1932

Abstract

The tools of next-generation sequencing (NGS) technology, such as targeted sequencing of candidate cancer genes and whole-exome and -genome sequencing, coupled with encouraging clinical results based on the use of targeted therapeutics and biomarker-guided clinical trials, are fueling further technological advancements of NGS technology. However, NGS data interpretation is associated with challenges that must be overcome to promote the techniques' effective integration into clinical oncology practice. Specifically, sequencing of a patient's tumor often yields 30–65 somatic variants, but most of these variants are “passenger” mutations that are phenotypically neutral and thus not targetable. Therefore, NGS data must be interpreted by multidisciplinary decision-support teams to determine mutation actionability and identify potential “drivers,” so that the treating physician can prioritize what clinical decisions can be pursued in order to provide cancer therapy that is personalized to the patient and his or her unique genome.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-102115-021556
2017-01-14
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/med/68/1/annurev-med-102115-021556.html?itemId=/content/journals/10.1146/annurev-med-102115-021556&mimeType=html&fmt=ahah

Literature Cited

  1. Kleihues P, Sobin LH. 1.  2000. World Health Organization classification of tumors. Cancer 88:2887 [Google Scholar]
  2. Gullapalli RR, Desai KV, Santana-Santos L. 2.  et al. 2012. Next generation sequencing in clinical medicine: challenges and lessons for pathology and biomedical informatics. J. Pathol. Inform. 3:40 [Google Scholar]
  3. Johnson A, Zeng J, Bailey AM. 3.  et al. 2015. The right drugs at the right time for the right patient: the MD Anderson precision oncology decision support platform. Drug Discov. Today 20:1433–38 [Google Scholar]
  4. Vogelstein B, Papadopoulos N, Velculescu VE. 4.  et al. 2013. Cancer genome landscapes. Science 339:1546–58 [Google Scholar]
  5. Lawrence MS, Stojanov P, Polak P. 5.  et al. 2013. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499:214–18 [Google Scholar]
  6. Wheeler DA, Wang LH. 6.  2013. From human genome to cancer genome: the first decade. Genome Res 23:1054–62 [Google Scholar]
  7. Fathiamini S, Johnson AM, Zeng J. 7.  et al. 2016. Automated identification of molecular effects of drugs (AIMED). J. Am. Med. Inform. Assoc. 23:758–65 [Google Scholar]
  8. Xu J, Lee HJ, Zeng J. 8.  et al. 2016. Extracting genetic alteration information for personalized cancer therapy from ClinicalTrials.gov. J. Am. Med. Inform. Assoc. 23:750–57 [Google Scholar]
  9. Dogruluk T, Tsang YH, Espitia M. 9.  et al. 2015. Identification of variant-specific functions of PIK3CA by rapid phenotyping of rare mutations. Cancer Res 75:5341–54 [Google Scholar]
  10. Sboner A, Mu XJ, Greenbaum D. 10.  et al. 2011. The real cost of sequencing: higher than you think. ! Genome Biol 12:125 [Google Scholar]
  11. 11.  2016. North American nuclear medicine/radiopharmaceuticals market by type (diagnostic (SPECT–technetium, PET–F-18), therapeutic (beta emitters—I-131, alpha emitters, brachytherapy—Y-90)), by application (oncology, cardiology)—forecasts to 2020 http://www.reportsnreports.com/reports/236047-north-american-nuclear-medicine-radiopharmaceuticals-stable-isotopes-market-spect-pet-radioisotopes-technetium-f-18-beta-alpha-radiation-therapy-i131-y-90-applications-cancer-oncology-cardiac-deuterium-c-13-forecast-to-2017.html
  12. 12.  2016. Next generation sequencing market: increasing NGS applications driving the market globally to 2020 http://www.prnewswire.com/news-releases/next-generation-sequencing-market-increasing-ngs-applications-driving-the-market-globally-to-2020-571967201.html
  13. Schweiger MR, Kerick M, Timmermann B, Isau M. 13.  2011. The power of NGS technologies to delineate the genome organization in cancer: from mutations to structural variations and epigenetic alterations. Cancer Metast. Rev. 30:199–210 [Google Scholar]
  14. Boland GM, Piha-Paul SA, Subbiah V. 14.  et al. 2015. Clinical next generation sequencing to identify actionable aberrations in a phase I program. Oncotarget 6:20099–110 [Google Scholar]
  15. Singh RR, Patel KP, Routbort MJ. 15.  et al. 2013. Clinical validation of a next-generation sequencing screen for mutational hotspots in 46 cancer-related genes. J. Mol. Diagnostics 15:607–22 [Google Scholar]
  16. Kanagal-Shamanna R, Portier BP, Singh RR. 16.  et al. 2014. Next-generation sequencing-based multi-gene mutation profiling of solid tumors using fine needle aspiration samples: promises and challenges for routine clinical diagnostics. Mod. Pathol. 27:314–27 [Google Scholar]
  17. Frampton GM, Fichtenholtz A, Otto GA. 17.  et al. 2013. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat. Biotechnol. 31:1023–31 [Google Scholar]
  18. Grasso C, Butler T, Rhodes K. 18.  et al. 2015. Assessing copy number alterations in targeted, amplicon-based next-generation sequencing data. J. Mol. Diagnostics 17:53–63 [Google Scholar]
  19. Mamanova L, Coffey AJ, Scott CE. 19.  et al. 2010. Target-enrichment strategies for next-generation sequencing. Nat. Methods 7:111–18 [Google Scholar]
  20. Barnard R, Futo V, Pecheniuk N. 20.  et al. 1998. PCR bias toward the wild-type k-ras and p53 sequences: implications for PCR detection of mutations and cancer diagnosis. Biotechniques 25:684–91 [Google Scholar]
  21. Hahn S, Garvin AM. Naro E, Holzgreve W. 21. , Di 1998. Allele drop-out can occur in alleles differing by a single nucleotide and is not alleviated by preamplification or minor template increments. Genet. Test 2:351–55 [Google Scholar]
  22. Gray PN, Dunlop CL, Elliott AM. 22.  2015. Not all next generation sequencing diagnostics are created equal: understanding the nuances of solid tumor assay design for somatic mutation detection. Cancers 7:1313–32 [Google Scholar]
  23. Kanagawa T. 23.  2003. Bias and artifacts in multitemplate polymerase chain reactions (PCR). J. Biosci. Bioeng. 96:317–23 [Google Scholar]
  24. Smith EN, Jepsen K, Khosroheidari M. 24.  et al. 2014. Biased estimates of clonal evolution and subclonal heterogeneity can arise from PCR duplicates in deep sequencing experiments. Genome Biol 15:420 [Google Scholar]
  25. Sawyer SL, Hartley T, Dyment DA. 25.  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]
  26. Redig AJ, Janne PA. 26.  2015. Basket trials and the evolution of clinical trial design in an era of genomic medicine. J. Clin. Oncol. 33:975–77 [Google Scholar]
  27. Weinstein JN, Collisson EA, Mills GB. 27.  et al. 2013. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45:1113–20 [Google Scholar]
  28. Kandoth C, McLellan MD, Vandin F. 28.  et al. 2013. Mutational landscape and significance across 12 major cancer types. Nature 502:333–39 [Google Scholar]
  29. Ciriello G, Miller ML, Aksoy BA. 29.  et al. 2013. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45:1127–33 [Google Scholar]
  30. Sastre L. 30.  2013. Exome sequencing: what clinicians need to know. Adv. Genom. Genet. 2014:415–27 [Google Scholar]
  31. Via M, Gignoux C, Burchard EG. 31.  2010. The 1000 Genomes Project: new opportunities for research and social challenges. Genome Med 2:3 [Google Scholar]
  32. Van Allen EM, Wagle N, Stojanov P. 32.  et al. 2014. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nat. Med. 20:682–88 [Google Scholar]
  33. Stratton MR, Campbell PJ, Futreal PA. 33.  2009. The cancer genome. Nature 458:719–24 [Google Scholar]
  34. Basanta D, Gatenby RA, Anderson AR. 34.  2012. Exploiting evolution to treat drug resistance: combination therapy and the double bind. Mol. Pharm. 9:914–21 [Google Scholar]
  35. Lee MJ, Ye AS, Gardino AK. 35.  et al. 2012. Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks. Cell 149:780–94 [Google Scholar]
  36. Nelander S, Wang W, Nilsson B. 36.  et al. 2008. Models from experiments: combinatorial drug perturbations of cancer cells. Mol. Syst. Biol. 4:216 [Google Scholar]
  37. Rahman N. 37.  2014. Realizing the promise of cancer predisposition genes. Nature 505:302–8 [Google Scholar]
  38. Hartmann LC, Sellers TA, Schaid DJ. 38.  et al. 2001. Efficacy of bilateral prophylactic mastectomy in BRCA1 and BRCA2 gene mutation carriers. J. Natl. Cancer Inst. 93:1633–37 [Google Scholar]
  39. Byrski T, Dent R, Blecharz P. 39.  et al. 2012. Results of a phase II open-label, non-randomized trial of cisplatin chemotherapy in patients with BRCA1-positive metastatic breast cancer. Breast Cancer Res. 14:R110 [Google Scholar]
  40. Turner NC, Tutt AN. 40.  2012. Platinum chemotherapy for BRCA1-related breast cancer: Do we need more evidence?. Breast Cancer Res. 14:115 [Google Scholar]
  41. Fong PC, Boss DS, Yap TA. 41.  et al. 2009. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 361:123–34 [Google Scholar]
  42. Bolton KL, Chenevix-Trench G, Goh C. 42.  et al. 2012. Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA 307:382–90 [Google Scholar]
  43. Hyman DM, Zhou Q, Iasonos A. 43.  et al. 2012. Improved survival for BRCA2-associated serous ovarian cancer compared with both BRCA-negative and BRCA1-associated serous ovarian cancer. Cancer 118:3703–9 [Google Scholar]
  44. Hennessy BTJ, Timms KM, Carey MS. 44.  et al. 2010. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. J. Clin. Oncol. 28:3570–76 [Google Scholar]
  45. Daly MB, Pilarski R, Axilbund JE. 45.  et al. 2014. Genetic/familial high-risk assessment: breast and ovarian, version 1.2014. J. Natl. Compr. Cancer Netw. 12:1326–38 [Google Scholar]
  46. Bancroft EK, Page EC, Castro E. 46.  et al. 2014. Targeted prostate cancer screening in BRCA1 and BRCA2 mutation carriers: results from the initial screening round of the IMPACT study. Eur. Urol. 66:489–99 [Google Scholar]
  47. Castro E, Goh C, Leongamornlert D. 47.  et al. 2015. Effect of BRCA mutations on metastatic relapse and cause-specific survival after radical treatment for localised prostate cancer. Eur. Urol. 68:186–93 [Google Scholar]
  48. Osorio A, de la Hoya M, Rodriguez-Lopez R. 48.  et al. 2002. Loss of heterozygosity analysis at the BRCA loci in tumor samples from patients with familial breast cancer. Int. J. Cancer 99:305–9 [Google Scholar]
  49. Spain BH, Larson CJ, Shihabuddin LS. 49.  et al. 1999. Truncated BRCA2 is cytoplasmic: implications for cancer-linked mutations. PNAS 96:13920–25 [Google Scholar]
  50. Dienstmann R, Dong F, Borger D. 50.  et al. 2014. Standardized decision support in next generation sequencing reports of somatic cancer variants. Mol. Oncol. 8:859–73 [Google Scholar]
  51. Servant N, Romejon J, Gestraud P. 51.  et al. 2014. Bioinformatics for precision medicine in oncology: principles and application to the SHIVA clinical trial. Front. Genet. 5:152 [Google Scholar]
  52. O'Donovan PJ, Livingston DM. 52.  2010. BRCA1 and BRCA2: breast/ovarian cancer susceptibility gene products and participants in DNA double-strand break repair. Carcinogenesis 31:961–67 [Google Scholar]
  53. Pennington KP, Swisher EM. 53.  2012. Hereditary ovarian cancer: beyond the usual suspects. Gynecol. Oncol. 124:347–53 [Google Scholar]
  54. Randall LM, Pothuri B. 54.  2016. The genetic prediction of risk for gynecologic cancers. Gynecol. Oncol. 141:10–16 [Google Scholar]
  55. Watson P, Vasen HF, Mecklin JP. 55.  et al. 2008. The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. Int. J. Cancer 123:444–49 [Google Scholar]
  56. Dudley JC, Lin MT, Le DT, Eshleman JR. 56.  2016. Microsatellite instability as a biomarker for PD-1 blockade. Clin. Cancer Res. 22:813–20 [Google Scholar]
  57. Schmeler KM, Lynch HT, Chen LM. 57.  et al. 2006. Prophylactic surgery to reduce the risk of gynecologic cancers in the Lynch syndrome. N. Engl. J. Med. 354:261–69 [Google Scholar]
  58. Le DT, Uram JN, Wang H. 58.  et al. 2015. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372:2509–20 [Google Scholar]
  59. Kan ZY, Jaiswal BS, Stinson J. 59.  et al. 2010. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466:869–73 [Google Scholar]
  60. 60.  2016. Patients with NTRK fusions respond to targeted therapies. Cancer Discov. 6:566–67 [Google Scholar]
  61. Hurwitz H, Hainsworth JD, Swanton C. 61.  et al. 2016. Targeted therapy for gastrointestinal (GI) tumors based on molecular profiles: early results from MyPathway, an open-label phase IIa basket study in patients with advanced solid tumors. J. Clin. Oncol. 34:Suppl. 4S Abstr 653 [Google Scholar]
  62. Janku F, Hong DS, Fu SQ. 62.  et al. 2014. Assessing PIK3CA and PTEN in early-phase trials with PI3K/AKT/mTOR inhibitors. Cell Rep 6:377–87 [Google Scholar]
  63. De Roock W, Claes B, Bernasconi D. 63.  et al. 2010. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 11:753–62 [Google Scholar]
  64. Bardelli A, Corso S, Bertotti A. 64.  et al. 2013. Amplification of the MET receptor drives resistance to anti-egfr therapies in colorectal cancer. Cancer Discov 3:658–73 [Google Scholar]
  65. Bertotti A, Papp E, Jones S. 65.  et al. 2015. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature 526:263–67 [Google Scholar]
  66. Lim SM, Kim HR, Cho EK. 66.  et al. 2016. Targeted sequencing identifies genetic alterations that confer primary resistance to EGFR tyrosine kinase inhibitor (Korean Lung Cancer Consortium). Oncotarget 736311–20
  67. Johnson DB, Menzies AM, Zimmer L. 67.  et al. 2015. Acquired BRAF inhibitor resistance: a multicenter meta-analysis of the spectrum and frequencies, clinical behaviour, and phenotypic associations of resistance mechanisms. Eur. J. Cancer 51:2792–99 [Google Scholar]
  68. Schwaederle M, Zhao M, Lee JJ. 68.  et al. 2015. Impact of precision medicine in diverse cancers: a meta-analysis of phase II clinical trials. J. Clin. Oncol. 33:3817–25 [Google Scholar]
  69. Prasad V, Fojo T, Brada M. 69.  2016. Precision oncology: origins, optimism, and potential. Lancet Oncol 17:E81–E86 [Google Scholar]
  70. Lopez-Chavez A, Thomas A, Rajan A. 70.  et al. 2015. Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarker-derived, multiarm, multihistology phase II basket trial. J. Clin. Oncol. 33:1000–7 [Google Scholar]
  71. Davis J. 71.  2016. Providers say they're ready to progress to precision medicine http://www.healthcareitnews.com/news/providers-say-theyre-ready-progress-precision-medicine
  72. 72.  2016. Healthcare gets personal https://dam.sap.com/mac/preview/a/67/UmEUnxHHnJJzOOlUwAmnyXggPmAJJzgxUPAEmOXHxcwxPEPc/open20160412042800.htm
  73. Stearns V, Park BH. 73.  2015. Gene mutation profiling of breast cancers for clinical decision making: drivers and passengers in the cart before the horse. JAMA Oncol 1:569–70 [Google Scholar]
/content/journals/10.1146/annurev-med-102115-021556
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