Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-06-08T12:24:42.844Z Has data issue: false hasContentIssue false

47 - Colon and rectal cancer

from Part 3.1 - Molecular pathology: carcinomas

Published online by Cambridge University Press:  05 February 2015

Erin M. Perchiniak
Affiliation:
Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA
Joanna Groden
Affiliation:
Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Colorectal cancer (CRC) represents roughly 10% of all new cancer cases worldwide each year, making it the third most common cancer and the second most common cause of cancer-related death. The National Cancer Institute estimates that in 2009 in the United States, there will be 106100 colon and 40870 rectal cancers newly diagnosed, and 49920 deaths. Colon cancer affects both sexes with equal frequency, although rectal cancer has nearly double its occurrence in men. The incidence of CRC worldwide varies greatly, with much higher rates in Western populations. Interestingly, incidence amongst immigrants to countries with higher rates of CRC rapidly reaches the rates of the host population, suggesting that diet and environment are major contributors to risk (1,2). A set of genetic alterations have been well defined as key regulators of colorectal cancer onset and progression. These genetic contributors to CRC are the focus of this chapter, along with current therapies for the treatment of CRC. Ongoing research efforts will further define the molecular events leading to CRC onset and improve personalized treatment options.

Staging and histopathology of CRC

Colon and rectal cancers are often discussed synonymously because they share many common factors. Most CRCs begin as benign polyps, or adenomas, anywhere in the gastrointestinal tract, from cecum to rectum, that then become invasive (3). Stage is defined by several factors, but most importantly by tumor size, degree of invasion, regional lymph node involvement, and distant metastasis. The tumor stage upon diagnosis largely predicts the survival outcomes and influences the particular course of treatment.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 547 - 556
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Haenszel, W. Cancer mortality among the foreign-born in the United States. Journal of the National Cancer Institute 1961;26:37–132.Google ScholarPubMed
McMichael, AJ, Giles, GG. Cancer in migrants to Australia: extending the descriptive epidemiological data. Cancer Research 1988;48:751–6.
Fearon, ER, Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990;61:759–67.CrossRef
Reya, T, Clevers, H. Wnt signalling in stem cells and cancer. Nature 2005;434:843–50.CrossRef
Muto, T, Bussey, HJ, Morson BC. The evolution of cancer of the colon and rectum. Cancer 1975;36:2251–70.CrossRef
Bird, RP, Good, CK. The significance of aberrant crypt foci in understanding the pathogenesis of colon cancer. Toxicology Letters 2000;112–113:395–402.
Muto, T, Kamiya, J, Sawada, T, et al. Small “flat adenoma” of the large bowel with special reference to its clinicopathologic features. Diseases of the Colon and Rectum 1985;28:847–51.CrossRef
Bisgaard, ML, Fenger, K, Bulow, S, Niebuhr, E, Mohr, J. Familial adenomatous polyposis (FAP): frequency, penetrance, and mutation rate. Human Mutation 1994;3:121–5.CrossRef
Lipton, L, Tomlinson, I. The genetics of FAP and FAP-like syndromes. Familial Cancer 2006;5:221–6.CrossRef
Lal, G, Gallinger, S. Familial adenomatous polyposis. Seminars in Surgical Oncology 2000;18:314–23.3.0.CO;2-9>CrossRef
Nishisho, I, Nakamura, Y, Miyoshi, Y, et al. Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science 1991;253:665–9.CrossRef
Groden, J, Thliveris, A, Samowitz, W, et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell 1991;66:589–600.CrossRef
Leppert, M, Dobbs, M, Scambler, P, et al. The gene for familial polyposis coli maps to the long arm of chromosome 5. Science 1987;238:1411–13.CrossRef
Bodmer, WF, Bailey, CJ, Bodmer, J, et al. Localization of the gene for familial adenomatous polyposis on chromosome 5. Nature 1987;328:614–16.CrossRef
Nieuwenhuis, MH, Vasen, HF. Correlations between mutation site in APC and phenotype of familial adenomatous polyposis (FAP): a review of the literature. Critical Reviews in Oncology and Hematology 2007;61:153–61.CrossRef
Miyoshi, Y, Nagase, H, Ando, H, et al. Somatic mutations of the APC gene in colorectal tumors: mutation cluster region in the APC gene. Human Molecular Genetics 1992;1:229–33.CrossRef
Powell, SM, Zilz, N, Beazer-Barclay, Y, et al. APC mutations occur early during colorectal tumorigenesis. Nature 1992;359:235–7.CrossRef
Nathke, IS. The adenomatous polyposis coli protein: the Achilles heel of the gut epithelium. Annual Review of Cell and Developmental Biology 2004;20:337–66.CrossRef
Nathke, I. Cytoskeleton out of the cupboard: colon cancer and cytoskeletal changes induced by loss of APC. Nature Reviews Cancer 2006;6:967–74.CrossRef
Watson, SA. Oncogenic targets of beta-catenin-mediated transcription in molecular pathogenesis of intestinal polyposis. Lancet 2001;357:572–3.CrossRef
Andreyev, HJ, Norman, AR, Cunningham, D, Oates, JR, Clarke, PA. Kirsten ras mutations in patients with colorectal cancer: the multicenter “RASCAL” study. Journal of the National Cancer Institute 1998;90:675–84.CrossRefGoogle ScholarPubMed
Downward, J. Targeting RAS signalling pathways in cancer therapy. Nature Reviews Cancer 2003;3:11–22.CrossRef
Repasky, GA, Chenette, EJ, Der, CJ. Renewing the conspiracy theory debate: does Raf function alone to mediate Ras oncogenesis? Trends in Cell Biology 2004;14:639–47.
Wee, S, Wiederschain, D, Maira, SM, et al. PTEN-deficient cancers depend on PIK3CB. Proceedings of the National Academy of Sciences USA 2008;105:13 057–62.
Cantley, LC, Neel, BG. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proceedings of the National Academy of Sciences USA 1999;96:4240–5.CrossRef
Samuels, Y, Diaz, LA, Jr., Schmidt-Kittler, O, et al. Mutant PIK3CA promotes cell growth and invasion of human cancer cells. Cancer Cell 2005;7:561–73.CrossRef
Russo, A, Bazan, V, Iacopetta, B, et al. The TP53 colorectal cancer international collaborative study on the prognostic and predictive significance of p53 mutation:influence of tumor site, type of mutation, and adjuvant treatment. Journal of Clinical Oncology 2005;23:7518–28.CrossRefGoogle ScholarPubMed
Munro, AJ, Lain, S, Lane, DP. P53 abnormalities and outcomes in colorectal cancer: a systematic review. British Journal of Cancer 2005;92:434–44.CrossRefGoogle ScholarPubMed
Popat, S, Houlston, RS. A systematic review and meta-analysis of the relationship between chromosome 18q genotype, DCC status and colorectal cancer prognosis. European Journal of Cancer 2005;41:2060–70.CrossRefGoogle ScholarPubMed
Boulay, JL, Mild, G, Lowy, A, et al. Smad4 is a predictive marker for 5-fluorouracil-based chemotherapy in patients with colorectal cancer. British Journal of Cancer 2002;87:630–4.CrossRefGoogle ScholarPubMed
Valle, L, Serena-Acedo, T, Liyanarachchi, S, et al. Germline allele-specific expression of TGFBR1 confers an increased risk of colorectal cancer. Science 2008;321:1361–5.CrossRef
Bardelli, A, Parsons, DW, Silliman, N, et al. Mutational analysis of the tyrosine kinome in colorectal cancers. Science 2003;300:949.CrossRef
Sjoblom, T, Jones, S, Wood, LD, et al. The consensus coding sequences of human breast and colorectal cancers. Science 2006;314:268–74.CrossRef
Chittenden, TW, Howe, EA, Culhane, AC, et al. Functional classification analysis of somatically mutated genes in human breast and colorectal cancers. Genomics 2008;91:508–11.CrossRef
Cardoso, J, Molenaar, L, de Menezes, RX, et al. Chromosomal instability in MYH- and APC-mutant adenomatous polyps. Cancer Research 2006;66:2514–19.CrossRef
Shih, IM, Zhou, W, Goodman, SN, et al. Evidence that genetic instability occurs at an early stage of colorectal tumorigenesis. Cancer Research 2001;61:818–22.
Stoler, DL, Chen, N, Basik, M, et al. The onset and extent of genomic instability in sporadic colorectal tumor progression. Proceedings of the National Academy of Sciences USA 1999;96:15 121–6.
Cahill, DP, Lengauer, C, Yu, J, et al. Mutations of mitotic checkpoint genes in human cancers. Nature 1998;392:300–3.CrossRef
Michel, LS, Liberal, V, Chatterjee, A, et al. MAD2 haplo-insufficiency causes premature anaphase and chromosome instability in mammalian cells. Nature 2001;409:355–9.CrossRef
Doxsey, S. The centrosome: a tiny organelle with big potential. Nature Genetics 1998;20:104–6.CrossRef
Fukasawa, K. Oncogenes and tumour suppressors take on centrosomes. Nature Reviews Cancer 2007;7:911–24.CrossRef
Pihan, GA, Purohit, A, Wallace, J, et al. Centrosome defects can account for cellular and genetic changes that characterize prostate cancer progression. Cancer Research 2001;61:2212–19.
Lingle, WL, Barrett, SL, Negron, VC, et al. Centrosome amplification drives chromosomal instability in breast tumor development. Proceedings of the National Academy of Sciences USA 2002;99:1978–83.CrossRef
Zhou, M, Sheldon, S, Akel, N, Killeen, AA. Chromosomal aneuploidy in leukemic blast crisis:a potential source of error in interpretation of bone marrow engraftment analysis by VNTR amplification. Molecular Diagnosis 1999;4:153–7.CrossRef
Zhou, H, Kuang, J, Zhong, L, et al. Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation. Nature Genetics 1998;20:189–93.CrossRef
Bischoff, JR, Anderson, L, Zhu, Y, et al. A homologue of Drosophila aurora kinase is oncogenic and amplified in human colorectal cancers. EMBO Journal 1998;17:3052–65.CrossRef
Fodde, R, Kuipers, J, Rosenberg, C, et al. Mutations in the APC tumour suppressor gene cause chromosomal instability. Nature Cell Biology 2001;3:433–8.CrossRef
Kaplan, KB, Burds, AA, Swedlow, JR, et al. A role for the adenomatous polyposis coli protein in chromosome segregation. Nature Cell Biology 2001;3:429–32.CrossRef
Zhang, J, Neisa, R, Mao, Y. Oncogenic adenomatous polyposis coli mutants impair the mitotic checkpoint through direct interaction with Mad2. Molecular Biology of the Cell 2009;20:2381–8.CrossRef
Sansom, OJ, Meniel, VS, Muncan, V, et al. Myc deletion rescues Apc deficiency in the small intestine. Nature 2007;446:676–9.CrossRef
Aoki, K, Aoki, M, Sugai, M, et al. Chromosomal instability by beta-catenin/TCF transcription in APC or beta-catenin mutant cells. Oncogene 2007;26:3511–20.CrossRef
Hadjihannas, MV, Bruckner, M, Jerchow, B, et al. Aberrant Wnt/beta-catenin signaling can induce chromosomal instability in colon cancer. Proceedings of the National Academy of Sciences USA 2006;103:10 747–52.
Loeb, LA. Mutator phenotype may be required for multistage carcinogenesis. Cancer Research 1991;51:3075–9.
Markowitz, S, Wang, J, Myeroff, L, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 1995;268:1336–8.CrossRef
Parsons, R, Myeroff, LL, Liu, B, et al. Microsatellite instability and mutations of the transforming growth factor beta type II receptor gene in colorectal cancer. Cancer Research 1995;55:5548–50.
Rampino, N, Yamamoto, H, Ionov, Y, et al. Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype. Science 1997;275:967–9.CrossRef
Yamamoto, H, Sawai, H, Perucho, M. Frameshift somatic mutations in gastrointestinal cancer of the microsatellite mutator phenotype. Cancer Research 1997;57:4420–6.
Malkhosyan, S, Rampino, N, Yamamoto, H, Perucho, M. Frameshift mutator mutations. Nature 1996;382:499–500.CrossRef
Deng, G, Bell, I, Crawley, S, et al. BRAF mutation is frequently present in sporadic colorectal cancer with methylated hMLH1, but not in hereditary nonpolyposis colorectal cancer. Clinical Cancer Research 2004;10:191–5.CrossRef
Rajagopalan, H, Bardelli, A, Lengauer, C, et al. Tumorigenesis:RAF/RAS oncogenes and mismatch-repair status. Nature 2002;418:934.CrossRef
Davies, H, Bignell, GR, Cox, C, et al. Mutations of the BRAF gene in human cancer. Nature 2002;417:949–54.CrossRef
Roth, AD, Tejpar, S, Delorenzi, M, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60–00 trial. Journal of Clinical Oncology 2010;28:466–74.CrossRefGoogle ScholarPubMed
Ogino, S, Nosho, K, Kirkner, GJ, et al. CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut 2009;58:90–6.CrossRef
Samowitz, WS, Sweeney, C, Herrick, J, et al. Poor survival associated with the BRAF V600E mutation in microsatellite-stable colon cancers. Cancer Research 2005;65:6063–9.CrossRef
Georgiades, IB, Curtis, LJ, Morris, RM, Bird, CC, Wyllie, AH. Heterogeneity studies identify a subset of sporadic colorectal cancers without evidence for chromosomal or microsatellite instability. Oncogene 1999;18:7933–40.CrossRef
Issa, JP. CpG island methylator phenotype in cancer. Nature Reviews Cancer 2004;4:988–93.CrossRef
Cheng, YW, Pincas, H, Bacolod, MD, et al. CpG island methylator phenotype associates with low-degree chromosomal abnormalities in colorectal cancer. Clinical Cancer Research 2008;14:6005–13.CrossRef
Goel, A, Nagasaka, T, Arnold, CN, et al. The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology 2007;132:127–38.CrossRef
Toyota, M, Ahuja, N, Ohe-Toyota, M, et al. CpG island methylator phenotype in colorectal cancer. Proceedings of the National Academy of Sciences USA 1999;96:8681–6.CrossRef
Forster, LF, Defres, S, Goudie, DR, Baty, DU, Carey, FA. An investigation of the Peutz-Jeghers gene (LKB1) in sporadic breast and colon cancers. Journal of Clinical Pathology 2000;53:791–3.CrossRefGoogle ScholarPubMed
Sanchez-Cespedes, M, Parrella, P, Esteller, M, et al. Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Research 2002;62:3659–62.
German, J. Bloom's syndrome. XX. The first 100 cancers. Cancer Genetics and Cytogenetics 1997;93:100–6.CrossRef
Lowy, AM, Kordich, JJ, Gismondi, V, et al. Numerous colonic adenomas in an individual with Bloom's syndrome. Gastroenterology 2001;121:435–9.CrossRef
Ellis, NA, Groden, J, Ye, TZ, et al. The Bloom's syndrome gene product is homologous to RecQ helicases. Cell 1995;83:655–66.CrossRef
Bhattacharyya, S, Keirsey, J, Russell, B, et al. Telomerase-associated protein 1, HSP90, and topoisomerase IIalpha associate directly with the BLM helicase in immortalized cells using ALT and modulate its helicase activity using telomeric DNA substrates. Journal of Biological Chemistry 2009;284:14 966–77.CrossRefGoogle ScholarPubMed
Su, LK, Kinzler, KW, Vogelstein, B, et al. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science 1992;256:668–70.CrossRef
Moser, AR, Pitot, HC, Dove, WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science 1990;247:322–4.CrossRef
de Wind, N, Dekker, M, Berns, A, Radman, M, te Riele, H. Inactivation of the mouse Msh2 gene results in mismatch repair deficiency, methylation tolerance, hyper-recombination, and predisposition to cancer. Cell 1995;82:321–30.CrossRef
Edelmann, W, Yang, K, Umar, A, et al. Mutation in the mismatch repair gene Msh6 causes cancer susceptibility. Cell 1997;91:467–77.CrossRef
Edelmann, W, Cohen, PE, Kane, M, et al. Meiotic pachytene arrest in MLH1-deficient mice. Cell 1996;85:1125–34.CrossRef
Baker, SM, Plug, AW, Prolla, TA, et al. Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over. Nature Genetics 1996;13:336–42.CrossRef
Prolla, TA, Baker, SM, Harris, AC, et al. Tumour susceptibility and spontaneous mutation in mice deficient in Mlh1, Pms1 and Pms2 DNA mismatch repair. Nature Genetics 1998;18:276–9.CrossRef
Baker, SM, Harris, AC, Tsao, JL, et al. Enhanced intestinal adenomatous polyp formation in Pms2-/-;Min mice. Cancer Research 1998;58:1087–9.
Edelmann, W, Yang, K, Kuraguchi, M, et al. Tumorigenesis in Mlh1 and Mlh1/Apc1638N mutant mice. Cancer Research 1999;59:1301–7.
Kuraguchi, M, Yang, K, Wong, E, et al. The distinct spectra of tumor-associated Apc mutations in mismatch repair-deficient Apc1638N mice define the roles of MSH3 and MSH6 in DNA repair and intestinal tumorigenesis. Cancer Research 2001;61:7934–42.
Reitmair, AH, Cai, JC, Bjerknes, M, et al. MSH2 deficiency contributes to accelerated APC-mediated intestinal tumorigenesis. Cancer Research 1996;56:2922–6.
Wei, K, Kucherlapati, R, Edelmann, W. Mouse models for human DNA mismatch-repair gene defects. Trends in Molecular Medicine 2002;8:346–53.CrossRef
Perchiniak, E, Groden, J. Mouse models of intestinal cancer. In: John D. Potter NML, editor. Genetics of Colorectal Cancer. New York:Springer;2009.
Efficacy of adjuvant fluorouracil and folinic acid in B2 colon cancer. International Multicentre Pooled Analysis of B2 Colon Cancer Trials (IMPACT B2) Investigators. Journal of Clinical Oncology 1999;17:1356–63.CrossRef
Gill, S, Loprinzi, CL, Sargent, DJ, et al. Pooled analysis of fluorouracil-based adjuvant therapy for stage II and III colon cancer:who benefits and by how much?Journal of Clinical Oncology 2004;22:1797–806.CrossRefGoogle ScholarPubMed
Mamounas, E, Wieand, S, Wolmark, N, et al. Comparative efficacy of adjuvant chemotherapy in patients with Dukes’ B versus Dukes’ C colon cancer:results from four National Surgical Adjuvant Breast and Bowel Project adjuvant studies (C-01, C-02, C-03, and C-04). Journal of Clinical Oncology 1999;17:1349–55.CrossRefGoogle Scholar
Van Cutsem, E, Twelves, C, Cassidy, J, et al. Oral capecitabine compared with intravenous fluorouracil plus leucovorin in patients with metastatic colorectal cancer:results of a large Phase III study. Journal of Clinical Oncology 2001;19:4097–106.CrossRefGoogle ScholarPubMed
Hoff, PM, Ansari, R, Batist, G, et al. Comparison of oral capecitabine versus intravenous fluorouracil plus leucovorin as first-line treatment in 605 patients with metastatic colorectal cancer:results of a randomized Phase III study. Journal of Clinical Oncology 2001;19:2282–92.CrossRefGoogle ScholarPubMed
Twelves, C, Wong, A, Nowacki, MP, et al. Capecitabine as adjuvant treatment for stage III colon cancer. New England Journal of Medicine 2005;352:2696–704.CrossRefGoogle ScholarPubMed
Andre, T, Boni, C, Mounedji-Boudiaf, L, et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. New England Journal of Medicine 2004;350:2343–51.CrossRefGoogle ScholarPubMed
Douillard, JY, Cunningham, D, Roth, AD, et al. Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer:a multicentre randomised trial. Lancet 2000;355:1041–7.CrossRef
Saltz, LB, Cox, JV, Blanke, C, et al. Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. Irinotecan Study Group. New England Journal of Medicine 2000;343:905–14.CrossRefGoogle ScholarPubMed
Cunningham, D, Humblet, Y, Siena, S, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. New England Journal of Medicine 2004;351:337–45.CrossRefGoogle ScholarPubMed
Van Cutsem, E, Peeters, M, Siena, S, et al. Open-label Phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. Journal of Clinical Oncology 2007;25:1658–64.CrossRefGoogle ScholarPubMed
Allegra, CJ, Jessup, JM, Somerfield, MR, et al. American Society of Clinical Oncology provisional clinical opinion:testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy. Journal of Clinical Oncology 2009;27:2091–6.CrossRefGoogle Scholar
Jhawer, M, Goel, S, Wilson, AJ, et al. PIK3CA mutation/PTEN expression status predicts response of colon cancer cells to the epidermal growth factor receptor inhibitor cetuximab. Cancer Research 2008;68:1953–61.CrossRef
Zhang, C, Bollag, G. Scaffold-based design of kinase inhibitors for cancer therapy. Current Opinion in Genetic Development 2010;20:79–86.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Colon and rectal cancer
    • By Erin M. Perchiniak, Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA, Joanna Groden, Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.048
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Colon and rectal cancer
    • By Erin M. Perchiniak, Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA, Joanna Groden, Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.048
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Colon and rectal cancer
    • By Erin M. Perchiniak, Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA, Joanna Groden, Department of Molecular Virology, Immunology, and Medical Genetics,he Ohio State University College of Medicine, Columbus, OH, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.048
Available formats
×