DOI QR코드

DOI QR Code

Comparison of the Genetic Alterations between Primary Colorectal Cancers and Their Corresponding Patient-Derived Xenograft Tissues

  • Yu, Sang Mi (Department of Microbiology, College of Medicine, The Catholic University of Korea) ;
  • Jung, Seung-Hyun (Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea) ;
  • Chung, Yeun-Jun (Department of Microbiology, College of Medicine, The Catholic University of Korea)
  • Received : 2018.05.08
  • Accepted : 2018.05.16
  • Published : 2018.06.30

Abstract

Patient-derived xenograft (PDX) models are useful tools for tumor biology research and testing the efficacy of candidate anticancer drugs targeting the druggable mutations identified in tumor tissue. However, it is still unknown how much of the genetic alterations identified in primary tumors are consistently detected in tumor tissues in the PDX model. In this study, we analyzed the genetic alterations of three primary colorectal cancers (CRCs) and matched xenograft tissues in PDX models using a next-generation sequencing cancer panel. Of the 17 somatic mutations identified from the three CRCs, 14 (82.4%) were consistently identified in both primary and xenograft tumors. The other three mutations identified in the primary tumor were not detected in the xenograft tumor tissue. There was no newly identified mutation in the xenograft tumor tissues. In addition to the somatic mutations, the copy number alteration profiles were also largely consistent between the primary tumor and xenograft tissue. All of these data suggest that the PDX tumor model preserves the majority of the key mutations detected in the primary tumor site. This study provides evidence that the PDX model is useful for testing targeted therapies in the clinical field and research on precision medicine.

Keywords

References

  1. Davey JW, Hohenlohe PA, Etter PD, Boone JQ, Catchen JM, Blaxter ML. Genome-wide genetic marker discovery and genotyping using next-generation sequencing. Nat Rev Genet 2011;12:499-510. https://doi.org/10.1038/nrg3012
  2. Han SW, Kim HP, Shin JY, Jeong EG, Lee WC, Lee KH, et al. Targeted sequencing of cancer-related genes in colorectal cancer using next-generation sequencing. PLoS One 2013;8: e64271. https://doi.org/10.1371/journal.pone.0064271
  3. Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 2010;363:1693-1703. https://doi.org/10.1056/NEJMoa1006448
  4. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat Rev Clin Oncol 2012;9:338-350. https://doi.org/10.1038/nrclinonc.2012.61
  5. Lee WS, Kim HY, Seok JY, Jang HH, Park YH, Kim SY, et al. Genomic profiling of patient-derived colon cancer xenograft models. Medicine (Baltimore) 2014;93:e298. https://doi.org/10.1097/MD.0000000000000298
  6. Daniel VC, Marchionni L, Hierman JS, Rhodes JT, Devereux WL, Rudin CM, et al. A primary xenograft model of small-cell lung cancer reveals irreversible changes in gene expression imposed by culture in vitro. Cancer Res 2009;69:3364-3373. https://doi.org/10.1158/0008-5472.CAN-08-4210
  7. John T, Kohler D, Pintilie M, Yanagawa N, Pham NA, Li M, et al. The ability to form primary tumor xenografts is predictive of increased risk of disease recurrence in early-stage non-small cell lung cancer. Clin Cancer Res 2011;17:134-141. https://doi.org/10.1158/1078-0432.CCR-10-2224
  8. Gu Z, Jiang J, Yan Y, Tan T, Yu T, Liu Z, et al. Evaluation of the correlations between patient-derived xenograft (PDX) mod- el-based mouse trials and cancer patient-based clinical trials. J Clin Oncol 2017;35:e23140. https://doi.org/10.1200/JCO.2017.35.15_suppl.e23140
  9. Fichtner I, Rolff J, Soong R, Hoffmann J, Hammer S, Sommer A, et al. Establishment of patient-derived non-small cell lung cancer xenografts as models for the identification of predictive biomarkers. Clin Cancer Res 2008;14:6456-6468. https://doi.org/10.1158/1078-0432.CCR-08-0138
  10. Choi SH, Jung SH, Chung YJ. Validation of customized cancer panel for detecting somatic mutations and copy number alterations. Genomics Inform 2017;15:136-141. https://doi.org/10.5808/GI.2017.15.4.136
  11. Tso KY, Lee SD, Lo KW, Yip KY. Are special read alignment strategies necessary and cost-effective when handling sequencing reads from patient-derived tumor xenografts? BMC Genomics 2014;15:1172. https://doi.org/10.1186/1471-2164-15-1172
  12. Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol 2011;29:24-26. https://doi.org/10.1038/nbt.1754
  13. Rathi V, Wright G, Constantin D, Chang S, Pham H, Jones K, et al. Clinical validation of the 50 gene AmpliSeq Cancer Panel V2 for use on a next generation sequencing platform using formalin fixed, paraffin embedded and fine needle aspiration tumour specimens. Pathology 2017;49:75-82. https://doi.org/10.1016/j.pathol.2016.08.016
  14. Jung SH, Kim MS, Lee SH, Park HC, Choi HJ, Maeng L, et al. Whole-exome sequencing identifies recurrent AKT1 mutations in sclerosing hemangioma of lung. Proc Natl Acad Sci U S A 2016;113:10672-10677. https://doi.org/10.1073/pnas.1606946113
  15. Jung KW, Won YJ, Oh CM, Kong HJ, Lee DH, Lee KH. Prediction of cancer incidence and mortality in Korea, 2017. Cancer Res Treat 2017;49:306-312. https://doi.org/10.4143/crt.2017.130
  16. Hao C, Wang L, Peng S, Cao M, Li H, Hu J, et al. Gene mutations in primary tumors and corresponding patient-derived xenografts derived from non-small cell lung cancer. Cancer Lett 2015;357:179-185. https://doi.org/10.1016/j.canlet.2014.11.024
  17. Choi YY, Lee JE, Kim H, Sim MH, Kim KK, Lee G, et al. Establishment and characterisation of patient-derived xeno- grafts as paraclinical models for gastric cancer. Sci Rep 2016;6:22172. https://doi.org/10.1038/srep22172