Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Whole exome sequencing in neurogenetic odysseys: An effective, cost- and time-saving diagnostic approach

  • Marta Córdoba,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Software, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina, Programa de Medicina de Precisión y Genómica Clínica, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina

  • Sergio Alejandro Rodriguez-Quiroga,

    Roles Data curation, Writing – review & editing

    Affiliation Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina

  • Patricia Analía Vega,

    Roles Data curation, Writing – review & editing

    Affiliation Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina

  • Valeria Salinas,

    Roles Writing – original draft

    Affiliations Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina, Programa de Medicina de Precisión y Genómica Clínica, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina

  • Josefina Perez-Maturo,

    Roles Writing – review & editing

    Affiliations Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina, Programa de Medicina de Precisión y Genómica Clínica, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina

  • Hernán Amartino,

    Roles Data curation

    Affiliation Servicio de Neurología Infantil, Hospital Universitario Austral, Buenos Aires, Argentina

  • Cecilia Vásquez-Dusefante,

    Roles Data curation

    Affiliation Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina

  • Nancy Medina,

    Roles Data curation, Formal analysis, Investigation

    Affiliation Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina

  • Dolores González-Morón,

    Roles Data curation, Investigation

    Affiliation Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina

  • Marcelo Andrés Kauffman

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – review & editing

    marcelokauffman@gmail.com

    Affiliations Consultorio de Neurogenética, Centro Universitario de Neurologia y Division Neurologia, Hospital J.M.Ramos Mejia, Facultad de Medicina, UBA, Buenos Aires, Argentina, Programa de Medicina de Precisión y Genómica Clínica, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina

Abstract

Background

Diagnostic trajectories for neurogenetic disorders frequently require the use of considerable time and resources, exposing patients and families to so-called “diagnostic odysseys”. Previous studies have provided strong evidence for increased diagnostic and clinical utility of whole-exome sequencing in medical genetics. However, specific reports assessing its utility in a setting such as ours- a neurogeneticist led academic group serving in a low-income country—are rare.

Objectives

To assess the diagnostic yield of WES in patients suspected of having a neurogenetic condition and explore the cost-effectiveness of its implementation in a research group located in an Argentinean public hospital.

Methods

This is a prospective study of the clinical utility of WES in a series of 40 consecutive patients selected from a Neurogenetic Clinic of a tertiary Hospital in Argentina. We evaluated patients retrospectively for previous diagnostic trajectories. Diagnostic yield, clinical impact on management and economic diagnostic burden were evaluated.

Results

We demonstrated the clinical utility of Whole Exome Sequencing in our patient cohort, obtaining a diagnostic yield of 40% (95% CI, 24.8%-55.2%) among a diverse group of neurological disorders. The average age at the time of WES was 23 (range 3–70). The mean time elapsed from symptom onset to WES was 11 years (range 3–42). The mean cost of the diagnostic workup prior to WES was USD 1646 (USD 1439 to 1853), which is 60% higher than WES cost in our center.

Conclusions

WES for neurogenetics proved to be an effective, cost- and time-saving approach for the molecular diagnosis of this heterogeneous and complex group of patients.

Introduction

Neurogenetic disorders are a frequent reason for medical consultation in neurology services. Clinical variability and genetic heterogeneity are a hallmark of these diseases. Their diagnostic approach requires extensive clinical, radiological and genetic evaluations. Moreover, many of these procedures are invasive and costly. However, despite the use of considerable time and resources, the diagnostic yield in this field has been disappointingly low. This etiologic search has been called a “diagnostic odyssey” for many families [1].

Whole Exome Sequencing (WES) has proved to be a valuable tool in medical genetics, for diagnostic and gene discovery purposes [24]. Although a diagnostic yield of about 30% in neurogenetic disorders can be extrapolated from the results of large series that have included other medical conditions as well [5], specific reports assessing its utility in a setting such as ours—a neurogeneticist led academic group serving in a low-income country—are rare. Therefore, there is still a necessity to assess its clinical utility and the feasibility of its implementation for neurogenetic diagnostic practice in less economic favorable locations where rational and effective use of resources is both an obligation and an opportunity for reducing inequalities [6, 7].

We are reporting here on our first 40 consecutive cases which were selected from our research-based laboratory for WES. We demonstrated the clinical utility of WES and the potential cost-effectiveness of WES as a single test by examining the number and types of tests that were performed prior to WES that add to the cost of diagnostic workups.

Materials and methods

Clinical samples

We included a consecutive series of 40 patients selected for WES from a Neurogenetic Clinic of a tertiary Hospital in Argentina. These patients were considered candidates for genomic studies according to the presence of typical findings of known neurogenetic diseases and/or hints of monogenic etiology such as familial aggregation or chronic and progressive course. We recorded perinatal and family history, likely inheritance model/s, disease progression characteristics, comorbidities, and studies performed before WES from each patient of our cohort. The diverse clinical features of this cohort are summarized in Table 1. Written informed Consent for WES was obtained from the patients and/or their family. The informed consent included the option to receive or not incidental findings according to ACMG recommendations. Internal review board (IRB) approval was obtained at Hospital JM Ramos Mejia. All methods were performed in accordance with the relevant guidelines and regulations.

thumbnail
Table 1. Demographic characteristics and clinical features of patients selected for WES (*).

https://doi.org/10.1371/journal.pone.0191228.t001

Whole exome sequencing and sanger confirmation

Genomic DNA was isolated from blood samples of each subject with the use of commercial kits. DNA sequencing libraries were constructed mostly by chemical fragmentation using commercial preparation kits. Exomes were enriched using different systems, being the vast majority of our cases processed with SureSelect Human All Exon v4 Kits (Agilent Technologies, Santa Clara, CA, USA). NGS sequencing runs were made in Illumina HiSeq 2500 systems as an outsourced service from Macrogen Inc (Korea) obtaining an average sequence coverage of more than 70X, with more than 97% of the target bases having at least 10X coverage. All standardized procedures were performed according to manufacturer’s instructions that have been widely mentioned in the literature [8, 9]. Clinically relevant variants, from proband and parental samples (whenever available), were confirmed by Sanger sequencing.

Data analysis and annotation

Sequence data in FastQ format were aligned to the human reference genome (GRCh37) using the Burrows-Wheeler Alignment Tool (BWA-MEM) [10]. Variants Calls were generated using GATK haplotype caller following the so called best practices [11]. The output vcf file was annotated at various levels using Annovar [12] (S1A Fig). Variants were prioritized according to inheritance model, population frequency, molecular function and effects of mutations, reported clinical effect, and optionally according to a list of genes associated with the disease under study. In that sense two in-house protocols were defined. One “molecular hypothesis free”, for patients presenting complex phenotypes without candidate genes. Another “molecular hypothesis targeted” for patients that shows a defined clinical syndrome with available candidate genes. (S1B Fig). Classification of variants followed previously published schemes [13] updated with recent recommendations and guidelines by the American College of Medical Genetics and Genomics and the Association for Molecular Pathology [14]. Joining variant level and clinical features information, we classified each WES study as positive if a pathogenic/likely pathogenic mutation in known disease gene was identified with positive phenotypic and inheritance overlap; undetermined if a pathogenic/likely pathogenic mutation in a putative candidate gene was identified with positive phenotypic and inheritance overlap or only one pathogenic/likely pathogenic mutation was identified with positive phenotypic overlap in a recessive disorder and negative in the rest of the cases. We paid special attention to reviews of previous work done in cases studied before the 2015 update, reanalyzing them according to the new schema. Details for each novel variant are presented in S1 Table.

Incidental findings were informed according to ACMG recommendations. Counseling to patients was performed by trained professionals.

Results

WES proved to be an effective, cost- and time-saving diagnostic approach in our setting. Sixteen WES satisfied criteria for a full molecular diagnosis (Table 2 and S1 Table), thus the overall diagnostic yield for WES in our series was 40% (S2 Fig, Yield). Among them, two WES were reclassified from original undetermined and negative categories after subsequent reanalysis identified pathogenic variants in genes not associated with human disorders at the time of original reports. A diverse group of neurological disorders were represented in the positive patients (Table 2). The average age at the time of WES was 23 (3–70). The mean time elapsed from symptom onset to WES was 11 years (range 3–42). The positive group included 9 patients with autosomal dominant disease and 7 with autosomal recessive disease. Different mutation types were observed in this cohort. Noteworthy, 56% of the mutations were novel, according to ExAC v3 database [9] (Fig 1). Although almost all of the molecular diagnoses were in nuclear genes, mitochondrial genome sequencing included in the WES test yielded one diagnosis (one individual with a missense mutation in MT-T8993G.

thumbnail
Table 2. Summary of patients with established molecular diagnosis by WES.

https://doi.org/10.1371/journal.pone.0191228.t002

thumbnail
Fig 1. Location and impact of novel variants identified by this study.

https://doi.org/10.1371/journal.pone.0191228.g001

As we mention in methods, a WES study is considered positive if pathogenic or likely pathogenic variants correspond to the phenotype and the mode of inheritance. We must recognize that only in case 33, this criterion is not strictly accomplished because the identified variant in ALS2 must be considered of unknown significance according to last ACMG criteria. However, we discussed this situation with the referring physician and the patient’s family and decided to consider the ALS2 variant likely causing the disease, despite acknowledging a higher uncertainty in diagnostic terms. According to this clinical decision, we included this case as a positive one into this work.

WES were defined as undetermined in two cases (5%). In one of them, we were able to identify only one pathogenic variant (NM_018082.5:c.1568T>A; p.V523Q) in POLR3B in a patient showing clinical features consistent with autosomal recessive POLR3-related disorders [15]. We hypothesize that the second missing allele is a large deletion/insertion or a deep intronic mutation. This case highlights current limitations of WES. In case 17, we found a heterozygous likely pathogenic variant (NM_030954.3:c.668C>A; p.A223N) in RNF170 gene. This gene was reported as a cause of sensory ataxia [16]. The patient’s phenotype corresponds to pure cerebellar ataxia.

Table 2 shows a summary of the impact that a definitive diagnosis obtained from WES had on our patients. The information obtained by means of WES ended the diagnostic odysseys, led to therapeutic trials in some cases and improved genetic counselling processes with more precise information.

As an exploratory approach to a monetary cost-analysis of WES in neurogenetic diseases, we recorded the number and type of complementary tests done by our patients before WES. The average cost of the “expendable” diagnostic workup prior to WES was USD 1646 (USD 1439 to 1853), which is 60% higher than WES cost in our center (USD 1000). Table 3 shows that several genetic and non-genetic assays considered unnecessary (e.g. repetitive neuroimages and non-genetic assays) and/or evitable (e.g. recurrent outpatients visits and single-gene testing) were performed in almost all of our patients. This often-unnecessary repetition of complementary studies might be a consequence of the extension in time of the so-called diagnostic odyssey (see before results about time at WES since symptom onset). A more conservative analysis that added up WES cost and stratified the cohort into solved and unsolved cases showed differences too. The average cost of the diagnostic work up (including WES, expendable and non-expendable procedures) in solved cases was USD 4572 (USD 4302 to 4842), whereas in unsolved cases was USD 4514 (USD 4289 to 4739). Avoiding expendable procedures, by means of WES, could reduce diagnostic work up expenses in about 39% (USD 2792; 95% CI, USD 2634–2950).

thumbnail
Table 3. Summary of procedures* and visits* performed during the Diagnostic Odysseys.

https://doi.org/10.1371/journal.pone.0191228.t003

Discussion

Applying WES to a representative sample of 40 patients suffering from neurogenetic diseases, we obtained an etiologic diagnostic yield of 40%. Furthermore, we were able to expand the phenotypic spectrum of known genes and identify new pathogenic variants in other genes. Two cases were illustrative of common themes in medical genomics [17, 18]. A non-sense mutation in GRIK2 caused a more complex phenotype than it was previously recognized for this gene. This gene encodes a glutamate receptor and was previously reported once in members of a consanguineous family segregating intellectual disability [19]. Our patient also presented with intellectual disability, epilepsy, dystonia, and behavioral problems of the autism spectrum [20]. Thus, we were able to extend the phenotypic spectrum associated with this gene. We also emphasize the finding of a mutation in KCNA2 in a patient with early onset epilepsy and ataxia. This variant was identified after periodic reanalysis of previously non-diagnostic WES. Mutations in KCNA2 were recently recognized as the cause of epileptic encephalopathies and early onset ataxia [21]. This information was unknown at the moment of the initial analysis, however, being available when this WES was reassessed, it led us to reinterpret this case. Recent reports have shown that systematic re-analysis of unsolved WES data lead to about 10% additional diagnoses [22].

Our preliminary cost-analysis lend support to the assertion made by others that WES is more cost-effective than other molecular diagnostic approaches based on single- or panel- gene analysis [2, 3]. However, our estimates ought to be interpreted with caution. The retrospective design precludes us to avoid biases during the classification of previous procedures as unnecessary or evitable. We acknowledge that some of them could certainly be useful for WES interpretation and should not be considered a complete cost to be saved by WES. Nevertheless, our findings are similar to other formal analyses in this subject [23], where an early implementation of WES in the diagnostic trajectory of suspected genetic conditions proves to be cost-effective by means of a reduction in the number of procedures and specialist visits [24]. Moreover, there are other diagnostic odysseys costs that are harder to represent in monetary terms but are not less important, such as time lost to the patient and family and quality of life decrement because of this loss. They deserve other type of formal economic studies that could even show more advantages for the use of WES in the diagnostic approach of complex diseases such as neurogenetic disorders.

The diagnostic yield in less restrictive adult and pediatric populations series ranged from 17 to 30% [4, 25]. Groups that included only patients showing phenotypes involving the nervous system reported higher diagnostic yields [2628]. Our results are comparable with these experiences and highlight the advantages of working as a personalized research group where phenotypic and genotypic information can be thoughtfully assessed in contrast to commercial diagnostic laboratories that only have access to focused, heterogeneous and often less informative clinical phenotypic reports filled by the external ordering physician. Although undirected next generation sequencing tests such as WES have proved powerful and useful in the diagnosis of several genetic conditions, a targeted approach based on multi-genic panels or even single-gene assays is still justified for patients presenting with well-defined phenotypes where a higher diagnostic yield might be expected because of better coverage and more favorable cost implications [29]. However, WES have the advantage over more focused approaches, when a more comprehensive solution is needed in those patients suffering from genetically and phenotypically heterogeneous conditions [30, 31].

WES for neurogenetics proved to be an effective, cost- and time-saving approach for the molecular diagnosis of this heterogeneous and complex group of patients. It reduces the long time that these patients must wait before getting a diagnosis thereby ending odysseys of many years, impacting on their medical management, and optimizing the genetic counseling for these families. Negative WES still remain a challenge, given the complexity of genomic data interpretation and the lack of a thorough knowledge of monogenic disorders.

Supporting information

S1 Fig. Data analysis and interpretation workflow.

https://doi.org/10.1371/journal.pone.0191228.s002

(TIFF)

S2 Fig. Characteristics and diagnostic yield of our cohort according to phenotype.

https://doi.org/10.1371/journal.pone.0191228.s003

(TIFF)

References

  1. 1. Carmichael N, Tsipis J, Windmueller G, Mandel L, Estrella E. "Is it going to hurt?": the impact of the diagnostic odyssey on children and their families. J Genet Couns. 2015;24(2):325–35. Epub 2014/10/04. pmid:25277096.
  2. 2. Valencia CA, Husami A, Holle J, Johnson JA, Qian Y, Mathur A, et al. Clinical Impact and Cost-Effectiveness of Whole Exome Sequencing as a Diagnostic Tool: A Pediatric Center's Experience. Front Pediatr. 2015;3:67. Epub 2015/08/19. pmid:26284228
  3. 3. Monroe GR, Frederix GW, Savelberg SM, de Vries TI, Duran KJ, van der Smagt JJ, et al. Effectiveness of whole-exome sequencing and costs of the traditional diagnostic trajectory in children with intellectual disability. Genet Med. 2016;18(9):949–56. Epub 2016/02/05. pmid:26845106.
  4. 4. Yang Y, Muzny DM, Xia F, Niu Z, Person R, Ding Y, et al. Molecular findings among patients referred for clinical whole-exome sequencing. JAMA. 2014;312(18):1870–9. Epub 2014/10/19. pmid:25326635
  5. 5. Retterer K, Juusola J, Cho MT, Vitazka P, Millan F, Gibellini F, et al. Clinical application of whole-exome sequencing across clinical indications. Genet Med. 2016;18(7):696–704. Epub 2015/12/04. pmid:26633542.
  6. 6. Kingsmore SF, Lantos JD, Dinwiddie DL, Miller NA, Soden SE, Farrow EG, et al. Next-generation community genetics for low- and middle-income countries. Genome Med. 2012;4(3):25. Epub 2012/03/31. pmid:22458566
  7. 7. Alwan A, Modell B. Recommendations for introducing genetics services in developing countries. Nat Rev Genet. 2003;4(1):61–8. Epub 2003/01/02. pmid:12509754.
  8. 8. Kozarewa I, Turner DJ. Amplification-free library preparation for paired-end Illumina sequencing. Methods Mol Biol. 2011;733:257–66. Epub 2011/03/25. pmid:21431776.
  9. 9. Margraf RL, Durtschi JD, Dames S, Pattison DC, Stephens JE, Voelkerding KV. Variant identification in multi-sample pools by illumina genome analyzer sequencing. J Biomol Tech. 2011;22(2):74–84. Epub 2011/07/09. pmid:21738440
  10. 10. Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv:13033997v1. 2013.
  11. 11. Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics. 2013;43:11.0.1–33. Epub 2014/11/29. pmid:25431634
  12. 12. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38(16):e164. Epub 2010/07/06. pmid:20601685
  13. 13. Richards CS, Bale S, Bellissimo DB, Das S, Grody WW, Hegde MR, et al. ACMG recommendations for standards for interpretation and reporting of sequence variations: Revisions 2007. Genet Med. 2008;10(4):294–300. Epub 2008/04/17. pmid:18414213.
  14. 14. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, 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):405–24. Epub 2015/03/06. pmid:25741868
  15. 15. Wolf NI, Vanderver A, van Spaendonk RM, Schiffmann R, Brais B, Bugiani M, et al. Clinical spectrum of 4H leukodystrophy caused by POLR3A and POLR3B mutations. Neurology. 2014;83(21):1898–905. Epub 2014/10/24. pmid:25339210
  16. 16. Wright FA, Lu JP, Sliter DA, Dupre N, Rouleau GA, Wojcikiewicz RJ. A Point Mutation in the Ubiquitin Ligase RNF170 That Causes Autosomal Dominant Sensory Ataxia Destabilizes the Protein and Impairs Inositol 1,4,5-Trisphosphate Receptor-mediated Ca2+ Signaling. J Biol Chem. 2015;290(22):13948–57. Epub 2015/04/18. pmid:25882839
  17. 17. Bettencourt C, Salpietro V, Efthymiou S, Chelban V, Hughes D, Pittman AM, et al. Genotype-phenotype correlations and expansion of the molecular spectrum of AP4M1-related hereditary spastic paraplegia. Orphanet J Rare Dis. 2017;12(1):172. Epub 2017/11/04. pmid:29096665.
  18. 18. Nambot S, Thevenon J, Kuentz P, Duffourd Y, Tisserant E, Bruel AL, et al. Clinical whole-exome sequencing for the diagnosis of rare disorders with congenital anomalies and/or intellectual disability: substantial interest of prospective annual reanalysis. Genet Med. 2017. Epub 2017/11/03. pmid:29095811.
  19. 19. Motazacker MM, Rost BR, Hucho T, Garshasbi M, Kahrizi K, Ullmann R, et al. A defect in the ionotropic glutamate receptor 6 gene (GRIK2) is associated with autosomal recessive mental retardation. Am J Hum Genet. 2007;81(4):792–8. Epub 2007/09/12. pmid:17847003
  20. 20. Cordoba M, Rodriguez S, Gonzalez Moron D, Medina N, Kauffman MA. Expanding the spectrum of Grik2 mutations: intellectual disability, behavioural disorder, epilepsy and dystonia. Clin Genet. 2015;87(3):293–5. Epub 2014/07/22. pmid:25039795.
  21. 21. Syrbe S, Hedrich UBS, Riesch E, Djemie T, Muller S, Moller RS, et al. De novo loss- or gain-of-function mutations in KCNA2 cause epileptic encephalopathy. Nat Genet. 2015;47(4):393–9. Epub 2015/03/10. pmid:25751627
  22. 22. Wenger AM, Guturu H, Bernstein JA, Bejerano G. Systematic reanalysis of clinical exome data yields additional diagnoses: implications for providers. Genet Med. 2017;19(2):209–14. Epub 2016/07/22. pmid:27441994.
  23. 23. Shashi V, McConkie-Rosell A, Rosell B, Schoch K, Vellore K, McDonald M, et al. The utility of the traditional medical genetics diagnostic evaluation in the context of next-generation sequencing for undiagnosed genetic disorders. Genet Med. 2014;16(2):176–82. Epub 2013/08/10. pmid:23928913.
  24. 24. Tan TY, Dillon OJ, Stark Z, Schofield D, Alam K, Shrestha R, et al. Diagnostic Impact and Cost-effectiveness of Whole-Exome Sequencing for Ambulant Children With Suspected Monogenic Conditions. JAMA Pediatr. 2017;171(9):855–62. Epub 2017/08/02. pmid:28759686.
  25. 25. Posey JE, Rosenfeld JA, James RA, Bainbridge M, Niu Z, Wang X, et al. Molecular diagnostic experience of whole-exome sequencing in adult patients. Genet Med. 2016;18(7):678–85. Epub 2015/12/04. pmid:26633545
  26. 26. Fogel BL, Satya-Murti S, Cohen BH. Clinical exome sequencing in neurologic disease. Neurol Clin Pract. 2016;6(2):164–76. Epub 2016/04/23. pmid:27104068
  27. 27. Srivastava S, Cohen JS, Vernon H, Baranano K, McClellan R, Jamal L, et al. Clinical whole exome sequencing in child neurology practice. Ann Neurol. 2014;76(4):473–83. Epub 2014/08/19. pmid:25131622.
  28. 28. Fogel BL, Lee H, Deignan JL, Strom SP, Kantarci S, Wang X, et al. Exome sequencing in the clinical diagnosis of sporadic or familial cerebellar ataxia. JAMA Neurol. 2014;71(10):1237–46. Epub 2014/08/19. pmid:25133958
  29. 29. Xue Y, Ankala A, Wilcox WR, Hegde MR. Solving the molecular diagnostic testing conundrum for Mendelian disorders in the era of next-generation sequencing: single-gene, gene panel, or exome/genome sequencing. Genet Med. 2015;17(6):444–51. Epub 2014/09/19. pmid:25232854.
  30. 30. Lionel AC, Costain G, Monfared N, Walker S, Reuter MS, Hosseini SM, et al. Improved diagnostic yield compared with targeted gene sequencing panels suggests a role for whole-genome sequencing as a first-tier genetic test. Genet Med. 2017. Epub 2017/08/05. pmid:28771251.
  31. 31. Sun Y, Ruivenkamp CA, Hoffer MJ, Vrijenhoek T, Kriek M, van Asperen CJ, et al. Next-generation diagnostics: gene panel, exome, or whole genome? Hum Mutat. 2015;36(6):648–55. Epub 2015/03/17. pmid:25772376.