-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.

527 Overcoming NOTCH1-Driven Chemoresistance in T-Cell Acute Lymphoblastic Leukemia Via Metabolic Intervention with Oxphos Inhibitor

Program: Oral and Poster Abstracts
Type: Oral
Session: 605. Molecular Pharmacology, Drug Resistance—Lymphoid and Other Diseases: Molecular pharmacology and drug resistance mechanisms in lymphoproliferative disorders
Hematology Disease Topics & Pathways:
Leukemia, ALL, apoptosis, Diseases, drug-drug interaction, Therapies, Combinations, Biological Processes, DNA damage, Lymphoid Malignancies, integrative -omics, metabolomics, proteomics, signal transduction
Monday, December 7, 2020: 7:45 AM

Natalia Baran, PhD, MD1, Alessia Lodi, PhD2*, Yogesh Dhungana, PhD3*, Shannon RENEE Sweeney, PhD4*, Pandey Renu, PhD4*, Shelley Herbrich, PhD5, Fieke W Hoff, BSc6*, Anna Skwarska, PhD5,7*, Mecit Kaplan, MS8*, Vinitha Mary Kuruvilla, MSc5*, Antonio Cavazos, MSc5*, Marc Warmoes, PhD9*, Shanti Rojas-Sutterlin, PhD10*, Andre Haman, PhD10*, Diogo Troggian Veiga, PhD11*, Ken Furudate, DDM, PhD5*, Helen Ma, MS5*, Karine Harutyunyan, PhD5*, Wentao Yang12*, Mihai Gagea, MD PhD13*, Di Du, PhD9*, May Daher14, Luciana Melo Garcia, MD8*, Sujan Piya, PhD15, Vivian Ruvolo, MS16*, Sriram Shanmugavelandy17*, Ningping Feng, PhD18*, Jason P Gay, MS18*, Ondrej Havranek, MD, PhD19, Jared Henderson, MS20*, Katarzyna Tomczak, PhD21*, Marcin Kaminski, PhD22*, Daniel Herranz, PharmD23, Adolfo Ferrando, M.D., PhD24*, Elias Jabbour, MD5, Maria E Di Francesco, PhD18*, David T. Teachey, MD25, Terzah M. Horton, MD, PhD26, Katayoun Rezvani, MD, PhD8, Jun J. Yang, PhD12, Steven M. Kornblau, MD16, Eric Davis, MD20*, Koichi Takahashi, MD, PhD5, Michael Andreeff, MD, PhD16, Joseph R Marszalek, PhD27*, Philip Lorenzi, PhD9, Jiyang Yu, Ph.D.28*, Stefano Tiziani, PhD2, Trang Hoang, PhD10 and Marina Konopleva, MD, PhD5

1Department of Leukemia, MD Anderson Cancer Center, Houston, TX
2Department of Nutritional Sciences & Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX
3Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN
4Dell Pediatric Research Institute (DPRI), The University of Texas at Austin, Austin, TX
5Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
6Department of Pediatric Oncology/ Hematology, University Medical Center Groningen, Groningen, Netherlands
7Department of Genetics, Medical University of Bialystok, Bialystok, Poland
8Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
9Dept. of Bioinformatics and Computational Biology and The Proteomics and Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX
10IRIC - University of Montreal, Montreal, QC, Canada
11The Jackson Laboratory for Genomic Medicine, Farmington, CT
12Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
13Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
14Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
15Department of Leukemia, The University of Texas MD Anderson Cancer Center, Pearland, TX
16Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
17Abbvie, Lake Bluff, IL
18Institute for Applied Cancer Science and Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX
19Institute of Biotechnology, Czech Academy of Sciences, Vestec, Czech Republic
20Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
21Department of Translational Molecular Pathology, UT M. D. Anderson Cancer Center, Houston, TX
22Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
23Cancer Institute of NJ, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
24Department of Pathology, Columbia University Medical Center, New York, NY
25Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
26Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
27Institute for Applied Cancer Science & Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX
28Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN

The inferior cure rate of T-cell acute lymphoblastic leukemia (T-ALL) is associated with inherent drug resistance. The activating NOTCH1 gene mutations have been reported to cause chemoresistance at the stem cell level1. Direct NOTCH1 inhibition has failed in clinical trials due to a narrow therapeutic window but targeting key oncogenic and metabolic pathways downstream of mutated NOTCH1 may offer novel approaches. We previously reported that rapid transformation of thymocytes at the DN3 differentiation stage into preleukemic stem cells (pre-LSC) requires elevated Notch1 in addition to the presence of Scl/Lmo11. Notably, we showed that cellular metabolism of NOTCH1-mutated T-ALLs depends on Oxidative Phosphorylation (OxPhos) and that OxPhos inhibition using the complex I inhibitor IACS-010759 (OxPhos-i) is efficacious in NOTCH1-mutated T-ALL patient derived xenografts (PDXs)2.

Here, we investigated the link between NOTCH1-mutated chemoresistance and OxPhos in pre-leukemic and leukemic cells, utilizing comprehensive molecular and functional assays.
We hypothesized that chemotherapy aided by OxPhos-i overcomes chemoresistance, depletes LSCs and combats T-ALL.

First, we analyzed the role of OxPhos in downstream Notch1 targets at the pre- and leukemic stage considering four stages of thymocyte differentiation (D1-D4), in a mouse model of human T-ALL1. Gene set enrichment analysis (GSEA) implicated increased expression of Notch1 target genes starting at DN1, and OxPhos target genes were the highest-ranked gene set at DN3.

Next, activation of Notch1 by its ligand DL4 and inhibition of OxPhos reduced viability of pre-LSCs, indicating that ligand-dependent activation of Notch1 signaling upregulates the OxPhos pathway and sensitizes pre-LSCs to OxPhos-i. To clarify the role of Notch1 signaling, we examined the effect of IACS-010759 on pre-leukemic thymocytes harboring LMO1, SCL-LMO1, NOTCH1, LMO1-NOTCH1 and SCL-LMO1-NOTCH1 with and without DL4 stimulation. We found that in the absence of DL4, only thymocytes harboring the Notch1 oncogene responded to OxPhos-i, whereas all DL4-stimulated thymocytes responded regardless of Notch1 status (Fig. 1a). In addition, at the leukemic stage, we found elevation of the OxPhos pathway driven by oncogenic Notch1 when we compared transcriptomes of SCL-LMO1 induced T-ALL in the presence or absence of the NOTCH1 oncogene.

In line with the murine T-ALL NOTCH1 model, we performed transcriptome analysis of two independent T-ALL patient cohorts prior to chemotherapy, COG TARGET ALL (n=263) and AALL1231 (n=75), comparing transcriptomes of NOTCH1-mutated vs NOTCH1-wt T-ALLs. We found co-segregation of NOTCH1 mutations with significant upregulation of OxPhos and TCA cycle genes and downregulation of apoptosis signaling. Aiming to reverse the NOTCH1-controlled anti-apoptotic program and chemoresistance, we next tested the combination of Vincristine, Dexamethasone and L-Asparaginase (VXL) with IACS-010759. When compared to vehicle, OxPhos-i or VXL alone, only the VXL-OxPhos-i treatment caused an energetic crisis indicated by decreased OCR and ECAR (Seahorse), which translated to a profound reduction of viability (CTG, flow cytometry) in T-ALL cell lines (n=9) and primary T-ALL samples (n=5). Additionally, the IACS-VXL combination in vivo resulted in pan-metabolic blockade, which caused metabolic shut-down and triggered early induction of apoptosis in leukemic cells in peripheral blood, spleen and bone marrow (Fig. 1b). Single cell Proteomic analysis (CyTOF) of spleen showed reduced expression of cell proliferation marker -ki67, c-myc, ERK and p38 proteins, and reduction in number of leukemic cells. Finally, this combination therapy resulted in reduced leukemia burden and extension of overall survival across all three aggressive NOTCH1-mutated T-ALL PDX models (p<0.0001) (Fig.1 c, d). In summary, we demonstrated that targeting OxPhos with IACS-010759 in combination with chemotherapy facilitates eradication of chemoresistant NOTCH1-driven T-ALL through direct targeting of the key metabolic regulators of OxPhos conferred by mutant NOTCH1 in T-ALL. Clinical trials rewiring metabolism by incorporation of OxPhos-i to standard-of-care therapy in patients with NOTCH1-mutated T-ALL are warranted to improve patients’ outcomes.

Disclosures: Jabbour: Pfizer: Other: Advisory role, Research Funding; Genentech: Other: Advisory role, Research Funding; BMS: Other: Advisory role, Research Funding; Takeda: Other: Advisory role, Research Funding; Amgen: Other: Advisory role, Research Funding; Adaptive Biotechnologies: Other: Advisory role, Research Funding; AbbVie: Other: Advisory role, Research Funding. Teachey: Sobi: Consultancy; Amgen: Consultancy; Janssen: Consultancy; La Roche: Consultancy. Rezvani: Takeda: Other: Licensing agreement; GemoAb: Membership on an entity's Board of Directors or advisory committees; Adicet Bio: Membership on an entity's Board of Directors or advisory committees; Virogen: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Other: Educational grant; Affimed: Other: Educational grant; Formula Pharma: Membership on an entity's Board of Directors or advisory committees. Andreeff: Daiichi-Sankyo; Jazz Pharmaceuticals; Celgene; Amgen; AstraZeneca; 6 Dimensions Capital: Consultancy; Daiichi-Sankyo; Breast Cancer Research Foundation; CPRIT; NIH/NCI; Amgen; AstraZeneca: Research Funding; Centre for Drug Research & Development; Cancer UK; NCI-CTEP; German Research Council; Leukemia Lymphoma Foundation (LLS); NCI-RDCRN (Rare Disease Clin Network); CLL Founcdation; BioLineRx; SentiBio; Aptose Biosciences, Inc: Membership on an entity's Board of Directors or advisory committees; Amgen: Research Funding. Lorenzi: Precision Pathways: Consultancy. Konopleva: Calithera: Research Funding; Kisoji: Consultancy; AbbVie: Consultancy, Research Funding; Sanofi: Research Funding; Genentech: Consultancy, Research Funding; F. Hoffmann La-Roche: Consultancy, Research Funding; Cellectis: Research Funding; Rafael Pharmaceutical: Research Funding; Eli Lilly: Research Funding; Reata Pharmaceutical Inc.;: Patents & Royalties: patents and royalties with patent US 7,795,305 B2 on CDDO-compounds and combination therapies, licensed to Reata Pharmaceutical; Agios: Research Funding; AstraZeneca: Research Funding; Ablynx: Research Funding; Forty-Seven: Consultancy, Research Funding; Amgen: Consultancy; Stemline Therapeutics: Consultancy, Research Funding; Ascentage: Research Funding.

*signifies non-member of ASH