We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Selinexor: a first-in-class SINE compound for treatment of relapsed refractory multiple myeloma

    Shambavi Richard

    Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York 10029, USA

    ,
    Joshua Richter

    Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York 10029, USA

    &
    Sundar Jagannath

    *Author for correspondence:

    E-mail Address: Sundar.Jagannath@mountsinai.org

    Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York 10029, USA

    Published Online:https://doi.org/10.2217/fon-2020-0054

    The progression of multiple myeloma is accompanied by complex cytogenetic and epigenetic alterations that include mutation or functional inactivation of tumor suppressor proteins and overexpression of oncoproteins. Patients whose myeloma is refractory to the three major classes of drugs including immunomodulatory agents, proteasome inhibitors and anti-CD38 monoclonal antibodies have a very poor prognosis. Drugs with novel mechanisms of action that can bypass resistance mechanisms are sorely needed for this group of patients. Selinexor represents a novel, oral agent with an innovative mechanism of action that offers a significant therapeutic advance in this group of heavily treated patients. Moreover, this novel mechanism may provide additional options for patients with less refractory disease.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Xu D , Farmer A , Collett G et al. Sequence and structural analyses of nuclear export signals in the NESdb database. Mol. Biol. Cell 18, 3677–3693 (2012).
    • 2. Gravina GL , Senapedis W , McCauley D et al. Nucleo-cytoplasmic transport as a therapeutic target of cancer. J. Hematol. Oncol. 7, 85 (2014).
    • 3. Culjkovic-Kraljacic B , Baguet A , Volpon L et al. The oncogene eIF4E reprograms the nuclear pore complex to promote mRNA export and oncogenic transformation. Cell Rep. 2(2), 207–215 (2012).
    • 4. Conforti F , Wang Y , Rodriguez JA et al. Molecular pathways: anticancer activity by inhibition of nucleocytoplasmic shuttling. Clin. Cancer Res. 21(20), 4508–4513 (2015).
    • 5. Noske A , Weichert W , Niesporek S et al. Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpo1 is a prognostic factor in human ovarian cancer. Cancer 112, 1733–1743 (2008).
    • 6. Kojima K , Kornblau SM , Ruvolo V et al. Prognostic impact and targeting of CRM1 in acute myeloid leukemia. Blood 121, 4166–4174 (2013).
    • 7. Turner JG , Dawson J , Sullivan DM et al. Nuclear export of proteins and drug resistance in cancer. Biochem. Pharmacol. 83(8), 1021–1032 (2012).
    • 8. Tai YT , Landesman Y , Acharya C et al. CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: molecular mechanisms and therapeutic implications. Leukemia 28(1), 155–165 (2014). •• Selinexor mechanism of action in multiple myeloma.
    • 9. Kumar SK , Lee JH , Lahuerta JJ et al. Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: a multicenter international myeloma working group study. Leukemia 26, 149–157 (2012).
    • 10. Kumar SK , Dimopoulos MA , Kastritis E et al. Natural history of relapsed myeloma, refractory to immunomodulatory drugs and proteasome inhibitors: a multicenter IMWG study. Leukemia 31, 2443–2448 (2017).
    • 11. Pick M , Vainstein V , Goldschmidt N et al. Daratumumab resistance is frequent in advanced stage multiple myeloma patients irrespective of CD38 expression and is related to dismal prognosis. Eur. J. Haematol. 100, 494–501 (2018).
    • 12. Ferlay J , Soerjomataram I , Dikshit R et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 136(5), E359–E386 (2015).
    • 13. Mahipal A , Malafa M . Importins and exportins as therapeutic targets in cancer. Pharmacol. Ther. 164, 135–143 (2016).
    • 14. Kutay U , Guttinger S . Leucine- rich nuclear-export signals: born to be weak. Trends Cell Biol. 15, 121–124 (2005).
    • 15. Sharpless NE , DePinho RA . Cancer biology: gone but not forgotten. Nature 445, 606–607 (2007).
    • 16. Li S , Fu J , Lu C et al. Elevated translation initiation factor eIF4E is an attractive therapeutic target in multiple myeloma. Mol. Cancer Ther. 15(4), 711–719 (2016).
    • 17. Culjkovic B , Topisirovic I , Skrabanek L et al. eIF4E is a central node of an RNA regulon that governs cellular proliferation. J. Cell Biol. 175(3), 415–426 (2006).
    • 18. Schmidt J , Braggio E , Korteum KM 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 27, 2357–2365 (2013).
    • 19. Tan DS , Bedard PL , Kuruvilla J et al. Promising SINEs for embargoing nuclear-cytoplasmic export as an anticancer strategy. Cancer Discov. 4, 527–537 (2014).
    • 20. Yoshimura M , Ishizawa J , Ruvolo V et al. Induction of p53-mediated transcription and apoptosis by exportin-1 (XPO1) inhibition in mantle cell lymphoma. Cancer Sci. 105, 795–801 (2014).
    • 21. Tiedemann RE , Zhu YX , Schmidt J et al. Identification of molecular vulnerabilities in human multiple myeloma cells by RNA interference lethality screening of the druggable genome. Cancer Res. 72, 757–768 (2012).
    • 22. Das A , Wei G , Parikh K et al. Selective inhibitors of nuclear export (SINE) in hematological malignancies. Exp. Hematol. Oncol. 4, 7 (2015).
    • 23. Allegra A , Innao V , Allegra AG et al. Selective inhibitors of nuclear export in the treatment of hematologic malignancies. Clin. Lymphoma Myeloma Leuk. 19(11), 689–698 (2019).
    • 24. Hing ZA , Fung HY , Ranganathan P et al. Next-generation XPO1 inhibitor shows improved efficacy and in vivo tolerability in hematological malignancies. Leukemia 30, 2364–2372 (2016).
    • 25. Neggers JE , Vanstreels E , Baloglu E et al. Heterozygous mutation of cysteine528 in XPO1 is sufficient for resistance to selective inhibitors of nuclear export. Oncotarget 7(42), 68842–68850 (2016).
    • 26. Neggers JE , Vercruysse T , Jacquemyn M et al. Identifying drugtarget selectivity of small-molecule CRM1/XPO1 inhibitors by CRISPR/Cas9 genome editing. Chem. Biol. 22, 107–116 (2015).
    • 27. Golomb L , Bublik DR , Wilder S et al. Importin 7 and exportin 1 link c-myc and p53 to regulation of ribosomal biogenesis. Mol. Cell. 45, 222–232 (2012).
    • 28. Tan M , Wettersten HI , Chu K et al. Novel inhibitors of nuclear transport cause cell cycle arrest and decrease cyst growth in ADPKD associated with decreased CDK4 levels. Am. J. Physiol. Renal Physiol. 307(11), 1179–1186 (2014).
    • 29. Gandhi UH , Senapedis W , Baloglu E et al. Clinical implications of targeting XPO1 mediated nuclear export in multiple myeloma. Clin. Lymphoma Myeloma Leuk. 18, 335–345 (2018).
    • 30. Nguyen KT , Holloway MP , Altura RA . The CRM1 nuclear export protein in normal development and disease. Int. J. Biochem. Mol. Biol. 3, 137–151 (2012).
    • 31. Senapedis WT , Baloglu E , Landesman Y . Clinical translation of nuclear export inhibitors in cancer. Semin. Cancer Biol. 27, 74–86 (2014).
    • 32. Stauber RH , Mann W , Knauer SK et al. Nuclear and cytoplasmic survivin: molecular mechanism, prognostic, and therapeutic potential. Cancer Res. 67, 5999–6002 (2007).
    • 33. Argueta C , Kashayp T , Klebanov B et al. Selinexor synergizes with dexamethasone to repress mTORC1 signaling and induce multiple myeloma cell death. Oncotarget 9(39), 25529–25544 (2018).
    • 34. Nair J , Musi E , Schwartz GK . Selinexor (KPT-330) induces tumor suppression through nuclear sequestration of IκB and down-regulation of survivin. Clin. Cancer Res. 23(15), 4301–4311 (2017).
    • 35. Kashyap T , Argueta C , Aboukameel A et al. Selinexor, a selective inhibitor of nuclear export (SINE) compound, acts through NF-κB deactivation and combines with proteasome inhibitors to synergistically induce tumor cell death. Oncotarget 7(48), 7888395 (2016).
    • 36. Kim J , McMillan E , Kim HS et al. XPO1-dependent nuclear export is a druggable vulnerability in KRAS-mutant lung cancer. Nature 538, 114–117 (2016).
    • 37. Chesi M , Matthews GM , Garbitt VM et al. Drug response in a genetically engineered mouse model of multiple myeloma is predictive of clinical efficacy. Blood 120, 376–385 (2012).
    • 38. Elloul S , Chang H , Klebanov B et al. Synergistic antitumor effect of selinexor, a selective inhibitor of nuclear export (SINE) compound and panobinostat in a mouse model ofmultiple myeloma. Cancer Res. 76, 4720 (2016).
    • 39. Abdi J , Chen G , Chang H et al. Drug resistance in multiple myeloma: latest findings and new concepts on molecular mechanisms. Oncotarget 4, 2186–2207 (2013).
    • 40. Turner JG , Dawson JL , Cubitt CL et al. Inhibition of CRM1-dependent nuclear exportsensitizes malignant cells to cytotoxic and targeted agents. Semin. Cancer Biol. 27, 62–73 (2014).
    • 41. Rosebeck S , Kandarpa M , Alonge MM et al. Effects of inhibition of XPO1/CRM1-dependent nuclear export by selinexor (KPT-330), alone and in combination withcarfilzomib (CFZ), on apoptosis and autophagy in multiple myeloma (MM). Blood 122, 279 (2013).
    • 42. Turner JG , Kashyap T , Dawson JL et al. XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IkBα and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget 7, 78896–78909 (2016). • Selinexor + proteasome inhibitor mechanism of action to overcome proteasome inhibitor resistance.
    • 43. Rosebeck S , Alonge MM , Kandarpa M et al. Synergistic myeloma cell death via novel intracellular activation of caspase-10-dependent apoptosis by carfilzomib and selinexor. Mol. Cancer Ther. 15, 60–71 (2016).
    • 44. Turner JG , Dawson J , Bauer A et al. Ixazomib combined with the nuclear export inhibitors selinexor or eltanexor for the treatment of multiple myeloma. Experimental and Molecular Therapeutics. AACR Annual Meeting Abstract. GA, USA, 285 (2019).
    • 45. Lu S , Wang J . The resistance mechanisms of proteasome inhibitor bortezomib. Biomark. Res. 1(1), 13 (2013).
    • 46. Markovina S , Callander NS , O'Connor SL et al. Bortezomib resistant nuclear factor- kappaB activity in multiple myeloma cells. Mol. Cancer Res. 6(8), 1356–1364 (2008).
    • 47. Carlson R , Kashyap T , Elloul S et al. In vitro and in vivo antimultiple myeloma activity of selinexor (KPT-330), an oral selective inhibitor of nuclear export (SINE™) compound, is enhanced through combination with standard anti-myeloma agents . ESH 2Nd International Conference on Multiple Myeloma. Athens, Greece (2014).
    • 48. Chanukupp , Paul D , Taunk K et al. XPO1 is a critical player for bortezomib resistance in multiple myeloma: a quantitative proteomic approach. J Proteomics 209, 103504 (2019).
    • 49. Turner JG , Dawson JL , Grant S et al. Treatment of acquired drug resistance in multiple myeloma by combination therapy with XPO1 and topoisomerase II inhibitors. J. Hematol. Oncol. 9, 73 (2016).
    • 50. Turner JG , Dawson JL , Cubitt C et al. Selinexor and melphalan combination therapy for the treatment of multiple myeloma. AACR Annual Meeting Abstract. PA, USA (2015).
    • 51. Adachi Y , Yanagida M . Higher order chromosome structure is affected by cold sensitive mutations in Schizosaccharomyces pombe gene crm1 + which encodes a 115-kDA protein preferentially localized in the nucleus and its periphery. J. Cell Biol. 108, 1195–1207 (1989).
    • 52. Fornerod M , Ohno M , Yoshida M et al. CRM1 is an export receptor for leucine-rich nuclear export signals. Cell 90, 1051–1060 (1997).
    • 53. Dong X , Biswas A , Chook YM . Structural basis for assembly and disassembly of the CRM1 nuclear export complex. Nat. Struct. Mol. Biol. 16, 558–560 (2009).
    • 54. Dong X , Biswas A , Suel KE et al. Structural basis for leucine-rich nuclear export signal recognition by CRM1. Nature 458, 1136–1141 (2009).
    • 55. Kalid O , Toledo Warshaviak D , Shechter S et al. Consensus induced fit docking (cIFD): methodology, validation, and application to the discovery of novel Crm1 inhibitors. J. Comput. Aided. Mol. Des. 26, 1217–1228 (2012).
    • 56. Wu XZ . A new classification system of anticancer drugs – based on cell biological mechanisms. Med. Hypotheses 66, 883–887 (2006).
    • 57. US FDA. Karyopharm Therapeutics (2020). www.accessdata.fda.gov
    • 58. Chen C , Siegel D , Gutierrez M et al. Safety and efficacy of selinexor in relapsed or refractory multiple myeloma and Waldenstrom macroglobulinemia. Blood 131(8), 855–863 (2018). • Dose finding Phase I trial that established biweekly dosing of selinexor.
    • 59. Chari A , Vogl DT , Gavriatopoulou M et al. Oral selinexor–dexamethasone for triple-class refractory multiple myeloma. N. Engl. J. Med. 381, 727–738 (2019). •• Pivotal trial leading to the US FDA approval of selinexor in relapsed, refractory multiple myeloma.
    • 60. Vogl DT , Dingli D , Cornell RF et al. Selective inhibition of nuclear export with oral selinexor for treatment of relapsed or refractory multiple myeloma. J. Clin. Oncol. 36(9), 859–866 (2018). • STORM trial Part I that established recommended Phase II dosing for pivotal Part II.
    • 61. Cornell R , Parameswaran H , Tang S et al. Real world vs. clinical trial outcomes of triple class refractory penta-exposed multiple myeloma (MM). 17th Int. Myeloma Work. 179–181, FP-106 (2019).
    • 62. Richardson PG , Jagannath S , Chari A et al. Overall survival (OS) with oral selinexor plus low dose dexamethasone (Sd) in patients with triple class refractory-multiple myeloma (TCR-MM). J. Clin. Oncol. 8014 (2019).
    • 63. Jakubowiak AJ , Jasielec JK , Rosenbaum CA et al. Phase I study of selinexor plus carfilzomib and dexamethasone for the treatment of relapsed/refractory multiple myeloma. Br. J. Haematol. 186, 549–560 (2019). • Phase I trial showing feasibility of SKd to re-establish disease control in relapsed, refractory multiple myeloma including in carfilzomib refractory patients.
    • 64. Bahlis NJ , Sutherland H , White D et al. Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. Blood 132(24), 2546–2554 (2018). • Selinexor + velcade + dexamethasone arm of STOMP trial establishing dose and activity leading to confirmatory ongoing Phase III BOSTON trial.
    • 65. Baz RC , Shain KH , Hussein MA et al. Phase I trial of the combination of selinexor, liposomal doxorubicin and dexamethasone for relapsed and refractory multiple myeloma. J. Clin. Oncol. 34, 8013 (2016).
    • 66. Gavriatopoulou M , Vogl DT , Nooka A et al. Effect of age on the safety and efficacy of selinexor in patients with relapsed refractory multiple myeloma: a post-hoc analysis of the STORM study. 17th Int. Myeloma Work. 183–184, FP-110 (2019).
    • 67. Vogl DT , Nooka A , Gavriatopoulou M et al. Improvements in renal function with selinexor in relapsed/refractory multiple myeloma: post-hoc analyses from the STORM study. 17th Int. Myeloma Work. 184–186, FP-111 (2019).
    • 68. Gandhi UH , Cornell RF , lakshman A et al. Outcomes of patients with multiple myeloma refractory to CD38 targeted monoclonal antibody therapy. Leukemia 33(9), 2266–2275 (2019).
    • 69. Knopf KB , Duh MS , Lafeuille M-H et al. Meta-analysis of the efficacy and safety of bortezomib re-treatment in patients with multiple myeloma. Clin. Lymphoma Myeloma Leuk. 14, 380–388 (2014).
    • 70. Gasparetto C , Schiller GJ , Collander NS et al. A Phase Ib/II study of selinexor, carfilzomib and dexamethasone (SKd) in relapsed/refractory multiple myeloma (RRMM). Blood 134(Suppl. 1), 3157 (2019).
    • 71. White D , LeBlanc R , Venner C et al. Safety and efficacy of the combination of selinexor, lenalidomide and dexamethasone (SRd) in patients with relapsed/refractory multiple myeloma (RRMM). 17th Int. Myeloma Work. 84–86, OAB-083 (2019).
    • 72. White DJ , Lentzsch S , gasparetto C et al. Safety and efficacy of the combination of selinexor, lenalidomide and dexamethasone (SRd) in patients with newly diagnosed multiple myeloma. Blood 134(Suppl. 1), 3165 (2019).
    • 73. Chen C , Bahlis N , Gasparetto C et al. Selinexor, pomalidomide and dexamethasone (SPd) in patients with relapsed or refractory multiple myeloma. Blood 134(Suppl.1), 141 (2019).
    • 74. Gasparetto CJ , Lentzsch S , Schiller GJ et al. Deep and durable responses with selinexor, daratumumab, and dexamethasone (SDd) in patients with multiple myeloma (MM) previously exposed to proteasome inhibitors and immunomodulatory drugs: results of Phase Ib study of SDd. EHA Library S1606 (2019).
    • 75. Chari A , Vogl DT , Jagannath S et al. Selinexor containing regimens for the treatment of patients with multiple myeloma refractory to chimeric antigen receptor T cell (CAR-T) therapy. Blood 134(Suppl.1), 1854 (2019).
    • 76. Machlus KR , Wu SK , Vijey P et al. Selinexor-induced thrombocytopenia results from inhibition of thrombopoietin signaling in early megakaryopoiesis. Blood 130, 1132–1143 (2017).
    • 77. Etchin J , Berezovskaya A , Conway AS et al. KPT-8602, a second generation inhibitor of XPO-1 mediated nuclear transport, is well tolerated and highly active against AML blasts and leukemia-initiating cells. Leukemia 31, 143–150 (2017).