Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Genet Med ; 26(5): 101076, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38258669

RESUMEN

PURPOSE: Genome sequencing (GS)-specific diagnostic rates in prospective tightly ascertained exome sequencing (ES)-negative intellectual disability (ID) cohorts have not been reported extensively. METHODS: ES, GS, epigenetic signatures, and long-read sequencing diagnoses were assessed in 74 trios with at least moderate ID. RESULTS: The ES diagnostic yield was 42 of 74 (57%). GS diagnoses were made in 9 of 32 (28%) ES-unresolved families. Repeated ES with a contemporary pipeline on the GS-diagnosed families identified 8 of 9 single-nucleotide variations/copy-number variations undetected in older ES, confirming a GS-unique diagnostic rate of 1 in 32 (3%). Episignatures contributed diagnostic information in 9% with GS corroboration in 1 of 32 (3%) and diagnostic clues in 2 of 32 (6%). A genetic etiology for ID was detected in 51 of 74 (69%) families. Twelve candidate disease genes were identified. Contemporary ES followed by GS cost US$4976 (95% CI: $3704; $6969) per diagnosis and first-line GS at a cost of $7062 (95% CI: $6210; $8475) per diagnosis. CONCLUSION: Performing GS only in ID trios would be cost equivalent to ES if GS were available at $2435, about a 60% reduction from current prices. This study demonstrates that first-line GS achieves higher diagnostic rate than contemporary ES but at a higher cost.


Asunto(s)
Secuenciación del Exoma , Exoma , Discapacidad Intelectual , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico , Masculino , Femenino , Exoma/genética , Secuenciación del Exoma/economía , Estudios de Cohortes , Pruebas Genéticas/economía , Pruebas Genéticas/métodos , Secuenciación Completa del Genoma/economía , Niño , Genoma Humano/genética , Variaciones en el Número de Copia de ADN/genética , Polimorfismo de Nucleótido Simple/genética , Preescolar
2.
Sci Rep ; 12(1): 609, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35022470

RESUMEN

Human exome sequencing is a classical method used in most medical genetic applications. The leaders in the field are the manufacturers of enrichment kits based on hybridization of cRNA or cDNA biotinylated probes specific for a genomic region of interest. Recently, the platforms manufactured by the Chinese company MGI Tech have become widespread in Europe and Asia. The reliability and quality of the obtained data are already beyond any doubt. However, only a few kits compatible with these sequencers can be used for such specific tasks as exome sequencing. We developed our own solution for library pre-capture pooling and exome enrichment with Agilent probes. In this work, using a set of the standard benchmark samples from the Platinum Genome collection, we demonstrate that the qualitative and quantitative parameters of our protocol which we called "RSMU_exome" exceed those of the MGI Tech kit. Our protocol allows for identifying more SNV and indels, generates fewer PCR duplicates, enables pooling of more samples in a single enrichment procedure, and requires less raw data to obtain results comparable with the MGI Tech's protocol. The cost of our protocol is also lower than that of MGI Tech's solution.


Asunto(s)
Sondas de ADN , Secuenciación del Exoma/normas , Composición de Base , Humanos , Mutación INDEL , Polimorfismo de Nucleótido Simple , Secuenciación del Exoma/economía
3.
Eur J Hum Genet ; 30(1): 117-125, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34690354

RESUMEN

This single-center study aims to determine the time, diagnostic procedure, and cost saving potential of early exome sequencing in a cohort of 111 individuals with genetically confirmed neurodevelopmental disorders. We retrospectively collected data regarding diagnostic time points and procedures from the individuals' medical histories and developed criteria for classifying diagnostic procedures in terms of requirement, followed by a cost allocation. All genetic variants were re-evaluated according to ACMG recommendations and considering the individuals' phenotype. Individuals who developed first symptoms of their underlying genetic disorder when Next Generation Sequencing (NGS) diagnostics were already available received a diagnosis significantly faster than individuals with first symptoms before this cutoff. The largest amount of potentially dispensable diagnostics was found in genetic, metabolic, and cranial magnetic resonance imaging examinations. Out of 407 performed genetic examinations, 296 (72.7%) were classified as potentially dispensable. The same applied to 36 (27.9%) of 129 cranial magnetic resonance imaging and 111 (31.8%) of 349 metabolic examinations. Dispensable genetic examinations accounted 302,947.07€ (90.2%) of the total 335,837.49€ in potentially savable costs in this cohort. The remaining 32,890.42€ (9.8%) are related to non-required metabolic and cranial magnetic resonance imaging diagnostics. On average, the total potentially savable costs in our study amount to €3,025.56 per individual. Cost savings by first tier exome sequencing lie primarily in genetic, metabolic, and cMRI testing in this German cohort, underscoring the utility of performing exome sequencing at the beginning of the diagnostic pathway and the potential for saving diagnostic costs and time.


Asunto(s)
Discapacidades del Desarrollo/genética , Secuenciación del Exoma/métodos , Pruebas Genéticas/métodos , Enfermedades Raras/genética , Espasmos Infantiles/genética , Adolescente , Niño , Preescolar , Costos y Análisis de Costo , Discapacidades del Desarrollo/patología , Pruebas Genéticas/economía , Humanos , Lactante , Enfermedades Raras/diagnóstico , Espasmos Infantiles/patología , Secuenciación del Exoma/economía , Adulto Joven
4.
Sci Rep ; 11(1): 19099, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34580403

RESUMEN

Exome sequencing (ES) is an important diagnostic tool for individuals with neurodevelopmental disorders (NDD) and/or multiple congenital anomalies (MCA). However, the cost of ES limits the test's accessibility for many patients. We evaluated the yield of publicly funded clinical ES, performed at a tertiary center in Israel, over a 3-year period (2018-2020). Probands presented with (1) moderate-to-profound global developmental delay (GDD)/intellectual disability (ID); or (2) mild GDD/ID with epilepsy or congenital anomaly; and/or (3) MCA. Subjects with normal chromosomal microarray analysis who met inclusion criteria were included, totaling 280 consecutive cases. Trio ES (proband and parents) was the default option. In 252 cases (90.0%), indication of NDD was noted. Most probands were males (62.9%), and their mean age at ES submission was 9.3 years (range 1 month to 51 years). Molecular diagnosis was reached in 109 probands (38.9%), mainly due to de novo variants (91/109, 83.5%). Disease-causing variants were identified in 92 genes, 15 of which were implicated in more than a single case. Male sex, families with multiple-affected members and premature birth were significantly associated with lower ES yield (p < 0.05). Other factors, including MCA and coexistence of epilepsy, autism spectrum disorder, microcephaly or abnormal brain magnetic resonance imaging findings, were not associated with the yield. To conclude, our findings support the utility of clinical ES in a real-world setting, as part of a publicly funded genetic workup for individuals with GDD/ID and/or MCA.


Asunto(s)
Anomalías Múltiples/diagnóstico , Secuenciación del Exoma/economía , Financiación Gubernamental , Pruebas Genéticas/economía , Trastornos del Neurodesarrollo/diagnóstico , Anomalías Múltiples/economía , Anomalías Múltiples/genética , Adolescente , Adulto , Niño , Preescolar , Análisis Costo-Beneficio , Estudios de Factibilidad , Femenino , Asesoramiento Genético/economía , Asesoramiento Genético/métodos , Asesoramiento Genético/estadística & datos numéricos , Pruebas Genéticas/métodos , Pruebas Genéticas/estadística & datos numéricos , Humanos , Lactante , Recién Nacido , Israel , Masculino , Edad Materna , Trastornos del Neurodesarrollo/economía , Trastornos del Neurodesarrollo/genética , Edad Paterna , Embarazo , Diagnóstico Prenatal/economía , Diagnóstico Prenatal/métodos , Evaluación de Programas y Proyectos de Salud , Estudios Retrospectivos , Centros de Atención Terciaria/economía , Centros de Atención Terciaria/estadística & datos numéricos , Secuenciación del Exoma/estadística & datos numéricos , Adulto Joven
5.
Nature ; 597(7877): 555-560, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497419

RESUMEN

The immune microenvironment influences tumour evolution and can be both prognostic and predict response to immunotherapy1,2. However, measurements of tumour infiltrating lymphocytes (TILs) are limited by a shortage of appropriate data. Whole-exome sequencing (WES) of DNA is frequently performed to calculate tumour mutational burden and identify actionable mutations. Here we develop T cell exome TREC tool (T cell ExTRECT), a method for estimation of T cell fraction from WES samples using a signal from T cell receptor excision circle (TREC) loss during V(D)J recombination of the T cell receptor-α gene (TCRA (also known as TRA)). TCRA T cell fraction correlates with orthogonal TIL estimates and is agnostic to sample type. Blood TCRA T cell fraction is higher in females than in males and correlates with both tumour immune infiltrate and presence of bacterial sequencing reads. Tumour TCRA T cell fraction is prognostic in lung adenocarcinoma. Using a meta-analysis of tumours treated with immunotherapy, we show that tumour TCRA T cell fraction predicts immunotherapy response, providing value beyond measuring tumour mutational burden. Applying T cell ExTRECT to a multi-sample pan-cancer cohort reveals a high diversity of the degree of immune infiltration within tumours. Subclonal loss of 12q24.31-32, encompassing SPPL3, is associated with reduced TCRA T cell fraction. T cell ExTRECT provides a cost-effective technique to characterize immune infiltrate alongside somatic changes.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/citología , Linfocitos T/metabolismo , Adenocarcinoma del Pulmón/diagnóstico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/terapia , Ácido Aspártico Endopeptidasas/genética , Estudios de Cohortes , Exoma/genética , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Mutación , Neoplasias/diagnóstico , Neoplasias/genética , Pronóstico , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Secuenciación del Exoma/economía
6.
J Mol Diagn ; 23(7): 894-905, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33962052

RESUMEN

Massively parallel sequencing has markedly improved mendelian diagnostic rates. This study assessed the effects of custom alterations to a diagnostic genomic bioinformatic pipeline in response to clinical need and derived practice recommendations relative to diagnostic rates and efficiency. The Genomic Annotation and Interpretation Application (GAIA) bioinformatics pipeline was designed to detect panel, exome, and genome sample integrity and prioritize gene variants in mendelian disorders. Reanalysis of selected negative cases was performed after improvements to the pipeline. GAIA improvements and their effect on sensitivity are described, including addition of a PubMed search for gene-disease associations not in the Online Mendelian Inheritance of Man database, inclusion of a process for calling low-quality variants (known as QPatch), and gene symbol nomenclature consistency checking. The new pipeline increased the diagnostic rate and reduced staff costs, resulting in a saving of US$844.34 per additional diagnosis. Recommendations for genomic analysis pipeline requirements are summarized. Clinically responsive bioinformatics pipeline improvements increase diagnostic sensitivity and increase cost-effectiveness.


Asunto(s)
Secuenciación del Exoma/métodos , Enfermedades Genéticas Congénitas/genética , Pruebas Genéticas/métodos , Genómica/métodos , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis Costo-Beneficio , Exoma , Pruebas Genéticas/economía , Genoma Humano , Genómica/economía , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Mutación INDEL , Fenotipo , Polimorfismo de Nucleótido Simple , Sensibilidad y Especificidad , Secuenciación del Exoma/economía
7.
Pan Afr Med J ; 38: 111, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33912281

RESUMEN

Millions of patients, with suspected complex neurogenetic disorders, living in resource limited regions around the world have no access to genetic testing despite the rapidly expanding availability and decreasing costs of genetic testing in first world nations. The barriers to increasing availability of genetic testing in resource limited nations are multifactorial but can be attributed, in large part, to a lack of awareness of the power of genetic testing to lead to a rapid, cost-effective, diagnosis that potentially will have profound clinical implications on treatment and patient outcomes. We report our experience with whole exome sequencing (WES) done for the first time in 5 patients of African descent with a suspected neurogenetic disorder living in a resource limited setting on the Eastern Caribbean island of Barbados. A diagnostic pathogenic mutation was found in 3 patients in the SCN1A, STXBP1 and SCN4A, who clinically were diagnosed with Dravet syndrome, Lennox-Gastaut syndrome, paramytonia and seizures respectively. A variant of undetermined significance was found in a patient with global developmental delays, hypotonia, with abnormal eye movements. In one patient WES was non-diagnostic. This result highlights the high yield of WES in carefully selected patients with a neurologic disease and the need for increase access to genetic testing in resource limited settings globally.


Asunto(s)
Secuenciación del Exoma/métodos , Pruebas Genéticas/métodos , Enfermedades del Sistema Nervioso/diagnóstico , Adulto , Barbados , Niño , Análisis Costo-Beneficio , Pruebas Genéticas/economía , Humanos , Lactante , Proteínas Munc18/genética , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/fisiopatología , Secuenciación del Exoma/economía , Adulto Joven
8.
Laryngoscope ; 131(7): E2371-E2377, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33382469

RESUMEN

OBJECTIVES/HYPOTHESIS: To assess the relative cost-effectiveness of exome sequencing for isolated congenital deafness compared with standard care. STUDY DESIGN: Incremental cost-effectiveness and cost-benefit analyses were undertaken from the perspective of the Australian healthcare system using an 18-year time horizon. METHODS: A decision tree was used to model the costs and outcomes associated with exome sequencing and standard care for infants presenting with isolated congenital deafness. RESULTS: Exome sequencing resulted in an incremental cost of AU$1,000 per child and an additional 30 diagnoses per 100 children tested. The incremental cost-effectiveness ratio was AU$3,333 per additional diagnosis. The mean societal willingness to pay for exome sequencing was estimated at AU$4,600 per child tested relative to standard care, resulting in a positive net benefit of AU$3,600. Deterministic and probabilistic sensitivity analyses confirmed the cost-effectiveness of exome sequencing. CONCLUSIONS: Our findings demonstrate the cost-effectiveness of exome sequencing in congenital hearing loss, through increased diagnostic rate and consequent improved process of care by reducing or ceasing diagnostic investigation or facilitating targeted further investigation. We recommend equitable funding for exome sequencing in infants presenting with isolated congenital hearing loss. LEVEL OF EVIDENCE: N/A. Laryngoscope, 131:E2371-E2377, 2021.


Asunto(s)
Análisis Costo-Beneficio , Secuenciación del Exoma/economía , Pruebas Genéticas/economía , Pérdida Auditiva Sensorineural/diagnóstico , Nivel de Atención/economía , Australia , Femenino , Pruebas Genéticas/métodos , Pruebas Genéticas/estadística & datos numéricos , Costos de la Atención en Salud/estadística & datos numéricos , Pérdida Auditiva Sensorineural/congénito , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/terapia , Humanos , Lactante , Masculino , Nivel de Atención/estadística & datos numéricos , Secuenciación del Exoma/estadística & datos numéricos
9.
Am J Med Genet A ; 182(7): 1601-1607, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32369273

RESUMEN

The utility of whole exome analysis has been extensively demonstrated in research settings, but its clinical utility as a first-tier genetic test has not been well documented from diagnostic and health economic standpoints in real-life clinical settings. We performed medical exome analyses focusing on a clinically interpretable portion of the genome (4,813 genes) as a first-tier genetic test for 360 consecutive patients visiting a genetics clinic at a tertiary children's hospital in Japan, over a 3-year period. Bioinformatics analyses were conducted using standard software. A molecular diagnosis was made in 171 patients involving a total of 107 causative genes. Among these 107 causative genes, 57 genes were classified as genes with potential organ-specific interventions and management strategies. Clinically relevant results were obtained in 26% of the total cohort and 54% of the patients with a definitive molecular diagnosis. Performing the medical exome analysis at the time of the initial visit to the tertiary center, rather than after visits to pertinent specialists, brain MRI examination, and G-banded chromosome testing, would have reduced the financial cost by 197 euros according to retrospective calculation under multiple assumption. The present study demonstrated a high diagnostic yield (47.5%) for singleton medical exome analysis as a first-tier test in a real-life setting. Medical exome analysis yielded clinically relevant information in a quarter of the total patient cohort. The application of genomic testing during the initial visit to a tertiary medical center could be a rational approach to the diagnosis of patients with suspected genetic disorders.


Asunto(s)
Secuenciación del Exoma/economía , Enfermedades Genéticas Congénitas/diagnóstico , Pruebas Genéticas/economía , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Encéfalo/diagnóstico por imagen , Preescolar , Estudios de Cohortes , Biología Computacional , Análisis Costo-Beneficio , Enfermedades Genéticas Congénitas/economía , Enfermedades Genéticas Congénitas/genética , Pruebas Genéticas/métodos , Humanos , Lactante , Japón , Imagen por Resonancia Magnética , Centros de Atención Terciaria , Secuenciación del Exoma/métodos
11.
Fetal Diagn Ther ; 47(7): 554-564, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31962312

RESUMEN

INTRODUCTION: In light of the prospective Prenatal Assessment of Genomes and Exomes (PAGE) study, this paper aimed to determine the additional costs of using exome sequencing (ES) alongside or in place of chromosomal microarray (CMA) in a fetus with an identified congenital anomaly. METHODS: A decision tree was populated using data from a prospective cohort of women undergoing invasive diagnostic testing. Four testing strategies were evaluated: CMA, ES, CMA followed by ES ("stepwise"); CMA and ES combined. RESULTS: When ES is priced at GBP 2,100 (EUR 2,407/USD 2,694), performing ES alone prenatally would cost a further GBP 31,410 (EUR 36,001/USD 40,289) per additional genetic diagnosis, whereas the stepwise would cost a further GBP 24,657 (EUR 28,261/USD 31,627) per additional genetic diagnosis. When ES is priced at GBP 966 (EUR 1,107/USD 1,239), performing ES alone prenatally would cost a further GBP 11,532 (EUR 13,217/USD 14,792) per additional genetic diagnosis, whereas the stepwise would cost a further additional GBP 11,639 (EUR 13,340/USD 14,929) per additional genetic diagnosis. The sub-group analysis suggests that performing stepwise on cases indicative of multiple anomalies at ultrasound scan (USS) compared to cases indicative of a single anomaly, is more cost-effective compared to using ES alone. DISCUSSION/CONCLUSION: Performing ES alongside CMA is more cost-effective than ES alone, which can potentially lead to improvements in pregnancy management. The direct effects of test results on pregnancy outcomes were not examined; therefore, further research is recommended to examine changes on the projected incremental cost-effectiveness ratios.


Asunto(s)
Análisis Costo-Beneficio/métodos , Secuenciación del Exoma/economía , Exoma/genética , Pruebas Genéticas/economía , Análisis de Secuencia por Matrices de Oligonucleótidos/economía , Ultrasonografía Prenatal/economía , Estudios de Cohortes , Árboles de Decisión , Femenino , Estudios de Seguimiento , Pruebas Genéticas/métodos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Embarazo , Estudios Prospectivos , Ultrasonografía Prenatal/métodos , Secuenciación del Exoma/métodos
12.
J Med Genet ; 57(7): 479-486, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31980565

RESUMEN

BACKGROUND: This study provides an integrated assessment of the economic and social impacts of genomic sequencing for the detection of monogenic disorders resulting in intellectual disability (ID). METHODS: Multiple knowledge bases were cross-referenced and analysed to compile a reference list of monogenic disorders associated with ID. Multiple literature searches were used to quantify the health and social costs for the care of people with ID. Health and social expenditures and the current cost of whole-exome sequencing and whole-genome sequencing were quantified in relation to the more common causes of ID and their impact on lifespan. RESULTS: On average, individuals with ID incur annual costs in terms of health costs, disability support, lost income and other social costs of US$172 000, accumulating to many millions of dollars over a lifetime. CONCLUSION: The diagnosis of monogenic disorders through genomic testing provides the opportunity to improve the diagnosis and management, and to reduce the costs of ID through informed reproductive decisions, reductions in unproductive diagnostic tests and increasingly targeted therapies.


Asunto(s)
Secuenciación del Exoma/economía , Genómica/economía , Discapacidad Intelectual/economía , Discapacidad Intelectual/genética , Costos de la Atención en Salud/estadística & datos numéricos , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/epidemiología
13.
Pediatr Int ; 62(5): 556-561, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31955471

RESUMEN

BACKGROUND: Genetic testing has enabled the diagnosis of multiple congenital anomalies and/or intellectual disabilities. However, because of the phenotypic variability in these disorders, selection of an appropriate genetic test can be difficult and complex. For clinical examination, particularly in clinical facilities, a simple and standardized system is needed. METHODS: We compared microarray comparative genomic hybridization and clinical exome sequencing with regard to diagnostic yield, cost, and time required to reach a definitive diagnosis. After first performing G-banding for 200 patients with multiple congenital anomalies and/or intellectual disability, as a subsequent genetic test, microarray and clinical exome sequencing were compared with regard to diagnostic yield, cost, and time required. RESULTS: There was no obvious difference in the diagnostic rate between the two methods; however, clinical exome sequencing was superior in terms of cost and time. In addition, clinical exome sequencing could sufficiently identify copy number variants, and even smaller copy number variants could be identified. CONCLUSIONS: Clinical exome sequencing should be implemented earlier as a genetic test for undiagnosed patients with multiple congenital anomalies and/or intellectual disabilities. Our results can be used to establish inspection methods in clinical facilities.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Pruebas Genéticas/métodos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Niño , Preescolar , Hibridación Genómica Comparativa/economía , Hibridación Genómica Comparativa/métodos , Variaciones en el Número de Copia de ADN , Pruebas Genéticas/economía , Humanos , Análisis por Micromatrices/métodos , Secuenciación del Exoma/economía , Secuenciación del Exoma/métodos
14.
Genet Med ; 22(2): 283-291, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31501586

RESUMEN

PURPOSE: Exome sequencing (ES) has the potential to improve management of congenital anomalies and neurodevelopmental disorders in fetuses, infants, and children. US payers are key stakeholders in patient access to ES. We examined how payers view insurance coverage and clinical utility of pediatric and prenatal ES. METHODS: We employed the framework approach of qualitative research to conduct this study. The study cohort represented 14 payers collectively covering 170,000,000 enrollees. RESULTS: Seventy-one percent of payers covered pediatric ES despite perceived insufficient evidence because they saw merit in available interventions or in ending the diagnostic odyssey. None covered prenatal ES, because they saw no merit. For pediatric ES, 50% agreed with expanded aspects of clinical utility (e.g., information utility), and 21% considered them sufficient for coverage. For prenatal ES, payers saw little utility until in utero interventions become available. CONCLUSION: The perceived merit of ES is becoming a factor in payers' coverage for serious diseases with available interventions, even when evidence is perceived insufficient. Payers' views on ES's clinical utility are expanding to include informational utility, aligning with the views of patients and other stakeholders. Our findings inform clinical research, patient advocacy, and policy-making, allowing them to be more relevant to payers.


Asunto(s)
Secuenciación del Exoma/economía , Cobertura del Seguro/economía , Diagnóstico Prenatal/economía , Adulto , Secuencia de Bases/genética , Mapeo Cromosómico/métodos , Exoma/genética , Femenino , Pruebas Genéticas/economía , Genómica/métodos , Humanos , Masculino , Persona de Mediana Edad , Formulación de Políticas , Investigación Cualitativa , Participación de los Interesados , Encuestas y Cuestionarios , Estados Unidos , Secuenciación del Exoma/métodos
15.
Genet Med ; 22(2): 292-300, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31462755

RESUMEN

PURPOSE: This study describes the cost trajectory of the standard diagnostic care pathway for children with suspected genetic disorders in British Columbia, Canada. METHODS: Average annual per-patient costs were estimated using medical records review and a caregiver survey for a cohort of 498 children referred to BC Children's and Women's Hospitals (C&W) with unexplained intellectual disability (the TIDE-BC study) and families enrolled in the CAUSES study, which offered diagnostic genome-wide sequencing (GWS; exome and genome sequencing) to 500 families of children with suspected genetic disorders. RESULTS: Direct costs peaked in the first year of patients' diagnostic odyssey, with an average of C$2257 per patient (95% confidence interval [CI] C$2074, C$2441) for diagnostic testing and C$631 (95% CI C$543, C$727) for specialist consultations at C&W. In subsequent years, direct costs accrued at a constant rate, with an estimated annual per-patient cost of C$511 (95% CI C$473, C$551) for diagnostic testing and C$334 (95% CI C$295, C$369) for consultations at C&W. Travel costs and caregiver productivity loss associated with attending diagnosis-related physician appointments averaged C$1907/family/year. CONCLUSIONS: The continuing long-term accrual of costs by undiagnosed patients suggests that economic evaluations of diagnostic GWS services should use longer time horizons than have typically been used.


Asunto(s)
Enfermedades Genéticas Congénitas/economía , Pruebas Genéticas/economía , Costos de la Atención en Salud/tendencias , Adulto , Colombia Británica/epidemiología , Cuidadores/economía , Cuidadores/psicología , Estudios de Cohortes , Análisis Costo-Beneficio , Exoma/genética , Femenino , Costos de la Atención en Salud/ética , Humanos , Discapacidad Intelectual/genética , Masculino , Análisis de Secuencia de ADN/economía , Secuenciación del Exoma/economía , Secuenciación del Exoma/métodos
16.
Eur J Hum Genet ; 27(12): 1791-1799, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31320747

RESUMEN

Diagnostic exome sequencing (ES) can be performed on the proband only (singleton; sES) or with additional samples, often including both biological parents with the proband (trio; tES). In this study we sought to compare the efficiencies of exome sequencing (ES) by trio (tES) versus singleton (sES) approach, determine costs, and identify factors to consider when deciding on optimal implementation strategies for the diagnosis of monogenic disorders. We undertook ES in 30 trios and analysed each proband's sES and tES data in parallel. Two teams were randomly allocated to either sES or tES analysis for each case and blinded to each other's work. Each task was timed and cost analyses were based on time taken and diagnostic yield. We modelled three scenarios to determine the factors to consider in the implementation of tES. sES diagnosed 11/30 (36.7%) cases and tES identified one additional diagnosis (12/30 (40.0%)). tES obviated the need for Sanger segregation, reduced the number of variants for curation, and had lower cost-per-diagnosis when considering analysis alone. When sequencing costs were included, tES nearly doubled the cost of sES. Reflexing to tES in those who remain undiagnosed after sES was cost-saving over tES in all as first-line. This approach requires a large differential in diagnostic yield between sES and tES for maximal benefit given current sequencing costs. tES may be preferable when scaling up laboratory throughput due to efficiency gains and opportunity cost considerations. Our findings are relevant to clinicians, laboratories and health services considering tES over sES.


Asunto(s)
Análisis Costo-Beneficio/economía , Secuenciación del Exoma/normas , Enfermedades Genéticas Congénitas/genética , Pruebas Genéticas/normas , Adolescente , Adulto , Niño , Preescolar , Exoma/genética , Femenino , Enfermedades Genéticas Congénitas/diagnóstico , Pruebas Genéticas/economía , Humanos , Lactante , Masculino , Padres , Análisis de Secuencia de ADN/economía , Análisis de Secuencia de ADN/normas , Secuenciación del Exoma/economía , Adulto Joven
17.
Hum Mutat ; 40(9): 1346-1363, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31209962

RESUMEN

Intellectual disability (ID) and autism spectrum disorder (ASD) are clinically and genetically heterogeneous diseases. Recent whole exome sequencing studies indicated that genes associated with different neurological diseases are shared across disorders and converge on common functional pathways. Using the Ion Torrent platform, we developed a low-cost next-generation sequencing gene panel that has been transferred into clinical practice, replacing single disease-gene analyses for the early diagnosis of individuals with ID/ASD. The gene panel was designed using an innovative in silico approach based on disease networks and mining data from public resources to score disease-gene associations. We analyzed 150 unrelated individuals with ID and/or ASD and a confident diagnosis has been reached in 26 cases (17%). Likely pathogenic mutations have been identified in another 15 patients, reaching a total diagnostic yield of 27%. Our data also support the pathogenic role of genes recently proposed to be involved in ASD. Although many of the identified variants need further investigation to be considered disease-causing, our results indicate the efficiency of the targeted gene panel on the identification of novel and rare variants in patients with ID and ASD.


Asunto(s)
Trastorno del Espectro Autista/diagnóstico , Biología Computacional/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Discapacidad Intelectual/diagnóstico , Adolescente , Adulto , Trastorno del Espectro Autista/genética , Niño , Preescolar , Comorbilidad , Simulación por Computador , Minería de Datos , Bases de Datos Genéticas , Diagnóstico Precoz , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Discapacidad Intelectual/genética , Masculino , Mutación , Secuenciación del Exoma/economía , Secuenciación del Exoma/métodos , Adulto Joven
18.
Genet Med ; 21(11): 2586-2593, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31110331

RESUMEN

PURPOSE: To undertake the first end-to-end cost-effectiveness analysis of exome sequencing (ES) in rare disease diagnosis. METHODS: A cohort of 80 infants who underwent ES and usual diagnostic care in parallel were used to model incremental cost and health outcomes (quality adjusted life-years, QALYs) attributable to ES diagnosis over a 20-year horizon. Three models were developed: (1) outcomes in patients only, (2) outcomes in patients and first-degree relatives as a result of cascade testing, and (3) outcomes in patients and first-degree relatives including parental reproductive outcomes. RESULTS: When the directly observed cost and health outcomes of the cohort participants were projected, the use of ES resulted in a gain of 7.39 QALYs and an incremental cost-effectiveness ratio (ICER) of AU$31,144.35 (i.e., cost per additional QALY gained). When cascade testing in first-degree relatives was added, cost-effectiveness increased, to a gain of 11.62 QALYs and an ICER of AU$20,839.57. When parental reproductive outcomes were added, cost-effectiveness increased again, with 36.00 QALYs gained and an ICER of AU$14,235.28. CONCLUSION: Use of ES in suspected monogenic disorders becomes increasingly cost-effective as the benefits of ES data reanalysis, cascade testing in first-degree relatives, and parental reproductive outcomes are incorporated into modeling.


Asunto(s)
Secuenciación del Exoma/economía , Enfermedades Raras/diagnóstico , Enfermedades Raras/economía , Simulación por Computador , Análisis Costo-Beneficio/economía , Análisis Costo-Beneficio/métodos , Exoma/genética , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Años de Vida Ajustados por Calidad de Vida
19.
Pediatr Neurol ; 99: 47-54, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30952489

RESUMEN

BACKGROUND: Whole exome sequencing on family trios gives the highest diagnostic yield, but high cost limits its application. Here, we performed proband-only clinical exome sequencing in a population of patients with neurodevelopmental disabilities and tested the diagnostic yield. METHODS: This observational, retrospective study included 108 unrelated patients with neurodevelopmental disabilities who underwent clinical exome sequencing at the outpatient clinics of the Severance Children's Hospital, Seoul, South Korea, between March 2017 and May 2018. Clinical exome sequencing targeted 4503 disease-causing genes. RESULTS: The overall diagnostic rate was 38.0% (41 of 108) when proband-only clinical exome sequencing was performed without additional parental testing. Four sequence variants were reclassified as likely pathogenic after parental testing, representing an additional 3.7% of the diagnostic yield. The final diagnostic rate was 41.7% (45 of 108). Of 45 patients with genetic abnormalities, a total of 38 sequence variations were detected in 33 (30.6%) patients with five homozygous cases, and 13 chromosomal copy number variants were detected in 12 (11.1%) patients. Novel variants of known causal genes for neurodevelopmental disabilities were detected in 18 (16.7%) patients. These were variants that could be reclassified as likely pathogenic if the de novo nature of the mutation was confirmed after testing of parental samples. CONCLUSIONS: Proband-only clinical exome sequencing is a practical diagnostic tool that may be implemented in the clinical setting for patients with neurodevelopmental disabilities. A cost-effective approach to neurodevelopmental disabilities would be a proband-only clinical exome sequencing followed by parental testing of selective candidate variants.


Asunto(s)
Secuenciación del Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Trastornos del Neurodesarrollo/genética , Anomalías Múltiples/epidemiología , Anomalías Múltiples/genética , Edad de Inicio , Causalidad , Niño , Preescolar , Aberraciones Cromosómicas , Comorbilidad , Consenso , Variaciones en el Número de Copia de ADN , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Lactante , Recien Nacido Prematuro , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/genética , Masculino , Anotación de Secuencia Molecular , Mutación , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/epidemiología , Padres , Linaje , República de Corea/epidemiología , Estudios Retrospectivos , Convulsiones/epidemiología , Convulsiones/genética , Secuenciación del Exoma/economía
20.
Mol Genet Genomic Med ; 7(4): e00575, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30793526

RESUMEN

BACKGROUND: Detailed analysis of imprinting center (IC) defects in individuals with Prader-Willi syndrome (PWS) is not readily available beyond chromosomal microarray (MA) analysis, and such testing is important for a more accurate diagnosis and recurrence risks. This is the first feasibility study of newly developed droplet digital polymerase chain reaction (ddPCR) examining DNA copy number differences in the PWS IC region of those with IC defects. METHODS: The study cohort included 17 individuals without 15q11-q13 deletions or maternal disomy but with IC defects as determined by genotype analysis showing biparental inheritance. Seven sets of parents and two healthy, unrelated controls were also analyzed. RESULTS: Copy number differences were distinguished by comparing the number of positive droplets detected by IC probes to those from a chromosome 15 reference probe, GABRß3. The ddPCR findings were compared to results from other methods including MA, and whole-exome sequencing (WES) with 100% concordance. The study also estimated the frequency of IC microdeletions and identified gene variants by WES that may impact phenotypes including CPT2 and NTRK1 genes. CONCLUSION: Droplet digital polymerase chain reaction is a cost-effective method that can be used to confirm the presence of microdeletions in PWS with impact on genetic counseling and recurrence risks for families.


Asunto(s)
Secuenciación del Exoma/métodos , Pruebas Genéticas/métodos , Impresión Genómica , Síndrome de Prader-Willi/genética , Adolescente , Adulto , Niño , Preescolar , Cromosomas Humanos Par 15/genética , Femenino , Eliminación de Gen , Pruebas Genéticas/economía , Pruebas Genéticas/normas , Humanos , Masculino , Síndrome de Prader-Willi/diagnóstico , Secuenciación del Exoma/economía , Secuenciación del Exoma/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...