476
Views
8
CrossRef citations to date
0
Altmetric
Review

Utility of droplet digital PCR and NGS-based CNV clinical assays in hearing loss diagnostics: current status and future prospects

& ORCID Icon
Pages 213-221 | Received 16 Nov 2020, Accepted 05 Feb 2021, Published online: 26 Feb 2021

References

  • Korver AM, Smith RJ, Van Camp G, et al. Congenital hearing loss. Nat Rev Dis Primers. 2017;3:16094.
  • DiStefano MT, Hemphill SE, Oza AM, et al. ClinGen expert clinical validity curation of 164 hearing loss gene-disease pairs. Genet Med. 2019;21(10):2239–2247.
  • Abou Tayoun AN, Al Turki SH, Oza AM, et al. Improving hearing loss gene testing: a systematic review of gene evidence toward more efficient next-generation sequencing-based diagnostic testing and interpretation. Genet Med. 2016;18(6):545–553.
  • Azaiez H, Booth KT, Ephraim SS, et al. Genomic landscape and mutational signatures of deafness-associated genes. Am J Hum Genet. 2018;103(4):484–497.
  • Shearer AE, Kolbe DL, Azaiez H, et al. Copy number variants are a common cause of non-syndromic hearing loss. Genome Med. 2014;6(5):37.
  • Sloan-Heggen CM, Bierer AO, Shearer AE, et al. Comprehensive genetic testing in the clinical evaluation of 1119 patients with hearing loss. Hum Genet. 2016;135(4):441–450.
  • Wilch E, Azaiez H, Fisher RA, et al. A novel DFNB1 deletion allele supports the existence of a distant cis-regulatory region that controls GJB2 and GJB6 expression. Clin Genet. 2010;78(3):267–274.
  • Snoeckx RL, Huygen PL, Feldmann D, et al. GJB2 mutations and degree of hearing loss: a multicenter study. Am J Hum Genet. 2005;77(6):945–957.
  • Shen J, Oza AM, Del Castillo I, et al. Consensus interpretation of the p.Met34Thr and p.Val37Ile variants in GJB2 by the ClinGen hearing loss expert panel. Genet Med. 2019;21(11):2442–2452.
  • Weil D, Blanchard S, Kaplan J, et al. Defective myosin VIIA gene responsible for Usher syndrome type 1B. Nature. 1995;374(6517):60–61.
  • Hildebrand MS, Thorne NP, Bromhead CJ, et al. Variable hearing impairment in a DFNB2 family with a novel MYO7A missense mutation. Clin Genet. 2010;77(6):563–571.
  • Bolz H, Bolz SS, Schade G, et al. Impaired calmodulin binding of myosin-7A causes autosomal dominant hearing loss (DFNA11. Hum Mutat. 2004;24(3):274–275.
  • Schraders M, Oostrik J, Huygen PL, et al. Mutations in PTPRQ are a cause of autosomal-recessive nonsyndromic hearing impairment DFNB84 and associated with vestibular dysfunction. Am J Hum Genet. 2010;86(4):604–610.
  • Eisenberger T, Di Donato N, Decker C, et al. A C-terminal nonsense mutation links PTPRQ with autosomal-dominant hearing loss, DFNA73. Genet Med. 2018;20(6):614–621.
  • Azaiez H, Booth KT, Bu F, et al. TBC1D24 mutation causes autosomal-dominant nonsyndromic hearing loss. Hum Mutat. 2014;35(7):819–823.
  • Rehman AU, Santos-Cortez RL, Morell RJ, et al. Mutations in TBC1D24, a gene associated with epilepsy, also cause nonsyndromic deafness DFNB86. Am J Hum Genet. 2014;94(1):144–152.
  • Oza AM, DiStefano MT, Hemphill SE, et al. Expert specification of the ACMG/AMP variant interpretation guidelines for genetic hearing loss. Hum Mutat. 2018;39(11):1593–1613.
  • Del Castillo FJ, Rodriguez-Ballesteros M, Alvarez A, et al. A novel deletion involving the connexin-30 gene, del(GJB6-d13s1854), found in trans with mutations in the GJB2 gene (connexin-26) in subjects with DFNB1 non-syndromic hearing impairment. J Med Genet. 2005;42(7):588–594.
  • Mandelker D, Amr SS, Pugh T, et al. Comprehensive diagnostic testing for stereocilin: an approach for analyzing medically important genes with high homology. J Mol Diagn. 2014;16(6):639–647.
  • Sugiyama K, Moteki H, Kitajiri SI, et al. Mid-frequency hearing loss is characteristic clinical feature of OTOA-associated hearing loss. Genes (Basel). 2019;10:9.
  • Yokota Y, Moteki H, Nishio SY, et al. Frequency and clinical features of hearing loss caused by STRC deletions. Sci Rep. 2019;9(1):4408.
  • 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.
  • Bademci G, Diaz-Horta O, Guo S, et al. Identification of copy number variants through whole-exome sequencing in autosomal recessive nonsyndromic hearing loss. Genet Test Mol Biomarkers. 2014;18(9):658–661.
  • Ellingford JM, Campbell C, Barton S, et al. Validation of copy number variation analysis for next-generation sequencing diagnostics. Eur J Hum Genet. 2017;25(6):719–724.
  • Haraksingh RR, Jahanbani F, Rodriguez-Paris J, et al. Exome sequencing and genome-wide copy number variant mapping reveal novel associations with sensorineural hereditary hearing loss. BMC Genomics. 2014;15:1155.
  • Sommen M, Schrauwen I, Vandeweyer G, et al. DNA diagnostics of hereditary hearing loss: a targeted resequencing approach combined with a mutation classification system. Hum Mutat. 2016;37(8):812–819.
  • Sheppard S, Biswas S, Li MH, et al. Utility and limitations of exome sequencing as a genetic diagnostic tool for children with hearing loss. Genet Med. 2018;20(12):1663–1676.
  • Hindson BJ, Ness KD, Masquelier DA, et al. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem. 2011;83(22):8604–8610.
  • Kim BJ, Oh DY, Han JH, et al. Significant Mendelian genetic contribution to pediatric mild-to-moderate hearing loss and its comprehensive diagnostic approach. Genet Med. 2020;22(6):1119–1128.
  • Del Castillo I, Villamar M, Moreno-Pelayo MA, et al. A deletion involving the connexin 30 gene in nonsyndromic hearing impairment. N Engl J Med. 2002;346(4):243–249.
  • Tayoun AN, Mason-Suares H, Frisella AL, et al. Targeted droplet-digital PCR as a tool for novel deletion discovery at the DFNB1 locus. Hum Mutat. 2016;37(1):119–126.
  • Moisan S, Le Nabec A, Quillevere A, et al. Characterization of GJB2 cis-regulatory elements in the DFNB1 locus. Hum Genet. 2019;138(11–12):1275–1286.
  • Francey LJ, Conlin LK, Kadesch HE, et al. Genome-wide SNP genotyping identifies the Stereocilin (STRC) gene as a major contributor to pediatric bilateral sensorineural hearing impairment. Am J Med Genet A. 2012;158A(2):298–308.
  • Vona B, Hofrichter MA, Neuner C, et al. DFNB16 is a frequent cause of congenital hearing impairment: implementation of STRC mutation analysis in routine diagnostics. Clin Genet. 2015;87(1):49–55.
  • Amr SS, Murphy E, Duffy E, et al. Allele-Specific droplet digital PCR combined with a next-generation sequencing-based algorithm for diagnostic copy number analysis in genes with high homology: proof of concept using stereocilin. Clin Chem. 2018;64(4):705–714.
  • Moteki H, Azaiez H, Sloan-Heggen CM, et al. Detection and confirmation of deafness-causing copy number variations in the STRC gene by massively parallel sequencing and comparative genomic hybridization. Ann Otol Rhinol Laryngol. 2016;125(11):918–923.
  • Ito T, Kawashima Y, Fujikawa T, et al. Rapid screening of copy number variations in STRC by droplet digital PCR in patients with mild-to-moderate hearing loss. Hum Genome Var. 2019;6:41.
  • Hoppman N, Aypar U, Brodersen P, et al. Genetic testing for hearing loss in the United States should include deletion/duplication analysis for the deafness/infertility locus at 15q15.3. Mol Cytogenet. 2013;6(1):19.
  • Knijnenburg J, Oberstein SA, Frei K, et al. A homozygous deletion of a normal variation locus in a patient with hearing loss from non-consanguineous parents. J Med Genet. 2009;46(6):412–417.
  • Pugh TJ, Amr SS, Bowser MJ, et al. VisCap: inference and visualization of germ-line copy-number variants from targeted clinical sequencing data. Genet Med. 2016;18(7):712–719.
  • Adey A, Morrison HG, Asan, et al. Rapid, low-input, low-bias construction of shotgun fragment libraries by high-density in vitro transposition. Genome Biol. 2010;11(12):R119.
  • Miller NA, Farrow EG, Gibson M, et al. A 26-hour system of highly sensitive whole genome sequencing for emergency management of genetic diseases. Genome Med. 2015;7:100.
  • Chen X, Schulz-Trieglaff O, Shaw R, et al. Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics. 2016;32(8):1220–1222.
  • 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.
  • Gross AM, Ajay SS, Rajan V, et al. Copy-number variants in clinical genome sequencing: deployment and interpretation for rare and undiagnosed disease. Genet Med. 2019;21(5):1121–1130.
  • Haraksingh RR, Abyzov A, Urban AE. Comprehensive performance comparison of high-resolution array platforms for genome-wide Copy Number Variation (CNV) analysis in humans. BMC Genomics. 2017;18(1):321.
  • Lindstrand A, Eisfeldt J, Pettersson M, et al. From cytogenetics to cytogenomics: whole-genome sequencing as a first-line test comprehensively captures the diverse spectrum of disease-causing genetic variation underlying intellectual disability. Genome Med. 2019;11(1):68.
  • Trost B, Walker S, Wang Z, et al. A comprehensive workflow for read depth-based identification of copy-number variation from whole-genome sequence data. Am J Hum Genet. 2018;102(1):142–155.
  • Zhou B, Ho SS, Zhang X, et al. Whole-genome sequencing analysis of CNV using low-coverage and paired-end strategies is efficient and outperforms array-based CNV analysis. J Med Genet. 2018;55(11):735–743.
  • Sineni CJ, Yildirim-Baylan M, Guo S, et al. A truncating CLDN9 variant is associated with autosomal recessive nonsyndromic hearing loss. Hum Genet. 2019;138(10):1071–1075.
  • Bademci G, Abad C, Incesulu A, et al. MPZL2 is a novel gene associated with autosomal recessive nonsyndromic moderate hearing loss. Hum Genet. 2018;137(6–7):479–486.
  • Mantere T, Kersten S, Hoischen A. Long-read sequencing emerging in medical genetics. Front Genet. 2019;10:426.
  • Van Dijk EL, Jaszczyszyn Y, Naquin D, et al. The third revolution in sequencing technology. Trends Genet. 2018;34(9):666–681.
  • Borras DM, Vossen R, Liem M, et al. Detecting PKD1 variants in polycystic kidney disease patients by single-molecule long-read sequencing. Hum Mutat. 2017;38(7):870–879.
  • Frans G, Meert W, Van Der Werff Ten Bosch J, et al. Conventional and single-molecule targeted sequencing method for specific variant detection in IKBKG while bypassing the IKBKGP1 pseudogene. J Mol Diagn. 2018;20(2):195–202.
  • 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.
  • Vollger MR, Logsdon GA, Audano PA, et al. Improved assembly and variant detection of a haploid human genome using single-molecule, high-fidelity long reads. Ann Hum Genet. 2020;84(2):125–140.
  • Mitsuhashi S, Matsumoto N. Long-read sequencing for rare human genetic diseases. J Hum Genet. 2020;65(1):11–19.
  • Koboldt DC. Best practices for variant calling in clinical sequencing. Genome Med. 2020;12(1):91.
  • Mu W, Lu HM, Chen J, et al. Sanger confirmation is required to achieve optimal sensitivity and specificity in next-generation sequencing panel testing. J Mol Diagn. 2016;18(6):923–932.
  • Baudhuin LM, Lagerstedt SA, Klee EW, et al. Confirming variants in next-generation sequencing panel testing by sanger sequencing. J Mol Diagn. 2015;17(4):456–461.
  • Strom SP, Lee H, Das K, et al. Assessing the necessity of confirmatory testing for exome-sequencing results in a clinical molecular diagnostic laboratory. Genet Med. 2014;16(7):510–515.
  • Lincoln SE, Truty R, Lin CF, et al. A rigorous interlaboratory examination of the need to confirm next-generation sequencing-detected variants with an orthogonal method in clinical genetic testing. J Mol Diagn. 2019;21(2):318–329.
  • Muzzey D, Kash S, Johnson JI, et al. Software-assisted manual review of clinical next-generation sequencing data: an alternative to routine sanger sequencing confirmation with equivalent results in >15,000 germline DNA screens. J Mol Diagn. 2019;21(2):296–306.
  • Pfundt R, Del Rosario M, Vissers L, 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.
  • Marshall CR, Chowdhury S, Taft RJ, 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.
  • Plagnol V, Curtis J, Epstein M, et al. A robust model for read count data in exome sequencing experiments and implications for copy number variant calling. Bioinformatics. 2012;28(21):2747–2754.
  • Yao R, Zhang C, Yu T, et al. Evaluation of three read-depth based CNV detection tools using whole-exome sequencing data. Mol Cytogenet. 2017;10:30.
  • Sanghvi RV, Buhay CJ, Powell BC, et al. Characterizing reduced coverage regions through comparison of exome and genome sequencing data across 10 centers. Genet Med. 2018;20(8):855–866.
  • Mandelker D, Schmidt RJ, 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.
  • Marchuk DS, Crooks K, Strande N, et al. Increasing the diagnostic yield of exome sequencing by copy number variant analysis. PLoS One. 2018;13(12):e0209185.
  • Hong CS, Singh LN, Mullikin JC, et al. Assessing the reproducibility of exome copy number variations predictions. Genome Med. 2016;8(1):82.
  • Kim HY, Choi JW, Lee JY, et al. Gene-based comparative analysis of tools for estimating copy number alterations using whole-exome sequencing data. Oncotarget. 2017;8(16):27277–27285.
  • Austin-Tse CA, Mandelker DL, Oza AM, et al. Analysis of intragenic USH2A copy number variation unveils broad spectrum of unique and recurrent variants. Eur J Med Genet. 2018;61(10):621–626.
  • Le Guedard S, Faugere V, Malcolm S, et al. Large genomic rearrangements within the PCDH15 gene are a significant cause of USH1F syndrome. Mol Vis. 2007;13:102–107.
  • Rosenberg C, Freitas EL, Uehara DT, et al. Genomic copy number alterations in non-syndromic hearing loss. Clin Genet. 2016;89(4):473–477.
  • Riggs ER, Andersen EF, Cherry AM, 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.
  • Abou Tayoun AN, Pesaran T, DiStefano MT, et al. Recommendations for interpreting the loss of function PVS1 ACMG/AMP variant criterion. Hum Mutat. 2018;39(11):1517–1524.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.