-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.

950 Targeting MM at the Nexus between Cell Cycle and Transcriptional Regulation Via CDK7 Inhibition

Program: Oral and Poster Abstracts
Session: 603. Oncogenes and Tumor Suppressors: Poster I
Hematology Disease Topics & Pathways:
multiple myeloma, Diseases, cell regulation, Biological Processes, Plasma Cell Disorders, Lymphoid Malignancies, Study Population, pathways, signal transduction
Saturday, December 5, 2020, 7:00 AM-3:30 PM

Yao Yao1*, Woojun D Park2*, Eugenio Morelli, MD3, Mehmet Kemal Samur, PhD4*, Nicholas P Kwiatkowski, PhD5*, Yan Xu, MD6*, Chandraditya Chakraborty, PhD7*, Behnam Nabet, phD8*, Marta Chesi, PhD9, Herve Avet-Loiseau10,11, Nathaniel Gray12*, Richard A. Young, PhD13,14*, Kenneth Anderson, MD6,15,16,17, Charles Y Lin, PhD18,19*, Nikhil C. Munshi, MD17,20,21,22 and Mariateresa Fulciniti, PhD6

1Dana Farber Cancer Institute, Boston, MA
2Department of Molecular and Human Genetics, Baylor College of Medicine, Huston, TX
3Department of Medical Oncology, DANA FARBER CANCER INSTITUTE, Boston, MA
4Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
5Biological Chemistry and Molecular Pharmacology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
6Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
7Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
8Dana farber cancer institute, Boston, MA
9Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ
10Unite de Genomique du Myelome, IUC-T Oncopole, Toulouse, France
11Universite Paul Sabatier, Toulouse, France
12Dana-Farber Cancer Institute, Boston, MA
13Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
14Whitehead Institute for Biomedical Research, Cambridge, MA
15Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
16Dana-Farber Cancer Institute, Medical Oncology, Harvard Medical School, Boston, MA
17Harvard Medical School, Boston, MA
18Dan L. Duncan Cancer Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
19Dan L. Duncan Cancer Center, Department of Molecular and Human Genetics, Baylor College of Medicine, HOUSTON, TX
20Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
21VA Boston Healthcare System, Boston, MA
22The LeBow Institute for Myeloma Therapeutic and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA

Deregulated transcription and cell cycle control are hallmarks of cancer that are especially frequent in multiple myeloma (MM). Largely non-overlapping sets of cyclin-dependent kinases (CDKs) regulate cell division and RNA polymerase II (Pol II)-dependent transcription; and targeting of cell cycle CDKs has been long pursued as an attractive therapeutic strategy. Among CDKs, CDK7 presents a unique therapeutic opportunity as it functions as a CDK activating kinase (CAK), licensing the activity of cell cycle CDKs, and also serves as a core component of the general transcription factor TFIIH.

Here we elucidated the biological role of CDK7 and its transcriptional regulatory landscape in MM, using genetic as well chemical approaches, including tools for CDK7 rapid protein degradation (dTAG) and the selective covalent inhibitor YKL-5-124 that targets a cysteine residue (C312) located outside of the kinase domain.

We have observed that CDK7 inhibition via YKL-5-124 robustly inhibited the phosphorylation of the CDK1, 2 and 4 activation loops in a representative panel of MM cell lines at concentrations as low as 50 nM. This reduction was not observed in MM cells expressing a resistant mutation in the reactive cysteine (C312S). Consistent with decrease of CAK activity, we observed G1 arrest and S phase loss after CDK7 inhibition, which was also associated with a rapid and transient loss of Ser2 and Ser5 phosphorylation of the RNA Pol2 C-terminal domain.

To understand the effect of CDK7 inhibition on MM cell growth and viability, we evaluated activity of YKL-5-124 across a large panel of 25 MM cell lines and observed a significant inhibition of MM cell proliferation, with a significantly lower IC50 compared to PHA-activated normal donor peripheral blood mononuclear cells (PBMCs), suggesting a specific sensitivity of MM cells to CDK7 inhibition. Longer exposure to YKL-5-124 caused apoptotic cell death in MM cells; however treatment with an inactive analog or in cells expressing the C312S mutation failed to inhibit MM cell proliferation, confirming that the antiproliferative potency of YKL-5-124 resides in its unique characteristic to covalently bind to C312 domain. Importantly, CDK7 inhibition impaired primary MM cells proliferation alone and when cultured in the presence of BM microenvironment. Selective pharmacological degradation of endogenously tagged CDK7 confirmed impact of CDK7 inhibition on MM cell proliferation via inhibition of CDK7 transcriptional and cell cycle activities. To complement the pharmacological studies, we have established MM cells to express inducible CRISPR/Cas9 constructs encoding 4 independent small guide RNAs targeting CDK7, resulting in the reduction of the abundance of CDK7 protein by 20–60% which was sufficient to inhibit MM cell viability over time, phenocopying pharmacologic inhibition of CDK7. These results support the view that CDK7 is a pharmacologically relevant target for MM.

Gene expression analysis after CDK7 inhibition in MM1S and H929 cells revealed that transcripts for only a subset of genes were substantially affected by treatment with low dose of YKL-5-124, showing a strong leading-edge enrichment for downregulation of E2F expression program, cell cycle, DNA damage, and MYC targets. We have indeed confirmed a potent reduction in phosphorylation of RB protein, with consequent decrease of E2F activity in MM cells confirmed using E2F-driven luciferase reporter. These data suggest significant role for CDK7 in the CDK-pRB-E2F pathway in MM, which was strengthened by the observation of a positive correlation between expression of CDK7 and expression of E2F target genes in primary MM cells (n=409).

Finally, we have evaluated the in vivo effect of CDK7 inhibition in several murine models of human MM. In the localized subcutaneous model, and the disseminated MM model where treatment with YKL-5-124 decreased tumor burden and improved survival. The effect of CDK7 inhibition explored in an aggressive, genetically engineered model of Myc-dependent MM, revealed evidence of response by decline in measurement of monotypic serum immunoglobulins.

In conclusion, our study demonstrates that CDK7 contributes to the ‘transcriptional addiction’ and the cell cycle deregulation frequently observed in MM and represents an attractive molecular vulnerability to be exploited therapeutically.

Disclosures: Anderson: Millenium-Takeda: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Sanofi-Aventis: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Oncopep and C4 Therapeutics.: Other: Scientific Founder of Oncopep and C4 Therapeutics.; Celgene: Membership on an entity's Board of Directors or advisory committees. Munshi: Takeda: Consultancy; Karyopharm: Consultancy; AbbVie: Consultancy; Amgen: Consultancy; Legend: Consultancy; Adaptive: Consultancy; Janssen: Consultancy; C4: Current equity holder in private company; OncoPep: Consultancy, Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties; BMS: Consultancy. Fulciniti: NIH: Research Funding.

*signifies non-member of ASH