Skip to main content
Top
Published in: Drug Safety 3/2017

01-03-2017 | Review Article

Hyperglycaemia Induced by Novel Anticancer Agents: An Undesirable Complication or a Potential Therapeutic Opportunity?

Author: Rashmi R. Shah

Published in: Drug Safety | Issue 3/2017

Login to get access

Abstract

Signalling pathways involving protein kinase, insulin-like growth factor 1, insulin receptors and the phosphoinositide 3 kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) system are critical in promoting oncogenesis. The use of anticancer agents that inhibit these pathways frequently results in hyperglycaemia, an on-target effect of these drugs. Hyperglycaemia induced by these agents denotes optimal inhibition of the desired pharmacological target. As hyperglycaemia can be treated successfully and effectively with metformin, managing this complication by reducing the dose of or discontinuing the anticancer drug may be counterproductive, especially if it is otherwise effective and clinically tolerated. The use of metformin to treat hyperglycaemia induced by anticancer drugs provides a valuable therapeutic opportunity of potentiating their clinical anticancer effects. Although evidence from randomised controlled trials is awaited, extensive preclinical evidence and clinical observational studies suggest that metformin has anticancer properties that improve overall survival in patients with diabetes and a variety of cancers. Metformin has also been reported to reverse resistance to epidermal growth factor receptor (EGFR)-inhibiting tyrosine kinase inhibitors. This review summarises briefly the role of the above signalling pathways in oncogenesis, the causal association between inhibition of these pathways and hyperglycaemia, and the effect of metformin on clinical outcomes resulting from its anticancer properties. The evidence reviewed herein, albeit almost exclusively from observational studies, provides support for a greater use of metformin not only in patients with cancer and diabetes or drug-induced hyperglycaemia but also potentially as an anticancer drug. However, prospective randomised controlled studies are needed in all these settings to better assess the effect on clinical outcomes of adding metformin to ongoing anticancer therapy.
Literature
1.
go back to reference Shah RR. Tyrosine kinase inhibitors-induced interstitial lung disease: clinical features, diagnostic challenges and therapeutic dilemmas. Drug Saf. 2016;39:1073–91.PubMedCrossRef Shah RR. Tyrosine kinase inhibitors-induced interstitial lung disease: clinical features, diagnostic challenges and therapeutic dilemmas. Drug Saf. 2016;39:1073–91.PubMedCrossRef
2.
go back to reference Wu P, Nielsen TE, Clausen MH. Small-molecule kinase inhibitors: an analysis of FDA-approved drugs. Drug Discov Today. 2016;21:5–10.PubMedCrossRef Wu P, Nielsen TE, Clausen MH. Small-molecule kinase inhibitors: an analysis of FDA-approved drugs. Drug Discov Today. 2016;21:5–10.PubMedCrossRef
3.
go back to reference Amir E, Seruga B, Martinez-Lopez J, Kwong R, Pandiella A, Tannock IF, et al. Oncogenic targets, magnitude of benefit, and market pricing of antineoplastic drugs. J Clin Oncol. 2011;29:2543–9.PubMedCrossRef Amir E, Seruga B, Martinez-Lopez J, Kwong R, Pandiella A, Tannock IF, et al. Oncogenic targets, magnitude of benefit, and market pricing of antineoplastic drugs. J Clin Oncol. 2011;29:2543–9.PubMedCrossRef
4.
go back to reference Xiao YY, Zhan P, Yuan DM, Liu HB, Lv TF, Song Y, et al. Chemotherapy plus multitargeted antiangiogenic tyrosine kinase inhibitors or chemotherapy alone in advanced NSCLC: a meta-analysis of randomized controlled trials. Eur J Clin Pharmacol. 2013;69:151–9.PubMedCrossRef Xiao YY, Zhan P, Yuan DM, Liu HB, Lv TF, Song Y, et al. Chemotherapy plus multitargeted antiangiogenic tyrosine kinase inhibitors or chemotherapy alone in advanced NSCLC: a meta-analysis of randomized controlled trials. Eur J Clin Pharmacol. 2013;69:151–9.PubMedCrossRef
5.
go back to reference Haspinger ER, Agustoni F, Torri V, Gelsomino F, Platania M, Zilembo N, et al. Is there evidence for different effects among EGFR-TKIs? Systematic review and meta-analysis of EGFR tyrosine kinase inhibitors (TKIs) versus chemotherapy as first-line treatment for patients harboring EGFR mutations. Crit Rev Oncol Hematol. 2015;94:213–27.PubMedCrossRef Haspinger ER, Agustoni F, Torri V, Gelsomino F, Platania M, Zilembo N, et al. Is there evidence for different effects among EGFR-TKIs? Systematic review and meta-analysis of EGFR tyrosine kinase inhibitors (TKIs) versus chemotherapy as first-line treatment for patients harboring EGFR mutations. Crit Rev Oncol Hematol. 2015;94:213–27.PubMedCrossRef
6.
go back to reference Liu J, Sheng Z, Zhang Y, Li G. The efficacy of epidermal growth factor receptor tyrosine kinase inhibitors for molecularly selected patients with non-small cell lung cancer: a meta-analysis of 30 randomized controlled trials. Target Oncol. 2016;11:49–58.PubMedCrossRef Liu J, Sheng Z, Zhang Y, Li G. The efficacy of epidermal growth factor receptor tyrosine kinase inhibitors for molecularly selected patients with non-small cell lung cancer: a meta-analysis of 30 randomized controlled trials. Target Oncol. 2016;11:49–58.PubMedCrossRef
7.
go back to reference Guetz GD, Landre T, Uzzan B, Chouahnia K, Nicolas P, Morere JF. Is there a survival benefit of first-line epidermal growth factor receptor tyrosine kinase inhibitor monotherapy versus chemotherapy in patients with advanced non-small-cell lung cancer? A meta-analysis. Target Oncol. 2016;11:41–7.PubMedCrossRef Guetz GD, Landre T, Uzzan B, Chouahnia K, Nicolas P, Morere JF. Is there a survival benefit of first-line epidermal growth factor receptor tyrosine kinase inhibitor monotherapy versus chemotherapy in patients with advanced non-small-cell lung cancer? A meta-analysis. Target Oncol. 2016;11:41–7.PubMedCrossRef
8.
go back to reference Shah DR, Shah RR, Morganroth J. Tyrosine kinase inhibitors: their on-target toxicities as potential indicators of efficacy. Drug Saf. 2013;36:413–26.PubMedCrossRef Shah DR, Shah RR, Morganroth J. Tyrosine kinase inhibitors: their on-target toxicities as potential indicators of efficacy. Drug Saf. 2013;36:413–26.PubMedCrossRef
19.
go back to reference Agostino NM, Chinchilli VM, Lynch CJ, Koszyk-Szewczyk A, Gingrich R, Sivik J, et al. Effect of the tyrosine kinase inhibitors (sunitinib, sorafenib, dasatinib, and imatinib) on blood glucose levels in diabetic and nondiabetic patients in general clinical practice. J Oncol Pharm Pract. 2011;17:197–202.PubMedCrossRef Agostino NM, Chinchilli VM, Lynch CJ, Koszyk-Szewczyk A, Gingrich R, Sivik J, et al. Effect of the tyrosine kinase inhibitors (sunitinib, sorafenib, dasatinib, and imatinib) on blood glucose levels in diabetic and nondiabetic patients in general clinical practice. J Oncol Pharm Pract. 2011;17:197–202.PubMedCrossRef
20.
go back to reference Fountas A, Diamantopoulos LN, Tsatsoulis A. Tyrosine kinase inhibitors and diabetes: a novel treatment paradigm? Trends Endocrinol Metab. 2015;26:643–56.PubMedCrossRef Fountas A, Diamantopoulos LN, Tsatsoulis A. Tyrosine kinase inhibitors and diabetes: a novel treatment paradigm? Trends Endocrinol Metab. 2015;26:643–56.PubMedCrossRef
21.
go back to reference Malek R, Davis SN. Tyrosine kinase inhibitors under investigation for the treatment of type II diabetes. Expert Opin Investig Drugs. 2016;25:287–96.PubMedCrossRef Malek R, Davis SN. Tyrosine kinase inhibitors under investigation for the treatment of type II diabetes. Expert Opin Investig Drugs. 2016;25:287–96.PubMedCrossRef
23.
go back to reference Villadolid J, Ersek JL, Fong MK, Sirianno L, Story ES. Management of hyperglycemia from epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) targeting T790M-mediated resistance. Transl Lung Cancer Res. 2015;4:576–83.PubMedPubMedCentral Villadolid J, Ersek JL, Fong MK, Sirianno L, Story ES. Management of hyperglycemia from epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) targeting T790M-mediated resistance. Transl Lung Cancer Res. 2015;4:576–83.PubMedPubMedCentral
24.
go back to reference Ma H, Zhang T, Shen H, Cao H, Du J. The adverse events profile of anti-IGF-1R monoclonal antibodies in cancer therapy. Br J Clin Pharmacol. 2014;77:917–28.PubMedCrossRef Ma H, Zhang T, Shen H, Cao H, Du J. The adverse events profile of anti-IGF-1R monoclonal antibodies in cancer therapy. Br J Clin Pharmacol. 2014;77:917–28.PubMedCrossRef
25.
go back to reference Hartog H, Wesseling J, Boezen HM, van der Graff WTA. The insulin-like growth factor 1 receptor in cancer: Old focus, new future. Eur J Cancer. 2007;43:1895–904.PubMedCrossRef Hartog H, Wesseling J, Boezen HM, van der Graff WTA. The insulin-like growth factor 1 receptor in cancer: Old focus, new future. Eur J Cancer. 2007;43:1895–904.PubMedCrossRef
26.
go back to reference Lundby A, Bolvig P, Hegelund AC, Hansen BF, Worm J, Lützen A, et al. Surface-expressed insulin receptors as well as IGF-I receptors both contribute to the mitogenic effects of human insulin and its analogues. J Appl Toxicol. 2015;35:842–50.PubMedCrossRef Lundby A, Bolvig P, Hegelund AC, Hansen BF, Worm J, Lützen A, et al. Surface-expressed insulin receptors as well as IGF-I receptors both contribute to the mitogenic effects of human insulin and its analogues. J Appl Toxicol. 2015;35:842–50.PubMedCrossRef
27.
go back to reference Vigneri R, Goldfine ID, Frittitta L. Insulin, insulin receptors, and cancer. J Endocrinol Invest. 2016;39:1365–76.PubMedCrossRef Vigneri R, Goldfine ID, Frittitta L. Insulin, insulin receptors, and cancer. J Endocrinol Invest. 2016;39:1365–76.PubMedCrossRef
28.
go back to reference Lu Y, Zi X, Zhao Y, Mascarenhas D, Pollak M. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). J Natl Cancer Inst. 2001;93:1852–7.PubMedCrossRef Lu Y, Zi X, Zhao Y, Mascarenhas D, Pollak M. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). J Natl Cancer Inst. 2001;93:1852–7.PubMedCrossRef
29.
go back to reference Browne BC, Crown J, Venkatesan N, Duffy MJ, Clynes M, Slamon D, et al. Inhibition of IGF1R activity enhances response to trastuzumab in HER-2-positive breast cancer cells. Ann Oncol. 2011;22:68–73.PubMedCrossRef Browne BC, Crown J, Venkatesan N, Duffy MJ, Clynes M, Slamon D, et al. Inhibition of IGF1R activity enhances response to trastuzumab in HER-2-positive breast cancer cells. Ann Oncol. 2011;22:68–73.PubMedCrossRef
31.
go back to reference Anastassiadis T, Duong-Ly KC, Deacon SW, Lafontant A, Ma H, Devarajan K, et al. A highly selective dual insulin receptor (IR)/insulin-like growth factor 1 receptor (IGF-1R) inhibitor derived from an extracellular signal-regulated kinase (ERK) inhibitor. J Biol Chem. 2013;288:28068–77.PubMedPubMedCentralCrossRef Anastassiadis T, Duong-Ly KC, Deacon SW, Lafontant A, Ma H, Devarajan K, et al. A highly selective dual insulin receptor (IR)/insulin-like growth factor 1 receptor (IGF-1R) inhibitor derived from an extracellular signal-regulated kinase (ERK) inhibitor. J Biol Chem. 2013;288:28068–77.PubMedPubMedCentralCrossRef
32.
go back to reference Jin M, Wang J, Buck E, Mulvihill MJ. Small-molecule ATP-competitive dual IGF-1R and insulin receptor inhibitors: structural insights, chemical diversity and molecular evolution. Future Med Chem. 2012;4:315–28.PubMedCrossRef Jin M, Wang J, Buck E, Mulvihill MJ. Small-molecule ATP-competitive dual IGF-1R and insulin receptor inhibitors: structural insights, chemical diversity and molecular evolution. Future Med Chem. 2012;4:315–28.PubMedCrossRef
33.
34.
go back to reference Allison M. Clinical setbacks reduce IGF-1 inhibitors to cocktail mixers. Nat Biotechnol. 2012;30:906–7.PubMedCrossRef Allison M. Clinical setbacks reduce IGF-1 inhibitors to cocktail mixers. Nat Biotechnol. 2012;30:906–7.PubMedCrossRef
35.
36.
37.
go back to reference Baselga J. Targeting the phosphoinositide-3 (PI3) kinase pathway in breast cancer. Oncologist. 2011;16(Suppl 1):12–9.PubMedCrossRef Baselga J. Targeting the phosphoinositide-3 (PI3) kinase pathway in breast cancer. Oncologist. 2011;16(Suppl 1):12–9.PubMedCrossRef
38.
go back to reference Subramani R, Lopez-Valdez R, Arumugam A, Nandy S, Boopalan T, Lakshmanaswamy R. Targeting insulin-like growth factor 1 receptor inhibits pancreatic cancer growth and metastasis. PLoS One. 2014;9(5):e97016.PubMedPubMedCentralCrossRef Subramani R, Lopez-Valdez R, Arumugam A, Nandy S, Boopalan T, Lakshmanaswamy R. Targeting insulin-like growth factor 1 receptor inhibits pancreatic cancer growth and metastasis. PLoS One. 2014;9(5):e97016.PubMedPubMedCentralCrossRef
40.
go back to reference Chia S, Gandhi S, Joy AA, Edwards S, Gorr M, Hopkins S, et al. Novel agents and associated toxicities of inhibitors of the pi3k/Akt/mtor pathway for the treatment of breast cancer. Curr Oncol. 2015;22:33–48.PubMedPubMedCentralCrossRef Chia S, Gandhi S, Joy AA, Edwards S, Gorr M, Hopkins S, et al. Novel agents and associated toxicities of inhibitors of the pi3k/Akt/mtor pathway for the treatment of breast cancer. Curr Oncol. 2015;22:33–48.PubMedPubMedCentralCrossRef
42.
go back to reference Mayer IA, Arteaga CL. The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med. 2016;67:11–28.PubMedCrossRef Mayer IA, Arteaga CL. The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med. 2016;67:11–28.PubMedCrossRef
43.
go back to reference Zhuang H, Bai J, Chang JY, Yuan Z, Wang P. mTOR inhibition reversed drug resistance after combination radiation with erlotinib in lung adenocarcinoma. Oncotarget. (Epub 2016 Oct 4). Zhuang H, Bai J, Chang JY, Yuan Z, Wang P. mTOR inhibition reversed drug resistance after combination radiation with erlotinib in lung adenocarcinoma. Oncotarget. (Epub 2016 Oct 4).
44.
go back to reference Li J, Dang Y, Gao J, Li Y, Zou J, Shen L. PI3K/AKT/mTOR pathway is activated after imatinib secondary resistance in gastrointestinal stromal tumors (GISTs). Med Oncol. 2015;32:111.PubMedCrossRef Li J, Dang Y, Gao J, Li Y, Zou J, Shen L. PI3K/AKT/mTOR pathway is activated after imatinib secondary resistance in gastrointestinal stromal tumors (GISTs). Med Oncol. 2015;32:111.PubMedCrossRef
46.
go back to reference Soria JC, Massard C, Lazar V, Ozoux ML, Mery-Mignard D, Deslandes A, et al. A dose finding, safety and pharmacokinetic study of AVE1642, an anti-insulin-like growth factor-1 receptor (IGF-1R/CD221) monoclonal antibody, administered as a single agent and in combination with docetaxel in patients with advanced solid tumours. Eur J Cancer. 2013;49:1799–807.PubMedCrossRef Soria JC, Massard C, Lazar V, Ozoux ML, Mery-Mignard D, Deslandes A, et al. A dose finding, safety and pharmacokinetic study of AVE1642, an anti-insulin-like growth factor-1 receptor (IGF-1R/CD221) monoclonal antibody, administered as a single agent and in combination with docetaxel in patients with advanced solid tumours. Eur J Cancer. 2013;49:1799–807.PubMedCrossRef
47.
go back to reference Schöffski P, Adkins D, Blay JY, Gil T, Elias AD, Rutkowski P, et al. An open-label, phase 2 study evaluating the efficacy and safety of the anti-IGF-1R antibody cixutumumab in patients with previously treated advanced or metastatic soft-tissue sarcoma or Ewing family of tumours. Eur J Cancer. 2013;49:3219–28.PubMedCrossRef Schöffski P, Adkins D, Blay JY, Gil T, Elias AD, Rutkowski P, et al. An open-label, phase 2 study evaluating the efficacy and safety of the anti-IGF-1R antibody cixutumumab in patients with previously treated advanced or metastatic soft-tissue sarcoma or Ewing family of tumours. Eur J Cancer. 2013;49:3219–28.PubMedCrossRef
48.
go back to reference Rajan A, Carter CA, Berman A, Cao L, Kelly RJ, Thomas A, et al. Cixutumumab for patients with recurrent or refractory advanced thymic epithelial tumours: a multicentre, open-label, phase 2 trial. Lancet Oncol. 2014;15:191–200.PubMedPubMedCentralCrossRef Rajan A, Carter CA, Berman A, Cao L, Kelly RJ, Thomas A, et al. Cixutumumab for patients with recurrent or refractory advanced thymic epithelial tumours: a multicentre, open-label, phase 2 trial. Lancet Oncol. 2014;15:191–200.PubMedPubMedCentralCrossRef
49.
go back to reference Weigel B, Malempati S, Reid JM, Voss SD, Cho SY, Chen HX, et al. Phase 2 trial of cixutumumab in children, adolescents, and young adults with refractory solid tumors: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2014;61:452–6.PubMedCrossRef Weigel B, Malempati S, Reid JM, Voss SD, Cho SY, Chen HX, et al. Phase 2 trial of cixutumumab in children, adolescents, and young adults with refractory solid tumors: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2014;61:452–6.PubMedCrossRef
50.
go back to reference Doi T, Shitara K, Kojima T, Yoshino T, Dontabhaktuni A, Rebscher H, et al. A phase I study evaluating cixutumumab, a type 1 insulin-like growth factor receptor inhibitor, given every 2 or 3 weeks in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2016;77:1253–62.PubMedCrossRef Doi T, Shitara K, Kojima T, Yoshino T, Dontabhaktuni A, Rebscher H, et al. A phase I study evaluating cixutumumab, a type 1 insulin-like growth factor receptor inhibitor, given every 2 or 3 weeks in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2016;77:1253–62.PubMedCrossRef
51.
go back to reference Busaidy NL, LoRusso P, Lawhorn K, Hess KR, Habra MA, Fu S, et al. The prevalence and impact of hyperglycemia and hyperlipidemia in patients with advanced cancer receiving combination treatment with the mammalian target of rapamycin inhibitor temsirolimus and insulin growth factor-receptor antibody cixutumumab. Oncologist. 2015;20:737–41.PubMedPubMedCentralCrossRef Busaidy NL, LoRusso P, Lawhorn K, Hess KR, Habra MA, Fu S, et al. The prevalence and impact of hyperglycemia and hyperlipidemia in patients with advanced cancer receiving combination treatment with the mammalian target of rapamycin inhibitor temsirolimus and insulin growth factor-receptor antibody cixutumumab. Oncologist. 2015;20:737–41.PubMedPubMedCentralCrossRef
52.
go back to reference Atzori F, Tabernero J, Cervantes A, Prudkin L, Andreu J, Rodríguez-Braun E, et al. A phase I pharmacokinetic and pharmacodynamic study of dalotuzumab (MK-0646), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res. 2011;17:6304–12.PubMedCrossRef Atzori F, Tabernero J, Cervantes A, Prudkin L, Andreu J, Rodríguez-Braun E, et al. A phase I pharmacokinetic and pharmacodynamic study of dalotuzumab (MK-0646), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res. 2011;17:6304–12.PubMedCrossRef
53.
go back to reference Doi T, Muro K, Yoshino T, Fuse N, Ura T, Takahari D, et al. Phase 1 pharmacokinetic study of MK-0646 (dalotuzumab), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in combination with cetuximab and irinotecan in Japanese patients with advanced colorectal cancer. Cancer Chemother Pharmacol. 2013;72:643–52.PubMedPubMedCentralCrossRef Doi T, Muro K, Yoshino T, Fuse N, Ura T, Takahari D, et al. Phase 1 pharmacokinetic study of MK-0646 (dalotuzumab), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in combination with cetuximab and irinotecan in Japanese patients with advanced colorectal cancer. Cancer Chemother Pharmacol. 2013;72:643–52.PubMedPubMedCentralCrossRef
54.
go back to reference Reidy-Lagunes DL, Vakiani E, Segal MF, Hollywood EM, Tang LH, Solit DB, et al. A phase 2 study of the insulin-like growth factor-1 receptor inhibitor MK-0646 in patients with metastatic, well-differentiated neuroendocrine tumors. Cancer. 2012;118:4795–800.PubMedCrossRef Reidy-Lagunes DL, Vakiani E, Segal MF, Hollywood EM, Tang LH, Solit DB, et al. A phase 2 study of the insulin-like growth factor-1 receptor inhibitor MK-0646 in patients with metastatic, well-differentiated neuroendocrine tumors. Cancer. 2012;118:4795–800.PubMedCrossRef
55.
go back to reference Haluska P, Worden F, Olmos D, Yin D, Schteingart D, Batzel GN, et al. Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother Pharmacol. 2010;65:765–73.PubMedCrossRef Haluska P, Worden F, Olmos D, Yin D, Schteingart D, Batzel GN, et al. Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother Pharmacol. 2010;65:765–73.PubMedCrossRef
56.
go back to reference von Mehren M, Britten CD, Pieslor P, Saville W, Vassos A, Harris S, et al. A phase 1, open-label, dose-escalation study of BIIB022 (anti-IGF-1R monoclonal antibody) in subjects with relapsed or refractory solid tumors. Invest New Drugs. 2014;32:518–25.CrossRef von Mehren M, Britten CD, Pieslor P, Saville W, Vassos A, Harris S, et al. A phase 1, open-label, dose-escalation study of BIIB022 (anti-IGF-1R monoclonal antibody) in subjects with relapsed or refractory solid tumors. Invest New Drugs. 2014;32:518–25.CrossRef
57.
go back to reference Titze MI, Schaaf O, Hofmann MH, Sanderson MP, Zahn SK, Quant J, et al. A comprehensive pharmacokinetic/pharmacodynamics analysis of the novel IGF1R/INSR inhibitor BI 893923 applying in vitro, in vivo and in silico modeling techniques. Cancer Chemother Pharmacol. 2016;77:1303–14.PubMedCrossRef Titze MI, Schaaf O, Hofmann MH, Sanderson MP, Zahn SK, Quant J, et al. A comprehensive pharmacokinetic/pharmacodynamics analysis of the novel IGF1R/INSR inhibitor BI 893923 applying in vitro, in vivo and in silico modeling techniques. Cancer Chemother Pharmacol. 2016;77:1303–14.PubMedCrossRef
58.
go back to reference Puzanov I, Lindsay CR, Goff L, Sosman J, Gilbert J, Berlin J, et al. A phase I study of continuous oral dosing of OSI-906, a dual inhibitor of insulin-like growth factor-1 and insulin receptors, in patients with advanced solid tumors. Clin Cancer Res. 2015;21:701–11.PubMedCrossRef Puzanov I, Lindsay CR, Goff L, Sosman J, Gilbert J, Berlin J, et al. A phase I study of continuous oral dosing of OSI-906, a dual inhibitor of insulin-like growth factor-1 and insulin receptors, in patients with advanced solid tumors. Clin Cancer Res. 2015;21:701–11.PubMedCrossRef
59.
go back to reference Schwartz GK, Dickson MA, LoRusso PM, Sausville EA, Maekawa Y, Watanabe Y, et al. Preclinical and first-in-human phase I studies of KW-2450, an oral tyrosine kinase inhibitor with insulin-like growth factor receptor-1/insulin receptor selectivity. Cancer Sci. 2016;107:499–506.PubMedPubMedCentralCrossRef Schwartz GK, Dickson MA, LoRusso PM, Sausville EA, Maekawa Y, Watanabe Y, et al. Preclinical and first-in-human phase I studies of KW-2450, an oral tyrosine kinase inhibitor with insulin-like growth factor receptor-1/insulin receptor selectivity. Cancer Sci. 2016;107:499–506.PubMedPubMedCentralCrossRef
60.
go back to reference Desai J, Solomon BJ, Davis ID, Lipton LR, Hicks R, Scott AM, et al. Phase I dose-escalation study of daily BMS-754807, an oral, dual IGF-1R/insulin receptor (IR) inhibitor in subjects with solid tumors. J Clin Oncol. 2010;28(Suppl):3104. Desai J, Solomon BJ, Davis ID, Lipton LR, Hicks R, Scott AM, et al. Phase I dose-escalation study of daily BMS-754807, an oral, dual IGF-1R/insulin receptor (IR) inhibitor in subjects with solid tumors. J Clin Oncol. 2010;28(Suppl):3104.
64.
go back to reference Yamanaka K, Petrulionis M, Lin S, Gao C, Galli U, Richter S, et al. Therapeutic potential and adverse events of everolimus for treatment of hepatocellular carcinoma—systematic review and meta-analysis. Cancer Med. 2013;2:862–71.PubMedPubMedCentralCrossRef Yamanaka K, Petrulionis M, Lin S, Gao C, Galli U, Richter S, et al. Therapeutic potential and adverse events of everolimus for treatment of hepatocellular carcinoma—systematic review and meta-analysis. Cancer Med. 2013;2:862–71.PubMedPubMedCentralCrossRef
65.
go back to reference Sivendran S, Agarwal N, Gartrell B, Ying J, Boucher KM, Choueiri TK, et al. Metabolic complications with the use of mTOR inhibitors for cancer therapy. Cancer Treat Rev. 2014;40:190–6.PubMedCrossRef Sivendran S, Agarwal N, Gartrell B, Ying J, Boucher KM, Choueiri TK, et al. Metabolic complications with the use of mTOR inhibitors for cancer therapy. Cancer Treat Rev. 2014;40:190–6.PubMedCrossRef
66.
go back to reference Lew S, Chamberlain RS. Risk of metabolic complications in patients with solid tumors treated with mTOR inhibitors: meta-analysis. Anticancer Res. 2016;36:1711–8.PubMed Lew S, Chamberlain RS. Risk of metabolic complications in patients with solid tumors treated with mTOR inhibitors: meta-analysis. Anticancer Res. 2016;36:1711–8.PubMed
67.
go back to reference Xu KY, Shameem R, Wu S. Risk of hyperglycemia attributable to everolimus in cancer patients: a meta-analysis. Acta Oncol. 2016;55(1196–203):68. Xu KY, Shameem R, Wu S. Risk of hyperglycemia attributable to everolimus in cancer patients: a meta-analysis. Acta Oncol. 2016;55(1196–203):68.
68.
go back to reference Sun Y, Rha S, Lee SH, Guo J, Ueda T, Qin S, et al. Phase II study of the safety and efficacy of temsirolimus in East Asian patients with advanced renal cell carcinoma. Jpn J Clin Oncol. 2012;42:836–44.PubMedCrossRef Sun Y, Rha S, Lee SH, Guo J, Ueda T, Qin S, et al. Phase II study of the safety and efficacy of temsirolimus in East Asian patients with advanced renal cell carcinoma. Jpn J Clin Oncol. 2012;42:836–44.PubMedCrossRef
69.
go back to reference Kruczek K, Ratterman M, Tolzien K, Sulo S, Lestingi TM, Nabhan C. A phase II study evaluating the toxicity and efficacy of single-agent temsirolimus in chemotherapy-naïve castration-resistant prostate cancer. Br J Cancer. 2013;109:1711–6.PubMedPubMedCentralCrossRef Kruczek K, Ratterman M, Tolzien K, Sulo S, Lestingi TM, Nabhan C. A phase II study evaluating the toxicity and efficacy of single-agent temsirolimus in chemotherapy-naïve castration-resistant prostate cancer. Br J Cancer. 2013;109:1711–6.PubMedPubMedCentralCrossRef
70.
go back to reference Oza AM, Pignata S, Poveda A, McCormack M, Clamp A, Schwartz B, et al. Randomized phase II trial of ridaforolimus in advanced endometrial carcinoma. J Clin Oncol. 2015;33:3576–82.PubMedCrossRef Oza AM, Pignata S, Poveda A, McCormack M, Clamp A, Schwartz B, et al. Randomized phase II trial of ridaforolimus in advanced endometrial carcinoma. J Clin Oncol. 2015;33:3576–82.PubMedCrossRef
71.
go back to reference Di Paolo S, Teutonico A, Leogrande D, Capobianco C, Schena PF. Chronic inhibition of mammalian target of rapamycin signaling downregulates insulin receptor substrates 1 and 2 and AKT activation: a crossroad between cancer and diabetes? J Am Soc Nephrol. 2006;17:2236–44.PubMedCrossRef Di Paolo S, Teutonico A, Leogrande D, Capobianco C, Schena PF. Chronic inhibition of mammalian target of rapamycin signaling downregulates insulin receptor substrates 1 and 2 and AKT activation: a crossroad between cancer and diabetes? J Am Soc Nephrol. 2006;17:2236–44.PubMedCrossRef
72.
go back to reference Crouthamel MC, Kahana JA, Korenchuk S, Zhang SY, Sundaresan G, Eberwein DJ, et al. Mechanism and management of AKT inhibitor-induced hyperglycemia. Clin Cancer Res. 2009;15:217–25.PubMedCrossRef Crouthamel MC, Kahana JA, Korenchuk S, Zhang SY, Sundaresan G, Eberwein DJ, et al. Mechanism and management of AKT inhibitor-induced hyperglycemia. Clin Cancer Res. 2009;15:217–25.PubMedCrossRef
73.
74.
go back to reference Khan KH, Wong M, Rihawi K, Bodla S, Morganstein D, Banerji U, et al. Hyperglycemia and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) inhibitors in phase I trials: incidence, predictive factors, and management. Oncologist. 2016;21:855–60.PubMedCrossRef Khan KH, Wong M, Rihawi K, Bodla S, Morganstein D, Banerji U, et al. Hyperglycemia and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) inhibitors in phase I trials: incidence, predictive factors, and management. Oncologist. 2016;21:855–60.PubMedCrossRef
75.
go back to reference Yap TA, Yan L, Patnaik A, Fearen I, Olmos D, Papadopoulos K, et al. First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors. J Clin Oncol. 2011;29:4688–95.PubMedCrossRef Yap TA, Yan L, Patnaik A, Fearen I, Olmos D, Papadopoulos K, et al. First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors. J Clin Oncol. 2011;29:4688–95.PubMedCrossRef
76.
go back to reference Tamura K, Hashimoto J, Tanabe Y, Kodaira M, Yonemori K, Seto T, et al. Safety and tolerability of AZD5363 in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2016;77:787–95.PubMedPubMedCentralCrossRef Tamura K, Hashimoto J, Tanabe Y, Kodaira M, Yonemori K, Seto T, et al. Safety and tolerability of AZD5363 in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2016;77:787–95.PubMedPubMedCentralCrossRef
77.
go back to reference Makker V, Recio FO, Ma L, Matulonis UA, Lauchle JO, Parmar H, et al. A multicenter, single-arm, open-label, phase 2 study of apitolisib (GDC-0980) for the treatment of recurrent or persistent endometrial carcinoma (MAGGIE study). Cancer. (Epub 2016 Sep 7). Makker V, Recio FO, Ma L, Matulonis UA, Lauchle JO, Parmar H, et al. A multicenter, single-arm, open-label, phase 2 study of apitolisib (GDC-0980) for the treatment of recurrent or persistent endometrial carcinoma (MAGGIE study). Cancer. (Epub 2016 Sep 7).
78.
go back to reference Aggarwal R, Grabowsky J, Strait N, Cockerill A, Munster P. Impact of patient ethnicity on the metabolic and immunologic effects of PI3K-mTOR pathway inhibition in patients with solid tumor malignancies. Cancer Chemother Pharmacol. 2014;74:359–65.PubMedPubMedCentralCrossRef Aggarwal R, Grabowsky J, Strait N, Cockerill A, Munster P. Impact of patient ethnicity on the metabolic and immunologic effects of PI3K-mTOR pathway inhibition in patients with solid tumor malignancies. Cancer Chemother Pharmacol. 2014;74:359–65.PubMedPubMedCentralCrossRef
79.
go back to reference Lee Y, Jung HS, Choi HJ, Kim MJ, Kim TM, Park KS, et al. Life-threatening hypoglycemia induced by a tyrosine kinase inhibitor in a patient with neuroendocrine tumor: a case report. Diabetes Res Clin Pract. 2011;93:e68–70.PubMedCrossRef Lee Y, Jung HS, Choi HJ, Kim MJ, Kim TM, Park KS, et al. Life-threatening hypoglycemia induced by a tyrosine kinase inhibitor in a patient with neuroendocrine tumor: a case report. Diabetes Res Clin Pract. 2011;93:e68–70.PubMedCrossRef
80.
go back to reference Demirci A, Bal O, Durnali A, Ekinci AŞ, Eşbah O, Alkiş N, et al. Sunitinib-induced severe hypoglycemia in a diabetic patient. J Oncol Pharm Pract. 2014;20:469–72.PubMedCrossRef Demirci A, Bal O, Durnali A, Ekinci AŞ, Eşbah O, Alkiş N, et al. Sunitinib-induced severe hypoglycemia in a diabetic patient. J Oncol Pharm Pract. 2014;20:469–72.PubMedCrossRef
81.
go back to reference Fountas A, Tigas S, Giotaki Z, Petrakis D, Pentheroudakis G, Tsatsoulis A. Severe resistant hypoglycemia in a patient with a pancreatic neuroendocrine tumor on sunitinib treatment. Hormones (Athens). 2015;14:438–41.PubMed Fountas A, Tigas S, Giotaki Z, Petrakis D, Pentheroudakis G, Tsatsoulis A. Severe resistant hypoglycemia in a patient with a pancreatic neuroendocrine tumor on sunitinib treatment. Hormones (Athens). 2015;14:438–41.PubMed
82.
go back to reference Ohn JH, Kim YG, Lee SH, Jung HS. Transformation of nonfunctioning pancreatic neuroendocrine carcinoma cells into insulin producing cells after treatment with sunitinib. Endocrinol Metab (Seoul). 2013;28:149–52.PubMedPubMedCentralCrossRef Ohn JH, Kim YG, Lee SH, Jung HS. Transformation of nonfunctioning pancreatic neuroendocrine carcinoma cells into insulin producing cells after treatment with sunitinib. Endocrinol Metab (Seoul). 2013;28:149–52.PubMedPubMedCentralCrossRef
83.
go back to reference Huda MS, Amiel SA, Ross P, Aylwin SJ. Tyrosine kinase inhibitor sunitinib allows insulin independence in long-standing type 1 diabetes. Diabetes Care. 2014;37:e87–8.PubMedCrossRef Huda MS, Amiel SA, Ross P, Aylwin SJ. Tyrosine kinase inhibitor sunitinib allows insulin independence in long-standing type 1 diabetes. Diabetes Care. 2014;37:e87–8.PubMedCrossRef
84.
go back to reference Thijs AM, Tack CJ, van der Graaf WT, Rongen GA, van Herpen CM. The early effect of sunitinib on insulin clearance in patients with metastatic renal cell carcinoma. Br J Clin Pharmacol. 2016;81:768–72.PubMedCrossRef Thijs AM, Tack CJ, van der Graaf WT, Rongen GA, van Herpen CM. The early effect of sunitinib on insulin clearance in patients with metastatic renal cell carcinoma. Br J Clin Pharmacol. 2016;81:768–72.PubMedCrossRef
85.
go back to reference Jalving M, Gietema JA, Lefrandt JD, de Jong S, Reyners AK, Gans RO, et al. Metformin: taking away the candy for cancer? Eur J Cancer. 2010;46:2369–80.PubMedCrossRef Jalving M, Gietema JA, Lefrandt JD, de Jong S, Reyners AK, Gans RO, et al. Metformin: taking away the candy for cancer? Eur J Cancer. 2010;46:2369–80.PubMedCrossRef
86.
go back to reference Tsilidis KK, Kasimis JC, Lopez DS, Ntzani EE, Ioannidis JP. Type 2 diabetes and cancer: umbrella review of meta-analyses of observational studies. BMJ. 2015;350:g7607.PubMedCrossRef Tsilidis KK, Kasimis JC, Lopez DS, Ntzani EE, Ioannidis JP. Type 2 diabetes and cancer: umbrella review of meta-analyses of observational studies. BMJ. 2015;350:g7607.PubMedCrossRef
88.
go back to reference Wojciechowska J, Krajewski W, Bolanowski M, Kręcicki T, Zatoński T. Diabetes and cancer: a review of current knowledge. Exp Clin Endocrinol Diabetes. 2016;124:263–75.PubMedCrossRef Wojciechowska J, Krajewski W, Bolanowski M, Kręcicki T, Zatoński T. Diabetes and cancer: a review of current knowledge. Exp Clin Endocrinol Diabetes. 2016;124:263–75.PubMedCrossRef
89.
go back to reference Mills KT, Bellows CF, Hoffman AE, Kelly TN, Gagliardi G. Diabetes mellitus and colorectal cancer prognosis: a meta-analysis. Dis Colon Rectum. 2013;56:1304–19.PubMedPubMedCentralCrossRef Mills KT, Bellows CF, Hoffman AE, Kelly TN, Gagliardi G. Diabetes mellitus and colorectal cancer prognosis: a meta-analysis. Dis Colon Rectum. 2013;56:1304–19.PubMedPubMedCentralCrossRef
90.
go back to reference Daugan M, Dufaÿ Wojcicki A, d’Hayer B, Boudy V. Metformin: an anti-diabetic drug to fight cancer. Pharmacol Res. 2016;113:675–85.PubMedCrossRef Daugan M, Dufaÿ Wojcicki A, d’Hayer B, Boudy V. Metformin: an anti-diabetic drug to fight cancer. Pharmacol Res. 2016;113:675–85.PubMedCrossRef
92.
go back to reference Nair V, Sreevalsan S, Basha R, Abdelrahim M, Abudayyeh A, Rodrigues Hoffman A, et al. Mechanism of metformin-dependent inhibition of mammalian target of rapamycin (mTOR) and Ras activity in pancreatic cancer: role of specificity protein (Sp) transcription factors. J Biol Chem. 2014;289:27692–701.PubMedPubMedCentralCrossRef Nair V, Sreevalsan S, Basha R, Abdelrahim M, Abudayyeh A, Rodrigues Hoffman A, et al. Mechanism of metformin-dependent inhibition of mammalian target of rapamycin (mTOR) and Ras activity in pancreatic cancer: role of specificity protein (Sp) transcription factors. J Biol Chem. 2014;289:27692–701.PubMedPubMedCentralCrossRef
93.
go back to reference Bhaw-Luximon A, Jhurry D. Metformin in pancreatic cancer treatment: from clinical trials through basic research to biomarker quantification. J Cancer Res Clin Oncol. 2016;142:2159–71.PubMedCrossRef Bhaw-Luximon A, Jhurry D. Metformin in pancreatic cancer treatment: from clinical trials through basic research to biomarker quantification. J Cancer Res Clin Oncol. 2016;142:2159–71.PubMedCrossRef
94.
go back to reference Liu H, Scholz C, Zang C, Schefe JH, Habbel P, Regierer AC, et al. Metformin and the mTOR inhibitor everolimus (RAD001) sensitize breast cancer cells to the cytotoxic effect of chemotherapeutic drugs in vitro. Anticancer Res. 2012;32:1627–37.PubMed Liu H, Scholz C, Zang C, Schefe JH, Habbel P, Regierer AC, et al. Metformin and the mTOR inhibitor everolimus (RAD001) sensitize breast cancer cells to the cytotoxic effect of chemotherapeutic drugs in vitro. Anticancer Res. 2012;32:1627–37.PubMed
95.
go back to reference Wang Y, Wei J, Li L, Fan C, Sun Y. Combined use of metformin and everolimus is synergistic in the treatment of breast cancer cells. Oncol Res. 2014;22:193–201.PubMedCrossRef Wang Y, Wei J, Li L, Fan C, Sun Y. Combined use of metformin and everolimus is synergistic in the treatment of breast cancer cells. Oncol Res. 2014;22:193–201.PubMedCrossRef
96.
go back to reference Dowling RJ, Niraula S, Stambolic V, Goodwin PJ. Metformin in cancer: translational challenges. J Mol Endocrinol. 2012;48:R31–43.PubMedCrossRef Dowling RJ, Niraula S, Stambolic V, Goodwin PJ. Metformin in cancer: translational challenges. J Mol Endocrinol. 2012;48:R31–43.PubMedCrossRef
97.
go back to reference Coperchini F, Leporati P, Rotondi M, Chiovato L. Expanding the therapeutic spectrum of metformin: from diabetes to cancer. J Endocrinol Invest. 2015;38:1047–55.PubMedCrossRef Coperchini F, Leporati P, Rotondi M, Chiovato L. Expanding the therapeutic spectrum of metformin: from diabetes to cancer. J Endocrinol Invest. 2015;38:1047–55.PubMedCrossRef
98.
go back to reference Zhang HH, Guo XL. Combinational strategies of metformin and chemotherapy in cancers. Cancer Chemother Pharmacol. 2016;78:13–26.PubMedCrossRef Zhang HH, Guo XL. Combinational strategies of metformin and chemotherapy in cancers. Cancer Chemother Pharmacol. 2016;78:13–26.PubMedCrossRef
99.
go back to reference Chae YK, Arya A, Malecek MK, Shin DS, Carneiro B, Chandra S, et al. Repurposing metformin for cancer treatment: current clinical studies. Oncotarget. 2016;7:40767–80.PubMedPubMedCentral Chae YK, Arya A, Malecek MK, Shin DS, Carneiro B, Chandra S, et al. Repurposing metformin for cancer treatment: current clinical studies. Oncotarget. 2016;7:40767–80.PubMedPubMedCentral
100.
go back to reference Li L, Han R, Xiao H, Lin C, Wang Y, Liu H, et al. Metformin sensitizes EGFR-TKI-resistant human lung cancer cells in vitro and in vivo through inhibition of IL-6 signaling and EMT reversal. Clin Cancer Res. 2014;20:2714–26.PubMedCrossRef Li L, Han R, Xiao H, Lin C, Wang Y, Liu H, et al. Metformin sensitizes EGFR-TKI-resistant human lung cancer cells in vitro and in vivo through inhibition of IL-6 signaling and EMT reversal. Clin Cancer Res. 2014;20:2714–26.PubMedCrossRef
101.
go back to reference Li L, Wang Y, Peng T, Zhang K, Lin C, Han R, et al. Metformin restores crizotinib sensitivity in crizotinib-resistant human lung cancer cells through inhibition of IGF1-R signaling pathway. Oncotarget. 2016;7:34442–52.PubMedPubMedCentral Li L, Wang Y, Peng T, Zhang K, Lin C, Han R, et al. Metformin restores crizotinib sensitivity in crizotinib-resistant human lung cancer cells through inhibition of IGF1-R signaling pathway. Oncotarget. 2016;7:34442–52.PubMedPubMedCentral
102.
go back to reference Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, et al. The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov. 2014;4:662–73.PubMedPubMedCentralCrossRef Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, et al. The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov. 2014;4:662–73.PubMedPubMedCentralCrossRef
103.
go back to reference Peng M, Huang Y, Tao T, Peng CY, Su Q, Xu W, et al. Metformin and gefitinib cooperate to inhibit bladder cancer growth via both AMPK and EGFR pathways joining at Akt and Erk. Sci Rep. 2016;6:28611.PubMedPubMedCentralCrossRef Peng M, Huang Y, Tao T, Peng CY, Su Q, Xu W, et al. Metformin and gefitinib cooperate to inhibit bladder cancer growth via both AMPK and EGFR pathways joining at Akt and Erk. Sci Rep. 2016;6:28611.PubMedPubMedCentralCrossRef
104.
go back to reference Ma J, Guo Y, Chen S, Zhong C, Xue Y, Zhang Y, et al. Metformin enhances tamoxifen-mediated tumor growth inhibition in ER-positive breast carcinoma. BMC Cancer. 2014;14:172.PubMedPubMedCentralCrossRef Ma J, Guo Y, Chen S, Zhong C, Xue Y, Zhang Y, et al. Metformin enhances tamoxifen-mediated tumor growth inhibition in ER-positive breast carcinoma. BMC Cancer. 2014;14:172.PubMedPubMedCentralCrossRef
105.
106.
go back to reference Franciosi M, Lucisano G, Lapice E, Strippoli GF, Pellegrini F, Nicolucci A. Metformin therapy and risk of cancer in patients with type 2 diabetes: systematic review. PLoS One. 2013;8:e71583.PubMedPubMedCentralCrossRef Franciosi M, Lucisano G, Lapice E, Strippoli GF, Pellegrini F, Nicolucci A. Metformin therapy and risk of cancer in patients with type 2 diabetes: systematic review. PLoS One. 2013;8:e71583.PubMedPubMedCentralCrossRef
107.
go back to reference Gandini S, Puntoni M, Heckman-Stoddard BM, Dunn BK, Ford L, DeCensi A, et al. Metformin and cancer risk and mortality: a systematic review and meta-analysis taking into account biases and confounders. Cancer Prev Res (Phila). 2014;7:867–85.PubMedPubMedCentralCrossRef Gandini S, Puntoni M, Heckman-Stoddard BM, Dunn BK, Ford L, DeCensi A, et al. Metformin and cancer risk and mortality: a systematic review and meta-analysis taking into account biases and confounders. Cancer Prev Res (Phila). 2014;7:867–85.PubMedPubMedCentralCrossRef
108.
go back to reference Lin HC, Kachingwe BH, Lin HL, Cheng HW, Uang YS, Wang LH. Effects of metformin dose on cancer risk reduction in patients with type 2 diabetes mellitus: a 6-year follow-up study. Pharmacotherapy. 2014;34:36–45.PubMedCrossRef Lin HC, Kachingwe BH, Lin HL, Cheng HW, Uang YS, Wang LH. Effects of metformin dose on cancer risk reduction in patients with type 2 diabetes mellitus: a 6-year follow-up study. Pharmacotherapy. 2014;34:36–45.PubMedCrossRef
109.
110.
go back to reference Tseng CH. Use of metformin and risk of kidney cancer in patients with type 2 diabetes. Eur J Cancer. 2016;52:19–25.PubMedCrossRef Tseng CH. Use of metformin and risk of kidney cancer in patients with type 2 diabetes. Eur J Cancer. 2016;52:19–25.PubMedCrossRef
111.
go back to reference Tseng CH. Response to Letter to the Editor on comments on “Use of metformin and risk of kidney cancer in patients with type 2 diabetes”. Eur J Cancer. 2016;61:159–60.PubMedCrossRef Tseng CH. Response to Letter to the Editor on comments on “Use of metformin and risk of kidney cancer in patients with type 2 diabetes”. Eur J Cancer. 2016;61:159–60.PubMedCrossRef
112.
go back to reference Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B, et al. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev Res (Phila). 2010;3:1451–61.PubMedCrossRef Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B, et al. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev Res (Phila). 2010;3:1451–61.PubMedCrossRef
113.
go back to reference Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JM. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care. 2009;32:1620–5.PubMedPubMedCentralCrossRef Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JM. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care. 2009;32:1620–5.PubMedPubMedCentralCrossRef
114.
go back to reference Wu L, Zhu J, Prokop LJ, Murad MH. Pharmacologic therapy of diabetes and overall cancer risk and mortality: a meta-analysis of 265 studies. Sci Rep. 2015;5:10147.PubMedPubMedCentralCrossRef Wu L, Zhu J, Prokop LJ, Murad MH. Pharmacologic therapy of diabetes and overall cancer risk and mortality: a meta-analysis of 265 studies. Sci Rep. 2015;5:10147.PubMedPubMedCentralCrossRef
115.
go back to reference Stevens RJ, Ali R, Bankhead CR, Bethel MA, Cairns BJ, Camisasca RP, et al. Cancer outcomes and all-cause mortality in adults allocated to metformin: systematic review and collaborative meta-analysis of randomised clinical trials. Diabetologia. 2012;55:2593–603.PubMedCrossRef Stevens RJ, Ali R, Bankhead CR, Bethel MA, Cairns BJ, Camisasca RP, et al. Cancer outcomes and all-cause mortality in adults allocated to metformin: systematic review and collaborative meta-analysis of randomised clinical trials. Diabetologia. 2012;55:2593–603.PubMedCrossRef
116.
go back to reference Golozar A, Liu S, Lin JA, Peairs K, Yeh HC. Does metformin reduce cancer risks? Methodologic considerations. Curr Diab Rep. 2016;16:4.PubMedCrossRef Golozar A, Liu S, Lin JA, Peairs K, Yeh HC. Does metformin reduce cancer risks? Methodologic considerations. Curr Diab Rep. 2016;16:4.PubMedCrossRef
117.
go back to reference Becker C, Jick SS, Meier CR, Bodmer M. Metformin and the risk of renal cell carcinoma: a case–control analysis. Eur J Cancer Prev. (Epub 2016 Mar 3). Becker C, Jick SS, Meier CR, Bodmer M. Metformin and the risk of renal cell carcinoma: a case–control analysis. Eur J Cancer Prev. (Epub 2016 Mar 3).
118.
go back to reference Becker C, Jick SS, Meier CR, Bodmer M. No evidence for a decreased risk of thyroid cancer in association with use of metformin or other antidiabetic drugs: a case–control study. BMC Cancer. 2015;15:719.PubMedPubMedCentralCrossRef Becker C, Jick SS, Meier CR, Bodmer M. No evidence for a decreased risk of thyroid cancer in association with use of metformin or other antidiabetic drugs: a case–control study. BMC Cancer. 2015;15:719.PubMedPubMedCentralCrossRef
119.
go back to reference Becker C, Jick SS, Meier CR, Bodmer M. Metformin and the risk of head and neck cancer: a case–control analysis. Diabetes Obes Metab. 2014;16:1148–54.PubMedCrossRef Becker C, Jick SS, Meier CR, Bodmer M. Metformin and the risk of head and neck cancer: a case–control analysis. Diabetes Obes Metab. 2014;16:1148–54.PubMedCrossRef
120.
go back to reference Becker C, Meier CR, Jick SS, Bodmer M. Case–control analysis on metformin and cancer of the esophagus. Cancer Causes Control. 2013;24:1763–70.PubMedCrossRef Becker C, Meier CR, Jick SS, Bodmer M. Case–control analysis on metformin and cancer of the esophagus. Cancer Causes Control. 2013;24:1763–70.PubMedCrossRef
121.
go back to reference Becker C, Jick SS, Meier CR, Bodmer M. Metformin and the risk of endometrial cancer: a case–control analysis. Gynecol Oncol. 2013;129:565–9.PubMedCrossRef Becker C, Jick SS, Meier CR, Bodmer M. Metformin and the risk of endometrial cancer: a case–control analysis. Gynecol Oncol. 2013;129:565–9.PubMedCrossRef
122.
go back to reference Yin M, Zhou J, Gorak EJ, Quddus F. Metformin is associated with survival benefit in cancer patients with concurrent type 2 diabetes: a systematic review and meta-analysis. Oncologist. 2013;18:1248–55.PubMedPubMedCentralCrossRef Yin M, Zhou J, Gorak EJ, Quddus F. Metformin is associated with survival benefit in cancer patients with concurrent type 2 diabetes: a systematic review and meta-analysis. Oncologist. 2013;18:1248–55.PubMedPubMedCentralCrossRef
123.
go back to reference Lin JJ, Gallagher EJ, Sigel K, Mhango G, Galsky MD, Smith CB, et al. Survival of patients with stage IV lung cancer with diabetes treated with metformin. Am J Respir Crit Care Med. 2015;191:448–54.PubMedPubMedCentralCrossRef Lin JJ, Gallagher EJ, Sigel K, Mhango G, Galsky MD, Smith CB, et al. Survival of patients with stage IV lung cancer with diabetes treated with metformin. Am J Respir Crit Care Med. 2015;191:448–54.PubMedPubMedCentralCrossRef
124.
go back to reference Chen H, Yao W, Chu Q, Han R, Wang Y, Sun J, et al. Synergistic effects of metformin in combination with EGFR-TKI in the treatment of patients with advanced non-small cell lung cancer and type 2 diabetes. Cancer Lett. 2015;369:97–102.PubMedCrossRef Chen H, Yao W, Chu Q, Han R, Wang Y, Sun J, et al. Synergistic effects of metformin in combination with EGFR-TKI in the treatment of patients with advanced non-small cell lung cancer and type 2 diabetes. Cancer Lett. 2015;369:97–102.PubMedCrossRef
125.
go back to reference Xu T, Liang G, Yang L, Zhang F. Prognosis of small cell lung cancer patients with diabetes treated with metformin. Clin Transl Oncol. 2015;17:819–24.PubMedCrossRef Xu T, Liang G, Yang L, Zhang F. Prognosis of small cell lung cancer patients with diabetes treated with metformin. Clin Transl Oncol. 2015;17:819–24.PubMedCrossRef
126.
go back to reference Kong F, Gao F, Liu H, Chen L, Zheng R, Yu J, et al. Metformin use improves the survival of diabetic combined small-cell lung cancer patients. Tumour Biol. 2015;36:8101–6.PubMedCrossRef Kong F, Gao F, Liu H, Chen L, Zheng R, Yu J, et al. Metformin use improves the survival of diabetic combined small-cell lung cancer patients. Tumour Biol. 2015;36:8101–6.PubMedCrossRef
127.
go back to reference Wink KC, Belderbos JS, Dieleman EM, Rossi M, Rasch CR, Damhuis RA, et al. Improved progression free survival for patients with diabetes and locally advanced non-small cell lung cancer (NSCLC) using metformin during concurrent chemoradiotherapy. Radiother Oncol. 2016;118:453–9.PubMedCrossRef Wink KC, Belderbos JS, Dieleman EM, Rossi M, Rasch CR, Damhuis RA, et al. Improved progression free survival for patients with diabetes and locally advanced non-small cell lung cancer (NSCLC) using metformin during concurrent chemoradiotherapy. Radiother Oncol. 2016;118:453–9.PubMedCrossRef
128.
go back to reference Wan G, Yu X, Chen P, Wang X, Pan D, Wang X, et al. Metformin therapy associated with survival benefit in lung cancer patients with diabetes. Oncotarget. 2016;7:35437–45.PubMedPubMedCentral Wan G, Yu X, Chen P, Wang X, Pan D, Wang X, et al. Metformin therapy associated with survival benefit in lung cancer patients with diabetes. Oncotarget. 2016;7:35437–45.PubMedPubMedCentral
129.
go back to reference Tian RH, Zhang YG, Wu Z, Liu X, Yang JW, Ji HL. Effects of metformin on survival outcomes of lung cancer patients with type 2 diabetes mellitus: a meta-analysis. Clin Transl Oncol. 2016;18:641–9.PubMedCrossRef Tian RH, Zhang YG, Wu Z, Liu X, Yang JW, Ji HL. Effects of metformin on survival outcomes of lung cancer patients with type 2 diabetes mellitus: a meta-analysis. Clin Transl Oncol. 2016;18:641–9.PubMedCrossRef
130.
go back to reference Sadeghi N, Abbruzzese JL, Yeung SC, Hassan M, Li D. Metformin use is associated with better survival of diabetic patients with pancreatic cancer. Clin Cancer Res. 2012;18:2905–12.PubMedPubMedCentralCrossRef Sadeghi N, Abbruzzese JL, Yeung SC, Hassan M, Li D. Metformin use is associated with better survival of diabetic patients with pancreatic cancer. Clin Cancer Res. 2012;18:2905–12.PubMedPubMedCentralCrossRef
131.
go back to reference Lee SH, Yoon SH, Lee HS, Chung MJ, Park JY, Park SW, et al. Can metformin change the prognosis of pancreatic cancer? Retrospective study for pancreatic cancer patients with pre-existing diabetes mellitus type 2. Dig Liver Dis. 2016;48:435–40.PubMedCrossRef Lee SH, Yoon SH, Lee HS, Chung MJ, Park JY, Park SW, et al. Can metformin change the prognosis of pancreatic cancer? Retrospective study for pancreatic cancer patients with pre-existing diabetes mellitus type 2. Dig Liver Dis. 2016;48:435–40.PubMedCrossRef
132.
go back to reference Cerullo M, Gani F, Chen SY, Canner J, Pawlik TM. Metformin use is associated with improved survival in patients undergoing resection for pancreatic cancer. J Gastrointest Surg. 2016;20:1572–80.PubMedCrossRef Cerullo M, Gani F, Chen SY, Canner J, Pawlik TM. Metformin use is associated with improved survival in patients undergoing resection for pancreatic cancer. J Gastrointest Surg. 2016;20:1572–80.PubMedCrossRef
133.
go back to reference Ambe CM, Mahipal A, Fulp J, Chen L, Malafa MP. Effect of metformin use on survival in resectable pancreatic cancer: a single-institution experience and review of the literature. PLoS One. 2016;11:e0151632.PubMedPubMedCentralCrossRef Ambe CM, Mahipal A, Fulp J, Chen L, Malafa MP. Effect of metformin use on survival in resectable pancreatic cancer: a single-institution experience and review of the literature. PLoS One. 2016;11:e0151632.PubMedPubMedCentralCrossRef
134.
go back to reference Keizman D, Ish-Shalom M, Sella A, Gottfried M, Maimon N, Peer A, et al. Metformin use and outcome of sunitinib treatment in patients with diabetes and metastatic renal cell carcinoma. Clin Genitourin Cancer. 2016;14:420–5.PubMedCrossRef Keizman D, Ish-Shalom M, Sella A, Gottfried M, Maimon N, Peer A, et al. Metformin use and outcome of sunitinib treatment in patients with diabetes and metastatic renal cell carcinoma. Clin Genitourin Cancer. 2016;14:420–5.PubMedCrossRef
135.
go back to reference Cheng JJ, Li H, Tan HS, Tan PH, Ng LG, Ng QS, et al. Metformin use in relation with survival outcomes of patients with renal cell carcinoma. Clin Genitourin Cancer. 2016;14:168–75.PubMedCrossRef Cheng JJ, Li H, Tan HS, Tan PH, Ng LG, Ng QS, et al. Metformin use in relation with survival outcomes of patients with renal cell carcinoma. Clin Genitourin Cancer. 2016;14:168–75.PubMedCrossRef
136.
go back to reference Ezewuiro O, Grushko TA, Kocherginsky M, Habis M, Hurteau JA, Mills KA, et al. Association of metformin use with outcomes in advanced endometrial cancer treated with chemotherapy. PLoS One. 2016;11:e0147145.PubMedPubMedCentralCrossRef Ezewuiro O, Grushko TA, Kocherginsky M, Habis M, Hurteau JA, Mills KA, et al. Association of metformin use with outcomes in advanced endometrial cancer treated with chemotherapy. PLoS One. 2016;11:e0147145.PubMedPubMedCentralCrossRef
137.
go back to reference Jacob L, Kostev K, Rathmann W, Kalder M. Impact of metformin on metastases in patients with breast cancer and type 2 diabetes. J Diabetes Complicat. 2016;30:1056–9.PubMedCrossRef Jacob L, Kostev K, Rathmann W, Kalder M. Impact of metformin on metastases in patients with breast cancer and type 2 diabetes. J Diabetes Complicat. 2016;30:1056–9.PubMedCrossRef
138.
go back to reference Xu H, Chen K, Jia X, Tian Y, Dai Y, Li D, et al. Metformin use is associated with better survival of breast cancer patients with diabetes: a meta-analysis. Oncologist. 2015;20:1236–44.PubMedPubMedCentralCrossRef Xu H, Chen K, Jia X, Tian Y, Dai Y, Li D, et al. Metformin use is associated with better survival of breast cancer patients with diabetes: a meta-analysis. Oncologist. 2015;20:1236–44.PubMedPubMedCentralCrossRef
139.
go back to reference Mei ZB, Zhang ZJ, Liu CY, Liu Y, Cui A, Liang ZL, et al. Survival benefits of metformin for colorectal cancer patients with diabetes: a systematic review and meta-analysis. PLoS One. 2014;9:e91818.PubMedPubMedCentralCrossRef Mei ZB, Zhang ZJ, Liu CY, Liu Y, Cui A, Liang ZL, et al. Survival benefits of metformin for colorectal cancer patients with diabetes: a systematic review and meta-analysis. PLoS One. 2014;9:e91818.PubMedPubMedCentralCrossRef
140.
go back to reference Ki YJ, Kim HJ, Kim MS, Park CM, Ko MJ, Seo YS, et al. Association between metformin use and survival in non-metastatic rectal cancer treated with a curative resection: a nationwide population study. Cancer Res Treat. (Epub 2016 Jul 4). Ki YJ, Kim HJ, Kim MS, Park CM, Ko MJ, Seo YS, et al. Association between metformin use and survival in non-metastatic rectal cancer treated with a curative resection: a nationwide population study. Cancer Res Treat. (Epub 2016 Jul 4).
142.
go back to reference Chong RW, Vasudevan V, Zuber J, Solomon SS. Metformin has a positive therapeutic effect on prostate cancer in patients with type 2 diabetes mellitus. Am J Med Sci. 2016;351:416–9.PubMedCrossRef Chong RW, Vasudevan V, Zuber J, Solomon SS. Metformin has a positive therapeutic effect on prostate cancer in patients with type 2 diabetes mellitus. Am J Med Sci. 2016;351:416–9.PubMedCrossRef
143.
go back to reference Rêgo DF, Pavan LM, Elias ST, De Luca Canto G, Guerra EN. Effects of metformin on head and neck cancer: a systematic review. Oral Oncol. 2015;51:416–22.PubMedCrossRef Rêgo DF, Pavan LM, Elias ST, De Luca Canto G, Guerra EN. Effects of metformin on head and neck cancer: a systematic review. Oral Oncol. 2015;51:416–22.PubMedCrossRef
144.
go back to reference Sayed R, Saad AS, El Wakeel L, Elkholy E, Badary O. Metformin addition to chemotherapy in stage IV non-small cell lung cancer: an open label randomized controlled study. Asian Pac J Cancer Prev. 2015;16:6621–6.PubMedCrossRef Sayed R, Saad AS, El Wakeel L, Elkholy E, Badary O. Metformin addition to chemotherapy in stage IV non-small cell lung cancer: an open label randomized controlled study. Asian Pac J Cancer Prev. 2015;16:6621–6.PubMedCrossRef
145.
go back to reference Menamin ÚC, Cardwell CR, Hughes CM, Murray LM. Metformin use and survival from lung cancer: a population-based cohort study. Lung Cancer. 2016;94:35–9.PubMedCrossRef Menamin ÚC, Cardwell CR, Hughes CM, Murray LM. Metformin use and survival from lung cancer: a population-based cohort study. Lung Cancer. 2016;94:35–9.PubMedCrossRef
146.
go back to reference Menamin ÚC, Murray LJ, Hughes CM, Cardwell CR. Metformin use and survival after colorectal cancer: a population-based cohort study. Int J Cancer. 2016;138:369–79.CrossRef Menamin ÚC, Murray LJ, Hughes CM, Cardwell CR. Metformin use and survival after colorectal cancer: a population-based cohort study. Int J Cancer. 2016;138:369–79.CrossRef
147.
go back to reference Kordes S, Pollak MN, Zwinderman AH, Mathôt RA, Weterman MJ, Beeker A, et al. Metformin in patients with advanced pancreatic cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol. 2015;16:839–47.PubMedCrossRef Kordes S, Pollak MN, Zwinderman AH, Mathôt RA, Weterman MJ, Beeker A, et al. Metformin in patients with advanced pancreatic cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol. 2015;16:839–47.PubMedCrossRef
148.
go back to reference Reni M, Dugnani E, Cereda S, Belli C, Balzano G, Nicoletti R, et al. (Ir)relevance of metformin treatment in patients with metastatic pancreatic cancer: an open-label, randomized phase II trial. Clin Cancer Res. 2016;22:1076–85.PubMedCrossRef Reni M, Dugnani E, Cereda S, Belli C, Balzano G, Nicoletti R, et al. (Ir)relevance of metformin treatment in patients with metastatic pancreatic cancer: an open-label, randomized phase II trial. Clin Cancer Res. 2016;22:1076–85.PubMedCrossRef
149.
go back to reference Hamieh L, McKay RR, Lin X, Moreira RB, Simantov R, Choueiri TK. The impact of metformin use on survival outcomes in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer. (Epub 2016 Jun 29). Hamieh L, McKay RR, Lin X, Moreira RB, Simantov R, Choueiri TK. The impact of metformin use on survival outcomes in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer. (Epub 2016 Jun 29).
150.
go back to reference Nayan M, Finelli A, Jewett MAS, Juurlink DN, Austin PC, Kulkarni GS, et al. Metformin use and kidney cancer outcomes in patients with diabetes: a propensity score analysis. Clin Genitourin Cancer. (Epub 2016 Jun 23). Nayan M, Finelli A, Jewett MAS, Juurlink DN, Austin PC, Kulkarni GS, et al. Metformin use and kidney cancer outcomes in patients with diabetes: a propensity score analysis. Clin Genitourin Cancer. (Epub 2016 Jun 23).
151.
go back to reference Shah RR. Comment on: Bono P, Oudard S, Bodrogi I, et al. Outcomes in patients with metastatic renal cell carcinoma who develop everolimus-related hyperglycemia and hypercholesterolemia: combined subgroup analyses of the RECORD-1 and REACT trials. Clin Genitourin Cancer. (Epub 2016 Jul 21). Shah RR. Comment on: Bono P, Oudard S, Bodrogi I, et al. Outcomes in patients with metastatic renal cell carcinoma who develop everolimus-related hyperglycemia and hypercholesterolemia: combined subgroup analyses of the RECORD-1 and REACT trials. Clin Genitourin Cancer. (Epub 2016 Jul 21).
152.
go back to reference Goodwin PJ, Parulekar WR, Gelmon KA, Shepherd LE, Ligibel JA, Hershman DL, et al. Effect of metformin vs placebo on and metabolic factors in NCIC CTG MA.32. J Natl Cancer Inst. 2015;107(3):djv006.PubMedPubMedCentralCrossRef Goodwin PJ, Parulekar WR, Gelmon KA, Shepherd LE, Ligibel JA, Hershman DL, et al. Effect of metformin vs placebo on and metabolic factors in NCIC CTG MA.32. J Natl Cancer Inst. 2015;107(3):djv006.PubMedPubMedCentralCrossRef
153.
go back to reference Hanprasertpong J, Jiamset I, Geater A, Peerawong T, Hemman W, Kornsilp S. The effect of metformin on oncological outcomes in patients with cervical cancer with type 2 diabetes mellitus. Int J Gynecol Cancer. (Epub 2016 Nov 17). Hanprasertpong J, Jiamset I, Geater A, Peerawong T, Hemman W, Kornsilp S. The effect of metformin on oncological outcomes in patients with cervical cancer with type 2 diabetes mellitus. Int J Gynecol Cancer. (Epub 2016 Nov 17).
154.
go back to reference Jayalath VH, Ireland C, Fleshner NE, Hamilton RJ, Jenkins DJ. The relationship between metformin and serum prostate-specific antigen levels. Prostate. 2016;76:1445–53.PubMedPubMedCentralCrossRef Jayalath VH, Ireland C, Fleshner NE, Hamilton RJ, Jenkins DJ. The relationship between metformin and serum prostate-specific antigen levels. Prostate. 2016;76:1445–53.PubMedPubMedCentralCrossRef
155.
go back to reference Sequist LV, Soria JC, Goldman JW, Wakelee HA, Gadgeel SM, Varga A, et al. Rociletinib in EGFR-mutated non-small-cell lung cancer. N Engl J Med. 2015;372:1700–9.PubMedCrossRef Sequist LV, Soria JC, Goldman JW, Wakelee HA, Gadgeel SM, Varga A, et al. Rociletinib in EGFR-mutated non-small-cell lung cancer. N Engl J Med. 2015;372:1700–9.PubMedCrossRef
156.
go back to reference Sequist LV, Soria JC, Camidge DR. Update to rociletinib data with the RECIST confirmed response rate. N Engl J Med. 2016;374:2296–7.PubMedCrossRef Sequist LV, Soria JC, Camidge DR. Update to rociletinib data with the RECIST confirmed response rate. N Engl J Med. 2016;374:2296–7.PubMedCrossRef
157.
go back to reference Bono P, Oudard S, Bodrogi I, Hutson TE, Escudier B, Machiels JP, et al. Outcomes in patients with metastatic renal cell carcinoma who develop everolimus-related hyperglycemia and hypercholesterolemia: combined subgroup analyses of the RECORD-1 and REACT trials. Clin Genitourin Cancer. 2016;14:406–14.PubMedPubMedCentralCrossRef Bono P, Oudard S, Bodrogi I, Hutson TE, Escudier B, Machiels JP, et al. Outcomes in patients with metastatic renal cell carcinoma who develop everolimus-related hyperglycemia and hypercholesterolemia: combined subgroup analyses of the RECORD-1 and REACT trials. Clin Genitourin Cancer. 2016;14:406–14.PubMedPubMedCentralCrossRef
158.
go back to reference Sequist LV, Rolfe L, Allen AR. Rociletinib in EGFR-mutated non-small-cell lung cancer. N Engl J Med. 2015;373:578–9.PubMedCrossRef Sequist LV, Rolfe L, Allen AR. Rociletinib in EGFR-mutated non-small-cell lung cancer. N Engl J Med. 2015;373:578–9.PubMedCrossRef
159.
go back to reference Gong Z, Aragaki AK, Chlebowski RT, Manson JE, Rohan TE, Chen C, et al. Diabetes, metformin and incidence of and death from invasive cancer in postmenopausal women: Results from the women’s health initiative. Int J Cancer. 2016;138:1915–27.PubMedCrossRef Gong Z, Aragaki AK, Chlebowski RT, Manson JE, Rohan TE, Chen C, et al. Diabetes, metformin and incidence of and death from invasive cancer in postmenopausal women: Results from the women’s health initiative. Int J Cancer. 2016;138:1915–27.PubMedCrossRef
160.
go back to reference Gartrell BA, Ying J, Sivendran S, Boucher KM, Choueiri TK, Sonpavde G, et al. Pulmonary complications with the use of mTOR inhibitors in targeted cancer therapy: a systematic review and meta-analysis. Target Oncol. 2014;9:195–204.PubMedCrossRef Gartrell BA, Ying J, Sivendran S, Boucher KM, Choueiri TK, Sonpavde G, et al. Pulmonary complications with the use of mTOR inhibitors in targeted cancer therapy: a systematic review and meta-analysis. Target Oncol. 2014;9:195–204.PubMedCrossRef
161.
go back to reference Willemsen AE, Grutters JC, Gerritsen WR, van Erp NP, van Herpen CM, Tol J. mTOR inhibitor-induced interstitial lung disease in cancer patients: Comprehensive review and a practical management algorithm. Int J Cancer. 2016;138:2312–21.PubMedCrossRef Willemsen AE, Grutters JC, Gerritsen WR, van Erp NP, van Herpen CM, Tol J. mTOR inhibitor-induced interstitial lung disease in cancer patients: Comprehensive review and a practical management algorithm. Int J Cancer. 2016;138:2312–21.PubMedCrossRef
162.
go back to reference Dolly SO, Wagner AJ, Bendell JC, Kindler HL, Krug LM, Seiwert TY, et al. Phase I study of apitolisib (GDC-0980), dual phosphatidylinositol-3-kinase and mammalian target of rapamycin kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res. 2016;22:2874–84.PubMedPubMedCentralCrossRef Dolly SO, Wagner AJ, Bendell JC, Kindler HL, Krug LM, Seiwert TY, et al. Phase I study of apitolisib (GDC-0980), dual phosphatidylinositol-3-kinase and mammalian target of rapamycin kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res. 2016;22:2874–84.PubMedPubMedCentralCrossRef
163.
164.
go back to reference Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45:1434–45.PubMedPubMedCentralCrossRef Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45:1434–45.PubMedPubMedCentralCrossRef
165.
go back to reference Li L, Huang W, Li K, Zhang K, Lin C, Han R, et al. Metformin attenuates gefitinib-induced exacerbation of pulmonary fibrosis by inhibition of TGF-β signaling pathway. Oncotarget. 2015;6:43605–19.PubMedPubMedCentral Li L, Huang W, Li K, Zhang K, Lin C, Han R, et al. Metformin attenuates gefitinib-induced exacerbation of pulmonary fibrosis by inhibition of TGF-β signaling pathway. Oncotarget. 2015;6:43605–19.PubMedPubMedCentral
166.
go back to reference Fasano M, Della Corte CM, Capuano A, Sasso FC, Papaccio F, Berrino L, et al. A multicenter, open-label phase II study of metformin with erlotinib in second-line therapy of stage IV non-small-cell lung cancer patients: treatment rationale and protocol dynamics of the METAL trial. Clin Lung Cancer. 2015;16:57–9.PubMedCrossRef Fasano M, Della Corte CM, Capuano A, Sasso FC, Papaccio F, Berrino L, et al. A multicenter, open-label phase II study of metformin with erlotinib in second-line therapy of stage IV non-small-cell lung cancer patients: treatment rationale and protocol dynamics of the METAL trial. Clin Lung Cancer. 2015;16:57–9.PubMedCrossRef
Metadata
Title
Hyperglycaemia Induced by Novel Anticancer Agents: An Undesirable Complication or a Potential Therapeutic Opportunity?
Author
Rashmi R. Shah
Publication date
01-03-2017
Publisher
Springer International Publishing
Published in
Drug Safety / Issue 3/2017
Print ISSN: 0114-5916
Electronic ISSN: 1179-1942
DOI
https://doi.org/10.1007/s40264-016-0485-y

Other articles of this Issue 3/2017

Drug Safety 3/2017 Go to the issue