Skip to main content
Genetics in Medicine Open logoLink to Genetics in Medicine Open
. 2024 Mar 20;2:101840. doi: 10.1016/j.gimo.2024.101840

Applications of genome sequencing as a single platform for clinical constitutional genetic testing

Yao Yang 1,2, Daniela del Gaudio 3, Avni Santani 4, Stuart A Scott 1,2,
PMCID: PMC11736070  PMID: 39822265

Abstract

The number of human disease genes has dramatically increased over the past decade, largely fueled by ongoing advances in sequencing technologies. In parallel, the number of available clinical genetic tests has also increased, including the utilization of exome sequencing for undiagnosed diseases. Although most clinical sequencing tests have been centered on enrichment-based multigene panels and exome sequencing, the continued improvements in performance and throughput of genome sequencing suggest that this technology is emerging as a potential platform for routine clinical genetic testing. A notable advantage is a single workflow with the opportunity to reflexively interrogate content as clinically indicated; however, challenges with implementing routine clinical genome sequencing still remain. This review is centered on evaluating the applications of genome sequencing as a single platform for clinical constitutional genetic testing, including its potential utility for diagnostic testing, carrier screening, cytogenomic molecular karyotyping, prenatal testing, mitochondrial genome interrogation, and pharmacogenomic and polygenic risk score testing.

Keywords: Clinical genome sequencing, Genome sequencing applications, Long-read sequencing, Medical genomics, Short-read sequencing

Introduction

High-throughput sequencing is a widely used technology for clinical constitutional genetic testing, which is commonly implemented as enrichment-based multigene panels for diagnostic testing,1 carrier screening,2 and enrichment-based exome sequencing for undiagnosed disease.3,4 However, genome sequencing is increasingly being considered as a potential platform for clinical testing at some institutions and laboratories.5,6 In addition to the increased likelihood of Mendelian disease diagnosis, the operational advantages with clinical genome sequencing include a single laboratory workflow and the opportunity to reflexively interrogate additional clinical content when clinically indicated.7, 8, 9 However, several challenges with implementing routine clinical genome sequencing still remain, including cost-effectiveness, computational infrastructure, and both logistical and ethical considerations.

Genome sequencing has technical advantages over enrichment-based sequencing, including less bias and more consistent coverage across coding regions,10 as well as improved capacity to detect copy-number variants (CNVs) and breakpoints,11 and ongoing improvements in repeat expansion detection.12 Moreover, genome sequencing inherently has advantages with interrogating noncoding variants for both gene/variant discovery and clinical testing,13 and computational algorithms are continually improving to facilitate the interrogation of challenging homologous regions14 and repeat expansions.15 Data have also emerged indicating that genome sequencing has a higher diagnostic yield compared with exome sequencing in pediatric and other clinical service lines,16, 17, 18 including rapid genome sequencing for critically ill children.19 Although the vast majority of clinical genome sequencing currently uses short-read sequencing platforms, long-read sequencing platforms continue to improve in performance and throughput, which has translated to higher variant calling accuracy in historically challenging genomic contexts.20

As the accessibility of genome sequencing for clinical laboratories continues to improve, resources and regulatory recommendations that support clinical genome sequencing are continually evolving, including design recommendations, analytical validation resources, and professional guideline statements (Table 121, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46). This review is centered on evaluating the applications of genome sequencing for routine clinical constitutional genetic testing, including its utility as a technology for diagnostic testing, carrier screening, cytogenomic molecular karyotyping, prenatal testing, mitochondrial genome interrogation, and pharmacogenomic and polygenic risk score testing. Of note, ethics evaluations and thorough cost-effectiveness analyses of clinical genome sequencing, as well as prenatal cell-free DNA screening, were considered outside the scope of this review.

Table 1.

Resources for clinical genome sequencing development and validation

Category Organization/Entity Year Resource Reference
Professional Guidelines
CDC 2012 Clinical sequencing quality recommendations Gargis et al21
AMP 2012 Clinical genome sequencing recommendations Schrijver et al22
ACMG 2013 Clinical sequencing laboratory standards Rehm et al23
CAP 2014 Clinical sequencing proficiency standards Schrijver et al24
CAP 2015 Clinical sequencing laboratory standards Aziz et al25
CDC 2015 Clinical sequencing informatics best practices Gargis et al26
CCMG 2015 Clinical genome sequencing for monogenic disease statement Boycott et al27
ESHG 2016 Clinical sequencing guidelines Matthijs et al28
CAP 2017 Clinical sequencing development and validation recommendations Santani et al29
CAP 2017 Clinical exome/genome sequencing development and validation recommendations Hegde et al30
AMP/CAP 2018 Clinical sequencing bioinformatic validation guidelines Roy et al31
CAP 2019 Clinical sequencing design and implementation Santani et al32
CCMG 2019 Clinical sequencing laboratory guidelines Hume et al33
ACMG 2020 Clinical sequencing test development guidelines Bean et al34
MGI 2020 Clinical genome sequencing validation recommendations Marshall et al35
MGI 2020 Clinical genome sequencing utility Hayeems et al36
ACMG 2021 Clinical exome and genome sequencing guideline Manickam et al37
MGI 2022 Clinical genome sequencing interpretation and reporting Austin-Tse et al38
ESHG 2022 Clinical genome sequencing implementation recommendations Souche et al39
CLSI 2023 Cinical laboratory sequencing standards and recommendations CLSI47
Analytical Validation and Reference Materials
GIAB/NIST 2016 Genome sequencing benchmarking reference material Zook et al40
GIAB/NIST 2018 Reference material benchmarking best practices Cleveland et al41
GIAB/NIST 2019 Small-variant benchmarking reference material Zook et al42
GA4GH 2019 Small-variant benchmarking best practices Krusche et al43
GIAB/NIST 2020 Deletion/insertion benchmarking reference material Zook et al44
ClinGen/GeT-RM 2021 Clinical genome sequencing in silico reference material Wilcox et al45
GIAB/NIST 2022 Benchmarking reference material for challenging medically relevant genes Wagner et al46

ACMG, American College of Medical Genetics and Genomics; AMP, Association for Molecular Pathology; CAP, College of American Pathologists; CCMG, Canadian College of Medical Geneticists; CDC, Centers for Disease Control and Prevention; ClinGen, Clinical Genome Resource; CLSI, Clinical and Laboratory Standards Institute; ESHG, European Society of Human Genetics; GA4GH, Global Alliance for Genomics and Health; GeT-RM, Genetic Testing Reference Materials Coordination Program; GIAB, Genome in a Bottle Consortium; MGI, Medical Genome Initiative; NIST, National Institute of Standards and Technology.

Clinical constitutional genome sequencing

Short- and long-read genome sequencing

Short-read sequencing is the most commonly implemented platform for genome sequencing, which is driven by its throughput and cost-effectiveness, as well as the accessibility of the platform infrastructure. Clinical constitutional short-read genome sequencing applications typically require an average autosomal depth of >30 or 40× (Figure 1)35,48,49; however, low-pass (∼0.5-5×) short-read sequencing has also been used for germline copy-number profiling, as well as small-variant genotyping when coupled with imputation. However, long-read genome sequencing has recently emerged as an important alternative,18,50,51 based on its improved interrogation of clinically significant genomic regions (Figure 148,49), including expansions, structural variation, homologous regions, and the human leukocyte antigen (HLA) locus, as well as its capacity for variant phasing.52,53 Both single-molecule real-time (SMRT; HiFi; Pacific Biosciences) and nanopore (Oxford Nanopore Technologies) long-read genome sequencing have been used for constitutional variant detection applications using both high-depth and low-pass sequencing.54, 55, 56, 57 As such, future iterations of clinical genome sequencing could be developed as 2 parallel workflows to strategically leverage the benefits of either high-depth or low-pass sequencing depending on the intended use and economics of the application.

Figure 1.

Figure 1

Illustration of germline genome architecture and sequencing accessibility, as represented by6 clinically significant genes. From top left to bottom right: ADAMTSL2 (NM_014694.4; geleophysic dysplasia), CYP2D6 (NM_000106.6; drug metabolism), IKBKG (NM_001099857.5; incontinentia pigmenti), KRT86 (NM_001320198.2; monilethrix), OTOA (NM_144672.4; sensorineural hearing loss), and STRC (NM_153700.2; sensorineural hearing loss) (GRCh38). From inner to outer circles: previously reported exon-level sequencing “dead zones” across the genome (lifted over to GRCh38 from GRCh37; black)48; enrichment-based short-read exome sequencing coverage (average 185.8× across all coding regions; red); short-read genome sequencing coverage (average 33.2× across all coding regions; dark blue); long-read HiFi genome sequencing coverage (average 27.5× across all coding regions; light blue); and gene transcripts (introns: light green; exons: dark green). Noncoding exons were excluded from transcript tracks. All sequencing data were acquired from publicly available GIAB/NIST reference material sample NA12878 (HG001), filtered with mapping and base quality scores greater than Q20, and Circos images generated using R package circlize.49

Clinical genome sequencing resources

Although sequencing depth is largely considered the most critical quality metric for clinical genome sequencing, other quality metrics and thresholds are necessary to establish and monitor when developing a clinical genome sequencing assay (eg, callability and mappability).35 Importantly, support for the implementation of clinical genome sequencing is currently available from several venues (Table 121, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46), including guidelines from the College of American Pathologists (CAP),25 the American College of Medical Genetics and Genomics (ACMG),37 the Association for Molecular Pathology (AMP),31 and related professional consortia21,29,32,58; benchmarking reference materials from the Genome in a Bottle (GIAB)/National Institute of Standards and Technology (NIST) consortium,42 the Global Alliance for Genomics and Health consortium,43 the Coriell Cell Repository/CDC Genetic Testing Reference Materials Coordination Program59; and clinical genome sequencing recommendations from the Medical Genome Initiative.35,36,38 The expansion of available resources, including large publicly available genomic databases that support variant interpretation (eg, The Genome Aggregation Database60 and ClinVar61) and classification62 (Table 260, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76), suggest that clinical laboratories will continue to adopt genomic-based diagnostic platforms in the future.

Table 2.

Resources for clinical genome sequencing variant interpretation

Category Organization/Entity Year Resource Reference
Germline Sequence Variants
HGMD 2003 Collection of published germline variants implicated in human inherited disease Stenson et al63
AMP/ACMG 2015 Standards and guidelines for the interpretation of sequence variants Richards et al62
ClinGen 2015 SVI Working Group recommendations on using the ACMG/AMP guidelines Rehm et al64
ClinGen 2015 SVI Expert-Panel-specified ACMG/AMP variant interpretation guidelines Rehm et al64
ClinVar 2016 Database of genetic variation and clinical interpretation Landrum et al61
gnomAD 2017 Aggregated exome and genome sequencing population database Karczewski et al60
CPIC 2017 Standardized pharmacogenomic allele function and phenotype terms Caudle et al65
OMIM 2019 Database of human genes and genetic disorders Amberger et al66
UK-ACGS 2020 Practice guidelines for variant classification in rare disease Ellard et al67
PharmGKB 2021 Comprehensive resource of pharmacogenomic variation and knowledge Whirl-Carrillo et al68
TOPMed 2021 Aggregated genome sequencing population database Taliun et al69
GenCC 2022 Mendelian disease gene curation database DiStefano et al70
Germline Structural Variants
DECIPHER 2009 Interactive web-based database of human genomic variation and phenotypes Firth et al71
DGV 2014 Curated collection of structural variations in the human genome MacDonald et al72
ClinGen 2018 Human gene dosage sensitivity map Riggs et al73
ACMG/ClinGen 2020 Technical standards for the interpretation and reporting of constitutional CNVs Riggs et al74
Mitochondrial Variants
MitoMap 1996 Compendium of variants in human mtDNA Kogelnik et al75
ClinGen (mtDNA) 2020 Adapted ACMG/AMP standards and guidelines for mtDNA variant interpretation McCormick et al76

ACGS, Association for Clinical Genomic Science; ACMG, American College of Medical Genetics and Genomics; AMP, Association for Molecular Pathology; ClinGen, Clinical Genome Resource; CPIC, Clinical Pharmacogenetics Implementation Consortium; DGV, Database of Genomic Variants; GenCC, Gene Curation Coalition; gnomAD, Genome Aggregation Database; HGMD, Human Gene Mutation Database; OMIM, Online Mendelian Inheritance in Man; PharmGKB, Pharmacogenomics Knowledgebase; TOPMed, Trans-Omics for Precision Medicine.

Clinical genome sequencing applications

Diagnostic panels and exome sequencing

High-throughput sequencing of targeted gene panels and exome sequencing has resulted in a dramatic improvement over the traditional diagnostic pathway, which often included a prolonged diagnostic odyssey due to low diagnostic yields from sequential gene testing for Mendelian diseases. The first demonstrated use of exome sequencing for the diagnosis of a rare genetic condition of unknown etiology was reported in 2010,77 which prompted a series of publications on the utility of targeted panels and exome sequencing for the diagnosis of pediatric and adult patients with genetic diseases. In parallel to the last decade of high-throughput sequencing-based gene discovery research, professional societies (eg, ACMG and AMP), and regulatory agencies (eg, CAP) have published important clinical guidelines on the use of high-throughput sequencing in the diagnostic setting (Table 121, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46). Additionally, selected societies and federally funded consortia dedicated to gene and variant curation (eg, Clinical Genome Resource [ClinGen]64 and Gene Curation Coalition70) have emerged as central resources for diagnostic testing implementation and variant interpretation (Table 260, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76).

Diagnostic constitutional sequencing is commonly implemented as multigene enrichment-based panels for specific clinical indications, which have the advantage of being optimized for quality and depth of coverage across targeted regions of interest. However, technical limitations include restricted updates to panel content, variant interpretation limited to coding regions (unless specifically targeted), and reduced sensitivity for detecting CNV breakpoints (and single exon copy-number changes).78 In contrast, enrichment-based exome sequencing aims to capture all protein-coding exons representing ∼2% of the genome. In addition, exome sequencing is often implemented as family or trio based, which can facilitate proband variant interpretation under autosomal dominant, recessive, and/or X-linked inheritance models.34 As the cost of sequencing has decreased, exome sequencing has been rapidly adopted as a first-tier test for undiagnosed conditions, particularly among patients with syndromic indications or for whom traditional diagnostic modalities have been uninformative. However, although exome sequencing is considered to have strong diagnostic utility, its overall diagnostic yield remains at ∼25% to 30% across indications (eg, neurodevelopmental disorders, multiple congenital anomalies, and congenital heart disease).3,79,80 Limitations of exome sequencing include the inability to detect disease-associated intronic variants if not specifically targeted, as well as some coding variants in difficult regions. An increasingly utilized application of exome sequencing among clinical laboratories is an “in silico enrichment exome” approach (ie, “exome slice”) that leverages a single underlying enrichment-based exome platform and laboratory workflow but bioinformatically restricts the clinical interpretation to specific diagnostic gene panel regions of interest.81, 82, 83

In contrast to enrichment-based exome sequencing, genome sequencing evaluates approximately 95% of nuclear and mitochondrial DNA, and it is increasingly being considered for diagnostic constitutional genetic testing.84 Diagnostic yield comparisons between genome and exome sequencing are increasingly being reported, which suggest an improvement that may be influenced by age of presentation and clinical indication.16, 17, 18,85,86 Moreover, rapid genome sequencing has become feasible for the diagnosis of critically ill newborns presenting with suspected genetic conditions in the NICU,87 and some national health care systems are currently implementing genome sequencing broadly to guide medical management among seriously ill children.88 Consistent with the “exome slice” approach, genome-based diagnostic panels (“genome slice”) can also be developed by clinical laboratories when implementing genome sequencing as a single workflow. This approach inherently has the added benefit of offering flexibility in additional reporting content for a variety of Mendelian diagnostic panels to guide patient care, clinical management, prognosis, and disease risk screening as needed (Figure 249). Moreover, as clinical RNA sequencing increasingly becomes available to resolve the functional significance of germline variants, the interrogation of noncoding variants by genome sequencing will likely have continued increasing value for diagnostic testing.89, 90, 91 However, this flexibility in reporting content is offset by the necessary computational infrastructure needed from clinical laboratories to develop, validate, and maintain a clinical genome sequencing workflow.

Figure 2.

Figure 2

Illustration of Mendelian disease genes and pharmacogenomic genes across the human genome (GRCh38). From inner to outer circles: genes defined by the US FDA Table of Pharmacogenetic Associations as having “supportive evidence for therapeutic management recommendations” (blue) and “potentially impacted”/ “pharmacokinetics only” (red); genes with CPIC evidence levels A, A/B, B (blue) and C, C/D, D (red); OMIM-defined recessive (light green), dominant (dark green), and X-linked (gray) disease genes with an established molecular etiology (phenotype mapping key: 3); and ClinGen-defined recessive (light green), dominant (dark green), and X-linked (gray) disease genes with “definitive” or “strong” evidence. All data were downloaded from primary sources and Circos images generated using R package circlize.49

Genome sequencing and carrier screening

In contrast to diagnostic genomic testing, carrier screening tests clinically asymptomatic individuals for pathogenic variants associated with autosomal recessive and X-linked disorders for the purpose of reproductive risk management.2,92 The American College of Obstetricians and Gynecologists (ACOG) recommends screening be offered to all pregnant women,92,93 ideally coupled with genetic counseling and performed preconception so that carrier couples can understand their reproductive risks and make informed decisions. ACOG currently recommends screening for cystic fibrosis, spinal muscular atrophy, and hemoglobinopathy for all pregnant women regardless of ancestry or ethnicity.14,92,94 However, screening for a limited number of additional recessive conditions is also recommended for reproductive women of certain ethnicities and/or those with a family history of a particular genetic disorder (eg, Tay-Sachs disease and fragile X syndrome).92,95 The ACMG has recently reported updated guidance on screening, which recommends a tiered system for panel gene content based on heterozygote frequency, among other important considerations related to implementation and result management.2

Screening technologies have evolved from targeted genotyping of pathogenic variants to high-throughput full-gene sequencing, as well as orthogonal copy-number techniques for selected recessive genes with recurrent deletion alleles. As such, pan-ethnic expanded screening can now test for hundreds of conditions simultaneously, which is more efficient and cost-effective than targeted screening. Although ACOG indicates that expanded screening is an acceptable strategy for prepregnancy and prenatal screening, a heterozygote frequency threshold of 1 in 100 has been recommended when incorporating genes/diseases into test panels to minimize patient anxiety as more heterozygotes are identified.93 As such, there is ongoing debate around the utility and rationale of expanded screening panels,96,97 as currently available commercial panels often include recessive conditions with low-frequency heterozygote disease alleles. However, these expanded panels with lower heterozygote frequencies are supported by recently reported data indicating that the recommended 1 in 100 carrier frequency threshold can lead to gene panels with significantly reduced detection of at-risk couples,98 particularly when considering multiethnic populations.99

Genome sequencing has potential utility for screening, including improved detection rates for certain conditions and the flexibility to interrogate additional disease genes or genomic regions when indicated. Of note, some recessive conditions with recurrent pathogenic CNV alleles (eg, spinal muscular atrophy) have variable detection rates when screened solely by enrichment-based sequencing panels without ancillary methods; however, bioinformatic tools have recently been developed that detect SMN1 and SMN2 deletion heterozygotes from enrichment-based targeted sequencing100 and genome sequencing with high accuracy.14 Preconception screening for 728 gene-disease pairs using genome sequencing has also recently been reported, which resulted in improved sensitivity compared with targeted screening for clinically significant variant detection, including CNVs and noncoding variants.101 Importantly, genome sequencing can theoretically identify carrier status across 3104 autosomal and X-linked recessive genes in the human genome, as defined by OMIM66 (Figure 249). This enhanced capability of disease gene inclusion by genome sequencing is counterbalanced by the ongoing debate over the clinical utility of expanded screening panels; however, to facilitate more informed and compartmentalized gene/disease panel development, a taxonomy framework has been proposed that categorizes potential gene content into lifespan limiting, serious, mild, unpredictable, and adult-onset,102 with the overarching aim of minimizing anxiety when detecting more heterozygotes for very rare and/or less severe conditions.

Genome sequencing and cytogenomic molecular karyotyping

Cytogenetic testing is arguably the original genomic platform, which interrogates chromosomal variation through metaphase cell preparations and microscopy-based karyotyping. Aneuploidies, large structural rearrangements (eg, translocations and inversions), and polyploidy can be detected by traditional cytogenetic testing; however, copy-number aberrations less than ∼5 Mb are below the limit of detection for high-resolution chromosome analyses. This prompted the implementation of clinical fluorescence in situ hybridization testing to interrogate recurrent constitutional deletions and duplications less than 5 Mb, as well as genome-wide chromosomal microarray (CMA) testing with resolutions defined by the density of oligonucleotide probes. CMA is now a common first-tier platform for copy-number testing across several clinical indications, and resolution typically ranges from ∼10 to 100 kb depending on CMA design and platform. Critical resources for CNV interpretation include the Database of Genomic Variants (DGV), which catalogs structural variation (>50 bp) in healthy population cohorts72; the ClinGen Dosage Sensitivity Map73 and DECIPHER,71 which catalog genomic variation in patient cohorts (Figure 349); and quantitative CNV classification recommendations have been reported by the ACMG (Table 2).60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76 In addition, the Genome Aggregation Database has increasingly incorporated structural variants identified by sequencing, including deletions, duplications, CNVs, insertions, inversions, and other variant types.103

Figure 3.

Figure 3

Illustration of structural variation across the human genome (GRCh38). From inner to outer circles: ClinGen genes and genomic regions with “sufficient” or “emerging” evidence curated as haploinsufficient (red) and triplosensitive (blue) and DGV Gold Standard deletions (red), duplications (blue), and “other” structural variants (as defined by DGV; light green) found in the general population. All data were downloaded from primary sources and Circos images generated using R package circlize.49

The implementation of constitutional CMA testing as an orthogonal platform for chromosome analysis has evolved the field of cytogenetics from low-resolution karyotyping to high-resolution molecular karyotyping. In addition to now being a routine clinical genetic test, CMA profiling of both patient population and general population cohorts has resulted in the discovery of significant copy-number variability across the human genome that previously was not appreciated, including a spectrum of benign, uncertain, pathogenic, and susceptibility CNVs. However, technical limitations of CMA testing include the inability to precisely identify CNV breakpoints and the genomic location(s) of copy-number gain material. Consistent with the ongoing improvements in both short- and long-read sequencing chemistries and throughput, bioinformatic algorithms that leverage local sequencing depth and/or read orientation have now enabled the identification of constitutional CNVs and other structural rearrangements by sequencing.

Enrichment-based diagnostic sequencing panels now routinely include CNV detection; however, given that the sequencing reads in these panels are localized to coding regions, precise reporting of CNV breakpoints is technically not possible. As such, the uniform coverage of genome sequencing provides an immediate improvement in CNV detection and breakpoint localization compared with enrichment-based sequencing. Many bioinformatic tools for constitutional copy-number detection from short-read sequencing data are currently available,104 as are long-read sequencing structural variant callers for both HiFi and nanopore sequencing.105 Importantly, analytical validation of CNV detection by genome sequencing can now be accomplished, in part, using the NA24385 (HG002) GIAB/NIST reference sample with a reported structural variant benchmarking truth set (Table 1).21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46

Notably, low-pass (∼5×) genome sequencing has been reported to have high accuracy and precision for clinically significant CNV detection compared with CMA analysis, which included enhanced sensitivity and cost-effectiveness,106 and the detection of absence of heterozygosity.107 Moreover, both short- and long-read genome sequencing are increasingly emerging as efficient tools for translocation breakpoint detection,108, 109, 110 underscoring the potential utility of genome sequencing for more comprehensive molecular karyotyping. Given the ongoing improvements and resources supporting CNV and structural variant detection by genome sequencing, molecular karyotyping by clinical genome sequencing platforms will likely supplant CMA and/or karyotyping in the future when workflow feasibility is justified.

Prenatal genome sequencing

Prenatal genetic testing has unique characteristics compared with postnatal constitutional testing because the specimens are procured from invasive procedures during pregnancy, including chorionic villus sampling and amniocentesis. Given the inherent fetal risk of these procedures and the sensitivities around prenatal genetic result interpretation and counseling, prenatal testing has historically been limited to targeted variant testing when parents are positive for a pathogenic variant, or sequencing panels, karyotyping, fluorescence in situ hybridization, and/or CMA testing for fetuses with abnormal ultrasound findings. The ACOG recommends CMA testing to any patient undergoing invasive diagnostic testing, as well as being the primary test for pregnancies with a fetal structural abnormality detected by ultrasound,111 including a recommendation “against” routine prenatal exome or genome sequencing.112 However, ACOG states that “prenatal exome sequencing may be reasonable for fetuses with multiple anomalies or in cases of recurrent fetal phenotypes with no diagnosis by karyotype or CMA.”112 Similarly, the ACMG recommends “considering exome sequencing only when specific genetic tests for a phenotype, including targeted sequencing, have failed to determine a diagnosis in a fetus with multiple congenital anomalies suggestive of a genetic disorder.”113

Despite the increased diagnostic rate of exome sequencing compared with targeted testing, a limitation for prenatal exome testing is the longer turnaround time, which can negatively affect reproductive decision making. Another major limitation is the higher number of variants of uncertain significance, which can be challenging for providers and patients to interpret, as well as lead to increased patient anxiety. However, rapid prenatal exome testing is currently available, and evidence is continuing to emerge, indicating a high diagnostic yield for fetuses with abnormal ultrasound findings (increased nuchal translucency, hydrops, intrauterine growth restriction, and/or congenital anomalies).114,115 As such, it is likely that as prenatal exome testing workflows become more efficient, including variant filtration and prioritization algorithms, that the relevant professional societies will report more supportive recommendations for prenatal exome testing under certain clinical contexts.

Less data are available on constitutional prenatal genome sequencing, which is also not commonly implemented clinically. However, there is potential with implementing genome sequencing prenatally because both coding and noncoding pathogenic variants can be interrogated, as well as the improved capacity to detect CNVs and balanced chromosomal abnormalities. This is counterbalanced by the understandable concerns about identifying more information than needed through prenatal genome sequencing, including more variants of uncertain significance, late-onset disease variants and susceptibility alleles, which could overwhelm providers, counselors, and patients.116 Similar to postnatal molecular karyotyping, the potential utility of prenatal genome sequencing is supported by the prenatal discovery of a balanced translocation that disrupted the CHD7 gene, which confirmed a CHARGE syndrome diagnosis,117 and subsequent cohort studies on additional prenatal subjects with apparently balanced chromosomal rearrangements and normal CMA results.118 More recently, low-pass (<1×) genome sequencing among 1023 women undergoing prenatal diagnosis identified clinically significant structural variants with enhanced resolution and increased sensitivity to detect mosaicism compared with CMA testing.119 Taken together, the increasing utilization of exome sequencing for prenatal testing and the continued technical improvements in genome sequencing suggest that in the future, prenatal genome sequencing may have clinical utility by coupling genome-wide CNV and structural variant detection with appropriate content and reporting restrictions. However, continued utility research and policy guidelines will be necessary before broadly adopting this approach to prenatal diagnosis, particularly concerning issues of equitable access, the necessary infrastructure for genetic counseling, policy development, fiscal sustainability, and associated ethical and psychosocial implications.

Genome sequencing and mitochondrial genetic testing

Mitochondria are directly involved in several important cellular processes, including oxidative phosphorylation (OXPHOS) energy production, apoptosis, cytosolic calcium level control, lipid homeostasis, steroid synthesis, innate immune response, and metabolic cell signaling. Importantly, all mitochondria harbor a circular 16.6 kb double-stranded DNA (mtDNA) that encodes 13 essential OXPHOS genes and the rRNAs and tRNAs necessary for their expression. The mtDNA is maternally transmitted and has a very high mutation rate, which can result in a spectrum of benign to pathogenic variants. Given that there are numerous mitochondria within a cell, mixed intracellular populations of wild-type and mutant mtDNAs can coexist (ie, heteroplasmy), and their proportion can vary across tissues. Most pathogenic mtDNA variants are heteroplasmic and the manifestation and severity of disease is directly related to the heteroplasmy levels in affected tissues.120 The thresholds for heteroplasmic disease vary across tissues; however, those tissues with high energy requirements are most commonly affected.

Mitochondrial disorders are genetically heterogeneous because they can be due to a defect in mtDNA or to pathogenic variants affecting OXPHOS complex subunits and/or assembly factors that are encoded by nuclear DNA (nDNA). As such, mitochondrial disorders can be sporadic, maternally inherited, or follow Mendelian inheritance.121 The most comprehensive mtDNA disease sequence and variant resources include MITOMAP,75 HmtDB,122 HmtVar,123 MtSNPscore,124 MSeqDR,125 and ClinVar, which provide curation resources for both mtDNA and mitochondrial nDNA variants. In addition, an international working group of the Mitochondrial Disease Sequence Data Resource Consortium recently adapted the ACMG/AMP constitutional variant interpretation guidelines62 to provide guidance on mtDNA variant classification (Table 2).60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76

Traditional diagnostic methods for the detection of mtDNA variants include Sanger sequencing of prioritized genes and alternative methods to accurately quantify mtDNA heteroplasmy,126 as well as large mtDNA deletion analysis by Southern blotting or custom CMA.127 However, high-throughput sequencing of the complete mtDNA and selected nDNA genes has greatly improved mitochondrial diagnostics by enabling more accurate quantification of mtDNA heteroplasmy and better detection of deletions.128 The clinical and genetic heterogeneity of mitochondrial disorders along with the growing number of implicated nuclear genes has led to exome sequencing as an effective first-tier platform for mitochondrial diagnostic testing129,130; however, most commercial exome capture kits do not contain probes that specifically interrogate the mitochondrial genome. Alternatively, “off-target” capture of the mitochondrial genome can be analyzed from exome sequencing data; however, the limitations of this approach include a greater depth of coverage needed for accurate quantification of low-level heteroplasmy and the inability to reliably identify mtDNA CNVs.131 The diagnostic yield of exome sequencing for mitochondrial disorders ranges from 35% to 70% depending on the patient population.132,133

Genome sequencing provides further potential for mitochondrial diagnostics based on consistent coverage and simultaneous sequencing of mtDNA and nDNA, inclusion of noncoding regions, superior mtDNA coverage, and better detection of structural variants.5,134 For example, a previous study on patients with suspected mitochondrial disease concluded that the diagnostic yield is at least equivalent to exome sequencing for known variants but with potential for improved yield because of identification of novel genes and variants.135 More recently, genome sequencing was applied to a cohort of 345 patients with suspected mitochondrial disease, which resulted in a probable diagnosis in 31% of cases, 38% of which were mitochondrial, and 63% were nuclear.136

Genome sequencing and pharmacogenomic testing

Pharmacogenomic testing is increasingly being considered for clinical implementation for selected medications with high levels of evidence (Figure 249).137 Important resources for pharmacogenomics include the Pharmacogenomics Knowledgebase (PharmGKB),68 practice guidelines from the Clinical Pharmacogenetics Implementation Consortium (CPIC),138 and the US FDA Table of Pharmacogenetic Associations139 (Table 260, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76). Importantly, the ACMG has recently reported a technical guideline on clinical pharmacogenomic testing and reporting, which includes recommendations when implementing exome- and genome-based pharmacogenomic testing,140 and the AMP continues to release recommendations on which alleles to include in clinical pharmacogenomic genotyping assays for multiple genes with available CPIC/DPWG guidelines.141, 142, 143, 144, 145, 146

Most clinical pharmacogenomic panels are based on targeted genotyping because of the cost-effectiveness of the platform, focused content selection, and relatively short turnaround time.147 However, a limitation of targeted genotyping is detecting structural variants and CNVs unless they are directly interrogated by specific molecular assays,148 which are increasingly identified as clinically significant forms of pharmacogenomic variation,149,150 particularly for CYP2D6.151, 152, 153 Pharmacogenomic full-gene sequencing154 and enrichment-based exome and genome sequencing have also been reported155; however, these strategies are not able to phase haplotypes and diplotypes compared with the reported long-read pharmacogenomic sequencing assays.156,157

Enrichment-based exome or genome sequencing is not currently cost-effective for routine pharmacogenomic testing; however, these platforms have been analytically assessed across pharmacogenomic genes with CPIC guidelines.155,158 Genome sequencing results in highly concordant variant calls compared with clinical pharmacogenomic genotyping; however, genes with known short-read sequencing alignment challenges (eg, CYP2D6, G6PD, and HLA) were the most common sources of discrepancy. Although assessment of CYP2D6 copy number was also not robust by genome sequencing, recent computational tools have reported improvements in inferring CYP2D6 CNVs and gene conversions from short-read sequencing data.159,160 In addition, star (∗) allele diplotyping tools are currently in development,161,162 which enable the translation of sequence variant calls to common pharmacogenomic nomenclature based on haplotype definitions from the Pharmacogene Variation (PharmVar) consortium.163 As such, interrogating pharmacogenomic variation by genome sequencing is currently feasible,164,165 including the clinically actionable HLA region,166 and it is expected that the necessary computational phasing and translation tools will continue to improve and enable clinical pharmacogenomic reporting from genome sequence data.

Genome sequencing and polygenic risk score testing

Fifteen years of genome-wide association study (GWAS) research has resulted in the discovery of numerous constitutional variants (common and rare) implicated in complex diseases and other human traits but with effect sizes that are individually small. However, the combinations of these predisposition variants for certain diseases are now emerging as potentially actionable polygenic risk scores (PRS), with the aim of providing useful information for disease risk stratification or prognosis.167 PRS algorithms are currently available that leverage thousands or millions of variants and weighted sums of allele counts to generate a single number that is proportional to the risk for a given disease; however, their clinical validity and utility are still being established, with the most robust PRS algorithms emerging for cardiovascular disease and certain types of cancer.168 In an effort to enable more standardized PRS research and facilitate translation into clinical care, the ClinGen Complex Disease Working Group and the Polygenic Score (PGS) Catalog recently reported Polygenic Risk Score Reporting Standards (PRS-RS) to inform PRS best practices and result reporting,169 and the ACMG has recently reported a “points to consider” perspective on the development of laboratory-developed PRS tests.170

PRS testing is inherently a genomic assay, which was initially driven by GWAS that used high-resolution single-nucleotide variant microarrays; however, as high-throughput sequencing technologies became more accessible, GWAS programs evolved to utilize exome and genome sequencing, including more cost-effective low-pass (∼1×) genome sequencing. As such, clinical genome sequencing is uniquely suited to support PRS testing because the consistent coverage at depths commonly implemented for clinical testing (>30×) can easily genotype single-nucleotide variants implemented in PRS algorithms. This conceptually suggests that, similar to optional pharmacogenomic reporting for diagnostic genome sequencing, PRS reporting for selected diseases could also be developed as a companion to Mendelian diagnostic testing when implementing a clinical genome platform. However, a more cost-effective strategy for independent PRS testing utilizes low-pass genome sequencing coupled with variant imputation, which has resulted in both efficient and accurate PRS prediction compared with both microarrays and high-depth genome sequencing.171

Despite the extensive research on PRS for many diseases and complex traits, clinical laboratory guidelines and best practices are only beginning to emerge. As such, development of clinical PRS tests (low-pass or high-depth sequencing) must follow the laboratory-developed test validation requirements from the CAP. The unique characteristics of PRS testing necessitate additional analytical considerations beyond variant calling accuracy and precision, particularly when pursuing low-pass genome sequencing. Several imputation methods are currently available to infer common variant genotype results from low-pass sequence data, and laboratories must assess which is the most accurate and robust for their assay. Moreover, a quantitative assessment of PRS prediction using specimens with orthogonally derived PRS values that are considered truth is also necessary to validate all elements of the low-pass sequencing PRS workflow. However, although PRS testing is increasingly being considered by progressive laboratories that pursue clinical genome sequencing, caution should be exercised when implementing across diverse populations because most PRS algorithms have been developed among predominantly European cohorts.172

Clinical genome sequencing limitations

Technical limitations

Despite the analytical improvements with uniform coverage and small-variant/CNV accuracy of genome sequencing compared with enrichment-based exome sequencing,173 genome sequencing does have some technical limitations. For example, short-read genome sequencing can have inconsistent CNV breakpoint identification, particularly for copy-number gains. Although the resolution of CNV detection is greater by genome sequencing compared with CMA testing, algorithms for CNV identification can result in imprecise breakpoint detection and false positives. This can be partially solved by filtering CNV calls using an internal database of genome sequencing-based CNVs; however, this effort inherently requires extra resources and a significant volume of internal data. Notably, long-read sequencing-based structural variant detection is rapidly emerging as a more accurate method for these constitutional events, particularly for complex structural variants.55,174,175

In addition, given the lower depths typically implemented with genome sequencing (∼30×), mosaic variant detection by genome sequencing is less sensitive compared with higher depth enrichment-based panel or exome sequencing. Moreover, mosaic CNVs are also problematic for genome sequencing, particularly for mosaic aneuploidy (eg, mosaic Turner syndrome).176 Another technical challenge for genome sequencing compared with enrichment-based sequencing is specimen types because interfering bacterial contamination in saliva samples is known to negatively affect certain genome sequencing quality metrics.177 In addition, although genome sequencing has proven performance for mitochondrial disease testing, significant differences in detecting low-level heteroplasmic variants has been reported between genome sequencing and targeted mtDNA sequencing.178

Economic limitations

Importantly, the costs of sequencing are continually evolving and are dependent on multiple factors, including platform and chemistry, desired depth and sample pooling strategy, laboratory procedures and overhead, and intended use. As such, thorough comparisons between enrichment-based panels, exome, and genome sequencing were also considered outside of the scope of this review. However, as one of the most notable factors driving the implementation of genome sequencing among clinical laboratories, direct reagent costs for short-read enrichment-based exome and genome sequencing (short and long read) at clinical-grade sequencing depths on high-throughput instruments can be currently estimated at ∼$100 and ∼$500, respectively. This does not include any computational, workflow, or technical effort considerations but does highlight an important barrier for clinical laboratories when implementing genome sequencing. However, higher throughput short- and long-read sequencing platforms have recently been released, indicating that individual genome sequencing reagent costs will only continue to decline.

Of note, the cost-effectiveness of genome sequencing for diagnosing critically ill infants and pediatric patients with suspected genetic disease has recently been reported to be potentially cost saving as a first-line diagnostic platform.179, 180, 181 However, although preliminary economic evaluations support the cost-saving potential of diagnostic clinical genome sequencing, multidisciplinary implementation research, including more robust outcome measurement and economic evaluation, is needed to demonstrate the cost-effectiveness of clinical genome sequencing.16,182 Moreover, although the reimbursement landscape for clinical genome sequencing has historically not been very favorable, this is also a rapidly evolving area that is now supported by a dedicated Current Procedural Terminology code for clinical genome sequencing (81425).

Conclusions and future directions

Genome sequencing is increasingly being considered as a single platform for genetic testing by clinical laboratories, which is based on the ongoing improvements in throughput, computational pipelines, variant calling and prioritization algorithms, structural variant callers, and the potential utility across many traditional constitutional genetic testing applications. Although professional guideline recommendations for genome sequencing are still emerging, substantial data currently exist supporting potential utility across diagnostic testing, carrier screening, molecular karyotyping, mitochondrial testing, and pharmacogenomic and polygenic risk score testing. This progressive approach is justifiably contrasted by important logistical and ethical considerations because the infrastructure and resources needed to implement clinical genome sequencing are significant. Development and validation of laboratory and bioinformatic procedures, as well as establishing a computational data management infrastructure, require investments that may not be feasible for many laboratories at this time; however, the efficiency of a single platform would undoubtedly result in significantly reduced long-term resources when developing “new” tests with a single genome sequencing workflow.

Although short-read sequencing bioinformatic tools are emerging to facilitate the accurate interrogation of historically challenging genomic regions, long-read genome sequencing inherently can outperform short-read sequencing across homologous regions, repeat expansions, and other difficult sequence contexts. As such, future iterations of clinical genome sequencing may leverage the benefits of both technologies by integrating the throughput of short-read sequencing with the improved phasing and variant calling accuracy of long-read sequencing; however, additional research and validation data are needed to establish the clinical feasibility of this approach. New technologies are also increasingly being implemented by clinical genomic laboratories, including optical genome mapping, which is based on fluorescently labeled high molecular weight DNA molecules. Optical genome mapping has recently been reported to have improved structural variant calling and molecular karyotyping compared with classic cytogenetic technologies,110,183 supporting its utility as an orthogonal cytogenomic platform. However, the rapid and ongoing improvements in both short- and long-read sequencing indicate that it is only a matter of time before genome sequencing as a single platform for clinical constitutional genetic testing is considered routine and not a progressive outlier.

Conflict of Interest

A.S. is a paid employee of LetsGetChecked, Monrovia, CA, and holds equity in LetsGetChecked, Opus Genomics, and PathFinder Health. All other authors declare no conflicts of interest.

Acknowledgments

Funding

S.A.S. was supported in part by the NIH/NHGRI/NICHD/NIDA grant U24 HG010615.

Author Information

Conceptualization: S.A.S.; Data Curation: Y.Y., D.d.G., A.S., S.A.S.; Visualization: Y.Y.; Writing-original draft: Y.Y., D.d.G., A.S., S.A.S.; Writing-review and editing: Y.Y., D.d.G., A.S., S.A.S.

Footnotes

The Article Publishing Charge (APC) for this article was paid by Stuart A. Scott.

References

  • 1.Fiala E.M., Jayakumaran G., Mauguen A., et al. Prospective pan-cancer germline testing using MSK-IMPACT informs clinical translation in 751 patients with pediatric solid tumors. Nat Cancer. 2021;2:357–365. doi: 10.1038/s43018-021-00172-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gregg A.R., Aarabi M., Klugman S., et al. Screening for autosomal recessive and X-linked conditions during pregnancy and preconception: a practice resource of the American College of Medical Genetics and Genomics (ACMG) Genet Med. 2021;23(10):1793–1806. doi: 10.1038/s41436-021-01203-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lee H., Deignan J.L., Dorrani N., et al. Clinical exome sequencing for genetic identification of rare Mendelian disorders. JAMA. 2014;312(18):1880–1887. doi: 10.1001/jama.2014.14604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Yang Y., Muzny D.M., Xia F., et al. Molecular findings among patients referred for clinical whole-exome sequencing. JAMA. 2014;312(18):1870–1879. doi: 10.1001/jama.2014.14601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Belkadi A., Bolze A., Itan Y., et al. Whole-genome sequencing is more powerful than whole-exome sequencing for detecting exome variants. Proc Natl Acad Sci U S A. 2015;112(17):5473–5478. doi: 10.1073/pnas.1418631112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Costain G., Walker S., Marano M., et al. Genome sequencing as a diagnostic test in children with unexplained medical complexity. JAMA Netw Open. 2020;3(9) doi: 10.1001/jamanetworkopen.2020.18109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bick D., Jones M., Taylor S.L., Taft R.J., Belmont J. Case for genome sequencing in infants and children with rare, undiagnosed or genetic diseases. J Med Genet. 2019;56(12):783–791. doi: 10.1136/jmedgenet-2019-106111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Rehm H.L. Evolving health care through personal genomics. Nat Rev Genet. 2017;18(4):259–267. doi: 10.1038/nrg.2016.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Costain G., Jobling R., Walker S., et al. Periodic reanalysis of whole-genome sequencing data enhances the diagnostic advantage over standard clinical genetic testing. Eur J Hum Genet. 2018;26(5):740–744. doi: 10.1038/s41431-018-0114-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lelieveld S.H., Spielmann M., Mundlos S., Veltman J.A., Gilissen C. Comparison of exome and genome sequencing technologies for the complete capture of protein-coding regions. Hum Mutat. 2015;36(8):815–822. doi: 10.1002/humu.22813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hehir-Kwa J.Y., Pfundt R., Veltman J.A. Exome sequencing and whole genome sequencing for the detection of copy number variation. Expert Rev Mol Diagn. 2015;15(8):1023–1032. doi: 10.1586/14737159.2015.1053467. [DOI] [PubMed] [Google Scholar]
  • 12.Dolzhenko E., van Vugt J.J.F.A., Shaw R.J., et al. Detection of long repeat expansions from PCR-free whole-genome sequence data. Genome Res. 2017;27(11):1895–1903. doi: 10.1101/gr.225672.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Walker S., Lamoureux S., Khan T., et al. Genome sequencing for detection of pathogenic deep intronic variation: a clinical case report illustrating opportunities and challenges. Am J Med Genet A. 2021;185(10):3129–3135. doi: 10.1002/ajmg.a.62389. [DOI] [PubMed] [Google Scholar]
  • 14.Chen X., Sanchis-Juan A., French C.E., et al. Spinal muscular atrophy diagnosis and carrier screening from genome sequencing data. Genet Med. 2020;22(5):945–953. doi: 10.1038/s41436-020-0754-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dolzhenko E., Deshpande V., Schlesinger F., et al. ExpansionHunter: a sequence-graph-based tool to analyze variation in short tandem repeat regions. Bioinformatics. 2019;35(22):4754–4756. doi: 10.1093/bioinformatics/btz431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Smith H.S., Swint J.M., Lalani S.R., et al. Clinical application of genome and exome sequencing as a diagnostic tool for pediatric patients: a scoping review of the literature. Genet Med. 2019;21(1):3–16. doi: 10.1038/s41436-018-0024-6. [DOI] [PubMed] [Google Scholar]
  • 17.Gilissen C., Hehir-Kwa J.Y., Thung D.T., et al. Genome sequencing identifies major causes of severe intellectual disability. Nature. 2014;511(7509):344–347. doi: 10.1038/nature13394. [DOI] [PubMed] [Google Scholar]
  • 18.Cohen A.S.A., Farrow E.G., Abdelmoity A.T., et al. Genomic answers for children: dynamic analyses of >1000 pediatric rare disease genomes. Genet Med. 2022;24(6):1336–1348. doi: 10.1016/j.gim.2022.02.007. [DOI] [PubMed] [Google Scholar]
  • 19.Dimmock D., Caylor S., Waldman B., et al. Project Baby Bear: rapid precision care incorporating rWGS in 5 California children’s hospitals demonstrates improved clinical outcomes and reduced costs of care. Am J Hum Genet. 2021;108(7):1231–1238. doi: 10.1016/j.ajhg.2021.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wenger A.M., Peluso P., Rowell W.J., et al. Accurate circular consensus long-read sequencing improves variant detection and assembly of a human genome. Nat Biotechnol. 2019;37(10):1155–1162. doi: 10.1038/s41587-019-0217-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gargis A.S., Kalman L., Berry M.W., et al. Assuring the quality of next-generation sequencing in clinical laboratory practice. Nat Biotechnol. 2012;30(11):1033–1036. doi: 10.1038/nbt.2403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Schrijver I., Aziz N., Farkas D.H., et al. Opportunities and challenges associated with clinical diagnostic genome sequencing: a report of the Association for Molecular Pathology. J Mol Diagn. 2012;14(6):525–540. doi: 10.1016/j.jmoldx.2012.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rehm H.L., Bale S.J., Bayrak-Toydemir P., et al. ACMG clinical laboratory standards for next-generation sequencing. Genet Med. 2013;15(9):733–747. doi: 10.1038/gim.2013.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Schrijver I., Aziz N., Jennings L.J., Richards C.S., Voelkerding K.V., Weck K.E. Methods-based proficiency testing in molecular genetic pathology. J Mol Diagn. 2014;16(3):283–287. doi: 10.1016/j.jmoldx.2014.02.002. [DOI] [PubMed] [Google Scholar]
  • 25.Aziz N., Zhao Q., Bry L., et al. College of American Pathologists’ laboratory standards for next-generation sequencing clinical tests. Arch Pathol Lab Med. 2015;139(4):481–493. doi: 10.5858/arpa.2014-0250-CP. [DOI] [PubMed] [Google Scholar]
  • 26.Gargis A.S., Kalman L., Bick D.P., et al. Good laboratory practice for clinical next-generation sequencing informatics pipelines. Nat Biotechnol. 2015;33(7):689–693. doi: 10.1038/nbt.3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Boycott K., Hartley T., Adam S., et al. The clinical application of genome-wide sequencing for monogenic diseases in Canada: position Statement of the Canadian College of Medical Geneticists. J Med Genet. 2015;52(7):431–437. doi: 10.1136/jmedgenet-2015-103144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Matthijs G., Souche E., Alders M., et al. Guidelines for diagnostic next-generation sequencing. Eur J Hum Genet. 2016;24(1):2–5. doi: 10.1038/ejhg.2015.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Santani A., Murrell J., Funke B., et al. Development and validation of targeted next-generation sequencing panels for detection of germline variants in inherited diseases. Arch Pathol Lab Med. 2017;141(6):787–797. doi: 10.5858/arpa.2016-0517-RA. [DOI] [PubMed] [Google Scholar]
  • 30.Hegde M., Santani A., Mao R., Ferreira-Gonzalez A., Weck K.E., Voelkerding K.V. Development and validation of clinical whole-exome and whole-genome sequencing for detection of germline variants in inherited disease. Arch Pathol Lab Med. 2017;141(6):798–805. doi: 10.5858/arpa.2016-0622-RA. [DOI] [PubMed] [Google Scholar]
  • 31.Roy S., Coldren C., Karunamurthy A., et al. Standards and guidelines for validating next-generation sequencing bioinformatics pipelines: a joint recommendation of the association for molecular pathology and the College of American Pathologists. J Mol Diagn. 2018;20(1):4–27. doi: 10.1016/j.jmoldx.2017.11.003. [DOI] [PubMed] [Google Scholar]
  • 32.Santani A., Simen B.B., Briggs M., et al. Designing and implementing NGS tests for inherited disorders: a practical framework with step-by-step guidance for clinical laboratories. J Mol Diagn. 2019;21(3):369–374. doi: 10.1016/j.jmoldx.2018.11.004. [DOI] [PubMed] [Google Scholar]
  • 33.Hume S., Nelson T.N., Speevak M., et al. CCMG practice guideline: laboratory guidelines for next-generation sequencing. J Med Genet. 2019;56(12):792–800. doi: 10.1136/jmedgenet-2019-106152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bean L.J.H., Funke B., Carlston C.M., et al. Diagnostic gene sequencing panels: from design to report-a technical standard of the American College of Medical Genetics and Genomics (ACMG) Genet Med. 2020;22(3):453–461. doi: 10.1038/s41436-019-0666-z. [DOI] [PubMed] [Google Scholar]
  • 35.Marshall C.R., Chowdhury S., Taft R.J., et al. Best practices for the analytical validation of clinical whole-genome sequencing intended for the diagnosis of germline disease. npj Genom Med. 2020;5:47. doi: 10.1038/s41525-020-00154-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hayeems R.Z., Dimmock D., Bick D., et al. Clinical utility of genomic sequencing: a measurement toolkit. npj Genom Med. 2020;5(1):56. doi: 10.1038/s41525-020-00164-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Manickam K., McClain M.R., Demmer L.A., et al. Exome and genome sequencing for pediatric patients with congenital anomalies or intellectual disability: an evidence-based clinical guideline of the American College of Medical Genetics and Genomics (ACMG) Genet Med. 2021;23(11):2029–2037. doi: 10.1038/s41436-021-01242-6. [DOI] [PubMed] [Google Scholar]
  • 38.Austin-Tse C.A., Jobanputra V., Perry D.L., et al. Best practices for the interpretation and reporting of clinical whole genome sequencing. npj Genom Med. 2022;7(1):27. doi: 10.1038/s41525-022-00295-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Souche E., Beltran S., Brosens E., et al. Recommendations for whole genome sequencing in diagnostics for rare diseases. Eur J Hum Genet. 2022;30(9):1017–1021. doi: 10.1038/s41431-022-01113-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zook J.M., Catoe D., McDaniel J., et al. Extensive sequencing of seven human genomes to characterize benchmark reference materials. Sci Data. 2016;3 doi: 10.1038/sdata.2016.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cleveland M.H., Zook J.M., Salit M., Vallone P.M. Determining performance metrics for targeted next-generation sequencing panels using reference materials. J Mol Diagn. 2018;20(5):583–590. doi: 10.1016/j.jmoldx.2018.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zook J.M., McDaniel J., Olson N.D., et al. An open resource for accurately benchmarking small variant and reference calls. Nat Biotechnol. 2019;37(5):561–566. doi: 10.1038/s41587-019-0074-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Krusche P., Trigg L., Boutros P.C., et al. Best practices for benchmarking germline small-variant calls in human genomes. Nat Biotechnol. 2019;37(5):555–560. doi: 10.1038/s41587-019-0054-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Zook J.M., Hansen N.F., Olson N.D., et al. A robust benchmark for detection of germline large deletions and insertions. Nat Biotechnol. 2020;38(11):1347–1355. doi: 10.1038/s41587-020-0538-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wilcox E., Harrison S.M., Lockhart E., et al. Creation of an expert curated variant list for clinical genomic test development and validation: a ClinGen and GeT-RM collaborative project. J Mol Diagn. 2021;23(11):1500–1505. doi: 10.1016/j.jmoldx.2021.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wagner J., Olson N.D., Harris L., et al. Curated variation benchmarks for challenging medically relevant autosomal genes. Nat Biotechnol. 2022;40(5):672–680. doi: 10.1038/s41587-021-01158-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Human CLSI. CLSI guideline MM09. 3rd ed. Clinical and Laboratory Standards Institute; 2023. Genetic and Genomic Testing Using Traditional and High-Throughput Nucleic Acid Sequencing Methods. [Google Scholar]
  • 48.Mandelker D., Schmidt R.J., Ankala A., et al. Navigating highly homologous genes in a molecular diagnostic setting: a resource for clinical next-generation sequencing. Genet Med. 2016;18(12):1282–1289. doi: 10.1038/gim.2016.58. [DOI] [PubMed] [Google Scholar]
  • 49.Gu Z., Gu L., Eils R., Schlesner M., Brors B. circlize Implements and enhances circular visualization in R. Bioinformatics. 2014;30(19):2811–2812. doi: 10.1093/bioinformatics/btu393. [DOI] [PubMed] [Google Scholar]
  • 50.Logsdon G.A., Vollger M.R., Eichler E.E. Long-read human genome sequencing and its applications. Nat Rev Genet. 2020;21(10):597–614. doi: 10.1038/s41576-020-0236-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Conlin L.K., Aref-Eshghi E., McEldrew D.A., Luo M., Rajagopalan R. Long-read sequencing for molecular diagnostics in constitutional genetic disorders. Hum Mutat. 2022;43(11):1531–1544. doi: 10.1002/humu.24465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ameur A., Kloosterman W.P., Hestand M.S. Single-molecule sequencing: towards clinical applications. Trends Biotechnol. 2019;37(1):72–85. doi: 10.1016/j.tibtech.2018.07.013. [DOI] [PubMed] [Google Scholar]
  • 53.Ardui S., Ameur A., Vermeesch J.R., Hestand M.S. Single molecule real-time (SMRT) sequencing comes of age: applications and utilities for medical diagnostics. Nucleic Acids Res. 2018;46(5):2159–2168. doi: 10.1093/nar/gky066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Merker J.D., Wenger A.M., Sneddon T., et al. Long-read genome sequencing identifies causal structural variation in a Mendelian disease. Genet Med. 2018;20(1):159–163. doi: 10.1038/gim.2017.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cretu Stancu M., van Roosmalen M.J., Renkens I., et al. Mapping and phasing of structural variation in patient genomes using nanopore sequencing. Nat Commun. 2017;8(1):1326. doi: 10.1038/s41467-017-01343-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Liang Q., Liu Y., Liu Y., et al. Comprehensive analysis of fragile X syndrome: full characterization of the FMR1 locus by long-read sequencing. Clin Chem. 2022;68(12):1529–1540. doi: 10.1093/clinchem/hvac154. [DOI] [PubMed] [Google Scholar]
  • 57.Zamora-Cánovas A., de la Morena-Barrio B., Marín-Quilez A., et al. Targeted long-read sequencing identifies and characterizes structural variants in cases of inherited platelet disorders. J Thromb Haemost. 2024;22(3):851–859. doi: 10.1016/j.jtha.2023.11.007. [DOI] [PubMed] [Google Scholar]
  • 58.Matthijs G., Souche E., Alders M., et al. Erratum: guidelines for diagnostic next-generation sequencing. Eur J Hum Genet. 2016;24(10):1515. doi: 10.1038/ejhg.2016.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Pratt V.M., Everts R.E., Aggarwal P., et al. Characterization of 137 genomic DNA reference materials for 28 pharmacogenetic genes: a GeT-RM collaborative project. J Mol Diagn. 2016;18(1):109–123. doi: 10.1016/j.jmoldx.2015.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Karczewski K.J., Francioli L.C., Tiao G., et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581(7809):434–443. doi: 10.1038/s41586-020-2308-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Landrum M.J., Lee J.M., Benson M., et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 2016;44(D1):D862–D868. doi: 10.1093/nar/gkv1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Richards S., Aziz N., Bale S., 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. Genet Med. 2015;17(5):405–424. doi: 10.1038/gim.2015.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Stenson P.D., Ball E.V., Mort M., et al. Human Gene Mutation Database (HGMD): 2003 update. Hum Mutat. 2003;21(6):577–581. doi: 10.1002/humu.10212. [DOI] [PubMed] [Google Scholar]
  • 64.Rehm H.L., Berg J.S., Brooks L.D., et al. ClinGen—the clinical genome resource. N Engl J Med. 2015;372(23):2235–2242. doi: 10.1056/NEJMsr1406261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Caudle K.E., Dunnenberger H.M., Freimuth R.R., et al. Standardizing terms for clinical pharmacogenetic test results: consensus terms from the Clinical Pharmacogenetics Implementation Consortium (CPIC) Genet Med. 2017;19(2):215–223. doi: 10.1038/gim.2016.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Amberger J.S., Bocchini C.A., Scott A.F., Hamosh A. OMIM.org: leveraging knowledge across phenotype-gene relationships. Nucleic Acids Res. 2019;47(D1):D1038–D1043. doi: 10.1093/nar/gky1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ellard S, Baple EL, Callaway A, et al. ACGS best practice guidelines for variant classification in rare disease 2020. In: Vol. 4.01.1-32. ACGS; April 2 2020.
  • 68.Whirl-Carrillo M., Huddart R., Gong L., et al. An evidence-based framework for evaluating pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2021;110(3):563–572. doi: 10.1002/cpt.2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Taliun D., Harris D.N., Kessler M.D., et al. Sequencing of 53,831 diverse genomes from the Nhlbi TOPMed Program. Nature. 2021;590(7845):290–299. doi: 10.1038/s41586-021-03205-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.DiStefano M.T., Goehringer S., Babb L., et al. The Gene Curation Coalition: a global effort to harmonize gene-disease evidence resources. Genet Med. 2022;24(8):1732–1742. doi: 10.1016/j.gim.2022.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Firth H.V., Richards S.M., Bevan A.P., et al. DECIPHER: database of chromosomal imbalance and phenotype in humans using Ensembl resources. Am J Hum Genet. 2009;84(4):524–533. doi: 10.1016/j.ajhg.2009.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.MacDonald J.R., Ziman R., Yuen R.K.C., Feuk L., Scherer S.W. The Database of Genomic Variants: a curated collection of structural variation in the human genome. Nucleic Acids Res. 2014;42(database issue):D986–D992. doi: 10.1093/nar/gkt958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Riggs E.R., Nelson T., Merz A., et al. Copy number variant discrepancy resolution using the ClinGen dosage sensitivity map results in updated clinical interpretations in ClinVar. Hum Mutat. 2018;39(11):1650–1659. doi: 10.1002/humu.23610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Riggs E.R., Andersen E.F., Cherry A.M., et al. Technical standards for the interpretation and reporting of constitutional copy-number variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics (ACMG) and the Clinical Genome Resource (ClinGen) Genet Med. 2020;22(2):245–257. doi: 10.1038/s41436-019-0686-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kogelnik A.M., Lott M.T., Brown M.D., Navathe S.B., Wallace D.C. MITOMAP: a human mitochondrial genome database. Nucleic Acids Res. 1996;24(1):177–179. doi: 10.1093/nar/24.1.177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.McCormick E.M., Lott M.T., Dulik M.C., et al. Specifications of the ACMG/AMP standards and guidelines for mitochondrial DNA variant interpretation. Hum Mutat. 2020;41(12):2028–2057. doi: 10.1002/humu.24107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ng S.B., Buckingham K.J., Lee C., et al. Exome sequencing identifies the cause of a Mendelian disorder. Nat Genet. 2010;42(1):30–35. doi: 10.1038/ng.499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Pfundt R., Del Rosario M., Vissers L.E.L.M., et al. Detection of clinically relevant copy-number variants by exome sequencing in a large cohort of genetic disorders. Genet Med. 2017;19(6):667–675. doi: 10.1038/gim.2016.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Retterer K., Juusola J., Cho M.T., et al. Clinical application of whole-exome sequencing across clinical indications. Genet Med. 2016;18(7):696–704. doi: 10.1038/gim.2015.148. [DOI] [PubMed] [Google Scholar]
  • 80.Yang Y., Muzny D.M., Reid J.G., et al. Clinical whole-exome sequencing for the diagnosis of Mendelian disorders. N Engl J Med. 2013;369(16):1502–1511. doi: 10.1056/NEJMoa1306555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Guan Q., Balciuniene J., Cao K., et al. AUDIOME: a tiered exome sequencing-based comprehensive gene panel for the diagnosis of heterogeneous nonsyndromic sensorineural hearing loss. Genet Med. 2018;20(12):1600–1608. doi: 10.1038/gim.2018.48. [DOI] [PubMed] [Google Scholar]
  • 82.Niazi R., Gonzalez M.A., Balciuniene J., Evans P., Sarmady M., Abou Tayoun A.N. The development and validation of clinical exome-based panels using ExomeSlicer: considerations and proof of concept using an epilepsy panel. J Mol Diagn. 2018;20(5):643–652. doi: 10.1016/j.jmoldx.2018.05.003. [DOI] [PubMed] [Google Scholar]
  • 83.SoRelle J.A., Funke B.H., Eno C.C., et al. Slice testing - considerations from ordering to reporting: a joint report of the Association for Molecular Pathology, College of American Pathologists, and National Society of Genetic Counselors. J Mol Diagn. 2024;26(3):159–167. doi: 10.1016/j.jmoldx.2023.11.008. [DOI] [PubMed] [Google Scholar]
  • 84.Rajagopalan R., Gilbert M.A., McEldrew D.A., et al. Genome sequencing increases diagnostic yield in clinically diagnosed Alagille syndrome patients with previously negative test results. Genet Med. 2021;23(2):323–330. doi: 10.1038/s41436-020-00989-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Alfares A., Aloraini T., Subaie L.A., et al. Whole-genome sequencing offers additional but limited clinical utility compared with reanalysis of whole-exome sequencing. Genet Med. 2018;20(11):1328–1333. doi: 10.1038/gim.2018.41. [DOI] [PubMed] [Google Scholar]
  • 86.Soden S.E., Saunders C.J., Willig L.K., et al. Effectiveness of exome and genome sequencing guided by acuity of illness for diagnosis of neurodevelopmental disorders. Sci Transl Med. 2014;6(265) doi: 10.1126/scitranslmed.3010076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Clark M.M., Hildreth A., Batalov S., et al. Diagnosis of genetic diseases in seriously ill children by rapid whole-genome sequencing and automated phenotyping and interpretation. Sci Transl Med. 2019;11(489) doi: 10.1126/scitranslmed.aat6177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Stark Z., Dolman L., Manolio T.A., et al. Integrating genomics into healthcare: a global responsibility. Am J Hum Genet. 2019;104(1):13–20. doi: 10.1016/j.ajhg.2018.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wojcik M.H., Reuter C.M., Marwaha S., et al. Beyond the exome: what’s next in diagnostic testing for Mendelian conditions. Am J Hum Genet. 2023;110(8):1229–1248. doi: 10.1016/j.ajhg.2023.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Postel M.D., Culver J.O., Ricker C., Craig D.W. Transcriptome analysis provides critical answers to the “variants of uncertain significance” conundrum. Hum Mutat. 2022;43(11):1590–1608. doi: 10.1002/humu.24394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Horton C., Hoang L., Zimmermann H., et al. Diagnostic outcomes of concurrent DNA and RNA sequencing in individuals undergoing hereditary cancer testing. JAMA Oncol. 2024;10(2):212–219. doi: 10.1001/jamaoncol.2023.5586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Committee opinion no. 691: carrier screening for genetic conditions. Obstet Gynecol. 2017;129(3):e41–e55. doi: 10.1097/AOG.0000000000001952. [DOI] [PubMed] [Google Scholar]
  • 93.Committee opinion no. 690: carrier screening in the age of genomic medicine. Obstet Gynecol. 2017;129(3):e35–e40. doi: 10.1097/AOG.0000000000001951. [DOI] [PubMed] [Google Scholar]
  • 94.Watson M.S., Cutting G.R., Desnick R.J., et al. Cystic fibrosis population carrier screening: 2004 revision of American College of Medical Genetics mutation panel. Genet Med. 2004;6(5):387–391. doi: 10.1097/01.gim.0000139506.11694.7c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Scott S.A., Edelmann L., Liu L., Luo M., Desnick R.J., Kornreich R. Experience with carrier screening and prenatal diagnosis for 16 Ashkenazi Jewish genetic diseases. Hum Mutat. 2010;31(11):1240–1250. doi: 10.1002/humu.21327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Wapner R.J., Biggio J.R. Commentary: expanded carrier screening: how much is too much? Genet Med. 2019;21(9):1927–1930. doi: 10.1038/s41436-019-0514-1. [DOI] [PubMed] [Google Scholar]
  • 97.Stevens B., Krstic N., Jones M., Murphy L., Hoskovec J. Finding middle ground in constructing a clinically useful expanded carrier screening panel. Obstet Gynecol. 2017;130(2):279–284. doi: 10.1097/AOG.0000000000002139. [DOI] [PubMed] [Google Scholar]
  • 98.Ben-Shachar R., Svenson A., Goldberg J.D., Muzzey D. A data-driven evaluation of the size and content of expanded carrier screening panels. Genet Med. 2019;21(9):1931–1939. doi: 10.1038/s41436-019-0466-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kaseniit K.E., Haque I.S., Goldberg J.D., Shulman L.P., Muzzey D. Genetic ancestry analysis on >93,000 individuals undergoing expanded carrier screening reveals limitations of ethnicity-based medical guidelines. Genet Med. 2020;22(10):1694–1702. doi: 10.1038/s41436-020-0869-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Feng Y., Ge X., Meng L., et al. The next generation of population-based spinal muscular atrophy carrier screening: comprehensive pan-ethnic SMN1 copy-number and sequence variant analysis by massively parallel sequencing. Genet Med. 2017;19(8):936–944. doi: 10.1038/gim.2016.215. [DOI] [PubMed] [Google Scholar]
  • 101.Punj S., Akkari Y., Huang J., et al. Preconception carrier screening by genome sequencing: results from the Clinical Laboratory. Am J Hum Genet. 2018;102(6):1078–1089. doi: 10.1016/j.ajhg.2018.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Himes P., Kauffman T.L., Muessig K.R., et al. Genome sequencing and carrier testing: decisions on categorization and whether to disclose results of carrier testing. Genet Med. 2017;19(7):803–808. doi: 10.1038/gim.2016.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Chen S., Francioli L.C., Goodrich J.K., et al. A genomic mutational constraint map using variation in 76,156 human genomes. Nature. 2024;625(7993):92–100. doi: 10.1038/s41586-023-06045-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kosugi S., Momozawa Y., Liu X., Terao C., Kubo M., Kamatani Y. Comprehensive evaluation of structural variation detection algorithms for whole genome sequencing. Genome Biol. 2019;20(1):117. doi: 10.1186/s13059-019-1720-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhou A., Lin T., Xing J. Evaluating nanopore sequencing data processing pipelines for structural variation identification. Genome Biol. 2019;20(1):237. doi: 10.1186/s13059-019-1858-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Chaubey A., Shenoy S., Mathur A., et al. Low-pass genome sequencing: validation and diagnostic utility from 409 clinical cases of low-pass genome sequencing for the detection of copy number variants to replace constitutional microarray. J Mol Diagn. 2020;22(6):823–840. doi: 10.1016/j.jmoldx.2020.03.008. [DOI] [PubMed] [Google Scholar]
  • 107.Dong Z., Chau M.H.K., Zhang Y., et al. Low-pass genome sequencing-based detection of absence of heterozygosity: validation in clinical cytogenetics. Genet Med. 2021;23(7):1225–1233. doi: 10.1038/s41436-021-01128-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Redin C., Brand H., Collins R.L., et al. The genomic landscape of balanced cytogenetic abnormalities associated with human congenital anomalies. Nat Genet. 2017;49(1):36–45. doi: 10.1038/ng.3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Dong Z., Ye L., Yang Z., et al. Balanced chromosomal rearrangement detection by low-pass whole-genome sequencing. Curr Protoc Hum Genet. 2018;96(8):18.1–8.18.16. doi: 10.1002/cphg.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Savara J., Novosád T., Gajdoš P., Kriegová E. Comparison of structural variants detected by optical mapping with long-read next-generation sequencing. Bioinformatics. 2021;37(20):3398–3404. doi: 10.1093/bioinformatics/btab359. [DOI] [PubMed] [Google Scholar]
  • 111.Practice bulletin no. 162: prenatal diagnostic testing for genetic disorders. Obstet Gynecol. 2016;127(5):e108–e122. doi: 10.1097/AOG.0000000000001405. [DOI] [PubMed] [Google Scholar]
  • 112.Committee on Genetics and the Society for Maternal-Fetal Medicine Committee opinion no.682: microarrays and next-generation sequencing technology: the use of advanced genetic diagnostic tools in obstetrics and gynecology. Obstet Gynecol. 2016;128(6):e262–e268. doi: 10.1097/AOG.0000000000001817. [DOI] [PubMed] [Google Scholar]
  • 113.Atwal P.S., Brennan M.L., Cox R., et al. Clinical whole-exome sequencing: are we there yet? Genet Med. 2014;16(9):717–719. doi: 10.1038/gim.2014.10. [DOI] [PubMed] [Google Scholar]
  • 114.Pauta M., Martinez-Portilla R.J., Borrell A. Diagnostic yield of exome sequencing in fetuses with an isolated increased nuchal translucency: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2022;59(1):26–32. doi: 10.1002/uog.23746. [DOI] [PubMed] [Google Scholar]
  • 115.Filges I., Miny P., Holzgreve W., Tercanli S. How genomics is changing the practice of prenatal testing. J Perinat Med. 2021;49(8):1003–1010. doi: 10.1515/jpm-2021-0220. [DOI] [PubMed] [Google Scholar]
  • 116.Donley G., Hull S.C., Berkman B.E. Prenatal whole genome sequencing: just because we can, should we? Hastings Cent Rep. 2012;42(4):28–40. doi: 10.1002/hast.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Talkowski M.E., Ordulu Z., Pillalamarri V., et al. Clinical diagnosis by whole-genome sequencing of a prenatal sample. N Engl J Med. 2012;367(23):2226–2232. doi: 10.1056/NEJMoa1208594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Ordulu Z., Kammin T., Brand H., et al. Structural chromosomal rearrangements require nucleotide-level resolution: lessons from next-generation sequencing in prenatal diagnosis. Am J Hum Genet. 2016;99(5):1015–1033. doi: 10.1016/j.ajhg.2016.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Wang H., Dong Z., Zhang R., et al. Low-pass genome sequencing versus chromosomal microarray analysis: implementation in prenatal diagnosis. Genet Med. 2020;22(3):500–510. doi: 10.1038/s41436-019-0634-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Gorman G.S., Chinnery P.F., DiMauro S., et al. Mitochondrial diseases. Nat Rev Dis Primers. 2016;2 doi: 10.1038/nrdp.2016.80. [DOI] [PubMed] [Google Scholar]
  • 121.DiMauro S., Schon E.A. Mitochondrial respiratory-chain diseases. N Engl J Med. 2003;348(26):2656–2668. doi: 10.1056/NEJMra022567. [DOI] [PubMed] [Google Scholar]
  • 122.Clima R., Preste R., Calabrese C., et al. HmtDB 2016: data update, a better performing query system and human mitochondrial DNA haplogroup predictor. Nucleic Acids Res. 2017;45(D1):D698–D706. doi: 10.1093/nar/gkw1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Preste R., Vitale O., Clima R., Gasparre G., Attimonelli M. HmtVar: a new resource for human mitochondrial variations and pathogenicity data. Nucleic Acids Res. 2019;47(D1):D1202–D1210. doi: 10.1093/nar/gky1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Bhardwaj A., Mukerji M., Sharma S., et al. MtSNPscore: a combined evidence approach for assessing cumulative impact of mitochondrial variations in disease. BMC Bioinformatics. August 27 2009;10(suppl 8):S7. doi: 10.1186/1471-2105-10-S8-S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Shen L., Attimonelli M., Bai R., et al. MSeqDR mvTool: a mitochondrial DNA Web and API resource for comprehensive variant annotation, universal nomenclature collation, and reference genome conversion. Hum Mutat. 2018;39(6):806–810. doi: 10.1002/humu.23422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bai R.K., Wong L.J.C. Detection and quantification of heteroplasmic mutant mitochondrial DNA by real-time amplification refractory mutation system quantitative PCR analysis: a single-step approach. Clin Chem. 2004;50(6):996–1001. doi: 10.1373/clinchem.2004.031153. [DOI] [PubMed] [Google Scholar]
  • 127.Chinault A.C., Shaw C.A., Brundage E.K., Tang L.Y., Wong L.J.C. Application of dual-genome oligonucleotide array-based comparative genomic hybridization to the molecular diagnosis of mitochondrial DNA deletion and depletion syndromes. Genet Med. 2009;11(7):518–526. doi: 10.1097/GIM.0b013e3181abd83c. [DOI] [PubMed] [Google Scholar]
  • 128.Cui H., Li F., Chen D., et al. Comprehensive next-generation sequence analyses of the entire mitochondrial genome reveal new insights into the molecular diagnosis of mitochondrial DNA disorders. Genet Med. 2013;15(5):388–394. doi: 10.1038/gim.2012.144. [DOI] [PubMed] [Google Scholar]
  • 129.Picardi E., Pesole G. Mitochondrial genomes gleaned from human whole-exome sequencing. Nat Methods. 2012;9(6):523–524. doi: 10.1038/nmeth.2029. [DOI] [PubMed] [Google Scholar]
  • 130.Falk M.J., Pierce E.A., Consugar M., et al. Mitochondrial disease genetic diagnostics: optimized whole-exome analysis for all MitoCarta nuclear genes and the mitochondrial genome. Discov Med. 2012;14(79):389–399. [PMC free article] [PubMed] [Google Scholar]
  • 131.Wagner M., Berutti R., Lorenz-Depiereux B., et al. Mitochondrial DNA mutation analysis from exome sequencing-A more holistic approach in diagnostics of suspected mitochondrial disease. J Inherit Metab Dis. 2019;42(5):909–917. doi: 10.1002/jimd.12109. [DOI] [PubMed] [Google Scholar]
  • 132.Pronicka E., Piekutowska-Abramczuk D., Ciara E., et al. New perspective in diagnostics of mitochondrial disorders: two years’ experience with whole-exome sequencing at a national paediatric centre. J Transl Med. 2016;14(1):174. doi: 10.1186/s12967-016-0930-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Theunissen T.E.J., Nguyen M., Kamps R., et al. Whole exome sequencing is the preferred strategy to identify the genetic defect in patients with a probable or possible mitochondrial cause. Front Genet. 2018;9:400. doi: 10.3389/fgene.2018.00400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Raymond F.L., Horvath R., Chinnery P.F. First-line genomic diagnosis of mitochondrial disorders. Nat Rev Genet. 2018;19(7):399–400. doi: 10.1038/s41576-018-0022-1. [DOI] [PubMed] [Google Scholar]
  • 135.Riley L.G., Cowley M.J., Gayevskiy V., et al. The diagnostic utility of genome sequencing in a pediatric cohort with suspected mitochondrial disease. Genet Med. 2020;22(7):1254–1261. doi: 10.1038/s41436-020-0793-6. [DOI] [PubMed] [Google Scholar]
  • 136.Schon K.R., Horvath R., Wei W., et al. Use of whole genome sequencing to determine genetic basis of suspected mitochondrial disorders: cohort study. BMJ. 2021;375 doi: 10.1136/bmj-2021-066288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Scott S.A. Personalizing medicine with clinical pharmacogenetics. Genet Med. December 2011;13(12):987–995. doi: 10.1097/GIM.0b013e318238b38c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Relling M.V., Klein T.E., Gammal R.S., Whirl-Carrillo M., Hoffman J.M., Caudle K.E. The clinical pharmacogenetics implementation consortium: 10 years later. Clin Pharmacol Ther. 2020;107(1):171–175. doi: 10.1002/cpt.1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Rubinstein W.S., Pacanowski M. Pharmacogenetic gene-drug associations: FDA perspective on what physicians need to know. Am Fam Physician. 2021;104(1):16–19. [PubMed] [Google Scholar]
  • 140.Tayeh M.K., Gaedigk A., Goetz M.P., et al. Clinical pharmacogenomic testing and reporting: a technical standard of the American College of Medical Genetics and Genomics (ACMG) Genet Med. 2022;24(4):759–768. doi: 10.1016/j.gim.2021.12.009. [DOI] [PubMed] [Google Scholar]
  • 141.Pratt V.M., Cavallari L.H., Del Tredici A.L., et al. Recommendations for clinical CYP2D6 genotyping allele selection: a joint consensus recommendation of the association for molecular pathology, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, and the European Society for Pharmacogenomics and Personalized Therapy. J Mol Diagn. 2021;23(9):1047–1064. doi: 10.1016/j.jmoldx.2021.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Pratt V.M., Cavallari L.H., Del Tredici A.L., et al. Recommendations for clinical warfarin genotyping allele selection: a report of the association for molecular pathology and the College of American Pathologists. J Mol Diagn. 2020;22(7):847–859. doi: 10.1016/j.jmoldx.2020.04.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Pratt V.M., Cavallari L.H., Del Tredici A.L., et al. Recommendations for clinical CYP2C9 genotyping allele selection: a joint recommendation of the association for molecular pathology and College of American Pathologists. J Mol Diagn. 2019;21(5):746–755. doi: 10.1016/j.jmoldx.2019.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Pratt V.M., Cavallari L.H., Fulmer M.L., et al. TPMT and NUDT15 genotyping recommendations: a joint consensus recommendation of the association for molecular pathology, Clinical Pharmacogenetics Implementation Consortium, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, European Society for Pharmacogenomics and Personalized Therapy, and Pharmacogenomics Knowledgebase. J Mol Diagn. 2022;24(10):1051–1063. doi: 10.1016/j.jmoldx.2022.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Pratt V.M., Cavallari L.H., Fulmer M.L., et al. CYP3A4 and CYP3A5 genotyping recommendations: a joint consensus recommendation of the association for molecular pathology, Clinical Pharmacogenetics Implementation Consortium, College of American Pathologists, Dutch Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association, European Society for Pharmacogenomics and Personalized Therapy, and Pharmacogenomics Knowledgebase. J Mol Diagn. 2023;25(9):619–629. doi: 10.1016/j.jmoldx.2023.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Pratt V.M., Del Tredici A.L., Hachad H., et al. Recommendations for clinical CYP2C19 genotyping allele selection: a report of the association for molecular pathology. J Mol Diagn. 2018;20(3):269–276. doi: 10.1016/j.jmoldx.2018.01.011. [DOI] [PubMed] [Google Scholar]
  • 147.Scott S.A., Scott E.R., Seki Y., et al. Development and analytical validation of a 29 gene clinical pharmacogenetic genotyping panel: multi-ethnic allele and copy number variant detection. Clin Transl Sci. 2021;14(1):204–213. doi: 10.1111/cts.12844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Turner A.J., Nofziger C., Ramey B.E., et al. PharmVar tutorial on CYP2D6 structural variation testing and recommendations on reporting. Clin Pharmacol Ther. 2023;114(6):1220–1237. doi: 10.1002/cpt.3044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Martis S., Mei H., Vijzelaar R., Edelmann L., Desnick R.J., Scott S.A. Multi-ethnic cytochrome-P450 copy number profiling: novel pharmacogenetic alleles and mechanism of copy number variation formation. Pharmacogenomics J. 2013;13(6):558–566. doi: 10.1038/tpj.2012.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Botton M.R., Lu X., Zhao G., et al. Structural variation at the CYP2C locus: characterization of deletion and duplication alleles. Hum Mutat. 2019;40(11):e37–e51. doi: 10.1002/humu.23855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Gaedigk A., Ndjountché L., Divakaran K., et al. Cytochrome P4502D6 (CYP2D6) gene locus heterogeneity: characterization of gene duplication events. Clin Pharmacol Ther. 2007;81(2):242–251. doi: 10.1038/sj.clpt.6100033. [DOI] [PubMed] [Google Scholar]
  • 152.Ramamoorthy A., Skaar T.C. Gene copy number variations: it is important to determine which allele is affected. Pharmacogenomics. 2011;12(3):299–301. doi: 10.2217/pgs.11.5. [DOI] [PubMed] [Google Scholar]
  • 153.Qiao W., Martis S., Mendiratta G., et al. Integrated CYP2D6 interrogation for multiethnic copy number and tandem allele detection. Pharmacogenomics. 2019;20(1):9–20. doi: 10.2217/pgs-2018-0135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Rasmussen-Torvik L.J., Almoguera B., Doheny K.F., et al. Concordance between research sequencing and clinical pharmacogenetic genotyping in the eMERGE-PGx study. J Mol Diagn. 2017;19(4):561–566. doi: 10.1016/j.jmoldx.2017.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Yang W., Wu G., Broeckel U., et al. Comparison of genome sequencing and clinical genotyping for pharmacogenes. Clin Pharmacol Ther. 2016;100(4):380–388. doi: 10.1002/cpt.411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Qiao W., Yang Y., Sebra R., et al. Long-read single molecule real-time full gene sequencing of cytochrome P450-2D6. Hum Mutat. 2016;37(3):315–323. doi: 10.1002/humu.22936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Botton M.R., Yang Y., Scott E.R., Desnick R.J., Scott S.A. Phased haplotype resolution of the SLC6A4 promoter using long-read single molecule real-time (SMRT) sequencing. Genes (Basel) 2020;11(11) doi: 10.3390/genes11111333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Cohn I., Paton T.A., Marshall C.R., et al. Genome sequencing as a platform for pharmacogenetic genotyping: a pediatric cohort study. npj Genom Med. 2017;2:19. doi: 10.1038/s41525-017-0021-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Lee S.B., Wheeler M.M., Thummel K.E., Nickerson D.A. Calling star alleles with stargazer in 28 pharmacogenes with whole genome sequences. Clin Pharmacol Ther. 2019;106(6):1328–1337. doi: 10.1002/cpt.1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Twesigomwe D., Drögemöller B.I., Wright G.E.B., et al. StellarPGx: a Nextflow pipeline for calling star alleles in cytochrome P450 genes. Clin Pharmacol Ther. 2021;110(3):741–749. doi: 10.1002/cpt.2173. [DOI] [PubMed] [Google Scholar]
  • 161.Sangkuhl K., Whirl-Carrillo M., Whaley R.M., et al. Pharmacogenomics clinical annotation tool (PharmCAT) Clin Pharmacol Ther. 2020;107(1):203–210. doi: 10.1002/cpt.1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Twist G.P., Gaedigk A., Miller N.A., et al. Constellation: a tool for rapid, automated phenotype assignment of a highly polymorphic pharmacogene, CYP2D6, from whole-genome sequences. npj Genom Med. 2016;1 doi: 10.1038/npjgenmed.2015.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Gaedigk A., Casey S.T., Whirl-Carrillo M., Miller N.A., Klein T.E. Pharmacogene Variation Consortium: a global resource and repository for pharmacogene variation. Clin Pharmacol Ther. 2021;110(3):542–545. doi: 10.1002/cpt.2321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Pan A., Scodellaro S., Khan T., et al. Pharmacogenetic profiling via genome sequencing in children with medical complexity. Pediatr Res. 2023;93(4):905–910. doi: 10.1038/s41390-022-02313-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Caspar S.M., Schneider T., Stoll P., Meienberg J., Matyas G. Potential of whole-genome sequencing-based pharmacogenetic profiling. Pharmacogenomics. 2021;22(3):177–190. doi: 10.2217/pgs-2020-0155. [DOI] [PubMed] [Google Scholar]
  • 166.Ka S., Lee S., Hong J., et al. HLAscan: genotyping of the HLA region using next-generation sequencing data. BMC Bioinformatics. 2017;18(1):258. doi: 10.1186/s12859-017-1671-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Lewis C.M., Vassos E. Polygenic risk scores: from research tools to clinical instruments. Genome Med. 2020;12(1):44. doi: 10.1186/s13073-020-00742-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Lambert S.A., Abraham G., Inouye M. Towards clinical utility of polygenic risk scores. Hum Mol Genet. 2019;28(R2):R133–R142. doi: 10.1093/hmg/ddz187. [DOI] [PubMed] [Google Scholar]
  • 169.Wand H., Lambert S.A., Tamburro C., et al. Improving reporting standards for polygenic scores in risk prediction studies. Nature. 2021;591(7849):211–219. doi: 10.1038/s41586-021-03243-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Reddi H.V., Wand H., Funke B., et al. Laboratory perspectives in the development of polygenic risk scores for disease: a points to consider statement of the American College of Medical Genetics and Genomics (ACMG) Genet Med. 2023;25(5) doi: 10.1016/j.gim.2023.100804. [DOI] [PubMed] [Google Scholar]
  • 171.Homburger J.R., Neben C.L., Mishne G., Zhou A.Y., Kathiresan S., Khera A.V. Low coverage whole genome sequencing enables accurate assessment of common variants and calculation of genome-wide polygenic scores. Genome Med. 2019;11(1):74. doi: 10.1186/s13073-019-0682-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Cavazos T.B., Witte J.S. Inclusion of variants discovered from diverse populations improves polygenic risk score transferability. HGG Adv. 2021;2(1) doi: 10.1016/j.xhgg.2020.100017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Meienberg J., Bruggmann R., Oexle K., Matyas G. Clinical sequencing: is WGS the better WES? Hum Genet. 2016;135(3):359–362. doi: 10.1007/s00439-015-1631-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Miller D.E., Sulovari A., Wang T., et al. Targeted long-read sequencing identifies missing disease-causing variation. Am J Hum Genet. 2021;108(8):1436–1449. doi: 10.1016/j.ajhg.2021.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.De Coster W., De Rijk P., De Roeck A., et al. Structural variants identified by Oxford nanopore PromethION sequencing of the human genome. Genome Res. 2019;29(7):1178–1187. doi: 10.1101/gr.244939.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Fang L., Kao C., Gonzalez M.V., et al. LinkedSV for detection of mosaic structural variants from linked-read exome and genome sequencing data. Nat Commun. 2019;10(1):5585. doi: 10.1038/s41467-019-13397-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Trost B., Walker S., Haider S.A., et al. Impact of DNA source on genetic variant detection from human whole-genome sequencing data. J Med Genet. 2019;56(12):809–817. doi: 10.1136/jmedgenet-2019-106281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Chen R., Aldred M.A., Xu W., et al. Comparison of whole genome sequencing and targeted sequencing for mitochondrial DNA. Mitochondrion. 2021;58:303–310. doi: 10.1016/j.mito.2021.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Yeung A., Tan N.B., Tan T.Y., et al. A cost-effectiveness analysis of genomic sequencing in a prospective versus historical cohort of complex pediatric patients. Genet Med. 2020;22(12):1986–1993. doi: 10.1038/s41436-020-0929-8. [DOI] [PubMed] [Google Scholar]
  • 180.Incerti D., Xu X.M., Chou J.W., Gonzaludo N., Belmont J.W., Schroeder B.E. Cost-effectiveness of genome sequencing for diagnosing patients with undiagnosed rare genetic diseases. Genet Med. 2022;24(1):109–118. doi: 10.1016/j.gim.2021.08.015. [DOI] [PubMed] [Google Scholar]
  • 181.Lavelle T.A., Feng X., Keisler M., et al. Cost-effectiveness of exome and genome sequencing for children with rare and undiagnosed conditions. Genet Med. 2022;24(6):1349–1361. doi: 10.1016/j.gim.2022.03.005. [DOI] [PubMed] [Google Scholar]
  • 182.Schwarze K., Buchanan J., Taylor J.C., Wordsworth S. Are whole-exome and whole-genome sequencing approaches cost-effective? A systematic review of the literature. Genet Med. 2018;20(10):1122–1130. doi: 10.1038/gim.2017.247. [DOI] [PubMed] [Google Scholar]
  • 183.Sahajpal N.S., Barseghyan H., Kolhe R., Hastie A., Chaubey A. Optical genome mapping as a next-generation cytogenomic tool for detection of structural and copy number variations for prenatal genomic analyses. Genes (Basel) 2021;12(3):398. doi: 10.3390/genes12030398. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Genetics in Medicine Open are provided here courtesy of Elsevier

RESOURCES