DOI QR코드

DOI QR Code

Clinical relevance of Lgr5 expression in colorectal cancer patients

  • Kim, Young Joo (Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine) ;
  • Kang, Dong Hyun (Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine) ;
  • Song, Geum Jong (Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine) ;
  • Ahn, Tae Sung (Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine) ;
  • Son, Myoung Won (Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine) ;
  • Lee, Moon Soo (Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine) ;
  • Baek, Moo-Jun (Department of Surgery, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine)
  • 투고 : 2018.07.31
  • 심사 : 2018.11.29
  • 발행 : 2018.12.31

초록

Purpose: Lgr5 is a well-known stem cell marker in colorectal cancer (CRC). This retrospective study evaluated the expressions of Lgr5 in CRC specimens, and examined whether these expressions were associated with survival outcomes. Methods: We used immunohistochemistry to retrospectively examine expressions of Lgr5 in paraffin-embedded specimens from 337 patients with CRC between January 2009 and December 2013. All clinicopathologic data were collected by retrospective review based on medical records. The correlation between its expression and clinicopathological data as well as clinical outcomes of patients was analyzed. Results: Low expression and high expression of Lgr5 in 337 patients were 175 (51.9%) and 162 (48.1%), respectively. There was no statistically significant difference in the association of Lgr5 expression with clinicopathologic factors (age, tumor location, lymphatic invasion, vascular invasion, perineural invasion, TNM stage, and differentiation). In the survival analysis, the high expression group of Lgr5 showed a better prognosis than the low expression group in disease-free survival (P=0.044). However, overall survival was not significantly different between the two groups (P=0.087). In multivariate analysis, we found that high expression of Lgr5 was independent prognostic factor for tumor relapse (hazard ratio, 0.601; 95% confidence interval, 0.388-0.929; P=0.022). Conclusion: In present study, high expression of Lgr5 is an independent predictor of favorable prognosis in patients with CRC. So, further well designed, prospective, large scale studies are needed to examine the value of Lgr5 as a prognostic biomarker for CRC.

키워드

과제정보

연구 과제 주관 기관 : Korea Health Industry Development Institute (KHID)

참고문헌

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424. https://doi.org/10.3322/caac.21492
  2. National Cancer Information Center. Caner statistics, 2017 [Internet]. Goyang (KR): National Cancer Information Center [cited 2018 Jul 10]. Available from: https://www.cancer.go.kr/lay1/S1T645C646/contents.do
  3. Jung KW, Won YJ, Oh CM, Kong HJ, Lee DH, Lee KH, et al. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2014. Cancer Res Treat 2017;49:292-305. https://doi.org/10.4143/crt.2017.118
  4. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018;68:7-30. https://doi.org/10.3322/caac.21442
  5. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, et al. Identification and expansion of human colon-cancer-initiating cells. Nature 2007;445:111-5. https://doi.org/10.1038/nature05384
  6. Morin PJ, Sparks AB, Korinek V, Barker N, Clevers H, Vogelstein B, et al. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 1997;275:1787-90. https://doi.org/10.1126/science.275.5307.1787
  7. Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, et al. APC mutations occur early during colorectal tumorigenesis. Nature 1992;359:235-7. https://doi.org/10.1038/359235a0
  8. de Lau W, Peng WC, Gros P, Clevers H. The R-spondin/Lgr5/Rnf43 module: regulator of Wnt signal strength. Genes Dev 2014;28:305-16. https://doi.org/10.1101/gad.235473.113
  9. Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M, et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 2009;457:608-11. https://doi.org/10.1038/nature07602
  10. Ziskin JL, Dunlap D, Yaylaoglu M, Fodor IK, Forrest WF, Patel R, et al. In situ validation of an intestinal stem cell signature in colorectal cancer. Gut 2013;62:1012-23. https://doi.org/10.1136/gutjnl-2011-301195
  11. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003;100:3983-8. https://doi.org/10.1073/pnas.0530291100
  12. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997;3:730-7. https://doi.org/10.1038/nm0797-730
  13. Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, et al. Identification of cells initiating human melanomas. Nature 2008;451:345-9. https://doi.org/10.1038/nature06489
  14. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, et al. Identification of pancreatic cancer stem cells. Cancer Res 2007;67:1030-7. https://doi.org/10.1158/0008-5472.CAN-06-2030
  15. O'Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007;445:106-10. https://doi.org/10.1038/nature05372
  16. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature 2004;432:396-401. https://doi.org/10.1038/nature03128
  17. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007;449:1003-7. https://doi.org/10.1038/nature06196
  18. de Sousa E Melo F, Colak S, Buikhuisen J, Koster J, Cameron K, de Jong JH, et al. Methylation of cancer-stem-cell-associated Wnt target genes predicts poor prognosis in colorectal cancer patients. Cell Stem Cell 2011;9:476-85. https://doi.org/10.1016/j.stem.2011.10.008
  19. Walker F, Zhang HH, Odorizzi A, Burgess AW. LGR5 is a negative regulator of tumourigenicity, antagonizes Wnt signalling and regulates cell adhesion in colorectal cancer cell lines. PLoS One 2011;6:e22733. https://doi.org/10.1371/journal.pone.0022733
  20. Melling N, Kowitz CM, Simon R, Bokemeyer C, Terracciano L, Sauter G, et al. High Ki67 expression is an independent good prognostic marker in colorectal cancer. J Clin Pathol 2016;69:209-14. https://doi.org/10.1136/jclinpath-2015-202985
  21. Anjomshoaa A, Nasri S, Humar B, McCall JL, Chatterjee A, Yoon HS, et al. Slow proliferation as a biological feature of colorectal cancer metastasis. Br J Cancer 2009;101:822-8. https://doi.org/10.1038/sj.bjc.6605229
  22. Yang L, Lin Y, Liu Z, Li Y, Liu Y, Liang R. Impact of LGR5 in colorectal cancer on overall and progression-free survival: a systematic review and meta-analysis. Int J Clin Exp Med 2016;9:10537-43.
  23. Jiang Y, Li W, He X, Zhang H, Jiang F, Chen Z. Lgr5 expression is a valuable prognostic factor for colorectal cancer: evidence from a meta-analysis. BMC Cancer 2015;15:948. https://doi.org/10.1186/s12885-015-1985-3
  24. Merlos-Suarez A, Barriga FM, Jung P, Iglesias M, Cespedes MV, Rossell D, et al. The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell 2011;8:511-24. https://doi.org/10.1016/j.stem.2011.02.020
  25. Yang L, Tang H, Kong Y, Xie X, Chen J, Song C, et al. LGR5 promotes breast cancer progression and maintains stem-like cells through activation of Wnt/beta-catenin signaling. Stem Cells 2015;33:2913-24. https://doi.org/10.1002/stem.2083
  26. Simon E, Petke D, Boger C, Behrens HM, Warneke V, Ebert M, et al. The spatial distribution of LGR5+ cells correlates with gastric cancer progression. PLoS One 2012;7:e35486. https://doi.org/10.1371/journal.pone.0035486
  27. de Sousa e Melo F, Kurtova AV, Harnoss JM, Kljavin N, Hoeck JD, Hung J, et al. A distinct role for Lgr5(+) stem cells in primary and metastatic colon cancer. Nature 2017;543:676-80. https://doi.org/10.1038/nature21713
  28. Fujii M, Sato T. Defining the role of Lgr5(+) stem cells in colorectal cancer: from basic research to clinical applications. Genome Med 2017;9:66. https://doi.org/10.1186/s13073-017-0460-y
  29. Junttila MR, Mao W, Wang X, Wang BE, Pham T, Flygare J, et al. Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Sci Transl Med 2015;7:314ra186. https://doi.org/10.1126/scitranslmed.aac7433
  30. Hutchins G, Southward K, Handley K, Magill L, Beaumont C, Stahlschmidt J, et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol 2011;29:1261-70. https://doi.org/10.1200/JCO.2010.30.1366