1
|
Wang B, Reville PK, Abbas HA. Therapeutic hurdles in acute myeloid leukemia: Leukemic stem cells, inflammation and immune dysfunction. Curr Opin Pharmacol 2025; 82:102526. [PMID: 40318269 DOI: 10.1016/j.coph.2025.102526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/07/2025]
Abstract
Acute myeloid leukemia (AML) is an aggressive and highly heterogeneous hematological malignancy characterized by clonal expansion and differentiation arrest in myeloid progenitor cells. Despite advancements in chemotherapy, allogeneic hematopoietic stem cell transplantation, and post-remission maintenance therapies, the long-term survival remains unsatisfactory with high rates of relapse and refractory. These therapeutic challenges are mediated by multiple factors, including the complexity of the cellular hierarchies in AML, the interaction of leukemic stem cells (LSCs) with the bone marrow niche, inflammation, and immune evasion mechanisms. Further, the absence of specific surface markers that distinguish LSCs from normal hematopoietic stem cells, together with LSCs' functional heterogeneity, complicates targeted treatment approaches. Immune dysfunction, including T cell exhaustion and immune suppression within the bone marrow niche contributes to therapy resistance. In this brief review, we aim to explore current challenges in AML therapy, focusing on LSC-driven resistance, immune evasion, and the need for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Bofei Wang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick K Reville
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Abbas
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Wang T, Zhao L, Ma H. Impact of secondary-type mutations on the prognosis of AML patients with NPM1 mutation: a systematic review and meta-analysis. Ann Hematol 2025:10.1007/s00277-025-06431-w. [PMID: 40434717 DOI: 10.1007/s00277-025-06431-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/22/2025] [Indexed: 05/29/2025]
Abstract
Nucleophosmin 1 (NPM1) mutation is commonly associated with a favorable prognosis in acute myeloid leukemia (AML). Conversely, secondary mutations such as those in ASXL1, RUNX1, EZH2, and SRSF2 are generally linked to poor outcomes. The combined prognostic impact of NPM1 and secondary mutations in AML patients remains unclear. This meta-analysis aimed to evaluate the prognostic significance of secondary mutations in AML patients harboring NPM1 mutation. A systematic literature search was conducted following PRISMA guidelines, identifying studies published up to June 2024 from databases such as PubMed, Web of Science, and the Cochrane Library. The inclusion criteria included adult AML patients with confirmed NPM1 mutation, detailed reporting of secondary mutations, and comparative prognostic outcomes. Fourteen high-quality studies from twelve publications were included, encompassing 4,022 patients who all carried NPM1 mutations; among these, 618 also harbored secondary mutations. Data extraction and quality assessment were performed independently by two researchers via the Newcastle-Ottawa Scale (NOS). Statistical analyses involved fixed-effects models due to low heterogeneity (I²=0% for OS and I²=35% for EFS/RFS). Publication bias and sensitivity analyses confirmed the robustness of the findings. Secondary mutations were not significantly associated with OS (HR = 1.16, 95% CI: 0.99-1.35, p = 0.07) or EFS/RFS (HR = 1.15, 95% CI: 0.96-1.38, p = 0.14) in the overall NPM1-mutated AML population. However, within the European LeukemiaNet (ELN) favorable prognosis group, the presence of secondary mutations was significantly associated with reduced OS (HR = 1.95, 95% CI: 1.39-2.73, p < 0.01). Subgroup analyses based on median age, geographical region, and allogeneic hematopoietic stem cell transplantation (allo-HSCT) rates did not reveal significant modifiers of the prognostic impact of secondary mutations. Secondary mutations do not significantly adversely affect OS or EFS/RFS in the general population of AML patients with NPM1 mutation. Notably, within the ELN favorable prognosis group, secondary mutations are associated with markedly poorer OS, highlighting the need for careful prognostic assessment and potential treatment strategy adjustments in this subset of patients.
Collapse
Affiliation(s)
- Tao Wang
- Department of Hematology and Institute of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Lei Zhao
- Department of Hematology and Institute of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Hongbing Ma
- Department of Hematology and Institute of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
3
|
Shaw R, Yoon JJ, Johnston H, Davidson MB, Siddon AJ, Shallis RM, Chen EC, Burkart M, Oh TS, Iyer SG, Madarang E, Muthiah C, Kassner J, Rampal RK, Guru Murthy GS, Bradley T, Abaza Y, Garcia JS, Gupta V, Pettit KM, Odenike O, Patel AA. Assessing AML-based risk criteria in patients with accelerated/blast-phase MPN treated with less-intensive therapy. Blood Adv 2025; 9:2453-2457. [PMID: 39969200 PMCID: PMC12144506 DOI: 10.1182/bloodadvances.2024015712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Reid Shaw
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | - James J. Yoon
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hannah Johnston
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | - Marta B. Davidson
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Alexa J. Siddon
- Departments of Laboratory Medicine and Pathology, Yale School of Medicine, New Haven, CT
| | - Rory M. Shallis
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Evan C. Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Madelyn Burkart
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Timothy S. Oh
- Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sunil G. Iyer
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| | - Ellen Madarang
- Department of Pharmacy, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | | | - Joshua Kassner
- Division of Hematology and Medical Oncology, Weill Department of Medicine, Cornell University, Ithaca, NY
| | - Raajit K. Rampal
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Terrence Bradley
- Department of Pharmacy, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Yasmin Abaza
- Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Vikas Gupta
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Kristen M. Pettit
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Michigan Medical School, Michigan Medicine, Ann Arbor, MI
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | - Anand A. Patel
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| |
Collapse
|
4
|
Kwon AJ, Cantu MD, Madanat YF, Hasserjian RP, Koduru P, Wang S, Tang G, Weinberg OK. Trisomy 8 in De Novo Acute Myeloid Leukemia Lacking MDS-Related Cytogenetics Does Not Significantly Influence Survival. Int J Lab Hematol 2025. [PMID: 40419386 DOI: 10.1111/ijlh.14504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/13/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025]
Abstract
INTRODUCTION The 2022 WHO and ICC classifications identify MDS-related cytogenetic abnormalities and secondary gene mutations (SM) that in de novo disease are diagnostic of myelodysplasia-related AML, which confers a poorer prognosis. While most MDS-related abnormalities overlap between the two classifications, trisomy 8 (+8) is unique to the ICC and has not been previously included as an MDS-related abnormality. In light of this, we sought to determine the prognostic significance of +8 as an MDS-related abnormality in patients with de novo AML lacking other MDS-related cytogenetics. METHODS We identified 337 patients with de novo AML lacking MDS-related cytogenetics other than +8 and analyzed clinicopathologic outcomes, overall survival (OS), and relapse-free survival (RFS). Two groups were identified: AML with SM (n = 195, 57.9%) and AML without SM (n = 142, 42.1%). Fifty-nine (17.5%) patients had +8; 39 (66.1%) of these had at least one SM, while 20 (33.9%) did not. RESULTS Among patients treated with induction or hypomethylating agents (n = 317), OS and RFS were significantly shorter in patients with SM than without (OS: p = 0.001, RFS: p = 0.0004) but not significantly different between patients with and without +8 (OS: p = 0.15, RFS: p = 0.35). Similarly, when cases were limited to those with SM (n = 179), no significant difference in OS or RFS was observed between patients with and without +8 (OS: p = 0.21, RFS: p = 0.30). There was no significant association between +8 and SM (p = 0.15). CONCLUSION In our cohort, unlike MDS-related SM, trisomy 8 does not influence OS or RFS, despite its inclusion in the ICC as an MDS-related abnormality.
Collapse
Affiliation(s)
- Adelaide J Kwon
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Miguel D Cantu
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Yazan F Madanat
- Department of Hematology and Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Robert P Hasserjian
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Prasad Koduru
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Sa Wang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guilin Tang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Olga K Weinberg
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
5
|
Allen B, Bottomly D, Köhnke T, Wang A, Lin HY, Johnson K, Kenna I, Streltsova A, Martin E, Chen R, Savoy L, Long N, Ryabinin P, Kurtz SE, Eide CA, Carlos A, Kaempf A, Liu T, Tognon C, Searles R, Piehowski PD, Gosline SJC, Agarwal A, Chang BH, Barton M, Druker BJ, McWeeney SK, Majeti R, Tyner JW, Zhang H. A CEBPB/IL-1β/TNF-α feedback loop drives drug resistance to venetoclax and MDM2 inhibitors in monocytic leukemia. Blood 2025; 145:2488-2506. [PMID: 40009487 DOI: 10.1182/blood.2024028239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRAT MDM2 inhibitors are promising therapeutics for acute myeloid leukemia (AML) with wild-type TP53. Through an integrated analysis of functional genomic data from primary patient samples, we found that an MDM2 inhibitor, idasanutlin, like venetoclax, is ineffective against monocytic leukemia (French-American-British [FAB] subtype M4/M5). To dissect the underlying resistance mechanisms, we explored both intrinsic and extrinsic factors. We found that monocytic leukemia cells express elevated levels of CEBPB, which promote monocytic differentiation, suppress CASP3 and CASP6, and upregulate MCL1, BCL2A1, and the interleukin (IL-1)/tumor necrosis factor alpha (TNF-α)/NF-κB pathway members, thereby conferring drug resistance to a broad range of MDM2 inhibitors, BH3 mimetics, and venetoclax combinations. In addition, aberrant monocytes in M4/M5 leukemia produce elevated levels of IL-1 and TNF-α, which promote monocytic differentiation and upregulate inflammatory cytokines and receptors, thereby extrinsically protecting leukemia blasts from venetoclax and MDM2 inhibition. Interestingly, IL-1β and TNF-α only increase CEBPB levels and protect M4/M5 cells from these drugs but not M0/M1 leukemia cells. Treatment with venetoclax and idasanutlin induces compensatory upregulation of CEBPB and the IL-1/TNF-α/NF-κB pathway independent of the FAB subtype, indicating drug-induced compensatory protection mechanisms. The combination of venetoclax or idasanutlin with inhibitors that block the IL-1/TNF-α pathway demonstrates synergistic cytotoxicity in M4/M5 AML. As such, we uncovered a targetable positive feedback loop that involves CEBPB, IL-1/TNF-α, and monocyte differentiation in M4/M5 leukemia and promotes both intrinsic and extrinsic drug resistance and drug-induced protection against venetoclax and MDM2 inhibitors.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors
- Sulfonamides/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Drug Resistance, Neoplasm/drug effects
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- CCAAT-Enhancer-Binding Protein-beta/metabolism
- CCAAT-Enhancer-Binding Protein-beta/genetics
- Leukemia, Monocytic, Acute/drug therapy
- Leukemia, Monocytic, Acute/metabolism
- Leukemia, Monocytic, Acute/pathology
- Leukemia, Monocytic, Acute/genetics
- Signal Transduction/drug effects
- Cell Line, Tumor
- Antineoplastic Agents/pharmacology
- Monocytes
Collapse
Affiliation(s)
- Basil Allen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Thomas Köhnke
- Division of Hematology, Department of Medicine, Cancer Institute and Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA
| | - Anthony Wang
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Hsin-Yun Lin
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Kara Johnson
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Isabel Kenna
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Anastatia Streltsova
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Emma Martin
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Reid Chen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Lindsey Savoy
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Nicola Long
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Peter Ryabinin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Stephen E Kurtz
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Christopher A Eide
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Amy Carlos
- Massively Parallel Sequencing Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Andy Kaempf
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Tingting Liu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Cristina Tognon
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Robert Searles
- Massively Parallel Sequencing Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Paul D Piehowski
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Sara J C Gosline
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Anupriya Agarwal
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Bill H Chang
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Michelle Barton
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR
| | - Brian J Druker
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Shannon K McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Cancer Institute and Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA
| | - Jeffrey W Tyner
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Haijiao Zhang
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| |
Collapse
|
6
|
Ishikawa C, Barreyro L, Sampson AM, Hueneman KM, Choi K, Philbrook SY, Choi I, Bolanos LC, Wunderlich M, Volk AG, Watowich SS, Greis KD, Starczynowski DT. Ubiquitin-conjugating enzyme UBE2N modulates proteostasis in immunoproteasome-positive acute myeloid leukemia. J Clin Invest 2025; 135:e184665. [PMID: 40371639 PMCID: PMC12077902 DOI: 10.1172/jci184665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/06/2025] [Indexed: 05/16/2025] Open
Abstract
Altered protein homeostasis through proteasomal degradation of ubiquitinated proteins is a hallmark of many cancers. Ubiquitination, coordinated by E1, E2, and E3 enzymes, involves up to 40 E2-conjugating enzymes in humans to specify substrates and ubiquitin linkages. In a screen for E2 dependencies in acute myeloid leukemia (AML), ubiquitin conjugating enzyme E2 N (UBE2N) emerged as the top candidate. To investigate UBE2N's role in AML, we characterized an enzymatically defective mouse model of UBE2N, revealing UBE2N's requirement in AML without an impact on normal hematopoiesis. Unlike other E2s, which mediate lysine-48 (K48) polyubiquitination and degradation of proteins, UBE2N primarily synthesizes K63-linked chains, stabilizing or altering protein function. Proteomic analyses and a whole-genome CRISPR-activation screen in pharmacologically and genetically UBE2N-inhibited AML cells unveiled a network of UBE2N-regulated proteins, many of which are implicated in cancer. UBE2N inhibition reduced their protein levels, leading to increased K48-linked ubiquitination and degradation through the immunoproteasome and revealing UBE2N activity is enriched in immunoproteasome-positive AML. Furthermore, an interactome screen identified tripartite motif-containing protein 21 (TRIM21) as the E3 ligase partnering with activated UBE2N in AML to modulate UBE2N-dependent proteostasis. In conclusion, UBE2N maintains proteostasis in AML by stabilizing target proteins through K63-linked ubiquitination and prevention of K48 ubiquitin-mediated degradation by the immunoproteasome. Thus, inhibition of UBE2N catalytic function suppresses leukemic cells through selective degradation of critical proteins in immunoproteasome-positive AML.
Collapse
Affiliation(s)
- Chiharu Ishikawa
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Laura Barreyro
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Avery M. Sampson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathleen M. Hueneman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sophia Y. Philbrook
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Issac Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lyndsey C. Bolanos
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrew G. Volk
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Kenneth D. Greis
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Daniel T. Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
- University of Cincinnati Cancer Center, Cincinnati, USA
| |
Collapse
|
7
|
Aakko S, Ylitalo A, Kuusanmäki H, Rannikko JH, Björkman M, Mandelin J, Heckman CA, Kontro M, Hollmén M. CLEVER-1 targeting antibody, bexmarilimab, supports HLA-DR expression and alters ex vivo responsiveness to azacitidine and venetoclax in myeloid malignancies. Sci Rep 2025; 15:16775. [PMID: 40369178 PMCID: PMC12078569 DOI: 10.1038/s41598-025-01675-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 05/07/2025] [Indexed: 05/16/2025] Open
Abstract
Treatment of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) requires new therapy options, especially for patients uneligible for intense chemotherapy or with relapsed or refractory disease. CLEVER-1 is a myeloid checkpoint protein, which can be targeted with a therapeutic function blocking antibody, bexmarilimab. Bexmarilimab has shown clinical efficacy in different solid tumors. Here, we show preclinical data demonstrating expression of CLEVER-1 on immature malignant myeloid cells and their derivates in MDS and AML bone marrow samples and AML cell lines. Highest CLEVER-1 levels were observed in AML with monocytic differentiation. Ex vivo treatment of AML/MDS bone marrow samples with bexmarilimab led to an increase in antigen-presenting human leukocyte antigen DR isotype (HLA-DR) molecule expression. Combination of bexmarilimab with current standard-of-care (SoC) drugs, azacitidine and venetoclax, showed potential for HLA-DR induction and enhanced killing of leukemic cells, respectively. Our non-clinical findings support the feasibility of CLEVER-1 inhibition in AML/MDS to induce antigen presentating molecule expression and potentially, an anti-leukemic effect together with SoC. Therapeutic targeting of CLEVER-1 with bexmarilimab is currently undergoing clinical investigation in the BEXMAB trial (NCT05428969).
Collapse
MESH Headings
- Humans
- Azacitidine/pharmacology
- Azacitidine/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/metabolism
- Myelodysplastic Syndromes/pathology
- Antibodies, Monoclonal, Humanized/pharmacology
- HLA-DR Antigens/metabolism
- Cell Line, Tumor
Collapse
Affiliation(s)
- Sofia Aakko
- Faron Pharmaceuticals Ltd, Turku, Finland
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arno Ylitalo
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre (BRIC) and Novo Nordisk Foundation Center For Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Jenna H Rannikko
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | | | | | - Caroline A Heckman
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mika Kontro
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Maija Hollmén
- Faron Pharmaceuticals Ltd, Turku, Finland.
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland.
| |
Collapse
|
8
|
Eckardt J, Hahn W, Ries RE, Chrost SD, Winter S, Stasik S, Röllig C, Platzbecker U, Müller‐Tidow C, Serve H, Baldus CD, Schliemann C, Schäfer‐Eckart K, Hanoun M, Kaufmann M, Burchert A, Schetelig J, Bornhäuser M, Wolfien M, Meshinchi S, Thiede C, Middeke JM. Age-stratified machine learning identifies divergent prognostic significance of molecular alterations in AML. Hemasphere 2025; 9:e70132. [PMID: 40337301 PMCID: PMC12056602 DOI: 10.1002/hem3.70132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/20/2025] [Accepted: 03/06/2025] [Indexed: 05/09/2025] Open
Abstract
Risk stratification in acute myeloid leukemia (AML) is driven by genetics, yet patient age substantially influences therapeutic decisions. To evaluate how age alters the prognostic impact of genetic mutations, we pooled data from 3062 pediatric and adult AML patients from multiple cohorts. Signaling pathway mutations dominated in younger patients, while mutations in epigenetic regulators, spliceosome genes, and TP53 alterations became more frequent with increasing age. Machine learning models were trained to identify prognostic variables and predict complete remission and 2-year overall survival, achieving area-under-the-curve scores of 0.801 and 0.791, respectively. Using Shapley (SHAP) values, we quantified the contribution of each variable to model decisions and traced their impact across six age groups: infants, children, adolescents/young adults, adults, seniors, and elderly. The highest contributions to model decisions among genetic variables were found for alterations of NPM1, CEBPA, inv(16), and t(8;21) conferring favorable risk and alterations of TP53, RUNX1, ASXL1, del(5q), -7, and -17 conferring adverse risk, while FLT3-ITD had an ambiguous role conferring favorable treatment responses yet poor overall survival. Age significantly modified the prognostic value of genetic alterations, with no single alteration consistently predicting outcomes across all age groups. Specific alterations associated with aging such as TP53, ASXL1, or del(5q) posed a disproportionately higher risk in younger patients. These results challenge uniform risk stratification models and highlight the need for context-sensitive AML treatment strategies.
Collapse
Affiliation(s)
- Jan‐Niklas Eckardt
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
- Else Kröner Fresenius Center for Digital HealthTUD Dresden University of TechnologyDresdenGermany
| | - Waldemar Hahn
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI) Dresden/LeipzigDresdenGermany
- Institute for Medical Informatics and BiometryTUD Dresden University of TechnologyDresdenGermany
| | - Rhonda E. Ries
- Translational Science and Therapeutics DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
- Division of Hematology and OncologySeattle Children's HospitalSeattleWashingtonUSA
| | - Szymon D. Chrost
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
| | - Susann Winter
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
| | - Sebastian Stasik
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
| | - Christoph Röllig
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious DiseaseUniversity of Leipzig Medical CenterLeipzigGermany
| | | | - Hubert Serve
- Department of Medicine 2, Hematology and OncologyGoethe University FrankfurtFrankfurtGermany
| | - Claudia D. Baldus
- Department of Hematology and OncologyUniversity Hospital Schleswig HolsteinKielGermany
| | | | - Kerstin Schäfer‐Eckart
- Department of Internal Medicine VParacelsus Medizinische Privatuniversität and University Hospital NürnbergNürnbergGermany
| | - Maher Hanoun
- Department of HematologyUniversity Hospital EssenEssenGermany
| | - Martin Kaufmann
- Department of Hematology, Oncology and Palliative CareRobert Bosch HospitalStuttgartGermany
| | - Andreas Burchert
- Department of Hematology, Oncology and ImmunologyPhilipps‐University MarburgMarburgGermany
| | - Johannes Schetelig
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
| | - Martin Bornhäuser
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
- German Cancer Consortium (DKTK), Partner Site DresdenGerman Cancer Research Center (DKFZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT), NCT/UCC Dresden, a partnership between DKFZFaculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz‐Zentrum Dresden‐Rossendorf (HZDR)Germany
| | - Markus Wolfien
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI) Dresden/LeipzigDresdenGermany
- Institute for Medical Informatics and BiometryTUD Dresden University of TechnologyDresdenGermany
| | - Soheil Meshinchi
- Translational Science and Therapeutics DivisionFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
- Division of Hematology and OncologySeattle Children's HospitalSeattleWashingtonUSA
| | - Christian Thiede
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
| | - Jan Moritz Middeke
- Department of Internal Medicine IUniversity Hospital Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
- Else Kröner Fresenius Center for Digital HealthTUD Dresden University of TechnologyDresdenGermany
| |
Collapse
|
9
|
Haring E, Buescher JM, Apostolova P. Metabolism in hematology: Technological advances open new perspectives on disease biology and treatment. Hemasphere 2025; 9:e70134. [PMID: 40390870 PMCID: PMC12086526 DOI: 10.1002/hem3.70134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 05/21/2025] Open
Abstract
The term metabolism refers to the multi-faceted biochemical reactions within a cell or an organism that occur to maintain energy homeostasis, cell growth, and oxidative balance. Cells possess a high metabolic plasticity, allowing them to adapt to the dynamic requirements of their functional state and environment. Deregulated cellular metabolism is a hallmark of many diseases, including benign and malignant hematological conditions. In the last decade, multiple technological innovations in the metabolism field have made in-depth metabolic analysis broadly applicable. Such studies are shedding new light on normal and malignant hematopoiesis and open avenues to a better understanding of the biology of hematological diseases. In this review, we will first give a brief overview of central metabolic processes. Furthermore, we discuss the most commonly used methods to study metabolism. We begin by elaborating on the use of next-generation sequencing to detect metabolism-related genomic mutations and study transcriptional signatures. Furthermore, we discuss methods for measuring protein expression, such as mass spectrometry (MS), flow cytometry, and cytometry time-of-flight. Next, we describe the use of nuclear magnetic resonance spectroscopy, MS, and flow cytometry for metabolite quantification. Finally, we highlight functional assays to probe metabolic pathways in real-time. We illustrate how these technologies and their combination have advanced our understanding of the role of metabolism. Our goal is to provide hematologists with a comprehensive guide to modern techniques in metabolism research, their benefits and disadvantages, and how they guide our understanding of disease and potentially future personalized therapy decisions.
Collapse
Affiliation(s)
- Eileen Haring
- Department of BiomedicineUniversity Hospital Basel, University of BaselBaselSwitzerland
| | - Joerg M. Buescher
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Petya Apostolova
- Department of BiomedicineUniversity Hospital Basel, University of BaselBaselSwitzerland
- Division of HematologyUniversity Hospital BaselBaselSwitzerland
| |
Collapse
|
10
|
González-Guerrero L, Castellet H, Martínez C, González N, Guijarro F, Lloveras N, Pratcorona M, Gich I, Berenguer-Molins P, Perera-Bel J, Zamora L, Mascaró M, Sampol A, Garcia-Guiñón A, Vives S, Tormo M, Arnan M, Villamor N, Nomdedéu JF. CD200 in acute myeloid leukemia: marked upregulation in CEBPA biallelic mutated cases. Diagn Pathol 2025; 20:56. [PMID: 40307896 PMCID: PMC12042300 DOI: 10.1186/s13000-025-01655-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/20/2025] [Indexed: 05/02/2025] Open
Abstract
CD200 is a glycoprotein that binds with its receptor CD200R, providing immunosuppressive signals to T and NK cells. CD200 is expressed by normal stem cells and progenitors committed to B-lymphopoiesis and myeloid development. CD200 biological relevance in acute leukemias is only partially understood.The study included a consecutive series of four hundred thirty-one patients with acute myeloid leukemia (AML). Immunophenotype was established by multiparametric flow cytometry, and the genetic diagnosis was performed by PCR-based methods and a targeted resequencing method covering 42 genes.66% of AML patients expressed CD200 being significantly associated with CD34 reactivity. The frequency of CD200 positivity was higher in cases with core-binding factor genetic lesions such as RUNX1-RUNX1T1 (81.3%) fusions and CBFB-MHY11 (63.2%) rearrangements and also with biallelic CEBPA mutations (100%). The molecular AML group with the lowest CD200 reactivity (19.1%) corresponded to AML with NPM1 mutations. RNA seq showed no uniform pattern of infiltrating cells in CEBPA mutated AML. Deconvolution analysis may be used to assess the immunoregulatory mechanisms of AML.CD200 expression could help identify the more immature compartment and, combined with other markers, single out CEPA-mutated AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/immunology
- Nucleophosmin
- Male
- Female
- Middle Aged
- Adult
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Mutation
- CCAAT-Enhancer-Binding Proteins/genetics
- Aged
- Up-Regulation
- Young Adult
- Adolescent
- Immunophenotyping
- Biomarkers, Tumor/genetics
- Aged, 80 and over
Collapse
Affiliation(s)
- Laura González-Guerrero
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Helena Castellet
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Clara Martínez
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Nuria González
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Francesca Guijarro
- Unitat d'Hematopatologia, Servei d'Anatomia Patológica, Hospital Clínic. IDIBAPS, Barcelona, Spain
| | - Natalia Lloveras
- Laboratori Hematologia. Hospital Dr. Josep Trueta. ICO Girona, Girona, Spain
| | - Marta Pratcorona
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Ignasi Gich
- Epidemiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Pau Berenguer-Molins
- Bioniformatics Unit (BU), MARData, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Júlia Perera-Bel
- Bioniformatics Unit (BU), MARData, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Lurdes Zamora
- Hematology Department, Hospital Germans Trias i Pujol. ICO Badalona. Institut Josep Carreras, Badalona, Spain
| | - Martí Mascaró
- Hematology Department, Hospital de Son Llàtzer, Palma de Mallorca, Spain
| | - Antonia Sampol
- Hematology Department, Hospital de Son Espases, Palma de Mallorca, Spain
| | | | - Susana Vives
- Hematology Department, Hospital Germans Trias i Pujol. ICO Badalona. Institut Josep Carreras, Badalona, Spain
| | - Mar Tormo
- Laboratorio de Hematología. Hospital Clínico de Valencia, Universidad de Valencia, Valencia, Spain
| | - Montserrat Arnan
- Clinical Hematology Department, ICO-Hospital Duran i Reynals. Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Neus Villamor
- Unitat d'Hematopatologia, Servei d'Anatomia Patológica, Hospital Clínic. IDIBAPS, Barcelona, Spain
| | - Josep F Nomdedéu
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain.
| |
Collapse
|
11
|
Wangulu C, Zhao D, Zhou Q, Wei C, Kumar R, Schimmer A, Chang H. Proposed Refinement of 2022 European LeukemiaNet Adverse-Risk Group of AML Patients Using a Real-World Cohort. Cancers (Basel) 2025; 17:1405. [PMID: 40361333 PMCID: PMC12070929 DOI: 10.3390/cancers17091405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/01/2025] [Accepted: 04/05/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: The 2022 European LeukemiaNet (ELN 2022) is a widely used genotypic risk classification tool for the treatment and prognostication of acute myeloid leukemia (AML) patients. Our study evaluates its effectiveness in categorizing adverse-risk AML patients on standard therapy based on their overall survival (OS). Methods: We conducted a retrospective study involving 256 AML patients. Results: Those in the ELN 2022 adverse-risk group had the shortest OS (p < 0.0001) and were predominantly characterized by myelodysplasia-related (MR) mutations, complex karyotype (CK), monosomal karyotype (MK), and TP53 mutation (TP53 Mut). Subclassification and analysis of this adverse-risk group based on the TP53 Mut status revealed a significantly shorter OS compared to the adverse TP53 wild-type (TP53 WT) counterparts (p = 0.0036). We propose refining the ELN 2022 adverse-risk group into two categories, adverse TP53 Mut and adverse TP53 WT groups, to represent adverse- and ultra-adverse-risk groups, respectively. We used an external validation dataset to confirm our findings. Conclusions: This refinement allows for a more accurate classification of these adverse-risk patients based on their clinical outcomes.
Collapse
Affiliation(s)
- Collins Wangulu
- Princess Margaret Cancer Biobank (PMCB), University Health Network, Toronto, ON M5G 2C4, Canada;
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1V4, Canada
| | - Davidson Zhao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1V4, Canada
- Department of Laboratory Hematology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Qianghua Zhou
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1V4, Canada
- Department of Laboratory Hematology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Cuihong Wei
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1V4, Canada
- Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Rajat Kumar
- Department of Hematology and Oncology, Princess Margaret Cancer Center, University Heath Network, Toronto, ON M5G 2C4, Canada
| | - Aaron Schimmer
- Department of Hematology and Oncology, Princess Margaret Cancer Center, University Heath Network, Toronto, ON M5G 2C4, Canada
| | - Hong Chang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1V4, Canada
- Department of Laboratory Hematology, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
12
|
Chow RD, Velu P, Deihimi S, Belman J, Youn A, Shah N, Luger SM, Carroll MP, Morrissette J, Bowman RL. Persistent postremission clonal hematopoiesis shapes the relapse trajectories of acute myeloid leukemia. Blood Adv 2025; 9:1888-1899. [PMID: 39938015 PMCID: PMC12008691 DOI: 10.1182/bloodadvances.2024015149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
ABSTRACT Mutations found in acute myeloid leukemia (AML) such as DNMT3A, TET2, and ASXL1 can be found in the peripheral blood of healthy adults, a phenomenon termed clonal hematopoiesis (CH). These mutations are thought to represent the earliest genetic events in the evolution of AML. Genomic studies on samples acquired at diagnosis, remission, and at relapse have demonstrated significant stability of CH mutations after induction chemotherapy. Meanwhile, later mutations in genes such as NPM1 and FLT3 have been shown to contract at remission, and in the case of FLT3 often are absent at relapse. We sought to understand how early CH mutations influence subsequent evolutionary trajectories throughout remission and relapse in response to induction chemotherapy. We assembled a retrospective cohort of patients diagnosed with de novo AML at our institution that underwent genomic sequencing at diagnosis, remission, and/or relapse (total N = 182 patients). FLT3 and NPM1 mutations were generally eliminated at complete remission but subsequently reemerged upon relapse, whereas DNMT3A, TET2, and ASXL1 mutations often persisted through remission. CH-related mutations exhibited distinct constellations of co-occurring genetic alterations, with NPM1 and FLT3 mutations enriched in DNMT3Amut AML, whereas CBL and SRSF2 mutations were enriched in TET2mut and ASXL1mut AML, respectively. In the case of NPM1 and FLT3 mutations, these differences vanished at the time of complete remission yet readily reemerged upon relapse, indicating the reproducible nature of these genetic interactions. Thus, CH-associated mutations that likely precede malignant transformation subsequently shape the evolutionary trajectories of AML through diagnosis, therapy, and relapse.
Collapse
Affiliation(s)
- Ryan D. Chow
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Priya Velu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Weill Cornell School of Medicine, Cornell University, New York, NY
| | - Safoora Deihimi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jonathan Belman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Angela Youn
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nisargbhai Shah
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Selina M. Luger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jennifer Morrissette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Robert L. Bowman
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
13
|
Kim WJ, Crosse EI, De Neef E, Etxeberria I, Sabio EY, Wang E, Bewersdorf JP, Lin KT, Lu SX, Belleville A, Fox N, Castro C, Zhang P, Fujino T, Lewis J, Rahman J, Zhang B, Winick JH, Lewis AM, Stanley RF, DeWolf S, Urben BM, Takizawa M, Krause T, Molina H, Chaligne R, Koppikar P, Molldrem J, Gigoux M, Merghoub T, Daniyan A, Chandran SS, Greenbaum BD, Klebanoff CA, Bradley RK, Abdel-Wahab O. Mis-splicing-derived neoantigens and cognate TCRs in splicing factor mutant leukemias. Cell 2025:S0092-8674(25)00399-X. [PMID: 40273911 DOI: 10.1016/j.cell.2025.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 02/06/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025]
Abstract
Mutations in RNA splicing factors are prevalent across cancers and generate recurrently mis-spliced mRNA isoforms. Here, we identified a series of bona fide neoantigens translated from highly stereotyped splicing alterations promoted by neomorphic, leukemia-associated somatic splicing machinery mutations. We utilized feature-barcoded peptide-major histocompatibility complex (MHC) dextramers to isolate neoantigen-reactive T cell receptors (TCRs) from healthy donors, patients with active myeloid malignancy, and following curative allogeneic stem cell transplant. Neoantigen-reactive CD8+ T cells were present in the blood of patients with active cancer and had a distinct phenotype from virus-reactive T cells with evidence of impaired cytotoxic function. T cells engineered with TCRs recognizing SRSF2 mutant-induced neoantigens arising from mis-splicing events in CLK3 and RHOT2 resulted in specific recognition and cytotoxicity of SRSF2-mutant leukemia. These data identify recurrent RNA mis-splicing events as sources of actionable public neoantigens in myeloid leukemias and provide proof of concept for genetically redirecting T cells to recognize these targets.
Collapse
Affiliation(s)
- Won Jun Kim
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Edie I Crosse
- Public Health Sciences and Basic Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Emma De Neef
- Public Health Sciences and Basic Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Erich Y Sabio
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Eric Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jan Philipp Bewersdorf
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | | | - Sydney X Lu
- Department of Medicine, Division of Hematology, Stanford University, Palo Alto, CA, USA
| | - Andrea Belleville
- Public Health Sciences and Basic Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nina Fox
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Cynthia Castro
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Pu Zhang
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Takeshi Fujino
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Jennifer Lewis
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Jahan Rahman
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Beatrice Zhang
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Jacob H Winick
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Alexander M Lewis
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Robert F Stanley
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | - Susan DeWolf
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | | | - Meril Takizawa
- Single-cell Analytics Innovation Laboratory, MSK, New York, NY, USA
| | - Tobias Krause
- Single-cell Analytics Innovation Laboratory, MSK, New York, NY, USA
| | - Henrik Molina
- Proteomics Resource Center, Rockefeller University, New York, NY, USA
| | - Ronan Chaligne
- Single-cell Analytics Innovation Laboratory, MSK, New York, NY, USA
| | - Priya Koppikar
- Department of Hematopoietic Biology and Malignancy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey Molldrem
- Department of Hematopoietic Biology and Malignancy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mathieu Gigoux
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Sandra and Edward Meyer Cancer Center, Weill Cornell Medical Center, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Sandra and Edward Meyer Cancer Center, Weill Cornell Medical Center, New York, NY, USA
| | - Anthony Daniyan
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA
| | | | - Benjamin D Greenbaum
- Computational Oncology, Department of Epidemiology and Biostatistics, MSK, New York, NY, USA
| | - Christopher A Klebanoff
- Human Oncology and Pathogenesis Program, MSK, New York, NY, USA; Parker Institute for Cancer Immunotherapy, New York, NY, USA.
| | - Robert K Bradley
- Public Health Sciences and Basic Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, USA.
| |
Collapse
|
14
|
Richards CJ, Wierenga AT, Brouwers-Vos AZ, Kyrloglou E, Dillingh LS, Mulder PP, Palasantzas G, Schuringa JJ, Roos WH. Elastic properties of leukemic cells linked to maturation stage and integrin activation. iScience 2025; 28:112150. [PMID: 40201128 PMCID: PMC11978321 DOI: 10.1016/j.isci.2025.112150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/06/2025] [Accepted: 02/27/2025] [Indexed: 04/10/2025] Open
Abstract
Acute myeloid leukemia (AML) remains challenging to cure. In addition to mutations that alter cell functioning, biophysical properties are modulated by external cues. In particular, membrane proteins that interact with the bone marrow niche can induce cellular changes. Here, we develop an atomic force microscopy (AFM) approach to measure non-adherent AML cell mechanical properties. The Young's modulus of the AML cell line, THP-1, increased in response to retronectin, whereas knock-out of the adhesion protein ITGB1 resulted in no response to retronectin. Confocal microscopy revealed different actin cytoskeleton morphologies for wild-type and ITGB1 knock-out cells exposed to retronectin. These results indicate that ITGB1 mediates stimuli-induced cellular mechanoresponses through cytoskeletal changes. We next used AFM to investigate the elastic properties of primary AML cells and found that more committed cells had lower Young's moduli than immature AMLs. Overall, this provides a platform for investigating the molecular mechanisms involved in leukemic cell mechanoresponse.
Collapse
Affiliation(s)
- Ceri J. Richards
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, 9747 AG Groningen, the Netherlands
| | - Albertus T.J. Wierenga
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Annet Z. Brouwers-Vos
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Emmanouil Kyrloglou
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Laura S. Dillingh
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, 9747 AG Groningen, the Netherlands
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
- Nanostructure Materials and Interfaces, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Patty P.M.F.A. Mulder
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Georgios Palasantzas
- Nanostructure Materials and Interfaces, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Wouter H. Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, 9747 AG Groningen, the Netherlands
| |
Collapse
|
15
|
Hosseini A, Dhall A, Ikonen N, Sikora N, Nguyen S, Shen Y, Amaral MLJ, Jiao A, Wallner F, Sergeev P, Lim Y, Yang Y, Vick B, Kawabata KC, Melnick A, Vyas P, Ren B, Jeremias I, Psaila B, Heckman CA, Blanco MA, Shi Y. Perturbing LSD1 and WNT rewires transcription to synergistically induce AML differentiation. Nature 2025:10.1038/s41586-025-08915-1. [PMID: 40240608 DOI: 10.1038/s41586-025-08915-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/18/2025] [Indexed: 04/18/2025]
Abstract
Impaired differentiation is a hallmark of myeloid malignancies1,2. Therapies that enable cells to circumvent the differentiation block, such as all-trans retinoic acid (ATRA) and arsenic trioxide (ATO), are by and large curative in acute promyelocytic leukaemia3, but whether 'differentiation therapy' is a generalizable therapeutic approach for acute myeloid leukaemia (AML) and beyond remains incompletely understood. Here we demonstrate that simultaneous inhibition of the histone demethylase LSD1 (LSD1i) and the WNT pathway antagonist GSK3 kinase4 (GSK3i) robustly promotes therapeutic differentiation of established AML cell lines and primary human AML cells, as well as reducing tumour burden and significantly extending survival in a patient-derived xenograft mouse model. Mechanistically, this combination promotes differentiation by activating genes in the type I interferon pathway via inducing expression of transcription factors such as IRF7 (LSD1i) and the co-activator β-catenin (GSK3i), and their selective co-occupancy at targets such as STAT1, which is necessary for combination-induced differentiation. Combination treatment also suppresses the canonical, pro-oncogenic WNT pathway and cell cycle genes. Analysis of datasets from patients with AML suggests a correlation between the combination-induced transcription signature and better prognosis, highlighting clinical potential of this strategy. Collectively, this combination strategy rewires transcriptional programs to suppress stemness and to promote differentiation, which may have important therapeutic implications for AML and WNT-driven cancers beyond AML.
Collapse
Affiliation(s)
- Amir Hosseini
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Abhinav Dhall
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nemo Ikonen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Natalia Sikora
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sylvain Nguyen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yuqi Shen
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM), University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | | | - Alan Jiao
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Felice Wallner
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philipp Sergeev
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Yuhua Lim
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yuanqin Yang
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Binje Vick
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital LMU Munich, Munich, Germany
| | - Kimihito Cojin Kawabata
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Ari Melnick
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Paresh Vyas
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM), University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Bing Ren
- Cell and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital LMU Munich, Munich, Germany
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Bethan Psaila
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM), University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.
| | - M Andrés Blanco
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yang Shi
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Lee CF, Pienta KJ, Amend SR. The involvement of cyclin-dependent kinase 7 (CDK7) and 9 (CDK9) in coordinating transcription and cell cycle checkpoint regulation. Cell Cycle 2025:1-13. [PMID: 40223539 DOI: 10.1080/15384101.2025.2485844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 04/15/2025] Open
Abstract
Cells regulate the expression of cell cycle-related genes, including cyclins essential for mitosis, through the transcriptional activity of the positive transcription elongation factor b (P-TEFb), a complex comprising CDK9, cyclin T, and transcription factors. P-TEFb cooperates with CDK7 to activate RNA polymerase. In response to DNA stress, the cell cycle shifts from mitosis to repair, triggering cell cycle arrest and the activation of DNA repair genes. This tight coordination between transcription, cell cycle progression, and DNA stress response is crucial for maintaining cellular integrity. Cyclin-dependent kinases CDK7 and CDK9 are central to both transcription and cell cycle regulation. CDK7 functions as the CDK-activating kinase (CAK), essential for activating other CDKs, while CDK9 acts as a critical integrator of signals from both the cell cycle and transcriptional machinery. This review elucidates the mechanisms by which CDK7 and CDK9 regulate the mitotic process and cell cycle checkpoints, emphasizing their roles in balancing cell growth, homeostasis, and DNA repair through transcriptional control.
Collapse
Affiliation(s)
- Cheng-Fan Lee
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - Kenneth J Pienta
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - Sarah R Amend
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| |
Collapse
|
17
|
Pan X, Huang C, Bai X, Li F. Causal relationship between breast cancer and acute myeloid leukemia based on two-sample bidirectional Mendelian randomization and transcriptome overlap analysis. Discov Oncol 2025; 16:492. [PMID: 40198525 PMCID: PMC11979033 DOI: 10.1007/s12672-025-02288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Breast cancer is the most prevalent malignancy and the leading cause of cancer-related deaths among women worldwide. Several case reports have shown that some breast cancer patients subsequently develop acute myeloid leukemia (AML) within a short period. However, the causal relationship and pathogenic mechanisms between breast cancer and AML remain incompletely understood. METHODS Mendelian randomization (MR) analyses were conducted to explore the bidirectional causal relationships between breast cancer and AML. Additionally, we applied the Bayesian Weighted Mendelian Randomization (BWMR) approach to validate the results of the MR analysis. Subsequently, we utilized RNA-seq data from various sources to explore the potential molecular signaling pathways between breast cancer and AML. RESULTS Both IVW method and BWMR approach demonstrated that data from three distinct sources consistently indicated breast cancer as a risk factor for AML, with all sources showing statistically significant results (all P < 0.05, Odds Ratios [ORs] > 1). Bioinformatic analyses suggested that extracellular vesicle functions and p53 signaling pathway may mediate molecular links between breast cancer and AML. Using machine learning, we identified 8 genes with high diagnostic efficacy for predicting the occurrence of AML in breast cancer patients. CONCLUSIONS MR analyses indicated a causal relationship between breast cancer and AML. Additionally, transcriptome analysis offered a theoretical basis for understanding the potential mechanisms and therapeutic targets of AML in breast cancer patients.
Collapse
Affiliation(s)
- Xin'an Pan
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Zheng Street, East Lake District, Nanchang City, 330006, Jiangxi Province, China
| | - Cuihan Huang
- The First Clinical Medical College of Nanchang University, Xuefu Road, Nanchang, 330006, Jiangxi, China
| | - Xinyi Bai
- School of Public, Health of Nanchang University, Xuefu Road, Nanchang, 330006, Jiangxi, China
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Zheng Street, East Lake District, Nanchang City, 330006, Jiangxi Province, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, China.
| |
Collapse
|
18
|
Park SJ, Cerella C, Kang JM, Byun J, Kum D, Orlikova-Boyer B, Lorant A, Schnekenburger M, Al-Mourabit A, Christov C, Lee J, Han BW, Diederich M. Tetrahydrobenzimidazole TMQ0153 targets OPA1 and restores drug sensitivity in AML via ROS-induced mitochondrial metabolic reprogramming. J Exp Clin Cancer Res 2025; 44:114. [PMID: 40197337 PMCID: PMC11974110 DOI: 10.1186/s13046-025-03372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a highly aggressive cancer with a 5-year survival rate of less than 35%. It is characterized by significant drug resistance and abnormal energy metabolism. Mitochondrial dynamics and metabolism are crucial for AML cell survival. Mitochondrial fusion protein optic atrophy (OPA)1 is upregulated in AML patients with adverse mutations and correlates with poor prognosis. METHOD This study investigated targeting OPA1 with TMQ0153, a tetrahydrobenzimidazole derivative, to disrupt mitochondrial metabolism and dynamics as a novel therapeutic approach to overcome treatment resistance. Effects of TMQ0153 treatment on OPA1 and mitofusin (MFN)2 protein levels, mitochondrial morphology, and function in AML cells. In this study, we examined reactive oxygen species (ROS) production, oxidative phosphorylation (OXPHOS) inhibition, mitochondrial membrane potential (MMP) depolarization, and apoptosis. Additionally, metabolic profiling was conducted to analyze changes in metabolic pathways. RESULTS TMQ0153 treatment significantly reduced OPA1 and mitofusin (MFN)2 protein levels and disrupted the mitochondrial morphology and function in AML cells. This increases ROS production and inhibits OXPHOS, MMP depolarization, and caspase-dependent apoptosis. Metabolic reprogramming was observed, shifting from mitochondrial respiration to glycolysis and impaired respiratory chain activity. Profiling revealed reduced overall metabolism along with changes in the glutathione (GSH)/oxidized glutathione (GSSG) and NAD⁺/NADH redox ratios. TMQ0153 treatment reduces tumor volume and weight in MV4-11 xenografts in vivo. Combination therapies with TMQ0153 and other AML drugs significantly reduced the leukemic burden and prolonged survival in NOD scid gamma (NSG) mice xenografted with U937-luc and MOLM-14-luc cells. CONCLUSION TMQ0153 targets mitochondrial dynamics by inhibiting OPA1, inducing metabolic reprogramming, and triggering apoptosis in AML cells. It enhances the efficacy of existing AML therapies and provides a promising combination treatment approach that exploits mitochondrial vulnerability and metabolic reprogramming to improve treatment outcomes in AML.
Collapse
MESH Headings
- Humans
- GTP Phosphohydrolases/metabolism
- GTP Phosphohydrolases/antagonists & inhibitors
- GTP Phosphohydrolases/genetics
- Reactive Oxygen Species/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Animals
- Mice
- Benzimidazoles/pharmacology
- Mitochondria/metabolism
- Mitochondria/drug effects
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Apoptosis/drug effects
- Oxidative Phosphorylation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Membrane Potential, Mitochondrial/drug effects
- Metabolic Reprogramming
Collapse
Affiliation(s)
- Su Jung Park
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Jin Mo Kang
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinyoung Byun
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - David Kum
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Barbora Orlikova-Boyer
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Anne Lorant
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Luxembourg Centre for Systems Biomedicine, Bioinformatics Core, Roudeneck, 1, Boulevard du Jazz, Esch-sur-Alzette, L-4370, Luxembourg
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Ali Al-Mourabit
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-Sur-Yvette, 91190, France
| | - Christo Christov
- Service d'Histologie, Faculté de Médicine, Université de Lorraine, and INSERM U1256 NGERE, 54000, Nancy, France
| | - Juyong Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Byung Woo Han
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Marc Diederich
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
19
|
Carlos JAEG, Tavares MT, Lima K, de Almeida LC, de Barros Waitman K, Costa-Lotufo LV, Parise-Filho R, Machado-Neto JA. Enhancing venetoclax efficacy in leukemia through association with HDAC inhibitors. Cell Death Discov 2025; 11:147. [PMID: 40188101 PMCID: PMC11972356 DOI: 10.1038/s41420-025-02446-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Epigenetic modifications significantly influence gene expression and play crucial roles in various biological processes, including carcinogenesis. This study investigates the effects of novel purine-benzohydroxamate compounds, particularly 4 f, as hybrid kinase/histone deacetylase (HDAC) inhibitors in hematological malignancies, focusing on acute myeloid leukemia (AML). Our results demonstrate that these compounds selectively reduce cell viability in blood cancer cells, with inhibitory concentration values indicating higher potency against neoplastic cells compared to normal leukocytes. Mechanistically, 4 f induces apoptosis and cell cycle arrest, promoting differentiation in leukemia cells, while effectively inhibiting HDAC activity. Furthermore, 4 f enhances the therapeutic efficacy of venetoclax, a BCL2 inhibitor, in AML models sensitive and resistant to this drug. The combination treatment significantly increases apoptosis and reduces cell viability, suggesting a synergistic effect that may overcome drug resistance. This study provides valuable insights into the potential of HDAC inhibitors, particularly 4 f, as a promising therapeutic strategy for treating resistant hematological malignancies. Our findings underscore the importance of further exploring hybrid kinase/HDAC inhibitors in combination therapies to improve outcomes in patients with acute leukemias and other hematological malignancies.
Collapse
Affiliation(s)
| | - Mauricio Temotheo Tavares
- Department of Pharmacy, Faculty of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Larissa Costa de Almeida
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Roberto Parise-Filho
- Department of Pharmacy, Faculty of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
20
|
Marhelava K, Fidyt K, Pepek M, Krawczyk M, Forcados C, Malinowska A, Swiderska B, Fernandez-Fuentes N, Czerwik N, Baranowska I, Krzywdzinska A, Sedek L, Slota L, Perkowski B, Villatoro A, Leray T, Lech-Maranda E, Menendez P, Inderberg EM, Wälchli S, Winiarska M, Firczuk M. LILRB1-directed CAR-T cells for the treatment of hematological malignancies. Leukemia 2025:10.1038/s41375-025-02580-z. [PMID: 40186066 DOI: 10.1038/s41375-025-02580-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/27/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025]
Abstract
CD19 CAR-T cells have established a new standard for relapsed/refractory B-cell malignancies. However, the treatment fails in 50% of patients, often due to CD19 antigen loss. Alternative immunotherapies targeting other antigens are being tested but show limited efficacy, especially in cases of lineage switching or loss of B-cell phenotype, highlighting the need for novel targets. Herein, we identified leukocyte-immunoglobulin-like-receptor-B1 (LILRB1, CD85j) as a novel target for CAR-T cells through cell surface proteomics on patient-derived samples of high-risk B-cell acute lymphoblastic leukemia (B-ALL). LILRB1, an immune inhibitory receptor, is normally expressed only on monocytes and B-cells. We observed stable LILRB1 expression in B-ALL and B-cell non-Hodgkin lymphoma (B-NHL), even after CD20/CD19-based immunotherapies. LILRB1 CAR-T cells showed antigen-specific antitumor activity in vitro against B-ALL/B-NHL cells, including those resistant to CD19 CAR-T-cells, and in vivo in B-ALL xenografts. Additionally, we identified LILRB1 in monocytic acute myeloid leukemia (AML) and demonstrated LILRB1 CAR-T cell cytotoxicity against AML cell lines in vitro and in vivo. These findings establish LILRB1 as a novel target for cancer immunotherapy and show evidence for the preclinical efficacy of LILRB1 CAR-T cells against haematological malignancies, including cases resistant to previous lines of immunotherapy, thus holding promise for further clinical development.
Collapse
Affiliation(s)
- Katsiaryna Marhelava
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Monika Pepek
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Marta Krawczyk
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Doctoral School of Translational Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Christopher Forcados
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Agata Malinowska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Bianka Swiderska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Natalia Czerwik
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Iwona Baranowska
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Krzywdzinska
- Laboratory of Immunophenotyping, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Lukasz Sedek
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Lukasz Slota
- Department of Pediatric Hematology and Oncology, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Bartosz Perkowski
- Department of Pediatric Hematology and Oncology, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Alicia Villatoro
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Thibault Leray
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ewa Lech-Maranda
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Instituto de Salud Carlos III, Madrid, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III, Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Institut de Recerca Hospital Sant Joan de Déu-Pediatric Cancer Center Barcelona (SJD-PCCB), Barcelona, Spain
| | - Else Marit Inderberg
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Magdalena Winiarska
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland.
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland.
| | - Malgorzata Firczuk
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland.
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
21
|
Giri AK, Lin J, Kyriakidis K, Tripathi G, Almusa H. Exome-wide association study reveals 7 functional variants associated with ex-vivo drug response in acute myeloid leukemia patients. BMC Med Genomics 2025; 18:64. [PMID: 40186177 PMCID: PMC11969768 DOI: 10.1186/s12920-025-02130-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer characterized by poor survival outcomes. Further, due to the extreme molecular heterogeneity of the disease, drug treatment response varies from patient to patient. The variability of drug response can cause unnecessary treatment in more than half of the patients with no or partial therapy responses leading to severe side effects, monetary as well as time loss. Understanding the genetic risk factors underlying the drug response in AML can help with improved prediction of treatment responses and identification of biomarkers in addition to mechanistic insights to monitor treatment response. Here, we report the results of the first Exome-Wide Association Study (EWAS) of ex-vivo drug response performed to date with 175 AML cases and 47 drugs. We used information from 55,423 germline exonic SNPs to perform the analysis. We identified exome-wide significant (p < 9.02 × 10- 7) associations for rs113985677 in CCIN with tamoxifen response, rs115400838 in TRMT5 with idelalisib response, rs11878277 in HDGFL2 with entinostat, and rs2229092 in LTA associated with vorinostat response. Further, using multivariate genome-wide association analysis, we identified the association of rs11556165 in ATRAID, and rs11236938 in TSKU with the combined response of all 47 drugs and 29 nonchemotherapy drugs at the genome-wide significance level (p < 5 × 10- 8). Additionally, a significant association of rs35704242 in NIBAN1 was associated with the combined response for nonchemotherapy medicines (p = 2.51 × 10- 8), and BI.2536, gefitinib, and belinostat were identified as the central traits. Our study represents the first EWAS to date on ex-vivo drug response in AML and reports 7 new associated loci that help to understand the anticancer drug response in AML patients.
Collapse
Affiliation(s)
- Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
- Foundation for the Finnish Cancer Institute (FCI), Tukholmankatu 8, Helsinki, 00290, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, PO Box 20, Helsinki, FI-00014, Finland.
| | - Jake Lin
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Konstantinos Kyriakidis
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Garima Tripathi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Butler JT, Yashar WM, Swords R. Breaking the Bone Marrow Barrier: Peripheral Blood as a Gateway to Measurable Residual Disease Detection in Acute Myelogenous Leukemia. Am J Hematol 2025; 100:638-651. [PMID: 39777414 PMCID: PMC11886496 DOI: 10.1002/ajh.27586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025]
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous disease with high rates of relapse after initial treatment. Identifying measurable residual disease (MRD) following initial therapy is essential to assess response, predict patient outcomes, and identify those in need of additional intervention. Currently, MRD analysis relies on invasive, serial bone marrow (BM) biopsies, which complicate sample availability and processing time and negatively impact patient experience. Additionally, finding a positive result can generate more questions than answers, causing anxiety for both the patient and the provider. Peripheral blood (PB) evaluation has shown promise in detecting MRD and is now recommended by the European Leukemia Net for AML for certain genetic abnormalities. PB-based sampling allows for more frequent testing intervals and better temporal resolution of malignant expansion while sparing patients additional invasive procedures. In this review, we will discuss the current state of PB testing for MRD evaluation with a focus on next-generation sequencing methodologies that are capable of MRD detection across AML subtypes.
Collapse
MESH Headings
- Humans
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/blood
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Bone Marrow/pathology
- High-Throughput Nucleotide Sequencing
Collapse
Affiliation(s)
- John T. Butler
- Radiation Medicine and Applied Science, Moores Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
| | - William M. Yashar
- Knight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
- Division of Oncologic Sciences, Department of MedicineOregon Health & Science UniversityPortlandOregonUSA
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| | - Ronan Swords
- Division of Oncologic Sciences, Department of MedicineOregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
23
|
Zhang R, Jin W, Wang K. Glycolysis-Driven Prognostic Model for Acute Myeloid Leukemia: Insights into the Immune Landscape and Drug Sensitivity. Biomedicines 2025; 13:834. [PMID: 40299448 PMCID: PMC12024913 DOI: 10.3390/biomedicines13040834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Acute myeloid leukemia (AML), a malignant blood disease, is caused by the excessive growth of undifferentiated myeloid cells, which disrupt normal hematopoiesis and may invade several organs. Given the high heterogeneity in prognosis, identifying stable prognostic biomarkers is crucial for improved risk stratification and personalized treatment strategies. Although glycolysis has been extensively studied in cancer, its prognostic significance in AML remains unclear. Methods: Glycolysis-related prognostic genes were identified by differential expression profiles. We modeled prognostic risk by least absolute shrinkage and selection operator (LASSO) regression and validated it by Kaplan-Meier (KM) survival analysis, receiver operating characteristic (ROC) curves, and independent datasets (BeatAML2.0, GSE37642, GSE71014). Mechanisms were further explored through immune microenvironment analysis and drug sensitivity scores. Results: Differential expression and survival correlation analysis across the genes associated with glycolysis revealed multiple glycolytic genes associated with the outcomes of AML. We constructed a seven-gene prognostic model (G6PD, TFF3, GALM, SOD1, NT5E, CTH, FUT8). Kaplan-Meier analysis demonstrated significantly reduced survival in high-risk patients (hazard ratio (HR) = 3.4, p < 0.01). The model predicted the 1-, 3-, and 5-year survival outcomes, achieving area under the curve (AUC) values greater than 0.8. Immune profiling indicated distinct cellular compositions between risk groups: high-risk patients exhibited elevated monocytes and neutrophils but reduced Th1 cell infiltration. Drug sensitivity analysis showed that high-risk patients exhibited resistance to crizotinib and lapatinib but were more sensitive to motesanib. Conclusions: We established a novel glycolysis-related gene signature for AML prognosis, enabling effective risk classification. Combined with immune microenvironment analysis and drug sensitivity analysis, we screened metabolic characteristics and identified an immune signature to provide deeper insight into AML. Our findings may assist in identifying new therapeutic targets and more effective personalized treatment regimes.
Collapse
Affiliation(s)
- Rongsheng Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China; (R.Z.); (W.J.)
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Wen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China; (R.Z.); (W.J.)
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China; (R.Z.); (W.J.)
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China
| |
Collapse
|
24
|
Rahimian E, Koochak M, Traikov S, Schroeder M, Brilloff S, Schäfer S, Kufrin V, Küchler S, Krüger A, Mirtschink P, Baretton G, Schröck E, Schewe DM, Ball CR, Bornhäuser M, Glimm H, Bill M, Wurm AA. A quiescence-like/TGF-β1-specific CRISPRi screen reveals drug uptake transporters as secondary targets of kinase inhibitors in AML. Drug Resist Updat 2025; 81:101242. [PMID: 40184725 DOI: 10.1016/j.drup.2025.101242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/26/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
Relapse in acute myeloid leukemia (AML) is driven by resistant subclones that survive chemotherapy. It is assumed that these resilient leukemic cells can modify their proliferative behavior by entering a quiescent-like state, similar to healthy hematopoietic stem cells (HSCs). These dormant cells can evade the effects of cytostatic drugs that primarily target actively dividing cells. Although quiescence has been extensively studied in healthy hematopoiesis and various solid cancers, its role in AML has remained unexplored. In this study, we applied an HSC-derived quiescence-associated gene signature to an AML patient cohort and found it to be strongly correlated with poor prognosis and active TGF-β signaling. In vitro treatment with TGF-β1 induces a quiescence-like phenotype, resulting in a G0 shift and reduced sensitivity to cytarabine. To find potential therapeutic targets that prevent AML-associated quiescence and improve response to cytarabine, we conducted a comprehensive CRISPR interference (CRISPRi) screen combined with TGF-β1 stimulation. This approach identified TGFBR1 inhibitors, like vactosertib, as effective agents for preventing the G0 shift in AML cell models. However, pretreatment with vactosertib unexpectedly induced complete resistance to cytarabine. To elucidate the underlying mechanism, we performed a multi-faceted approach combining a second CRISPRi screen, liquid chromatography-tandem mass spectrometry (LC-MS/MS), and in silico analysis. Our findings revealed that TGFBR1 inhibitors unintentionally target the nucleoside transporter SLC29A1 (ENT1), leading to reduced intracellular cytarabine levels. Importantly, we found that this drug interaction is not unique to TGFBR1 inhibitors, but extends to other clinically significant kinase inhibitors, such as the FLT3 inhibitor midostaurin. These findings may have important implications for optimizing combination therapies in AML treatment.
Collapse
Affiliation(s)
- Elahe Rahimian
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Masoud Koochak
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Sofia Traikov
- Institute for Clinical Chemistry and Laboratory Medicine, TU Dresden, Dresden, Germany
| | - Michael Schroeder
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Silke Brilloff
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Silvia Schäfer
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Vida Kufrin
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Sandra Küchler
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany
| | - Alexander Krüger
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases Dresden (NCT), NCT/UCC Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory Medicine, TU Dresden, Dresden, Germany
| | - Gustavo Baretton
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases Dresden (NCT), NCT/UCC Dresden, a partnership between German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Institute for Pathology, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Evelin Schröck
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus at TUD Dresden University of Technology and Faculty of Medicine of TUD Dresden University of Technology, Dresden, Germany; ERN GENTURIS, Hereditary Cancer Syndrome Center Dresden, Germany; National Center for Tumor Diseases (NCT), NCT/UCC Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; German Cancer Consortium (DKTK), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Denis M Schewe
- Department of Pediatric Hematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Claudia R Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; TUD Dresden University of Technology, Faculty of Biology, Dresden, Germany
| | - Martin Bornhäuser
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Internal Medicine I, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marius Bill
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; Department of Internal Medicine I, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Alexander A Wurm
- Mildred Scheel Early Career Center, National Center for Tumor Diseases (NCT/UCC) Dresden, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Department of Pediatric Hematology and Oncology, University Hospital Dresden, Dresden, Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany.
| |
Collapse
|
25
|
Luo K, Chen J, Wang W, Hui Y, Qiu S, Liu B, Mi Y, Wang J, Wei H. Dynamin2 mutations in newly diagnosed acute myeloid leukemia: clinical characteristics, and prognostic significance. Exp Hematol Oncol 2025; 14:42. [PMID: 40119466 PMCID: PMC11927327 DOI: 10.1186/s40164-025-00628-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/02/2025] [Indexed: 03/24/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous myeloid malignancy which can be classified by genetic aberrations. To evaluate the impact of the dynamin 2 mutation in AML, we systematically assessed the characteristics and prognostic of DNM2 mutated patients in AML. In 912 AML patients, 20 somatic mutations in the DNM2 gene were identified among the 18 DNM2 mutated AML patients (2%). Of the mutation events, 60% (12/20) were in the dynamin central region of DNM2. DNM2mutations were preferentially occurred in AML with CEBPA mutation (11/18, 61.1%), or RUNX1::RUNX1T1 fusion gene (6/18, 33.3%). DNM2 mutations were associated with better overall survival (P = 0.028), event-free survival (P = 0.0093) and trends towards better relapse-free survival (P = 0.08), which seems potentially attribute to its coexisting with CEBPA mutation and RUNX1::RUNX1T1 fusion gene. Our study demonstrated the clinical characteristics and the role of DNM2 mutations in AML, which might facilitate understanding the pathogenesis of AML.
Collapse
Affiliation(s)
- Kunpeng Luo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jiayuan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Wenting Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yan Hui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Bingcheng Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yingchang Mi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Hui Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Rd 288, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
26
|
Kenmogne VL, Takundwa MM, Nweke EE, Monchusi B, Dube P, Maher H, Du Toit J, Philip-Cherian V, Fru PN, Thimiri Govinda Raj DB. The first-in-Africa ex vivo drug sensitivity testing platform identifies novel drug combinations for South African leukaemia patient cohort. Sci Rep 2025; 15:9160. [PMID: 40097584 PMCID: PMC11914478 DOI: 10.1038/s41598-025-93634-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
In South Africa, leukemia remains a major health concern, posing significant challenges in treatment due to its varied subtypes. There is an unmet need for a testing pipeline that can identify drug effects on patient samples in an ex-vivo setting. Using the pilot study with South African patient samples, this paper reports the development of a drug-sensitivity testing pipeline for studying the drug effects in leukemia patient-derived cells. Forty-one (41) patients with Acute myeloid leukemia (AML) (n = 7), Chronic myelogenous leukemia (CML) (n = 30), and Chronic lymphocytic leukemia (CLL) (n = 4) were recruited for this study. Thirty (30) FDA-approved drugs were utilized for single drug sensitivity screening (DSS) on leukemia patient-derived cells with drug concentrations (1-1000 nM). The single DSS showed a distinct sensitivity pattern with different profiles among patients of the same subtype, confirming the need for precision therapy. This study observed irinotecan, used in solid tumour treatment, demonstrated efficacy in PBMCs in many patient samples compared to conventional leukemia drugs such as nilotinib. For drug combination studies, ten clinically relevant drugs were selected and tested based on the results of single drug sensitivity tests. This pilot study marks a crucial stride towards revolutionizing leukemia treatment in South Africa through an innovative ex vivo drug sensitivity testing platform. This pioneering initiative forms the basis for tailored and effective treatment options holding promise for more personalized treatment. Further exploration and validation of these findings could significantly contribute to cancer precision medicine efforts in South Africa.
Collapse
Affiliation(s)
- V L Kenmogne
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - M M Takundwa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - E E Nweke
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - B Monchusi
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - P Dube
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - H Maher
- Wits Donald Gordon Medical Centre, Johannesburg, South Africa
| | - J Du Toit
- Wits Donald Gordon Medical Centre, Johannesburg, South Africa
| | - V Philip-Cherian
- Department of Haematology, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - P N Fru
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - D B Thimiri Govinda Raj
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa.
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
| |
Collapse
|
27
|
Fiskus W, Mill CP, Piel J, Collins M, Hentemann M, Cuglievan B, Birdwell CE, Das K, Hou H, Davis JA, Jain A, Malovannaya A, Kadia TM, Daver N, Sasaki K, Takahashi K, Hammond D, Reville PK, Flores LB, Loghavi S, Su X, DiNardo CD, Bhalla KN. Superior preclinical efficacy of co-treatment with BRG1/BRM and FLT3 inhibitor against AML cells with FLT3 mutations. Blood Cancer J 2025; 15:40. [PMID: 40089460 PMCID: PMC11910597 DOI: 10.1038/s41408-025-01251-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/22/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025] Open
Abstract
Although treatment with standard frontline therapies, including a FLT3 inhibitor (FLT3i) reduces AML burden and achieves clinical remissions, most patients with AML with FLT3 mutation relapse due to therapy-resistant stem/progenitor cells. The core ATPases, BRG1 (SMARCA4) and BRM (SMARCA2) of the canonical (c) BAF (BRG1/BRM-associated factor) complex is a dependency in AML cells, including those harboring FLT3 mutations. We have previously reported that treatment with FHD-286, a BRG1/BRM ATPases inhibitor, induces differentiation and loss of viability of AML stem/progenitor cells. Findings of present studies demonstrate that treatment with FHD-286 induces lethality in AML cells, regardless of sensitivity or resistance to FLT3i. This efficacy is associated with the induction of gene-expression perturbations responsible for growth inhibition, differentiation, as well as a reduced AML-initiating potential of the AML cells. Additionally, co-treatment with FHD-286 and FLT3i exerts superior pre-clinical efficacy against AML cells and patient-derived (PD) xenograft (PDX) models of AML with FLT3 mutations.
Collapse
Affiliation(s)
- Warren Fiskus
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Christopher P Mill
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | | | | | | | - Branko Cuglievan
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Kaberi Das
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hanxi Hou
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - John A Davis
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Antrix Jain
- Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Tapan M Kadia
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Naval Daver
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Koji Sasaki
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Koichi Takahashi
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Danielle Hammond
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Patrick K Reville
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Lauren B Flores
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sanam Loghavi
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaoping Su
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Courtney D DiNardo
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kapil N Bhalla
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
28
|
Soto CA, Lesch ML, Becker JL, Sharipol A, Khan A, Schafer XL, Becker MW, Munger JC, Frisch BJ. Elevated Lactate in the AML Bone Marrow Microenvironment Polarizes Leukemia-Associated Macrophages via GPR81 Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.13.566874. [PMID: 39185193 PMCID: PMC11343108 DOI: 10.1101/2023.11.13.566874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Interactions between acute myeloid leukemia (AML) and the bone marrow microenvironment (BMME) are critical to leukemia progression and chemoresistance. In the solid tumor microenvironment, altered metabolite levels contribute to cancer progression. We performed a metabolomic analysis of AML patient bone marrow serum, revealing increased metabolites compared to age- and sex-matched controls. The most highly elevated metabolite in the AML BMME was lactate. Lactate signaling in solid tumors induces immunosuppressive tumor-associated macrophages and correlates with poor prognosis. This has not yet been studied in the leukemic BMME. Herein, we describe the role of lactate in the polarization of leukemia-associated macrophages (LAMs). Using a murine AML model of blast crisis chronic myelogenous leukemia (bcCML), we characterize the suppressive phenotype of LAMs by surface markers, transcriptomics, and cytokine profiling. Then, mice genetically lacking GPR81, the extracellular lactate receptor, were used to demonstrate GPR81 signaling as a mechanism of both the polarization of LAMs and the direct support of leukemia cells. Furthermore, elevated lactate diminished the function of hematopoietic progenitors and reduced stromal support for normal hematopoiesis. We report microenvironmental lactate as a mechanism of AML-induced immunosuppression and leukemic progression, thus identifying GPR81 signaling as an exciting and novel therapeutic target for treating this devastating disease.
Collapse
Affiliation(s)
- Celia A Soto
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Maggie L Lesch
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Jennifer L Becker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Azmeer Sharipol
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester School of Medicine, Rochester, NY, USA
| | - Amal Khan
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Xenia L Schafer
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Joshua C Munger
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY, USA
| | - Benjamin J Frisch
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester School of Medicine, Rochester, NY, USA
| |
Collapse
|
29
|
Hong J, Sui P, Li Y, Xu KY, Lee JH, Wang J, Chen S, Zhang P, Wingate N, Noor A, Yuan Y, Hromas R, Zhou H, Hamamoto K, Su R, Yin CC, Ye F, Quesada AE, Chen J, Huang S, Zhou D, You MJ, Yang FC, Wang J, Xu M. PSPC1 exerts an oncogenic role in AML by regulating a leukemic transcription program in cooperation with PU.1. Cell Stem Cell 2025; 32:463-478.e6. [PMID: 39954676 DOI: 10.1016/j.stem.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 11/01/2024] [Accepted: 01/16/2025] [Indexed: 02/17/2025]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy characterized by the blockage of myeloid cell differentiation and uncontrolled proliferation of immature myeloid cells. Here, we show that paraspeckle component 1 (PSPC1) is aberrantly overexpressed and associated with poor survival in AML patients. Using human AML cells and mouse models, we demonstrate that PSPC1 is not required for normal hematopoiesis, but it is critical and essential for AML cells to maintain their leukemic characteristics. PSPC1 loss induces robust differentiation, suppresses proliferation, and abolishes leukemogenesis in diverse AML cells. Mechanistically, PSPC1 exerts a pro-leukemia effect by regulating a unique leukemic transcription program via cooperative chromatin binding with PU.1 and activation of tumor-promoting genes, including NDC1, which is not previously implicated in AML. Our findings uncover a unique and crucial role of PSPC1 dependency in AML and highlight its potential as a promising therapeutic target for AML.
Collapse
Affiliation(s)
- Juyeong Hong
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pinpin Sui
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ying Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kerryn Y Xu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ji-Hoon Lee
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Juan Wang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Shi Chen
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Peng Zhang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Noah Wingate
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Asra Noor
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Yaxia Yuan
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Robert Hromas
- Department of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Hongwei Zhou
- Department of Medicine, Columbia Center for Human Development and Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Karina Hamamoto
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - C Cameron Yin
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Fengxi Ye
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Andrés E Quesada
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Suming Huang
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Daohong Zhou
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - M James You
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Feng-Chun Yang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| | - Jianlong Wang
- Department of Medicine, Columbia Center for Human Development and Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Mingjiang Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
30
|
Xu S, Hong J, Dongye M, Lin J, Xue R, Huang Z, Xu J, Zhang Y, Leung AYH, Shen J, Zhang W, Liu W. The synergistic effect of c-Myb hyperactivation and Pu.1 deficiency induces Pelger-Huët anomaly and promotes sAML. Proc Natl Acad Sci U S A 2025; 122:e2416121122. [PMID: 40020188 PMCID: PMC11892618 DOI: 10.1073/pnas.2416121122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/28/2025] [Indexed: 03/12/2025] Open
Abstract
Approximately 30% of patients with myelodysplastic syndrome (MDS) progress to secondary acute myeloid leukemia (sAML) via accumulating gene mutations. Genomic analyses reveal a complex interplay among mutant genes, with co-occurring and mutually exclusive patterns. Hyperactivation of c-MYB and deficiency of PU.1 have been linked to myeloid disorders. We report a case of AML with concurrent PU.1 and c-MYB mutations, exhibiting early onset, high blast count, chemo-resistance, indicating high-risk features, along with elevated Pelger-Huët anomaly (PHA). However, the synergistic mechanism of c-MYB and PU.1 in sAML remains unclear. Using c-Myb-hyperactivation and Pu.1-deficient double-strain (c-mybhyper;pu.1G242D/G242D) zebrafish, we investigated MDS/sAML progression. Surprisingly, the double mutant exhibited a distinct type of neutrophil resembling clinical PHA cells and demonstrated a higher rate of MDS/sAML transformation. Further expression analysis revealed reduced lmnb1 expression in double-mutant zebrafish. Knockdown of lmnb1 resulted in PHA and increased blast cells, while overexpression of lmnb1 in c-mybhyper;pu.1G242D/G242D reduced PHA cell level. This suggests that c-Myb hyperactivation and Pu.1 deficiency synergistically reduce lmnb1 expression, inducing the development of PHA-like neutrophils and promoting MDS/sAML progression in zebrafish. Moreover, coadministration of cell cycle inhibitor cytarabine (Ara-C) and the differential inducer all-trans retinoic acid (ATRA) could effectively relieve the neutrophil expansion and PHA symptoms in c-mybhyper;pu.1G242D/G242D zebrafish. Our findings revealed that c-Myb hyperactivation and Pu.1 deficiency played a synergistic role in sAML development and suggests a phenotypic association between the emergence of PH-like cells and the transformation to sAML. Furthermore, c-mybhyper;pu.1G242D/G242D zebrafish might serve as a suitable sAML model for drug screening.
Collapse
Affiliation(s)
- Song’en Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| | - Jiaxin Hong
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| | - Meimei Dongye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou510060, China
| | - Jiehao Lin
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| | - Rongtao Xue
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Zhibin Huang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| | - Anskar Yu-Hung Leung
- Division of Hematology, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Special Administrative Region of China
| | - Juan Shen
- Department of Marine Pharmacy, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou510006, China
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou510006, China
| | - Wenqing Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| | - Wei Liu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou510006, China
| |
Collapse
|
31
|
Sturgeon CM, Wagenblast E, Izzo F, Papapetrou EP. The Crossroads of Clonal Evolution, Differentiation Hierarchy, and Ontogeny in Leukemia Development. Blood Cancer Discov 2025; 6:94-109. [PMID: 39652739 PMCID: PMC11876951 DOI: 10.1158/2643-3230.bcd-24-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/19/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
SIGNIFICANCE In recent years, remarkable technological advances have illuminated aspects of the pathogenesis of myeloid malignancies-yet outcomes for patients with these devastating diseases have not significantly improved. We posit that a synthesized view of the three dimensions through which hematopoietic cells transit during their healthy and diseased life-clonal evolution, stem cell hierarchy, and ontogeny-promises high yields in new insights into disease pathogenesis and new therapeutic avenues.
Collapse
Affiliation(s)
- Christopher M. Sturgeon
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elvin Wagenblast
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pediatrics, Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Franco Izzo
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eirini P. Papapetrou
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
32
|
Yun J. Reclassification of Acute Myeloid Leukemia According to the 2022 World Health Organization Classification and the International Consensus Classification Using Open-Source Data. Ann Lab Med 2025; 45:170-177. [PMID: 39676421 PMCID: PMC11788709 DOI: 10.3343/alm.2024.0194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
Background In 2022, the revised WHO classification and International Consensus Classification (ICC) for myeloid neoplasms were published. We examined the impact of these guidelines on AML diagnoses alongside the 2022 European LeukemiaNet (ELN) recommendations on risk stratification. Methods We included 450 adult patients with newly diagnosed AML (non-acute promyelocytic leukemia) from the cBioPortal open-source dataset. Diagnoses and risk stratifications were revised based on the new guidelines and compared with the 2017 WHO classification. Survival analyses were performed using Cox regression. Results Among the patients included, 190 (42.2%) had consistent diagnoses across the three classifications, whereas 225 (50.0%) had inconsistent diagnoses. The two major WHO 2017 subtypes, AML not otherwise specified (AML-NOS) and AML with myelodysplasia-related changes (AML-MRC), were further subdivided according to the WHO 2022 and ICC. The ICC had the highest prognostication power among the three classifications. Subgroup analysis according to the different definitions of myelodysplasia-related AML and the introduction of AML with mutated TP53 (AML-TP53) showed that the differentiation of AML-TP53 was beneficial. The update from ELN 2017 to ELN 2022 resulted in significant transitions in a subset of patients. The updated diagnostic classification and ELN risk stratification (i.e., the ICC and ELN 2022) showed a straightforward relationship. Conclusions This study presents an integrative comparative analysis of past and current guidelines for AML diagnosis and risk classification based on open-source data. The ICC diagnostic criteria are clinically significant for determining AML prognosis. In line with the changing treatment paradigm for AML, future research is needed to continuously validate diagnostic and risk stratification systems.
Collapse
Affiliation(s)
- Jiwon Yun
- Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
33
|
Gurashi K, Wang YH, Amaral FMR, Spence K, Cant R, Yao CY, Lin CC, Wirth C, Wedge DC, Montalban-Bravo G, Colla S, Tien HF, Somervaille TCP, Batta K, Wiseman DH. An integrative multiparametric approach stratifies putative distinct phenotypes of blast phase chronic myelomonocytic leukemia. Cell Rep Med 2025; 6:101933. [PMID: 39892394 PMCID: PMC11866517 DOI: 10.1016/j.xcrm.2025.101933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/19/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Approximately 30% of patients with chronic myelomonocytic leukemia (CMML) undergo transformation to a chemo-refractory blastic phase (BP-CMML). Seeking novel therapeutic approaches, we profiled blast transcriptomes from 42 BP-CMMLs, observing extensive transcriptional heterogeneity and poor alignment to current acute myeloid leukemia (AML) classifications. BP-CMMLs display distinctive transcriptomic profiles, including enrichment for quiescence and variability in drug response signatures. Integrating clinical, immunophenotype, and transcriptome parameters, Random Forest unsupervised clustering distinguishes immature and mature subtypes characterized by differential expression of transcriptional modules, oncogenes, apoptotic regulators, and patterns of surface marker expression. Subtypes differ in predicted response to AML drugs, validated ex vivo in primary samples. Iteratively refined stratification resolves a classification structure comprising five subtypes along a maturation spectrum, predictive of response to novel agents including consistent patterns for receptor tyrosine kinase (RTK), cyclin-dependent kinase (CDK), mechanistic target of rapamycin (MTOR), and mitogen-activated protein kinase (MAPK) inhibitors. Finally, we generate a prototype decision tree to stratify BP-CMML with high specificity and sensitivity, requiring validation but with potential clinical applicability to guide personalized drug selection for improved outcomes.
Collapse
Affiliation(s)
- Kristian Gurashi
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Yu-Hung Wang
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Fabio M R Amaral
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Katherine Spence
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Rachel Cant
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Chi-Yuan Yao
- The National University Hospital of Taiwan, Taipei, Taiwan
| | - Chien-Chin Lin
- The National University Hospital of Taiwan, Taipei, Taiwan
| | - Christopher Wirth
- Wedge Group, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - David C Wedge
- Wedge Group, Manchester Cancer Research Centre, University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester, UK
| | | | - Simona Colla
- Departments of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Hwei-Fang Tien
- The National University Hospital of Taiwan, Taipei, Taiwan
| | - Tim C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK; The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Kiran Batta
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| | - Daniel H Wiseman
- Epigenetic of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, UK; The Christie Hospital NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
34
|
Bao X, Chen Y, Chang J, Du J, Yang C, Wu Y, Sha Y, Li M, Chen S, Yang M, Liu SB. Machine learning-based bulk RNA analysis reveals a prognostic signature of 13 cell death patterns and potential therapeutic target of SMAD3 in acute myeloid leukemia. BMC Cancer 2025; 25:273. [PMID: 39955536 PMCID: PMC11830216 DOI: 10.1186/s12885-025-13658-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Dysregulation or abnormality of the programmed cell death (PCD) pathway is closely related to the occurrence and development of many tumors, including acute myeloid leukemia (AML). Studying the abnormal characteristics of PCD pathway-related molecular markers can provide a basis for prognosis prediction and targeted drug design in AML patients. METHODS A total of 1394 genes representing 13 different PCD pathways were examined in AML patients and healthy donors. The upregulated genes were analyzed for their ability to predict overall survival (OS) individually, and these prognostic genes were subsequently combined to construct a PCD-related prognostic signature via an integrated approach consisting of 101 models based on ten machine learning algorithms. RNA transcriptome and clinical data from multiple AML cohorts (TCGA-AML, GSE106291, GSE146173 and Beat AML) were obtained to develop and validate the AML prognostic model. RESULTS A total of 214 upregulated PCD-related genes were identified in AML patients, 39 of which were proven to be prognostic genes in the training cohort. On the basis of the average C-index and number of model genes identified from the machine learning combinations, a PCD index was developed and validated for predicting AML OS. A prognostic nomogram was then generated and validated on the basis of the PCD index, age and ELN risk stratification in the Beat AML cohort and the GSE146173 cohort, revealing satisfactory predictive power (AUC values ≥ 0.7). With different mutation patterns, a higher PCD index was associated with a worse OS. The PCD index was significantly related to higher scores for immunosuppressive cells and mature leukemia cell subtypes. As the gene most closely related to the PCD index, the expression of SMAD3 was further validated in vitro. AML cells harboring KMT2A rearrangements were more sensitive to the SMAD3 inhibitor SIS3, and the expression of the autophagy-related molecular marker LC3 was increased in KMT2A-rearranged cell lines after SIS3 monotherapy and combined treatment. CONCLUSION The PCD index and SMAD3 gene expression levels have potential prognostic value and can be used in targeted therapy for AML, and these findings can lead to the development of effective strategies for the combined treatment of high-risk AML patients.
Collapse
Affiliation(s)
- Xiebing Bao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yao Chen
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Jie Chang
- School of Public Health, Medical College of Soochow University, Suzhou, 215123, China
| | - Jiahui Du
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Chen Yang
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Yijie Wu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Yu Sha
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Ming Li
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Minfeng Yang
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong, 226019, China.
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| | - Song-Bai Liu
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China.
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China.
| |
Collapse
|
35
|
Yuan Q, Duren Z. Inferring gene regulatory networks from single-cell multiome data using atlas-scale external data. Nat Biotechnol 2025; 43:247-257. [PMID: 38609714 PMCID: PMC11825371 DOI: 10.1038/s41587-024-02182-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 02/26/2024] [Indexed: 04/14/2024]
Abstract
Existing methods for gene regulatory network (GRN) inference rely on gene expression data alone or on lower resolution bulk data. Despite the recent integration of chromatin accessibility and RNA sequencing data, learning complex mechanisms from limited independent data points still presents a daunting challenge. Here we present LINGER (Lifelong neural network for gene regulation), a machine-learning method to infer GRNs from single-cell paired gene expression and chromatin accessibility data. LINGER incorporates atlas-scale external bulk data across diverse cellular contexts and prior knowledge of transcription factor motifs as a manifold regularization. LINGER achieves a fourfold to sevenfold relative increase in accuracy over existing methods and reveals a complex regulatory landscape of genome-wide association studies, enabling enhanced interpretation of disease-associated variants and genes. Following the GRN inference from reference single-cell multiome data, LINGER enables the estimation of transcription factor activity solely from bulk or single-cell gene expression data, leveraging the abundance of available gene expression data to identify driver regulators from case-control studies.
Collapse
Affiliation(s)
- Qiuyue Yuan
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, SC, USA
| | - Zhana Duren
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, SC, USA.
| |
Collapse
|
36
|
Gerritsen M, In 't Hout FEM, Knops R, Mandos BLR, Decker M, Ripperger T, van der Reijden BA, Martens JHA, Jansen JH. Acute myeloid leukemia associated RUNX1 variants induce aberrant expression of transcription factor TCF4. Leukemia 2025; 39:520-523. [PMID: 39668235 DOI: 10.1038/s41375-024-02470-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 12/14/2024]
Affiliation(s)
- Mylène Gerritsen
- Department of Laboratory medicine, Laboratory of Hematology, Radboud University medical center, Nijmegen, The Netherlands
| | - Florentien E M In 't Hout
- Department of Laboratory medicine, Laboratory of Hematology, Radboud University medical center, Nijmegen, The Netherlands
- Department of hematology, Radboud University medical center, Nijmegen, The Netherlands
| | - Ruth Knops
- Department of Laboratory medicine, Laboratory of Hematology, Radboud University medical center, Nijmegen, The Netherlands
| | - Bas L R Mandos
- Department of Laboratory medicine, Laboratory of Hematology, Radboud University medical center, Nijmegen, The Netherlands
| | - Melanie Decker
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Tim Ripperger
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Bert A van der Reijden
- Department of Laboratory medicine, Laboratory of Hematology, Radboud University medical center, Nijmegen, The Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Radboud University, Nijmegen, The Netherlands
| | - Joop H Jansen
- Department of Laboratory medicine, Laboratory of Hematology, Radboud University medical center, Nijmegen, The Netherlands.
| |
Collapse
|
37
|
Hegde M, Ahmad MH, Mulet Lazaro R, Sugita M, Li R, Hu K, Gebhard C, Guzman ML, Bushweller JH, Zhu LJ, Brehm M, Wolfe SA, Delwel R, Castilla LH. The co-receptor Neuropilin-1 enhances proliferation in inv(16) acute myeloid leukemia via VEGF signaling. Leukemia 2025; 39:360-370. [PMID: 39572712 DOI: 10.1038/s41375-024-02471-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 02/06/2025]
Abstract
Oncogenic programs regulate the proliferation and maintenance of cancer stem cells, and can define pharmacologic dependencies. In acute myeloid leukemia (AML) with the chromosome inversion 16 (inv(16)), the fusion oncoprotein CBFβ::MYH11 regulates pathways associated with leukemia stem cell activity. Here we demonstrate that expression of Neuropilin-1 (NRP1) is regulated by the fusion oncoprotein, and promotes AML expansion. Mechanistically, we show that the NRP1 locus has open chromatin in inv(16) AML, and that CBFβ::MYH11 modulates the local function of the transcription factors ERG, GATA2 and RUNX1 to sustain NRP1 levels. We found that ERG activates NRP1 expression, and that CBFβ::MYH11 knockdown represses ERG expression, thereby allowing the repressive activity of GATA2/RUNX1 at three NRP1 enhancers. Functionally, we demonstrate that NRP1 enhances the expansion of leukemic cells in vitro and in mice, and that this activity is dependent on its VEGFR-associated FV/FVIII domain. Finally, we show that treatment with VEGF inhibitor axitinib reduces AML cell growth and delays median leukemia latency in vivo. Our findings reveal that the NRP1/VEGF axis mediates proliferation in inv(16) AML blasts, and suggest that targeting NRP1 function could be promising in combination AML therapy.
Collapse
MESH Headings
- Neuropilin-1/genetics
- Neuropilin-1/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Humans
- Cell Proliferation
- Animals
- Mice
- Signal Transduction
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Chromosome Inversion
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Chromosomes, Human, Pair 16/genetics
- Transcriptional Regulator ERG
- Cell Line, Tumor
Collapse
Affiliation(s)
- Mahesh Hegde
- Department of Molecular, Cell and Cancer Biology; University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, USA
| | - Mohd H Ahmad
- Department of Molecular, Cell and Cancer Biology; University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, USA
| | - Roger Mulet Lazaro
- Department of Hematology, Erasmus University Medical Center and Oncode Institute, Rotterdam, The Netherlands
| | - Mayumi Sugita
- Department of Medicine, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Rui Li
- Department of Molecular, Cell and Cancer Biology; University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, USA
| | - Kai Hu
- Department of Molecular, Cell and Cancer Biology; University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, USA
| | - Claudia Gebhard
- Department of Internal Medicine III and Regensburg Centre for Interventional Immunology, University Hospital Regensburg, Regensburg, Germany
| | - Monica L Guzman
- Department of Medicine, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - John H Bushweller
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Lihua J Zhu
- Department of Molecular, Cell and Cancer Biology; University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, USA
| | - Michael Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology; University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, USA
| | - Ruud Delwel
- Department of Hematology, Erasmus University Medical Center and Oncode Institute, Rotterdam, The Netherlands
| | - Lucio H Castilla
- Department of Molecular, Cell and Cancer Biology; University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, USA.
| |
Collapse
|
38
|
Umeda M, Hiltenbrand R, Michmerhuizen NL, Barajas JM, Thomas ME, Arthur B, Walsh MP, Song G, Ma J, Westover T, Kumar A, Pölönen P, Mecucci C, Di Giacomo D, Locatelli F, Masetti R, Bertuccio SN, Pigazzi M, Pruett-Miller SM, Pounds S, Rubnitz J, Inaba H, Papadopoulos KP, Wick MJ, Iacobucci I, Mullighan CG, Klco JM. Fusion oncoproteins and cooperating mutations define disease phenotypes in NUP98-rearranged leukemia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.21.25320683. [PMID: 39974131 PMCID: PMC11838931 DOI: 10.1101/2025.01.21.25320683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Leukemias with NUP98 rearrangements exhibit heterogeneous phenotypes correlated to fusion partners, whereas the mechanism responsible for this heterogeneity is poorly understood. Through genome-wide mutational and transcriptional analyses of 177 NUP98-rearranged leukemias, we show that cooperating alterations are associated with differentiation status even among leukemias sharing the same NUP98 fusions, such as NUP98::KDM5A acute megakaryocytic leukemia with RB1 loss or T-cell acute lymphoblastic leukemia with NOTCH1 mutations. CUT&RUN profiling reveals that NUP98 fusion oncoproteins directly regulate differentiation-related genes, with binding patterns also influenced by differentiation stage. Using in vitro models, we show RB1 loss cooperates with NUP98::KDM5A by blocking terminal differentiation toward platelets and expanding megakaryocyte-like cells, whereas WT1 frameshifts skew differentiation toward dormant lympho-myeloid primed progenitor cells and cycling granulocyte-monocyte progenitor cells. NUP98::KDM5A models with RB1 or WT1 alterations have different sensitivities to menin inhibition, suggesting cellular differentiation stage-specific resistant mechanism against menin inhibitors with clinical implications for NUP98-rearranged leukemia.
Collapse
Affiliation(s)
- Masayuki Umeda
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Ryan Hiltenbrand
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | | | - Juan M. Barajas
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Melvin E. Thomas
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Bright Arthur
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Michael P Walsh
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Guangchun Song
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Jing Ma
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Tamara Westover
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Amit Kumar
- Center of Excellence for Leukemia Studies (CELS), St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Petri Pölönen
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Cristina Mecucci
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy
| | - Danika Di Giacomo
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Franco Locatelli
- Department of Onco-Haematology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - Riccardo Masetti
- Pediatric Oncology and Hematology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | | - Martina Pigazzi
- Department of Women’s and Children’s Health, Onco-hematology lab and clinic, University of Padova, Padova, Italy
| | - Shondra M. Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, TN, US
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Jeffrey Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | | | - Michael J. Wick
- XenoSTART/ The START Center for Cancer Research, San Antonio, TX, US
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
- Center of Excellence for Leukemia Studies (CELS), St. Jude Children’s Research Hospital, Memphis, TN, US
| | - Jeffery M. Klco
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, US
- Center of Excellence for Leukemia Studies (CELS), St. Jude Children’s Research Hospital, Memphis, TN, US
| |
Collapse
|
39
|
Marchand V, Laplane L, Valensi L, Plo I, Aglave M, Silvin A, Pasquier F, Porteu F, Vainchenker W, Selimoglu-Buet D, Droin N, Raslova H, Marcel V, Diaz JJ, Fontenay M, Solary E. Monocytes generated by interleukin-6-treated human hematopoietic stem and progenitor cells secrete calprotectin that inhibits erythropoiesis. iScience 2025; 28:111522. [PMID: 39811665 PMCID: PMC11732210 DOI: 10.1016/j.isci.2024.111522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/02/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Elevated circulating levels of calprotectin (CAL), the S100A8/A9 heterodimer, are biomarkers of severe systemic inflammation. Here, we investigate the effects of CAL on early human hematopoiesis. CAL demonstrates limited impact on gene expression in stem and progenitor cells, in contrast with interleukin-6 (IL6), which promotes the expression of the S100A8 and S100A9 genes in hematopoietic progenitors and the generation of monocytes that release CAL. The main target of CAL is an erythroid-megakaryocyte progenitor (EMP) subset. CAL prevents both erythropoietin-driven differentiation of healthy progenitors and JAK2-V617F-driven erythropoiesis. In the context of JAK2-V617F, CAL also promotes the expression of S100A8 and S100A9 genes in monocytes. The signature of CAL effects is detected in the bone marrow progenitors of patients with myeloid malignancy or severe infection. These results position CAL as a mediator of IL6 effects on triggering anemia during inflammation, an effect that is amplified in the context of JAK2-V617F-driven hematopoiesis.
Collapse
Affiliation(s)
- Valentine Marchand
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Lucie Laplane
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- CNRS 8590, Université Paris 1 Panthéon-Sorbonne, Paris, France
| | - Louis Valensi
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Isabelle Plo
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Marine Aglave
- AMMICa, INSERM US 23, CNRS UMS 3655, Gustave Roussy Cancer Center, Villejuif, France
| | - Aymeric Silvin
- INSERM U1108, Gustave Roussy Cancer Center, Villejuif, France
| | - Florence Pasquier
- Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Françoise Porteu
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - William Vainchenker
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Nathalie Droin
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- AMMICa, INSERM US 23, CNRS UMS 3655, Gustave Roussy Cancer Center, Villejuif, France
| | - Hana Raslova
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Virginie Marcel
- Inserm U1052, CNRS UMR5286 Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jean-Jacques Diaz
- Inserm U1052, CNRS UMR5286 Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Michaela Fontenay
- Université Paris Cité, Institut Cochin, CNRS UMR 8104, INSERM U1016, Paris, France
- Laboratory of Excellence for Red Blood Cells, GR-Ex, Paris, France
| | - Eric Solary
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| |
Collapse
|
40
|
Kong T, Laranjeira ABA, Letson CT, Yu L, Lin S, Fowles JS, Fisher DAC, Ng S, Yang W, He F, Youn M, Mark K, Jose AS, Liu J, Kim AB, Cox MJ, Fulbright MC, Jayanthan A, Los G, Rentschler SL, Ding L, Sakamoto KM, Dunn SE, Challen GA, Oh ST. RSK1 is an exploitable dependency in myeloproliferative neoplasms and secondary acute myeloid leukemia. Nat Commun 2025; 16:492. [PMID: 39820365 PMCID: PMC11739599 DOI: 10.1038/s41467-024-55643-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
Myeloid malignancies are heterogenous disorders characterized by distinct molecular drivers but share convergence of oncogenic signaling pathways and propagation by ripe pro-inflammatory niches. Here, we establish a comprehensive transcriptional atlas across the spectrum of myeloproliferative neoplasms (MPN) and secondary acute myeloid leukemia (sAML) through RNA-sequencing of 158 primary samples encompassing CD34+ hematopoietic stem/progenitor cells and CD14+ monocytes. Supported by mass cytometry (CyTOF) profiling, we reveal aberrant networks of PI3K/AKT/mTOR signalling and NFκB-mediated hyper-inflammation. Combining ATAC-Seq, CUT&Tag, RNA-seq, and CyTOF, we demonstrate that targeting of ribosomal protein S6 kinase A1 (RSK1) suppresses NFκB activation and diminishes pro-inflammatory mediators including tumor necrosis factor (TNF) associated with MPN disease severity and transformation. We further evaluate a therapeutic approach utilizing a first-in-class RSK inhibitor, PMD-026, currently in Phase 2 development for breast cancer, for use in myeloid malignancies. Treatment with PMD-026 suppressed disease burden across seven syngeneic and patient-derived xenograft leukemia mouse models spanning the spectrum of driver and disease-modifying mutations. These findings uncover a therapeutic avenue for a conserved dependency across MPN and sAML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/genetics
- Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/metabolism
- Myeloproliferative Disorders/drug therapy
- Myeloproliferative Disorders/pathology
- Animals
- Mice
- Signal Transduction/drug effects
- Female
- NF-kappa B/metabolism
- Male
- Xenograft Model Antitumor Assays
- Hematopoietic Stem Cells/metabolism
Collapse
Affiliation(s)
- Tim Kong
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Angelo B A Laranjeira
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher T Letson
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - LaYow Yu
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Shuyang Lin
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jared S Fowles
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel A C Fisher
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sherwin Ng
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Fan He
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Minyoung Youn
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Kailen Mark
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Ana San Jose
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Jingxian Liu
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Alexander B Kim
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Bursky Center for Human Immunology & Immunotherapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Maggie J Cox
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mary C Fulbright
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Aarthi Jayanthan
- Phoenix Molecular Designs, Vancouver, BC, Canada, San Diego, CA, USA
| | - Gerrit Los
- Phoenix Molecular Designs, Vancouver, BC, Canada, San Diego, CA, USA
| | - Stacey L Rentschler
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Center for Noninvasive Cardiac Radioablation, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Li Ding
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Kathleen M Sakamoto
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Sandra E Dunn
- Phoenix Molecular Designs, Vancouver, BC, Canada, San Diego, CA, USA
| | - Grant A Challen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen T Oh
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Bursky Center for Human Immunology & Immunotherapy, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
41
|
Ladikou EE, Sharp K, Simoes FA, Jones JR, Burley T, Stott L, Vareli A, Kennedy E, Vause S, Chevassut T, Devi A, Ashworth I, Ross DM, Hartmann TN, Mitchell SA, Pepper CJ, Best G, Pepper AGS. A Novel In Vitro Model of the Bone Marrow Microenvironment in Acute Myeloid Leukemia Identifies CD44 and Focal Adhesion Kinase as Therapeutic Targets to Reverse Cell Adhesion-Mediated Drug Resistance. Cancers (Basel) 2025; 17:135. [PMID: 39796762 PMCID: PMC11719579 DOI: 10.3390/cancers17010135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Acute myeloid leukemia (AML) is an aggressive neoplasm. Although most patients respond to induction therapy, they commonly relapse due to recurrent disease in the bone marrow microenvironment (BMME). So, the disruption of the BMME, releasing tumor cells into the peripheral circulation, has therapeutic potential. METHODS Using both primary donor AML cells and cell lines, we developed an in vitro co-culture model of the AML BMME. We used this model to identify the most effective agent(s) to block AML cell adherence and reverse adhesion-mediated treatment resistance. RESULTS We identified that anti-CD44 treatment significantly increased the efficacy of cytarabine. However, some AML cells remained adhered, and transcriptional analysis identified focal adhesion kinase (FAK) signaling as a contributing factor; the adhered cells showed elevated FAK phosphorylation that was reduced by the FAK inhibitor, defactinib. Importantly, we demonstrated that anti-CD44 and defactinib were highly synergistic at diminishing the adhesion of the most primitive CD34high AML cells in primary autologous co-cultures. CONCLUSIONS Taken together, we identified anti-CD44 and defactinib as a promising therapeutic combination to release AML cells from the chemoprotective AML BMME. As anti-CD44 is already available as a recombinant humanized monoclonal antibody, the combination of this agent with defactinib could be rapidly tested in AML clinical trials.
Collapse
Affiliation(s)
- Eleni E. Ladikou
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Kim Sharp
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Fabio A. Simoes
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - John R. Jones
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Thomas Burley
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Lauren Stott
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Aimilia Vareli
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Emma Kennedy
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Sophie Vause
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Timothy Chevassut
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
- Department of Haematology, Brighton and Sussex University Hospital Trust, Brighton BN2 5BE, UK;
| | - Amarpreet Devi
- Department of Haematology, Brighton and Sussex University Hospital Trust, Brighton BN2 5BE, UK;
| | - Iona Ashworth
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - David M. Ross
- Department of Haematology, Flinders Medical Centre, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.M.R.); (G.B.)
| | - Tanja Nicole Hartmann
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany;
| | - Simon A. Mitchell
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Chris J. Pepper
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| | - Giles Best
- Department of Haematology, Flinders Medical Centre, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.M.R.); (G.B.)
| | - Andrea G. S. Pepper
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PX, UK; (K.S.); (F.A.S.); (J.R.J.); (T.B.); (L.S.); (A.V.); (E.K.); (S.V.); (T.C.); (I.A.); (S.A.M.); (C.J.P.); (A.G.S.P.)
| |
Collapse
|
42
|
Safa-Tahar-Henni S, Páez Martinez K, Gress V, Esparza N, Roques É, Bonnet-Magnaval F, Bilodeau M, Gagné V, Bresson E, Cardin S, El-Hachem N, Iasenza I, Alzial G, Boivin I, Nakamichi N, Soufflet AC, Mirela Pascariu C, Duchaine J, Mathien S, Bonneil É, Eppert K, Marinier A, Sauvageau G, Deblois G, Thibault P, Hébert J, Eaves CJ, Cellot S, Barabé F, Wilhelm BT. Comparative small molecule screening of primary human acute leukemias, engineered human leukemia and leukemia cell lines. Leukemia 2025; 39:29-41. [PMID: 39472547 PMCID: PMC11717705 DOI: 10.1038/s41375-024-02400-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/14/2024] [Accepted: 08/28/2024] [Indexed: 01/11/2025]
Abstract
Targeted therapeutics for high-risk cancers remain an unmet medical need. Here we report the results of a large-scale screen of over 11,000 molecules for their ability to inhibit the survival and growth in vitro of human leukemic cells from multiple sources including patient samples, de novo generated human leukemia models, and established human leukemic cell lines. The responses of cells from de novo models were most similar to those of patient samples, both of which showed striking differences from the cell-line responses. Analysis of differences in subtype-specific therapeutic vulnerabilities made possible by the scale of this screen enabled the identification of new specific modulators of apoptosis, while also highlighting the complex polypharmacology of anti-leukemic small molecules such as shikonin. These findings introduce a new platform for uncovering new therapeutic options for high-risk human leukemia, in addition to reinforcing the importance of the test sample choice for effective drug discovery.
Collapse
Affiliation(s)
- Safia Safa-Tahar-Henni
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Karla Páez Martinez
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Verena Gress
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Nayeli Esparza
- Centre de recherche en infectiologie du CHUL, Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Élodie Roques
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Florence Bonnet-Magnaval
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Mélanie Bilodeau
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Valérie Gagné
- Laboratory for High Throughput Biology, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
| | - Eva Bresson
- Centre de recherche en infectiologie du CHUL, Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Sophie Cardin
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Nehme El-Hachem
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Isabella Iasenza
- Centre for Translational Biology, McGill University Heath Centre Research Institute, Montréal, QC, Canada
| | - Gabriel Alzial
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Metabolic and Epigenetic Alterations in Cancer Research unit, Montréal, QC, Canada
| | - Isabel Boivin
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Molecular Genetics of Stem Cells Research Unit, Montréal, QC, Canada
| | - Naoto Nakamichi
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Anne-Cécile Soufflet
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
| | - Cristina Mirela Pascariu
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Proteomics and Bioanalytical Mass Spectrometry Research Unit, Montréal, QC, Canada
| | - Jean Duchaine
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- High throughput screening platform, Montréal, QC, Canada
| | - Simon Mathien
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- High throughput screening platform, Montréal, QC, Canada
| | - Éric Bonneil
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Proteomics and Bioanalytical Mass Spectrometry Research Unit, Montréal, QC, Canada
| | - Kolja Eppert
- Centre for Translational Biology, McGill University Heath Centre Research Institute, Montréal, QC, Canada
- Department of Pediatrics, McGill University, Montréal, QC, Canada
| | - Anne Marinier
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Medicinal Chemistry/Drug Discovery Unit, Montréal, QC, Canada
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Guy Sauvageau
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Molecular Genetics of Stem Cells Research Unit, Montréal, QC, Canada
- Institut universitaire d'hémato-oncologie et de thérapie cellulaire, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Geneviève Deblois
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Faculty of Medicine, Faculty of Pharmacy, University of Montréal, Montréal, QC, Canada
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Proteomics and Bioanalytical Mass Spectrometry Research Unit, Montréal, QC, Canada
- Department of Chemistry, Faculty of Arts and Sciences, Université de Montréal, Montréal, QC, Canada
| | - Josée Hébert
- Institut universitaire d'hémato-oncologie et de thérapie cellulaire, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Sonia Cellot
- Unité de recherche en immuno-hémato-oncologie Charles-Bruneau, Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| | - Frédéric Barabé
- Centre de recherche en infectiologie du CHUL, Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada.
- Department of Medicine, Université Laval, Quebec City, QC, Canada.
| | - Brian T Wilhelm
- Laboratory for High Throughput Biology, Montréal, QC, Canada.
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
43
|
Arafeh R, Shibue T, Dempster JM, Hahn WC, Vazquez F. The present and future of the Cancer Dependency Map. Nat Rev Cancer 2025; 25:59-73. [PMID: 39468210 DOI: 10.1038/s41568-024-00763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/30/2024]
Abstract
Despite tremendous progress in the past decade, the complex and heterogeneous nature of cancer complicates efforts to identify new therapies and therapeutic combinations that achieve durable responses in most patients. Further advances in cancer therapy will rely, in part, on the development of targeted therapeutics matched with the genetic and molecular characteristics of cancer. The Cancer Dependency Map (DepMap) is a large-scale data repository and research platform, aiming to systematically reveal the landscape of cancer vulnerabilities in thousands of genetically and molecularly annotated cancer models. DepMap is used routinely by cancer researchers and translational scientists and has facilitated the identification of several novel and selective therapeutic strategies for multiple cancer types that are being tested in the clinic. However, it is also clear that the current version of DepMap is not yet comprehensive. In this Perspective, we review (1) the impact and current uses of DepMap, (2) the opportunities to enhance DepMap to overcome its current limitations, and (3) the ongoing efforts to further improve and expand DepMap.
Collapse
Affiliation(s)
- Rand Arafeh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | | | | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
44
|
Tomska K, Scheinost S, Kivioja J, Kummer S, Do THL, Zenz T. Lymphoma and Leukemia Cell Vulnerabilities and Resistance Identified by Compound Library Screens. Methods Mol Biol 2025; 2865:259-272. [PMID: 39424728 DOI: 10.1007/978-1-0716-4188-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Response to anticancer agents is often restricted to subsets of patients. Recognition of factors underlying this heterogeneity and identification of biomarkers associated with response to drugs would greatly improve the efficacy of drug treatment. Platforms that can comprehensively map cellular response to compounds in high-throughput provide a unique tool to identify associated biomarkers and provide hypotheses for mechanisms underlying variable response. Such screens can be performed on cell lines and short-term cultures of primary cells to take advantage of the respective models' strength, which include, e.g., the ability to silence genes or introduce somatic mutations to cell lines. Cohorts of patient samples represent the natural diversity of cancers, including rarer mutations and combinatorial patterns of mutations that are often absent from existing cell lines. We here summarize a simple and scalable method for the measurement of viability after drug exposure based on ATP measurements as a surrogate for viability, which we use to measure and understand drug response in cell lines and primary cells.
Collapse
Affiliation(s)
- Katarzyna Tomska
- Department of Molecular Therapy in Hematology and Oncology, DKFZ & NCT Heidelberg, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Scheinost
- Department of Translational Oncology, German Cancer Research Center DKFZ & NCT Heidelberg, Heidelberg, Germany
| | - Jarno Kivioja
- Department of Medical Oncology and Hematology, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Sandra Kummer
- Department of Medical Oncology and Hematology, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Thi Huong Lan Do
- Department of Medical Oncology and Hematology, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital Zürich and University of Zürich, Zürich, Switzerland.
| |
Collapse
|
45
|
Dong Y, Yue T, Wang X, Huo Q, Li W, Zhang S, Zhao Y, Li D. MS4A3 regulates hematopoietic myeloid differentiation through ROS/TGF-β/p38MAPK pathway. Int Immunopharmacol 2024; 143:113578. [PMID: 39532018 DOI: 10.1016/j.intimp.2024.113578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
The hematopoietic homeostasis relies on the intricate regulation of hematopoietic stem cells during their proliferation and differentiation. Myeloid differentiation disorders can lead to chronic myeloid leukemia and acute myeloid leukemia. Previous studies have shown increased expression of MS4A3 in myeloid cells, suggesting that MS4A3 may play a critical role in hematopoietic myeloid differentiation. However, the underlying mechanism and its role in hematopoietic myeloid differentiation require further elucidation. In this study, using K562 cell lines with MS4A3 over-expression (oeMS4A3) and MS4A3 knockdown (shMS4A3), we demonstrated that the overexpression of MS4A3 resulted in an augmented skewing towards myeloid differentiation and cell cycle arrest at G0/G1. In addition, inhibition of ROS, TGF-β, and p38MAPK in oeMS4A3 K562 cells attenuated the skewing of myeloid differentiation. Furthermore, in vivo experiments revealed a slight myeloid differentiation suppression tendency in MS4A3 knockout mice. Taken together, we show that MS4A3 overexpression promote myeloid differentiation skewing through the activation of the ROS/p38MAPK/TGFβ pathway. This study underscored the role of MS4A3 in the hematopoietic myeloid differentiation.
Collapse
Affiliation(s)
- Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Tongpeng Yue
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Xinyue Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Qidong Huo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Wenxuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Shiyi Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
46
|
Kim D, Sutamtewagul G, Yu Y. FLT3 and NPM1 mRNA expression-based risk stratification of de novo acute Myeloid Leukemia. Leuk Res Rep 2024; 23:100494. [PMID: 39834628 PMCID: PMC11743808 DOI: 10.1016/j.lrr.2024.100494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Prognostication of acute myeloid leukemia (AML) at initial diagnosis relies on identification of pre-determined underlying genetic abnormalities. Nevertheless, the disease course of AML remains highly unpredictable and robust reliable prognostic biomarkers for newly diagnosed AML are lacking. We retrospectively explored two publicly available AML RNA-Seq datasets and found that inferior overall survival was associated with high-FLT3 and low-NPM1 transcript levels ("FLT3 high/NPM1 low") compared to low-FLT3 and high-NPM1 transcript levels ("FLT3 low/NPM1 high") in adult de novo AML patients, with a hazard ratio for death of at least 2. Transcript level-dependent differential overall survival was independent from the underlying FLT3 or NPM1 genotypes. Our two-gene RNA expression-based de novo AML risk stratification may supplement and fine-tune traditional genetic aberration-based prognostication methods.
Collapse
Affiliation(s)
- Donghyun Kim
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| | - Grerk Sutamtewagul
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| | - Yeonhwa Yu
- Independent Researcher, Gimpo-si, Gyeonggi-do 10090, South Korea
| |
Collapse
|
47
|
Pawar AS, Somers P, Alex A, George SS, Antony C, Verner R, White-Brown SK, Khera M, Mendoza-Figueroa MS, Liu KF, Morrissette JJD, Paralkar VR. Leukemia-mutated proteins PHF6 and PHIP form a chromatin complex that represses acute myeloid leukemia stemness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.625909. [PMID: 39677666 PMCID: PMC11642813 DOI: 10.1101/2024.11.29.625909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Myeloid leukemias are heterogeneous cancers with diverse mutations, sometimes in genes with unclear roles and unknown functional partners. PHF6 and PHIP are two poorly-understood chromatin-binding proteins recurrently mutated in acute myeloid leukemia (AML). PHF6 mutations are associated with poorer outcomes, while PHIP was recently identified as the most common selective mutation in Black patients in AML. Here, we show that PHF6 is a transcriptional repressor that suppresses a stemness gene network, and that PHF6 missense mutations, classified by current clinical algorithms as variants of unknown significance, produce unstable or non-functional protein. We present multiple lines of evidence converging on a critical mechanistic connection between PHF6 and PHIP. We show that PHIP loss phenocopies PHF6 loss, and that PHF6 requires PHIP to occupy chromatin and exert its downstream transcriptional program. Our work unifies PHF6 and PHIP, two disparate leukemia-mutated proteins, into a common functional complex that suppresses AML stemness.
Collapse
Affiliation(s)
- Aishwarya S Pawar
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Biomedical Graduate Studies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Patrick Somers
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Aleena Alex
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Subin S George
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Charles Antony
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Roman Verner
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sanese K White-Brown
- Center for Personalized Diagnostics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mohit Khera
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - María Saraí Mendoza-Figueroa
- Department of Biochemistry & Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kathy Fange Liu
- Department of Biochemistry & Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jennifer J D Morrissette
- Center for Personalized Diagnostics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Vikram R Paralkar
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
48
|
Li LX, Aguilar B, Gennari JH, Qin G. LM-Merger: A workflow for merging logical models with an application to gene regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612961. [PMID: 39345612 PMCID: PMC11429764 DOI: 10.1101/2024.09.13.612961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Motivation Gene regulatory network (GRN) models provide mechanistic understanding of genetic interactions that regulate gene expression and, consequently, influence cellular behavior. Dysregulated gene expression plays a critical role in disease progression and treatment response, making GRN models a promising tool for precision medicine. While researchers have built many models to describe specific subsets of gene interactions, more comprehensive models that cover a broader range of genes are challenging to build. This necessitates the development of automated approaches for merging existing models. Results We present LM-Merger, a workflow for semi-automatically merging logical GRN models. The workflow consists of five main steps: (a) model identification, (b) model standardization and annotation, (c) model verification, (d) model merging, and (d) model evaluation. We demonstrate the feasibility and benefit of this workflow with two pairs of published models pertaining to acute myeloid leukemia (AML). The integrated models were able to retain the predictive accuracy of the original models, while expanding coverage of the biological system. Notably, when applied to a new dataset, the integrated models outperformed the individual models in predicting patient response. This study highlights the potential of logical model merging to advance systems biology research and our understanding of complex diseases. Availability and implementation The workflow and accompanying tools, including modules for model standardization, automated logical model merging, and evaluation, are available at https://github.com/IlyaLab/LogicModelMerger/.
Collapse
Affiliation(s)
- Luna Xingyu Li
- Institute for Systems Biology, Seattle, WA 98109, United States of America
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, United States of America
| | - Boris Aguilar
- Institute for Systems Biology, Seattle, WA 98109, United States of America
| | - John H Gennari
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, United States of America
| | - Guangrong Qin
- Institute for Systems Biology, Seattle, WA 98109, United States of America
| |
Collapse
|
49
|
Yang X, Wang Q, Sun Y, Zhang Z, Wuchty S, Liang Z, Dong Y. Genomic differences of patients with hematologic malignancies in different age groups. Commun Biol 2024; 7:1630. [PMID: 39643631 PMCID: PMC11624270 DOI: 10.1038/s42003-024-07293-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024] Open
Abstract
Hematologic malignancies cause significant morbidity/mortality in both children and young adults (CYAs) as well as older adults (OAs). Yet their biological underpinnings remain inadequately understood. Here, we analyzed clinical and genomic disparities between CYAs and OAs in various hematologic malignancies. We found substantial differences in clinical features such as patient sex, ethnicity, metastasis rates, and tumor subtypes. Genomically, most CYA hematologic malignancies indicated lower mutational burden. Subsequently, we identified differentially mutated genes (DMGs) with varying mutation rates between CYAs and OAs, noting fewer mutations in CYAs for most genes such as TP53, TET2, and DNMT3A. In contrast, several DMGs (i.e., NRAS, KRAS, SMARCA4, ID3, PTPN11, WT1, and KIT) were overrepresented in CYAs. We further investigated human protein interacting partners of these identified DMGs that were highly mutated in CYAs/OAs, respectively, and found significant differences in network topological and functional roles. Notably, CYA malignancies demonstrated extensive copy number alterations (CNAs) and more driver gene fusions. In particular, four CNA differential genes (i.e., ARID1B, MYB, TP53, and ESR1) were overrepresented as amplifications and deletions in CYAs and OAs, respectively. Ultimately, we demonstrated a landscape comparative view of clinically actionable genetic events in CYAs and OAs, providing clues for age-related personalized treatment.
Collapse
Affiliation(s)
- Xiaodi Yang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Qian Wang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Yuhua Sun
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Ziding Zhang
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Stefan Wuchty
- Department of Computer Science, University of Miami, Miami, FL, USA
- Department of Biology, University of Miami, Miami, FL, USA
- Institute of Data Science and Computation, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Zeyin Liang
- Department of Hematology, Peking University First Hospital, Beijing, China.
| | - Yujun Dong
- Department of Hematology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
50
|
Karathanasis N, Papasavva PL, Oulas A, Spyrou GM. Combining clinical and molecular data for personalized treatment in acute myeloid leukemia: A machine learning approach. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108432. [PMID: 39316958 DOI: 10.1016/j.cmpb.2024.108432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND AND OBJECTIVE The standard of care in Acute Myeloid Leukemia patients has remained essentially unchanged for nearly 40 years. Due to the complicated mutational patterns within and between individual patients and a lack of targeted agents for most mutational events, implementing individualized treatment for AML has proven difficult. We reanalysed the BeatAML dataset employing Machine Learning algorithms. The BeatAML project entails patients extensively characterized at the molecular and clinical levels and linked to drug sensitivity outputs. Our approach capitalizes on the molecular and clinical data provided by the BeatAML dataset to predict the ex vivo drug sensitivity for the 122 drugs evaluated by the project. METHODS We utilized ElasticNet, which produces fully interpretable models, in combination with a two-step training protocol that allowed us to narrow down computations. We automated the genes' filtering step by employing two metrics, and we evaluated all possible data combinations to identify the best training configuration settings per drug. RESULTS We report a Pearson correlation across all drugs of 0.36 when clinical and RNA sequencing data were combined, with the best-performing models reaching a Pearson correlation of 0.67. When we trained using the datasets in isolation, we noted that RNA Sequencing data (Pearson: 0.36) attained three times the predictive power of whole exome sequencing data (Pearson: 0.11), with clinical data falling somewhere in between (Pearson 0.26). Lastly, we present a paradigm of clinical significance. We used our models' prediction as a drug sensitivity score to rank an individual's expected response to treatment. We identified 78 patients out of 89 (88 %) that the proposed drug was more potent than the administered one based on their ex vivo drug sensitivity data. CONCLUSIONS In conclusion, our reanalysis of the BeatAML dataset using Machine Learning algorithms demonstrates the potential for individualized treatment prediction in Acute Myeloid Leukemia patients, addressing the longstanding challenge of treatment personalization in this disease. By leveraging molecular and clinical data, our approach yields promising correlations between predicted drug sensitivity and actual responses, highlighting a significant step forward in improving therapeutic outcomes for AML patients.
Collapse
Affiliation(s)
- Nestoras Karathanasis
- Bioinformatics Department, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, 2371 Ayios Dometios, Nicosia, Cyprus.
| | - Panayiota L Papasavva
- Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, 2371 Ayios Dometios, Nicosia, Cyprus
| | - Anastasis Oulas
- Bioinformatics Department, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, 2371 Ayios Dometios, Nicosia, Cyprus
| | - George M Spyrou
- Bioinformatics Department, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, 2371 Ayios Dometios, Nicosia, Cyprus
| |
Collapse
|