-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

171 Salicylates Potentiate and Broaden CRM1 Inhibitor Anti-Tumor Activity Via S-Phase Arrest and Impaired DNA-Damage RepairClinically Relevant Abstract

Program: Oral and Poster Abstracts
Type: Oral
Session: 625. Lymphoma: Pre-Clinical—Chemotherapy and Biologic Agents: Novel Approaches to Overcome Resistance
Hematology Disease Topics & Pathways:
Follicular Lymphoma, multiple myeloma, Leukemia, Marginal Zone Lymphoma, drug-drug interaction, Diseases, CLL, Lymphoma (any), Mantle Cell Lymphoma, Therapies, Combinations, Non-Hodgkin Lymphoma, DLBCL, B-Cell Lymphoma, T-Cell Lymphoma, Plasma Cell Disorders, Lymphoid Malignancies, Clinically relevant
Saturday, December 5, 2020: 12:00 PM

Jithma P. Abeykoon, MD1,2, Xiaosheng Wu, MD1*, Kevin E. Nowakowski1*, Surendra Dasari, PhD3*, Jonas Paludo, MD1, S. John Weroha2*, Chunling Hu, PhD2*, Jann Sarkaria2*, Ann Mladek2*, Jessica Phillips, PhD2*, Andrew L. Feldman, MD4, Aishwarya Ravindran, MBBS5, Rebecca L. King, MD4, Mary J. Stenson1*, Michelle Manske, MS, BS1*, Fergus Couch2*, Mrinal M. Patnaik, MD, MBBS1 and Thomas E. Witzig, MD1

1Division of Hematology, Mayo Clinic, Rochester, MN
2Mayo Clinic, Rochester, MN
3Department of Health Sciences Research, Mayo Clinic, Rochester, MN
4Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
5Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN

Chromosome region maintenance protein1 (CRM1) mediates protein export from the nucleus and is a new target for anti-cancer therapeutics. Broader application of KPT-330 (selinexor), a first in class CRM1 inhibitor recently approved for multiple myeloma and diffuse large B-cell lymphoma (DLBCL), has been limited by substantial adverse effects (AEs). To address this clinical problem, we focused on identifying novel strategies to boost the potency, reduce toxicity, and broaden the applicability of CRM1 inhibitors to a wider range of malignancies.

We discovered that salicylates could markedly enhance the anti-tumor activity of CRM1 inhibitors by extending the mechanisms of action beyond CRM1 inhibition. KPT-330 was chosen as the prototypical CRM1 inhibitor given its current FDA approval status and characterized pharmacokinetics; and choline salicylate (CS) was chosen as the prototypical salicylate given its favorable pharmacokinetics and reduced antiplatelet, renal, neurological and gastrointestinal AEs in humans compared to other salicylates. By using cell lines belonging to different hematologic malignancies and solid tumors, we demonstrated ex vivo that the combination of KPT-330 and CS (K+CS) could induce unique and significant antitumor effect at much lower dose of KPT-330 (at 25% of the dose used in the clinic), thereby potentially mitigating prohibitive clinical AEs (Figure 1a-d, and e-g). This significant synergetic antitumor effect observed with K+CS ex vivo was also validated in vivo by using an NSG mouse model of mantle cell lymphoma (Figure 1h). Moreover, the K+CS combination did not show this potent toxic effect on non-malignant cells in vivo and was safe without inducing toxicity to normal organs in NSG mice.

Mechanistically, protein profiling through mass spectroscopy revealed that K+CS uniquely affects the cellular pathways of DNA damage repair, DNA synthesis and cell cycle progression. Studies involving immunoblotting, cell cycle analysis, immunofluorescence microscopy assessing nucleocytoplasmic molecular export and DNA damage, reporter assay to assess homologous recombination repair proficiency and immunohistochemistry showed that, compared to KPT-330 treatment alone, K+CS decreased the expression of CRM1, Rad51 and thymidylate synthase proteins in vitro and in vivo, leading to more efficient inhibition of CRM1-mediated nuclear export, impairment of DNA-damage repair, reduced pyrimidine synthesis, and importantly a unique cell cycle arrest in S-phase, thus leading to cell apoptosis. These effects on cellular proteins and pathways were unique to K+CS treatment and were not observed with KPT-330 or CS single agent treatment. Pathway analyses through RNA sequencing also paralleled the findings of proteomic studies thus further validating the unique effect of K+CS treatment on the aforementioned cellular pathways.

Importantly, K+CS treatment exerted unique and significant antitumor effect ex vivo on primary malignant cells obtained from patient’s with high-risk hematologic malignancies such as double hit DLBCL, BTK and BCL2 inhibitor resistant mantle cell lymphoma, TP53 deleted/mutated chronic lymphocytic leukemia and high-risk multiple myeloma thus signifying its broader applicability as a treatment option for these aggressive hematologic malignancies where there is a dire need to find new treatment strategies (Figure 1i).

In summary, we describe a unique all-oral drug combination with a novel constellation of mechanisms of action on cellular pathways that are exploited by cancer cells. K+CS represents a new class of therapy for multiple cancer types and will stimulate future investigations to exploit DNA-damage repair and nucleocytoplasmic transport for therapy of blood cancers.

Disclosures: Witzig: Incyte: Consultancy; Acerta: Research Funding; Karyopharm Therapeutics: Research Funding; Immune Design: Research Funding; AbbVie: Consultancy; Spectrum: Consultancy; Celgene: Consultancy, Research Funding; MorphSys: Consultancy.

Previous Abstract | Next Abstract >>
*signifies non-member of ASH