REFERENCES

1. Morel E, Mehrpour M, Botti J, Dupont N, Hamaï A, et al. Autophagy: a druggable process. Annu Rev Pharmacol Toxicol 2017;57:375-98.

2. Saha S, Panigrahi DP, Patil S, Bhutia SK. Autophagy in health and disease: a comprehensive review. Biomed Pharmacother 2018;104:485-95.

3. Masui A, Hamada M, Kameyama H, Wakabayashi K, Takasu A, et al. Autophagy as a survival mechanism for squamous cell carcinoma cells in endonuclease g-mediated apoptosis. PLoS One 2016;11:1-19.

4. Towers CG, Thorburn A. Therapeutic targeting of autophagy. EBioMedicine 2016;14:15-23.

5. Li W, Yang Q, Mao Z. Chaperone-mediated autophagy: machinery, regulation and biological consequences. Cell Mol Life Sci 2011;68:749-63.

6. Li WW, Li J, Bao JK. Microautophagy: lesser-known self-eating. Cell Mol Life Sci 2012;69:1125-36.

7. Yoshii SR, Mizushima N. Monitoring and measuring autophagy. Int J Mol Sci 2017;18:1-13.

8. Nascimbeni AC, Giordano F, Dupont N, Grasso D, Vaccaro MI, et al. ER-plasma membrane contact sites contribute to autophagosome biogenesis by regulation of local PI3P synthesis. EMBO J 2017;36:2018-33.

9. Nakamura S, Yoshimori T. New insights into autophagosome-lysosome fusion. J Cell Sci 2017;130:1209-16.

10. Klionsky DJ, Eskelinen EL, Deretic V. Autophagosomes, phagosomes, autolysosomes, phagolysosomes, autophagolysosomes... wait, I’m confused. Autophagy 2014;10:549-51.

11. Fang Y, Tan J, Zhang Q. Signaling pathways and mechanisms of hypoxia-induced autophagy in the animal cells. Cell Biol Int 2015;39:891-8.

12. Thorburn A. Autophagy and disease. J Biol Chem 2018;293:5425-30.

13. Eskelinen EL, Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta - Mol Cell Res 2009;1793:664-73.

14. Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol 2018;19:349-64.

15. Amaravadi R, Kimmelman AC, White E. Recent insights into the function of autophagy in cancer. Genes Dev 2016;30:1913-30.

16. Zachari M, Ganley IG. The mammalian ULK1 complex and autophagy initiation. Essays Biochem 2017;61:585-96.

17. Tanida I. Autophagy basics. Microbiol Immunol 2011;55:1-11.

18. Dunlop EA, Hunt DK, Acosta-Jaquez HA, Fingar DC, Tee AR. ULK1 inhibits mTORC1 signaling, promotes multisite raptor phosphorylation and hinders substrate binding. Autophagy 2011;7:737-47.

19. Roach PJ. AMPK -> ULK1 -> autophagy. Mol Cell Biol 2011;31:3082-4.

20. Lin S-Y, Li TY, Liu Q, Zhang C, Li X, et al. GSK3-TIP60-ULK1 signaling pathway links growth factor deprivation to autophagy. Science 2012;336:477-81.

21. Pyo JO, Nah J, Jung YK. Molecules and their functions in autophagy. Exp Mol Med 2012;44:73-80.

22. Mercer TJ, Gubas A, Tooze SA. A molecular perspective of mammalian autophagosome biogenesis. J Biol Chem 2018;293:5386-95.

23. Kang R, Zeh HJ, Lotze MT, Tang D. The beclin 1 network regulates autophagy and apoptosis. Cell Death Differ 2011;18:571-80.

24. Kim JH, Hong SB, Lee JK, Han S, Roh KH, et al. Insights into autophagosome maturation revealed by the structures of ATG5 with its interacting partners. Autophagy 2015;11:75-87.

25. Sakoh-Nakatogawa M, Matoba K, Asai E, Kirisako H, Ishii J, et al. Atg12-Atg5 conjugate enhances E2 activity of Atg3 by rearranging its catalytic site. Nat Struct Mol Biol 2013;20:433-9.

26. Lao Y, Xu N. Autophagy in cancer chemoprevention: identifcation of novel autophagy modulators with anticancer potential. Compr Toxicol Second Ed 2016;1379:151-63.

27. Manni D. Oxidation-dependent regulation of the selective autophagy receptor SQSTM1/p62. Available from: https://theses.ncl.ac.uk/dspace/handle/10443/3675 [Last accessed on 3 Apr 2019].

28. Weidberg H, Shvets E, Shpilka T, Shimron F, Shinder V, et al. LC3 and GATE-16/GABARAP subfamilies are both essential yet act differently in autophagosome biogenesis. EMBO J 2010;29:1792-802.

29. Fader CM, Sánchez D, Furlán M, Colombo MI. Induction of autophagy promotes fusion of multivesicular bodies with autophagic vacuoles in K562 cells. Traffic 2008;9:230-50.

30. Lee JA, Beigneux A, Ahmad ST, Young SG, Gao FB. Escrt-III dysfunction causes autophagosome accumulation and neurodegeneration. Curr Biol 2007;17:1561-7.

31. Li C, White SH, Warren LK, Wohlgemuth SE. Skeletal muscle from aged American Quarter Horses shows impairments in mitochondrial biogenesis and expression of autophagy markers. Exp Gerontol 2018;102:19-27.

32. Mawatwal S, Behura A, Ghosh A, Kidwai S, Mishra A, et al. Calcimycin mediates mycobacterial killing by inducing intracellular calcium-regulated autophagy in a P2RX7 dependent manner. Biochim Biophys Acta - Gen Subj 2017;1861:3190-200.

33. Settembre C, Fraldi A, Rubinsztein DC, Ballabio A. Lysosomal storage diseases as disorders of autophagy. Autophagy 2008;4:113-4.

34. Cai Y, Arikkath J, Yang L, Guo ML, Periyasamy P, et al. Interplay of endoplasmic reticulum stress and autophagy in neurodegenerative disorders. Autophagy 2016;12:225-44.

35. Singh SS, Vats S, Chia AYQ, Tan TZ, Deng S, et al. Dual role of autophagy in hallmarks of cancer. Oncogene 2018;37:1142-58.

36. Comel A, Sorrentino G, Capaci V, Del Sal G. The cytoplasmic side of p53’s oncosuppressive activities. FEBS Lett 2014;588:2600-9.

37. Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med 2013;368:651-62.

38. Takahashi Y, Coppola D, Matsushita N, Cualing HD, Sun M, et al. Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nat Cell Biol 2007;9:1142-51.

39. Coppola D, Khalil F, Eschrich SA, Boulware D, Yeatman T, et al. Down-regulation of bax-interacting factor-1 in colorectal adenocarcinoma. Cancer 2008;113:2665-70.

40. He S, Zhao Z, Yang Y, O’Connell D, Zhang X, et al. Truncating mutation in the autophagy gene UVRAG confers oncogenic properties and chemosensitivity in colorectal cancers. Nat Commun 2015;6:7839.

41. Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, et al. Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature 2015;524:361-5.

42. Fulda S. Autophagy in cancer therapy. Front Oncol 2017;7:128.

43. Thorburn A. Autophagy and its effects: making sense of double-edged swords. PLoS Biol 2014;12:e1001967.

44. Yun CW, Lee SH. The roles of autophagy in cancer. Int J Mol Sci 2018;19:3466.

45. Koustas E, Papavassiliou AG, Karamouzis MV. The role of autophagy in the treatment of BRAF mutant colorectal carcinomas differs based on microsatellite instability status. PLoS One 2018;13:e0207227.

46. Boutouja F, Brinkmeier R, Mastalski T, Magraoui F, Platta HW. Regulation of the tumor-suppressor beclin 1 by distinct ubiquitination cascades. Int J Mol Sci 2017;18:E2541.

47. Miracco C, Cosci E, Oliveri G, Luzi P, Pacenti L, et al. Protein and mRNA expression of autophagy gene beclin 1 in human brain tumours. Int J Oncol 2007;30:429-36.

48. Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in cancer: therapeutic implications. Mol Cancer Ther 2011;10:1533-41.

49. Kang MR, Kim MS, Oh JE, Kim YR, Song SY, et al. Frameshift mutations of autophagy-related genes ATG2B, ATG5, ATG9B and ATGI2 in gastric and colorectal cancers with microsatellite instability. J Pathol 2009;217:702-6.

50. Lipinski MM, Hoffman G, Ng A, Zhou W, Py BF, et al. A genome-wide siRNA screen reveals multiple mTORC1 independent signaling pathways regulating autophagy under normal nutritional conditions. Dev Cell 2010;18:1041-52.

51. Tang F, Hu P, Yang Z, Xue C, Gong J, et al. SBI0206965, a novel inhibitor of Ulk1, suppresses non-small cell lung cancer cell growth by modulating both autophagy and apoptosis pathways. Oncol Rep 2017;37:3449-58.

52. Bellot G, Garcia-Medina R, Gounon P, Chiche J, Roux D, et al. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol 2009;29:2570-81.

53. Karnoub AE, Weinberg RA. Ras oncogenes: split personalities. Nat Rev Mol Cell Biol 2008;9:517-31.

54. Ahn JH, Lee M. Autophagy-dependent survival of mutant B-Raf melanoma cells selected for resistance to apoptosis induced by inhibitors against oncogenic B-Raf. Biomol Ther 2013;21:114-20.

55. Russo M, Russo GL. Autophagy inducers in cancer. Biochem Pharmacol 2018;153:51-61.

56. He S, Li Q, Jiang X, Lu X, Feng F, et al. Design of small molecule autophagy modulators: a promising druggable strategy. J Med Chem 2018;61:4656-87.

57. Kwitkowski VE, Prowell TM, Ibrahim A, Farrell AT, Justice R, et al. FDA approval summary: temsirolimus as treatment for advanced renal cell carcinoma. Oncologist 2010;15:428-35.

58. Koschmann C, Farooqui Z, Kasaian K, Cao X, Zamler D, et al. Multi-focal sequencing of a diffuse intrinsic pontine glioma establishes PTEN loss as an early event. NPJ Precis Oncol 2017;1:32.

59. Klionsky DJ, Eskelinen EL, Deretic V. Autophagosomes, phagosomes, autolysosomes, phagolysosomes, autophagolysosomes… wait, I’m confused. Autophagy 2014;10:549-51.

60. Wymann MP, Bulgarelli-Leva G, Zvelebil MJ, Pirola L, Vanhaesebroeck B. Wortmannin inactivates phosphoinositide 3-Kinase by covalent modification of Lys-802, a residue involved in the phosphate transfer reaction. Mol Cell Biol 1996;16:1722-33.

61. McNamara CR, Degterev A. Small-molecule inhibitors of the PI3K signaling network. Future Med Chem 2011;3:549-65.

62. Gharbi SI, Zvelebil MJ, Shuttleworth SJ, Hancox T, Saghir N, et al. Exploring the specificity of the PI3K family inhibitor LY294002. Biochem J 2007;404:15-21.

63. Harding JJ, Bauer TM, Tan DSW, Bedard PL, Rodon J, et al. Characterization and phase I study of CLR457, an orally bioavailable pan-class I PI3-kinase inhibitor. Invest New Drugs 2019;37:271-81.

64. Knight SD, Adams ND, Burgess JL, Chaudhari AM, Darcy MG, et al. Discovery of GSK2126458, a highly potent inhibitor of PI3K and the mammalian target of rapamycin. ACS Med Chem Lett 2010;1:39-43.

65. Basu D, Salgado CM, Bauer B, Khakoo Y, Patel JR, et al. The dual PI3K/mToR inhibitor omipalisib/GSK2126458 inhibits clonogenic growth in oncogenically-transformed cells from neurocutaneous melanocytosis. Cancer Genomics and Proteomics 2018;15:239-48.

66. Liu Q, Xu C, Kirubakaran S, Zhang X, Hur W, et al. Characterization of torin2, an ATP-competitive inhibitor of mTOR, ATM, and ATR. Cancer Res 2013;73:2574-86.

67. Nelson EE, Guyer AE. The development of the ventral prefrontal cortex and social flexibility. Dev Cogn Neurosci 2011;1:233-45.

68. Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, et al. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res 2010;70:288-98.

69. Wang L, Zhu YR, Wang S, Zhao S. Autophagy inhibition sensitizes WYE-354-induced anti-colon cancer activity in vitro and in vivo. Tumor Biol 2016;37:11743-52.

70. Park S, Chapuis N, Bardet V, Tamburini J, Gallay N, et al. PI-103, a dual inhibitor of class IA phosphatidylinositide 3-kinase and mTOR, has antileukemic activity in AML. Leukemia 2008;22:1698-706.

71. Mallon R, Feldberg LR, Lucas J, Chaudhary I, Dehnhardt C, et al. Antitumor efficacy of PKI-587, a highly potent dual PI3K/mTOR kinase inhibitor. Clin Cancer Res 2011;17:3193-203.

72. Powles T, Lackner MR, Oudard S, Escudier B, Ralph C, et al. Randomized open-label phase II trial of apitolisib (GDC-0980), a novel inhibitor of the PI3K/mammalian target of rapamycin pathway, versus everolimus in patients with metastatic renal cell carcinoma. J Clin Oncol 2016;34:1660-8.

73. Simioni C, Cani A, Martelli AM, Zauli G, Alameen AA, et al. The novel dual PI3K/mTOR inhibitor NVP-BGT226 displays cytotoxic activity in both normoxic and hypoxic hepatocarcinoma cells. Oncotarget 2015;6:17147-60.

74. Li H, Jin X, Zhang Z, Xing Y, Kong X. Inhibition of autophagy enhances apoptosis induced by the PI3K/AKT/mTor inhibitor NVP-BEZ235 in renal cell carcinoma cells. Cell Biochem Funct 2013;31:427-33.

75. Lazarus MB, Novotny CJ, Shokat KM. Structure of the human autophagy initiating kinase ULK1 in complex with potent inhibitors. ACS Chem Biol 2015;10:257-61.

76. Martin KR, Celano SL, Solitro AR, Gunaydin H, Scott M, et al. A potent and selective ULK1 inhibitor suppresses autophagy and sensitizes cancer cells to nutrient stress. iScience 2018;8:74-84.

77. Limpert AS, Lambert LJ, Bakas NA, Bata N, Brun SN, et al. Autophagy in cancer: regulation by small molecules. Trends Pharmacol Sci 2018;39:53-63.

78. Lazarus MB, Shokat KM. Discovery and structure of a new inhibitor scaffold of the autophagy initiating kinase ULK1. Bioorganic Med Chem 2015;23:5483-8.

79. Matsunaga K, Morita E, Saitoh T, Akira S, Ktistakis NT, et al. Autophagy requires endoplasmic reticulum targeting of the PI3-kinase complex via Atg14L. J Cell Biol 2010;190:511-21.

80. Honda A, Harrington E, Cornella-Taracido I, Furet P, Knapp MS, et al. Potent, selective, and orally bioavailable inhibitors of VPS34 provide chemical tools to modulate autophagy in vivo. ACS Med Chem Lett 2016;7:72-6.

81. Dowdle WE, Nyfeler B, Nagel J, Elling RA, Liu S, et al. Selective VPS34 inhibitor blocks autophagy and uncovers a role for NCOA4 in ferritin degradation and iron homeostasis in vivo. Nat Cell Biol 2014;16:1069-79.

82. Pasquier B, El-Ahmad Y, Filoche-Rommé B, Dureuil C, Fassy F, et al. Discovery of (2 S)-8-[(3 R)-3-methylmorpholin-4-yl]-1-(3-methyl-2-oxobutyl)-2-(trifluoromethyl)-3,4-dihydro-2 H -pyrimido[1,2- a ]pyrimidin-6-one: a novel potent and selective inhibitor of Vps34 for the treatment of solid tumors. J Med Chem 2015;58:376-400.

83. Manic G, Obrist F, Kroemer G, Vitale I, Galluzzi L. Chloroquine and hydroxychloroquine for cancer therapy. Mol Cell Oncol 2014;1:e29911.

84. Shi TT, Yu XX, Yan LJ, Xiao HT. Research progress of hydroxychloroquine and autophagy inhibitors on cancer. Cancer Chemother Pharmacol 2017;79:287-94.

85. Mauthe M, Orhon I, Rocchi C, Zhou X, Luhr M, et al. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy 2018;14:1435-55.

86. Chude CI, Amaravadi RK. Targeting autophagy in cancer: update on clinical trials and novel inhibitors. Int J Mol Sci 2017;18:E1279.

87. Xu R, Ji Z, Xu C, Zhu J. The clinical value of using chloroquine or hydroxychloroquine as autophagy inhibitors in the treatment of cancers: a systematic review and meta-analysis. Medicine (Baltimore) 2018;97:e12912.

88. Amaravadi RK, Winkler JD. Lys05: a new lysosomal autophagy inhibitor. Autophagy 2012;8:1383-4.

89. McAfee Q, Zhang Z, Samanta A, Levi SM, Ma XH, et al. Autophagy inhibitor Lys05 has single-agent antitumor activity and reproduces the phenotype of a genetic autophagy deficiency. Proc Natl Acad Sci 2012;109:8253-8.

90. Kasi PD, Tamilselvam R, Skalicka-Woźniak K, Nabavi SF, Daglia M, et al. Molecular targets of curcumin for cancer therapy: an updated review. Tumor Biol 2016;37:13017-28.

91. Zhu Y, Bu S. Curcumin induces autophagy, apoptosis, and cell cycle arrest in human pancreatic cancer cells. Evidence-Based Complement Altern Med 2017;2017:5787218.

92. Xiao K, Jiang J, Guan C, Dong C, Wang G, et al. Curcumin induces autophagy via activating the AMPK signaling pathway in lung adenocarcinoma cells. J Pharmacol Sci 2013;123:102-9.

93. Kobori M, Takahashi Y, Sakurai M, Akimoto Y, Tsushida T, et al. Quercetin suppresses immune cell accumulation and improves mitochondrial gene expression in adipose tissue of diet-induced obese mice. Mol Nutr Food Res 2016;60:300-12.

94. Lou M, Zhang LN, Ji PG, Feng FQ, Liu JH, et al. Quercetin nanoparticles induced autophagy and apoptosis through AKT/ERK/Caspase-3 signaling pathway in human neuroglioma cells: In vitro and in vivo. Biomed Pharmacother 2016;84:1-9.

95. Liu Y, Gong W, Yang ZY, Zhou XS, Gong C, et al. Quercetin induces protective autophagy and apoptosis through ER stress via the p-STAT3/Bcl-2 axis in ovarian cancer. Apoptosis 2017;22:544-57.

96. Hwang JJ, Kuruvilla J, Mendelson D, Pishvaian MJ, Deeken JF, et al. Phase I dose finding studies of obatoclax (GX15-070), a small molecule Pan-BCL-2 family antagonist, in patients with advanced solid tumors or lymphoma. Clin Cancer Res 2010;16:4038-45.

97. Basit F, Cristofanon S, Fulda S. Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ 2013;20:1161-73.

98. Yang Y, Chen S, Zhang Y, Lin X, Song Y, et al. Induction of autophagy by spermidine is neuroprotective via inhibition of caspase 3-mediated Beclin 1 cleavage. Cell Death Dis 2017;8:e2738.

99. Sacitharan PK, Lwin S, Gharios GB, Edwards JR. Spermidine restores dysregulated autophagy and polyamine synthesis in aged and osteoarthritic chondrocytes via EP300. Exp Mol Med 2018;50:123.

100. Welsh PA, Sass-Kuhn S, Prakashagowda C, McCloskey D, Feith D. Spermine synthase overexpression in vivo does not increase susceptibility to DMBA/TPA skin carcinogenesis or Min-Apc intestinal tumorigenesis. Cancer Biol Ther 2012;13:358-68.

101. Matsui TA, Murata H, Sakabe T, Sowa Y, Horie N, et al. Sulforaphane induces cell cycle arrest and apoptosis in murine osteosarcoma cells in vitro and inhibits tumor growth in vivo. Oncol Rep 2007;18:1263-8.

102. Xiao D, Powolny AA, Antosiewicz J, Hahm ER, Bommareddy A, et al. Cellular responses to cancer chemopreventive agent D,L-sulforaphane in human prostate cancer cells are initiated by mitochondrial reactive oxygen species. Pharm Res 2009;26:1729-38.

103. Liu H, Smith AJ, Ball SS, Bao Y, Bowater RP, et al. Sulforaphane promotes ER stress, autophagy, and cell death: implications for cataract surgery. J Mol Med 2017;95:553-64.

104. Wiczk A, Hofman D, Konopa G, Herman-Antosiewicz A. Sulforaphane, a cruciferous vegetable-derived isothiocyanate, inhibits protein synthesis in human prostate cancer cells. Biochim Biophys Acta - Mol Cell Res 2012;1823:1295-305.

105. Cohen MH, Johnson JR, Pazdur R. Food and drug administration drug approval summary: temozolomide plus radiation therapy for the treatment of newly diagnosed glioblastoma multiforme. Clin Cancer Res 2005;11:6767-71.

106. Würstle S, Schneider F, Ringel F, Gempt J, Lämmer F, et al. Temozolomide induces autophagy in primary and established glioblastoma cells in an EGFR independent manner. Oncol Lett 2017;14:322-8.

107. Dai C, Zhang B, Liu X, Ma S, Yang Y, et al. Inhibition of PI3K/AKT/mTOR pathway enhances temozolomide-induced cytotoxicity in pituitary adenoma cell lines in vitro and xenografted pituitary adenoma in female nude mice. Endocrinology 2013;154:1247-59.

108. Chen B, Xiao F, Li B, Xie B, Zhou J, et al. The role of epithelial-mesenchymal transition and IGF-1R expression in prediction of gefitinib activity as the second-line treatment for advanced nonsmall-cell lung cancer. Cancer Invest 2013;31:454-60.

109. Zhao ZQ, Yu ZY, Li J, Ouyang XN. Gefitinib induces lung cancer cell autophagy and apoptosis via blockade of the PI3K/AKT/mTOR pathway. Oncol Lett 2016;12:63-8.

110. Sugita S, Ito K, Yamashiro Y, Moriya S, Che XF, et al. EGFR-independent autophagy induction with gefitinib and enhancement of its cytotoxic effect by targeting autophagy with clarithromycin in non-small cell lung cancer cells. Biochem Biophys Res Commun 2015;461:28-34.

111. Dowling RJO, Goodwin PJ, Stambolic V. Understanding the benefit of metformin use in cancer treatment. BMC Med 2011;9:33.

112. Feng Y, Ke C, Tang Q, Dong H, Zheng X, et al. Metformin promotes autophagy and apoptosis in esophageal squamous cell carcinoma by downregulating Stat3 signaling. Cell Death Dis 2014;5:1-12.

113. Wang Y, Xu W, Yan Z, Zhao W, Mi J, et al. Metformin induces autophagy and G0/G1 phase cell cycle arrest in myeloma by targeting the AMPK/mTORC1 and mTORC2 pathways. J Exp Clin Cancer Res 2018;37:1-12.

114. Field-smith A, Morgan GJ, Davies FE. Bortezomib (velcadetrade mark) in the treatment of multiple myeloma. Ther Clin Risk Manag 2006;2:271-9.

115. Kao C, Chao A, Tsai CL, Chuang WC, Huang WP, et al. Bortezomib enhances cancer cell death by blocking the autophagic flux through stimulating ERK phosphorylation. Cell Death Dis 2014;5:e1510.

116. Yang X, Srivastava R, Howell SH, Bassham DC. Activation of autophagy by unfolded proteins during endoplasmic reticulum stress. Plant J 2016;85:83-95.

117. Akbarzadeh L, Moini Zanjani T, Sabetkasaei M. Comparison of anticancer effects of carbamazepine and valproic acid. Iran Red Crescent Med J 2016;18:e37230.

118. Kania E, Pająk B, O’Prey J, Sierra Gonzalez P, Litwiniuk A, et al. Verapamil treatment induces cytoprotective autophagy by modulating cellular metabolism. FEBS J 2017;284:1370-87.

119. Shimizu T, Ono T, Yoshida T, Cho F, Goto N. Morphometrical study of physical growth in infant cynomolgus monkeys using multivariate analysis. Jikken Dobutsu 1988;37:145-51.

120. Tan Q, Joshua AM, Wang M, Bristow RG, Wouters BG, et al. Up-regulation of autophagy is a mechanism of resistance to chemotherapy and can be inhibited by pantoprazole to increase drug sensitivity. Cancer Chemother Pharmacol 2017;79:959-69.

121. Tan Q, Joshua AM, Saggar JK, Yu M, Wang M, et al. Effect of pantoprazole to enhance activity of docetaxel against human tumour xenografts by inhibiting autophagy. Br J Cancer 2015;112:832-40.

122. Cao Y, Chen M, Tang D, Yan H, Ding X, et al. The proton pump inhibitor pantoprazole disrupts protein degradation systems and sensitizes cancer cells to death under various stresses article. Cell Death Dis 2018;9:604.

123. Lu Y, Liu LL, Liu SS, Fang ZG, Zou Y, et al. Celecoxib suppresses autophagy and enhances cytotoxicity of imatinib in imatinib-resistant chronic myeloid leukemia cells. J Transl Med 2016;14:270.

Journal of Cancer Metastasis and Treatment
ISSN 2454-2857 (Online) 2394-4722 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/