1
|
Liu J, Zhao XS, Chang YJ, Qin YZ, Jiang Q, Jiang H, Zhang XH, Xu LP, Wang Y, Lv M, Liu KY, Huang XJ, Zhao XY. Monitoring the KMT2A gene post-chemotherapy independently predicts the relapse and survival risk after allogeneic haematopoietic stem cell transplantation. Br J Haematol 2025. [PMID: 40081934 DOI: 10.1111/bjh.20036] [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: 10/22/2024] [Accepted: 02/21/2025] [Indexed: 03/16/2025]
Abstract
This study evaluated the kinetics of KMT2A-r during chemotherapy and its impact on allogeneic haematopoietic stem cell transplantation (allo-HSCT) outcomes. KMT2A-r was assessed post-induction (MRD1), after the first (MRD2) and second (MRD3) consolidations and pre-transplant (MRD4) in 52 patients with acute myeloid leukaemia (AML). KMT2A-r significantly decreased from diagnosis to MRD2 (p < 0.001 for diagnosis vs. MRD1; p = 0.019 for MRD1 vs. MRD2). The incidence of KMT2A-r negativity (57.5%) peaked at MRD2. KMT2A-r status at each time point significantly affected post-transplant outcomes. Cluster analysis identified four KMT2A-r kinetic profiles: persistently negative (-/-), turned negative at transplant (+/-), turned positive at transplant (-/+) and persistently positive (+/+). The (-/-) group had the best outcomes, with a cumulative incidence of relapse (CIR) of 13.0%, overall survival (OS) of 82.0% and leukaemia-free survival (LFS) of 81.7%. The (+/+) group had the worst prognosis, with a CIR of 58.8%, OS of 29.4% and LFS of 23.5%. KMT2A dynamics were an independent risk factor for CIR (Hazard ratio [HR] = 11.070, 95%CI 2.395-51.165, p = 0.002), LFS (HR = 9.316, 95%CI 2.656-32.668, p < 0.001) and OS (HR = 7.172, 95%CI 1.999-25.730, p = 0.003). In conclusion, KMT2A-r status after chemotherapy and its kinetics are significant HSCT prognostic indicators.
Collapse
Affiliation(s)
- Jing Liu
- 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, Peking University, Beijing, China
| | - Xiao-Su 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, Peking University, Beijing, China
| | - Ying-Jun Chang
- 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, Peking University, Beijing, China
| | - Ya-Zhen Qin
- 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, Peking University, Beijing, China
| | - Qian Jiang
- 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, Peking University, Beijing, China
| | - Hao Jiang
- 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, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- 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, Peking University, Beijing, China
| | - Lan-Ping Xu
- 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, Peking University, Beijing, China
| | - Yu Wang
- 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, Peking University, Beijing, China
| | - Meng Lv
- 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, Peking University, Beijing, China
| | - Kai-Yan Liu
- 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, Peking University, Beijing, China
| | - Xiao-Jun Huang
- 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, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiang-Yu 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, Peking University, Beijing, China
| |
Collapse
|
2
|
Wang L, Chen Y, Zang M, Zhou J, Xiao M, Fu H, Mo X, Wang F, Han W, Zhang Y, Yan C, Wang Z, Han T, Lv M, Chen H, Chen Y, Chen Y, Wang J, Wang Y, Xu L, Liu K, Huang X, Zhang X. Molecular measurable residual disease before transplantation independently predicts survival and relapse risk in adult lysine methyltransferase 2a-rearranged acute myeloid leukemia. Cancer 2025; 131:e35717. [PMID: 39760486 DOI: 10.1002/cncr.35717] [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: 10/03/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Patients with lysine methyltransferase 2a (KMT2A)-rearranged (KMT2A-r) acute myeloid leukemia (AML) are assigned to intermediate-risk and adverse-risk categories at diagnosis. However, the value of molecular measurable residual disease (MRD) status in patients who have KMT2A-r AML before allogeneic hematopoietic stem cell transplantation (allo-HSCT) in adult cohorts has rarely been evaluated. METHODS Patients with KMT2A-r AML who achieved complete remission and subsequently underwent allo-HSCT between January 2015 and January 2023 were included in this analysis. Real-time quantitative polymerase chain reaction was used to detect molecular MRD in bone marrow samples. The end points were overall survival (OS), leukemia-free survival (LFS), the cumulative incidence of relapse (CIR), and nonrelapse mortality (NRM). RESULTS Pretransplantation molecular MRD was identified in 52 of 125 patients (42%) with KMT2A-r AML. The presence of KMT2A-r MRD was associated with inferior 3-year OS (51% vs. 82%; p < .001), LFS (42% vs. 81%; p < .001), CIR (33% vs. 12%; p < .001), and NRM (11% vs. 5%; p = .12). In multivariate models, molecular MRD status before transplantation independently predicted OS, LFS, and CIR. The survival of adult patients with KMT2A-r AML was heterogeneous, depending on the KMT2A translocation partners, and was more favorable in patients who had t(9;11) and t(10;11) than in those who had t(11;19) and t(6;11). In addition, flow cytometry-based MRD analysis conferred no additional prognostic value to the results of molecular MRD status. CONCLUSIONS Residual KMT2A-r before allo-HSCT independently predicts the risk of survival and relapse, and donor lymphocyte infusion or posttransplantation maintenance therapies should be considered for patients who have AML with detectable molecular MRD.
Collapse
Affiliation(s)
- Lulu Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuxiu Chen
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Mengtong Zang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Jianying Zhou
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Mengyu Xiao
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Haixia Fu
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiaodong Mo
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Fengrong Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Wei Han
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuanyuan Zhang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Chenhua Yan
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Zhidong Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Tingting Han
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Meng Lv
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Huan Chen
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuhong Chen
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yao Chen
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Jingzhi Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yu Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Lanping Xu
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Kaiyan Liu
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiaojun Huang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiaohui Zhang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| |
Collapse
|
3
|
Guo X, Atla S, Nyalata S, Alugubelli YR, Chen PHC, Xu S, Liu WR. Prioritization of Eleven-Nineteen-Leukemia Inhibitors as Orally Available Drug Candidates for Acute Myeloid Leukemia. J Med Chem 2024; 67:20100-20117. [PMID: 39530508 PMCID: PMC11613437 DOI: 10.1021/acs.jmedchem.4c01337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
Acute myeloid leukemia (AML) is the second most prevalent and fatal form of leukemia. The growth of AML cells harboring oncogenic MLL rearrangements relies on the YEATS domain-containing protein ENL. Many small molecule inhibitors targeting ENL have been developed. To prioritize these inhibitors for in vivo studies, a NanoBRET system was introduced to evaluate their cellular permeability and potency. This screening identified inhibitor 13 as a promising candidate. This inhibitor has remarkable metabolic stability and potent antiproliferative effects on MLL-fusion leukemia cell lines. In AML-xenografted mice, inhibitor 13 significantly improved survival. Subsequent optimization efforts led to the development of SR-C-107 (R), which exhibited strong activity against AML both at the cellular level (CC50 (MOLM-13): 1.25 ± 0.18 μM; CC50 (MV4-11): 0.81 ± 0.15 μM) and in vivo. These findings establish SR-C-107 (R) as a compelling candidate for AML treatment and lay the groundwork for the development of next-generation AML inhibitors.
Collapse
Affiliation(s)
- Xuejiao
Shirley Guo
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Sandeep Atla
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Satyanarayana Nyalata
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Yugendar R. Alugubelli
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Peng-Hsun Chase Chen
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Shiqing Xu
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
- Department
of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas 77843, United States
| | - Wenshe Ray Liu
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
- Department
of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas 77843, United States
- Institute
of Biosciences and Technology and Department of Translational Medical
Sciences, College of Medicine, Texas A&M
University, Houston, Texas 77030, United States
- Department
of Biochemistry and Biophysics, Texas A&M
University, College Station, Texas 77843, United States
- Department
of Cell Biology and Genetics, College of Medicine, Texas A&M University, Bryan, Texas 77807, United States
| |
Collapse
|
4
|
Demoen L, Matthijssens F, Reunes L, Palhais B, Lintermans B, T’Sas S, Fijalkowski I, Taminau J, Akele MZ, Van Belle S, Taghon T, Deforce D, Van Nieuwerburgh F, Berx G, Ntziachristos P, Debyser Z, Durinck K, Pieters T, Goossens S, Van Vlierberghe P. A dual role for PSIP1/LEDGF in T cell acute lymphoblastic leukemia. SCIENCE ADVANCES 2024; 10:eado6765. [PMID: 39485844 PMCID: PMC11529709 DOI: 10.1126/sciadv.ado6765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy. Current intensified therapeutic protocols coincide with severe side effects, and no salvage therapy is available for primary therapy-resistant or relapsed patients. This highlights the need to identify new therapeutic targets in T-ALL. PSIP1, dispensable for normal hematopoiesis, is a dependency factor in KMT2A-rearranged myeloid leukemia. Nonetheless, loss-of-function mutations suggest a tumor suppressor role for PSIP1 in T-ALL. Here, we demonstrate that the loss of Psip1 accelerates T-ALL initiation in mice which we correlated with reduced H3K27me3 binding. Contrastingly, loss of PSIP1 impaired cell proliferation in several T-ALL cell lines. In cell lines, PSIP1 down-regulation leads to a reduction of COX20, an assembly factor of the cytochrome c oxidase in the mitochondria, and to a reduction in mitochondrial respiration. This indicates that PSIP1 can exert a dual role in the context of T-ALL, either as a tumor suppressor gene during tumor initiation or as a dependency factor in tumor maintenance.
Collapse
Affiliation(s)
- Lisa Demoen
- Lab of Normal and Malignant Hematopoiesis, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Filip Matthijssens
- Lab of Normal and Malignant Hematopoiesis, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Lindy Reunes
- Lab of Normal and Malignant Hematopoiesis, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Bruno Palhais
- Lab of Normal and Malignant Hematopoiesis, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Béatrice Lintermans
- Lab of Normal and Malignant Hematopoiesis, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| | - Sara T’Sas
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Unit for Translational Research in Oncology, Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - Igor Fijalkowski
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Leukemia Therapy Resistance Laboratory, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Joachim Taminau
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Department Biomedical Molecular Biology, 9000 Ghent University, Ghent, Belgium
| | - Muluembet Z. Akele
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
| | - Siska Van Belle
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
| | - Tom Taghon
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- T Cell Team Taghon, Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Ghent University, 9000 Ghent, Belgium
| | | | - Geert Berx
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Department Biomedical Molecular Biology, 9000 Ghent University, Ghent, Belgium
| | - Panagiotis Ntziachristos
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Leukemia Therapy Resistance Laboratory, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Zeger Debyser
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
| | - Kaat Durinck
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Pediatric Precision Oncology Lab, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Tim Pieters
- Lab of Normal and Malignant Hematopoiesis, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Unit for Translational Research in Oncology, Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
- Leukemia Therapy Resistance Laboratory, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Unit for Translational Research in Oncology, Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - Pieter Van Vlierberghe
- Lab of Normal and Malignant Hematopoiesis, Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
5
|
Yin L, Wan L, Zhang Y, Hua S, Shao X. Recent Developments and Evolving Therapeutic Strategies in KMT2A-Rearranged Acute Leukemia. Cancer Med 2024; 13:e70326. [PMID: 39428967 PMCID: PMC11491690 DOI: 10.1002/cam4.70326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/09/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Rearrangements of the histone-lysine-N-methyltransferase (KMT2A), previously referred to as mixed-lineage leukemia (MLL), are among the most common chromosomal abnormalities in patients with acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), involving numerous different fusion partners. KMT2A-rearranged (KMT2A-r) leukemia is characterized by a rapid onset, aggressive progression, and significantly worse prognosis compared to non-KMT2A-r leukemias. Even with contemporary chemotherapeutic treatments and hematopoietic stem cell transplantations (HSCT), patients with KMT2A-r leukemia typically experience poor outcomes and limited responses to these therapies. OBJECTIVES This review aims to consolidate recent studies on the general gene characteristics and associated mechanisms of KMT2A-r acute leukemia, as well as the cytogenetics, immunophenotype, clinical presentation, and risk stratification of both KMT2A-r-AML and KMT2A-r-ALL. Particularly, the treatment targets in KMT2A-r acute leukemia are examined. METHODS A comprehensive review was carried out by systematically synthesizing existing literature on PubMed, using the combination of the keywords 'KMT2A-rearranged acute leukemia', 'lymphoblastic leukemia', 'myeloid leukemia', and 'therapy'. The available studies were screened for selection based on quality and relevance. CONCLUSIONS Studies indicate that KMT2A rearrangements are present in over 70% of infant leukemia cases, approximately 10% of adult AML cases, and numerous instances of secondary acute leukemias, making it a disease of critical concern to clinicians and researchers alike. The future of KMT2A-r acute leukemia research is characterized by an expanding knowledge of the disease's biology, with an emphasis on personalized therapies, immunotherapies, genomic advancements, and innovative therapeutic combinations. The overarching aim is to enhance patient outcomes, lessen the disease burden, and elevate the quality of life for those affected. Ongoing research and clinical trials in this area continue to offer promising opportunities for refining treatment strategies and improving patient prognosis.
Collapse
Affiliation(s)
- Lei Yin
- Department of Clinical LaboratoryChildren's Hospital of Soochow UniversitySuzhouChina
| | - Lin Wan
- Department of PediatricsThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Youjian Zhang
- Department of Clinical LaboratoryChildren's Hospital of Soochow UniversitySuzhouChina
| | - Shenghao Hua
- Department of Clinical LaboratoryChildren's Hospital of Soochow UniversitySuzhouChina
| | - Xuejun Shao
- Department of Clinical LaboratoryChildren's Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
6
|
Xu J, Zhong A, Zhang S, Chen M, Zhang L, Hang X, Zheng J, Wu B, Deng X, Pan X, Wang Z, Qi L, Shi K, Li S, Wang Y, Wang M, Chen X, Zhang Q, Liu P, Gale RP, Chen C, Liu Y, Niu T. KMT2D Deficiency Promotes Myeloid Leukemias which Is Vulnerable to Ribosome Biogenesis Inhibition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206098. [PMID: 37142882 PMCID: PMC10323629 DOI: 10.1002/advs.202206098] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/17/2023] [Indexed: 05/06/2023]
Abstract
KMT2C and KMT2D are the most frequently mutated epigenetic genes in human cancers. While KMT2C is identified as a tumor suppressor in acute myeloid leukemia (AML), the role of KMT2D remains unclear in this disease, though its loss promotes B cell lymphoma and various solid cancers. Here, it is reported that KMT2D is downregulated or mutated in AML and its deficiency, through shRNA knockdown or CRISPR/Cas9 editing, accelerates leukemogenesis in mice. Hematopoietic stem and progenitor cells and AML cells with Kmt2d loss have significantly enhanced ribosome biogenesis and consistently, enlarged nucleolus, increased rRNA and protein synthesis rates. Mechanistically, it is found that KMT2D deficiency leads to the activation of the mTOR pathway in both mouse and human AML cells. Kmt2d directly regulates the expression of Ddit4, a negative regulator of the mTOR pathway. Consistent with the abnormal ribosome biogenesis, it is shown that CX-5461, an inhibitor of RNA polymerase I, significantly restrains the growth of AML with Kmt2d loss in vivo and extends the survival of leukemic mice. These studies validate KMT2D as a de facto tumor suppressor in AML and reveal an unprecedented vulnerability to ribosome biogenesis inhibition.
Collapse
Grants
- 82130007 National Natural Science Foundation of China
- 2022M722272 China Postdoctoral Science Foundation
- 2018RZ0140 Sichuan Science and Technology Program
- 2022SCUH0037 "From 0 to 1" Innovation Project of Sichuan University
- 19HXFH030 Incubation Program for Clinical Trials, West China Hospital, Sichuan University
- ZYJC21007 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21009 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYGD22012 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- National Institute of Health Research Biomedical Research Centre
- 2023HXBH019 Post-Doctor Research Project, West China Hospital, Sichuan University
- 2023SCU12073 Post-Doctor Research Project of Sichuan University
- National Natural Science Foundation of China
- China Postdoctoral Science Foundation
Collapse
Affiliation(s)
- Jing Xu
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Ailing Zhong
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Shan Zhang
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Mei Chen
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Lanxin Zhang
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Xiaohang Hang
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Jianan Zheng
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Baohong Wu
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Xintong Deng
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Xiangyu Pan
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Zhongwang Wang
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Lu Qi
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Kaidou Shi
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Shujun Li
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Yiyun Wang
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Manli Wang
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Xuelan Chen
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Qi Zhang
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Pengpeng Liu
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Robert Peter Gale
- Centre for HematologyImperial College of ScienceTechnology and MedicineLondonSW7 2BXUK
- Department of Hematologic OncologySun Yat‐sen Cancer CenterGuangzhou510060China
| | - Chong Chen
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Yu Liu
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Ting Niu
- Department of HematologyInstitute of HematologyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
7
|
Bouligny IM, Maher KR, Grant S. Mechanisms of myeloid leukemogenesis: Current perspectives and therapeutic objectives. Blood Rev 2023; 57:100996. [PMID: 35989139 PMCID: PMC10693933 DOI: 10.1016/j.blre.2022.100996] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic neoplasm which results in clonal proliferation of abnormally differentiated hematopoietic cells. In this review, mechanisms contributing to myeloid leukemogenesis are summarized, highlighting aberrations of epigenetics, transcription factors, signal transduction, cell cycling, and the bone marrow microenvironment. The mechanisms contributing to AML are detailed to spotlight recent findings that convey clinical impact. The applications of current and prospective therapeutic targets are accentuated in addition to reviews of treatment paradigms stratified for each characteristic molecular lesion - with a focus on exploring novel treatment approaches and combinations to improve outcomes in AML.
Collapse
Affiliation(s)
- Ian M Bouligny
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Keri R Maher
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
8
|
Matthews AH, Pratz KW, Carroll MP. Targeting Menin and CD47 to Address Unmet Needs in Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:5906. [PMID: 36497385 PMCID: PMC9735817 DOI: 10.3390/cancers14235906] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 12/02/2022] Open
Abstract
After forty years of essentially unchanged treatment in acute myeloid leukemia (AML), innovation over the past five years has been rapid, with nine drug approvals from 2016 to 2021. Increased understanding of the molecular changes and genetic ontology of disease have led to targeting mutations in isocitrate dehydrogenase, FMS-like tyrosine kinase 3 (FLT3), B-cell lymphoma 2 and hedgehog pathways. Yet outcomes remain variable; especially in defined molecular and genetic subgroups such as NPM1 (Nucleophosmin 1) mutations, 11q23/KMT2A rearranged and TP53 mutations. Emerging therapies seek to address these unmet needs, and all three of these subgroups have promising new therapeutic approaches. Here, we will discuss the normal biological roles of menin in acute leukemia, notably in KMT2A translocations and NPM1 mutation, as well as current drug development. We will also explore how CD47 inhibition may move immunotherapy into front-line settings and unlock new treatment strategies in TP53 mutated disease. We will then consider how these new therapeutic advances may change the management of AML overall.
Collapse
Affiliation(s)
- Andrew H. Matthews
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Keith W. Pratz
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 715 Biomedical Research Building II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Konoplev S, Wang X, Tang G, Li S, Wang W, Xu J, Pierce SA, Jia F, Jorgensen JL, Ravandi F, Issa GC, Medeiros LJ, Wang SA. Comprehensive immunophenotypic study of acute myeloid leukemia with KMT2A (MLL) rearrangement in adults: A single-institution experience. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2022; 102:123-133. [PMID: 34964255 DOI: 10.1002/cyto.b.22051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Acute myeloid leukemia (AML) with KMT2A (MLL) rearrangement is known for monocytic or myelomonocytic differentiation, but the full immunophenotypic spectrum and dynamic changes of the immunophenotype in this genetically defined disease have not been systematically studied. METHODS We reviewed the immunophenotype, karyotype, and mutations at the time of initial diagnosis and relapse of adults with AML with KMT2A rearrangement in our institution between 2007 and 2020. RESULTS We identified 102 patients: 44 men and 58 women with a median age of 52 years (range, 18-87). Forty-three patients were considered to be therapy-related. Twenty-four out of 64 patients relapsed from complete remission after induction therapy, 34 had persistent/progressive disease, and 58 patients died with a median overall survival of 17 months. We detected five immunophenotypes: immature monocytic (38%); myelomonocytic (22%); myeloblastic (22%); mature monocytic (10%); and acute promyelocytic (APL)-like (8%). By chromosomal breakpoints, we presumed 11 different partners; t(9;11) (p22;q23)/MLLT3-KMT2A was the most common rearrangement (n = 56, 55%), followed by t(6;11) (q27;q23)/AFDN-KMT2A (n = 13,13%). Patients with t(6;11) (q27;q23)/AFDN-KMT2A preferentially showed a myeloblastic phenotype (p = 0.026). Mutations were detected in 39/64 (61%) cases, and RAS pathway (NRAS/KRAS/PTPN11) was involved in 26/64 (41%) cases. None of the APL-like cases had mutations detected. At the time of disease relapse, 10/24 (42%) showed major immunophenotypic change, and 7/10 cases gained additional cytogenetic and/or molecular alterations. CONCLUSION The immunophenotype of AML with KMT2A rearrangement is more diverse than previously recognized, with a substantial subset showing no evidence of monocytic differentiation. Major immunophenotype change is common at the time of relapse.
Collapse
Affiliation(s)
- Sergej Konoplev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiaoqiong Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jie Xu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sherry A Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fuli Jia
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey L Jorgensen
- Department of Hematopathology, 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
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
10
|
Gottardi M, Simonetti G, Sperotto A, Nappi D, Ghelli Luserna di Rorà A, Padella A, Norata M, Giannini MB, Musuraca G, Lanza F, Cerchione C, Martinelli G. Therapeutic Targeting of Acute Myeloid Leukemia by Gemtuzumab Ozogamicin. Cancers (Basel) 2021; 13:cancers13184566. [PMID: 34572794 PMCID: PMC8469571 DOI: 10.3390/cancers13184566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematological malignancy characterized by genetic and clinical heterogeneity and high mortality. Despite the recent introduction of novel pharmaceutical agents in hemato-oncology, few advancements have been made in AML for decades. In the last years, the therapeutic options have rapidly changed, with the approval of innovative compounds that provide new opportunities, together with new challenges for clinicians: among them, on 1 September, 2017 the Food and Drug Administration granted approval for Gemtuzumab Ozogamicin (GO) in combination with daunorubicin and cytarabine for the treatment of adult patients affected by newly diagnosed CD33+ AML. Benefits of GO-based regimens were also reported in the pre- and post-transplantation settings. Moreover, several biomarkers of GO response have been suggested, including expression of CD33 and multidrug resistance genes, cytogenetic and molecular profiles, minimal residual disease and stemness signatures. Among them, elevated CD33 expression on blast cells and non-adverse cytogenetic or molecular risk represent largely validated predictors of good response.
Collapse
Affiliation(s)
- Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV, IRCCS, 31033 Padua, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Alessandra Sperotto
- Hematology and Transplant Center Unit, Dipartimento di Area Medica (DAME), Udine University Hospital, 33100 Udine, Italy
| | - Davide Nappi
- Department of Hematology and Cell Bone Marrow Transplantation (CBMT), Ospedale di Bolzano, 39100 Bolzano, Italy
| | - Andrea Ghelli Luserna di Rorà
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Antonella Padella
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Marianna Norata
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Maria Benedetta Giannini
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Gerardo Musuraca
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Francesco Lanza
- Hematology Unit & Romagna Transplant Network, Ravenna Hospital, 48121 Ravenna, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola (FC), Italy
| |
Collapse
|
11
|
Gene Transcription as a Therapeutic Target in Leukemia. Int J Mol Sci 2021; 22:ijms22147340. [PMID: 34298959 PMCID: PMC8304797 DOI: 10.3390/ijms22147340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022] Open
Abstract
Blood malignancies often arise from undifferentiated hematopoietic stem cells or partially differentiated stem-like cells. A tight balance of multipotency and differentiation, cell division, and quiescence underlying normal hematopoiesis requires a special program governed by the transcriptional machinery. Acquisition of drug resistance by tumor cells also involves reprogramming of their transcriptional landscape. Limiting tumor cell plasticity by disabling reprogramming of the gene transcription is a promising strategy for improvement of treatment outcomes. Herein, we review the molecular mechanisms of action of transcription-targeted drugs in hematological malignancies (largely in leukemia) with particular respect to the results of clinical trials.
Collapse
|
12
|
CD33 Expression and Gentuzumab Ozogamicin in Acute Myeloid Leukemia: Two Sides of the Same Coin. Cancers (Basel) 2021; 13:cancers13133214. [PMID: 34203180 PMCID: PMC8268215 DOI: 10.3390/cancers13133214] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Roughly 85–90% of adult and pediatric acute myeloid leukemia (AML) are CD33-positive. Gemtuzumab ozogamicin (GO), a humanized murine IgG4 anti-CD33 antibody, is the first target therapy approved in AML therapeutic scenario. This review focuses on current biological information and clinical data from several studies investigating the use of GO in patients with AML. Over the years, flow cytometry, cytogenetics, molecular techniques, and genotyping studies of CD33 SNPs have provided a comprehensive analysis of promising biomarkers for GO responses and have potentially helped to identify subgroups of patients that may benefit from GO addition to standard chemotherapies. Increased understanding of molecular mutations, altered intracellular pathways, and their potential relationship with CD33 expression may open new therapeutic landscapes based on combinatorial regimens in an AML scenario. Abstract Acute myeloid leukemia (AML), the most frequent acute leukemia in adults, has been historically treated with infusional cytarabine (ara-c) + daunorubicin (3 + 7) for at least 40 years. The first “target therapy” to be introduced was the monoclonal anti-CD33 gemtuzumab ozogamicin (GO) in 2004. Unfortunately, in 2010 it was voluntarily withdrawn from the market both for safety reasons related to potential liver toxicity and veno-occlusive disease (VOD) and because clinical studies failed to confirm the clinical benefit during induction and maintenance. Seven years later, GO was re-approved based on new data, including insights into its mechanism of action on its target receptor CD33 expressed on myeloid cells. The present review focuses on current biological information and clinical data from several studies investigating GO. Cytogenetic, molecular, and immunophenotypic data are now able to predict the potential positive advantages of GO, with the exception of high-risk AML patients who do not seem to benefit. GO can be considered a ‘repurposed drug’ that could be beneficial for some patients with AML, mostly in combination with new drugs already approved or currently in testing.
Collapse
|
13
|
Venney D, Mohd-Sarip A, Mills KI. The Impact of Epigenetic Modifications in Myeloid Malignancies. Int J Mol Sci 2021; 22:5013. [PMID: 34065087 PMCID: PMC8125972 DOI: 10.3390/ijms22095013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/19/2022] Open
Abstract
Myeloid malignancy is a broad term encapsulating myeloproliferative neoplasms (MPN), myelodysplastic syndrome (MDS) and acute myeloid leukaemia (AML). Initial studies into genomic profiles of these diseases have shown 2000 somatic mutations prevalent across the spectrum of myeloid blood disorders. Epigenetic mutations are emerging as critical components of disease progression, with mutations in genes controlling chromatin regulation and methylation/acetylation status. Genes such as DNA methyltransferase 3A (DNMT3A), ten eleven translocation methylcytosine dioxygenase 2 (TET2), additional sex combs-like 1 (ASXL1), enhancer of zeste homolog 2 (EZH2) and isocitrate dehydrogenase 1/2 (IDH1/2) show functional impact in disease pathogenesis. In this review we discuss how current knowledge relating to disease progression, mutational profile and therapeutic potential is progressing and increasing understanding of myeloid malignancies.
Collapse
Affiliation(s)
| | | | - Ken I Mills
- Patrick G Johnston Center for Cancer Research, Queens University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (D.V.); (A.M.-S.)
| |
Collapse
|
14
|
Chen S, Zeiser R. Novel Biomarkers for Outcome After Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2020; 11:1854. [PMID: 33013836 PMCID: PMC7461883 DOI: 10.3389/fimmu.2020.01854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/09/2020] [Indexed: 12/29/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a well-established curative treatment for various malignant hematological diseases. However, its clinical success is substantially limited by major complications including graft-vs.-host disease (GVHD) and relapse of the underlying disease. Although these complications are known to lead to significant morbidity and mortality, standardized pathways for risk stratification of patients undergoing allo-HSCT are lacking. Recent advances in the development of diagnostic and prognostic tools have allowed the identification of biomarkers in order to predict outcome after allo-HSCT. This review will provide a summary of clinically relevant biomarkers that have been studied to predict the development of acute GVHD, the responsiveness of affected patients to immunosuppressive treatment and the risk of non-relapse mortality. Furthermore, biomarkers associated with increased risk of relapse and subsequent mortality will be discussed.
Collapse
Affiliation(s)
- Sophia Chen
- Department of Immunology, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, New York, NY, United States.,Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Fenwarth L, Fournier E, Cheok M, Boyer T, Gonzales F, Castaigne S, Boissel N, Lambert J, Dombret H, Preudhomme C, Duployez N. Biomarkers of Gemtuzumab Ozogamicin Response for Acute Myeloid Leukemia Treatment. Int J Mol Sci 2020; 21:E5626. [PMID: 32781546 PMCID: PMC7460695 DOI: 10.3390/ijms21165626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 08/03/2020] [Indexed: 11/27/2022] Open
Abstract
Gemtuzumab ozogamicin (GO, Mylotarg®) consists of a humanized CD33-targeted antibody-drug conjugated to a calicheamicin derivative. Growing evidence of GO efficacy in acute myeloid leukemia (AML), demonstrated by improved outcomes in CD33-positive AML patients across phase I to III clinical trials, led to the Food and Drug Administration (FDA) approval on 1 September 2017 in CD33-positive AML patients aged 2 years and older. Discrepancies in GO recipients outcome have raised significant efforts to characterize biomarkers predictive of GO response and have refined the subset of patients that may strongly benefit from GO. Among them, CD33 expression levels, favorable cytogenetics (t(8;21), inv(16)/t(16;16), t(15;17)) and molecular alterations, such as NPM1, FLT3-internal tandem duplications and other signaling mutations, represent well-known candidates. Additionally, in depth analyses including minimal residual disease monitoring, stemness expression (LSC17 score), mutations or single nucleotide polymorphisms in GO pathway genes (CD33, ABCB1) and molecular-derived scores, such as the recently set up CD33_PGx6_Score, represent promising markers to enhance GO response prediction and improve patient management.
Collapse
Affiliation(s)
- Laurène Fenwarth
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (E.F.); (M.C.); (F.G.); (C.P.); (N.D.)
| | - Elise Fournier
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (E.F.); (M.C.); (F.G.); (C.P.); (N.D.)
| | - Meyling Cheok
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (E.F.); (M.C.); (F.G.); (C.P.); (N.D.)
| | - Thomas Boyer
- Laboratory of Hematology, CHU Amiens, F-80054 Amiens, France;
| | - Fanny Gonzales
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (E.F.); (M.C.); (F.G.); (C.P.); (N.D.)
| | - Sylvie Castaigne
- Department of Hematology, CH Versailles, F-78157 Le Chesnay, France; (S.C.); (J.L.)
| | - Nicolas Boissel
- Adolescent and Young Adult Hematology Unit, Hôpital Saint-Louis, AP-HP, Université de Paris, F-75010 Paris, France;
| | - Juliette Lambert
- Department of Hematology, CH Versailles, F-78157 Le Chesnay, France; (S.C.); (J.L.)
| | - Hervé Dombret
- Department of Hematology, Hôpital Saint-Louis, AP-HP, Université de Paris, F-75010 Paris, France;
| | - Claude Preudhomme
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (E.F.); (M.C.); (F.G.); (C.P.); (N.D.)
| | - Nicolas Duployez
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (E.F.); (M.C.); (F.G.); (C.P.); (N.D.)
| |
Collapse
|
16
|
Noura M, Matsuo H, Koyama A, Adachi S, Masutani H. TXNIP induces growth arrest and enhances ABT263-induced apoptosis in mixed-lineage leukemia-rearranged acute myeloid leukemia cells. FEBS Open Bio 2020; 10:1532-1541. [PMID: 32511893 PMCID: PMC7396447 DOI: 10.1002/2211-5463.12908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/26/2020] [Accepted: 06/04/2020] [Indexed: 01/20/2023] Open
Abstract
Thioredoxin‐interacting protein (TXNIP) has been widely recognized as a tumor suppressor in various cancers, including liver, breast, and thyroid cancers. Although TXNIP is epigenetically silenced in acute myeloid leukemia (AML) cells, as in many cancer cells, its role in leukemogenesis remains elusive. Mixed‐lineage leukemia (MLL) gene rearrangements in AML are associated with poor prognosis, and the development of a new treatment method is eagerly anticipated. In this study, we first reveal that lower expression of TXNIP is correlated with shortened overall survival periods in AML patients. Moreover, we demonstrated that TXNIP overexpression significantly suppresses proliferation in AML cells harboring MLL fusion genes. TXNIP promotes autophagy by increasing expression of the autophagy protein, Beclin 1, and lipidation of LC3B. We also show that TXNIP overexpression combined with ABT263, a potent inhibitor of Bcl‐2 and Bcl‐xL, is highly effective at inducing cell death in MLL‐rearranged (MLL‐r) AML cells. In summary, this study provides insights into the molecular mechanism of TXNIP‐mediated tumor suppression and furthermore underscores the potential of TXNIP as a promising therapeutic target for MLL‐r AML.
Collapse
Affiliation(s)
- Mina Noura
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Clinical Laboratory Sciences, Tenri Health Care University, Tenri, Japan
| | - Hidemasa Matsuo
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Asami Koyama
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Masutani
- Department of Clinical Laboratory Sciences, Tenri Health Care University, Tenri, Japan
| |
Collapse
|
17
|
Wang SY, Cheng WY, Mao YF, Zhu YM, Liu FJ, Ma TT, Shen Y. Genetic alteration patterns and clinical outcomes of elderly and secondary acute myeloid leukemia. Hematol Oncol 2019; 37:456-463. [PMID: 31348835 PMCID: PMC6899678 DOI: 10.1002/hon.2656] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/16/2019] [Accepted: 07/20/2019] [Indexed: 12/19/2022]
Abstract
To illustrate the clinical and genetic features of elderly and secondary acute myeloid leukemia (AML) patients, we compared 145 elderly AML (e‐AML) and 55 secondary AML (s‐AML) patients with 451 young de novo AML patients. Both e‐AML and s‐AML patients showed lower white blood cell (WBC) and bone marrow (BM) blasts at diagnosis. NPM1, DNMT3A, and IDH2 mutations were more common while biallelic CEBPA and IDH1 mutations were less seen in e‐AML patients. s‐AML patients carried a higher frequency of KMT2A‐AF9. In treatment response and survival, e/s‐AML conferred a lower complete remission (CR) rate and shorter duration of event‐free survival (EFS) and overall survival (OS) compared with young patients. In multivariate analysis, s‐AML was an independent risk factor for OS but not EFS in the whole cohort. Importantly, intensive therapy tended to improve the survival of e/s‐AML patients without increasing the risk of early death, and hematopoietic stem cell transplantation (HSCT) could rescue the prognosis of s‐AML, which should be recommended for the treatment of fit patients.
Collapse
Affiliation(s)
- Shi-Yang Wang
- Shanghai Institute of Hematology, Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Yan Cheng
- Shanghai Institute of Hematology, Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Fei Mao
- Shanghai Institute of Hematology, Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Mei Zhu
- Shanghai Institute of Hematology, Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu-Jia Liu
- Shanghai Institute of Hematology, Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting-Ting Ma
- Shanghai Institute of Hematology, Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Shen
- Shanghai Institute of Hematology, Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Bussaglia E, Antón R, Nomdedéu JF, Fuentes-Prior P. TET2 missense variants in human neoplasia. A proposal of structural and functional classification. Mol Genet Genomic Med 2019; 7:e00772. [PMID: 31187595 PMCID: PMC6625141 DOI: 10.1002/mgg3.772] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background The human TET2 gene plays a pivotal role in the epigenetic regulation of normal and malignant hematopoiesis. Somatic TET2 mutations have been repeatedly identified in age‐related clonal hematopoiesis and in myeloid neoplasms ranging from acute myeloid leukemia (AML) to myeloproliferative neoplasms. However, there have been no attempts to systematically explore the structural and functional consequences of the hundreds of TET2 missense variants reported to date. Methods We have sequenced the TET2 gene in 189 Spanish AML patients using Sanger sequencing and NGS protocols. Next, we performed a thorough bioinformatics analysis of TET2 protein and of the expected impact of all reported TET2 missense variants on protein structure and function, exploiting available structure‐and‐function information as well as 3D structure prediction tools. Results We have identified 38 TET2 allelic variants in the studied patients, including two frequent SNPs: p.G355D (10 cases) and p.I1762V (28 cases). Four of the detected mutations are reported here for the first time: c.122C>T (p.P41L), c.4535C>G (p.A1512G), c.4760A>G (p.D1587G), and c.5087A>T (p.Y1696F). We predict a complex multidomain architecture for the noncatalytic regions of TET2, and in particular the presence of well‐conserved α+β globular domains immediately preceding and following the actual catalytic unit. Further, we provide a rigorous interpretation of over 430 missense SNVs that affect the TET2 catalytic domain, and we hypothesize explanations for ~700 additional variants found within the regulatory regions of the protein. Finally, we propose a systematic classification of all missense mutants and SNPs reported to date into three major categories (severe, moderate, and mild), based on their predicted structural and functional impact. Conclusions The proposed classification of missense TET2 variants would help to assess their clinical impact on human neoplasia and may guide future structure‐and‐function investigations of TET family members.
Collapse
Affiliation(s)
- Elena Bussaglia
- Hematology Department and Diagnostic Hematology Group, Barcelona, Spain
| | - Rosa Antón
- Molecular Bases of Disease, The Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Josep F Nomdedéu
- Hematology Department and Diagnostic Hematology Group, Barcelona, Spain
| | - Pablo Fuentes-Prior
- Molecular Bases of Disease, The Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
19
|
Wang YM, Mo JQ, Kuo DJ, Wong V. MLL rearranged acute lymphoblastic leukaemia presenting as a maxillary sinus mass with a discordant immunophenotypic profile from the bone marrow. BMJ Case Rep 2019; 12:12/2/e227400. [PMID: 30772833 DOI: 10.1136/bcr-2018-227400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We describe an unusual case of pre-B lymphoblastic leukaemia presenting with a unilateral maxillary sinus mass in which biopsies of the primary mass and the bone marrow demonstrated conflicting immunophenotyping results. The extramedullary mass was consistent with a precursor B-cell malignancy, while the bone marrow was initially reported as a possible mature B-cell malignancy. The treatments for the two are fundamentally different, which necessitated a delay in the initiation of his chemotherapy until a clear diagnosis was made. Mixed lineage leukaemia gene rearrangement was confirmed by fluorescence in situ hybridisation in both the primary mass and bone marrow, which unified the diagnosis as pre-B acute lymphoblastic leukaemia given the common cytogenetic feature.
Collapse
Affiliation(s)
- Yunzu Michele Wang
- Department of Pediatric Hematology-Oncology, University of California San Diego, San Diego, California, USA
| | - Jun Qin Mo
- Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Dennis John Kuo
- Department of Pediatric Hematology-Oncology, University of California San Diego, San Diego, California, USA
| | - Victor Wong
- Department of Pediatric Hematology-Oncology, University of California San Diego, San Diego, California, USA
| |
Collapse
|
20
|
Shahjahani M, Hadad EH, Azizidoost S, Nezhad KC, Shahrabi S. Complex karyotype in myelodysplastic syndromes: Diagnostic procedure and prognostic susceptibility. Oncol Rev 2019; 13:389. [PMID: 30858933 PMCID: PMC6379782 DOI: 10.4081/oncol.2019.389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 01/03/2019] [Indexed: 12/22/2022] Open
Abstract
Complex karyotype (CK) is a poor prognosis factor in hematological malignancies. Studies have shown that the presence of CK in myelodysplastic syndrome (MDS) can be associated with MDS progression to acute myeloid leukemia. The goal of this review was to examine the relationship between different types of CK with MDS, as well as its possible role in the deterioration and progression of MDS to leukemia. The content used in this paper has been obtained by a PubMed and Google Scholar search of English language papers (1975-2018) using the terms complex karyotype and myelodysplastic syndromes. A single independent abnormality can be associated with a good prognosis. However, the coexistence of a series of abnormalities can lead to CK, which is associated with the deterioration of MDS and its progression to leukemia. Therefore, CK may be referred to as a prognostic factor in MDS. The detection of independent cytogenetic disorders that altogether can result in CK could be used as a prognostic model for laboratory and clinical use.
Collapse
Affiliation(s)
- Mohammad Shahjahani
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
| | - Elham Homaei Hadad
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
| | - Shirin Azizidoost
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
| | | | - Saeid Shahrabi
- Thalassemia & Hemoglobinopathy Research center, research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
21
|
Abstract
Immunophenotyping by multiparameter flow cytometry is a rapid and efficient technique to simultaneously assess and correlate multiple individual cell properties like size and internal complexity along with antigen expression in a population of cells. This method is utilized for rapid characterization of the blasts and classification of acute myeloid leukemia (AML), in both the peripheral blood (PB) and bone marrow (BM). This technique is not only useful in the initial diagnosis but also in monitoring and determining prognosis of the disease through minimal residual disease (MRD) testing. This chapter provides an overview of procedures for specimen processing, staining, and immunophenotyping of AML and describes the principles of data analysis for AML classification and MRD testing.
Collapse
|
22
|
Cui P, Zhang Y, Cui M, Li Z, Ma G, Wang R, Wang N, Huang S, Gao J. Leukemia cells impair normal hematopoiesis and induce functionally loss of hematopoietic stem cells through immune cells and inflammation. Leuk Res 2018; 65:49-54. [PMID: 29306107 DOI: 10.1016/j.leukres.2018.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/21/2017] [Accepted: 01/01/2018] [Indexed: 11/20/2022]
Abstract
Bone marrow (BM) failure is often seen in leukemia patients, indicating an abnormal hematopoietic process. However, hematopoiesis in leukemic milieus is largely unknown. In the present study, we utilized one of the most frequent leukemogenic translocations MLL-AF9 to induce leukemia and investigated the hematopoiesis and the activity of hematopoietic stem and progenitor cells (HSPCs) in a leukemic milieu. We found that the phenotypes of the non-leukemic population in leukemic BM were drastically different than normal BM, including blockage of differentiation and a drastically reduced Lin-/Sca+/c-kit+ (LSK) population that contains all HSPCs in leukemic BM. Further, transplantation assays demonstrated that stem cell function of HSPCs from leukemic BM was significantly compromised. Intriguingly, BM from a patient-derived xenograft leukemia model and from immunocompromised mice transplanted with murine MLL-AF9 cells, showed comparable percentage of hematopoietic stem cells (HSCs) to normal controls, indicating that an immunocompetent microenvironment is critical for leukemia-induced loss of HSPCs. Mechanistically, we found that the non-leukemic cells from leukemic BM possessed a more inflammatory profile than either leukemic cells or normal BM counterparts. Co-culturing or co-transplantation with non-leukemic cells from leukemic BM impaired the stem cell function of normal HSPCs in vitro and in vivo respectively, suggesting that the highly inflammatory non-leukemic population in leukemic BM not only is functionally abnormal but displayed a 'leukemia-like' characteristic. Finally, we tested the effect of the anti-inflammation drug diclofenac on leukemia mice. However, no phenotypic changes of HSPCs were observed upon diclofenac treatment due to only mild repression of inflammatory genes by diclofenac, further indicating that inflammation is a powerful negative regulator of HSPCs. Together, our results suggest that leukemia impairs normal hematopoiesis and inflammation as well as immune cells play a critical role in leukemia-induced BM failure.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Coculture Techniques
- Diclofenac/pharmacology
- Hematopoiesis
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/pathology
- Humans
- Immunocompetence
- Inflammation/immunology
- Inflammation/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Myeloid-Lymphoid Leukemia Protein/genetics
- Oncogene Proteins, Fusion/genetics
- Tumor Microenvironment
Collapse
Affiliation(s)
- Ping Cui
- Department of Pathology, Cangzhou Medical College, China.
| | - Yuhua Zhang
- Department of Pathology, Cangzhou Medical College, China
| | - Maoxiang Cui
- Department of Pathology, Cangzhou Medical College, China
| | - Zhihong Li
- Department of Pathology, Cangzhou Medical College, China
| | - Guang Ma
- Department of Pathology, Cangzhou Medical College, China
| | - Rufeng Wang
- Department of Pathology, Cangzhou Medical College, China
| | - Ning Wang
- Department of Pathology, Cangzhou Medical College, China
| | - Shujuan Huang
- Department of Pathology, Cangzhou Medical College, China
| | - Jie Gao
- Department of Pathology, Cangzhou Medical College, China
| |
Collapse
|
23
|
|
24
|
Laing AA, Harrison CJ, Gibson BE, Keeshan K. Unlocking the potential of anti-CD33 therapy in adult and childhood acute myeloid leukemia. Exp Hematol 2017; 54:40-50. [DOI: 10.1016/j.exphem.2017.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 10/19/2022]
|
25
|
Upregulation of CD11b and CD86 through LSD1 inhibition promotes myeloid differentiation and suppresses cell proliferation in human monocytic leukemia cells. Oncotarget 2017; 8:85085-85101. [PMID: 29156705 PMCID: PMC5689595 DOI: 10.18632/oncotarget.18564] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 06/02/2017] [Indexed: 12/11/2022] Open
Abstract
LSD1 (Lysine Specific Demethylase1)/KDM1A (Lysine Demethylase 1A), a flavin adenine dinucleotide (FAD)-dependent histone H3K4/K9 demethylase, sustains oncogenic potential of leukemia stem cells in primary human leukemia cells. However, the pro-differentiation and anti-proliferation effects of LSD1 inhibition in acute myeloid leukemia (AML) are not yet fully understood. Here, we report that small hairpin RNA (shRNA) mediated LSD1 inhibition causes a remarkable transcriptional activation of myeloid lineage marker genes (CD11b/ITGAM and CD86), reduction of cell proliferation and decrease of clonogenic ability of human AML cells. Cell surface expression of CD11b and CD86 is significantly and dynamically increased in human AML cells upon sustained LSD1 inhibition. Chromatin immunoprecipitation and quantitative PCR (ChIP-qPCR) analyses of histone marks revealed that there is a specific increase of H3K4me2 modification and an accompanied increase of H3K4me3 modification at the respective CD11b and CD86 promoter region, whereas the global H3K4me2 level remains constant. Consistently, inhibition of LSD1 in vivo significantly blocks tumor growth and induces a prominent increase of CD11b and CD86. Taken together, our results demonstrate the anti-tumor properties of LSD1 inhibition on human AML cell line and mouse xenograft model. Our findings provide mechanistic insights into the LSD1 functions in controlling both differentiation and proliferation in AML.
Collapse
|
26
|
Lee JM, Kim IS, Lee JN, Park SH, Kim HH, Chang CL, Lee EY, Kim HR, Oh SH, Song SA. Acute Myeloid Leukemia With MLL Rearrangement and CD4+/CD56+ Expression can be Misdiagnosed as Blastic Plasmacytoid Dendritic Cell Neoplasm: Two Case Reports. Ann Lab Med 2017; 36:494-7. [PMID: 27374717 PMCID: PMC4940495 DOI: 10.3343/alm.2016.36.5.494] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/27/2016] [Accepted: 06/02/2016] [Indexed: 11/22/2022] Open
Affiliation(s)
- Ju Mee Lee
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - In Suk Kim
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea.
| | - Jeong Nyeo Lee
- Department of Laboratory Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea.
| | - Sang Hyuk Park
- Department of Laboratory Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Hyung Hoi Kim
- Department of Laboratory Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Chulhun L Chang
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Eun Yup Lee
- Department of Laboratory Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Hye Ran Kim
- Department of Laboratory Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Seung Hwan Oh
- Department of Laboratory Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Sae Am Song
- Department of Laboratory Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
27
|
Expression of immunoproteasome genes is regulated by cell-intrinsic and -extrinsic factors in human cancers. Sci Rep 2016; 6:34019. [PMID: 27659694 PMCID: PMC5034284 DOI: 10.1038/srep34019] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/06/2016] [Indexed: 01/07/2023] Open
Abstract
Based on transcriptomic analyses of thousands of samples from The Cancer Genome Atlas, we report that expression of constitutive proteasome (CP) genes (PSMB5, PSMB6, PSMB7) and immunoproteasome (IP) genes (PSMB8, PSMB9, PSMB10) is increased in most cancer types. In breast cancer, expression of IP genes was determined by the abundance of tumor infiltrating lymphocytes and high expression of IP genes was associated with longer survival. In contrast, IP upregulation in acute myeloid leukemia (AML) was a cell-intrinsic feature that was not associated with longer survival. Expression of IP genes in AML was IFN-independent, correlated with the methylation status of IP genes, and was particularly high in AML with an M5 phenotype and/or MLL rearrangement. Notably, PSMB8 inhibition led to accumulation of polyubiquitinated proteins and cell death in IPhigh but not IPlow AML cells. Co-clustering analysis revealed that genes correlated with IP subunits in non-M5 AMLs were primarily implicated in immune processes. However, in M5 AML, IP genes were primarily co-regulated with genes involved in cell metabolism and proliferation, mitochondrial activity and stress responses. We conclude that M5 AML cells can upregulate IP genes in a cell-intrinsic manner in order to resist cell stress.
Collapse
|
28
|
Prada-Arismendy J, Arroyave JC, Röthlisberger S. Molecular biomarkers in acute myeloid leukemia. Blood Rev 2016; 31:63-76. [PMID: 27639498 DOI: 10.1016/j.blre.2016.08.005] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. The pathophysiology of this disease is just beginning to be understood at the cellular and molecular level, and currently cytogenetic markers are the most important for risk stratification and treatment of AML patients. However, with the advent of new technologies, the detection of other molecular markers such as point mutations and characterization of epigenetic and proteomic profiles, have begun to play an important role in how the disease is approached. Recent evidence shows that the identification of new AML biomarkers contributes to a better understanding of the molecular basis of the disease, is significantly useful in screening, diagnosis, prognosis and monitoring of AML, as well as the possibility of predicting each individual's response to treatment. This review summarizes the most relevant molecular (genetic, epigenetic, and protein) biomarkers associated with acute myeloid leukemia and discusses their clinical importance in terms of risk prediction, diagnosis and prognosis.
Collapse
MESH Headings
- Biomarkers, Tumor
- DNA Methylation
- Disease Susceptibility
- Epigenesis, Genetic
- Genetic Variation
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Mutation
- Prognosis
Collapse
Affiliation(s)
- Jeanette Prada-Arismendy
- Grupo de Investigación e Innovación Biomédica, Instituto Tecnológico Metropolitano, Medellín, Colombia.
| | - Johanna C Arroyave
- Grupo de Investigación e Innovación Biomédica, Instituto Tecnológico Metropolitano, Medellín, Colombia
| | - Sarah Röthlisberger
- Grupo de Investigación e Innovación Biomédica, Instituto Tecnológico Metropolitano, Medellín, Colombia
| |
Collapse
|
29
|
Nomdedéu JF, Puigdecanet E, Bussaglia E, Hernández JJ, Carricondo M, Estivill C, Martí-Tutusaus JM, Tormo M, Zamora L, Serrano E, Perea G, de Llano MPQ, García A, Sánchez-Ortega I, Ribera JM, Nonell L, Aventin A, Solé F, Brunet MS, Sierra J. Feasibility of the AML profiler (Skyline™ Array) for patient risk stratification in a multicentre trial: a preliminary comparison with the conventional approach. Hematol Oncol 2016; 35:778-788. [PMID: 27140599 DOI: 10.1002/hon.2304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/15/2016] [Accepted: 03/31/2016] [Indexed: 12/11/2022]
Abstract
Deoxyribonucleic acid microarrays allow researchers to measure mRNA levels of thousands of genes in a single experiment and could be useful for diagnostic purposes in patients with acute myeloid leukaemia (AML). We assessed the feasibility of the AML profiler (Skyline™ Array) in genetic stratification of patients with de novo AML and compared the results with those obtained using the standard cytogenetic and molecular approach. Diagnostic bone marrow from 31 consecutive de novo AML cases was used to test MLL-PTD, FLT3-ITD and TKD, NPM1 and CEBPAdm mutations. Purified RNA was used to assess RUNX1-RUNX1T1, PML-RARα and CBFβ-MYH11 rearrangements. RNA remnants underwent gene expression profiling analysis using the AML profiler, which detects chromosomal aberrations: t(8;21), t(15;17), inv(16), mutations (CEBPAdm, ABD-NPM1) and BAALC and EVI1 expression. Thirty cases were successfully analysed with both methods. Five cases had FLT3-ITD. In one case, a t(8;21) was correctly detected by both methods. Four cases had inv(16); in one, the RNA quality was unsatisfactory and it was not hybridized, and in the other three, the AML profiler detected the genetic lesion - this being a rare type I translocation in one case. Two cases with acute promyelocytic leukaemia were diagnosed by both methods. Results for NPM1 mutations were concordant in all but two cases (2/11, non-ABD mutations). Analysis of costs and turnaround times showed that the AML profiler was no more expensive than the conventional molecular approach. These results suggest that the AML profiler could be useful in multicentre trials to rapidly identify patients with AML with a good prognosis. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Josep F Nomdedéu
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Eulalia Puigdecanet
- Servei d'Analisi de Microarrays, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Elena Bussaglia
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Maite Carricondo
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Camino Estivill
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Mar Tormo
- Hematology Department, Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Lurdes Zamora
- Hematology Department, Institut Recerca contra la Leucemia Josep Carreras (IJC), ICO Badalona Hospital Germans Trias i Pujol, Badalona, Spain
| | - Elena Serrano
- Bioinformatic Platform, IIB Sant Pau, Barcelona, Spain
| | - Granada Perea
- Laboraotory Service, UDIAT-CD, Parc Taulí Hospital Universitari, Sabadell, Spain
| | | | - Antoni García
- Hematology Department, Hospital Arnau de Vilanova, Lleida, Spain
| | | | - Josep Maria Ribera
- Hematology Department, Institut Recerca contra la Leucemia Josep Carreras (IJC), ICO Badalona Hospital Germans Trias i Pujol, Badalona, Spain
| | - Lara Nonell
- Servei d'Analisi de Microarrays, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Anna Aventin
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francesc Solé
- Hematology Department, Institut Recerca contra la Leucemia Josep Carreras (IJC), ICO Badalona Hospital Germans Trias i Pujol, Badalona, Spain
| | - Maria Salut Brunet
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jorge Sierra
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
30
|
Hou HA, Tien HF. Mutations in epigenetic modifiers in acute myeloid leukemia and their clinical utility. Expert Rev Hematol 2016; 9:447-69. [DOI: 10.1586/17474086.2016.1144469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
31
|
Cao L, Wang N, Pan J, Hu S, Zhao W, He H, Wang Y, Gu G, Chai Y. Clinical significance of microRNA-34b expression in pediatric acute leukemia. Mol Med Rep 2016; 13:2777-84. [PMID: 26861642 DOI: 10.3892/mmr.2016.4876] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 12/03/2015] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to explore the function of miR‑34b promoter methylation in cell proliferation in children's acute leukemia. Quantitative PCR and methylation‑specific PCR were performed to measure the levels of miR‑34b and its promoter methylation in normal cells, eight leukemia cell lines as well as primary leukemic cells isolated from patients newly diagnosed with acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML) and mixed lymphocytic lymphoma. miR‑34b levels in leukemia cell lines and primary leukemic cells were significantly lower than those in normal cells. The miR‑34b promoter was found to be methylated in all leukemia cell lines, 24 of 31 ALL patients and 8 of 19 AML patients, but not in the 23 normal controls. miR‑34b expression and methylation of its promoter were not associated with most clinical parameters assessed; however, miR‑34b levels in prednisone‑sensitive ALL were significantly different from those in insensitive ALL. A cell counting kit‑8 assay showed that transfection of miR‑34b mimics into K562 cells inhibited their proliferation. Furthermore, treatment with the demethylating agent 5‑aza‑2‑deoxycytidine significantly enhanced miR‑34b expression levels and decreased the methylation status of its promoter in HL‑60 and K562 cells. In conclusion, the results of the present study indicated that in pediatric leukemia cells and leukemia cell lines, the expression of miR‑34b is inhibited by methylation of its promoter, which impairs the restraining effects of miR‑34b on cell proliferation. It was also indicated that the expression of miR‑34b in ALL patients may affect their response to early treatments.
Collapse
Affiliation(s)
- Lan Cao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Na Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Jian Pan
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Shaoyan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Wenli Zhao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Hailong He
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Yi Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Guixiong Gu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Yihuan Chai
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| |
Collapse
|
32
|
García-Dabrio MC, Hoyos M, Brunet S, Tormo M, Ribera JM, Esteve J, Gallardo D, Duarte RF, de Llano MPQ, Bargay J, Martí-Tutusaus JM, Heras I, Garcia A, Salamero O, Aventin A, Lecrevisse Q, Orfao A, Sierra J, Nomdedéu JF. Complex measurements may be required to establish the prognostic impact of immunophenotypic markers in AML. Am J Clin Pathol 2015; 144:484-92. [PMID: 26276779 DOI: 10.1309/ajcprl6xsvfmlh9v] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES The prognostic impact of immunophenotypic markers in acute myeloid leukemia (AML) is controversial. METHODS We retrospectively analyzed the value of CD34, CD117, CD7, and CD123 expression in a consecutive series of 592 adult patients with de novo AML. RESULTS CD34+ measured as a percentage (≥2.88%) and CD34 mean fluorescence intensity (MFI) (≥146.79, arbitrary units [AU]) expression had a prognostic impact in terms of overall survival (OS; P = .005, P = .003), leukemia-free survival (LFS; P = .011, P < .001), and cumulative incidence of relapse (CIR; P = .014, P =. 001). The percentage of CD117+ cells (61.29%) was associated with shorter LFS (P =. 043), and CD117 MFI (≥284.01 AU) was associated with a shorter OS (P =. 033) and LFS (P =. 028). In the multivariate analysis, high CD34 MFI retained the independent value as predictor of LFS and CIR (P =. 012; hazard ratio [HR], 1.59; 95% confidence interval [CI], 1.11-2.28 and P =. 045; HR, 1.58; 95% CI, 1.01-2.46). CONCLUSIONS CD34 positivity threshold with prognostic relevance is low (3% positive cells). Immunophenotypic findings in AML probably could only be fully exploited after a complex analysis that takes into account unconventional thresholds and the MFI.
Collapse
Affiliation(s)
| | - Montserrat Hoyos
- Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Universitat Autònoma de Barcelona, Spain
| | - Salut Brunet
- Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Universitat Autònoma de Barcelona, Spain
| | - Mar Tormo
- Department of Hematology, Hospital Clínic, Valencia, Spain
| | - Josep-Maria Ribera
- Department of Hematology, Hospital ICO Germans Trias i Pujol, Badalona, Spain
| | - Jordi Esteve
- Department of Hematology, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - David Gallardo
- Department of Hematology, Hospital ICO Hospital Josep Trueta, Girona, Spain
| | - Rafael F. Duarte
- Department of Hematology, Hospital ICO Duran i Reynalds, L’Hospitalet, Barcelona, Spain
| | | | - Joan Bargay
- Department of Hematology, Hospital Sont Llatzer, Palma de Mallorca, Spain
| | | | - Inmaculada Heras
- Department of Hematology, Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Antoni Garcia
- Department of Hematology, Hospital Arnau de Vilanova, Lleida, Spain
| | - Olga Salamero
- Department of Hematology, Hospital de la Vall d’ Hebrón, Barcelona, Spain; and
| | | | - Quentin Lecrevisse
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain, on behalf of the Spanish CETLAM Group
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca (USAL) and Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain, on behalf of the Spanish CETLAM Group
| | - Jorge Sierra
- Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Universitat Autònoma de Barcelona, Spain
| | | |
Collapse
|
33
|
Hagag AA, Shebl SS, El-Fadaly NH. Frequency of 11q23/MLL gene rearrangement in Egyptian childhood acute myeloblastic leukemia: Biologic and clinical significance. South Asian J Cancer 2014; 3:206-8. [PMID: 25422805 PMCID: PMC4236697 DOI: 10.4103/2278-330x.142964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Molecular cytogenetic abnormalities involving 11q23 are among the most common cytogenetic abnormalities in acute myeloid leukemia (AML) patients. AIM OF THE WORK we aimed to evaluate the frequency of MLL/AF9 fusion gene in de novo AML patients, its impact on clinical features, and its prognostic significance. PATIENTS AND METHODS Twenty-eight children patients with AML and 20 healthy controls were subjected to complete clinical examination and laboratory investigations including, complete hemogram and bone marrow (BM) examination. Diagnosis was based on FAB morphologic and immunophenotypic criteria. Detection of (MLL/AF9) fusion gene was assessed by dual color fluorescent in situ hybridization (FISH). Follow-up were carried out clinically and by blast count in BM, and response to therapy to detect the outcome of the disease. RESULTS The incidence of MLL-fusion gene MLL/AF9 in AML cases was about (6/28) (21%). Four patients with MLL/AF9 fusion gene were newly diagnosed, two cases were at relapse and no patient at remission showed positivity. As regard the clinical outcome, five out of six MLL positive cases died, three of them during induction and two during relapse. The FAB AML subtypes with MLL/AF9 fusion were one M2, three M4, and two M5. CONCLUSION MLL-fusion gene MLL/AF9 was found in about 21% of studied AML patients when assessed by FISH technique and this is of high clinical relevance as most of these abnormalities have been associated with poor prognosis.
Collapse
Affiliation(s)
| | - Shebl S Shebl
- Department of Pediatrics, Pediatric Hematology-Oncology Unit, Egypt
| | | |
Collapse
|
34
|
Gaikwad A, Bonifant CL, Cubbage M, Goltsova T, Mudannayake M, Ringrose J, Punia J, Lopez-Terrada D, Sheehan AM. Detection of Lymphoid and Myeloid Lineages in Infantile B-Cell Acute Lymphoblastic Leukemia With Mixed-Lineage Leukemia Rearrangement by Use of Flow Cytometry and Cytogenetic Analysis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2014; 14 Suppl:S2-5. [DOI: 10.1016/j.clml.2014.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/03/2014] [Accepted: 06/04/2014] [Indexed: 11/25/2022]
|
35
|
Chandran P, Kavalakatt A, Malarvizhi GL, Vasanthakumari DRVN, Retnakumari AP, Sidharthan N, Pavithran K, Nair S, Koyakutty M. Epigenetics targeted protein-vorinostat nanomedicine inducing apoptosis in heterogeneous population of primary acute myeloid leukemia cells including refractory and relapsed cases. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:721-32. [DOI: 10.1016/j.nano.2013.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 09/23/2013] [Accepted: 09/24/2013] [Indexed: 12/17/2022]
|
36
|
Ikoma MRV, Sandes AF, Thiago LS, Cavalcanti Júnior GB, Lorand-Metze IGH, Costa ES, Pimenta G, Santos-Silva MC, Bacal NS, Yamamoto M, Souto EX. First proposed panels on acute leukemia for four-color immunophenotyping by flow cytometry from the Brazilian group of flow cytometry-GBCFLUX. CYTOMETRY PART B-CLINICAL CYTOMETRY 2014; 88:194-203. [DOI: 10.1002/cyto.b.21175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Maura R. V. Ikoma
- Hospital Amaral Carvalho; Laboratório de Citometria de Fluxo do Hemonúcleo Regional de Jau; São Paulo Brazil
| | - Alex F. Sandes
- Division of Hematology; Fleury Group; São Paulo Brazil
- Division of Hematology and Blood Transfusion Medicine; Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP-EPM); Brazil
| | - Leandro S. Thiago
- Brazilian National Cancer Institute (INCa), Cancer Research Center; Rio de Janeiro Brazil
| | | | | | - Elaine S. Costa
- Pediatric Institute IPPMG, Universidade Federal do Rio de Janeiro (UFRJ); Rio de Janeiro Brazil
| | - Glicinia Pimenta
- Universidade Federal do Rio de Janeiro; Brazil
- Laboratório Diagnósticos da América; Rio de Janeiro Brazil
| | | | - Nydia S. Bacal
- Hospital Israelita Albert Einstein; São Paulo Brazil
- Centro de Hematologia de; São Paulo
| | - Mihoko Yamamoto
- Division of Hematology and Blood Transfusion Medicine; Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP-EPM); Brazil
| | | | | |
Collapse
|
37
|
Chemical genomic screening identifies LY294002 as a modulator of glucocorticoid resistance in MLL-rearranged infant ALL. Leukemia 2013; 28:761-9. [PMID: 23958920 DOI: 10.1038/leu.2013.245] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 08/05/2013] [Accepted: 08/07/2013] [Indexed: 11/09/2022]
Abstract
Successful treatment results for MLL-rearranged Acute Lymphoblastic Leukemia (ALL) in infants remain difficult to achieve. Significantly contributing to therapy failure is poor response to glucocorticoids (GCs), like prednisone. Thus, overcoming resistance to these drugs may be a crucial step towards improving prognosis. We defined a gene signature that accurately discriminates between prednisolone-resistant and prednisolone-sensitive MLL-rearranged infant ALL patient samples. In the current study, we applied Connectivity Map analysis to perform an in silico screening for agents capable of reversing the prednisolone-resistance profile and induce sensitivity. These analyses revealed that LY294002, a PI3K inhibitor, would potentially fulfill this task. Subsequent validation experiments demonstrated that indeed LY294002, and other known PI3K inhibitors, markedly sensitized otherwise resistant MLL-rearranged ALL cells to prednisolone in vitro. Using quantitative RT-PCR analyses, we validated the modulating effects of the PI3K inhibitors on the expression of the genes present in our prednisolone-resistance profile. Interestingly, prednisolone-sensitizing actions may be mediated by inhibition of FCGR1B. Moreover, only high-level expression of FCGR1B showed to be predictive for a poor prognosis and shRNA-mediated knock-down of FCGR1B led to in vitro prednisolone sensitization. Thus, implementing FDA-approved PI3K inhibitors in current treatments may potentially improve the GC response and prognosis in patients with MLL-rearranged ALL.
Collapse
|
38
|
Takahashi S. Epigenetic aberrations in myeloid malignancies (Review). Int J Mol Med 2013; 32:532-8. [PMID: 23760684 DOI: 10.3892/ijmm.2013.1417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/16/2013] [Indexed: 11/05/2022] Open
Abstract
The development of novel technologies, such as massively parallel DNA sequencing, has led to the identification of several novel recurrent gene mutations, such as DNA methyltransferase (Dnmt)3a, ten-eleven-translocation oncogene family member 2 (TET2), isocitrate dehydrogenase (IDH)1/2, additional sex comb-like 1 (ASXL1), enhancer of zeste homolog 2 (EZH2) and ubiquitously transcribed tetratricopeptide repeat X chromosome (UTX) mutations in acute myeloid leukemia (AML) and other myeloid malignancies. These findings strongly suggest a link between recurrent genetic alterations and aberrant epigenetic regulations, resulting from an abnormal DNA methylation and histone modification status. This review focuses on the current findings of aberrant epigenetic signatures by these newly described genetic alterations. Moreover, epigenetic aberrations resulting from transcription factor aberrations, such as mixed lineage leukemia (MLL) rearrangement, ecotropic viral integration site 1 (Evi1) overexpression, chromosomal translocations and the downregulation of PU.1 are also described.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- Division of Hematology, Kitasato University School of Allied Health Sciences; Division of Molecular Hematology, Kitasato University Graduate School of Medical Sciences, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
39
|
De Santis GC, Benicio MTL, Oliveira LC, Falcão RP, Rego EM. Genetic mutations in patients with acute myeloid leukemia and leukostasis. Acta Haematol 2013; 130:95-7. [PMID: 23548579 DOI: 10.1159/000346442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/06/2012] [Indexed: 01/05/2023]
Affiliation(s)
- Gil C De Santis
- Center for Cell-Based Therapy, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | | | | |
Collapse
|
40
|
Abstract
Transcription factors critical for normal hematopoietic stem cell functions are frequently mutated in acute leukemia leading to an aberrant re-programming of normal hematopoietic progenitor/stem cells into leukemic stem cells. Among them, re-arrangements of the mixed lineage leukemia gene (MLL), including chimeric fusion, partial tandem duplication (PTD), amplification and internal exonic deletion, represent one of the most common recurring oncogenic events and associate with very poor prognosis in human leukemias. Extensive research on wild type MLL and MLL-fusions has significant advanced our knowledge about their functions in normal and malignant hematopoiesis, which also provides a framework for the underlying pathogenic role of MLL re-arrangements in human leukemias. In contrast, research progress on MLL-PTD, MLL amplification and internal exonic deletion remains stagnant, in particular for the last two abnormalities where mouse model is not yet available. In this article, we will review the key features of both wild-type and re-arranged MLL proteins with particular focuses on MLL-PTD and MLL amplification for their roles in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Bon Ham Yip
- Leukemia and Stem Cell Biology Lab, Department of Haematological Medicine, King's College London, Denmark Hill, London SE5 9NU, UK
| | | |
Collapse
|
41
|
Infant acute leukemia with lineage switch at relapse expressing a novel t(4;11)(q21;q23) MLL-AF4 fusion transcript. REV ROMANA MED LAB 2013. [DOI: 10.2478/rrlm-2013-0017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
42
|
Mll partial tandem duplication and Flt3 internal tandem duplication in a double knock-in mouse recapitulates features of counterpart human acute myeloid leukemias. Blood 2012; 120:1130-6. [PMID: 22674806 DOI: 10.1182/blood-2012-03-415067] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The MLL-partial tandem duplication (PTD) associates with high-risk cytogenetically normal acute myeloid leukemia (AML). Concurrent presence of FLT3-internal tandem duplication (ITD) is observed in 25% of patients with MLL-PTD AML. However, mice expressing either Mll-PTD or Flt3-ITD do not develop AML, suggesting that 2 mutations are necessary for the AML phenotype. Thus, we generated a mouse expressing both Mll-PTD and Flt3-ITD. Mll(PTD/WT):Flt3(ITD/WT) mice developed acute leukemia with 100% penetrance, at a median of 49 weeks. As in human MLL-PTD and/or the FLT3-ITD AML, mouse blasts exhibited normal cytogenetics, decreased Mll-WT-to-Mll-PTD ratio, loss of the Flt3-WT allele, and increased total Flt3. Highlighting the adverse impact of FLT3-ITD dosage on patient survival, mice with homozygous Flt3-ITD alleles, Mll(PTD/WT):Flt3(ITD/ITD), demonstrated a nearly 30-week reduction in latency to overt AML. Here we demonstrate, for the first time, that Mll-PTD contributes to leukemogenesis as a gain-of-function mutation and describe a novel murine model closely recapitulating human AML.
Collapse
|
43
|
Ruggeri A, Michel G, Dalle JH, Caniglia M, Locatelli F, Campos A, de Heredia CD, Mohty M, Hurtado JMP, Bierings M, Bittencourt H, Mauad M, Purtill D, Cunha R, Kabbara N, Gluckman E, Labopin M, Peters C, Rocha V. Impact of pretransplant minimal residual disease after cord blood transplantation for childhood acute lymphoblastic leukemia in remission: an Eurocord, PDWP–EBMT analysis. Leukemia 2012; 26:2455-61. [DOI: 10.1038/leu.2012.123] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
44
|
Abstract
Acute myeloid leukemia (AML) is a highly heterogenous disease with multiple signaling pathways contributing to its pathogenesis. A key driver of AML is the FMS-like tyrosine kinase receptor-3 (FLT3). Activating mutations in FLT3, primarily the FLT3-internal tandem duplication (FLT3-ITD), are associated with decreased progression-free and overall survival. Identification of the importance of FLT3-ITD and the FLT3 pathway in the prognosis of patients with AML has stimulated efforts to develop therapeutic inhibitors of FLT3. Although these inhibitors have shown promising antileukemic activity, they have had limited efficacy to date as single agents and may require use in combination with cytotoxic chemotherapies. Here, we review clinical and preclinical results for the clinically mature FLT3 inhibitors currently in development. We conclude that multitargeted FLT3 inhibitors may have more utility earlier in the course of disease, when in vitro evidence suggests that AML cells are less dependent on FLT3 signaling, perhaps because of upregulation of multiple other signaling pathways. More potent agents may have greater utility in relapsed and heavily pretreated patients, in whom high levels of circulating FLT3 ligand may necessitate use of an agent with a very favorable pharmacokinetic/pharmacodynamic profile. Novel combination regimens are also discussed.
Collapse
|
45
|
Nomdedéu J, Hoyos M, Carricondo M, Esteve J, Bussaglia E, Estivill C, Ribera JM, Duarte R, Salamero O, Gallardo D, Pedro C, Aventin A, Brunet S, Sierra J. Adverse impact of IDH1 and IDH2 mutations in primary AML: experience of the Spanish CETLAM group. Leuk Res 2012; 36:990-7. [PMID: 22520341 DOI: 10.1016/j.leukres.2012.03.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/13/2012] [Accepted: 03/23/2012] [Indexed: 02/02/2023]
Abstract
The study of genetic lesions in AML cells is helpful to define the prognosis of patients with this disease. This study analyzed the frequency and clinical impact of recently described gene alterations, isocitrate dehydrogenase 1 (IDH1) and isocitrate dehydrogenase 2 (IDH2) mutations, in a series of homogeneously treated patients with primary (de novo) AML. Two-hundred and seventy-five patients enrolled in the CETLAM 2003 protocol were analyzed. IDH1 and IDH2 mutations were investigated by well-established melting curve-analysis and direct sequencing (R140 IDH2 mutations). To establish the percentage of the mutated allele a pyrosequencing method was used. Patients were also studied for NPM, FLT3, MLL, CEBPA, TET2 and WT1 mutations. IDH1 or IDH2 mutations were identified in 23.3% AML cases and in 22.5% of those with a normal karyotype. In this latter group, mutations were associated with short overall survival. This adverse effect was even more evident in patients with the NPM or CEBPA mutated/FLT3 wt genotype. In all the cases analyzed, the normal allele was detected, suggesting that both mutations act as dominant oncogenes. No adverse clinical impact was observed in cases with TET2 mutations. IDH1 and IDH2 mutations are common genetic alterations in normal karyotype AML. Favourable genotype NPM or CEBPA mutated/FLT3 wt can be further categorized according to the IDH1 and IDH2 mutational status.
Collapse
Affiliation(s)
- J Nomdedéu
- Hematology Department, Hosptial de Santa Creu i Sant Pau, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Takahashi S. Current findings for recurring mutations in acute myeloid leukemia. J Hematol Oncol 2011; 4:36. [PMID: 21917154 PMCID: PMC3180439 DOI: 10.1186/1756-8722-4-36] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/14/2011] [Indexed: 12/21/2022] Open
Abstract
The development of acute myeloid leukemia (AML) is a multistep process that requires at least two genetic abnormalities for the development of the disease. The identification of genetic mutations in AML has greatly advanced our understanding of leukemogenesis. Recently, the use of novel technologies, such as massively parallel DNA sequencing or high-resolution single-nucleotide polymorphism arrays, has allowed the identification of several novel recurrent gene mutations in AML. The aim of this review is to summarize the current findings for the identification of these gene mutations (Dnmt, TET2, IDH1/2, NPM1, ASXL1, etc.), most of which are frequently found in cytogenetically normal AML. The cooperative interactions of these molecular aberrations and their interactions with class I/II mutations are presented. The prognostic and predictive significances of these aberrations are also reviewed.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- Division of Molecular Hematology, Kitasato University Graduate School of Medical Sciences and Division of Hematology, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Minami-ku, Sagamihara, Japan.
| |
Collapse
|
47
|
Balgobind BV, Zwaan CM, Pieters R, Van den Heuvel-Eibrink MM. The heterogeneity of pediatric MLL-rearranged acute myeloid leukemia. Leukemia 2011; 25:1239-48. [DOI: 10.1038/leu.2011.90] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
48
|
Identification of T-lymphocytic leukemia-initiating stem cells residing in a small subset of patients with acute myeloid leukemic disease. Blood 2011; 117:7112-20. [PMID: 21562049 DOI: 10.1182/blood-2011-01-329078] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Xenotransplantation of acute myeloid leukemia (AML) into immunodeficient mice has been critical for understanding leukemogenesis in vivo and defining self-renewing leukemia-initiating cell subfractions (LICs). Although AML-engraftment capacity is considered an inherent property of LICs, substrains of NOD/SCID mice that possess additional deletions such as the IL2Rγc(null) (NSG) have been described as a more sensitive recipient to assay human LIC function. Using 23 AML-patient samples, 39% demonstrated no detectable engraftment in NOD/SCID and were categorized as AMLs devoid of LICs. However, 33% of AML patients lacking AML-LICs were capable of engrafting NSG recipients, but produced a monoclonal T-cell proliferative disorder similar to T-ALL. These grafts demonstrated self-renewal capacity as measured by in vivo serial passage and were restricted to CD34-positive fraction, and were defined as LICs. Molecular analysis for translocations in MLL genes indicated that these AML patient-derived LICs all expressed the MLL-AFX1 fusion product. Our results reveal that the in vivo human versus xenograft host microenvironment dictates the developmental capacity of human LICs residing in a small subset of patients diagnosed with AML harboring MLL mutations. These findings have implications both for the basic biology of CSC function, and for the use of in vivo models of the leukemogenic process in preclinical or diagnostic studies.
Collapse
|
49
|
Balgobind BV, Hollink IH, Reinhardt D, van Wering ER, de Graaf SS, Baruchel A, Stary J, Beverloo HB, de Greef GE, Pieters R, Zwaan CM, van den Heuvel-Eibrink MM. Low frequency of MLL-partial tandem duplications in paediatric acute myeloid leukaemia using MLPA as a novel DNA screenings technique. Eur J Cancer 2010; 46:1892-9. [DOI: 10.1016/j.ejca.2010.02.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 02/15/2010] [Accepted: 02/16/2010] [Indexed: 11/25/2022]
|
50
|
Nomdedeu J, Bussaglia E, Villamor N, Martinez C, Esteve J, Tormo M, Estivill C, Queipo MP, Guardia R, Carricondo M, Hoyos M, Llorente A, Juncà J, Gallart M, Domingo A, Bargay J, Mascaró M, Moraleda JM, Florensa L, Ribera JM, Gallardo D, Brunet S, Aventin A, Sierra J. Immunophenotype of acute myeloid leukemia with NPM mutations: prognostic impact of the leukemic compartment size. Leuk Res 2010; 35:163-8. [PMID: 20542566 DOI: 10.1016/j.leukres.2010.05.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 05/12/2010] [Accepted: 05/17/2010] [Indexed: 11/26/2022]
Abstract
NPM mutations are the most common genetic abnormalities found in non-promyelocytic AML. NPM-positive patients usually show a normal karyotype, a peculiar morphologic appearance with frequent monocytic traits and good prognosis in the absence of an associated FLT3 mutation. This report describes the immunophenotypic and genetic characteristics of a consecutive series of NPM-mutated de novo AML patients enroled in the CETLAM trial. Eighty-three patients were included in the study. Complete immunophenotype was obtained using multiparametric flow cytometry. Associated genetic lesions (FLT3, MLL, CEBPA and WT1 mutations) were studied by standardized methods. Real-time PCR was employed to assess the minimal residual status. The most common pattern was CD34-CD15+ and HLA-DR+. Small CD34 populations with immunophenotypic aberrations (CD15 and CD19 coexpression, abnormal SSC) were detected even in CD34 negative samples. Nearly all cases expressed CD33 (strong positivity), CD13 and CD117, and all were CD123+. The stem cell marker CD110 was also positive in most cases. Biologic parameters such as a high percentage of intermediate CD45+ (blast gate) (>75% nucleated cells), CD123+ and FLT3-ITD mutations were associated with a poor outcome. Quantitative PCR positivity had no prognostic impact either after induction or at the end of chemotherapy. Only PCR positivity (greater than 10 copies) detected in patients in haematological remission was associated with an increased relapse rate. Further studies are required to determine whether the degree of leukemic stem cell expansion (CD45+CD123+cells) increases the risk of acquisition of FLT3-ITD and/or provides selective advantages.
Collapse
Affiliation(s)
- J Nomdedeu
- Department of Hematology and Laboratory, Hospital de la Santa Creu I Sant Pau, Avda Sant Antoni M Claret 167, 08025 Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|