Generic placeholder image

Current Signal Transduction Therapy

Editor-in-Chief

ISSN (Print): 1574-3624
ISSN (Online): 2212-389X

Review Article

Kinase Phosphorylation-based Mechanisms of PARP Inhibitor Resistance During Synthetic Lethal Oncotherapy

Author(s): Eriko Osaki and Shinya Mizuno*

Volume 15, Issue 1, 2020

Page: [12 - 23] Pages: 12

DOI: 10.2174/1574362413666180724134204

Abstract

Background: Poly-(ADP-Ribose) Polymerase (PARP) plays a central role in recovery from single-strand DNA (ssDNA) damage via base excision repair. When PARP activity is inhibited by a NAD+ mimetic analog, ssDNA is converted into a Double-Strand Break (DSB) during the S-phase in a cell cycle. However, the DSB site is repaired in a process of Homologous Recombination (HR) that is derived by genes such as BRCA1/2, PALB2, and RAD51. Under conditions of HR dysfunction, including mutations of BRCA1/2 (called BRCAness), PARP inhibitor (PARPi) induces “synthetic lethality” in BRCAness-specific cancer cells. Indeed, clinical trials using forms of PARPi that include olaparib, veliparib and rucaparib, have revealed that PARP inhibition produces a dramatic effect that actually arrests cancer progression. Its clinical efficiency is limited, however, due to the acquisition of PARPi resistance during long-term use of this inhibitor. Thus, it is important to elucidate the mechanisms of PARPi resistance.

Methods: We searched the scientific literature published in PubMed, with a special focus on kinase phosphorylation that is involved in acquiring PARPi resistance. We also summarized the possible molecular events for recovering HR system, a key event for acquiring PARPi resistance.

Results: CDK1 is a critical kinase for 5’-3’ DNA end resection, which is important for generating ssDNA for recruiting HR-priming factors. CDK12 is necessary for the transcription of HR-driver genes, such as BRCA1, BRCA2, RAD51 and ATR via the phosphorylation of RNA Pol-II. PLK-1 participates in driving HR via the phosphorylation of RAD51. The PI3K-AKT-mTOR signaling cascade is involved in BRCA1 induction via an ETS1 transcriptional pathway. Even under ATMdeficient conditions, the ATR-CHK1 axis compensates for loss in the DNA damage response, which results in HR recovery. The HGF receptor Met tyrosine kinase is responsible for promoting DNA repair by activating the PARP catalytic domain.

Conclusion: These kinase-based signaling pathways are biologically important for understanding the compensatory system of HR, whereas inactivation of these kinases has shown promise for the release of PARPi resistance. Several lines of preclinical studies have demonstrated the potential use of kinase inhibitors to enhance PARPi sensitivity. We emphasize the clinical importance of chemical inhibitors as adjuvant drugs to block critical kinase activities and prevent the possible PARPi resistance.

Keywords: ATR, checkpoint kinases, DNA damage response, homologous recombination, genome instability, synthetic lethal therapy.

Graphical Abstract
[1]
Rajawat J, Shukla N, Mishra DP. Therapeutic targeting of Poly(ADP-Ribose) Polymerase-1 (PARP1) in cancer: Current developments, therapeutic strategies, and future opportunities. Med Res Rev 2017; 37(6): 1461-91.
[http://dx.doi.org/10.1002/med.21442] [PMID: 28510338]
[2]
Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer 2016; 16(2): 110-20.
[http://dx.doi.org/10.1038/nrc.2015.21] [PMID: 26775620]
[3]
Nickoloff JA, Jones D, Lee SH, Williamson EA, Hromas R. Drugging the cancers addicted to DNA repair. J Natl Cancer Inst 2017; 109(11)djx059
[http://dx.doi.org/10.1093/jnci/djx059] [PMID: 28521333]
[4]
Ledermann J, Harter P, Gourley C, et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med 2012; 366(15): 1382-92.
[http://dx.doi.org/10.1056/NEJMoa1105535] [PMID: 22452356]
[5]
Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med 2017; 377(6): 523-33.
[http://dx.doi.org/10.1056/NEJMoa1706450] [PMID: 28578601]
[6]
Ferrara R, Simionato F, Ciccarese C, et al. The development of PARP as a successful target for cancer therapy. Expert Rev Anticancer Ther 2018; 18(2): 161-75.
[http://dx.doi.org/10.1080/14737140.2018.1419870] [PMID: 29260919]
[7]
Incorvaia L, Passiglia F, Rizzo S, et al. “Back to a false normality”: new intriguing mechanisms of resistance to PARP inhibitors. Oncotarget 2017; 8(14): 23891-904.
[http://dx.doi.org/10.18632/oncotarget.14409] [PMID: 28055979]
[8]
Lawlor D, Martin P, Busschots S, et al. PARP inhibitors as P-glyoprotein substrates. J Pharm Sci 2014; 103(6): 1913-20.
[http://dx.doi.org/10.1002/jps.23952] [PMID: 24700236]
[9]
Ira G, Pellicioli A, Balijja A, et al. DNA end resection, homologous recombination and DNA damage checkpoint activation require CDK1. Nature 2004; 431(7011): 1011-7.
[http://dx.doi.org/10.1038/nature02964] [PMID: 15496928]
[10]
Huertas P, Cortés-Ledesma F, Sartori AA, Aguilera A, Jackson SP. CDK targets Sae2 to control DNA-end resection and homologous recombination. Nature 2008; 455(7213): 689-92.
[http://dx.doi.org/10.1038/nature07215] [PMID: 18716619]
[11]
Chen X, Niu H, Chung WH, et al. Cell cycle regulation of DNA double-strand break end resection by Cdk1-dependent Dna2 phosphorylation. Nat Struct Mol Biol 2011; 18(9): 1015-9.
[http://dx.doi.org/10.1038/nsmb.2105] [PMID: 21841787]
[12]
Huertas P, Jackson SP. Human CtIP mediates cell cycle control of DNA end resection and double strand break repair. J Biol Chem 2009; 284(14): 9558-65.
[http://dx.doi.org/10.1074/jbc.M808906200] [PMID: 19202191]
[13]
Zhang Y, Shim EY, Davis M, Lee SE. Regulation of repair choice: Cdk1 suppresses recruitment of end joining factors at DNA breaks. DNA Repair (Amst) 2009; 8(10): 1235-41.
[http://dx.doi.org/10.1016/j.dnarep.2009.07.007] [PMID: 19699692]
[14]
Johnson N, Cai D, Kennedy RD, et al. Cdk1 participates in BRCA1-dependent S phase checkpoint control in response to DNA damage. Mol Cell 2009; 35(3): 327-39.
[http://dx.doi.org/10.1016/j.molcel.2009.06.036] [PMID: 19683496]
[15]
Johnson N, Li YC, Walton ZE, et al. Compromised CDK1 activity sensitizes BRCA-proficient cancers to PARP inhibition. Nat Med 2011; 17(7): 875-82.
[http://dx.doi.org/10.1038/nm.2377] [PMID: 21706030]
[16]
Chen X, Niu H, Yu Y, et al. Enrichment of Cdk1-cyclins at DNA double-strand breaks stimulates Fun30 phosphorylation and DNA end resection. Nucleic Acids Res 2016; 44(6): 2742-53.
[http://dx.doi.org/10.1093/nar/gkv1544] [PMID: 26801641]
[17]
Palermo V, Rinalducci S, Sanchez M, et al. CDK1 phosphorylates WRN at collapsed replication forks. Nat Commun 2016; 7(9): 12880.
[http://dx.doi.org/10.1038/ncomms12880] [PMID: 27634057]
[18]
Lu H, Shamanna RA, de Freitas JK, et al. Cell cycle-dependent phosphorylation regulates RECQL4 pathway choice and ubiquitination in DNA double-strand break repair. Nat Commun 2017; 8(1): 2039.
[http://dx.doi.org/10.1038/s41467-017-02146-3] [PMID: 29229926]
[19]
Raghavan P, Tumati V, Yu L, et al. AZD5438, an inhibitor of Cdk1, 2, and 9, enhances the radiosensitivity of non-small cell lung carcinoma cells. Int J Radiat Oncol Biol Phys 2012; 84(4): e507-14.
[http://dx.doi.org/10.1016/j.ijrobp.2012.05.035] [PMID: 22795803]
[20]
Xia Q, Cai Y, Peng R, Wu G, Shi Y, Jiang W. The CDK1 inhibitor RO3306 improves the response of BRCA-proficient breast cancer cells to PARP inhibition. Int J Oncol 2014; 44(3): 735-44.
[http://dx.doi.org/10.3892/ijo.2013.2240] [PMID: 24378347]
[21]
Bartkowiak B, Liu P, Phatnani HP, et al. CDK12 is a transcription elongation-associated CTD kinase, the metazoan ortholog of yeast Ctk1. Genes Dev 2010; 24(20): 2303-16.
[http://dx.doi.org/10.1101/gad.1968210] [PMID: 20952539]
[22]
Blazek D, Kohoutek J, Bartholomeeusen K, et al. The Cyclin K/Cdk12 complex maintains genomic stability via regulation of expression of DNA damage response genes. Genes Dev 2011; 25(20): 2158-72.
[http://dx.doi.org/10.1101/gad.16962311] [PMID: 22012619]
[23]
Liang K, Gao X, Gilmore JM, et al. Characterization of human cyclin-dependent kinase 12 (CDK12) and CDK13 complexes in C-terminal domain phosphorylation, gene transcription, and RNA processing. Mol Cell Biol 2015; 35(6): 928-38.
[http://dx.doi.org/10.1128/MCB.01426-14] [PMID: 25561469]
[24]
Bajrami I, Frankum JR, Konde A, et al. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res 201 74(1): 287-97.
[25]
Joshi PM, Sutor SL, Huntoon CJ, Karnitz LM. Ovarian cancer-associated mutations disable catalytic activity of CDK12, a kinase that promotes homologous recombination repair and resistance to cisplatin and poly(ADP-ribose) polymerase inhibitors. J Biol Chem 2014; 289(13): 9247-53.
[http://dx.doi.org/10.1074/jbc.M114.551143] [PMID: 24554720]
[26]
Ekumi KM, Paculova H, Lenasi T, et al. Ovarian carcinoma CDK12 mutations misregulate expression of DNA repair genes via deficient formation and function of the Cdk12/CycK complex. Nucleic Acids Res 2015; 43(5): 2575-89.
[http://dx.doi.org/10.1093/nar/gkv101] [PMID: 25712099]
[27]
Johnson SF, Cruz C, Greifenberg AK, et al. CDK12 inhibition reverses de novo and acquired PARP inhibitor resistance in BRCA wild-type and mutated models of triple-negative breast cancer. Cell Rep 2016; 17(9): 2367-81.
[http://dx.doi.org/10.1016/j.celrep.2016.10.077] [PMID: 27880910]
[28]
Zhang T, Kwiatkowski N, Olson CM, et al. Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat Chem Biol 2016; 12(10): 876-84.
[http://dx.doi.org/10.1038/nchembio.2166] [PMID: 27571479]
[29]
Iniguez AB, Stolte B, Wang EJ, et al. EWS/FLI confers tumor cell synthetic lethality to CDK12 inhibition in Ewing sarcoma. Cancer Cell 2018; 33(2): 202-216.e6.
[http://dx.doi.org/10.1016/j.ccell.2017.12.009] [PMID: 29358035]
[30]
Johannes JW, Denz CR, Su N, et al. Structure-based design of selective noncovalent CDK12 Inhibitors. ChemMedChem 2018; 13(3): 231-5.
[http://dx.doi.org/10.1002/cmdc.201700695] [PMID: 29266803]
[31]
Gutteridge RE, Ndiaye MA, Liu X, Ahmad N. Plk1 Inhibitors in cancer therapy: From laboratory to clinics. Mol Cancer Ther 2016; 15(7): 1427-35.
[http://dx.doi.org/10.1158/1535-7163.MCT-15-0897] [PMID: 27330107]
[32]
Kumar S, Sharma G, Chakraborty C, Sharma AR, Kim J. Regulatory functional territory of PLK-1 and their substrates beyond mitosis. Oncotarget 2017; 8(23): 37942-62.
[http://dx.doi.org/10.18632/oncotarget.16290] [PMID: 28415805]
[33]
Lee M, Kim IS, Park KC, Kim JS, Baek SH, Kim KI. Mitosis-specific phosphorylation of Mis18α by Aurora B kinase enhances kinetochore recruitment of polo-like kinase 1. Oncotarget 2017; 9(2): 1563-76.
[PMID: 29416714]
[34]
Saatci Ö, Borgoni S, Akbulut Ö, et al. Targeting PLK1 overcomes T-DM1 resistance via CDK1-dependent phosphorylation and inactivation of Bcl-2/xL in HER2-positive breast cancer. Oncogene 2018; 37(17): 2251-69.
[http://dx.doi.org/10.1038/s41388-017-0108-9] [PMID: 29391599]
[35]
Dasmahapatra G, Patel H, Nguyen T, Attkisson E, Grant S. PLK1 inhibitors synergistically potentiate HDAC inhibitor lethality in imatinib mesylate-sensitive or -resistant BCR/ABL+ leukemia cells in vitro and in vivo. Clin Cancer Res 2013; 19(2): 404-14.
[http://dx.doi.org/10.1158/1078-0432.CCR-12-2799] [PMID: 23204129]
[36]
Moudry P, Watanabe K, Wolanin KM, et al. TOPBP1 regulates RAD51 phosphorylation and chromatin loading and determines PARP inhibitor sensitivity. J Cell Biol 2016; 212(3): 281-8.
[http://dx.doi.org/10.1083/jcb.201507042] [PMID: 26811421]
[37]
Sourisseau T, Maniotis D, McCarthy A, et al. Aurora-A expressing tumour cells are deficient for homology-directed DNA double strand-break repair and sensitive to PARP inhibition. EMBO Mol Med 2010; 2(4): 130-42.
[http://dx.doi.org/10.1002/emmm.201000068] [PMID: 20373286]
[38]
Li J, Wang R, Kong Y, et al. Targeting Plk1 to enhance efficacy of olaparib in castration-resistant prostate cancer. Mol Cancer Ther 2017; 16(3): 469-79.
[http://dx.doi.org/10.1158/1535-7163.MCT-16-0361] [PMID: 28069876]
[39]
Ellis PM, Leighl NB, Hirsh V, et al. A Randomized, Open-label phase II trial of volasertib as monotherapy and in combination with standard-dose pemetrexed compared with pemetrexed monotherapy in second-line treatment for non-small-cell lung cancer. Clin Lung Cancer 2015; 16(6): 457-65.
[http://dx.doi.org/10.1016/j.cllc.2015.05.010] [PMID: 26100229]
[40]
Van den Bossche J, Lardon F, Deschoolmeester V, et al. Spotlight on volasertib: preclinical and clinical evaluation of a promising Plk1 inhibitor. Med Res Rev 2016; 36(4): 749-86.
[http://dx.doi.org/10.1002/med.21392] [PMID: 27140825]
[41]
Park JE, Hymel D, Burke TR Jr, Lee KS. Current progress and future perspectives in the development of anti-polo-like kinase 1 therapeutic agents. F1000 Res 2017; 6: 1024.
[http://dx.doi.org/10.12688/f1000research.11398.1] [PMID: 28721210]
[42]
Li Z, Li J, Kong Y, Yan S, Ahmad N, Liu X. Plk1 Phosphorylation of Mre11 antagonizes the DNA damage response. Cancer Res 2017; 77(12): 3169-80.
[http://dx.doi.org/10.1158/0008-5472.CAN-16-2787] [PMID: 28512243]
[43]
Cardnell RJ, Feng Y, Diao L, et al. Proteomic markers of DNA repair and PI3K pathway activation predict response to the PARP inhibitor BMN 673 in small cell lung cancer. Clin Cancer Res 2013; 19(22): 6322-8.
[http://dx.doi.org/10.1158/1078-0432.CCR-13-1975] [PMID: 24077350]
[44]
Kawahara N, Ogawa K, Nagayasu M, Kimura M, Sasaki Y, Kobayashi H. Candidate synthetic lethality partners to PARP inhibitors in the treatment of ovarian clear cell cancer. Biomed Rep 2017; 7(5): 391-9.
[http://dx.doi.org/10.3892/br.2017.990] [PMID: 29109859]
[45]
Kimbung S, Biskup E, Johansson I, et al. Co-targeting of the PI3K pathway improves the response of BRCA1 deficient breast cancer cells to PARP1 inhibition. Cancer Lett 2012; 319(2): 232-41.
[http://dx.doi.org/10.1016/j.canlet.2012.01.015] [PMID: 22266096]
[46]
Juvekar A, Burga LN, Hu H, et al. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov 2012; 2(11): 1048-63.
[http://dx.doi.org/10.1158/2159-8290.CD-11-0336] [PMID: 22915751]
[47]
Ibrahim YH, García-García C, Serra V, et al. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov 2012; 2(11): 1036-47.
[http://dx.doi.org/10.1158/2159-8290.CD-11-0348] [PMID: 22915752]
[48]
Jang NY, Kim DH, Cho BJ, et al. Radiosensitization with combined use of olaparib and PI-103 in triple-negative breast cancer. BMC Cancer 2015; 15: 89.
[http://dx.doi.org/10.1186/s12885-015-1090-7] [PMID: 25884663]
[49]
Cardnell RJ, Feng Y, Mukherjee S, et al. Activation of the PI3K/mTOR pathway following PARP inhibition in small cell lung cancer. PLoS One 2016; 11(4)e0152584
[http://dx.doi.org/10.1371/journal.pone.0152584] [PMID: 27055253]
[50]
Sun CK, Zhang F, Xiang T, et al. Phosphorylation of ribosomal protein S6 confers PARP inhibitor resistance in BRCA1-deficient cancers. Oncotarget 2014; 5(10): 3375-85.
[http://dx.doi.org/10.18632/oncotarget.1952] [PMID: 24831086]
[51]
Matulonis UA, Wulf GM, Barry WT, et al. Phase I dose escalation study of the PI3kinase pathway inhibitor BKM120 and the oral poly (ADP ribose) Polymerase (PARP) inhibitor olaparib for the treatment of high-grade serous ovarian and breast cancer. Ann Oncol 2017; 28(3): 512-8.
[http://dx.doi.org/10.1093/annonc/mdw672] [PMID: 27993796]
[52]
De P, Sun Y, Carlson JH, Friedman LS, Leyland-Jones BR, Dey N. Doubling down on the PI3K-AKT-mTOR pathway enhances the antitumor efficacy of PARP inhibitor in triple negative breast cancer model beyond BRCA-ness. Neoplasia 2014; 16(1): 43-72.
[http://dx.doi.org/10.1593/neo.131694] [PMID: 24563619]
[53]
Philip CA, Laskov I, Beauchamp MC, et al. Inhibition of PI3K-AKT-mTOR pathway sensitizes endometrial cancer cell lines to PARP inhibitors. BMC Cancer 2017; 17(1): 638.
[http://dx.doi.org/10.1186/s12885-017-3639-0] [PMID: 28886696]
[54]
Wang D, Wang M, Jiang N, et al. Effective use of PI3K inhibitor BKM120 and PARP inhibitor Olaparib to treat PIK3CA mutant ovarian cancer. Oncotarget 2016; 7(11): 13153-66.
[http://dx.doi.org/10.18632/oncotarget.7549] [PMID: 26909613]
[55]
Sun Y, Gallacchi D, Zhang EY, et al. Rapamycin-resistant poly (ADP-ribose) polymerase-1 overexpression is a potential therapeutic target in lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2014; 51(6): 738-49.
[http://dx.doi.org/10.1165/rcmb.2014-0033OC] [PMID: 24874429]
[56]
Smith J, Tho LM, Xu N, Gillespie DA. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res 2010; 108: 73-112.
[http://dx.doi.org/10.1016/B978-0-12-380888-2.00003-0] [PMID: 21034966]
[57]
Buisson R, Niraj J, Rodrigue A, et al. Coupling of homologous recombination and the checkpoint by ATR. Mol Cell 2017; 65(2): 336-46.
[http://dx.doi.org/10.1016/j.molcel.2016.12.007] [PMID: 28089683]
[58]
Jones SE, Fleuren EDG, Frankum J, et al. ATR is a therapeutic target in synovial sarcoma. Cancer Res 2017; 77(24): 7014-26.
[http://dx.doi.org/10.1158/0008-5472.CAN-17-2056] [PMID: 29038346]
[59]
Schmitt A, Knittel G, Welcker D, et al. ATM deficiency is associated with sensitivity to PARP1- and ATR-inhibitors in lung adenocarcinoma. Cancer Res 2017; 77(11): 3040-56.
[http://dx.doi.org/10.1158/0008-5472.CAN-16-3398] [PMID: 28363999]
[60]
Perkhofer L, Schmitt A, Romero Carrasco MC, et al. ATM defi-ciency generating genomic instability sensitizes pancreatic ductal adenocarcinoma cells to therapy-induced DNA damage. Cancer Res 2017; 77(20): 5576-90.
[http://dx.doi.org/10.1158/0008-5472.CAN-17-0634] [PMID: 28790064]
[61]
Yazinski SA, Comaills V, Buisson R, et al. ATR inhibition disrupts rewired homologous recombination and fork protection pathways in PARP inhibitor-resistant BRCA-deficient cancer cells. Genes Dev 2017; 31(3): 318-32.
[http://dx.doi.org/10.1101/gad.290957.116] [PMID: 28242626]
[62]
Krajewska M, Fehrmann RS, Schoonen PM, et al. ATR inhibition preferentially targets homologous recombination-deficient tumor cells. Oncogene 2015; 34(26): 3474-81.
[http://dx.doi.org/10.1038/onc.2014.276] [PMID: 25174396]
[63]
Kim H, George E, Ragland R, et al. Targeting the ATR/CHK1 axis with PARP inhibition results in tumor regression in BRCA-mutant ovarian cancer models. Clin Cancer Res 2017; 23(12): 3097-108.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-2273] [PMID: 27993965]
[64]
Yin Y, Shen Q, Zhang P, et al. Chk1 inhibition potentiates the therapeutic efficacy of PARP inhibitor BMN673 in gastric cancer. Am J Cancer Res 2017; 7(3): 473-83.
[PMID: 28401005]
[65]
Ahmed SU, Carruthers R, Gilmour L, Yildirim S, Watts C, Chalmers AJ. Selective inhibition of parallel DNA damage response pathways optimizes radiosensitization of glioblastoma stem-like cells. Cancer Res 2015; 75(20): 4416-28.
[http://dx.doi.org/10.1158/0008-5472.CAN-14-3790] [PMID: 26282173]
[66]
Abu-Sanad A, Wang Y, Hasheminasab F, et al. Simultaneous inhibition of ATR and PARP sensitizes colon cancer cell lines to irinotecan. Front Pharmacol 2015; 6: 147.
[http://dx.doi.org/10.3389/fphar.2015.00147] [PMID: 26257651]
[67]
Huntoon CJ, Flatten KS, Wahner Hendrickson AE, et al. ATR inhibition broadly sensitizes ovarian cancer cells to chemotherapy independent of BRCA status. Cancer Res 2013; 73(12): 3683-91.
[http://dx.doi.org/10.1158/0008-5472.CAN-13-0110] [PMID: 23548269]
[68]
Mu Y, Lou J, Srivastava M, et al. SLFN11 inhibits checkpoint maintenance and homologous recombination repair. EMBO Rep 2016; 17(1): 94-109.
[http://dx.doi.org/10.15252/embr.201540964] [PMID: 26658330]
[69]
Murai J, Feng Y, Yu GK, et al. Resistance to PARP inhibitors by SLFN11 inactivation can be overcome by ATR inhibition. Oncotarget 2016; 7(47): 76534-50.
[http://dx.doi.org/10.18632/oncotarget.12266] [PMID: 27708213]
[70]
Rundle S, Bradbury A, Drew Y, et al. Targeting the ATR-CHK1 axis in cancer therapy. Cancers 2017; 9(5)
[71]
Cooper CS, Park M, Blair DG, et al. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature 1984; 311(5981): 29-33.
[http://dx.doi.org/10.1038/311029a0] [PMID: 6590967]
[72]
Bottaro DP, Rubin JS, Faletto DL, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science 1991; 251(4995): 802-4.
[http://dx.doi.org/10.1126/science.1846706] [PMID: 1846706]
[73]
Mizuno S, Matsumoto K, Nakamura T. HGF as a renotrophic and anti-fibrotic regulator in chronic renal disease. Front Biosci 2008; 13: 7072-86.
[http://dx.doi.org/10.2741/3211] [PMID: 18508717]
[74]
Nakamura T, Mizuno S. The discovery of hepatocyte growth factor (HGF) and its significance for cell biology, life sciences and clinical medicine. Proc Jpn Acad, Ser B, Phys Biol Sci 2010; 86(6): 588-610.
[http://dx.doi.org/10.2183/pjab.86.588] [PMID: 20551596]
[75]
Mizuno S, Nakamura T. HGF-MET cascade, a key target for inhibiting cancer metastasis: The impact of NK4 discovery on cancer biology and therapeutics. Int J Mol Sci 2013; 14(1): 888-919.
[http://dx.doi.org/10.3390/ijms14010888] [PMID: 23296269]
[76]
Fan S, Ma YX, Wang JA, et al. The cytokine hepatocyte growth factor/scatter factor inhibits apoptosis and enhances DNA repair by a common mechanism involving signaling through phosphatidyl inositol 3′ kinase. Oncogene 2000; 19(18): 2212-23.
[http://dx.doi.org/10.1038/sj.onc.1203566] [PMID: 10822371]
[77]
Dai L, Trillo-Tinoco J, Cao Y, et al. Targeting HGF/c-MET induces cell cycle arrest, DNA damage, and apoptosis for primary effusion lymphoma. Blood 2015; 126(26): 2821-31.
[http://dx.doi.org/10.1182/blood-2015-07-658823] [PMID: 26531163]
[78]
Henneman L, van Miltenburg MH, Michalak EM, et al. Selective resistance to the PARP inhibitor olaparib in a mouse model for BRCA1-deficient metaplastic breast cancer. Proc Natl Acad Sci USA 2015; 112(27): 8409-14.
[http://dx.doi.org/10.1073/pnas.1500223112] [PMID: 26100884]
[79]
Du Y, Yamaguchi H, Wei Y, et al. Blocking c-Met-mediated PARP1 phosphorylation enhances anti-tumor effects of PARP inhibitors. Nat Med 2016; 22(2): 194-201.
[http://dx.doi.org/10.1038/nm.4032] [PMID: 26779812]
[80]
Shen Y, Aoyagi-Scharber M, Wang B. Trapping Poly(ADP-Ribose) Polymerase. J Pharmacol Exp Ther 2015; 353(3): 446-57.
[http://dx.doi.org/10.1124/jpet.114.222448] [PMID: 25758918]
[81]
Funakoshi Y, Mukohara T, Tomioka H, et al. Excessive MET signaling causes acquired resistance and addiction to MET inhibitors in the MKN45 gastric cancer cell line. Invest New Drugs 2013; 31(5): 1158-68.
[http://dx.doi.org/10.1007/s10637-013-9959-2] [PMID: 23568717]
[82]
Medová M, Aebersold DM, Blank-Liss W, et al. MET Inhibition Results in DNA breaks and synergistically sensitizes tumor cells to DNA-damaging agents potentially by breaching a damage-induced checkpoint arrest. Genes Cancer 2010; 1(10): 1053-62.
[http://dx.doi.org/10.1177/1947601910388030] [PMID: 21779429]
[83]
Song N, Che X, Xu L, et al. A novel function of hepatocyte growth factor in the activation of checkpoint kinase 1 phosphorylation in colon cancer cells. Mol Cell Biochem 2017; 436(1-2): 29-38.
[http://dx.doi.org/10.1007/s11010-017-3075-0] [PMID: 28573382]
[84]
Mihailidou C, Karamouzis MV, Schizas D, Papavassiliou AG. Co-targeting c-Met and DNA double-strand breaks (DSBs): Therapeutic strategies in BRCA-mutated gastric carcinomas. Biochimie 2017; 142: 135-43.
[http://dx.doi.org/10.1016/j.biochi.2017.09.001] [PMID: 28890386]
[85]
Kim Y, Kim A, Sharip A, et al. Reverse the resistance to PARP inhibitors. Int J Biol Sci 2017; 13(2): 198-208.
[http://dx.doi.org/10.7150/ijbs.17240] [PMID: 28255272]
[86]
Thomas C, Ji Y, Lodhi N, et al. Non-NAD-Like poly(ADP-Ribose) polymerase-1 inhibitors effectively eliminate cancer in vivo. EBioMedicine 2016; 13: 90-8.
[http://dx.doi.org/10.1016/j.ebiom.2016.10.001] [PMID: 27727003]
[87]
Dufour R, Daumar P, Mounetou E, et al. BCRP and P-gp relay overexpression in triple negative basal-like breast cancer cell line: a prospective role in resistance to Olaparib. Sci Rep 2015; 5: 12670.
[http://dx.doi.org/10.1038/srep12670] [PMID: 26234720]
[88]
Rottenberg S, Jaspers JE, Kersbergen A, et al. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. Proc Natl Acad Sci USA 2008; 105(44): 17079-84.
[http://dx.doi.org/10.1073/pnas.0806092105] [PMID: 18971340]
[89]
Vaidyanathan A, Sawers L, Gannon AL, et al. ABCB1 (MDR1) induction defines a common resistance mechanism in paclitaxel- and olaparib-resistant ovarian cancer cells. Br J Cancer 2016; 115(4): 431-41.
[http://dx.doi.org/10.1038/bjc.2016.203] [PMID: 27415012]
[90]
Jaspers JE, Kersbergen A, Boon U, et al. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov 2013; 3(1): 68-81.
[http://dx.doi.org/10.1158/2159-8290.CD-12-0049] [PMID: 23103855]
[91]
Xu G, Chapman JR, Brandsma I, et al. REV7 counteracts DNA double-strand break resection and affects PARP inhibition. Nature 2015; 521(7553): 541-4.
[http://dx.doi.org/10.1038/nature14328] [PMID: 25799992]
[92]
Isono M, Niimi A, Oike T, et al. BRCA1 directs the repair pathway to homologous recombination by promoting 53BP1 dephosphorylation. Cell Rep 2017; 18(2): 520-32.
[http://dx.doi.org/10.1016/j.celrep.2016.12.042] [PMID: 28076794]

© 2024 Bentham Science Publishers | Privacy Policy