REQUEST FOR PROJECT TEAM MEMBER APPLICATIONS FOR CONDUCTING CLINICAL TRIALS USING SELINEXOR (NSC 781780)

The National Cancer Institute (NCI) Cancer Therapy Evaluation Program (CTEP) is accepting Project Team Member Applications for a project using selinexor, a selective small-molecule inhibitor of the nuclear export protein XPO1 being developed by CTEP as an anticancer agent in collaboration with Karyopharm Therapeutics. Selinexor inhibits nuclear export by binding the nuclear export protein XPO1, leading to the accumulation of tumor suppressor proteins in the . This is believed to reinitiate and amplify their tumor suppressor function and is believed to lead to the selective induction of in cancer cells, while largely sparing normal cells; the precise mechanism by which XPO1 inhibition leads to cancer cell death is uncertain. Selinexor (XPOVIO) is approved for treatment of patients with relapsed or refractory diffuse large B-cell and, in combination with , for treatment of patients with relapsed or refractory .

At the present time, the preliminary CTEP drug development plan is for Phase I development of combinations of selinexor with venetoclax, venetoclax plus DNA methyltransferase inhibitors, and venetoclax plus flt-3 inhibitors in myeloid malignancy; Phase I combinations of selinexor with proteasome inhibitors in soft tissue sarcomas, and Phase I combinations of selinexor with temozolomide in glioblastoma multiforme. Preclinical models suggest synergistic activity of these combinations in these disease settings. The role of the project team is to evaluate all available evidence to modify and refine this initial plan or suggest an alternative project.

It is anticipated that CTEP will activate 3 – 6 different combination trials with selinexor. The project team will include:

1. Clinician Scientists with expertise in phase I and II studies and with an interest in myeloid malignancies, sarcoma, and glioblastoma (fill out Part A of the attached Application; Clinician Scientists must belong to a qualifying NCI grant funded institution as defined at the end of this letter); 2. Translational scientists with an interest in biomarker development in nuclear export, tumor suppressor genes and/or apoptosis, and acquired mechanisms of resistance (fill out Part B of the attached Application and see the submission instructions at the end of this letter); and 3. Basic scientists with expertise in nuclear export and mechanisms of carcinogenesis or apotosis (fill out Part C of the attached Application and see the submission instructions at the end of this letter).

Prospective team members may apply for multiple roles using a single application form by completing all the appropriate Parts. The project team will be recruited nationally and will prioritize the research questions regarding selinexor in combination trials, including prioritization of biomarker studies. It is anticipated that the clinicians on the drug project team will be tasked with writing the Letters of Intent describing the study design, based upon the team’s recommendations, for CTEP approval, and that these clinicians will ultimately lead the clinical studies. It is also anticipated that other extramural members of the drug project team will stay involved in the subsequent design and execution of the proposed trials. It is anticipated that the project team will complete its work in 6 – 8 weeks or less.

Background/Rationale Exportin 1 (XPO1) is the major nuclear export protein of tumor suppressor proteins (TSPs; e.g., p53, pRb, IκB, p27, p21, FOXOs) and eIF4E-bound proto- mRNAs (e.g., c-Myc, Bcl2, Bcl6, BclxL). Elevated XPO1 expression inactivates TSPs by mislocalization, enhances proto-oncoprotein translation, and correlates with poor patient prognosis in a variety of cancers, including multiple myeloma, soft tissue sarcoma, and glioblastoma.

Mechanism of Action Selinexor is an oral small-molecule selective inhibitor of XPO1. This inhibition leads to the accumulation and reactivation of TSPs in the cell nucleus and is also associated with blockage of proto-oncoprotein translation and of DNA damage repair. Ultimately, inhibition of XPO1 by selinexor is believed to lead to the selective induction of apoptosis in cancer cells, while largely sparing normal cells. Synergy has been reported in vitro and in vivo in combination studies of selinexor plus DNA damage-inducing therapies, proteasome inhibitors, and conventional .

Nonclinical Studies of Selinexor and Analogues Selinexor and/or its congeners have shown broad anti-tumor activity in acute myeloid (AML) cell lines and murine xenograft models of AML and glioblastoma multiforme (GBM), both as single agents and in combination with sorafenib, quizartinib, nutlin-3a, venetoclax, decitabine, temozolomide, and radiation. In in vitro AML models, selinexor treatment led to reductions of flt-3, c- kit and MCL-1 expression; enhanced therapeutic index in killing AML cells versus normal hematopoietic cells; and AML cell differentiation. In xenograft models of AML and diffuse large B cell lymphoma (DLBCL), combinations of selinexor with venetoclax synergized. In vitro augmentation of activity with MDM2 inhibition was demonstrated in p53 wild-type AML. In flt-3 ITD AML xenografts, flt-3 inhibitors augmented activity; synergy was particularly impressive with quizartinib. In animal models of glioblastoma multiforme, addition of selinexor to radiation alone improved survival, and selinexor synergized with temozolomide in inhibition of tumor growth. Selinexor appears to be particularly active against NPM-1 mutated leukemia: selinexor restored nuclear localization of the cytoplasmically mis-localized mutant NPM1 protein and restored HOX expression programs. In multiple myeloma, selinexor inhibited bone marrow stromal cell protection and osteoclast differentiation.

Clinical Studies of Selinexor Selinexor (XPOVIO) has been approved by the Food and Drug Administration (FDA) for treatment of patients with relapsed or refractory diffuse large B-cell lymphoma and, in combination with dexamethasone, for treatment of patients with relapsed or refractory multiple myeloma. Clinical trials are currently testing Selinexor in Phase II and III clinical development, alone or in combination regimens, for the treatment of patients with liposarcoma, endometrial cancer, diffuse large B-cell lymphoma, and glioblastoma. The following active studies are being conducted as part of the Karyopharm clinical development program for selinexor (Table 1):

Table 1: Active Karyopharm-sponsored selinexor listings on ClinicalTrials.gov

NCT Number Phase Agent(s) Disease/ Sponsor Study Planned Abstract Indication Start- Accrual End NCT02091245 1 Selinexor Relapsed/ Karyopharm and 3/2014 – 16 Place et al., refractory Dana-Farber 6/2020 2018 childhood NCT02228525 2 Selinexor Myelodysplastic Karyopharm, Sloan 8/2014 – 26 Taylor et al., syndrome Kettering, MD 8/2021 2018 Anderson, and Columbia University

Page 2 of 7

NCT Number Phase Agent(s) Disease/ Sponsor Study Planned Abstract Indication Start- Accrual End NCT02186834 1/2 Selinexor + Relapsed/ Karyopharm and H. 9/2014 – 28 Baz et al., liposomal refractory Lee Moffitt Cancer 12/2020 2016 doxorubicin + multiple Center dexamethasone myeloma NCT02227251 2 Selinexor Diffuse large B- Karyopharm 11/2014 129 Maerevoet et cell lymphoma – al., 2020 12/2020

NCT02403310 1 Selinexor + Leukemia Karyopharm and H. 6/2015 – 21 Sweet et al., daunorubicin + Lee Moffitt Cancer 12/2020 2020 cytarabine Center

NCT02343042 1/2 Selinexor + Multiple Karyopharm 10/2015 437 Bahlis et al. dexamethasone myeloma – 5/2020 2018 + backbones (, , , pomalidomide + bortezomib, , , ixazomib, or pomalidomide + elotuzumab) NCT02471911 1 Selinexor + Diffuse large B- Karyopharm and 11/2015 23 RICE (rituximab, cell lymphoma Weill-Cornell – 1/2021 etoposide, carboplatin, ifosfamide, dexamethasone) NCT02606461 2/3 Selinexor vs. Advanced Karyopharm 12/2015 334 Gounder et placebo liposarcoma – al., 2018 11/2020

NCT02831686 1 Selinexor + Multiple Karyopharm, Sloan 7/2016 – 18 Salcedo et ixazomib + myeloma Kettering, and 7/2020 al., 2020 dexamethasone Millennium Pharmaceuticals NCT02741388 1 Selinexor + R- B-cell lymphoma Karyopharm and 10/2016 60 DHAOx Lymphoma Academic – 6/2021 (rituximab, Research dexamethasone, Organisation oxaliplatin, cytarabine); selinexor + R- GDP (rituximab, dexamethasone, gemcitabine, cisplatin) NCT03042819 1 Selinexor + Soft tissue Karyopharm and 5/2017 – 25 Malone et doxorubicin sarcoma University Health 11/2020 al., 2018 Network, Toronto

NCT03110562 3 Selinexor + Multiple Karyopharm 5/2017 – 402 bortezomib + myeloma 6/2020 dexamethasone vs. bortezomib + dexamethasone NCT02780609 1/2 Selinexor + high- Multiple Karyopharm and H. 6/2017 – 46 Nishihori et dose melphalan myeloma Lee Moffitt Cancer 8/2022 al., 2019 + Center dexamethasone + hematopoietic cell transplant

Page 3 of 7

NCT Number Phase Agent(s) Disease/ Sponsor Study Planned Abstract Indication Start- Accrual End NCT03466827 2 Selinexor Thymic cancers Karyopharm, Institut 10/2017 25 Curie, Gustave – 7/2020 Roussy, Hospices Civils de Lyon, and GSO Global NCT03555422 3 Selinexor vs. Endometrial Karyopharm, BGOG, 1/2018 – 192 placebo cancer NOGGO, MITO, 3/2023 GEICO, CEEGOG, and ISGO NCT03095612 1/2 Selinexor + Non-small cell Karyopharm and UT 3/2018 – 59 docetaxel lung cancer Southwestern 5/2021

NCT03193437 2 Selinexor Thymic cancers Karyopharm, 4/2018 – 25 Georgetown U., and 12/2021 Hackensack Meridian Health NCT03627403 2 Selinexor Myelofibrosis Karyopharm and U. of 3/2019 – 56 Utah 3/2025

NCT03955783 1 Selinexor + Diffuse large B- Karyopharm, AbbVie 6/2019 – 78 venetoclax cell lymphoma; and Vanderbilt-Ingram 1/2023 ; non- Hodgkin’s lymphoma NCT04256707 1 Selinexor; Non-small cell Karyopharm 1/2020 – 38 Selinexor + lung cancer; 12/2023 docetaxel; Selinexor + pembrolizumab NCT04349098 2 Selinexor vs. Severe COVID- Karyopharm 4/2020 – 230 placebo 19 infection 8/2020

NCT04421378 1/2 Selinexor + Glioblastoma Karyopharm 6/2020 – 402 radiation vs. multiforme 1/2023 temozolomide + radiation; Selinexor + temozolomide + radiation vs. temozolomide + radiation; Selinexor + lomustine vs. lomustine

CTEP’s Plans for Selinexor Development The current CTEP development plan focuses on Phase I or Phase II studies of combinations of selinexor with appropriate agents in myeloid malignancy, glioblastoma, and soft tissue sarcomas. Selinexor is FDA-approved in multiple myeloma and has shown activity in a wide variety of non- Hodgkins lymphoma, including T cell lymphoma.

Correlative Studies of Interest to CTEP The downstream effects of XPO1 inhibition following clinical administration of selinexor have not been well studied. Correlative studies are expected to be designed with focus on the appropriate targets in the individual clinical trials. An example might be nuclear relocation of NPM1 protein in cases of NPM1-mutated AML; p53 expression in combinations with MDM2 inhibitors. Identification of the proteins whose nuclear accumulation triggers apoptosis and the mechanism by which that occurs are of interest.

Page 4 of 7

Selinexor Project Team Selection, Composition, and Tasks The selinexor drug project team will meet regularly by WebEx to review the in vitro, in vivo, and clinical data regarding selinexor and selinexor drug combinations. The team will determine promising strategies, appropriate biomarkers to evaluate the strategies, and clinical trial designs to test the strategies. The project team will be composed of intramural and extramural members. The extramural members will include clinician-scientists with experience in Phase I and II studies in myeloid malignancy, soft tissue sarcoma, and brain tumors; translational scientists with expertise in biomarker development; and basic scientists with expertise in nuclear transport, tumor suppressor genes, and mechanisms of apoptosis. Since the clinician scientists selected for the project team will be expected to lead the clinical trials that come out of this process, the evaluation criteria for the clinician scientists will include not only clinical trial expertise but also their documented record of accrual to early phase studies in one or more of the above-mentioned tumor types.

Questions regarding this request for applications may be addressed to Steven Gore, M.D., Medical Officer, Investigational Drug Branch, CTEP, DCTD, NCI (phone: 240-276-6565; e-mail: [email protected]).

CTEP recognizes the importance of encouraging and supporting young investigators as they embark upon a clinical cancer research career. CTEP highly encourages Career Development Applications (CrDAs) from these investigators and their mentors to participate as Project Team members and to develop Career Development Letters of Intent (CrDLs) after conclusion of Project Team activities.

Project Team Member Applications (PTMAs) should contain a clear indication of the applicant’s desired role on the Selinexor Project Team (clinician scientist, translational scientist, or basic scientist). The PTMA should also be accompanied by an NIH Biosketch containing a personal statement customized to this project. The PTMAs should be sent to the Protocol and Information Office (PIO) at the address below by 5:00 PM Eastern Time (ET), August 4, 2020. The most recent version of the PTMA form, which has been distributed with this communication, must be used. PTMAs should be submitted electronically to:

PIO, CTEP/DCTD/NCI E-mail: [email protected]

Please note that Clinician Scientists may only participate through association with the ETCTN, an NCTN Group, or a consortium (see below), and will need to submit the PTMA through their ETCTN LAO’s Coordinating Center or the Group/Consortium Operations office, as applicable. That organization will then need to submit the Clinician’s application to PIO on your behalf to confirm that they are in support of the proposal. All submissions funded by NCTN organizations should include a Letter of Support from the Group Chair. Please allow sufficient time for your organization’s review. Qualifying clinical institutions include: • ETCTN Participating Institution (under UM1 grant) • NCTN Group member institution (under U10 grant; Alliance, COG, ECOG-ACRIN, NRG Oncology, or SWOG) • Institutional affiliation with the Pediatric Brain Tumor Consortium (PBTC), Adult Brain Tumor Consortium (ABTC), or Cancer Immunotherapy Trials Network (CITN)

Basic and Translational Scientists who belong to a participating ETCTN institution (Lead Academic Organization [LAO] or Affiliated Organization [AO]) must submit applications through your LAO’s Coordinating Center. Please allow enough time for your organization’s review. Basic and

Page 5 of 7

Translational Scientists from non-ETCTN-affiliated institutions may submit their applications directly to PIO.

Bibliography

Bahlis, N.J., H. Sutherland, D. White, et al. (2018). Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. Blood. 132:2546-2554.

Baz, R.C., H.H. Shain, M. Alsina, et al. (2016). Phase I trial of the combination of selinexor (SEL), liposomal doxorubicin (DOX) and dexamethasone (Dex) for relapsed and refractory multiple myeloma (RRMM). J Clin Oncol. 34:Abstract 8013.

Bertucci, F., P. Finetti, and D. Birnbaum. (2016). XPO1, Therapeutic… and Prognostic Target in Sarcomas. Oncoscience. 3:143-144.

Brunetti, L., M.C. Gundry, D. Sorcini, et al. (2018). Mutant NPM1 Maintains the Leukemic State Through HOX Expression. Cancer Cell. 34:499-512.

Carlson, R.O., W. Senapedis, T. Kashyap, et al. (2014). The combination of selinexor (KPT-330), a selective inhibitor of nuclear export (SINE), & the FLT3 inhibitor quizartinib shows anti-tumor activity in acute myeloid leukemia (AML) in vitro and in vivo. 19th European Hematology Association Congress. Abstract P796.

Cooperrider, J.H., N. Fulton, A.S. Artz, et al. (2020). Phase I Trial of Maintenance Selinexor After Allogeneic Transplantation for Patients With Acute Myeloid Leukemia and Myelodysplastic Syndrome. Bone Marrow Transplant. Online ahead of print. PMID: 32376970.

Etchin, J., Q. Sun, A. Kentsis, et al. (2013). Antileukemic activity of nuclear export inhibitors that spare normal hematopoietic cells. Leukemia. 27:66-74.

Etchin, J., J. Montero, A. Berezovskaya, et al. (2016). Activity of a selective inhibitor of nuclear export, selinexor (KPT-330), against AML-initiating cells engrafted into immunosuppressed NSG mice. Leukemia. 30:190-199.

Gounder, M.M., N. Somaiah, S. Attia, et al. (2018). Phase 2 results of selinexor in advanced de- differentiated (DDLS) liposarcoma (SEAL) study: A phase 2/3, randomized, double blind, placebo controlled cross-over study. J Clin Oncol. 36:Abstract 11512.

Gu, X., Q. Ebrahem, R.Z. Mahfouz, et al. (2018). Leukemogenic Nucleophosmin Mutation Disrupts the Transcription Factor Hub That Regulates Granulomonocytic Fates. J Clin Invest. 128:4260- 4279.

Jakubowiak, A.J., J.K. Jasielec, C.A. Rosenbaum, et al. (2019). Phase 1 Study of Selinexor Plus Carfilzomib and Dexamethasone for the Treatment of relapsed/refractory Multiple Myeloma. Br J Haemtol. 186:549-560.

Kojima, K., S.M. Kornblau, V. Ruvolo, et al. (2013). Prognostic Impact and Targeting of CRM1 in Acute Myeloid Leukemia. Blood. 121-4166-4174.

Leudtke, D.A., Y. Su, S. Liu, et al. (2018). Inhibition of XPO1 Enhances Cell Death Induced by ABT- 199 in Acute Myeloid Leukaemia via Mcl-1. J Cell Mol Med. 22:6099-6111.

Page 6 of 7

Liu, X., Y. Chong, Y. Tu, et al. (2016). CRM1/XPO1 is associated with clinical outcome in glioma and represents a therapeutic target by perturbing multiple core pathways. J. Hematol Oncol. 9:108.

Maerevoet, M., J.M. Zijlstra, G. Follows, et al. (2020). Survival among patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) treated with single agent oral selinexor in the SADAL study. 25th European Hematology Association Congress. Abstract EP1260.

Malone, E.R., E. al-Ezzi, A.A. Gupta, et al. (2018). Phase 1b study of selinexor, a first in class selective inhibitor of nuclear export (SINE) compound, in combination with doxorubicin in patients (pts) with locally advanced or metastatic soft tissue sarcoma (STS). J Clin Oncol. 36:Abstract 11562.

Nishihori, T., M. Alsina, J. Ochoa, et al. (2019). The Result of a Phase 1 Study of Selinexor in Combination with High-Dose Melphalan and Autologous Hematopoietic Cell Transplantation for Multiple Myeloma. Blood. 134:3314.

Pardee, T.S., K.C. Wood, K.H. Lin, et al. (2019). Selinexor in Combination with Induction and Consolidation Therapy in Older Adults with AML Is Highly Active. Blood. 134:1388.

Place, A.E., T.M. Blonquist, E. Stieglitz, et al. (2018). Phase I Study of the Selinexor in Relapsed/Refractory Childhood Acute Leukemia. Blood. 132:1405.

Ranganathan, P., X. Yu, C. Na, et al. (2012). Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia. Blood. 120:1765-1773.

Ranganathan, P., X. Yu, R. Santhanam, et al. (2015). Decitabine Priming Enhances the Antileukemic Effects of Exportin 1 (XPO1) Selective Inhibitor Selinexor in Acute Myeloid Leukemia. Blood. 125:2689-2692.

Ranganathan, P., T. Kashyap, X. Yu, et al. (2016). XPO1 Inhibition Using Selinexor Synergizes With Chemotherapy in Acute Myeloid Leukemia by Targeting DNA Repair and Restoring Topoisomerase IIα to the Nucleus. Clin Cancer Res. 22:6142-6152.

Salcedo, M., N. Lendvai, D. Mastey, et al. (2020). Phase I Study of Selinexor, Ixazomib, and Low- dose Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma. Clin Lymphoma Myeloma Leuk. 20-198-200.

Sweet, K., R. Komrokji, E. Padron, et al. (2020). Phase I Clinical Trial of Selinexor in Combination With Daunorubicin and Cytarabine in Previously Untreated Poor-Risk Acute Myeloid Leukemia. Clin Cancer Res. 26:54-60.

Tai, Y.-T., Y. Landesman, C. Acharya, et al. (2014). CRM1 Inhibition Induces Tumor Cell Cytotoxicity and Impairs Osteoclastogenesis in Multiple Myeloma: Molecular Mechanisms and Therapeutic Implications. Leukemia. 28:155-165.

Taylor, J., M. Coleman, K. Alvarez, et al. (2018). Selinexor, a First-in-Class XPO1 Inhibitor, Is Efficacious and Tolerable in Patients with Myelodysplastic Syndromes Refractory to Hypomethylating Agents. Blood. 132:233.

Zhang, W., C. Ly, J. Ishizawa, et al. (2018). Combinatorial Targeting of XPO1 and FLT3 Exerts Synergistic Anti-Leukemia Effects Through Induction of Differentiation and Apoptosis in FLT3- mutated Acute Myeloid Leukemias: From Concept to Clinical Trial. Haematologica. 103:1642-1653.

Page 7 of 7