1
|
Doll ES, Mahal J, Alex K, Lerch SP, Kölker S, Schaaf CP, Winkler EC, Ditzen B. Tension between the need for certainty and numerous uncertainties-A focus group study on various perspectives on a potential genomic newborn screening program in Germany. J Genet Couns 2025; 34:e70004. [PMID: 40377246 DOI: 10.1002/jgc4.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 05/18/2025]
Abstract
The advancement of genome sequencing technology and its potential application in newborn screening is being discussed in various countries. Genomic newborn screening (gNBS) can provide parents with information about their child's genetic susceptibility for known disorders. However, it also presents ethical and psychosocial challenges. This study was carried out with a view toward the possible introduction of gNBS in Germany. Due to the existing challenges, it is crucial to understand different perspectives of relevant groups in Germany before implementing gNBS. Four online focus groups were conducted with parents, patient representatives, and healthcare professionals to explore perceived opportunities and challenges, as well as needs regarding a potential gNBS program. Discussions with altogether 24 participants were semi-structured using a pre-defined interview guide. Sessions were audio-visually recorded and transcripts were analyzed using a structuring qualitative content analysis combining both deductive and inductive methods. Participants expressed positive views about gNBS but also reservations about a gNBS program and posed requirements for operating conditions. One salient theme that emerged was hope for certainty through gNBS in the face of numerous uncertainties. The study complements the perspective of patient representatives, providing further insight into the subject matter. This is a valuable contribution as they possess a comprehensive understanding of the medical, psychological, and ethical considerations involved. Patient representatives placed particular emphasis on the advantages of avoiding a diagnostic odyssey and the significance of support systems. The results provide first insights into different views on gNBS in Germany. These views can inform the potential preparation of a gNBS program in Germany, particularly with regard to information and consent requirements. Implications for practice, such as informing and educating parents about gNBS during pregnancy, can be derived from the study.
Collapse
Affiliation(s)
- Elena Sophia Doll
- Ruprecht-Karls University Heidelberg, Heidelberg, Germany
- Institute of Medical Psychology, Heidelberg University Hospital, Heidelberg, Germany
| | - Julia Mahal
- Ruprecht-Karls University Heidelberg, Heidelberg, Germany
- Institute of Medical Psychology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karla Alex
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Section Translational Medical Ethics, Department of Medical Oncology, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Seraina Petra Lerch
- Ruprecht-Karls University Heidelberg, Heidelberg, Germany
- Institute of Medical Psychology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Medical Psychology, University Medicine Greifswald, Greifswald, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Greifswald/Rostock, Greifswald, Germany
| | - Stefan Kölker
- Heidelberg University, Medical Faculty, Center for Child and Adolescent Medicine, Division of Child Neurology and Metabolic Medicine, Heidelberg, Germany
| | - Christian P Schaaf
- Institute of Human Genetics, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- German Centre for Mental Health, Partner Site Heidelberg-Mannheim-Ulm, Mannheim, Germany
| | - Eva C Winkler
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Section Translational Medical Ethics, Department of Medical Oncology, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Beate Ditzen
- Ruprecht-Karls University Heidelberg, Heidelberg, Germany
- Institute of Medical Psychology, Heidelberg University Hospital, Heidelberg, Germany
- German Centre for Mental Health, Partner Site Heidelberg-Mannheim-Ulm, Mannheim, Germany
| |
Collapse
|
2
|
Mu JL, Sun M, Li YL, Li PP, Zou H. Urgent need for newborn targeted sequencing screening technology in Shandong Province, China. World J Pediatr 2025:10.1007/s12519-025-00907-5. [PMID: 40299190 DOI: 10.1007/s12519-025-00907-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/30/2025]
Affiliation(s)
- Jia-Lin Mu
- Jinan Maternal and Child Care Hospital, No.2, Jingsan Road, Jianguo Xiao, Shizhong District, Jinan, China
| | - Meng Sun
- Jinan Maternal and Child Care Hospital, No.2, Jingsan Road, Jianguo Xiao, Shizhong District, Jinan, China
| | - Yu-Lin Li
- Jinan Maternal and Child Care Hospital, No.2, Jingsan Road, Jianguo Xiao, Shizhong District, Jinan, China
| | - Pan-Pan Li
- Jinan Maternal and Child Care Hospital, No.2, Jingsan Road, Jianguo Xiao, Shizhong District, Jinan, China
| | - Hui Zou
- Jinan Maternal and Child Care Hospital, No.2, Jingsan Road, Jianguo Xiao, Shizhong District, Jinan, China.
| |
Collapse
|
3
|
Moody L, Clarke S, Compton M, Hughson-Gill R, Boardman F, Clark C, Holder P, Bonham JR, Chudleigh J. Development of an Online Scenario-Based Tool to Enable Research Participation and Public Engagement in Cystic Fibrosis Newborn Screening: Mixed Methods Study. J Particip Med 2025; 17:e59686. [PMID: 40053726 PMCID: PMC11926439 DOI: 10.2196/59686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/06/2024] [Accepted: 11/27/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Newborn screening aims to identify babies affected by rare but serious genetic conditions. As technology advances, there is the potential to expand the newborn screening program following evaluation of the likely benefits and drawbacks. To inform these decisions, it is important to consider the family experience of screening and the views of the public. Engaging in public dialogue can be difficult. The conditions, screening processes, and associated moral and ethical considerations are complex. OBJECTIVE This study aims to develop a stand-alone online resource to enable a range of stakeholders to understand whether and how next-generation sequencing should be incorporated into the CF screening algorithm. METHODS Around 4 development workshops with policymakers, parents, and other stakeholders informed the design of an interactive activity, including the structure, content, and questions posed. Stakeholders were recruited to take part in the development workshops via purposeful and snowball sampling methods to achieve a diversity of views across roles and organizations, with email invitations sent to representative individuals with lived, clinical, and academic experience related to CF and screening. Ten stakeholders informed the development process including those with lived experience of CF (2/10, 20%), clinicians (2/10, 20%), and representatives from relevant government, charity, and research organizations (6/10, 60%). Vignettes constructed using interview data and translated into scripts were recorded to provide short films to represent and provoke consideration of families' experiences. Participants were recruited (n=6, adults older than 18 years) to test the resulting resource. Study advertisements were circulated via physical posters and digital newsletters to recruit participants who self-identified as having a reading difficulty or having English as a second language. RESULTS An open access online resource, "Cystic Fibrosis Newborn Screening: You Decide," was developed and usability and acceptability tested to provide the "user" (eg, a parent, the general public, or a health care professional) with an interactive scenario-based presentation of the potential outcomes of extended genetic testing, allowing them to visualize the impact on families. This included a learning workbook that explains key concepts and processes. The resulting tool facilitates public engagement with and understanding of complex genetic and screening concepts. CONCLUSIONS Online resources such as the one developed during this work have the potential to help people form considered views and facilitate access to the perspectives of parents and the wider public on genetic testing. These may be otherwise difficult to obtain but are of importance to health care professionals and policymakers. TRIAL REGISTRATION ClinicalTrials.gov NCT06299566; https://clinicaltrials.gov/study/NCT06299566.
Collapse
Affiliation(s)
- Louise Moody
- Centre for Arts, Memory and Communities, Coventry University, Coventry, United Kingdom
| | - Samantha Clarke
- Centre for Arts, Memory and Communities, Coventry University, Coventry, United Kingdom
| | - Matt Compton
- Centre for Arts, Memory and Communities, Coventry University, Coventry, United Kingdom
| | - Rachael Hughson-Gill
- Centre for Arts, Memory and Communities, Coventry University, Coventry, United Kingdom
| | - Felicity Boardman
- Warwick Medical School, Warwick University, Coventry, United Kingdom
| | - Corinna Clark
- Warwick Medical School, Warwick University, Coventry, United Kingdom
| | - Pru Holder
- Florence Nightingale Faculty of Nursing, Midwifery & Palliative Care, King's College London, London, United Kingdom
| | - James R Bonham
- Pharmacy, Diagnostics and Genetics, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom
| | - Jane Chudleigh
- Florence Nightingale Faculty of Nursing, Midwifery & Palliative Care, King's College London, London, United Kingdom
| |
Collapse
|
4
|
Blom M, Soomann M, Soler-Palacín P, Šedivá A, Stray-Pedersen A, Zetterström R, Speckmann C, Gennery AR, van der Burg M. Newborn screening for SCID and severe T lymphocytopenia in Europe. J Allergy Clin Immunol 2025; 155:377-386. [PMID: 39510364 DOI: 10.1016/j.jaci.2024.10.018] [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: 05/31/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
Initiation of newborn screening (NBS) programs in Europe dates back to the 1960s. One of the most recent expansions of NBS programs was the addition of severe combined immunodeficiency (SCID) based on detection of T-cell receptor excision circles (TRECs). In this review, we present an overview of the current situation in Europe. To avoid a biased overview based on only published results, a 37-item survey on TREC-based NBS was sent to representatives of 46 European countries. With a response rate of 83%, we collected data of 38 countries. Seventeen of the 38 European countries that have completed the survey have nationally or regionally implemented TREC-based NBS. The survey results emphasize similarities and differences as well as common practices and challenges in TREC-based NBS. Because TRECs are a general surrogate marker for severe T lymphocytopenia, conditions other than SCID are also identified. Therefore, the initial definition of the target disease as "SCID" might need to be reconsidered and extended to "SCID and severe T lymphocytopenia." Even though complete harmonization of TREC-based NBS programs across Europe will remain challenging, collaboration and close partnerships will help in the move toward universal TREC-based screening for all newborns, resulting in more infants with SCID and severe T lymphocytopenia being detected each year.
Collapse
Affiliation(s)
- Maartje Blom
- Laboratory for Paediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarja Soomann
- Division of Immunology and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Pere Soler-Palacín
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Children's Hospital, Vall d'Hebron Barcelona Hospital, Barcelona, Spain
| | - Anna Šedivá
- Department of Immunology, 2nd Faculty of Medicine Charles University and Motol University Hospital, Prague, Czech Republic
| | - Asbjørg Stray-Pedersen
- Norwegian National Unit for Newborn Screening, Division of Pediatric and Adolescent Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Rolf Zetterström
- Center for Inherited Metabolic Diseases, Karolinska University Hospital and Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Carsten Speckmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, and Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Mirjam van der Burg
- Laboratory for Paediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
5
|
Drole Torkar A, Klinc A, Remec ZI, Rankovic B, Bartolj K, Bertok S, Colja S, Cuk V, Debeljak M, Kozjek E, Repic Lampret B, Mlinaric M, Mohar Hajnsek T, Perko D, Stajer K, Tesovnik T, Trampuz D, Ulaga B, Kovac J, Battelino T, Zerjav Tansek M, Groselj U. Sudden Death of a Four-Day-Old Newborn Due to Mitochondrial Trifunctional Protein/Long-Chain 3-Hydroxyacyl-CoA Dehydrogenase Deficiencies and a Systematic Literature Review of Early Deaths of Neonates with Fatty Acid Oxidation Disorders. Int J Neonatal Screen 2025; 11:9. [PMID: 39982343 PMCID: PMC11843868 DOI: 10.3390/ijns11010009] [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: 11/05/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
Mitochondrial trifunctional protein (MTP) and long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiencies have been a part of the Slovenian newborn screening (NBS) program since 2018. We describe a case of early lethal presentation of MTPD/LCHADD in a term newborn. The girl was born after an uneventful pregnancy and delivery, and she was discharged home at the age of 3 days, appearing well. At the age of 4 days, she was found without signs of life. Resuscitation was not successful. The NBS test performed using tandem mass spectrometry (MS/MS) showed a positive screen for MTPD/LCHADD. Genetic analysis performed on a dried blood spot (DBS) sample identified two heterozygous variants in the HADHA gene: a nucleotide duplication introducing a premature termination codon (p.Arg205Ter) and a nucleotide substitution (p.Glu510Gln). Post-mortem studies showed massive macro-vesicular fat accumulation in the liver and, to a smaller extent, in the heart, consistent with MTPD/LCHADD. A neonatal acute cardiac presentation resulting in demise was suspected. We conducted a systematic literature review of early neonatal deaths within 14 days postpartum attributed to confirmed fatty acid oxidation disorders (FAODs), which are estimated to account for 5% of sudden infant deaths. We discuss the pitfalls of the NBS for MTPD/LCHADD.
Collapse
Affiliation(s)
- Ana Drole Torkar
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, Ljubljana University Medical Center, Bohoriceva 20, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
| | - Ana Klinc
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
| | - Ziga Iztok Remec
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Branislava Rankovic
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Klara Bartolj
- Novo Mesto General Hospital, Smihelska cesta 1, 8000 Novo Mesto, Slovenia
| | - Sara Bertok
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, Ljubljana University Medical Center, Bohoriceva 20, 1000 Ljubljana, Slovenia
| | - Sara Colja
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Vanja Cuk
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Marusa Debeljak
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Eva Kozjek
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Barbka Repic Lampret
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Matej Mlinaric
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, Ljubljana University Medical Center, Bohoriceva 20, 1000 Ljubljana, Slovenia
| | | | - Daša Perko
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Katarina Stajer
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, Ljubljana University Medical Center, Bohoriceva 20, 1000 Ljubljana, Slovenia
| | - Tine Tesovnik
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Domen Trampuz
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Blanka Ulaga
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Jernej Kovac
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, Ljubljana University Medical Center, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, Ljubljana University Medical Center, Bohoriceva 20, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
| | - Mojca Zerjav Tansek
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, Ljubljana University Medical Center, Bohoriceva 20, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
| | - Urh Groselj
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, Ljubljana University Medical Center, Bohoriceva 20, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (A.K.); (J.K.)
| |
Collapse
|
6
|
Tagliacollo VA, Tan M, Reis RE, Gaia R, Carrijo V, Ranuzi M, Craig JM, Albert JS. Time-calibrated phylogeny of neotropical freshwater fishes. FRONTIERS IN BIOINFORMATICS 2024; 4:1433995. [PMID: 39711580 PMCID: PMC11659281 DOI: 10.3389/fbinf.2024.1433995] [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: 05/16/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Neotropical Freshwater Fish (NFF) fauna exhibits the greatest phenotypic disparity and species richness among all continental aquatic vertebrate faunas, with more than 6,345 species distributed across the mostly tropical regions of Central and South America. The last two decades have seen a proliferation of molecular phylogenies, often at the species level, covering almost all 875 valid NFF genera. This study presents the most comprehensive genome-wide, time-calibrated phylogenetic hypothesis of NFF species to date, based on DNA sequences generated over decades through the collaborative efforts of the multinational ichthyological research community. Our purpose is to build and curate an extensive molecular dataset allowing researchers to evaluate macroevolutionary hypotheses in the NFF while facilitating continuous refinement and expansion. Using thousands of DNA sequences from dozens of studies, we compiled a supermatrix of 51 markers for 5,984 taxa, representing 3,167 NFF species. Based on this dataset, we built the most species-rich time-calibrated phylogeny of the NFF taxa to date, summarizing the collective efforts of the ichthyological research community since the midpoint of the last century. We provide a summary review of this remarkable evolutionary history and hope this dataset provides a framework for forthcoming studies of the NFF fauna, documenting compelling, emergent patterns in the world's most diverse continental vertebrate fauna.
Collapse
Affiliation(s)
- Victor A. Tagliacollo
- Institute of Biology, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Milton Tan
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Roberto E. Reis
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ronielson Gaia
- Institute of Biology, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Virgilio Carrijo
- Institute of Biology, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Marcelo Ranuzi
- Institute of Biology, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Jack M. Craig
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, United States
| | - James S. Albert
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA, United States
| |
Collapse
|
7
|
Tutty E, Archibald AD, Downie L, Gaff C, Lunke S, Vears DF, Stark Z, Best S. Key informant perspectives on implementing genomic newborn screening: a qualitative study guided by the Action, Actor, Context, Target, Time framework. Eur J Hum Genet 2024; 32:1599-1605. [PMID: 38907005 PMCID: PMC11606939 DOI: 10.1038/s41431-024-01650-7] [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: 12/13/2023] [Revised: 05/01/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
Newborn screening (NBS) programmes are highly successful, trusted, public health interventions. Genomic sequencing offers the opportunity to increase the benefits of NBS by screening infants for a greater number and variety of childhood-onset conditions. This study aimed to describe who needs to do what, when, and for whom to deliver genomic newborn screening (gNBS) and capture perceived implementation barriers and enablers. 'Key informants' (individuals involved in the delivery of NBS) were interviewed. The Actor, Action, Context, Time and Target framework guided data collection and analysis. Participants (N = 20) identified new Actions required to deliver gNBS (educating healthcare providers, longitudinal psychosocial support), NBS Actions needing modification (obtaining consent) and NBS Actions that could be adopted for gNBS (prompt referral pathways). Obtaining consent in a prenatal Context was a source of some disagreement. The Time to disclose high chance results was raised as a key consideration in gNBS programme design. Genetic counsellors were identified as key Actors in results management, but workforce limitations may be a barrier. Online decision support tools were an enabler to offering gNBS. The implementation of gNBS will require behaviour changes from HCPs delivering NBS. Findings can inform how to deliver gNBS at population-scale.
Collapse
Affiliation(s)
- Erin Tutty
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Alison D Archibald
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
- Victorian Clinical Genetics Service, Melbourne, VIC, Australia
| | - Lilian Downie
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
- Victorian Clinical Genetics Service, Melbourne, VIC, Australia
| | - Clara Gaff
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Genomics Health Alliance, Melbourne, VIC, Australia
- WEHI, Melbourne, VIC, 3052, Australia
| | - Sebastian Lunke
- Victorian Clinical Genetics Service, Melbourne, VIC, Australia
- Australian Genomics, Melbourne, VIC, Australia
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Danya F Vears
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Law School, University of Melbourne, Melbourne, VIC, Australia
| | - Zornitza Stark
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
- Victorian Clinical Genetics Service, Melbourne, VIC, Australia
- Australian Genomics, Melbourne, VIC, Australia
| | - Stephanie Best
- Australian Genomics, Melbourne, VIC, Australia.
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- School of Health Sciences, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
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
|
9
|
Chen C, Zhang C, Wu DW, Wang BY, Xiao R, Huang XL, Yang X, Gao ZG, Yang RL. Comprehensive newborn screening for severe combined immunodeficiency, X-linked agammaglobulinemia, and spinal muscular atrophy: the Chinese experience. World J Pediatr 2024; 20:1270-1282. [PMID: 39500858 PMCID: PMC11634924 DOI: 10.1007/s12519-024-00846-7] [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: 01/25/2024] [Accepted: 09/18/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Newborn screening (NBS) for severe combined immunodeficiency (SCID), X-linked agammaglobulinemia (XLA), and spinal muscular atrophy (SMA) enables early diagnosis and intervention, significantly improving patient outcomes. Advances in real-time polymerase chain reaction (PCR) technology have been instrumental in facilitating their inclusion in NBS programs. METHODS We employed multiplex real-time PCR to simultaneously detect T-cell receptor excision circles (TRECs), kappa-deleting recombination excision circles (KRECs), and the absence of the survival motor neuron (SMN) 1 gene in dried blood spots from 103,240 newborns in Zhejiang Province, China, between July 2021 and December 2022. RESULTS Of all the samples, 122 were requested further evaluation. After flow cytometry evaluation and/or genetic diagnostics, we identified one patient with SCID, two patients with XLA, nine patients with SMA [one of whom also had Wiskott-Aldrich Syndrome (WAS)], and eight patients with other medical conditions. The positive predictive values (PPVs) of NBS for SCID, XLA, and SMA were 2.44%, 2.78%, and 100%, respectively. The estimated prevalence rates in the Chinese population were 1 in 103,240 for SCID, 1 in 51,620 for XLA, and 1 in 11,471 for SMA. CONCLUSION This study represents the first large-scale screening in mainland China using a TREC/KREC/SMN1 multiplex assay, providing valuable epidemiological data. Our findings suggest that this multiplex assay is an effective screening method for SCID, XLA, and SMA, potentially supporting the universal implementation of NBS programs across China.
Collapse
Affiliation(s)
- Chi Chen
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chao Zhang
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ding-Wen Wu
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Bing-Yi Wang
- National Engineering Laboratory for Key Technology of Birth Defect Control and Prevention, Screening and Diagnostic R and D Center, Hangzhou, China
| | - Rui Xiao
- National Engineering Laboratory for Key Technology of Birth Defect Control and Prevention, Screening and Diagnostic R and D Center, Hangzhou, China
| | - Xiao-Lei Huang
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xin Yang
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhi-Gang Gao
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Ru-Lai Yang
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
10
|
Alhusseini N, Almuhanna Y, Alabduljabbar L, Alamri S, Altayeb M, Askar G, Alsaadoun N, Ateq K, AlEissa MM. International Newborn Screening: Where Are We in Saudi Arabia? J Epidemiol Glob Health 2024; 14:638-644. [PMID: 38922570 PMCID: PMC11442708 DOI: 10.1007/s44197-024-00263-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Newborn screening (NBS) programs are believed to play an important role in the decrease of infant mortality rates in many countries. This is achieved through offering early detection and treatment of many genetic as well as metabolic disorders prior to the onset of symptoms. Our paper examines NBS across seven diverse nations: Saudi Arabia, the United States, Japan, Singapore, Canada, Australia, and the United Kingdom. This paper discusses the diseases screened for by each country, latest additions, as well as future recommendations, when applicable. Employing a comparative approach, we conducted a comprehensive review of the most recent published literature on NBS programs in each country and subsequently examined their latest implemented NBS guidelines as outlined on their respective official government health sector websites. We then reviewed the economic feasibility of each of these programs and factors that affect implementation and overall benefit. While all six countries employ well-developed programs, variations are observed. Those variations are mainly attributed to disparities in access, resource scarcity, financial availability, as well as ethical and cultural considerations. From a local perspective, we recommend conducting further population-based studies to assess the epidemiological data in relation to the disease burden on the country's economy. Moreover, we recommend updating national and international guidelines to contain a more comprehensive approach on policies, operation, and sustainability to deliver a service through the lens of value-based healthcare.
Collapse
Affiliation(s)
| | - Yara Almuhanna
- Medical School, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Soaad Alamri
- Medical School, Alfaisal University, Riyadh, Saudi Arabia
| | - Maryam Altayeb
- Medical School, Alfaisal University, Riyadh, Saudi Arabia
| | - Ghadi Askar
- Medical School, Alfaisal University, Riyadh, Saudi Arabia
| | - Noor Alsaadoun
- Medical School, Alfaisal University, Riyadh, Saudi Arabia
| | - Khadijah Ateq
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Science, Riyadh, Saudi Arabia.
| | - Mariam M AlEissa
- Medical School, Alfaisal University, Riyadh, Saudi Arabia
- Public Health Authority, Public health Lab, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Tresbach RH, Sperb-Ludwig F, Ligabue-Braun R, Bitencourt FHD, Tonon T, Souza CFMD, Poswar FDO, Leite MEDQ, Amorim T, Porta G, Seda Neto J, Miura IK, Steiner CE, Martins AM, Pessoa ALS, Ribeiro EM, Schwartz IVD. Maple syrup urine disease diagnosis in Brazilian patients by massive parallel sequencing. Mol Genet Metab 2024; 143:108569. [PMID: 39270351 DOI: 10.1016/j.ymgme.2024.108569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Biallelic pathogenic variants cause maple syrup urine disease (MSUD) in one of the branched-chain α-keto acid dehydrogenase (BCKDH) complex genes (BCKDHA, BCKDHB, DBT, DLD, and PPM1K) leading to the accumulation of leucine, isoleucine, and valine. This study aimed to perform a molecular diagnosis of Brazilian patients with MSUD using gene panels and massive parallel sequencing. Eighteen Brazilian patients with a biochemical diagnosis of MSUD were analyzed by massive parallel sequencing in the Ion PGM Torrent Server using a gene panel with the BCKDHA, BCKDHB, and DBT genes. The American College of Medical Genetics and Genomics guidelines were used to determine variant pathogenicity. Thirteen patients had both variants found by massive parallel sequencing, whereas 3 patients had only one variant found. In 2 patients, the variants were not found by this analysis. These 5 patients required additional Sanger sequencing to confirm their genotype. Twenty-five pathogenic variants were identified in the 3 MSUD-related genes (BCKDHA, BCKDHB, and DBT). Most variants were present in the BCKDHB gene, and no common variants were found. Nine novel variants were observed: c.922 A > G, c.964C > A, and c.1237 T > C in the BCKDHA gene; and c.80_90dup, c.384delA, c.478 A > T, c.528C > G, c.977 T > C, and c.1039-2 A > G in the BCKDHB gene. All novel variants were classified as pathogenic. Molecular modeling of the novel variants indicated that the binding of monomers was affected in the BCKDH complex tetramer, which could lead to a change in the stability and activity of the enzyme. Massive parallel sequencing with targeted gene panels seems to be a cost-effective method that can provide a molecular diagnosis of MSUD.
Collapse
Affiliation(s)
- Rafael Hencke Tresbach
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fernanda Sperb-Ludwig
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Rodrigo Ligabue-Braun
- Graduate Program in Biological Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil; Department of Pharmacosciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Fernanda Hendges de Bitencourt
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Tássia Tonon
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carolina Fischinger Moura de Souza
- Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Fabiano de Oliveira Poswar
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Maria Efigênia de Queiroz Leite
- Newborn Screening Reference Center - Association of Parents and Friends of People with Disabilities (APAE), Salvador, BA, Brazil
| | - Tatiana Amorim
- Newborn Screening Reference Center - Association of Parents and Friends of People with Disabilities (APAE), Salvador, BA, Brazil
| | - Gilda Porta
- Pedro de Alcântara Children's Institute - Hospital das Clínicas, Medical School, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - João Seda Neto
- Department of Hepatology and Liver Transplantation, Hospital Sírio-Libanês, São Paulo, SP, Brazil
| | - Irene Kazumi Miura
- Department of Hepatology and Liver Transplantation, Hospital Sírio-Libanês, São Paulo, SP, Brazil
| | - Carlos Eduardo Steiner
- Department of Translational Medicine, School of Medical Sciences, Universidade Estadual de Campinas (Unicamp), Campinas, SP, Brazil
| | - Ana Maria Martins
- Reference Center for Inborn Errors of Metabolism, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
| | - André Luiz Santos Pessoa
- Hospital Infantil Albert Sabin, Fortaleza, CE, Brazil; Department of Pediatrics, Universidade Estadual do Ceará (UECE), Fortaleza, CE, Brazil
| | | | - Ida Vanessa Doederlein Schwartz
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Clinical Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; InRaras, National Institute of Rare Diseases, Brazil
| |
Collapse
|
12
|
Kumarasamy G, Khairiz K, Chang WL, Aye TT, Ali A. Paving the way in implementation of SCID newborn screening in developing nations: feasibility study and strategies to move forward in Malaysia. Front Immunol 2024; 15:1400247. [PMID: 38983864 PMCID: PMC11231083 DOI: 10.3389/fimmu.2024.1400247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
Early diagnosis and effective management of Primary immunodeficiency diseases (PIDs), particularly severe combined immunodeficiency (SCID), play a crucial role in minimizing associated morbidities and mortality. Newborn screening (NBS) serves as a valuable tool in facilitating these efforts. Timely detection and diagnosis are essential for swiftly implementing isolation measures and ensuring prompt referral for definitive treatment, such as allogeneic hematopoietic stem cell transplantation. The utilization of comprehensive protocols and screening assays, including T cell receptor excision circles (TREC) and kappa-deleting recombination excision circles (KREC), is essential in facilitating early diagnosis of SCID and other PIDs, but their successful application requires clinical expertise and proper implementation strategy. Unfortunately, a notable challenge arises from insufficient funding for the treatment of PIDs. To address these issues, a collaborative approach is imperative, involving advancements in technology, a well-functioning healthcare system, and active engagement from stakeholders. The integration of these elements is essential for overcoming the existing challenges in NBS for PIDs. By fostering synergy between technology providers, healthcare professionals, and governmental stakeholders, we can enhance the efficiency and effectiveness of early diagnosis and intervention, ultimately improving outcomes for individuals with PIDs.
Collapse
Affiliation(s)
- Gaayathri Kumarasamy
- Arcadia Life Sciences, Hive 5, Taman Teknologi Malaysian Research Accelerator for Technology & Innovation (MRANTI), Bukit Jalil, Kuala Lumpur, Malaysia
| | - Khayrin Khairiz
- Arcadia Life Sciences, Hive 5, Taman Teknologi Malaysian Research Accelerator for Technology & Innovation (MRANTI), Bukit Jalil, Kuala Lumpur, Malaysia
| | - Wai Leng Chang
- Department of Pediatric, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Research Center, Hospital Tunku Ampuan Besar Tuanku Aishah Rohani, Universiti Kebangsaan Malaysia (UKM) Specialist Children's Hospital, Kuala Lumpur, Malaysia
| | - Thin Thin Aye
- Arcadia Life Sciences, Hive 5, Taman Teknologi Malaysian Research Accelerator for Technology & Innovation (MRANTI), Bukit Jalil, Kuala Lumpur, Malaysia
| | - Adli Ali
- Department of Pediatric, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Research Center, Hospital Tunku Ampuan Besar Tuanku Aishah Rohani, Universiti Kebangsaan Malaysia (UKM) Specialist Children's Hospital, Kuala Lumpur, Malaysia
- Institute of IR4.0, Universiti Kebangsaan Malaysia, Bangi, Malaysia
- Infection and Immunology Health and Advanced Medicine Cluster, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
de Frutos F, Caraballo Ramos I, Martínez Chaves V, Corral Azor AM, Berchíd Débdi MS, García-Pavía P. Results of a population screening program for hereditary transthyretin amyloidosis. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024; 77:498-500. [PMID: 38220055 DOI: 10.1016/j.rec.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024]
Affiliation(s)
- Fernando de Frutos
- Unidad de Insuficiencia Cardiaca y Cardiopatías Familiares, Servicio de Cardiología, Hospital Universitario Puerta de Hierro, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | | | | | | | | | - Pablo García-Pavía
- Unidad de Insuficiencia Cardiaca y Cardiopatías Familiares, Servicio de Cardiología, Hospital Universitario Puerta de Hierro, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
14
|
Linga BG, Mohammed SGAA, Farrell T, Rifai HA, Al-Dewik N, Qoronfleh MW. Genomic Newborn Screening for Pediatric Cancer Predisposition Syndromes: A Holistic Approach. Cancers (Basel) 2024; 16:2017. [PMID: 38893137 PMCID: PMC11171256 DOI: 10.3390/cancers16112017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
As next-generation sequencing (NGS) has become more widely used, germline and rare genetic variations responsible for inherited illnesses, including cancer predisposition syndromes (CPSs) that account for up to 10% of childhood malignancies, have been found. The CPSs are a group of germline genetic disorders that have been identified as risk factors for pediatric cancer development. Excluding a few "classic" CPSs, there is no agreement regarding when and how to conduct germline genetic diagnostic studies in children with cancer due to the constant evolution of knowledge in NGS technologies. Various clinical screening tools have been suggested to aid in the identification of individuals who are at greater risk, using diverse strategies and with varied outcomes. We present here an overview of the primary clinical and molecular characteristics of various CPSs and summarize the existing clinical genomics data on the prevalence of CPSs in pediatric cancer patients. Additionally, we discuss several ethical issues, challenges, limitations, cost-effectiveness, and integration of genomic newborn screening for CPSs into a healthcare system. Furthermore, we assess the effectiveness of commonly utilized decision-support tools in identifying patients who may benefit from genetic counseling and/or direct genetic testing. This investigation highlights a tailored and systematic approach utilizing medical newborn screening tools such as the genome sequencing of high-risk newborns for CPSs, which could be a practical and cost-effective strategy in pediatric cancer care.
Collapse
Affiliation(s)
- BalaSubramani Gattu Linga
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
- Translational and Precision Medicine Research, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
| | | | - Thomas Farrell
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
| | - Hilal Al Rifai
- Neonatal Intensive Care Unit (NICU), Newborn Screening Unit, Department of Pediatrics and Neonatology, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
| | - Nader Al-Dewik
- Department of Research, Women’s Wellness and Research Center, Hamad Medical Corporation (HMC), P.O. Box 3050, Doha 0974, Qatar
- Translational and Precision Medicine Research, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
- Neonatal Intensive Care Unit (NICU), Newborn Screening Unit, Department of Pediatrics and Neonatology, Women’s Wellness and Research Center (WWRC), Hamad Medical Corporation (HMC), Doha 0974, Qatar
- Genomics and Precision Medicine (GPM), College of Health & Life Science (CHLS), Hamad Bin Khalifa University (HBKU), Doha 0974, Qatar
- Faculty of Health and Social Care Sciences, Kingston University and St George’s University of London, Kingston upon Thames, Surrey, London KT1 2EE, UK
| | - M. Walid Qoronfleh
- Healthcare Research & Policy Division, Q3 Research Institute (QRI), Ann Arbor, MI 48197, USA
| |
Collapse
|
15
|
Beigh M, Vagher J, Codden R, Maese LD, Cook S, Gammon A. Newborn Screening for Li-Fraumeni Syndrome: Patient Perspectives. RESEARCH SQUARE 2024:rs.3.rs-4351728. [PMID: 38798617 PMCID: PMC11118696 DOI: 10.21203/rs.3.rs-4351728/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Li-Fraumeni syndrome (LFS) is an inherited cancer predisposition syndrome with an estimated prevalence of 1 in 3,000-5,000 individuals. LFS poses a significant cancer risk throughout the lifespan, with notable cancer susceptibility in childhood. Despite being predominantly inherited, up to 20% of cases arise de novo. Surveillance protocols facilitate the reduction of mortality and morbidity through early cancer detection. While newborn screening (NBS) has proven effective in identifying newborns with rare genetic conditions, even those occurring as rarely as 1 in 185,000, its potential for detecting inherited cancer predispositions remains largely unexplored. Methods This survey-based study investigates perspectives toward NBS for LFS among individuals with and parents of children with LFS receiving care at single comprehensive cancer center in the U.S. Results All participants unanimously supported NBS for LFS (n = 24). Reasons included empowerment (83.3%), control (66.7%), and peace of mind (54.2%), albeit with concerns about anxiety (62.5%) and devastation (50%) related to receiving positive results. Participants endorsed NBS as beneficial for cancer detection and prevention (91.7%), research efforts (87.5%), and family planning (79.2%) but voiced apprehensions about the financial cost of cancer surveillance (62.5%), emotional burdens (62.5%), and insurance coverage and discrimination (54.2%). Approximately 83% of respondents believed that parental consent should be required to screen newborns for LFS. Conclusion This study revealed strong support for NBS for LFS despite the recognition of various perceived benefits and risks. These findings underscore the complex interplay between clinical, psychosocial, and ethical factors in considering NBS for LFS from the perspective of the LFS community.
Collapse
Affiliation(s)
| | | | - Rachel Codden
- Division of Epidemiology, Department of Internal Medicine, University of Utah
| | | | - Sabina Cook
- Utah Department of Health and Human Services
| | | |
Collapse
|
16
|
Solis L, Van Coillie S, Bonham JR, Hauck F, Hammarström L, Staal FJT, Lim B, Pergent M, Prévot J. Newborn Screening Today and Tomorrow: A Brief Report from the International Primary Immunodeficiencies Congress. Int J Neonatal Screen 2024; 10:30. [PMID: 38651395 PMCID: PMC11036269 DOI: 10.3390/ijns10020030] [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: 02/22/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 04/25/2024] Open
Abstract
This article presents the report of the session on "Newborn Screening for Primary Immunodeficiencies-Now What?" organised during the International Primary Immunodeficiency Congress (IPIC) held in November 2023. This clinical conference was organised by the International Patient Organisation for Primary Immunodeficiencies (IPOPI), the global patient organisation advocating for primary immunodeficiencies (PIDs) in patients. The session aimed at exploring the advances in newborn screening (NBS) for severe combined immunodeficiency, starting with the common practice and inserting the discussion into the wider perspective of genomics whilst taking into consideration the ethical aspects of screening as well as incorporating families and the public into the discussions, so as to ensure that NBS for treatable rare disorders continues to be one of the major public health advances of the 20th century.
Collapse
Affiliation(s)
- Leire Solis
- IPOPI, BE-1050 Brussels, Belgium; (L.S.); (S.V.C.); (B.L.); (M.P.)
| | | | - James R. Bonham
- International Society of Neonatal Screening, Reigerskamp 273, 3607 HP Maarssen, The Netherlands;
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, Ludwig-Maximilians-Universität München, DE-80337 Munich, Germany;
| | - Lennart Hammarström
- Department of Medical Biophysics and Biochemistry, Karolinska Instituet, SE-17177 Stockholm, Sweden;
| | - Frank J. T. Staal
- Departments of Immunology and Pediatrics, Leiden University Medical School, 2300 RC Leiden, The Netherlands;
| | - Bruce Lim
- IPOPI, BE-1050 Brussels, Belgium; (L.S.); (S.V.C.); (B.L.); (M.P.)
| | - Martine Pergent
- IPOPI, BE-1050 Brussels, Belgium; (L.S.); (S.V.C.); (B.L.); (M.P.)
| | - Johan Prévot
- IPOPI, BE-1050 Brussels, Belgium; (L.S.); (S.V.C.); (B.L.); (M.P.)
| |
Collapse
|
17
|
Gupte SA, Kurup M, Jangam SM, Arora P, Shah SS. Newborn Genetic Screening: Significance in Early Diagnosis of an Infant with Mitochondrial DNA Depletion Syndrome-6. J Obstet Gynaecol India 2024; 74:176-178. [PMID: 38707873 PMCID: PMC11065833 DOI: 10.1007/s13224-023-01770-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 05/07/2023] [Indexed: 05/07/2024] Open
Affiliation(s)
- Sanjay A. Gupte
- Gupte Hospital, Postgraduate Institution and Centre of Research in Reproduction, Pune, Maharashtra India
- Greenarray Genomic Research and Solutions, a division of Accurate Diagnostics Pvt. Ltd., Kothrud, Pune, Maharashtra India
| | - Manju Kurup
- Greenarray Genomic Research and Solutions, a division of Accurate Diagnostics Pvt. Ltd., Kothrud, Pune, Maharashtra India
| | - Shweta M. Jangam
- Gupte Hospital, Postgraduate Institution and Centre of Research in Reproduction, Pune, Maharashtra India
| | - Preeti Arora
- Greenarray Genomic Research and Solutions, a division of Accurate Diagnostics Pvt. Ltd., Kothrud, Pune, Maharashtra India
| | - Sarjan S. Shah
- Greenarray Genomic Research and Solutions, a division of Accurate Diagnostics Pvt. Ltd., Kothrud, Pune, Maharashtra India
| |
Collapse
|
18
|
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
|
19
|
Vela-Amieva M, Alcántara-Ortigoza MA, González-del Angel A, Ibarra-González I, Fernández-Hernández L, Guillén-López S, López-Mejía L, Fernández-Lainez C. In Silico Structural Protein Evaluation of the Phenylalanine Hydroxylase p.(Tyr77His) Variant Associated with Benign Hyperphenylalaninemia as Identified through Mexican Newborn Screening. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1865. [PMID: 38136067 PMCID: PMC10742057 DOI: 10.3390/children10121865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023]
Abstract
Hyperphenylalaninemia (HPA), which includes phenylketonuria (PKU), is a genetic autosomal recessive disorder arising from a deficiency in the enzyme named phenylalanine hydroxylase (PAH). Affected patients can experience severe and irreversible neurological impairments when phenylalanine (Phe) blood concentration exceeds 360 μmol/L (6 mg/dL). Here, we describe a female HPA patient who was born in Mexico to Cuban non-consanguineous parents and identified by newborn screening, and who bears the previously unreported PAH NM_000277.3(PAH):c.[229T>C];[1222C>T] or p.[Tyr77His];[Arg408Trp] genotype. At diagnosis, the patient showed a Phe blood level of 321 μmol/L (5.3 mg/dL), indicative of mild HPA. Neither of the PAH variants found in this patient had been previously reported in the mutational PAH spectrum of the Mexican population. The c.229T>C or p.(Tyr77His) PAH variant was previously related to mild HPA in the Swedish population. Our in silico structural analysis and molecular docking showed that mutated His 77 residue is located in the allosteric site of PAH at the interface of the two monomers. The PDBsum in silico tool predicted that this variant would cause minimal structural disturbance of the protein interface in the presence of Phe at the allosteric site. Docking studies revealed that these structural changes might be attenuated by the allosteric effect of Phe. Given the classic PKU phenotype conditioned by the "Celtic" or c.[1222C>T] or p.(Arg408Trp) PAH variant, which is the second variant in this patient, we propose that p.(Tyr77His) has a hypomorphic feature that could explain her mild HPA phenotype. Our results show the importance of following up on cases detected by NBS and the value of genetic studies and in silico tools that aid in the establishment of correct therapeutic strategies.
Collapse
Affiliation(s)
- Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico; (M.V.-A.); (S.G.-L.); (L.L.-M.)
| | - Miguel Angel Alcántara-Ortigoza
- Laboratorio de Biología Molecular, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico; (M.A.A.-O.); (A.G.-d.A.); (L.F.-H.)
- Centro de Alta Especialidad en Genética Humana DNA-GEN S.C., Ciudad de México 14070, Mexico
| | - Ariadna González-del Angel
- Laboratorio de Biología Molecular, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico; (M.A.A.-O.); (A.G.-d.A.); (L.F.-H.)
- Centro de Alta Especialidad en Genética Humana DNA-GEN S.C., Ciudad de México 14070, Mexico
| | - Isabel Ibarra-González
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas UNAM, Ciudad de México 04510, Mexico;
| | - Liliana Fernández-Hernández
- Laboratorio de Biología Molecular, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico; (M.A.A.-O.); (A.G.-d.A.); (L.F.-H.)
| | - Sara Guillén-López
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico; (M.V.-A.); (S.G.-L.); (L.L.-M.)
| | - Lizbeth López-Mejía
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico; (M.V.-A.); (S.G.-L.); (L.L.-M.)
| | - Cynthia Fernández-Lainez
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico; (M.V.-A.); (S.G.-L.); (L.L.-M.)
| |
Collapse
|
20
|
Szalai C. Arguments for and against the whole-genome sequencing of newborns. Am J Transl Res 2023; 15:6255-6263. [PMID: 37969196 PMCID: PMC10641337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/11/2023] [Indexed: 11/17/2023]
Abstract
Recent decades have brought enormous progress in both genetics and genomics, as well as in information technology (IT). The sequence of the human genome is now known, and although our knowledge is far from complete, great progress has been made in understanding how the genome works. With the developments in storage capacity, artificial intelligence, and learning algorithms, we are now able to learn and interpret complex systems such as the human genome in a very short time. Perhaps the most important goal of learning about the human genome is to understand diseases better: how they develop; how their processes can be prevented or slowed down; and after diseases have developed, how they can be cured or their symptoms alleviated. The vast majority of diseases have a genetic background, i.e., genes, sequence variations, and gene-gene interactions play a role in most diseases to a greater or lesser extent. Accordingly, the first step is to discover which genes, or genomic variants, cause or contribute to the development of a particular disease in a given patient. Given that an individual's genome remains virtually unchanged throughout their life (with one or two exceptions, such as in the case of cancer, which is caused by somatic mutations), it might be considered advantageous to sequence the genome of every person at birth. In this paper, we set out to show the possible benefits of sequencing the entire genome of every human being at birth, while also discussing the main arguments against it.
Collapse
Affiliation(s)
- Csaba Szalai
- Department of Genetics, Cell and Immunobiology, Semmelweis University1089 Budapest, Hungary
- Heim Pál Children’s Hospital1089 Budapest, Hungary
| |
Collapse
|
21
|
Barseghyan H, Pang AWC, Clifford B, Serrano MA, Chaubey A, Hastie AR. Comparative Benchmarking of Optical Genome Mapping and Chromosomal Microarray Reveals High Technological Concordance in CNV Identification and Additional Structural Variant Refinement. Genes (Basel) 2023; 14:1868. [PMID: 37895217 PMCID: PMC10667989 DOI: 10.3390/genes14101868] [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: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
The recommended practice for individuals suspected of a genetic etiology for disorders including unexplained developmental delay/intellectual disability (DD/ID), autism spectrum disorders (ASD), and multiple congenital anomalies (MCA) involves a genetic testing workflow including chromosomal microarray (CMA), Fragile-X testing, karyotype analysis, and/or sequencing-based gene panels. Since genomic imbalances are often found to be causative, CMA is recommended as first tier testing for many indications. Optical genome mapping (OGM) is an emerging next generation cytogenomic technique that can detect not only copy number variants (CNVs), triploidy and absence of heterozygosity (AOH) like CMA, but can also define the location of duplications, and detect other structural variants (SVs), including balanced rearrangements and repeat expansions/contractions. This study compares OGM to CMA for clinically reported genomic variants, some of these samples also have structural characterization by fluorescence in situ hybridization (FISH). OGM was performed on IRB approved, de-identified specimens from 55 individuals with genomic abnormalities previously identified by CMA (61 clinically reported abnormalities). SVs identified by OGM were filtered by a control database to remove polymorphic variants and against an established gene list to prioritize clinically relevant findings before comparing with CMA and FISH results. OGM results showed 100% concordance with CMA findings for pathogenic variants and 98% concordant for all pathogenic/likely pathogenic/variants of uncertain significance (VUS), while also providing additional insight into the genomic structure of abnormalities that CMA was unable to provide. OGM demonstrates equivalent performance to CMA for CNV and AOH detection, enhanced by its ability to determine the structure of the genome. This work adds to an increasing body of evidence on the analytical validity and ability to detect clinically relevant abnormalities identified by CMA. Moreover, OGM identifies translocations, structures of duplications and complex CNVs intractable by CMA, yielding additional clinical utility.
Collapse
Affiliation(s)
- Hayk Barseghyan
- Bionano, San Diego, CA 92121, USA; (H.B.); (A.W.C.P.); (B.C.); (A.C.)
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA
- Genomics and Precision Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | | | - Benjamin Clifford
- Bionano, San Diego, CA 92121, USA; (H.B.); (A.W.C.P.); (B.C.); (A.C.)
| | | | - Alka Chaubey
- Bionano, San Diego, CA 92121, USA; (H.B.); (A.W.C.P.); (B.C.); (A.C.)
| | - Alex R. Hastie
- Bionano, San Diego, CA 92121, USA; (H.B.); (A.W.C.P.); (B.C.); (A.C.)
| |
Collapse
|
22
|
Vianey-Saban C, Guffon N, Fouilhoux A, Acquaviva C. Fifty years of research on mitochondrial fatty acid oxidation disorders: The remaining challenges. J Inherit Metab Dis 2023; 46:848-873. [PMID: 37530674 DOI: 10.1002/jimd.12664] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
Since the identification of the first disorder of mitochondrial fatty acid oxidation defects (FAOD) in 1973, more than 20 defects have been identified. Although there are some differences, most FAOD have similar clinical signs, which are mainly due to energy depletion and toxicity of accumulated metabolites. However, some of them have an unusual clinical phenotype or specific clinical signs. This manuscript focuses on what we have learnt so far on the pathophysiology of these disorders, which present with clinical signs that are not typical of categorical FAOD. It also highlights that some disorders have not yet been identified and tries to make assumptions to explain why. It also deals with new treatments under consideration in FAOD, including triheptanoin and similar anaplerotic substrates, ketone body treatments, RNA and gene therapy approaches. Finally, it suggests challenges for the diagnosis of FAOD in the coming years, both for symptomatic patients and for those diagnosed through newborn screening. The ultimate goal would be to identify all the patients born with FAOD and ensure for them the best possible quality of life.
Collapse
Affiliation(s)
- Christine Vianey-Saban
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Nathalie Guffon
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Alain Fouilhoux
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Cécile Acquaviva
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| |
Collapse
|
23
|
Guardado M, Steurer M, Chapin C, Hernandez RD, Ballard PL, Torgerson D. The Urinary Metabolomic Fingerprint in Extremely Preterm Infants on Total Parenteral Nutrition vs. Enteral Feeds. Metabolites 2023; 13:971. [PMID: 37755251 PMCID: PMC10537655 DOI: 10.3390/metabo13090971] [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: 07/28/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
Total Parenteral Nutrition (TPN), which uses intravenous administration of nutrients, minerals and vitamins, is essential for sustaining premature infants until they transition to enteral feeds, but there is limited information on metabolomic differences between infants on TPN and enteral feeds. We performed untargeted global metabolomics on urine samples collected between 23-30 days of life from 314 infants born <29 weeks gestational age from the TOLSURF and PROP cohorts. Principal component analysis across all metabolites showed a separation of infants solely on TPN compared to infants who had transitioned to enteral feeds, indicating global metabolomic differences between infants based on feeding status. Among 913 metabolites that passed quality control filters, 609 varied in abundance between infants on TPN vs. enteral feeds at p < 0.05. Of these, 88% were in the direction of higher abundance in the urine of infants on enteral feeds. In a subset of infants in a longitudinal analysis, both concurrent and delayed changes in metabolite levels were observed with the initiation of enteral feeds. These infants had higher concentrations of essential amino acids, lipids, and vitamins, which are necessary for growth and development, suggesting the nutritional benefit of an enteral feeding regimen.
Collapse
Affiliation(s)
- Miguel Guardado
- Biological and Medical Informatics Graduate Program, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94134, USA
- Department of Epidemiology and Biostatistics, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94158, USA;
- Department of Bioengineering and Therapeutic Sciences, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94134, USA;
| | - Martina Steurer
- Department of Pediatrics, School of Medicine, Mission Bay & Parnassus Campuses, University of California, San Francisco, CA 94158, USA; (M.S.); (C.C.); (P.L.B.)
| | - Cheryl Chapin
- Department of Pediatrics, School of Medicine, Mission Bay & Parnassus Campuses, University of California, San Francisco, CA 94158, USA; (M.S.); (C.C.); (P.L.B.)
| | - Ryan D. Hernandez
- Department of Bioengineering and Therapeutic Sciences, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94134, USA;
| | - Philip L. Ballard
- Department of Pediatrics, School of Medicine, Mission Bay & Parnassus Campuses, University of California, San Francisco, CA 94158, USA; (M.S.); (C.C.); (P.L.B.)
| | - Dara Torgerson
- Department of Epidemiology and Biostatistics, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94158, USA;
| |
Collapse
|
24
|
Tin O, Zübarioğlu T, Cansever MŞ, Kıykım E, Aktuğlu-Zeybek Ç. Maternal Inborn Errors of Metabolism Detected in Expanded Newborn Metabolic Screening. Turk Arch Pediatr 2023; 58:382-387. [PMID: 37317575 PMCID: PMC10441140 DOI: 10.5152/turkarchpediatr.2023.23009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/01/2023] [Indexed: 06/16/2023]
Abstract
OBJECTIVE Pathologic results in expanded metabolic screening tests may be due to the medications, inappropriate sampling methods, or the maternal originated inborn errors of metabolism. The aim of this study is to identify mothers with inborn errors of metabolism through the pathologic expanded metabolic screening results of their babies. MATERIALS AND METHODS Babies who were under 1 year of age and had a pathologic result of an expanded newborn screening for inborn errors of metabolism and their mothers were included in this retrospective single-centered study. Data of expanded metabolic screening results of both babies and their mothers were recorded. Clinical and laboratory findings relevant to suspected inborn errors of metabolism due to the pathologic screening results analysis were also noted for the mothers. RESULTS Seventeen babies and their mothers were enrolled. Expanded metabolic screening results were found compatible with inborn errors of metabolism in 4 (23.5%) of 17 mothers. Two of these mothers were diagnosed with 3-methylcrotonyl-CoA carboxylase deficiency and 2 mothers were diagnosed with glutaric aciduria type 1. CONCLUSION Inborn errors of metabolism can present in any period of life, and this is the first study to address the importance of metabolic screening via tandem mass spectrometry in terms of early diagnosis of inborn errors of metabolism not only in pediatric aged patients but also in adulthood in Turkey. The performance of expanded metabolic screening tests may be an important step in terms of detecting maternal inborn errors of metabolism that are not diagnosed until adulthood.
Collapse
Affiliation(s)
- Oğuzhan Tin
- Department of Pediatrics, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| | - Tanyel Zübarioğlu
- Division of Nutrition and Metabolism, Department of Pediatrics, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| | - Mehmet Şerif Cansever
- Division of Medical Laboratory Techniques, Department of Medical Documentation and Techniques, İstanbul University-Cerrahpaşa, The Vocational School of Health Services, İstanbul, Turkey
| | - Ertuğrul Kıykım
- Division of Nutrition and Metabolism, Department of Pediatrics, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| | - Çiğdem Aktuğlu-Zeybek
- Division of Nutrition and Metabolism, Department of Pediatrics, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| |
Collapse
|
25
|
White S, Mossfield T, Fleming J, Barlow-Stewart K, Ghedia S, Dickson R, Richards F, Bombard Y, Wiley V. Expanding the Australian Newborn Blood Spot Screening Program using genomic sequencing: do we want it and are we ready? Eur J Hum Genet 2023; 31:703-711. [PMID: 36935418 PMCID: PMC10250371 DOI: 10.1038/s41431-023-01311-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/12/2022] [Accepted: 01/31/2023] [Indexed: 03/21/2023] Open
Abstract
Since the introduction of genome sequencing in medicine, the factors involved in deciding how to integrate this technology into population screening programs such as Newborn Screening (NBS) have been widely debated. In Australia, participation in NBS is not mandatory, but over 99.9% of parents elect to uptake this screening. Gauging stakeholder attitudes towards potential changes to NBS is vital in maintaining this high participation rate. The current study aimed to determine the knowledge and attitudes of Australian parents and health professionals to the incorporation of genomic sequencing into NBS programs. Participants were surveyed online in 2016 using surveys adapted from previous studies. The majority of parents (90%) self-reported some knowledge of NBS, with 77% expressing an interest in NBS using the new technology. This was significantly lower than those who would utilise NBS using current technologies (99%). Although, many health professionals (62%) felt that new technologies should currently not be used as an adjunct to NBS, 79% foresaw the use of genomic sequencing in NBS by 2026. However, for genomic sequencing to be considered, practical and technical challenges as well as parent information needs were identified including the need for accurate interpretation of data; pre-and post-test counselling; and appropriate parental consent and opt-out process. Therefore, although some support for implementing genomic sequencing into Australian NBS does exist, there is a need for further investigation into the ethical, social, legal and practical implications of introducing this new technology as a replacement to current NBS methods.
Collapse
Affiliation(s)
- Stephanie White
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
- Department of Clinical Genetics, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Tamara Mossfield
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
- Genea, Sydney CBD, Sydney, NSW, Australia
| | - Jane Fleming
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia.
| | - Kristine Barlow-Stewart
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
| | - Sondhya Ghedia
- Department of Clinical Genetics, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rebecca Dickson
- Genea, Sydney CBD, Sydney, NSW, Australia
- Royal Hospital for Women, Sydney, NSW, Australia
| | - Fiona Richards
- Department of Clinical Genetics, Children's Hospital, Westmead, Sydney, NSW, Australia
| | - Yvonne Bombard
- Genomics Health Services Research Program, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Veronica Wiley
- NSW Newborn Screening Programme, The Children's Hospital at Westmead, Sydney, NSW, Australia
| |
Collapse
|
26
|
Babcock SJ, Flores-Marin D, Thiagarajah JR. The genetics of monogenic intestinal epithelial disorders. Hum Genet 2023; 142:613-654. [PMID: 36422736 PMCID: PMC10182130 DOI: 10.1007/s00439-022-02501-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/23/2022] [Indexed: 11/27/2022]
Abstract
Monogenic intestinal epithelial disorders, also known as congenital diarrheas and enteropathies (CoDEs), are a group of rare diseases that result from mutations in genes that primarily affect intestinal epithelial cell function. Patients with CoDE disorders generally present with infantile-onset diarrhea and poor growth, and often require intensive fluid and nutritional management. CoDE disorders can be classified into several categories that relate to broad areas of epithelial function, structure, and development. The advent of accessible and low-cost genetic sequencing has accelerated discovery in the field with over 45 different genes now associated with CoDE disorders. Despite this increasing knowledge in the causal genetics of disease, the underlying cellular pathophysiology remains incompletely understood for many disorders. Consequently, clinical management options for CoDE disorders are currently limited and there is an urgent need for new and disorder-specific therapies. In this review, we provide a general overview of CoDE disorders, including a historical perspective of the field and relationship to other monogenic disorders of the intestine. We describe the genetics, clinical presentation, and known pathophysiology for specific disorders. Lastly, we describe the major challenges relating to CoDE disorders, briefly outline key areas that need further study, and provide a perspective on the future genetic and therapeutic landscape.
Collapse
Affiliation(s)
- Stephen J Babcock
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA
| | - David Flores-Marin
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
27
|
Lin S, Nateqi J, Weingartner-Ortner R, Gruarin S, Marling H, Pilgram V, Lagler FB, Aigner E, Martin AG. An artificial intelligence-based approach for identifying rare disease patients using retrospective electronic health records applied for Pompe disease. Front Neurol 2023; 14:1108222. [PMID: 37153672 PMCID: PMC10160659 DOI: 10.3389/fneur.2023.1108222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
Objective We retrospectively screened 350,116 electronic health records (EHRs) to identify suspected patients for Pompe disease. Using these suspected patients, we then describe their phenotypical characteristics and estimate the prevalence in the respective population covered by the EHRs. Methods We applied Symptoma's Artificial Intelligence-based approach for identifying rare disease patients to retrospective anonymized EHRs provided by the "University Hospital Salzburg" clinic group. Within 1 month, the AI screened 350,116 EHRs reaching back 15 years from five hospitals, and 104 patients were flagged as probable for Pompe disease. Flagged patients were manually reviewed and assessed by generalist and specialist physicians for their likelihood for Pompe disease, from which the performance of the algorithms was evaluated. Results Of the 104 patients flagged by the algorithms, generalist physicians found five "diagnosed," 10 "suspected," and seven patients with "reduced suspicion." After feedback from Pompe disease specialist physicians, 19 patients remained clinically plausible for Pompe disease, resulting in a specificity of 18.27% for the AI. Estimating from the remaining plausible patients, the prevalence of Pompe disease for the greater Salzburg region [incl. Bavaria (Germany), Styria (Austria), and Upper Austria (Austria)] was one in every 18,427 people. Phenotypes for patient cohorts with an approximated onset of symptoms above or below 1 year of age were established, which correspond to infantile-onset Pompe disease (IOPD) and late-onset Pompe disease (LOPD), respectively. Conclusion Our study shows the feasibility of Symptoma's AI-based approach for identifying rare disease patients using retrospective EHRs. Via the algorithm's screening of an entire EHR population, a physician had only to manually review 5.47 patients on average to find one suspected candidate. This efficiency is crucial as Pompe disease, while rare, is a progressively debilitating but treatable neuromuscular disease. As such, we demonstrated both the efficiency of the approach and the potential of a scalable solution to the systematic identification of rare disease patients. Thus, similar implementation of this methodology should be encouraged to improve care for all rare disease patients.
Collapse
Affiliation(s)
- Simon Lin
- Science Department, Symptoma GmbH, Vienna, Austria
- Department of Internal Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Jama Nateqi
- Science Department, Symptoma GmbH, Vienna, Austria
- Department of Internal Medicine, Paracelsus Medical University, Salzburg, Austria
| | | | | | | | - Vinzenz Pilgram
- Medical and Information Technology - MIT, University Hospital Salzburg (SALK), Salzburg, Austria
| | - Florian B. Lagler
- Medical and Information Technology - MIT, University Hospital Salzburg (SALK), Salzburg, Austria
- Department of Pediatrics and Institute for Inherited Metabolic Diseases, Paracelsus Medical University, Salzburg, Austria
| | - Elmar Aigner
- Department of Internal Medicine, Paracelsus Medical University, Salzburg, Austria
- Medical and Information Technology - MIT, University Hospital Salzburg (SALK), Salzburg, Austria
| | - Alistair G. Martin
- Science Department, Symptoma GmbH, Vienna, Austria
- *Correspondence: Alistair G. Martin
| |
Collapse
|
28
|
Cnpy3 2xHA mice reveal neuronal expression of Cnpy3 in the brain. J Neurosci Methods 2023; 383:109730. [PMID: 36280087 DOI: 10.1016/j.jneumeth.2022.109730] [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: 08/09/2022] [Revised: 10/04/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Identification of biallelic CNPY3 mutations in patients with epileptic encephalopathy and abnormal electroencephalography findings of Cnpy3 knock-out mice have indicated that the loss of CNPY3 function causes neurological disorders such as epilepsy. However, the basic property of CNPY3 in the brain remains unclear. NEW METHOD We generated C-terminal 2xHA-tag knock-in Cnpy3 mice by i-GONAD in vivo genome editing system to investigate the expression and function of Cnpy3 in the mouse brain. RESULTS 2xHA-tagged Cnpy3 was confirmed by immunoblot analysis using anti-HA and CNPY3 antibodies, although HA tagging caused the decreased Cnpy3 protein level. Immunohistochemical analysis of Cnpy32xHA knock-in mice showed that Cnpy3-2xHA was predominantly expressed in the neuron. In addition, Cnpy3 and Cnpy3-2xHA were both localized in the endoplasmic reticulum and synaptosome and showed age-dependent expression changes in the brain. COMPARISON WITH EXISTING METHODS Conventional Cnpy3 antibodies could not allow us to investigate the distribution of Cnpy3 expression in the brain, while HA-tagging revealed the expression of CNPY3 in neuronal cells. CONCLUSIONS Taken together, we demonstrated that Cnpy32xHA knock-in mice would be useful to further elucidate the property of Cnpy3 in brain function and neurological disorders.
Collapse
|
29
|
Veys K, Zadora W, Hohenfellner K, Bockenhauer D, Janssen MCH, Niaudet P, Servais A, Topaloglu R, Besouw M, Novo R, Haffner D, Kanzelmeyer N, Pape L, Wühl E, Harms E, Awan A, Sikora P, Ariceta G, van den Heuvel B, Levtchenko E. Outcome of infantile nephropathic cystinosis depends on early intervention, not genotype: A multicenter sibling cohort study. J Inherit Metab Dis 2023; 46:43-54. [PMID: 36117148 DOI: 10.1002/jimd.12562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/01/2022] [Accepted: 09/16/2022] [Indexed: 01/19/2023]
Abstract
Infantile nephropathic cystinosis (INC) is an inheritable lysosomal storage disorder characterized by lysosomal cystine accumulation, progressive kidney disease, and multiple extrarenal complications (ERCs). Cysteamine postpones the onset of end-stage kidney disease (ESKD) and reduces the incidence of ERCs; however, cysteamine is generally initiated upon establishment of the renal Fanconi syndrome (FS) and partial loss of kidney function, whereas data on long-term effects of cysteamine administered from neonatal age are lacking. An international multicenter retrospective cohort study of siblings with INC was set up to investigate the outcome in relation to age at initiation of cysteamine versus CTNS genotype, with attention to patients treated with cysteamine from neonatal age. None of the siblings treated from neonatal age (n = 9; age 10 ± 6 years) had reached ESKD, while 22% of their index counterparts (n = 9; age 14 ± 5 years) had commenced renal replacement therapy. Siblings treated with cysteamine from the onset of symptoms at a younger age compared with their index counterparts, reached ESKD at a significant older age (13 ± 3 vs. 10 ± 3 years, p = 0.002). In contrast, no significant difference in ERCs was observed between sibling and index patients, independently from the age at initiation of cysteamine. The CTNS genotype had no impact on the overall outcome in this cohort. In INC, presymptomatic treatment with cysteamine results in a better renal outcome in comparison to treatment initiated from the onset of symptoms. This justifies including cystinosis into newborn screening programs. SYNOPSIS: In infantile nephropathic cystinosis, presymptomatic treatment with cysteamine improves the renal outcome which justifies the inclusion of cystinosis into newborn screening programs.
Collapse
Affiliation(s)
- Koenraad Veys
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Development & Regeneration, Laboratory of Pediatric Nephrology, KU Leuven, Leuven, Belgium
| | - Ward Zadora
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | | | - Detlef Bockenhauer
- Department of Pediatric Nephrology, Great Ormond Street Hospital for Sick Children NHS Foundation Trust (GOSH) and Department of Renal Medicine, University College London, London, UK
| | - Mirian C H Janssen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Patrick Niaudet
- Department of Pediatric Nephrology, Hôpital Necker-Enfants Malades, Paris, France
| | - Aude Servais
- Department of Adult Nephrology and Transplantation, Hôpital Necker, Paris, France
| | - Rezan Topaloglu
- Department of Pediatric Nephrology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Martine Besouw
- Department of Pediatric Nephrology, University of Groningen, Groningen, The Netherlands
| | - Robert Novo
- Pediatric Nephrology, Hôpital Jeanne de Flandre, University Hospital Lille, Lille, France
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Nele Kanzelmeyer
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Lars Pape
- Department of Pediatrics, University Hospital Essen, Essen, Germany
| | - Elke Wühl
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Erik Harms
- Children's University Hospital Münster, Münster, Germany
| | - Atif Awan
- Paediatric Nephrology and Transplantation, Temple Street Children's University Hospital, Dublin, Ireland
| | - Przemyslaw Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Gema Ariceta
- Department of Pediatric Nephrology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Bert van den Heuvel
- Department of Development & Regeneration, Laboratory of Pediatric Nephrology, KU Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Development & Regeneration, Laboratory of Pediatric Nephrology, KU Leuven, Leuven, Belgium
| |
Collapse
|
30
|
Kalkman S, Dondorp W. The case for screening in early life for 'non-treatable' disorders: ethics, evidence and proportionality. A report from the Health Council of the Netherlands. Eur J Hum Genet 2022; 30:1155-1158. [PMID: 35132176 PMCID: PMC9554018 DOI: 10.1038/s41431-022-01055-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/25/2021] [Accepted: 01/20/2022] [Indexed: 12/15/2022] Open
Abstract
In the Netherlands, the call to add 'non-treatable' disorders to the newborn bloodspot screening programme has found a sympathetic ear with the Government. In 2019, the Health Council of the Netherlands was formally asked for advice on the conditions under which bloodspot screening for such disorders might be offered. Here we present the reasoning and the recommendations of the resulting report, and briefly discuss its reception. The report holds on to the classical view that screening must benefit the child, but argues for a wider account of child benefit than only in terms of substantial health gains. However, screening for 'non-treatable' disorders would still require evidence of a favourable benefits to harm ratio. The report presents a framework for such screening, but concludes that apart perhaps from Duchenne Muscular Dystrophy (DMD), no or only very few 'non-treatable' disorders would at present meet its criteria. Setting up a screening programme that might benefit only a small percentage of families struggling with uncertainty about their child's diagnosis would not seem proportional. Instead, the Government is advised to invest in a better infrastructure for early referral, testing and care. The reaction to the report from proponents of such screening shows that the dividing line in the debate is not about whether screening neonates for 'non-treatable' disorders is acceptable in itself. It is rather whether such screening should be regarded as catering to a parental 'right to know', or as a public health service that should be subject to standards of evidence and proportionality.
Collapse
Affiliation(s)
- Shona Kalkman
- Health Council of the Netherlands, The Hague, the Netherlands
| | - Wybo Dondorp
- Dept of Health, Ethics & Society, School for Public Health & Primary Care (CAPHRI), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
31
|
Ferket BS, Baldwin Z, Murali P, Pai A, Mittendorf KF, Russell HV, Chen F, Lynch FL, Lich KH, Hindorff LA, Savich R, Slavotinek A, Smith HS, Gelb BD, Veenstra DL. Cost-effectiveness frameworks for comparing genome and exome sequencing versus conventional diagnostic pathways: A scoping review and recommended methods. Genet Med 2022; 24:2014-2027. [PMID: 35833928 PMCID: PMC9997042 DOI: 10.1016/j.gim.2022.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Methodological challenges have limited economic evaluations of genome sequencing (GS) and exome sequencing (ES). Our objective was to develop conceptual frameworks for model-based cost-effectiveness analyses (CEAs) of diagnostic GS/ES. METHODS We conducted a scoping review of economic analyses to develop and iterate with experts a set of conceptual CEA frameworks for GS/ES for prenatal testing, early diagnosis in pediatrics, diagnosis of delayed-onset disorders in pediatrics, genetic testing in cancer, screening of newborns, and general population screening. RESULTS Reflecting on 57 studies meeting inclusion criteria, we recommend the following considerations for each clinical scenario. For prenatal testing, performing comparative analyses of costs of ES strategies and postpartum care, as well as genetic diagnoses and pregnancy outcomes. For early diagnosis in pediatrics, modeling quality-adjusted life years (QALYs) and costs over ≥20 years for rapid turnaround GS/ES. For hereditary cancer syndrome testing, modeling cumulative costs and QALYs for the individual tested and first/second/third-degree relatives. For tumor profiling, not restricting to treatment uptake or response and including QALYs and costs of downstream outcomes. For screening, modeling lifetime costs and QALYs and considering consequences of low penetrance and GS/ES reanalysis. CONCLUSION Our frameworks can guide the design of model-based CEAs and ultimately foster robust evidence for the economic value of GS/ES.
Collapse
Affiliation(s)
- Bart S Ferket
- Institute for Healthcare Delivery Science, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Zach Baldwin
- The Comparative Health Outcomes, Policy and Economics (CHOICE) Institute, School of Pharmacy, University of Washington, Seattle, WA
| | - Priyanka Murali
- Division of Medical Genetics, Department of Medicine, University of Washington Medical Center, University of Washington, Seattle, WA
| | - Akila Pai
- Institute for Healthcare Delivery Science, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kathleen F Mittendorf
- Department of Translational and Applied Genomics (TAG), Kaiser Permanente Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Heidi V Russell
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX; Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - Flavia Chen
- Program in Bioethics, University of California San Francisco, San Francisco, CA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA
| | | | - Kristen Hassmiller Lich
- Department of Health Policy and Management, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lucia A Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Renate Savich
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS; Division of Neonatology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Anne Slavotinek
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Hadley Stevens Smith
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX
| | - Bruce D Gelb
- Departments of Pediatrics and Genetics & Genomic Sciences, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - David L Veenstra
- The Comparative Health Outcomes, Policy and Economics (CHOICE) Institute, School of Pharmacy, University of Washington, Seattle, WA
| |
Collapse
|
32
|
Dondorp W, de Wert G. Towards responsible ctDNA-based multi-cancer screening: a preliminary exploration and discussion of ethically relevant aspects. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:235-243. [PMID: 39697487 PMCID: PMC11648474 DOI: 10.20517/evcna.2022.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/07/2022] [Accepted: 08/12/2022] [Indexed: 12/20/2024]
Abstract
While testing for easily accessible biomarkers in the circulation ("liquid biopsy") has found its way to clinical cancer care, a further expected development is its use as a "universal" early detection test in population screening for cancer. A promising marker for such screening is circulating cell-free fragments of tumor DNA, shed into the circulation during tumor cell turnover. Several blood-based "multicancer early detection (MCED) tests" have recently been developed - but still need validation in large-scale studies involving non-patient populations. In this paper, we proactively explore the ethical aspects of this development. We refer to an often quoted synthesis of the internationally accepted framework of principles for responsible screening as first drawn up for the World Health Organisation (WHO) by Wilson and Junger 50 years ago and further developed and fine-tuned ever since. As our analysis suggests, some specific ethical issues and concerns about potential MCED screening connect to the fact that cancer is not just one disease. As a consequence, not all findings will have the same clinical utility. We discuss this against the background of earlier debates pertaining to broad scope forms of screening in other contexts, specifically newborn and reproductive genetic screening. We highlight the guidance provided by some of the criteria from the screening framework that seems most relevant in this connection: the need for screening objectives to be defined at the outset, the need for mechanisms to minimize potential risks, and the requirement that, for those participating in the screening, the overall benefits outweigh the harm.
Collapse
Affiliation(s)
- Wybo Dondorp
- Department of Health, Ethics & Society (HES), School for Oncology and Reproduction (GROW), School for Public Health and Primary Care (CAPHRI), Maastricht University, HA Maastricht 6229, the Netherlands
| | | |
Collapse
|
33
|
Sustar U, Groselj U, Khan SA, Shafi S, Khan I, Kovac J, Bizjan BJ, Battelino T, Sadiq F. A homozygous variant in the GPIHBP1 gene in a child with severe hypertriglyceridemia and a systematic literature review. Front Genet 2022; 13:983283. [PMID: 36051701 PMCID: PMC9424485 DOI: 10.3389/fgene.2022.983283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Due to nonspecific symptoms, rare dyslipidaemias are frequently misdiagnosed, overlooked, and undertreated, leading to increased risk for severe cardiovascular disease, pancreatitis and/or multiple organ failures before diagnosis. Better guidelines for the recognition and early diagnosis of rare dyslipidaemias are urgently required. Methods: Genomic DNA was isolated from blood samples of a Pakistani paediatric patient with hypertriglyceridemia, and from his parents and siblings. Next-generation sequencing (NGS) was performed, and an expanded dyslipidaemia panel was employed for genetic analysis. Results: The NGS revealed the presence of a homozygous missense pathogenic variant c.230G>A (NM_178172.6) in exon 3 of the GPIHBP1 (glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1) gene resulting in amino acid change p.Cys77Tyr (NP_835466.2). The patient was 5.5 years old at the time of genetic diagnosis. The maximal total cholesterol and triglyceride levels were measured at the age of 10 months (850.7 mg/dl, 22.0 mmol/L and 5,137 mg/dl, 58.0 mmol/L, respectively). The patient had cholesterol deposits at the hard palate, eruptive xanthomas, lethargy, poor appetite, and mild splenomegaly. Both parents and sister were heterozygous for the familial variant in the GPIHBP1 gene. Moreover, in the systematic review, we present 62 patients with pathogenic variants in the GPIHBP1 gene and clinical findings, associated with hyperlipoproteinemia. Conclusion: In a child with severe hypertriglyceridemia, we identified a pathogenic variant in the GPIHBP1 gene causing hyperlipoproteinemia (type 1D). In cases of severe elevations of plasma cholesterol and/or triglycerides genetic testing for rare dyslipidaemias should be performed as soon as possible for optimal therapy and patient management.
Collapse
Affiliation(s)
- Ursa Sustar
- Department of Endocrinology, Diabetes and Metabolism, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Urh Groselj
- Department of Endocrinology, Diabetes and Metabolism, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Urh Groselj, ; Fouzia Sadiq,
| | - Sabeen Abid Khan
- Department of Paediatrics, Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Saeed Shafi
- Department of Anatomy, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Iqbal Khan
- Department of Vascular Surgery, Shifa International Hospital, Islamabad, Pakistan
- Department of Vascular Surgery, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Jernej Kovac
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Barbara Jenko Bizjan
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolism, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Fouzia Sadiq
- Directorate of Research, Shifa Tameer-e-Millat University, Islamabad, Pakistan
- *Correspondence: Urh Groselj, ; Fouzia Sadiq,
| |
Collapse
|
34
|
Elhawary NA, AlJahdali IA, Abumansour IS, Elhawary EN, Gaboon N, Dandini M, Madkhali A, Alosaimi W, Alzahrani A, Aljohani F, Melibary EM, Kensara OA. Genetic etiology and clinical challenges of phenylketonuria. Hum Genomics 2022; 16:22. [PMID: 35854334 PMCID: PMC9295449 DOI: 10.1186/s40246-022-00398-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/08/2022] [Indexed: 02/08/2023] Open
Abstract
This review discusses the epidemiology, pathophysiology, genetic etiology, and management of phenylketonuria (PKU). PKU, an autosomal recessive disease, is an inborn error of phenylalanine (Phe) metabolism caused by pathogenic variants in the phenylalanine hydroxylase (PAH) gene. The prevalence of PKU varies widely among ethnicities and geographic regions, affecting approximately 1 in 24,000 individuals worldwide. Deficiency in the PAH enzyme or, in rare cases, the cofactor tetrahydrobiopterin results in high blood Phe concentrations, causing brain dysfunction. Untreated PKU, also known as PAH deficiency, results in severe and irreversible intellectual disability, epilepsy, behavioral disorders, and clinical features such as acquired microcephaly, seizures, psychological signs, and generalized hypopigmentation of skin (including hair and eyes). Severe phenotypes are classic PKU, and less severe forms of PAH deficiency are moderate PKU, mild PKU, mild hyperphenylalaninaemia (HPA), or benign HPA. Early diagnosis and intervention must start shortly after birth to prevent major cognitive and neurological effects. Dietary treatment, including natural protein restriction and Phe-free supplements, must be used to maintain blood Phe concentrations of 120-360 μmol/L throughout the life span. Additional treatments include the casein glycomacropeptide (GMP), which contains very limited aromatic amino acids and may improve immunological function, and large neutral amino acid (LNAA) supplementation to prevent plasma Phe transport into the brain. The synthetic BH4 analog, sapropterin hydrochloride (i.e., Kuvan®, BioMarin), is another potential treatment that activates residual PAH, thus decreasing Phe concentrations in the blood of PKU patients. Moreover, daily subcutaneous injection of pegylated Phe ammonia-lyase (i.e., pegvaliase; PALYNZIQ®, BioMarin) has promised gene therapy in recent clinical trials, and mRNA approaches are also being studied.
Collapse
Affiliation(s)
- Nasser A. Elhawary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, P.O. Box 57543, Mecca, 21955 Saudi Arabia
| | - Imad A. AlJahdali
- Department of Community Medicine, College of Medicine, Umm Al-Qura University, P.O. Box 57543, Mecca, 21955 Saudi Arabia
| | - Iman S. Abumansour
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, P.O. Box 57543, Mecca, 21955 Saudi Arabia
| | - Ezzeldin N. Elhawary
- Faculty of Medicine, MS Genomic Medicine Program, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Nagwa Gaboon
- Department of Clinical Genetics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohammed Dandini
- Department of Laboratory and Blood Bank, Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Abdulelah Madkhali
- Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Wafaa Alosaimi
- Department of Hematology, Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Abdulmajeed Alzahrani
- Department of Laboratory and Blood Bank at Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Fawzia Aljohani
- Department of Pediatric Clinics, Maternity and Children Hospital, King Salman Medical City, Madinah, Saudi Arabia
| | - Ehab M. Melibary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, P.O. Box 57543, Mecca, 21955 Saudi Arabia
| | - Osama A. Kensara
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Umm Al-Qura University, Jeddah, Saudi Arabia
- Department of Biochemistry, Batterjee Medical College, Jeddah, Saudi Arabia
| |
Collapse
|
35
|
Bick D, Ahmed A, Deen D, Ferlini A, Garnier N, Kasperaviciute D, Leblond M, Pichini A, Rendon A, Satija A, Tuff-Lacey A, Scott RH. Newborn Screening by Genomic Sequencing: Opportunities and Challenges. Int J Neonatal Screen 2022; 8:40. [PMID: 35892470 PMCID: PMC9326745 DOI: 10.3390/ijns8030040] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 12/11/2022] Open
Abstract
Newborn screening for treatable disorders is one of the great public health success stories of the twentieth century worldwide. This commentary examines the potential use of a new technology, next generation sequencing, in newborn screening through the lens of the Wilson and Jungner criteria. Each of the ten criteria are examined to show how they might be applied by programmes using genomic sequencing as a screening tool. While there are obvious advantages to a method that can examine all disease-causing genes in a single assay at an ever-diminishing cost, implementation of genomic sequencing at scale presents numerous challenges, some which are intrinsic to screening for rare disease and some specifically linked to genomics-led screening. In addition to questions specific to routine screening considerations, the ethical, communication, data management, legal, and social implications of genomic screening programmes require consideration.
Collapse
Affiliation(s)
- David Bick
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Arzoo Ahmed
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Dasha Deen
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Alessandra Ferlini
- Medical Genetics Unit, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | | | - Dalia Kasperaviciute
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Mathilde Leblond
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Amanda Pichini
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Augusto Rendon
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Aditi Satija
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Alice Tuff-Lacey
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| | - Richard H. Scott
- Genomics England Ltd., Dawson Hall, Charterhouse Square, Barbican, London EC1M 6BQ, UK; (A.A.); (D.D.); (D.K.); (M.L.); (A.P.); (A.R.); (A.S.); (A.T.-L.); (R.H.S.)
| |
Collapse
|
36
|
Sustar U, Groselj U, Trebusak Podkrajsek K, Mlinaric M, Kovac J, Thaler M, Drole Torkar A, Skarlovnik A, Battelino T, Hovnik T. Early Discovery of Children With Lysosomal Acid Lipase Deficiency With the Universal Familial Hypercholesterolemia Screening Program. Front Genet 2022; 13:936121. [PMID: 35903350 PMCID: PMC9314654 DOI: 10.3389/fgene.2022.936121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023] Open
Abstract
Lysosomal acid lipase deficiency (LAL-D) is an autosomal recessive lysosomal storage disorder, caused by homozygous or compound heterozygous pathogenic variants in the LIPA gene. Clinically, LAL-D is under- and misdiagnosed, due to similar clinical and laboratory findings with other cholesterol or liver misfunctions. As a part of the Slovenian universal familial hypercholesterolemia (FH) screening, LAL-D is screened as a secondary condition among other rare dyslipidemias manifesting with hypercholesterolemia. Out of 669 children included, three were positive for a homozygous disease-causing splicing variant NM_000235.4: c.894G > A (NP_000226.2:p. Gln298Gln) in the LIPA gene (NG_008194.1). The mean age by the diagnosis of LAL-D was 9.8 ± 0.9 years. Moreover, all three LAL-D-positive children had an important elevation of transaminases and decreased activity of the lysosomal acid lipase enzyme. Abdominal MRI in all children detected an enlarged liver but a normal-sized spleen. In conclusion, universal FH screening algorithms with the confirmatory genetic analysis in the pediatric population enable also rare dyslipidemia detection at an early age. An important clinical criterion for differentiation between FH and the LAL-D-positive children has elevated transaminase levels (AST and ALT). In all three LAL-D positive children, an improvement in cholesterol and transaminase levels and steatosis of the liver has been seen after early treatment initiation.
Collapse
Affiliation(s)
- Ursa Sustar
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Urh Groselj
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Urh Groselj, ; Tinka Hovnik,
| | - Katarina Trebusak Podkrajsek
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
| | - Matej Mlinaric
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jernej Kovac
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Martin Thaler
- Department of Radiology, University Children’s Hospital Ljubljana, Ljubljana, Slovenia
| | - Ana Drole Torkar
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ajda Skarlovnik
- Department of Vascular Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tinka Hovnik
- Clinical Institute of Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Urh Groselj, ; Tinka Hovnik,
| |
Collapse
|
37
|
Korotkov E, Zaytsev K, Fedorov A. Use of 6 Nucleotide Length Words to Study the Complexity of Gene Sequences from Different Organisms. ENTROPY (BASEL, SWITZERLAND) 2022; 24:632. [PMID: 35626518 PMCID: PMC9141341 DOI: 10.3390/e24050632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022]
Abstract
In this paper, we attempted to find a relation between bacteria living conditions and their genome algorithmic complexity. We developed a probabilistic mathematical method for the evaluation of k-words (6 bases length) occurrence irregularity in bacterial gene coding sequences. For this, the coding sequences from different bacterial genomes were analyzed and as an index of k-words occurrence irregularity, we used W, which has a distribution similar to normal. The research results for bacterial genomes show that they can be divided into two uneven groups. First, the smaller one has W in the interval from 170 to 475, while for the second it is from 475 to 875. Plants, metazoan and virus genomes also have W in the same interval as the first bacterial group. We suggested that second bacterial group coding sequences are much less susceptible to evolutionary changes than the first group ones. It is also discussed to use the W index as a biological stress value.
Collapse
Affiliation(s)
- Eugene Korotkov
- Institute of Bioengineering, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Konstantin Zaytsev
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia; (K.Z.); (A.F.)
| | - Alexey Fedorov
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia; (K.Z.); (A.F.)
| |
Collapse
|
38
|
Powell SN, Byfield G, Bennetone A, Frantz AM, Harrison LK, James-Crook ER, Osborne H, Owens TH, Shaw JL, O’Daniel J, Milko LV. Parental Guidance Suggested: Engaging Parents as Partners in Research Studies of Genomic Screening for a Pediatric Population. Front Genet 2022; 13:867030. [PMID: 35401676 PMCID: PMC8990237 DOI: 10.3389/fgene.2022.867030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 01/21/2023] Open
Abstract
Recent advances in genomic sequencing and genomic medicine are reshaping the landscape of clinical care. As a screening modality, genetic sequencing has the potential to dramatically expand the clinical utility of newborn screening (NBS), though significant barriers remain regarding ethical, legal, and social implications (ELSI) and technical and evidentiary challenges. Stakeholder-informed implementation research is poised to grapple with many of these barriers, and parents are crucial stakeholders in this process. We describe the formation and activities of a Community Research Board (CRB) composed of parents with diverse backgrounds assembled to participate in an ongoing research partnership with genomic and public health researchers at the University of North Carolina. The mission of the CRB is to provide insight into parental perspectives regarding the prospect of adding genomic sequencing to NBS and collaboratively develop strategies to ensure its equitable uptake. We describe how these contributions can improve the accessibility of research and recruitment methods and promote trust and inclusivity within diverse communities to maximize the societal benefit of population genomic screening in healthy children.
Collapse
Affiliation(s)
- Sabrina N. Powell
- Program for Precision Medicine in Health Care, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Grace Byfield
- Program for Precision Medicine in Health Care, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | - Annabelle M. Frantz
- Program for Precision Medicine in Health Care, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Langston K. Harrison
- Program for Precision Medicine in Health Care, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | - Heather Osborne
- Community Research Board Member, Mooresville, NC, United States
| | | | | | - Julianne O’Daniel
- Program for Precision Medicine in Health Care, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Laura V. Milko
- Program for Precision Medicine in Health Care, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States,*Correspondence: Laura V. Milko,
| |
Collapse
|
39
|
Shibuya Y, Kumar KK, Mader MMD, Yoo Y, Ayala LA, Zhou M, Mohr MA, Neumayer G, Kumar I, Yamamoto R, Marcoux P, Liou B, Bennett FC, Nakauchi H, Sun Y, Chen X, Heppner FL, Wyss-Coray T, Südhof TC, Wernig M. Treatment of a genetic brain disease by CNS-wide microglia replacement. Sci Transl Med 2022; 14:eabl9945. [PMID: 35294256 PMCID: PMC9618306 DOI: 10.1126/scitranslmed.abl9945] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Hematopoietic cell transplantation after myeloablative conditioning has been used to treat various genetic metabolic syndromes but is largely ineffective in diseases affecting the brain presumably due to poor and variable myeloid cell incorporation into the central nervous system. Here, we developed and characterized a near-complete and homogeneous replacement of microglia with bone marrow cells in mice without the need for genetic manipulation of donor or host. The high chimerism resulted from a competitive advantage of scarce donor cells during microglia repopulation rather than enhanced recruitment from the periphery. Hematopoietic stem cells, but not immediate myeloid or monocyte progenitor cells, contained full microglia replacement potency equivalent to whole bone marrow. To explore its therapeutic potential, we applied microglia replacement to a mouse model for Prosaposin deficiency, which is characterized by a progressive neurodegeneration phenotype. We found a reduction of cerebellar neurodegeneration and gliosis in treated brains, improvement of motor and balance impairment, and life span extension even with treatment started in young adulthood. This proof-of-concept study suggests that efficient microglia replacement may have therapeutic efficacy for a variety of neurological diseases.
Collapse
Affiliation(s)
- Yohei Shibuya
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin K Kumar
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Marius Marc-Daniel Mader
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Yongjin Yoo
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,These authors contributed equally
| | - Luis Angel Ayala
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mu Zhou
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Gernot Neumayer
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ishan Kumar
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryo Yamamoto
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul Marcoux
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Benjamin Liou
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA,Division of Stem Cell Therapy, Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ying Sun
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Frank L. Heppner
- Department of Neuropathology, Cluster of Excellence, NeuroCure, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany,Department of Neuropathology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany,Cluster of Excellence, NeuroCure, Charitéplatz 1, 10117 Berlin, Germany,Berlin Institute of Health (BIH), 10117 Berlin, Germany,German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA,Veterans Administration Palo Alto Healthcare System, Palo Alto, CA 94304, USA
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA,Lead Contact,Correspondence:
| |
Collapse
|
40
|
Malgorzata M, Maria S, Michał W. Genetic testing-whether to allow complete freedom? Direct to consumer tests versus genetic tests for medical purposes. J Appl Genet 2022; 63:119-126. [PMID: 34826052 PMCID: PMC8755658 DOI: 10.1007/s13353-021-00670-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/04/2021] [Accepted: 11/12/2021] [Indexed: 10/26/2022]
Abstract
Direct-to-consumer tests opened the opportunity of genetic testing without medical supervision, e.g., without medical referral and medical interpretation of the results. Thus, these approaches allow for free access to information concerning individual genetic profile increasing the area of personal freedom, but also posing the risk of false (positive and negative) or misinterpreted results along with health and psychological negative consequences. The paper discusses medical and non-medical applications of DTC, exploring also the legal framework implemented by European states and organizations. These legal acts strive to control the developing DTC market through such basic principles as patient protection, informed consent, medical information confidentiality, and the rights to know and to refuse knowledge about one's genetic predispositions.
Collapse
Affiliation(s)
- Madej Malgorzata
- Institute of Political Science, University of Wroclaw, Wrocław, Poland
| | - Sąsiadek Maria
- Department of Genetics, Medical University of Wroclaw, Wrocław, Poland.
| | - Witt Michał
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
41
|
An Assessment of Traditional and Genomic Screening in Newborns and their Applicability for Africa. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
42
|
Wu X, Yang Y, Zhou L, Long W, Yu B. Are We Ready for Newborn Genetic Screening? A Cross-Sectional Survey of Healthcare Professionals in Southeast China. Front Pediatr 2022; 10:875229. [PMID: 35601442 PMCID: PMC9120836 DOI: 10.3389/fped.2022.875229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/06/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES To understand the knowledge, attitude, willingness, and ability of healthcare professionals working in newborn screening (NBS) centers regarding newborn genetic screening (nGS). METHODS The questionnaire consisted of four sections with 27 questions and the data were collected by the WJX platform. All participants accessed the questionnaire by scanning a specific QR code with their mobile phones. Two researchers independently completed the summary and analysis. RESULTS A total of 258 valid questionnaires were collected from 43 NBS centers in six provinces of southeast China. In total, 209 (81.01%) participants were interested in nGS, and almost all participants (97.67%) thought that nGS was necessary in China. About 89.53% of participants thought that it could be used to effectively expand the diseases that could be screened, but 72.87% also worried about the inability to provide genetic counseling. About 55.34% suggested that nGS and tandem mass spectrometry (TMS) screening could be applied in a unite screening mode. The higher the institution and personal education levels, the higher the interest healthcare professionals displayed toward nGS. However, they also showed greater concern about the inability to provide genetic counseling and ethical issues. If a center had engaged in TMS screening, its staff would have been more likely to believe that nGS had great advantages. In addition, most participants had ethical concerns, such as "the psychological burden caused by carrying information regarding adult morbidity risk." CONCLUSION Most participants were interested and considered nGS necessary. The inability to provide genetic counseling may be the primary impediment to clinical practice. Three important influencing factors were level of education, institution level, and engagement in TMS screening.
Collapse
Affiliation(s)
- Xian Wu
- Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Yuqi Yang
- Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Lingna Zhou
- Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Wei Long
- Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Bin Yu
- Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| |
Collapse
|
43
|
Scarpa M, Bonham JR, Dionisi-Vici C, Prevot J, Pergent M, Meyts I, Mahlaoui N, Schielen PC. Newborn screening as a fully integrated system to stimulate equity in neonatal screening in Europe. Lancet Reg Health Eur 2022; 13:100311. [PMID: 35199083 PMCID: PMC8841274 DOI: 10.1016/j.lanepe.2022.100311] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
44
|
Suhaimi SA, Zulkipli IN, Ghani H, Abdul-Hamid MRW. Applications of next generation sequencing in the screening and diagnosis of thalassemia: A mini-review. Front Pediatr 2022; 10:1015769. [PMID: 36245713 PMCID: PMC9557073 DOI: 10.3389/fped.2022.1015769] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Thalassemias are a group of inherited blood disorders that affects 5-7% of the world population. Comprehensive screening strategies are essential for the management and prevention of this disorder. Today, many clinical and research laboratories have widely utilized next-generation sequencing (NGS) technologies to identify diseases, from germline and somatic disorders to infectious diseases. Yet, NGS application in thalassemia is limited and has just recently surfaced due to current demands in seeking alternative DNA screening tools that are more efficient, versatile, and cost-effective. This review aims to understand the several aspects of NGS technology, including its most current and expanding uses, advantages, and limitations, along with the issues and solutions related to its integration into routine screening and diagnosis of thalassemias. Hitherto, NGS has been a groundbreaking technology that offers tremendous improvements as a diagnostic tool for thalassemia in terms of its higher throughput, accuracy, and adaptability. The superiority of NGS in detecting rare variants, solving complex hematological problems, and providing non-invasive alternatives to neonatal diagnosis cannot be overlooked. However, several pitfalls still preclude its use as a stand-alone technique over conventional methods.
Collapse
Affiliation(s)
| | | | - Hazim Ghani
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | | |
Collapse
|
45
|
Excluding embryos with two novel mutations in FREM2 gene by the next-generation sequencing-based single nucleotide polymorphism haplotyping. Aging (Albany NY) 2021; 13:24786-24794. [PMID: 34837691 PMCID: PMC8660615 DOI: 10.18632/aging.203715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/27/2021] [Indexed: 11/25/2022]
Abstract
Fraser syndrome is a rare autosomal recessive malformation disorder. It is characterized by cryptophthalmos, syndactyly, urinary tract abnormalities and ambiguous genitalia. This condition is due to homozygous or heterozygous mutations in the FRAS1, FREM1, FREM2, and GRIP1 genes. In the present study, we recruited a Chinese family with Fraser syndrome. Two novel mutations c.7542_7543insG and c.2689C>T in the FREM2 gene were detected in this Fraser syndrome family by PCR-based sequencing. The next-generation sequencing-based single nucleotide polymorphism haplotyping method was applied to exclude these two mutations in 9 blastocysts obtained from the patient. After obtaining consent and informing the risk, the patient received in vitro fertilization and embryo transfer treatment with an embryo carrying a heterozygous mutation. Finally, she delivered a healthy baby without any complications on March 17, 2019. In conclusion, we first reported two novel mutations in the FREM2 gene associated with the risk of Fraser syndrome. Moreover, we described a next-generation sequencing-based single nucleotide polymorphism haplotyping method to select the ‘right’ embryos from patients with Fraser syndrome for in vitro fertilization and embryo transfer treatment.
Collapse
|
46
|
Lund AM, Wibrand F, Skogstrand K, Bækvad-Hansen M, Gregersen N, Andresen BS, Hougaard DM, Dunø M, Olsen RKJ. Use of Molecular Genetic Analyses in Danish Routine Newborn Screening. Int J Neonatal Screen 2021; 7:ijns7030050. [PMID: 34449524 PMCID: PMC8395600 DOI: 10.3390/ijns7030050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 12/20/2022] Open
Abstract
Historically, the analyses used for newborn screening (NBS) were biochemical, but increasingly, molecular genetic analyses are being introduced in the workflow. We describe the application of molecular genetic analyses in the Danish NBS programme and show that second-tier molecular genetic testing is useful to reduce the false positive rate while simultaneously providing information about the precise molecular genetic variant and thus informing therapeutic strategy and easing providing information to parents. When molecular genetic analyses are applied as second-tier testing, valuable functional data from biochemical methods are available and in our view, such targeted NGS technology should be implemented when possible in the NBS workflow. First-tier NGS technology may be a promising future possibility for disorders without a reliable biomarker and as a general approach to increase the adaptability of NBS for a broader range of genetic diseases, which is important in the current landscape of quickly evolving new therapeutic possibilities. However, studies on feasibility, sensitivity, and specificity are needed as well as more insight into what views the general population has towards using genetic analyses in NBS. This may be sensitive to some and could have potentially negative consequences for the NBS programme.
Collapse
Affiliation(s)
- Allan Meldgaard Lund
- Center for Inherited Metabolic Disorders, Departments of Clinical Genetics and Pediatrics, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Correspondence: ; Fax: +45-35454072
| | - Flemming Wibrand
- Metabolic Laboratory, Department of Clinical Genetics, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
| | - Kristin Skogstrand
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institute, 2300 Copenhagen, Denmark; (K.S.); (M.B.-H.); (D.M.H.)
| | - Marie Bækvad-Hansen
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institute, 2300 Copenhagen, Denmark; (K.S.); (M.B.-H.); (D.M.H.)
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, 8200 Aarhus, Denmark; (N.G.); (R.K.J.O.)
| | - Brage Storstein Andresen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark;
| | - David M. Hougaard
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institute, 2300 Copenhagen, Denmark; (K.S.); (M.B.-H.); (D.M.H.)
| | - Morten Dunø
- Molecular Genetics Laboratory, Department of Clinical Genetics, Copenhagen University Hospital, 2100 Copenhagen, Denmark;
| | - Rikke Katrine Jentoft Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, 8200 Aarhus, Denmark; (N.G.); (R.K.J.O.)
| |
Collapse
|
47
|
Dima V. Actualities in neonatal endocrine and metabolic screening. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2021; 17:416-421. [PMID: 35342476 PMCID: PMC8919488 DOI: 10.4183/aeb.2021.416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Neonatal metabolic screening has proven to be an important tool for the early detection of innate metabolic errors. Despite the fact that simple and effective methods of testing for metabolic diseases have been identified since the middle of the twentieth century, no consensus has been reached so far on the content of neonatal metabolic screening panels. There are large differences between countries in the number of metabolic diseases identified through national metabolic screening programs, ranging from zero to several tens, the most common testing being for phenylketonuria and congenital hypothyroidism (including in Romania). Given the fact that rare but treatable diseases have been identified in recent decades, reducing the financial burden on the health system, it would be useful to include them in the national neonatal metabolic screening program.
Collapse
Affiliation(s)
- V. Dima
- Correspondence to: Vlad Dima MD, “Filantropia” Clinical Hospital of Obstetrics and Gynecology, 11-13 Ion Mihalache Blvd., Bucharest, 011171, Romania, E-mail:
| |
Collapse
|