Skip to main content

Advertisement

Log in

Role of Apoptosis in Colon Cancer Biology, Therapy, and Prevention

  • Molecular Biology (S Anant, Section Editor)
  • Published:
Current Colorectal Cancer Reports

Abstract

Deregulation of apoptosis is a hallmark of human cancer and contributes to therapeutic resistance. Recent advances in cancer genomics have revealed a myriad of alterations in key pathways that directly or indirectly increase tumor cell survival. This review outlines the pathways of apoptosis in mammalian cells, and highlights the common alterations of apoptosis regulators found in colon cancer, the role of apoptosis, and underlying mechanisms in colon cancer treatment and prevention, including recent advances in investigational agents, such as kinase inhibitors, proteasome inhibitors, heat shock protein 90 inhibitors, BH3 mimetics, tumor necrosis factor related apoptosis-inducing ligand, and inhibitor of apoptosis protein antagonists. The topics also include novel concepts as well as opportunities and challenges for drug discovery and combination therapy by exploring cancer-specific genetic defects, and therefore selective induction of apoptosis in cancer cells. Although the emphasis is on colon cancer, the main theme and many of the aspects are applicable to other solid tumors.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1

Similar content being viewed by others

References

Papers of particular interests, published recently, have been highlighted as • Of importance •• Of major importance

  1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  PubMed  CAS  Google Scholar 

  2. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012;62(1):10–29.

    Article  PubMed  Google Scholar 

  3. Ashkenazi A. Directing cancer cells to self-destruct with pro-apoptotic receptor agonists. Nat Rev Drug Discov. 2008;7(12):1001–12.

    Article  PubMed  CAS  Google Scholar 

  4. Bagnoli M, Canevari S, Mezzanzanica D. Cellular FLICE-inhibitory protein (c-FLIP) signalling: a key regulator of receptor-mediated apoptosis in physiologic context and in cancer. Int J Biochem Cell Biol. 2010;42(2):210–3.

    Article  PubMed  CAS  Google Scholar 

  5. Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 2007;26(9):1324–37.

    Article  PubMed  CAS  Google Scholar 

  6. Vousden KH, Prives C. Blinded by the light: the growing complexity of p53. Cell. 2009;137(3):413–31.

    Article  PubMed  CAS  Google Scholar 

  7. Joza N, Susin SA, Daugas E, et al. Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death. Nature. 2001;410(6828):549–54.

    Article  PubMed  CAS  Google Scholar 

  8. Wang X, Yang C, Chai J, Shi Y, Xue D. Mechanisms of AIF-mediated apoptotic DNA degradation in Caenorhabditis elegans. Science. 2002;298(5598):1587–92.

    Article  PubMed  CAS  Google Scholar 

  9. •• Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz Jr LA, Kinzler KW. Cancer genome landscapes. Science. 2013;339(6127):1546–58. This review summarizes key genetic alterations from recent genomics studies on human cancers, including colorectal cancer.

    Article  PubMed  CAS  Google Scholar 

  10. Yu J, Zhang L. The transcriptional targets of p53 in apoptosis control. Biochem Biophys Res Commun. 2005;331(3):851–8.

    Article  PubMed  CAS  Google Scholar 

  11. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–99.

    Article  PubMed  CAS  Google Scholar 

  12. •• McCubrey JA, Steelman LS, Kempf CR, et al. Therapeutic resistance resulting from mutations in Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR signaling pathways. J Cell Physiol. 2011;226(11):2762–81. This review summarizes key findings on the therapeutic responses to various targeted agents and mutations in key survival pathways in human cancer, including colorectal cancer.

    Article  PubMed  CAS  Google Scholar 

  13. 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(5302):967–9.

    Article  PubMed  CAS  Google Scholar 

  14. Ewings KE, Wiggins CM, Cook SJ. Bim and the pro-survival Bcl-2 proteins: opposites attract. ERK repels. Cell Cycle. 2007;6(18):2236–40.

    Article  PubMed  CAS  Google Scholar 

  15. Dudgeon C, Wang P, Sun X, et al. PUMA induction by FoxO3a mediates the anticancer activities of the broad-range kinase inhibitor UCN-01. Mol Cancer Ther. 2010;9(11):2893–902.

    Article  PubMed  CAS  Google Scholar 

  16. Ming L, Sakaida T, Yue W, Jha A, Zhang L, Yu J. Sp1 and p73 activate PUMA following serum starvation. Carcinogenesis. 2008;29:1878–84.

    Article  PubMed  CAS  Google Scholar 

  17. Sun Q, Ming L, Thomas SM, et al. PUMA mediates EGFR tyrosine kinase inhibitor-induced apoptosis in head and neck cancer cells. Oncogene. 2009;18(28):2348–57.

    Article  Google Scholar 

  18. •• Qiu W, Carson-Walter EB, Kuan SF, Zhang L, Yu J. PUMA suppresses intestinal tumorigenesis in mice. Cancer Res. 2009;69(12):4999–5006. This study showed that blocked apoptosis increases intestinal cancer initiation and invasiveness in mice.

    Article  PubMed  CAS  Google Scholar 

  19. Johnstone RW, Frew AJ, Smyth MJ. The TRAIL apoptotic pathway in cancer onset, progression and therapy. Nat Rev Cancer. 2008;8(10):782–98.

    Article  PubMed  CAS  Google Scholar 

  20. Fulda S, Vucic D. Targeting IAP proteins for therapeutic intervention in cancer. Nat Rev Drug Discov. 2012;11(2):109–24.

    Article  PubMed  CAS  Google Scholar 

  21. Wilson TR, McLaughlin KM, McEwan M, et al. c-FLIP: a key regulator of colorectal cancer cell death. Cancer Res. 2007;67(12):5754–62.

    Article  PubMed  CAS  Google Scholar 

  22. Xiang G, Wen X, Wang H, Chen K, Liu H. Expression of X-linked inhibitor of apoptosis protein in human colorectal cancer and its correlation with prognosis. J Surg Oncol. 2009;100(8):708–12.

    Article  PubMed  CAS  Google Scholar 

  23. Krajewska M, Kim H, Kim C, et al. Analysis of apoptosis protein expression in early-stage colorectal cancer suggests opportunities for new prognostic biomarkers. Clin Cancer Res. 2005;11(15):5451–61.

    Article  PubMed  CAS  Google Scholar 

  24. Endo K, Kohnoe S, Watanabe A, et al. Clinical significance of Smac/DIABLO expression in colorectal cancer. Oncol Rep. 2009;21(2):351–5.

    PubMed  Google Scholar 

  25. Qiu W, Liu H, Sebastini A, et al. An apoptosis-independent role of SMAC in tumor suppression. Oncogene. 2013;32(19):2380–9.

    Article  PubMed  CAS  Google Scholar 

  26. Yu J, Zhang L. Apoptosis in human cancer cells. Curr Opin Oncol. 2004;16(1):19–24.

    Article  PubMed  Google Scholar 

  27. Martini M, Vecchione L, Siena S, Tejpar S, Bardelli A. Targeted therapies: how personal should we go? Nat Rev Clin Oncol. 2012;9(2):87–97.

    Article  CAS  Google Scholar 

  28. Yu J, Zhang L. PUMA, a potent killer with or without p53. Oncogene. 2008;27 Suppl 1:S71–83.

    Article  PubMed  CAS  Google Scholar 

  29. Qiu W, Carson-Walter EB, Liu H, et al. PUMA regulates intestinal progenitor cell radiosensitivity and gastrointestinal syndrome. Cell Stem Cell. 2008;2(6):576–83.

    Article  PubMed  CAS  Google Scholar 

  30. Yu H, Shen H, Yuan Y, et al. Deletion of Puma protects hematopoietic stem cells and confers long-term survival in response to high-dose gamma-irradiation. Blood. 2010;115(17):3472–80.

    Article  PubMed  CAS  Google Scholar 

  31. Olsson M, Vakifahmetoglu H, Abruzzo PM, Hogstrand K, Grandien A, Zhivotovsky B. DISC-mediated activation of caspase-2 in DNA damage-induced apoptosis. Oncogene. 2009;28(18):1949–59.

    Article  PubMed  CAS  Google Scholar 

  32. Bunz F, Hwang PM, Torrance C, et al. Disruption of p53 in human cancer cells alters the responses to therapeutic agents. J Clin Invest. 1999;104(3):263–9.

    Article  PubMed  CAS  Google Scholar 

  33. Moll UM, Wolff S, Speidel D, Deppert W. Transcription-independent pro-apoptotic functions of p53. Curr Opin Cell Biol. 2005;17(6):631–6.

    Article  PubMed  CAS  Google Scholar 

  34. Dotsch V, Bernassola F, Coutandin D, Candi E, Melino G. p63 and p73, the ancestors of p53. Cold Spring Harb Perspect Biol. 2010;2(9):a004887.

    Article  PubMed  CAS  Google Scholar 

  35. Gudkov AV, Komarova EA. Pathologies associated with the p53 response. Cold Spring Harb Perspect Biol. 2010;2(7):a001180.

    Article  PubMed  Google Scholar 

  36. Gong Y, Somwar R, Politi K, et al. Induction of BIM is essential for apoptosis triggered by EGFR kinase inhibitors in mutant EGFR-dependent lung adenocarcinomas. PLoS Med. 2007;4(10):e294.

    Article  PubMed  Google Scholar 

  37. Costa DB, Halmos B, Kumar A, et al. BIM mediates EGFR tyrosine kinase inhibitor-induced apoptosis in lung cancers with oncogenic EGFR mutations. PLoS Med. 2007;4(10):1669–79. discussion 1680.

    Article  PubMed  CAS  Google Scholar 

  38. Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science. 2004;305(5687):1163–7.

    Article  PubMed  CAS  Google Scholar 

  39. Selvakumaran M, Yao KS, Feldman MD, O'Dwyer PJ. Antitumor effect of the angiogenesis inhibitor bevacizumab is dependent on susceptibility of tumors to hypoxia-induced apoptosis. Biochem Pharmacol. 2008;75(3):627–38.

    Article  PubMed  CAS  Google Scholar 

  40. Mizobe T, Ogata Y, Murakami H, et al. Efficacy of the combined use of bevacizumab and irinotecan as a postoperative adjuvant chemotherapy in colon carcinoma. Oncol Rep. 2008;20(3):517–23.

    PubMed  CAS  Google Scholar 

  41. Kargi A, Yalcin AD, Erin N, Savas B, Polat HH, Gorczynski RM. IL8 and serum soluble TRAIL levels following anti-VEGF monoclonal antibody treatment in patients with metastatic colon cancer. Clin Lab. 2012;58(5–6):501–5.

    PubMed  CAS  Google Scholar 

  42. Bisgin A, Kargi A, Yalcin AD, et al. Increased serum sTRAIL levels were correlated with survival in bevacizumab-treated metastatic colon cancer. BMC Cancer. 2012;12:58.

    Article  PubMed  CAS  Google Scholar 

  43. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356(2):125–34.

    Article  PubMed  CAS  Google Scholar 

  44. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–90.

    Article  PubMed  CAS  Google Scholar 

  45. Yu C, Bruzek LM, Meng XW, et al. The role of Mcl-1 downregulation in the proapoptotic activity of the multikinase inhibitor BAY 43–9006. Oncogene. 2005;24(46):6861–9.

    Article  PubMed  CAS  Google Scholar 

  46. Liu L, Cao Y, Chen C, et al. Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 2006;66(24):11851–8.

    Article  PubMed  CAS  Google Scholar 

  47. • Dudgeon C, Peng R, Wang P, Sebastiani A, Yu J, Zhang L. Inhibiting oncogenic signaling by sorafenib activates PUMA via GSK3β and NF-κB to suppress tumor cell growth. Oncogene. 2012;31:4848–58. This study showed that sorafenib induces apoptosis in colon cancer cells by inducing the BH3-only protein PUMA through the NF-κB pathway.

    Article  PubMed  CAS  Google Scholar 

  48. Zhang W, Konopleva M, Ruvolo VR, et al. Sorafenib induces apoptosis of AML cells via Bim-mediated activation of the intrinsic apoptotic pathway. Leukemia. 2008;22(4):808–18.

    Article  PubMed  CAS  Google Scholar 

  49. Mao M, Tian F, Mariadason JM, et al. Resistance to BRAF inhibition in BRAF-mutant colon cancer can be overcome with PI3K inhibition or demethylating agents. Clin Cancer Res. 2013;19(3):657–67.

    Article  PubMed  CAS  Google Scholar 

  50. Greger JG, Eastman SD, Zhang V, et al. Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol Cancer Ther. 2012;11(4):909–20.

    Article  PubMed  CAS  Google Scholar 

  51. • Prahallad A, Sun C, Huang S, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483(7387):100–3. This study showed that insensitivity of colon cancer to the BRAF inhibitor vemurafenib is caused by feedback activation of EGFR and can be overcome by EGFR inhibitors.

    Article  PubMed  CAS  Google Scholar 

  52. Sun J, Sun Q, Brown MF, et al. The multi-targeted kinase inhibitor sunitinib induces apoptosis in colon cancer cells via PUMA. PLoS One. 2012;7(8):e43158.

    Article  PubMed  CAS  Google Scholar 

  53. • Zheng X, He K, Zhang L, Yu J. Crizotinib induces PUMA-dependent apoptosis in colon cancer cells. Mol Cancer Ther. 2013;12(5):777–86. This study showed that the MET/anaplastic lymphoma kinase inhibitor crizotinib promotes PUMA-dependent apoptosis through both p53-dependent and p53-independent mechanisms in colon cancer cells.

    Article  PubMed  CAS  Google Scholar 

  54. Yu J, Tiwari S, Steiner P, Zhang L. Differential apoptotic response to the proteasome inhibitor bortezomib (VELCADETM, PS-341) in Bax-deficient and p21-deficient colon cancer cells. Cancer Biol Ther. 2003;2(6):694–9.

    PubMed  CAS  Google Scholar 

  55. Milano A, Iaffaioli RV, Caponigro F. The proteasome: a worthwhile target for the treatment of solid tumours? Eur J Cancer. 2007;43(7):1125–33.

    Article  PubMed  CAS  Google Scholar 

  56. Neckers L, Workman P. Hsp90 molecular chaperone inhibitors: are we there yet? Clin Cancer Res. 2012;18(1):64–76.

    Article  PubMed  CAS  Google Scholar 

  57. Vaseva AV, Yallowitz AR, Marchenko ND, Xu S, Moll UM. Blockade of Hsp90 by 17AAG antagonizes MDMX and synergizes with nutlin to induce p53-mediated apoptosis in solid tumors. Cell Death Dis. 2011;2:e156.

    Article  PubMed  CAS  Google Scholar 

  58. Lippman SM. The future of molecular-targeted cancer chemoprevention. Gastroenterology. 2008;135(6):1834–41.

    Article  PubMed  CAS  Google Scholar 

  59. Rao CV, Reddy BS. NSAIDs and chemoprevention. Curr Cancer Drug Targets. 2004;4(1):29–42.

    Article  PubMed  CAS  Google Scholar 

  60. Gupta RA, Dubois RN. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2. Nat Rev Cancer. 2001;1(1):11–21.

    Article  PubMed  CAS  Google Scholar 

  61. Sun SY, Hail Jr N, Lotan R. Apoptosis as a novel target for cancer chemoprevention. J Natl Cancer Inst. 2004;96(9):662–72.

    Article  PubMed  CAS  Google Scholar 

  62. Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B. Role of BAX in the apoptotic response to anticancer agents. Science. 2000;290(5493):989–92.

    Article  PubMed  CAS  Google Scholar 

  63. Kohli M, Yu J, Seaman C, et al. SMAC/Diablo-dependent apoptosis induced by nonsteroidal antiinflammatory drugs (NSAIDs) in colon cancer cells. Proc Natl Acad Sci U S A. 2004;101(48):16897–902.

    Article  PubMed  CAS  Google Scholar 

  64. Bank A, Wang P, Du C, Yu J, Zhang L. SMAC mimetics sensitize nonsteroidal anti-inflammatory drug-induced apoptosis by promoting caspase-3-mediated cytochrome c release. Cancer Res. 2008;68(1):276–84.

    Article  PubMed  CAS  Google Scholar 

  65. •• Qiu W, Wang X, Leibowitz B, et al. Chemoprevention by nonsteroidal anti-inflammatory drugs eliminates oncogenic intestinal stem cells via SMAC-dependent apoptosis. Proc Natl Acad Sci U S A. 2010;107(46):20027–32. This study used a mouse tumor model to show that intestinal stem cells containing gatekeeper mutations are the key target for chemoprevention.

    Article  PubMed  CAS  Google Scholar 

  66. Meyskens Jr FL, McLaren CE, Pelot D, et al. Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: a randomized placebo-controlled, double-blind trial. Cancer Prev Res (Phila). 2008;1(1):32–8.

    Article  CAS  Google Scholar 

  67. Barker N, Ridgway RA, van Es JH, et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 2008;457(7229):608–11.

    Article  PubMed  Google Scholar 

  68. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132(1):27–42.

    Article  PubMed  CAS  Google Scholar 

  69. Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010;22(2):124–31.

    Article  PubMed  CAS  Google Scholar 

  70. Degenhardt K, Mathew R, Beaudoin B, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 2006;10(1):51–64.

    Article  PubMed  CAS  Google Scholar 

  71. White E, DiPaola RS. The double-edged sword of autophagy modulation in cancer. Clin Cancer Res. 2009;15(17):5308–16.

    Article  PubMed  Google Scholar 

  72. Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 2007;8(9):741–52.

    Article  PubMed  CAS  Google Scholar 

  73. Oberstein A, Jeffrey PD, Shi Y. Crystal structure of the Bcl-XL-beclin 1 peptide complex: beclin 1 is a novel BH3-only protein. J Biol Chem. 2007;282(17):13123–32.

    Article  PubMed  CAS  Google Scholar 

  74. Pattingre S, Tassa A, Qu X, et al. Bcl-2 antiapoptotic proteins inhibit beclin 1-dependent autophagy. Cell. 2005;122(6):927–39.

    Article  PubMed  CAS  Google Scholar 

  75. Yousefi S, Perozzo R, Schmid I, et al. Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol. 2006;8(10):1124–32.

    Article  PubMed  CAS  Google Scholar 

  76. Djavaheri-Mergny M, Maiuri MC, Kroemer G. Cross talk between apoptosis and autophagy by caspase-mediated cleavage of beclin 1. Oncogene. 2010;29(12):1717–9.

    Article  PubMed  CAS  Google Scholar 

  77. Li H, Wang P, Sun Q, et al. Following cytochrome c release, autophagy is inhibited during chemotherapy-induced apoptosis by caspase 8-mediated cleavage of beclin 1. Cancer Res. 2011;71(10):3625–34. This study used a knock-in approach to demonstrate a cross-talk mechanism between chemotherapy-induced apoptosis and autophagy.

    Article  PubMed  CAS  Google Scholar 

  78. Wang L, Du F, Wang X. TNF-alpha induces two distinct caspase-8 activation pathways. Cell. 2008;133(4):693–703.

    Article  PubMed  CAS  Google Scholar 

  79. Petersen SL, Wang L, Yalcin-Chin A, et al. Autocrine TNFα signaling renders human cancer cells susceptible to Smac-mimetic-induced apoptosis. Cancer Cell. 2007;12(5):445–56.

    Article  PubMed  CAS  Google Scholar 

  80. Kaiser WJ, Upton JW, Long AB, et al. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature. 2011;471(7338):368–72.

    Article  PubMed  CAS  Google Scholar 

  81. •• Oberst A, Dillon CP, Weinlich R, et al. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature. 2011;471(7338):363–7. This study provided mechanistic insight into how apoptosis and programmed necrosis are co-regulated, sharing components of the extrinsic apoptotic pathway.

    Article  PubMed  CAS  Google Scholar 

  82. Zhang L, Ming L, Yu J. BH3 mimetics to improve cancer therapy; mechanisms and examples. Drug Resist Updates. 2007;10(6):207–17.

    Article  CAS  Google Scholar 

  83. Oltersdorf T, Elmore SW, Shoemaker AR, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005;435(7042):677–81.

    Article  PubMed  CAS  Google Scholar 

  84. van Delft MF, Wei AH, Mason KD, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell. 2006;10(5):389–99.

    Article  PubMed  Google Scholar 

  85. Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68(9):3421–8.

    Article  PubMed  CAS  Google Scholar 

  86. Wang S. Design of small-molecule Smac mimetics as IAP antagonists. Curr Top Microbiol Immunol. 2011;348:89–113.

    Article  PubMed  CAS  Google Scholar 

  87. Sun Q, Zheng X, Zhang L, Yu J. Smac modulates chemosensitivity in head and neck cancer cells through the mitochondrial apoptotic pathway. Clin Cancer Res. 2011;17(8):2361–72.

    Article  PubMed  CAS  Google Scholar 

  88. Hartwell LH, Szankasi P, Roberts CJ, Murray AW, Friend SH. Integrating genetic approaches into the discovery of anticancer drugs. Science. 1997;278(5340):1064–8.

    Article  PubMed  CAS  Google Scholar 

  89. Luo J, Emanuele MJ, Li D, et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell. 2009;137(5):835–48.

    Article  PubMed  CAS  Google Scholar 

  90. Scholl C, Frohling S, Dunn IF, et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell. 2009;137(5):821–34.

    Article  PubMed  CAS  Google Scholar 

  91. Singh A, Sweeney MF, Yu M, et al. TAK1 inhibition promotes apoptosis in KRAS-dependent colon cancers. Cell. 2012;148(4):639–50.

    Article  PubMed  CAS  Google Scholar 

  92. Steckel M, Molina-Arcas M, Weigelt B, et al. Determination of synthetic lethal interactions in KRAS oncogene-dependent cancer cells reveals novel therapeutic targeting strategies. Cell Res. 2012;22(8):1227–45.

    Article  PubMed  CAS  Google Scholar 

  93. • Corcoran RB, Cheng KA, Hata AN, et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. Cancer Cell. 2013;23(1):121–8. This study showed that co-targeting Bcl-XL and MEK triggered a synthetic lethal interaction in KRAS mutant colorectal tumors.

    Article  PubMed  CAS  Google Scholar 

  94. Yang D, Liu H, Goga A, Kim S, Yuneva M, Bishop JM. Therapeutic potential of a synthetic lethal interaction between the MYC proto-oncogene and inhibition of aurora-B kinase. Proc Natl Acad Sci U S A. 2010;107(31):13836–41.

    Article  PubMed  CAS  Google Scholar 

  95. Wang Y, Quon KC, Knee DA, Nesterov A, Kraft AS. RAS, MYC, and sensitivity to tumor necrosis factor-alpha-related apoptosis-inducing ligand-induced apoptosis. Cancer Res. 2005;65(4):1615–6. author reply 1616–7.

    Article  PubMed  CAS  Google Scholar 

  96. Nieminen AI, Partanen JI, Hau A, Klefstrom J. c-Myc primed mitochondria determine cellular sensitivity to TRAIL-induced apoptosis. EMBO J. 2007;26(4):1055–67.

    Article  PubMed  CAS  Google Scholar 

  97. Ashworth A. A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J Clin Oncol. 2008;26(22):3785–90.

    Article  PubMed  CAS  Google Scholar 

  98. Martin SA, McCabe N, Mullarkey M, et al. DNA polymerases as potential therapeutic targets for cancers deficient in the DNA mismatch repair proteins MSH2 or MLH1. Cancer Cell. 2010;17(3):235–48.

    Article  PubMed  CAS  Google Scholar 

  99. Martin SA, Hewish M, Sims D, Lord CJ, Ashworth A. Parallel high-throughput RNA interference screens identify PINK1 as a potential therapeutic target for the treatment of DNA mismatch repair-deficient cancers. Cancer Res. 2011;71(5):1836–48.

    Article  PubMed  CAS  Google Scholar 

  100. • Chan DA, Giaccia AJ. Harnessing synthetic lethal interactions in anticancer drug discovery. Nat Rev Drug Discov. 2011;10(5):351–64. This review discusses the potential use of synthetic lethal interactions for discovery of new anticancer drugs.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

Research in the authors’ laboratories is supported by National Institutes of Health grants CA106348, CA121105, and CA172136 and the V Foundation for Cancer Research (L.Z.), and American Cancer Society grant RSG-10-124-10-CCE and National Institutes of Health grants CA129829 and U01DK085570 (J.Y.).

Compliance with Ethics Guidelines

Conflict of Interest

Lin Zhang has received compensation for US patents 5,695,937, 5,866,330, 6,383,743, 6,746,845, and 6,333,152, and has received royalties from Johns Hopkins University for the preparation of genetically engineered cells.

Jian Yu has received compensation for US patent WO 02/064790, and has received royalties from Johns Hopkins University for the preparation of genetically engineered cells.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jian Yu.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Zhang, L., Yu, J. Role of Apoptosis in Colon Cancer Biology, Therapy, and Prevention. Curr Colorectal Cancer Rep 9, 331–340 (2013). https://doi.org/10.1007/s11888-013-0188-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11888-013-0188-z

Keywords

Navigation