Next-generation phenotyping integrated in a national framework for patients with ultra-rare disorders improves genetic diagnostics and yields new molecular findings

Most individuals with rare diseases initially consult their primary care physician. For a subset of rare diseases, efficient diagnostic pathways are available. However, ultra-rare diseases often require both expert clinical knowledge and comprehensive genetic diagnostics, which poses structural challenges for public healthcare systems. To address these challenges within Germany, a novel structured diagnostic concept, based on multidisciplinary expertise at established university hospital centers for rare diseases (CRDs), was evaluated in the three year prospective study TRANSLATE NAMSE. A key goal of TRANSLATE NAMSE was to assess the clinical value of exome sequencing (ES) in the ultra-rare disease population. The aims of the present study were to perform a systematic investigation of the phenotypic and molecular genetic data of TRANSLATE NAMSE patients who had undergone ES in order to determine the yield of both ultra-rare diagnoses and novel gene-disease associations; and determine whether the complementary use of machine learning and artificial intelligence (AI) tools improved diagnostic effectiveness and efficiency. ES was performed for 1,577 patients (268 adult and 1,309 pediatric). Molecular genetic diagnoses were established in 499 patients (74 adult and 425 pediatric). A total of 370 distinct molecular genetic causes were established. The majority of these concerned known disorders, most of which were ultra-rare. During the diagnostic process, 34 novel and 23 candidate genotype-phenotype associations were delineated, mainly in individuals with neurodevelopmental disorders. To determine the likelihood that ES will lead to a molecular diagnosis in a given patient, based on the respective clinical features only, we developed a statistical framework called YieldPred. The genetic data of a subcohort of 224 individuals that also gave consent to the computer-assisted analysis of their facial images were processed with the AI tool Prioritization of Exome Data by Image Analysis (PEDIA) and showed superior performance in variant prioritization. The present analyses demonstrated that the novel structured diagnostic concept facilitated the identification of ultra-rare genetic disorders and novel gene-disease associations on a national level and that the machine learning and AI tools improved diagnostic effectiveness and efficiency for ultra-rare genetic disorders.

Heinen | Heiko | Kehrer | D. Horn | S. Mundlos | M. Spielmann | E. Schröck | T. Meitinger | T. Strom | L. Schöls | M. Holtgrewe | J. Hentschel | Alexej Knaus | J. Pantel | Tzung-Chien Hsieh | C. Ott | M. Sturm | Wagner | Nadja Ehmke | T. Haack | Nazanin Mirza-Schreiber | Hoffjan | Shahida Moosa | C. Kubisch | E. Graf | N. Donato | K. Cremer | M. Hempel | H. Engels | R. Betz | F. Hauck | A. Kaindl | I. Kurth | F. Kaiser | K. Platzer | T. Brunet | H. Graessner | O. Riess | C. Schramm | G. Demidov | R. Betz | S. Holzhauer | T. Rothoeft | J. Lemke | R. Jamra | Y. Hellenbroich | C. Rudolph | M. Elbracht | C. Grasemann | F. Brinkmann | K. Oexle | E. Mangold | P. Weydt | A. Kuechler | S. Hoffjan | C. Bührer | R. Berutti | C. Schuetz | K. Hinderhofer | C. Weiss | J. Hoefele | U. Kotzaeridou | F. Kraft | V. Schäfer | C. Schlein | P. Tacik | O. Kimmich | L. Zeltner | M. Krenn | C. Makowski | A. Bevot | Grigull | P. Bufler | U. Reuner | A. Muntau | L. Kaufmann | M. Rohlfs | Kobeleva | F. Brinkmann | Martje G. Pauly | Rebecca Herzog | M. Zawada | Martin | Klein | Magdalena Danyel | Hannah Klinkhammer | H. Lesmann | S. Sivalingam | A. Schmidt | S. Peters | M. Kreiß | C. Perne | Tim Bender | Luisa Averdunk | Ulrich Schatz | N. Weinhold | T. Kallinich | J. Neugebauer | C. Knopp | M. Mücke | J. Körholz | Martin Munteanu | Matias | K. Riedhammer | H. Nguyen | Réka | Britta Hanker | Christoph | Alexander | S. Beck-Wödl | A. Grüters-Kieslich | N. Weinhold | Castro-Goméz | Felix Boschann | Karakostas | K. Grundmann | M. Grobe-Einsler | Weigand | Kreiss | M. Schülke | Eva | V. Strehlow | Nóra | D. Choukair | Mańka | Fabian Brand | Franziska Rillig | Theresia Herget | A. Tibelius | J. Magg | M. Pamela | S. Schröder | M. Nöthen | Demet Önder | Sabine | C. Stieber | Eva M. C. Schwaibold | Matar | E. Schlapakow | Matthias Begemann | Aude-Annick Suter | N. Kaiser | M. Brugger | M. A. Mensah | C. Buehrer | C. Stoltenburg | F. Boschann | Christiane | Pantelis | S. Schwartzmann | Maximilian | Christian | D. Westphal | Markus | G. Hoffmann | Heike | T. Bäumer | Katja Lohmann | Katharina Mayerhanser | Martina | Sheetal Kumar | Ingo Borggraefe | Annekatrin Ripke | Henrike | Okun | U. Kornak | Sarah Bernsen | S. Moosa | A. Boesch | H. Sczakiel | H. Klinkhammer | A. Knaus | L. Averdunk | M. Danyel | T. Bender | M. Schuelke | Tanita Kretschmer | N. Matar | C. Weiss | J. Neugebauer | H. P. Nguyen | T. Klockgether | R. Hirtz | C. Weiler-Normann | Christina Weiler-Normann | M. Grobe‐Einsler | W. Müller-Felber | Reinhard Berner | I. Hüning | Jasmin Lisfeld | André | P. M. Krawitz | Sczakiel | Annemarie Bösch | -. Aude | Annick Suter | Tillmann Kallinich | Lorenz | Spier Isabel | H. André | Bender Tim | Alexandra Marzena Morawiec | Sergio | Ahmad Aziz | Xenia | Min | A. Lee-Kirsch | Amalia-Mihaela Hanßke | Kiewert Cordula | K. Ullrich | Groffmann | P. Schaaf | Bettendorf | Münchau | Kovacs | Vill Katharina | Krude | P. Krawitz | B. Tim | Riccardo Berutti | Henrike | Julia Körholz | Florian Kraft | Melanie Brugger | Matar

[1]  Tzung-Chien Hsieh,et al.  Improving Deep Facial Phenotyping for Ultra-rare Disorder Verification Using Model Ensembles , 2022, 2023 IEEE/CVF Winter Conference on Applications of Computer Vision (WACV).

[2]  M. Posada de la Paz,et al.  Diagnostic delay in rare diseases: data from the Spanish rare diseases patient registry , 2022, Orphanet Journal of Rare Diseases.

[3]  Nadja Ehmke,et al.  Novel mutation and expanding phenotype in IRF2BP2 deficiency. , 2022, Rheumatology.

[4]  Alexej Knaus,et al.  Next‐generation phenotyping contributing to the identification of a 4.7 kb deletion in KANSL1 causing Koolen‐de Vries syndrome , 2022, Human mutation.

[5]  The Genomes-Project-Pilot-Investigators 100,000 genomes pilot on rare-disease diagnosis in health care – preliminary report , 2022, Yearbook of Paediatric Endocrinology.

[6]  K. Platzer,et al.  AutoCaSc: Prioritizing candidate genes for neurodevelopmental disorders , 2022, Human mutation.

[7]  J. Lupski,et al.  A reverse genetics and genomics approach to gene paralog function and disease: Myokymia and the juxtaparanode. , 2022, American journal of human genetics.

[8]  M. Nöthen,et al.  The Interdisciplinary Diagnosis of Rare Diseases-Results of the Translate-NAMSE Project. , 2022, Deutsches Arzteblatt international.

[9]  M. Posada de la Paz,et al.  Diagnostic Process in Rare Diseases: Determinants Associated with Diagnostic Delay , 2022, International journal of environmental research and public health.

[10]  M. Nöthen,et al.  GestaltMatcher facilitates rare disease matching using facial phenotype descriptors , 2022, Nature Genetics.

[11]  C. Klein,et al.  An Integrated clinical pathway for diagnosis, treatment and care of rare diseases: model, operating procedures, and results of the project TRANSLATE-NAMSE funded by the German Federal Joint Committee , 2021, Orphanet Journal of Rare Diseases.

[12]  J. Gécz,et al.  Bi-allelic variants in SPATA5L1 lead to intellectual disability, spastic-dystonic cerebral palsy, epilepsy, and hearing loss. , 2021, American journal of human genetics.

[13]  C. Gieger,et al.  Blood DNA methylation provides an accurate biomarker of KMT2B-related dystonia and predicts onset. , 2021, Brain : a journal of neurology.

[14]  J. Lupski,et al.  High prevalence of multilocus pathogenic variation in neurodevelopmental disorders in the Turkish population. , 2021, American journal of human genetics.

[15]  S. Wortmann,et al.  A spoonful of L‐fucose—an efficient therapy for GFUS‐CDG, a new glycosylation disorder , 2021, EMBO molecular medicine.

[16]  D. Seelow,et al.  Biallelic truncating variants in ATP9A cause a novel neurodevelopmental disorder involving postnatal microcephaly and failure to thrive , 2021, Journal of Medical Genetics.

[17]  H. Kanegane,et al.  Heterozygous OAS1 gain-of-function variants cause an autoinflammatory immunodeficiency , 2021, Science Immunology.

[18]  H. Fröhlich,et al.  CADA: phenotype-driven gene prioritization based on a case-enriched knowledge graph , 2021, medRxiv.

[19]  J. Shendure,et al.  CADD-Splice—improving genome-wide variant effect prediction using deep learning-derived splice scores , 2021, Genome Medicine.

[20]  T. Strom,et al.  Impaired complex I repair causes recessive Leber’s hereditary optic neuropathy , 2021, The Journal of clinical investigation.

[21]  D. Horn,et al.  Biallelic truncating variants in MAPKAPK5 cause a new developmental disorder involving neurological, cardiac, and facial anomalies combined with synpolydactyly , 2021, Genetics in Medicine.

[22]  J. Pantel,et al.  Drop of Prevalence after Population Expansion: A lower prevalence for recessive disorders in a random mating population is a transient phenomenon during and after a growth phase , 2020, bioRxiv.

[23]  H. Hakonarson,et al.  Pathogenic variants in SMARCA5, a chromatin remodeler, cause a range of syndromic neurodevelopmental features , 2020, Science Advances.

[24]  M. Caulfield,et al.  An online compendium of treatable genetic disorders , 2020, American journal of medical genetics. Part C, Seminars in medical genetics.

[25]  T. Meitinger,et al.  A homozygous truncating variant in CCDC186 in an individual with epileptic encephalopathy , 2020, Annals of clinical and translational neurology.

[26]  Golder N Wilson,et al.  Variants in the SK2 channel gene (KCNN2) lead to dominant neurodevelopmental movement disorders. , 2020, Brain : a journal of neurology.

[27]  R. Mägi,et al.  The landscape of autosomal-recessive pathogenic variants in European populations reveals phenotype-specific effects , 2020, bioRxiv.

[28]  M. Hurles,et al.  Evaluating variants classified as pathogenic in ClinVar in the DDD Study , 2020, Genetics in Medicine.

[29]  Rebecca C. Spillmann,et al.  Mutation-specific pathophysiological mechanisms define different neurodevelopmental disorders associated with SATB1 dysfunction , 2020, bioRxiv.

[30]  David R. Murdock,et al.  Transcriptome-directed analysis for Mendelian disease diagnosis overcomes limitations of conventional genomic testing. , 2020, The Journal of clinical investigation.

[31]  Justyna A. Karolak,et al.  Low-level parental somatic mosaic SNVs in exomes from a large cohort of trios with diverse suspected Mendelian conditions , 2020, Genetics in Medicine.

[32]  Keith W. Muir,et al.  Whole-genome sequencing of patients with rare diseases in a national health system , 2020, Nature.

[33]  D. Seelow,et al.  VarFish: comprehensive DNA variant analysis for diagnostics and research , 2020, Nucleic Acids Res..

[34]  Melissa J. Landrum,et al.  ClinVar: improvements to accessing data , 2019, Nucleic Acids Res..

[35]  Ryan L. Collins,et al.  The mutational constraint spectrum quantified from variation in 141,456 humans , 2020, Nature.

[36]  William J. Astle,et al.  Whole genome sequencing of a sporadic primary immunodeficiency cohort , 2018, Nature.

[37]  Tudor I. Oprea,et al.  Therapies for rare diseases: therapeutic modalities, progress and challenges ahead , 2019, Nature Reviews Drug Discovery.

[38]  Kiely N. James,et al.  A Randomized, Controlled Trial of the Analytic and Diagnostic Performance of Singleton and Trio, Rapid Genome and Exome Sequencing in Ill Infants. , 2019, American journal of human genetics.

[39]  A. Olry,et al.  Estimating cumulative point prevalence of rare diseases: analysis of the Orphanet database , 2019, European Journal of Human Genetics.

[40]  S. Antonarakis Carrier screening for recessive disorders , 2019, Nature Reviews Genetics.

[41]  Tomas W. Fitzgerald,et al.  Clinically-relevant postzygotic mosaicism in parents and children with developmental disorders in trio exome sequencing data , 2019, Nature Communications.

[42]  T. Meitinger,et al.  Mitochondrial DNA mutation analysis from exome sequencing—A more holistic approach in diagnostics of suspected mitochondrial disease , 2019, Journal of inherited metabolic disease.

[43]  M. Nöthen,et al.  PEDIA: prioritization of exome data by image analysis , 2018, Genetics in Medicine.

[44]  Jae Min Lee,et al.  Molecular diagnosis of hereditary spherocytosis by multi-gene target sequencing in Korea: matching with osmotic fragility test and presence of spherocyte , 2019, Orphanet Journal of Rare Diseases.

[45]  Srinivas Aluru,et al.  Efficient Architecture-Aware Acceleration of BWA-MEM for Multicore Systems , 2019, 2019 IEEE International Parallel and Distributed Processing Symposium (IPDPS).

[46]  Jean M. Davidson,et al.  Identification of rare-disease genes using blood transcriptome sequencing and large control cohorts , 2019, Nature Medicine.

[47]  T. Wienker,et al.  Genetics of intellectual disability in consanguineous families , 2019, Molecular Psychiatry.

[48]  Borut Peterlin,et al.  PEDIA: prioritization of exome data by image analysis , 2018, Genetics in Medicine.

[49]  Tomas W. Fitzgerald,et al.  Making new genetic diagnoses with old data: iterative reanalysis and reporting from genome-wide data in 1,133 families with developmental disorders , 2018, Genetics in Medicine.

[50]  James D Stephenson,et al.  Quantifying the contribution of recessive coding variation to developmental disorders , 2017, Science.

[51]  Christine M. Cutillo,et al.  Future of Rare Diseases Research 2017–2027: An IRDiRC Perspective , 2017, Clinical and translational science.

[52]  Z. Ning,et al.  Split-Read Indel and Structural Variant Calling Using PINDEL. , 2018, Methods in molecular biology.

[53]  Tudor Groza,et al.  Matchmaker Exchange , 2017, Current protocols in human genetics.

[54]  Peter N. Robinson,et al.  International Cooperation to Enable the Diagnosis of All Rare Genetic Diseases , 2017, American journal of human genetics.

[55]  J. Michael Cherry,et al.  Evaluating the clinical validity of gene-disease associations: an evidence-based framework developed by the Clinical Genome Resource , 2017, bioRxiv.

[56]  Frank Klawonn,et al.  Diagnostic needs for rare diseases and shared prediagnostic phenomena: Results of a German-wide expert Delphi survey , 2017, PloS one.

[57]  Clara Gaff,et al.  Prospective comparison of the cost-effectiveness of clinical whole-exome sequencing with that of usual care overwhelmingly supports early use and reimbursement , 2017, Genetics in Medicine.

[58]  Donna M. Muzny,et al.  Resolution of Disease Phenotypes Resulting from Multilocus Genomic Variation , 2017, The New England journal of medicine.

[59]  Francesco Muntoni,et al.  Improving genetic diagnosis in Mendelian disease with transcriptome sequencing , 2016, Science Translational Medicine.

[60]  Brent S. Pedersen,et al.  Who’s Who? Detecting and Resolving Sample Anomalies in Human DNA Sequencing Studies with Peddy , 2016, bioRxiv.

[61]  F. Cunningham,et al.  The Ensembl Variant Effect Predictor , 2016, bioRxiv.

[62]  Yali Xue,et al.  BCFtools/RoH: a hidden Markov model approach for detecting autozygosity from next-generation sequencing data , 2016, Bioinform..

[63]  C. Webber,et al.  Systematic Phenomics Analysis Deconvolutes Genes Mutated in Intellectual Disability into Biologically Coherent Modules. , 2016, American journal of human genetics.

[64]  W. Chung,et al.  Clinical application of whole-exome sequencing across clinical indications , 2015, Genetics in Medicine.

[65]  Harry Hemingway,et al.  Health and population effects of rare gene knockouts in adult humans with related parents , 2015, Science.

[66]  Orion J. Buske,et al.  The Matchmaker Exchange: A Platform for Rare Disease Gene Discovery , 2015, Human mutation.

[67]  Bale,et al.  Standards and Guidelines for the Interpretation of Sequence Variants: A Joint Consensus Recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology , 2015, Genetics in Medicine.

[68]  M. Stephens,et al.  An Estimate of the Average Number of Recessive Lethal Mutations Carried by Humans , 2014, Genetics.

[69]  Stephan J Sanders,et al.  A framework for the interpretation of de novo mutation in human disease , 2014, Nature Genetics.

[70]  Damian Smedley,et al.  Jannovar: A Java Library for Exome Annotation , 2014, Human mutation.

[71]  D. G. MacArthur,et al.  Guidelines for investigating causality of sequence variants in human disease , 2014, Nature.

[72]  J. Shendure,et al.  A general framework for estimating the relative pathogenicity of human genetic variants , 2014, Nature Genetics.

[73]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[74]  Nicholas W. Wood,et al.  A robust model for read count data in exome sequencing experiments and implications for copy number variant calling , 2012, Bioinform..

[75]  Richard M Myers,et al.  Genomic patterns of homozygosity in worldwide human populations. , 2012, American journal of human genetics.

[76]  Heng Li,et al.  A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data , 2011, Bioinform..

[77]  M. DePristo,et al.  A framework for variation discovery and genotyping using next-generation DNA sequencing data , 2011, Nature Genetics.

[78]  E. van Binsbergen,et al.  Array analysis and karyotyping: workflow consequences based on a retrospective study of 36,325 patients with idiopathic developmental delay in the Netherlands. , 2009, European journal of medical genetics.

[79]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[80]  Jurg Ott,et al.  Genome‐wide autozygosity mapping in human populations , 2009, Genetic epidemiology.

[81]  A. Ballabio,et al.  Safety of arylsulfatase A overexpression for gene therapy of metachromatic leukodystrophy. , 2007, Human gene therapy.

[82]  A. Chakravarti,et al.  On consanguineous marriages and the genetic load , 2004, Human Genetics.