Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Sulfonylurea | Review

Cancer and diabetes: the interlinking metabolic pathways and repurposing actions of antidiabetic drugs

Authors: Ahmed Olatunde, Manisha Nigam, Rahul Kunwar Singh, Abhaya Shikhar Panwar, Abdulwahab Lasisi, Fahad A. Alhumaydhi, Vijay Jyoti kumar, Abhay Prakash Mishra, Javad Sharifi-Rad

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Cancers are regarded as one of the main causes of death and result in high health burden worldwide. The management of cancer include chemotherapy, surgery and radiotherapy. The chemotherapy, which involves the use of chemical agents with cytotoxic actions is utilised as a single treatment or combined treatment. However, these managements of cancer such as chemotherapy poses some setbacks such as cytotoxicity on normal cells and the problem of anticancer drug resistance. Therefore, the use of other therapeutic agents such as antidiabetic drugs is one of the alternative interventions used in addressing some of the limitations in the use of anticancer agents. Antidiabetic drugs such as sulfonylureas, biguanides and thiazolidinediones showed beneficial and repurposing actions in the management of cancer, thus, the activities of these drugs against cancer is attributed to some of the metabolic links between the two disorders and these includes hyperglycaemia, hyperinsulinemia, inflammation, and oxidative stress as well as obesity. Furthermore, some studies showed that the use of antidiabetic drugs could serve as risk factors for the development of cancerous cells particularly pancreatic cancer. However, the beneficial role of these chemical agents overweighs their detrimental actions in cancer management. Hence, the present review indicates the metabolic links between cancer and diabetes and the mechanistic actions of antidiabetic drugs in the management of cancers.
Literature
1.
go back to reference Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA A Cancer J Clin. 2013;63(1):11–30.CrossRef Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA A Cancer J Clin. 2013;63(1):11–30.CrossRef
2.
go back to reference Shafiei-Irannejad V, Samadi N, Salehi R, Yousefi B, Zarghami N. New insights into antidiabetic drugs: possible applications in cancer treatment. Chem Biol Drug Des. 2017;90(6):1056–66.PubMedCrossRef Shafiei-Irannejad V, Samadi N, Salehi R, Yousefi B, Zarghami N. New insights into antidiabetic drugs: possible applications in cancer treatment. Chem Biol Drug Des. 2017;90(6):1056–66.PubMedCrossRef
3.
go back to reference Nasiri M, Zarghami N, Koshki KN, Mollazadeh M, Moghaddam MP, Yamchi MR, Esfahlan RJ, Barkhordari A, Alibakhshi A. Curcumin and silibinin inhibit telomerase expression in T47D human breast cancer cells. Asian Pac J Cancer Prev. 2013;14(6):3449–53.PubMedCrossRef Nasiri M, Zarghami N, Koshki KN, Mollazadeh M, Moghaddam MP, Yamchi MR, Esfahlan RJ, Barkhordari A, Alibakhshi A. Curcumin and silibinin inhibit telomerase expression in T47D human breast cancer cells. Asian Pac J Cancer Prev. 2013;14(6):3449–53.PubMedCrossRef
4.
go back to reference Nejati-Koshki K, Zarghami N, Pourhassan-Moghaddam M, Rahmati-Yamchi M, Mollazade M, Nasiri M, Esfahlan RJ, Barkhordari A, Tayefi-Nasrabadi H. Inhibition of leptin gene expression and secretion by silibinin: possible role of estrogen receptors. Cytotechnology. 2012;64(6):719–26.PubMedPubMedCentralCrossRef Nejati-Koshki K, Zarghami N, Pourhassan-Moghaddam M, Rahmati-Yamchi M, Mollazade M, Nasiri M, Esfahlan RJ, Barkhordari A, Tayefi-Nasrabadi H. Inhibition of leptin gene expression and secretion by silibinin: possible role of estrogen receptors. Cytotechnology. 2012;64(6):719–26.PubMedPubMedCentralCrossRef
5.
go back to reference Yousefi B, Zarghami N, Samadi N, Majidinia M. Peroxisome proliferator-activated receptors and their ligands in cancer drug- resistance: Opportunity or challenge. Anticancer Agents Med Chem. 2016;16(12):1541–8.PubMedCrossRef Yousefi B, Zarghami N, Samadi N, Majidinia M. Peroxisome proliferator-activated receptors and their ligands in cancer drug- resistance: Opportunity or challenge. Anticancer Agents Med Chem. 2016;16(12):1541–8.PubMedCrossRef
6.
go back to reference Currie CJ, Poole CD, Gale EAM. The influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia. 2009;52(9):1766–77.PubMedCrossRef Currie CJ, Poole CD, Gale EAM. The influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia. 2009;52(9):1766–77.PubMedCrossRef
7.
go back to reference Evans JMM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005;330(7503):1304–5.PubMedPubMedCentralCrossRef Evans JMM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005;330(7503):1304–5.PubMedPubMedCentralCrossRef
8.
go back to reference Huxley R, Ansary-Moghaddam A, Berrington de González A, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer. 2005;92(11):2076–83.PubMedPubMedCentralCrossRef Huxley R, Ansary-Moghaddam A, Berrington de González A, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer. 2005;92(11):2076–83.PubMedPubMedCentralCrossRef
10.
go back to reference Azar M, Lyons TJ. Diabetes, insulin treatment, and cancer risk: what is the evidence? F1000 Med Rep. 2009;2(4):1–4. Azar M, Lyons TJ. Diabetes, insulin treatment, and cancer risk: what is the evidence? F1000 Med Rep. 2009;2(4):1–4.
11.
go back to reference Smith U, Gale EA. Does diabetes therapy influence the risk of cancer? Diabetologia. 2009;52(9):1699–708.PubMedCrossRef Smith U, Gale EA. Does diabetes therapy influence the risk of cancer? Diabetologia. 2009;52(9):1699–708.PubMedCrossRef
12.
go back to reference Swerdlow AJ, Laing SP, Qiao Z, Slater SD, Burden AC, Botha JL, Waugh NR, Morris AD, Gatling W, Gale EA, et al. Cancer incidence and mortality in patients with insulin-treated diabetes: a UK cohort study. Br J Cancer. 2005;92(11):2070–5.PubMedPubMedCentralCrossRef Swerdlow AJ, Laing SP, Qiao Z, Slater SD, Burden AC, Botha JL, Waugh NR, Morris AD, Gatling W, Gale EA, et al. Cancer incidence and mortality in patients with insulin-treated diabetes: a UK cohort study. Br J Cancer. 2005;92(11):2070–5.PubMedPubMedCentralCrossRef
13.
go back to reference Shu X, Ji J, Li X, Sundquist J, Sundquist K, Hemminki K. Cancer risk among patients hospitalized for Type 1 diabetes mellitus: a population-based cohort study in Sweden. Diabet Med. 2010;27(7):791–7.PubMedCrossRef Shu X, Ji J, Li X, Sundquist J, Sundquist K, Hemminki K. Cancer risk among patients hospitalized for Type 1 diabetes mellitus: a population-based cohort study in Sweden. Diabet Med. 2010;27(7):791–7.PubMedCrossRef
14.
go back to reference Gordon-Dseagu VL, Shelton N, Mindell JS. Epidemiological evidence of a relationship between type-1 diabetes mellitus and cancer: a review of the existing literature. Int J Cancer. 2013;132(3):501–8.PubMedCrossRef Gordon-Dseagu VL, Shelton N, Mindell JS. Epidemiological evidence of a relationship between type-1 diabetes mellitus and cancer: a review of the existing literature. Int J Cancer. 2013;132(3):501–8.PubMedCrossRef
15.
go back to reference Harding JL, Shaw JE, Peeters A, Cartensen B, Magliano DJ. Cancer risk among people with type 1 and type 2 diabetes: Disentangling true associations, detection bias, and reverse causation. Diabetes Care. 2015;38(2):264–70.PubMedCrossRef Harding JL, Shaw JE, Peeters A, Cartensen B, Magliano DJ. Cancer risk among people with type 1 and type 2 diabetes: Disentangling true associations, detection bias, and reverse causation. Diabetes Care. 2015;38(2):264–70.PubMedCrossRef
16.
go back to reference Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R. Diabetes and cancer. Endocr Relat Cancer. 2009;16(4):1103–23.PubMedCrossRef Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R. Diabetes and cancer. Endocr Relat Cancer. 2009;16(4):1103–23.PubMedCrossRef
17.
go back to reference Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG, Yee D. Diabetes and cancer: a consensus report. Diabetes Care. 2010;33(7):1674–85.PubMedPubMedCentralCrossRef Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG, Yee D. Diabetes and cancer: a consensus report. Diabetes Care. 2010;33(7):1674–85.PubMedPubMedCentralCrossRef
18.
go back to reference Yeh HC, Platz EA, Wang NY, Visvanathan K, Helzlsouer KJ, Brancati FL. A prospective study of the associations between treated diabetes and cancer outcomes. Diabetes Care. 2012;35(1):113–8.PubMedCrossRef Yeh HC, Platz EA, Wang NY, Visvanathan K, Helzlsouer KJ, Brancati FL. A prospective study of the associations between treated diabetes and cancer outcomes. Diabetes Care. 2012;35(1):113–8.PubMedCrossRef
19.
go back to reference Friberg E, Orsini N, Mantzoros CS, Wolk A. Diabetes mellitus and risk of endometrial cancer: a meta-analysis. Diabetologia. 2007;50(7):1365–74.PubMedCrossRef Friberg E, Orsini N, Mantzoros CS, Wolk A. Diabetes mellitus and risk of endometrial cancer: a meta-analysis. Diabetologia. 2007;50(7):1365–74.PubMedCrossRef
20.
go back to reference Larsson SC, Orsini N, Wolk A. Diabetes mellitus and risk of colorectal cancer: a meta-analysis. J Natl Cancer Inst. 2005;97(22):1679–87.PubMedCrossRef Larsson SC, Orsini N, Wolk A. Diabetes mellitus and risk of colorectal cancer: a meta-analysis. J Natl Cancer Inst. 2005;97(22):1679–87.PubMedCrossRef
21.
go back to reference Larsson SC, Mantzoros CS, Wolk A. Diabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer. 2007;121(4):856–62.PubMedCrossRef Larsson SC, Mantzoros CS, Wolk A. Diabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer. 2007;121(4):856–62.PubMedCrossRef
22.
go back to reference Larsson SC, Orsini N, Brismar K, Wolk A. Diabetes mellitus and risk of bladder cancer: a meta-analysis. Diabetologia. 2006;49(12):2819–23.PubMedCrossRef Larsson SC, Orsini N, Brismar K, Wolk A. Diabetes mellitus and risk of bladder cancer: a meta-analysis. Diabetologia. 2006;49(12):2819–23.PubMedCrossRef
23.
go back to reference Larsson SC, Wolk A. Diabetes mellitus and incidence of kidney cancer: a meta-analysis of cohort studies. Diabetologia. 2011;54(5):1013–8.PubMedCrossRef Larsson SC, Wolk A. Diabetes mellitus and incidence of kidney cancer: a meta-analysis of cohort studies. Diabetologia. 2011;54(5):1013–8.PubMedCrossRef
24.
go back to reference Mitri J, Castillo J, Pittas AG. Diabetes and risk of Non-Hodgkin’s lymphoma: a meta-analysis of observational studies. Diabetes Care. 2008;31(12):2391–7.PubMedPubMedCentralCrossRef Mitri J, Castillo J, Pittas AG. Diabetes and risk of Non-Hodgkin’s lymphoma: a meta-analysis of observational studies. Diabetes Care. 2008;31(12):2391–7.PubMedPubMedCentralCrossRef
25.
go back to reference Kasper JS, Giovannucci E. A meta-analysis of diabetes mellitus and the risk of prostate cancer. Cancer Epidemiol Prev Biomark. 2006;15(11):2056–62.CrossRef Kasper JS, Giovannucci E. A meta-analysis of diabetes mellitus and the risk of prostate cancer. Cancer Epidemiol Prev Biomark. 2006;15(11):2056–62.CrossRef
26.
go back to reference Weiderpass E, Ye W, Vainio H, Kaaks R, Adami HO. Diabetes mellitus and ovarian cancer (Sweden). Cancer Causes Control. 2002;13(8):759–64.PubMedCrossRef Weiderpass E, Ye W, Vainio H, Kaaks R, Adami HO. Diabetes mellitus and ovarian cancer (Sweden). Cancer Causes Control. 2002;13(8):759–64.PubMedCrossRef
27.
go back to reference Hall GC, Roberts CM, Boulis M, Mo J, MacRae KD. Diabetes and the risk of lung cancer. Diabetes Care. 2005;28(3):590–4.PubMedCrossRef Hall GC, Roberts CM, Boulis M, Mo J, MacRae KD. Diabetes and the risk of lung cancer. Diabetes Care. 2005;28(3):590–4.PubMedCrossRef
28.
go back to reference Luo J, Chlebowski R, Wactawski-Wende J, Schlecht NF, Tinker L, Margolis KL. Diabetes and lung cancer among postmenopausal women. Diabetes Care. 2012;35(7):1485–91.PubMedPubMedCentralCrossRef Luo J, Chlebowski R, Wactawski-Wende J, Schlecht NF, Tinker L, Margolis KL. Diabetes and lung cancer among postmenopausal women. Diabetes Care. 2012;35(7):1485–91.PubMedPubMedCentralCrossRef
29.
go back to reference Lee J-Y, Jeon I, Lee JM, Yoon J-M, Park SM. Diabetes mellitus as an independent risk factor for lung cancer: a meta-analysis of observational studies. Eur J Cancer. 2013;49(10):2411–23.PubMedCrossRef Lee J-Y, Jeon I, Lee JM, Yoon J-M, Park SM. Diabetes mellitus as an independent risk factor for lung cancer: a meta-analysis of observational studies. Eur J Cancer. 2013;49(10):2411–23.PubMedCrossRef
30.
go back to reference Kurishima K, Watanabe H, Ishikawa H, Satoh H, Hizawa N. Survival of patients with lung cancer and diabetes mellitus. Mol Clin Oncol. 2017;6(6):907–10.PubMedPubMedCentralCrossRef Kurishima K, Watanabe H, Ishikawa H, Satoh H, Hizawa N. Survival of patients with lung cancer and diabetes mellitus. Mol Clin Oncol. 2017;6(6):907–10.PubMedPubMedCentralCrossRef
31.
go back to reference Tseng C-H. Higher risk of mortality from lung cancer in Taiwanese people with diabetes. Diabetes Res Clin Pract. 2013;102(3):193–201.PubMedCrossRef Tseng C-H. Higher risk of mortality from lung cancer in Taiwanese people with diabetes. Diabetes Res Clin Pract. 2013;102(3):193–201.PubMedCrossRef
32.
go back to reference Wan G, Yu X, Chen P, Wang X, Pan D, Wang X, Li L, Cai X, Cao F. Metformin therapy associated with survival benefit in lung cancer patients with diabetes. Oncotarget. 2016;7(23):35437–45.PubMedPubMedCentralCrossRef Wan G, Yu X, Chen P, Wang X, Pan D, Wang X, Li L, Cai X, Cao F. Metformin therapy associated with survival benefit in lung cancer patients with diabetes. Oncotarget. 2016;7(23):35437–45.PubMedPubMedCentralCrossRef
33.
go back to reference Yi ZH, Luther Y, Xiong GH, Ni YL, Yun F, Chen J, Yang Z, Zhang Q, Kuang YM, Zhu YC. Association between diabetes mellitus and lung cancer: meta-analysis. Eur J Clin Investig. 2020;50(10):e13332.CrossRef Yi ZH, Luther Y, Xiong GH, Ni YL, Yun F, Chen J, Yang Z, Zhang Q, Kuang YM, Zhu YC. Association between diabetes mellitus and lung cancer: meta-analysis. Eur J Clin Investig. 2020;50(10):e13332.CrossRef
34.
go back to reference Yamagata H, Kiyohara Y, Nakamura S, Kubo M, Tanizaki Y, Matsumoto T, Tanaka K, Kato I, Shirota T, Iida M. Impact of fasting plasma glucose levels on gastric cancer incidence in a general Japanese population: the hisayama study. Diabetes Care. 2005;28(4):789–94.PubMedCrossRef Yamagata H, Kiyohara Y, Nakamura S, Kubo M, Tanizaki Y, Matsumoto T, Tanaka K, Kato I, Shirota T, Iida M. Impact of fasting plasma glucose levels on gastric cancer incidence in a general Japanese population: the hisayama study. Diabetes Care. 2005;28(4):789–94.PubMedCrossRef
35.
go back to reference Johnson JA, Carstensen B, Witte D, Bowker SL, Lipscombe L, Renehan AG. Diabetes and cancer (1): evaluating the temporal relationship between type 2 diabetes and cancer incidence. Diabetologia. 2012;55(6):1607–18.PubMedCrossRef Johnson JA, Carstensen B, Witte D, Bowker SL, Lipscombe L, Renehan AG. Diabetes and cancer (1): evaluating the temporal relationship between type 2 diabetes and cancer incidence. Diabetologia. 2012;55(6):1607–18.PubMedCrossRef
36.
go back to reference Glicksman AS, Laird Myers WP, Rawson RW. Diabetes mellitus and carbohydrate metabolism in patients with cancer. Med Clin North Am. 1956;40(3):887–900.PubMedCrossRef Glicksman AS, Laird Myers WP, Rawson RW. Diabetes mellitus and carbohydrate metabolism in patients with cancer. Med Clin North Am. 1956;40(3):887–900.PubMedCrossRef
37.
go back to reference Kim JY, Lee YS, Jo G, Shin M-J. Glycated hemoglobin and cancer risk in korean adults: results from Korean genome and epidemiology study. Clin Nutr Res. 2018;7(3):170.PubMedPubMedCentralCrossRef Kim JY, Lee YS, Jo G, Shin M-J. Glycated hemoglobin and cancer risk in korean adults: results from Korean genome and epidemiology study. Clin Nutr Res. 2018;7(3):170.PubMedPubMedCentralCrossRef
38.
go back to reference Johnson JA, Bowker SL. Intensive glycaemic control and cancer risk in type 2 diabetes: a meta-analysis of major trials. Diabetologia. 2010;54(1):25–31.PubMedCrossRef Johnson JA, Bowker SL. Intensive glycaemic control and cancer risk in type 2 diabetes: a meta-analysis of major trials. Diabetologia. 2010;54(1):25–31.PubMedCrossRef
39.
go back to reference Shikata K, Ninomiya T, Kiyohara Y. Diabetes mellitus and cancer risk: review of the epidemiological evidence. Cancer Sci. 2013;104(1):9–14.PubMedCrossRef Shikata K, Ninomiya T, Kiyohara Y. Diabetes mellitus and cancer risk: review of the epidemiological evidence. Cancer Sci. 2013;104(1):9–14.PubMedCrossRef
40.
go back to reference Schulze PC, Yoshioka J, Takahashi T, He Z, King GL, Lee RT. Hyperglycemia promotes oxidative stress through inhibition of thioredoxin function by thioredoxin-interacting protein. J Biol Chem. 2004;279(29):30369–74.PubMedCrossRef Schulze PC, Yoshioka J, Takahashi T, He Z, King GL, Lee RT. Hyperglycemia promotes oxidative stress through inhibition of thioredoxin function by thioredoxin-interacting protein. J Biol Chem. 2004;279(29):30369–74.PubMedCrossRef
41.
go back to reference Turturro F, Friday E, Welbourne T. Hyperglycemia regulates thioredoxin-ROS activity through induction of thioredoxin-interacting protein (TXNIP) in metastatic breast cancer-derived cells MDA-MB-231. BMC Cancer. 2007;7(1):1–7.CrossRef Turturro F, Friday E, Welbourne T. Hyperglycemia regulates thioredoxin-ROS activity through induction of thioredoxin-interacting protein (TXNIP) in metastatic breast cancer-derived cells MDA-MB-231. BMC Cancer. 2007;7(1):1–7.CrossRef
42.
go back to reference Dunn LL, Simpson PJL, Prosser HC, Lecce L, Yuen GSC, Buckle A, Sieveking DP, Vanags LZ, Lim PR, Chow RWY, et al. A critical role for thioredoxin-interacting protein in diabetes-related impairment of angiogenesis. Diabetes. 2013;63(2):675–87.PubMedCrossRef Dunn LL, Simpson PJL, Prosser HC, Lecce L, Yuen GSC, Buckle A, Sieveking DP, Vanags LZ, Lim PR, Chow RWY, et al. A critical role for thioredoxin-interacting protein in diabetes-related impairment of angiogenesis. Diabetes. 2013;63(2):675–87.PubMedCrossRef
43.
go back to reference Becker S, Dossus L, Kaaks R. Obesity related hyperinsulinaemia and hyperglycaemia and cancer development. Arch Physiol Biochem. 2009;115(2):86–96.PubMedCrossRef Becker S, Dossus L, Kaaks R. Obesity related hyperinsulinaemia and hyperglycaemia and cancer development. Arch Physiol Biochem. 2009;115(2):86–96.PubMedCrossRef
44.
go back to reference Schröter D, Höhn A. Role of advanced glycation end products in carcinogenesis and their therapeutic implications. Curr Pharm Des. 2019;24(44):5245–51.CrossRef Schröter D, Höhn A. Role of advanced glycation end products in carcinogenesis and their therapeutic implications. Curr Pharm Des. 2019;24(44):5245–51.CrossRef
45.
go back to reference Sebekova K, Wagner Z, Schupp N, Boor P. Genomic damage and malignancy in end-stage renal failure: do advanced glycation end products contribute? Kidney Blood Press Res. 2007;30(1):56–66.PubMedCrossRef Sebekova K, Wagner Z, Schupp N, Boor P. Genomic damage and malignancy in end-stage renal failure: do advanced glycation end products contribute? Kidney Blood Press Res. 2007;30(1):56–66.PubMedCrossRef
46.
go back to reference Tan ALY, Forbes JM, Cooper ME. AGE, RAGE, and ROS in diabetic nephropathy. Semin Nephrol. 2007;27(2):130–43.PubMedCrossRef Tan ALY, Forbes JM, Cooper ME. AGE, RAGE, and ROS in diabetic nephropathy. Semin Nephrol. 2007;27(2):130–43.PubMedCrossRef
47.
48.
go back to reference Yamagishi SI, Imaizumi T. Diabetic vascular complications: Pathophysiology, biochemical basis and potential therapeutic strategy. Curr Pharm Des. 2005;11(18):2279–99.PubMedCrossRef Yamagishi SI, Imaizumi T. Diabetic vascular complications: Pathophysiology, biochemical basis and potential therapeutic strategy. Curr Pharm Des. 2005;11(18):2279–99.PubMedCrossRef
49.
go back to reference Warburg O. The metabolism of carcinoma cells. J Cancer Res. 1925;9(1):148–63.CrossRef Warburg O. The metabolism of carcinoma cells. J Cancer Res. 1925;9(1):148–63.CrossRef
50.
go back to reference Krone CA, Ely JT. Controlling hyperglycemia as an adjunct to cancer therapy. Integr Cancer Ther. 2005;4(1):25–31.PubMedCrossRef Krone CA, Ely JT. Controlling hyperglycemia as an adjunct to cancer therapy. Integr Cancer Ther. 2005;4(1):25–31.PubMedCrossRef
51.
go back to reference Durak-Kozica M, Paszek E, Stepien EL. Role of the Wnt signaling pathway in the development of endothelial disorders in response to hyperglycaemia. Expert Rev Mol Med. 2019;21:e7.PubMedCrossRef Durak-Kozica M, Paszek E, Stepien EL. Role of the Wnt signaling pathway in the development of endothelial disorders in response to hyperglycaemia. Expert Rev Mol Med. 2019;21:e7.PubMedCrossRef
52.
go back to reference García-Jiménez C, García-Martínez JM, Chocarro-Calvo A, De la Vieja A. A new link between diabetes and cancer: enhanced WNT/β-catenin signaling by high glucose. J Mol Endocrinol. 2014;52(1):R51–66.PubMedCrossRef García-Jiménez C, García-Martínez JM, Chocarro-Calvo A, De la Vieja A. A new link between diabetes and cancer: enhanced WNT/β-catenin signaling by high glucose. J Mol Endocrinol. 2014;52(1):R51–66.PubMedCrossRef
53.
go back to reference Heuson JC, Legros N, Heimann R. Influence of insulin administration on growth of the 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in intact, oophorectomized, and hypophysectomized rats. Cancer Res. 1972;32(2):233–8.PubMed Heuson JC, Legros N, Heimann R. Influence of insulin administration on growth of the 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in intact, oophorectomized, and hypophysectomized rats. Cancer Res. 1972;32(2):233–8.PubMed
54.
go back to reference Frasca F, Pandini G, Sciacca L, Pezzino V, Squatrito S, Belfiore A, Vigneri R. The role of insulin receptors and IGF-I receptors in cancer and other diseases. Arch Physiol Biochem. 2008;114(1):23–37.PubMedCrossRef Frasca F, Pandini G, Sciacca L, Pezzino V, Squatrito S, Belfiore A, Vigneri R. The role of insulin receptors and IGF-I receptors in cancer and other diseases. Arch Physiol Biochem. 2008;114(1):23–37.PubMedCrossRef
55.
go back to reference Cox ME, Gleave ME, Zakikhani M, Bell RH, Piura E, Vickers E, Cunningham M, Larsson O, Fazli L, Pollak M. Insulin receptor expression by human prostate cancers. Prostate. 2009;69(1):33–40.PubMedCrossRef Cox ME, Gleave ME, Zakikhani M, Bell RH, Piura E, Vickers E, Cunningham M, Larsson O, Fazli L, Pollak M. Insulin receptor expression by human prostate cancers. Prostate. 2009;69(1):33–40.PubMedCrossRef
56.
go back to reference Law JH, Habibi G, Hu K, Masoudi H, Wang MYC, Stratford AL, Park E, Gee JMW, Finlay P, Jones HE, et al. Phosphorylated insulin-like growth factor-I/insulin receptor is present in all breast cancer subtypes and is related to poor survival. Can Res. 2008;68(24):10238–46.CrossRef Law JH, Habibi G, Hu K, Masoudi H, Wang MYC, Stratford AL, Park E, Gee JMW, Finlay P, Jones HE, et al. Phosphorylated insulin-like growth factor-I/insulin receptor is present in all breast cancer subtypes and is related to poor survival. Can Res. 2008;68(24):10238–46.CrossRef
57.
go back to reference Werner H, Weinstein D, Bentov I. Similarities and differences between insulin and IGF-I: structures, receptors, and signaling pathways. Arch Physiol Biochem. 2008;114(1):17–22.PubMedCrossRef Werner H, Weinstein D, Bentov I. Similarities and differences between insulin and IGF-I: structures, receptors, and signaling pathways. Arch Physiol Biochem. 2008;114(1):17–22.PubMedCrossRef
58.
go back to reference Soos MA, Field CE, Siddle K. Purified hybrid insulin/insulin-like growth factor-I receptors bind insulin-like growth factor-I, but not insulin, with high affinity. Biochem J. 1993;290(2):419–26.PubMedPubMedCentralCrossRef Soos MA, Field CE, Siddle K. Purified hybrid insulin/insulin-like growth factor-I receptors bind insulin-like growth factor-I, but not insulin, with high affinity. Biochem J. 1993;290(2):419–26.PubMedPubMedCentralCrossRef
59.
go back to reference Clemmons DR, Maile LA, Ling Y, Yarber J, Busby WH. Role of the integrin αVβ3 in mediating increased smooth muscle cell responsiveness to IGF-I in response to hyperglycemic stress. Growth Hormon IGF Res. 2007;17(4):265–70.CrossRef Clemmons DR, Maile LA, Ling Y, Yarber J, Busby WH. Role of the integrin αVβ3 in mediating increased smooth muscle cell responsiveness to IGF-I in response to hyperglycemic stress. Growth Hormon IGF Res. 2007;17(4):265–70.CrossRef
60.
go back to reference Giovannucci E. Insulin, insulin-like growth factors and colon cancer: a review of the evidence. J Nutr. 2001;131(11):3109S-3120S.PubMedCrossRef Giovannucci E. Insulin, insulin-like growth factors and colon cancer: a review of the evidence. J Nutr. 2001;131(11):3109S-3120S.PubMedCrossRef
61.
go back to reference Powell DR, Suwanichkul A, Cubbage ML, DePaolis LA, Snuggs MB, Lee PD. Insulin inhibits transcription of the human gene for insulin-like growth factor-binding protein-1. J Biol Chem. 1991;266(28):18868–76.PubMedCrossRef Powell DR, Suwanichkul A, Cubbage ML, DePaolis LA, Snuggs MB, Lee PD. Insulin inhibits transcription of the human gene for insulin-like growth factor-binding protein-1. J Biol Chem. 1991;266(28):18868–76.PubMedCrossRef
62.
go back to reference Renehan AG, Frystyk J, Flyvbjerg A. Obesity and cancer risk: the role of the insulin-IGF axis. Trends Endocrinol Metab. 2006;17(8):328–36.PubMedCrossRef Renehan AG, Frystyk J, Flyvbjerg A. Obesity and cancer risk: the role of the insulin-IGF axis. Trends Endocrinol Metab. 2006;17(8):328–36.PubMedCrossRef
63.
go back to reference Weinstein D, Simon M, Yehezkel E, Laron Z, Werner H. Insulin analogues display IGF-I-like mitogenic and anti-apoptotic activities in cultured cancer cells. Diabetes Metab Res Rev. 2009;25(1):41–9.PubMedCrossRef Weinstein D, Simon M, Yehezkel E, Laron Z, Werner H. Insulin analogues display IGF-I-like mitogenic and anti-apoptotic activities in cultured cancer cells. Diabetes Metab Res Rev. 2009;25(1):41–9.PubMedCrossRef
64.
go back to reference Pollak M. Insulin and insulin-like growth factor signaling in neoplasia. Nat Rev Cancer. 2008;8(12):915–28.PubMedCrossRef Pollak M. Insulin and insulin-like growth factor signaling in neoplasia. Nat Rev Cancer. 2008;8(12):915–28.PubMedCrossRef
65.
go back to reference Renehan AG, Howell A. Preventing cancer, cardiovascular disease, and diabetes. The Lancet. 2005;365(9469):1449–51.CrossRef Renehan AG, Howell A. Preventing cancer, cardiovascular disease, and diabetes. The Lancet. 2005;365(9469):1449–51.CrossRef
66.
go back to reference Lipscombe LL, Lega IC. Review: Diabetes, obesity, and cancer—pathophysiology and clinical implications. Endocr Rev. 2020;41(1):33–52.CrossRef Lipscombe LL, Lega IC. Review: Diabetes, obesity, and cancer—pathophysiology and clinical implications. Endocr Rev. 2020;41(1):33–52.CrossRef
67.
68.
go back to reference Nam SY, Lee EJ, Kim KR, Cha BS, Song YD, Lim SK, Lee HC, Huh KB. Effect of obesity on total and free insulin-like growth factor (IGF)-1, and their relationship to IGF-binding protein (BP)-1, IGFBP-2, IGFBP-3, insulin, and growth hormone. Int J Obes. 1997;21(5):355–9.CrossRef Nam SY, Lee EJ, Kim KR, Cha BS, Song YD, Lim SK, Lee HC, Huh KB. Effect of obesity on total and free insulin-like growth factor (IGF)-1, and their relationship to IGF-binding protein (BP)-1, IGFBP-2, IGFBP-3, insulin, and growth hormone. Int J Obes. 1997;21(5):355–9.CrossRef
69.
go back to reference Kulik G, Klippel A, Weber MJ. Antiapoptotic signaling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol Cell Biol. 1997;17(3):1595–606.PubMedPubMedCentralCrossRef Kulik G, Klippel A, Weber MJ. Antiapoptotic signaling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol Cell Biol. 1997;17(3):1595–606.PubMedPubMedCentralCrossRef
70.
go back to reference Frystyk J. Free insulin-like growth factors—measurements and relationships to growth hormone secretion and glucose homeostasis. Growth Hormon IGF Res. 2004;14(5):337–75.CrossRef Frystyk J. Free insulin-like growth factors—measurements and relationships to growth hormone secretion and glucose homeostasis. Growth Hormon IGF Res. 2004;14(5):337–75.CrossRef
71.
go back to reference Lai GY, Giovannucci EL, Pollak MN, Peskoe SB, Stampfer MJ, Willett WC, Platz EA. Association of C-peptide and leptin with prostate cancer incidence in the Health Professionals Follow-up Study. Cancer Causes Control. 2014;25(5):625–32.PubMedPubMedCentralCrossRef Lai GY, Giovannucci EL, Pollak MN, Peskoe SB, Stampfer MJ, Willett WC, Platz EA. Association of C-peptide and leptin with prostate cancer incidence in the Health Professionals Follow-up Study. Cancer Causes Control. 2014;25(5):625–32.PubMedPubMedCentralCrossRef
72.
go back to reference Stevens VL, Jacobs EJ, Sun J, Gapstur SM. No association of plasma levels of adiponectin and C-peptide with risk of aggressive prostate cancer in the cancer prevention study ii nutrition cohort. Cancer Epidemiol Biomark Prev. 2014;23(5):890–2.CrossRef Stevens VL, Jacobs EJ, Sun J, Gapstur SM. No association of plasma levels of adiponectin and C-peptide with risk of aggressive prostate cancer in the cancer prevention study ii nutrition cohort. Cancer Epidemiol Biomark Prev. 2014;23(5):890–2.CrossRef
73.
go back to reference Ma J, Li H, Giovannucci E, Mucci L, Qiu W, Nguyen PL, Gaziano JM, Pollak M, Stampfer MJ. Prediagnostic body-mass index, plasma C-peptide concentration, and prostate cancer-specific mortality in men with prostate cancer: a long-term survival analysis. Lancet Oncol. 2008;9(11):1039–47.PubMedPubMedCentralCrossRef Ma J, Li H, Giovannucci E, Mucci L, Qiu W, Nguyen PL, Gaziano JM, Pollak M, Stampfer MJ. Prediagnostic body-mass index, plasma C-peptide concentration, and prostate cancer-specific mortality in men with prostate cancer: a long-term survival analysis. Lancet Oncol. 2008;9(11):1039–47.PubMedPubMedCentralCrossRef
74.
75.
go back to reference Parida S, Siddharth S, Sharma D. Adiponectin, obesity, and cancer: clash of the bigwigs in health and disease. Int J Mol Sci. 2019;20(10):2519.PubMedCentralCrossRef Parida S, Siddharth S, Sharma D. Adiponectin, obesity, and cancer: clash of the bigwigs in health and disease. Int J Mol Sci. 2019;20(10):2519.PubMedCentralCrossRef
76.
77.
go back to reference Stattin P, Lukanova A, Biessy C, Söderberg S, Palmqvist R, Kaaks R, Olsson T, Jellum E. Obesity and colon cancer: does leptin provide a link? Int J Cancer. 2004;109(1):149–52.PubMedCrossRef Stattin P, Lukanova A, Biessy C, Söderberg S, Palmqvist R, Kaaks R, Olsson T, Jellum E. Obesity and colon cancer: does leptin provide a link? Int J Cancer. 2004;109(1):149–52.PubMedCrossRef
78.
go back to reference Wu MH, Chou YC, Chou WY, Hsu GC, Chu CH, Yu CP, Yu JC, Sun CA. Circulating levels of leptin, adiposity and breast cancer risk. Br J Cancer. 2009;100(4):578–82.PubMedPubMedCentralCrossRef Wu MH, Chou YC, Chou WY, Hsu GC, Chu CH, Yu CP, Yu JC, Sun CA. Circulating levels of leptin, adiposity and breast cancer risk. Br J Cancer. 2009;100(4):578–82.PubMedPubMedCentralCrossRef
79.
go back to reference Mantzoros CS, Bolhke K, Moschos S, Cramer DW. Leptin in relation to carcinomaIn situ of the breast: a study of pre-menopausal cases and controls. Int J Cancer. 1999;80(4):523–6.PubMedCrossRef Mantzoros CS, Bolhke K, Moschos S, Cramer DW. Leptin in relation to carcinomaIn situ of the breast: a study of pre-menopausal cases and controls. Int J Cancer. 1999;80(4):523–6.PubMedCrossRef
81.
go back to reference Lee JY, Sohn KH, Rhee SH, Hwang D. Saturated fatty acids, but not unsaturated fatty acids, induce the expression of cyclooxygenase-2 mediated through toll-like receptor 4. J Biol Chem. 2001;276(20):16683–9.CrossRefPubMed Lee JY, Sohn KH, Rhee SH, Hwang D. Saturated fatty acids, but not unsaturated fatty acids, induce the expression of cyclooxygenase-2 mediated through toll-like receptor 4. J Biol Chem. 2001;276(20):16683–9.CrossRefPubMed
82.
go back to reference Ramos EJB, Xu Y, Romanova I, Middleton F, Chen C, Quinn R, Inui A, Das U, Meguid MM. Is obesity an inflammatory disease? Surgery. 2003;134(2):329–35.PubMedCrossRef Ramos EJB, Xu Y, Romanova I, Middleton F, Chen C, Quinn R, Inui A, Das U, Meguid MM. Is obesity an inflammatory disease? Surgery. 2003;134(2):329–35.PubMedCrossRef
83.
go back to reference Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK, Du B, Brogi E, Crawford CB, Kopelovich L, et al. Inflammation and increased aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev Res. 2011;4(7):1021–9.CrossRef Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK, Du B, Brogi E, Crawford CB, Kopelovich L, et al. Inflammation and increased aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev Res. 2011;4(7):1021–9.CrossRef
84.
go back to reference Wu Q, Arner E, Forrest ARR, Ehrlund A, Mejhert N, Itoh M, Kawaji H, Lassmann T, Laurencikiene J, Rydén M, et al. Ceruloplasmin is a novel adipokine which is overexpressed in adipose tissue of obese subjects and in obesity-associated cancer cells. PLoS ONE. 2014;9(3):e80274.CrossRef Wu Q, Arner E, Forrest ARR, Ehrlund A, Mejhert N, Itoh M, Kawaji H, Lassmann T, Laurencikiene J, Rydén M, et al. Ceruloplasmin is a novel adipokine which is overexpressed in adipose tissue of obese subjects and in obesity-associated cancer cells. PLoS ONE. 2014;9(3):e80274.CrossRef
85.
go back to reference Yang W, Yan H-X, Chen L, Liu Q, He Y-Q, Yu L-X, Zhang S-H, Huang D-D, Tang L, Kong X-N, et al. Wnt/β-catenin signaling contributes to activation of normal and tumorigenic liver progenitor cells. Can Res. 2008;68(11):4287–95.CrossRef Yang W, Yan H-X, Chen L, Liu Q, He Y-Q, Yu L-X, Zhang S-H, Huang D-D, Tang L, Kong X-N, et al. Wnt/β-catenin signaling contributes to activation of normal and tumorigenic liver progenitor cells. Can Res. 2008;68(11):4287–95.CrossRef
86.
go back to reference Martin-Orozco E, Sanchez-Fernandez A, Ortiz-Parra I, Ayala-San Nicolas M. WNT signaling in tumors: the way to evade drugs and immunity. Front Immunol. 2019;10:2854.PubMedPubMedCentralCrossRef Martin-Orozco E, Sanchez-Fernandez A, Ortiz-Parra I, Ayala-San Nicolas M. WNT signaling in tumors: the way to evade drugs and immunity. Front Immunol. 2019;10:2854.PubMedPubMedCentralCrossRef
88.
go back to reference Welters HJ, Kulkarni RN. Wnt signaling: relevance to β-cell biology and diabetes. Trends Endocrinol Metab. 2008;19(10):349–55.PubMedCrossRef Welters HJ, Kulkarni RN. Wnt signaling: relevance to β-cell biology and diabetes. Trends Endocrinol Metab. 2008;19(10):349–55.PubMedCrossRef
89.
go back to reference Monga SP. β-catenin signaling and roles in liver homeostasis, injury, and tumorigenesis. Gastroenterology. 2015;148(7):1294–310.PubMedCrossRef Monga SP. β-catenin signaling and roles in liver homeostasis, injury, and tumorigenesis. Gastroenterology. 2015;148(7):1294–310.PubMedCrossRef
90.
go back to reference Duchartre Y, Kim Y-M, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol. 2016;99:141–9.PubMedCrossRef Duchartre Y, Kim Y-M, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol. 2016;99:141–9.PubMedCrossRef
91.
go back to reference Seifert JRK, Mlodzik M. Frizzled/PCP signaling: a conserved mechanism regulating cell polarity and directed motility. Nat Rev Genet. 2007;8(2):126–38.PubMedCrossRef Seifert JRK, Mlodzik M. Frizzled/PCP signaling: a conserved mechanism regulating cell polarity and directed motility. Nat Rev Genet. 2007;8(2):126–38.PubMedCrossRef
92.
go back to reference Chocarro-Calvo A, García-Martínez Jose M, Ardila-González S, De la Vieja A, García-Jiménez C. Glucose-induced β-catenin acetylation enhances Wnt signaling in cancer. Mol Cell. 2013;49(3):474–86.PubMedCrossRef Chocarro-Calvo A, García-Martínez Jose M, Ardila-González S, De la Vieja A, García-Jiménez C. Glucose-induced β-catenin acetylation enhances Wnt signaling in cancer. Mol Cell. 2013;49(3):474–86.PubMedCrossRef
93.
go back to reference Krzeslak A, Wojcik-Krowiranda K, Forma E, Jozwiak P, Romanowicz H, Bienkiewicz A, Brys M. Expression of GLUT1 and GLUT3 glucose transporters in endometrial and breast cancers. Pathol Oncol Res. 2012;18(3):721–8.PubMedCrossRef Krzeslak A, Wojcik-Krowiranda K, Forma E, Jozwiak P, Romanowicz H, Bienkiewicz A, Brys M. Expression of GLUT1 and GLUT3 glucose transporters in endometrial and breast cancers. Pathol Oncol Res. 2012;18(3):721–8.PubMedCrossRef
95.
96.
go back to reference Hao H-X, Xie Y, Zhang Y, Charlat O, Oster E, Avello M, Lei H, Mickanin C, Liu D, Ruffner H, et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature. 2012;485(7397):195–200.PubMedCrossRef Hao H-X, Xie Y, Zhang Y, Charlat O, Oster E, Avello M, Lei H, Mickanin C, Liu D, Ruffner H, et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature. 2012;485(7397):195–200.PubMedCrossRef
97.
go back to reference Koo B-K, Spit M, Jordens I, Low TY, Stange DE, van de Wetering M, van Es JH, Mohammed S, Heck AJR, Maurice MM, et al. Tumor suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature. 2012;488(7413):665–9.PubMedCrossRef Koo B-K, Spit M, Jordens I, Low TY, Stange DE, van de Wetering M, van Es JH, Mohammed S, Heck AJR, Maurice MM, et al. Tumor suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature. 2012;488(7413):665–9.PubMedCrossRef
98.
go back to reference Chiarini F, Paganelli F, Martelli AM, Evangelisti C. The role played by Wnt/β-catenin signaling pathway in acute lymphoblastic leukemia. Int J Mol Sci. 2020;21(3):1098.PubMedCentralCrossRef Chiarini F, Paganelli F, Martelli AM, Evangelisti C. The role played by Wnt/β-catenin signaling pathway in acute lymphoblastic leukemia. Int J Mol Sci. 2020;21(3):1098.PubMedCentralCrossRef
99.
go back to reference Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, Chaudhuri S, Guan Y, Janakiraman V, Jaiswal BS, et al. Recurrent R-spondin fusions in colon cancer. Nature. 2012;488(7413):660–4.PubMedPubMedCentralCrossRef Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, Chaudhuri S, Guan Y, Janakiraman V, Jaiswal BS, et al. Recurrent R-spondin fusions in colon cancer. Nature. 2012;488(7413):660–4.PubMedPubMedCentralCrossRef
100.
go back to reference Zhang L, Song Y, Ling Z, Li Y, Ren X, Yang J, Wang Z, Xia J, Zhang W, Cheng B. R-spondin 2-LGR4 system regulates growth, migration and invasion, epithelial-mesenchymal transition and stem-like properties of tongue squamous cell carcinoma via Wnt/β-catenin signaling. EBioMedicine. 2019;44:275–88.PubMedPubMedCentralCrossRef Zhang L, Song Y, Ling Z, Li Y, Ren X, Yang J, Wang Z, Xia J, Zhang W, Cheng B. R-spondin 2-LGR4 system regulates growth, migration and invasion, epithelial-mesenchymal transition and stem-like properties of tongue squamous cell carcinoma via Wnt/β-catenin signaling. EBioMedicine. 2019;44:275–88.PubMedPubMedCentralCrossRef
101.
go back to reference Wu J, Jiao Y, Dal Molin M, Maitra A, de Wilde RF, Wood LD, Eshleman JR, Goggins MG, Wolfgang CL, Canto MI, et al. Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitin-dependent pathways. Proc Natl Acad Sci. 2011;108(52):21188–93.PubMedPubMedCentralCrossRef Wu J, Jiao Y, Dal Molin M, Maitra A, de Wilde RF, Wood LD, Eshleman JR, Goggins MG, Wolfgang CL, Canto MI, et al. Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitin-dependent pathways. Proc Natl Acad Sci. 2011;108(52):21188–93.PubMedPubMedCentralCrossRef
104.
go back to reference Tian T, Li X, Zhang J. mTOR signaling in cancer and mTOR Inhibitors in solid tumor targeting therapy. Int J Mol Sci. 2019;20(3):755.PubMedCentralCrossRef Tian T, Li X, Zhang J. mTOR signaling in cancer and mTOR Inhibitors in solid tumor targeting therapy. Int J Mol Sci. 2019;20(3):755.PubMedCentralCrossRef
105.
go back to reference Kaur A, Sharma S. Mammalian target of rapamycin (mTOR) as a potential therapeutic target in various diseases. Inflammopharmacology. 2017;25(3):293–312.PubMedCrossRef Kaur A, Sharma S. Mammalian target of rapamycin (mTOR) as a potential therapeutic target in various diseases. Inflammopharmacology. 2017;25(3):293–312.PubMedCrossRef
107.
108.
go back to reference Dufour M, Dormond-Meuwly A, Demartines N, Dormond O. Targeting the mammalian target of Rapamycin (mTOR) in cancer therapy: Lessons from past and future perspectives. Cancers. 2011;3(2):2478–500.PubMedPubMedCentralCrossRef Dufour M, Dormond-Meuwly A, Demartines N, Dormond O. Targeting the mammalian target of Rapamycin (mTOR) in cancer therapy: Lessons from past and future perspectives. Cancers. 2011;3(2):2478–500.PubMedPubMedCentralCrossRef
109.
111.
go back to reference García-Martínez Juan M, Alessi Dario R. mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1). Biochem J. 2008;416(3):375–85.PubMedCrossRef García-Martínez Juan M, Alessi Dario R. mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1). Biochem J. 2008;416(3):375–85.PubMedCrossRef
113.
go back to reference Hanahan D, Weinberg Robert A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef Hanahan D, Weinberg Robert A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef
114.
go back to reference Grabiner BC, Nardi V, Birsoy K, Possemato R, Shen K, Sinha S, Jordan A, Beck AH, Sabatini DM. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict Rapamycin sensitivity. Cancer Discov. 2014;4(5):554–63.PubMedPubMedCentralCrossRef Grabiner BC, Nardi V, Birsoy K, Possemato R, Shen K, Sinha S, Jordan A, Beck AH, Sabatini DM. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict Rapamycin sensitivity. Cancer Discov. 2014;4(5):554–63.PubMedPubMedCentralCrossRef
115.
go back to reference Lim K-H, Counter CM. Reduction in the requirement of oncogenic Ras signaling to activation of PI3K/AKT pathway during tumor maintenance. Cancer Cell. 2005;8(5):381–92.PubMedCrossRef Lim K-H, Counter CM. Reduction in the requirement of oncogenic Ras signaling to activation of PI3K/AKT pathway during tumor maintenance. Cancer Cell. 2005;8(5):381–92.PubMedCrossRef
116.
117.
go back to reference Takei Y, Saga Y, Mizukami H, Takayama T, Ohwada M, Ozawa K, Suzuki M. Overexpression of PTEN in ovarian cancer cells suppresses i.p. dissemination and extends survival in mice. Mol Cancer Ther. 2008;7(3):704–11.PubMedCrossRef Takei Y, Saga Y, Mizukami H, Takayama T, Ohwada M, Ozawa K, Suzuki M. Overexpression of PTEN in ovarian cancer cells suppresses i.p. dissemination and extends survival in mice. Mol Cancer Ther. 2008;7(3):704–11.PubMedCrossRef
119.
go back to reference Milella M, Falcone I, Conciatori F, Cesta Incani U, Del Curatolo A, Inzerilli N, Nuzzo CMA, Vaccaro V, Vari S, Cognetti F, et al. PTEN: multiple functions in human malignant tumors. Front Oncol. 2015;5:24.PubMedPubMedCentralCrossRef Milella M, Falcone I, Conciatori F, Cesta Incani U, Del Curatolo A, Inzerilli N, Nuzzo CMA, Vaccaro V, Vari S, Cognetti F, et al. PTEN: multiple functions in human malignant tumors. Front Oncol. 2015;5:24.PubMedPubMedCentralCrossRef
120.
go back to reference Showkat M, Beigh MA, Andrabi KI. mTOR signaling in protein translation regulation: implications in cancer genesis and therapeutic interventions. Mol Biol Int. 2014;2014:1–14.CrossRef Showkat M, Beigh MA, Andrabi KI. mTOR signaling in protein translation regulation: implications in cancer genesis and therapeutic interventions. Mol Biol Int. 2014;2014:1–14.CrossRef
121.
go back to reference Mannucci E. Insulin therapy and cancer in type 2 diabetes. ISRN Endocrinol. 2012;2012:1–12.CrossRef Mannucci E. Insulin therapy and cancer in type 2 diabetes. ISRN Endocrinol. 2012;2012:1–12.CrossRef
122.
go back to reference Joung KH, Jeong J-W, Ku BJ. The association between type 2 diabetes mellitus and women cancer: the epidemiological evidences and putative mechanisms. Biomed Res Int. 2015;2015:1–12.CrossRef Joung KH, Jeong J-W, Ku BJ. The association between type 2 diabetes mellitus and women cancer: the epidemiological evidences and putative mechanisms. Biomed Res Int. 2015;2015:1–12.CrossRef
123.
125.
go back to reference Bishayee A. The inflammation and liver cancer. In: Aggarwal BB, Sung B, Gupta SC, editors. Inflammation and cancer, vol. 816. Swetzerland: Springer, Basel; 2014. p. 401–35.CrossRef Bishayee A. The inflammation and liver cancer. In: Aggarwal BB, Sung B, Gupta SC, editors. Inflammation and cancer, vol. 816. Swetzerland: Springer, Basel; 2014. p. 401–35.CrossRef
126.
go back to reference Hausmann S, Kong B, Michalski C, Erkan M, Friess H. The role of inflammation in pancreatic cancer. In: Aggarwal BB, Sung B, Gupta SC, editors. Inflammation and cancer, vol. 816. Swetzerland: Springer, Basel; 2014. p. 129–51.CrossRef Hausmann S, Kong B, Michalski C, Erkan M, Friess H. The role of inflammation in pancreatic cancer. In: Aggarwal BB, Sung B, Gupta SC, editors. Inflammation and cancer, vol. 816. Swetzerland: Springer, Basel; 2014. p. 129–51.CrossRef
127.
128.
go back to reference Naugler WE, Karin M. The wolf in sheep’s clothing: the role of interleukin-6 in immunity, inflammation and cancer. Trends Mol Med. 2008;14(3):109–19.PubMedCrossRef Naugler WE, Karin M. The wolf in sheep’s clothing: the role of interleukin-6 in immunity, inflammation and cancer. Trends Mol Med. 2008;14(3):109–19.PubMedCrossRef
130.
go back to reference Jung IH, Choi JH-K, Chung Y-Y, Lim G-L, Park Y-N, Park SW. Predominant activation of JAK/STAT3 pathway by interleukin-6 is implicated in hepatocarcinogenesis. Neoplasia. 2015;17(7):586–97.PubMedPubMedCentralCrossRef Jung IH, Choi JH-K, Chung Y-Y, Lim G-L, Park Y-N, Park SW. Predominant activation of JAK/STAT3 pathway by interleukin-6 is implicated in hepatocarcinogenesis. Neoplasia. 2015;17(7):586–97.PubMedPubMedCentralCrossRef
131.
go back to reference Ando M, Uehara I, Kogure K, Asano Y, Nakajima W, Abe Y, Kawauchi K, Tanaka N. Interleukin 6 enhances glycolysis through expression of the glycolytic enzymes hexokinase 2 and 6-phosphofructo-2-kinase/Fructose-2,6-bisphosphatase-3. J Nippon Med Sch. 2010;77(2):97–105.PubMedCrossRef Ando M, Uehara I, Kogure K, Asano Y, Nakajima W, Abe Y, Kawauchi K, Tanaka N. Interleukin 6 enhances glycolysis through expression of the glycolytic enzymes hexokinase 2 and 6-phosphofructo-2-kinase/Fructose-2,6-bisphosphatase-3. J Nippon Med Sch. 2010;77(2):97–105.PubMedCrossRef
132.
go back to reference Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Investig. 2003;112(12):1796–808.PubMedPubMedCentralCrossRef Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Investig. 2003;112(12):1796–808.PubMedPubMedCentralCrossRef
133.
134.
go back to reference Shimizu M, Tanaka T, Moriwaki H. Obesity and hepatocellular carcinoma: targeting obesity-related inflammation for chemoprevention of liver carcinogenesis. Semin Immunopathol. 2012;35(2):191–202.PubMedCrossRef Shimizu M, Tanaka T, Moriwaki H. Obesity and hepatocellular carcinoma: targeting obesity-related inflammation for chemoprevention of liver carcinogenesis. Semin Immunopathol. 2012;35(2):191–202.PubMedCrossRef
135.
go back to reference Fève B, Bastard J-P. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat Rev Endocrinol. 2009;5(6):305–11.PubMedCrossRef Fève B, Bastard J-P. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat Rev Endocrinol. 2009;5(6):305–11.PubMedCrossRef
136.
go back to reference Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.PubMedCrossRef Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.PubMedCrossRef
137.
go back to reference Takamori H, Oades ZG, Hoch RC, Burger M, Schraufstatter IU. Autocrine growth effect of IL-8 and GRO? On a human pancreatic cancer cell line, capan-1. Pancreas. 2000;21(1):52–6.PubMedCrossRef Takamori H, Oades ZG, Hoch RC, Burger M, Schraufstatter IU. Autocrine growth effect of IL-8 and GRO? On a human pancreatic cancer cell line, capan-1. Pancreas. 2000;21(1):52–6.PubMedCrossRef
138.
go back to reference Prokopchuk O, Liu Y, Henne-Bruns D, Kornmann M. Interleukin-4 enhances proliferation of human pancreatic cancer cells: evidence for autocrine and paracrine actions. Br J Cancer. 2005;92(5):921–8.PubMedPubMedCentralCrossRef Prokopchuk O, Liu Y, Henne-Bruns D, Kornmann M. Interleukin-4 enhances proliferation of human pancreatic cancer cells: evidence for autocrine and paracrine actions. Br J Cancer. 2005;92(5):921–8.PubMedPubMedCentralCrossRef
139.
go back to reference Barbier L, Ferhat M, Salamé E, Robin A, Herbelin A, Gombert J-M, Silvain C, Barbarin A. Interleukin-1 family cytokines: keystones in liver inflammatory diseases. Front Immunol. 2014;2019:10. Barbier L, Ferhat M, Salamé E, Robin A, Herbelin A, Gombert J-M, Silvain C, Barbarin A. Interleukin-1 family cytokines: keystones in liver inflammatory diseases. Front Immunol. 2014;2019:10.
140.
go back to reference McKay CJ, Glen P, McMillan DC. Chronic inflammation and pancreatic cancer. Best Pract Res Clin Gastroenterol. 2008;22(1):65–73.PubMedCrossRef McKay CJ, Glen P, McMillan DC. Chronic inflammation and pancreatic cancer. Best Pract Res Clin Gastroenterol. 2008;22(1):65–73.PubMedCrossRef
142.
go back to reference Nikolaou K, Sarris M, Talianidis I. Molecular pathways: the complex roles of inflammation pathways in the development and treatment of liver cancer. Clin Cancer Res. 2013;19(11):2810–6.PubMedCrossRef Nikolaou K, Sarris M, Talianidis I. Molecular pathways: the complex roles of inflammation pathways in the development and treatment of liver cancer. Clin Cancer Res. 2013;19(11):2810–6.PubMedCrossRef
145.
go back to reference Rial NS, Choi K, Nguyen T, Snyder B, Slepian MJ. Nuclear factor kappa B (NF-κB): a novel cause for diabetes, coronary artery disease and cancer initiation and promotion? Med Hypotheses. 2012;78(1):29–32.PubMedCrossRef Rial NS, Choi K, Nguyen T, Snyder B, Slepian MJ. Nuclear factor kappa B (NF-κB): a novel cause for diabetes, coronary artery disease and cancer initiation and promotion? Med Hypotheses. 2012;78(1):29–32.PubMedCrossRef
146.
go back to reference Seif F, Khoshmirsafa M, Aazami H, Mohsenzadegan M, Sedighi G, Bahar M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun Signal. 2017;15(1):1–13.CrossRef Seif F, Khoshmirsafa M, Aazami H, Mohsenzadegan M, Sedighi G, Bahar M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun Signal. 2017;15(1):1–13.CrossRef
147.
go back to reference Haricharan S, Li Y. STAT signaling in mammary gland differentiation, cell survival and tumorigenesis. Mol Cell Endocrinol. 2014;382(1):560–9.PubMedCrossRef Haricharan S, Li Y. STAT signaling in mammary gland differentiation, cell survival and tumorigenesis. Mol Cell Endocrinol. 2014;382(1):560–9.PubMedCrossRef
148.
go back to reference Miyatsuka T. Persistent expression of PDX-1 in the pancreas causes acinar-to-ductal metaplasia through Stat3 activation. Genes Dev. 2006;20(11):1435–40.PubMedPubMedCentralCrossRef Miyatsuka T. Persistent expression of PDX-1 in the pancreas causes acinar-to-ductal metaplasia through Stat3 activation. Genes Dev. 2006;20(11):1435–40.PubMedPubMedCentralCrossRef
149.
go back to reference Fukuda A, Wang Sam C, Morris John P, Folias Alexandra E, Liou A, Kim Grace E, Akira S, Boucher Kenneth M, Firpo Matthew A, Mulvihill Sean J, et al. Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell. 2011;19(4):441–55.PubMedPubMedCentralCrossRef Fukuda A, Wang Sam C, Morris John P, Folias Alexandra E, Liou A, Kim Grace E, Akira S, Boucher Kenneth M, Firpo Matthew A, Mulvihill Sean J, et al. Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell. 2011;19(4):441–55.PubMedPubMedCentralCrossRef
150.
go back to reference Mashili F, Chibalin AV, Krook A, Zierath JR. Constitutive STAT3 phosphorylation contributes to skeletal muscle insulin resistance in type 2 diabetes. Diabetes. 2012;62(2):457–65.PubMedCrossRef Mashili F, Chibalin AV, Krook A, Zierath JR. Constitutive STAT3 phosphorylation contributes to skeletal muscle insulin resistance in type 2 diabetes. Diabetes. 2012;62(2):457–65.PubMedCrossRef
151.
go back to reference Berasain C, Ujue Latasa M, Urtasun R, Goñi S, Elizalde M, Garcia-Irigoyen O, Azcona M, Prieto J, Ávila MA. Epidermal growth factor receptor (EGFR) crosstalks in liver cancer. Cancers. 2011;3(2):2444–61.PubMedPubMedCentralCrossRef Berasain C, Ujue Latasa M, Urtasun R, Goñi S, Elizalde M, Garcia-Irigoyen O, Azcona M, Prieto J, Ávila MA. Epidermal growth factor receptor (EGFR) crosstalks in liver cancer. Cancers. 2011;3(2):2444–61.PubMedPubMedCentralCrossRef
152.
go back to reference Berasain C, Castillo J, Prieto J, Avila MA. New molecular targets for hepatocellular carcinoma: the ErbB1 signaling system. Liver Int. 2007;27(2):174–85.PubMedCrossRef Berasain C, Castillo J, Prieto J, Avila MA. New molecular targets for hepatocellular carcinoma: the ErbB1 signaling system. Liver Int. 2007;27(2):174–85.PubMedCrossRef
154.
go back to reference Grapa CM, Mocan T, Gonciar D, Zdrehus C, Mosteanu O, Pop T, Mocan L. Epidermal growth factor receptor and its role in pancreatic cancer treatment mediated by nanoparticles. Int J Nanomed. 2019;14:9693–706.CrossRef Grapa CM, Mocan T, Gonciar D, Zdrehus C, Mosteanu O, Pop T, Mocan L. Epidermal growth factor receptor and its role in pancreatic cancer treatment mediated by nanoparticles. Int J Nanomed. 2019;14:9693–706.CrossRef
155.
go back to reference Tzeng C-WD, Frolov A, Frolova N, Jhala NC, Howard JH, Buchsbaum DJ, Vickers SM, Heslin MJ, Arnoletti JP. Epidermal growth factor receptor (EGFR) is highly conserved in pancreatic cancer. Surgery. 2007;141(4):464–9.PubMedCrossRef Tzeng C-WD, Frolov A, Frolova N, Jhala NC, Howard JH, Buchsbaum DJ, Vickers SM, Heslin MJ, Arnoletti JP. Epidermal growth factor receptor (EGFR) is highly conserved in pancreatic cancer. Surgery. 2007;141(4):464–9.PubMedCrossRef
157.
go back to reference Konishi A, Berk BC. Epidermal growth factor receptor transactivation is regulated by glucose in vascular smooth muscle cells. J Biol Chem. 2003;278(37):35049–56.PubMedCrossRef Konishi A, Berk BC. Epidermal growth factor receptor transactivation is regulated by glucose in vascular smooth muscle cells. J Biol Chem. 2003;278(37):35049–56.PubMedCrossRef
158.
go back to reference Li R, Uttarwar L, Gao B, Charbonneau M, Shi Y, Chan JSD, Dubois CM, Krepinsky JC. High glucose up-regulates ADAM17 through HIF-1α in mesangial cells. J Biol Chem. 2015;290(35):21603–14.PubMedPubMedCentralCrossRef Li R, Uttarwar L, Gao B, Charbonneau M, Shi Y, Chan JSD, Dubois CM, Krepinsky JC. High glucose up-regulates ADAM17 through HIF-1α in mesangial cells. J Biol Chem. 2015;290(35):21603–14.PubMedPubMedCentralCrossRef
159.
go back to reference Zhang M-Z, Wang Y, Paueksakon P, Harris RC. Epidermal growth factor receptor inhibition slows progression of diabetic nephropathy in association with a decrease in endoplasmic reticulum stress and an increase in autophagy. Diabetes. 2014;63(6):2063–72.PubMedPubMedCentralCrossRef Zhang M-Z, Wang Y, Paueksakon P, Harris RC. Epidermal growth factor receptor inhibition slows progression of diabetic nephropathy in association with a decrease in endoplasmic reticulum stress and an increase in autophagy. Diabetes. 2014;63(6):2063–72.PubMedPubMedCentralCrossRef
160.
go back to reference Li Z, Li Y, Overstreet JM, Chung S, Niu A, Fan X, Wang S, Wang Y, Zhang M-Z, Harris RC. Inhibition of epidermal growth factor receptor activation is associated with improved diabetic nephropathy and insulin resistance in type 2 diabetes. Diabetes. 2018;67(9):1847–57.PubMedPubMedCentralCrossRef Li Z, Li Y, Overstreet JM, Chung S, Niu A, Fan X, Wang S, Wang Y, Zhang M-Z, Harris RC. Inhibition of epidermal growth factor receptor activation is associated with improved diabetic nephropathy and insulin resistance in type 2 diabetes. Diabetes. 2018;67(9):1847–57.PubMedPubMedCentralCrossRef
161.
go back to reference Sheng L, Bayliss G, Zhuang S. Epidermal growth factor receptor: a potential therapeutic target for diabetic kidney disease. Front Pharmacol. 2021;11:2232.CrossRef Sheng L, Bayliss G, Zhuang S. Epidermal growth factor receptor: a potential therapeutic target for diabetic kidney disease. Front Pharmacol. 2021;11:2232.CrossRef
162.
go back to reference Khansari N, Shakiba Y, Mahmoudi M. Chronic inflammation and oxidative stress as a major cause of age- related diseases and cancer. Recent Pat Inflamm Allergy Drug Discov. 2009;3(1):73–80.PubMedCrossRef Khansari N, Shakiba Y, Mahmoudi M. Chronic inflammation and oxidative stress as a major cause of age- related diseases and cancer. Recent Pat Inflamm Allergy Drug Discov. 2009;3(1):73–80.PubMedCrossRef
164.
go back to reference Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Testa G, Cacciatore F, Bonaduce D, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757–72.PubMedPubMedCentralCrossRef Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Testa G, Cacciatore F, Bonaduce D, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757–72.PubMedPubMedCentralCrossRef
165.
go back to reference Pitocco D, Zaccardi F, Di Stasio E, Romitelli F, Santini SA, Zuppi C, Ghirlanda G. Oxidative stress, nitric oxide, and diabetes. Rev Diabet Stud. 2010;7(1):15–25.PubMedPubMedCentralCrossRef Pitocco D, Zaccardi F, Di Stasio E, Romitelli F, Santini SA, Zuppi C, Ghirlanda G. Oxidative stress, nitric oxide, and diabetes. Rev Diabet Stud. 2010;7(1):15–25.PubMedPubMedCentralCrossRef
166.
go back to reference Volpe CMO, Villar-Delfino PH, dos Anjos PMF, Nogueira-Machado JA. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018;9(2):1–9.CrossRef Volpe CMO, Villar-Delfino PH, dos Anjos PMF, Nogueira-Machado JA. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018;9(2):1–9.CrossRef
167.
go back to reference Zhang Y, Peng T, Zhu H, Zheng X, Zhang X, Jiang N, Cheng X, Lai X, Shunnar A, Singh M, et al. Prevention of hyperglycemia-induced myocardial apoptosis by gene silencing of Toll-like receptor-4. J Transl Med. 2010;8(1):1–8.CrossRef Zhang Y, Peng T, Zhu H, Zheng X, Zhang X, Jiang N, Cheng X, Lai X, Shunnar A, Singh M, et al. Prevention of hyperglycemia-induced myocardial apoptosis by gene silencing of Toll-like receptor-4. J Transl Med. 2010;8(1):1–8.CrossRef
168.
go back to reference Matough FA, Budin SB, Hamid ZA, Alwahaibi N, Mohamed J. The role of oxidative stress and antioxidants in diabetic complications = دور الإجهاد التأكسدي و المواد المضادة للأكسدة في مضاعفات مرض السكري. Sultan Qaboos Univ Med J. 2012;12(1):5–18.PubMedPubMedCentralCrossRef Matough FA, Budin SB, Hamid ZA, Alwahaibi N, Mohamed J. The role of oxidative stress and antioxidants in diabetic complications = دور الإجهاد التأكسدي و المواد المضادة للأكسدة في مضاعفات مرض السكري. Sultan Qaboos Univ Med J. 2012;12(1):5–18.PubMedPubMedCentralCrossRef
169.
go back to reference Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014;20(7):1126–67.PubMedPubMedCentralCrossRef Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014;20(7):1126–67.PubMedPubMedCentralCrossRef
170.
go back to reference Fiorentino TV, Prioletta A, Zuo P, Folli F. Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases. Curr Pharm Des. 2013;19(32):5695–703.PubMedCrossRef Fiorentino TV, Prioletta A, Zuo P, Folli F. Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases. Curr Pharm Des. 2013;19(32):5695–703.PubMedCrossRef
171.
go back to reference Abdel-Moneim A, El-Senousy Waled M, Abdel-Latif M, Khalil Rehab G. Association between antioxidant enzyme activities and enterovirus-infected type 1 diabetic children. Med Princ Pract. 2018;27(1):86–91.PubMedPubMedCentralCrossRef Abdel-Moneim A, El-Senousy Waled M, Abdel-Latif M, Khalil Rehab G. Association between antioxidant enzyme activities and enterovirus-infected type 1 diabetic children. Med Princ Pract. 2018;27(1):86–91.PubMedPubMedCentralCrossRef
173.
go back to reference Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radical Biol Med. 2010;49(11):1603–16.CrossRef Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radical Biol Med. 2010;49(11):1603–16.CrossRef
174.
go back to reference Federico A, Morgillo F, Tuccillo C, Ciardiello F, Loguercio C. Chronic inflammation and oxidative stress in human carcinogenesis. Int J Cancer. 2007;121(11):2381–6.PubMedCrossRef Federico A, Morgillo F, Tuccillo C, Ciardiello F, Loguercio C. Chronic inflammation and oxidative stress in human carcinogenesis. Int J Cancer. 2007;121(11):2381–6.PubMedCrossRef
175.
go back to reference Kim Y-W, West XZ, Byzova TV. Inflammation and oxidative stress in angiogenesis and vascular disease. J Mol Med. 2013;91(3):323–8.PubMedCrossRef Kim Y-W, West XZ, Byzova TV. Inflammation and oxidative stress in angiogenesis and vascular disease. J Mol Med. 2013;91(3):323–8.PubMedCrossRef
176.
go back to reference Stingl J. Estrogen and progesterone in normal mammary gland development and in cancer. Horm Cancer. 2010;2(2):85–90.PubMedCrossRef Stingl J. Estrogen and progesterone in normal mammary gland development and in cancer. Horm Cancer. 2010;2(2):85–90.PubMedCrossRef
177.
go back to reference Armaiz-Pena GN, Mangala LS, Spannuth WA, Lin YG, Jennings NB, Nick AM, Langley RR, Schmandt R, Lutgendorf SK, Cole SW, et al. Estrous cycle modulates ovarian carcinoma growth. Clin Cancer Res. 2009;15(9):2971–8.PubMedPubMedCentralCrossRef Armaiz-Pena GN, Mangala LS, Spannuth WA, Lin YG, Jennings NB, Nick AM, Langley RR, Schmandt R, Lutgendorf SK, Cole SW, et al. Estrous cycle modulates ovarian carcinoma growth. Clin Cancer Res. 2009;15(9):2971–8.PubMedPubMedCentralCrossRef
178.
go back to reference Chuffa LGDA, Lupi-Júnior LA, Costa AB, Amorim JPDA, Seiva FRF. The role of sex hormones and steroid receptors on female reproductive cancers. Steroids. 2017;118:93–108.PubMedCrossRef Chuffa LGDA, Lupi-Júnior LA, Costa AB, Amorim JPDA, Seiva FRF. The role of sex hormones and steroid receptors on female reproductive cancers. Steroids. 2017;118:93–108.PubMedCrossRef
179.
go back to reference Key TJ, Appleby Pn Fau - Reeves, G. K., Reeves Gk Fau - Roddam A, Roddam A Fau - Dorgan JF, Dorgan Jf Fau - Longcope C, Longcope C Fau - Stanczyk FZ, Stanczyk Fz Fau - Stephenson HE, Jr., Stephenson He Jr Fau - Falk RT, Falk Rt Fau - Miller R, Miller R Fau - Schatzkin A, Schatzkin A Fau - Allen DS et al: Body mass index, serum sex hormones, and breast cancer risk in postmenopausal women. JNCI J Natl Cancer Inst 2003; 95(16):1218–1226. Key TJ, Appleby Pn Fau - Reeves, G. K., Reeves Gk Fau - Roddam A, Roddam A Fau - Dorgan JF, Dorgan Jf Fau - Longcope C, Longcope C Fau - Stanczyk FZ, Stanczyk Fz Fau - Stephenson HE, Jr., Stephenson He Jr Fau - Falk RT, Falk Rt Fau - Miller R, Miller R Fau - Schatzkin A, Schatzkin A Fau - Allen DS et al: Body mass index, serum sex hormones, and breast cancer risk in postmenopausal women. JNCI J Natl Cancer Inst 2003; 95(16):1218–1226.
180.
go back to reference Ferguson RD, Gallagher EJ, Scheinman EJ, Damouni R, LeRoith D. Chapter two—the epidemiology and molecular mechanisms linking obesity, diabetes, and cancer. In: Litwack G, editor. Vitamins & hormones, vol. 93. Academic Press; 2013. p. 51–98. Ferguson RD, Gallagher EJ, Scheinman EJ, Damouni R, LeRoith D. Chapter two—the epidemiology and molecular mechanisms linking obesity, diabetes, and cancer. In: Litwack G, editor. Vitamins & hormones, vol. 93. Academic Press; 2013. p. 51–98.
181.
go back to reference Garg SK, Maurer H, Reed K, Selagamsetty R. Diabetes and cancer: two diseases with obesity as a common risk factor. Diabetes Obes Metab. 2013;16(2):97–110.PubMedPubMedCentralCrossRef Garg SK, Maurer H, Reed K, Selagamsetty R. Diabetes and cancer: two diseases with obesity as a common risk factor. Diabetes Obes Metab. 2013;16(2):97–110.PubMedPubMedCentralCrossRef
182.
go back to reference Bonovas S, Filioussi K, Tsantes A. Diabetes mellitus and risk of prostate cancer: a meta-analysis. Diabetologia. 2004;47(6):1071–8.PubMedCrossRef Bonovas S, Filioussi K, Tsantes A. Diabetes mellitus and risk of prostate cancer: a meta-analysis. Diabetologia. 2004;47(6):1071–8.PubMedCrossRef
183.
go back to reference Hara N. Prostate carcinogenesis with diabetes and androgen-deprivation-therapy-related diabetes: an update. Exp Diabetes Res. 2012;2012:1–8.CrossRef Hara N. Prostate carcinogenesis with diabetes and androgen-deprivation-therapy-related diabetes: an update. Exp Diabetes Res. 2012;2012:1–8.CrossRef
184.
go back to reference Tseng C-H, Chen C-J, Landolph JR. Diabetes and cancer: Epidemiological, clinical, and experimental perspectives. Exp Diabetes Res. 2012;2012:1–2. Tseng C-H, Chen C-J, Landolph JR. Diabetes and cancer: Epidemiological, clinical, and experimental perspectives. Exp Diabetes Res. 2012;2012:1–2.
186.
go back to reference Arcidiacono B, Iiritano S, Nocera A, Possidente K, Nevolo MT, Ventura V, Foti D, Chiefari E, Brunetti A. Insulin resistance and cancer risk: an overview of the pathogenetic mechanisms. Exp Diabetes Res. 2012;2012:1–12.CrossRef Arcidiacono B, Iiritano S, Nocera A, Possidente K, Nevolo MT, Ventura V, Foti D, Chiefari E, Brunetti A. Insulin resistance and cancer risk: an overview of the pathogenetic mechanisms. Exp Diabetes Res. 2012;2012:1–12.CrossRef
187.
go back to reference Hernandez AV, Pasupuleti V, Benites-Zapata VA, Thota P, Deshpande A, Perez-Lopez FR. Insulin resistance and endometrial cancer risk: a systematic review and meta-analysis. Eur J Cancer. 2015;51(18):2747–58.PubMedCrossRef Hernandez AV, Pasupuleti V, Benites-Zapata VA, Thota P, Deshpande A, Perez-Lopez FR. Insulin resistance and endometrial cancer risk: a systematic review and meta-analysis. Eur J Cancer. 2015;51(18):2747–58.PubMedCrossRef
188.
go back to reference Farahani H, Mahmoudi T, Asadi A, Nobakht H, Dabiri R, Hamta A. Insulin resistance and colorectal cancer risk: The role of elevated plasma resistin levels. J Gastrointest Cancer. 2019;51(2):478–83.CrossRef Farahani H, Mahmoudi T, Asadi A, Nobakht H, Dabiri R, Hamta A. Insulin resistance and colorectal cancer risk: The role of elevated plasma resistin levels. J Gastrointest Cancer. 2019;51(2):478–83.CrossRef
189.
go back to reference Saraei P, Asadi I, Kakar MA, Moradi-Kor N. The beneficial effects of metformin on cancer prevention and therapy: a comprehensive review of recent advances. Cancer Manage Res. 2019;11:3295–313.CrossRef Saraei P, Asadi I, Kakar MA, Moradi-Kor N. The beneficial effects of metformin on cancer prevention and therapy: a comprehensive review of recent advances. Cancer Manage Res. 2019;11:3295–313.CrossRef
191.
go back to reference Xu K, Sun G, Li M, Chen H, Zhang Z, Qian X, Li P, Xu L, Huang W, Wang X. Glibenclamide targets sulfonylurea receptor 1 to inhibit p70S6K activity and upregulate KLF4 expression to suppress non-small cell lung carcinoma. Mol Cancer Ther. 2019;18(11):2085–96.PubMedCrossRef Xu K, Sun G, Li M, Chen H, Zhang Z, Qian X, Li P, Xu L, Huang W, Wang X. Glibenclamide targets sulfonylurea receptor 1 to inhibit p70S6K activity and upregulate KLF4 expression to suppress non-small cell lung carcinoma. Mol Cancer Ther. 2019;18(11):2085–96.PubMedCrossRef
192.
go back to reference Corigliano DM, Syed R, Messineo S, Lupia A, Patel R, Reddy CVR, Dubey PK, Colica C, Amato R, De Sarro G, et al. Indole and 2,4-Thiazolidinedione conjugates as potential anticancer modulators. PeerJ. 2018;6:e5386.PubMedPubMedCentralCrossRef Corigliano DM, Syed R, Messineo S, Lupia A, Patel R, Reddy CVR, Dubey PK, Colica C, Amato R, De Sarro G, et al. Indole and 2,4-Thiazolidinedione conjugates as potential anticancer modulators. PeerJ. 2018;6:e5386.PubMedPubMedCentralCrossRef
193.
go back to reference Sola D, Rossi L, Schianca GPC, Maffioli P, Bigliocca M, Mella R, Corlianò F, Fra GP, Bartoli E, Derosa G. State of the art paper Sulfonylureas and their use in clinical practice. Arch Med Sci. 2015;4:840–8.CrossRef Sola D, Rossi L, Schianca GPC, Maffioli P, Bigliocca M, Mella R, Corlianò F, Fra GP, Bartoli E, Derosa G. State of the art paper Sulfonylureas and their use in clinical practice. Arch Med Sci. 2015;4:840–8.CrossRef
194.
go back to reference Avery M, Mizuno C, Chittiboyina A, Kurtz T, Pershadsingh H. Type 2 diabetes and oral antihyperglycemic drugs. Curr Med Chem. 2008;15(1):61–74.PubMedCrossRef Avery M, Mizuno C, Chittiboyina A, Kurtz T, Pershadsingh H. Type 2 diabetes and oral antihyperglycemic drugs. Curr Med Chem. 2008;15(1):61–74.PubMedCrossRef
195.
196.
go back to reference Núñez M, Medina V, Cricco G, Croci M, Cocca C, Rivera E, Bergoc R, Martín G. Glibenclamide inhibits cell growth by inducing G0/G1 arrest in the human breast cancer cell line MDA-MB-231. BMC Pharmacol Toxicol. 2013;14(1):1–13.CrossRef Núñez M, Medina V, Cricco G, Croci M, Cocca C, Rivera E, Bergoc R, Martín G. Glibenclamide inhibits cell growth by inducing G0/G1 arrest in the human breast cancer cell line MDA-MB-231. BMC Pharmacol Toxicol. 2013;14(1):1–13.CrossRef
197.
go back to reference Tuccori M, Filion KB, Yin H, Yu OH, Platt RW, Azoulay L. Pioglitazone use and risk of bladder cancer: population based cohort study. BMJ. 2016;352:i1541.PubMedPubMedCentralCrossRef Tuccori M, Filion KB, Yin H, Yu OH, Platt RW, Azoulay L. Pioglitazone use and risk of bladder cancer: population based cohort study. BMJ. 2016;352:i1541.PubMedPubMedCentralCrossRef
198.
go back to reference Roy S, Mahapatra AD, Mohammad T, Gupta P, Alajmi MF, Hussain A, Rehman MT, Datta B, Hassan MI. Design and development of novel urea, sulfonyltriurea, and sulfonamide derivatives as potential inhibitors of sphingosine kinase 1. Pharmaceuticals. 2020;13(6):118.PubMedCentralCrossRef Roy S, Mahapatra AD, Mohammad T, Gupta P, Alajmi MF, Hussain A, Rehman MT, Datta B, Hassan MI. Design and development of novel urea, sulfonyltriurea, and sulfonamide derivatives as potential inhibitors of sphingosine kinase 1. Pharmaceuticals. 2020;13(6):118.PubMedCentralCrossRef
199.
go back to reference Hendriks AM, Schrijnders D, Kleefstra N, de Vries EGE, Bilo HJG, Jalving M, Landman GWD: Sulfonylurea derivatives and cancer, friend or foe? European Journal of Pharmacology 2019, 861:172598. Hendriks AM, Schrijnders D, Kleefstra N, de Vries EGE, Bilo HJG, Jalving M, Landman GWD: Sulfonylurea derivatives and cancer, friend or foe? European Journal of Pharmacology 2019, 861:172598.
200.
go back to reference Payen L, Delugin L, Courtois A, Trinquart Y, Guillouzo A, Fardel O. The sulphonylurea glibenclamide inhibits multidrug resistance protein (MRP1) activity in human lung cancer cells. Br J Pharmacol. 2001;132(3):778–84.PubMedPubMedCentralCrossRef Payen L, Delugin L, Courtois A, Trinquart Y, Guillouzo A, Fardel O. The sulphonylurea glibenclamide inhibits multidrug resistance protein (MRP1) activity in human lung cancer cells. Br J Pharmacol. 2001;132(3):778–84.PubMedPubMedCentralCrossRef
201.
go back to reference Qian X, Li J, Ding J, Wang Z, Duan L, Hu G. Glibenclamide exerts an antitumor activity through reactive oxygen species–c-jun NH(2)-terminal kinase pathway in human gastric cancer cell line MGC-803. Biochem Pharmacol. 2008;76(12):1705–15.PubMedCrossRef Qian X, Li J, Ding J, Wang Z, Duan L, Hu G. Glibenclamide exerts an antitumor activity through reactive oxygen species–c-jun NH(2)-terminal kinase pathway in human gastric cancer cell line MGC-803. Biochem Pharmacol. 2008;76(12):1705–15.PubMedCrossRef
202.
go back to reference Suzuki Y, Inoue T, Murai M, Suzuki-Karasaki M, Ochiai T, Ra C. Depolarization potentiates TRAIL-induced apoptosis in human melanoma cells: role for ATP-sensitive K+ channels and endoplasmic reticulum stress. Int J Oncol. 2012;41(2):465–75.PubMedPubMedCentralCrossRef Suzuki Y, Inoue T, Murai M, Suzuki-Karasaki M, Ochiai T, Ra C. Depolarization potentiates TRAIL-induced apoptosis in human melanoma cells: role for ATP-sensitive K+ channels and endoplasmic reticulum stress. Int J Oncol. 2012;41(2):465–75.PubMedPubMedCentralCrossRef
203.
go back to reference Yasukagawa T, Niwa Y, Simizu S, Umezawa K. Suppression of cellular invasion by glybenclamide through inhibited secretion of platelet-derived growth factor in ovarian clear cell carcinoma ES-2 cells. FEBS Lett. 2012;586(10):1504–9.PubMedCrossRef Yasukagawa T, Niwa Y, Simizu S, Umezawa K. Suppression of cellular invasion by glybenclamide through inhibited secretion of platelet-derived growth factor in ovarian clear cell carcinoma ES-2 cells. FEBS Lett. 2012;586(10):1504–9.PubMedCrossRef
205.
206.
go back to reference Draznin B. Mechanism of the mitogenic influence of hyperinsulinemia. Diabetol Metab Syndr. 2011;3(1):1–3.CrossRef Draznin B. Mechanism of the mitogenic influence of hyperinsulinemia. Diabetol Metab Syndr. 2011;3(1):1–3.CrossRef
207.
go back to reference Hjartaker A, Langseth H, Weiderpass E. Obesity and diabetes epidemics: cancer repercussions. In: Berstein LM, Santen RJ, editors. Innovative endocrinology of cancer. New York, NY: Springer; 2008. p. 72–93.CrossRef Hjartaker A, Langseth H, Weiderpass E. Obesity and diabetes epidemics: cancer repercussions. In: Berstein LM, Santen RJ, editors. Innovative endocrinology of cancer. New York, NY: Springer; 2008. p. 72–93.CrossRef
208.
go back to reference Onitilo AA, Stankowski RV, Berg RL, Engel JM, Glurich I, Williams GM, Doi SA. Type 2 diabetes mellitus, glycemic control, and cancer risk. Eur J Cancer Prev. 2014;23(2):134–40.PubMedCrossRef Onitilo AA, Stankowski RV, Berg RL, Engel JM, Glurich I, Williams GM, Doi SA. Type 2 diabetes mellitus, glycemic control, and cancer risk. Eur J Cancer Prev. 2014;23(2):134–40.PubMedCrossRef
209.
go back to reference Boyle P, Boniol M, Koechlin A, Robertson C, Valentini F, Coppens K, Fairley LL, Boniol M, Zheng T, Zhang Y, et al. Diabetes and breast cancer risk: a meta-analysis. Br J Cancer. 2012;107(9):1608–17.PubMedPubMedCentralCrossRef Boyle P, Boniol M, Koechlin A, Robertson C, Valentini F, Coppens K, Fairley LL, Boniol M, Zheng T, Zhang Y, et al. Diabetes and breast cancer risk: a meta-analysis. Br J Cancer. 2012;107(9):1608–17.PubMedPubMedCentralCrossRef
210.
go back to reference Chae YK, Arya A, Malecek M-K, Shin DS, Carneiro B, Chandra S, Kaplan J, Kalyan A, Altman JK, Platanias L, et al. Repurposing metformin for cancer treatment: current clinical studies. Oncotarget. 2016;7(26):40767–80.PubMedPubMedCentralCrossRef Chae YK, Arya A, Malecek M-K, Shin DS, Carneiro B, Chandra S, Kaplan J, Kalyan A, Altman JK, Platanias L, et al. Repurposing metformin for cancer treatment: current clinical studies. Oncotarget. 2016;7(26):40767–80.PubMedPubMedCentralCrossRef
211.
go back to reference Lai S-W, Liao K-F, Chen P-C, Tsai P-Y, Hsieh DPH, Chen C-C. Antidiabetes drugs correlate with decreased risk of lung cancer: a population-based observation in Taiwan. Clin Lung Cancer. 2012;13(2):143–8.PubMedCrossRef Lai S-W, Liao K-F, Chen P-C, Tsai P-Y, Hsieh DPH, Chen C-C. Antidiabetes drugs correlate with decreased risk of lung cancer: a population-based observation in Taiwan. Clin Lung Cancer. 2012;13(2):143–8.PubMedCrossRef
212.
go back to reference Geraldine N, Marc A, Carla T, Chantal M, Stefaan B, Welcome W, Frank B. Relation between diabetes, metformin treatment and the occurrence of malignancies in a Belgian primary care setting. Diabetes Res Clin Pract. 2012;97(2):331–6.PubMedCrossRef Geraldine N, Marc A, Carla T, Chantal M, Stefaan B, Welcome W, Frank B. Relation between diabetes, metformin treatment and the occurrence of malignancies in a Belgian primary care setting. Diabetes Res Clin Pract. 2012;97(2):331–6.PubMedCrossRef
213.
go back to reference Vissers PAJ, Cardwell CR, van de Poll-Franse LV, Young IS, Pouwer F, Murray LJ. The association between glucose-lowering drug use and mortality among breast cancer patients with type 2 diabetes. Breast Cancer Res Treat. 2015;150(2):427–37.PubMedCrossRef Vissers PAJ, Cardwell CR, van de Poll-Franse LV, Young IS, Pouwer F, Murray LJ. The association between glucose-lowering drug use and mortality among breast cancer patients with type 2 diabetes. Breast Cancer Res Treat. 2015;150(2):427–37.PubMedCrossRef
214.
go back to reference Bosco JLF, Antonsen S, Sørensen HT, Pedersen L, Lash TL. Metformin and incident breast cancer among diabetic women: a population-based case–control study in Denmark. Cancer Epidemiol Biomark Prev. 2011;20(1):101–11.CrossRef Bosco JLF, Antonsen S, Sørensen HT, Pedersen L, Lash TL. Metformin and incident breast cancer among diabetic women: a population-based case–control study in Denmark. Cancer Epidemiol Biomark Prev. 2011;20(1):101–11.CrossRef
215.
go back to reference Rothermundt C, Hayoz S, Templeton AJ, Winterhalder R, Strebel RT, Bärtschi D, Pollak M, Lui L, Endt K, Schiess R, et al. Metformin in chemotherapy-naive castration-resistant prostate cancer: a multicenter phase 2 trial (SAKK 08/09). Eur Urol. 2014;66(3):468–74.PubMedCrossRef Rothermundt C, Hayoz S, Templeton AJ, Winterhalder R, Strebel RT, Bärtschi D, Pollak M, Lui L, Endt K, Schiess R, et al. Metformin in chemotherapy-naive castration-resistant prostate cancer: a multicenter phase 2 trial (SAKK 08/09). Eur Urol. 2014;66(3):468–74.PubMedCrossRef
216.
go back to reference Zakikhani M, Dowling R, Fantus IG, Sonenberg N, Pollak M. Metformin Is an AMP kinase–dependent growth inhibitor for breast cancer cells. Can Res. 2006;66(21):10269–73.CrossRef Zakikhani M, Dowling R, Fantus IG, Sonenberg N, Pollak M. Metformin Is an AMP kinase–dependent growth inhibitor for breast cancer cells. Can Res. 2006;66(21):10269–73.CrossRef
217.
go back to reference Pollak MN. Investigating metformin for cancer prevention and treatment: the end of the beginning. Cancer Discov. 2012;2(9):778–90.PubMedCrossRef Pollak MN. Investigating metformin for cancer prevention and treatment: the end of the beginning. Cancer Discov. 2012;2(9):778–90.PubMedCrossRef
218.
go back to reference Quinn BJ, Kitagawa H, Memmott RM, Gills JJ, Dennis PA. Repositioning metformin for cancer prevention and treatment. Trends Endocrinol Metab. 2013;24(9):469–80.PubMedCrossRef Quinn BJ, Kitagawa H, Memmott RM, Gills JJ, Dennis PA. Repositioning metformin for cancer prevention and treatment. Trends Endocrinol Metab. 2013;24(9):469–80.PubMedCrossRef
219.
go back to reference Deng X-S, Wang S, Deng A, Liu B, Edgerton SM, Lind SE, Wahdan-Alaswad R, Thor AD. Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle. 2012;11(2):367–76.PubMedCrossRef Deng X-S, Wang S, Deng A, Liu B, Edgerton SM, Lind SE, Wahdan-Alaswad R, Thor AD. Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle. 2012;11(2):367–76.PubMedCrossRef
220.
go back to reference Buzzai M, Jones RG, Amaravadi RK, Lum JJ, DeBerardinis RJ, Zhao F, Viollet B, Thompson CB. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Can Res. 2007;67(14):6745–52.CrossRef Buzzai M, Jones RG, Amaravadi RK, Lum JJ, DeBerardinis RJ, Zhao F, Viollet B, Thompson CB. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Can Res. 2007;67(14):6745–52.CrossRef
221.
go back to reference Huang X, Wullschleger S, Shpiro N, McGuire Victoria A, Sakamoto K, Woods Yvonne L, McBurnie W, Fleming S, Alessi Dario R. Important role of the LKB1–AMPK pathway in suppressing tumorigenesis in PTEN-deficient mice. Biochem J. 2008;412(2):211–21.PubMedCrossRef Huang X, Wullschleger S, Shpiro N, McGuire Victoria A, Sakamoto K, Woods Yvonne L, McBurnie W, Fleming S, Alessi Dario R. Important role of the LKB1–AMPK pathway in suppressing tumorigenesis in PTEN-deficient mice. Biochem J. 2008;412(2):211–21.PubMedCrossRef
222.
go back to reference Shaw RJ. LKB1 and AMP-activated protein kinase control of mTOR signaling and growth. Acta Physiol. 2009;196(1):65–80.CrossRef Shaw RJ. LKB1 and AMP-activated protein kinase control of mTOR signaling and growth. Acta Physiol. 2009;196(1):65–80.CrossRef
223.
go back to reference Hart PC, Chiyoda T, Liu X, Weigert M, Curtis M, Chiang C-Y, Loth R, Lastra R, McGregor SM, Locasale JW, et al. SPHK1 Is a novel target of metformin in ovarian cancer. Mol Cancer Res. 2019;17(4):870–81.PubMedPubMedCentralCrossRef Hart PC, Chiyoda T, Liu X, Weigert M, Curtis M, Chiang C-Y, Loth R, Lastra R, McGregor SM, Locasale JW, et al. SPHK1 Is a novel target of metformin in ovarian cancer. Mol Cancer Res. 2019;17(4):870–81.PubMedPubMedCentralCrossRef
224.
225.
go back to reference Wang L, Li K, Lin X, Yao Z, Wang S, Xiong X, Ning Z, Wang J, Xu X, Jiang Y, et al. Metformin induces human esophageal carcinoma cell pyroptosis by targeting the miR-497/PELP1 axis. Cancer Lett. 2019;450:22–31.PubMedCrossRef Wang L, Li K, Lin X, Yao Z, Wang S, Xiong X, Ning Z, Wang J, Xu X, Jiang Y, et al. Metformin induces human esophageal carcinoma cell pyroptosis by targeting the miR-497/PELP1 axis. Cancer Lett. 2019;450:22–31.PubMedCrossRef
226.
go back to reference Wang T, McNeill AM, Chen Y, Senderak M, Shankar RR. Metformin prescription patterns among US adolescents aged 10–19 years: 2009–2013. J Clin Pharm Ther. 2016;41(2):229–36.PubMedCrossRef Wang T, McNeill AM, Chen Y, Senderak M, Shankar RR. Metformin prescription patterns among US adolescents aged 10–19 years: 2009–2013. J Clin Pharm Ther. 2016;41(2):229–36.PubMedCrossRef
227.
go back to reference Hanawa S, Mitsuhashi A, Shozu M. Antitumor effects of metformin via indirect inhibition of protein phosphatase 2A in patients with endometrial cancer. PLoS ONE. 2018;13(2):e0192759.PubMedPubMedCentralCrossRef Hanawa S, Mitsuhashi A, Shozu M. Antitumor effects of metformin via indirect inhibition of protein phosphatase 2A in patients with endometrial cancer. PLoS ONE. 2018;13(2):e0192759.PubMedPubMedCentralCrossRef
228.
go back to reference Wu L, Zhou B, Oshiro-Rapley N, Li M, Paulo JA, Webster CM, Mou F, Kacergis MC, Talkowski ME, Carr CE, et al. An ancient, unified mechanism for metformin growth inhibition in C. elegans and cancer. Cell. 2016;167(7):1705-1718.e1713.PubMedPubMedCentralCrossRef Wu L, Zhou B, Oshiro-Rapley N, Li M, Paulo JA, Webster CM, Mou F, Kacergis MC, Talkowski ME, Carr CE, et al. An ancient, unified mechanism for metformin growth inhibition in C. elegans and cancer. Cell. 2016;167(7):1705-1718.e1713.PubMedPubMedCentralCrossRef
229.
go back to reference Lu C-C, Chiang J-H, Tsai F-J, Hsu Y-M, Juan Y-N, Yang J-S, Chiu H-Y. Metformin triggers the intrinsic apoptotic response in human AGS gastric adenocarcinoma cells by activating AMPK and suppressing mTOR/AKT signaling. Int J Oncol. 2019;54:1271–81.PubMedPubMedCentral Lu C-C, Chiang J-H, Tsai F-J, Hsu Y-M, Juan Y-N, Yang J-S, Chiu H-Y. Metformin triggers the intrinsic apoptotic response in human AGS gastric adenocarcinoma cells by activating AMPK and suppressing mTOR/AKT signaling. Int J Oncol. 2019;54:1271–81.PubMedPubMedCentral
230.
go back to reference Tseng H-W, Li S-C, Tsai K-W. Metformin treatment suppresses melanoma cell growth and motility through modulation of microRNA expression. Cancers. 2019;11(2):209.PubMedCentralCrossRef Tseng H-W, Li S-C, Tsai K-W. Metformin treatment suppresses melanoma cell growth and motility through modulation of microRNA expression. Cancers. 2019;11(2):209.PubMedCentralCrossRef
231.
go back to reference Xie J, Ye J, Cai Z, Luo Y, Zhu X, Deng Y, Feng Y, Liang Y, Liu R, Han Z, et al. GPD1 enhances the anticancer effects of metformin by synergistically increasing total cellular glycerol-3-phosphate. Can Res. 2020;80(11):2150–62.CrossRef Xie J, Ye J, Cai Z, Luo Y, Zhu X, Deng Y, Feng Y, Liang Y, Liu R, Han Z, et al. GPD1 enhances the anticancer effects of metformin by synergistically increasing total cellular glycerol-3-phosphate. Can Res. 2020;80(11):2150–62.CrossRef
232.
go back to reference Liu M, Zhang Z, Wang H, Chen X, Jin C. Activation of AMPK by metformin promotes renal cancer cell proliferation under glucose deprivation through its interaction with PKM2. Int J Biol Sci. 2019;15(3):617–27.PubMedPubMedCentralCrossRef Liu M, Zhang Z, Wang H, Chen X, Jin C. Activation of AMPK by metformin promotes renal cancer cell proliferation under glucose deprivation through its interaction with PKM2. Int J Biol Sci. 2019;15(3):617–27.PubMedPubMedCentralCrossRef
233.
go back to reference Ma L, Wei J, Wan J, Wang W, Wang L, Yuan Y, Yang Z, Liu X, Ming L. Low glucose and metformin-induced apoptosis of human ovarian cancer cells is connected to ASK1 via mitochondrial and endoplasmic reticulum stress-associated pathways. J Exp Clin Cancer Res. 2019;38(1):1–14.CrossRef Ma L, Wei J, Wan J, Wang W, Wang L, Yuan Y, Yang Z, Liu X, Ming L. Low glucose and metformin-induced apoptosis of human ovarian cancer cells is connected to ASK1 via mitochondrial and endoplasmic reticulum stress-associated pathways. J Exp Clin Cancer Res. 2019;38(1):1–14.CrossRef
234.
go back to reference Fröhlich E, Wahl R. Chemotherapy and chemoprevention by thiazolidinediones. Biomed Res Int. 2015;2015:1–14.CrossRef Fröhlich E, Wahl R. Chemotherapy and chemoprevention by thiazolidinediones. Biomed Res Int. 2015;2015:1–14.CrossRef
235.
go back to reference Koffarnus RL, Wargo KA, Phillippe HM. Rivoglitazone: a new thiazolidinedione for the treatment of type 2 diabetes mellitus. Ann Pharmacother. 2013;47(6):877–85.PubMedCrossRef Koffarnus RL, Wargo KA, Phillippe HM. Rivoglitazone: a new thiazolidinedione for the treatment of type 2 diabetes mellitus. Ann Pharmacother. 2013;47(6):877–85.PubMedCrossRef
236.
237.
go back to reference Nanjan MJ, Mohammed M, Prashantha Kumar BR, Chandrasekar MJN. Thiazolidinediones as antidiabetic agents: a critical review. Bioorg Chem. 2018;77:548–67.PubMedCrossRef Nanjan MJ, Mohammed M, Prashantha Kumar BR, Chandrasekar MJN. Thiazolidinediones as antidiabetic agents: a critical review. Bioorg Chem. 2018;77:548–67.PubMedCrossRef
238.
go back to reference Soccio Raymond E, Chen Eric R, Lazar Mitchell A. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab. 2014;20(4):573–91.PubMedPubMedCentralCrossRef Soccio Raymond E, Chen Eric R, Lazar Mitchell A. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab. 2014;20(4):573–91.PubMedPubMedCentralCrossRef
239.
go back to reference Colca JR. The TZD insulin sensitizer clue provides a new route into diabetes drug discovery. Expert Opin Drug Discov. 2015;10(12):1259–70.PubMedCrossRef Colca JR. The TZD insulin sensitizer clue provides a new route into diabetes drug discovery. Expert Opin Drug Discov. 2015;10(12):1259–70.PubMedCrossRef
240.
go back to reference Lebovitz HE. Thiazolidinediones: the forgotten diabetes medications. Curr Diabetes Rep. 2019;19(12):1–13.CrossRef Lebovitz HE. Thiazolidinediones: the forgotten diabetes medications. Curr Diabetes Rep. 2019;19(12):1–13.CrossRef
241.
go back to reference Imchen T, Manasse J, Min K-W, Baek SJ. Characterization of PPAR dual ligand MCC-555 in AOM-induced colorectal tumorigenesis. Exp Toxicol Pathol. 2013;65(6):919–24.PubMedPubMedCentralCrossRef Imchen T, Manasse J, Min K-W, Baek SJ. Characterization of PPAR dual ligand MCC-555 in AOM-induced colorectal tumorigenesis. Exp Toxicol Pathol. 2013;65(6):919–24.PubMedPubMedCentralCrossRef
242.
go back to reference Tseng C-H. Rosiglitazone reduces breast cancer risk in Taiwanese female patients with type 2 diabetes mellitus. Oncotarget. 2017;8(2):3042–8.PubMedCrossRef Tseng C-H. Rosiglitazone reduces breast cancer risk in Taiwanese female patients with type 2 diabetes mellitus. Oncotarget. 2017;8(2):3042–8.PubMedCrossRef
244.
go back to reference Du R, Lin L, Cheng D, Xu Y, Xu M, Chen Y, Wang W, Bi Y, Li D, Lu J. Thiazolidinedione therapy and breast cancer risk in diabetic women: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2018;34(2):e2961.CrossRef Du R, Lin L, Cheng D, Xu Y, Xu M, Chen Y, Wang W, Bi Y, Li D, Lu J. Thiazolidinedione therapy and breast cancer risk in diabetic women: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2018;34(2):e2961.CrossRef
245.
go back to reference Alzhrani ZMM, Alam MM, Neamatallah T, Nazreen S. Design, synthesis and in vitro antiproliferative activity of new thiazolidinedione-1,3,4-oxadiazole hybrids as thymidylate synthase inhibitors. J Enzyme Inhib Med Chem. 2020;35(1):1116–23.PubMedPubMedCentralCrossRef Alzhrani ZMM, Alam MM, Neamatallah T, Nazreen S. Design, synthesis and in vitro antiproliferative activity of new thiazolidinedione-1,3,4-oxadiazole hybrids as thymidylate synthase inhibitors. J Enzyme Inhib Med Chem. 2020;35(1):1116–23.PubMedPubMedCentralCrossRef
246.
go back to reference Lyles BE, Akinyeke TO, Moss PE, Stewart LV. Thiazolidinediones regulate expression of cell cycle proteins in human prostate cancer cells via PPARγ-dependent and PPARγ-independent pathways. Cell Cycle. 2009;8(2):268–77.PubMedCrossRef Lyles BE, Akinyeke TO, Moss PE, Stewart LV. Thiazolidinediones regulate expression of cell cycle proteins in human prostate cancer cells via PPARγ-dependent and PPARγ-independent pathways. Cell Cycle. 2009;8(2):268–77.PubMedCrossRef
247.
go back to reference Kazberuk A, Zareba I, Palka J, Surazynski A. A novel plausible mechanism of NSAIDs-induced apoptosis in cancer cells: the implication of proline oxidase and peroxisome proliferator-activated receptor. Pharmacol Rep. 2020;72(5):1152–60.PubMedPubMedCentralCrossRef Kazberuk A, Zareba I, Palka J, Surazynski A. A novel plausible mechanism of NSAIDs-induced apoptosis in cancer cells: the implication of proline oxidase and peroxisome proliferator-activated receptor. Pharmacol Rep. 2020;72(5):1152–60.PubMedPubMedCentralCrossRef
248.
249.
go back to reference Kotlinowski J, Jozkowicz A. PPAR gamma and angiogenesis: endothelial cells perspective. J Diabetes Res. 2016;2016:1–11.CrossRef Kotlinowski J, Jozkowicz A. PPAR gamma and angiogenesis: endothelial cells perspective. J Diabetes Res. 2016;2016:1–11.CrossRef
250.
go back to reference Kim SY, Kim MS, Lee MK, Kim JS, Yi HK, Nam SY, Lee DY, Hwang PH. PPARγ induces growth inhibition and apoptosis through upregulation of insulin-like growth factor-binding protein-3 in gastric cancer cells. Braz J Med Biol Res. 2015;48(3):226–33.PubMedPubMedCentralCrossRef Kim SY, Kim MS, Lee MK, Kim JS, Yi HK, Nam SY, Lee DY, Hwang PH. PPARγ induces growth inhibition and apoptosis through upregulation of insulin-like growth factor-binding protein-3 in gastric cancer cells. Braz J Med Biol Res. 2015;48(3):226–33.PubMedPubMedCentralCrossRef
251.
go back to reference Luo S, Wang J, Ma Y, Yao Z, Pan H. PPARγ inhibits ovarian cancer cells proliferation through upregulation of miR-125b. Biochem Biophys Res Commun. 2015;462(2):85–90.PubMedCrossRef Luo S, Wang J, Ma Y, Yao Z, Pan H. PPARγ inhibits ovarian cancer cells proliferation through upregulation of miR-125b. Biochem Biophys Res Commun. 2015;462(2):85–90.PubMedCrossRef
252.
go back to reference Nazim UMD, Moon J-H, Lee Y-J, Seol J-W, Park S-Y. PPARγ activation by troglitazone enhances human lung cancer cells to TRAIL-induced apoptosis via autophagy flux. Oncotarget. 2017;8(16):26819–31.PubMedPubMedCentralCrossRef Nazim UMD, Moon J-H, Lee Y-J, Seol J-W, Park S-Y. PPARγ activation by troglitazone enhances human lung cancer cells to TRAIL-induced apoptosis via autophagy flux. Oncotarget. 2017;8(16):26819–31.PubMedPubMedCentralCrossRef
253.
go back to reference Moss PE, Lyles BE, Stewart LV. The PPARγ ligand ciglitazone regulates androgen receptor activation differently in androgen-dependent versus androgen-independent human prostate cancer cells. Exp Cell Res. 2010;316(20):3478–88.PubMedPubMedCentralCrossRef Moss PE, Lyles BE, Stewart LV. The PPARγ ligand ciglitazone regulates androgen receptor activation differently in androgen-dependent versus androgen-independent human prostate cancer cells. Exp Cell Res. 2010;316(20):3478–88.PubMedPubMedCentralCrossRef
254.
go back to reference Dąbrowski M, Szymańska-Garbacz E, Miszczyszyn Z, Dereziński T, Czupryniak L. Antidiabetic medications use and cancer risk in type 2 diabetes. Clin Diabetol. 2017;6(1):17–25.CrossRef Dąbrowski M, Szymańska-Garbacz E, Miszczyszyn Z, Dereziński T, Czupryniak L. Antidiabetic medications use and cancer risk in type 2 diabetes. Clin Diabetol. 2017;6(1):17–25.CrossRef
255.
go back to reference Kowall B, Stang A, Rathmann W, Kostev K. No reduced risk of overall, colorectal, lung, breast, and prostate cancer with metformin therapy in diabetic patients: database analyses from Germany and the UK. Pharmacoepidemiol Drug Saf. 2015;24(8):865–74.PubMedCrossRef Kowall B, Stang A, Rathmann W, Kostev K. No reduced risk of overall, colorectal, lung, breast, and prostate cancer with metformin therapy in diabetic patients: database analyses from Germany and the UK. Pharmacoepidemiol Drug Saf. 2015;24(8):865–74.PubMedCrossRef
256.
go back to reference Gallagher EJ, LeRoith D. Diabetes, antihyperglycemic medications and cancer risk. Curr Opin Endocrinol Diabetes Obes. 2013;20(5):485–94.PubMedCrossRef Gallagher EJ, LeRoith D. Diabetes, antihyperglycemic medications and cancer risk. Curr Opin Endocrinol Diabetes Obes. 2013;20(5):485–94.PubMedCrossRef
257.
go back to reference Belfiore A, Malaguarnera R. Insulin receptor and cancer. Endocr Relat Cancer. 2011;18(4):R125–47.PubMedCrossRef Belfiore A, Malaguarnera R. Insulin receptor and cancer. Endocr Relat Cancer. 2011;18(4):R125–47.PubMedCrossRef
258.
go back to reference Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Investig. 2001;108(8):1167–74.PubMedPubMedCentralCrossRef Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Investig. 2001;108(8):1167–74.PubMedPubMedCentralCrossRef
259.
go back to reference Lee DY, Yu JH, Park S, Han K, Kim NH, Yoo HJ, Choi KM, Baik SH, Kim NH, Seo JA. The influence of diabetes and antidiabetic medications on the risk of pancreatic cancer: a nationwide population-based study in Korea. Sci Rep. 2018;8(1):1–8. Lee DY, Yu JH, Park S, Han K, Kim NH, Yoo HJ, Choi KM, Baik SH, Kim NH, Seo JA. The influence of diabetes and antidiabetic medications on the risk of pancreatic cancer: a nationwide population-based study in Korea. Sci Rep. 2018;8(1):1–8.
261.
go back to reference Sciacca L, Moli RL, Vigneri R. Insulin analogs and cancer. Front Endocrinol. 2012;3:21.CrossRef Sciacca L, Moli RL, Vigneri R. Insulin analogs and cancer. Front Endocrinol. 2012;3:21.CrossRef
262.
go back to reference Pocock SJ, Smeeth L. Insulin glargine and malignancy: an unwarranted alarm. The Lancet. 2009;374(9689):511–3.CrossRef Pocock SJ, Smeeth L. Insulin glargine and malignancy: an unwarranted alarm. The Lancet. 2009;374(9689):511–3.CrossRef
Metadata
Title
Cancer and diabetes: the interlinking metabolic pathways and repurposing actions of antidiabetic drugs
Authors
Ahmed Olatunde
Manisha Nigam
Rahul Kunwar Singh
Abhaya Shikhar Panwar
Abdulwahab Lasisi
Fahad A. Alhumaydhi
Vijay Jyoti kumar
Abhay Prakash Mishra
Javad Sharifi-Rad
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-02202-5

Other articles of this Issue 1/2021

Cancer Cell International 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine