Nguyen
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Anna-Kaisa Niemi
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA; Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Danny Oh
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Muhammed Saad
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA
| | - Gunter Scharer
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Jennifer Schleit
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | | | - Erica Sanford
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Laurie D Smith
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Mary J Willis
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Kristen Wigby
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Rebecca Reimers
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA; Rady Children's Hospital, San Diego, CA 92123, USA; Scripps Research Translational Institute, La Jolla, CA 92037, USA
| |
Collapse
|
19
|
Halley MC, Olson NW, Ashley EA, Goldenberg AJ, Tabor HK. A Just Genomics Needs an ELSI of Translation. Hastings Cent Rep 2024; 54 Suppl 2:S126-S135. [PMID: 39707956 PMCID: PMC11801241 DOI: 10.1002/hast.4938] [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: 12/23/2024]
Abstract
The rapid advances in genomics over the last decade have come to fruition amid intense public discussions of justice in medicine and health care. While much emphasis has been placed on increasing diversity in genomics research participation, an overly narrow focus on recruitment eschews recognition of the disparities in health care that will ultimately shape access to the benefits of genomic medicine. In this essay, we suggest that achieving a just genomics, both now and in the future, requires an explicit ELSI of translation-normative and pragmatic scholarship that embraces the interconnectedness of research and clinical care and centers the obligations of researchers, institutions, and funders to mitigate inequities throughout the translational pipeline. We propose core principles to guide an ELSI of translation and to ensure that this work balances the value of the generalizable knowledge that genomics research generates and the value of the individuals and communities who make this research possible.
Collapse
|
20
|
Cope HL, Milko LV, Jalazo ER, Crissman BG, Foreman AKM, Powell BC, deJong NA, Hunter JE, Boyea BL, Forsythe AN, Wheeler AC, Zimmerman RS, Suchy SF, Begtrup A, Langley KG, Monaghan KG, Kraczkowski C, Hruska KS, Kruszka P, Kucera KS, Berg JS, Powell CM, Peay HL. A systematic framework for selecting gene-condition pairs for inclusion in newborn sequencing panels: Early Check implementation. Genet Med 2024; 26:101290. [PMID: 39375994 PMCID: PMC11625596 DOI: 10.1016/j.gim.2024.101290] [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: 05/13/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
PURPOSE Research is underway worldwide to investigate the feasibility, acceptability, and utility of sequencing-based newborn screening. Different methods have been used to select gene-condition pairs for screening, leading to highly inconsistent gene lists across studies. METHODS Early Check developed and utilized actionability-based frameworks for evaluating gene-condition pairs for inclusion in newborn panels (panel 1-high actionability, panel 2-possible actionability). A previously developed framework, the Age-based Semi Quantitative Metric (ASQM), was adapted. Increasing ASQM scores, with a maximum of 15, suggest greater actionability. Wilcoxon tests were performed to compare panel 1 gene-condition pairs on the Recommended Uniform Screening Panel (RUSP) with non-RUSP pairs. RESULTS In our first round of assessment, Early Check identified 178 gene-condition pairs for inclusion in panel 1 and 29 for panel 2. Median ASQM scores of RUSP conditions on panel 1 was 12 (range 4 to 15) and non-RUSP was 13 (range 9 to 15). Median scores for panel 2 was 10 (range 6 to 14). CONCLUSION The Early Check frameworks provide a transparent, semiquantitative, and reproducible methodology for selecting gene-condition pairs for newborn screening sequencing pilot studies that may inform future integration of genomic sequencing into population-level newborn screening. Collaborative efforts among newborn sequencing studies to establish shared criteria is needed to enhance cross-study comparisons.
Collapse
Affiliation(s)
- Heidi L Cope
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC.
| | - Laura V Milko
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Elizabeth R Jalazo
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Blythe G Crissman
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC
| | | | - Bradford C Powell
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Neal A deJong
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jessica Ezzell Hunter
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - Beth Lincoln Boyea
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - Ana N Forsythe
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - Anne C Wheeler
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC
| | | | | | | | | | | | | | | | | | - Katerina S Kucera
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - Jonathan S Berg
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Cynthia M Powell
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Holly L Peay
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC
| |
Collapse
|
21
|
Kazan HH, Karaca M, Akan G, Özgen Ö, Tuncel G, Özketen AÇ, Balcı MC, Körbeyli HK, Atalar F, Gökçay GF. Oxford nanopore sequencing-based assay for BTD gene screening: Design, clinical validation, and variant frequency assessment in the Turkish population. Gene 2024; 928:148782. [PMID: 39033936 DOI: 10.1016/j.gene.2024.148782] [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: 05/29/2024] [Revised: 06/28/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Biotinidase deficiency (BTD) is an autosomal recessive disorder characterized by impaired recycling of the water-soluble vitamin biotin which leads to a spectrum of clinical manifestations ranging from mild to severe, including mainly neurological and cutaneous symptoms. Biotin supplementation is a cornerstone of treatment, but diagnosis often relies on measuring serum enzyme activity, which needs to be confirmed by genetic analysis. Thus, molecular methods become necessary in the differential diagnosis of BTD. Accordingly, countries with a high-incidence have implemented next-generation sequencing (NGS) techniques to newborn screening programs for BT. Nevertheless, NGS platforms, while well-established, present challenges in cost, labor, accessibility, and duration for newborn screening programs targeting BTD, therefore these limitations necessitate the exploration of alternative systems to ensure efficient and widespread screening. Here, third-generation sequencing platforms, notably Oxford Nanopore Technology (ONT), present promising solutions to the associated challenges. Hence, in the present study, we aimed to develop an ONT-based assay for the screening of BTD gene. After designing and optimizing primers for long-PCR using reference DNA, we assessed the performance of the ONT assay in BTD patients previously diagnosed by enzyme assay and confirmed using Illumina-based sequencing. The results demonstrate a strong correlation between the two methods, indicating the reliability of the ONT-based assay. Moreover, this first in-house single gene testing specifically tailored for BTD successfully detected previously known genetic variants with high sequencing depths, affirming the effectiveness of ONT-based sequencing in human genetics.
Collapse
Affiliation(s)
- Hasan Hüseyin Kazan
- Department of Medical Biology, Gulhane Faculty of Medicine, University of Health Sciences, Ankara, Turkey
| | - Meryem Karaca
- Division of Pediatric Nutrition and Metabolism, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Gökçe Akan
- DESAM Institute, Near East University, Mersin 10, Turkey
| | - Özge Özgen
- Rare Diseases Research Laboratory, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Gülten Tuncel
- DESAM Institute, Near East University, Mersin 10, Turkey
| | | | - Mehmet Cihan Balcı
- Division of Pediatric Nutrition and Metabolism, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Hüseyin Kutay Körbeyli
- Division of Pediatric Nutrition and Metabolism, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Fatmahan Atalar
- Rare Diseases Research Laboratory, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey; Department of Rare Diseases, Child Health Institute, Istanbul University, Istanbul, Turkey.
| | - Gülden Fatma Gökçay
- Division of Pediatric Nutrition and Metabolism, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
22
|
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
|
23
|
Vrščaj E, Dangouloff T, Osredkar D, Servais L. Newborn screening programs for spinal muscular atrophy worldwide in 2023. J Neuromuscul Dis 2024; 11:1180-1189. [PMID: 39973463 DOI: 10.1177/22143602241288095] [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] [Indexed: 02/21/2025]
Abstract
BACKGROUND Spinal muscular atrophy is a rare, genetic neuromuscular disorder. Disease-modifying therapies, when administered early, have shown improved outcomes, leading to the implementation of numerous newborn screening programs for spinal muscular atrophy. OBJECTIVE The aim of this study was to evaluate the progress in implementing newborn screening for spinal muscular atrophy and therapy accessibility worldwide, after the first paper published in 2021. METHODS We conducted a survey, contacted experts from 143 countries worldwide, gathered responses from 86 experts from 80 countries. RESULTS By 2023, 31 countries reported established programs, 33 in the beginning of the year 2024; identifying approximately 1176 cases of spinal muscular atrophy. Additionally, the availability of disease-modifying therapies has expanded. At least one therapy is now accessible in 62 countries. Challenges, such as lack of governmental support, resource constraints, and varying healthcare priorities continue to impede implementation in some countries. CONCLUSIONS The data shows a significant increase in the implementation of newborn screening programs since 2021. The experts are still expressing a strong need for equitable access to standard of care for all the patients globally. Despite all setbacks, collaborative efforts have played a crucial role in newborn screening for spinal muscular atrophy implementation and currently 7% of world newborns are being screened, projections indicate an estimated 18% screening rate by 2028.
Collapse
Affiliation(s)
- Eva Vrščaj
- Department of Pediatric Neurology, University Children's Hospital, University Medical Centre Ljubljana, Slovenia
| | - Tamara Dangouloff
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, University Hospital Liège & University of Liège, Belgium
| | - Damjan Osredkar
- Department of Pediatric Neurology, University Children's Hospital, University Medical Centre Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, Slovenia
| | - Laurent Servais
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Pediatrics, University Hospital Liège & University of Liège, Belgium
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, UK
| |
Collapse
|
24
|
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
|
25
|
Schnabel-Besson E, Mütze U, Dikow N, Hörster F, Morath MA, Alex K, Brennenstuhl H, Settegast S, Okun JG, Schaaf CP, Winkler EC, Kölker S. Wilson and Jungner Revisited: Are Screening Criteria Fit for the 21st Century? Int J Neonatal Screen 2024; 10:62. [PMID: 39311364 PMCID: PMC11417796 DOI: 10.3390/ijns10030062] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
Driven by technological innovations, newborn screening (NBS) panels have been expanded and the development of genomic NBS pilot programs is rapidly progressing. Decisions on disease selection for NBS are still based on the Wilson and Jungner (WJ) criteria published in 1968. Despite this uniform reference, interpretation of the WJ criteria and actual disease selection for NBS programs are highly variable. A systematic literature search [PubMED search "Wilson" AND "Jungner"; last search 16.07.22] was performed to evaluate the applicability of the WJ criteria for current and future NBS programs and the need for adaptation. By at least two reviewers, 105 publications (systematic literature search, N = 77; manual search, N = 28) were screened for relevant content and, finally, 38 publications were evaluated. Limited by the study design of qualitative text analysis, no statistical evaluation was performed, but a structured collection of reported aspects of criticism and proposed improvements was instead collated. This revealed a set of general limitations of the WJ criteria, such as imprecise terminology, lack of measurability and objectivity, missing pediatric focus, and absent guidance on program management. Furthermore, it unraveled specific aspects of criticism on clinical, diagnostic, therapeutic, and economical aspects. A major obstacle was found to be the incompletely understood natural history and phenotypic diversity of rare diseases prior to NBS implementation, resulting in uncertainty about case definition, risk stratification, and indications for treatment. This gap could be closed through the systematic collection and evaluation of real-world evidence on the quality, safety, and (cost-)effectiveness of NBS, as well as the long-term benefits experienced by screened individuals. An integrated NBS public health program that is designed to continuously learn would fulfil these requirements, and a multi-dimensional framework for future NBS programs integrating medical, ethical, legal, and societal perspectives is overdue.
Collapse
Affiliation(s)
- Elena Schnabel-Besson
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Ulrike Mütze
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Nicola Dikow
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Friederike Hörster
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Marina A. Morath
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Karla Alex
- Section Translational Medical Ethics, Department of Medical Oncology, National Center for Tumor Diseases (NCT), Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Heiko Brennenstuhl
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Sascha Settegast
- Section Translational Medical Ethics, Department of Medical Oncology, National Center for Tumor Diseases (NCT), Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Jürgen G. Okun
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Christian P. Schaaf
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Eva C. Winkler
- Section Translational Medical Ethics, Department of Medical Oncology, National Center for Tumor Diseases (NCT), Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| | - Stefan Kölker
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
26
|
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
|
27
|
Khera HK, Mishra R. Nucleic Acid Based Testing (NABing): A Game Changer Technology for Public Health. Mol Biotechnol 2024; 66:2168-2200. [PMID: 37695473 DOI: 10.1007/s12033-023-00870-4] [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: 04/28/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023]
Abstract
Timely and accurate detection of the causal agent of a disease is crucial to restrict suffering and save lives. Mere symptoms are often not enough to detect the root cause of the disease. Better diagnostics applied for screening at a population level and sensitive detection assays remain the crucial component of disease surveillance which may include clinical, plant, and environmental samples, including wastewater. The recent advances in genome sequencing, nucleic acid amplification, and detection methods have revolutionized nucleic acid-based testing (NABing) and screening assays. A typical NABing assay consists of three modules: isolation of the nucleic acid from the collected sample, identification of the target sequence, and final reading the target with the help of a signal, which may be in the form of color, fluorescence, etc. Here, we review current NABing assays covering the different aspects of all three modules. We also describe the frequently used target amplification or signal amplification procedures along with the variety of applications of this fast-evolving technology and challenges in implementation of NABing in the context of disease management especially in low-resource settings.
Collapse
Affiliation(s)
- Harvinder Kour Khera
- Tata Institute for Genetics and Society, New inStem Building NCBS Campus, GKVK Post, Bellary Road, Bengaluru, 560065, India.
| | - Rakesh Mishra
- Tata Institute for Genetics and Society, New inStem Building NCBS Campus, GKVK Post, Bellary Road, Bengaluru, 560065, India.
- CSIR-Centre for Cellular and Molecular Biology, Uppal Rd, IICT Colony, Habsiguda, Hyderabad, Telangana, 500007, India.
| |
Collapse
|
28
|
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
|
29
|
Baple EL, Scott RH, Banka S, Buchanan J, Fish L, Wynn S, Wilkinson D, Ellard S, MacArthur DG, Stark Z. Exploring the benefits, harms and costs of genomic newborn screening for rare diseases. Nat Med 2024; 30:1823-1825. [PMID: 38898121 DOI: 10.1038/s41591-024-03055-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Affiliation(s)
- Emma L Baple
- RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK.
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK.
| | - Richard H Scott
- Great Ormond Street Hospital for Children, London, UK.
- UCL Great Ormond Street Institute of Child Health, London, UK.
- Genomics England, London, UK.
| | - Siddharth Banka
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
| | - James Buchanan
- Health Economics and Policy Research Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Sarah Wynn
- Unique-Rare Chromosome Disorder Support Group, Oxted, UK
| | - Dominic Wilkinson
- Faculty of Philosophy, Oxford Uehiro Centre for Practical Ethics, University of Oxford, Oxford, UK
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- John Radcliffe Hospital, Oxford University NHS Foundation Trust, Oxford, UK
- Centre for Biomedical Ethics, National University of, Singapore, Singapore
| | - Sian Ellard
- Exeter Genomics Laboratory, South West Genomic Laboratory Hub, Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Daniel G MacArthur
- Centre for Population Genomics, Garvan Institute of Medical Research, and UNSW Sydney, 384 Victoria Street, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Zornitza Stark
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Australian Genomics, Melbourne, Victoria, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
30
|
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
|
31
|
Shah N, Brlek P, Bulić L, Brenner E, Škaro V, Skelin A, Projić P, Shah P, Primorac D. Genomic sequencing for newborn screening: current perspectives and challenges. Croat Med J 2024; 65:261-267. [PMID: 38868972 PMCID: PMC11157259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/17/2024] [Indexed: 06/14/2024] Open
Abstract
Traditional newborn screening (NBS) serves as a critical tool in identifying conditions that may impact a child's health from an early stage. Newborn sequencing (NBSeq), the comprehensive analysis of an infant's genome, holds immense promise for revolutionizing health care throughout the lifespan. NBSeq allows for early detection of genetic disease risk and precision personalized medicine. The rapid evolution of DNA sequencing technologies and increasing affordability have spurred numerous endeavors to explore the potential of whole-genome sequencing in newborn screening. However, this transformative potential cannot be realized without challenges. Ethical aspects must be carefully navigated to safeguard individual rights and maintain public trust. Moreover, genomic data interpretation poses complex challenges due to its amount, the presence of variants of uncertain significance, and the dynamic nature of our understanding of genetics. Implementation hurdles, including cost, infrastructure, and specialized expertise, also present barriers to the widespread adoption of NBSeq. Addressing these challenges requires collaboration among clinicians, researchers, policymakers, ethicists, and stakeholders across various sectors. Robust frameworks for informed consent, data protection, and governance are essential. Advances in bioinformatics, machine learning, and genomic interpretation are crucial for translation into actionable clinical insights. Scalability and improving downstream health care access are vital for equitability, particularly in underserved communities. By fostering interdisciplinary collaboration, advancing technology and infrastructure, and upholding ethical principles, we can unlock the full potential of NBSeq as a tool for precision medicine and pave the way toward a future where every child has the opportunity for a healthier, genomics-informed start to life.
Collapse
Affiliation(s)
| | - Petar Brlek
- Petar Brlek, St. Catherine Specialty Hospital, Ul. kneza Branimira 71E, 10000, Zagreb, Croatia,
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Shao X, Steiner R, Peterson AL. Newborn screening for lipid disorders. Curr Opin Lipidol 2024; 35:149-156. [PMID: 38408035 DOI: 10.1097/mol.0000000000000928] [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] [Indexed: 02/28/2024]
Abstract
PURPOSE OF REVIEW Newborn screening is one of the most successful public health programs of the last century and offers unparalleled access to universal screening for a variety of metabolic and other disorders. Interest in development of newborn screening for lipid disorders has intensified in recent years. Screening newborns for lipid disorders has important implications for the health of the newborn as well as their relatives, and in the case of more common lipid disorders like familial hypercholesterolemia, could have important public health implications. RECENT FINDINGS Recent studies have demonstrated feasibility of measuring biomarkers for heterozygous familial hypercholesterolemia from newborn screening dried blood spot specimens. Another lipid disorder, cerebrotendinous xanthomatosis, is currently amenable to newborn screening utilizing currently available assays. New research in next-generation sequencing as a primary screen in newborns will also identify both common and rare lipid disorders in newborns. SUMMARY Historically, newborn screening for lipid disorders was not done for many reasons, but new research has developed testing methods that may successfully identify common and rare lipid disorders. This will impact the health of the newborn but could also impact family members and public health.
Collapse
Affiliation(s)
- Xiangqiang Shao
- Department of Pediatrics, Division of Genetics and Metabolism
| | - Robert Steiner
- Department of Pediatrics, Division of Genetics and Metabolism
| | - Amy L Peterson
- Department of Pediatrics, Division of Pediatric Cardiology, University of Wisconsin School of Medicine and Public Health. Madison, Wisconsin, USA
| |
Collapse
|
33
|
Phanthunane C, Pongcharoen S, Pannarunothai S, Roboon J, Phanthunane P, Nontarak J. Precision medicine in Asia enhanced by next-generation sequencing: Implications for Thailand through a scoping review and interview study. Clin Transl Sci 2024; 17:e13868. [PMID: 38924657 PMCID: PMC11197108 DOI: 10.1111/cts.13868] [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: 10/10/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Next-generation sequencing (NGS) significantly enhances precision medicine (PM) by offering personalized approaches to diagnosis, treatment, and prevention of unmet medical needs. Little is known about the current situation of PM in Asia. Thus, we aimed to conduct an overview of the progress and gaps in PM in Asia and enrich it with in-depth insight into the possibilities of future PM in Thailand. This scoping review focused on Asian countries starting with non-cancer studies, including rare and undiagnosed diseases (RUDs), non-communicable diseases (NCDs), infectious diseases (IDs), and pharmacogenomics, with a focus on NGS. Subsequent in-depth interviews with experts in Thailand were performed, and a thematic analysis served as the main qualitative methodology. Out of 2898 searched articles, 387 studies were included after the review. Although most of the studies focused on cancer, 89 (23.0%) studies were related to RUDs (17.1%), NCDs (2.8%), IDs (1.8%), and pharmacogenomics (1.3%). Apart from medicine and related sciences, the studies were mostly composed of PM (61.8%), followed by genetics medicine and bioinformatics. Interestingly, 28% of articles were conducted exclusively within the fields of medicine and related sciences, emphasizing interdisciplinary integration. The experts emphasized the need for sustainability-driven political will, nurturing collaboration, reinforcing computational infrastructure, and expanding the bioinformatic workforce. In Asia, developments of NGS have made remarkable progress in PM. Thailand has extended PM beyond cancer and focused on clinical implementation. We summarized the PM challenges, including equity and efficiency targeting, guided research funding, sufficient sample size, integrated collaboration, computational infrastructure, and sufficient trained human resources.
Collapse
Affiliation(s)
- Chumut Phanthunane
- Division of Medical OncologyChulabhorn Hospital, Chulabhorn Royal AcademyBangkokThailand
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of MedicineNaresuan UniversityPhitsanulokThailand
| | | | - Jureepon Roboon
- Department of Anatomy, Faculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
- Centre of Excellence in Medical BiotechnologyNaresuan UniversityPhitsanulokThailand
| | - Pudtan Phanthunane
- Department of Economics, Faculty of Business, Economics and CommunicationsNaresuan UniversityPhitsanulokThailand
| | - Jiraluck Nontarak
- Department of Epidemiology, Faculty of Public HealthMahidol UniversityBangkokThailand
| |
Collapse
|
34
|
Wang D, Zhang J, Yang R, Zhang D, Wang M, Yu C, Yang J, Huang W, Liu S, Tang S, He X. Disease spectrum, prevalence, genetic characteristics of inborn errors of metabolism in 21,840 hospitalized infants in Chongqing, China, 2017-2022. Front Genet 2024; 15:1395988. [PMID: 38863445 PMCID: PMC11165094 DOI: 10.3389/fgene.2024.1395988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/08/2024] [Indexed: 06/13/2024] Open
Abstract
Inborn errors of metabolism (IEMs) are uncommon. Although some studies have explored the distribution and characteristics of IEMs in newborns, the impact of these disorders on hospitalized newborns remains unclear. In this study, we gathered data from 21,840 newborn patients admitted for various medical conditions at the Children's Hospital of Chongqing Medical University from January 2017 and December 2022. Liquid chromatography-tandem mass spectrometry (LC-MS/MS), gas chromatography-mass spectrometry (GC-MS/MS), and genetic analysis were used to elucidate the disease spectrum, incidence rate, and genetic characteristics of IEMs in hospitalized newborns. The results revealed that the incidence of IEMs in hospitalized newborns was 1/377 (58/21,840), with a higher incidence in full-term infants (1/428) than in premature infants (1/3,120). Among the diagnosed genetic metabolic diseases, organic acid metabolism disorders (1/662), amino acid metabolism disorders (1/950), and fatty acid oxidation disorders (1/10,920) were the most prevalent. Methylmalonic acidemia (MMA), especially the isolated form, emerged as the most common IEM, while neonatal intrahepatic cholestasis caused by citrin deficiency (NICCD) and ornithine transcarbamylase deficiency (OTCD) were prevalent in premature infants. Of the 58 confirmed cases of IEMs, 72 variants were identified, of which 31.94% (23/72) had not been reported previously. This study contributes to understanding the incidence and clinical features of IEMs in hospitalized newborns, offering more efficient strategies for screening and diagnosing these disorders.
Collapse
Affiliation(s)
- Dongjuan Wang
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Juan Zhang
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Yang
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Dayong Zhang
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Wang
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Chaowen Yu
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Jingli Yang
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Wenxia Huang
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Shan Liu
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Shi Tang
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan He
- Center for Clinical Molecular Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
35
|
Therrell BL, Padilla CD, Borrajo GJC, Khneisser I, Schielen PCJI, Knight-Madden J, Malherbe HL, Kase M. Current Status of Newborn Bloodspot Screening Worldwide 2024: A Comprehensive Review of Recent Activities (2020-2023). Int J Neonatal Screen 2024; 10:38. [PMID: 38920845 PMCID: PMC11203842 DOI: 10.3390/ijns10020038] [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: 02/06/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 06/27/2024] Open
Abstract
Newborn bloodspot screening (NBS) began in the early 1960s based on the work of Dr. Robert "Bob" Guthrie in Buffalo, NY, USA. His development of a screening test for phenylketonuria on blood absorbed onto a special filter paper and transported to a remote testing laboratory began it all. Expansion of NBS to large numbers of asymptomatic congenital conditions flourishes in many settings while it has not yet been realized in others. The need for NBS as an efficient and effective public health prevention strategy that contributes to lowered morbidity and mortality wherever it is sustained is well known in the medical field but not necessarily by political policy makers. Acknowledging the value of national NBS reports published in 2007, the authors collaborated to create a worldwide NBS update in 2015. In a continuing attempt to review the progress of NBS globally, and to move towards a more harmonized and equitable screening system, we have updated our 2015 report with information available at the beginning of 2024. Reports on sub-Saharan Africa and the Caribbean, missing in 2015, have been included. Tables popular in the previous report have been updated with an eye towards harmonized comparisons. To emphasize areas needing attention globally, we have used regional tables containing similar listings of conditions screened, numbers of screening laboratories, and time at which specimen collection is recommended. Discussions are limited to bloodspot screening.
Collapse
Affiliation(s)
- Bradford L. Therrell
- Department of Pediatrics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
- National Newborn Screening and Global Resource Center, Austin, TX 78759, USA
| | - Carmencita D. Padilla
- Department of Pediatrics, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines;
| | - Gustavo J. C. Borrajo
- Detección de Errores Congénitos—Fundación Bioquímica Argentina, La Plata 1908, Argentina;
| | - Issam Khneisser
- Jacques LOISELET Genetic and Genomic Medical Center, Faculty of Medicine, Saint Joseph University, Beirut 1104 2020, Lebanon;
| | - Peter C. J. I. Schielen
- Office of the International Society for Neonatal Screening, Reigerskamp 273, 3607 HP Maarssen, The Netherlands;
| | - Jennifer Knight-Madden
- Caribbean Institute for Health Research—Sickle Cell Unit, The University of the West Indies, Mona, Kingston 7, Jamaica;
| | - Helen L. Malherbe
- Centre for Human Metabolomics, North-West University, Potchefstroom 2531, South Africa;
- Rare Diseases South Africa NPC, The Station Office, Bryanston, Sandton 2021, South Africa
| | - Marika Kase
- Strategic Initiatives Reproductive Health, Revvity, PL10, 10101 Turku, Finland;
| |
Collapse
|
36
|
Lynch F, Best S, Gaff C, Downie L, Archibald AD, Gyngell C, Goranitis I, Peters R, Savulescu J, Lunke S, Stark Z, Vears DF. Australian public perspectives on genomic newborn screening: which conditions should be included? Hum Genomics 2024; 18:45. [PMID: 38720401 PMCID: PMC11077791 DOI: 10.1186/s40246-024-00611-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: 02/13/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Implementing genomic sequencing into newborn screening programs allows for significant expansion in the number and scope of conditions detected. We sought to explore public preferences and perspectives on which conditions to include in genomic newborn screening (gNBS). METHODS We recruited English-speaking members of the Australian public over 18 years of age, using social media, and invited them to participate in online focus groups. RESULTS Seventy-five members of the public aged 23-72 participated in one of fifteen focus groups. Participants agreed that if prioritisation of conditions was necessary, childhood-onset conditions were more important to include than later-onset conditions. Despite the purpose of the focus groups being to elicit public preferences, participants wanted to defer to others, such as health professionals or those with a lived experience of each condition, to make decisions about which conditions to include. Many participants saw benefit in including conditions with no available treatment. Participants agreed that gNBS should be fully publicly funded. CONCLUSION How many and which conditions are included in a gNBS program will be a complex decision requiring detailed assessment of benefits and costs alongside public and professional engagement. Our study provides support for implementing gNBS for treatable childhood-onset conditions.
Collapse
Affiliation(s)
- Fiona Lynch
- Biomedical Ethics Research Group, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Melbourne Law School, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Stephanie Best
- Sir Peter MacCallum Cancer Centre Dept of Oncology, University of Melbourne, Melbourne, VIC, 3052, Australia
- Australian Genomics, Melbourne, VIC, 3052, Australia
- School of Health Sciences, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Clara Gaff
- Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Melbourne Genomics, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Lilian Downie
- Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3052, Australia
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Alison D Archibald
- Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3052, Australia
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Christopher Gyngell
- Biomedical Ethics Research Group, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Ilias Goranitis
- Australian Genomics, Melbourne, VIC, 3052, Australia
- Economics of Genomics and Precision Medicine Unit, Centre for Health Policy, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Riccarda Peters
- Economics of Genomics and Precision Medicine Unit, Centre for Health Policy, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Julian Savulescu
- Biomedical Ethics Research Group, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Melbourne Law School, The University of Melbourne, Melbourne, VIC, 3052, Australia
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Uehiro Chair of Practical Ethics, The Oxford Uehiro Centre for Practical Ethics, Oxford University, Oxford, OX1 1PT, UK
| | - Sebastian Lunke
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Pathology, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Zornitza Stark
- Australian Genomics, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3052, Australia
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Danya F Vears
- Biomedical Ethics Research Group, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia.
- Melbourne Law School, The University of Melbourne, Melbourne, VIC, 3052, Australia.
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3052, Australia.
- Centre for Biomedical Ethics and Law, Department of Public Health and Primary Care, KU Leuven, Leuven, 3000, Belgium.
| |
Collapse
|
37
|
Downie L, Bouffler SE, Amor DJ, Christodoulou J, Yeung A, Horton AE, Macciocca I, Archibald AD, Wall M, Caruana J, Lunke S, Stark Z. Gene selection for genomic newborn screening: Moving toward consensus? Genet Med 2024; 26:101077. [PMID: 38275146 DOI: 10.1016/j.gim.2024.101077] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
PURPOSE Gene selection for genomic newborn screening (gNBS) underpins the validity, acceptability, and ethical application of this technology. Existing gNBS gene lists are highly variable despite being based on shared principles of gene-disease validity, treatability, and age of onset. This study aimed to curate a gNBS gene list that builds upon existing efforts and provide a core consensus list of gene-disease pairs assessed by multiple expert groups worldwide. METHODS Our multidisciplinary expert team curated a gene list using an open platform and multiple existing curated resources. We included severe treatable disorders with age of disease onset <5 years with established gene-disease associations and reliable variant detection. We compared the final list with published lists from 5 other gNBS projects to determine consensus genes and to identify areas of discrepancy. RESULTS We reviewed 1279 genes and 604 met our inclusion criteria. Metabolic conditions comprised the largest group (25%), followed by immunodeficiencies (21%) and endocrine disorders (15%). We identified 55 consensus genes included by all 6 gNBS research projects. Common reasons for discrepancy included variable definitions of treatability and strength of gene-disease association. CONCLUSION We have identified a consensus gene list for gNBS that can be used as a basis for systematic harmonization efforts internationally.
Collapse
Affiliation(s)
- Lilian Downie
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | | | - David J Amor
- Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - John Christodoulou
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Alison Yeung
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Ari E Horton
- Victorian Heart Institute, Monash University, Melbourne, VIC, Australia; Public Health Genomics, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Ivan Macciocca
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Alison D Archibald
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Meghan Wall
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Jade Caruana
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Sebastian Lunke
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia; Australian Genomics, Melbourne, VIC, Australia.
| |
Collapse
|
38
|
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
|
39
|
Rips J, Halstuk O, Fuchs A, Lang Z, Sido T, Gershon-Naamat S, Abu-Libdeh B, Edvardson S, Salah S, Breuer O, Hadhud M, Eden S, Simon I, Slae M, Damseh NS, Abu-Libdeh A, Eskin-Schwartz M, Birk OS, Varga J, Schueler-Furman O, Rosenbluh C, Elpeleg O, Yanovsky-Dagan S, Mor-Shaked H, Harel T. Unbiased phenotype and genotype matching maximizes gene discovery and diagnostic yield. Genet Med 2024; 26:101068. [PMID: 38193396 DOI: 10.1016/j.gim.2024.101068] [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: 08/11/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024] Open
Abstract
PURPOSE Widespread application of next-generation sequencing, combined with data exchange platforms, has provided molecular diagnoses for countless families. To maximize diagnostic yield, we implemented an unbiased semi-automated genematching algorithm based on genotype and phenotype matching. METHODS Rare homozygous variants identified in 2 or more affected individuals, but not in healthy individuals, were extracted from our local database of ∼12,000 exomes. Phenotype similarity scores (PSS), based on human phenotype ontology terms, were assigned to each pair of individuals matched at the genotype level using HPOsim. RESULTS 33,792 genotype-matched pairs were discovered, representing variants in 7567 unique genes. There was an enrichment of PSS ≥0.1 among pathogenic/likely pathogenic variant-level pairs (94.3% in pathogenic/likely pathogenic variant-level matches vs 34.75% in all matches). We highlighted founder or region-specific variants as an internal positive control and proceeded to identify candidate disease genes. Variant-level matches were particularly helpful in cases involving inframe indels and splice region variants beyond the canonical splice sites, which may otherwise have been disregarded, allowing for detection of candidate disease genes, such as KAT2A, RPAIN, and LAMP3. CONCLUSION Semi-automated genotype matching combined with PSS is a powerful tool to resolve variants of uncertain significance and to identify candidate disease genes.
Collapse
Affiliation(s)
- Jonathan Rips
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Orli Halstuk
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Adina Fuchs
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Ziv Lang
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Tal Sido
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | | | - Bassam Abu-Libdeh
- Department of Pediatrics & Genetics, Makassed Hospital & Al-Quds Medical School, E. Jerusalem, Palestine
| | - Simon Edvardson
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; Pediatric Neurology Unit, Hadassah Medical Center, Jerusalem, Israel
| | - Somaya Salah
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Oded Breuer
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; Pediatric Pulmonology and CF Unit, Department of Pediatrics, Hadassah Medical Center, Jerusalem, Israel
| | - Mohamad Hadhud
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; Pediatric Pulmonology and CF Unit, Department of Pediatrics, Hadassah Medical Center, Jerusalem, Israel
| | - Sharon Eden
- Institute of Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Itamar Simon
- Institute of Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Mordechai Slae
- Pediatric Gastroenterology Unit, Department of Pediatrics, Hadassah Medical Center, Jerusalem, Israel
| | - Nadirah S Damseh
- Department of Pediatrics & Genetics, Makassed Hospital & Al-Quds Medical School, E. Jerusalem, Palestine
| | - Abdulsalam Abu-Libdeh
- Department of Pediatrics & Genetics, Makassed Hospital & Al-Quds Medical School, E. Jerusalem, Palestine; Division of Pediatric Endocrinology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Marina Eskin-Schwartz
- The Morris Kahn Laboratory of Human Genetics at the National Institute of Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; Genetics Institute, Soroka University Medical Center, Beer-Sheva, Israel
| | - Ohad S Birk
- The Morris Kahn Laboratory of Human Genetics at the National Institute of Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; Genetics Institute, Soroka University Medical Center, Beer-Sheva, Israel
| | - Julia Varga
- Microbiology and Molecular Genetics, Institute for Biomedical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ora Schueler-Furman
- Microbiology and Molecular Genetics, Institute for Biomedical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Orly Elpeleg
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | - Hagar Mor-Shaked
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Tamar Harel
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| |
Collapse
|
40
|
Fehlings DL, Zarrei M, Engchuan W, Sondheimer N, Thiruvahindrapuram B, MacDonald JR, Higginbotham EJ, Thapa R, Behlim T, Aimola S, Switzer L, Ng P, Wei J, Danthi PS, Pellecchia G, Lamoureux S, Ho K, Pereira SL, de Rijke J, Sung WWL, Mowjoodi A, Howe JL, Nalpathamkalam T, Manshaei R, Ghaffari S, Whitney J, Patel RV, Hamdan O, Shaath R, Trost B, Knights S, Samdup D, McCormick A, Hunt C, Kirton A, Kawamura A, Mesterman R, Gorter JW, Dlamini N, Merico D, Hilali M, Hirschfeld K, Grover K, Bautista NX, Han K, Marshall CR, Yuen RKC, Subbarao P, Azad MB, Turvey SE, Mandhane P, Moraes TJ, Simons E, Maxwell G, Shevell M, Costain G, Michaud JL, Hamdan FF, Gauthier J, Uguen K, Stavropoulos DJ, Wintle RF, Oskoui M, Scherer SW. Comprehensive whole-genome sequence analyses provide insights into the genomic architecture of cerebral palsy. Nat Genet 2024; 56:585-594. [PMID: 38553553 DOI: 10.1038/s41588-024-01686-x] [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: 12/23/2022] [Accepted: 02/13/2024] [Indexed: 04/17/2024]
Abstract
We performed whole-genome sequencing (WGS) in 327 children with cerebral palsy (CP) and their biological parents. We classified 37 of 327 (11.3%) children as having pathogenic/likely pathogenic (P/LP) variants and 58 of 327 (17.7%) as having variants of uncertain significance. Multiple classes of P/LP variants included single-nucleotide variants (SNVs)/indels (6.7%), copy number variations (3.4%) and mitochondrial mutations (1.5%). The COL4A1 gene had the most P/LP SNVs. We also analyzed two pediatric control cohorts (n = 203 trios and n = 89 sib-pair families) to provide a baseline for de novo mutation rates and genetic burden analyses, the latter of which demonstrated associations between de novo deleterious variants and genes related to the nervous system. An enrichment analysis revealed previously undescribed plausible candidate CP genes (SMOC1, KDM5B, BCL11A and CYP51A1). A multifactorial CP risk profile and substantial presence of P/LP variants combine to support WGS in the diagnostic work-up across all CP and related phenotypes.
Collapse
Affiliation(s)
- Darcy L Fehlings
- Division of Developmental Paediatrics, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mehdi Zarrei
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Worrawat Engchuan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Neal Sondheimer
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Jeffrey R MacDonald
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Edward J Higginbotham
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ritesh Thapa
- Division of Developmental Paediatrics, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Tarannum Behlim
- Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Sabrina Aimola
- Division of Developmental Paediatrics, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Lauren Switzer
- Division of Developmental Paediatrics, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Pamela Ng
- Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - John Wei
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Prakroothi S Danthi
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Giovanna Pellecchia
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sylvia Lamoureux
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Karen Ho
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sergio L Pereira
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jill de Rijke
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Wilson W L Sung
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alireza Mowjoodi
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jennifer L Howe
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Thomas Nalpathamkalam
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Roozbeh Manshaei
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Ted Rogers Centre for Heart Research, Cardiac Genome Clinic, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Siavash Ghaffari
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joseph Whitney
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rohan V Patel
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Omar Hamdan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rulan Shaath
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brett Trost
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shannon Knights
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Grandview Children's Centre, Oshawa, Ontario, Canada
| | - Dawa Samdup
- Department of Pediatrics, Queen's University, Kingston, Ontario, Canada
| | - Anna McCormick
- Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
| | - Carolyn Hunt
- Grandview Children's Centre, Oshawa, Ontario, Canada
| | - Adam Kirton
- Department of Pediatrics, Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Anne Kawamura
- Division of Developmental Paediatrics, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ronit Mesterman
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Jan Willem Gorter
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Nomazulu Dlamini
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Neurology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Neurosciences and Mental Health Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniele Merico
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Deep Genomics Inc., Toronto, Ontario, Canada
- Vevo Therapeutics Inc., San Francisco, CA, USA
| | - Murto Hilali
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kyle Hirschfeld
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kritika Grover
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nelson X Bautista
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kara Han
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christian R Marshall
- Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ryan K C Yuen
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Padmaja Subbarao
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Piush Mandhane
- Faculty of Medicine & Dentistry, Pediatrics Department, University of Alberta, Edmonton, Alberta, Canada
| | - Theo J Moraes
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Program in Translation Medicine & Division of Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, Section of Allergy and Clinical Immunology, University of Manitoba, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - George Maxwell
- Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - Michael Shevell
- Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
- Departments of Pediatrics and Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Gregory Costain
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jacques L Michaud
- Departments of Pediatrics and Neurosciences, Université de Montréal, Montréal, Québec, Canada
- CHU Sainte-Justine Azrieli Research Center, Montréal, Québec, Canada
| | - Fadi F Hamdan
- CHU Sainte-Justine Azrieli Research Center, Montréal, Québec, Canada
- Department of Pediatrics, Université de Montréal, Montréal, Québec, Canada
| | - Julie Gauthier
- CHU Sainte-Justine Azrieli Research Center, Montréal, Québec, Canada
- Department of Pediatrics, Université de Montréal, Montréal, Québec, Canada
| | - Kevin Uguen
- CHU Sainte-Justine Azrieli Research Center, Montréal, Québec, Canada
| | - Dimitri J Stavropoulos
- Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Richard F Wintle
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maryam Oskoui
- Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
- Departments of Pediatrics and Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Kariyawasam DS, Scarfe J, Meagher C, Farrar MA, Bhattacharya K, Carter SM, Newson AJ, Otlowski M, Watson J, Millis N, Norris S. 'Integrating Ethics and Equity with Economics and Effectiveness for newborn screening in the genomic age: A qualitative study protocol of stakeholder perspectives. PLoS One 2024; 19:e0299336. [PMID: 38527031 PMCID: PMC10962853 DOI: 10.1371/journal.pone.0299336] [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: 02/14/2024] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Newborn bloodspot screening is a well-established population health initiative that detects serious, childhood-onset, treatable conditions to improve health outcomes. With genomic technologies advancing rapidly, many countries are actively discussing the introduction of genomic assays into newborn screening programs. While adding genomic testing to Australia's newborn screening program could improve outcomes for infants and families, it must be considered against potential harms, ethical, legal, equity and social implications, and economic and health system impacts. We must ask not only 'can' we use genomics to screen newborns?' but 'should we'?' and 'how much should health systems invest in genomic newborn screening?'. METHODS This study will use qualitative methods to explore understanding, priorities, concerns and expectations of genomic newborn screening among parents/carers, health professionals/scientists, and health policy makers across Australia. In-depth, semi-structured interviews will be held with 30-40 parents/carers recruited via hospital and community settings, 15-20 health professionals/scientists, and 10-15 health policy makers. Data will be analysed using inductive content analysis. The Sydney Children's Hospital Network Human Research Ethics Committee approved this study protocol [2023/ETH02371]. The Standards for Reporting Qualitative Research will guide study planning, conduct and reporting. DISCUSSION Few studies have engaged a diverse range of stakeholders to explore the implications of genomics in newborn screening in a culturally and genetically diverse population, nor in a health system underpinned by universal health care. As the first study within a multi-part research program, findings will be used to generate new knowledge on the risks and benefits and importance of ethical, legal, social and equity implications of genomic newborn screening from the perspective of key stakeholders. As such it will be the foundation on which child and family centered criteria can be developed to inform health technology assessments and drive efficient and effective policy decision-making on the implementation of genomics in newborn screening.
Collapse
Affiliation(s)
- Didu S. Kariyawasam
- Department of Neurology, Sydney Children’s Hospital Network, Sydney, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Joanne Scarfe
- Faculty of Medicine and Health, Sydney School of Public Health, Menzies Centre for Health Policy & Economics, The University of Sydney, Camperdown, New South Wales, Australia
| | - Christian Meagher
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michelle A. Farrar
- Department of Neurology, Sydney Children’s Hospital Network, Sydney, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kaustav Bhattacharya
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Genetic Metabolic Disorders Service, Sydney Children’s Hospital Network, Randwick and Westmead, New South Wales, Australia
- Faculty of Medicine and Health, Discipline of Genomics, Sydney University, Westmead, New South Wales, Australia
| | - Stacy M. Carter
- Australian Centre for Health Engagement, Evidence and Values, School of Health and Society, The University of Wollongong, Wollongong, New South Wales, Australia
| | - Ainsley J. Newson
- Sydney Health Ethics, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Margaret Otlowski
- Centre for Law and Genetics, Faculty of Law, College of Arts, Law and Education, University of Tasmania, Tasmania, Australia
| | - Jo Watson
- HTA Consumer Consultative Committee, Department of Health & Aged Care, Canberra, Australian Capital Territory, Australia
| | | | - Sarah Norris
- Faculty of Medicine and Health, Sydney School of Public Health, Menzies Centre for Health Policy & Economics, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
42
|
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
|
43
|
Brunelli L, Chan K, Tabery J, Binford W, Brower A. A Children's Rights Framework for Genomic Medicine: Newborn Screening as a Use Case. MEDICAL RESEARCH ARCHIVES 2024; 12:5167. [PMID: 39220179 PMCID: PMC11364257 DOI: 10.18103/mra.v12i3.5167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The year 2023 marked the 60th anniversary of screening newborns in the United States for diseases that benefit from early identification and intervention. All around the world, the goal of NBS is to facilitate timely diagnosis and management to improve individual health outcomes in all newborns regardless of their place of birth, economic circumstances, ability to pay for treatment, and access to healthcare. Advances in technology to screen and treat disease have led to a rapid increase in the number of screened conditions, and innovations in genomics are expected to exponentially expand this number further. A system where all newborns are screened, coupled with rapid technological innovation, provides a unique opportunity to improve pediatric health outcomes and advance children's rights, including the unique rights of sick and disabled children. This is especially timely as we approach the 100th anniversary of the 1924 Geneva Declaration of the Rights of the Child, which includes children's right to healthcare, and the 1989 United Nations Convention on the Rights of the Child that expanded upon this aspect and affirmed each child's right to the highest attainable standard of health. In this manuscript, we provide background on the evolving recognition of the rights of children and the foundational rights to healthcare and non-discrimination, provide two examples that highlight issues to access and equity in newborn screening that may limit a child's right to healthcare and best possible outcomes, detail ways the current approach to newborn screening advances the rights of the child, and finally, propose that the incorporation of genomics into newborn screening presents a useful case study to recognize and uphold the rights of every child.
Collapse
Affiliation(s)
- Luca Brunelli
- Division of Neonatology, Department of Pediatrics, Spencer Fox Eccles School of Medicine, University of Utah
| | - Kee Chan
- American College of Medical Genetics and Genomics
| | | | - Warren Binford
- University of Colorado, School of Medicine, CU Law School
| | - Amy Brower
- American College of Medical Genetics and Genomics
- Munroe-Meyer Institute, University of Nebraska Medical Center
- Creighton University School of Medicine
| |
Collapse
|
44
|
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
|
45
|
Singh G, Alser M, Denolf K, Firtina C, Khodamoradi A, Cavlak MB, Corporaal H, Mutlu O. RUBICON: a framework for designing efficient deep learning-based genomic basecallers. Genome Biol 2024; 25:49. [PMID: 38365730 PMCID: PMC10870431 DOI: 10.1186/s13059-024-03181-2] [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: 04/24/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Nanopore sequencing generates noisy electrical signals that need to be converted into a standard string of DNA nucleotide bases using a computational step called basecalling. The performance of basecalling has critical implications for all later steps in genome analysis. Therefore, there is a need to reduce the computation and memory cost of basecalling while maintaining accuracy. We present RUBICON, a framework to develop efficient hardware-optimized basecallers. We demonstrate the effectiveness of RUBICON by developing RUBICALL, the first hardware-optimized mixed-precision basecaller that performs efficient basecalling, outperforming the state-of-the-art basecallers. We believe RUBICON offers a promising path to develop future hardware-optimized basecallers.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland
- Research and Advanced Development, AMD, Longmont, USA
| | - Mohammed Alser
- Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland
| | | | - Can Firtina
- Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland.
| | | | - Meryem Banu Cavlak
- Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland
| | - Henk Corporaal
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Onur Mutlu
- Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
46
|
Boerwinkle VL, Appavu B, Cediel EG, Erklaurer J, Lalgudi Ganesan S, Gibbons C, Hahn C, LaRovere KL, Moberg D, Natarajan G, Molteni E, Reuther WR, Slomine BS. Common Data Elements for Disorders of Consciousness: Recommendations from the Working Group in the Pediatric Population. Neurocrit Care 2024; 40:65-73. [PMID: 38062304 DOI: 10.1007/s12028-023-01870-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND The fundamental gap obstructing forward progress of evidenced-based care in pediatric and neonatal disorders of consciousness (DoC) is the lack of defining consensus-based terminology to perform comparative research. This lack of shared nomenclature in pediatric DoC stems from the inherently recursive dilemma of the inability to reliably measure consciousness in the very young. However, recent advancements in validated clinical examinations and technologically sophisticated biomarkers of brain activity linked to future abilities are unlocking this previously formidable challenge to understanding the DoC in the developing brain. METHODS To address this need, the first of its kind international convergence of an interdisciplinary team of pediatric DoC experts was organized by the Neurocritical Care Society's Curing Coma Campaign. The multidisciplinary panel of pediatric DoC experts proposed pediatric-tailored common data elements (CDEs) covering each of the CDE working groups including behavioral phenotyping, biospecimens, electrophysiology, family and goals of care, neuroimaging, outcome and endpoints, physiology and big Data, therapies, and pediatrics. RESULTS We report the working groups' pediatric-focused DoC CDE recommendations and disseminate CDEs to be used in studies of pediatric patients with DoC. CONCLUSIONS The CDEs recommended support the vision of progressing collaborative and successful internationally collaborative pediatric coma research.
Collapse
Affiliation(s)
- Varina L Boerwinkle
- Department of Neurology, University of North Carolina in Chapel Hill, Chapel Hill, NC, USA.
| | - Brian Appavu
- Department of Child Health and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Emilio Garzon Cediel
- Department of Neurology, University of North Carolina in Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer Erklaurer
- Divisions of Critical Care Medicine and Child Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Saptharishi Lalgudi Ganesan
- Departments of Paediatrics and Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Christie Gibbons
- Brain Injury Association of America Family Advocate, Phoenix, AZ, USA
| | - Cecil Hahn
- Department of Paediatrics (Neurology), The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Kerri L LaRovere
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dick Moberg
- Moberg Analytics, Inc., Philadelphia, PA, USA
| | - Girija Natarajan
- Discipline of Pediatrics, Children's Hospital of Michigan and Hutzel Women's Hospital, Central Michigan University, Mount Pleasant, MI, USA
| | - Erika Molteni
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - William R Reuther
- Department of Neurology, University of North Carolina in Chapel Hill, Chapel Hill, NC, USA
| | - Beth S Slomine
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
47
|
Solomon BD. The future of commercial genetic testing. Curr Opin Pediatr 2023; 35:615-619. [PMID: 37218641 PMCID: PMC10667560 DOI: 10.1097/mop.0000000000001260] [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: 05/24/2023]
Abstract
PURPOSE OF REVIEW There are thousands of different clinical genetic tests currently available. Genetic testing and its applications continue to change rapidly for multiple reasons. These reasons include technological advances, accruing evidence about the impact and effects of testing, and many complex financial and regulatory factors. RECENT FINDINGS This article considers a number of key issues and axes related to the current and future state of clinical genetic testing, including targeted versus broad testing, simple/Mendelian versus polygenic and multifactorial testing models, genetic testing for individuals with high suspicion of genetic conditions versus ascertainment through population screening, the rise of artificial intelligence in multiple aspects of the genetic testing process, and how developments such as rapid genetic testing and the growing availability of new therapies for genetic conditions may affect the field. SUMMARY Genetic testing is expanding and evolving, including into new clinical applications. Developments in the field of genetics will likely result in genetic testing becoming increasingly in the purview of a very broad range of clinicians, including general paediatricians as well as paediatric subspecialists.
Collapse
Affiliation(s)
- Benjamin D. Solomon
- Medical Genetics Branch, National Human Genome Research Institute, United States of America
| |
Collapse
|
48
|
Lev A, Somech R, Somekh I. Newborn screening for severe combined immunodeficiency and inborn errors of immunity. Curr Opin Pediatr 2023; 35:692-702. [PMID: 37707504 DOI: 10.1097/mop.0000000000001291] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
PURPOSE OF REVIEW Severe combined immune deficiency (SCID) is the most devastating genetic disease of the immune system with an unfavorable outcome unless diagnosed early in life. Newborn screening (NBS) programs play a crucial role in facilitating early diagnoses and timely interventions for affected infants. RECENT FINDINGS SCID marked the pioneering inborn error of immunity (IEI) to undergo NBS, a milestone achieved 15 years ago through the enumeration of T-cell receptor excision circles (TRECs) extracted from Guthrie cards. This breakthrough has revolutionized our approach to SCID, enabling not only presymptomatic identification and prompt treatments (including hematopoietic stem cell transplantation), but also enhancing our comprehension of the global epidemiology of SCID. SUMMARY NBS is continuing to evolve with the advent of novel diagnostic technologies and treatments. Following the successful implementation of SCID-NBS programs, a call for the early identification of additional IEIs is the next step, encompassing a broader spectrum of IEIs, facilitating early diagnoses, and preventing morbidity and mortality.
Collapse
Affiliation(s)
- Atar Lev
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | | | | |
Collapse
|
49
|
Djafar JV, Smith NJ, Johnson AM, Bhattacharya K, Ardern-Holmes SL, Ellaway C, Dale RC, D'Silva AM, Kariyawasam DS, Grattan S, Kandula T, Lewis K, Mohammed SS, Farrar MA. Characterizing Common Phenotypes Across the Childhood Dementia Disorders: A Cross-sectional Study From Two Australian Centers. Pediatr Neurol 2023; 149:75-83. [PMID: 37806042 DOI: 10.1016/j.pediatrneurol.2023.09.006] [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: 06/07/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Childhood dementias are a group of rare pediatric conditions characterized by progressive neurocognitive decline. Quantifying and characterising phenotypes to identify similarities between specific conditions is critical to inform opportunities to optimize care and advance research. METHODS This cross-sectional study recruited primary caregivers of children (<18 years) living with a dementia syndrome from neurology and metabolic clinics in Sydney and Adelaide, Australia. Sociodemographic and clinical data were collated. Behavior, eating, sleep, pain, and neurological disability were assessed using validated tools, including Strengths and Difficulties, Child Eating Behaviour, and Children's Sleep Habits questionnaires and visual analog of pain and modified Rankin scales. Data were analyzed with descriptive statistics. RESULTS Among 45 children with 23 different dementia syndromes, the modified Rankin Scale demonstrated at least moderate neurological disability and functional dependence in 82% (37/45). Families reported delays in receiving an accurate diagnosis following initial symptoms (mean: 1.6 ± 1.4 years, range: 0-5 years). The most prevalent phenotypes included communication, comprehension, or recall difficulties (87%, 39/45); disturbances in sleep (80%, 36/45); appetite changes (74%, 29/39); mobility issues (53%, 24/45); and hyperactive behavior (53%, 21/40). Behavioral problems had a "high" or "very high" impact on everyday family life in 73% (24/33). CONCLUSIONS Childhood dementia disorders share substantial behavioral, motor, sensory, and socioemotional symptoms, resulting in high care needs, despite their vast heterogeneity in age of onset and progression. Considering their unifying characteristics under one collective term is an opportunity to improve treatment, provide quality care, and accelerate research.
Collapse
Affiliation(s)
- Jason V Djafar
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Nicholas J Smith
- Department of Neurology and Clinical Neurophysiology, Women's and Children's Health Network, Adelaide, Australia; Discipline of Paediatrics, School of Medicine, The University of Adelaide, Adelaide, Australia
| | - Alexandra M Johnson
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia; Department of Neurology, Sydney Children's Hospital Network, Sydney, Australia
| | - Kaustuv Bhattacharya
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Sydney, Australia
| | | | - Carolyn Ellaway
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Sydney, Australia
| | - Russell C Dale
- Department of Neurology, Sydney Children's Hospital Network, Sydney, Australia
| | - Arlene M D'Silva
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Didu S Kariyawasam
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia; Department of Neurology, Sydney Children's Hospital Network, Sydney, Australia
| | - Sarah Grattan
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia
| | - Tejaswi Kandula
- Department of Neurology, Sydney Children's Hospital Network, Sydney, Australia
| | - Katherine Lewis
- Department of Neurology, Sydney Children's Hospital Network, Sydney, Australia
| | - Shekeeb S Mohammed
- Department of Neurology, Sydney Children's Hospital Network, Sydney, Australia
| | - Michelle A Farrar
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia; Department of Neurology, Sydney Children's Hospital Network, Sydney, Australia.
| |
Collapse
|
50
|
Abstract
Rare diseases are a leading cause of infant mortality and lifelong disability. To improve outcomes, timely diagnosis and effective treatments are needed. Genomic sequencing has transformed the traditional diagnostic process, providing rapid, accurate and cost-effective genetic diagnoses to many. Incorporating genomic sequencing into newborn screening programmes at the population scale holds the promise of substantially expanding the early detection of treatable rare diseases, with stored genomic data potentially benefitting health over a lifetime and supporting further research. As several large-scale newborn genomic screening projects launch internationally, we review the challenges and opportunities presented, particularly the need to generate evidence of benefit and to address the ethical, legal and psychosocial issues that genomic newborn screening raises.
Collapse
Affiliation(s)
- Zornitza Stark
- Australian Genomics, Melbourne, Victoria, Australia.
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
| | - Richard H Scott
- Great Ormond Street Hospital for Children, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
- Genomics England, London, UK
| |
Collapse
|