Treatment of acquired drug resistance in multiple myeloma by combination therapy with XPO1 and topoisomerase II inhibitors

Joel G Turner, Jana L Dawson, Steven Grant, Kenneth H Shain, William S Dalton, Yun Dai, Mark Meads, Rachid Baz, Michael Kauffman, Sharon Shacham, Daniel M Sullivan, Joel G Turner, Jana L Dawson, Steven Grant, Kenneth H Shain, William S Dalton, Yun Dai, Mark Meads, Rachid Baz, Michael Kauffman, Sharon Shacham, Daniel M Sullivan

Abstract

Background: Acquired drug resistance is the greatest obstacle to the successful treatment of multiple myeloma (MM). Despite recent advanced treatment options such as liposomal formulations, proteasome inhibitors, immunomodulatory drugs, myeloma-targeted antibodies, and histone deacetylase inhibitors, MM is still considered an incurable disease.

Methods: We investigated whether the clinical exportin 1 (XPO1) inhibitor selinexor (KPT-330), when combined with pegylated liposomal doxorubicin (PLD) or doxorubicin hydrochloride, could overcome acquired drug resistance in multidrug-resistant human MM xenograft tumors, four different multidrug-resistant MM cell lines, or ex vivo MM biopsies from relapsed/refractory patients. Mechanistic studies were performed to assess co-localization of topoisomerase II alpha (TOP2A), DNA damage, and siRNA knockdown of drug targets.

Results: Selinexor was found to restore sensitivity of multidrug-resistant 8226B25, 8226Dox6, 8226Dox40, and U266PSR human MM cells to doxorubicin to levels found in parental myeloma cell lines. NOD/SCID-γ mice challenged with drug-resistant or parental U266 human MM and treated with selinexor/PLD had significantly decreased tumor growth and increased survival with minimal toxicity. Selinexor/doxorubicin treatment selectively induced apoptosis in CD138/light-chain-positive MM cells without affecting non-myeloma cells in ex vivo-treated bone marrow aspirates from newly diagnosed or relapsed/refractory MM patients. Selinexor inhibited XPO1-TOP2A protein complexes (proximity ligation assay), preventing nuclear export of TOP2A in both parental and multidrug-resistant MM cell lines. Selinexor/doxorubicin treatment significantly increased DNA damage (comet assay/γ-H2AX) in both parental and drug-resistant MM cells. TOP2A knockdown reversed both the anti-tumor effect and significantly reduced DNA damage induced by selinexor/doxorubicin treatment.

Conclusions: The combination of an XPO1 inhibitor and liposomal doxorubicin was highly effective against acquired drug resistance in in vitro MM models, in in vivo xenograft studies, and in ex vivo samples obtained from patients with relapsed/refractory myeloma. This drug combination synergistically induced TOP2A-mediated DNA damage and subsequent apoptosis. In addition, based on our preclinical data, we have initiated a phase I/II study with the XPO1 inhibitor selinexor and PLD (ClinicalTrials.gov NCT02186834). Initial results from both preclinical and clinical trials have shown significant promise for this drug combination for the treatment of MM.

Keywords: Acquired drug resistance; Liposomal doxorubicin; Mouse models; Multiple myeloma; Relapsed/refractory myeloma; XPO1 inhibition.

Figures

Fig. 1
Fig. 1
XPO1 inhibition sensitizes drug-resistant human MM cell lines to doxorubicin (DOX). Human 8226B25 (a), U266PSR (b), 8226Dox6 (c), and 8226Dox40 (d) drug-resistant and parental MM cell lines were treated concurrently for 20 h with selinexor (300 nM) or KOS-2464 (10 nM) +/− doxorubicin (2 μM) and assayed for apoptosis by flow cytometry (activated caspase 3) (n = 3). XPO1 inhibitors sensitized drug-resistant cells to DOX compared with single-agent treatment
Fig. 2
Fig. 2
Selinexor and KOS-2464 sensitize newly diagnosed and relapsed patient MM cells to doxorubicin. Bone marrow mononuclear cells were isolated and treated with selinexor or KOS-2464 +/− doxorubicin and fluorescently labeled with antibodies against activated caspase 3, CD138, and light chain (kappa or lambda). Newly diagnosed (n = 19) and relapsed (n = 22) CD-138/light-chain double-positive MM patient samples were all sensitized by selinexor and KOS-2464 to doxorubicin (P ≤ 1.37 × 10−8) versus single-agent treatment as shown by increased apoptosis (a, c). Non-myeloma CD138/light-chain double-negative patient cells were not sensitized to apoptosis by XPO1 inhibitors (b, d)
Fig. 3
Fig. 3
In vivo studies. PLD/selinexor treatment reduced drug-resistant U266PSR (a) and parental U266 (b) tumor growth compared with single-agent treatment with PLD (P = 0.001) or selinexor (P = 0.009) in drug-resistant U266PSR (a) and PLD (P = 0.0003) or selinexor (P = 0.0079) in parental U266 (b). c, d: Selinexor/PLD significantly improved survival in U266PSR-challenged mice compared with single-agent selinexor (P = 0.0095) or PLD (P = 0.0018) (c) and in U266 compared with selinexor (P = 0.0002) or PLD (P = 0.0024) (d)
Fig. 4
Fig. 4
Selinexor inhibits XPO1-TOP2A binding. Parental H929, U266, and 8226 and drug-resistant U266PSR, 8226Dox6, and 8226B25 plateau-density human MM cells (3 × 106/ml) were treated with selinexor (300 nM), cytospun, and assayed for intracellular co-localization of XPO1 and TOP2A by proximity ligation assay. A red fluorescent signal was generated only when XPO1 and TOP2A were in close proximity (<40 nm). a Selinexor blocked proximity co-localization of XPO1 and TOP2A. Inset, selinexor treatment did not affect XPO1 or TOP2A protein levels at 4 h as shown by Western blot. b Analysis of the number of XPO1-TOP2A foci showed that selinexor significantly decreased the number of foci in the nucleus and whole cells of both parental and drug-resistant MM cells
Fig. 5
Fig. 5
Comet DNA fragmentation assay. High-density human 8226 and U226 and drug-resistant cell lines 8226Dox6, 8226B25, and U266PSR MM (2 × 106/mL) were treated with selinexor (100 nM) followed by doxorubicin (5 μM). DNA fragmentation (double-strand breaks) was measured by the neutral comet assay. Doxorubicin or selinexor, when used as single agents, increased DNA double-strand cleavage compared with untreated controls. However, selinexor + doxorubicin further increased DNA fragmentation over doxorubicin or selinexor alone
Fig. 6
Fig. 6
Mechanism for selinexor and doxorubicin synergy. a Parental MM cell lines 8226 and U226 and drug-resistant 8226B25, 8226Dox6, and U266PSR cells were treated with doxorubicin (2 μM), selinexor (300 nM), or the combination for 20 h. Both parental and drug-resistant cell lines, when treated with the combination of selinexor and doxorubicin, had significantly increased DNA damage as shown by increased phospho-H2AX (Ser139) expression (n = 4). b SiRNA knockdown of TOP2A resulted in a substantial decrease in DNA damage (phospho-H2AX (Ser139)) and apoptosis, suggesting that selinexor and doxorubicin induced DNA damage, and subsequent apoptosis is TOP2A dependent. Inset, TOP2A knockdown (Western blot)

References

    1. Turner JG, Sullivan DM. CRM1-mediated nuclear export of proteins and drug resistance in cancer. Curr Med Chem. 2008;15:2648–55. doi: 10.2174/092986708786242859.
    1. Turner JG, Dawson J, Sullivan DM. Nuclear export of proteins and drug resistance in cancer. Biochem Pharmacol. 2012;83:1021–32. doi: 10.1016/j.bcp.2011.12.016.
    1. Turner JG, Marchion DC, Dawson JL, Emmons MF, Hazlehurst LA, Washausen P, Sullivan DM. Human multiple myeloma cells are sensitized to topoisomerase II inhibitors by CRM1 inhibition. Cancer Res. 2009;69:6899–905. doi: 10.1158/0008-5472.CAN-09-0484.
    1. Turner JG, Dawson J, Cubitt CL, Baz R, Sullivan DM. Inhibition of CRM1-dependent nuclear export sensitizes malignant cells to cytotoxic and targeted agents. Sem Cancer Biol. 2014;27C:62–73. doi: 10.1016/j.semcancer.2014.03.001.
    1. Turner JG, Dawson J, Emmons MF, Cubitt CL, Kauffman M, Shacham S, Hazlehurst LA, Sullivan DM. CRM1 inhibition sensitizes drug resistant human myeloma cells to topoisomerase II and proteasome inhibitors both in vitro and ex vivo. J Cancer. 2013;4:614–25. doi: 10.7150/jca.7080.
    1. Cheng Y, Holloway MP, Nguyen K, McCauley D, Landesman Y, Kauffman MG, Shacham S, Altura RA. XPO1 (CRM1) inhibition represses STAT3 activation to drive a survivin-dependent oncogenic switch in triple-negative breast cancer. Mol Cancer Ther. 2014;13:675–86. doi: 10.1158/1535-7163.MCT-13-0416.
    1. Miyake T, Pradeep S, Wu SY, Rupaimoole R, Zand B, Wen Y, Gharpure KM, Nagaraja AS, Hu W, Cho MS, et al. XPO1/CRM1 inhibition causes antitumor effects by mitochondrial accumulation of eIF5A. Clin Cancer Res. 2015;21:3286–97. doi: 10.1158/1078-0432.CCR-14-1953.
    1. Green AL, Ramkissoon SH, McCauley D, Jones K, Perry JA, Hsu JH, Ramkissoon LA, Maire CL, Hubbell-Engler B, Knoff DS, et al. Preclinical antitumor efficacy of selective exportin 1 inhibitors in glioblastoma. Neuro Oncol. 2015;17:697–707. doi: 10.1093/neuonc/nou303.
    1. Zheng Y, Gery S, Sun H, Shacham S, Kauffman M, Koeffler HP. KPT-330 inhibitor of XPO1-mediated nuclear export has anti-proliferative activity in hepatocellular carcinoma. Cancer Chemother Pharmacol. 2014;74:487–95. doi: 10.1007/s00280-014-2495-8.
    1. Inoue H, Kauffman M, Shacham S, Landesman Y, Yang J, Evans CP, Weiss RH. CRM1 blockade by selective inhibitors of nuclear export attenuates kidney cancer growth. J Urol. 2013;189(6):2317–26. doi: 10.1016/j.juro.2012.10.018.
    1. Wettersten HI, Landesman Y, Friedlander S, Shacham S, Kauffman M, Weiss RH. Specific inhibition of the nuclear exporter exportin-1 attenuates kidney cancer growth. PLoS One. 2014;9:e113867. doi: 10.1371/journal.pone.0113867.
    1. Ranganathan P, Yu X, Na C, Santhanam R, Shacham S, Kauffman M, Walker A, Klisovic R, Blum W, Caligiuri M, et al. Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia. Blood. 2012;120:1765–73. doi: 10.1182/blood-2012-04-423160.
    1. Tai YT, Landesman Y, Acharya C, Calle Y, Zhong MY, Cea M, Tannenbaum D, Cagnetta A, Reagan M, Munshi AA, 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.
    1. Lapalombella R, Sun Q, Williams K, Tangeman L, Jha S, Zhong Y, Goettl V, Mahoney E, Berglund C, Gupta S, et al. Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia. Blood. 2012;120:4621–34. doi: 10.1182/blood-2012-05-429506.
    1. Hing ZA, Mantel R, Beckwith KA, Guinn D, Williams E, Smith LL, Williams K, Johnson AJ, Lehman AM, Byrd JC, et al. Selinexor is effective in acquired resistance to ibrutinib and synergizes with ibrutinib in chronic lymphocytic leukemia. Blood. 2015;125:3128–32. doi: 10.1182/blood-2015-01-621391.
    1. Etchin J, Montero J, Berezovskaya A, Le BT, Kentsis A, Christie AL, Conway AS, Chen WC, Reed C, Mansour MR, et al. Activity of a selective inhibitor of nuclear export, selinexor (KPT-330), against AML-initiating cells engrafted into immunosuppressed NSG mice. Leukemia. 2016;30(1):190–9. doi: 10.1038/leu.2015.194.
    1. Sun H, Hattori N, Chien W, Sun Q, Sudo M, E-Ling GL, Ding L, Lim SL, Shacham S, Kauffman M, et al. KPT-330 has antitumour activity against non-small cell lung cancer. Br J Cancer. 2014;111:281–91. doi: 10.1038/bjc.2014.260.
    1. Zhang K, Wang M, Tamayo AT, Shacham S, Kauffman M, Lee J, Zhang L, Ou Z, Li C, Sun L, et al. Novel selective inhibitors of nuclear export CRM1 antagonists for therapy in mantle cell lymphoma. Exp Hemat. 2013;41:67–78. doi: 10.1016/j.exphem.2012.09.002.
    1. Tabe Y, Kojima K, Yamamoto S, Sekihara K, Matsushita H, Davis RE, Wang Z, Ma W, Ishizawa J, Kazuno S, et al. Ribosomal biogenesis and translational flux inhibition by the selective inhibitor of nuclear export (SINE) XPO1 antagonist KPT-185. PLoS One. 2015;10:e0137210. doi: 10.1371/journal.pone.0137210.
    1. Salas Fragomeni RA, Chung HW, Landesman Y, Senapedis W, Saint-Martin JR, Tsao H, Flaherty KT, Shacham S, Kauffman M, Cusack JC. CRM1 and BRAF inhibition synergize and induce tumor regression in BRAF-mutant melanoma. Mol Cancer Ther. 2013;12:1171–9. doi: 10.1158/1535-7163.MCT-12-1171.
    1. 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;132:2780–90. doi: 10.1038/jid.2012.233.
    1. De Cesare M, Cominetti D, Doldi V, Lopergolo A, Deraco M, Gandellini P, Friedlander S, Landesman Y, Kauffman MG, Shacham S, et al. Anti-tumor activity of selective inhibitors of XPO1/CRM1-mediated nuclear export in diffuse malignant peritoneal mesothelioma: the role of survivin. Oncotarget. 2015;6:13119–32. doi: 10.18632/oncotarget.3761.
    1. Azmi AS, Al-Katib A, Aboukameel A, McCauley D, Kauffman M, Shacham S, Mohammad RM. Selective inhibitors of nuclear export for the treatment of non-Hodgkin’s lymphomas. Haematologica. 2013;98:1098–106. doi: 10.3324/haematol.2012.074781.
    1. Azmi AS, Aboukameel A, Bao B, Sarkar FH, Philip PA, Kauffman M, Shacham S, Mohammad RM. Selective inhibitors of nuclear export block pancreatic cancer cell proliferation and reduce tumor growth in mice. Gastroenterology. 2013;144:447–56. doi: 10.1053/j.gastro.2012.10.036.
    1. Gao J, Azmi AS, Aboukameel A, Kauffman M, Shacham S, Abou-Samra AB, Mohammad RM. Nuclear retention of Fbw7 by specific inhibitors of nuclear export leads to Notch1 degradation in pancreatic cancer. Oncotarget. 2014;5:3444–54. doi: 10.18632/oncotarget.1813.
    1. Gravina GL, Tortoreto M, Mancini A, Addis A, Di Cesare E, Lenzi A, Landesman Y, McCauley D, Kauffman M, Shacham S, 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 Jematol Oncol. 2014;7:46. doi: 10.1186/1756-8722-7-46.
    1. Schmidt J, Braggio E, Kortuem KM, Egan JB, Zhu YX, Xin CS, Tiedemann RE, Palmer SE, Garbitt VM, McCauley D, 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;27:2357–65. doi: 10.1038/leu.2013.172.
    1. Gabizon AA. Pegylated liposomal doxorubicin: metamorphosis of an old drug into a new form of chemotherapy. Cancer Invest. 2001;19:424–36. doi: 10.1081/CNV-100103136.
    1. Orlowski RZ, Nagler A, Sonneveld P, Blade J, Hajek R, Spencer A, San Miguel J, Robak T, Dmoszynska A, Horvath N, et al. Randomized phase III study of pegylated liposomal doxorubicin plus bortezomib compared with bortezomib alone in relapsed or refractory multiple myeloma: combination therapy improves time to progression. J Clin Oncol. 2007;25:3892–901. doi: 10.1200/JCO.2006.10.5460.
    1. Baz RC, Shain KH, Hussein MA, Lee JH, Sullivan DM, Oliver EF, Nardelli LA, Nodzon LA, Zhao X, Ochoa-Bayona JL, et al. Phase II study of pegylated liposomal doxorubicin, low-dose dexamethasone, and lenalidomide in patients with newly diagnosed multiple myeloma. Am J Hematol. 2014;89:62–7. doi: 10.1002/ajh.23587.
    1. Chen S, Dai Y, Pei XY, Myers J, Wang L, Kramer LB, Garnett M, Schwartz DM, Su F, Simmons GL, et al. CDK inhibitors upregulate BH3-only proteins to sensitize human myeloma cells to BH3 mimetic therapies. Cancer Res. 2012;72:4225–37. doi: 10.1158/0008-5472.CAN-12-1118.
    1. Chen S, Zhang Y, Zhou L, Leng Y, Lin H, Kmieciak M, Pei XY, Jones R, Orlowski RZ, Dai Y, Grant S. A Bim-targeting strategy overcomes adaptive bortezomib-resistance in multiple myeloma through a novel link between autophagy and apoptosis. Blood. 2014;124(17):2687–97. doi: 10.1182/blood-2014-03-564534.
    1. Pei XY, Dai Y, Felthousen J, Chen S, Takabatake Y, Zhou L, Youssefian LE, Sanderson MW, Bodie WW, Kramer LB, et al. Circumvention of Mcl-1-dependent drug resistance by simultaneous Chk1 and MEK1/2 inhibition in human multiple myeloma cells. PLoS One. 2014;9:e89064. doi: 10.1371/journal.pone.0089064.
    1. Dalton WS, Durie BG, Alberts DS, Gerlach JH, Cress AE. Characterization of a new drug-resistant human myeloma cell line that expresses P-glycoprotein. Cancer Res. 1986;46:5125–30.
    1. Bellamy WT, Dorr RT, Dalton WS, Alberts DS. Direct relation of DNA lesions in multidrug-resistant human myeloma cells to intracellular doxorubicin concentration. Cancer Res. 1988;48:6360–4.
    1. Dalton WS, Grogan TM, Rybski JA, Scheper RJ, Richter L, Kailey J, Broxterman HJ, Pinedo HM, Salmon SE. Immunohistochemical detection and quantitation of P-glycoprotein in multiple drug-resistant human myeloma cells: association with level of drug resistance and drug accumulation. Blood. 1989;73:747–52.
    1. Marx V. Cell-line authentication demystified. Nat Meth. 2014;11:483–8. doi: 10.1038/nmeth.2932.
    1. Soderberg O, Gullberg M, Jarvius M, Ridderstrale K, Leuchowius KJ, Jarvius J, Wester K, Hydbring P, Bahram F, Larsson LG, Landegren U. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Meth. 2006;3:995–1000. doi: 10.1038/nmeth947.
    1. Olive PL, Wlodek D, Banath JP. DNA double-strand breaks measured in individual cells subjected to gel electrophoresis. Cancer Res. 1991;51:4671–6.
    1. Engel R, Valkov NI, Gump JL, Hazlehurst L, Dalton WS, Sullivan DM. The cytoplasmic trafficking of DNA topoisomerase IIalpha correlates with etoposide resistance in human myeloma cells. Exp Cell Res. 2004;295:421–31. doi: 10.1016/j.yexcr.2004.01.012.
    1. Olive PL, Banath JP, Durand RE. Detection of etoposide resistance by measuring DNA damage in individual Chinese hamster cells. J Natl Canc Inst. 1990;82:779–83. doi: 10.1093/jnci/82.9.779.
    1. Olive PL, Banath JP, Durand RE. Heterogeneity in radiation-induced DNA damage and repair in tumor and normal cells measured using the “comet” assay. Radiat Res. 1990;122:86–94. doi: 10.2307/3577587.
    1. Giles GI, Sharma RP. Topoisomerase enzymes as therapeutic targets for cancer chemotherapy. Med Chem. 2005;1:383–94. doi: 10.2174/1573406054368738.
    1. Barcelo F, Martorell J, Gavilanes F, Gonzalez-Ros JM. Equilibrium binding of daunomycin and adriamycin to calf thymus DNA. Temperature and ionic strength dependence of thermodynamic parameters. Biochem Pharmacol. 1988;37:2133–8. doi: 10.1016/0006-2952(88)90571-0.
    1. Tewey KM, Rowe TC, Yang L, Halligan BD, Liu LF. Adriamycin-induced DNA damage mediated by mammalian DNA topoisomerase II. Science. 1984;226:466–8. doi: 10.1126/science.6093249.
    1. Valkov NI, Gump JL, Engel R, Sullivan DM. Cell density-dependent VP-16 sensitivity of leukaemic cells is accompanied by the translocation of topoisomerase IIalpha from the nucleus to the cytoplasm. Br J Haematol. 2000;108:331–45. doi: 10.1046/j.1365-2141.2000.01832.x.
    1. Baz RC, Shain KH, Hussein MA, Lee JH, Sullivan DM, Oliver EF, Nardelli LA, Nodzon LA, Zhao X, Ochoa-Bayona JL, et al. Phase I trial of the combination of selinexor, liposomal doxorubicin and dexamethasone for relapsed and refractory multiple myeloma. J Clin Oncol. 2016;34:8013.

Source: PubMed

3
Suscribir