1
|
Qi Y, Zhu X, Han J, Yan Y, Cui M, Hao Y, Yang L, Dai W, Wu H, Tao Y, He Q, Yu C, Liu F, Fan F. Discovery of cmpd D6 (FH-001) as a efficiency enhancement and myelosuppression degradation small-molecule fms-like tyrosine kinase 3 inhibitor for the treatment of FLT3-ITD positive acute myeloid leukemia. Eur J Pharmacol 2025; 996:177541. [PMID: 40154570 DOI: 10.1016/j.ejphar.2025.177541] [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: 12/17/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
AML is the most common and lethal type of leukemia. The mutant of FLT3 kinase is the most common mutation in AML. Based on the structure analysis and deuteration modification of the cmpd 18 (CHMFL-FLT3-122), a potent and orally available FLT3 Kinase inhibitor, cmpd D6 (FH-001) was found, which demonstrated a remarkable inhibitory effect on the proliferation of FLT3 - ITD positive AML cancer cell lines. Specifically, it effectively suppressed the growth of the MV4-11 cell line (IC50 = 42.8 nM versus 17.1 nM). Similarly, notable inhibitory activity was observed in the MOLM-13 (IC50 = 20.8 nM versus 53.9 nM). More importantly, the IC50 of cmpd D6 to inhibit FLT3 kinase was 338.689 nM and the IC50 to inhibit c-KIT kinase was 3006.042 nM, which were much lower than the IC50 of cmpd 18 to the two kinases, indicating that cmpd D6 may effectively avoid the synthetic lethal myelosuppression toxicity caused by FLT3/c-KIT double inhibition. Pharmacokinetic experiments showed that the deuterated cmpd D6 could prolong the half-life (T1/2 = 4.333 h versus 3.646 h) and improve bioavailability (F = 42.51 % versus 35.93 %). Pharmacodynamic experiments of the three models showed that cmpd D6 (12.5 mg/kg) could significantly inhibit tumor growth compared with cmpd 18, and had no obvious toxicity. Based on the above results, cmpd D6 is a potential candidate drug for the future treatment of FLT3-ITD positive AML.
Collapse
Affiliation(s)
- Yanan Qi
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Xinyi Zhu
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Jingjing Han
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Yuanyuan Yan
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Mengting Cui
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Yanmei Hao
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Lin Yang
- Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; PersonGen BioTherapeutics (Suzhou) Co., Ltd., China; PersonGen.Anke Cellular Therapeutics Co., Ltd., Hefei, Anhui, 230088, China
| | - Wenting Dai
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Hongyan Wu
- Institute of Biomedical Technology, Jiangsu Vocational College of Medicine, 283 South Jiefang Road, Yancheng, 224005, Jiangsu, China
| | - Yu Tao
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Qiwei He
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Chen Yu
- Department of lntegrated TCM & Western Medicine, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Fang Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China.
| | - Fangtian Fan
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China.
| |
Collapse
|
2
|
Lu J, Bhakta P, Hwang H, Lachowiez C, Apostolidou E. Clinical Outcomes of Patients With Newly Diagnosed Acute Myeloid Leukemia Receiving Treatment in a Safety-Net Hospital System. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:349-356. [PMID: 39799045 DOI: 10.1016/j.clml.2024.12.009] [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: 10/31/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND 'Standard of care' therapies for adult acute myeloid leukemia (AML) have yielded 5-year overall survival (OS) rates of 30%-45 %. Risk stratification and novel targeted therapies have improved 5-year OS rates to >75 % for certain groups in specialized centers. PATIENTS AND METHODS This is a retrospective cohort analysis of outcomes in patients ≥18 years with newly diagnosed AML treated between 2005 and 2019 in the Harris Health County, Safety-Net Hospital System in Houston, TX. RESULTS 192 patients were identified. Median age was 52 years, 52 % were male and 57 % identified as Hispanic. Most patients were uninsured or indigent, receiving care under the county's financial assistance programs (62 %). Of the 184 response-assessable patients, 139 achieved composite complete remission (CRc) (76 %). 182 patients had indications for HCT and only 25 patients received HCT (14 %), with main reasons including noncitizenship status and financial/insurance constraints. The 5-year OS rate in the entire cohort was 30 % (35 % in patients <60 years and 16 % if ≥60 years), with 92 % of deaths attributed to AML-related complications. Early death (<4 weeks) rate was 2 %. Secondary, adverse-risk AML, and uninsured status all portended significantly worse OS rates, per multivariate analysis. Patients with indications for HCT who received this modality fared significantly better than those who did not receive it (5-year OS 54 % vs. 21 %). CONCLUSIONS Optimizing AML remission induction regimens, reducing medication costs, ensuring timely administration of AML directed therapies, enhancing equity and diversity in clinical trials, and addressing socioeconomic factors may improve leukemia care for underserved patients.
Collapse
Affiliation(s)
- Jason Lu
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX
| | - Preeya Bhakta
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX
| | - Hyunsoo Hwang
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX
| | - Curtis Lachowiez
- Division of Hematology/Medical Oncology, Oregon Health and Science University, Portland, OR
| | - Effrosyni Apostolidou
- Section of Benign Hematology, Department of Internal Medicine, MD Anderson Cancer Center, Houston, TX.
| |
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
|
Monchusi B, Dube P, Takundwa MM, Kenmogne VL, Malise T, Thimiri Govinda Raj DB. Combination Therapies in Drug Repurposing: Personalized Approaches to Combatting Leukaemia and Multiple Myeloma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40279000 DOI: 10.1007/5584_2025_863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Despite advances in cancer research, treating malignancies remains challenging due to issues like drug resistance, disease heterogeneity, and the limited efficacy of current therapies, particularly in relapsed or refractory cases. In recent years, several drugs originally approved for non-cancer indications have shown potential in cancer treatment, demonstrating anti-proliferative, anti-metastatic, and immunomodulatory effects. Drug repurposing has shown immense promise due to well-established safety profiles and mechanisms of action of the compounds. However, the implementation is fraught with clinical, logistical, regulatory, and ethical challenges, especially in diseases such as leukaemia and multiple myeloma. This chapter examines the treatment challenges in leukaemia and multiple myeloma, focusing on the role of drug repurposing in addressing therapeutic resistance and disease variability. It highlights the potential of personalized, tailored combination therapies, using repurposed drug components, to offer more effective, targeted, and cost-efficient treatment strategies, overcoming resistance and improving patient outcomes.
Collapse
Affiliation(s)
- B Monchusi
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - P Dube
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - M M Takundwa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - V L Kenmogne
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - T Malise
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - D B Thimiri Govinda Raj
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
5
|
George BM, Eleftheriou M, Yankova E, Perr J, Chai P, Nestola G, Almahayni K, Evans S, Damaskou A, Hemberger H, Lebedenko CG, Rak J, Yu Q, Bapcum E, Russell J, Bagri J, Volk RF, Spiekermann M, Stone RM, Giotopoulos G, Huntly BJP, Baxter J, Camargo F, Liu J, Zaro BW, Vassiliou GS, Möckl L, de la Rosa J, Flynn RA, Tzelepis K. Treatment of acute myeloid leukemia models by targeting a cell surface RNA-binding protein. Nat Biotechnol 2025:10.1038/s41587-025-02648-2. [PMID: 40269321 DOI: 10.1038/s41587-025-02648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
Immunotherapies for acute myeloid leukemia (AML) and other cancers are limited by a lack of tumor-specific targets. Here we discover that RNA-binding proteins and glycosylated RNAs (glycoRNAs) form precisely organized nanodomains on cancer cell surfaces. We characterize nucleophosmin (NPM1) as an abundant cell surface protein (csNPM1) on a variety of tumor types. With a focus on AML, we observe csNPM1 on blasts and leukemic stem cells but not on normal hematopoietic stem cells. We develop a monoclonal antibody to target csNPM1, which exhibits robust anti-tumor activity in multiple syngeneic and xenograft models of AML, including patient-derived xenografts, without observable toxicity. We find that csNPM1 is expressed in a mutation-agnostic manner on primary AML cells and may therefore offer a general strategy for detecting and treating AML. Surface profiling and in vivo work also demonstrate csNPM1 as a target on solid tumors. Our data suggest that csNPM1 and its neighboring glycoRNA-cell surface RNA-binding protein (csRBP) clusters may serve as an alternative antigen class for therapeutic targeting or cell identification.
Collapse
Affiliation(s)
- Benson M George
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Eleftheriou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Eliza Yankova
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK
| | - Jonathan Perr
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Peiyuan Chai
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Gianluca Nestola
- Department of Physics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Karim Almahayni
- Department of Physics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Siân Evans
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Aristi Damaskou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Helena Hemberger
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Charlotta G Lebedenko
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Justyna Rak
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Qi Yu
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Ece Bapcum
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - James Russell
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Jaana Bagri
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Regan F Volk
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | | | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George Giotopoulos
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Brian J P Huntly
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Joanna Baxter
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Fernando Camargo
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Jie Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Balyn W Zaro
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - George S Vassiliou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Leonhard Möckl
- Max Planck Institute for the Science of Light, Erlangen, Germany
- Faculty of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Faculty of Sciences, Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jorge de la Rosa
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Cambridge Institute of Science, Altos Labs, Cambridge, UK
| | - Ryan A Flynn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Konstantinos Tzelepis
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Milner Therapeutics Institute, University of Cambridge, Cambridge, UK.
- Wellcome Trust Sanger Institute, Hinxton, UK.
| |
Collapse
|
6
|
Leber B, Ruiz MT, Elgendy H, Pettersson F, Prebet T, Vigil CE, Parikh RC, Korgaonkar S, Bello F, Davis KL, Gaugler L, Strocchia M, Sieluk J, Li Y, Schuh AC. Real-world treatment patterns and outcomes with oral azacitidine maintenance therapy in patients with acute myeloid leukemia. Cancer 2025; 131:e35845. [PMID: 40233158 DOI: 10.1002/cncr.35845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 04/17/2025]
Abstract
INTRODUCTION This study describes baseline and clinical characteristics, treatment patterns, survival, and safety outcomes of patients with acute myeloid leukemia (AML) who received oral azacitidine (oral-AZA) maintenance therapy in Canada following its approval in 2021. METHODS A retrospective, observational medical record review was conducted of patients with AML in remission after induction therapy and who initiated treatment with oral-AZA between March 2021 and July 2023 in Canada. Real-world relapse-free survival and overall survival outcomes were estimated using Kaplan-Meier methodology. RESULTS Data from 119 patients were analyzed. The median age at oral-AZA initiation was 62.5 years. Most patients had favorable (39.5%) or intermediate (39.5%) genetic risk per the 2017/2022 European LeukemiaNet classification. Nearly all patients (99.2%) received cytarabine-based induction regimens. A total of 55.5% of patients received consolidation therapy, with a median of two cycles. After a median follow-up of 9.4 months, 68.1% of all patients were still receiving oral-AZA at last follow-up. After oral-AZA treatment, 21.0% of patients relapsed. Rates of real-world relapse-free survival and overall survival at 12 months from oral-AZA initiation were 66.9% and 74.5%, respectively. During oral-AZA treatment, 67.2% of patients experienced ≥1 adverse event. Concomitant antiemetic treatment was received by 78.2% of patients. CONCLUSION These findings provide real-world evidence further supporting the use of oral-AZA as a standard-of-care maintenance therapy in current routine clinical practice for patients with AML in remission who do not receive hematopoietic stem cell transplantation. These results may inform a broader clinical audience because of the inclusion of patients with diverse demographic and clinical characteristics.
Collapse
Affiliation(s)
- Brian Leber
- McMaster University, Hamilton, Ontario, Canada
| | | | - Hany Elgendy
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | | | | | - Rohan C Parikh
- RTI Health Solutions, Research Triangle Park, North Carolina, USA
| | | | - Fareedat Bello
- RTI Health Solutions, Research Triangle Park, North Carolina, USA
| | - Keith L Davis
- RTI Health Solutions, Research Triangle Park, North Carolina, USA
| | - Lona Gaugler
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Jan Sieluk
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Yeran Li
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | |
Collapse
|
7
|
Métois A, Bordeleau ME, Theret L, Hajmirza A, Moujaber O, Spinella JF, Chagraoui J, Mayotte N, Boivin I, Audemard É, Aubert L, Lisi V, Khakipoor B, Farah A, Bonneil É, Robert A, Lippens J, Moraitis A, Béliveau F, Feghaly A, Boucher G, Marcotte R, Gendron P, Thibault P, Lemieux S, Richard-Carpentier G, Lavallée VP, Hébert J, Roux PP, Sauvageau G. IL1RAP is an immunotherapeutic target for normal karyotype triple-mutated acute myeloid leukemia. Biomark Res 2025; 13:61. [PMID: 40229904 PMCID: PMC11995633 DOI: 10.1186/s40364-025-00769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/16/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Surface antigens of potential clinical significance remain under-characterized in AML. The European Leukemia Network classifies normal karyotype AML (NK-AML) mutated for NPM1 (NPM1c) as a distinct entity associated with favorable outcomes if not associated with FLT3-ITD mutation. A subset of NPM1c NK-AML shows additional mutations in 2 genes: FLT3 (FLT3-ITD) and DNMT3 A. These leukemias, also referred to as NK triple mutated AML (NKt-AML), are particularly difficult to eradicate with current treatment options. Therefore, novel therapies are necessary that use proteins specifically expressed at the surface. METHODS In order to identify surface antigens for immunotherapy in NKt-AML, an extensive multi-omic analysis was conducted on primary AML samples. Surface proteome enrichment was performed on 100 primary AML samples, twelve of which were NKt-AML. Transcriptome analysis was carried out on the 691 primary AML samples, and single-cell RNA sequencing was conducted on 23 primary AML samples. RESULTS Herein, using multi-omics data from the Leucegene collection, we identify IL1RAP as a promising antigen for this AML subgroup. We demonstrate that IL1RAP is expressed at the surface of primitive AML cells reminiscent of leukemic stem cells in NKt-AML primary human AML specimens, while showing relatively low expression levels in normal bone marrow HSCs. Furthermore, results indicate that elevated IL1RAP expression associates with poor overall and relapse-free survival in the Leucegene cohort of AML patients and predicts nonresponse to hematopoietic stem cell transplantation. Finally, we show that IL1RAP protein is internalized following exposure to specific antibodies, suggesting that IL1RAP represents an interesting target for antibody-drug conjugate development in NKt-AML. CONCLUSIONS IL1RAP exhibits preferential expression within NKt-AML, correlating with diminished overall survival rates and diminished responsiveness to hematopoietic stem cell transplantation. Moreover, internalization of IL1RAP presents a promising avenue for immunotherapeutic intervention.
Collapse
Affiliation(s)
- Arnaud Métois
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Marie-Eve Bordeleau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Louis Theret
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Azadeh Hajmirza
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Ossama Moujaber
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Jean-François Spinella
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Jalila Chagraoui
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Nadine Mayotte
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Isabel Boivin
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Éric Audemard
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Léo Aubert
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Véronique Lisi
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Québec, H3 T 1 C5, Canada
| | - Banafsheh Khakipoor
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Québec, H3 T 1 C5, Canada
| | - Azer Farah
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Québec, H3 T 1 C5, Canada
| | - Éric Bonneil
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Alma Robert
- Medical Devices Research Center, National Research Council Canada, Montréal, Québec, H4P 2R2, Canada
| | - Julie Lippens
- Medical Devices Research Center, National Research Council Canada, Montréal, Québec, H4P 2R2, Canada
| | - Anna Moraitis
- Medical Devices Research Center, National Research Council Canada, Montréal, Québec, H4P 2R2, Canada
| | - François Béliveau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, Québec, H1 T 2M4, Canada
| | - Albert Feghaly
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Geneviève Boucher
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Richard Marcotte
- Human Health Therapeutic Research Center National Research Council Canada, Montréal, Québec, H4P 2R2, Canada
| | - Patrick Gendron
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Pierre Thibault
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Department of Chemistry, Faculty of Arts and Science, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Sébastien Lemieux
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Guillaume Richard-Carpentier
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2 C1, Canada
- Department of Medicine, Division of Medical Oncology and Hematology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1 A8, Canada
| | - Vincent-Philippe Lavallée
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, Québec, H3 T 1 C5, Canada
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Hematology and Oncology Division, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec, H3 T 1 C5, Canada
| | - Josée Hébert
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, Québec, H1 T 2M4, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Québec, H1 T 2M4, Canada
| | - Philippe P Roux
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada
| | - Guy Sauvageau
- The Leucegene Project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada.
- Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, Québec, H1 T 2M4, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3 T 1 J4, Canada.
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Québec, H1 T 2M4, Canada.
| |
Collapse
|
8
|
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
|
9
|
Guan Z, Zhang Z, Wang K, Qiao S, Ma T, Wu L. Targeting myeloid cells for hematological malignancies: the present and future. Biomark Res 2025; 13:59. [PMID: 40205623 PMCID: PMC11983845 DOI: 10.1186/s40364-025-00775-1] [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: 01/17/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
Hematological malignancies are a diverse group of cancers that originate in the blood and bone marrow and are characterized by the abnormal proliferation and differentiation of hematopoietic cells. Myeloid blasts, which are derived from normal myeloid progenitors, play a central role in these diseases by disrupting hematopoiesis and driving disease progression. In addition, other myeloid cells, including tumor-associated macrophages and myeloid-derived suppressor cells, adapt dynamically to the tumor microenvironment, where they can promote immune evasion and resistance to treatment. This review explores the unique characteristics and pathogenic mechanisms of myeloid blasts, the immunosuppressive roles of myeloid cells, and their complex interactions within the TME. Furthermore, we highlight emerging therapeutic approaches targeting myeloid cells, focusing on strategies to reprogram their functions, inhibit their suppressive effects, or eliminate pathological populations altogether, as well as the latest preclinical and clinical trials advancing these approaches. By integrating insights from these studies, we aim to provide a comprehensive understanding of the roles of myeloid cells in hematological malignancies and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Zihui Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Peking University First Hospital, Beijing, 100034, China
| | - Zhengqi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Kaiyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shukai Qiao
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Teng Ma
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
| | - Lina Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
10
|
Pienkowski T, Golonko A, Bolkun L, Wawrzak-Pienkowska K, Szczerbinski L, Kretowski A, Ciborowski M, Lewandowski W, Priebe W, Swislocka R. Investigation into biased signaling, glycosylation, and drug vulnerability of acute myeloid leukemia. Pharmacol Ther 2025; 270:108848. [PMID: 40194743 DOI: 10.1016/j.pharmthera.2025.108848] [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: 09/03/2024] [Revised: 01/22/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025]
Abstract
Understanding and harnessing biased signaling offers significant potential for developing novel therapeutic strategies or enhancing existing treatments. By managing biased signaling, it is possible to minimize adverse effects, including toxicity, and to optimize therapeutic outcomes by selectively targeting beneficial pathways. In the context of acute myeloid leukemia (AML), a highly aggressive blood cancer characterized by the rapid proliferation of abnormal myeloid cells in the bone marrow and blood, the dysregulation of these signaling pathways, particularly those involving G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs), significantly contributes to disease progression and therapeutic resistance. Traditional therapies for AML often struggle with resistance and toxicity, leading to poor patient outcomes. However, by exploiting the concept of biased signaling, researchers may be able to design drugs that selectively activate pathways that inhibit cancer cell growth while avoiding those that contribute to resistance or toxicity. Glycosylation, a key post-translational modification (PTM), plays a crucial role in biased signaling by altering receptor conformation and ligand-binding affinity, thereby affecting the outcome of biased signaling. Chemokine receptors like CXCR4, which are often overexpressed and heavily glycosylated in AML, serve as targets for therapeutic intervention. By externally inducing or inhibiting specific PTMs, it may be possible to further refine therapeutic strategies, unlocking new possibilities for developing more effective and less toxic treatments. This review highlights the importance of understanding the dynamic relationship between glycosylation and biased signaling in AML, which is essential for the development of more effective treatments and overcoming drug resistance, ultimately leading to better patient outcomes.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Aleksandra Golonko
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland; Waclaw Dabrowski Institute of Agricultural and Food Biotechnology State Research Institute, Rakowiecka 36, 02-532 Warsaw, Poland.
| | - Lukasz Bolkun
- Department of Hematology, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Katarzyna Wawrzak-Pienkowska
- Department of Gastroenterology, Hepatology and Internal Diseases, Voivodeship Hospital in Bialystok, 15-278 Bialystok, Poland; Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Lukasz Szczerbinski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Wlodzimierz Lewandowski
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1901 East Rd., Houston, TX 77054, USA
| | - Renata Swislocka
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland
| |
Collapse
|
11
|
Giri AK, Lin J, Kyriakidis K, Tripathi G, Almusa H. Exome-wide association study reveals 7 functional variants associated with ex-vivo drug response in acute myeloid leukemia patients. BMC Med Genomics 2025; 18:64. [PMID: 40186177 PMCID: PMC11969768 DOI: 10.1186/s12920-025-02130-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer characterized by poor survival outcomes. Further, due to the extreme molecular heterogeneity of the disease, drug treatment response varies from patient to patient. The variability of drug response can cause unnecessary treatment in more than half of the patients with no or partial therapy responses leading to severe side effects, monetary as well as time loss. Understanding the genetic risk factors underlying the drug response in AML can help with improved prediction of treatment responses and identification of biomarkers in addition to mechanistic insights to monitor treatment response. Here, we report the results of the first Exome-Wide Association Study (EWAS) of ex-vivo drug response performed to date with 175 AML cases and 47 drugs. We used information from 55,423 germline exonic SNPs to perform the analysis. We identified exome-wide significant (p < 9.02 × 10- 7) associations for rs113985677 in CCIN with tamoxifen response, rs115400838 in TRMT5 with idelalisib response, rs11878277 in HDGFL2 with entinostat, and rs2229092 in LTA associated with vorinostat response. Further, using multivariate genome-wide association analysis, we identified the association of rs11556165 in ATRAID, and rs11236938 in TSKU with the combined response of all 47 drugs and 29 nonchemotherapy drugs at the genome-wide significance level (p < 5 × 10- 8). Additionally, a significant association of rs35704242 in NIBAN1 was associated with the combined response for nonchemotherapy medicines (p = 2.51 × 10- 8), and BI.2536, gefitinib, and belinostat were identified as the central traits. Our study represents the first EWAS to date on ex-vivo drug response in AML and reports 7 new associated loci that help to understand the anticancer drug response in AML patients.
Collapse
Affiliation(s)
- Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
- Foundation for the Finnish Cancer Institute (FCI), Tukholmankatu 8, Helsinki, 00290, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, PO Box 20, Helsinki, FI-00014, Finland.
| | - Jake Lin
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Konstantinos Kyriakidis
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Garima Tripathi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Chen EC, Shimony S, Luskin MR, Stone RM. Biology and Management of Acute Myeloid Leukemia With Mutated NPM1. Am J Hematol 2025; 100:652-665. [PMID: 39901865 DOI: 10.1002/ajh.27600] [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: 10/28/2024] [Revised: 12/27/2024] [Accepted: 01/05/2025] [Indexed: 02/05/2025]
Abstract
Mutations in nucleophosmin 1 (NPM1) are diseased-defining genetic alterations encountered in approximately one-third of cases of acute myeloid leukemia (AML). A mutation in NPM1 confers a more favorable prognosis; however, clinical outcomes of NPM1-mutated AML (NPM1 mut AML) are diverse due to the heterogeneity of disease biology, patient characteristics, and treatment received. Research over the last two decades has dramatically expanded our understanding of the biology of NPM1 mut AML and led to the development of new therapeutic approaches and strategies for monitoring measurable residual disease (MRD). Here, we review NPM1 mut AML with a practical focus on the current treatment landscape, the role of MRD in guiding management, and emerging therapies, including menin inhibitors.
Collapse
Affiliation(s)
- Evan C Chen
- Adult Leukemia Program, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Shai Shimony
- Adult Leukemia Program, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Marlise R Luskin
- Adult Leukemia Program, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Richard M Stone
- Adult Leukemia Program, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Bruzzese A, Martino EA, Labanca C, Mendicino F, Lucia E, Olivito V, Rossi T, Neri A, Morabito F, Vigna E, Gentile M. Advances and Challenges in Quizartinib-Based FLT3 Inhibition for Acute Myeloid Leukemia: Mechanisms of Resistance and Prospective Combination Therapies. Eur J Haematol 2025; 114:584-595. [PMID: 39763167 PMCID: PMC11880963 DOI: 10.1111/ejh.14383] [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: 11/05/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 03/06/2025]
Abstract
FLT3 mutations are among the most common genetic alterations in acute myeloid leukemia (AML) and are associated with poor prognosis. Significant advancements have been made in developing FLT3 inhibitors (FLT3Is), such as quizartinib, which have improved treatment outcomes in both newly diagnosed and relapsed/refractory AML. Resistance to FLT3Is remains a major clinical challenge, driven by diverse mechanisms including FLT3 point mutations, cellular escape pathways, and the influence of the bone marrow microenvironment. Sustained STAT5 phosphorylation, AXL upregulation, and CXCR4 signaling have been identified as key factors in FLT3I resistance. Additionally, metabolic adaptations have been shown to support the survival of FLT3I-resistant cells. Ongoing clinical trials are investigating various combination regimens, including quizartinib with chemotherapy, Bcl-2 inhibitors, hypomethylating agents, and immune-modulatory drugs, with promising preliminary results. The European LeukemiaNet 2022 guidelines recommend incorporating FLT3Is into treatment regimens; however, questions remain regarding the best timing for the administration of each FLT3I. Additional studies are required to determine the optimal FLT3I-based combinations, reduce resistance emergence, and improve outcomes. This review highlights the current state of FLT3I therapy, ongoing challenges with resistance, and future directions in optimizing treatment for FLT3-mutated AML, focusing on quizartinib.
Collapse
Affiliation(s)
| | | | | | | | - Eugenio Lucia
- Hematology Unit, Azienda Ospedaliera AnnunziataCosenzaItaly
| | | | - Teresa Rossi
- Laboratorio di Ricerca Traslazionale Azienda USL‐IRCSSReggio EmiliaEmilia‐RomagnaItaly
| | - Antonino Neri
- Scientific Directorate IRCCS of Reggio EmiliaReggio EmiliaEmilia‐RomagnaItaly
| | | | - Ernesto Vigna
- Hematology Unit, Azienda Ospedaliera AnnunziataCosenzaItaly
| | - Massimo Gentile
- Hematology Unit, Azienda Ospedaliera AnnunziataCosenzaItaly
- Department of Pharmacy, Health and Nutritional ScienceUniversity of CalabriaRendeItaly
| |
Collapse
|
14
|
Nanni J, Azzali I, Papayannidis C, Mulè A, Audisio E, Martelli MP, Scappini B, Chiusolo P, Cambò B, Candoni A, Lunghi M, Albano F, Olivieri A, Fracchiolla N, Bernardi M, Romani C, Rigolin GM, Giannini MB, Bocchia M, Todisco E, Cilloni D, Bochicchio MT, Ottaviani E, Mattei A, Zamagni F, Valli I, Volpi R, Marconi G, Petracci E, Martinelli G. Upfront intensive treatment analysis of the Italian Cohort Study on FLT3-mutated AML patients (FLAM): The impact of a FLT3 inhibitor addition to standard chemotherapy in the real-life setting. Cancer 2025; 131:e35824. [PMID: 40159434 PMCID: PMC11955083 DOI: 10.1002/cncr.35824] [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: 01/08/2025] [Accepted: 01/29/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND The addition of a FLT3 inhibitor (FLT3i) to standard chemotherapy to treat fit newly diagnosed (ND) patients with FLT3-mutated acute myeloid leukemia (AML) represents the standard of care resulting from clinical trial results. However, evidence regarding FLT3i adoption in routine clinical practice is still scarce. METHODS Clinical data are reported from 394 ND patients with FLT3-mutated AML enrolled in the retrospective observational Italian Cohort Study on FLT3-mutated patients with AML and treated with an upfront intensive regimen with (FLT3i group, n = 92) or without (CT group, n = 302) the addition of a FLT3i. RESULTS With a median follow-up time of 34.5 months, an effectiveness benefit obtained by FLT3i incorporation both in terms of overall survival (median, 34.9 in the FLT3i vs 12.7 months in the CT group, p < .01) and relapse-free survival (median, 18.9 in the FLT3i vs 7.6 months in the CT group, p = .01) was documented, with a higher composite complete remission rate (75.4% in the FLT3i vs 62.4% in the CT group, p = .052). FLT3i benefit seemed to be independent from the transplant rate. CONCLUSIONS In conclusion, the benefit of FLT3i addition to upfront intensive treatment in newly diagnosed FLT3-mutated AML patients was confirmed in a large, real-life cohort study.
Collapse
Affiliation(s)
- Jacopo Nanni
- Dipartimento di Scienze Mediche e ChirurgicheIstituto di Ematologia “Seràgnoli”University of BolognaBolognaItaly
| | - Irene Azzali
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| | - Cristina Papayannidis
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
| | - Antonino Mulè
- U.O.C. di OncoematologiaA.O.O.R. Villa Sofia – CervelloPalermoItaly
| | - Ernesta Audisio
- SC Ematologia 2Dipartmento di Ematologia e OncologiaAO Città della Salute e della ScienzaTorinoItaly
| | - Maria Paola Martelli
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and Santa Maria della Misericordia HospitalPerugiaItaly
| | | | - Patrizia Chiusolo
- Dipartimento di Scienze Radiologiche ed EmatologicheUniversità Cattolica del Sacro CuoreRomaItaly
| | - Benedetta Cambò
- Department of Medicine and SurgeryHematology and BMT UnitUniversity of ParmaParmaItaly
| | - Anna Candoni
- Clinica Ematologica Azienda Sanitaria Universitaria Integrata di UdineUdineItaly
| | - Monia Lunghi
- Division of HematologyDepartment of Translational MedicineUniversità del Piemonte OrientaleNovaraItaly
| | - Francesco Albano
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe‐J)Hematology and Stem Cell Transplantation UnitUniversity of Bari “Aldo Moro”BariItaly
| | | | - Nicola Fracchiolla
- Fondazione IRCCS Ca’ Granda‐Ospedale Maggiore Policlinico di MilanoMilanoItaly
| | - Massimo Bernardi
- IRCCSOspedale San Raffaele s.r.l U.O. Ematologia e TMO MilanoMilanoItaly
| | - Claudio Romani
- SC Ematologia e CTMOAzienda Ospedaliera BrotzuCagliariItaly
| | | | | | | | - Elisabetta Todisco
- European Institute of OncologyMilanoItaly
- S.C. di Ematologia e Trapianto di Cellule Staminali EmopoieticheOspedale di Busto ArsizioASST Valle OlonaBusto Arsizio (VA)Italy
| | - Daniela Cilloni
- Dipartimento di Scienze Cliniche e BiologicheUniversità di TorinoS.S.D Terapia onco‐ematologica intensiva e trapianto CSEAOU San Luigi GonzagaOrbassano (TO)Italy
| | | | - Emanuela Ottaviani
- Dipartimento di Scienze Mediche e ChirurgicheIstituto di Ematologia “Seràgnoli”University of BolognaBolognaItaly
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
| | - Agnese Mattei
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| | - Federica Zamagni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| | - Irene Valli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| | - Roberta Volpi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| | - Giovanni Marconi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| | - Elisabetta Petracci
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| | | | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST)Meldola (FC)Italy
| |
Collapse
|
15
|
Senapati J, Kadia TM, Daver NG, DiNardo CD, Borthakur G, Ravandi F, Kantarjian HM. Therapeutic horizon of acute myeloid leukemia: Success, optimism, and challenges. Cancer 2025; 131:e35806. [PMID: 40105906 DOI: 10.1002/cncr.35806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 03/21/2025]
Abstract
Focused research in acute myeloid leukemia (AML) biology and treatment has led to the identification of new therapeutic targets and several new drug approvals over the last decade. Progressive improvements in response and survival have mirrored these improvements in treatment options. Traditionally adverse subtypes such as FLT3-internal tandem duplication-positive AML now have better outcomes with potent FLT3 inhibitors, and menin inhibitors in KMT2A-rearranged and other MEIS/HOX-dependent leukemias hold promise toward improving outcomes. More patients with AML are now able to undergo a consolidative allogeneic hematopoietic stem cell transplantation (HSCT), and the rates of nonrelapse mortality with or without HSCT have also decreased. Comprehensive genomic interrogation of AML has elucidated mechanisms of response and resistance to treatments, which has enabled more precise decision algorithms and better prognostication. Deep levels of measurable residual disease assessment in some AML subsets hold the potential to dynamically modify treatment on the basis of these responses. Improving frontline intensive and low-intensity therapies, by incorporating venetoclax and other targeted agents, is the most important intervention to improve AML outcomes. Despite these developments, a sizeable percentage of AML, such as AML with TP53 or MECOM aberrations, postmyeloproliferative neoplasm AML, and so forth, remains as subsets without significant improvement in outcomes and no targeted options. Evolving strategies with natural killer cell-based approaches, novel antibody-drug conjugates, bispecific T-cell engagers, and engineered chimeric antigen receptor T-cell therapies are being evaluated, and may fill the therapeutic vacuum for some of the high-risk AML subtypes.
Collapse
Affiliation(s)
- Jayastu Senapati
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Zhou Q, Li Z, Zhao P, Guan Y, Chu H, Xi Y. FLT3 inhibition upregulates OCT4/NANOG to promote maintenance and TKI resistance of FLT3-ITD + acute myeloid leukemia. Oncogenesis 2025; 14:7. [PMID: 40157912 PMCID: PMC11954930 DOI: 10.1038/s41389-025-00553-6] [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: 08/10/2024] [Revised: 02/09/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025] Open
Abstract
Up to 30% of acute myeloid leukemia (AML) patients face unfavorable outcomes due to the FMS-like receptor tyrosine kinase-3 (FLT3) internal tandem duplication (ITD) mutation. Although FLT3 inhibitors show encouraging outcomes in treatment, they fail to eliminate leukemia stem cells, the origin of persistent and resistant lesions. Exploration of the mechanism in FLT3-ITD+ AML maintenance and chemoresistance is crucial for the development of novel therapeutic approaches. The manifestation of pluripotency transcription factors (TFs) and their link to clinical outcomes have been documented in various tumors. This study investigates the correlation between core pluripotency TF and treatment in AML. We discovered that FLT3 inhibition induced upregulation of OCT4 and NANOG in FLT3-ITD+ AML cells. Subsequently, we demonstrated that downregulation of OCT4 or NANOG inhibited cell growth, promoted apoptosis, and induced G0/G1 cell cycle phase arrest in FLT3-ITD+ AML cells. Knockdown of OCT and NANOG inhibited tumor growth in a mouse tumor model. OCT4 promotes the malignant biological behavior of FLT3-ITD+ AML by enhancing the abnormal FLT3 signaling pathway through transcriptional activation of NANOG. Importantly, downregulation of OCT4 or NANOG increased responsiveness to FLT3-tyrosine kinase inhibitor (TKI) (Gilteritinib), implying that OCT4 and NANOG may contribute to TKI resistance in FLT3-ITD+ AML. Our study verifies the involvement of OCT4/NANOG in regulating TKI sensitivity and targeting them may improve the cytotoxicity of FLT3-TKIs in FLT3-ITD+ AML.
Collapse
Affiliation(s)
- Qi Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Zijian Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Pingping Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yongyu Guan
- Clinical laboratory, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou, China
| | - Huiyuan Chu
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yaming Xi
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
17
|
Wiese W, Galita G, Siwecka N, Rozpędek-Kamińska W, Slupianek A, Majsterek I. Endoplasmic Reticulum Stress in Acute Myeloid Leukemia: Pathogenesis, Prognostic Implications, and Therapeutic Strategies. Int J Mol Sci 2025; 26:3092. [PMID: 40243748 PMCID: PMC11988921 DOI: 10.3390/ijms26073092] [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: 01/03/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy that poses a significant therapeutic challenge due to its high recurrence rate and demanding treatment regimens. Increasing evidence suggests that endoplasmic reticulum (ER) stress and downstream activation of the unfolded protein response (UPR) pathway play a key role in the pathogenesis of AML. ER stress is triggered by the accumulation of misfolded or unfolded proteins within the ER. This causes activation of the UPR to restore cellular homeostasis. However, the UPR can shift from promoting survival to inducing apoptosis under prolonged or excessive stress conditions. AML cells can manipulate the UPR pathway to evade apoptosis, promoting tumor progression and resistance against various therapeutic strategies. This review provides the current knowledge on ER stress in AML and its prognostic and therapeutic implications.
Collapse
MESH Headings
- Humans
- Endoplasmic Reticulum Stress
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/diagnosis
- Unfolded Protein Response
- Prognosis
- Apoptosis
- Animals
- Signal Transduction
Collapse
Affiliation(s)
- Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Grzegorz Galita
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Artur Slupianek
- Office of the Vice President for Research, Temple University, Philadelphia, PA 19140, USA
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| |
Collapse
|
18
|
Kazmi F, Shrestha N, Liu TFD, Foord T, Heesen P, Booth S, Dodwell D, Lord S, Yeoh KW, Blagden SP. Next-generation sequencing for guiding matched targeted therapies in people with relapsed or metastatic cancer. Cochrane Database Syst Rev 2025; 3:CD014872. [PMID: 40122129 PMCID: PMC11930395 DOI: 10.1002/14651858.cd014872.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
BACKGROUND Matched targeted therapies (MTT) given alone or in combination with systemic anti-cancer therapies have delivered proven survival benefit for many people with newly diagnosed cancer. However, there is little evidence of their effectiveness in the recurrent or late-stage setting. With this uncertainty, alongside the perception that late-stage cancers are too genetically heterogenous or too mutationally diverse to benefit from matched targeted therapies, next-generation sequencing (NGS) of tumours in people with refractory cancer remains a low priority. As a result, next-generation sequencing testing of recurrent or late-stage disease is discouraged. We lack evidence to support the utility of next generation sequencing in guiding matched targeted therapies in this setting. OBJECTIVES To evaluate the benefits and harms of matched targeted therapies in people with advanced cancers in randomised controlled trials. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, ClinicalTrials.gov, and the World Health Organisation International Clinical Trials Registry Platform (WHO-ICTRP) search portal up to 30th October 2024. We also screened reference lists of included studies and also the publications that cited these studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) that had enroled participants with advanced/refractory solid or haematological cancers who had progressed through at least one line of standard anti-cancer systemic therapy. To be eligible, all participants should have received matched targeted therapy based on next-generation sequencing carried out on their tumour (tumour tissue, blood or bone marrow). DATA COLLECTION AND ANALYSIS We systematically searched medical databases (e.g. MEDLINE, Embase) and trial registers for randomised controlled trials (RCTs). Outcomes of interest were progression-free survival (PFS), overall survival (OS), overall response rates (ORR), serious (grade 3 or 4) adverse events (AEs) and quality of life (QOL). We used a random-effects model to pool outcomes across studies and compared predefined subgroups using interaction tests. Grading of Recommendations Assessment, Development and Evaluation (GRADE) assessment of certainty was used to evaluate the quality of evidence. MAIN RESULTS We identified a total of 37 studies, out of which 35 studies (including 9819 participants) were included in the meta-analysis. All included studies compared a matched targeted therapy intervention to standard-of-care treatment, non-matched targeted therapies or no treatment (best supportive care): Matched targeted therapy versus standard-of-care treatment Matched targeted therapy (MTT) compared with standard systematic therapy probably reduces the risk of disease progression by 34% (hazard ratio (HR) = 0.66, 95% confidence interval (CI) 0.59 to 0.74; 14 studies, 3848 participants; moderate-certainty evidence). However, MTT might have little to no difference in risk of death (HR = 0.85, 95% CI 0.75 to 0.97; 14 studies, 3848 participants; low-certainty evidence) and may increase overall response rates (low-certainty evidence). There was no clear evidence of a difference in severe (grade 3/4) adverse events between matched targeted therapy and standard-of-care treatment (low-certainty evidence). There was limited evidence of a difference in quality of life between groups (very low-certainty of evidence). Matched targeted therapy in combination with standard-of-care treatment versus standard-of-care treatment alone Matched targeted therapy in combination with standard-of-care treatment compared with standard-of-care treatment alone probably reduces the risk of disease progression by 39% (HR = 0.61, 95% CI 0.53-0.70, 14 studies, 2,637 participants; moderate-certainty evidence) and risk of death by 21% (HR = 0.79, 95% CI 0.70 to 0.89; 11 studies, 2575 participants, moderate-certainty evidence). The combination of MTT and standard-of-care treatment may also increase overall response rates (low-certainty evidence). There was limited evidence of a difference in the incidence of severe adverse events (very low-certainty evidence) and quality of life between the groups (very low-certainty of evidence). Matched targeted therapy versus non-matched targeted therapy Matched targeted therapy compared with non-matched targeted therapy probably reduces the risk of disease progression by 24% (HR = 0.76, 95% CI 0.64 to 0.89; 3 studies, 1568 participants; moderate-certainty evidence) and may reduce the risk of death by 25% (HR = 0.75, 95% CI 0.65 to 0.86, 1307 participants; low-certainty evidence). There was little to no effect on overall response rates between MTT and non-MTT. There was no clear evidence of a difference in overall response rates (low-certainty evidence) and severe adverse events between MTT and non-MTT (low-certainty evidence). None of the studies comparing MTT and non-MTT reported quality of life. Matched targeted therapy versus best supportive care Matched targeted therapy compared with the best supportive care (BSC) i.e. no active treatment probably reduces the risk of disease progression by 63% (HR 0.37, 95% CI 0.28 to 0.50; 4 studies, 858 participants; moderate-certainty evidence). There was no clear evidence of a difference in overall survival between groups (HR = 0.88, 95% CI 0.73 to 1.06, 3 studies, 783 participants; low-certainty evidence). There was no clear evidence of a difference in overall response rates (very low-certainty of evidence) and incidence of severe adverse events (very low-certainty of evidence) between the groups. Quality of life was reported in a single study but did not provide composite scores. Risk of bias The overall risk of bias was judged low for eight studies, unclear for two studies, and the remaining 27 studies were high risk. AUTHORS' CONCLUSIONS Matched targeted therapies guided by next-generation sequencing in people with advanced cancer prolongs the time before cancer progresses compared to standard therapies. However, there is limited evidence to suggest that it prolongs overall survival, improves the quality of life or increases adverse events. Importantly, this review supports equitable access to next-generation sequencing technology for all people with advanced cancer and offers them the opportunity to access genotype-matched targeted therapies.
Collapse
Affiliation(s)
- Farasat Kazmi
- Department of Oncology, University of Oxford, Oxford, UK
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Nipun Shrestha
- Health Evidence Synthesis, Recommendations and Impact (HESRI), School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Tik Fung Dave Liu
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | | | | | - Stephen Booth
- Department of Haematology, Royal Berkshire Hospital, Reading, UK
| | - David Dodwell
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Oxford, UK
| | - Kheng-Wei Yeoh
- Radiation Oncology, National Cancer Centre, Singapore, Singapore
| | | |
Collapse
|
19
|
Tecik M, Adan A. Concurrent inhibition of FLT3 and sphingosine kinase-1 triggers synergistic cytotoxicity in midostaurin resistant FLT3-ITD positive acute myeloid leukemia cells via blocking FLT3/STAT5A signaling to induce apoptosis. J Chemother 2025:1-17. [PMID: 40119531 DOI: 10.1080/1120009x.2025.2478340] [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: 10/28/2024] [Revised: 02/11/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025]
Abstract
The FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) is one of the most frequent mutations observed in acute myeloid leukemia (AML) which contributes to disease progression and unfavorable prognosis. Midostaurin, a small FLT3 inhibitor (FLT3I), is clinically approved. However, patients generally possess acquired resistance when midostaurin used alone. Shifting the balance in the sphingolipid rheostat toward anti-apoptotic sphingosine kinase-1 (SK-1) or glucosylceramide synthase (GCS) is related to therapy resistance in cancer, however, their role in midostaurin resistant FLT3-ITD positive AML has not been previously investigated. We generated midostaurin resistant MV4-11 and MOLM-13 cell lines which showed increased IC50 values compared to their sensitive partner cells. SK-1 is overexpressed in resistant cells while GCS remains unchanged. Subsequent pharmacological targeting of SK-1 in resistant cells decreased SK-1 protein level, inhibited cell proliferation and showed additive or synergistic effect on cell growth, as confirmed by the Chou-Talalay combination index, and induced G0/G1 arrest (PI staining by flow cytometry). Cotreatment (SKI-II plus midostaurin) triggered apoptosis via phosphatidylserine exposure (annexin V/PI double staining). Mechanistically, induction of the intrinsic pathway of apoptosis was confirmed as increased activating cleavages of caspase-3 and PARP and increased Bax/Bcl-2 ratios. Activating phosphorylations of FLT3 (at tyrosine residue 591) and STAT5A (at tyrosine residue 694) dramatically inhibited in resistant cells treated with the combination. In conclusion, midostaurin resistance could be reversed by dual SK-1 and FLT3 inhibition in midostaurin resistant AML cell lines, providing the first evidence of a novel treatment approach to re-sensitize FLT3-ITD positive AML.
Collapse
Affiliation(s)
- Melisa Tecik
- Bioengineering Program, Graduate School of Engineering and Science, Abdullah Gul University, Kayseri, Türkiye
| | - Aysun Adan
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, Türkiye
| |
Collapse
|
20
|
Issa GC, Stein EM, DiNardo CD. How I treat acute myeloid leukemia with differentiation therapy. Blood 2025; 145:1251-1259. [PMID: 38976876 PMCID: PMC11952016 DOI: 10.1182/blood.2024024008] [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: 05/17/2024] [Revised: 06/10/2024] [Accepted: 06/29/2024] [Indexed: 07/10/2024] Open
Abstract
ABSTRACT An increasing number of acute myeloid leukemia (AML) therapeutics have been developed, not as cytotoxic therapies but rather as targeted agents able to restore the aberrant and leukemogenic "block" in normal differentiation. All-trans retinoic acid and arsenic trioxide are classic examples of differentiating agents for treatment of acute promyelocytic leukemia (APL); newer therapies functioning through differentiation include isocitrate dehydrogenase 1 and 2 inhibitors, FMS-like tyrosine kinase 3 inhibitors, and menin inhibitors. The terminal differentiation of leukemic blasts via differentiating-agent therapy can lead to a constellation of signs and symptoms, originally referred to as "retinoic acid syndrome" and now termed "differentiation syndrome" (DS), characterized predominantly by systemic inflammatory response system-like features of dyspnea, pulmonary infiltrates, pleural and pericardial effusions, unexplained fevers, hypotension, edema, and renal insufficiency. DS in patients with newly diagnosed APL is generally straightforward to identify; however, DS in patients with multiply relapsed AML can be more challenging to diagnose, due to nonspecific signs and symptoms that can be mistakenly attributed to infectious etiologies or the underlying refractory leukemia itself. Prompt consideration of DS, rapid initiation of systemic corticosteroids, and early cytoreduction in the setting of concomitant hyperleukocytosis are essential for optimal management.
Collapse
Affiliation(s)
- Ghayas C. Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Eytan M. Stein
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
21
|
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
|
22
|
Leotta S, Giammarco S, Mariotti J. Editorial: Allogenic hematopoietic cell transplant in hematological malignancies: controversies and perspective. Front Oncol 2025; 15:1582751. [PMID: 40177246 PMCID: PMC11961914 DOI: 10.3389/fonc.2025.1582751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Affiliation(s)
- Salvatore Leotta
- Azienda Ospedaliero-Universitaria Policlinico “G-Rodolico”- San Marco - Catania, Catania, Italy
| | - Sabrina Giammarco
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Agostino Gemelli University Policlinic Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Jacopo Mariotti
- Humanitas Cancer Center, Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
23
|
Bazinet A, Bataller A, Kadia T, Daver N, Short NJ, Yilmaz M, Sasaki K, DiNardo CD, Borthakur GM, Issa G, Bouligny I, Pierce S, Garcia-Manero G, Ravandi F, Kantarjian HM. A retrospective study of outcomes across time and treatment regimens in newly diagnosed, FMS-like tyrosine kinase 3 (FLT3)-mutated acute myeloid leukemia. Cancer 2025; 131:e35813. [PMID: 40097915 DOI: 10.1002/cncr.35813] [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: 11/08/2024] [Revised: 01/13/2025] [Accepted: 02/11/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND FMS-like tyrosine kinase 3 (FLT3) mutations, either internal tandem duplications (FLT3-ITD) or tyrosine kinase domain (FLT3-TKD), are common in acute myeloid leukemia (AML). FLT3-ITD confers an adverse prognosis. METHODS The authors performed a retrospective study including 619 patients to evaluate outcomes in newly diagnosed FLT3-mutated AML across treatment regimens. RESULTS In patients with FLT3-ITD-mutated AML who received intensive chemotherapy (IC), the addition of a FLT3 inhibitor (FLT3i) was associated with trends toward improved relapse-free survival (median 32.3 vs. 14.3 months with vs. without a FLT3i; p = .055) and overall survival (OS; 35.5 vs. 18.9 months with vs. without a FLT3i; p = .098). In patients with FLT3-ITD mutations who received low-intensity (LIT) regimens, triplets (LIT plus a FLT3i plus venetoclax) were associated with significantly longer OS (19.1 months) compared with those who received other treatment combinations (11.2 months with LIT alone, 9.2 months with LIT plus FLT3i, and 10.3 months with LIT plus venetoclax). Patients with FLT3-ITD plus NPM1 co-mutations who received any therapy had a trend toward improved OS (2-year OS: 47% vs. 33%; p = .087). The FLT3-ITD allelic ratio; IDH1, IDH2, WT1, RUNX1, and myelodysplastic syndrome-related mutations; and adverse cytogenetics had no significant impact on OS. In landmark analyses, allogeneic stem cell transplantation was associated with a trend toward improved OS in patients with FLT3-ITD mutations who received IC (52.6 vs. 22.7 months with versus without allogeneic stem cell transplantation; p = .076) and a marked improvement in OS in those who received LIT (38.6 vs. 14.0 months with vs. without allogeneic stem cell transplantation; p < .0001). CONCLUSIONS A FLT3i and allogeneic stem cell transplantation are key treatment modalities for patients who have FLT3-mutated AML. LIT-based triplets are promising in IC-ineligible patients.
Collapse
MESH Headings
- Humans
- fms-Like Tyrosine Kinase 3/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/mortality
- Retrospective Studies
- Female
- Middle Aged
- Male
- Nucleophosmin
- Aged
- Mutation
- Adult
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Young Adult
- Treatment Outcome
- Aged, 80 and over
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Sulfonamides/therapeutic use
- Adolescent
- Hematopoietic Stem Cell Transplantation
- Prognosis
- Tandem Repeat Sequences/genetics
Collapse
Affiliation(s)
- Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gautam M Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ghayas Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ian Bouligny
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
24
|
Hong J, Sui P, Li Y, Xu KY, Lee JH, Wang J, Chen S, Zhang P, Wingate N, Noor A, Yuan Y, Hromas R, Zhou H, Hamamoto K, Su R, Yin CC, Ye F, Quesada AE, Chen J, Huang S, Zhou D, You MJ, Yang FC, Wang J, Xu M. PSPC1 exerts an oncogenic role in AML by regulating a leukemic transcription program in cooperation with PU.1. Cell Stem Cell 2025; 32:463-478.e6. [PMID: 39954676 DOI: 10.1016/j.stem.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 11/01/2024] [Accepted: 01/16/2025] [Indexed: 02/17/2025]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy characterized by the blockage of myeloid cell differentiation and uncontrolled proliferation of immature myeloid cells. Here, we show that paraspeckle component 1 (PSPC1) is aberrantly overexpressed and associated with poor survival in AML patients. Using human AML cells and mouse models, we demonstrate that PSPC1 is not required for normal hematopoiesis, but it is critical and essential for AML cells to maintain their leukemic characteristics. PSPC1 loss induces robust differentiation, suppresses proliferation, and abolishes leukemogenesis in diverse AML cells. Mechanistically, PSPC1 exerts a pro-leukemia effect by regulating a unique leukemic transcription program via cooperative chromatin binding with PU.1 and activation of tumor-promoting genes, including NDC1, which is not previously implicated in AML. Our findings uncover a unique and crucial role of PSPC1 dependency in AML and highlight its potential as a promising therapeutic target for AML.
Collapse
Affiliation(s)
- Juyeong Hong
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pinpin Sui
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ying Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kerryn Y Xu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ji-Hoon Lee
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Juan Wang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Shi Chen
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Peng Zhang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Noah Wingate
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Asra Noor
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Yaxia Yuan
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Robert Hromas
- Department of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Hongwei Zhou
- Department of Medicine, Columbia Center for Human Development and Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Karina Hamamoto
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - C Cameron Yin
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Fengxi Ye
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Andrés E Quesada
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Suming Huang
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Daohong Zhou
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - M James You
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Feng-Chun Yang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| | - Jianlong Wang
- Department of Medicine, Columbia Center for Human Development and Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Mingjiang Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
25
|
Jamy O, Bourne G, Mudd TW, Thigpen H, Bhatia R. Revisiting the Role of Day 14 Bone Marrow Biopsy in Acute Myeloid Leukemia. Cancers (Basel) 2025; 17:900. [PMID: 40075747 PMCID: PMC11899312 DOI: 10.3390/cancers17050900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
In recent years, the practice of routinely obtaining day 14 bone marrow biopsies during AML intensive induction therapy has been scrutinized. While current guidelines recommend obtaining mid-induction biopsies to gauge early response to treatment and guide potential changes in future management, concerns have been raised that these biopsies may not be as prognostically accurate as hoped and subsequently may result in additional and unwarranted chemotherapy toxicity in select patients. In this review, our goal is to summarize the most recent evidence surrounding day 14 bone marrow biopsies that have been published and clarify the utility of this currently recommended practice. Here, we review major developments in mid-induction biopsy in AML, along with ongoing and future planned studies in this area, outlining the limitations of available data and our future goals.
Collapse
Affiliation(s)
- Omer Jamy
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Garrett Bourne
- Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Todd William Mudd
- Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Haley Thigpen
- Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ravi Bhatia
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
26
|
Tracy PD, Delgado R, Al-Akdi A, Pilichowska M, Zhou Z. Extramedullary immunoprivileged sites as a niche for residual and relapsed FLT3-ITD mutated AML: an unmet clinical need. Leuk Lymphoma 2025; 66:548-552. [PMID: 39607871 DOI: 10.1080/10428194.2024.2431880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024]
Affiliation(s)
- Philip D Tracy
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Ruben Delgado
- Department of Pathology, Tufts Medical Center, Boston, MA, USA
| | - Ahmad Al-Akdi
- Department of Pathology, Tufts Medical Center, Boston, MA, USA
| | | | - Zheng Zhou
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
27
|
Anabtawi N, Nicolet D, Alotaibi N, Buelow DR, Orwick S, Gregory T, Raj R, Coleman K, Kolitz JE, Powell BL, Blum WG, Baer MR, Byrd JC, Stock W, Uy GL, Mrózek K, Eisfeld AK, Cheng X, Baker SD, Blachly JS. Prognostic, biological, and structural implications of FLT3-JMD point mutations in acute myeloid leukemia: an analysis of Alliance studies. Leukemia 2025; 39:623-631. [PMID: 39806020 PMCID: PMC11879849 DOI: 10.1038/s41375-024-02498-y] [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: 11/20/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025]
Abstract
The FLT3 gene frequently undergoes mutations in acute myeloid leukemia (AML), with internal tandem duplications (ITD) and tyrosine kinase domain (TKD) point mutations (PMs) being most common. Recently, PMs and deletions in the FLT3 juxtamembrane domain (JMD) have been identified, but their biological and clinical significance remains poorly understood. We analyzed 1660 patients with de novo AML and found FLT3-JMD mutations, mostly PMs, in 2% of the patients. Patients with FLT3-JMD mutations had a higher relapse rate and shorter disease-free survival than those with FLT3-TKD, whereas their relapse rate, disease-free and overall survival were not significantly different from those of FLT3-ITD-positive patients. In vitro experiments showed that FLT3-JMD PMs transformed hematopoietic cells and responded well to type I and II FLT3 inhibitors. Molecular dynamics simulations were used to explore the conformational changes of JMD PMs relative to wild-type FLT3. These mutations exhibited constrained domain motions with wider gate openings, potentially enhancing drug binding. Altered residue interactions and structural changes shed light on their unique functional mechanisms, with increased allosteric pathways suggesting reduced interactions with other residues. We conclude that patients with FLT3-JMD PMs represent uncommon but important subset with distinct molecular and biological features, and may benefit from FLT3 inhibitors.
Collapse
Affiliation(s)
- Nadeen Anabtawi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Deedra Nicolet
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Alliance Statistics and Data Management Center, The Ohio State University, Columbus, OH, USA
| | - Najla Alotaibi
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Daelynn R Buelow
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Shelley Orwick
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Thomas Gregory
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ruchika Raj
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Kennedy Coleman
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Jonathan E Kolitz
- Monter Cancer Center, Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY, USA
- Monter Cancer Center, North Shore University Hospital and Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY, USA
| | - Bayard L Powell
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, NC, USA
| | - William G Blum
- Department of Hematology and Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Maria R Baer
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - John C Byrd
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wendy Stock
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Geoffrey L Uy
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Krzysztof Mrózek
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Ann-Kathrin Eisfeld
- The 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, Columbus, OH, USA
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Sharyn D Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA.
| | - James S Blachly
- The 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, Columbus, OH, USA.
| |
Collapse
|
28
|
Marshalek JP, Epistola R, Tomassetti S. Real-world treatment outcomes from a retrospective cohort of patients with acute myeloid leukemia from an urban safety net hospital. J Oncol Pharm Pract 2025; 31:182-189. [PMID: 38321873 PMCID: PMC11898379 DOI: 10.1177/10781552231225398] [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: 08/21/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024]
Abstract
IntroductionWhile continual advancements in acute myeloid leukemia have augmented response rates and survival, outcomes in clinical trials may not correlate with real-world practice as trials may underrepresent individuals with comorbidities, decreased performance status, and older age. Additionally, clinical trials may underrepresent certain ethnicities, and disparities based on ethnicity, socioeconomic status, and insurance have been demonstrated in acute myeloid leukemia.MethodsWe performed a retrospective chart review of adult patients with acute myeloid leukemia who were treated at Harbor-UCLA from 2014 to 2022 to examine patient characteristics, management patterns, and outcomes in a safety net hospital setting.ResultsThe median age was 56 years old (range 18-84). In regards to risk stratification, 22%, 33%, and 41% had favorable, intermediate, and adverse risk acute myeloid leukemia, respectively. The most common induction regimens included 7 + 3 (55%), azacitidine (10%), azacitidine + venetoclax (7%), and 7 + 3 + midostaurin (7%). The complete remission rate was 51%. Among patients who received intensive induction chemotherapy, 15% underwent re-induction with a second cycle, 51% received consolidation therapy, and 5% received maintenance therapy with a targeted agent. Overall, 12% of patients received allogeneic stem cell transplant. Median overall survival was 12.2 months, and 5-year overall survival was 18%.ConclusionsSuboptimal response rates and survival in this population may be related to low rates of re-induction and allogeneic transplant in addition to high rates of adverse cytogenetics, secondary acute myeloid leukemia, and supportive care only. Efforts to increase access to clinical trials, novel therapies, and transplants for diverse and underinsured populations are essential.
Collapse
Affiliation(s)
- Joseph P Marshalek
- Division of Hematology/Oncology, Department of Internal Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Raisa Epistola
- Division of Hematology/Oncology, Department of Internal Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sarah Tomassetti
- Division of Hematology/Oncology, Department of Internal Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
29
|
Han Yu P, Yan Zhang Z, Yuan Kang Y, Huang P, Yang C, Naranmandura H. Acute myeloid leukemia with t(8;21) translocation: Molecular pathogenesis, potential therapeutics and future directions. Biochem Pharmacol 2025; 233:116774. [PMID: 39864466 DOI: 10.1016/j.bcp.2025.116774] [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: 10/29/2024] [Revised: 12/18/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous and aggressive blood cancer. Genetic abnormalities, such as the t(8;21) rearrangement, play a significant role in AML onset. This rearrangement leads to the formation of the RUNX1/RUNX1T1 fusion protein, disrupting gene regulation and genomic stability, ultimately causing full-blown leukemia. Despite a generally favorable prognosis, t(8;21) patients face relapse and chemotherapy resistance, particularly when harboring cooperating mutations. While advances in cellular genetics and molecular biology have improved AML treatment, there are currently no specific targeted therapies against RUNX1/RUNX1T1. Therefore, investigating targeted therapies for this AML subtype holds promise for patients. This review explores the complex landscape of t(8;21) AML, unravels the molecular mechanisms of RUNX1/RUNX1T1-driven leukemogenesis, and discusses recent advancements in target therapies including small molecule drugs and PROTAC. Our goal is to develop more effective and less toxic strategies for managing t(8;21) AML patients.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Translocation, Genetic/genetics
- Chromosomes, Human, Pair 8/genetics
- Chromosomes, Human, Pair 21/genetics
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/metabolism
- RUNX1 Translocation Partner 1 Protein/genetics
- RUNX1 Translocation Partner 1 Protein/metabolism
- RUNX1 Translocation Partner 1 Protein/antagonists & inhibitors
- Antineoplastic Agents/therapeutic use
- Animals
Collapse
Affiliation(s)
- Pei Han Yu
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ze Yan Zhang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Yuan Kang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Huang
- Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Chang Yang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China.
| | - Hua Naranmandura
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
30
|
Boluda B, Rodriguez-Veiga R, Sargas C, Ayala R, Larráyoz MJ, Chillón MC, Soria-Saldise E, Bilbao C, de la Torre EP, Navarro I, Martinez-Cuadron D, Gil C, Bernal T, Bergua J, Algarra L, Tormo M, Martínez-Sanchez P, Carrillo-Cruz E, Serrano J, Alonso-Domínguez JM, García R, Amigo ML, Herrera-Puente P, Sayas MJ, Lavilla-Rubira E, García-Pérez MJ, Morán J, Pérez-Santaolalla E, Alonso-Vence N, Oliva A, López JA, Barrios M, García-Fortes M, Olave MT, Labrador J, Martínez-López J, Calasanz MJ, García-Sanz R, Pérez-Simón JA, Gómez-Casares MT, Sánchez-Garcia J, Mendizabal Y, Barragán E, Montesinos P. Conventional PCR Versus Next Generation Sequencing for Diagnosis of FLT3, IDH and NPM1 Mutations in Acute Myeloid Leukemia: Results of the PETHEMA PCR-LMA Study. Cancers (Basel) 2025; 17:854. [PMID: 40075701 PMCID: PMC11898636 DOI: 10.3390/cancers17050854] [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: 01/13/2025] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES This PETHEMA PCR-LMA study aimed to evaluate whether mutations detected by NGS (VAF cut-off of ≥5%) correlate with NPM1, FLT3-ITD, FLT3-TKD, IDH1, and IDH2 mutations detected using conventional PCR (analytical sensitivity 3%) in a nationwide network of seven reference laboratories. METHODS Between 2019 and 2021, 1685 adult AML patients with at least one centralized sample (NGS or PCR) at primary diagnosis or relapse/refractory episode were included. RESULTS During this period, 1288 paired NGS/PCR samples (1094 at diagnosis, 103 at relapse and 88 at refractoriness) were analyzed. Considering PCR the gold-standard, for NPM1 NGS sensitivity was 98.5% and specificity 98.9%, for FLT3-ITD 73.8% and 99.6%, for FLT3-TKD 84.5% and 99.3%, for IDH1 98.7% and 98.7%, and for IDH2 99.1% and 97.7%, respectively. Overall concordance rate of positive results between NGS (and PCR was 95% (262/276) for NPM1, 72% (149/206) for FLT3-ITD, 74% (49/66) for FLT3-TKD, 87% (77/89) for IDH1 and 84% (107/127) for IDH2. Overall, median days from sample reception until report were 7 for PCR and 28 for NGS. CONCLUSIONS This study shows high concordance between NPM1 and IDH results using PCR and NGS. However, sensible important discrepancies are observed for FLT3 mutations. In our context, rapid screening for these druggable mutations should be performed by conventional PCR.
Collapse
Affiliation(s)
- Blanca Boluda
- Hematology Department, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (R.R.-V.); (I.N.); (D.M.-C.); (Y.M.); (P.M.)
| | - Rebeca Rodriguez-Veiga
- Hematology Department, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (R.R.-V.); (I.N.); (D.M.-C.); (Y.M.); (P.M.)
| | - Claudia Sargas
- Molecular Biology Unit, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (C.S.); (E.B.)
| | - Rosa Ayala
- Hematology Department, Hospital Universitario 12 de Octubre, CNIO, Complutense University, 28041 Madrid, Spain; (R.A.); (P.M.-S.); (J.M.-L.)
| | - María J. Larráyoz
- CIMA LAB Diagnostics, Universidad de Navarra, 31008 Pamplona, Spain; (M.J.L.); (M.J.C.)
| | - María Carmen Chillón
- Hospital Universitario de Salamanca (HUS/IBSAL), CIBERONC and Center for Cancer Research-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.C.C.); (R.G.-S.)
| | - Elena Soria-Saldise
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina (IBIS/CSIC/CIBERONC), Universidad de Sevilla, 41013 Sevilla, Spain; (E.S.-S.); (E.C.-C.); (J.A.P.-S.)
| | - Cristina Bilbao
- Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Spain; (C.B.); (M.T.G.-C.)
| | - Esther Prados de la Torre
- IMIBIC, Hematology, Hospital Universitario Reina Sofía, UCO, 14004 Córdoba, Spain; (E.P.d.l.T.); (J.S.); (J.S.-G.)
| | - Irene Navarro
- Hematology Department, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (R.R.-V.); (I.N.); (D.M.-C.); (Y.M.); (P.M.)
| | - David Martinez-Cuadron
- Hematology Department, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (R.R.-V.); (I.N.); (D.M.-C.); (Y.M.); (P.M.)
- CIBERONC Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Gil
- Hospital General Universitario de Alicante, 03010 Alicante, Spain;
| | - Teresa Bernal
- Hospital Universitario Central de Asturias, Instituto Universitario (IUOPA), Instituto de Investigación del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Juan Bergua
- Hospital Universitario San Pedro de Alcántara, 10003 Cáceres, Spain;
| | - Lorenzo Algarra
- Hospital Universitario General de Albacete, 02008 Albacete, Spain;
| | - Mar Tormo
- Hematology Department, Hospital Clínico Universitario-INCLIVA, 46010 Valencia, Spain;
| | - Pilar Martínez-Sanchez
- Hematology Department, Hospital Universitario 12 de Octubre, CNIO, Complutense University, 28041 Madrid, Spain; (R.A.); (P.M.-S.); (J.M.-L.)
| | - Estrella Carrillo-Cruz
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina (IBIS/CSIC/CIBERONC), Universidad de Sevilla, 41013 Sevilla, Spain; (E.S.-S.); (E.C.-C.); (J.A.P.-S.)
| | - Josefina Serrano
- IMIBIC, Hematology, Hospital Universitario Reina Sofía, UCO, 14004 Córdoba, Spain; (E.P.d.l.T.); (J.S.); (J.S.-G.)
| | | | - Raimundo García
- Hospital Universitari General de Castelló, 12004 Castellón, Spain;
| | - Maria Luz Amigo
- Hospital Universitario Morales Messeguer, 300008 Murcia, Spain;
| | | | | | | | | | - Julia Morán
- Hospital U. Puerta del Mar, 11009 Cadiz, Spain;
| | | | | | - Ana Oliva
- Hospital Universitario Nuestra Señora de Candelaria, 38010 Tenerife, Spain;
| | | | | | | | | | - Jorge Labrador
- Department of Hematology, Hospital University Burgos, 09006 Burgos, Spain;
| | - Joaquín Martínez-López
- Hematology Department, Hospital Universitario 12 de Octubre, CNIO, Complutense University, 28041 Madrid, Spain; (R.A.); (P.M.-S.); (J.M.-L.)
| | - María J. Calasanz
- CIMA LAB Diagnostics, Universidad de Navarra, 31008 Pamplona, Spain; (M.J.L.); (M.J.C.)
| | - Ramón García-Sanz
- Hospital Universitario de Salamanca (HUS/IBSAL), CIBERONC and Center for Cancer Research-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (M.C.C.); (R.G.-S.)
| | - José A. Pérez-Simón
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina (IBIS/CSIC/CIBERONC), Universidad de Sevilla, 41013 Sevilla, Spain; (E.S.-S.); (E.C.-C.); (J.A.P.-S.)
| | - María T. Gómez-Casares
- Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Spain; (C.B.); (M.T.G.-C.)
| | - Joaquín Sánchez-Garcia
- IMIBIC, Hematology, Hospital Universitario Reina Sofía, UCO, 14004 Córdoba, Spain; (E.P.d.l.T.); (J.S.); (J.S.-G.)
| | - Yolanda Mendizabal
- Hematology Department, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (R.R.-V.); (I.N.); (D.M.-C.); (Y.M.); (P.M.)
| | - Eva Barragán
- Molecular Biology Unit, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (C.S.); (E.B.)
- CIBERONC Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pau Montesinos
- Hematology Department, Hospital Universitari i Politécnic-IIS La Fe, 46026 Valencia, Spain; (R.R.-V.); (I.N.); (D.M.-C.); (Y.M.); (P.M.)
- CIBERONC Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
31
|
Costa A, Scalzulli E, Carmosino I, Ielo C, Bisegna ML, Martelli M, Breccia M. Clinical and biological advances of critical complications in acute myeloid leukemia. Leuk Lymphoma 2025; 66:400-419. [PMID: 39582141 DOI: 10.1080/10428194.2024.2425051] [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: 06/12/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/26/2024]
Abstract
Managing acute myeloid leukemia (AML) and its critical complications requires understanding the complex interplay between disease biology, treatment strategies, and patient characteristics. Complications like sepsis, acute respiratory failure (ARF), hyperleukocytosis, coagulopathy, tumor lysis syndrome (TLS) and central nervous system (CNS) involvement present unique challenges needing precise evaluation and tailored interventions. Venetoclax-induced TLS and differentiation syndrome (DS) from IDH1/IDH2 or menin inhibitors highlight the need for ongoing research and innovative approaches. As the microbiological landscape evolves and new therapeutic agents emerge, adapting strategies to mitigate harmful pharmacological interactions is crucial. Advances in understanding the genetic profiles of patients with hyperleukocytosis contribute to better-targeted therapeutic strategies. Effective AML management relies on collaborative efforts from hematologists, specialized services, and intensive care units (ICUs). This review analyzes recent data on critical AML complications, identifies areas for further investigation, and proposes ways to advance clinical research and enhance patient care strategies.
Collapse
Affiliation(s)
- Alessandro Costa
- Hematology Unit, Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Emilia Scalzulli
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Ida Carmosino
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Claudia Ielo
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Maria Laura Bisegna
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Maurizio Martelli
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| |
Collapse
|
32
|
Rao M, Luo W, Luo C, Wu B, Xu T, Wei Z, Deng H, Li K, Zhou D. Prognostic factors and outcomes in pediatric acute myeloid leukemia: a comprehensive bibliometric analysis of global research trends. Front Oncol 2025; 15:1466818. [PMID: 40034590 PMCID: PMC11873564 DOI: 10.3389/fonc.2025.1466818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pediatric AML prognosis research has advanced significantly, yet gaps in understanding genetic and molecular interactions persist. Despite improved outcomes, relapse/refractory cases and personalized treatment integration remain critical clinical challenges. Objective To analyze the global research landscape on pediatric AML prognosis, highlight influential components and collaborations, and identify major potential research trends. Methods Publications on pediatric AML prognosis research from 1999 to 2023 were retrieved from the Clarivate Analytics Web of Science Core Collection (WoSCC) database. Bibliometric analysis was conducted using CiteSpace and VOSviewer to identify leading countries, prominent institutions, high-impact journals, key research categories, influential authors, and emerging research topics. Results The bibliometric analysis encompassed 924 publications, with St. Jude Children's Research Hospital emerging as the most prolific institution. The United States leads globally in terms of countries, institutions, journals, and authors. Todd A. Alonzo ranks highest in publication volume, while U. Creutzig leads in citations. The top research categories were Oncology, Hematology, and Pediatrics. Key research topics included genomics, transcriptomics, epigenomics, targeted therapies, immune therapy, and integrative diagnostic approaches. Conclusion This bibliometric analysis highlights significant advancements in pediatric AML prognosis over the past 25 years, driven by the integration of genetic markers, immunological insights, transcriptomics, and epigenomics, which have collectively transformed risk stratification and treatment strategies. Overcoming challenges, such as discovering new therapeutic targets and enhancing treatment combinations, will depend on global collaboration and advanced technologies to propel the field forward.
Collapse
Affiliation(s)
- Mingliang Rao
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenna Luo
- Department of Laboratory Medicine, Heyuan People’s Hospital, Heyuan, China
| | - Caiju Luo
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baojing Wu
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tiantian Xu
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ziqian Wei
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haolan Deng
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kejing Li
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dunhua Zhou
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Klauer LK, Rejeski HA, Ugur S, Rackl E, Abdulmajid J, Fischer Z, Pepeldjiyska E, Frischhut A, Schmieder N, Völker A, Rank A, Schmid C, Schmohl J, Amberger DC, Schmetzer HM. Leukemia-Derived Dendritic Cells Induce Anti-Leukemic Effects Ex Vivo in AML Independently of Patients' Clinical and Biological Features. Int J Mol Sci 2025; 26:1700. [PMID: 40004163 PMCID: PMC11855365 DOI: 10.3390/ijms26041700] [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: 12/25/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
New therapies are highly needed to stabilize remission in patients with acute myeloid leukemia (AML). This study investigates the value of dendritic cells derived from leukemic blasts (DCleu) to enhance anti-leukemic immunity after T-cell-enriched mixed lymphocyte cultures (MLCs). We correlated induced anti-leukemic activity with patient data, including biological, clinical and prognostic factors. Additionally, we correlated the frequencies of DC/DCleu and leukemic-specific T cells with the achieved anti-leukemic activity after MLC. We show that mature DC/DCleu can be generated using the immunomodulating Kit-M, which contains granulocyte-macrophage colony-stimulating-factor (GM-CSF) and prostaglandin E1 (PGE1), without inducing blast proliferation from leukemic whole blood (WB) samples. Activated leukemia-specific immune and memory cells increased after MLC with Kit-M-pretreated WB, leading to improved blast lysis. Enhanced anti-leukemic activity positively correlated with the frequencies of generated DC/DCleu, proliferating leukemic-specific T cells and memory T cells, but not with leukemic blast counts, hemoglobin levels or platelet counts at diagnosis. No correlation was found between improved blast lysis and patients' prognostic data, including age, gender, ELN risk groups, disease stage and response to induction chemotherapy. These findings underscore the potential of DC/DCleu to evoke robust immune responses and potential immunological memory against AML. Overall, this innovative approach could pave the way for the development of improved immunotherapeutic strategies that function in vivo.
Collapse
Affiliation(s)
- Lara Kristina Klauer
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Hazal Aslan Rejeski
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Selda Ugur
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Elias Rackl
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Joudi Abdulmajid
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
- Faculty of Biology, University Bielefeld, 33615 Bielefeld, Germany
| | - Zuzanna Fischer
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Elena Pepeldjiyska
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Annalena Frischhut
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Nicolas Schmieder
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| | - Antje Völker
- Department of Statistics, Ludwig-Maximilian-University Munich, 80539 Munich, Germany
| | - Andreas Rank
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
- Department of Haematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Christoph Schmid
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
- Department of Haematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Jörg Schmohl
- Department of Haematology and Oncology, Diakonie-Klinikum, 70176 Stuttgart, Germany
| | - Daniel Christoph Amberger
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- First Department of Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Helga Maria Schmetzer
- Department of Medicine III, University Hospital of Ludwig-Maximilian-University Munich, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), 80539 Munich, Germany
| |
Collapse
|
34
|
Lazana I, Papathanassiou M, Konstantellos I, Tzenou T, Kopsaftopoulou A, Liga M, Violaki V, Kyriazopoulou L, Gkirkas K, Papalexandri A, Plata E, Michalis E, Leonidopoulou T, Garofalaki M, Sioni A, Tziotziou I, Lalayanni C, Kiousiafes D, Vassilakopoulos TP, Kapsali E, Spyridonidis A, Baltadakis I, Angelopoulou M, Sakellari I, Tsirigotis P. The Prognostic Impact of Additional Molecular and Cytogenetic Abnormalities on AML Patients with NPM1- and/or FLT3-ITD Mutations Receiving Intensive Chemotherapy: Real-World Data from the Greek Registry. Cancers (Basel) 2025; 17:667. [PMID: 40002262 PMCID: PMC11853380 DOI: 10.3390/cancers17040667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objective: The prognostic impact of additional cytogenetic aberrations and molecular abnormalities (such as MDS-related mutations, mutations in myeloid genes and the KRAS/NRAS mutations) in patients with NPM1- and/or FLT3-ITD-mutated AML remains elusive. Methods: This retrospective, multicentre study of real-world data aimed to investigate the impact of these mutations and cytogenetic abnormalities on the prognosis of patients with NPM1- and/or FLT3-ITD-mutated AML, treated with intensive chemotherapy. Results: In a cohort of 161 patients, the only parameters identified to affect the outcomes (EFS and OS) were the age of the patient, primary refractory disease, the presence of a NPM1 mutation and the use of allogenic stem cell transplantation (allo-SCT) within the first complete remission. More specifically, ages below the median conferred significantly improved outcomes, whereas primary refractory disease exhibited a negative correlation with the EFS and OS. Subsequent subgroup analysis, stratifying patients into three groups (Group 1: NPM1mutated/FLT3wt; Group 2: NPM1mutated/FLT3mutated; Group 3: NPM1wt/FLT3mutated). revealed that allo-SCT in CR1 improved the outcomes (EFS and OS) in Groups 2 and 3, but had no additional impact in Group 1. Conclusions: Age, primary refractory disease and allogenic stem cell transplantation in the first complete response were found to have a prognostic impact on outcomes, Interestingly, no significant association was detected between the poor prognostic cytogenetic abnormalities or the presence of additional mutations in myeloid genes, MDS-related genes or KRAS/NRAS genes and the outcomes in any group of patients.
Collapse
Affiliation(s)
- Ioanna Lazana
- Hematology Division, 2nd Department of Internal Medicine, “ATTIKON” General University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.L.); (I.K.); (K.G.)
| | - Maria Papathanassiou
- Hematology and Bone Marrow Transplantation Department, Papanikolaou General Hospital, 56403 Thessaloniki, Greece; (M.P.); (A.P.); (C.L.); (I.S.)
| | - Ioannis Konstantellos
- Hematology Division, 2nd Department of Internal Medicine, “ATTIKON” General University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.L.); (I.K.); (K.G.)
| | - Tatiana Tzenou
- Hematology-Lymphoma and Bone Marrow Transplantation Department, Evaggelismos General Hospital, 10676 Athens, Greece; (T.T.); (M.G.); (I.T.); (I.B.)
| | - Anastasia Kopsaftopoulou
- Hematology and Bone Marrow Transplantation Department, LAIKON General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (E.P.); (T.P.V.); (M.A.)
| | - Maria Liga
- Hematology and Bone Marrow Transplantation Department, University Hospital of Patras, Rio, 26504 Patras, Greece; (M.L.); (A.S.)
| | - Vasiliki Violaki
- Department of Hematology, General Hospital of Chania, 73300 Chania, Greece;
| | - Lydia Kyriazopoulou
- Department of Hematology, University Hospital of Ioannina, 45500 Ioannina, Greece; (L.K.); (E.K.)
| | - Konstantinos Gkirkas
- Hematology Division, 2nd Department of Internal Medicine, “ATTIKON” General University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.L.); (I.K.); (K.G.)
| | - Apostolia Papalexandri
- Hematology and Bone Marrow Transplantation Department, Papanikolaou General Hospital, 56403 Thessaloniki, Greece; (M.P.); (A.P.); (C.L.); (I.S.)
| | - Eleni Plata
- Hematology and Bone Marrow Transplantation Department, LAIKON General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (E.P.); (T.P.V.); (M.A.)
| | - Evrydiki Michalis
- Department of Hematology, Genimatas General Hospital, 11527 Athens, Greece;
| | - Theoni Leonidopoulou
- Department of Hematology, Sismanogleion General Hospital, 15126 Athens, Greece; (T.L.); (D.K.)
| | - Maria Garofalaki
- Hematology-Lymphoma and Bone Marrow Transplantation Department, Evaggelismos General Hospital, 10676 Athens, Greece; (T.T.); (M.G.); (I.T.); (I.B.)
| | - Anastasia Sioni
- Department of Hematology, Agios Savvas Cancer Center, 11522 Athens, Greece;
| | - Irene Tziotziou
- Hematology-Lymphoma and Bone Marrow Transplantation Department, Evaggelismos General Hospital, 10676 Athens, Greece; (T.T.); (M.G.); (I.T.); (I.B.)
| | - Chrysavgi Lalayanni
- Hematology and Bone Marrow Transplantation Department, Papanikolaou General Hospital, 56403 Thessaloniki, Greece; (M.P.); (A.P.); (C.L.); (I.S.)
| | - Dimitrios Kiousiafes
- Department of Hematology, Sismanogleion General Hospital, 15126 Athens, Greece; (T.L.); (D.K.)
| | - Theodoros P. Vassilakopoulos
- Hematology and Bone Marrow Transplantation Department, LAIKON General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (E.P.); (T.P.V.); (M.A.)
| | - Eleni Kapsali
- Department of Hematology, University Hospital of Ioannina, 45500 Ioannina, Greece; (L.K.); (E.K.)
| | - Alexandros Spyridonidis
- Hematology and Bone Marrow Transplantation Department, University Hospital of Patras, Rio, 26504 Patras, Greece; (M.L.); (A.S.)
| | - Ioannis Baltadakis
- Hematology-Lymphoma and Bone Marrow Transplantation Department, Evaggelismos General Hospital, 10676 Athens, Greece; (T.T.); (M.G.); (I.T.); (I.B.)
| | - Maria Angelopoulou
- Hematology and Bone Marrow Transplantation Department, LAIKON General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (E.P.); (T.P.V.); (M.A.)
| | - Ioanna Sakellari
- Hematology and Bone Marrow Transplantation Department, Papanikolaou General Hospital, 56403 Thessaloniki, Greece; (M.P.); (A.P.); (C.L.); (I.S.)
| | - Panagiotis Tsirigotis
- Hematology Division, 2nd Department of Internal Medicine, “ATTIKON” General University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.L.); (I.K.); (K.G.)
| |
Collapse
|
35
|
Gang M, Othus M, Walter RB. Significance of Measurable Residual Disease in Patients Undergoing Allogeneic Hematopoietic Cell Transplantation for Acute Myeloid Leukemia. Cells 2025; 14:290. [PMID: 39996762 PMCID: PMC11853423 DOI: 10.3390/cells14040290] [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: 01/21/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) remains an important curative-intent treatment for many patients with acute myeloid leukemia (AML), but AML recurrence after allografting is common. Many factors associated with relapse after allogeneic HCT have been identified over the years. Central among these is measurable ("minimal") residual disease (MRD) as detected by multiparameter flow cytometry, quantitative polymerase chain reaction, and/or next-generation sequencing. Demonstration of a strong, independent prognostic role of pre- and early post-HCT MRD has raised hopes MRD could also serve as a predictive biomarker to inform treatment decision-making, with emerging data indicating the potential value to guide candidacy assessment for allografting as a post-remission treatment strategy, the selection of conditioning intensity, use of small molecule inhibitors as post-HCT maintenance therapy, and preemptive infusion of donor lymphocytes. Monitoring for leukemia recurrence after HCT and surrogacy for treatment response are other considerations for the clinical use of MRD data. In this review, we will outline the current landscape of MRD as a biomarker for patients with AML undergoing HCT and discuss areas of uncertainty and ongoing research.
Collapse
Affiliation(s)
- Margery Gang
- Hematology and Oncology Fellowship Program, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA 98109, USA;
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA;
| | - Roland B. Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
36
|
Gyan E, Minden MD, Kubo K, Rambaldi A, Juliusson G, Jädersten M, Kelly RJ, Szerafin L, He W, Gill SC, Hill JE, Chen C, Delgado D, Hasabou N. Maintenance therapy with the FMS-like tyrosine kinase 3 inhibitor gilteritinib in patients with FMS-like tyrosine kinase 3-internal tandem duplication acute myeloid leukemia: A phase 2 study. Cancer 2025; 131:e35746. [PMID: 39945223 PMCID: PMC11822735 DOI: 10.1002/cncr.35746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/05/2024] [Accepted: 12/09/2024] [Indexed: 02/16/2025]
Abstract
BACKGROUND The GOSSAMER phase 2 study assessed the FMS-like tyrosine kinase 3 (FLT3) inhibitor gilteritinib as maintenance therapy in patients with FLT3-internal tandem duplication (FLT3-ITD) acute myeloid leukemia (AML) in first complete remission without previous hematopoietic stem cell transplantation (HSCT). METHODS Patients had to be within 2 months of their last consolidation cycle and have completed the recommended number of cycles per local practice. FLT3 inhibitors were allowed only during induction and/or consolidation. The primary end point was relapse-free survival (RFS). Secondary end points included overall survival (OS), event-free survival, and measurable residual disease (MRD). RESULTS In total, 98 patients were randomized (gilteritinib, n = 63; placebo, n = 35). RFS was not significantly different between the arms (hazard ratio, 0.74; 95% confidence interval, 0.41-1.34; p = .16). RFS rates for the gilteritinib and placebo arms were 68.5% and 55.3% at 1 year, 51.8% and 44.9% at 2 years, and 41.2% and 40.8% at 3 years, respectively. OS was not significantly different between the arms but may have been affected by subsequent AML therapies after discontinuation. In patients who received subsequent therapy (gilteritinib, 46.8%; placebo, 60.0%), a higher percentage of placebo-treated (57.1%) versus gilteritinib-treated patients (27.6%) underwent HSCT. At the end of treatment, 96.4% of gilteritinib-treated and 85.7% of placebo-treated patients had undetectable MRD. Relapsed placebo-treated (86.7%) versus gilteritinib-treated patients (34.8%) had a greater FLT3 mutational burden. No new significant safety concerns were noted. CONCLUSIONS The primary end point was not achieved; however, an observed trend toward potential benefit was noted in patients with FLT3-ITD AML who had not undergone prior HSCT.
Collapse
Affiliation(s)
- Emmanuel Gyan
- Department of Hematology and Cell TherapyUniversity Hospital of ToursUMR Institut National de la Santé et de la Recherche Médicale (INSERM) 1079 N2COx, CIC INSERM U1415ToursFrance
| | - Mark D. Minden
- Department of Medical Oncology and HematologyPrincess Margaret HospitalTorontoOntarioCanada
| | - Kohmei Kubo
- Department of HematologyAomori Prefectural Central HospitalAomoriJapan
| | - Alessandro Rambaldi
- Department of Oncology‐HematologyUniversity of Milan and Azienda Socio Sanitaria Territoriale Papa Giovanni XXIIIBergamoItaly
| | | | - Martin Jädersten
- Department of HematologyKarolinska University HospitalStockholmSweden
- Department of Medicine, HuddingeCenter for Hematology and Regenerative MedicineKarolinska InstitutetStockholmSweden
| | | | - László Szerafin
- Department of HematologyHospitals of County Szabolcs‐Szatmár‐Bereg and University Teaching HospitalNyíregyházaHungary
| | - Wensheng He
- Medical & DevelopmentAstellas Pharma Inc.NorthbrookIllinoisUSA
| | - Stanley C. Gill
- Medical & DevelopmentAstellas Pharma Inc.NorthbrookIllinoisUSA
| | - Jason E. Hill
- Medical & DevelopmentAstellas Pharma Inc.NorthbrookIllinoisUSA
| | - Caroline Chen
- Medical & DevelopmentAstellas Pharma Inc.NorthbrookIllinoisUSA
| | - David Delgado
- Medical & DevelopmentAstellas Pharma Inc.NorthbrookIllinoisUSA
| | - Nahla Hasabou
- Medical & DevelopmentAstellas Pharma Inc.NorthbrookIllinoisUSA
| |
Collapse
|
37
|
Mezzanotte V, Paterno G, Cerroni I, De Marchi L, Taka K, Buzzatti E, Mallegni F, Meddi E, Moretti F, Buccisano F, Maurillo L, Palmieri R, Gurnari C, Venditti A, Del Principe MI. Use of Primary Prophylaxis with G-CSF in Acute Myeloid Leukemia Patients Undergoing Intensive Chemotherapy Does Not Affect Quality of Response. J Clin Med 2025; 14:1254. [PMID: 40004785 PMCID: PMC11856925 DOI: 10.3390/jcm14041254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The objective of our study was to evaluate the safety and efficacy of granulocyte colony-stimulating factor (G-CSF) as primary prophylaxis in adult patients with acute myeloid leukemia (AML) undergoing intensive chemotherapy. Methods: We retrospectively analyzed 112 AML patients treated with intensive chemotherapy at Fondazione Policlinico Tor Vergata in Rome between January 2014 and March 2024. Patients were divided into G-CSF and non-G-CSF (nG-CSF) groups. We assessed the incidence of neutropenia, its severity and duration; duration of hospitalization and its costs; incidence of febrile neutropenia (FN) and septic shock; duration of antibiotic therapy (ABT) and antifungal therapy (AFT); complete remission (CR) rates; measurable residual disease (MRD) status; relapse rates; and outcomes. Results: G-CSF administration significantly reduced the duration of neutropenia (median 14 vs. 18 days, p < 0.05) and length of hospitalization (median 28 vs. 35 days, p < 0.05), in both induction and consolidation therapy. There were no significant differences in CR rates (73% vs. 67%, p = 0.64), MRD negativity achievement (52% vs. 48%, p = 0.68), leukemia relapse rates (43% vs. 62%, p = 0.14), or overall survival (OS) (median 16.7 vs. 12.3 months, p = 0.3) between G-CSF and nG-CSF groups. Thanks to a shorter hospitalization, the use of G-CSF led to €300,000 in savings over the last 4 years. Conclusions: Our findings support the safety of G-CSF in AML patients, demonstrating no adverse impact on treatment response. G-CSF abbreviated the duration of neutropenia and hospitalization, highlighting its potential clinical and cost-effective role in AML treatment.
Collapse
Affiliation(s)
- Valeria Mezzanotte
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | | | - Ilaria Cerroni
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Lucrezia De Marchi
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Kristian Taka
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Elisa Buzzatti
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Flavia Mallegni
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Elisa Meddi
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Federico Moretti
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Francesco Buccisano
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Luca Maurillo
- Hematology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Raffaele Palmieri
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Carmelo Gurnari
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| | - Maria Ilaria Del Principe
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (V.M.); (I.C.); (L.D.M.); (K.T.); (E.B.); (F.M.); (E.M.); (F.M.); (F.B.); (R.P.); (C.G.); (M.I.D.P.)
| |
Collapse
|
38
|
Wei BH, Tsai XCH, Sun KJ, Lo MY, Hung SY, Chou WC, Tien HF, Hou HA, Chen CY. Annotation-free deep learning for predicting gene mutations from whole slide images of acute myeloid leukemia. NPJ Precis Oncol 2025; 9:35. [PMID: 39900774 PMCID: PMC11791072 DOI: 10.1038/s41698-025-00804-0] [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: 07/31/2024] [Accepted: 01/02/2025] [Indexed: 02/05/2025] Open
Abstract
The rapid development of deep learning has revolutionized medical image processing, including analyzing whole slide images (WSIs). Despite the demonstrated potential for characterizing gene mutations directly from WSIs in certain cancers, challenges remain due to image resolution and reliance on manual annotations for acute myeloid leukemia (AML). We, therefore, propose a deep learning model based on multiple instance learning (MIL) with ensemble techniques to predict gene mutations from AML WSIs. Our model predicts NPM1 mutations and FLT3-ITD without requiring patch-level or cell-level annotations. Using a dataset of 572 WSIs, the largest database with both WSI and genetic mutation information, our model achieved an AUC of 0.90 ± 0.08 for NPM1 and 0.80 ± 0.10 for FLT3-ITD in the testing cohort. Additionally, we found that blasts are pivotal indicators for gene mutation predictions, with their proportions varying between mutated and standard WSIs, highlighting the clinical potential of AML WSI analysis.
Collapse
Affiliation(s)
- Bo-Han Wei
- Center for Advanced Computing and Imaging in Biomedicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
- Department of Biomechatronics Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Xavier Cheng-Hong Tsai
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No.1, Changde St., Zhongzheng Dist., Taipei, 100229, Taiwan
- Department of Medical Education and Research, National Taiwan University Hospital Yunlin Branch, No. 579, Sec. 2, Yunlin Rd., Douliu City, Yunlin County, 640203, Taiwan
- Department of Hematological Oncology, National Taiwan University Cancer Center, No.57, Ln. 155, Sec. 3, Keelung Rd., Da'an Dist., Taipei City, 106, Taiwan
| | - Kuo-Jui Sun
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No.1, Changde St., Zhongzheng Dist., Taipei, 100229, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, No.1, Changde St., Zhongzheng Dist., Taipei, 100229, Taiwan
| | - Min-Yen Lo
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, No. 579, Sec. 2, Yunlin Rd., Douliu City, Yunlin County, 640203, Taiwan
| | - Sheng-Yu Hung
- Department of Hematological Oncology, National Taiwan University Cancer Center, No.57, Ln. 155, Sec. 3, Keelung Rd., Da'an Dist., Taipei City, 106, Taiwan
| | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No.1, Changde St., Zhongzheng Dist., Taipei, 100229, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, No.1, Changde St., Zhongzheng Dist., Taipei, 100229, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No.1, Changde St., Zhongzheng Dist., Taipei, 100229, Taiwan
- Department of Internal Medicine, Far-Eastern Memorial Hospital, New Taipei City, Taiwan, No. 21, Section 2, Nanya S. Road, Banqiao District, New Taipei City, 220, Taiwan
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, No.1, Changde St., Zhongzheng Dist., Taipei, 100229, Taiwan
| | - Chien-Yu Chen
- Center for Advanced Computing and Imaging in Biomedicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.
- Department of Biomechatronics Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.
- Genome and Systems Biology Degree Program, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.
- Smart Medicine and Health Informatics Program, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.
- Center for Computational and Systems Biology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.
| |
Collapse
|
39
|
Yu S, Yang S, Hu L, Duan W, Zhao T, Qin Y, Wang Y, Lai Y, Shi H, Tang F, Sun Y, Jia J, Wang J, Lu S, Fu Q, Jiang H, Xu L, Wang Y, Zhang X, Huang X, Jiang Q. Genetic abnormalities predict outcomes in patients with core binding factor acute myeloid leukemia. Ann Hematol 2025; 104:997-1006. [PMID: 39966122 PMCID: PMC11971224 DOI: 10.1007/s00277-024-06182-0] [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: 08/05/2024] [Accepted: 12/31/2024] [Indexed: 02/20/2025]
Abstract
Research on the comprehensive integration of clinical and genomic characteristics in patients with core binding factor acute myeloid leukemia (CBF-AML) is limited. Clinical and genomic data from consecutive patients with CBF-AML were reviewed. A Cox regression model was used to identify the variables associated with event-free survival (EFS), relapse-free survival (RFS) and overall survival (OS). A total of 346 CBF-AML patients (211 with RUNX1::RUNX1T1 and 135 with CBFB::MYH11) were included in this study. In the RUNX1::RUNX1T1 cohort, multivariate analyses revealed that KDM6A mutations were significantly associated with poor RFS (hazard ratio = 3.1 [1.4, 7.1], p = 0.007) and OS (HR = 11.5 [3.6, 37.0], p < 0.001); FLT3-TKD mutations, poor OS (HR = 4.9 [1.7, 14.3], p = 0.004); KIT mutation VAF > 25%, poor RFS (KITwt as ref, HR = 2.5 [1.1, 5.3], p = 0.022); ASXL1 mutations, favorable EFS (HR = 0.4 [0.2, 0.9], p = 0.016) and OS (HR = 0.2 [0.03, 0.8], p = 0.028). In the CBFB::MYH11 cohort, multivariate analyses revealed that a high mutation burden was significantly associated with inferior OS (HR = 1.4 [1.1, 1.8], p = 0.018); FLT3-ITD mutations, inferior OS (HR = 6.8 [1.3, 36.0], p = 0.024). In addition, increasing age, nonintensive chemotherapy, and high MRD levels predict poor outcomes in the RUNX1::RUNX1T1 cohort. In addition to the adverse impact of high KIT mutation burden and FLT3-ITD or FLT3-TKD mutations on prognosis in CBF-AML, KDM6A mutations predicted poor outcomes in patients with RUNX1::RUXN1T1; however, ASXL1 mutations, favourable outcomes; high mutation burden, poor outcomes in those with CBFB::MYH11.
Collapse
Affiliation(s)
- Shunjie Yu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Sen Yang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Lijuan Hu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Wenbing Duan
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Ting Zhao
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Yazhen Qin
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Yazhe Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Yueyun Lai
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Hongxia Shi
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Feifei Tang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Yuqian Sun
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Jinsong Jia
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Jing Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Shengye Lu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Qiang Fu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Hao Jiang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Lanping Xu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Yu Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Xiaohui Zhang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
| | - Xiaojun Huang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Qian Jiang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University, No. 11 Xizhimen South St, Beijing, China.
- Peking University People's Hospital, Qingdao, China.
| |
Collapse
|
40
|
Xiong L, Beyer D, Liu N, Lehmann T, Neugebauer S, Schaeuble S, Sommerfeld O, Ernst P, Svensson CM, Nietzsche S, Scholl S, Bruns T, Gaßler N, Gräler MH, Figge MT, Panagiotou G, Bauer M, Press AT. Targeting protein kinase C-α prolongs survival and restores liver function in sepsis: Evidence from preclinical models. Pharmacol Res 2025; 212:107581. [PMID: 39761839 DOI: 10.1016/j.phrs.2025.107581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
Sepsis is a life-threatening organ failure resulting from a poorly regulated infection response. Organ dysfunction includes hepatic involvement, weakening the immune system due to excretory liver failure, and metabolic dysfunction, increasing the death risk. Although experimental studies correlated excretory liver functionality with immune performance and survival rates in sepsis, the proteins and pathways involved remain unclear. This study identified protein kinase C-α (PKCα) as a novel target for managing excretory liver function during sepsis. Using a preclinical murine sepsis model, we found that both PKCα knockout and the use of a PKCα-inhibitor midostaurin successfully restored liver function without hindering the host's response or ability to clear the pathogen, highlighting PKCα's vital role in excretory liver failure. In septic animals, both approaches significantly boosted survival rates. Midostaurin is the clinically approved active pharmaceutical ingredient in Rydapt, approved for the adjuvant treatment of FTL3-mutated AML. Here, it reduced plasma bile acids and related inflammation in those patients, opening a translational avenue for therapeutics in sepsis. Conclusively, our research underscores the significance of PKCα in controlling excretory liver function during inflammation. This suggests that targeting this protein could restore liver function without compromising the immune system, thereby decreasing sepsis mortality and supporting the recent paradigm that the liver is a hub for the host response to infection that might, in the future, result in novel host-directed therapies supporting the current state-of-the-art intensive care medicine in patients with sepsis-associated liver failure.
Collapse
Affiliation(s)
- Ling Xiong
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Dustin Beyer
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Na Liu
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Tina Lehmann
- Jena University Hospital, Electron Microscopy Center, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sophie Neugebauer
- Jena University Hospital, Institute of Clinical Chemistry and Laboratory Diagnostics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sascha Schaeuble
- Department of Microbiome Dynamics at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Oliver Sommerfeld
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Philipp Ernst
- Jena University Hospital, Clinic for Internal Medicine II, Department of Hematology and Internal Oncology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Carl-Magnus Svensson
- Research Group Applied Systems Biology at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Sandor Nietzsche
- Jena University Hospital, Electron Microscopy Center, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sebastian Scholl
- Jena University Hospital, Clinic for Internal Medicine II, Department of Hematology and Internal Oncology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Tony Bruns
- University Hospital RWTH Aachen, Department of Medicine III, Aachen, Germany
| | - Nikolaus Gaßler
- Jena University Hospital, Section of Pathology, Institute of Forensic Medicine, Friedrich-Schiller-University Jena, Jena, Germany
| | - Markus H Gräler
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Marc Thilo Figge
- Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany; Research Group Applied Systems Biology at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany; Friedrich-Schiller-University Jena, Institute of Microbiology, Faculty of Biological Sciences, Jena, Germany; Friedrich-Schiller-University Jena, Cluster of Excellence Balance of the Microverse, Jena, Germany
| | - Gianni Panagiotou
- Department of Microbiome Dynamics at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany; Friedrich-Schiller-University Jena, Institute of Microbiology, Faculty of Biological Sciences, Jena, Germany; Friedrich-Schiller-University Jena, Cluster of Excellence Balance of the Microverse, Jena, Germany
| | - Michael Bauer
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Adrian T Press
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany; Friedrich-Schiller-University Jena, Faculty of Medicine, Jena, Germany.
| |
Collapse
|
41
|
Huo W, Shen Y, Huang J, Yang Y, Fan S, Zhao X, Wen Q, Wang L, Jiang C, Cao Y, Mo X, Xu Y, Hu X. Allogeneic hematopoietic stem cell transplantation could overcome the poor prognosis of DNMT3A mutNPM1 mutFLT3-ITD mut in acute myeloid leukemia: real-world multicenter analysis in China. Front Med 2025; 19:90-100. [PMID: 39643798 DOI: 10.1007/s11684-024-1091-5] [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: 02/22/2024] [Accepted: 05/17/2024] [Indexed: 12/09/2024]
Abstract
The cooccurrence of NPM1, FLT3-ITD, and DNMT3A mutations (i.e., triple mutation) is related to dismal prognosis in patients with acute myeloid leukemia (AML) receiving chemotherapy alone. In this multicenter retrospective cohort study, we aimed to identify whether allogeneic hematopoietic stem cell transplantation (allo-HSCT) could overcome the poor prognosis of DNMT3AmutNPM1mutFLT3-ITDmut AML across four transplant centers in China. Fifty-three patients with triple-mutated AML receiving allo-HSCT in complete remission were enrolled. The 1.5-year probabilities of relapse, leukemia-free survival, and overall survival after allo-HSCT were 11.9%, 80.3%, and 81.8%, respectively. Multivariate analysis revealed that more than one course of induction chemotherapy and allo-HSCT beyond CR1 were associated with poor survival. To our knowledge, this work is the largest study to explore the up-to-date undefined role of allo-HSCT in patients with triple-mutated AML. Our real-world data suggest that allo-HSCT could overcome the poor prognosis of DNMT3AmutNPM1mutFLT3-ITDmut in AML.
Collapse
Affiliation(s)
- Wenxuan Huo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Yifan Shen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215006, China
| | - Jiayu Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yang Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuang Fan
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Xiaosu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, 2019RU029, Beijing, 10044, China
| | - Qi Wen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Luxiang Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chuanhe Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaodong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China.
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, 2019RU029, Beijing, 10044, China.
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China.
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215006, China.
| | - Xiaoxia Hu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
42
|
Zhou Q, Guan Y, Zhao P, Chu H, Xi Y. Combined anti-leukemic effect of gilteritinib and GSK-J4 in FLT3-ITD + acute myeloid leukemia. Transl Oncol 2025; 52:102271. [PMID: 39813767 PMCID: PMC11783125 DOI: 10.1016/j.tranon.2025.102271] [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: 09/11/2024] [Revised: 12/25/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025] Open
Abstract
Gilteritinib treats acute myeloid leukemia (AML) with the FMS-like receptor tyrosine kinase-3 (FLT3) internal tandem duplication (ITD) mutation. Dysregulation of histone modification affects the genesis and progression of AML. Strategies targeting key histone regulators have not been applied to the treatment of AML. Lysine demethylase 6B (KDM6B) is dysregulated in a variety of cancers and regulates the expression of oncogenes, which has potential in anticancer therapy. We explored whether GSK-J4 (an inhibitor of the demethylase KDM6B) has an anti-leukemic effect in the gilteritinib treatment of FLT3-ITD+ AML and the effect of gilteritinib combined with GSK-J4 in leukemia. In our study, we evaluated the anti-leukemic effect of GSK-J4 in gilteritinib therapy through in vitro and in vivo experiments. The results revealed that the combined treatment of gilteritinib and GSK-J4 has greater anti-proliferation and pro-apoptosis effects than gilteritinib alone. Gilteritinib and GSK-J4 performed synergistically to arrest the cell cycle. Gilteritinib mainly induces cell cycle phase arrest at the S or G0/G1, and GSK-J4 inhibits the cell cycle progression in the S phase and reduces cell viability by reducing the expression of key regulatory factors from the G1 phase to the S phase. At the same time, GSK-J4 enhances the expression of apoptosis-related proteins (Bax and cleavage caspase-9). In addition, gilteritinib or GSK-J4 monotherapy increases reactive oxygen species (ROS) production, and the combination has a synergistic effect, accelerating leukemic cell death. Our study provides proof that the combined therapy of gilteritinib and GSK-J4 has a synergistic antileukemic effect on FLT3-ITD+ AML.
Collapse
Affiliation(s)
- Qi Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Yongyu Guan
- Clinical laboratory, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou 730000, China
| | - Pingping Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Huiyuan Chu
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Yaming Xi
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; Department of Hematology, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
43
|
Shaffer BC, Kebriaei P, de Lima M, Jimenez Jimenez AM. Measurable residual disease testing and allogeneic hematopoietic cell transplantation for AML: adapting Pre-MEASURE to clinical practice. Bone Marrow Transplant 2025; 60:128-134. [PMID: 39562717 PMCID: PMC11810777 DOI: 10.1038/s41409-024-02481-2] [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: 09/18/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Measurable residual disease (MRD) testing in patients with acute myelogenous leukemia (AML) represents a heterogenous assessment process designed to quantify leukemia-specific biomarkers that are not ascertainable by routine pathologic evaluation. The most common tools used to assess MRD are multiparameter flow cytometry (MPFC), and polymerase chain reaction (PCR) based tools, including quantitative or digital droplet PCR (qPCR, ddPCR), or next-generation sequencing (NGS) technologies. Collectively, MRD assessments have become an important clinical tool in the management of patients with AML. Despite progress, significant questions remain with respect to the appropriate timing, frequency, and methodology of MRD assessment, and whether or how to adapt therapy based on MRD results. Recent data from the Pre-MEASURE study, a retrospective cohort analysis of error corrected NGS based MRD assessment prior to allogeneic hematopoietic cell transplantation (alloHCT) in patients with AML, provides additional key information with respect to the emerging role of NGS-based technology in MRD assessment. In the context of this review, we evaluate the Pre-MEASURE study as well as other recent, high-quality assessments of MRD in AML. Our focus is to provide a practical assessment of the use of emerging MRD technologies in patients with AML with an emphasis on the role of peri-transplant MRD for the practicing clinician.
Collapse
Affiliation(s)
- Brian C Shaffer
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcos de Lima
- Division of Hematology and Oncology, and Stem Cell Transplant Program, Case Western Reserve University, Cleveland, OH, USA
| | - Antonio M Jimenez Jimenez
- Division of Transplantation and Cell Therapy, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
44
|
Tolu SS, Viny AD, Amengual JE, Pro B, Bates SE. Getting the right combination to break the epigenetic code. Nat Rev Clin Oncol 2025; 22:117-133. [PMID: 39623073 DOI: 10.1038/s41571-024-00972-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 01/26/2025]
Abstract
Rapid advances in the field of epigenetics have facilitated the development of novel therapeutics targeting epigenetic mechanisms that are hijacked by cancer cells to support tumour growth and progression. Several epigenetic agents have been approved by the FDA for the treatment of cancer; however, the efficacy of these drugs is dependent on the underlying biology and drivers of the disease, with inherent differences between solid tumours and haematological malignancies. The efficacy of epigenetic drugs as single agents remains limited across most cancer types, which has spurred the clinical development of combination therapies, with the hope of attaining synergistic activity and/or overcoming treatment resistance. In this Review we discuss clinical advances that have been achieved with the use of epigenetic agents in combination with chemotherapies, immunotherapies or other targeted agents, including epigenetic-epigenetic combinations, as well as limitations and challenges associated with these combinatorial strategies. So far, the success of combination therapies targeting epigenetic mechanisms has generally been confined to haematological malignancies, with limited efficacy observed in patients with solid tumours. Nevertheless, this Review captures the field of epigenetic combination therapies across the spectra of haematology and oncology, highlighting opportunities for precision therapy to effectively harness the potential of epigenetic agents and produce meaningful improvements in clinical outcomes.
Collapse
Affiliation(s)
- Seda S Tolu
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| | - Aaron D Viny
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Jennifer E Amengual
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Barbara Pro
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Susan E Bates
- Division of Hematology and Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
45
|
Saadh MJ, K Abdulsahib W, Ashurova D, Sanghvi G, Ballal S, Sharma R, Kumar Pathak P, Aman S, Kumar A, Feez Sead F, Chaitanya MVNL. FLT3-mutated AML: immune evasion through exosome-mediated mechanisms and innovative combination therapies targeting immune escape. Expert Rev Anticancer Ther 2025; 25:143-150. [PMID: 39885639 DOI: 10.1080/14737140.2025.2461632] [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: 12/14/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/01/2025]
Abstract
INTRODUCTION Acute Myeloid Leukemia is a heterogeneous hematological malignancy characterized by the uncontrolled proliferation of abnormal myeloid cells. Besides several other genetic abnormalities developed in AML, FLT3 mutations are significant due to their worse prognostic impacts and therapeutic resistance. As a result, these mutations enable AML cells to develop mechanisms for evading immune surveillance. AREAS COVERED This review discusses the ways of immune escape of FLT3-mutated AML cells. A literature search was conducted on PubMed, Scopus, and Web of Science databases, covering articles published between 2010 and 2024 with related keywords. The discussion covers AML cells' downregulation of immune recognition markers, expression of immune checkpoint proteins, and establishment of an immunosuppressive tumor microenvironment. Specific attention is given to small extracellular vesicles and their participation in immune escape. The focus is on exosome-mediated pathways and possible combination therapies. EXPERT OPINION FLT3 mutations in AML represent a formidable therapeutic challenge due to their crucial role in immune evasion. Exosomes are major players in these processes. Combination therapies targeting the exosome pathway could significantly improve these patients' immune recognition and overall outcomes. Understanding the underlying mechanisms, including targeted therapies, will be required to transcend existing therapeutic limitations and push newer strategies in treatment.
Collapse
MESH Headings
- Humans
- Exosomes/genetics
- Exosomes/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- fms-Like Tyrosine Kinase 3/genetics
- Mutation
- Tumor Microenvironment/immunology
- Tumor Escape/genetics
- Molecular Targeted Therapy
- Animals
- Prognosis
Collapse
Affiliation(s)
| | - Waleed K Abdulsahib
- Department of Pharmacology and Toxicology, College of Pharmacy, Al Farahidi University, Baghdad, Iraq
| | - Dilfuza Ashurova
- Department of Propedeutics of Pediatric Diseases and Hematology, Tashkent Pediatric Medical Institute, Tashkent, Uzbekistan
| | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science Marwadi University, Marwadi University Research Center, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, India
| | - Rsk Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Shankhyan Aman
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia, Ekaterinburg, Russia
- Department of Technical Sciences, Western Caspian University, Baku, Azerbaijan
| | - Fadhil Feez Sead
- Department of Dentistry, College of Dentistry, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Diwaniyah, Iraq
| | - M V N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
46
|
Joudinaud R, Boudry A, Fenwarth L, Geffroy S, Salson M, Dombret H, Berthon C, Pigneux A, Lebon D, Peterlin P, Bouzy S, Flandrin-Gresta P, Tavernier E, Carre M, Tondeur S, Haddaoui L, Itzykson R, Bertoli S, Bidet A, Delabesse E, Hunault M, Récher C, Preudhomme C, Duployez N, Dumas PY. Midostaurin shapes macroclonal and microclonal evolution of FLT3-mutated acute myeloid leukemia. Blood Adv 2025; 9:365-374. [PMID: 39418643 PMCID: PMC11787458 DOI: 10.1182/bloodadvances.2024014672] [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: 08/28/2024] [Revised: 09/19/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
ABSTRACT Despite the use of midostaurin (MIDO) with intensive chemotherapy (ICT) as frontline treatment for Fms-like tyrosine kinase 3 (FLT3)-mutated acute myeloid leukemia (AML), complete remission rates are close to 60% to 70%, and relapses occur in >40% of cases. Here, we studied the molecular mechanisms underlying refractory/relapsed (R/R) disease in patients with FLT3-mutated AML. We conducted a retrospective and multicenter study involving 150 patients with R/R AML harboring FLT3-internal tandem duplication (ITD) (n = 130) and/or FLT3-tyrosine kinase domain mutation (n = 26) at diagnosis assessed by standard methods. Patients were treated with ICT + MIDO (n = 54) or ICT alone (n = 96) according to the diagnosis date and label of MIDO. The evolution of FLT3 clones and comutations was analyzed in paired diagnosis-R/R samples by targeted high-throughput sequencing. Using a dedicated algorithm for FLT3-ITD detection, 189 FLT3-ITD microclones (allelic ratio [AR] of <0.05) and 225 macroclones (AR ≥ 0.05) were detected at both time points. At R/R disease, the rate of FLT3-ITD persistence was lower in patients treated with ICT + MIDO than in patients not receiving MIDO (68% vs 87.5%; P = .011). In patients receiving ICT + MIDO, detection of multiple FLT3-ITD clones was associated with a higher FLT3-ITD persistence rate at R/R disease (multiple clones: 88% vs single clones: 57%; P = .049). If only 24% of FLT3-ITD microclones detected at diagnosis were retained at relapse, 43% became macroclones. Together, these results identify parameters influencing the fitness of FLT3-ITD clones.
Collapse
Affiliation(s)
- Romane Joudinaud
- INSERM UMR1277, Centre National de la Recherche Scientifique UMR9020-CANTHER, Lille University Hospital, Université de Lille, Lille, France
- Hematology Laboratory, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Augustin Boudry
- Hematology Laboratory, Centre Hospitalier Universitaire de Lille, Lille, France
- ULR 2694 Metrics, Centre Hospitalier Universitaire de Lille, Université de Lille, Lille, France
| | - Laurène Fenwarth
- INSERM UMR1277, Centre National de la Recherche Scientifique UMR9020-CANTHER, Lille University Hospital, Université de Lille, Lille, France
- Hematology Laboratory, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Sandrine Geffroy
- INSERM UMR1277, Centre National de la Recherche Scientifique UMR9020-CANTHER, Lille University Hospital, Université de Lille, Lille, France
- Hematology Laboratory, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Mikaël Salson
- UMR 9189 Cristal, Centrale Lille, Centre National de la Recherche Scientifique, Université de Lille, Lille, France
| | - Hervé Dombret
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Céline Berthon
- INSERM UMR1277, Centre National de la Recherche Scientifique UMR9020-CANTHER, Lille University Hospital, Université de Lille, Lille, France
- Hematology Department, Centre Hospitalier Universitaire Lille, Lille, France
| | - Arnaud Pigneux
- Service d’Hématologie Clinique et de Thérapie Cellulaire, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - Delphine Lebon
- Hematology Department, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Pierre Peterlin
- Service d'hématologie clinique, Nantes University Hospital, Nantes, France
| | - Simon Bouzy
- Hematology Biology, Nantes University Hospital, Nantes, France
| | | | - Emmanuelle Tavernier
- Département d'hématologie clinique, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France
| | - Martin Carre
- Département d'hématologie, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Sylvie Tondeur
- Laboratoire de Génétique des hémopathies, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble, Grenoble, France
| | - Lamya Haddaoui
- French Innovative Leukemia Organization Tumor Bank, Pitié-Salpêtrière Hospital, Paris, France
| | - Raphael Itzykson
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, Centre National de la Recherche Scientifique, Université Paris Cité, Paris, France
| | - Sarah Bertoli
- Service d’Hématologie, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire Toulouse, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Audrey Bidet
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - Eric Delabesse
- Laboratoire d'Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Mathilde Hunault
- Département des Maladies du Sang, Centre Hospitalier Universitaire Angers, INSERM, Centre National de la Recherche Scientifique, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers, Fédération Hospitalo-Universitaire Grand Ouest Against Leukemia, Université d'Angers, Université de Nantes, Angers, France
| | - Christian Récher
- Service d’Hématologie, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire Toulouse, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Claude Preudhomme
- INSERM UMR1277, Centre National de la Recherche Scientifique UMR9020-CANTHER, Lille University Hospital, Université de Lille, Lille, France
- Hematology Laboratory, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Nicolas Duployez
- INSERM UMR1277, Centre National de la Recherche Scientifique UMR9020-CANTHER, Lille University Hospital, Université de Lille, Lille, France
- Hematology Laboratory, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Pierre-Yves Dumas
- Service d’Hématologie Clinique et de Thérapie Cellulaire, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| |
Collapse
|
47
|
Sieberer H, Luciano M, Amend D, Blöchl C, Eglseer A, Steinkellner A, Rieser S, Andosch A, Steiner P, Hummer L, Krenn PW, Dang HH, Huber CG, Aberger F, Neuper T, Horejs-Hoeck J. Inhibition of NLRP3 enhances pro-apoptotic effects of FLT3 inhibition in AML. Cell Commun Signal 2025; 23:53. [PMID: 39875995 PMCID: PMC11773904 DOI: 10.1186/s12964-025-02046-w] [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: 09/30/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
FLT3 mutations occur in approximately 25% of all acute myeloid leukemia (AML) patients. While several FLT3 inhibitors have received FDA approval, their use is currently limited to combination therapies with chemotherapy, as resistance occurs, and efficacy decreases when the inhibitors are used alone. Given the highly heterogeneous nature of AML, there is an urgent need for novel targeted therapies that address the disease from multiple angles. Recent research has identified the NLRP3 inflammasome as a potential new driver in AML. Here, we investigated the efficacy of different NLRP3 inhibitors in targeting AML cells in vitro. Our findings reveal that NLRP3 inhibition induces cell cycle arrest as well as signs of senescence in multiple AML cell lines. In contrast, NLRP3 inhibition selectively induced apoptosis in FLT3 mutant AML cell lines, but not in FLT3 wild-type AML cells. Moreover, we show that NLRP3 inhibition impairs FLT3 signaling by reducing both FLT3 expression as well as downstream signaling in FLT3 mutant cells. A database analysis revealed a strong positive correlation between FLT3 and NLRP3 in cancer, which was particularly evident in AML patients. Strikingly, the simultaneous inhibition of NLRP3 and FLT3 markedly enhanced apoptosis in FLT3-ITD mutant AML cells, but not in FLT3 wild-type cells. In summary, this study reveals a promising combined therapeutic strategy specifically targeting NLRP3/FLT3-ITD positive AML blasts in vitro, highlighting a potential new avenue for AML treatment.
Collapse
MESH Headings
- Humans
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Apoptosis/drug effects
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- Cell Line, Tumor
- Signal Transduction/drug effects
- Mutation
Collapse
Affiliation(s)
- Helene Sieberer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Michela Luciano
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Diana Amend
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Constantin Blöchl
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Anna Eglseer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Alina Steinkellner
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
| | - Sebastian Rieser
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Ancuela Andosch
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Philip Steiner
- Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, Linz, 4020, Austria
| | - Laura Hummer
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Peter W Krenn
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Hieu-Hoa Dang
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Christian G Huber
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Fritz Aberger
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Theresa Neuper
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria
- Cancer Cluster Salzburg, Salzburg, 5020, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria.
- Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, 5020, Austria.
- Cancer Cluster Salzburg, Salzburg, 5020, Austria.
| |
Collapse
|
48
|
Hochman MJ, Muniz JP, Papadantonakis N. Precision Medicine in Myeloid Neoplasia: Challenges and Opportunities. J Pers Med 2025; 15:49. [PMID: 39997326 PMCID: PMC11856194 DOI: 10.3390/jpm15020049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
High-risk myeloid neoplasms encompass a group of hematologic malignancies known to cause significant cytopenias, which are accompanied by the risk of end-organ damage. They tend to have an aggressive clinical course and limit life expectancy in the absence of effective treatments. The adoption of precision medicine approaches has been limited by substantive diversity in somatic mutations, limited fraction of patients with targetable genetic lesions, and the prolonged turnaround times of pertinent genetic tests. Efforts to incorporate targeted agents into first-line treatment, rapidly determine pre-treatment molecular or cytogenetic aberrations, and evaluate functional vulnerabilities ex vivo hold promise for advancing the use of precision medicine in these malignancies. Given the relative accessibility of malignant cells from blood and bone marrow, precision medicine strategies hold great potential to shape future standard-of-care approaches to patients with high-risk myeloid malignancies. This review aims to summarize the development of the targeted therapies currently available to treat these blood cancers, most notably acute myeloid leukemia, and also evaluate future opportunities and challenges related to the integration of personalized approaches.
Collapse
Affiliation(s)
- Michael J. Hochman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Joshua P. Muniz
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Aflac Cancer & Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30329, USA
| | - Nikolaos Papadantonakis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
49
|
Battaglia G, Lazzarotto D, Tanasi I, Gurrieri C, Forlani L, Mauro E, Capraro F, Ciotti G, De Bellis E, Callegari C, Tosoni L, Fanin M, Morelli GL, Simio C, Skert C, Gottardi M, Zaja F, Toffoletti E, Damiani D, Fanin R, Tiribelli M. New Combination Regimens vs. Fludarabine, Cytarabine, and Idarubicin in the Treatment of Intermediate- or Low-Risk Nucleophosmin-1-Mutated Acute Myeloid Leukemia: A Retrospective Analysis from 7 Italian Centers. J Clin Med 2025; 14:700. [PMID: 39941372 PMCID: PMC11818901 DOI: 10.3390/jcm14030700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Nucleophosmin-1 (NPM1) mutation accounts for 30% of acute myeloid leukemia (AML) cases and defines either low- or intermediate-risk AML, depending on FLT3-ITD mutation. New combination regimens (NCRs), adding midostaurin and gemtuzumab ozogamicin (GO) to the 3 + 7 scheme, are commonly used, though there are no data that compare NCRs with intensive induction chemotherapy. Methods: To evaluate the efficacy and safety of NCRs and FLAI in NPM1+ AML, we retrospectively analyzed 125 patients treated with FLAI (n = 53) or NCRs (n = 72) at seven Italian Centers. Results: The median age was 61 years and 51/125 (41%) were FLT3-ITD+. The complete remission (CR) rate was 77%, slightly better with NCRs (83% vs. 68%; p = 0.054). NCRs yielded a superior median overall survival (OS) (not reached (NR) vs. 27.3 months; p = 0.002), though the median event-free survival (EFS) was similar (NR vs. 20.5 months; p = 0.07). In low-risk AML, CR was higher in NCRs (94% vs. 72%, p = 0.02), as were median OS (NR vs. 41.6 months; p = 0.0002) and EFS (NR vs. 17.8 months; p = 0.0085). In intermediate-risk AML (FLT3-ITD+), there were no differences in CR (60% vs. 71%; p = 0.5), OS (p = 0.27), or EFS (p = 0.86); only allogeneic transplantation improved OS (NR vs. 13.4 months; p = 0.005), regardless of induction regimen. The safety profile was similar, except for delayed platelet recovery with FLAI (22 vs. 18 days; p = 0.0024) and higher-grade II-IV gastrointestinal toxicity with NCRs (43% vs. 18.8%; p = 0.0066). Conclusions: Our data suggest the superiority of NCRs over FLAI in low-risk patients, while all outcomes were comparable in intermediate-risk patients, a setting in which only transplants positively impacted on survival.
Collapse
Affiliation(s)
- Giulia Battaglia
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
| | - Davide Lazzarotto
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
| | - Ilaria Tanasi
- Hematology Unit, Department of Engineering for Innovation Medicine, Azienda Ospedaliera Universitaria Integrata di Verona, 37126 Verona, Italy
| | - Carmela Gurrieri
- Hematology Unit, Azienda Ospedale-Università and University of Padova, 35128 Padua, Italy
| | - Laura Forlani
- Hematology Unit, Azienda Ospedale-Università and University of Padova, 35128 Padua, Italy
| | - Endri Mauro
- Hematology Section, Dipartimento di Medicina Specialistica, Ca’ Foncello Hospital, 31100 Treviso, Italy
| | - Francesca Capraro
- Hematology Unit, Azienda Ulss3 Serenissima, Ospedale dell’Angelo, 30174 Venice, Italy (C.S.)
| | - Giulia Ciotti
- Department of Oncology, UOC Oncohematology, Istituto Oncologico Veneto (IOV) IRCCS, 35128 Padova, Italy (M.G.)
| | - Eleonora De Bellis
- Hematology Unit, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy (F.Z.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Chiara Callegari
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
| | - Luca Tosoni
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
| | - Matteo Fanin
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
| | - Gian Luca Morelli
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
| | - Claudia Simio
- Hematology Unit, Department of Engineering for Innovation Medicine, Azienda Ospedaliera Universitaria Integrata di Verona, 37126 Verona, Italy
| | - Cristina Skert
- Hematology Unit, Azienda Ulss3 Serenissima, Ospedale dell’Angelo, 30174 Venice, Italy (C.S.)
| | - Michele Gottardi
- Department of Oncology, UOC Oncohematology, Istituto Oncologico Veneto (IOV) IRCCS, 35128 Padova, Italy (M.G.)
| | - Francesco Zaja
- Hematology Unit, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy (F.Z.)
| | - Eleonora Toffoletti
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Daniela Damiani
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Renato Fanin
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and BMT, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy; (G.B.); (R.F.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
50
|
Tjahjono E, Daneman MR, Meika B, Revtovich AV, Kirienko NV. Mitochondrial abnormalities as a target of intervention in acute myeloid leukemia. Front Oncol 2025; 14:1532857. [PMID: 39902131 PMCID: PMC11788353 DOI: 10.3389/fonc.2024.1532857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy; it is the most common acute leukemia in adults. AML prognosis is often poor, and relapse often occurs after initial remission. Recurrent genetic abnormalities underlying this disease and the presence of leukemic stem cells complicate disease treatment. However, the complex metabolic reprogramming that enables the unrestrained cell growth seen in these cells may also be their Achilles' heel. In these cells, mitophagy operates as a double-edged sword. On one hand, it provides a source of building blocks for further cell division and serves as a method for removing damaged organelles, promoting cell survival. However, the profound metabolic changes to mitochondria also render these organelles more sensitive to damage and place them precariously close to excess mitophagic activation. This review discusses the dual role mitophagy plays in AML survival, the importance of targeting mitophagy to treat AML, and current progress in the area. The discovery and mechanism of action of multiple compounds that were used to inhibit or stimulate mitophagy and their effects on AML survival are also described. Further, we explore the combination strategy of mitophagy-targeting compounds with existing and/or novel chemotherapeutics to eradicate AML and discuss strategies to uncover new drug targets and novel mitochondria-targeting drugs.
Collapse
|