-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.

1591 Deconvoluting Clonal and Cellular Architecture in IDH-Mutant Acute Myeloid Leukemia

Program: Oral and Poster Abstracts
Session: 617. Acute Myeloid Leukemias: Biomarkers, Molecular Markers and Minimal Residual Disease in Diagnosis and Prognosis: Poster I
Hematology Disease Topics & Pathways:
Research, Acute Myeloid Malignancies, adult, Translational Research, CHIP, genomics, hematopoiesis, Diseases, Therapies, Myeloid Malignancies, Biological Processes, Study Population, Human
Saturday, December 9, 2023, 5:30 PM-7:30 PM

Maria Sirenko, PhD1,2,3,4,5, Soobeom Lee2,4,5,6*, Zhengxi Sun2,4,5*, Ronan Chaligne, PhD7*, Georgios Asimomitis3,8*, Charlotte K Brierley, bmbch9, Elsa Bernard, PhD3*, Sheng F. Cai, MD, PhD10*, Robert M Myers, PhD11, Bettina Nadorp, PhD4,12*, Max F Levine3*, Dylan Domenico3*, Juan E Arango Ossa3*, Juan Santiago Medina Martinez3*, Kamal Menghrajani, MD10, Audrey Lasry, PhD4*, Alice Mims, MD13,14, Helee Desai13,14*, Andrea Laganson, MS13,14*, Christopher Famulare, MS15*, Minal Patel, BA15*, Gerard Lozanski, MD16*, Sanam Loghavi, MD17, Kelly L. Bolton, MD10,18, Aaron D. Viny, MD, MS19,20, Mikhail Roshal, MD, PhD21*, Ross L Levine, MD22, Eytan M. Stein10, Dan A. Landau, MD, PhD23,24,25,26*, Ann-Kathrin Eisfeld, MD*14,26,27,28, Iannis Aifantis, PhD4,26,29 and Elli Papaemmanuil, PhD3,26

1Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY
2Contributed equally, *
3Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
4Department of Pathology, NYU Grossman School of Medicine, New York, NY
5Laura & Isaac Perlmutter Cancer Center, NYU Langone, New York, NY
6Department of Biology, New York University, New York
7Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
8Biomedical Systems Laboratory, Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece
9Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM) and NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
10Leukemia Service, Department of Medicine, Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
11Tri-Institutional MD-PhD Program, Weill Cornell Medicine, Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY
12Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, NY
13Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH
14Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
15Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
16Department of Pathology, The Ohio State University, Columbus, OH
17Hematopathology, MD Anderson Cancer Center, Houston, TX
18Department of Medicine, Division of Oncology, Section of Stem Cell Biology, Washington University School of Medicine, Saint Louis, MO, Washington University In St Louis, St Louis, MO
19Leukemia Service, Department of Medicine, Center for Hematologic Malignancies,, Memorial Sloan Kettering Cancer Center, New York, NY
20Columbia Stem Cell Initiative, Columbia University, New York, NY
21Memorial Sloan Kettering Cancer Center, New York
22Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
23New York Genome Center, New York
24Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York
25Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York
26Corresponding author, #
27Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University, Columbus, OH
28Pelotonia Institute for Immuno-Oncology, OSUCCC – James, The Ohio State University, Columbus, OH
29New York University, Grossman School of Medicine, New York, NY

Introduction

Mutations in Isocitrate dehydrogenase 1/2 (IDH) occur in 20% of acute myeloid leukemias (AML) and frequently co-occur with DNMT3A, NPM1, SRSF2, and NRAS. In clonal hematopoiesis (CH), IDH mutations confer high risk of AML transformation. Recently developed methods linking single cell genotype to single cell transcriptomic profiles and cell lineage offer the opportunity to define the transcriptomic consequences of each mutation across leukemic cell lineages and clonal phylogenies. Here, we develop an integrative single cell laboratory and analytical framework to study the genetic, clonal and cellular heterogeneity of IDH mutated CH and AML, across disease stages (diagnosis and relapse) and deliver a high-resolution map of IDH-mutant AML.

Methods

Bone marrow or peripheral blood mononuclear samples from 15 AML patients, 2 CH individuals, and 12 unaffected age-matched donors were profiled by single cell RNA-sequencing. For the AML cases, a median of 3 samples per patient were profiled under treatment with IDH inhibitor (IDHi) monotherapy (n=5 patients), IDHi in combination with intensive chemotherapy (NCT02632708, n=6) or in combination with hypomethylating agent (n=2). This dataset comprised 259,610 cells across AML (n=190,133), CH (n=16,705) and normal controls (n=52,772). Genotyping of Transcriptomes (Nam et al., 2019) (for IDH1/2, NPM1, SRSF2, and NRAS mutations) across 6 AML patients enabled genotyping 23% of cells at diagnosis for at least one clone-defining mutation.

Results

Through meta-analysis of 3,653 diagnostic AML samples profiled by a targeted DNA panel (Papaemmanuil et al., 2016; Tazi et al., 2022), we identified DNMT3A, SRSF2 and NPM1 as critical co-operative mutations in IDH-mutant AML. Amongst the 15 AML patients, 6 had IDH1 mutation, 8 had IDH2 mutations, and 1 had both IDH1 and IDH2. The co-occurring mutations in DNMT3A (n=11 patients), NPM1 (n=5), SRSF2 (n=5) reflected the genomic landscape of IDH-mutant AML.

At diagnosis, IDH-mutant AML was enriched in immature hematopoietic stem and progenitor-like blasts. Compared to normal hematopoiesis, IDH-mutant AML blasts upregulated inflammatory response markers including TNFɑ/NFκB-signaling and downregulated proliferation-associated pathways including G2M Checkpoint, and MYC and E2F Targets, and DNA damage response.

To investigate how co-mutations modify the IDH-mutant phenotype, we established an integrative single–cell profiling approach to assign single cells to distinct phylogenetic subclones and extract clone-specific transcriptional programs. SRSF2 and NPM1 co-mutations modulated the AML blast phenotype, towards erythroid (SRSF2) or granulomonocytic lineage (NPM1). NRAS- or SRSF2-mutant subclones defined distinct gene expression-derived clusters and upregulated proliferation and metabolic pathways. Furthermore, NPM1 co-mutated blasts upregulated KMT2A-target genes.

Next, we tracked clonal responses to combination IDHi and chemotherapy using GoT and targeted DNA sequencing. In 4 cases, the ancestral clone, defined by IDH and DNMT3A mutations, retained clonal dominance in both mature and immature myeloid compartments at complete response (median 4 weeks). Conversely, subclones defined by mutations in NPM1, NRAS, or SRSF2, were either reduced or not detectable (limit of detection 2%). In two cases, mutations were not detected after therapy.

In patients who relapsed after IDHi monotherapy treatment, blasts upregulated stemness, fatty acid metabolism, OxPHOS, and inflammation at relapse compared to diagnosis. Furthermore, MHC Class II was downregulated at relapse, pointing to immune evasion of AML as a potential relapse mechanism.

Finally, we investigated the role of IDH2 mutations at CH. IDH-mutant CH-derived monocytes upregulated inflammation-associated pathways, mirroring the phenotypes observed in IDH-mutant AML at diagnosis.

Conclusion

We defined how IDH mutations and their frequent co-mutations dysregulated hematopoiesis. We leveraged state-of-the-art single cell techniques to assign leukemic cells to phylogenetic subclones and delineated the role of serially acquired co-mutations in shaping lineage bias and transcriptional programs. Furthermore, we defined genetic, clonal and transcriptional changes at three stages of the AML disease process: diagnosis, remission, and relapse under IDH inhibitor therapy.





Disclosures: Cai: Imago Biosciences: Consultancy, Current equity holder in private company. Levine: Isabl Inc: Current Employment. Medina Martinez: Isabl Inc.: Current Employment, Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees. Menghrajani: Gilead: Consultancy. Mims: Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees. Patel: Isabl Inc: Current Employment. Loghavi: Recordati/ EUSA Pharma: Consultancy; Blueprint Medicine: Consultancy; Abbvie: Consultancy; Gerson Lehrman Group: Consultancy; QualWorld: Consultancy; Guidepoint: Consultancy; Caris Diagnostics: Consultancy; Astellas: Research Funding; Amgen: Research Funding; Abbvie: Current equity holder in publicly-traded company; Daiichi Sankyo: Consultancy. Bolton: Servier: Research Funding; GoodCell: Membership on an entity's Board of Directors or advisory committees. Viny: Arima Genomics: Membership on an entity's Board of Directors or advisory committees. Roshal: Celgene: Other: Provision of services; Auron Therapeutics: Other: Ownership/Equity interests; Provision of services; Physicians' Education Resource: Other: Provision of services; Roche: Other: Funding; Beat AML: Other: Funding; NGM: Other: Funding. Levine: Prelude: Membership on an entity's Board of Directors or advisory committees; C4 Therapeutics: Membership on an entity's Board of Directors or advisory committees; Isoplexis: Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy; Qiagen: Membership on an entity's Board of Directors or advisory committees; Zentalis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Incyte: Consultancy; AstraZeneca: Consultancy, Honoraria; Lilly: Honoraria; Roche: Honoraria; Novartis: Consultancy; Auron: Membership on an entity's Board of Directors or advisory committees; Ajax: Membership on an entity's Board of Directors or advisory committees, Research Funding; Mission Bio: Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria. Stein: Genentech: Consultancy; Abbvie: Consultancy; Neoleukin: Consultancy; Calithera: Consultancy; Genesis: Consultancy; Eisai: Research Funding; Novartis: Consultancy; Syros: Consultancy; Menarini: Consultancy; Jazz: Consultancy; OnCusp: Consultancy; Daiichi: Consultancy; Foghorn: Consultancy; CTI Biopharma: Consultancy; Aptose: Consultancy; Servier: Consultancy; Syndax: Consultancy; Gilead: Consultancy; Agios: Consultancy; Janssen: Consultancy; Bristol Myers Squib: Consultancy, Research Funding; PinotBio: Consultancy; Astellas: Consultancy; Ono Pharma: Consultancy; Blueprint: Consultancy. Landau: BMS: Research Funding; Mission Bio: Membership on an entity's Board of Directors or advisory committees; 10X Genomics: Research Funding; Pangea: Membership on an entity's Board of Directors or advisory committees; Alethiomics: Membership on an entity's Board of Directors or advisory committees; Illumina: Consultancy, Research Funding; C2i Genomics: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Consultancy; Abbvie: Consultancy; Ultima Genomics: Research Funding. Eisfeld: Karyopharm Therapeutics: Other: spouse employment; Astra Zeneca: Honoraria, Other: CEI Advisory Board; OncLive: Honoraria. Aifantis: Foresite Labs: Consultancy; AstraZeneca: Research Funding.

*signifies non-member of ASH