Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

Deciphering mechanisms of drug sensitivity and resistance to Selective Inhibitor of Nuclear Export (SINE) compounds

Authors: Marsha Crochiere, Trinayan Kashyap, Ori Kalid, Sharon Shechter, Boris Klebanov, William Senapedis, Jean-Richard Saint-Martin, Yosef Landesman

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Exportin 1 (XPO1) is a well-characterized nuclear export protein whose expression is up-regulated in many types of cancers and functions to transport key tumor suppressor proteins (TSPs) from the nucleus. Karyopharm Therapeutics has developed a series of small-molecule Selective Inhibitor of Nuclear Export (SINE) compounds, which have been shown to block XPO1 function both in vitro and in vivo. The drug candidate, selinexor (KPT-330), is currently in Phase-II/IIb clinical trials for treatment of both hematologic and solid tumors. The present study sought to decipher the mechanisms that render cells either sensitive or resistant to treatment with SINE compounds, represented by KPT-185, an early analogue of KPT-330.

Methods

Using the human fibrosarcoma HT1080 cell line, resistance to SINE was acquired over a period of 10 months of constant incubation with increasing concentration of KPT-185. Cell viability was assayed by MTT. Immunofluorescence was used to compare nuclear export of TSPs. Fluorescence activated cell sorting (FACS), quantitative polymerase chain reaction (qPCR), and immunoblots were used to measure effects on cell cycle, gene expression, and cell death. RNA from naïve and drug treated parental and resistant cells was analyzed by Affymetrix microarrays.

Results

Treatment of HT1080 cells with gradually increasing concentrations of SINE resulted in > 100 fold decrease in sensitivity to SINE cytotoxicity. Resistant cells displayed prolonged cell cycle, reduced nuclear accumulation of TSPs, and similar changes in protein expression compared to parental cells, however the magnitude of the protein expression changes were more significant in parental cells. Microarray analyses comparing parental to resistant cells indicate that a number of key signaling pathways were altered in resistant cells including expression changes in genes involved in adhesion, apoptosis, and inflammation. While the patterns of changes in transcription following drug treatment are similar in parental and resistant cells, the extent of response was more robust in the parental cells.

Conclusions

These results suggest that SINE resistance is conferred by alterations in signaling pathways downstream of XPO1 inhibition. Modulation of these pathways could potentially overcome the resistance to nuclear export inhibitors.
Appendix
Available only for authorised users
Literature
3.
go back to reference Noske A, Weichert W, Niesporek S, Roske A, Buckendahl AC, Koch I, et al. Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpo1 is a prognostic factor in human ovarian cancer. Cancer. 2008;112(8):1733–43. doi:10.1002/cncr.23354.PubMedCrossRef Noske A, Weichert W, Niesporek S, Roske A, Buckendahl AC, Koch I, et al. Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpo1 is a prognostic factor in human ovarian cancer. Cancer. 2008;112(8):1733–43. doi:10.​1002/​cncr.​23354.PubMedCrossRef
4.
go back to reference Yao Y, Dong Y, Lin F, Zhao H, Shen Z, Chen P, et al. The expression of CRM1 is associated with prognosis in human osteosarcoma. Oncol Rep. 2009;21(1):229–35.PubMed Yao Y, Dong Y, Lin F, Zhao H, Shen Z, Chen P, et al. The expression of CRM1 is associated with prognosis in human osteosarcoma. Oncol Rep. 2009;21(1):229–35.PubMed
5.
go back to reference Huang WY, Yue L, Qiu WS, Wang LW, Zhou XH, Sun YJ. Prognostic value of CRM1 in pancreas cancer. Clin Invest Med. 2009;32(6):E315.PubMed Huang WY, Yue L, Qiu WS, Wang LW, Zhou XH, Sun YJ. Prognostic value of CRM1 in pancreas cancer. Clin Invest Med. 2009;32(6):E315.PubMed
6.
go back to reference van der Watt PJ, Maske CP, Hendricks DT, Parker MI, Denny L, Govender D, et al. The Karyopherin proteins, Crm1 and Karyopherin beta1, are overexpressed in cervical cancer and are critical for cancer cell survival and proliferation. Int J Cancer. 2009;124(8):1829–40. doi:10.1002/ijc.24146.PubMedCrossRef van der Watt PJ, Maske CP, Hendricks DT, Parker MI, Denny L, Govender D, et al. The Karyopherin proteins, Crm1 and Karyopherin beta1, are overexpressed in cervical cancer and are critical for cancer cell survival and proliferation. Int J Cancer. 2009;124(8):1829–40. doi:10.​1002/​ijc.​24146.PubMedCrossRef
8.
go back to reference Pathria G, Wagner C, Wagner SN. Inhibition of CRM1-mediated nucleocytoplasmic transport: triggering human melanoma cell apoptosis by perturbing multiple cellular pathways. J Invest Dermatol. 2012;132(12):2780–90. doi:10.1038/jid.2012.233.PubMedCrossRef Pathria G, Wagner C, Wagner SN. Inhibition of CRM1-mediated nucleocytoplasmic transport: triggering human melanoma cell apoptosis by perturbing multiple cellular pathways. J Invest Dermatol. 2012;132(12):2780–90. doi:10.​1038/​jid.​2012.​233.PubMedCrossRef
11.
go back to reference Schmidt J, Braggio E, Kortuem KM, Egan JB, Zhu YX, Xin CS, et al. Genome-wide studies in multiple myeloma identify XPO1/CRM1 as a critical target validated using the selective nuclear export inhibitor KPT-276. Leukemia. 2013. doi:10.1038/leu.2013.172.PubMedCentral Schmidt J, Braggio E, Kortuem KM, Egan JB, Zhu YX, Xin CS, et al. Genome-wide studies in multiple myeloma identify XPO1/CRM1 as a critical target validated using the selective nuclear export inhibitor KPT-276. Leukemia. 2013. doi:10.​1038/​leu.​2013.​172.PubMedCentral
12.
go back to reference Tai YT, Landesman Y, Acharya C, Calle Y, Zhong MY, Cea M, et al. CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: molecular mechanisms and therapeutic implications. Leukemia. 2014;28(1):155–65. doi:10.1038/leu.2013.115.PubMedCrossRef Tai YT, Landesman Y, Acharya C, Calle Y, Zhong MY, Cea M, et al. CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: molecular mechanisms and therapeutic implications. Leukemia. 2014;28(1):155–65. doi:10.​1038/​leu.​2013.​115.PubMedCrossRef
13.
go back to reference Walker CJ, Oaks JJ, Santhanam R, Neviani P, Harb JG, Ferenchak G, et al. Preclinical and clinical efficacy of XPO1/CRM1 inhibition by the karyopherin inhibitor KPT-330 in Ph + leukemias. Blood. 2013. doi:10.1182/blood-2013-04-495374. Walker CJ, Oaks JJ, Santhanam R, Neviani P, Harb JG, Ferenchak G, et al. Preclinical and clinical efficacy of XPO1/CRM1 inhibition by the karyopherin inhibitor KPT-330 in Ph + leukemias. Blood. 2013. doi:10.​1182/​blood-2013-04-495374.
14.
go back to reference Kanai M, Hanashiro K, Kim SH, Hanai S, Boulares AH, Miwa M, et al. Inhibition of Crm1-p53 interaction and nuclear export of p53 by poly(ADP-ribosyl)ation. Nat Cell Biol. 2007;9(10):1175–83. doi:10.1038/ncb1638.PubMedCrossRef Kanai M, Hanashiro K, Kim SH, Hanai S, Boulares AH, Miwa M, et al. Inhibition of Crm1-p53 interaction and nuclear export of p53 by poly(ADP-ribosyl)ation. Nat Cell Biol. 2007;9(10):1175–83. doi:10.​1038/​ncb1638.PubMedCrossRef
19.
go back to reference Vogt PK, Jiang H, Aoki M. Triple layer control: phosphorylation, acetylation and ubiquitination of FOXO proteins. Cell Cycle. 2005;4(7):908–13.PubMedCrossRef Vogt PK, Jiang H, Aoki M. Triple layer control: phosphorylation, acetylation and ubiquitination of FOXO proteins. Cell Cycle. 2005;4(7):908–13.PubMedCrossRef
20.
go back to reference Huang TT, Kudo N, Yoshida M, Miyamoto S. A nuclear export signal in the N-terminal regulatory domain of IkappaBalpha controls cytoplasmic localization of inactive NF-kappaB/IkappaBalpha complexes. Proc Natl Acad Sci U S A. 2000;97(3):1014–9.PubMedPubMedCentralCrossRef Huang TT, Kudo N, Yoshida M, Miyamoto S. A nuclear export signal in the N-terminal regulatory domain of IkappaBalpha controls cytoplasmic localization of inactive NF-kappaB/IkappaBalpha complexes. Proc Natl Acad Sci U S A. 2000;97(3):1014–9.PubMedPubMedCentralCrossRef
25.
go back to reference Hamamoto T, Gunji S, Tsuji H, Beppu T. Leptomycins A and B, new antifungal antibiotics. I. Taxonomy of the producing strain and their fermentation, purification and characterization. J Antibiot (Tokyo). 1983;36(6):639–45.CrossRef Hamamoto T, Gunji S, Tsuji H, Beppu T. Leptomycins A and B, new antifungal antibiotics. I. Taxonomy of the producing strain and their fermentation, purification and characterization. J Antibiot (Tokyo). 1983;36(6):639–45.CrossRef
26.
go back to reference Nishi K, Yoshida M, Fujiwara D, Nishikawa M, Horinouchi S, Beppu T. Leptomycin B targets a regulatory cascade of crm1, a fission yeast nuclear protein, involved in control of higher order chromosome structure and gene expression. J Biol Chem. 1994;269(9):6320–4.PubMed Nishi K, Yoshida M, Fujiwara D, Nishikawa M, Horinouchi S, Beppu T. Leptomycin B targets a regulatory cascade of crm1, a fission yeast nuclear protein, involved in control of higher order chromosome structure and gene expression. J Biol Chem. 1994;269(9):6320–4.PubMed
27.
go back to reference Kudo N, Matsumori N, Taoka H, Fujiwara D, Schreiner EP, Wolff B, et al. Leptomycin B inactivates CRM1/exportin 1 by covalent modification at a cysteine residue in the central conserved region. Proc Natl Acad Sci U S A. 1999;96(16):9112–7.PubMedPubMedCentralCrossRef Kudo N, Matsumori N, Taoka H, Fujiwara D, Schreiner EP, Wolff B, et al. Leptomycin B inactivates CRM1/exportin 1 by covalent modification at a cysteine residue in the central conserved region. Proc Natl Acad Sci U S A. 1999;96(16):9112–7.PubMedPubMedCentralCrossRef
30.
go back to reference Van Neck T, Pannecouque C, Vanstreels E, Stevens M, Dehaen W, Daelemans D. Inhibition of the CRM1-mediated nucleocytoplasmic transport by N-azolylacrylates: structure-activity relationship and mechanism of action. Bioorg Med Chem. 2008;16(21):9487–97. doi:10.1016/j.bmc.2008.09.051.PubMedCrossRef Van Neck T, Pannecouque C, Vanstreels E, Stevens M, Dehaen W, Daelemans D. Inhibition of the CRM1-mediated nucleocytoplasmic transport by N-azolylacrylates: structure-activity relationship and mechanism of action. Bioorg Med Chem. 2008;16(21):9487–97. doi:10.​1016/​j.​bmc.​2008.​09.​051.PubMedCrossRef
34.
36.
go back to reference Etchin J, Sanda T, Mansour MR, Kentsis A, Montero J, Le BT, et al. KPT-330 inhibitor of CRM1 (XPO1)-mediated nuclear export has selective anti-leukaemic activity in preclinical models of T-cell acute lymphoblastic leukaemia and acute myeloid leukaemia. Br J Haematol. 2013;161(1):117–27. doi:10.1111/bjh.12231.PubMedPubMedCentralCrossRef Etchin J, Sanda T, Mansour MR, Kentsis A, Montero J, Le BT, et al. KPT-330 inhibitor of CRM1 (XPO1)-mediated nuclear export has selective anti-leukaemic activity in preclinical models of T-cell acute lymphoblastic leukaemia and acute myeloid leukaemia. Br J Haematol. 2013;161(1):117–27. doi:10.​1111/​bjh.​12231.PubMedPubMedCentralCrossRef
38.
go back to reference Zhong Y, El-Gamal D, Dubovsky JA, Beckwith KA, Harrington BK, Williams KE, et al. Selinexor suppresses downstream effectors of B-cell activation, proliferation and migration in chronic lymphocytic leukemia cells. Leukemia. 2014;28(5):1158–63. doi:10.1038/leu.2014.9.PubMedPubMedCentralCrossRef Zhong Y, El-Gamal D, Dubovsky JA, Beckwith KA, Harrington BK, Williams KE, et al. Selinexor suppresses downstream effectors of B-cell activation, proliferation and migration in chronic lymphocytic leukemia cells. Leukemia. 2014;28(5):1158–63. doi:10.​1038/​leu.​2014.​9.PubMedPubMedCentralCrossRef
41.
go back to reference Gao J, Azmi AS, Aboukameel A, Kauffman M, Shacham S, Abou-Samra AB, et al. Nuclear retention of Fbw7 by specific inhibitors of nuclear export leads to Notch1 degradation in pancreatic cancer. Oncotarget. 2014;5(11):3444–54.PubMedPubMedCentralCrossRef Gao J, Azmi AS, Aboukameel A, Kauffman M, Shacham S, Abou-Samra AB, et al. Nuclear retention of Fbw7 by specific inhibitors of nuclear export leads to Notch1 degradation in pancreatic cancer. Oncotarget. 2014;5(11):3444–54.PubMedPubMedCentralCrossRef
44.
go back to reference Sun H, Hattori N, Chien W, Sun Q, Sudo M, E-Ling GL, et al. KPT-330 has antitumour activity against non-small cell lung cancer. Br J Cancer. 2014;111(2):281–91.PubMedPubMedCentralCrossRef Sun H, Hattori N, Chien W, Sun Q, Sudo M, E-Ling GL, et al. KPT-330 has antitumour activity against non-small cell lung cancer. Br J Cancer. 2014;111(2):281–91.PubMedPubMedCentralCrossRef
49.
go back to reference Gravina GL, Tortoreto M, Mancini A, Addis A, Di Cesare E, Lenzi A, et al. XPO1/CRM1-selective inhibitors of nuclear export (SINE) reduce tumor spreading and improve overall survival in preclinical models of prostate cancer (PCa). J Hematol Oncol. 2014;7:46. doi:10.1186/1756-8722-7-46.PubMedPubMedCentralCrossRef Gravina GL, Tortoreto M, Mancini A, Addis A, Di Cesare E, Lenzi A, et al. XPO1/CRM1-selective inhibitors of nuclear export (SINE) reduce tumor spreading and improve overall survival in preclinical models of prostate cancer (PCa). J Hematol Oncol. 2014;7:46. doi:10.​1186/​1756-8722-7-46.PubMedPubMedCentralCrossRef
50.
go back to reference Mendonca J, Sharma A, Kim HS, Hammers H, Meeker A, De Marzo A, et al. Selective inhibitors of nuclear export (SINE) as novel therapeutics for prostate cancer. Oncotarget. 2014;5(15):6102–12.PubMedPubMedCentralCrossRef Mendonca J, Sharma A, Kim HS, Hammers H, Meeker A, De Marzo A, et al. Selective inhibitors of nuclear export (SINE) as novel therapeutics for prostate cancer. Oncotarget. 2014;5(15):6102–12.PubMedPubMedCentralCrossRef
52.
go back to reference Rasheed S, Nelson-Rees WA, Toth EM, Arnstein P, Gardner MB. Characterization of a newly derived human sarcoma cell line (HT-1080). Cancer. 1974;33(4):1027–33.PubMedCrossRef Rasheed S, Nelson-Rees WA, Toth EM, Arnstein P, Gardner MB. Characterization of a newly derived human sarcoma cell line (HT-1080). Cancer. 1974;33(4):1027–33.PubMedCrossRef
58.
go back to reference Mahon FX, Deininger MW, Schultheis B, Chabrol J, Reiffers J, Goldman JM, et al. Selection and characterization of BCR-ABL positive cell lines with differential sensitivity to the tyrosine kinase inhibitor STI571: diverse mechanisms of resistance. Blood. 2000;96(3):1070–9.PubMed Mahon FX, Deininger MW, Schultheis B, Chabrol J, Reiffers J, Goldman JM, et al. Selection and characterization of BCR-ABL positive cell lines with differential sensitivity to the tyrosine kinase inhibitor STI571: diverse mechanisms of resistance. Blood. 2000;96(3):1070–9.PubMed
59.
go back to reference Wang NN, Zhao LJ, Wu LN, He MF, Qu JW, Zhao YB, et al. Mechanistic analysis of taxol-induced multidrug resistance in an ovarian cancer cell line. Asian Pac J Cancer Prev. 2013;14(9):4983–8.PubMedCrossRef Wang NN, Zhao LJ, Wu LN, He MF, Qu JW, Zhao YB, et al. Mechanistic analysis of taxol-induced multidrug resistance in an ovarian cancer cell line. Asian Pac J Cancer Prev. 2013;14(9):4983–8.PubMedCrossRef
65.
go back to reference Okuda H, Kobayashi A, Xia B, Watabe M, Pai SK, Hirota S, et al. Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of breast cancer stem-like cells with macrophages and stromal cells. Cancer Res. 2012;72(2):537–47. doi:10.1158/0008-5472.CAN-11-1678.PubMedCrossRef Okuda H, Kobayashi A, Xia B, Watabe M, Pai SK, Hirota S, et al. Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of breast cancer stem-like cells with macrophages and stromal cells. Cancer Res. 2012;72(2):537–47. doi:10.​1158/​0008-5472.​CAN-11-1678.PubMedCrossRef
70.
72.
77.
go back to reference Sharpe JC, Abel PD, Gilbertson JA, Brawn P, Foster CS. Modulated expression of human leucocyte antigen class I and class II determinants in hyperplastic and malignant human prostatic epithelium. Br J Urol. 1994;74(5):609–16.PubMedCrossRef Sharpe JC, Abel PD, Gilbertson JA, Brawn P, Foster CS. Modulated expression of human leucocyte antigen class I and class II determinants in hyperplastic and malignant human prostatic epithelium. Br J Urol. 1994;74(5):609–16.PubMedCrossRef
78.
go back to reference Reubi JC, Gugger M, Waser B, Schaer JC. Y(1)-mediated effect of neuropeptide Y in cancer: breast carcinomas as targets. Cancer Res. 2001;61(11):4636–41.PubMed Reubi JC, Gugger M, Waser B, Schaer JC. Y(1)-mediated effect of neuropeptide Y in cancer: breast carcinomas as targets. Cancer Res. 2001;61(11):4636–41.PubMed
80.
go back to reference Florio M, Hernandez MC, Yang H, Shu HK, Cleveland JL, Israel MA. Id2 promotes apoptosis by a novel mechanism independent of dimerization to basic helix-loop-helix factors. Mol Cell Biol. 1998;18(9):5435–44.PubMedPubMedCentralCrossRef Florio M, Hernandez MC, Yang H, Shu HK, Cleveland JL, Israel MA. Id2 promotes apoptosis by a novel mechanism independent of dimerization to basic helix-loop-helix factors. Mol Cell Biol. 1998;18(9):5435–44.PubMedPubMedCentralCrossRef
82.
83.
go back to reference Liu C, Adamson E, Mercola D. Transcription factor EGR-1 suppresses the growth and transformation of human HT-1080 fibrosarcoma cells by induction of transforming growth factor beta 1. Proc Natl Acad Sci U S A. 1996;93(21):11831–6.PubMedPubMedCentralCrossRef Liu C, Adamson E, Mercola D. Transcription factor EGR-1 suppresses the growth and transformation of human HT-1080 fibrosarcoma cells by induction of transforming growth factor beta 1. Proc Natl Acad Sci U S A. 1996;93(21):11831–6.PubMedPubMedCentralCrossRef
84.
go back to reference Su L, Cheng H, Sampaio AV, Nielsen TO, Underhill TM. EGR1 reactivation by histone deacetylase inhibitors promotes synovial sarcoma cell death through the PTEN tumor suppressor. Oncogene. 2010;29(30):4352–61. doi:10.1038/onc.2010.204.PubMedCrossRef Su L, Cheng H, Sampaio AV, Nielsen TO, Underhill TM. EGR1 reactivation by histone deacetylase inhibitors promotes synovial sarcoma cell death through the PTEN tumor suppressor. Oncogene. 2010;29(30):4352–61. doi:10.​1038/​onc.​2010.​204.PubMedCrossRef
85.
go back to reference Del Bufalo D, Di Castro V, Biroccio A, Varmi M, Salani D, Rosano L, et al. Endothelin-1 protects ovarian carcinoma cells against paclitaxel-induced apoptosis: requirement for Akt activation. Mol Pharmacol. 2002;61(3):524–32.PubMedCrossRef Del Bufalo D, Di Castro V, Biroccio A, Varmi M, Salani D, Rosano L, et al. Endothelin-1 protects ovarian carcinoma cells against paclitaxel-induced apoptosis: requirement for Akt activation. Mol Pharmacol. 2002;61(3):524–32.PubMedCrossRef
87.
go back to reference Kaluza D, Kroll J, Gesierich S, Manavski Y, Boeckel JN, Doebele C, et al. Histone deacetylase 9 promotes angiogenesis by targeting the antiangiogenic microRNA-17-92 cluster in endothelial cells. Arterioscler Thromb Vasc Biol. 2013;33(3):533–43. doi:10.1161/ATVBAHA.112.300415.PubMedCrossRef Kaluza D, Kroll J, Gesierich S, Manavski Y, Boeckel JN, Doebele C, et al. Histone deacetylase 9 promotes angiogenesis by targeting the antiangiogenic microRNA-17-92 cluster in endothelial cells. Arterioscler Thromb Vasc Biol. 2013;33(3):533–43. doi:10.​1161/​ATVBAHA.​112.​300415.PubMedCrossRef
88.
89.
93.
go back to reference Kuang SQ, Tong WG, Yang H, Lin W, Lee MK, Fang ZH, et al. Genome-wide identification of aberrantly methylated promoter associated CpG islands in acute lymphocytic leukemia. Leukemia. 2008;22(8):1529–38. doi:10.1038/leu.2008.130.PubMedCrossRef Kuang SQ, Tong WG, Yang H, Lin W, Lee MK, Fang ZH, et al. Genome-wide identification of aberrantly methylated promoter associated CpG islands in acute lymphocytic leukemia. Leukemia. 2008;22(8):1529–38. doi:10.​1038/​leu.​2008.​130.PubMedCrossRef
Metadata
Title
Deciphering mechanisms of drug sensitivity and resistance to Selective Inhibitor of Nuclear Export (SINE) compounds
Authors
Marsha Crochiere
Trinayan Kashyap
Ori Kalid
Sharon Shechter
Boris Klebanov
William Senapedis
Jean-Richard Saint-Martin
Yosef Landesman
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1790-z

Other articles of this Issue 1/2015

BMC Cancer 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

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

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