1551
|
Steckel NK, Groth C, Mikesch JH, Trenschel R, Ottinger H, Kordelas L, Mueller-Tidow C, Schliemann C, Reicherts C, Albring JC, Silling G, Schmidt E, Berdel WE, Lenz G, Ditschkowski M, Beelen DW, Stelljes M. High-dose melphalan-based sequential conditioning chemotherapy followed by allogeneic haematopoietic stem cell transplantation in adult patients with relapsed or refractory acute myeloid leukaemia. Br J Haematol 2018; 180:840-853. [PMID: 29468631 DOI: 10.1111/bjh.15137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/04/2017] [Indexed: 01/21/2023]
Abstract
Considering the unsatisfactory results of salvage therapies for patients with relapsed/refractory acute myeloid leukaemia (R/R-AML), their value before allogeneic haematopoietic stem cell transplantation (HSCT) remains questionable. However, direct allogeneic HSCT following established conditioning regimens applied in patients with R/R-AML during active disease has been equally disappointing. In this retrospective observational study, high-dose melphalan, as part of a sequential preparative regimen, followed by a total body irradiation (4 × 2 Gy)-based or a treosulfan-based dose-adapted conditioning therapy for allogeneic HSCT was administered to 292 adult patients (median age 56 years, range 17-74) with primary refractory (144 patients), secondary refractory (97 patients) or relapsed AML (51 patients). Overall survival rates at 3 years were 34%, 29% and 41%, respectively. Risk factors associated with an inferior survival were higher age, transplantation from a human leucocyte antigen-mismatched donor and high disease burden. Patients transplanted with blast infiltration <20% showed a notable survival rate of 51% at 3 years. In particular, patients with primary refractory AML showed a more favourable outcome when transplanted early during their disease course. Thus, high-dose melphalan-based sequential conditioning chemotherapy followed by an allogeneic HSCT is feasible and enables long-term remission to be achieved in a substantial proportion of patients with active R/R-AML.
Collapse
Affiliation(s)
- Nina K Steckel
- Department of Bone Marrow Transplantation, West German Cancer Centre, University Hospital of Essen, Essen, Germany
| | - Christoph Groth
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany
| | - Rudolf Trenschel
- Department of Bone Marrow Transplantation, West German Cancer Centre, University Hospital of Essen, Essen, Germany
| | - Hellmut Ottinger
- Department of Bone Marrow Transplantation, West German Cancer Centre, University Hospital of Essen, Essen, Germany
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, West German Cancer Centre, University Hospital of Essen, Essen, Germany
| | | | | | - Christian Reicherts
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany
| | - Joern C Albring
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany
| | - Gerda Silling
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany
| | - Eva Schmidt
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, Muenster, Germany
| | - Georg Lenz
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, Muenster, Germany.,Translational Oncology, University Hospital Muenster, Muenster, Germany
| | - Markus Ditschkowski
- Department of Bone Marrow Transplantation, West German Cancer Centre, University Hospital of Essen, Essen, Germany
| | - Dietrich W Beelen
- Department of Bone Marrow Transplantation, West German Cancer Centre, University Hospital of Essen, Essen, Germany
| | - Matthias Stelljes
- Department of Medicine A, University Hospital of Muenster, Muenster, Germany
| |
Collapse
|
1552
|
Brunner AM. No Mutation Left Behind: The Impact of Reporting Recurrent Genetic Abnormalities on Outcomes of Patients with Acute Myeloid Leukemia. Acta Haematol 2018; 139:128-130. [PMID: 29444505 DOI: 10.1159/000487071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 01/22/2018] [Indexed: 11/19/2022]
|
1553
|
Abstract
Treatment regimens for acute myeloid leukemia (AML) have remained largely unchanged until recently. Molecular advances have opened the door to targeted therapies, many of which are in late-phase clinical trials. As new therapeutic opportunities arise, it is appropriate to review key aspects of clinical trial design, statistical interpretation of outcomes, and methods of data reporting. Complete remission and overall survival (OS) are common primary endpoints in early-phase AML clinical trials. OS and event-free survival are frequent primary endpoints in phase 3 trials. Clinical trials are designed to address the primary endpoint using prespecified α and power levels. Interpretation of additional endpoints (eg, secondary endpoints and subgroup analyses) must be viewed in light of a trial's statistical design. Furthermore, variations in reporting of endpoints must be considered in order to understand trial outcomes. Time-to-event endpoints are typically reported using Kaplan-Meier curves, which are visually informative. Statistical data derived from these curves can be complex, and a variety of factors may impact interpretation. The purpose of this review is to discuss the nuances of common AML trial endpoints and their data presentation to better inform evaluation and understanding of clinical trial data.
Collapse
Affiliation(s)
- Bruno C Medeiros
- Department of Medicine, Stanford University School of Medicine, 875 Blake Wilbur Dr, Stanford, CA, USA.
| |
Collapse
|
1554
|
Ciurea SO. Allogeneic stem cell transplantation for FLT3 mutated acute myeloid leukemia in first complete remission: does age really matter? Haematologica 2018; 103:191-193. [PMID: 29386373 DOI: 10.3324/haematol.2017.186346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Stefan O Ciurea
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
1555
|
Baron F, Ruggeri A, Beohou E, Labopin M, Mohty M, Sanz J, Vigouroux S, Furst S, Bosi A, Chevallier P, Cornelissen JJ, Michallet M, Sierra J, Karakasis D, Savani BN, Gluckman E, Nagler A. Occurrence of graft-versus-host disease increases mortality after umbilical cord blood transplantation for acute myeloid leukaemia: a report from Eurocord and the ALWP of the EBMT. J Intern Med 2018; 283:178-189. [PMID: 28977716 DOI: 10.1111/joim.12696] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The efficacy of umbilical cord blood transplantation (UCBT) as treatment for acute myeloid leukaemia (AML) relies on immune-mediated graft-versus-leukaemia effects. Previous studies have suggested a strong association between graft-versus-host disease (GVHD) occurrence and graft-versus-leukaemia effects after allogeneic hematopoietic cell transplantation. METHODS Here, we evaluated the kinetics of relapse rate in correlation with GVHD occurrence after UCBT. The kinetics of relapse rate over time in correlation to GVHD occurrence were assessed by calculating the relapse rate per patient-year within sequential 90-day intervals. The impact of GVHD on relapse and mortality was further studied in multivariate Cox models handling GVHD as a time-dependent covariate. RESULTS The study included data from 1068 patients given single (n = 567) or double (n = 501) UCBT. The proportion of patients with grade II, III and IV acute GVHD was 20%, 7% and 4%, respectively. At 2 years, the cumulative incidence of chronic GVHD was 42%, the cumulative incidence of relapse was 32%, and overall survival was 32% as well. Relapse rates declined gradually over time during the first 30 months after transplantation. There was a possible suggestion that grade II-IV acute (HR = 0.8, P = 0.1) and chronic (HR = 0.65, P = 0.1) GVHD decreased relapse risk. However, grade II-IV acute GVHD significantly increased early (the first 18 months after UCBT) mortality (HR = 1.3, P = 0.02), whilst chronic GVHD increased each early (HR = 2.7, P < 0.001) and late (HR = 4.9, P < 0.001) mortality after UCBT. CONCLUSIONS The occurrence of grade II-IV acute or chronic GVHD each increases overall mortality after UCBT for AML mitigating the possible graft-versus-leukemia effect of GVHD.
Collapse
Affiliation(s)
- F Baron
- Laboratory of Hematology, GIGA-I3, University of Liege, Liege, Belgium
| | - A Ruggeri
- Eurocord, Hospital Saint Louis, AP-HP, IUH University Paris VII, Paris, France.,Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Université Pierre & Marie Curie and INSERM UMRs U938
| | - E Beohou
- EBMT Paris Office, Hospital Saint Antoine, Paris, France
| | - M Labopin
- EBMT Paris Office, Hospital Saint Antoine, Paris, France
| | - M Mohty
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Université Pierre & Marie Curie and INSERM UMRs U938
| | - J Sanz
- Servicio de Hematologia, Hospital Universitario La Fe, Valencia, Spain
| | - S Vigouroux
- Department of Hematology, University Hospital of Bordeaux, Bordeaux, France
| | - S Furst
- Department of Hematology, Institut Paoli Calmettes, Marseille, France
| | - A Bosi
- Hematology Unit, AOU Careggi, Florence, Italy
| | - P Chevallier
- Department of Hematology, CHU Nantes, Nantes, France
| | - J J Cornelissen
- Erasmus MC Cancer Institute, Department of Hematology, Rotterdam, The Netherlands
| | - M Michallet
- Department of Hematology, Centre Hospitalier Lyon-Sud, Lyon, France
| | - J Sierra
- Hematology Department, IIB Sant Pau and Josep Carreras Leukemia Research Institutes, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - D Karakasis
- Department of Hematology and Lymphomas, Evangelismos Hospital, Athens, Greece
| | - B N Savani
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - E Gluckman
- Eurocord, Hospital Saint Louis, AP-HP, France Monacord, Centre Scientifique de Monaco, IUH University Paris VII, Monaco city, Monaco
| | - A Nagler
- EBMT Paris Office, Hospital Saint Antoine, Paris, France.,Division of Hematology and Bone Marrow Transplantation, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| |
Collapse
|
1556
|
Sandmaier BM, Khaled S, Oran B, Gammon G, Trone D, Frankfurt O. Results of a phase 1 study of quizartinib as maintenance therapy in subjects with acute myeloid leukemia in remission following allogeneic hematopoietic stem cell transplant. Am J Hematol 2018; 93:222-231. [PMID: 29090473 PMCID: PMC6585789 DOI: 10.1002/ajh.24959] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 01/09/2023]
Abstract
FLT3-ITD-mutated acute myeloid leukemia (AML) has very high risk of relapse and is associated with poor outcome following allogeneic hematopoietic-cell transplant (allo-HCT). This two-part, phase 1, multicenter, open-label, sequential-group, dose-escalation study aimed to determine dose-limiting toxicities (DLTs), maximum tolerated dose (MTD), and safety/tolerability of quizartinib, a selective and highly potent FLT3 inhibitor, when administered as maintenance therapy after allo-HCT. Thirteen subjects with documented FLT3-ITD-mutated AML in morphological remission following allo-HCT received one of two quizartinib dihydrochloride dose levels (DL): 40 mg/d (DL1; n = 7) and 60 mg/d (DL2; n = 6), administered orally in 28-day cycles for up to 24 cycles. Median age of participants was 43 years. All subjects received human leukocyte antigen (HLA)-matched allo-HCT. One subject treated at DL1 and 1 treated at DL2 had DLTs that required drug interruption (grade 3 gastric hemorrhage and grade 3 anemia, respectively). Ten subjects (77%) received quizartinib for >1 year; 5 (38%) completed 24 cycles. Four subjects (31%) discontinued quizartinib due to adverse events. One subject (8%) experienced relapse during cycle 1 and discontinued treatment. Most common grade 3/4 adverse events were neutropenia (23%), anemia (15%), leukopenia (15%), lymphopenia (15%), and thrombocytopenia (15%). This study demonstrated acceptable tolerability and early evidence of reduced relapse rate following allo-HCT with quizartinib maintenance compared to historical cohorts. No MTD was identified, but 60 mg daily was selected as highest dose for continuous daily administration based on randomized comparison of daily 30 and 60 mg doses in relapsed/refractory AML.
Collapse
Affiliation(s)
- Brenda M. Sandmaier
- Fred Hutchinson Cancer Research CenterSeattleWashington
- University of Washington School of MedicineSeattleWashington
| | | | - Betül Oran
- The University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Guy Gammon
- Independent consultantSan DiegoCalifornia
| | | | | |
Collapse
|
1557
|
Altman JK, Foran JM, Pratz KW, Trone D, Cortes JE, Tallman MS. Phase 1 study of quizartinib in combination with induction and consolidation chemotherapy in patients with newly diagnosed acute myeloid leukemia. Am J Hematol 2018; 93:213-221. [PMID: 29139135 PMCID: PMC6586071 DOI: 10.1002/ajh.24974] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/09/2017] [Accepted: 11/12/2017] [Indexed: 11/27/2022]
Abstract
Novel therapies have potential to improve outcomes in patients with acute myeloid leukemia (AML) harboring FLT3‐ITD mutations that have high risk of relapse and poor survival following standard of care (SOC) cytarabine/anthracycline‐based induction/consolidation chemotherapy. Quizartinib is a selective and highly potent FLT3 inhibitor that has shown strong single‐agent activity in relapsed or refractory (R/R) AML. This phase 1, open‐label, sequential group dose‐escalation trial (NCT 01390337) is the first evaluating safety and tolerability of quizartinib in combination with SOC chemotherapy in newly diagnosed AML (ndAML). Nineteen patients unselected for FLT3 mutational status received one of three quizartinib dihydrochloride dose levels (DL): 60 mg/d for 7 days (DL1; n = 6), 60 mg/d for 14 days (DL2; n = 7), and 40 mg/d for 14 days (DL‐1; n = 6); administered orally starting on day 4 of chemotherapy. Median age was 43.8 years. Ten patients completed induction and consolidation. Three patients experienced dose‐limiting toxicities (DLTs): 2 at DL2 (1 pericardial effusion; 1 febrile neutropenia, decreased platelet count, and QT prolongation); 1 at DL‐1 (pericarditis). Maximum tolerated dose (MTD) was identified as DL‐1. Most common grade 3/4 adverse events were febrile neutropenia (47%), neutropenia (42%), thrombocytopenia (32%), and anemia (26%). There were no apparent additional toxicities with addition of quizartinib to chemotherapy although grade ≤1 QT prolongation was observed at MTD. Sixteen patients (84%) achieved a response; 14 (74%) composite complete response; 2 (11%) morphologic leukemia‐free state. The phase 3 QuANTUM‐First trial (NCT02668653) is further evaluating the effect of quizartinib plus SOC chemotherapy in ndAML FLT3‐ITD mutated patients.
Collapse
Affiliation(s)
- Jessica K. Altman
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Chicago Illinois
| | - James M. Foran
- Division of Hematology and Medical Oncology; Mayo Clinic Florida; Jacksonville Florida
| | - Keith W. Pratz
- Oncology, Johns Hopkins University School of Medicine; Baltimore Maryland
| | | | - Jorge E. Cortes
- Department of Leukemia, Division of Cancer Medicine; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Martin S. Tallman
- Leukemia Service, Department of Medicine; Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College; New York New York
| |
Collapse
|
1558
|
Kayser S, Levis MJ. Advances in targeted therapy for acute myeloid leukaemia. Br J Haematol 2018; 180:484-500. [PMID: 29193012 PMCID: PMC5801209 DOI: 10.1111/bjh.15032] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/21/2022]
Abstract
In the past few years, research in the underlying pathogenic mechanisms of acute myeloid leukaemia (AML) has led to remarkable advances in our understanding of the disease. Cytogenetic and molecular aberrations are the most important factors in determining response to chemotherapy as well as long-term outcome, but beyond prognostication are potential therapeutic targets. Our increased understanding of the pathogenesis of AML, facilitated by next-generation sequencing, has spurred the development of new compounds in the treatment of AML, particularly the creation of small molecules that target the disease on a molecular level. Various new agents, such as tyrosine kinase inhibitors, immune checkpoint inhibitors, monoclonal or bispecific T-cell engager antibodies, metabolic and pro-apoptotic agents are currently investigated within clinical trials. The highest response rates are often achieved when new molecularly targeted therapies are combined with standard chemotherapy. Presented here is an overview of novel therapies currently being evaluated in AML.
Collapse
Affiliation(s)
- Sabine Kayser
- Department of Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Haematology/Oncology, German Cancer Research Centre (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Mark J. Levis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
1559
|
Tyrosine kinase inhibitors and immune checkpoint blockade in allogeneic hematopoietic cell transplantation. Blood 2018; 131:1073-1080. [PMID: 29358177 DOI: 10.1182/blood-2017-10-752154] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 01/16/2018] [Indexed: 12/27/2022] Open
Abstract
Advances in the prevention of graft-versus-host disease (GVHD) and opportunistic infection have improved survival after allogeneic hematopoietic cell transplantation (allo-HCT) in the past decade. However, few inroads have been made into the treatment or prevention of relapse of the underlying malignancy for which allo-HCT is being performed. The introduction of US Food and Drug Administration-approved agents with significant activity in a variety of hematologic malignancies provides an opportunity to evaluate these interventions in the allo-HCT setting. Some of the most promising new agents include tyrosine kinase inhibitors (TKIs) directed at bcr-abl, kinase inhibitors targeting fms-like tyrosine kinase 3, and immune checkpoint inhibitors blocking both CTLA4 and PD-1. Data have emerged indicating potential efficacy of these agents in preventing or treating relapse, though definitive evidence remains elusive. However, potential toxicity can be considerable, highlighting the need for further clinical trials to define the therapeutic window. This review explores the immunologic and clinical consequence of treatment with both TKIs and checkpoint inhibitors in the peri- and post-allo-HCT setting.
Collapse
|
1560
|
Prognostic analysis according to the 2017 ELN risk stratification by genetics in adult acute myeloid leukemia patients treated in the Japan Adult Leukemia Study Group (JALSG) AML201 study. Leuk Res 2018; 66:20-27. [PMID: 29360622 DOI: 10.1016/j.leukres.2018.01.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/19/2017] [Accepted: 01/14/2018] [Indexed: 01/24/2023]
Abstract
Many genetic alterations that are associated with the prognosis of acute myeloid leukemia (AML) have been identified, and several risk stratification systems based on the genetic status have been recommended. The European LeukemiaNet (ELN) first proposed the risk stratification system for AML in 2010 (ELN-2010), and recently published the revised system (ELN-2017). We validated the long-term prognosis and clinical characteristics of each ELN-2017 risk category in Japanese adult AML patients who were treated in the Japan Adult Leukemia Study Group (JALSG) AML-201 study. We demonstrated that the 3-risk category system of the ELN-2017 successfully discriminated the overall survival and complete remission rates in our cohort in comparison with the 4-risk category of the ELN-2010. However, there were still genetic categories in which stratification of patients into favorable or intermediate risk categories was controversial; the low allelic ratio of FLT3-ITD was not necessarily associated with a better prognosis in patients with FLT3-ITD, and cytogenetic abnormalities may affect the prognosis in patients with favorable genetic lesions such as NPM1 and CEBPA mutations. As many molecular targeting agents, such as FLT3 inhibitors, have been developed, we must continue to modify the genetic risk stratification system to match the progression of therapeutic strategies.
Collapse
|
1561
|
Puente-Moncada N, Costales P, Antolín I, Núñez LE, Oro P, Hermosilla MA, Pérez-Escuredo J, Ríos-Lombardía N, Sanchez-Sanchez AM, Luño E, Rodríguez C, Martín V, Morís F. Inhibition of FLT3 and PIM Kinases by EC-70124 Exerts Potent Activity in Preclinical Models of Acute Myeloid Leukemia. Mol Cancer Ther 2018; 17:614-624. [DOI: 10.1158/1535-7163.mct-17-0530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/19/2017] [Accepted: 12/15/2017] [Indexed: 11/16/2022]
|
1562
|
Wang JX. [How I treat FLT3-ITD positive acute myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:1-4. [PMID: 29551024 PMCID: PMC7343119 DOI: 10.3760/cma.j.issn.0253-2727.2018.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Indexed: 12/20/2022]
Affiliation(s)
- J X Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| |
Collapse
|
1563
|
Risk-stratified therapy for children with FLT3-ITD-positive acute myeloid leukemia: results from the JPLSG AML-05 study. Int J Hematol 2018; 107:586-595. [DOI: 10.1007/s12185-017-2395-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 12/26/2017] [Accepted: 12/27/2017] [Indexed: 02/04/2023]
|
1564
|
Ghobrial IM, Detappe A, Anderson KC, Steensma DP. The bone-marrow niche in MDS and MGUS: implications for AML and MM. Nat Rev Clin Oncol 2018; 15:219-233. [PMID: 29311715 DOI: 10.1038/nrclinonc.2017.197] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Several haematological malignancies, including multiple myeloma (MM) and acute myeloid leukaemia (AML), have well-defined precursor states that precede the development of overt cancer. MM is almost always preceded by monoclonal gammopathy of undetermined significance (MGUS), and at least a quarter of all patients with myelodysplastic syndromes (MDS) have disease that evolves into AML. In turn, MDS are frequently anteceded by clonal haematopoiesis of indeterminate potential (CHIP). The acquisition of additional genetic and epigenetic alterations over time clearly influences the increasingly unstable and aggressive behaviour of neoplastic haematopoietic clones; however, perturbations in the bone-marrow microenvironment are increasingly recognized to have key roles in initiating and supporting oncogenesis. In this Review, we focus on the concept that the haematopoietic neoplasia-microenvironment relationship is an intimate rapport between two partners, provide an overview of the evidence supporting a role for the bone-marrow niche in promoting neoplasia, and discuss the potential for niche-specific therapeutic targets.
Collapse
Affiliation(s)
- Irene M Ghobrial
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Alexandre Detappe
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Kenneth C Anderson
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - David P Steensma
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
1565
|
El Fakih R, Rasheed W, Hawsawi Y, Alsermani M, Hassanein M. Targeting FLT3 Mutations in Acute Myeloid Leukemia. Cells 2018; 7:cells7010004. [PMID: 29316714 PMCID: PMC5789277 DOI: 10.3390/cells7010004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 01/04/2018] [Indexed: 01/08/2023] Open
Abstract
The FMS-like tyrosine kinase 3 (FLT3) pathway has an important role in cellular proliferation, survival, and differentiation. Acute myeloid leukemia (AML) patients with mutated FLT3 have a large disease burden at presentation and a dismal prognosis. A number of FLT3 inhibitors have been developed over the years. The first-generation inhibitors are largely non-specific, while the second-generation inhibitors are more specific and more potent. These inhibitors are used to treat patients with FLT3-mutated AML in virtually all disease settings including induction, consolidation, maintenance, relapse, and after hematopoietic cell transplantation (HCT). In this article, we will review the use of FLT3 inhibitors in AML.
Collapse
Affiliation(s)
- Riad El Fakih
- King Faisal Specialist Hospital and Research Center Riyadh, Riyadh 11211, Saudi Arabia.
| | - Walid Rasheed
- King Faisal Specialist Hospital and Research Center Riyadh, Riyadh 11211, Saudi Arabia.
| | - Yousef Hawsawi
- King Faisal Specialist Hospital and Research Center Riyadh, Riyadh 11211, Saudi Arabia.
| | - Maamoun Alsermani
- King Faisal Specialist Hospital and Research Center Riyadh, Riyadh 11211, Saudi Arabia.
| | - Mona Hassanein
- King Faisal Specialist Hospital and Research Center Riyadh, Riyadh 11211, Saudi Arabia.
| |
Collapse
|
1566
|
Pan Z, Yang M, Huang K, Büsche G, Glage S, Ganser A, Li Z. Flow cytometric characterization of acute leukemia reveals a distinctive "blast gate" of murine T-lymphoblastic leukemia/lymphoma. Oncotarget 2018; 9:2320-2328. [PMID: 29416774 PMCID: PMC5788642 DOI: 10.18632/oncotarget.23410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/05/2017] [Indexed: 11/28/2022] Open
Abstract
Immunophenotypic analysis using multiparameter flow cytometry is an indispensable tool for diagnosis and management of acute leukemia. Mouse models have been widely used for medical research for more than 100 years and are indispensable for leukemia research. However, immunophenotypic analysis of murine leukemia was not always performed in published studies, and blast gating for isolation of blasts was shown only in very few studies. No systemic characterization of all types of murine acute leukemia in large cohorts by flow cytometry has been reported. In this study, we used flow cytometry to comprehensively characterize murine acute leukemia in a large cohort of mice. We found that murine T-lymphoblastic leukemia/lymphoma (T-ALL) exhibits a distinctive “blast gate” (CD45bright) with CD45/side scatter gating that differs from the “blast gate” (CD45dim) of human T-ALL. By contrast, murine B-lymphoblastic leukemia and acute myeloid leukemia show the same blast region (CD45dim) as human leukemia. Using blast cell gating, we for first time detected T-ALL development in FLT3-ITD knock-in mice (incidence: 23%). These leukemic cells were selectively killed by the FLT3 inhibitors crenolanib and midostaurin in vitro. These data suggest that FLT3-ITD plays a potential role in the pathogenesis of T-ALL and that FLT3-ITD inhibition is a therapeutic option in the management of patients with T-ALL. Our gating strategy for immunophenotypic analysis can be used for leukemogenesis and preclinical gene therapy studies in mice and may improve the quality of such analyses.
Collapse
Affiliation(s)
- Zengkai Pan
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Min Yang
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Kezhi Huang
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guntram Büsche
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Silke Glage
- Institute of Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Zhixiong Li
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
1567
|
Yang X, Wang J. Precision therapy for acute myeloid leukemia. J Hematol Oncol 2018; 11:3. [PMID: 29301553 PMCID: PMC5755341 DOI: 10.1186/s13045-017-0543-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/19/2017] [Indexed: 01/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is a molecularly and clinically heterogeneous disease. Despite advances in understanding the pathogenesis of AML, the standard therapy remained nearly unchanged over the past three decades. With the poor survival for older patients and high relapse rate, multiple studies are ongoing to address this important issue. Novel therapies for AML, including the refinements of conventional cytotoxic chemotherapies and genetic and epigenetic targeted drugs, as well as immunotherapies, have been developed in recent years. Here, we present a mechanism-based review of some promising new drugs with clinical efficacy, focus on targeted drugs that are most potential to pave the road to success, and put forward the major challenges in promoting the precision therapy for AML.
Collapse
Affiliation(s)
- Xue Yang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| |
Collapse
|
1568
|
Yogarajah M, Stone RM. A concise review of BCL-2 inhibition in acute myeloid leukemia. Expert Rev Hematol 2018; 11:145-154. [DOI: 10.1080/17474086.2018.1420474] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Meera Yogarajah
- Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Richard M. Stone
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
1569
|
Brunner AM, Graubert TA. Genomics in childhood acute myeloid leukemia comes of age. Nat Med 2018; 24:7-9. [DOI: 10.1038/nm.4469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
1570
|
Schlenk RF, Kayser S. Midostaurin: A Multiple Tyrosine Kinases Inhibitor in Acute Myeloid Leukemia and Systemic Mastocytosis. Recent Results Cancer Res 2018; 212:199-214. [PMID: 30069632 DOI: 10.1007/978-3-319-91439-8_10] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Midostaurin (PKC412, Rydapt®) is an oral multiple tyrosine kinase inhibitor. Main targets are the kinase domain receptor, vascular endothelial-, platelet derived-, and fibroblast growth factor receptor, stem cell factor receptor c-KIT, as well as mutated and wild-type FLT3 kinases. Midostaurin was approved by the Food and Drug Administration (FDA) and the European Medical Agency (EMA) for acute myeloid leukemia with activating FLT3 mutations in combination with intensive induction and consolidation therapy as well as aggressive systemic mastocytosis (ASM), systemic mastocytosis with associated hematological neoplasm (SM-AHN) or mast cell leukemia (MCL). Several clinical trials are active or are planned to further investigate the role of midostaurin in myeloid malignancies and mastocytosis.
Collapse
Affiliation(s)
- Richard F Schlenk
- NCT-Trial Center, German Cancer Research Center, Heidelberg, Germany.
| | - Sabine Kayser
- Department of Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
1571
|
Weisberg E, Sattler M, Manley PW, Griffin JD. Spotlight on midostaurin in the treatment of FLT3-mutated acute myeloid leukemia and systemic mastocytosis: design, development, and potential place in therapy. Onco Targets Ther 2017; 11:175-182. [PMID: 29343975 PMCID: PMC5749544 DOI: 10.2147/ott.s127679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The Fms-like tyrosine kinase-3 (FLT3; fetal liver kinase-2; human stem cell tyrosine kinase-1; CD135) is a class III receptor tyrosine kinase that is normally involved in regulating the proliferation, differentiation, and survival of both hematopoietic cells and dendritic cells. Mutations leading it to be constitutively activated make it an oncogenic driver in ~30% of acute myeloid leukemia (AML) patients where it is associated with poor prognosis. The prevalence of oncogenic FLT3 and the dependency on its constitutively activated kinase activity for leukemia growth make this protein an attractive target for therapeutic intervention. Of the numerous small molecule inhibitors under clinical investigation for the treatment of oncogenic FLT3-positive AML, the N-benzoyl-staurosporine, midostaurin (CGP41251; PKC412; Rydapt®; Novartis Pharma AG, Basel, Switzerland), is the first to be approved by the US Food and Drug Administration for the treatment, in combination with standard chemotherapy, of newly diagnosed adult AML patients who harbor mutations in FLT3. Here, we describe the early design of midostaurin, the preclinical discovery of its activity against oncogenic FLT3, and its subsequent clinical development as a therapeutic agent for FLT3 mutant-positive AML.
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology, Dana-Farber Cancer Institute.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paul W Manley
- Department of Oncology, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - James D Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
1572
|
Short NJ, Kantarjian H, Jabbour E. Reply to ABCG2 overexpression and deoxyadenosine analogue activity in acute myeloid leukemia. Cancer 2017; 123:4935-4936. [PMID: 29053172 DOI: 10.1002/cncr.31036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/30/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
1573
|
Abstract
Activating mutations of FLT3 occur in about 30% of acute myeloid leukemia (AML) cases and are associated with relapse and poor prognosis. Midostaurin is the first drug approved for AML since 2000, and the first multi-kinase inhibitor approved for the FLT3-mutant subtype. To view this Bench to Bedside, open or download the PDF.
Collapse
|
1574
|
Poiré X, Labopin M, Polge E, Passweg J, Craddock C, Blaise D, Cornelissen JJ, Volin L, Russell NH, Socié G, Michallet M, Fegueux N, Chevallier P, Brecht A, Hunault-Berger M, Mohty M, Esteve J, Nagler A. Allogeneic stem cell transplantation benefits for patients ≥ 60 years with acute myeloid leukemia and FLT3 internal tandem duplication: a study from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Haematologica 2017; 103:256-265. [PMID: 29242299 PMCID: PMC5792270 DOI: 10.3324/haematol.2017.178251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/07/2017] [Indexed: 11/17/2022] Open
Abstract
Intermediate-risk cytogenetic acute myeloid leukemia with an internal tandem duplication of FLT3 (FLT3-ITD) is associated with a high risk of relapse, and is now a standard indication for allogeneic stem cell transplantation. Nevertheless, most studies supporting this strategy have been performed in young patients. To address the benefit of allogeneic transplantation in the elderly, we made a selection from the European Society for Blood and Marrow Transplantation registry of de novo intermediate-risk cytogenetic acute myeloid leukemia harboring FLT3-ITD in patients aged 60 or over and transplanted from a related or unrelated donor between January 2000 and December 2015. Two hundred and ninety-one patients were identified. Most patients received a reduced-intensity conditioning (82%), while donors consisted of an unrelated donor in 161 (55%) patients. Two hundred and twelve patients received their transplantation in first remission, 37 in second remission and 42 in a more advanced stage of the disease. The 2-year leukemia-free survival rate was 56% in patients in first remission, 22% in those in second remission and 10% in patients with active disease, respectively (P<0.005). Non-relapse mortality for the entire cohort was 20%. In multivariate analysis, disease status at transplantation was the most powerful predictor of worse leukemia-free survival, graft-versus-host disease and relapse-free survival, and overall survival. In this elderly population, age was not associated with outcome. Based on the current results, allogeneic transplantation translates into a favorable outcome in fit patients ≥ 60 with FLT3-ITD acute myeloid leukemia in first remission, similarly to current treatment recommendations for younger patients.
Collapse
Affiliation(s)
- Xavier Poiré
- Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Myriam Labopin
- Acute Leukemia Working Party of the EBMT.,Service d'Hématologie, Hôpital Saint-Antoine, Paris, France
| | - Emmanuelle Polge
- Acute Leukemia Working Party of the EBMT.,Service d'Hématologie, Hôpital Saint-Antoine, Paris, France
| | | | - Charles Craddock
- Center for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - Didier Blaise
- Programme de Transplantation et Thérapie Cellulaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, France
| | - Jan J Cornelissen
- Daniel den Hoed Cancer Centre, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Liisa Volin
- Stem Cell Transplantation Unit, HUH Comprehensive Cancer Center, Helsinki, Finland
| | | | - Gérard Socié
- Department of Hematology, Hôpital Saint-Louis, Paris, France
| | | | - Nathalie Fegueux
- Département d'Hématologie Clinique, CHU Lapeyronie, Montpellier, France
| | | | - Arne Brecht
- Deutsche Klinik für Diagnostik, KMT Zentrum, Wiesbaden, Germany
| | | | - Mohamad Mohty
- Acute Leukemia Working Party of the EBMT.,Service d'Hématologie, Hôpital Saint-Antoine, Paris, France
| | - Jordi Esteve
- Hematology Department, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - Arnon Nagler
- Acute Leukemia Working Party of the EBMT.,Chaim Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
1575
|
Prospective randomization of post-remission therapy comparing autologous peripheral blood stem cell transplantation versus high-dose cytarabine consolidation for acute myelogenous leukemia in first remission. Int J Hematol 2017; 107:468-477. [PMID: 29243031 DOI: 10.1007/s12185-017-2389-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/14/2022]
Abstract
We prospectively compared outcomes of autologous stem cell transplantation (ASCT) versus high-dose cytarabine (HiDAC) consolidation as post-remission therapy for favorable- and intermediate-risk acute myelogenous leukemia (AML) in first complete remission (CR1). Two-hundred-forty patients under 65 years with AML-M1, M2, M4, or M5 subtypes were enrolled. After induction, 153 patients did not undergo randomization, while the remaining 87 who achieved CR1 were prospectively randomized to HiDAC (n = 45) or ASCT arm (n = 42). In the HiDAC arm, 43 patients completed three cycles of HiDAC, whereas in ASCT arm 22 patients completed two cycles of consolidation consisting of intermediate-dose cytarabine plus mitoxantrone or etoposide followed by ASCT. The three-year disease-free survival (DFS) rate was 41% in HiDAC and 55% in ASCT arm (p = 0.25). Three-year overall survival (OS) rates were 77 and 68% (p = 0.67). Incidence of relapse was 54 and 41% (p = 0.22). There was no significant difference in nonrelapse mortality between two arms (p = 0.88). Patients in the ASCT arm tended to have higher DFS rates and lower relapse rates than patients in HiDAC; however, there was no significant improvement in OS in patients with favorable- and intermediate-risk AML in CR1. Patients with AML are not benefited by the intensified chemotherapy represented by ASCT.
Collapse
|
1576
|
Seligson ND, Hobbs ALV, Leonard JM, Mills EL, Evans AG, Goorha S. Evaluating the Impact of the Addition of Cladribine to Standard Acute Myeloid Leukemia Induction Therapy. Ann Pharmacother 2017; 52:439-445. [PMID: 29241342 DOI: 10.1177/1060028017749214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Treatment for acute myeloid leukemia (AML) has remained relatively unchanged over the past few decades. Although recent drug approvals have provided an increase in the number of treatment options in AML, further optimization of standard induction therapy is still necessary. The most commonly utilized induction options have been well studied, but there is a paucity of literature comparing the combination of idarubicin with cytarabine and cladribine. OBJECTIVE To assess the clinical effectiveness of the addition of cladribine to idarubicin and cytarabine (7+3 IA) induction therapy in the treatment of AML. METHODS This retrospective, propensity score-matched cohort study evaluated 37 patients with previously untreated AML who received either 7+3 IA or idarubicin, cytarabine, and cladribine (7+3+5 IAC) as induction therapy. The primary end point of this study was complete response (CR), with secondary end points including hospital length of stay (LOS), and adverse event rates. RESULTS After propensity score matching, odds of reaching CR in the 7+3+5 IAC cohort were increased by 33% (95% CI = 1.09-1.55; P < 0.01) compared with the 7+3 IA cohort. Patients who received cladribine were also found to have a reduction in hospital LOS by 3.5 days (95% CI = 0.07-6.85; P = 0.045) without an increase in adverse event rates. CONCLUSION The addition of cladribine to the 7+3 IA regimen may improve clinical outcomes when used as initial induction therapy, without increasing the incidence of adverse event rates.
Collapse
Affiliation(s)
- Nathan D Seligson
- 1 The Ohio State University, Columbus, OH, USA.,2 The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | | | - Amy G Evans
- 3 Baptist Memorial Hospital - Memphis, Memphis, TN, USA
| | | |
Collapse
|
1577
|
Kim M, Williams S. Midostaurin in Combination With Standard Chemotherapy for Treatment of Newly Diagnosed FMS-Like Tyrosine Kinase 3 (FLT3) Mutation–Positive Acute Myeloid Leukemia. Ann Pharmacother 2017; 52:364-369. [DOI: 10.1177/1060028017747900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Miryoung Kim
- The Arthur G James Cancer Hospital at The Ohio State University, Columbus, OH, USA
| | - Sherry Williams
- The Arthur G James Cancer Hospital at The Ohio State University, Columbus, OH, USA
| |
Collapse
|
1578
|
Perl AE. The role of targeted therapy in the management of patients with AML. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:54-65. [PMID: 29222237 PMCID: PMC6142552 DOI: 10.1182/asheducation-2017.1.54] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Drug therapy for acute myeloid leukemia (AML) is finally undergoing major changes in 2017. This is due to the US Food and Drug Administration's approval of several new, targeted agents (midostaurin, enasidenib, and gemtuzumab ozogamicin). Paired with the recent approval of a novel liposomal formulation of daunorubicin/cytarabine (CPX-351/Vyxeos), the standard of care is changing rapidly in AML for subgroups. This review will focus on currently approved agents and promising novel agents in development and will highlight controversial areas in targeted treatment.
Collapse
Affiliation(s)
- Alexander E Perl
- Division of Hematology/Oncology, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
1579
|
Soiffer RJ, Chen YB. Pharmacologic agents to prevent and treat relapse after allogeneic hematopoietic cell transplantation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:699-707. [PMID: 29222324 PMCID: PMC6142540 DOI: 10.1182/asheducation-2017.1.699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Affiliation(s)
- Robert J. Soiffer
- Dana-Farber Cancer Institute, Boston, MA
- Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Yi-Bin Chen
- Harvard Medical School, Boston, MA; and
- Massachusetts General Hospital, Boston, MA
| |
Collapse
|
1580
|
Linder K, Iragavarapu C, Liu D. SETBP1 mutations as a biomarker for myelodysplasia /myeloproliferative neoplasm overlap syndrome. Biomark Res 2017; 5:33. [PMID: 29225884 PMCID: PMC5718013 DOI: 10.1186/s40364-017-0113-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022] Open
Abstract
Myelodysplasia (MDS) /myeloproliferative neoplasm (MPN) overlap syndrome has been described since the 2001 WHO classification as disorders that have both proliferative and dysplastic changes simultaneously. Specific disorders include chronic myelomonocytic leukemia (CMML), juvenile myelomonocytic leukemia (JMML), BCR-ABL negative atypical chronic myeloid leukemia (aCML) and unclassifiable MDS/MPN (MPN/MDS-U). Recurrent gene mutations in these conditions have been described. Among them, SETBP1 mutations have been identified in up to 32% of aCML, 24% of JMML, 18% of CMML and 10% of MDS/MPN-U patients. The mutation hotspot lies in the amino acid residues 858–871 in the SETBP1 protein. SETBP1 mutations in MDS/MPN overlap syndrome is associated with accelerated transformation to leukemia and poor prognosis. In this review, we summarized the latest data on the role of SETBP1 mutations in the overlap syndrome. SETBP1 mutations may serve as a biomarker for the diagnosis and poor prognosis of the overlap syndrome.
Collapse
Affiliation(s)
- Katherine Linder
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| | - Chaitanya Iragavarapu
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| | - Delong Liu
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| |
Collapse
|
1581
|
Lei H, Wang W, Wu Y. Targeting oncoproteins for degradation by small molecules in myeloid leukemia. Leuk Lymphoma 2017; 59:2297-2304. [DOI: 10.1080/10428194.2017.1403600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Weiwei Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
1582
|
FLT3-ITD induces expression of Pim kinases through STAT5 to confer resistance to the PI3K/Akt pathway inhibitors on leukemic cells by enhancing the mTORC1/Mcl-1 pathway. Oncotarget 2017; 9:8870-8886. [PMID: 29507660 PMCID: PMC5823622 DOI: 10.18632/oncotarget.22926] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/15/2017] [Indexed: 12/20/2022] Open
Abstract
FLT3-ITD is the most frequent tyrosine kinase mutation in acute myeloid leukemia (AML) associated with poor prognosis. We previously reported that activation of STAT5 confers resistance to PI3K/Akt inhibitors on the FLT3-ITD-positive AML cell line MV4-11 and 32D cells driven by FLT3-ITD (32D/ITD) but not by FLT3 mutated in the tyrosine kinase domain (32D/TKD). Here, we report the involvement of Pim kinases expressed through STAT5 activation in acquisition of this resistance. The specific pan-Pim kinase inhibitor AZD1208 as well as PIM447 in combination with the PI3K inhibitor GDC-0941 or the Akt inhibitor MK-2206 cooperatively downregulated the mTORC1/4EBP1 pathway, formation of the eIF4E/eIF4G complex, and Mcl-1 expression leading to activation of Bak and Bax to induce caspase-dependent apoptosis synergistically in these cells. These cooperative effects were enhanced or inhibited by knock down of mTOR or expression of its activated mutant, respectively. Overexpression of Mcl-1 conferred the resistance on 32D/ITD cells to combined inhibition of the PI3K/Akt pathway and Pim kinases, while the Mcl-1-specific BH3 mimetic A-1210477 conquered the resistance of MV4-11 cells to GDC-0941. Furthermore, overexpression of Pim-1 in 32D/TKD enhanced the mTORC1/Mcl-1 pathway and partially protected it from the PI3K/Akt inhibitors or the FLT3 inhibitor gilteritinib to confer the resistance to PI3K/Akt inhibitors. Finally, AZD1208 and GDC-0941 cooperatively inhibited the mTORC1/Mcl-1 pathway and reduced viable cell numbers of primary AML cells from some FLT3-ITD positive cases. Thus, Pim kinases may protect the mTORC1/4EBP1/Mcl-1 pathway to confer the resistance to the PI3K/Akt inhibitors on FLT3-ITD cells and represent promising therapeutic targets.
Collapse
|
1583
|
Herschbein L, Liesveld JL. Dueling for dual inhibition: Means to enhance effectiveness of PI3K/Akt/mTOR inhibitors in AML. Blood Rev 2017; 32:235-248. [PMID: 29276026 DOI: 10.1016/j.blre.2017.11.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/10/2017] [Accepted: 11/30/2017] [Indexed: 01/04/2023]
Abstract
The phosphatidylinositol 3-kinase/protein kinase B (Akt)/mechanistic target of rapamycin (PI3K/Akt/mTOR) pathway is amplified in 60-80% of patients with acute myelogenous leukemia (AML). Since this complex pathway is crucial to cell functions such as growth, proliferation, and survival, inhibition of this pathway would be postulated to inhibit leukemia initiation and propagation. Inhibition of the mTORC1 pathway has met with limited success in AML due to multiple resistance mechanisms including direct insensitivity of the mTORC1 complex, feedback activation of the PI3k/Akt signaling network, insulin growth factor-1 (IGF-1) activation of PI3K, and others. This review explores the role of mTOR inhibition in AML, mechanisms of resistance, and means to improve outcomes through use of dual mTORC1/2 inhibitors or dual TORC/PI3K inhibitors. How these inhibitors interface with currently available therapies in AML will require additional preclinical experiments and conduct of well-designed clinical trials.
Collapse
Affiliation(s)
- Lauren Herschbein
- Department of Medicine, The James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA.
| | - Jane L Liesveld
- Department of Medicine, The James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
1584
|
Which new agents will be incorporated into frontline therapy in acute myeloid leukemia? Best Pract Res Clin Haematol 2017; 30:312-316. [DOI: 10.1016/j.beha.2017.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
1585
|
Manley PW, Weisberg E, Sattler M, Griffin JD. Midostaurin, a Natural Product-Derived Kinase Inhibitor Recently Approved for the Treatment of Hematological Malignancies Published as part of the Biochemistry series "Biochemistry to Bedside". Biochemistry 2017; 57:477-478. [PMID: 29188995 DOI: 10.1021/acs.biochem.7b01126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Paul W Manley
- Novartis Institutes of Biomedical Research , CH 4002 Basel, Switzerland
| | - Ellen Weisberg
- Department of Medical Oncology, Dana Farber Cancer Institute , Boston, Massachusetts 02215, United States.,Department of Medicine, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Martin Sattler
- Department of Medical Oncology, Dana Farber Cancer Institute , Boston, Massachusetts 02215, United States.,Department of Medicine, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - James D Griffin
- Department of Medical Oncology, Dana Farber Cancer Institute , Boston, Massachusetts 02215, United States.,Department of Medicine, Harvard Medical School , Boston, Massachusetts 02115, United States
| |
Collapse
|
1586
|
A novel irreversible FLT3 inhibitor, FF-10101, shows excellent efficacy against AML cells with FLT3 mutations. Blood 2017; 131:426-438. [PMID: 29187377 DOI: 10.1182/blood-2017-05-786657] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/03/2017] [Indexed: 11/20/2022] Open
Abstract
An activating mutation of Fms-like tyrosine kinase 3 (FLT3) is the most frequent genetic alteration associated with poor prognosis in acute myeloid leukemia (AML). Although many FLT3 inhibitors have been clinically developed, no first-generation inhibitors have demonstrated clinical efficacy by monotherapy, due to poor pharmacokinetics or unfavorable safety profiles possibly associated with low selectivity against FLT3 kinase. Recently, a selective FLT3 inhibitor, quizartinib, demonstrated favorable outcomes in clinical studies. However, several resistant mutations emerged during the disease progression. To overcome these problems, we developed a novel FLT3 inhibitor, FF-10101, designed to possess selective and irreversible FLT3 inhibition. The co-crystal structure of FLT3 protein bound to FF-10101 revealed the formation of a covalent bond between FF-10101 and the cysteine residue at 695 of FLT3. The unique binding brought high selectivity and inhibitory activity against FLT3 kinase. FF-10101 showed potent growth inhibitory effects on human AML cell lines harboring FLT3 internal tandem duplication (FLT3-ITD), MOLM-13, MOLM-14, and MV4-11, and all tested types of mutant FLT3-expressing 32D cells including quizartinib-resistant mutations at D835, Y842, and F691 residues in the FLT3 kinase domain. In mouse subcutaneous implantation models, orally administered FF-10101 showed significant growth inhibitory effect on FLT3-ITD-D835Y- and FLT3-ITD-F691L-expressing 32D cells. Furthermore, FF-10101 potently inhibited growth of primary AML cells harboring either FLT3-ITD or FLT3-D835 mutation in vitro and in vivo. These results indicate that FF-10101 is a promising agent for the treatment of patients with AML with FLT3 mutations, including the activation loop mutations clinically identified as quizartinib-resistant mutations.
Collapse
|
1587
|
Pharmacologic agents to prevent and treat relapse after allogeneic hematopoietic cell transplantation. Blood Adv 2017; 1:2473-2482. [PMID: 29296897 DOI: 10.1182/bloodadvances.2017009894] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/28/2017] [Indexed: 01/01/2023] Open
|
1588
|
Gallogly MM, Perl AE, Lazarus HM. Midostaurin and emerging FLT3 inhibitors for the treatment of adults with newly diagnosed acute myeloid leukemia with the FLT3 mutation. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1406798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Molly M. Gallogly
- University Hospitals Cleveland Medical Center, Department of Medicine, Division of Hematology and Oncology, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Alexander E. Perl
- University of Pennsylvania, Division of Hematology Oncology, Perelman Center for Advanced Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
1589
|
Stansfield LC, Pollyea DA. Midostaurin: A New Oral Agent Targeting FMS-Like Tyrosine Kinase 3-Mutant Acute Myeloid Leukemia. Pharmacotherapy 2017; 37:1586-1599. [PMID: 28976600 DOI: 10.1002/phar.2039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Acute myeloid leukemia (AML), a clonal hematologic malignancy that results in bone marrow failure, is the most common acute leukemia in adults (median age of diagnosis 67 yrs), and treatment options, especially in the elderly population, are limited. Induction chemotherapy with 7 + 3, the combination of continuous-infusion cytarabine and intermittent dosing of an anthracycline administered over 7 and 3 days, respectively, has remained the standard of care since its introduction in 1973 in the United States. Midostaurin is a first-generation FMS-like tyrosine kinase 3 (FLT3) inhibitor (TKI) that was approved by the U.S. Food and Drug Administration in April 2017 for the treatment of FLT3-mutant AML. We performed a search of the PubMed database (January 1990-January 2017) to review pertinent clinical trials of midostaurin. Phase I, II, and III trials reported in English evaluating the safety and efficacy of midostaurin in patients with AML or myelodysplastic syndrome were included. The ClinicalTrials.gov database was also searched for ongoing trials. In the only phase III trial that has been conducted to date, midostaurin demonstrated significant improvement compared with placebo in overall and event-free survival in patients aged 18-59 years with newly diagnosed FLT3-mutant AML treated with standard induction chemotherapy. The median overall survival for patients randomized to the midostaurin group was 74.7 months versus 25.6 months in the placebo group (hazard ratio [HR] 0.78, 95% confidence interval [CI] 0.63-0.96, p=0.009). Median event-free survival was 8.2 months with midostaurin compared with 3.0 months with placebo (HR 0.78, 95% CI 0.66-0.93, p=0.002). In addition to being evaluated in combination with conventional chemotherapy, midostaurin has been studied as monotherapy, in combination with the hypomethylating agents azacitidine and decitabine, and as single-agent maintenance. Studies evaluating midostaurin in the maintenance setting after allogeneic stem cell transplantation are underway. Midostaurin is the first oral multitargeted TKI to improve overall survival in patients with FLT3-mutant AML and represents an important addition to the limited armamentarium against AML.
Collapse
Affiliation(s)
| | - Daniel A Pollyea
- Division of Hematology, University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|
1590
|
|
1591
|
Hypomethylating agents for treatment and prevention of relapse after allogeneic blood stem cell transplantation. Int J Hematol 2017; 107:138-150. [DOI: 10.1007/s12185-017-2364-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 11/08/2017] [Indexed: 12/14/2022]
|
1592
|
The role of targeted therapy in the management of patients with AML. Blood Adv 2017; 1:2281-2294. [PMID: 29296877 DOI: 10.1182/bloodadvances.2017009829] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/12/2017] [Indexed: 12/25/2022] Open
Abstract
Drug therapy for acute myeloid leukemia (AML) is finally undergoing major changes in 2017. This is due to the US Food and Drug Administration's approval of several new, targeted agents (midostaurin, enasidenib, and gemtuzumab ozogamicin). Paired with the recent approval of a novel liposomal formulation of daunorubicin/cytarabine (CPX-351/Vyxeos), the standard of care is changing rapidly in AML for subgroups. This review will focus on currently approved agents and promising novel agents in development and will highlight controversial areas in targeted treatment.
Collapse
|
1593
|
Targeting c-KIT (CD117) by dasatinib and radotinib promotes acute myeloid leukemia cell death. Sci Rep 2017; 7:15278. [PMID: 29127384 PMCID: PMC5681687 DOI: 10.1038/s41598-017-15492-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/27/2017] [Indexed: 12/13/2022] Open
Abstract
Dasatinib and radotinib are oral BCR-ABL tyrosine kinase inhibitors that were developed as drugs for the treatment of chronic myeloid leukemia. We report here that the c-KIT (CD117) targeting with dasatinib and radotinib promotes acute myeloid leukemia (AML) cell death, and c-KIT endocytosis is essential for triggering c-KIT-positive AML cell death by dasatinib and radotinib during the early stages. In addition, dasatinib and radotinib reduce heat shock protein 90β (HSP90β) expression and release Apaf-1 in c-KIT-positive AML cells. Finally, this activates a caspase-dependent apoptotic pathway in c-KIT-positive AML cells. Moreover, the inhibition of c-KIT endocytosis by dynamin inhibitor (DY) reversed cell viability and c-KIT expression by dasatinib and radotinib. HSP90β expression was recovered by DY in c-KIT-positive AML cells as well. Furthermore, the effect of radotinib on c-KIT and HSP90β showed the same pattern in a xenograft animal model using HEL92.1.7 cells. Therefore, dasatinib and radotinib promote AML cell death by targeting c-KIT. Taken together, these results indicate that dasatinib and radotinib treatment have a potential role in anti-leukemic therapy on c-KIT-positive AML cells.
Collapse
|
1594
|
|
1595
|
Inhibition of SDF-1-induced migration of oncogene-driven myeloid leukemia by the L-RNA aptamer (Spiegelmer), NOX-A12, and potentiation of tyrosine kinase inhibition. Oncotarget 2017; 8:109973-109984. [PMID: 29299123 PMCID: PMC5746358 DOI: 10.18632/oncotarget.22409] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/25/2017] [Indexed: 01/06/2023] Open
Abstract
Resistance to targeted tyrosine kinase inhibitors (TKI) remains a challenge for the treatment of myeloid leukemias. Following treatment with TKIs, the bone marrow microenvironment has been found to harbor a small pool of surviving leukemic CD34+ progenitor cells. The long-term survival of these leukemic cells has been attributed, at least in part, to the protective effects of bone marrow stroma. We found that the NOX-A12 'Spiegelmer', an L-enantiomeric RNA oligonucleotide that inhibits SDF-1α, showed in vitro and in vivo activity against BCR-ABL- and FLT3-ITD-dependent leukemia cells. NOX-A12 was sufficient to suppress SDF-1-induced migration in vitro. The combination of NOX-A12 with TKIs reduced cell migration in the same in vitro model of SDF-1-induced chemotaxis to a greater extent than either drug alone, suggesting positive cooperativity as a result of the SDF-1 blocking function of NOX-A12 and cytotoxicity resulting from targeted oncogenic kinase inhibition. These results are consistent with our in vivo findings using a functional pre-clinical mouse model of chronic myeloid leukemia (CML), whereby we demonstrated the ability of NOX-A12, combined with the ABL kinase inhibitor, nilotinib, to reduce the leukemia burden in mice to a greater extent than either agent alone. Overall, the data support the idea of using SDF-1 inhibition in combination with targeted kinase inhibition to override drug resistance in oncogene-driven leukemia to significantly diminish or eradicate residual leukemic disease.
Collapse
|
1596
|
|
1597
|
Luskin MR, DeAngelo DJ. Midostaurin/PKC412 for the treatment of newly diagnosed FLT3 mutation-positive acute myeloid leukemia. Expert Rev Hematol 2017; 10:1033-1045. [PMID: 29069942 DOI: 10.1080/17474086.2017.1397510] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with inadequate treatment options. Approximately one-third of cases have a FLT3-ITD or FLT3-TKD mutation which leads to constitutive tyrosine kinase activation which contributes to leukemogenesis. The FLT3-ITD mutation is associated with a particularly poor prognosis. Midostaurin is a multi-kinase inhibitor active against the FLT3 receptor. Midostaurin was approved by the US FDA in April 2017 for treatment of newly diagnosed FLT3-mutant AML in combination with chemotherapy. Areas covered: Standard treatment of FLT3-mutant AML and outcomes. Early clinical development of midostaurin including pharmacokinetics and metabolism. The development of midostaurin in FLT3-mutant AML is then outlined including review of the phase I, II, and III trials of midostaurin as a single agent and in combination with chemotherapy. Expert commentary: The approval of midostaurin represents the first new therapy for AML in several decades. It is also the first targeted therapy approved for AML. Future studies will focus on defining mechanisms of resistance to midostaurin as well as establishing the role of midostaurin in combination with hypomethylating agents and as maintenance therapy. Second generation, more potent and selective FLT3 inhibitors are also in development; these agents need to be compared to midostaurin.
Collapse
Affiliation(s)
- Marlise R Luskin
- a Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA.,b Harvard Medical School , Boston , MA , USA
| | - Daniel J DeAngelo
- a Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA.,b Harvard Medical School , Boston , MA , USA
| |
Collapse
|
1598
|
Murphy T, Yee KWL. Cytarabine and daunorubicin for the treatment of acute myeloid leukemia. Expert Opin Pharmacother 2017; 18:1765-1780. [DOI: 10.1080/14656566.2017.1391216] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Tracy Murphy
- Division of Medical Oncology and Hematology, University Health Network – Princess Margaret Cancer Centre, Toronto, Canada
| | - Karen W. L. Yee
- Division of Medical Oncology and Hematology, University Health Network – Princess Margaret Cancer Centre, Toronto, Canada
| |
Collapse
|
1599
|
Midostaurin, enasidenib, CPX-351, gemtuzumab ozogamicin, and venetoclax bring new hope to AML. Blood 2017; 130:2469-2474. [PMID: 29051180 DOI: 10.1182/blood-2017-08-784066] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022] Open
Abstract
In 2017, 4 drugs received US Food and Drug Administration marketing approval for acute myeloid leukemia (AML) treatment: targeted therapies for mutant FLT3 and IDH2, a liposomal cytarabine-daunorubicin formulation for therapy-related AML and AML with myelodysplasia-related changes, and resurgence of an antibody-drug conjugate designed to target CD33. Promising results also emerged for the BCL-2 inhibitor venetoclax combined with low-intensity therapy in older patients unfit for intensive chemotherapy. This quintet of new drugs is likely to reshape the therapeutic landscape of AML.
Collapse
|
1600
|
Long-term survival of sorafenib-treated FLT3-ITD-positive acute myeloid leukaemia patients relapsing after allogeneic stem cell transplantation. Eur J Cancer 2017; 86:233-239. [PMID: 29055209 DOI: 10.1016/j.ejca.2017.09.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/03/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Fms-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD)-positive acute myeloid leukaemia (AML) relapsing after allogeneic stem cell transplantation (allo-SCT) has a dismal prognosis with limited therapeutic options. FLT3-ITD kinase inhibition is a reasonable but palliative experimental treatment alternative in this situation. Information on long-term outcome is not available. METHODS We performed a long-term follow-up analysis of a previously reported cohort of 29 FLT3-ITD-positive AML patients, which were treated in relapse after allo-SCT with sorafenib monotherapy. FINDINGS With a median follow-up of 7.5 years, 6 of 29 patients (21%) are still alive. Excluding one patient who received a second allo-SCT, five patients (17%) achieved sustained complete remissions with sorafenib. Four of these patients are in treatment-free remission for a median of 4.4 years. INTERPRETATION Sorafenib may enable cure of a proportion of very poor risk FLT3-ITD-positive AML relapsing after allo-SCT.
Collapse
|