The XPO1 Inhibitor KPT-8602 Synergizes with Dexamethasone in Acute Lymphoblastic Leukemia

Purpose: KPT-8602 (Eltanexor) is a second-generation exportin-1 (XPO1) inhibitor with potent activity against acute lymphoblastic leukemia (ALL) in preclinical models and with minimal effects on normal cells. In this study, we evaluated whether KPT-8602 would synergize with dexamethasone, vincristine, or doxorubicin, three drugs currently used for the treatment of ALL. Experimental Design: First, we searched for the most synergistic combination of KPT-8602 with dexamethasone, vincristine, or doxorubicin in vitro in both B-ALL and T-ALL cell lines using proliferation and apoptosis as a readout. Next, we validated this synergistic effect by treatment of clinically relevant B- and T-ALL patient-derived xenograft models in vivo. Finally, we performed RNA-sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) to determine the mechanism of synergy. Results: KPT-8602 showed strong synergism with dexamethasone on human B-ALL and T-ALL cell lines as well as in vivo in three patient-derived ALL xenografts. Compared with single-drug treatment, the drug combination caused increased apoptosis and led to histone depletion. Mechanistically, integration of ChIP-seq and RNA-seq data revealed that addition of KPT-8602 to dexamethasone enhanced the activity of the glucocorticoid receptor (NR3C1) and led to increased inhibition of E2F-mediated transcription. We observed strong inhibition of E2F target genes related to cell cycle, DNA replication, and transcriptional regulation. Conclusions: Our preclinical study demonstrates that KPT-8602 enhances the effects of dexamethasone to inhibit B-ALL and T-ALL cells via NR3C1- and E2F-mediated transcriptional complexes, allowing to achieve increased dexamethasone effects for patients.

[1]  E. Baloglu,et al.  Venetoclax response is enhanced by selective inhibitor of nuclear export compounds in hematologic malignancies. , 2020, Blood advances.

[2]  Y. Ohkawa,et al.  Chromatin-bound CRM1 recruits SET-Nup214 and NPM1c onto HOX clusters causing aberrant HOX expression in leukemia cells , 2019, eLife.

[3]  M. Dimopoulos,et al.  Oral Selinexor-Dexamethasone for Triple-Class Refractory Multiple Myeloma. , 2019, The New England journal of medicine.

[4]  G. Leone,et al.  The broken cycle: E2F dysfunction in cancer , 2019, Nature Reviews Cancer.

[5]  J. Shah,et al.  Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. , 2018, Blood.

[6]  F. Anwer,et al.  Clinical Response and Tolerability of Selinexor in Acute Myeloid Leukemia and Other Hematologic Malignancies - a Systematic Review , 2018, Blood.

[7]  E. Chang,et al.  Duffy Antigen Phenotyping Is a Useful and Clinically Available Test for Benign Ethnic Neutropenia , 2018, Blood.

[8]  D. Daelemans,et al.  Targeting the XPO1-dependent nuclear export of E2F7 reverses anthracycline resistance in head and neck squamous cell carcinomas , 2018, Science Translational Medicine.

[9]  Margaret S. Lee,et al.  Selinexor synergizes with dexamethasone to repress mTORC1 signaling and induce multiple myeloma cell death , 2018, Oncotarget.

[10]  Denis Thieffry,et al.  RSAT 2018: regulatory sequence analysis tools 20th anniversary , 2018, Nucleic Acids Res..

[11]  E. Baloglu,et al.  Clinical Implications of Targeting XPO1‐mediated Nuclear Export in Multiple Myeloma , 2018, Clinical lymphoma, myeloma & leukemia.

[12]  T. Burki Selinexor and dexamethasone in multiple myeloma. , 2018, The Lancet Oncology.

[13]  S. Aerts,et al.  Defining the molecular basis of oncogenic cooperation between TAL1 expression and Pten deletion in T-ALL using a novel pro-T-cell model system , 2017, Leukemia.

[14]  R. Garzon,et al.  A phase 1 clinical trial of single-agent selinexor in acute myeloid leukemia. , 2017, Blood.

[15]  T Terwilliger,et al.  Acute lymphoblastic leukemia: a comprehensive review and 2017 update , 2017, Blood Cancer Journal.

[16]  J. Taunton,et al.  JAK/STAT pathway inhibition overcomes IL7-induced glucocorticoid resistance in a subset of human T-cell acute lymphoblastic leukemias , 2017, Leukemia.

[17]  J. Cools,et al.  The genetics and molecular biology of T-ALL. , 2017, Blood.

[18]  C. Hofmeister,et al.  A Phase 1/2 Study of the Second Generation Selective Inhibitor of Nuclear Export (SINE) Compound, KPT-8602, in Patients with Relapsed Refractory Multiple Myeloma , 2016 .

[19]  Andrew P. Stubbs,et al.  IL-7 Receptor Mutations and Steroid Resistance in Pediatric T cell Acute Lymphoblastic Leukemia: A Genome Sequencing Study , 2016, PLoS medicine.

[20]  Jonathan L. Schmid-Burgk,et al.  The Second-Generation Exportin-1 Inhibitor KPT-8602 Demonstrates Potent Activity against Acute Lymphoblastic Leukemia , 2016, Clinical Cancer Research.

[21]  A. Azmi,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 , 2016, Oncotarget.

[22]  A. Look,et al.  KPT-8602, a second-generation inhibitor of XPO1-mediated nuclear export, is well tolerated and highly active against AML blasts and leukemia-initiating cells , 2016, Leukemia.

[23]  J. Byrd,et al.  Next generation XPO1 inhibitor shows improved efficacy and in vivo tolerability in hematologic malignancies , 2015, Leukemia.

[24]  C. Riccardi,et al.  GILZ as a Mediator of the Anti-Inflammatory Effects of Glucocorticoids , 2015, Front. Endocrinol..

[25]  M. Relling,et al.  Antileukemic Efficacy of Continuous vs Discontinuous Dexamethasone in Murine Models of Acute Lymphoblastic Leukemia , 2015, PloS one.

[26]  Stein Aerts,et al.  i-cisTarget 2015 update: generalized cis-regulatory enrichment analysis in human, mouse and fly , 2015, Nucleic Acids Res..

[27]  J. Byrd,et al.  Preliminary Evidence Of Anti Tumor Activity Of Selinexor (KPT-330) In a Phase I Trial Ofa First-In-Class Oral Selective Inhibitor Of Nuclear Export (SINE) In Patients (pts) With Relapsed / Refractory Non Hodgkin’s Lymphoma (NHL) and Chronic Lymphocytic Leukemia (CLL) , 2013 .

[28]  C. Burd,et al.  Chromatin architecture defines the glucocorticoid response , 2013, Molecular and Cellular Endocrinology.

[29]  J. Cidlowski,et al.  The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. , 2013, The Journal of allergy and clinical immunology.

[30]  J. Roganović Acute Lymphoblastic Leukemia in Children , 2013, Journal of Indian Academy of Oral Medicine and Radiology.

[31]  A. Letai,et al.  KPT‐330 inhibitor of CRM1 (XPO1)‐mediated nuclear export has selective anti‐leukaemic activity in preclinical models of T‐cell acute lymphoblastic leukaemia and acute myeloid leukaemia , 2013, British journal of haematology.

[32]  Yuh Min Chook,et al.  Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia. , 2012, Blood.

[33]  A. Ferrando,et al.  The molecular basis of T cell acute lymphoblastic leukemia. , 2012, The Journal of clinical investigation.

[34]  Gregory E. Crawford,et al.  The Hypersensitive Glucocorticoid Response Specifically Regulates Period 1 and Expression of Circadian Genes , 2012, Molecular and Cellular Biology.

[35]  S. Aerts,et al.  i-cisTarget: an integrative genomics method for the prediction of regulatory features and cis-regulatory modules , 2012, Nucleic acids research.

[36]  D. Sullivan,et al.  Nuclear export of proteins and drug resistance in cancer. , 2012, Biochemical pharmacology.

[37]  C. Libert,et al.  On the Trail of the Glucocorticoid Receptor: Into the Nucleus and Back , 2012, Traffic.

[38]  Kiran C. Bobba,et al.  The genetic basis of early T-cell precursor acute lymphoblastic leukaemia , 2012, Nature.

[39]  M. Cowley,et al.  Evaluation of the NOD/SCID xenograft model for glucocorticoid-regulated gene expression in childhood B-cell precursor acute lymphoblastic leukemia , 2011, BMC Genomics.

[40]  K. Ness,et al.  Adverse effects of treatment in childhood acute lymphoblastic leukemia: general overview and implications for long-term cardiac health , 2011, Expert review of hematology.

[41]  C. Pui,et al.  Glucocorticoid use in acute lymphoblastic leukaemia. , 2010, The Lancet. Oncology.

[42]  M. Ohno,et al.  Cajal body surveillance of U snRNA export complex assembly , 2010, The Journal of cell biology.

[43]  T. Chou Drug combination studies and their synergy quantification using the Chou-Talalay method. , 2010, Cancer research.

[44]  Richard M Myers,et al.  Genomic determination of the glucocorticoid response reveals unexpected mechanisms of gene regulation. , 2009, Genome research.

[45]  C. Riccardi,et al.  GILZ mediates the antiproliferative activity of glucocorticoids by negative regulation of Ras signaling. , 2007, The Journal of clinical investigation.

[46]  R. Crazzolara,et al.  Identification of glucocorticoid-response genes in children with acute lymphoblastic leukemia. , 2006, Blood.

[47]  T. Karpova,et al.  Crm1 is a mitotic effector of Ran-GTP in somatic cells , 2005, Nature Cell Biology.

[48]  C. Paul,et al.  PHAX and CRM1 are required sequentially to transport U3 snoRNA to nucleoli. , 2004, Molecular cell.

[49]  R. Haché,et al.  Nuclear Export of the Glucocorticoid Receptor Is Accelerated by Cell Fusion-dependent Release of Calreticulin* , 2003, Journal of Biological Chemistry.

[50]  M. Garabedian,et al.  Deciphering the Phosphorylation “Code” of the Glucocorticoid Receptor in Vivo * 210 , 2002, The Journal of Biological Chemistry.

[51]  N. Krett,et al.  Mechanisms of glucocorticoid-mediated apoptosis in hematological malignancies. , 2002, Clinical cancer research : an official journal of the American Association for Cancer Research.

[52]  D. Livingston,et al.  E2F4 Is Exported from the Nucleus in a CRM1-Dependent Manner , 2001, Molecular and Cellular Biology.

[53]  T. Hope,et al.  An N‐terminal nuclear export signal is required for the nucleocytoplasmic shuttling of IκBα , 1999 .

[54]  W. Liu,et al.  Glucocorticoid-induced cell death requires autoinduction of glucocorticoid receptor expression in human leukemic T cells. , 1999, Cancer research.

[55]  R. Haché,et al.  Discrimination between NL1- and NL2-Mediated Nuclear Localization of the Glucocorticoid Receptor , 1999, Molecular and Cellular Biology.

[56]  P. Herrlich,et al.  IκBα‐independent downregulation of NF‐κB activity by glucocorticoid receptor , 1997, The EMBO journal.

[57]  A. Baldwin,et al.  Role of Transcriptional Activation of IκBα in Mediation of Immunosuppression by Glucocorticoids , 1995, Science.

[58]  C. Pui,et al.  Glucocorticoid use in acute lymphoblastic leukemia: comparison of prednisone and dexamethasone , 2012 .

[59]  R. Kofler,et al.  Research resource: transcriptional response to glucocorticoids in childhood acute lymphoblastic leukemia. , 2012, Molecular endocrinology.

[60]  M. P. Holloway,et al.  The CRM1 nuclear export protein in normal development and disease. , 2012, International journal of biochemistry and molecular biology.

[61]  K. Borden,et al.  mRNA export and cancer , 2012, Wiley interdisciplinary reviews. RNA.