1932

Abstract

Exome sequencing (ES) and genome sequencing (GS) have radically transformed the diagnostic approach to undiagnosed rare/ultrarare Mendelian diseases. Next-generation sequencing (NGS), the technology integral for ES, GS, and most large (100+) gene panels, has enabled previously unimaginable diagnoses, changes in medical management, new treatments, and accurate reproductive risk assessments for patients, as well as new disease gene discoveries. Yet, challenges remain, as most individuals remain undiagnosed with current NGS. Improved NGS technology has resulted in long-read sequencing, which may resolve diagnoses in some patients who do not obtain a diagnosis with current short-read ES and GS, but its effectiveness is unclear, and it is expensive. Other challenges that persist include the resolution of variants of uncertain significance, the urgent need for patients with ultrarare disorders to have access to therapeutics, the need for equity in patient access to NGS-based testing, and the study of ethical concerns. However, the outlook for undiagnosed disease resolution is bright, due to continual advancements in the field.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-042921-110721
2023-01-27
2024-04-27
Loading full text...

Full text loading...

/deliver/fulltext/med/74/1/annurev-med-042921-110721.html?itemId=/content/journals/10.1146/annurev-med-042921-110721&mimeType=html&fmt=ahah

Literature Cited

  1. 1.
    Chong JX, Buckingham KJ, Jhangiani SN et al. 2015. The genetic basis of Mendelian phenotypes: discoveries, challenges, and opportunities. Am. J. Hum. Genet. 97:199–215
    [Google Scholar]
  2. 2.
    Eurordis 2005. Rare Diseases: Understanding This Public Health Priority Paris, France: Eur. Organ. Rare Dis. https://www.eurordis.org/wp-content/uploads/2009/12/princeps_document-EN.pdf
  3. 3.
    Kumar P, Radhakrishnan J, Chowdhary MA, Giampietro PF. 2001. Prevalence and patterns of presentation of genetic disorders in a pediatric emergency department. Mayo Clin. Proc. 76:777–83
    [Google Scholar]
  4. 4.
    Shashi V, McConkie-Rosell A, Rosell B et al. 2014. The utility of the traditional medical genetics diagnostic evaluation in the context of next-generation sequencing for undiagnosed genetic disorders. Genet. Med. 16:176–82
    [Google Scholar]
  5. 5.
    Christianson A, Howson CP, Modell B. 2006. March of Dimes global report on birth defects: the hidden toll of dying and disabled children Rep. March of Dimes Birth Defects Found. White Plains, NY:
  6. 6.
    Costa T, Scriver CR, Childs B. 1985. The effect of Mendelian disease on human health: a measurement. Am. J. Med. Genet. 21:231–42
    [Google Scholar]
  7. 7.
    Gahl WA, Markello TC, Toro C et al. 2012. The National Institutes of Health Undiagnosed Diseases Program: insights into rare diseases. Genet. Med. 14:51–59
    [Google Scholar]
  8. 8.
    Graungaard AH, Skov L. 2006. Why do we need a diagnosis? A qualitative study of parents’ experiences, coping and needs, when the newborn child is severely disabled. Child 33:296–307
    [Google Scholar]
  9. 9.
    Rosenthal ET, Biesecker LG, Biesecker BB. 2001. Parental attitudes toward a diagnosis in children with unidentified multiple congenital anomaly syndromes. Am. J. Med. Genet. 103:106–14
    [Google Scholar]
  10. 10.
    Global Genes 2021. Rare disease facts Global Genes Aliso Viejo, CA: https://globalgenes.org/rare-disease-facts/
  11. 11.
    Bamshad MJ, Ng SB, Bigham AW et al. 2011. Exome sequencing as a tool for Mendelian disease gene discovery. Nat. Rev. Genet. 12:745–55
    [Google Scholar]
  12. 12.
    Bamshad MJ, Nickerson DA, Chong JX. 2019. Mendelian gene discovery: fast and furious with no end in sight. Am. J. Hum. Genet. 105:448–55
    [Google Scholar]
  13. 13.
    Need AC, Shashi V, Hitomi Y et al. 2012. Clinical application of exome sequencing in undiagnosed genetic conditions. J. Med. Genet. 49:353–61
    [Google Scholar]
  14. 14.
    Lee H, Deignan JL, Dorrani N et al. 2014. Clinical exome sequencing for genetic identification of rare Mendelian disorders. JAMA 312:1880–87
    [Google Scholar]
  15. 15.
    Yang Y, Muzny DM, Reid JG et al. 2013. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. New Engl. . J. Med. 369:1502–11
    [Google Scholar]
  16. 16.
    Zhu X, Petrovski S, Xie P et al. 2015. Whole-exome sequencing in undiagnosed genetic diseases: interpreting 119 trios. Genet. Med. 17:774–81
    [Google Scholar]
  17. 17.
    Yang Y, Muzny DM, Xia F et al. 2014. Molecular findings among patients referred for clinical whole-exome sequencing. JAMA 312:1870–79
    [Google Scholar]
  18. 18.
    Stark Z, Schofield D, Martyn M et al. 2019. Does genomic sequencing early in the diagnostic trajectory make a difference? A follow-up study of clinical outcomes and cost-effectiveness. Genet. Med. 21:173–80
    [Google Scholar]
  19. 19.
    Shickh S, Mighton C, Uleryk E et al. 2021. The clinical utility of exome and genome sequencing across clinical indications: a systematic review. Hum. Genet. 140:1403–16
    [Google Scholar]
  20. 20.
    Petrovski S, Shashi V, Petrou S et al. 2015. Exome sequencing results in successful riboflavin treatment of a rapidly progressive neurological condition. Cold Spring Harb. Mol. Case Stud. 1:a000257
    [Google Scholar]
  21. 21.
    Mikati MA, Jiang Y, Carboni M et al. 2015. Quinidine in the treatment of KCNT1-positive epilepsies. Ann. Neurol. 78:995–99
    [Google Scholar]
  22. 22.
    Willig LK, Petrikin JE, Smith LD et al. 2015. Whole-genome sequencing for identification of Mendelian disorders in critically ill infants: a retrospective analysis of diagnostic and clinical findings. Lancet Respir. Med. 3:377–87
    [Google Scholar]
  23. 23.
    Saunders CJ, Miller NA, Soden SE et al. 2012. Rapid whole-genome sequencing for genetic disease diagnosis in neonatal intensive care units. Sci. Transl. Med. 4:154ra35
    [Google Scholar]
  24. 24.
    Kingsmore SF, Dinwiddie DL, Miller NA et al. 2011. Adopting orphans: comprehensive genetic testing of Mendelian diseases of childhood by next-generation sequencing. Expert Rev. Mol. Diagn. 11:855–68
    [Google Scholar]
  25. 25.
    Farnaes L, Hildreth A, Sweeney NM et al. 2018. Rapid whole-genome sequencing decreases infant morbidity and cost of hospitalization. NPJ Genom. Med. 3:10
    [Google Scholar]
  26. 26.
    Soden SE, Saunders CJ, Willig LK et al. 2014. Effectiveness of exome and genome sequencing guided by acuity of illness for diagnosis of neurodevelopmental disorders. Sci. Transl. Med. 6:265ra168
    [Google Scholar]
  27. 27.
    Schoch K, Esteves C, Bican A, Spillmann R, Cope H et al. 2020. Clinical sites of the Undiagnosed Diseases Network: unique contributions to genomic medicine and science. Genet. Med. 23:259–71
    [Google Scholar]
  28. 28.
    Shashi V, Schoch K, Spillmann R et al. 2019. A comprehensive iterative approach is highly effective in diagnosing individuals who are exome negative. Genet. Med. 21:161–72
    [Google Scholar]
  29. 29.
    Gilissen C, Hehir-Kwa JY, Thung DT et al. 2014. Genome sequencing identifies major causes of severe intellectual disability. Nature 511:344–47
    [Google Scholar]
  30. 30.
    Lander ES, Linton LM, Birren B et al. 2001. Initial sequencing and analysis of the human genome. Nature 409:860–921
    [Google Scholar]
  31. 31.
    Venter JC, Adams MD, Myers EW et al. 2001. The sequence of the human genome. Science 291:1304–51
    [Google Scholar]
  32. 32.
    Pareek CS, Smoczynski R, Tretyn A. 2011. Sequencing technologies and genome sequencing. J. Appl. Genet. 52:413–35
    [Google Scholar]
  33. 33.
    Ng SB, Buckingham KJ, Lee C et al. 2010. Exome sequencing identifies the cause of a mendelian disorder. Nat. Genet. 42:30–35
    [Google Scholar]
  34. 34.
    Vissers LE, de Ligt J, Gilissen C et al. 2010. A de novo paradigm for mental retardation. Nat. Genet. 42:1109–12
    [Google Scholar]
  35. 35.
    Lupski JR. 2007. Genomic rearrangements and sporadic disease. Nat. Genet. 39:S43–47
    [Google Scholar]
  36. 36.
    Sawyer SL, Hartley T, Dyment DA et al. 2016. Utility of whole-exome sequencing for those near the end of the diagnostic odyssey: time to address gaps in care. Clin. Genet. 89:275–84
    [Google Scholar]
  37. 37.
    Adams DR, Eng CM. 2018. Next-generation sequencing to diagnose suspected genetic disorders. N. Engl. J. Med. 379:1353–62
    [Google Scholar]
  38. 38.
    Richards S, Aziz N, Bale S et al. 2015. 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. 17:405–24
    [Google Scholar]
  39. 39.
    Shashi V, McConkie-Rosell A, Schoch K et al. 2016. Practical considerations in the clinical application of whole-exome sequencing. Clin. Genet. 89:173–81
    [Google Scholar]
  40. 40.
    Baldridge D, Heeley J, Vineyard M et al. 2017. The Exome Clinic and the role of medical genetics expertise in the interpretation of exome sequencing results. Genet. Med. 9:1040–48
    [Google Scholar]
  41. 41.
    Retterer K, Juusola J, Cho MT et al. 2016. Clinical application of whole-exome sequencing across clinical indications. Genet. Med. 18:696–704
    [Google Scholar]
  42. 42.
    Stavropoulos DJ, Merico D, Jobling R et al. 2016. Whole genome sequencing expands diagnostic utility and improves clinical management in pediatric medicine. NPJ Genom. Med. 1:15012
    [Google Scholar]
  43. 43.
    Mattick JS, Dinger M, Schonrock N, Cowley M. 2018. Whole genome sequencing provides better diagnostic yield and future value than whole exome sequencing. Med. J. Aust. 209:197–99
    [Google Scholar]
  44. 44.
    Rauch A, Wieczorek D, Graf E et al. 2012. Range of genetic mutations associated with severe non-syndromic sporadic intellectual disability: an exome sequencing study. Lancet 380:1674–82
    [Google Scholar]
  45. 45.
    Manickam K, McClain MR, Demmer LA et al. 2021. 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. 23:2029–37
    [Google Scholar]
  46. 46.
    Murdock DR, Rosenfeld JA, Lee B. 2022. What has the Undiagnosed Diseases Network taught us about the clinical applications of genomic testing?. Annu. Rev. Med. 73:575–85
    [Google Scholar]
  47. 47.
    Belkadi A, Bolze A, Itan Y et al. 2015. Whole-genome sequencing is more powerful than whole-exome sequencing for detecting exome variants. PNAS 112:5473–78
    [Google Scholar]
  48. 48.
    Deignan JL, Chung WK, Kearney HM et al. 2019. Points to consider in the reevaluation and reanalysis of genomic test results: a statement of the American College of Medical Genetics and Genomics (ACMG). Genet. Med. 21:1267–70
    [Google Scholar]
  49. 49.
    Liu P, Meng L, Normand EA et al. 2019. Reanalysis of clinical exome sequencing data. N. Engl. J. Med. 380:2478–80
    [Google Scholar]
  50. 50.
    Wenger AM, Guturu H, Bernstein JA, Bejerano G. 2017. Systematic reanalysis of clinical exome data yields additional diagnoses: implications for providers. Genet. Med. 19:209–14
    [Google Scholar]
  51. 51.
    Need AC, Shashi V, Schoch K et al. 2017. The importance of dynamic re-analysis in diagnostic whole exome sequencing. J. Med. Genet. 54:155–56
    [Google Scholar]
  52. 52.
    Green RC, Berg JS, Grody WW et al. 2013. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet. Med. 15:565–74
    [Google Scholar]
  53. 53.
    Miller DT, Lee K, Abul-Husn NS et al. 2022. ACMG SF v3.1 list for reporting of secondary findings in clinical exome and genome sequencing: a policy statement of the American College of Medical Genetics and Genomics (ACMG). Genet. Med. 24:1407–14
    [Google Scholar]
  54. 54.
    Delanne J, Nambot S, Chassagne A et al. 2019. Secondary findings from whole-exome/genome sequencing evaluating stakeholder perspectives. A review of the literature. Eur. J. Med. Genet. 62:103529
    [Google Scholar]
  55. 55.
    ACMG Board of Directors 2013. Points to consider for informed consent for genome/exome sequencing. Genet. Med. 15:748–49
    [Google Scholar]
  56. 56.
    Eno C, Bayrak-Toydemir P, Bean L et al. 2019. Misattributed parentage as an unanticipated finding during exome/genome sequencing: current clinical laboratory practices and an opportunity for standardization. Genet. Med. 21:861–66
    [Google Scholar]
  57. 57.
    Cohen ASA, Farrow EG, Abdelmoity AT et al. 2022. Genomic answers for children: dynamic analyses of >1000 pediatric rare disease genomes. Genet. Med. 24:1336–48
    [Google Scholar]
  58. 58.
    Bertier G, Hétu M, Joly Y. 2016. Unsolved challenges of clinical whole-exome sequencing: a systematic literature review of end-users’ views. BMC Med. Genom. 9:52
    [Google Scholar]
  59. 59.
    Kim Y-E, Ki C-S, Jang M-A. 2019. Challenges and considerations in sequence variant interpretation for Mendelian disorders. Ann. Lab. Med. 39:421–29
    [Google Scholar]
  60. 60.
    Schoch K, Tan QK, Stong N et al. 2020. Alternative transcripts in variant interpretation: the potential for missed diagnoses and misdiagnoses. Genet. Med. 22:1269–75
    [Google Scholar]
  61. 61.
    Landrum MJ, Lee JM, Benson M et al. 2018. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 46:D1062–67
    [Google Scholar]
  62. 62.
    Rehm HL, Berg JS, Brooks LD et al. 2015. ClinGen—the Clinical Genome Resource. N. Engl. J. Med. 372:2235–42
    [Google Scholar]
  63. 63.
    Sobreira N, Schiettecatte F, Valle D, Hamosh A. 2015. GeneMatcher: a matching tool for connecting investigators with an interest in the same gene. Hum. Mutat. 36:928–30
    [Google Scholar]
  64. 64.
    Azzariti DR, Hamosh A. 2020. Genomic data sharing for novel Mendelian disease gene discovery: the Matchmaker Exchange. Annu. Rev. Genom. Hum. Genet. 21:305–26
    [Google Scholar]
  65. 65.
    Posey JE, O'Donnell-Luria AH, Chong JX et al. 2019. Insights into genetics, human biology and disease gleaned from family based genomic studies. Genet. Med. 21:798–812
    [Google Scholar]
  66. 66.
    Shashi V, Geist J, Lee Y et al. 2019. Heterozygous variants in MYBPC1 are associated with an expanded neuromuscular phenotype beyond arthrogryposis. Hum. Mutat. 40:1115–26
    [Google Scholar]
  67. 67.
    Sullivan JA, Stong N, Baugh EH et al. 2020. A pathogenic variant in the SETBP1 hotspot results in a forme-fruste Schinzel-Giedion syndrome. Am. J. Med. Genet. A 182:1947–51
    [Google Scholar]
  68. 68.
    Baldridge D, Spillmann RC, Wegner DJ et al. 2020. Phenotypic expansion of KMT2D-related disorder: beyond Kabuki syndrome. Am. J. Med. Genet. A 182:1053–65
    [Google Scholar]
  69. 69.
    Marcogliese PC, Shashi V, Spillmann RC et al. 2018. IRF2BPL is associated with neurological phenotypes. Am. J. Hum. Genet. 103:245–60
    [Google Scholar]
  70. 70.
    Blanchard MG, Willemsen MH, Walker JB et al. 2015. De novo gain-of-function and loss-of-function mutations of SCN8A in patients with intellectual disabilities and epilepsy. J. Med. Genet. 52:330–37
    [Google Scholar]
  71. 71.
    Iglesias A, Anyane-Yeboa K, Wynn J et al. 2014. The usefulness of whole-exome sequencing in routine clinical practice. Genet. Med. 16:922–31
    [Google Scholar]
  72. 72.
    Splinter K, Adams DR, Bacino CA et al. 2018. Effect of genetic diagnosis on patients with previously undiagnosed disease. N. Engl. J. Med. 379:2131–39
    [Google Scholar]
  73. 73.
    Petrovski S, Shashi V, Petrou S et al. 2015. Sustained therapeutic response to riboflavin in a child with a progressive neurological condition, diagnosed by whole exome sequencing. Cold Spring Harb. Mol. Case Stud. 1:a000265
    [Google Scholar]
  74. 74.
    Bi Y, Might M, Vankayalapati H, Kuberan B. 2017. Repurposing of proton pump inhibitors as first identified small molecule inhibitors of endo-β-N-acetylglucosaminidase (ENGase) for the treatment of NGLY1, a rare genetic disease. Bioorg. Med. Chem. Lett. 27:2962–66
    [Google Scholar]
  75. 75.
    Hope KA, Berman AR, Peterson RT, Chow CY. 2022. An in vivo drug repurposing screen and transcriptional analyses reveals the serotonin pathway and GSK3 as major therapeutic targets for NGLY1 deficiency. PLOS Genet. 18:e1010228
    [Google Scholar]
  76. 76.
    Kim J, Hu C, Moufawad El Achkar C et al. 2019. Patient-customized oligonucleotide therapy for a rare genetic disease. N. Engl. J. Med. 381:1644–52
    [Google Scholar]
  77. 77.
    Vissers L, van Nimwegen KJM, Schieving JH et al. 2017. A clinical utility study of exome sequencing versus conventional genetic testing in pediatric neurology. Genet. Med. 19:1055–63
    [Google Scholar]
  78. 78.
    Tan TY, Dillon OJ, Stark Z et al. 2017. Diagnostic impact and cost-effectiveness of whole-exome sequencing for ambulant children with suspected monogenic conditions. JAMA Pediatr. 171:855–62
    [Google Scholar]
  79. 79.
    Hayeems RZ, Bhawra J, Tsiplova K et al. 2017. Care and cost consequences of pediatric whole genome sequencing compared to chromosome microarray. Eur. J. Hum. Genet. 25:1303–12
    [Google Scholar]
  80. 80.
    Schwarze K, Buchanan J, Taylor JC, Wordsworth S. 2018. Are whole-exome and whole-genome sequencing approaches cost-effective? A systematic review of the literature. Genet. Med. 20:1122–30
    [Google Scholar]
  81. 81.
    Incerti D, Xu XM, Chou JW et al. 2022. Cost-effectiveness of genome sequencing for diagnosing patients with undiagnosed rare genetic diseases. Genet. Med. 24:109–18
    [Google Scholar]
  82. 82.
    Nurchis MC, Riccardi MT, Radio FC et al. 2022. Incremental net benefit of whole genome sequencing for newborns and children with suspected genetic disorders: systematic review and meta-analysis of cost-effectiveness evidence. Health Policy 126:337–45
    [Google Scholar]
  83. 83.
    McConkie-Rosell A, Hooper SR, Pena LDM et al. 2018. Psychosocial profiles of parents of children with undiagnosed diseases: managing well or just managing?. J. Genet. Counsel. 27:935–46
    [Google Scholar]
  84. 84.
    Spillmann RC, McConkie-Rosell A, Pena L et al. 2017. A window into living with an undiagnosed disease: illness narratives from the Undiagnosed Diseases Network. Orphanet J. Rare Dis. 12:71
    [Google Scholar]
  85. 85.
    McConkie-Rosell A, Pena LD, Schoch K et al. 2016. Not the end of the odyssey: parental perceptions of whole exome sequencing (WES) in pediatric undiagnosed disorders. J. Genet. Counsel. 25:1019–31
    [Google Scholar]
  86. 86.
    Halley MC, Young JL, Fernandez L et al. 2022. Perceived utility and disutility of genomic sequencing for pediatric patients: perspectives from parents with diverse sociodemographic characteristics. Am. J. Med. Genet. A 188:1088–101
    [Google Scholar]
  87. 87.
    van Dijk EL, Jaszczyszyn Y, Naquin D, Thermes C. 2018. The third revolution in sequencing technology. Trends Genet. 34:666–81
    [Google Scholar]
  88. 88.
    Brown CG, Clarke J 2016. Nanopore development at Oxford Nanopore. Nat. Biotechnol. 34:810–11
    [Google Scholar]
  89. 89.
    Quail MA, Smith M, Coupland P et al. 2012. A tale of three next generation sequencing platforms: comparison of Ion Torrent, Pacific Biosciences and Illumina MiSeq sequencers. BMC Genom. 13:341
    [Google Scholar]
  90. 90.
    Nurk S, Koren S, Rhie A et al. 2022. The complete sequence of a human genome. Science 376:44–53
    [Google Scholar]
  91. 91.
    Merker JD, Wenger AM, Sneddon T et al. 2018. Long-read genome sequencing identifies causal structural variation in a Mendelian disease. Genet. Med. 20:159–63
    [Google Scholar]
  92. 92.
    Seaby EG, Rehm HL, O'Donnell-Luria A 2021. Strategies to uplift novel Mendelian gene discovery for improved clinical outcomes. Front. Genet. 12:674295
    [Google Scholar]
  93. 93.
    Might M, Wilsey M. 2014. The shifting model in clinical diagnostics: how next-generation sequencing and families are altering the way rare diseases are discovered, studied, and treated. Genet. Med. 16:736–37
    [Google Scholar]
  94. 94.
    Titgemeyer SC, Schaaf CP. 2022. Facebook support groups for pediatric rare diseases: cross-sectional study to investigate opportunities, limitations, and privacy concerns. JMIR Pediatr. Parent 5:e31411
    [Google Scholar]
  95. 95.
    Berg JS, Powell CM. 2015. Potential uses and inherent challenges of using genome-scale sequencing to augment current newborn screening. Cold Spring Harbor. . Perspect. Med. 5:a023150
    [Google Scholar]
  96. 96.
    Smogavec M, Gerykova Bujalkova M, Lehner R et al. 2022. Singleton exome sequencing of 90 fetuses with ultrasound anomalies revealing novel disease-causing variants and genotype-phenotype correlations. Eur. J. Hum. Genet. 30:428–38
    [Google Scholar]
  97. 97.
    Stark Z, Dolman L, Manolio TA et al. 2019. Integrating genomics into healthcare: a global responsibility. Am. J. Hum. Genet. 104:13–20
    [Google Scholar]
/content/journals/10.1146/annurev-med-042921-110721
Loading
/content/journals/10.1146/annurev-med-042921-110721
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error