CAR: chimeric antigen receptor; DLBCL: diffuse large B cell lymphoma; RR: relapsed and refractory; BCMA: B cell maturation antigen; GCB: germinal center B-cell; NHL: non-Hodgkin lymphoma. *Trial status provided is according to information on clinicaltrials.gov as of (Date—01/23/2025); respective trial sponsor should be contacted for further clarification/information
TK and CJW are employees and stockholders of Karyopharm Therapeutics. All the other authors declare no conflicts of interest.
Ethical approval
Not applicable.
Consent to participate
Not applicable.
Consent to publication
Not applicable.
Availability of data and materials
Not applicable.
Funding
This work was supported by Karyopharm Therapeutics and the John Goldman Fellowship Follow-up Award from Leukaemia UK (to MDB). Karyopharm Therapeutics reviewed the manuscript for scientific accuracy prior to submission but had no role in study design, data collection, analysis, interpretation, or writing of the manuscript; the other provider of funds had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.
References
Yang Y, Guo L, Chen L, Gong B, Jia D, Sun Q. Nuclear transport proteins: structure, function, and disease relevance.Signal Transduct Target Ther. 2023;8:425. [DOI] [PubMed] [PMC]
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation.Cell. 2011;144:646–74. [DOI] [PubMed]
Sun Q, Chen X, Zhou Q, Burstein E, Yang S, Jia D. Inhibiting cancer cell hallmark features through nuclear export inhibition.Signal Transduct Target Ther. 2016;1:16010. [DOI] [PubMed] [PMC]
Kashyap T, Argueta C, Unger T, 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;9:30773–86. [DOI] [PubMed] [PMC]
Stommel JM, Marchenko ND, Jimenez GS, Moll UM, Hope TJ, Wahl GM. A leucine-rich nuclear export signal in the p53 tetramerization domain: regulation of subcellular localization and p53 activity by NES masking.EMBO J. 1999;18:1660–72. [DOI] [PubMed] [PMC]
Biggs WH 3rd, Meisenhelder J, Hunter T, Cavenee WK, Arden KC. Protein kinase B/Akt-mediated phosphorylation promotes nuclear exclusion of the winged helix transcription factor FKHR1.Proc Natl Acad Sci U S A. 1999;96:7421–6. [DOI] [PubMed] [PMC]
Yoshimura M, Ishizawa J, Ruvolo V, Dilip A, Quintás-Cardama A, McDonnell TJ, et al. Induction of p53-mediated transcription and apoptosis by exportin-1 (XPO1) inhibition in mantle cell lymphoma.Cancer Sci. 2014;105:795–801. [DOI] [PubMed] [PMC]
Marullo R, Rutherford SC, Revuelta MV, Zamponi N, Culjkovic-Kraljacic B, Kotlov N, et al. XPO1 Enables Adaptive Regulation of mRNA Export Required for Genotoxic Stress Tolerance in Cancer Cells.Cancer Res. 2024;84:101–17. [DOI] [PubMed] [PMC]
Alt JR, Cleveland JL, Hannink M, Diehl JA. Phosphorylation-dependent regulation of cyclin D1 nuclear export and cyclin D1-dependent cellular transformation.Genes Dev. 2000;14:3102–14. [DOI] [PubMed] [PMC]
Jiao W, Datta J, Lin HM, Dundr M, Rane SG. Nucleocytoplasmic shuttling of the retinoblastoma tumor suppressor protein via Cdk phosphorylation-dependent nuclear export.J Biol Chem. 2006;281:38098–108. [DOI] [PubMed]
Thomas F, Kutay U. Biogenesis and nuclear export of ribosomal subunits in higher eukaryotes depend on the CRM1 export pathway.J Cell Sci. 2003;116:2409–19. [DOI] [PubMed]
Tabe Y, Kojima K, Yamamoto S, Sekihara K, Matsushita H, Davis RE, 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] [PubMed] [PMC]
Bai B, Moore HM, Laiho M. CRM1 and its ribosome export adaptor NMD3 localize to the nucleolus and affect rRNA synthesis.Nucleus. 2013;4:315–25. [DOI] [PubMed] [PMC]
Azizian NG, Li Y. XPO1-dependent nuclear export as a target for cancer therapy.J Hematol Oncol. 2020;13:61. [DOI] [PubMed] [PMC]
García-Santisteban I, Arregi I, Alonso-Mariño M, Urbaneja MA, Garcia-Vallejo JJ, Bañuelos S, et al. A cellular reporter to evaluate CRM1 nuclear export activity: functional analysis of the cancer-related mutant E571K.Cell Mol Life Sci. 2016;73:4685–99. [DOI] [PubMed] [PMC]
Miloudi H, Bohers É, Guillonneau F, Taly A, Gibouin VC, Viailly PJ, et al. XPO1E571K Mutation Modifies Exportin 1 Localisation and Interactome in B-cell Lymphoma.Cancers (Basel). 2020;12:2829. [DOI] [PubMed] [PMC]
Ishizawa J, Kojima K, Hail N Jr, Tabe Y, Andreeff M. Expression, function, and targeting of the nuclear exporter chromosome region maintenance 1 (CRM1) protein.Pharmacol Ther. 2015;153:25–35. [DOI] [PubMed] [PMC]
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;96:9112–7. [DOI] [PubMed] [PMC]
Komiyama K, Okada K, Tomisaka S, Umezawa I, Hamamoto T, Beppu T. Antitumor activity of leptomycin B.J Antibiot (Tokyo). 1985;38:427–9. [DOI] [PubMed]
Newlands ES, Rustin GJ, Brampton MH. Phase I trial of elactocin.Br J Cancer. 1996;74:648–9. [DOI] [PubMed] [PMC]
Mutka SC, Yang WQ, Dong SD, Ward SL, Craig DA, Timmermans PB, et al. Identification of nuclear export inhibitors with potent anticancer activity in vivo.Cancer Res. 2009;69:510–7. [DOI] [PubMed] [PMC]
Sun Q, Carrasco YP, Hu Y, Guo X, Mirzaei H, Macmillan J, et al. Nuclear export inhibition through covalent conjugation and hydrolysis of Leptomycin B by CRM1.Proc Natl Acad Sci U S A. 2013;110:1303–8. [DOI] [PubMed] [PMC]
Wang AY, Liu H. The past, present, and future of CRM1/XPO1 inhibitors.Stem Cell Investig. 2019;6:6. [DOI] [PubMed] [PMC]
Lee Y, Pei J, Baumhardt JM, Chook YM, Grishin NV. Structural prerequisites for CRM1-dependent nuclear export signaling peptides: accessibility, adapting conformation, and the stability at the binding site.Sci Rep. 2019;9:6627. [DOI] [PubMed] [PMC]
Schlicher L, Green LG, Romagnani A, Renner F. Small molecule inhibitors for cancer immunotherapy and associated biomarkers - the current status.Front Immunol. 2023;14:1297175. [DOI] [PubMed] [PMC]
Galluzzi L, Humeau J, Buqué A, Zitvogel L, Kroemer G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors.Nat Rev Clin Oncol. 2020;17:725–41. [DOI] [PubMed]
Afolabi LO, Bi J, Li X, Adeshakin AO, Adeshakin FO, Wu H, et al. Synergistic Tumor Cytolysis by NK Cells in Combination With a Pan-HDAC Inhibitor, Panobinostat.Front Immunol. 2021;12:701671. [DOI] [PubMed] [PMC]
Shimizu R, Kikuchi J, Wada T, Ozawa K, Kano Y, Furukawa Y. HDAC inhibitors augment cytotoxic activity of rituximab by upregulating CD20 expression on lymphoma cells.Leukemia. 2010;24:1760–8. [DOI] [PubMed]
Bobrowicz M, Dwojak M, Pyrzynska B, Stachura J, Muchowicz A, Berthel E, et al. HDAC6 inhibition upregulates CD20 levels and increases the efficacy of anti-CD20 monoclonal antibodies.Blood. 2017;130:1628–38. [DOI] [PubMed]
Chang MC, Cheng HI, Hsu K, Hsu YN, Kao CW, Chang YF, et al. NKG2A Down-Regulation by Dasatinib Enhances Natural Killer Cytotoxicity and Accelerates Effective Treatment Responses in Patients With Chronic Myeloid Leukemia.Front Immunol. 2019;9:3152. [DOI] [PubMed] [PMC]
Larmonier N, Janikashvili N, LaCasse CJ, Larmonier CB, Cantrell J, Situ E, et al. Imatinib mesylate inhibits CD4+ CD25+ regulatory T cell activity and enhances active immunotherapy against BCR-ABL- tumors.J Immunol. 2008;181:6955–63. [DOI] [PubMed] [PMC]
Liang S, Tran E, Du X, Dong J, Sudholz H, Chen H, et al. A small molecule inhibitor of PTP1B and PTPN2 enhances T cell anti-tumor immunity.Nat Commun. 2023;14:4524. [DOI] [PubMed] [PMC]
Carlsten M, Namazi A, Reger R, Levy E, Berg M, St Hilaire C, et al. Bortezomib sensitizes multiple myeloma to NK cells via ER-stress-induced suppression of HLA-E and upregulation of DR5.Oncoimmunology. 2018;8:e1534664. [DOI] [PubMed] [PMC]
Marullo R, Revuelta MV, Zamponi N, Rutherford SC, Martin P, Inghirami G, et al. XPO1 Relieves MYC-Induced Replication Stress Limiting the Immunogenicity of DLBCL Cells.Blood. 2020;136. [DOI]
Kotlov N, Bagaev A, Revuelta MV, Phillip JM, Cacciapuoti MT, Antysheva Z, et al. Clinical and Biological Subtypes of B-cell Lymphoma Revealed by Microenvironmental Signatures.Cancer Discov. 2021;11:1468–89. [DOI] [PubMed] [PMC]
Zhao L, Luo B, Wang L, Chen W, Jiang M, Zhang N. Pan-cancer analysis reveals the roles of XPO1 in predicting prognosis and tumorigenesis.Transl Cancer Res. 2021;10:4664–79. [DOI] [PubMed] [PMC]
Li X, Wang L. EP16.01-021 Less Immune Cell Infiltration and Worse Prognosis for NSCLC Patients with XPO1 Copy Number Alterations.J Thor Oncol. 2022;17:S564–5. [DOI]
Lin DC, Hao JJ, Nagata Y, Xu L, Shang L, Meng X, et al. Genomic and molecular characterization of esophageal squamous cell carcinoma.Nat Genet. 2014;46:467–73. [DOI] [PubMed] [PMC]
Puente XS, Pinyol M, Quesada V, Conde L, Ordóñez GR, Villamor N, et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia.Nature. 2011;475:101–5. [DOI] [PubMed] [PMC]
Walker JS, Hing ZA, Harrington B, Baumhardt J, Ozer HG, Lehman A, et al. Recurrent XPO1 mutations alter pathogenesis of chronic lymphocytic leukemia.J Hematol Oncol. 2021;14:17. [DOI] [PubMed] [PMC]
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 Discov. 2019;9:1452–67. [DOI] [PubMed] [PMC]
Baumhardt JM, Walker JS, Lee Y, Shakya B, Brautigam CA, Lapalombella R, et al. Recognition of nuclear export signals by CRM1 carrying the oncogenic E571K mutation.Mol Biol Cell. 2020;31:1879–91. [DOI] [PubMed] [PMC]
Arruga F, Gyau BB, Iannello A, Vitale N, Vaisitti T, Deaglio S. Immune Response Dysfunction in Chronic Lymphocytic Leukemia: Dissecting Molecular Mechanisms and Microenvironmental Conditions.Int J Mol Sci. 2020;21:1825. [DOI] [PubMed] [PMC]
Meng W, Gao SJ. Targeting XPO1 enhances innate immune response and inhibits KSHV lytic replication during primary infection by nuclear stabilization of the p62 autophagy adaptor protein.Cell Death Dis. 2021;12:29. [DOI] [PubMed] [PMC]
Liao Y, Ke X, Deng T, Qin Q. The Second-Generation XPO1 Inhibitor Eltanexor Inhibits Human Cytomegalovirus (HCMV) Replication and Promotes Type I Interferon Response.Front Microbiol. 2021;12:675112. [DOI] [PubMed] [PMC]
Perwitasari O, Johnson S, Yan X, Howerth E, Shacham S, Landesman Y, et al. Verdinexor, a novel selective inhibitor of nuclear export, reduces influenza a virus replication in vitro and in vivo.J Virol. 2014;88:10228–43. [DOI] [PubMed] [PMC]
Kashyap T, Murray J, Walker CJ, Chang H, Tamir S, Hou B, et al. Selinexor, a novel selective inhibitor of nuclear export, reduces SARS-CoV-2 infection and protects the respiratory system in vivo.Antiviral Res. 2021;192:105115. [DOI] [PubMed] [PMC]
Ostuni R, Kratochvill F, Murray PJ, Natoli G. Macrophages and cancer: from mechanisms to therapeutic implications.Trends Immunol. 2015;36:229–39. [DOI] [PubMed]
Dehne N, Mora J, Namgaladze D, Weigert A, Brüne B. Cancer cell and macrophage cross-talk in the tumor microenvironment.Curr Opin Pharmacol. 2017;35:12–9. [DOI] [PubMed]
Wang S, Wang J, Chen Z, Luo J, Guo W, Sun L, et al. Targeting M2-like tumor-associated macrophages is a potential therapeutic approach to overcome antitumor drug resistance.NPJ Precis Oncol. 2024;8:31. [DOI] [PubMed] [PMC]
Mirlekar B. Tumor promoting roles of IL-10, TGF-β, IL-4, and IL-35: Its implications in cancer immunotherapy.SAGE Open Med. 2022;10:20503121211069012. [DOI] [PubMed] [PMC]
Rodriguez-Garcia A, Lynn RC, Poussin M, Eiva MA, Shaw LC, O’Connor RS, et al. CAR-T cell-mediated depletion of immunosuppressive tumor-associated macrophages promotes endogenous antitumor immunity and augments adoptive immunotherapy.Nat Commun. 2021;12:877. [DOI] [PubMed] [PMC]
Kady N, Wang CG, Wolfe A, Maine I, Abdelrahman S, Murga-Zamalloa CA, et al. Xpo-1 Antagonism Impairs CSF-1R Expression and Depletes Lymphoma-Associated Macrophages in T-Cell Lymphomas.Blood. 2023;142:1647. [DOI]
Jiménez I, Carabia J, Bobillo S, Palacio C, Abrisqueta P, Pagès C, et al. Repolarization of tumor infiltrating macrophages and increased survival in mouse primary CNS lymphomas after XPO1 and BTK inhibition.J Neurooncol. 2020;149:13–25. [DOI] [PubMed] [PMC]
Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH, Rüttinger D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy.J Immunother Cancer. 2017;5:53. [DOI] [PubMed] [PMC]
Uddin MH, Al-Hallak MN, Khan HY, Aboukameel A, Li Y, Bannoura SF, et al. Molecular analysis of XPO1 inhibitor and gemcitabine-nab-paclitaxel combination in KPC pancreatic cancer mouse model.Clin Transl Med. 2023;13:e1513. [DOI] [PubMed] [PMC]
Farren MR, Hennessey RC, Shakya R, Elnaggar O, Young G, Kendra K, et al. The Exportin-1 Inhibitor Selinexor Exerts Superior Antitumor Activity when Combined with T-Cell Checkpoint Inhibitors.Mol Cancer Ther. 2017;16:417–27. [DOI] [PubMed] [PMC]
Daneshmandi S, Yan Q, Choi JE, Katsuta E, MacDonald CR, Goruganthu M, et al. Exportin 1 governs the immunosuppressive functions of myeloid-derived suppressor cells in tumors through ERK1/2 nuclear export.Cell Mol Immunol. 2024;21:873–91. [DOI] [PubMed] [PMC]
Middelburg J, Ghaffari S, Schoufour TAW, Sluijter M, Schaap G, Göynük B, et al. The MHC-E peptide ligands for checkpoint CD94/NKG2A are governed by inflammatory signals, whereas LILRB1/2 receptors are peptide indifferent.Cell Rep. 2023;42:113516. [DOI] [PubMed]
Fisher JG, Walker CJ, Doyle AD, Johnson PW, Forconi F, Cragg MS, et al. Selinexor Enhances NK Cell Activation Against Malignant B Cells via Downregulation of HLA-E.Front Oncol. 2021;11:785635. [DOI] [PubMed] [PMC]
Fisher JG, Doyle ADP, Graham LV, Sonar S, Sale B, Henderson I, et al. XPO1 inhibition sensitises CLL cells to NK cell mediated cytotoxicity and overcomes HLA-E expression.Leukemia. 2023;37:2036–49. [DOI] [PubMed] [PMC]
Gavriatopoulou M, Chari A, Chen C, Bahlis N, Vogl DT, Jakubowiak A, et al. Integrated safety profile of selinexor in multiple myeloma: experience from 437 patients enrolled in clinical trials.Leukemia. 2020;34:2430–40. [DOI] [PubMed] [PMC]
Baron S, Rashal T, Vaisman D, Elhasid R, Shukrun R. Selinexor, a selective inhibitor of nuclear export, inhibits human neutrophil extracellular trap formation in vitro.Front Pharmacol. 2022;13:1030991. [DOI] [PubMed] [PMC]
Masucci MT, Minopoli M, Del Vecchio S, Carriero MV. The Emerging Role of Neutrophil Extracellular Traps (NETs) in Tumor Progression and Metastasis.Front Immunol. 2020;11:1749. [DOI] [PubMed] [PMC]
Wu M, Gui H, Feng Z, Xu H, Li G, Li M, et al. KPT-330, a potent and selective CRM1 inhibitor, exhibits anti-inflammation effects and protection against sepsis.Biochem Biophys Res Commun. 2018;503:1773–9. [DOI] [PubMed]
Ali H, Maher K, Kishtagari A, Mohan SR, Prchal JT, Chai Y, et al. Selinexor-Driven Regulation of Proinflammatory Cytokines May Lead to Stabilization of Hematologic Parameters and Bone Marrow Function in Patients with Myelofibrosis: Case Study from the Phase 1 Sentry Trial.Blood. 2024;144:6676. [DOI]
Tantravahi S, Patel A, Yap J, Walker C, Ellero A, Rets A, et al. Long-Term Response to Selinexor in Patients With Myelofibrosis and Refractory or Intolerant to JAK Inhibitors: Follow-Up Results of a Single-Center, Phase II, Investigator-Initiated Trial (IIT).Clin Lymphoma Myeloma Leuk. 2024;24:S441. [DOI]
Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L, et al. The multifaceted nature of IL-10: regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions.Front Immunol. 2023;14:1161067. [DOI] [PubMed] [PMC]
Mostafa-Hedeab G, Al-Kuraishy HM, Al-Gareeb AI, Welson NN, El-Saber Batiha G, Conte-Junior CA. Selinexor and COVID-19: The Neglected Warden.Front Pharmacol. 2022;13:884228. [DOI] [PubMed] [PMC]
Blunt MD, Khakoo SI. Harnessing natural killer cell effector function against cancer.Immunother Adv. 2023;4:ltad031. [DOI] [PubMed] [PMC]
Tyler PM, Servos MM, de Vries RC, Klebanov B, Kashyap T, Sacham S, et al. Clinical Dosing Regimen of Selinexor Maintains Normal Immune Homeostasis and T-cell Effector Function in Mice: Implications for Combination with Immunotherapy.Mol Cancer Ther. 2017;16:428–39. [DOI] [PubMed] [PMC]
Lapalombella R, Sun Q, Williams K, Tangeman L, Jha S, Zhong Y, et al. Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia.Blood. 2012;120:4621–34. [DOI] [PubMed] [PMC]
Borst L, van der Burg SH, van Hall T. The NKG2A-HLA-E Axis as a Novel Checkpoint in the Tumor Microenvironment.Clin Cancer Res. 2020;26:5549–56. [DOI] [PubMed]
Kamiya T, Seow SV, Wong D, Robinson M, Campana D. Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells.J Clin Invest. 2019;129:2094–106. [DOI] [PubMed] [PMC]
Fisher JG, Doyle ADP, Graham LV, Khakoo SI, Blunt MD. Disruption of the NKG2A:HLA-E Immune Checkpoint Axis to Enhance NK Cell Activation against Cancer.Vaccines (Basel). 2022;10:1993. [DOI] [PubMed] [PMC]
Gustafson KS, Ginder GD. Interferon-gamma induction of the human leukocyte antigen-E gene is mediated through binding of a complex containing STAT1alpha to a distinct interferon-gamma-responsive element.J Biol Chem. 1996;271:20035–46. [DOI] [PubMed]
Zheng H, Guan X, Meng X, Tong Y, Wang Y, Xie S, et al. IFN-γ in ovarian tumor microenvironment upregulates HLA-E expression and predicts a poor prognosis.J Ovarian Res. 2023;16:229. [DOI] [PubMed] [PMC]
Bowles JA, Wang SY, Link BK, Allan B, Beuerlein G, Campbell MA, et al. Anti-CD20 monoclonal antibody with enhanced affinity for CD16 activates NK cells at lower concentrations and more effectively than rituximab.Blood. 2006;108:2648–54. [DOI] [PubMed] [PMC]
Kohrt HE, Sagiv-Barfi I, Rafiq S, Herman SE, Butchar JP, Cheney C, et al. Ibrutinib antagonizes rituximab-dependent NK cell-mediated cytotoxicity.Blood. 2014;123:1957–60. [DOI] [PubMed] [PMC]
Marin D, Li Y, Basar R, Rafei H, Daher M, Dou J, et al. Safety, efficacy and determinants of response of allogeneic CD19-specific CAR-NK cells in CD19+ B cell tumors: a phase 1/2 trial.Nat Med. 2024;30:772–84. [DOI] [PubMed] [PMC]
André P, Denis C, Soulas C, Bourbon-Caillet C, Lopez J, Arnoux T, et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells.Cell. 2018;175:1731–43.e13. [DOI] [PubMed] [PMC]
Kaulfuss M, Mietz J, Fabri A, Vom Berg J, Münz C, Chijioke O. The NK cell checkpoint NKG2A maintains expansion capacity of human NK cells.Sci Rep. 2023;13:10555. [DOI] [PubMed] [PMC]
Encinas J, García-Ortiz A, Maroto-Martín E, Castellano E, Oliva R, Alonso Fernández R, et al. S256: HLA-E/NKG2A CHECKPOINT DRIVES MULTIPLE MYELOMA RESISTANCE TO CAR-NK THERAPY.HemaSphere. 2023;7:e16745e9.
Croom-Perez TJ, Robles-Carrillo LD, Dieffenthaller TA, Copik AJ. Abstract 2819: Suppression of NKG2A mediated inhibition in ex vivo expanded natural killer cells increases their cytotoxicity.Cancer Res. 2022;82:2819–9. [DOI]
Kang Y, Neff J, Gasparetto C, Wang X, Ellero A, Walker C. P-396 Investigation of T-cell fitness and mechanisms of drug resistance in selinexor treated patients with relapsed/refractory multiple myeloma.Clin Lymphoma Myeloma Leuk. 2023;23:S259.
Binder AF, Walker CJ, Mark TM, Baljevic M. Impacting T-cell fitness in multiple myeloma: potential roles for selinexor and XPO1 inhibitors.Front Immunol. 2023;14:1275329. [DOI] [PubMed] [PMC]
Stiff PJ, Mehrotra S, Potkul RK, Banerjee S, Walker C, Drakes ML. Selinexor in Combination with Decitabine Attenuates Ovarian Cancer in Mice.Cancers (Basel). 2023;15:4541. [DOI] [PubMed] [PMC]
Klein C, Brinkmann U, Reichert JM, Kontermann RE. The present and future of bispecific antibodies for cancer therapy.Nat Rev Drug Discov. 2024;23:301–19. [DOI] [PubMed]
Martini S, Figini M, Croce A, Frigerio B, Pennati M, Gianni AM, et al. Selinexor Sensitizes TRAIL-R2-Positive TNBC Cells to the Activity of TRAIL-R2xCD3 Bispecific Antibody.Cells. 2020;9:2231. [DOI] [PubMed] [PMC]
van Montfoort N, Borst L, Korrer MJ, Sluijter M, Marijt KA, Santegoets SJ, et al. NKG2A Blockade Potentiates CD8 T Cell Immunity Induced by Cancer Vaccines.Cell. 2018;175:1744–55.e15. [DOI] [PubMed] [PMC]
Borst L, Sluijter M, Sturm G, Charoentong P, Santegoets SJ, van Gulijk M, et al. NKG2A is a late immune checkpoint on CD8 T cells and marks repeated stimulation and cell division.Int J Cancer. 2022;150:688–704. [DOI] [PubMed] [PMC]
Abd Hamid M, Wang RZ, Yao X, Fan P, Li X, Chang XM, et al. Enriched HLA-E and CD94/NKG2A Interaction Limits Antitumor CD8+ Tumor-Infiltrating T Lymphocyte Responses.Cancer Immunol Res. 2019;7:1293–306. [DOI] [PubMed]
Salomé B, Sfakianos JP, Ranti D, Daza J, Bieber C, Charap A, et al. NKG2A and HLA-E define an alternative immune checkpoint axis in bladder cancer.Cancer Cell. 2022;40:1027–43.e9. [DOI] [PubMed] [PMC]
Bossard C, Bézieau S, Matysiak-Budnik T, Volteau C, Laboisse CL, Jotereau F, et al. HLA-E/β2 microglobulin overexpression in colorectal cancer is associated with recruitment of inhibitory immune cells and tumor progression.Int J Cancer. 2012;131:855–63. [DOI] [PubMed]
Wang X, Xiong H, Ning Z. Implications of NKG2A in immunity and immune-mediated diseases.Front Immunol. 2022;13:960852. [DOI] [PubMed] [PMC]
Cazzetta V, Bruni E, Terzoli S, Carenza C, Franzese S, Piazza R, et al. NKG2A expression identifies a subset of human Vδ2 T cells exerting the highest antitumor effector functions.Cell Rep. 2021;37:109871. [DOI] [PubMed]
Jabri B, Selby JM, Negulescu H, Lee L, Roberts AI, Beavis A, et al. TCR specificity dictates CD94/NKG2A expression by human CTL.Immunity. 2002;17:487–99. [DOI] [PubMed]
Braud VM, Aldemir H, Breart B, Ferlin WG. Expression of CD94-NKG2A inhibitory receptor is restricted to a subset of CD8+ T cells.Trends Immunol. 2003;24:162–4. [DOI] [PubMed]
Andrews LP, Butler SC, Cui J, Cillo AR, Cardello C, Liu C, et al. LAG-3 and PD-1 synergize on CD8+ T cells to drive T cell exhaustion and hinder autocrine IFN-γ-dependent anti-tumor immunity.Cell. 2024;187:4355–72.e22. [DOI] [PubMed] [PMC]
Sun C, Xu J, Huang Q, Huang M, Wen H, Zhang C, et al. High NKG2A expression contributes to NK cell exhaustion and predicts a poor prognosis of patients with liver cancer.Oncoimmunology. 2016;6:e1264562. [DOI] [PubMed] [PMC]
Ruella M, Korell F, Porazzi P, Maus MV. Mechanisms of resistance to chimeric antigen receptor-T cells in haematological malignancies.Nat Rev Drug Discov. 2023;22:976–95. [DOI] [PubMed] [PMC]
Cao X, Jin X, Zhang X, Utsav P, Zhang Y, Guo R, et al. Small-Molecule Compounds Boost CAR-T Cell Therapy in Hematological Malignancies.Curr Treat Options Oncol. 2023;24:184–211. [DOI] [PubMed] [PMC]
Uslu U, Castelli S, June CH. CAR T cell combination therapies to treat cancer.Cancer Cell. 2024;42:1319–25. [DOI] [PubMed]
Wang S, Sellner L, Wang L, Sauer T, Neuber B, Gong W, et al. Combining selective inhibitors of nuclear export (SINEs) with chimeric antigen receptor (CAR) T cells for CD19-positive malignancies.Oncol Rep. 2021;46:170. [DOI] [PubMed] [PMC]
Stadel R, Liu R, Landesman Y, Wald D, Vasanna SH, de Lima MJG. Sequential Administration of Selinexor then CD19 CAR-T Cells Exhibits Enhanced Efficacy in a Mouse Model of Human Non-Hodgkin's Lymphoma.Blood. 2022;140:7413–4. [DOI]
Luo W, Xu J, Li C, Tang L, Li Y, Wang X, et al. Selinexor Reduces the Immunosuppressive Properties of Macrophages and Synergizes with CD19 CAR-T Cells Against B-Cell Lymphoma.Blood. 2024;144:3420. [DOI]
Zhang Y, Zong X, Jia S, Li J, Geng H, Zeng L, et al. Preliminary Data from a First‐in Human Phase Ii Study of Sequential Use of Selinexor and Cd19 Cart Therapy in Patients with Relapsed or Refractory B‐Cell Non‐Hodgkin Lymphoma.Hematol Oncol. 2023;41:784. [DOI]
Wang D, Fu H, Que Y, Ruan H, Xu M, Long X, et al. A novel two-step administration of XPO-1 inhibitor may enhance the effect of anti-BCMA CAR-T in relapsed/refractory extramedullary multiple myeloma.J Transl Med. 2023;21:812. [DOI] [PubMed] [PMC]
Gill SK, Biran N, Phull P, Vesole DH, Siegel DS, Parmar H. Sequential Administration of Selinexor and CAR-T Therapy in Relapsed/Refractory Multiple Myeloma.Blood. 2023;142:6930. [DOI]
Good CR, Aznar MA, Kuramitsu S, Samareh P, Agarwal S, Donahue G, et al. An NK-like CAR T cell transition in CAR T cell dysfunction.Cell. 2021;184:6081–100.e26. [DOI] [PubMed] [PMC]
Abid H, Wu JF, Abid MB. Risk for infections with selinexor in patients with relapsed/refractory multiple myeloma: a systematic review of clinical trials.Eur J Cancer. 2021;154:7–10. [DOI] [PubMed]
Stephens DM, Huang Y, Ruppert AS, Walker JS, Canfield D, Cempre CB, et al. Selinexor Combined with Ibrutinib Demonstrates Tolerability and Safety in Advanced B-Cell Malignancies: A Phase I Study.Clin Cancer Res. 2022;28:3242–7. [DOI] [PubMed] [PMC]
Cornell RF, Baz R, Richter JR, Rossi A, Vogl DT, Chen C, et al. A phase 1 clinical trial of oral eltanexor in patients with relapsed or refractory multiple myeloma.Am J Hematol. 2022;97:E54–8. [DOI] [PubMed]
Hing ZA, Fung HY, Ranganathan P, Mitchell S, El-Gamal D, Woyach JA, et al. Next-generation XPO1 inhibitor shows improved efficacy and in vivo tolerability in hematological malignancies.Leukemia. 2016;30:2364–72. [DOI] [PubMed] [PMC]