Drug Res (Stuttg) 2016; 66(11): 571-579
DOI: 10.1055/s-0042-111821
Review
© Georg Thieme Verlag KG Stuttgart · New York

Downregulation of Notch Signaling Pathway as an Effective Chemosensitizer for Cancer Treatment

M. Majidinia
1   Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
2   Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
,
E. Alizadeh
3   Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
,
B. Yousefi
1   Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
,
M. Akbarzadeh
4   Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran
,
N. Zarghami
5   Hematology and Oncology Research Center, Tabriz University of Medical Science, Tabriz, Iran
› Author Affiliations
Further Information

Publication History

received 04 June 2016

accepted 01 July 2016

Publication Date:
04 October 2016 (online)

Abstract

Despite remarkable progress in cancer treatment, development of drug resistance is still a big burden to eliminate all tumor cells and a mean cause for tumor recurrence. Recent studies have been revealed the contribution of many signaling pathways in acquisition of resistance to chemotherapy. Because of its potential in maintaining the balance between cell proliferation and apoptosis, Notch signaling pathway has mean relevance to various aspects of cancer biology, from cancer stem cells to tumor immunity to multidrug resistance. Therefore, Notch signaling pathway is an attractive target for cancer therapy because targeting Notch signaling could overcome multi drug resistance (MDR). This article will provide a brief overview of the published evidences in support of Notch targeting in reverting multidrug resistance as a safer and novel approach for the improvement of tumor treatment.

 
  • References

  • 1 Previs RA, Coleman RL, Harris AL et al. Molecular pathways: translational and therapeutic implications of the Notch signaling pathway in cancer. Clinical Cancer Research 2015; 21: 955-961
  • 2 Yen W-C, Fischer MM, Axelrod F et al. Targeting Notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clinical Cancer Research 2015; 21: 2084-2095
  • 3 Takebe N, Miele L, Harris PJ et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nature reviews Clinical oncology 2015; 12: 445-464
  • 4 Kangsamaksin T, Murtomaki A, Kofler NM et al. NOTCH decoys that selectively block DLL/NOTCH or JAG/NOTCH disrupt angiogenesis by unique mechanisms to inhibit tumor growth. Cancer discovery 2015; 5: 182-197
  • 5 Yuan X, Wu H, Xu H et al. Notch signaling: An emerging therapeutic target for cancer treatment. Cancer letters 2015; 369: 20-27
  • 6 D’Angelo RC, Ouzounova M, Davis A et al. Notch reporter activity in breast cancer cell lines identifies a subset of cells with stem cell activity. Molecular cancer therapeutics 2015; 14: 779-787
  • 7 Ramos P, Bentires-Alj M. Mechanism-based cancer therapy: resistance to therapy, therapy for resistance. Oncogene 2015; 34: 3617-3626
  • 8 Kimball J. The notch signaling pathway. Kimball’s Biology Pages Retrieved on April 2014; 26
  • 9 Lobry C, Oh P, Mansour MR et al. Notch signaling: switching an oncogene to a tumor suppressor. Blood 2014; 123: 2451-2459
  • 10 Geisler F, Strazzabosco M. Emerging roles of Notch signaling in liver disease. Hepatology 2015; 61: 382-392
  • 11 Groeneweg JW, Foster R, Growdon WB et al. Notch signaling in serous ovarian cancer. Journal of ovarian research 2014; 7: 1
  • 12 Yousefi B, Rahmati M, Ahmadi Y. The roles of p53R2 in cancer progression based on the new function of mutant p53 and cytoplasmic p21. Life Sciences 2014; 99: 14-17
  • 13 Ayaz F, Osborne BA. Non-canonical notch signaling in cancer and immunity. Frontiers in oncology 2014; 4: 435
  • 14 Miele L, Golde T, Osborne B. Notch signaling in cancer. Current molecular medicine 2006; 6: 905-918
  • 15 Rizzo P, Osipo C, Foreman K et al. Rational targeting of Notch signaling in cancer. Oncogene 2008; 27: 5124-5131
  • 16 Yousefi B, Zarghami N, Samadi N et al. Peroxisome Proliferator-activated Receptors and their ligands in cancer drug resistance: opportunity or challenge. Anticancer Agents Med Chem 2016; 16
  • 17 McCubrey JA, Abrams SL, Fitzgerald TL et al. Roles of signaling pathways in drug resistance, cancer initiating cells and cancer progression and metastasis. Advances in biological regulation 2015; 57: 75-101
  • 18 Lobry C, Oh P, Aifantis I. Oncogenic and tumor suppressor functions of Notch in cancer: it’s NOTCH what you think. The Journal of experimental medicine 2011; 208: 1931-1935
  • 19 Ranganathan P, Weaver KL, Capobianco AJ. Notch signalling in solid tumours: a little bit of everything but not all the time. Nature Reviews Cancer 2011; 11: 338-351
  • 20 Aithal MG, Rajeswari N. Role of Notch signalling pathway in cancer and its association with DNA methylation. Journal of genetics 2013; 92: 667-675
  • 21 Radtke F, Raj K. The role of Notch in tumorigenesis: oncogene or tumour suppressor?. Nature Reviews Cancer 2003; 3: 756-767
  • 22 Fabbri G, Rasi S, Rossi D et al. Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation. The Journal of experimental medicine 2011; 208: 1389-1401
  • 23 Yousefi B, Samadi N, Baradaran B et al. Peroxisome proliferator-activated receptor ligands and their role in chronic myeloid leukemia: Therapeutic strategies. Chem Biol Drug Des 2016; 88: 17-25
  • 24 Nguyen B-C, Lefort K, Mandinova A et al. Cross-regulation between Notch and p63 in keratinocyte commitment to differentiation. Genes & development 2006; 20: 1028-1042
  • 25 Guo S, Liu M, Gonzalez-Perez RR. Role of Notch and its oncogenic signaling crosstalk in breast cancer. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer 2011; 1815: 197-213
  • 26 Takebe N, Nguyen D, Yang SX. Targeting notch signaling pathway in cancer: clinical development advances and challenges. Pharmacology & therapeutics 2014; 141: 140-149
  • 27 Brechbiel J, Miller-Moslin K, Adjei AA. Crosstalk between hedgehog and other signaling pathways as a basis for combination therapies in cancer. Cancer treatment reviews 2014; 40: 750-759
  • 28 Androutsellis-Theotokis A, Leker RR, Soldner F et al. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature 2006; 442: 823-826
  • 29 Zhao X, Malhotra GK, Lele SM et al. Telomerase-immortalized human mammary stem/progenitor cells with ability to self-renew and differentiate. Proceedings of the National Academy of Sciences 2010; 107: 14146-14151
  • 30 Ayyanan A, Civenni G, Ciarloni L et al. Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells by a Notch-dependent mechanism. Proceedings of the National Academy of Sciences of the United States of America 2006; 103: 3799-3804
  • 31 Wang Z, Ahmad A, Li Y et al. Emerging roles of PDGF-D signaling pathway in tumor development and progression. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer 2010; 1806: 122-130
  • 32 Niimi H, Pardali K, Vanlandewijck M et al. Notch signaling is necessary for epithelial growth arrest by TGF-β. The Journal of cell biology 2007; 176: 695-707
  • 33 Cojoc M, Mäbert K, Muders MH, Dubrovska A. (eds.). A role for cancer stem cells in therapy resistance: cellular and molecular mechanisms. Seminars in cancer biology. 2015. Elsevier;
  • 34 Justilien V, Fields AP. Molecular pathways: novel approaches for improved therapeutic targeting of Hedgehog signaling in cancer stem cells. Clinical Cancer Research 2015; 21: 505-513
  • 35 Ajani JA, Song S, Hochster HS, Steinberg IB. (eds.). Cancer stem cells: the promise and the potential. Seminars in oncology. 2015. Elsevier;
  • 36 Fan X, Matsui W, Khaki L et al. Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumors. Cancer research 2006; 66: 7445-7452
  • 37 Farnie G, Clarke RB. Mammary stem cells and breast cancer – role of Notch signalling. Stem cell reviews 2007; 3: 169-175
  • 38 Pannuti A, Foreman K, Rizzo P et al. Targeting Notch to target cancer stem cells. Clinical Cancer Research 2010; 16: 3141-3152
  • 39 Eyler CE, Rich JN. Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. Journal of Clinical Oncology 2008; 26: 2839-2845
  • 40 Conde J, Jesús M, Baptista PV. Nanomaterials for reversion of multidrug resistance in cancer: a new hope for an old idea?. Multidrug Resistance in Cancer: Pharmacological Strategies from Basic Research to Clinical Issues 2015; 74
  • 41 Cort A, Ozben T. Natural product modulators to overcome multidrug resistance in cancer. Nutrition and cancer 2015; 67: 411-423
  • 42 Nigam SK. What do drug transporters really do?. Nature Reviews Drug Discovery 2015; 14: 29-44
  • 43 Di C, Zhao Y. Multiple drug resistance due to resistance to stem cells and stem cell treatment progress in cancer (Review). Experimental and therapeutic medicine 2015; 9: 289-293
  • 44 Ferreira RJ, Ferreira MJU, dos Santos DJ. Reversing cancer multidrug resistance: insights into the efflux by ABC transports from in silico studies. Wiley Interdisciplinary Reviews: Computational Molecular Science 2015; 5: 27-55
  • 45 Zhao L, Liu W, Xiao J et al. The role of exosomes and “exosomal shuttle microRNA” in tumorigenesis and drug resistance. Cancer letters 2015; 356: 339-346
  • 46 Kathawala RJ, Gupta P, Ashby CR et al. The modulation of ABC transporter-mediated multidrug resistance in cancer: A review of the past decade. Drug Resistance Updates 2015; 18: 1-17
  • 47 Wilkens S. Structure and mechanism of ABC transporters. F1000prime reports 2015; 7: 14
  • 48 Kim B, Stephen SL, Hanby AM et al. Chemotherapy induces Notch1-dependent MRP1 up-regulation, inhibition of which sensitizes breast cancer cells to chemotherapy. BMC cancer 2015; 15: 1
  • 49 Cho S, Lu M, He X et al. Notch1 regulates the expression of the multidrug resistance gene ABCC1/MRP1 in cultured cancer cells. Proceedings of the National Academy of Sciences 2011; 108: 20778-20783
  • 50 Zoni E, van der Pluijm G, Gray PC et al. Epithelial plasticity in cancer: unmasking a microRNA network for TGF-β-, Notch-, and Wnt-mediated EMT. Journal of oncology 2015; 2015
  • 51 Satoh K, Hamada S, Shimosegawa T. Involvement of epithelial to mesenchymal transition in the development of pancreatic ductal adenocarcinoma. Journal of gastroenterology 2015; 50: 140-146
  • 52 Yang Z, Guo L, Liu D et al. Acquisition of resistance to trastuzumab in gastric cancer cells is associated with activation of IL-6/STAT3/Jagged-1/Notch positive feedback loop. Oncotarget 2015; 6: 5072-5087
  • 53 Capaccione KM, Pine SR. The Notch signaling pathway as a mediator of tumor survival. Carcinogenesis 2013; bgt127
  • 54 Olsauskas-Kuprys R, Zlobin A, Osipo C. Gamma secretase inhibitors of Notch signaling. Onco Targets Ther 2013; 6: 943-955
  • 55 Dittmer J, Leyh B. (eds.). The impact of tumor stroma on drug response in breast cancer. Seminars in cancer biology. 2015. Elsevier;
  • 56 Florea A-M, Büsselberg D. Breast cancer and possible mechanisms of therapy resistance. Journal of Local and Global Health Science 2013;
  • 57 Li S-Y, Sun R, Wang H-X et al. Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells. Journal of Controlled Release 2015; 205: 7-14
  • 58 Acar A, Simões BM, Clarke RB et al. A role for Notch signalling in breast cancer and endocrine resistance. Stem Cells International 2016; 2016
  • 59 Zang S, Chen F, Dai J et al. RNAi-mediated knockdown of Notch-1 leads to cell growth inhibition and enhanced chemosensitivity in human breast cancer. Oncology reports 2010; 23: 893-899
  • 60 Yun J, Pannuti A, Espinoza I et al. Crosstalk between PKCα and Notch-4 in endocrine-resistant breast cancer cells. Oncogenesis 2013; 2: e60
  • 61 Magnifico A, Albano L, Campaner S et al. Tumor-initiating cells of HER2-positive carcinoma cell lines express the highest oncoprotein levels and are sensitive to trastuzumab. Clinical Cancer Research 2009; 15: 2010-2021
  • 62 Osipo C, Patel P, Rizzo P et al. ErbB-2 inhibition activates Notch-1 and sensitizes breast cancer cells to a γ-secretase inhibitor. Oncogene 2008; 27: 5019-5032
  • 63 Rizzo P, Miao H, D’Souza G et al. Cross-talk between notch and the estrogen receptor in breast cancer suggests novel therapeutic approaches. Cancer research 2008; 68: 5226-5235
  • 64 Schott AF, Landis MD, Dontu G et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clinical Cancer Research 2013; 19: 1512-1524
  • 65 Bhola NE, Jansen VM, Koch JP et al. Treatment of triple-negative breast cancer with TORC1/2 inhibitors sustains a drug-resistant and Notch-dependent cancer stem cell population. Cancer research 2016; 76: 440-452
  • 66 Waddell N, Pajic M, Patch A-M et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015; 518: 495-501
  • 67 Long J, Zhang Y, Yu X et al. Overcoming drug resistance in pancreatic cancer. Expert opinion on therapeutic targets 2011; 15: 817-828
  • 68 Wang Z, Ahmad A, Li Y et al. Targeting notch to eradicate pancreatic cancer stem cells for cancer therapy. Anticancer research 2011; 31: 1105-1113
  • 69 Avila JL, Kissil JL. Notch signaling in pancreatic cancer: oncogene or tumor suppressor?. Trends in molecular medicine 2013; 19: 320-327
  • 70 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 research 2009; 69: 2400-2407
  • 71 Güngör 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 research 2011; 71: 5009-5019
  • 72 Yao J, Qian C. Inhibition of Notch3 enhances sensitivity to gemcitabine in pancreatic cancer through an inactivation of PI3K/Akt-dependent pathway. Medical Oncology 2010; 27: 1017-1022
  • 73 Kehoe S, Hook J, Nankivell M et al. Primary chemotherapy versus primary surgery for newly diagnosed advanced ovarian cancer (CHORUS): an open-label, randomised, controlled, non-inferiority trial. The Lancet 2015; 386: 249-257
  • 74 Zhang J, Yin X, Xu C et al. The histone deacetylase SIRT6 inhibits ovarian cancer cell proliferation via down-regulation of Notch 3 expression. Eur Rev Med Pharmacol Sci 2015; 19: 818-824
  • 75 Alniaimi AN, Demorest-Hayes K, Alexander VM et al. Increased Notch1 expression is associated with poor overall survival in patients with ovarian cancer. International Journal of Gynecological Cancer 2015; 25: 208-213
  • 76 McAuliffe SM, Morgan SL, Wyant GA et al. Targeting Notch, a key pathway for ovarian cancer stem cells, sensitizes tumors to platinum therapy. Proceedings of the National Academy of Sciences 2012; 109: E2939-E2948
  • 77 Wang M, Ma X, Wang J et al. Pretreatment with the γ-secretase inhibitor DAPT sensitizes drug-resistant ovarian cancer cells to cisplatin by downregulation of Notch signaling. International journal of oncology 2014; 44: 1401-1409
  • 78 Park JT, Chen X, Trope CG et al. Notch3 overexpression is related to the recurrence of ovarian cancer and confers resistance to carboplatin. The American journal of pathology 2010; 177: 1087-1094
  • 79 Mine T, Matsueda S, Gao H et al. Created Gli-1 duplex short-RNA (i-Gli-RNA) eliminates CD44Hi progenitors of taxol-resistant ovarian cancer cells. Oncology reports 2010; 23: 1537-1543
  • 80 Karimian A, Ahmadi Y, Yousefi B. Multiple functions of p21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair (Amst) 2016; 42: 63-71
  • 81 Fender AW, Nutter JM, Fitzgerald TL et al. Notch-1 promotes stemness and epithelial to mesenchymal transition in colorectal cancer. Journal of cellular biochemistry 2015; 116: 2517-2527
  • 82 Meng RD, Shelton CC, Li Y-M et al. γ-Secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 signaling pathway in colon cancer cells resulting in enhanced chemosensitivity. Cancer research 2009; 69: 573-582
  • 83 Akiyoshi T, Nakamura M, Yanai K et al. γ-Secretase inhibitors enhance taxane-induced mitotic arrest and apoptosis in colon cancer cells. Gastroenterology 2008; 134: 131-144
  • 84 Yuan X, Wu H, Xu H et al. Meta-analysis reveals the correlation of Notch signaling with non-small cell lung cancer progression and prognosis. Scientific reports 2015; 5 Article number: 10338 (2015) doi: 10.1038/srep10338
  • 85 Liu Y-P, Yang C-J, Huang M-S et al. Cisplatin selects for multidrug-resistant CD133+ cells in lung adenocarcinoma by activating Notch signaling. Cancer research 2013; 73: 406-416
  • 86 Xie M, Zhang L, He CS et al. Activation of notch-1 enhances epithelial-mesenchymal transition in gefitinib-acquired resistant lung cancer cells. Journal of cellular biochemistry 2012; 113: 1501-1513
  • 87 Xie M, He C-S, Wei S-H et al. Notch-1 contributes to epidermal growth factor receptor tyrosine kinase inhibitor acquired resistance in non-small cell lung cancer in vitro and in vivo. European Journal of Cancer 2013; 49: 3559-3572
  • 88 Xie M, He J, He C et al. γ secretase inhibitor BMS-708163 reverses resistance to EGFR inhibitor via the PI3K/Akt pathway in lung cancer. Journal of cellular biochemistry 2015; 116: 1019-1027
  • 89 James ND, Spears MR, Clarke NW et al. Survival with newly diagnosed metastatic prostate cancer in the “Docetaxel Era”: data from 917 patients in the control arm of the STAMPEDE Trial (MRC PR08, CRUK/06/019). European urology 2015; 67: 1028-1038
  • 90 James N, Mason M. Docetaxel and/or zoledronic acid for hormone-naïve prostate cancer: First survival results from STAMPEDE. J Clin Oncol 2015; 33 (supplabstr 5001)
  • 91 Ryan CJ, Smith MR, Fizazi K et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. The Lancet Oncology 2015; 16: 152-160
  • 92 Carvalho FL, Simons BW, Eberhart CG et al. Notch signaling in prostate cancer: a moving target. The Prostate 2014; 74: 933-945
  • 93 Ye Q-F, Zhang Y-C, Peng X-Q et al. Silencing Notch-1 induces apoptosis and increases the chemosensitivity of prostate cancer cells to docetaxel through Bcl-2 and Bax. Oncology letters 2012; 3: 879-884
  • 94 Domingo-Domenech J, Vidal SJ, Rodriguez-Bravo V et al. Suppression of acquired docetaxel resistance in prostate cancer through depletion of notch-and hedgehog-dependent tumor-initiating cells. Cancer cell 2012; 22: 373-388
  • 95 López-Guerra M, Xargay-Torrent S, Rosich L et al. The γ-secretase inhibitor PF-03084014 combined with fludarabine antagonizes migration, invasion and angiogenesis in NOTCH1-mutated CLL cells. Leukemia 2015; 29: 96-106
  • 96 Liu MX, Siu MK, Liu SS et al. Epigenetic silencing of microRNA-199b-5p is associated with acquired chemoresistance via activation of JAG1-Notch1 signaling in ovarian cancer. Oncotarget 2014; 5: 944
  • 97 Schmitz S, Ang KK, Vermorken J et al. Targeted therapies for squamous cell carcinoma of the head and neck: current knowledge and future directions. Cancer treatment reviews 2014; 40: 390-404
  • 98 Yousefi B, Samadi N, Ahmadi Y. Akt and p53R2, partners that dictate the progression and invasiveness of cancer. Journal of hepatology 2009; 50: 969-979
  • 99 Xu D, Hu J, De Bruyne E et al. Dll1/Notch activation contributes to bortezomib resistance by upregulating CYP1A1 in multiple myeloma. Biochemical and biophysical research communications 2012; 428: 518-524