Open access peer-reviewed chapter

Autophagy as a Therapeutic Target in Gastrointestinal Cancer

Written By

Michiko Shintani

Submitted: 25 December 2014 Reviewed: 15 September 2015 Published: 16 December 2015

DOI: 10.5772/61523

From the Edited Volume

Cell Death - Autophagy, Apoptosis and Necrosis

Edited by Tobias M. Ntuli

Chapter metrics overview

2,015 Chapter Downloads

View Full Metrics

Abstract

Autophagy is a bulk protein and organelle degradation system and is an important homeostatic cellular recycling mechanism. The following kinds are the three types of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. In general, the term “autophagy” indicates macroautophagy. Autophagy is mediated by double-membrane-bound structures called autophagosomes. During the autophagic process, cytoplasmic components are sequestered and engulfed by autophagosomes. Autophagosomes then fuse with lysosomes to form autolysosomes where the sequestered components are digested by lysosomal hydrolases. Microtubule-associated protein 1 light chain 3 (LC3) is an autophagosomal ortholog of the yeast protein ATG8. Autophagy stimulates the upregulation of LC3 expression, and a cytosolic form of LC3 (LC3-I) is conjugated to phosphatidylethanolamine to form LC3-II which is recruited to autophagosomal membranes. Subsequently, LC3-II is degraded by lysosomal hydrolases after the fusion of autophagosomes with lysosomes. Therefore, LC3 is a specific marker of autophagosome formation. Additionally, beclin 1, the mammalian ortholog of the yeast protein ATG6, has been known to play a crucial role in autophagy. Beclin 1 acts in conjunction with the phosphoinositide-3 kinase pathway to enhance the formation of the autophagic vacuole.

Keywords

  • Gastric carcinoma
  • colorectal carcinoma
  • immunohistochemistry
  • cancer therapy

1. Introduction

Autophagy is a bulk protein and organelle degradation system and is an important homeostatic cellular recycling mechanism. The following are the three types of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. In general, macroautophagy is believed to be the major type of autophagy. Autophagy is mediated by double-membrane-bound structures called autophagosomes [13]. During the autophagic process, cytoplasmic components are sequestered and engulfed by autophagosomes. Autophagosomes then fuse with lysosomes to form autolysosomes where the sequestered components are digested by lysosomal hydrolases. Microtubule-associated protein 1 light chain 3 (LC3) is an autophagosomal ortholog of the yeast protein ATG8. LC3 exists in two forms, LC3-I and LC3-II. LC3-I is localized in the cytoplasm. Autophagy stimulates the upregulation of LC3 expression, and LC3-I conjugates with phosphatidylethanolamine to form LC3-II. LC3-II binds to autophagosomes and it is degraded by lysosomal hydrolases after the fusion of autophagosomes with lysosomes [46]. Therefore, LC3 is a specific marker of autophagosome formation. The autophagic pathway includes several phases: initiation, vesicle elongation, maturation, fusion, and degradation (Figure 1). Recent studies have suggested that additional membranes are derived from the Golgi complex, mitochondria, and plasma membrane; however, this phenomenon has not been confirmed [12, 710].

Figure 1.

Cellular mechanism of autophagy. The autophagic pathway includes several phases: initiation, vesicle elongation, maturation, fusion, and degradation.

Current studies are examining the molecular regulation and function of autophagy. Additionally, autophagy is believed to play a role in various diseases such as cancer, infectious diseases, cardiovascular diseases, metabolic diseases, pulmonary diseases, and neurodegenerative disorders [1120]. Recently, in clinical trials, several autophagic inhibitors, including hydroxychloroquine and chloroquine, have been examined as targets in diseases. In cancer, these autophagic components are being studied to enhance chemotherapeutic efficacy. Thus, autophagy is now an important and widely studied topic in human health and disease [1115].

Advertisement

2. Detection of autophagy using immunohistochemistry

The role of autophagy in cancer development and progression has been studied by investigating autophagy-related proteins, LC3, beclin 1, and p62 using immunohistochemistry [2123].

2.1. LC3

LC3 is an autophagosomal ortholog of the yeast protein ATG8 and is a specific marker of autophagosome formation. LC3-I is localized in the cytoplasm whereas LC3-II binds to autophagosomes. LC3 is presently used as an autophagy marker [2126].

2.2. Beclin 1

Beclin 1 is the mammalian homolog of the yeast protein ATG6 and it has a central role in autophagy. The expression of beclin 1 has been reported in tumors such as breast, ovarian, prostate, lung, brain, stomach, and colorectal tumors. Beclin 1 may play a role in the tumorigenesis and/or progression of human cancers. However, beclin 1 has several physiological functions other than autophagy [2122, 2731].

2.3. p62

p62/SQSTM1 (p62) is an autophagy substrate protein which accumulates in autophagy-deficient cells after metabolic stress. p62 accumulation leads to mitochondrial damage, oxidative stress, and DNA damage. Additionally, p62 accumulation has been strongly suggested to result in cancer development [23, 32].

Advertisement

3. Autophagy and tumor suppression/progression

Recently, several studies have suggested that dysregulation of autophagy plays a critical role in tumorigenesis. Liang et al. (1999) reported that beclin 1 can inhibit tumorigenesis and that its levels decrease in human breast carcinoma [27]. Qu et al. (2003) presented the genetic evidence for the role of autophagy genes in tumor suppression. They suggested that beclin 1 is a haploinsufficient tumor-suppressor gene, and that mutation of beclin 1 or other autophagy genes may contribute to the pathogenesis of human cancers [28]. Autophagy may prevent normal cells from developing into tumor cells; however, it may also protect cancer cells by providing nutritional support. Yang et al. (2011) investigated the effects of autophagy in stressed and unstressed colon cancer cells. They found that, in unstressed cells, the inhibition of autophagy was associated with a significant growth advantage but, in biologically stressed cells, the inhibition of autophagy markedly reduced cell viability compared to that observed in controls. Therefore, they suggested that autophagy has a dual role in colon cancer cells; it is pro-survival under biological stress and pro-death under normal conditions [33].

Li et al. (2009) reported the dual role of autophagy in colon cancer. They found that the inhibition of autophagy enhances 5-fluorouracil-induced colon cancer cell apoptosis and improves the chemotherapeutic effect of 5-fluorouracil. This result indicates that autophagy plays a role in protecting some cells from chemotherapy-induced death [34].

Autophagy may play an important role in maintaining normal cellular homeostasis and may prevent normal cells from developing into cancer cells. However, autophagy is a cellular recycling mechanism and is active during metabolic stress [35–36]. Additionally, it may prevent cell death in tumor cells (apoptosis or autophagic cell death). Thus, autophagy has a role in both the suppression of cancer initiation and the promotion of cancer growth [27–28, 33–34].

Advertisement

4. Autophagy as a therapeutic target in gastrointestinal carcinomas

Autophagy has a role in both tumor promotion and tumor suppression. In cancer therapy, using the effect of autophagy, cell survival is inhibited or cell death is promoted by inhibition or induction [3741].

Recently, autophagy has been used for cancer therapy in aggressive cancers. Particularly, a clinical trial of cancer therapy involving the combination of an autophagy regulator with conventional anticancer agents or radiation therapy has been performed. In colorectal cancer, a combination of an autophagy inhibitor (hydroxychloroquine), oxaliplatin, leucovorin, 5-fluorouracil, and bevacizumab has been used. Several studies have reported the influence of apoptosis and autophagy on each other in cancer cells after chemotherapy [4245].

Therapeutic agents in gastrointestinal cancer are summarized in Tables 1 and 2 [4657].

Advertisement

5. Conclusion

Autophagy, an intracellular process involved in removing and recycling cellular components, may play a role in both protecting and promoting cancer cell death under different stress situations. The role of autophagy in tumorigenesis is controversial, because it can either protect or promote cell death. Recently, autophagy has been used in cancer therapy for aggressive cancers. Particularly, a clinical trial of cancer therapy involving the combination of an autophagy regulator with conventional anticancer agents or radiation therapy has been performed [5, 12, 3741].

Our previous study suggested that different cell-death pathways are activated in gastric and colorectal carcinomas and the extrinsic and intrinsic apoptotic pathways could be mutually regulated in gastric adenocarcinomas. In contrast, in colorectal carcinomas, autophagy may function as a cellular guardian to prevent caspase-9-dependent apoptosis (intrinsic apoptotic pathway). LC3 positivity was less frequent in gastric adenocarcinomas than in colorectal adenocarcinomas [25]. Therefore, we suggested that LC3 expression in colorectal carcinomas is likely to aid cancer therapy. The detection of apoptosis and autophagy activity may help predict the treatment effect in colorectal cancer.

Presently, the anticancer agents that induce apoptosis are mainly being used. The development of a drug that induces autophagic cell death is expected in the near future. Therefore, the identification of a marker for determining the autophagic effect of drugs is important. Additionally, the development of a regulator specific for autophagy is needed.

Alisertib (ALS) Yuan et al. [46]
ALS exerts potent inhibitory effects on cell proliferation, and inducing effects on cell-cycle arrest, mitochondria-dependent apoptosis, and autophagy with the involvement of PI3K/Akt/mTOR, p38 MAPK, and AURKA-mediated signaling pathways in AGS and NCI-N78 cells.
Compound 1 Chun et al. [47]
Compound 1 effectively inhibits the growth of AGS cells by inducing apoptosis, as well as autophagy. Apoptosis after compound 1 treatment is associated with activation of caspases, release of cytochrome c, and an increased ratio of Bax/Bcl-2. Autophagy with compound 1 treatment is indicated by LC3-II protein expression.
Klotho gene (klotho protein) Xie et al. [48]
Klotho is a tumor suppressor in gastric cancer. Restoration of klotho gene expression significantly inhibits cell proliferation, induces cell apoptosis, and increases LC3-I/LC3-II expression in gastric cancer cells. Klotho regulates IGF-1R phosphorylation and the subsequent activation of IRS-1/PI3K/Akt/mTOR signaling, tumor cell proliferation, apoptosis, and autophagy.
Evodiamine Rasul et al. [49]
Evodiamine significantly inhibits the proliferation of SGC-7901 cells and induces G2/M phase cell cycle arrest. Furthermore, both autophagy and apoptosis are activated during the evodiamine-induced death of SGC-7901 cells. Evodiamine is an effective natural compound for the treatment of gastric cancer, and it may be used in in vivo studies of monotherapies or combined antitumor therapies.

Table 1.

Therapeutic agents in gastric cancer

Melphalan, bortezomib, and rapamycin Song et al. [50]
Melphalan triggers apoptosis, bortezomib induces apoptosis and autophagy, and rapamycin induces autophagy. The combination treatment induces synergistic apoptosis, which is mediated through an increase in caspase activation in human colon cancer cell lines. Mitochondrial dysfunction induced by this combination treatment has been linked with altered cellular metabolism, which induces adenosine monophosphate-activated protein kinase (AMPK) activation. AMPK-induced apoptosis, through an interplay between autophagy and apoptosis, is triggered by this combination treatment.
DENSpm Çoker-Gürkan et al. [51]
The effect of DENSpm, a polyamine analog, on cell death differs according to the p53 protein expression profile. In addition, DENSpm-induced autophagy may be critical in drug resistance in colon cancer cells.
Apigenin (4',5,7-trihydroxyflavone) Lee et al. [52]
Apigenin is a natural flavonoid with apoptosis- and autophagy-inducing effects in HCT116 colon cancer cells. Autophagy plays a cytoprotective role in apigenin-induced apoptosis, and the combination of apigenin and an autophagy inhibitor may be a promising strategy for colon cancer.
Compound K (20-O-(β-D-glucopyranosyl)-20(S)-protopanaxadiol) Kim et al. [53]
Compound K stimulates autophagy as well as apoptosis by disrupting the interaction between Atg6 and Bcl-2. The induction of autophagy and apoptosis by compound K is mediated through reactive oxygen species generation and c-Jun NH2-terminal kinase activation in human colon cancer cells.
LBH589 Gandesiri et al. [54]
The histone deacetylase inhibitor (HDACi) LBH589 has been verified as an effective anticancer agent. Death-associated protein kinase (DAPK) induces autophagy in response to HDACi treatment. In autophagy-deficient cells, DAPK plays an essential role in committing cells to HDACi-induced apoptosis.
MS-275 Zhan et al. [55]
MS-275 is a synthetic benzamide derivative of an HDACi. p38 MAP kinase plays a vital role in the switch from autophagy to apoptosis in MS-275-induced human colon cancer cells. High expression of p38 induces cell autophagy, but low expression results in apoptosis. MS-275 may be a promising clinical chemotherapeutic agent with multiple effects.
11'-deoxyverticillin A (C42) Zhang et al. [56]
C42 is an epipolythiodioxopiperazine. C42 enhances the cellular autophagic process, which requires both PARP and receptor-interacting protein 1 participation, and this precedes and possibly augments caspase-dependent apoptotic cell death.
Sulforaphane (SUL) Nishikawa et al. [57]
SUL, a type of isothiocyanate and a pro-apoptotic agent, triggers the induction of autophagy by endothelial cells, similar to cancer cells, and the inhibition of autophagy potentiates the pro-apoptotic effect. This suggests the possible use of autophagy inhibitors in combination with anti-angiogenic agents.

Table 2.

Therapeutic agents in colorectal cancer

References

  1. 1. Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med 2013;368:651–62. doi: 10.1056/NEJMra1205406.
  2. 2. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell 2011;147:728–41. doi: 10.1016/j.cell.2011.10.026.
  3. 3. Ohsumi Y. Historical landmarks of autophagy research. Cell Res 2014;24(1):9–23. doi: 10.1038/cr.2013.169.
  4. 4. Chen S, Rehman SK, Zhang W, Wen A, Yao L, Zhang J. Autophagy is a therapeutic target in anticancer drug resistance. Biochim Biophys Acta 2010 Dec;1806(2):220–9. doi:10.1016/j.bbcan.2010.07.003.
  5. 5. Mathew R, Karantza-Wadsworth V, White E. Role of autophagy in cancer. Nat Rev Cancer 2007;7(12):961–7. doi:10.1038/nrc2254
  6. 6. Nakatogawa H, Suzuki K, Kamada Y, Ohsumi Y. Dynamics and diversity in autophagy mechanisms: lessons from yeast. Nat Rev Mol Cell Biol 2009.;10(7):458–67. doi: 10.1038/nrm2708.
  7. 7. Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol 2011;27:107–32. doi: 10.1146/annurev-cellbio-092910-154005.
  8. 8. Ravikumar B, Moreau K, Jahreiss L, Puri C, Rubinsztein DC. Plasma membrane contributes to the formation of pre-autophagosomal structures. Nat Cell Biol 2010;12(8):747–57. doi: 10.1038/ncb2078.
  9. 9. Hailey DW, Rambold AS, Satpute-Krishnan P, Mitra K, Sougrat R, Kim PK, Lippincott-Schwartz J. Mitochondria supply membranes for autophagosome biogenesis during starvation. Cell 2010;141(4):656–67. doi: 10.1016/j.cell.2010.04.009.
  10. 10. Tooze SA, Yoshimori T. The origin of the autophagosomal membrane. Nat Cell Biol 2010;12(9):831–5. doi: 10.1038/ncb0910-831.
  11. 11. Mah LY, Ryan KM. Autophagy and cancer. Cold Spring Harb Perspect Biol 2012;4(1):a008821. doi: 10.1101/cshperspect.a008821.
  12. 12. Cheong H, Lu C, Lindsten T, Thompson CB. Therapeutic targets in cancer cell metabolism and autophagy. Nat Biotechnol 2012;30(7):671–8. doi: 10.1038/nbt.2285.
  13. 13. White E. Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer 2012;12(6):401–10. doi: 10.1038/nrc3262.
  14. 14. Rubinsztein DC, Codogno P, Levine B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat Rev Drug Discov 2012;11(9):709–30. doi: 10.1038/nrd3802.
  15. 15. Janku F, McConkey DJ, Hong DS, Kurzrock R. Autophagy as a target for anticancer therapy. Nat Rev Clin Oncol 2011;8(9):528–39. doi: 10.1038/nrclinonc.2011.71.
  16. 16. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 2006;441(7095):885–9.
  17. 17. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, Tanaka K. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 2006;441(7095):880–4.
  18. 18. Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, Nishida K, Hori M, Mizushima N, Otsu K. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med 2007;13(5):619–24.
  19. 19. Campoy E, Colombo MI. Autophagy in intracellular bacterial infection. Biochim Biophys Acta 2009;1793(9):1465–77. doi: 10.1016/j.bbamcr.
  20. 20. Yang Z, Klionsky DJ. An overview of the molecular mechanism of autophagy. Curr Top Microbiol Immunol 2009;335:1–32. doi: 10.1007/978-3-642-00302-8_1.
  21. 21. Martinet W, Schrijvers DM, Timmermans JP, Bult H, De Meyer GR. Immunohistochemical analysis of macroautophagy: recommendations and limitations. Autophagy 2013;9(3):386–402. doi: 10.4161/auto.22968.
  22. 22. Chen Y, Li X, Wu X, He C, Guo L, Zhang S, Xiao Y, Guo W, Tan B. Autophagy-related proteins LC3 and Beclin-1 impact the efficacy of chemoradiation on esophageal squamous cell carcinoma. Pathol Res Pract 2013;209(9):562–7. doi: 10.1016/j.prp.2013.06.006.
  23. 23. Lee HS, Daniels BH, Salas E, Bollen AW, Debnath J, Margeta M. Clinical utility of LC3 and p62 immunohistochemistry in diagnosis of drug-induced autophagic vacuolar myopathies: a case-control study. PLoS One 2012;7(4):e36221. doi: 10.1371/journal.pone.0036221.
  24. 24. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y, Yoshimori T. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 2000;19(21):5720–8. doi: 10.1093/emboj/19.21.5720
  25. 25. Shintani M, Sangawa A, Yamao N, Miyake T, Kamoshida S. Immunohistochemical analysis of cell death pathways in gastrointestinal adenocarcinoma. Biomed Res 2011;32(6):379–86. doi:org/10.2220/biomedres.32.379
  26. 26. Tanida I, Ueno T, Kominami E. LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol 2004;36(12):2503–18. doi:10.1016/j.biocel.2004.05.009.
  27. 27. Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, Levine B. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999;402(6762):672–6.
  28. 28. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, Cattoretti G, Levine B.J Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. Clin Invest 2003;112(12):1809–20.
  29. 29. Fu LL, Cheng Y, Liu B. Beclin-1:autophagic regulator and therapeutic target in cancer. Int J Biochem Cell Biol 2013;45(5):921–4. doi: 10.1016/j.biocel.2013.02.007. Epub 2013 Feb 16.
  30. 30. Yue Z, Jin S, Yang C, Levine AJ, Heintz N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci USA 2003;100(25):15077–82.
  31. 31. Ahn CH, Jeong EG, Lee JW, Kim MS, Kim SH, Kim SS, Yoo NJ, Lee SH. Expression of beclin-1, an autophagy-related protein, in gastric and colorectal cancers. APMIS 2007;115(12):1344–9. doi: 10.1111/j.1600-0463.2007.00858.x.
  32. 32. Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, Bray K, Reddy A, Bhanot G, Gelinas C, Dipaola RS, Karantza-Wadsworth V, White E. Autophagy suppresses tumorigenesis through elimination of p62. Cell 2009;137(6):1062–75.
  33. 33. Yang SY, Winslet MC. Dual role of autophagy in colon cancer cell survival. Ann Surg Oncol 2011;18(3):S239. doi: 10.1245/s10434-011-1789-x. Epub 2011 May 21.
  34. 34. Li J, Hou N, Faried A, Tsutsumi S, Takeuchi T, Kuwano H. Inhibition of autophagy by 3-MA enhances the effect of 5-FU-induced apoptosis in colon cancer cells. Ann Surg Oncol 2009;16(3):761–71. doi: 10.1245/s10434-008-0260-0.
  35. 35. Levine B, Klionsky DJ. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Cell 2004;6(4):463–77. doi:10.1016/S1534-5807(04)00099-1.
  36. 36. Baehrecke EH. Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol 2005;6(6):505–10.
  37. 37. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res 2007;13(24):7271–9.
  38. 38. Turcotte S, Giaccia AJ. Targeting cancer cells through autophagy for anticancer therapy. Curr Opin Cell Biol 2010;22(2):246–51.
  39. 39. Edinger AL, Thompson CB. Death by design: apoptosis, necrosis and autophagy. Curr Opin Cell Biol 2004;16(6):663–9. doi:10.1016/j.ceb.2004.09.011.
  40. 40. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gélinas C, Fan Y, Nelson DA, Jin S, White E. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006;10(1):51–64. doi: 10.1016/j.ccr.2006.06.001.
  41. 41. 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. doi:10.1038/nrm2239.
  42. 42. Park JM, Huang S, Wu TT, Foster NR, Sinicrope FA. Prognostic impact of Beclin 1, p62/sequestosome 1 and LC3 protein expression in colon carcinomas from patients receiving 5-fluorouracil as adjuvant chemotherapy. Cancer Biol Ther 2013;14(2):100–7. doi: 10.4161/cbt.22954.
  43. 43. Li J, Hou N, Faried A, Tsutsumi S, Kuwano H. Inhibition of autophagy augments 5-fluorouracil chemotherapy in human colon cancer in vitro and in vivo model. Eur J Cancer 2010;46(10):1900–9. doi: 10.1016/j.ejca.2010.02.021.
  44. 44. Bijnsdorp IV, Peters GJ, Temmink OH, Fukushima M, Kruyt FA. Differential activation of cell death and autophagy results in an increased cytotoxic potential for trifluorothymidine compared to 5-fluorouracil in colon cancer cells. Int J Cancer 2010;126(10):2457–68. doi: 10.1002/ijc.24943.
  45. 45. Huang S, Sinicrope FA. Celecoxib-induced apoptosis is enhanced by ABT-737 and by inhibition of autophagy in human colorectal cancer cells. Autophagy 2010;6(2):256–69. Epub 2010 Feb 6.
  46. 46. Yuan CX, Zhou ZW, Yang YX, He ZX, Zhang X, Wang D, Yang T, Wang NJ, Zhao RJ, Zhou SF. Inhibition of mitotic Aurora kinase A by alisertib induces apoptosis and autophagy of human gastric cancer AGS and NCI-N78 cells. Drug Des Develop Ther 2015;9:487–508. doi: 10.2147/DDDT.S74127.
  47. 47. Chun J, Kang M, Kim YS. A triterpenoid saponin from Adenophora triphylla var. japonica suppresses the growth of human gastric cancer cells via regulation of apoptosis and autophagy. Tumour Biol 2014;35(12):12021–30. doi: 10.1007/s13277-014-2501-0.
  48. 48. Xie B, Zhou J, Shu G, Liu DC, Zhou J, Chen J, Yuan L. Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int 2013;13(1):18. doi: 10.1186/1475-2867-13-18.
  49. 49. Rasul A, Yu B, Zhong L, Khan M, Yang H, Ma T. Cytotoxic effect of evodiamine in SGC-7901 human gastric adenocarcinoma cells via simultaneous induction of apoptosis and autophagy. Oncol Rep 2012;27(5):1481–7. doi: 10.3892/or.2012.1694.
  50. 50. Song X, Kim SY, Zhang L, Tang D, Bartlett DL, Kwon YT, Lee YJ. Role of AMP-activated protein kinase in cross-talk between apoptosis and autophagy in human colon cancer. Cell Death Dis 2014;5:e1504. doi: 10.1038/cddis.2014.463.
  51. 51. Çoker-Gürkan A, Arisan ED, Obakan P, Palavan-Unsal N. Lack of functional p53 renders DENSpm-induced autophagy and apoptosis in time dependent manner in colon cancer cells. Amino Acids 2015;47(1):87–100. doi: 10.1007/s00726-014-1851-7.
  52. 52. Lee Y, Sung B, Kang YJ, Kim DH, Jang JY, Hwang SY, Kim M, Lim HS, Yoon JH, Chung HY, Kim ND. Apigenin-induced apoptosis is enhanced by inhibition of autophagy formation in HCT116 human colon cancer cells. Int J Oncol 2014;44(5):1599–606. doi: 10.3892/ijo.2014.2339.
  53. 53. Kim AD, Kang KA, Kim HS, Kim DH, Choi YH, Lee SJ, Kim HS, Hyun JW. A ginseng metabolite, compound K, induces autophagy and apoptosis via generation of reactive oxygen species and activation of JNK in human colon cancer cells. Cell Death Dis 2013;4:e750. doi: 10.1038/cddis.2013.273.
  54. 54. Gandesiri M, Chakilam S, Ivanovska J, Benderska N, Ocker M, Di Fazio P, Feoktistova M, Gali-Muhtasib H, Rave-Fränk M, Prante O, Christiansen H, Leverkus M, Hartmann A, Schneider-Stock R. DAPK plays an important role in panobinostat-induced autophagy and commits cells to apoptosis under autophagy deficient conditions. Apoptosis 2012;17(12):1300–15. doi: 10.1007/s10495-012-0757-7.
  55. 55. Zhan Y, Gong K, Chen C, Wang H, Li W. P38 MAP kinase functions as a switch in MS-275-induced reactive oxygen species-dependent autophagy and apoptosis in human colon cancer cells. Free Radic Biol Med 2012;53(3):532–43. doi: 10.1016/j.freeradbiomed.2012.05.018.
  56. 56. Zhang N, Chen Y, Jiang R, Li E, Chen X, Xi Z, Guo Y, Liu X, Zhou Y, Che Y, Jiang X. PARP and RIP 1 are required for autophagy induced by 11'-deoxyverticillin A, which precedes caspase-dependent apoptosis. Autophagy 2011;7(6):598–612.
  57. 57. Nishikawa T, Tsuno NH, Okaji Y, Sunami E, Shuno Y, Sasaki K, Hongo K, Kaneko M, Hiyoshi M, Kawai K, Kitayama J, Takahashi K, Nagawa H. The inhibition of autophagy potentiates anti-angiogenic effects of sulforaphane by inducing apoptosis. Angiogenesis 2010;13(3):227–38. doi: 10.1007/s10456-010-9180-2.

Written By

Michiko Shintani

Submitted: 25 December 2014 Reviewed: 15 September 2015 Published: 16 December 2015