1
|
Dong J, Konopleva M. Preclinical targeting of leukemia-initiating cells in the development future biologics for acute myeloid leukemia. Expert Opin Ther Targets 2025:1-15. [PMID: 40304258 DOI: 10.1080/14728222.2025.2500417] [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: 11/16/2024] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Leukemia-initiating cells (LICs) are a critical subset of cells driving acute myeloid leukemia (AML) relapse and resistance to therapy. They possess unique properties, including metabolic, epigenetic, and microenvironmental dependencies, making them promising therapeutic targets. AREAS COVERED This review summarizes preclinical advances in targeting AML LICs, including strategies to exploit metabolic vulnerabilities, such as the reliance on oxidative phosphorylation (OXPHOS), through the use of mitochondrial inhibitors; target epigenetic regulators like DOT1L (Disruptor of Telomeric Silencing 1-like) to disrupt LIC survival mechanisms; develop immunotherapies, including CAR (chimeric antigen receptor) T-cell therapy, and bispecific antibodies; and disrupt LIC interactions with the bone marrow microenvironment by inhibiting supportive niches. EXPERT OPINION LIC-targeted therapies hold significant promise for revolutionizing AML treatment by reducing relapse rates and improving long-term outcomes. However, challenges such as LIC heterogeneity, therapy resistance, and associated toxicity persist. Recent studies have illuminated the distinct biological characteristics of LICs, advancing our understanding of their behavior and vulnerabilities. These insights offer new opportunities to target LICs at earlier disease stages and to explore combination therapies with other targeted treatments, ultimately enhancing therapeutic efficacy and improving patient outcomes.
Collapse
Affiliation(s)
- Jiaxin Dong
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marina Konopleva
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Shu W, Yang Q, He D, Li Y, Le J, Cai Q, Dai H, Luo L, Chen B, Gong Y, Jin D. Impact of KIT mutation on efficacy of venetoclax and hypomethylating agents in newly diagnosed acute myeloid leukemia. Eur J Med Res 2025; 30:354. [PMID: 40312469 PMCID: PMC12046753 DOI: 10.1186/s40001-025-02637-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: 03/02/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND The combination of venetoclax (VEN) with hypomethylating agents (HMAs) has emerged as a new standard treatment for older or unfit patients with acute myeloid leukemia (AML). However, the predictive factors for VEN/HMA efficacy remain unclear. In our study, we performed the first analysis of the impact of KIT mutations on therapeutic outcomes in newly diagnosed AML patients undergoing VEN/HMA regimens. METHODS In this retrospective study, we included 16 KIT-mutant AML patients receiving VEN/HMA (Cohort A), 141 KIT-wild-type AML patients receiving VEN/HMA (Cohort B), and 69 KIT-mutant AML patients receiving intensive chemotherapy (IC) (Cohort C). We compared the differences in therapeutic efficacy among the different cohorts. Furthermore, we conducted multivariate analyses in patients receiving VEN/HMA to identify factors influencing therapeutic outcomes. RESULTS Compared to Cohort B, Cohort A exhibited significantly lower overall response rate (ORR) (18.8% vs. 72.3%, p < 0.001) and measurable residual disease (MRD) negativity rate (18.8% vs. 68.1%, p < 0.001), with a shorter median event-free survival (EFS) (1.9 months vs. 7.8 months, p < 0.001). No significant difference in overall survival (OS) was observed. Among KIT-mutant patients, IC showed superior ORR (78.3% vs. 18.8%, p < 0.001), MRD negativity rate (75.4% vs. 18.8%, p < 0.001), and EFS (12.2 months vs. 1.9 months, p < 0.001) compared to VEN/HMA. No significant difference in OS was observed between the two cohorts. Multivariate analysis confirmed KIT mutations as an independent predictor of lower ORR (OR 0.020, 95% CI 0.002-0.211, p = 0.001) and shorter EFS (HR 6.318, 95% CI 2.659-15.012, p < 0.001). CONCLUSIONS Our findings suggest that KIT mutations are associated with poor response and shorter EFS in AML patients treated with VEN/HMA, highlighting the importance of KIT mutation status in risk stratification and treatment selection.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/diagnosis
- Sulfonamides/therapeutic use
- Sulfonamides/administration & dosage
- Female
- Male
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Retrospective Studies
- Mutation
- Middle Aged
- Aged
- Proto-Oncogene Proteins c-kit/genetics
- Adult
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Aged, 80 and over
- Treatment Outcome
- DNA Methylation/drug effects
Collapse
Affiliation(s)
- Wenxiu Shu
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Qianqian Yang
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Donghua He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yi Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jing Le
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Qianqian Cai
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Hui Dai
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Liufei Luo
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Bingrong Chen
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Yuan Gong
- Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, 550001, China
| | - Dian Jin
- Department of Hematology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China.
| |
Collapse
|
3
|
Shimony S, Stahl M, Stone RM. Acute Myeloid Leukemia: 2025 Update on Diagnosis, Risk-Stratification, and Management. Am J Hematol 2025; 100:860-891. [PMID: 39936576 PMCID: PMC11966364 DOI: 10.1002/ajh.27625] [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: 11/22/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/13/2025]
Abstract
DISEASE OVERVIEW Acute myeloid leukemia (AML) is a bone marrow stem cell cancer that is often fatal despite available treatments. Diagnosis, risk assessment, monitoring, and therapeutic management of AML have changed dramatically in the last decade due to increased pathophysiologic understanding, improved assessment technology, and the addition of at least 12 approved therapies. DIAGNOSIS The diagnosis is based on the presence of immature leukemia cells in the blood, and/or bone marrow or less often in extra-medullary tissues. New biological insights have been integrated into recent classification systems. RISK ASSESSMENT The European Leukemia Network has published risk classification algorithms for both intensively and non-intensively treated patients based on cytogenetic and on molecular findings. Prognostic factors may differ based on the therapeutic approach. MONITORING Our increasing ability to quantify lower levels of measurable residual disease (MRD) potentially allows better response assessment, as well as dynamic monitoring of disease status. The incorporation of MRD findings into therapeutic decision-making is rapidly evolving. RISK ADAPTED THERAPY The availability of 12 newly approved agents has been welcomed; however, optimal strategies incorporating newer agents into therapeutic algorithms are debated. The overarching approach integrates patient and caregiver goals of care, comorbidities, and disease characteristics.
Collapse
Affiliation(s)
- Shai Shimony
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Maximilian Stahl
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Richard M. Stone
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| |
Collapse
|
4
|
Zhang X, Han Y, Qiu H, Han M, Sun A, Xue S, Jin Z, Miao M, Wang Y, Fu C, Tang X, Chen S, Li C, Bai L, Lin Z, Chen J, Han H, Chen J, Wu D. Cladribine Added to Idarubicin and Cytarabine as an Induction Regimen for Patients with De Novo Acute Myeloid Leukemia: A Multicenter, Randomized Phase III Trial. Clin Cancer Res 2025; 31:1407-1414. [PMID: 40008901 DOI: 10.1158/1078-0432.ccr-24-2437] [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: 09/02/2024] [Revised: 11/24/2024] [Accepted: 02/05/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE To assess the efficacy and safety of an induction regimen composed of idarubicin, cytarabine, and cladribine (IAC) in patients with de novo acute myeloid leukemia (AML). PATIENTS AND METHODS Adult patients with newly diagnosed AML were randomized to the IAC group (cladribine 5 mg/m2/day for 5 days, idarubicin 8 mg/m2/day for 3 days, and cytarabine 100 mg/m2/day for 7 days) and the IA group (idarubicin 12 mg/m2/day for 3 days and cytarabine 100 mg/m2/day for 7 days) at a 1:2 ratio. The primary endpoint was complete remission (CR) after induction. Secondary endpoints included 2-year overall survival (OS), disease-free survival, and cumulative incidence of relapse. RESULTS A total of 618 adult patients with newly diagnosed AML were enrolled. The overall CR rate was 80.5% in the IAC group compared with 72.4% in the IA group (P = 0.029). The 2-year OS was 81.3% in the IAC group compared with 70.0% in the IA group (P = 0.011). Patients on the IAC regimen achieved a higher CR rate compared to those on the IA regimen, particularly in those with adverse risk (69.8% vs. 49.1%, P = 0.008), 2-year OS (80.1% vs. IA 58.1%, P = 0.014), and disease-free survival (78.8% vs. 51.3%, P = 0.009). In the subgroup of patients older than 45 years of age, the IAC regimen exerted better CR (77.1% vs. 62.6%, P = 0.033) and 2-year OS (74.7% vs. IA 55.0%, P = 0.019). There were no differences in chemotherapy-related toxicities between the groups. CONCLUSIONS Cladribine added to the IA regimen was safe and effective in de novo AML. Patients with adverse risk or those between 45 and 60 years of age might benefit significantly on both response and survival with the IAC regimen.
Collapse
Affiliation(s)
- Xiang Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yue Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Huiying Qiu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Miaoxinqi Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Aining Sun
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengli Xue
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengming Jin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Miao Miao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chengcheng Fu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaowen Tang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caixia Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lian Bai
- Canglang Hospital of Suzhou, Suzhou, China
| | - Zhihong Lin
- Hygeia Suzhou Yongding Hospital, Suzhou, China
| | - Jun Chen
- Hygeia Suzhou Yongding Hospital, Suzhou, China
| | - Haohao Han
- Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Santos de Macedo BG, Albuquerque de Melo M, Pereira-Martins DA, Machado-Neto JA, Traina F. A common ground: an in silico assessment of the sources of intrinsic ex vivo resistance to venetoclax in acute myeloid leukemia. Hematol Transfus Cell Ther 2025; 47:103758. [PMID: 40222279 PMCID: PMC12019820 DOI: 10.1016/j.htct.2025.103758] [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: 08/06/2024] [Revised: 12/18/2024] [Accepted: 01/15/2025] [Indexed: 04/15/2025] Open
Abstract
Venetoclax is a promising alternative for patients with acute myeloid leukemia who are considered unfit for conventional chemotherapy; however, its employment still faces challenges mostly related to drug resistance. Here, we provide further biological mechanisms underlying the previously described and potentially novel intrinsic sources of poor response to venetoclax departing from ex vivo response data. Acute myeloid leukemia data including FLT3 mutation status, gene expression data, and ex vivo response data were extracted from the publicly available BeatAML 1.0 study database and aided sample categorization that supported differential gene expression analysis that, in turn, supported gene set enrichment analysis. CIBERSORTx-based bulk RNA sequencing deconvolution of BeatAML 1.0 data allowed us to categorize samples according to their cell type content. We observed that inflammation-related gene sets, such as cytokines and inflammatory response, NLRP3 inflammasome activation, and activation of adaptive immune response, were concordantly positively enriched across all the conditions reported to be associated with poor ex vivo venetoclax response, whereas samples from good ex vivo responders' mostly enriched gene sets related to mitochondrial activity, and early myeloid progenitor cell molecular programs. Besides the alternative reliance on BCL2A1, we highlight inflammation as a common element present across multiple sources of venetoclax ex vivo response modulation in acute myeloid leukemia samples. Hence, a potential key modulator for venetoclax response.
Collapse
Affiliation(s)
- Brunno Gilberto Santos de Macedo
- Department of Medical Images, Hematology, and Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Manuela Albuquerque de Melo
- Department of Medical Images, Hematology, and Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | | | - João Agostinho Machado-Neto
- Department of Medical Images, Hematology, and Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil; Department of Pharmacology, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Fabiola Traina
- Department of Medical Images, Hematology, and Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
6
|
Rodriguez-Sevilla JJ, Ganan-Gomez I, Kumar B, Thongon N, Ma F, Chien KS, Kim YJ, Yang H, Loghavi S, Tan R, Adema V, Li Z, Tanaka T, Uryu H, Kanagal-Shamanna R, Al-Atrash G, Bejar R, Banerjee PP, Lynn Cha S, Montalban-Bravo G, Dougherty M, Fernandez Laurita MC, Wheeler N, Jia B, Papapetrou EP, Izzo F, Dueñas DE, McAllen S, Gu Y, Todisco G, Ficara F, Della Porta MG, Jain A, Takahashi K, Clise-Dwyer K, Halene S, Bertilaccio MTS, Garcia-Manero G, Daher M, Colla S. Natural killer cells' functional impairment drives the immune escape of pre-malignant clones in early-stage myelodysplastic syndromes. Nat Commun 2025; 16:3450. [PMID: 40216768 PMCID: PMC11992119 DOI: 10.1038/s41467-025-58662-0] [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: 09/19/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Dissecting the preneoplastic disease states' biological mechanisms that precede tumorigenesis can lead to interventions that can slow down disease progression and/or mitigate disease-related comorbidities. Myelodysplastic syndromes (MDS) cannot be cured by currently available pharmacological therapies, which fail to eradicate aberrant hematopoietic stem cells (HSCs), most of which are mutated by the time of diagnosis. Here, we sought to elucidate how MDS HSCs evade immune surveillance and expand in patients with clonal cytopenias of undetermined significance (CCUS), the pre-malignant stage of MDS. We used multi-omic single-cell approaches and functional in vitro studies to show that immune escape at disease initiation is mainly mediated by mutant, dysfunctional natural killer (NK) cells with impaired cytotoxic capability against cancer cells. Preclinical in vivo studies demonstrated that injecting NK cells from healthy donors efficiently depleted CCUS mutant cells while allowing normal cells to regenerate hematopoiesis. Our findings suggest that early intervention with adoptive cell therapy can prevent or delay the development of MDS.
Collapse
Affiliation(s)
| | - Irene Ganan-Gomez
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Bijender Kumar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natthakan Thongon
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Feiyang Ma
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kelly S Chien
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Yi J Kim
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Yang
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roselyn Tan
- Moores Cancer Center, University of California San Diego, Moores Cancer Center, San Diego, CA, USA
| | - Vera Adema
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Zongrui Li
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Tomoyuki Tanaka
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Hidetaka Uryu
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rafael Bejar
- Moores Cancer Center, University of California San Diego, Moores Cancer Center, San Diego, CA, USA
| | - Pinaki Prosad Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sophia Lynn Cha
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Max Dougherty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Claudina Fernandez Laurita
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noelle Wheeler
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Baosen Jia
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Franco Izzo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniela E Dueñas
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Salome McAllen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yiqian Gu
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Gabriele Todisco
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Francesca Ficara
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
- Istituto di Ricerca Genetica e Biomedica, National Research Council, 20090, Milan, Italy
| | - Matteo Giovanni Della Porta
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Abhinav Jain
- Department of Epigenetics and Molecular Carcinogenesis, Center for Cancer Epigenetics, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of MD Anderson Cancer Center, Houston, TX, USA
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Simona Colla
- Department of Leukemia, The University of MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
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] [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
|
8
|
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
|
9
|
Xu JJ, Scoca V, Chen Y, Zhan YA, Fisher A, Udoh EO, Fernando S, Alija B, Pantazi J, Sudunagunta V, Stewart E, Galang AMD, Williams M, Bhagat G, Gebhard C, Visconte V, Ondrejka S, Delwel R, Hu M, Koche R, Viny AD. Enhanced FLI1 accessibility mediates STAG2-mutant leukemogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646632. [PMID: 40236115 PMCID: PMC11996548 DOI: 10.1101/2025.04.01.646632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Transcription factors (TFs) influencing cell fate can be dysregulated in cancer. FLI1 is crucial for hematopoietic stem/progenitor cell (HSPC) function, with STAG2 regulating FLI1 target accessibility. STAG2 depletion enhances HSPC self-renewal, but its role in leukemic transformation is unclear. We uncovered that STAG2 loss maintains FLI1 target accessibility in murine HSPCs and enhances FLI1 binding in NPM1c leukemia. In our Stag2/Npm1c/+ murine model, myeloid-biased HSPCs with increased FLI1 accessibility are reservoirs for transformation, leading to a fully penetrant leukemia. STAG2 deleted NPM1c cell lines exhibit increased chromatin accessibility and chromatin-looping of key stem and leukemia genes including FLI1-target genes CD34 and MEN1. Similarly, enrichment for a CD34+ immunophenotype was observed in co-mutant leukemia patients. STAG2 deficient cells show increased chromatin-bound MENIN and increased sensitivity to MENIN inhibition. Our findings demonstrate that altered chromatin architecture can co-opt oncogenic TF signaling, such as FLI1, as a hallmark of leukemogenesis. Key Findings Loss of STAG2 results in aberrant increased accessibility at FLI1 targets in mouse and human hematopoietic stem and progenitor cellsIncreased accessibility results in an increased fraction of chromatin-bound FLI1, which overlap with NPM1c targets in STAG2 NPM1c AML cellsStag2 Npm1c co-mutation leads to dysplastic murine AML phenotype arising from myeloid biased progenitors that exhibit increased Fli1 target accessibilityIn addition to higher chromatin-bound FLI1, co-mutant cells have higher chromatin-bound MENIN, including at the HOXA cluster, rendering cells highly sensitive to MENIN inhibition. Statement of Significance Here, we identify enhanced FLI1 chromatin accessibility as a driver of stemness and leukemic transformation in STAG2 mutant leukemia. Through comprehensive in vivo and in vitro modeling, we demonstrate that altered chromatin architecture can co-opt oncogenic TF activity, like FLI1, to drive divergent leukemia development and therapeutic response.
Collapse
|
10
|
Jiao CQ, Hu C, Sun MH, Li Y, Wu C, Xu F, Zhang L, Huang FR, Zhou JJ, Dai JF, Ruan M, Wang WC, Liu QS, Ge J. Targeting METTL3 mitigates venetoclax resistance via proteasome-mediated modulation of MCL1 in acute myeloid leukemia. Cell Death Dis 2025; 16:233. [PMID: 40169588 PMCID: PMC11962166 DOI: 10.1038/s41419-025-07560-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025]
Abstract
Venetoclax, a selective BCL2 inhibitor, is extensively utilized in clinical settings for the treatment of acute myeloid leukemia (AML). However, its efficacy is often compromised by the development of drug resistance. Hence, identification of potential venetoclax combination treatment strategies is imperative to overcome this acquired resistance. In this study, we discovered that inhibition of METTL3 can synergistically enhance the anti-leukemic efficacy of venetoclax, and is capable of overcoming venetoclax resistance in in vivo experiments and various venetoclax resistance models. Mechanistic study revealed that STM2457 augmented venetoclax activity by downregulating MCL1 and MYC, thereby increasing apoptosis in leukemia cells induced by venetoclax. Further investigation demonstrated that STM2457 promotes the ubiquitination and subsequent protein degradation of MCL1 primarily through pharmaceutically targeting METTL3. Moreover, through molecular docking-based virtual screening, we identified isoliquiritigenin as a potential novel small molecule natural product targeting METTL3, which exhibited potential effects as an anti-leukemic agent.
Collapse
MESH Headings
- Humans
- Sulfonamides/pharmacology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Methyltransferases/metabolism
- Methyltransferases/antagonists & inhibitors
- Drug Resistance, Neoplasm/drug effects
- Proteasome Endopeptidase Complex/metabolism
- Animals
- Cell Line, Tumor
- Mice
- Apoptosis/drug effects
- Antineoplastic Agents/pharmacology
- Mice, Nude
Collapse
Affiliation(s)
- Chang-Qing Jiao
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Chen Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Meng-Hua Sun
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Yan Li
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Chao Wu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Fei Xu
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Lei Zhang
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Fu-Rong Huang
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Jun-Jie Zhou
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Ji-Fei Dai
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Min Ruan
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China
| | - Wen-Chao Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Qing-Song Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Jian Ge
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China & Anhui Medical University, Hefei, 230032, China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| |
Collapse
|
11
|
Vogler M, Braun Y, Smith VM, Westhoff MA, Pereira RS, Pieper NM, Anders M, Callens M, Vervliet T, Abbas M, Macip S, Schmid R, Bultynck G, Dyer MJ. The BCL2 family: from apoptosis mechanisms to new advances in targeted therapy. Signal Transduct Target Ther 2025; 10:91. [PMID: 40113751 PMCID: PMC11926181 DOI: 10.1038/s41392-025-02176-0] [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: 11/07/2024] [Revised: 12/21/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The B cell lymphoma 2 (BCL2) protein family critically controls apoptosis by regulating the release of cytochrome c from mitochondria. In this cutting-edge review, we summarize the basic biology regulating the BCL2 family including canonical and non-canonical functions, and highlight milestones from basic research to clinical applications in cancer and other pathophysiological conditions. We review laboratory and clinical development of BH3-mimetics as well as more recent approaches including proteolysis targeting chimeras (PROTACs), antibody-drug conjugates (ADCs) and tools targeting the BH4 domain of BCL2. The first BCL2-selective BH3-mimetic, venetoclax, showed remarkable efficacy with manageable toxicities and has transformed the treatment of several hematologic malignancies. Following its success, several chemically similar BCL2 inhibitors such as sonrotoclax and lisaftoclax are currently under clinical evaluation, alone and in combination. Genetic analysis highlights the importance of BCL-XL and MCL1 across different cancer types and the possible utility of BH3-mimetics targeting these proteins. However, the development of BH3-mimetics targeting BCL-XL or MCL1 has been more challenging, with on-target toxicities including thrombocytopenia for BCL-XL and cardiac toxicities for MCL1 inhibitors precluding clinical development. Tumor-specific BCL-XL or MCL1 inhibition may be achieved by novel targeting approaches using PROTACs or selective drug delivery strategies and would be transformational in many subtypes of malignancy. Taken together, we envision that the targeting of BCL2 proteins, while already a success story of translational research, may in the foreseeable future have broader clinical applicability and improve the treatment of multiple diseases.
Collapse
Affiliation(s)
- Meike Vogler
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) partner site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany.
| | - Yannick Braun
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Victoria M Smith
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Raquel S Pereira
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Nadja M Pieper
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Marius Anders
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Manon Callens
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Tim Vervliet
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Maha Abbas
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Ralf Schmid
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Institute for Structural and Chemical Biology, University of Leicester, Leicester, UK
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Martin Js Dyer
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
12
|
Decker M, Huang BJ, Ware T, Boone C, Tang M, Ybarra J, Ballapuram AC, Taran KA, Chen PY, Amendáriz M, Leung CJ, Harris M, Tjoa K, Hongo H, Abelson S, Rivera J, Ngo N, Herbst DM, Suciu RM, Guijas C, Sedighi K, Andalis T, Roche E, Xie B, Liu Y, Smith CC, Stieglitz E, Niphakis MJ, Cravatt BF, Shannon K. In vivo validation of the palmitoylation cycle as a therapeutic target in NRAS-mutant cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644389. [PMID: 40166265 PMCID: PMC11957127 DOI: 10.1101/2025.03.20.644389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Normal and oncogenic Ras proteins are functionally dependent on one or more lipid modifications1,2. Whereas K-Ras4b farnesylation is sufficient for stable association with the plasma membrane, farnesylated H-Ras, K-Ras4a, and N-Ras traffic to the Golgi where they must undergo palmitoylation before regulated translocation to cell membranes. N-Ras palmitoylation by the DHHC family of palmitoyl acyl transferases (PATs) and depalmitoylation by ABHD17 serine hydrolases is a dynamic process that is essential for the growth of acute myeloid leukemias (AMLs) harboring oncogenic NRAS mutations3-6. Here, we have tested whether co-targeting ABHD17 enzymes and Ras signal output would cooperatively inhibit the proliferation and survival of NRAS-mutant AMLs while sparing normal tissues that retain K-Ras4b function. We show that ABD778, a potent and selective ABHD17 inhibitor with in vivo activity, selectively reduces the growth of NRAS-mutant AML cells in vitro and is synergistic with the allosteric MEK inhibitor PD0325901 (PD901)7,8. Similarly, ABD778 and PD901 significantly extended the survival of recipient mice transplanted with three independent primary mouse AMLs harboring an oncogenic Nras G12D driver mutation. Resistant leukemias that emerged during continuous drug treatment acquired by-pass mutations that confer adaptive drug resistance and increase mitogen activated protein kinase (MAPK) signal output. ABD778 augmented the anti-leukemia activity of the pan-PI3 kinase inhibitor pictilisib9, the K/N-RasG12C inhibitor sotorasib10, and the FLT3 inhibitor gilteritinib11. Co-treatment with ABD778 and gilteritinib restored drug sensitivity in a patient-derived xenograft model of adaptive resistance to FLT3 inhibition. These data validate the palmitoylation cycle as a promising therapeutic target in AML and support exploring it in other NRAS-mutant cancers.
Collapse
Affiliation(s)
- Matthew Decker
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin J. Huang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Timothy Ware
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher Boone
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle Tang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Ybarra
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Katrine A. Taran
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Pan-Yu Chen
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Marcos Amendáriz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Camille J. Leung
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Max Harris
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Karensa Tjoa
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Hongo
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Sydney Abelson
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jose Rivera
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Nhi Ngo
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | | | - Radu M. Suciu
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Carlos Guijas
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Kimia Sedighi
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Taylor Andalis
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Elysia Roche
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Boer Xie
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Yunlong Liu
- Center for Center for Medical Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Catherine C. Smith
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Elliot Stieglitz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Kevin Shannon
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Wei AH, Loo S, Daver N. How I treat patients with AML using azacitidine and venetoclax. Blood 2025; 145:1237-1250. [PMID: 39316723 DOI: 10.1182/blood.2024024009] [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: 05/03/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
ABSTRACT Venetoclax (VEN) received full approval in October 2020 for use in older patients who are unfit with acute myeloid leukemia (AML) combined with either hypomethylating agents or low-dose cytarabine. This ended a semicentennial of stalled clinical progress and initiated a new treatment option with proven capacity to enhance response and prolong survival in older patients with AML. Despite widespread use of azacitidine-VEN (AZA-VEN), there is increasing appreciation that this regimen is myelosuppressive and associated with a higher risk of infectious complications than AZA alone. Key principles of initial management include prevention of tumor lysis syndrome in patients at high risk and minimizing infectious complications during induction. In the postremission phase, limiting cumulative marrow suppression by allowing sufficient time between cycles for optimal marrow recovery and truncating the duration of VEN exposure for those with delayed blood count recovery have emerged as important axioms of effective care. This article casts a clinical spotlight on important challenges and dilemmas encountered in practice. We also outline a structured framework to assist in the safe management of AZA-VEN in the clinic.
Collapse
Affiliation(s)
- Andrew H Wei
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Melbourne, Australia
| | - Sun Loo
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Melbourne, Australia
- Department of Haematology, Northern Hospital, Epping, VIC, Australia
| | - Naval Daver
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
14
|
Shi X, Li M, Liu Z, Tiessen J, Li Y, Zhou J, Zhu Y, Mahesula S, Ding Q, Tan L, Feng M, Kageyama Y, Hara Y, Tao JJ, Luo X, Patras KA, Lorenzi PL, Huang S, Stevens AM, Takahashi K, Issa GC, Samee MAH, Agathocleous M, Nakada D. Guanine nucleotide biosynthesis blockade impairs MLL complex formation and sensitizes leukemias to menin inhibition. Nat Commun 2025; 16:2641. [PMID: 40102405 PMCID: PMC11920272 DOI: 10.1038/s41467-025-57544-9] [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: 06/13/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
Targeting the dependency of MLL-rearranged (MLLr) leukemias on menin with small molecule inhibitors has opened new therapeutic strategies for these poor-prognosis diseases. However, the rapid development of menin inhibitor resistance calls for combinatory strategies to improve responses and prevent resistance. Here we show that leukemia stem cells (LSCs) of MLLr acute myeloid leukemia (AML) exhibit enhanced guanine nucleotide biosynthesis, the inhibition of which leads to myeloid differentiation and sensitization to menin inhibitors. Mechanistically, targeting inosine monophosphate dehydrogenase 2 (IMPDH2) reduces guanine nucleotides and rRNA transcription, leading to reduced protein expression of LEDGF and menin. Consequently, the formation and chromatin binding of the MLL-fusion complex is impaired, reducing the expression of MLL target genes. Inhibition of guanine nucleotide biosynthesis or rRNA transcription further suppresses MLLr AML when combined with a menin inhibitor. Our findings underscore the requirement of guanine nucleotide biosynthesis in maintaining the function of the LEDGF/menin/MLL-fusion complex and provide a rationale to target guanine nucleotide biosynthesis to sensitize MLLr leukemias to menin inhibitors.
Collapse
MESH Headings
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Myeloid-Lymphoid Leukemia Protein/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/antagonists & inhibitors
- Humans
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Animals
- Mice
- IMP Dehydrogenase/metabolism
- IMP Dehydrogenase/antagonists & inhibitors
- IMP Dehydrogenase/genetics
- Cell Line, Tumor
- Gene Expression Regulation, Leukemic/drug effects
Collapse
Affiliation(s)
- Xiangguo Shi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Department of Molecular and Precision Medicine, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Minhua Li
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zian Liu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan Tiessen
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuan Li
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jing Zhou
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yudan Zhu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Swetha Mahesula
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Qing Ding
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mengdie Feng
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuki Kageyama
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yusuke Hara
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jacob J Tao
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xuan Luo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Alexandra M Stevens
- Section of Hematology/Oncology, Department of Pediatrics, Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michalis Agathocleous
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Daisuke Nakada
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Baronas D, Zvirblyte J, Norvaisis S, Leonaviciene G, Goda K, Mikulenaite V, Kaseta V, Sablauskas K, Griskevicius L, Juzenas S, Mazutis L. High-throughput single cell -omics using semi-permeable capsules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.642805. [PMID: 40166174 PMCID: PMC11957016 DOI: 10.1101/2025.03.14.642805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Biological systems are inherently complex and heterogeneous. Deciphering this complexity increasingly relies on high-throughput analytical methods and tools that efficiently probe the cellular phenotype and genotype. While recent advancements have enabled various single-cell -omics assays, their broader applications are inherently limited by the challenge of efficiently conducting multi-step biochemical assays while retaining various biological analytes. Extending on our previous work (1) here we present a versatile technology based on semi-permeable capsules (SPCs), tailored for a variety of high-throughput nucleic acid assays, including digital PCR, genome sequencing, single-cell RNA-sequencing (scRNA-Seq) and FACS-based isolation of individual transcriptomes based on nucleic acid marker of interest. Being biocompatible, the SPCs support single-cell cultivation and clonal expansion over long periods of time - a fundamental limitation of droplet microfluidics systems. Using SPCs we perform scRNA-Seq on white blood cells from patients with hematopoietic disorders and demonstrate that capsule-based sequencing approach (CapSeq) offers superior transcript capture, even for the most challenging cell types. By applying CapSeq on acute myeloid leukemia (AML) samples, we uncover notable changes in transcriptomes of mature granulocytes and monocytes associated with blast and progenitor cell phenotypes. Accurate representation of the entirety of the cellular heterogeneity of clinical samples, driving new insights into the malfunctioning of the innate immune system, and ability to clonally expand individual cells over long periods of time, positions SPC technology as customizable, highly sensitive and broadly applicable tool for easy-to-use, scalable single-cell -omics applications.
Collapse
Affiliation(s)
- Denis Baronas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Justina Zvirblyte
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Simonas Norvaisis
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Greta Leonaviciene
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Karolis Goda
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vincenta Mikulenaite
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vytautas Kaseta
- State Research Institute Centre for Innovative Medicine, Department of Stem Cell Biology, Vilnius, Lithuania
| | - Karolis Sablauskas
- Hematology, Oncology and Transfusion Medicine Center, National Cancer Center, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
- Institute of Data Science and Digital Technologies, Vilnius University, Vilnius, Lithuania
| | - Laimonas Griskevicius
- Hematology, Oncology and Transfusion Medicine Center, National Cancer Center, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Simonas Juzenas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Linas Mazutis
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Department of Molecular Biology, Umea University, Sweden
| |
Collapse
|
16
|
Carraway HE, Brunner AM, Lai CE, Luskin MR, Park J, Perl AE, Stein EM, Wang ES, Zeidan AM, Zeidner JF, Komrokji R. Advancing the Management of CH, MDS, and AML From the First Bridging the Gaps in Leukemia, Lymphoma, and Multiple Myeloma Conference. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025:S2152-2650(25)00082-5. [PMID: 40187939 DOI: 10.1016/j.clml.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 04/07/2025]
Abstract
PURPOSE The management of myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) have evolved substantially in recent years with the development of targeted therapies and novel nontargeted approaches. However, many questions remain about how to best use current therapies, and there is a large unmet need for effective therapies, particularly for patients with higher-risk MDS, AML, and those with MDS/AML relapsed/refractory (R/R) to prior therapy. METHODS AND RESULTS A panel of experts was assembled to discuss current controversies and unanswered questions in the care of patients with MDS and AML. Workshop topics included: molecular testing and new classification systems, clonal hematopoiesis, treatment of MDS (lower-risk and higher-risk), frontline treatment of AML, treatment of special populations, treatment of R/R AML, and novel approaches. CONCLUSIONS We identified many areas of ongoing controversy in the diagnosis and management of MDS and AML related to classification and risk assessment, treatment selection, sequencing of therapies, and monitoring of responses. Many clinical trials are ongoing to further improve outcomes for patients with MDS and AML, and we noted potential areas of debate related to study design, selection of endpoints, and assessment of responses. The controversies and gaps in knowledge identified by this panel will inform a follow-up conference in 2025 that will employ a modified Delphi method with a goal of developing and publishing formal consensus recommendations that can provide actionable guidance to clinicians in practice.
Collapse
Affiliation(s)
- Hetty E Carraway
- Division of Hematologic Oncology and Blood Disorders, Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH; Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH.
| | - Andrew M Brunner
- Leukemia Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Catherine E Lai
- Leukemia Clinical Research Unit, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA
| | | | - Jae Park
- Cellular Therapy Service, Deparment of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander E Perl
- Leukemia Clinical Research Unit, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA
| | - Eytan M Stein
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eunice S Wang
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Amer M Zeidan
- Department of Internal Medicine, Yale University, New Haven, CT
| | - Joshua F Zeidner
- Division of Hematology, Department of Medicine, University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Rami Komrokji
- Malignant Hematology Department, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
17
|
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
|
18
|
Terra R, Éthier V, Busque L, Morin-Quintal A, D'Angelo G, Hébert J, Wang X, Lépine G, LeBlanc R, Bergeron J. Improved identification of clinically relevant Acute Leukemia subtypes using standardized EuroFlow panels versus non-standardized approach. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2025; 108:116-127. [PMID: 39538364 DOI: 10.1002/cyto.b.22213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/23/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Rare acute leukemia (AL) components or subtypes such as blastic plasmacytoid dendritic cell neoplasm (BPDCN) or early T-cell precursor acute Lymphoblastic Leukemia (ETP-ALL) can be difficult to detect by routine flow cytometry due to their immunophenotypes overlapping with other poorly differentiated AL. We hypothesized that using standardized EuroFlow™ Consortium approach could better diagnose such entities among cases that previously classified as acute myeloid leukemia (AML)-M0, AML with minimal differentiation, AML with myelodysplasia-related changes without further lineage differentiation, and AL of ambiguous lineage. In order to confirm this hypothesis and assess whether these AL subtypes such as BPDCN and ETP-ALL had previously gone undetected, we reanalyzed 49 banked cryopreserved sample cases using standardized EuroFlow™ Consortium panels. We also performed target sequencing to capture the mutational commonalities between these AL subtypes. Reanalysis led to revised or refined diagnoses for 23 cases (47%). Of these, five diagnoses were modified, uncovering 3 ETP-ALL and 2 typical BPDCN cases. In 12 AML cases, a variable proportion of immature plasmacytoid dendritic cell and/or monocytic component was newly identified. In one AML case, we have identified a megakaryoblastic differentiation. Finally, in five acute lymphoblastic leukemia (ALL) cases, we were able to more precisely determine the maturation stage. The application of standardized EuroFlow flow cytometry immunophenotyping improves the diagnostic accuracy of ALs and could impact treatment decisions.
Collapse
Affiliation(s)
- Rafik Terra
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Hôpital Maisonneuve-Rosemont, Montréal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Vincent Éthier
- Division of Hematology and Oncology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
- Département de médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Lambert Busque
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Hôpital Maisonneuve-Rosemont, Montréal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
- Hematology-Oncology and Cell Therapy University Institute, Hôpital Maisonneuve-Rosemont Research Center, Université de Montréal, Montreal, Quebec, Canada
| | - Ariane Morin-Quintal
- Département d'hémato-oncologie, Hôpital du Sacré-Coeur-de, Montréal, Quebec, Canada
| | - Giovanni D'Angelo
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Hôpital Maisonneuve-Rosemont, Montréal, Quebec, Canada
| | - Josée Hébert
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Hôpital Maisonneuve-Rosemont, Montréal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
- Hematology-Oncology and Cell Therapy University Institute, Hôpital Maisonneuve-Rosemont Research Center, Université de Montréal, Montreal, Quebec, Canada
| | - Xuehai Wang
- Department of Pathology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Guylaine Lépine
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Hôpital Maisonneuve-Rosemont, Montréal, Quebec, Canada
| | - Richard LeBlanc
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Hôpital Maisonneuve-Rosemont, Montréal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Julie Bergeron
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Hôpital Maisonneuve-Rosemont, Montréal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
- Hematology-Oncology and Cell Therapy University Institute, Hôpital Maisonneuve-Rosemont Research Center, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
19
|
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
|
20
|
Selheim F, Aasebø E, Reikvam H, Bruserud Ø, Hernandez-Valladares M. Proteomic Comparison of Acute Myeloid Leukemia Cells and Normal CD34 + Bone Marrow Cells: Studies of Leukemia Cell Differentiation and Regulation of Iron Metabolism/Ferroptosis. Proteomes 2025; 13:11. [PMID: 39982321 PMCID: PMC11843884 DOI: 10.3390/proteomes13010011] [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: 01/02/2025] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive bone marrow malignancy that can be cured only by intensive chemotherapy possibly combined with allogeneic stem cell transplantation. We compared the pretreatment proteomic profiles of AML cells derived from 50 patients at the time of first diagnosis with normal CD34+ bone marrow cells. A comparison based on all AML and CD34+ normal cell populations identified 121 differentially abundant proteins that showed at least 2-fold differences, and these proteins included several markers of neutrophil differentiation (e.g., TLR2, the integrins ITGM and ITGX, and downstream mediators including RHO GTPase, S100A8, S100A9, S100A22). However, the expression of these 121 proteins varied between patients, and a subset of 28 patients was characterized by increased long-term AML-free survival, signs of myeloid AML cell differentiation, and favorable genetic abnormalities. These two main patient subsets (28 with differentiation versus 22 with fewer signs of differentiation) also differed with regard to the phosphorylation of 16 differentially abundant proteins. Furthermore, we also classified our patients based on their expression of 16 proteins involved in the regulation of iron metabolism/ferroptosis and showing differential expression when comparing AML cells and normal CD34+ cells. Among the 22 patients with less favorable prognosis, we could then identify a genetically heterogeneous subset characterized by adverse prognosis (i.e., death from primary resistance/relapse) and an iron metabolism/ferroptosis protein profile showing similarities with normal CD34+ cells. We conclude that proteomic profiles differ between AML and normal CD34+ cells; especially, proteomic differences reflecting differentiation and regulation of iron metabolism/ferroptosis are associated with risk of relapse after intensive conventional therapy.
Collapse
Affiliation(s)
- Frode Selheim
- Proteomics Unit of University of Bergen (PROBE), University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; (F.S.); (M.H.-V.)
| | - Elise Aasebø
- Acute Leukemia Research Group, Department of Clinical Science, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; (E.A.); (H.R.)
| | - Håkon Reikvam
- Acute Leukemia Research Group, Department of Clinical Science, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; (E.A.); (H.R.)
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5009 Bergen, Norway
| | - Øystein Bruserud
- Acute Leukemia Research Group, Department of Clinical Science, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; (E.A.); (H.R.)
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5009 Bergen, Norway
| | - Maria Hernandez-Valladares
- Proteomics Unit of University of Bergen (PROBE), University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; (F.S.); (M.H.-V.)
- Department of Physical Chemistry, University of Granada, Avenida de la Fuente Nueva S/N, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| |
Collapse
|
21
|
Fonseca R, Zhu YX, Bruins LA, Ahmann J, de Bonolo Campos C, Braggio E, Chen X, Arribas M, Darvish S, Welsh S, Meermeier E, Mangalaparthi KK, Kandasamy RK, Ahmann G, Wiedmeier-Nutor JE, Pandey A, Chesi M, Bergsagel PL, Fonseca R. Exploring BCL2 regulation and upstream signaling transduction in venetoclax resistance in multiple myeloma: potential avenues for therapeutic intervention. Blood Cancer J 2025; 15:10. [PMID: 39910038 PMCID: PMC11799149 DOI: 10.1038/s41408-025-01215-x] [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: 08/23/2024] [Revised: 12/21/2024] [Accepted: 01/21/2025] [Indexed: 02/07/2025] Open
Abstract
Investigating venetoclax (VTX) resistance in multiple myeloma (MM) is crucial for the development of novel therapeutic strategies to tackle resistance. We conducted a multi-omic characterization of established VTX-resistant isogenic human myeloma cell lines (HMCL) and primary MM patient samples pre- and post-VTX treatment. Transcriptomic and proteomic analysis revealed that resistance was largely associated with BCL-2 family protein dysregulation, including upregulation of anti-apoptotic proteins such as MCL-1, BCL-XL, BCL-2, and downregulation of pro-apoptotic members. Notably, the re-introduction of BIM into resistant cells restored VTX sensitivity and synergized with MCL-1 inhibitors. Upstream signaling pathways, including growth factor receptor tyrosine kinase (RTK) and phosphoinositide-3-kinase (PI3K) were implicated in this dysregulation. Simultaneous inhibition of MCL-1, BCL-XL, and upstream PI3K, RTK (FGF, EGF, and IGF1) mediated signaling enhanced VTX sensitivity. Post-translational modifications of MCL-1, particularly its stabilization via acetylation and phosphorylation, were investigated, although their inhibition only marginally increased VTX sensitivity. Lastly, the inhibition of AURKA and mitochondrial respiration also improved VTX sensitivity in some resistant HMCLs. Our findings suggest that combining VTX with MCL-1 and BCL-XL inhibitors or PIK3/RTK inhibitors holds potential for overcoming resistance. The study illustrates the importance of understanding molecular determinants of resistance to develop tailored therapeutic strategies.
Collapse
Affiliation(s)
| | - Yuan Xiao Zhu
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - Laura A Bruins
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - Joseph Ahmann
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | | | - Esteban Braggio
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - Xianfeng Chen
- Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Mariano Arribas
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - Susie Darvish
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - Seth Welsh
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - Erin Meermeier
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | | | - Richard K Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Greg Ahmann
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | | | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Marta Chesi
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - P Leif Bergsagel
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA
| | - Rafael Fonseca
- Division of Hematology and Medical Oncology, Mayo Clinic, AZ, USA.
| |
Collapse
|
22
|
Xu Y, Li D, Wang N, Ge B, Meng C, Zhao M, Lin Z, Li M, Yuan Y, Cai Y, Shi L, Gao S, Ye H. miR-182 promoter hypermethylation predicts the better outcome of AML patients treated with AZA + VEN in a real-world setting. Clin Epigenetics 2025; 17:18. [PMID: 39910681 PMCID: PMC11800541 DOI: 10.1186/s13148-025-01823-1] [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: 11/01/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND 5-Azacytidine (AZA) combined with the BCL2 inhibitor Venetoclax (VEN) is the standard treatment for elderly acute myeloid leukemia (AML) patients or those who are unfit for intensive chemotherapy (elderly or unfit AML). However, an effective and rapid predictive biomarker to predict treatment outcome remains elusive. METHODS miR-182 promoter methylation was measured in 94 AZA + VEN-treated elderly or unfit AML patients and 20 normal controls (NCs) samples. To determine whether miR-182 promoter methylation is a predictive marker of clinical outcomes in AZA + VEN-treated AML patients in a real-world setting, we analyzed and compared the complete remission (CR)/CR with incomplete hematologic recovery (CRi) rate, overall survival (OS), and leukemia free-survival (LFS) across different methylation groups: miR-182 promoter hypomethylation (median value < 20.21%) and hypermethylation (> 20.21%) in a retrospective study. RESULTS The average methylation frequency was markedly higher in 94 AZA + VEN-treated elderly or unfit AML patients than that in 20 NCs. However, some AML patients (11.7%) still presented low miR-182 promoter methylation (< 10%). The average time to obtain CR/CRi was shorter in AML patients with miR-182 promoter hypermethylation than AML with hypomethylation. Moreover, the median OS and LFS were longer in AML patients with miR-182 promoter hypermethylation than AML with hypomethylation. Finally, the area under the curve (AUC) for 1-year mortality was 0.831, for 2-year was 0.788, and for 3-year was 0.800. CONCLUSIONS AML patients with miR-182 promoter hypermethylation have better outcomes. miR-182 promoter methylation is a predictive biomarker for AZA + VEN-treated AML patients.
Collapse
Affiliation(s)
- Yilan Xu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Danyang Li
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
- Department of Blood Transfusion, The First Affiliated Hospital of Ningbo University, Ningbo, 315099, Zhejiang Province, China
| | - Na Wang
- Health Care Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, Zhejiang Province, China
| | - Bei Ge
- Infection Control Department, Eye Hospital, Wenzhou Medical University, 270 Xueyuanxi Road, Wenzhou, Zhejiang Province, China
| | - Chen Meng
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Min Zhao
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Zihan Lin
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Min Li
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Yigang Yuan
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Yue Cai
- Department of Clinical Medicine, Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang Province, China
| | - Liuzhi Shi
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Shenmeng Gao
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China.
| | - Haige Ye
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, 1 Xuefubei Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
23
|
Falini B, Sorcini D, Perriello VM, Sportoletti P. Functions of the native NPM1 protein and its leukemic mutant. Leukemia 2025; 39:276-290. [PMID: 39690184 DOI: 10.1038/s41375-024-02476-4] [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: 09/10/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
The nucleophosmin (NPM1) gene encodes for the most abundant nucleolar protein. Thanks to its property to act as histone chaperone and to shuttle between the nucleus and cytoplasm, the NPM1 protein is involved in multiple cellular function that are here extensively reviewed and include the formation of the nucleolus through liquid-liquid phase separation, regulation of ribosome biogenesis and transport, control of DNA repair and centrosome duplication as well as response to nucleolar stress. NPM1 is mutated in about 30-35% of adult acute myeloid leukemia (AML). Due to its unique biological and clinical features, NPM1-mutated AML is regarded as a distinct leukemia entity in the WHO 5th edition and ICC classifications of myeloid malignancies. The NPM1 mutant undergoes changes at the C-terminus of the protein that leads to its delocalization in the cytoplasm of the leukemic cells. Here, we focus also on its biological functions discussing the murine models of NPM1 mutations and the various mechanisms that occur at cytoplasmic and nuclear levels to promote and maintain NPM1-mutated AML.
Collapse
Affiliation(s)
- Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy.
| | - Daniele Sorcini
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Vincenzo Maria Perriello
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Paolo Sportoletti
- Institute of Hematology and Center for Hemato-Oncological research (CREO), University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
24
|
Yabushita T, Goyama S. Nucleic acid metabolism: the key therapeutic target for myeloid tumors. Exp Hematol 2025; 142:104693. [PMID: 39647658 DOI: 10.1016/j.exphem.2024.104693] [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: 09/26/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
Nucleic acid analogs, including cytarabine, decitabine, and azacitidine, have significantly advanced therapeutic approaches for myeloid tumors over the past five decades. Nucleic acid metabolism is a crucial pathway driving myeloid tumorigenesis, with emerging evidence indicating that myeloid tumors are particularly dependent on the de novo nucleotide synthesis pathway, underscoring its potential as a therapeutic target. This review provides a comprehensive overview of nucleic acid metabolism, focusing on de novo nucleotide synthesis. We then described the range of clinically utilized agents targeting nucleic acid metabolism and discussed our recent findings on the nonepigenetic actions of decitabine, as well as the therapeutic effects of inosine monophosphate dehydrogenase (IMPDH) inhibitors in the treatment of myeloid tumors.
Collapse
Affiliation(s)
- Tomohiro Yabushita
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Susumu Goyama
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan.
| |
Collapse
|
25
|
Amaya ML. A novel therapy with a rational design for AML. Blood 2025; 145:457-458. [PMID: 39883443 DOI: 10.1182/blood.2024027063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
|
26
|
Kytölä S, Vänttinen I, Ruokoranta T, Partanen A, Holopainen A, Saad J, Kuusisto MEL, Koskela S, Räty R, Itälä-Remes M, Västrik I, Suvela M, Parsons A, Porkka K, Wennerberg K, Heckman CA, Jalkanen T, Huttunen T, Ettala P, Rimpiläinen J, Siitonen T, Pyörälä M, Kuusanmäki H, Kontro M. Ex vivo venetoclax sensitivity predicts clinical response in acute myeloid leukemia in the prospective VenEx trial. Blood 2025; 145:409-421. [PMID: 39357056 DOI: 10.1182/blood.2024024968] [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: 04/11/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 10/04/2024] Open
Abstract
ABSTRACT The B-cell lymphoma 2 inhibitor venetoclax has shown promise for treating acute myeloid leukemia (AML). However, identifying patients likely to respond remains a challenge, especially for those with relapsed/refractory (R/R) disease. We evaluated the utility of ex vivo venetoclax sensitivity testing to predict treatment responses to venetoclax-azacitidine in a prospective, multicenter, phase 2 trial. The trial recruited 104 participants with previously untreated (n = 48), R/R (n = 39), or previously treated secondary AML (sAML) (n = 17). The primary end point was complete remission or complete remission with incomplete hematologic recovery (CR/CRi) rate in ex vivo sensitive trial participants during the first 3 therapy cycles. The key secondary end points included the correlations between ex vivo drug sensitivity, responses, and survival. Venetoclax sensitivity was successfully assessed in 102 of 104 participants, with results available within a median of 3 days from sampling. In previously untreated AML, ex vivo sensitivity corresponded to an 85% (34/40) CR/CRi rate, with a median overall survival (OS) of 28.7 months, compared with 5.5 months for ex vivo resistant patients (P = .002). For R/R/sAML, ex vivo sensitivity resulted in a 62% CR/CRi rate (21/34) and median OS of 9.7 vs 3.3 months for ex vivo resistant patients (P < .001). In univariate and multivariate analysis, ex vivo venetoclax sensitivity was the strongest predictor for a favorable treatment response and survival. This trial demonstrates the feasibility of integrating ex vivo drug testing into clinical practice to identify patients with AML, particularly in the R/R setting, who benefit from venetoclax. This trial was registered at www.clinicaltrials.gov as #NCT04267081.
Collapse
MESH Headings
- Humans
- Sulfonamides/therapeutic use
- Sulfonamides/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Male
- Female
- Middle Aged
- Aged
- Adult
- Prospective Studies
- Aged, 80 and over
- Antineoplastic Agents/therapeutic use
- Treatment Outcome
- Remission Induction
- Drug Resistance, Neoplasm
Collapse
Affiliation(s)
- Sari Kytölä
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ida Vänttinen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tanja Ruokoranta
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Anu Partanen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | | | - Joseph Saad
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Sirpa Koskela
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Riikka Räty
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Maija Itälä-Remes
- Department of Clinical Hematology and Stem Cell Transplantations, Turku University Hospital, Turku, Finland
| | - Imre Västrik
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Minna Suvela
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Alun Parsons
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kimmo Porkka
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Krister Wennerberg
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | - Pia Ettala
- Department of Clinical Hematology and Stem Cell Transplantations, Turku University Hospital, Turku, Finland
| | - Johanna Rimpiläinen
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Timo Siitonen
- Department of Hematology, University of Oulu, Oulu, Finland
| | - Marja Pyörälä
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Mika Kontro
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| |
Collapse
|
27
|
Koolivand Z, Bahreini F, Rayzan E, Rezaei N. Inducing apoptosis in acute myeloid leukemia; mechanisms and limitations. Heliyon 2025; 11:e41355. [PMID: 39811307 PMCID: PMC11730532 DOI: 10.1016/j.heliyon.2024.e41355] [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: 05/13/2023] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Acute myeloid leukemia is the expansion of leukemic stem cells which might originate from a stem cell or a progenitor which has acquired self-renewal capacity. An aggregation of leukemic blasts in bone marrow, peripheral blood, and extramedullary tissue will result in acute myeloid leukemia. The main difficulty in treating acute myeloid leukemia is multidrug resistance, leading to treatment failure. This unfortunate phenomenon is practically elevated because of apoptosis inhibition in tumor cells. Two general apoptotic pathways are the Bcl-2 regulated pathway (the intrinsic pathway) and the death receptor pathway. Deficiencies in each of these apoptotic pathways can cause the usual resistance mechanism in this disease. This article reviews and highlights different antiapoptotic pathways, currently-used treatments, and new findings in this field, which may lead to the development of treatment methods for acute myeloid leukemia.
Collapse
Affiliation(s)
- Zahra Koolivand
- Faculty of Pharmacy and Pharmaceutical Sciences, Islamic Azad University, Tehran Medical Sciences University (IAUTMU), Tehran, Iran
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farbod Bahreini
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Elham Rayzan
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Brown FC, Wang X, Birkinshaw R, Chua CC, Morley T, Kasapgil S, Pomilio G, Blombery P, Huang DCS, Czabotar P, Priore SF, Yang G, Carroll M, Wei AH, Perl AE. Acquired BCL2 variants associated with venetoclax resistance in acute myeloid leukemia. Blood Adv 2025; 9:127-131. [PMID: 39374584 PMCID: PMC11742564 DOI: 10.1182/bloodadvances.2024014446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/29/2024] [Indexed: 10/09/2024] Open
Affiliation(s)
- Fiona C. Brown
- Division of Blood Cells and Blood Cancer, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Xin Wang
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Richard Birkinshaw
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Division of Structural Biology, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Chong Chyn Chua
- Division of Blood Cells and Blood Cancer, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Thomas Morley
- Division of Blood Cells and Blood Cancer, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Sila Kasapgil
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Division of Structural Biology, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Giovanna Pomilio
- Division of Blood Cells and Blood Cancer, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Piers Blombery
- Department of Pathology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Sir Peter Maccallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Haematology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - David C. S. Huang
- Division of Blood Cells and Blood Cancer, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Peter Czabotar
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Division of Structural Biology, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Salvatore F. Priore
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Guang Yang
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Martin Carroll
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
- Leukemia Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Andrew H. Wei
- Division of Blood Cells and Blood Cancer, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Hematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Alexander E. Perl
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
- Leukemia Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
29
|
Goulart H, Kantarjian H, Pemmaraju N, Daver N, DiNardo CD, Rausch CR, Ravandi F, Kadia TM. Venetoclax-Based Combination Regimens in Acute Myeloid Leukemia. Blood Cancer Discov 2025; 6:23-37. [PMID: 39565177 PMCID: PMC11707511 DOI: 10.1158/2643-3230.bcd-24-0171] [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: 07/05/2024] [Revised: 08/22/2024] [Accepted: 09/30/2024] [Indexed: 11/21/2024] Open
Abstract
SIGNIFICANCE In recent years, there has been tremendous interest surrounding the integration of venetoclax into both non-intensive and intensive chemotherapy regimens for AML. However, with this increasing utilization of venetoclax, considerable questions surrounding key issues such as dosing strategies and the practicality of venetoclax administration have arisen. This review highlights the evolution of venetoclax-based regimens in AML and provides a commentary on notable practical considerations when utilizing this agent.
Collapse
Affiliation(s)
- Hannah Goulart
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naveen Pemmaraju
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney D. DiNardo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caitlin R. Rausch
- Division of Pharmacy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tapan M. Kadia
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
30
|
Kawakatsu R, Tadagaki K, Yamasaki K, Kuwahara Y, Yoshida T. Valproic Acid Enhances Venetoclax Efficacy in Targeting Acute Myeloid Leukemia. Diseases 2025; 13:10. [PMID: 39851474 PMCID: PMC11764158 DOI: 10.3390/diseases13010010] [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: 11/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a common and aggressive form of leukemia, yet current treatment strategies remain insufficient. Venetoclax, a BH3-mimetic approved for AML treatment, induces Bcl-2-dependent apoptosis, though its therapeutic efficacy is still limited. Therefore, new strategies to enhance the effect of venetoclax are highly sought. Valproic acid (VPA), commonly used for epilepsy, has also been studied for potential applications in AML treatment. METHODS AML cells were treated with venetoclax, with or without VPA. Cell viability was assessed using the trypan blue dye exclusion assay, while cell cycle progression was analyzed by flow cytometry. The expression of pro-apoptotic proteins Bax and Bak was measured by RT-qPCR. RESULTS Venetoclax and VPA individually had only mild effects on AML cell proliferation. However, their combination significantly inhibited cell growth and triggered pronounced cell death. This combination also led to the cleavage of poly (ADP-ribose) polymerase (PARP), a substrate of caspases, indicating activation of apoptosis. VPA treatment upregulated the expression of Bax and Bak, further supporting apoptosis induction. The cell death induced by the venetoclax-VPA combination was predominantly apoptotic, as confirmed by the near-complete blockade of cell death by a pan-caspase inhibitor. CONCLUSIONS Our study demonstrates that VPA enhances venetoclax-induced apoptosis in AML cell lines, providing a novel role for VPA and suggesting a promising combinatory strategy for AML treatment. These findings offer valuable insights into potential clinical applications of venetoclax and VPA in AML management.
Collapse
Affiliation(s)
- Renshi Kawakatsu
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kenjiro Tadagaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kenta Yamasaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yasumichi Kuwahara
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Kyoto Pediatric Community-Based Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Tatsushi Yoshida
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
31
|
Niscola P, Gianfelici V, Giovannini M, Piccioni D, Mazzone C, de Fabritiis P. Menin Inhibitors: New Targeted Therapies for Specific Genetic Subtypes of Difficult-to-Treat Acute Leukemias. Cancers (Basel) 2025; 17:142. [PMID: 39796769 PMCID: PMC11720583 DOI: 10.3390/cancers17010142] [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: 11/30/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Menin (MEN1) is a well-recognized powerful tumor promoter in acute leukemias (AL) with KMT2A rearrangements (KMT2Ar, also known as MLL) and mutant nucleophosmin 1 (NPM1m) acute myeloid leukemia (AML). MEN1 is essential for sustaining leukemic transformation due to its interaction with wild-type KMT2A and KMT2A fusion proteins, leading to the dysregulation of KMT2A target genes. MEN1 inhibitors (MIs), such as revumenib, ziftomenib, and other active small molecules, represent a promising new class of therapies currently under clinical development. By disrupting the MEN1-KMT2Ar complex, a group of proteins involved in chromatin remodeling, MIs induce apoptosis and differentiation AL expressing KMT2Ar or NPM1m AML. Phase I and II clinical trials have evaluated MIs as standalone treatments and combined them with other synergistic drugs, yielding promising results. These trials have demonstrated notable response rates with manageable toxicities. Among MIs, ziftomenib received orphan drug and breakthrough therapy designations from the European Medicines Agency in January 2024 and the Food and Drug Administration (FDA) in April 2024, respectively, for treating R/R patients with NPM1m AML. Additionally, in November 2024, the FDA approved revumenib for treating R/R patients with KMT2Ar-AL. This review focuses on the pathophysiology of MI-sensitive AL, primarily AML. It illustrates data from clinical trials and discusses the emergence of resistance mechanisms. In addition, we outline future directions for the use of MIs and emphasize the need for further research to fully realize the potential of these novel compounds, especially in the context of specific genetic subtypes of challenging AL.
Collapse
Affiliation(s)
- Pasquale Niscola
- Hematology Unit, S. Eugenio Hospital (ASL Roma 2), 00122 Rome, Italy; (V.G.); (M.G.); (D.P.); (C.M.); (P.d.F.)
| | | | | | | | | | | |
Collapse
|
32
|
Minhajuddin M, Winters A, Ye H, Pei S, Stevens B, Gillen A, Engel K, Gipson S, Ransom M, Amaya M, Inguva A, Gasparetto M, Althoff MJ, Miller R, Shelton I, Tolison H, Krug A, Culp-Hill R, D'Alessandro A, Sherbenou DW, Pollyea DA, Smith C, Jordan CT. Lysosomal acid lipase A modulates leukemia stem cell response to venetoclax/tyrosine kinase inhibitor combination therapy in blast phase chronic myeloid leukemia. Haematologica 2025; 110:103-116. [PMID: 38934082 PMCID: PMC11694110 DOI: 10.3324/haematol.2023.284716] [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: 11/22/2023] [Accepted: 06/20/2024] [Indexed: 06/28/2024] Open
Abstract
The treatment of blast phase chronic myeloid leukemia (bpCML) remains a challenge due, at least in part, to drug resistance of leukemia stem cells (LSC). Recent clinical evidence suggests that the BCL-2 inhibitor venetoclax in combination with ABL-targeting tyrosine kinase inhibitors can eradicate bpCML LSC. In this study, we employed preclinical models of bpCML to investigate the efficacy and underlying mechanism of LSC-targeting with combinations of venetoclax/tyrosine kinase inhibitors. Transcriptional analysis of LSC exposed to venetoclax and dasatinib revealed upregulation of genes involved in lysosomal biology, in particular lysosomal acid lipase A (LIPA), a regulator of free fatty acids. Metabolomic analysis confirmed increased levels of free fatty acids in response to treatment with venetoclax/dasatinib. Pretreatment of leukemia cells with bafilomycin, a specific lysosome inhibitor, or genetic perturbation of LIPA, resulted in increased sensitivity of leukemia cells to venetoclax/dasatinib, implicating LIPA in treatment resistance. Importantly, venetoclax/dasatinib treatment did not affect normal stem cell function, suggesting a leukemia-specific response. These results demonstrate that venetoclax/dasatinib is a LSC-selective regimen in bpCML and that disrupting LIPA and fatty acid transport enhances the response to venetoclax/ dasatinib when targeting LSC, providing a rationale for exploring lysosomal disruption as an adjunctive therapeutic strategy to prolong disease remission.
Collapse
MESH Headings
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Humans
- Sulfonamides/pharmacology
- Sulfonamides/administration & dosage
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/administration & dosage
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Mice
- Blast Crisis/drug therapy
- Blast Crisis/pathology
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Tyrosine Kinase Inhibitors
Collapse
Affiliation(s)
- Mohd Minhajuddin
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO.
| | - Amanda Winters
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Haobin Ye
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghaii
| | - Shanshan Pei
- Liangzhu Laboratory, Zhejiang University Medical Center, Zhejiang
| | - Brett Stevens
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Austin Gillen
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Krysta Engel
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Stephanie Gipson
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Monica Ransom
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Maria Amaya
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Anagha Inguva
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Maura Gasparetto
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Mark J Althoff
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Regan Miller
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Ian Shelton
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Hunter Tolison
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Anna Krug
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Daniel W Sherbenou
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Daniel A Pollyea
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Clayton Smith
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
33
|
Cierpicki T, Grembecka J. Targeting Protein-Protein Interactions in Hematologic Malignancies. ANNUAL REVIEW OF PATHOLOGY 2025; 20:275-301. [PMID: 39854187 DOI: 10.1146/annurev-pathmechdis-031521-033231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Over the last two decades, there have been extensive efforts to develop small-molecule inhibitors of protein-protein interactions (PPIs) as novel therapeutics for cancer, including hematologic malignancies. Despite the numerous challenges associated with developing PPI inhibitors, a significant number of them have advanced to clinical studies in hematologic patients in recent years. The US Food and Drug Administration approval of the very first PPI inhibitor, venetoclax, demonstrated the real clinical value of blocking protein-protein interfaces. In this review, we discuss the most successful examples of PPI inhibitors that have reached clinical studies in patients with hematologic malignancies. We also describe the challenges of blocking PPIs with small molecules, clinical resistance to such compounds, and the lessons learned from the development of successful PPI inhibitors. Overall, this review highlights the remarkable success and substantial promise of blocking PPIs in hematologic malignancies.
Collapse
Affiliation(s)
- Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA; ,
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA; ,
| |
Collapse
|
34
|
Yu M, Nah GSS, Krishnan V, Sulaimi FNB, Ng KP, Wang C, Bhatt S, Chuah C, Bergstrom DE, Ong ST. The BIM deletion polymorphism potentiates the survival of leukemia stem and progenitor cells and impairs response to targeted therapies. Leukemia 2025; 39:134-143. [PMID: 39438588 DOI: 10.1038/s41375-024-02418-0] [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: 11/27/2023] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
One sixth of human cancers harbor pathogenic germline variants, but few studies have established their functional contribution to cancer outcomes. Here, we developed a humanized mouse model harboring a common East Asian polymorphism, the BIM deletion polymorphism (BDP), which confers resistance to oncogenic kinase inhibitors through generation of non-apoptotic splice isoforms. However, despite its clear role in mediating bulk resistance in patients, the BDP's role in cancer stem and progenitor cells, which initiate disease and possess altered BCL-2 rheostats compared to differentiated tumor cells, remains unknown. To study the role of the BDP in leukemia initiation, we crossed the BDP mouse into a chronic myeloid leukemia (CML) model. We found that the BDP greatly enhanced the fitness of CML cells with a three-fold greater competitive advantage, leading to more aggressive disease. The BDP conferred almost complete resistance to cell death induced by imatinib in CML stem and progenitor cells (LSPCs). Using BH3 profiling, we identified a novel therapeutic vulnerability of BDP LSPCs to MCL-1 antagonists, which we confirmed in primary human LSPCs, and in vivo. Our findings demonstrate the impact of human polymorphisms on the survival of LSPCs and highlight their potential as companion diagnostics for tailored therapies.
Collapse
MESH Headings
- Animals
- Bcl-2-Like Protein 11/genetics
- Mice
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Polymorphism, Genetic
- Drug Resistance, Neoplasm/genetics
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Apoptosis
- Cell Survival/genetics
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Molecular Targeted Therapy
Collapse
Affiliation(s)
- Mengge Yu
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Giselle Sek Suan Nah
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Vaidehi Krishnan
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | | | - King Pan Ng
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
- KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Chuqi Wang
- Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Charles Chuah
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore
| | | | - S Tiong Ong
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore.
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, 27708, USA.
| |
Collapse
|
35
|
Tremblay D, Csizmar C, DiNardo CD, Ball S, Rippel N, Hammond D, Kadia TM, Ravandi F, Chien K, Van Hyfte G, Mazumdar M, Saliba A, Mangaonkar A, Lasho T, Al-Kali A, Kremyanskaya M, Feld J, Silverman LR, Komrokji R, Mascarenhas J, Padron E, Garcia-Manero G, Sallman DA, Patnaik MM, Montalban-Bravo G. Venetoclax in combination with hypomethylating agents in chronic myelomonocytic leukemia: a propensity score matched multicenter cohort study. Leukemia 2025; 39:257-260. [PMID: 39533069 DOI: 10.1038/s41375-024-02466-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | | | | | - Somedeb Ball
- Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Noa Rippel
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Grace Van Hyfte
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhu Mazumdar
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - Marina Kremyanskaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Feld
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lewis R Silverman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
36
|
Sango J, Carcamo S, Sirenko M, Maiti A, Mansour H, Ulukaya G, Tomalin LE, Cruz-Rodriguez N, Wang T, Olszewska M, Olivier E, Jaud M, Nadorp B, Kroger B, Hu F, Silverman L, Chung SS, Wagenblast E, Chaligne R, Eisfeld AK, Demircioglu D, Landau DA, Lito P, Papaemmanuil E, DiNardo CD, Hasson D, Konopleva M, Papapetrou EP. RAS-mutant leukaemia stem cells drive clinical resistance to venetoclax. Nature 2024; 636:241-250. [PMID: 39478230 PMCID: PMC11618090 DOI: 10.1038/s41586-024-08137-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/30/2024] [Indexed: 12/06/2024]
Abstract
Cancer driver mutations often show distinct temporal acquisition patterns, but the biological basis for this, if any, remains unknown. RAS mutations occur invariably late in the course of acute myeloid leukaemia, upon progression or relapsed/refractory disease1-6. Here, by using human leukaemogenesis models, we first show that RAS mutations are obligatory late events that need to succeed earlier cooperating mutations. We provide the mechanistic explanation for this in a requirement for mutant RAS to specifically transform committed progenitors of the myelomonocytic lineage (granulocyte-monocyte progenitors) harbouring previously acquired driver mutations, showing that advanced leukaemic clones can originate from a different cell type in the haematopoietic hierarchy than ancestral clones. Furthermore, we demonstrate that RAS-mutant leukaemia stem cells (LSCs) give rise to monocytic disease, as observed frequently in patients with poor responses to treatment with the BCL2 inhibitor venetoclax. We show that this is because RAS-mutant LSCs, in contrast to RAS-wild-type LSCs, have altered BCL2 family gene expression and are resistant to venetoclax, driving clinical resistance and relapse with monocytic features. Our findings demonstrate that a specific genetic driver shapes the non-genetic cellular hierarchy of acute myeloid leukaemia by imposing a specific LSC target cell restriction and critically affects therapeutic outcomes in patients.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Cell Lineage/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Monocytes/metabolism
- Monocytes/drug effects
- Mutation
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- ras Proteins/metabolism
- ras Proteins/genetics
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Granulocytes
- Clone Cells/metabolism
- Clone Cells/pathology
- Stem Cells/metabolism
- Stem Cells/pathology
- Recurrence
Collapse
Affiliation(s)
- Junya Sango
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saul Carcamo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Sirenko
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hager Mansour
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gulay Ulukaya
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lewis E Tomalin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nataly Cruz-Rodriguez
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tiansu Wang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Malgorzata Olszewska
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emmanuel Olivier
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manon Jaud
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bettina Nadorp
- Department of Medicine, Division of Precision Medicine, NYU Grossman School of Medicine, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Benjamin Kroger
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Feng Hu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lewis Silverman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen S Chung
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elvin Wagenblast
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronan Chaligne
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ann-Kathrin Eisfeld
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Deniz Demircioglu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan A Landau
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Piro Lito
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Hasson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Konopleva
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
37
|
Chamberlin J, Gillen A, Quinlan A. Improved characterization of 3' single-cell RNA-seq libraries with paired-end avidity sequencing. NAR Genom Bioinform 2024; 6:lqae175. [PMID: 39703419 PMCID: PMC11655283 DOI: 10.1093/nargab/lqae175] [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: 08/21/2024] [Revised: 11/12/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
Prevailing poly(dT)-primed 3' single-cell RNA-seq protocols generate barcoded cDNA fragments containing the reverse transcriptase priming site or in principle the polyadenylation site. Direct sequencing across this site was historically difficult because of DNA sequencing errors induced by the homopolymeric primer at the 'barcode' end. Here, we evaluate the capability of 'avidity base chemistry' DNA sequencing from Element Biosciences to sequence through the primer and enable accurate paired-end read alignment and precise quantification of polyadenylation sites. We find that the Element Aviti instrument sequences through the thymine homopolymer into the subsequent cDNA sequence without detectable loss of accuracy. The additional sequence enables direct and independent assignment of reads to polyadenylation sites, which bypasses the complexities and limitations of conventional approaches but does not consistently improve read mapping rates compared to single-end alignment. We also characterize low-level artifacts and demonstrate necessary adjustments to adapter trimming and sequence alignment regardless of platform, particularly in the context of extended read lengths. Our analyses confirm that Element avidity sequencing is an effective alternative to Illumina sequencing for standard single-cell RNA-seq, particularly for polyadenylation site measurement but do not rule out the potential for similar performance from other emerging platforms.
Collapse
Affiliation(s)
- John T Chamberlin
- Department of Biomedical Informatics, University of Utah School of Medicine, 421 Wakara Way #140, Salt Lake City, UT 84112, USA
| | - Austin E Gillen
- RNA Bioscience Initiative, University of Colorado School of Medicine, 12801 E 17th Ave, Aurora, CO 80045, USA
- Division of Hematology, University of Colorado School of Medicine, 12700 East 19th Ave, Aurora, CO 80045, USA
- Rocky Mountain Regional VA Medical Center, 1700 N Wheeling St, Aurora, CO 80045, USA
| | - Aaron R Quinlan
- Department of Biomedical Informatics, University of Utah School of Medicine, 421 Wakara Way #140, Salt Lake City, UT 84112, USA
- Department of Human Genetics, University of Utah School of Medicine, 15 N 2030 E, Salt Lake City, UT 84112, USA
| |
Collapse
|
38
|
Tatarata QZ, Wang Z, Konopleva M. BCL-2 inhibition in acute myeloid leukemia: resistance and combinations. Expert Rev Hematol 2024; 17:935-946. [PMID: 39552410 DOI: 10.1080/17474086.2024.2429604] [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: 06/19/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION The introduction of venetoclax has revolutionized the treatment landscape of acute myeloid leukemia, offering new therapeutic opportunities. However, the clinical response to venetoclax varies significantly between patients, with many experiencing limited duration of response. AREAS COVERED Identified resistance mechanisms include both intrinsic and acquired resistance to VEN. The former is associated with cell lineage and differentiation state. The latter includes dependency on alternative BCL-2 family anti-apoptotic protein(s) mediated by genetic, epigenetic, or post-translational mechanisms, mitochondrial and metabolic involvement, as well as microenvironment. Understanding these mechanisms is crucial for optimizing venetoclax-based therapies and enhancing treatment outcomes for patients with acute myeloid leukemia. This review aims to elucidate the primary mechanisms underlying resistance to venetoclax and explore current therapeutic strategies to overcome this challenge. EXPERT OPINION In patients with venetoclax resistance, alternative options include targeted combination therapies tailored to individual cases based on cytogenetics and prior treatments. Many of these therapies require further clinical investigation to validate their safety and efficacy.
Collapse
Affiliation(s)
- Qi Zhang Tatarata
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- The Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Zhe Wang
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
39
|
Patel SB, Moskop DR, Jordan CT, Pietras EM. Understanding MDS stem cells: Advances and limitations. Semin Hematol 2024; 61:409-419. [PMID: 39472255 DOI: 10.1053/j.seminhematol.2024.09.007] [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: 06/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
In work spanning several decades, extensive studies have focused on the properties of malignant stem cells that drive the pathogenesis of acute myeloid leukemia (AML). However, relatively little attention has been devoted to several serious myeloid malignancies that occur prior to the onset of frank leukemia, including myelodysplastic syndrome (MDS). Like leukemia, MDS is hypothesized to arise from a pool of immature malignant stem and progenitor cells (MDS-SCs) that serve as a reservoir for disease evolution and progression1. While multiple studies have sought to identify and characterize the biology and vulnerabilities of MDS-SCs, yet translation of scientific concepts to therapeutically impactful regimens has been limited. Here, we evaluate the currently known properties of MDS-SCs as well as the post-transcriptional mechanisms that drive MDS pathogenesis at a stem and progenitor level. We highlight limits and gaps in our characterization and understanding of MDS-SCs and address the extent to which the properties of MDS-SC are (and can be) inferred from the characterization of LSCs.
Collapse
Affiliation(s)
- Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Daniel R Moskop
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| |
Collapse
|
40
|
Cui J, Chen X, Li C, Yan Q, Yuan G. Reduced duration and dosage of venetoclax is efficient in newly diagnosed patients with acute myeloid leukemia. Hematology 2024; 29:2293512. [PMID: 38095287 DOI: 10.1080/16078454.2023.2293512] [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: 06/27/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
OBJECTIVES The combination of Venetoclax (VEN) and Azacitidine (AZA) increases survival outcomes and yields excellent responses in patients with acute myeloid leukemia (AML). However, dose reduction (or discontinuation) is commonly encountered due to therapy-related toxicity. Thus, this study aimed to investigate the efficiency and safety of a lower dosage of venetoclax for the treatment of AML. METHODS This observational study analyzed the characteristics and outcomes of newly diagnosed AML patients who received 100 mg VEN combined with AZA for 14 days at our institution. RESULTS A total of 36 patients were enrolled, and the median age at diagnosis was 64 years. After a median follow-up of 15 (range 4-29) months, the median overall survival (OS) and progression-free survival (PFS) for the whole cohort were 17 (4-29) months and 12 (1-28) months, respectively. Meanwhile, the overall response rate (ORR) was 69.4%, and the CRc rate was 66.7% in the whole cohort. Subgroup analysis revealed that NPM1 mutations and FAB-M5 subtype were associated with higher response rates, whereas the adverse ELN risk group was predictive of an inferior response. Moreover, ASXL1, NPM1, and IDH1/2 mutations negatively impacted PFS. DISCUSSION Our study optimized the administration of venetoclax plus azacytidine for the treatment of AML patients. Response rates were favorable, with median survival in agreement with the findings of earlier reports, offering valuable insights for optimizing VEN-based regimens. CONCLUSION In summary, the VEN combination regimen is effective for the treatment of newly diagnosed AML patients in the real world despite VEN dose reductions .
Collapse
Affiliation(s)
- Jingying Cui
- School of Medicine, Wuhan University of Science and Technology, Wuhan, People's Republic of China
- Department of Hematology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Xuexing Chen
- Department of Hematology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Chunfang Li
- Department of Hematology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Qiong Yan
- Department of Hematology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Guolin Yuan
- Department of Hematology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| |
Collapse
|
41
|
Zheng S, Tong Y, Yang L, Chen J, Tan Y. The prognostic significance of genetics in acute myeloid leukemia under venetoclax-based treatment. Ann Hematol 2024; 103:5019-5033. [PMID: 39467855 DOI: 10.1007/s00277-024-06050-x] [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: 05/27/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024]
Abstract
Acute myeloid leukemia (AML) is the most prevalent hematologic malignancy in adults. In 2022, the European LeukemiaNet (ELN) has updated its prognostic system that incorporates cytogenetics and molecular genetics based on data from patients undergoing intensive chemotherapy (IC). Recently, a risk stratification framework has been established for hypomethylating agents (HMA)-based low-intensity treatment (LIT) to fill the gaps in stratification for this treatment modality, but this needs further refinement. Venetoclax (VEN), a BH3 mimetic, targets BCL-2 to modulate apoptosis and metabolism in AML cells. Its combination with HMA or low-dose cytarabine (LDAC) has been shown to enhance the response rates and prolong the survival outcomes of older or unfit patients with AML. In this review, we delved into the prognostic significance of FLT3-ITD and IDH mutations when used in combination with VEN and HMA, as well as in conjunction with their specific inhibitors. We also explored the role of VEN in NPM1-mutated AML and its efficacy in splicing factor mutations AML. Additionally, we examined the response rates and survival outcomes of CBF-AML when treated with a VEN-based regimen. Moving forward, it is imperative that risk stratification for LIT becomes more nuanced to better align with the requirements of personalized diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Shan Zheng
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Department of Hematology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yuxin Tong
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Department of Hematology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Linlin Yang
- Department of Hematology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Jiayi Chen
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Department of Hematology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yamin Tan
- Department of Hematology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
- Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
42
|
Kashima E, Sugimoto Y, Nagaharu K, Ohya E, Ikejiri M, Watanabe Y, Kageyama S, Oka K, Tawara I. Venetoclax is effective for chronic myelomonocytic leukemia blastic transformation with RUNX1 mutation. Hematology 2024; 29:2392908. [PMID: 39163269 DOI: 10.1080/16078454.2024.2392908] [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: 01/15/2024] [Accepted: 08/10/2024] [Indexed: 08/22/2024] Open
Abstract
Background: Chronic myelomonocytic leukemia is a clonal hematological disorder with an inherent risk of transformation to acute myeloid leukemia. Recently, there has been exponential discovery of molecular abnormalities in patients with chronic myelomonocytic leukemia. Some of these mutations independently contribute to a higher risk of transformation and result in inferior overall survival. Treatment strategies for patients undergoing blastic transformation in chronic myelomonocytic leukemia, especially after progressing on hypomethylating agents, are currently limited.Case presentation: We present a case of a 70-year-old male patient with chronic myelomonocytic leukemia blastic transformation with RUNX1 mutation following azacitidine monotherapy. Notably, he achieved hematological complete remission after the first course of venetoclax plus azacitidine, leading to the disappearance of RUNX1 mutation. We performed serial assessments of molecular analysis by next generation sequencing throughout his clinical course.Conclusion: The presence of RUNX1 mutation is associated with higher response rates to venetoclax-based combination therapies in chronic myelomonocytic leukemia with blastic transformation. Our findings suggest that even after azacitidine monotherapy, venetoclax plus azacitidine is effective in targeting leukemic clones harboring RUNX1 mutations. Furthermore, we emphasize the significance of molecular analysis, including next-generation sequencing, in providing insights into the detailed dynamics of clonal evolution and guiding treatment decisions.
Collapse
Affiliation(s)
- Emiko Kashima
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yuka Sugimoto
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Keiki Nagaharu
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
- Department of Hematology, Yokkaichi Municipal Hospital, Yokkaichi, Japan
| | - Eiko Ohya
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
- Department of Hematology, Matsusaka Chuo General Hospital, Matsusaka, Japan
| | - Makoto Ikejiri
- Department of Clinical Laboratory, Mie University Hospital, Tsu, Japan
| | | | | | - Koji Oka
- Department of Hematology, Suzuka Kaisei Hospital, Suzuka, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
43
|
Bonilla G, Morris A, Kundu S, DuCasse A, Kirkpatrick G, Jeffries NE, Chetal K, Yvanovich EE, Milosevic J, Zhao T, Xia J, Barghout R, Scadden D, Mansour MK, Kingston RE, Sykes DB, Mercier FE, Sadreyev RI. Leukemia aggressiveness is driven by chromatin remodeling and expression changes of core regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582846. [PMID: 38496490 PMCID: PMC10942317 DOI: 10.1101/2024.02.29.582846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Molecular mechanisms driving clonal aggressiveness in leukemia are not fully understood. We tracked and analyzed MLL-rearranged leukemic clones independently evolving towards higher aggressiveness. More aggressive subclones lost their growth differential ex vivo but restored it upon secondary transplantation, suggesting molecular memory of aggressiveness. Development of aggressiveness was associated with clone-specific gradual modulation of chromatin states and expression levels across the genome, with a surprising preferential trend of reversing the earlier changes between normal and leukemic progenitors. To focus on the core aggressiveness program, we identified genes with consistent changes of expression and chromatin marks that were maintained in vivo and ex vivo in both clones. Overexpressing selected core genes (Smad1 as aggressiveness driver, Irx5 and Plag1 as suppressors) affected leukemic progenitor growth in the predicted way and had convergent downstream effects on central transcription factors and repressive epigenetic modifiers, suggesting a broader regulatory network of leukemic aggressiveness.
Collapse
|
44
|
Iyer P, Jasdanwala SS, Wang Y, Bhatia K, Bhatt S. Decoding Acute Myeloid Leukemia: A Clinician's Guide to Functional Profiling. Diagnostics (Basel) 2024; 14:2560. [PMID: 39594226 PMCID: PMC11593197 DOI: 10.3390/diagnostics14222560] [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: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is a complex clonal disorder characterized by clinical, genetic, metabolomic, and epigenetic heterogeneity resulting in the uncontrolled proliferation of aberrant blood-forming precursor cells. Despite advancements in the understanding of the genetic, metabolic, and epigenetic landscape of AML, it remains a significant therapeutic challenge. Functional profiling techniques, such as BH3 profiling (BP), gene expression profiling (GEP), proteomics, metabolomics, drug sensitivity/resistance testing (DSRT), CRISPR/Cas9, and RNAi screens offer valuable insights into the functional behavior of leukemia cells. BP evaluates the mitochondrial response to pro-apoptotic BH3 peptides, determining a cell's apoptotic threshold and its reliance on specific anti-apoptotic proteins. This knowledge can pinpoint vulnerabilities in the mitochondria-mediated apoptotic pathway in leukemia cells, potentially informing treatment strategies and predicting therapeutic responses. GEP, particularly RNA sequencing, evaluates the transcriptomic landscape and identifies gene expression alterations specific to AML subtypes. Proteomics and metabolomics, utilizing mass spectrometry and nuclear magnetic resonance (NMR), provide a detailed view of the active proteins and metabolic pathways in leukemia cells. DSRT involves exposing leukemia cells to a panel of chemotherapeutic and targeted agents to assess their sensitivity or resistance profiles and potentially guide personalized treatment strategies. CRISPR/Cas9 and RNAi screens enable systematic disruption of genes to ascertain their roles in leukemia cell survival and proliferation. These techniques facilitate precise disease subtyping, uncover novel biomarkers and therapeutic targets, and provide a deeper understanding of drug-resistance mechanisms. Recent studies utilizing functional profiling have identified specific mutations and gene signatures associated with aggressive AML subtypes, aberrant signaling pathways, and potential opportunities for drug repurposing. The integration of multi-omics approaches, advances in single-cell sequencing, and artificial intelligence is expected to refine the precision of functional profiling and ultimately improve patient outcomes in AML. This review highlights the diverse landscape of functional profiling methods and emphasizes their respective advantages and limitations. It highlights select successes in how these methods have further advanced our understanding of AML biology, identifies druggable targets that have improved outcomes, delineates challenges associated with these techniques, and provides a prospective view of the future where these techniques are likely to be increasingly incorporated into the routine care of patients with AML.
Collapse
Affiliation(s)
- Prasad Iyer
- Children’s Blood and Cancer Centre, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Shaista Shabbir Jasdanwala
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| | - Yuhan Wang
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| | - Karanpreet Bhatia
- Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore; (S.S.J.); (Y.W.); (S.B.)
| |
Collapse
|
45
|
Drucker M, Lee D, Zhang X, Kain B, Bowman M, Nicolet D, Wang Z, Stone RM, Mrózek K, Carroll AJ, Starczynowski DT, Levine RL, Byrd JC, Eisfeld AK, Salomonis N, Grimes HL, Bowman RL, Miles LA. Genotype-immunophenotype relationships in NPM1-mutant AML clonal evolution uncovered by single cell multiomic analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623033. [PMID: 39605444 PMCID: PMC11601460 DOI: 10.1101/2024.11.11.623033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Acute myeloid leukemia (AML) is a multi-clonal disease, existing as a milieu of clones with unique but related genotypes as initiating clones acquire subsequent mutations. However, bulk sequencing cannot fully capture AML clonal architecture or the clonal evolution that occurs as patients undergo therapy. To interrogate clonal evolution, we performed simultaneous single cell molecular profiling and immunophenotyping on 43 samples from 32 NPM1-mutant AML patients at different stages of disease. Here we show that diagnosis and relapsed AML samples display similar clonal architecture patterns, but signaling mutations can drive increased clonal diversity specifically at relapse. We uncovered unique genotype-immunophenotype relationships regardless of disease state, suggesting leukemic lineage trajectories can be hard-wired by the mutations present. Analysis of longitudinal samples from patients on therapy identified dynamic clonal, transcriptomic, and immunophenotypic changes. Our studies provide resolved understanding of leukemic clonal evolution and the relationships between genotype and cell state in leukemia biology.
Collapse
Affiliation(s)
- Morgan Drucker
- Division of Hematology/Oncology, Cancer & Blood Disease Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati OH USA
| | - Darren Lee
- University of Cincinnati College of Medicine, Cincinnati OH USA
| | - Xuan Zhang
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati OH USA
| | - Bailee Kain
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati OH USA
| | - Michael Bowman
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA USA
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, OH USA
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus OH USA
| | - Zhe Wang
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati OH USA
| | | | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, OH USA
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus OH USA
| | - Andrew J. Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL USA
| | - Daniel T. Starczynowski
- Division of Experimental Hematology & Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati OH USA
- Department of Pediatrics, University of Cincinnati, Cincinnati OH USA
- University of Cincinnati Cancer Center, Cincinnati OH USA
| | - Ross L. Levine
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John C. Byrd
- University of Cincinnati Cancer Center, Cincinnati OH USA
- Department of Internal Medicine, University of Cincinnati, Cincinnati OH USA
| | - Ann-Kathrin Eisfeld
- The Ohio State University Comprehensive Cancer Center, Columbus, OH USA
- Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus OH USA
- Division of Hematology Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati, Cincinnati OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - H. Leighton Grimes
- Department of Pediatrics, University of Cincinnati, Cincinnati OH USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati OH USA
| | - Robert L. Bowman
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA USA
| | - Linde A. Miles
- Division of Experimental Hematology & Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati OH USA
- Department of Pediatrics, University of Cincinnati, Cincinnati OH USA
- University of Cincinnati Cancer Center, Cincinnati OH USA
| |
Collapse
|
46
|
Chen T, Zhang Y, Zhang D, Zhou H. Immune-based subgroups uncover diverse tumor immunogenicity and implications for prognosis and precision therapy in acute myeloid leukemia. Front Immunol 2024; 15:1451486. [PMID: 39582863 PMCID: PMC11581856 DOI: 10.3389/fimmu.2024.1451486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/26/2024] Open
Abstract
Background Although a considerable proportion of acute myeloid leukemia (AML) patients achieve remission through chemotherapy, relapse remains a recurring and significant event leading to treatment failure. This study aims to investigate the immune landscape in AML and its potential implications for prognosis and chemo-/immune-therapy. Methods Integrated analyses based on multiple sequencing datasets of AML were performed. Various algorithms estimated immune infiltration in AML samples. A subgroup prediction model was developed, and comprehensive bioinformatics and machine learning algorithms were applied to compare immune-based subgroups in relation to clinical features, mutational landscapes, immune characterizations, drug sensitivities, and cellular hierarchies at the single-cell level. Results Two immune-based AML subgroups, G1 and G2, were identified. G1 demonstrated higher immune infiltration, a more monocytic phenotype, increased proportions of monocytes/macrophages, and higher FLT3, DNMT3A, and NPM1 mutation frequencies. It was associated with a poorer prognosis, lower proportions of various immune cell types and a lower T cell infiltration score (TIS). AML T-cell-based immunotherapy target antigens, including CLEC12A, Folate receptor β, IL1RAP and TIM3, showed higher expression levels in G1, while CD117, CD244, CD96, WT and TERT exhibited higher expression levels in G2. G1 samples demonstrated higher sensitivity to elesclomol and panobinostat but increased resistance to venetoclax compared to G2 samples. Moreover, we observed a positive correlation between sample immune infiltration and sample resistance to elesclomol and panobinostat, whereas a negative correlation was found with venetoclax resistance. Conclusion Our study enriches the current AML risk stratification and provides guidance for precision medicine in AML.
Collapse
Affiliation(s)
| | | | | | - Hebing Zhou
- Department of Hematology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
47
|
Zhou X, Zhang L, Aryal S, Veasey V, Tajik A, Restelli C, Moreira S, Zhang P, Zhang Y, Hope KJ, Zhou Y, Cheng C, Bhatia R, Lu R. Epigenetic regulation of noncanonical menin targets modulates menin inhibitor response in acute myeloid leukemia. Blood 2024; 144:2018-2032. [PMID: 39158067 PMCID: PMC11561541 DOI: 10.1182/blood.2023023644] [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: 12/18/2023] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
ABSTRACT Menin inhibitors that disrupt the menin-MLL interaction hold promise for treating specific acute myeloid leukemia (AML) subtypes, including those with KMT2A rearrangements (KMT2A-r), yet resistance remains a challenge. Here, through systematic chromatin-focused CRISPR screens, along with genetic, epigenetic, and pharmacologic studies in a variety of human and mouse KMT2A-r AML models, we uncovered a potential resistance mechanism independent of canonical menin-MLL targets. We show that a group of noncanonical menin targets, which are bivalently cooccupied by active menin and repressive H2AK119ub marks, are typically downregulated after menin inhibition. Loss of polycomb repressive complex 1.1 (PRC1.1) subunits, such as polycomb group ring finger 1 (PCGF1) or BCL6 corepressor (BCOR), leads to menin inhibitor resistance by epigenetic reactivation of these noncanonical targets, including MYC. Genetic and pharmacological inhibition of MYC can resensitize PRC1.1-deficient leukemia cells to menin inhibition. Moreover, we demonstrate that leukemia cells with the loss of PRC1.1 subunits exhibit reduced monocytic gene signatures and are susceptible to BCL2 inhibition, and that combinational treatment with venetoclax overcomes the resistance to menin inhibition in PRC1.1-deficient leukemia cells. These findings highlight the important roles of PRC1.1 and its regulated noncanonical menin targets in modulating the menin inhibitor response and provide potential strategies to treat leukemia with compromised PRC1.1 function.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Epigenesis, Genetic/drug effects
- Mice
- Animals
- Gene Expression Regulation, Leukemic/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Cell Line, Tumor
- Polycomb Repressive Complex 1/genetics
- Polycomb Repressive Complex 1/metabolism
- Polycomb Repressive Complex 1/antagonists & inhibitors
- Sulfonamides/pharmacology
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Xinyue Zhou
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| | - Lixia Zhang
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sajesan Aryal
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| | - Virginia Veasey
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| | - Amanda Tajik
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Cecilia Restelli
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Steven Moreira
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Pengcheng Zhang
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| | - Yanfeng Zhang
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- Department of Genetics, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| | - Kristin J. Hope
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Yang Zhou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham, Birmingham, AL
| | - Changde Cheng
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| | - Ravi Bhatia
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| | - Rui Lu
- Division of Hematology/Oncology, Department of Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL
| |
Collapse
|
48
|
Azimi N, Deshpande AJ. Broken brakes: PRC loss foils menin inhibition. Blood 2024; 144:1979-1980. [PMID: 39509120 PMCID: PMC11561538 DOI: 10.1182/blood.2024026297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Affiliation(s)
- Nahal Azimi
- Sanford Burnham Prebys Medical Discovery Institute
| | | |
Collapse
|
49
|
Loghavi S. SOHO State of the Art Updates and Next Questions-WHO Classification of Acute Myeloid Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:752-758. [PMID: 38866644 DOI: 10.1016/j.clml.2024.05.001] [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: 04/11/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 06/14/2024]
Abstract
The 5th edition of the World Health Organization (WHO) classification of Hematolymphoid tumors provides a hierarchically-driven catalog of hematologic neoplasms and introduces a series of changes to the classification of acute myeloid leukemia (AML). Emphasizing molecular genetic findings, it expands the category of "acute myeloid leukemias with defining genetic abnormalities" while retaining the morphologically defined category of AML for cases that do not harbor disease-defining genetic drivers. The updates to the classification of AML provide refined definitions and diagnostic criteria based on clinicopathologic parameters and molecular genetic findings, emphasizing therapeutically and/or prognostically actionable biomarkers. This review provides an overview of the WHO 5th classification for AML with practical considerations for applying this classification system.
Collapse
Affiliation(s)
- Sanam Loghavi
- Department of Hematopathology, MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
50
|
Driessen A, Unger S, Nguyen AP, Ries RE, Meshinchi S, Kreutmair S, Alberti C, Sumazin P, Aplenc R, Redell MS, Becher B, Rodríguez Martínez M. Identification of single-cell blasts in pediatric acute myeloid leukemia using an autoencoder. Life Sci Alliance 2024; 7:e202402674. [PMID: 39191488 PMCID: PMC11358707 DOI: 10.26508/lsa.202402674] [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: 02/23/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
Pediatric acute myeloid leukemia (AML) is an aggressive blood cancer with a poor prognosis and high relapse rate. Current challenges in the identification of immunotherapy targets arise from patient-specific blast immunophenotypes and their change during disease progression. To overcome this, we present a new computational research tool to rapidly identify malignant cells. We generated single-cell flow cytometry profiles of 21 pediatric AML patients with matched samples at diagnosis, remission, and relapse. We coupled a classifier to an autoencoder for anomaly detection and classified malignant blasts with 90% accuracy. Moreover, our method assigns a developmental stage to blasts at the single-cell level, improving current classification approaches based on differentiation of the dominant phenotype. We observed major immunophenotype and developmental stage alterations between diagnosis and relapse. Patients with KMT2A rearrangement had more profound changes in their blast immunophenotypes at relapse compared to patients with other molecular features. Our method provides new insights into the immunophenotypic composition of AML blasts in an unbiased fashion and can help to define immunotherapy targets that might improve personalized AML treatment.
Collapse
Affiliation(s)
- Alice Driessen
- Data and AI Research, IBM Research Europe, Zürich, Switzerland
- ETH Zürich, Zürich, Switzerland
| | - Susanne Unger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - An-Phi Nguyen
- Data and AI Research, IBM Research Europe, Zürich, Switzerland
| | - Rhonda E Ries
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Stefanie Kreutmair
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Chiara Alberti
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Pavel Sumazin
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Richard Aplenc
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michele S Redell
- Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, USA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|