Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The path to metastatic mouse models of colorectal cancer

Abstract

The study and comprehension of the molecular mechanisms underlying cancer biology strongly rely on mouse modeling. An ideal mouse model should have molecular, histopathological, and etiological characteristics as close as possible to those of the corresponding human tumors. Among solid tumors, colorectal cancer (CRC) is one of the malignancies that best suits reproduction in an animal model: it evolves through a progressive set of molecular events and is generally associated with a precise histopathology and a neat cataloging of stages and grades. The development of refined CRC mouse models over several decades has seen them recently evolve toward sophisticated systems that ever more closely approximate the human pathology, with different models addressing different human CRC subtypes. In particular, a metastatic CRC model has been seen as a “holy grail” in this field, and we describe in this review the path taken to achieve metastatic models and discuss the path forward.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1

Similar content being viewed by others

References

  1. Groden J, Thliveris A, Samowitz W, Carlson M, Gelbert L, Albertsen H, et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell. 1991;66:589–600.

    Article  CAS  Google Scholar 

  2. Nishisho I, Nakamura Y, Miyoshi Y, Miki Y, Ando H, Horii A, et al. Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science. 1991;253:665–9.

    Article  CAS  Google Scholar 

  3. Kinzler K, Nilbert M, Su L, Vogelstein B, Bryan T, Levy D, et al. Identification of FAP locus genes from chromosome 5q21. Science. 1991;253:661–5.

    Article  CAS  Google Scholar 

  4. 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.

    Article  CAS  Google Scholar 

  5. Cottrell S, Bicknell D, Kaklamanis L, Bodmer WF. Molecular analysis of APC mutations in familial adenomatous polyposis and sporadic colon carcinomas. Lancet. 1992;340:626–30.

    Article  CAS  Google Scholar 

  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.

    Article  CAS  Google Scholar 

  7. Moser AR, Luongo C, Gould KA, McNeley MK, Shoemaker AR, Dove WF. ApcMin: a mouse model for intestinal and mammary tumorigenesis. Eur J Cancer. 1995;31A:1061–4.

    Article  CAS  Google Scholar 

  8. Smits R, Kartheuser A, Jagmohan-Changur S, Leblanc V, Breukel C, de Vries A, et al. Loss of Apc and the entire chromosome 18 but absence of mutations at the Ras and Tp53 genes in intestinal tumors from Apc1638N, a mouse model for Apc-driven carcinogenesis. Carcinogenesis. 1997;18:321–7.

    Article  CAS  Google Scholar 

  9. Shibata H, Toyama K, Shioya H, Ito M, Hirota M, Hasegawa S, et al. Rapid colorectal adenoma formation initiated by conditional targeting of the Apc gene. Science. 1997;278:120–3.

    Article  CAS  Google Scholar 

  10. Robanus-Maandag EC, Koelink PJ, Breukel C, Salvatori DC, Jagmohan-Changur SC, Bosch CA, et al. A new conditional Apc-mutant mouse model for colorectal cancer. Carcinogenesis. 2010;31:946–52.

    Article  CAS  Google Scholar 

  11. Faller WJ, Jackson TJ, Knight JR, Ridgway RA, Jamieson T, Karim SA, et al. mTORC1-mediated translational elongation limits intestinal tumour initiation and growth. Nature. 2015;517:497–500.

    Article  CAS  Google Scholar 

  12. Andreu P, Colnot S, Godard C, Gad S, Chafey P, Niwa-Kawakita M, et al. Crypt-restricted proliferation and commitment to the Paneth cell lineage following Apc loss in the mouse intestine. Development. 2005;132:1443–51.

    Article  CAS  Google Scholar 

  13. Khazaie K, Zadeh M, Khan MW, Bere P, Gounari F, Dennis K, et al. Abating colon cancer polyposis by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc Natl Acad Sci USA. 2012;109:10462–7.

    Article  CAS  Google Scholar 

  14. Halberg RB, Waggoner J, Rasmussen K, White A, Clipson L, Prunuske AJ, et al. Long-lived Min mice develop advanced intestinal cancers through a genetically conservative pathway. Cancer Res. 2009;69:5768–75.

    Article  CAS  Google Scholar 

  15. Kwong LN, Weiss KR, Haigis KM, Dove WF. Atm is a negative regulator of intestinal neoplasia. Oncogene. 2008;27:1013–8.

    Article  CAS  Google Scholar 

  16. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61:759–67.

    Article  CAS  Google Scholar 

  17. Kwong LN, Dove WF. APC and its modifiers in colon cancer. Adv Exp Med Biol. 2009;656:85–106.

    Article  CAS  Google Scholar 

  18. McIntyre RE, Buczacki SJ, Arends MJ, Adams DJ. Mouse models of colorectal cancer as preclinical models. Bioessays. 2015;37:909–20.

    Article  Google Scholar 

  19. Taketo MM, Edelmann W. Mouse models of colon cancer. Gastroenterology. 2009;136:780–98.

    Article  CAS  Google Scholar 

  20. Zeineldin M, Neufeld KL. More than two decades of Apc modeling in rodents. Biochim Biophys Acta. 2013;1836:80–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 1995;268:1336–8.

    Article  CAS  Google Scholar 

  22. Miyaki M, Iijima T, Konishi M, Sakai K, Ishii A, Yasuno M, et al. Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis. Oncogene. 1999;18:3098–103.

    Article  CAS  Google Scholar 

  23. Takaku K, Oshima M, Miyoshi H, Matsui M, Seldin MF, Taketo MM. Intestinal tumorigenesis in compound mutant mice of both Dpc4 (Smad4) and Apc genes. Cell. 1998;92:645–56.

    Article  CAS  Google Scholar 

  24. Hamamoto T, Beppu H, Okada H, Kawabata M, Kitamura T, Miyazono K, et al. Compound disruption of smad2 accelerates malignant progression of intestinal tumors in apc knockout mice. Cancer Res. 2002;62:5955–61.

    CAS  PubMed  Google Scholar 

  25. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–7.

  26. Haigis KM, Kendall KR, Wang Y, Cheung A, Haigis MC, Glickman JN, et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet. 2008;40:600–8.

    Article  CAS  Google Scholar 

  27. Olivier M, Eeles R, Hollstein M, Khan MA, Harris CC, Hainaut P. The IARC TP53 database: new online mutation analysis and recommendations to users. Hum Mutat. 2002;19:607–14.

    Article  CAS  Google Scholar 

  28. Clarke AR, Cummings MC, Harrison DJ. Interaction between murine germline mutations in P53 and Apc predisposes to pancreatic neoplasia but not to increased intestinal malignancy. Oncogene. 1995;11:1913–20.

    CAS  PubMed  Google Scholar 

  29. Harvey M, McArthur MJ, Montgomery CA Jr, Butel JS, Bradley A, Donehower LA. Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice. Nat Genet. 1993;5:225–9.

    Article  CAS  Google Scholar 

  30. Halberg RB, Katzung DS, Hoff PD, Moser AR, Cole CE, Lubet RA, et al. Tumorigenesis in the multiple intestinal neoplasia mouse: redundancy of negative regulators and specificity of modifiers. Proc Natl Acad Sci USA. 2000;97:3461–6.

    Article  CAS  Google Scholar 

  31. Kwong LN, Shedlovsky A, Biehl BS, Clipson L, Pasch CA, Dove WF. Identification of Mom7, a novel modifier of Apc(Min/+) on mouse chromosome 18. Genetics. 2007;176:1237–44.

    Article  CAS  Google Scholar 

  32. Xue Y, Johnson R, Desmet M, Snyder PW, Fleet JC. Generation of a transgenic mouse for colorectal cancer research with intestinal cre expression limited to the large intestine. Mol Cancer Res. 2010;8:1095–104.

    Article  CAS  Google Scholar 

  33. Hinoi T, Akyol A, Theisen BK, Ferguson DO, Greenson JK, Williams BO, et al. Mouse model of colonic adenoma-carcinoma progression based on somatic Apc inactivation. Cancer Res. 2007;67:9721–30.

    Article  CAS  Google Scholar 

  34. Hung KE, Maricevich MA, Richard LG, Chen WY, Richardson MP, Kunin A, et al. Development of a mouse model for sporadic and metastatic colon tumors and its use in assessing drug treatment. Proc Natl Acad Sci USA. 2010;107:1565–70.

    Article  CAS  Google Scholar 

  35. Boutin AT, Liao WT, Wang M, Hwang SS, Karpinets TV, Cheung H, et al. Oncogenic Kras drives invasion and maintains metastases in colorectal cancer. Genes Dev. 2017;31:370–82.

    Article  CAS  Google Scholar 

  36. Sottoriva A, Kang H, Ma Z, Graham TA, Salomon MP, Zhao J, et al. A big bang model of human colorectal tumor growth. Nat Genet. 2015;47:209–16.

    Article  CAS  Google Scholar 

  37. Beach R, Chan AO, Wu TT, White JA, Morris JS, Lunagomez S, et al. BRAF mutations in aberrant crypt foci and hyperplastic polyposis. Am J Pathol. 2005;166:1069–75.

    Article  CAS  Google Scholar 

  38. Yang S, Farraye FA, Mack C, Posnik O, O’Brien MJ. BRAF and KRAS mutations in hyperplastic polyps and serrated adenomas of the colorectum: relationship to histology and CpG island methylation status. Am J Surg Pathol. 2004;28:1452–9.

    Article  Google Scholar 

  39. Kucherlapati MH, Lee K, Nguyen AA, Clark AB, Hou H Jr., Rosulek A, et al. An Msh2 conditional knockout mouse for studying intestinal cancer and testing anticancer agents. Gastroenterology. 2010;138:993–1002.e1.

    Article  Google Scholar 

  40. Reitmair AH, Cai JC, Bjerknes M, Redston M, Cheng H, Pind MT, et al. MSH2 deficiency contributes to accelerated APC-mediated intestinal tumorigenesis. Cancer Res. 1996;56:2922–6.

    CAS  PubMed  Google Scholar 

  41. Carragher LA, Snell KR, Giblett SM, Aldridge VS, Patel B, Cook SJ, et al. V600EBraf induces gastrointestinal crypt senescence and promotes tumour progression through enhanced CpG methylation of p16INK4a. EMBO Mol Med. 2010;2:458–71.

    Article  CAS  Google Scholar 

  42. Rad R, Cadinanos J, Rad L, Varela I, Strong A, Kriegl L, et al. A genetic progression model of Braf(V600E)-induced intestinal tumorigenesis reveals targets for therapeutic intervention. Cancer Cell. 2013;24:15–29.

    Article  CAS  Google Scholar 

  43. Parsons DW, Wang TL, Samuels Y, Bardelli A, Cummins JM, DeLong L, et al. Colorectal cancer: mutations in a signalling pathway. Nature. 2005;436:792.

    Article  CAS  Google Scholar 

  44. Goel A, Arnold CN, Niedzwiecki D, Carethers JM, Dowell JM, Wasserman L, et al. Frequent inactivation of PTEN by promoter hypermethylation in microsatellite instability-high sporadic colorectal cancers. Cancer Res. 2004;64:3014–21.

    Article  CAS  Google Scholar 

  45. Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology. 2008;135:1079–99.

    Article  CAS  Google Scholar 

  46. Yu M, Trobridge P, Wang Y, Kanngurn S, Morris SM, Knoblaugh S, et al. Inactivation of TGF-beta signaling and loss of PTEN cooperate to induce colon cancer in vivo. Oncogene. 2014;33:1538–47.

    Article  CAS  Google Scholar 

  47. Trobridge P, Knoblaugh S, Washington MK, Munoz NM, Tsuchiya KD, Rojas A, et al. TGF-beta receptor inactivation and mutant Kras induce intestinal neoplasms in mice via a beta-catenin-independent pathway. Gastroenterology. 2009;136:1680–8.e7.

    Article  CAS  Google Scholar 

  48. Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, et al. Recurrent R-spondin fusions in colon cancer. Nature. 2012;488:660–4.

    Article  CAS  Google Scholar 

  49. 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.

    Article  Google Scholar 

  50. Han T, Schatoff EM, Murphy C, Zafra MP, Wilkinson JE, Elemento O, et al. R-Spondin chromosome rearrangements drive Wnt-dependent tumour initiation and maintenance in the intestine. Nat Commun. 2017;8:15945.

    Article  CAS  Google Scholar 

  51. Schwitalla S, Ziegler PK, Horst D, Becker V, Kerle I, Begus-Nahrmann Y, et al. Loss of p53 in enterocytes generates an inflammatory microenvironment enabling invasion and lymph node metastasis of carcinogen-induced colorectal tumors. Cancer Cell. 2013;23:93–106.

    Article  CAS  Google Scholar 

  52. Pan Q, Lou X, Zhang J, Zhu Y, Li F, Shan Q, et al. Genomic variants in mouse model induced by azoxymethane and dextran sodium sulfate improperly mimic human colorectal cancer. Sci Rep. 2017;7:25.

    Article  Google Scholar 

  53. Sanchez-Rivera FJ, Jacks T. Applications of the CRISPR-Cas9 system in cancer biology. Nat Rev Cancer. 2015;15:387–95.

    Article  CAS  Google Scholar 

  54. Platt RJ, Chen S, Zhou Y, Yim MJ, Swiech L, Kempton HR, et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell. 2014;159:440–55.

    Article  CAS  Google Scholar 

  55. Roper J, Tammela T, Cetinbas NM, Akkad A, Roghanian A, Rickelt S, et al. In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat Biotechnol. 2017;35:569–76.

    Article  CAS  Google Scholar 

  56. Dow LE, O’Rourke KP, Simon J, Tschaharganeh DF, van Es JH, Clevers H, et al. Apc restoration promotes cellular differentiation and reestablishes crypt homeostasis in colorectal. Cancer Cell. 2015;161:1539–52.

    CAS  Google Scholar 

  57. O’Rourke KP, Loizou E, Livshits G, Schatoff EM, Baslan T, Manchado E, et al. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat Biotechnol. 2017;35:577–82.

    Article  Google Scholar 

  58. Velcich A, Yang W, Heyer J, Fragale A, Nicholas C, Viani S, et al. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science. 2002;295:1726–9.

    Article  CAS  Google Scholar 

  59. Zhu Y, Richardson JA, Parada LF, Graff JM. Smad3 mutant mice develop metastatic colorectal cancer. Cell. 1998;94:703–14.

    Article  CAS  Google Scholar 

  60. Leystra AA, Deming DA, Zahm CD, Farhoud M, Olson TJ, Hadac JN, et al. Mice expressing activated PI3K rapidly develop advanced colon cancer. Cancer Res. 2012;72:2931–6.

    Article  CAS  Google Scholar 

  61. Maggio-Price L, Treuting P, Zeng W, Tsang M, Bielefeldt-Ohmann H, Iritani BM. Helicobacter infection is required for inflammation and colon cancer in SMAD3-deficient mice. Cancer research. 2006;66:828–38.

    Article  CAS  Google Scholar 

  62. Chen HJ, Wei Z, Sun J, Bhattacharya A, Savage DJ, Serda R, et al. A recellularized human colon model identifies cancer driver genes. Nat Biotechnol. 2016;34:845–51.

    Article  Google Scholar 

Download references

Acknowledgements

We thank Lukas E Dow (Weill Cornell Medicine) for critical review of the manuscript and expert editorial assistance. The figure was produced, in part, by using Servier Medical Art collection.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lawrence N. Kwong.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Romano, G., Chagani, S. & Kwong, L.N. The path to metastatic mouse models of colorectal cancer. Oncogene 37, 2481–2489 (2018). https://doi.org/10.1038/s41388-018-0155-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-018-0155-x

This article is cited by

Search

Quick links