1
|
Masood A, Iqbal Q, Ehsan H, Davis JA, Hansen DK, Hashmi H. Efficacy and safety of selinexor-based regimens for relapsed/refractory multiple myeloma: a systematic review of literature. Ann Hematol 2022; 101:2601-2610. [PMID: 36214853 DOI: 10.1007/s00277-022-04999-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/29/2022] [Indexed: 11/30/2022]
Abstract
With the incorporation of novel agents in earlier lines of therapy, an increasing number of multiple myeloma patients are refractory to traditional classes of drugs. Selinexor in combination with dexamethasone has emerged as a viable option for heavily pretreated triple-class relapsed and refractory multiple myeloma (RRMM). In this systematic review, we analyzed available literature on the role of selinexor in RRMM. The Boston trial demonstrated that selinexor when combined with dexamethasone and bortezomib is associated with a better depth and duration of response without excessive toxicity, compared with bortezomib and dexamethasone alone. Similarly, selinexor in combination with carfilzomib and dexamethasone was found to have a durable response and tolerable safety profile in both carfilzomib-naive and carfilzomib refractory RRMM patients. Selinexor in combination with IMiDs (lenalidomide and pomalidomide) as well as CD38 monoclonal antibodies (daratumumab) also have promising results. Selinexor combination therapy is both safe and effective for patients with pretreated RRMM.
Collapse
Affiliation(s)
- Adeel Masood
- University of Texas, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Qamar Iqbal
- Internal Medicine - Tidal Health Peninsula Regional, 100 East Carroll Street, Salisbury, MD, 21801, USA
| | - Hamid Ehsan
- Levine Cancer Institute/Atrium Health, 1021 Morehead Medical Drive, Charlotte, NC, 28262, USA
| | - James A Davis
- Hematology/Oncology, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA
| | - Doris K Hansen
- Department of Blood and Marrow Transplant and Cellular Immunotherapy (BMT CI), H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Hamza Hashmi
- Hematology/Oncology, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA
| |
Collapse
|
2
|
Balasubramanian SK, Azmi AS, Maciejewski J. Selective inhibition of nuclear export: a promising approach in the shifting treatment paradigms for hematological neoplasms. Leukemia 2022; 36:601-612. [PMID: 35091658 PMCID: PMC8885406 DOI: 10.1038/s41375-021-01483-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/04/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
Novel targeted therapeutics alone or in rational combinations are likely to dominate the future management of various hematological neoplasms. However, the challenges currently faced are the molecular heterogeneity in driver lesions and genetic plasticity leading to multiple resistance pathways. Thus, progress has overall been gradual. For example, despite the advent of targeted agents against actionable drivers like FLT3 in acute myeloid leukemia (AML), the prognosis remains suboptimal in newly diagnosed and dismal in the relapsed/refractory (R/R) setting, due to other molecular abnormalities contributing to inherent and acquired treatment resistance. Nuclear export inhibitors are of keen interest because they can inhibit several active tumorigenic processes simultaneously and also synergize with other targeted drugs and chemotherapy. XPO1 (or CRM1, chromosome maintenance region 1) is one of the most studied exportins involved in transporting critical cargoes, including tumor suppressor proteins like p27, p53, and RB1. Apart from the TSP cargo transport and its role in drug resistance, XPO1 inhibition results in retention of master transcription factors essential for cell differentiation, cell survival, and autophagy, rendering cells more susceptible to the effects of other antineoplastic agents, including targeted therapies. This review will dissect the role of XPO1 inhibition in hematological neoplasms, focusing on mechanistic insights gleaned mainly from work with SINE compounds. Future potential combinatorial strategies will be discussed.
Collapse
Affiliation(s)
| | - Asfar S Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, USA
| | - Jaroslaw Maciejewski
- Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, USA.
| |
Collapse
|
3
|
Turner JG, Cui Y, Bauer AA, Dawson JL, Gomez JA, Kim J, Cubitt CL, Nishihori T, Dalton WS, Sullivan DM. Melphalan and Exportin 1 Inhibitors Exert Synergistic Antitumor Effects in Preclinical Models of Human Multiple Myeloma. Cancer Res 2020; 80:5344-5354. [PMID: 33023948 PMCID: PMC7718436 DOI: 10.1158/0008-5472.can-19-0677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 08/25/2020] [Accepted: 10/01/2020] [Indexed: 01/22/2023]
Abstract
High-dose chemotherapy with melphalan followed by autologous transplantation is a first-line treatment for multiple myeloma. Here, we present preclinical evidence that this treatment may be significantly improved by the addition of exportin 1 inhibitors (XPO1i). The XPO1i selinexor, eltanexor, and KOS-2464 sensitized human multiple myeloma cells to melphalan. Human 8226 and U266 multiple myeloma cell lines and melphalan-resistant cell lines (8226-LR5 and U266-LR6) were highly sensitized to melphalan by XPO1i. Multiple myeloma cells from newly diagnosed and relapsed/refractory multiple myeloma patients were also sensitized by XPO1i to melphalan. In NOD/SCIDγ mice challenged with either parental 8226 or U266 multiple myeloma and melphalan-resistant multiple myeloma tumors, XPO1i/melphalan combination treatments demonstrated stronger synergistic antitumor effects than single-agent melphalan with minimal toxicity. Synergistic cell death resulted from increased XPO1i/melphalan-induced DNA damage in a dose-dependent manner and decreased DNA repair. In addition, repair of melphalan-induced DNA damage was inhibited by selinexor, which decreased melphalan-induced monoubiquitination of FANCD2 in multiple myeloma cells. Knockdown of FANCD2 was found to replicate the effect of selinexor when used with melphalan, increasing DNA damage (γH2AX) by inhibiting DNA repair. Thus, combination therapies that include selinexor or eltanexor with melphalan may have the potential to improve treatment outcomes of multiple myeloma in melphalan-resistant and newly diagnosed patients. The combination of selinexor and melphalan is currently being investigated in the context of high-dose chemotherapy and autologous transplant (NCT02780609). SIGNIFICANCE: Inhibition of exportin 1 with selinexor synergistically sensitizes human multiple myeloma to melphalan by inhibiting Fanconi anemia pathway-mediated DNA repair.
Collapse
Affiliation(s)
- Joel G Turner
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Yan Cui
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Alexis A Bauer
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jana L Dawson
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Juan A Gomez
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Christopher L Cubitt
- Translational Research Core, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taiga Nishihori
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - William S Dalton
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Daniel M Sullivan
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
4
|
Richard S, Richter J, Jagannath S. Selinexor: a first-in-class SINE compound for treatment of relapsed refractory multiple myeloma. Future Oncol 2020; 16:1331-1350. [PMID: 32511022 DOI: 10.2217/fon-2020-0054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- 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
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York 10029, USA
| |
Collapse
|
5
|
Walker JS, Garzon R, Lapalombella R. Selinexor for advanced hematologic malignancies. Leuk Lymphoma 2020; 61:2335-2350. [DOI: 10.1080/10428194.2020.1775210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Janek S. Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ramiro Garzon
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
6
|
van Erp AEM, van Houdt L, Hillebrandt-Roeffen MHS, van Bree NFHN, Flucke UE, Mentzel T, Shipley J, Desar IME, Fleuren EDG, Versleijen-Jonkers YMH, van der Graaf WTA. Olaparib and temozolomide in desmoplastic small round cell tumors: a promising combination in vitro and in vivo. J Cancer Res Clin Oncol 2020; 146:1659-1670. [PMID: 32279088 PMCID: PMC7256072 DOI: 10.1007/s00432-020-03211-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/03/2020] [Indexed: 12/15/2022]
Abstract
Purpose Desmoplastic small round cell tumors (DSRCTs) are highly malignant and very rare soft tissue sarcomas with a high unmet need for new therapeutic options. Therefore, we examined poly(ADP-ribose) polymerase 1 (PARP1) and Schlafen-11 (SLFN11) expression in DSRCT tumor tissue and the combination of PARP inhibitor olaparib with the alkylating agent temozolomide (TMZ) in a preclinical DSRCT model. Methods PARP1 and SLFN11 have been described as predictive biomarkers for response to PARP inhibition. Expression of PARP1 and SLFN11 was assessed in 16 and 12 DSRCT tumor tissue samples, respectively. Effects of single-agent olaparib, and olaparib and TMZ combination treatment were examined using the preclinical JN-DSRCT-1 model. In vitro, single-agent and combination treatment effects on cell viability, the cell cycle, DNA damage and apoptosis were examined. Olaparib and TMZ combination treatment was also assessed in vivo. Results PARP1 and SLFN11 expression was observed in 100% and 92% of DSRCT tumor tissues, respectively. Olaparib treatment reduced cell viability and cell migration in a dose-dependent manner in vitro. Drug synergy between olaparib and TMZ was observed in vitro and in vivo. Combination treatment led to a cell-cycle arrest and induction of DNA damage and apoptosis, even when combined at low dosages. Conclusion We show high PARP1 and SLFN11 expression in DSRCT tumor material and antitumor effects following olaparib and TMZ combination treatment in a preclinical DSRCT model. This suggests that olaparib and TMZ combination treatment could be a potential treatment option for DSRCTs. Electronic supplementary material The online version of this article (10.1007/s00432-020-03211-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anke E M van Erp
- Department of Medical Oncology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Laurens van Houdt
- Department of Medical Oncology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - Niek F H N van Bree
- Department of Medical Oncology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Uta E Flucke
- Department of Pathology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - Janet Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, UK
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Emmy D G Fleuren
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Yvonne M H Versleijen-Jonkers
- Department of Medical Oncology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Medical Oncology, The Netherlands Cancer Institute-Van Leeuwenhoek, 1066 CX, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Sudalagunta P, Silva MC, Canevarolo RR, Alugubelli RR, DeAvila G, Tungesvik A, Perez L, Gatenby R, Gillies R, Baz R, Meads MB, Shain KH, Silva AS. A pharmacodynamic model of clinical synergy in multiple myeloma. EBioMedicine 2020; 54:102716. [PMID: 32268267 PMCID: PMC7136599 DOI: 10.1016/j.ebiom.2020.102716] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/03/2020] [Accepted: 02/28/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Multiagent therapies, due to their ability to delay or overcome resistance, are a hallmark of treatment in multiple myeloma (MM). The growing number of therapeutic options in MM requires high-throughput combination screening tools to better allocate treatment, and facilitate personalized therapy. METHODS A second-order drug response model was employed to fit patient-specific ex vivo responses of 203 MM patients to single-agent models. A novel pharmacodynamic model, developed to account for two-way combination effects, was tested with 130 two-drug combinations. We have demonstrated that this model is sufficiently parameterized by single-agent and fixed-ratio combination responses, by validating model estimates with ex vivo combination responses for different concentration ratios, using a checkerboard assay. This new model reconciles ex vivo observations from both Loewe and BLISS synergy models, by accounting for the dimension of time, as opposed to focusing on arbitrary time-points or drug effect. Clinical outcomes of patients were simulated by coupling patient-specific drug combination models with pharmacokinetic data. FINDINGS Combination screening showed 1 in 5 combinations (21.43% by LD50, 18.42% by AUC) were synergistic ex vivo with statistical significance (P < 0.05), but clinical synergy was predicted for only 1 in 10 combinations (8.69%), which was attributed to the role of pharmacokinetics and dosing schedules. INTERPRETATION The proposed framework can inform clinical decisions from ex vivo observations, thus providing a path toward personalized therapy using combination regimens. FUNDING This research was funded by the H. Lee Moffitt Cancer Center Physical Sciences in Oncology (PSOC) Grant (1U54CA193489-01A1) and by H. Lee Moffitt Cancer Center's Team Science Grant. This work has been supported in part by the PSOC Pilot Project Award (5U54CA193489-04), the Translational Research Core Facility at the H. Lee Moffitt Cancer Center & Research Institute, an NCI-designated Comprehensive Cancer Center (P30-CA076292), the Pentecost Family Foundation, and Miles for Moffitt Foundation.
Collapse
Affiliation(s)
- Praneeth Sudalagunta
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Dr, SRB 4th 24011, Tampa, FL 33612, USA
| | - Maria C Silva
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Dr, SRB 4th 24011, Tampa, FL 33612, USA
| | - Rafael R Canevarolo
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Dr, SRB 4th 24011, Tampa, FL 33612, USA
| | - Raghunandan Reddy Alugubelli
- Department of Collaborative Data Services Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Gabriel DeAvila
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Alexandre Tungesvik
- Department of Internal Medicine, USF Health Morsani College of Medicine, Tampa, FL 33612, USA
| | - Lia Perez
- Department of Blood and Marrow Transplantation Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Robert Gatenby
- Department of Diagnostic Imaging, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Robert Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Dr, SRB 4th 24011, Tampa, FL 33612, USA
| | - Rachid Baz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Mark B Meads
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kenneth H Shain
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ariosto S Silva
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Dr, SRB 4th 24011, Tampa, FL 33612, USA.
| |
Collapse
|
8
|
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; 19:689-698. [PMID: 31543372 DOI: 10.1016/j.clml.2019.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/08/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
Abstract
The correct localization of molecules between nucleus and cytoplasm is fundamental for cellular homeostasis and is controlled by a bidirectional transport system. Exportin 1 (XPO1) regulates the passage of numerous cancer-related proteins. In this review, we summarize the development of a novel class of antitumor agents, known as selective inhibitors of nuclear export (SINEs). We report results of preclinical studies and clinical trials, and discuss the mechanism of action of SINEs and their effects in multiple myeloma, non-Hodgkin lymphomas, lymphoblastic leukemia, and acute and chronic myeloid leukemia. In the future, the numerous experimental studies currently underway will allow us to define the role of SINEs and will possibly permit these substances to be introduced into daily clinical practice.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi," University of Messina, Messina, Italy.
| | - Vanessa Innao
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi," University of Messina, Messina, Italy
| | - Andrea Gaetano Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi," University of Messina, Messina, Italy
| | - Rossana Leanza
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi," University of Messina, Messina, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi," University of Messina, Messina, Italy
| |
Collapse
|
9
|
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; 18:335-345. [PMID: 29610030 DOI: 10.1016/j.clml.2018.03.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/26/2018] [Accepted: 03/08/2018] [Indexed: 12/30/2022]
Abstract
Multiple myeloma (MM) is a malignancy of plasma cells that is typically chronic, and relapse is common. Current therapeutic strategies include combination and sequential treatments with corticosteroids, alkylating agents, proteasomal inhibitors, immunomodulators, and monoclonal antibodies. These drugs prolong survival but ultimately become ineffective. Exportin 1 (XPO1), a nuclear export protein, is overexpressed in MM cells, and knockdown studies have suggested that XPO1 is essential for MM cell survival. Selective inhibitor of nuclear export (SINE) compounds are novel, orally bioavailable class of agents that specifically inhibit XPO1. Selinexor (KPT-330) is the first-in-human SINE compound. Early phase clinical trials have established the safety profile of this agent and have shown promising efficacy in combination with low-dose dexamethasone and other anti-MM agents. The combination of selinexor and dexamethasone has demonstrated activity in "penta-refractory" MM, (ie, MM refractory to the 5 most active anti-MM agents currently used in treatment). We have reviewed the available data on the molecular implications of XPO1 inhibition in MM. We also reviewed the pertinent early phase clinical data with SINE compounds and discuss management strategies for common toxicities encountered with use of selinexor.
Collapse
Affiliation(s)
- Ujjawal H Gandhi
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN
| | | | | | | | - Ajai Chari
- Division of Hematology and Oncology, Mount Sinai Hospital, New York, NY
| | - Dan Vogl
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA
| | - Robert F Cornell
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
10
|
Turner JG, Dawson JL, Grant S, Shain KH, Dalton WS, Dai Y, Meads M, Baz R, Kauffman M, Shacham S, Sullivan DM. Treatment of acquired drug resistance in multiple myeloma by combination therapy with XPO1 and topoisomerase II inhibitors. J Hematol Oncol 2016; 9:73. [PMID: 27557643 PMCID: PMC4997728 DOI: 10.1186/s13045-016-0304-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/18/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Acquired drug resistance is the greatest obstacle to the successful treatment of multiple myeloma (MM). Despite recent advanced treatment options such as liposomal formulations, proteasome inhibitors, immunomodulatory drugs, myeloma-targeted antibodies, and histone deacetylase inhibitors, MM is still considered an incurable disease. METHODS We investigated whether the clinical exportin 1 (XPO1) inhibitor selinexor (KPT-330), when combined with pegylated liposomal doxorubicin (PLD) or doxorubicin hydrochloride, could overcome acquired drug resistance in multidrug-resistant human MM xenograft tumors, four different multidrug-resistant MM cell lines, or ex vivo MM biopsies from relapsed/refractory patients. Mechanistic studies were performed to assess co-localization of topoisomerase II alpha (TOP2A), DNA damage, and siRNA knockdown of drug targets. RESULTS Selinexor was found to restore sensitivity of multidrug-resistant 8226B25, 8226Dox6, 8226Dox40, and U266PSR human MM cells to doxorubicin to levels found in parental myeloma cell lines. NOD/SCID-γ mice challenged with drug-resistant or parental U266 human MM and treated with selinexor/PLD had significantly decreased tumor growth and increased survival with minimal toxicity. Selinexor/doxorubicin treatment selectively induced apoptosis in CD138/light-chain-positive MM cells without affecting non-myeloma cells in ex vivo-treated bone marrow aspirates from newly diagnosed or relapsed/refractory MM patients. Selinexor inhibited XPO1-TOP2A protein complexes (proximity ligation assay), preventing nuclear export of TOP2A in both parental and multidrug-resistant MM cell lines. Selinexor/doxorubicin treatment significantly increased DNA damage (comet assay/γ-H2AX) in both parental and drug-resistant MM cells. TOP2A knockdown reversed both the anti-tumor effect and significantly reduced DNA damage induced by selinexor/doxorubicin treatment. CONCLUSIONS The combination of an XPO1 inhibitor and liposomal doxorubicin was highly effective against acquired drug resistance in in vitro MM models, in in vivo xenograft studies, and in ex vivo samples obtained from patients with relapsed/refractory myeloma. This drug combination synergistically induced TOP2A-mediated DNA damage and subsequent apoptosis. In addition, based on our preclinical data, we have initiated a phase I/II study with the XPO1 inhibitor selinexor and PLD (ClinicalTrials.gov NCT02186834). Initial results from both preclinical and clinical trials have shown significant promise for this drug combination for the treatment of MM.
Collapse
Affiliation(s)
- Joel G. Turner
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Jana L. Dawson
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA USA
| | - Kenneth H. Shain
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - William S. Dalton
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
- M2Gen® Biotechnologies, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Yun Dai
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA USA
| | - Mark Meads
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | - Rachid Baz
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
| | | | | | - Daniel M. Sullivan
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL USA
- H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612 USA
| |
Collapse
|