Open access

Introductory Chapter: Oral Selinexor, a Selective Inhibitor of Nuclear Export in the Treatment of Patients with Multiple Myeloma Refractory to Proteasome Inhibitors, Immunomodulatory Agents and Monoclonal Antibodies

Written By

Ota Fuchs

Published: 28 July 2021

DOI: 10.5772/intechopen.96945

From the Edited Volume

Multiple Myeloma

Edited by Ota Fuchs

Chapter metrics overview

323 Chapter Downloads

View Full Metrics

1. Introduction

The export of proteins from the nucleus to the cytoplasm plays an important role in the development of cancer and drug resistance [1, 2, 3]. The major mammalian nuclear export receptor protein is exportin 1 (XPO1, also known as chromosomal maintenance 1/ CRM1/) [1, 2, 3, 4, 5]. The crystal structure of this protein showed a complex with the Ran protein (Ras-related nuclear protein) bound to GTP [6, 7]. XPO1 interacts also with nucleoporins in the nuclear pore complex and transports multiple tumor suppressor proteins (eg p53, FOXO, p21 pRB, BRCA1/2), growth regulators, and oncoprotein mRNAs (eg c-myc, Bcl-xL, MDM2, cyclins) containing a leucine rich nuclear export signal (NES) (Figure 1) [8]. XPO1 is also involved in regulation of cytoplasmic localization and translation of c-myc and other oncoprotein mRNAs (eg cyclin D1, Bcl-6, Mdm2, and Pim) through complexing with eukaryotic initiation factor 4E (eIF4E) [9]. The XPO1 protein level is increased in many types of cancer including multiple myeloma [10, 11, 12, 13]. As a result of the increased nuclear-cytoplasmic transport in cancer cells, an elevated level of multiple tumor suppressor proteins and oncoproteins in the cytoplasm leads to advanced disease, resistance to therapy, and poor survival. Thus, XPO1 is a promising cancer drug target. Leptomycin B (LMB) is a Streptomyces metabolite that inhibits the function of XPO1 in NES-dependent nuclear export of proteins [14]. However, clinical studies found serious side effects of LMB. In order to find a more specific inhibitor of XPO1 without side effects, many natural and synthetic compounds have been tested. These compounds include selinexor (KPT-330, XPOVIO™), verdinexor (KPT-335), KPT-185, KPT-276, KPT-251, and KPT-8602 [15, 16, 17, 18]. These agents are a family of small molecules that block nuclear export through covalent binding to cysteine 528 (Cys528) in the cargo proteins NES-binding pocket of exportin1 and contribute to cancer cell death [15]. All these drugs have been developed by Karyopharm Therapeutics Inc., Natick, MA.

Figure 1.

Exportin 1-mediated nuclear export in multiple myeloma; Abbreviations: Ran-GDP and Ran-GTP – GDP or GTP bound Ras related factor; NPC – nuclear pore complex; SINE - selective inhibitor of nuclear export; FOXO – a subgroup of the Forkhead family of transcription factors, p53 – a tumor suppressor protein and transcription factor; XPO1 – exportin 1, also known as chromosomal maintenance 1 (CRM1); (A) XPO1 transports nuclear proteins out of the nucleus. Cargo proteins such as FOXO or p53 that are marked for export from the nucleus bind a pocket in XPO1 in the presence of the activated small G-protein, Ran. The active Ran-GTP-XPO1-cargo complex is exported from the nucleus through the nuclear pore complex driven by the concentration gradient of Ran-GTP across the nuclear membrane. Once in the cytoplasm, Ran-GTP is hydrolyzed to Ran-GDP, and the XPO1-cargo complex dissociates. (B) SINE compounds (Hexagons) bind to XPO1-Cys528 and occupy the cargo-binding pocket of XPO1 and prevent formation of the Ran-GTP-XPO1-cargo complex. The result is increased nuclear localization of tumor suppressor cargo proteins and upregulation of their transcriptional activity [15].

Advertisement

2. Selinexor-a small molecule exportin 1 inhibitor

The structural formula of selinexor is shown in Figure 2. Selinexor is a first member of small molecule oral inhibitors of exportin 1 developed for the treatment of cancer. Selinexor in combination with a synthetic glucocorticoid dexamethasone was approved by the FDA (U.S. Food and Drug Administration) on July 3, 2019 for the treatment of adult patients with relapsed or refractory multiple myeloma (RRMM) who have received at least four prior therapies. Selinexor synergizes with dexamethasone and inhibits the mTOR pathway and subsequently induces cell death in multiple myeloma cells [19]. Selinexor increases the expression of glucocorticoid receptor and in combination with dexamethasone stimulates transcriptional activity of the glucocoticoid receptor [19]. Selinexor is studied in clinical trials also in many hematological and solid cancers [20, 21, 22, 23, 24]. The treatment with selinexor in preclinical and clinical studies resulted in nuclear localization of tumor suppressor proteins (eg p53 and FOXO3A), induced apoptosis and decreased proliferation. Selinexor reduces the expression of DNA damage repair proteins and sensitizes cancer cells to DNA damaging agents [25]. Selinexor blocks the transcription factor NF-κB and induces ribosomal stressby disruption of ribosomal subunits assembly [26].

Figure 2.

Chemical structure of selinexor (alternative names: ATG-010, KPT-330, ONO7705, XPOVIO, CRM1 nuclear export inhibitor). Chemical name: (Z)-3-(3-(3,5-bis(trifluoromethyl)phenyl-1H-1,2,4-triazol-1-yl)-N´-(pyrazin-2-yl)acrylohydrazide.

Selinexor is orally bioavailable with a mean half-life 6–8 h after a single dose. Selinexor pharmacokinetics are not significantly affected by age, sex, ethnicity, renal impairment or mild hepatic impairment. Most frequent adverse events associated with selinexor treatment are thrombocytopenia, fatigue, nausea, anemia, decreased appetite, decreased weight, diarrhoea, vomiting, hyponatremia, neutropenia, leukopenia, constipation, dyspnoea, and upper respiratory tract infection.

Advertisement

3. The phase II STORM trial with selinexor plus low-dose dexamethasone in patients with multiple myeloma pretreated with bortezomib, carfilzomib, lenalidomide, pomalidomide, daratumumab, and an alkylating agent

Selinexor demonstrated small single-agent activity with an overall response rate achieved in 4% (2/57 heavily pre-treated patients with RRMM (about six prior therapies)) [27]. The response was considerably increased from 4 to 50% (6/12) when selinexor was combined with dexamethasone in a phase I trial in patients with advanced hematological malignancies (NCT 01607892) [27]. The phase II STORM trial (NCT02336815) with selinexor and dexamethasone combination in heavily pre-treated patients with RRMM had relatively quick responses. The primary endpoint was overall response. Patients were given twice weekly oral doses of selinexor (80 mg) and dexamethasone (20 mg) in 28-day cycles [28, 29]. An overall response was recorded in 21% patients (16/78) or 26% (32/122) and median duration of response was 5 months. Patients required a lot of supportive care to mange many side effects. The most common adverse event was thrombocytopenia.

Advertisement

4. A multicenter, open-label, phase 1b/2, dose escalation trial STOMP in patients with relapsed or refractory multiple myeloma with a median of three prior therapies

The STOMP trial (NCT02343042) is a five arms study of selinexor, dexamethasone and either lenalidomide, pomalidomide, bortezomib, carfilzomib or daratumumab for the treatment of relapsed or refractory multiple myeloma with median of three prior therapies in order to evaluate the safety, tolerability and efficacy of these combinations, determining the maximum tolerated dose, the recommended phase 2 dose, overall response rate (ORR), and progression-free survival (PFS) [23, 24, 26]. Individual arms were described in abstracts No. 726, 1366, and 1393 on ASH 2020 meeting.

Advertisement

5. The randomized open-label, phase III international BOSTON trial in patients with relapsed or refractory multiple myeloma with a median of two prior therapies

The combination of selinexor and bortezomib once per week plus dexamethasone twice per week (SVd) was compared with bortezomib twice per week in combination with dexamethasone four times per week for the first six months and one half of this dose thereafter (Vd) [30]. Median PFS was longer with selinexor treatment: 13.93 months versus 9.46 months in the Vd group. The improvements in survival and response rates with selinexor were associated with higher rates of adverse events [30].

This work was supported by the project for conceptual development of research organisation No 00023736 (Institute of Hematology and Blood Transfusion) from the Ministry of Health of the Czech Republic.

References

  1. 1. Turner JG, Sullivan DM. CRM1 mediated nuclear export of proteins and drug resistance in cancer. Current Medicinal Chemistry. 2008; 15(26): 2648-2655. DOI: 10.2174/092986708786242859
  2. 2. Nguen KT, Holloway MP, Altura RA. The CRM1 nuclear export protein in normal development and disease. International Journal of Biochemistry and Molecular Biology. 2012; 3(2): 137-151. PMID: 22773955/ISSN: 2152-4114/ IJBM 1204001
  3. 3. Turner JG, Dawson J, Cubitt CL, Baz R, Sullivan DM. Inhibition of CRM1-dependent nuclear export sensitizes malignant cells to cytotoxic and targeted agents. Seminars in Cance Biology. 2014; 27: 62-73. DOI: 10.1016/j.semcancer.2014.03.001
  4. 4. Gravina GL, Senapedis W, Mc Cauley D, Baloglu E, Shacham S, Festuccia C. Nucleo-cytoplasmic transport as a therapeutic target of cancer. Journal of Hematology & Oncology. 2014; 7: 85. DOI: 10.1186/s13045-014-0085-1
  5. 5. Lu C, Figueroa JA, Liu Z, Konala V, Aulakh A, Verma R, et al. Nuclear export as a novel therapeutic target: the CRM1 connection. Current Cancer Drug Targets. 2015; 15(7): 575-592. DOI: 10.2174/156800961507150828223554
  6. 6. Monecke T, Güttler T, Neumann P, Dickmanns A, Görlich D, Ficner R. Crystal structure of the nuclear export receptor CRM1 in complex with Snurportin1 and RanGTP. Science. 2009; 324(5930): 1087-1091. DOI: 10.1126/science.1173388
  7. 7. Monecke T, Dickmanns A, Ficner R. Allosteric control of the exportin CRM1 unraveled by crystal structure analysis. FEBS Journal. 2014; 281(18): 4179-4194. DOI: 10.1111/febs.12842
  8. 8. Fung HY, Chook YM. Atomic basis of CRM1-cargo recognition, release and inhibition. Seminars in Cancer Biology. 2014; 27: 52-61. DOI: 10.1016/semcancer.2014.03.002
  9. 9. Volpon L, Culjkovic-Kraljacic B, Sohn HS, Blanchet-Cohen A, Osborne MJ, Borden KLB. A biochemical framework for eIF4E-dependent mRNA export and nuclear recycling of the export machinery. RNA. 2017; 23(6): 927-937. DOI: 10.1261/rna.060137.116
  10. 10. 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; 27(12): 2357-2365. DOI: 10.1038/leu.2013.172
  11. 11. Muqbil I, Azmi AS, Mohammad RM. Nuclear export inhibition for pancreatic cancer therapy. Cancers (Basel). 2018; 10(5): 138. DOI: 103390/cancers10050138
  12. 12. Chanukuppa V, Paul D, Taunk K, Chatterjee T, Sharma S, Kumar S, et al. XPO1 is a critical player for bortezomib resistance in multiple myeloma: A quantitative proteomic approach. Journal of Proteomics. 2019; 209: 103504. DOI: 10.1016/j.prot.2019.103504
  13. 13. Taylor J, Sendino M, Gorelick AN, Pastore A, Chang MT, Penson AV, et al. Altered nuclear export signal recognition as a driver of oncogenesis. Cancer Discovery. 2019; 9(10): 1452-1467. DOI: 10.1158/2159-8290.CD-19-0298
  14. 14. Kudo N, Wolff B, Sekimoto T, Schreiner EP, Yoneda Y, Yanagida M, et al. Leptomycin B inhibition of signal-mediated nuclear export by direct binding to CRM1. Experimental Cell Research. 1998; 242(2): 540-547. DOI: 10.1006/excr.1998.4136
  15. 15. Parikh K, Cang S, Sekhri A, Liu D. Selective inhibitors of nuclear export (SINE)-a novel class of anti-cancer agents. Journal of Hematology & Oncology. 2014; 7: 78. DOI: 10.1186/s13045-014-0078-0
  16. 16. Gandhi UH, Senapedis W, Baloglu E, Unger TJ, Chari A, Vogl D, Cornell RF. Clinical implications of targeting XPO1-mediated nuclear export in multiple myeloma. Clinical Lymphoma, Myeloma & Leukemia. 2018; 18(5): 335-345. DOI: 10.1016/j.clml.2018.03.003
  17. 17. Allegra A, Innao V, Allegra AG, Leanza R, Musolino C. Selective inhibitors of nuclear export in the treatment of hematologic malignancies. Clinical Lymphoma, Myeloma & Leukemia. 2019; 19(11): 689-698. DOI: 10.1016/j.clml.2019.08.011
  18. 18. Nachmias B, Schimmer AD. Targeting nuclear import and export in hematological malignancies. Leukemia. 2020; 34(11): 2875-2886. DOI: 10.1038/s41375-020-0958-y
  19. 19. Argueta C, Kashyap T, Klebanov B, Unger TJ, Guo C, Harrington S, et al. Selinexor synergizes with dexamethasone to repress mTORC1 signaling and induce multiple myeloma cell death. Oncotarget. 2018; 9(39): 25529-25544. DOI: 10.18632/oncotarget.25368
  20. 20. Syed YY. Selinexor: first global approval. Drugs. 2019; 79(13): 1485-1494. DOI: 10.1007/s40265-019-01188-9
  21. 21. Peterson TJ, Orozco J, Buege M. Selinexor: A first-in-class nuclear export inhibitor for management of multiply relapsed multiple myeloma. Annals of Pharmacotherapy. 2020; 54(6): 577-582. DOI: 10.1177/1060028019892643
  22. 22. Podar K, Shah J, Chari A, Richardson PG, Jagannath S. Selinexor for the treatment of multiple myeloma. Expert Oinion on Pharmacotherapy. 2020; 21(4): 398-408. DOI: 10.1080/14656566.2019.1707184
  23. 23. Richter J, Madduri D, Richard S, Chari A. Selinexor in relapsed/refractory multiple myeloma. Therapeutic Advances in Hematology. 2020; 11: 1-10. DOI: 10.1177/2040620720930629
  24. 24. Benkova K, Mihalyova J, Hajek R, Jelinek T. Selinexor, selective inhibitor of nuclear export: unselective bullet for blood cancers. Blood Reviews. 2020; on line ahead of print Sep 15; 100758. DOI: 10.1016/j.blre.2020.100758
  25. 25. Kashyap T, Argueta C, Unger TJ, Klebanov B, Debler S, Senapedis W, et al. Selinexor reduces the expression of DNA damage repair proteins and sensitizes cancer cells to DNA damaging agents. Oncotarget. 2018; 9(56): 30773-30786. DOI: 10.18632/oncotarget.25637
  26. 26. Bahlis NJ, Sutherland H, White D, Sebag M, Lentzsch S, Kotb R, et al. Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. Blood. 2018; 132(24): 2546-2554. DOI: 10.1182/blood-2018-06-858852
  27. 27. Chen C, Siegel D, Guttierrez M, Jacoby M, Hofmeister CC, Gabrail N, et al. Safety and efficacy of selinexor in relapsed or refractory multiple myeloma and Waldenstrom macroglobulinemia. Blood. 2018; 131(8): 855-863. DOI: 10.1182/blood-2017-08-797886
  28. 28. Vogl DT, Dingli D, Cornell RF, Huff CA, Jagannath S, Bhutani D, et al. Selective inhibition of nuclear export with oral selinexor for treatment of relapsed or refractory multiple myeloma. Journal of Clinical Oncology. 2018; 36(9): 859-866. DOI: 10.1200/JCO.2017.75.5207
  29. 29. Chari A, Vogl DT, Gavriatopoulou M, Naoka AK, Yee AJ, Huff CA, et al. Oral selinexor-dexamethasone for triple class refractory multiple myeloma. The New England Journal of Medicine 2019; 381(8): 727-738. DOI: 10.1056/NEJMoa1903455
  30. 30. Grosicki S, Simonova M, Spicka I, Pour L, Kriachok I, Gavriatopoulou M, et al. Once-per-week selinexor, bortezomib, and dexamethasone versus twice-per-week bortezomib dexamethasone in patients with multiple myeloma (BOSTON): a randomised, open-label, phase 3 trial. Lancet. 2020; 396(10262): 1563-1573. DOI: 10.1016/S0140-6736(20)32292-3

Written By

Ota Fuchs

Published: 28 July 2021