Selinexor, bortezomib, and dexamethasone versus bortezomib and dexamethasone in previously treated multiple myeloma: Outcomes by cytogenetic risk

Shambavi Richard, Ajai Chari, Sosana Delimpasi, Maryana Simonova, Ivan Spicka, Ludek Pour, Iryna Kriachok, Meletios A Dimopoulos, Halyna Pylypenko, Holger W Auner, Xavier Leleu, Ganna Usenko, Roman Hajek, Reuben Benjamin, Tuphan Kanti Dolai, Dinesh Kumar Sinha, Christopher P Venner, Mamta Garg, Don Ambrose Stevens, Hang Quach, Sundar Jagannath, Phillipe Moreau, Moshe Levy, Ashraf Badros, Larry D Anderson Jr, Nizar J Bahlis, Thierry Facon, Maria Victoria Mateos, Michele Cavo, Hua Chang, Yosef Landesman, Yi Chai, Melina Arazy, Jatin Shah, Sharon Shacham, Michael G Kauffman, Sebastian Grosicki, Paul G Richardson, Shambavi Richard, Ajai Chari, Sosana Delimpasi, Maryana Simonova, Ivan Spicka, Ludek Pour, Iryna Kriachok, Meletios A Dimopoulos, Halyna Pylypenko, Holger W Auner, Xavier Leleu, Ganna Usenko, Roman Hajek, Reuben Benjamin, Tuphan Kanti Dolai, Dinesh Kumar Sinha, Christopher P Venner, Mamta Garg, Don Ambrose Stevens, Hang Quach, Sundar Jagannath, Phillipe Moreau, Moshe Levy, Ashraf Badros, Larry D Anderson Jr, Nizar J Bahlis, Thierry Facon, Maria Victoria Mateos, Michele Cavo, Hua Chang, Yosef Landesman, Yi Chai, Melina Arazy, Jatin Shah, Sharon Shacham, Michael G Kauffman, Sebastian Grosicki, Paul G Richardson

Abstract

In the phase 3 BOSTON study, patients with multiple myeloma (MM) after 1-3 prior regimens were randomized to once-weekly selinexor (an oral inhibitor of exportin 1 [XPO1]) plus bortezomib-dexamethasone (XVd) or twice-weekly bortezomib-dexamethasone (Vd). Compared with Vd, XVd was associated with significant improvements in median progression-free survival (PFS), overall response rate (ORR), and lower rates of peripheral neuropathy, with trends in overall survival (OS) favoring XVd. In BOSTON, 141 (35.1%) patients had MM with high-risk (presence of del[17p], t[4;14], t[14;16], or ≥4 copies of amp1q21) cytogenetics (XVd, n = 70; Vd, n = 71), and 261 (64.9%) exhibited standard-risk cytogenetics (XVd, n = 125; Vd, n = 136). Among patients with high-risk MM, median PFS was 12.91 months for XVd and 8.61 months for Vd (HR, 0.73 [95% CI, (0.4673, 1.1406)], p = 0.082), and ORRs were 78.6% and 57.7%, respectively (OR 2.68; p = 0.004). In the standard-risk subgroup, median PFS was 16.62 months for XVd and 9.46 months for Vd (HR 0.61; p = 0.004), and ORRs were 75.2% and 64.7%, respectively (OR 1.65; p = 0.033). The safety profiles of XVd and Vd in both subgroups were consistent with the overall population. These data suggest that selinexor can confer benefits to patients with MM regardless of cytogenetic risk. ClinicalTrials.gov identifier: NCT03110562.

Conflict of interest statement

Ajai Chari reports grants and personal fees from Janssen, Celgene, Novartis, Amgen, Seattle Genetics, and Millenium/Takeda; personal fees from Bristol Myers Squibb, Karyopharm, Sanofi, Oncopeptides, Antengene, Glaxo Smith Kline, Secura Bio, and Shattuch Labs.

Ivan Spicka reports personal fees from Janssen‐Cilag, Takeda, Sanofi Aventis and Novartis; personal fees and non‐financial support from Colgene, BMS and Amgen.

Iryna Kriachok reports a consulting role, an advisory role, and a speaker's bureau role for Takeda, Janssen, Roche, Abbvie and MSD; Travel support by Takeda, MSD, Roche, Abbvie and Janssen.

Holger W. Auner reports an advisory role for Takeda and Karyopharm; grant from Amgen; and a speaker's bureau role for Janssen.

Roman Hajek has had a consultant or advisory relationship with Janssen, Amgen, Celgene, AbbVie, BMS, Novartis, PharmaMar, and Takeda; has received honoraria from Janssen, Amgen, Celgene, BMS, PharmaMar, and Takeda; has received research funding from Janssen, Amgen, Celgene, BMS, Novartis, and Takeda.

Christopher P. Venner has received honoraria from BMS/Celgene, Janssen, Sanofi, Amgen, GSK, and Takeda.

Mamta Garg reports support for attending conferences from Takeda; an advisory role for Amgen, Takeda, Jansen, Novartis and Celgene; and a speaker's bureau role for Janssen.

Hang Quach reports grants from and an advisory board role for Amgen, Celgene, Karyopharm, GlaxoSmithKline; non‐financial support and research drug supply from Sanofi; an advisory board role for Janssen Cilag and Specialized therapeutics.

Sundar Jagannath reports consulting services for AbbVie, Bristol‐Myers Squibb, Janssen Pharmaceuticals, Merck & Co. PM reports personal fees from Celgene, Amgen, Takeda, Janssen and Abbvie.

Moshe Levy reports receiving consulting fees and lecture fees from Takeda, Celgene, Seattle Genetics, AbbVie, Jazz Pharmaceuticals, Gilead Sciences, Bristol‐Myers Squibb, Amgen, Spectrum Pharmaceuticals, and Janssen.

Larry D. Anderson, Jr. reports honoraria from advisory board activity from the following: GSK, Amgen, Janssen, BMS/Celgene, Karyopharm, and Oncopeptides.

Nizar J. Bahlis reports grants and personal fees from Celgene; personal fees from Janssen, Amgen, Takeda, Abbvie, GSK and Karyopharm.

Thierry Facon reports an advisory board role for Karyopharm, Amgen, Roche and Oncopeptides; an advisory board role and a speaker bureau role for Janssen, Celgene/BMS, and Takeda.

Maria Victoria Mateos has served as member of advisory boards or received honoraria from Janssen, BMS‐Celgene, Takeda, Amgen, Sanofi, Oncopeptides, GSK, Adaptive, Pfizer, Regeneron, Roche and Sea‐Gen.

Hua Chang, Yosef Landesman, Yi Chai, Melina Arazy, Jatin Shah and Michael G. Kauffman are salaried employees and stockholders of Karyopharm Therapeutics Inc.

Sharon Shacham reports being employed by and owning stock in Karyopharm Therapeutics, holding patents (8999996, 9079865, 9714226, PCT/US12/048319, and I574957) on hydrazide‐containing nuclear transport modulators and uses, and holding pending patents (PCT/US12/048319, 499/2012, PI20102724, and 2012000928) on hydrazide‐containing nuclear transport modulators and uses.

Paul G. Richardson reports receiving grant support and honoraria from Oncopeptides, Celgene, and Takeda, grant support from Bristol‐Myers Squibb, and honoraria from Amgen, Janssen, and Karyopharm Therapeutics.

© 2021 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.

Figures

FIGURE 1
FIGURE 1
Progression‐free survival. Patients with A, high‐risk and B, standard‐risk cytogenetics. CI, confidence interval; HR, hazard ratio; PFS, progression‐free survival; Vd, bortezomib and dexamethasone; XVd, selinexor, bortezomib, and dexamethasone [Color figure can be viewed at wileyonlinelibrary.com]

References

    1. Hanamura I, Stewart JP, Huang Y, et al. Frequent gain of chromosome band 1q21 in plasma‐cell dyscrasias detected by fluorescence in situ hybridization: incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem‐cell transplantation. Blood. 2006;108:1724‐1732.
    1. Fonseca R, Bergsagel PL, Drach J, et al. International myeloma working group molecular classification of multiple myeloma: spotlight review. Leukemia. 2009;23:2210‐2221.
    1. Sonneveld P, Avet‐Loiseau H, Lonial S, et al. Treatment of multiple myeloma with high‐risk cytogenetics: a consensus of the International Myeloma Working Group. Blood. 2016;127:2955‐2962.
    1. Shah V, Sherborne AL, Walker BA, et al. Prediction of outcome in newly diagnosed myeloma: a meta‐analysis of the molecular profiles of 1905 trial patients. Leukemia. 2018;32:102‐110.
    1. Avet‐Loiseau H, Durie BGM, Cavo M, et al. Combining fluorescent in situ hybridization data with ISS staging improves risk assessment in myeloma: An International Myeloma Working Group collaborative project. Leukemia. 2013;27:711‐717.
    1. Gandhi UH, Senapedis W, Baloglu E, et al. Clinical implications of targeting XPO1‐mediated nuclear export in multiple myeloma. Clin Lymphoma Myeloma Leuk. 2018;18:335‐380.
    1. Chari A, Vogl DT, Gavriatopoulou M, et al. Oral selinexor–dexamethasone for triple‐class refractory multiple myeloma. N Engl J Med. 2019;381:727‐738.
    1. Azizian NG, Azizian NG, Li Y, Li Y. XPO1‐dependent nuclear export as a target for cancer therapy. J Hematol Oncol. 2020;13:1‐9.
    1. Gravina GL, Senapedis W, McCauley D, Baloglu E, Shacham S, Festuccia C. Nucleo‐cytoplasmic transport as a therapeutic target of cancer. J Hematol Oncol. 2014;7:85. 10.1186/s13045-014-0085-1
    1. 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. 2014;28:155‐165.
    1. Chanukuppa V, Paul D, Taunk K, et al. XPO1 is a critical player for bortezomib resistance in multiple myeloma: a quantitative proteomic approach. J Proteomics. 2019;209:103504.
    1. Bhutani M, Zhang Q, Friend R, et al. Investigation of a gene signature to predict response to immunomodulatory derivatives for patients with multiple myeloma: an exploratory, retrospective study using microarray datasets from prospective clinical trials. Lancet Haematol. 2017;4:e443‐e451.
    1. Culjkovic‐Kraljacic B, Baguet A, Volpon L, Amri A, Borden KLB. The oncogene eIF4E reprograms the nuclear pore complex to promote mRNA export and oncogenic transformation. Cell Rep. 2012;2:207‐215.
    1. Jardin F, Pujals A, Pelletier L, et al. Recurrent mutations of the exportin 1 gene (XPO1) and their impact on selective inhibitor of nuclear export compounds sensitivity in primary mediastinal B‐cell lymphoma. Am J Hematol. 2016;91:923‐930.
    1. 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. 2012;45:222‐232.
    1. Schmidt J, Braggio E, Kortuem 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. 2013;27:2357‐2365.
    1. Turner JG, Kashyap T, Dawson JL, et al. XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IκBα and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget. 2016;7:78896‐78909.
    1. Bahlis NJ, Sutherland H, White D, et al. Selinexor plus low‐dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. Blood. 2018;132:2546‐2554.
    1. Grosicki S, Simonova M, Picka I, et al. Once‐weekly selinexor, bortezomib, and dexamethasone versus twice‐weekly bortezomib and dexamethasone in patients with multiple myeloma (BOSTON): a randomised, open‐label phase 3 trial. Lancet. 2020;396:1563‐1573.
    1. XPOVIO® (selinexor) , package insert. 2020. . Accessed February 1, 2021.
    1. Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016;17:e328‐e346.
    1. Fonseca R, Oken MM, Greipp PR. The t(4;14)(p16.3;q32) is strongly associated with chromosome 13 abnormalities in both multiple myeloma and monoclonal gammopathy of undetermined significance. Blood. 2001;98:1271‐1272.
    1. Fonseca R, Barlogie B, Bataille R, et al. Genetics and cytogenetics of multiple myeloma. Cancer Research. 2004;64:1546‐1558.
    1. An G, Li Z, Tai YT, et al. The impact of clone size on the prognostic value of chromosome aberrations by fluorescence in situ hybridization in multiple myeloma. Clin Cancer Res. 2015;21:2148‐2156.
    1. Hanamura I. Gain/amplification of chromosome arm 1q21 in multiple myeloma. Cancers (Basel). 2021;13:1‐16.
    1. Mateos M, Gavriatopoulou M, Facon T, et al. Effect of prior treatments on selinexor, bortezomib, and dexamethasone in previously treated multiple myeloma. J Hematol Oncol. 2021;14(1):1‐5.
    1. Weisel K, Spencer A, Lentzsch S, et al. Daratumumab, bortezomib, and dexamethasone in relapsed or refractory multiple myeloma: subgroup analysis of CASTOR based on cytogenetic risk. J Hematol Oncol. 2020;13:115. 10.1186/s13045-020-00948-5
    1. An G, Xu Y, Shi L, et al. Chromosome 1q21 gains confer inferior outcomes in multiple myeloma treated with bortezomib but copy number variation and percentage of plasma cells involved have no additional prognostic value. Haematologica. 2014;99:353‐359.
    1. Locher M, Steurer M, Jukic E, et al. The prognostic value of additional copies of 1q21 in multiple myeloma depends on the primary genetic event. Am J Hematol. 2020;95:1562‐1571.
    1. Richardson PG, Oriol A, Beksac M, et al. Pomalidomide, bortezomib, and dexamethasone for patients with relapsed or refractory multiple myeloma previously treated with lenalidomide (OPTIMISMM): a randomised, open‐label, phase 3 trial. Lancet Oncol. 2019;20:781‐794.
    1. 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. 2016;7:78883‐78895.
    1. Argueta C, Kashyap T, Klebanov B, et al. Selinexor synergizes with dexamethasone to repress mTORC1 signaling and induce multiple myeloma cell death. Oncotarget. 2018;9:25529‐25544.
    1. 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. 2016;9:73.
    1. Liu Y, Chen C, Xu Z, et al. Deletions linked to TP53 loss drive cancer through p53‐independent mechanisms. Nature. 2016;531:471‐475.
    1. Nakayama R, Zhang YX, Czaplinski JT, et al. Preclinical activity of selinexor, an inhibitor of XPO1, in sarcoma. Oncotarget. 2016;7:16581‐16592.
    1. Kalff A, Spencer A. The t(4;14) translocation and FGFR3 overexpression in multiple myeloma: prognostic implications and current clinical strategies. Blood Cancer J. 2012;2:e89‐e89.
    1. Senapedis WT, Baloglu E, Landesman Y. Clinical translation of nuclear export inhibitors in cancer. Semin Cancer Biol. 2014;27:74‐86.
    1. Cao S, Liu J, Song L, Ma X. The protooncogene c‐Maf is an essential transcription factor for IL‐10 gene expression in macrophages. J Immunol. 2005;174:3484‐3492.
    1. Wang AY, Liu H. The past, present, and future of CRM1/XPO1 inhibitors. Stem Cell Investig. 2019;6:6. 10.21037/sci.2019.02.03
    1. Annunziata CM, Hernandez L, Davis RE, et al. A mechanistic rationale for MEK inhibitor therapy in myeloma based on blockade of MAF oncogene expression. Blood. 2011;117:2396‐2404.

Source: PubMed

3
Tilaa