AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.1 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Promising molecular mechanisms responsible for gemcitabine resistance in cancer

Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, China
Division of Hematology and Oncology, Department of Pediatrics, Wells Center for Pediatric Research, Indiana University Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

Gemcitabine is the first-line treatment for pancreatic ductual adenocarcinoma (PDAC) as well as acts against a wide range of other solid tumors. Patients usually have a good initial response to gemcitabine-based chemotherapy but would eventually develop resistance. To improve survival and prognosis of cancer patients, better understanding of the mechanisms responsible for gemcitabine resistance and discovery of new therapeutic strategies are in great need. Amounting evidence indicate that the developmental pathways, such as Hedgehog (Hh), Wnt and Notch, become reactivated in gemcitabine-resistant cancer cells. Thus, the strategies for targeting these pathways may sensitize cancer cells to gemcitabine treatment. In this review, we will summarize recent development in this area of research and discuss strategies to overcome gemcitabine resistance. Given the cross-talk between these three developmental signaling pathways, designing clinical trials using a cocktail of inhibitory agents targeting all these pathways may be more effective. Ultimately, our hope is that targeting these developmental pathways may be an effective way to improve the gemcitabine treatment outcome in cancer patients.

References

1

Bianchi V, Borella S, Calderazzo F, Ferraro P, Chieco Bianchi L, Reichard P. Inhibition of ribonucleotide reductase by 2’-substituted deoxycytidine analogs: possible application in AIDS treatment. Proc Natl Acad Sci U. S. A. 1994;91:8403-8407.

2

Gandhi V, Plunkett W. Modulatory activity of 2’,2’-difluorodeoxycytidine on the phosphorylation and cytotoxicity of arabinosyl nucleosides. Cancer Res. 1990;50:3675-3680.

3

Hertel LW, Boder GB, Kroin JS, et al. Evaluation of the antitumor activity of gemcitabine (2′,2′-difluoro-2′-deoxycytidine). Cancer Res. 1990;50:4417-4422.

4

Huang P, Chubb S, Hertel LW, Grindey GB, Plunkett W. Action of 2’,2’-difluorodeoxycytidine on DNA synthesis. Cancer Res. 1991;51:6110-6117.

5

Burris 3rd HA, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol - Off J Am Soc Clin Oncol. 1997;15:2403-2413.

6

Sandler AB, Nemunaitis J, Denham C, et al. Phase Ⅲ trial of gemcitabine plus cisplatin versus cisplatin alone in patients with locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol - Off J Am Soc Clin Oncol. 2000;18:122-130.

7

Nagourney RA, Flam M, Link J, et al. Carboplatin plus gemcitabine repeating doublet therapy in recurrent breast cancer. Clin Breast Cancer. 2008;8:432-435.

8

von der Maase H, Hansen SW, Roberts JT, et al. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase Ⅲ study. J Clin Oncol. 2000;18:3068-3077.

9

Pfisterer J, Plante M, Vergote I, et al. Gemcitabine plus carboplatin compared with carboplatin in patients with platinum-sensitive recurrent ovarian cancer: an intergroup trial of the AGO-OVAR, the NCIC CTG, and the EORTC GCG. J Clin Oncol. 2006;24:4699-4707.

10

Rauchwerger DR, Firby PS, Hedley DW, Moore MJ. Equilibrative-sensitive nucleoside transporter and its role in gemcitabine sensitivity. Cancer Res. 2000;60:6075-6079.

11

Spratlin J, Sangha R, Glubrecht D, et al. The absence of human equilibrative nucleoside transporter 1 is associated with reduced survival in patients with gemcitabine-treated pancreas adenocarcinoma. Clin Cancer Res. 2004;10:6956-6961.

12

Mini E, Nobili S, Caciagli B, Landini I, Mazzei T. Cellular pharmacology of gemcitabine. Ann Oncol. 2006;17:v7-12.

13

Heinemann V, Xu YZ, Chubb S, et al. Cellular elimination of 2′,2′-difluorodeoxycytidine 5′-triphosphate: a mechanism of self-potentiation. Cancer Res. 1992;52:533-539.

14

Zhou BS, Tsai P, Ker R, et al. Overexpression of transfected human ribonucleotide reductase M2 subunit in human cancer cells enhances their invasive potential. Clin Exp Metastasis. 1998;16:43-49.

15

Zhou J, Oliveira P, Li X, Chen Z, Bepler G. Modulation of the ribonucleotide reductase-antimetabolite drug interaction in cancer cell lines. J Nucleic Acids. 2010;2010:597098.

16

Costantino CL, Witkiewicz AK, Kuwano Y, et al. The role of HuR in gemcitabine efficacy in pancreatic cancer: HuR Up-regulates the expression of the gemcitabine metabolizing enzyme deoxycytidine kinase. Cancer Res. 2009;69:4567-4572.

17

Chen M, Xue X, Wang F, et al. Expression and promoter methylation analysis of ATP-binding cassette genes in pancreatic cancer. Oncol Rep. 2012;27:265-269.

18

Zinzi L, Contino M, Cantore M, Capparelli E, Leopoldo M, Colabufo NA. ABC transporters in CSCs membranes as a novel target for treating tumor relapse. Front Pharmacol. 2014;5:163.

19

Quint K, Tonigold M, Di Fazio P, et al. Pancreatic cancer cells surviving gemcitabine treatment express markers of stem cell differentiation and epithelial-mesenchymal transition. Int J Oncol. 2012;41:2093-2102.

20

Arlt A, Gehrz A, Muerkoster S, et al. Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene. 2003;22:3243-3251.

21

Pan X, Arumugam T, Yamamoto T, et al. Nuclear factor-kappaB p65/relA silencing induces apoptosis and increases gemcitabine effectiveness in a subset of pancreatic cancer cells. Clin Cancer Res - Off J Am Assoc Cancer Res. 2008;14:8143-8151.

22

Jung H, Kim JS, Kim WK, et al. Intracellular annexin A2 regulates NF-kappaB signaling by binding to the p50 subunit: implications for gemcitabine resistance in pancreatic cancer. Cell Death Dis. 2015;6:e1606.

23

Yu Y, Wang J, Xia N, Li B, Jiang X. Maslinic acid potentiates the antitumor activities of gemcitabine in vitro and in vivo by inhibiting NF-kappaB-mediated survival signaling pathways in human gallbladder cancer cells. Oncol Rep. 2015;33:1683-1690.

24

, McDunn JE, Kashiwagi H, et al. Targeting AKT with the proapoptotic peptide, TAT-CTMP: a novel strategy for the treatment of human pancreatic adenocarcinoma. Int J Cancer J. 2009;125:942-951.

25

Trehoux S, Duchene B, Jonckheere N, Van Seuningen I. The MUC1 oncomucin regulates pancreatic cancer cell biological properties and chemoresistance. Implication of p42-44 MAPK, Akt, Bcl-2 and MMP13 pathways. Biochem Biophysical Res Commun. 2015;456:757-762.

26

Yang XL, Lin FJ, Guo YJ, Shao ZM, Ou ZL. Gemcitabine resistance in breast cancer cells regulated by PI3K/AKT-mediated cellular proliferation exerts negative feedback via the MEK/MAPK and mTOR pathways. Onco Targets Ther. 2014;7:1033-1042.

27

Wang R, Cheng L, Xia J, Wang Z, Wu Q, Wang Z. Gemcitabine resistance is associated with epithelial-mesenchymal transition and induction of HIF-1alpha in pancreatic cancer cells. Curr Cancer Drug Targets. 2014;14:407-417.

28

Kinzler KW, Vogelstein B. The GLI gene encodes a nuclear protein which binds specific sequences in the human genome. Mol Cell Biol. 1990;10:634-642.

29

Sasaki H, Hui C, Nakafuku M, Kondoh H. A binding site for Gli proteins is essential for HNF-3beta floor plate enhancer activity in transgenics and can respond to Shh in vitro. Development. 1997;124:1313-1322.

30

Jenkins D. Hedgehog signalling: emerging evidence for non-canonical pathways. Cell Signal. 2009;21:1023-1034.

31

LoRusso PM, Rudin CM, Reddy JC, et al. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clin Cancer Res - Off J Am Assoc Cancer Res. 2011;17:2502-2511.

32

An Y, Cai B, Chen J, et al. MAP3K10 promotes the proliferation and decreases the sensitivity of pancreatic cancer cells to gemcitabine by upregulating Gli-1 and Gli-2. Cancer Lett. 2013;329:228-235.

33

Xin Y, Shen XD, Cheng L, Hong DF, Chen B. Perifosine inhibits S6K1-Gli1 signaling and enhances gemcitabine-induced anti-pancreatic cancer efficiency. Cancer Chemother Pharmacol. 2014;73:711-719.

34

Peng Z, Ji Z, Mei F, Lu M, Ou Y, Cheng X. Lithium inhibits tumorigenic potential of PDA cells through targeting hedgehog-GLI signaling pathway. PLoS One. 2013;8:e61457.

35

Xu M, Li L, Liu Z, et al. ABCB2 (TAP1) as the downstream target of SHH signaling enhances pancreatic ductal adenocarcinoma drug resistance. Cancer Lett. 2013;333:152-158.

36

Gu D, Liu H, Su GH, et al. Combining hedgehog signaling inhibition with focal irradiation on reduction of pancreatic cancer metastasis. Mol Cancer Ther. 2013;12:1038-1048.

37

Inaguma S, Kasai K, Hashimoto M, Ikeda H. GLI1 modulates EMT in pancreatic cancer–letter. Cancer Res. 2012;72:3702-3703 [author reply 3704e3705].

38

Liu S, Dontu G, Mantle ID, et al. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006;66:6063-6071.

39

Song Z, Yue W, Wei B, et al. Sonic hedgehog pathway is essential for maintenance of cancer stem-like cells in human gastric cancer. PLoS One. 2011;6:e17687.

40

Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol. 2011;8:97-106.

41

Tanaka H, Nakamura M, Kameda C, et al. The Hedgehog signaling pathway plays an essential role in maintaining the CD44+CD24-/low subpopulation and the side population of breast cancer cells. Anticancer Res. 2009;29:2147-2157.

42

Takahashi T, Kawakami K, Mishima S, et al. Cyclopamine induces eosinophilic differentiation and upregulates CD44 expression in myeloid leukemia cells. Leukemia Res. 2011;35:638-645.

43

Bar EE, Chaudhry A, Lin A, et al. Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells. 2007;25:2524-2533.

44

Li C, Heidt DG, Dalerba P, et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67:1030-1037.

45

Visbal AP, LaMarca HL, Villanueva H, et al. Altered differentiation and paracrine stimulation of mammary epithelial cell proliferation by conditionally activated Smoothened. Dev Biol. 2011;352:116-127.

46

Su W, Meng F, Huang L, Zheng M, Liu W, Sun H. Sonic hedgehog maintains survival and growth of chronic myeloid leukemia progenitor cells through beta-catenin signaling. Exp Hematol. 2012;40:418-427.

47

Balic A, Sorensen MD, Trabulo SM, et al. Chloroquine targets pancreatic cancer stem cells via inhibition of CXCR4 and hedgehog signaling. Mol Cancer Ther. 2014;13:1758-1771.

48

Han JB, Sang F, Chang JJ, et al. Arsenic trioxide inhibits viability of pancreatic cancer stem cells in culture and in a xenograft model via binding to SHH-Gli. Onco Targets Ther. 2013;6:1129-1138.

49

Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781-810.

50

Niehrs C. The complex world of WNT receptor signalling. Nat Rev Mol Cell Biol. 2012;13:767-779.

51

Veeman MT, Axelrod JD, Moon RT. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev Cell. 2003;5:367-377.

52

Janikova M, Skarda J. Differentiation pathways in carcinogenesis and in chemo- and radioresistance. Neoplasma. 2012;59:6-17.

53

Zhang HH, Zhang ZY, Che CL, Mei YF, Shi YZ. Array analysis for potential biomarker of gemcitabine identification in non-small cell lung cancer cell lines. Int J Clin Exp Pathology. 2013;6:1734-1746.

54

Griesmann H, Ripka S, Pralle M, et al. WNT5A-NFAT signaling mediates resistance to apoptosis in pancreatic cancer. Neoplasia. 2013;15:11-22.

55

Jung DB, Yun M, Kim EO, et al. The heparan sulfate mimetic PG545 interferes with Wnt/beta-catenin signaling and significantly suppresses pancreatic tumorigenesis alone and in combination with gemcitabine. Oncotarget. 2015;6:4992-5004.

56

Humbert M, Casteran N, Letard S, et al. Masitinib combined with standard gemcitabine chemotherapy: in vitro and in vivo studies in human pancreatic tumour cell lines and ectopic mouse model. PLoS One. 2010;5:e9430.

57

Niess H, Camaj P, Renner A, et al. Side population cells of pancreatic cancer show characteristics of cancer stem cells responsible for resistance and metastasis. Target Oncol. 2015;10:215-227.

58

Sanchez-Tillo E, Fanlo L, Siles L, et al. The EMT activator ZEB1 promotes tumor growth and determines differential response to chemotherapy in mantle cell lymphoma. Cell Death Differ. 2014;21:247-257.

59

Fiuza UM, Arias AM. Cell and molecular biology of Notch. J Endocrinol. 2007;194:459-474.

60

Meng RD, Shelton CC, Li YM, et al. gamma-Secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 signaling pathway in colon cancer cells resulting in enhanced chemosensitivity. Cancer Res. 2009;69:573-582.

61

Wang Z, Li Y, Ahmad A, et al. Targeting Notch signaling pathway to overcome drug resistance for cancer therapy. Biochimica Biophysica Acta. 2010;1806:258-267.

62

Wang Z, Li Y, Kong D, et al. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009;69:2400-2407.

63

Eto K, Kawakami H, Kuwatani M, et al. Human equilibrative nucleoside transporter 1 and Notch3 can predict gemcitabine effects in patients with unresectable pancreatic cancer. Br J Cancer. 2013;108:1488-1494.

64

Du X, Wang YH, Wang ZQ, et al. Down-regulation of Notch1 by small interfering RNA enhances chemosensitivity to gemcitabine in pancreatic cancer cells through activating apoptosis activity. J Zhejiang Univ Med Sci. 2014;43:313-318.

65

Gungor C, Zander H, Effenberger KE, et al. Notch signaling activated by replication stress-induced expression of midkine drives epithelial-mesenchymal transition and chemoresistance in pancreatic cancer. Cancer Res. 2011;71:5009-5019.

66

Yao J, Qian C. Inhibition of Notch3 enhances sensitivity to gemcitabine in pancreatic cancer through an inactivation of PI3K/Akt-dependent pathway. Med Oncol. 2010;27:1017-1022.

67

Cook N, Frese KK, Bapiro TE, et al. Gamma secretase inhibition promotes hypoxic necrosis in mouse pancreatic ductal adenocarcinoma. J Exp Med. 2012;209:437-444.

68

Du X, Zhao YP, Zhang TP, et al. Alteration of the intrinsic apoptosis pathway is involved in Notch-induced chemoresistance to gemcitabine in pancreatic cancer. Archives Med Res. 2014;45:15-20.

69

Lomberk G, Urrutia R. Primers on molecular pathways–notch. Pancreatol – Off J Int Assoc Pancreatol. 2008;8:103-104.

70

Mizuma M, Rasheed ZA, Yabuuchi S, et al. The gamma secretase inhibitor MRK-003 attenuates pancreatic cancer growth in preclinical models. Mol Cancer Ther. 2012;11:1999-2009.

71

Lee JY, Song SY, Park JY. Notch pathway activation is associated with pancreatic cancer treatment failure. Pancreatology. 2014;14:48-53.

72

Cao F, Li J, Sun H, Liu S, Cui Y, Li F. HES 1 is essential for chemoresistance induced by stellate cells and is associated with poor prognosis in pancreatic cancer. Oncol Rep. 2015;33:1883-1889.

73

Yen WC, Fischer MM, Hynes M, et al. Anti-DLL4 has broad spectrum activity in pancreatic cancer dependent on targeting DLL4-Notch signaling in both tumor and vasculature cells. Clin Cancer Res. 2012;18:5374-5386.

74

Kallifatidis G, Labsch S, Rausch V, et al. Sulforaphane increases drug-mediated cytotoxicity toward cancer stem-like cells of pancreas and prostate. Mol Ther. 2011;19:188-195.

75

Rabadan MA, Cayuso J, Le Dreau G, et al. Jagged2 controls the generation of motor neuron and oligodendrocyte progenitors in the ventral spinal cord. Cell Death Differ. 2012;19:209-219.

76

Chen X, Stoeck A, Lee SJ, Shih Ie M, Wang MM, Wang TL. Jagged1 expression regulated by Notch3 and Wnt/beta-catenin signaling pathways in ovarian cancer. Oncotarget. 2010;1:210-218.

77

Schreck KC, Taylor P, Marchionni L, et al. The Notch target Hes1 directly modulates Gli1 expression and Hedgehog signaling: a potential mechanism of therapeutic resistance. Clin Cancer Res. 2010;16:6060-6070.

78

He J, Sheng T, Stelter AA, et al. Suppressing Wnt signaling by the hedgehog pathway through sFRP-1. J Biol Chem. 2006;281:35598-35602.

79

Ramaswamy B, Lu Y, Teng KY, et al. Hedgehog signaling is a novel therapeutic target in tamoxifen-resistant breast cancer aberrantly activated by PI3K/AKT pathway. Cancer Res. 2012;72:5048-5059.

Genes & Diseases
Pages 299-306
Cite this article:
Jia Y, Xie J. Promising molecular mechanisms responsible for gemcitabine resistance in cancer. Genes & Diseases, 2015, 2(4): 299-306. https://doi.org/10.1016/j.gendis.2015.07.003

275

Views

1

Downloads

114

Crossref

N/A

Web of Science

115

Scopus

0

CSCD

Altmetrics

Received: 14 June 2015
Accepted: 20 July 2015
Published: 30 July 2015
© 2015, Chongqing Medical University

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return