1
|
Cremer A, Enssle JC, Pfaff S, Kouidri K, Lang F, Brandts C, Zeiher A, Cremer S, Steffen B, Serve H, Bug G. Treatment with midostaurin and other FLT3 targeting inhibitors is associated with an increased risk of cardiovascular adverse events in patients who underwent allogeneic hematopoietic stem cell transplantation with FLT3-mutated AML. Ann Hematol 2023; 102:2903-2908. [PMID: 37552323 PMCID: PMC10492676 DOI: 10.1007/s00277-023-05396-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/29/2023] [Indexed: 08/09/2023]
Abstract
The addition of midostaurin to standard chemotherapy has improved survival in patients with FLT3-mutated AML. However, the impact of midostaurin and other FLT3 inhibitors (FLT3i) on cardiovascular adverse events (CAEs) has not been studied in patients who underwent allogeneic hematopoietic stem cell transplantation in a real-world setting. We reviewed 132 patients with AML who were treated with intensive induction therapy and consecutive allogeneic stem cell transplantation at our institution (42 FLT3-mutated AML and 90 with FLT3 wildtype). We identified treatment with midostaurin and/or FLT3i as an independent risk factor for CAEs not resulting in higher non-relapse mortality (NRM) or impaired overall survival (OS). Hence, close monitoring for CAEs is warranted for these patients.
Collapse
Affiliation(s)
- Anjali Cremer
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany.
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany.
| | - Julius C Enssle
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
| | - Saskia Pfaff
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Khouloud Kouidri
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Fabian Lang
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Christian Brandts
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University, Frankfurt, Germany
| | - Andreas Zeiher
- Department of Medicine, Cardiology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Sebastian Cremer
- Department of Medicine, Cardiology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
| | - Björn Steffen
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
| | - Hubert Serve
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Frankfurt Am Main, Germany
| | - Gesine Bug
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt Am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt Am Main, Germany
| |
Collapse
|
2
|
Perrone S, Ottone T, Zhdanovskaya N, Molica M. How acute myeloid leukemia (AML) escapes from FMS-related tyrosine kinase 3 (FLT3) inhibitors? Still an overrated complication? CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:223-238. [PMID: 37457126 PMCID: PMC10344728 DOI: 10.20517/cdr.2022.130] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/21/2023] [Accepted: 04/11/2023] [Indexed: 07/18/2023]
Abstract
FMS-related tyrosine kinase 3 (FLT3) mutations, present in about 25%-30% of acute myeloid leukemia (AML) patients, constitute one of the most frequently detected mutations in these patients. The binding of FLT3L to FLT3 activates the phosphatidylinositol 3-kinase (PI3K) and RAS pathways, producing increased cell proliferation and the inhibition of apoptosis. Two types of FLT3 mutations exist: FLT3-ITD and FLT3-TKD (point mutations in D835 and I836 or deletion of codon I836). A class of drugs, tyrosine-kinase inhibitors (TKI), targeting mutated FLT3, is already available with 1st and 2nd generation molecules, but only midostaurin and gilteritinib are currently approved. However, the emergence of resistance or the selection of clones not responding to FLT3 inhibitors has become an important clinical dilemma, as the duration of clinical responses is generally limited to a few months. This review analyzes the insights into mechanisms of resistance to TKI and poses a particular view on the clinical relevance of this phenomenon. Has resistance been overlooked? Indeed, FLT3 inhibitors have significantly contributed to reducing the negative impact of FLT3 mutations on the prognosis of AML patients who are no longer considered at high risk by the European LeukemiaNet (ELN) 2022. Finally, several ongoing efforts to overcome resistance to FLT3-inhibitors will be presented: new generation FLT3 inhibitors in monotherapy or combined with standard chemotherapy, hypomethylating drugs, or IDH1/2 inhibitors, Bcl2 inhibitors; novel anti-human FLT3 monoclonal antibodies (e.g., FLT3/CD3 bispecific antibodies); FLT3-CAR T-cells; CDK4/6 kinase inhibitor (e.g., palbociclib).
Collapse
Affiliation(s)
- Salvatore Perrone
- Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, Latina 04100, Italy
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, the University of Rome “Tor Vergata”, Rome 00100 Italy
- Neuro-Oncohematology, Santa Lucia Foundation, I.R.C.C.S., Rome 00100, Italy
| | - Nadezda Zhdanovskaya
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome 00161, Italy
| | - Matteo Molica
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, Rome 00144, Italy
| |
Collapse
|
3
|
Midostaurin in patients with acute myeloid leukemia and FLT3-TKD mutations: a subanalysis from the RATIFY trial. Blood Adv 2021; 4:4945-4954. [PMID: 33049054 DOI: 10.1182/bloodadvances.2020002904] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/21/2020] [Indexed: 12/21/2022] Open
Abstract
The results from the RATIFY trial (ClinicalTrials.gov: NCT00651261; CALGB 10603) showed that midostaurin combined with standard chemotherapy significantly improved outcomes in patients with FMS-like tyrosine kinase 3 (FLT3)-mutated acute myeloid leukemia (AML), compared with placebo. In this post hoc subgroup analysis from the trial, we evaluated the impact of midostaurin in 163 patients with FLT3-tyrosine kinase domain (TKD) mutations. At a median follow-up of 60.7 months (95% CI, 55.0-70.8), the 5-year event-free survival (EFS) rate was significantly higher in patients treated with midostaurin than in those treated with placebo (45.2% vs 30.1%; P = .044). A trend toward improved disease-free survival was also observed with midostaurin (67.3% vs 53.4%; P = .089), whereas overall survival (OS) was similar in the 2 groups. Patients with AML and NPM1mut/FLT3-TKDmut or core binding factor (CBF)-rearranged/FLT3-TKDmut genotypes had significantly prolonged OS with or without censoring at hematopoietic cell transplantation (HCT), compared with NPM1WT/CBF-negative AMLs. The multivariable model for OS and EFS adjusted for allogeneic HCT in first complete remission as a time-dependent covariable, revealed NPM1 mutations and CBF rearrangements as significant favorable factors. These data show that NPM1 mutations or CBF rearrangements identify favorable prognostic groups in patients with FLT3-TKD AMLs, independent of other factors, also in the context of midostaurin treatment.
Collapse
|
4
|
Staudt D, Murray HC, McLachlan T, Alvaro F, Enjeti AK, Verrills NM, Dun MD. Targeting Oncogenic Signaling in Mutant FLT3 Acute Myeloid Leukemia: The Path to Least Resistance. Int J Mol Sci 2018; 19:ijms19103198. [PMID: 30332834 PMCID: PMC6214138 DOI: 10.3390/ijms19103198] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
The identification of recurrent driver mutations in genes encoding tyrosine kinases has resulted in the development of molecularly-targeted treatment strategies designed to improve outcomes for patients diagnosed with acute myeloid leukemia (AML). The receptor tyrosine kinase FLT3 is the most commonly mutated gene in AML, with internal tandem duplications within the juxtamembrane domain (FLT3-ITD) or missense mutations in the tyrosine kinase domain (FLT3-TKD) present in 30–35% of AML patients at diagnosis. An established driver mutation and marker of poor prognosis, the FLT3 tyrosine kinase has emerged as an attractive therapeutic target, and thus, encouraged the development of FLT3 tyrosine kinase inhibitors (TKIs). However, the therapeutic benefit of FLT3 inhibition, particularly as a monotherapy, frequently results in the development of treatment resistance and disease relapse. Commonly, FLT3 inhibitor resistance occurs by the emergence of secondary lesions in the FLT3 gene, particularly in the second tyrosine kinase domain (TKD) at residue Asp835 (D835) to form a ‘dual mutation’ (ITD-D835). Individual FLT3-ITD and FLT3-TKD mutations influence independent signaling cascades; however, little is known about which divergent signaling pathways are controlled by each of the FLT3 specific mutations, particularly in the context of patients harboring dual ITD-D835 mutations. This review provides a comprehensive analysis of the known discrete and cooperative signaling pathways deregulated by each of the FLT3 specific mutations, as well as the therapeutic approaches that hold the most promise of more durable and personalized therapeutic approaches to improve treatments of FLT3 mutant AML.
Collapse
Affiliation(s)
- Dilana Staudt
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia.
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Heather C Murray
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia.
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Tabitha McLachlan
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia.
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Frank Alvaro
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
- John Hunter Children's Hospital, Faculty of Health and Medicine, University of Newcastle, New Lambton Heights, NSW 2305, Australia.
| | - Anoop K Enjeti
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
- Calvary Mater Hospital, Hematology Department, Waratah, NSW 2298, Australia.
- NSW Health Pathology North, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia.
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia.
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia.
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health & Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
5
|
Abstract
Acute myeloid leukemia (AML) is a complex clonal disorder. The disease is characterized by chromosomal and molecular abnormalities that propagate and expand the abnormal clone(s). The main goal of therapy is to achieve and ultimately maintain a complete remission. In the younger AML patient (less than 60 years of age), there has been a standardization of the initial therapy with the 3 + 7 regimen, consisting of an anthracycline and cytarabine combination. Recent intensification of the anthracycline has led to improved remission and survival outcomes in these patients. Prognosis and therapy in this disease is driven by cytogenetic studies and the additional molecular information that is gathered at the time of diagnosis. With the finding of potential targetable lesions within these molecular aberrancies, new treatments are emerging to deepen remissions and ultimately improve survival.
Collapse
|
6
|
Duployez N, Marceau-Renaut A, Boissel N, Petit A, Bucci M, Geffroy S, Lapillonne H, Renneville A, Ragu C, Figeac M, Celli-Lebras K, Lacombe C, Micol JB, Abdel-Wahab O, Cornillet P, Ifrah N, Dombret H, Leverger G, Jourdan E, Preudhomme C. Comprehensive mutational profiling of core binding factor acute myeloid leukemia. Blood 2016; 127:2451-9. [PMID: 26980726 PMCID: PMC5457131 DOI: 10.1182/blood-2015-12-688705] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/08/2016] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukemia (AML) with t(8;21) or inv(16) have been recognized as unique entities within AML and are usually reported together as core binding factor AML (CBF-AML). However, there is considerable clinical and biological heterogeneity within this group of diseases, and relapse incidence reaches up to 40%. Moreover, translocations involving CBFs are not sufficient to induce AML on its own and the full spectrum of mutations coexisting with CBF translocations has not been elucidated. To address these issues, we performed extensive mutational analysis by high-throughput sequencing in 215 patients with CBF-AML enrolled in the Phase 3 Trial of Systematic Versus Response-adapted Timed-Sequential Induction in Patients With Core Binding Factor Acute Myeloid Leukemia and Treating Patients with Childhood Acute Myeloid Leukemia with Interleukin-2 trials (age, 1-60 years). Mutations in genes activating tyrosine kinase signaling (including KIT, N/KRAS, and FLT3) were frequent in both subtypes of CBF-AML. In contrast, mutations in genes that regulate chromatin conformation or encode members of the cohesin complex were observed with high frequencies in t(8;21) AML (42% and 18%, respectively), whereas they were nearly absent in inv(16) AML. High KIT mutant allele ratios defined a group of t(8;21) AML patients with poor prognosis, whereas high N/KRAS mutant allele ratios were associated with the lack of KIT or FLT3 mutations and a favorable outcome. In addition, mutations in epigenetic modifying or cohesin genes were associated with a poor prognosis in patients with tyrosine kinase pathway mutations, suggesting synergic cooperation between these events. These data suggest that diverse cooperating mutations may influence CBF-AML pathophysiology as well as clinical behavior and point to potential unique pathogenesis of t(8;21) vs inv(16) AML.
Collapse
MESH Headings
- Adolescent
- Adult
- Alleles
- Cell Cycle Proteins/genetics
- Child
- Child, Preschool
- Chromatin/genetics
- Chromatin/ultrastructure
- Chromosomal Proteins, Non-Histone/genetics
- Chromosome Inversion
- Chromosomes, Human, Pair 16/genetics
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 8/genetics
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factors/genetics
- DNA Mutational Analysis
- DNA, Neoplasm/genetics
- Female
- Genetic Association Studies
- High-Throughput Nucleotide Sequencing
- Humans
- Infant
- Leukemia, Myeloid, Acute/genetics
- Male
- Middle Aged
- Mutation
- Oncogene Proteins, Fusion/genetics
- Prognosis
- RUNX1 Translocation Partner 1 Protein
- Translocation, Genetic
- Young Adult
- Cohesins
Collapse
Affiliation(s)
- Nicolas Duployez
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire (CHU) Lille, Lille, France; Cancer Research Institute, INSERM Unité Mixte de Recherche (UMR)-S 1172, Lille, France
| | - Alice Marceau-Renaut
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire (CHU) Lille, Lille, France; Cancer Research Institute, INSERM Unité Mixte de Recherche (UMR)-S 1172, Lille, France
| | - Nicolas Boissel
- Department of Hematology, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Arnaud Petit
- Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Maxime Bucci
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire (CHU) Lille, Lille, France
| | - Sandrine Geffroy
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire (CHU) Lille, Lille, France; Cancer Research Institute, INSERM Unité Mixte de Recherche (UMR)-S 1172, Lille, France
| | | | - Aline Renneville
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire (CHU) Lille, Lille, France; Cancer Research Institute, INSERM Unité Mixte de Recherche (UMR)-S 1172, Lille, France
| | - Christine Ragu
- Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Martin Figeac
- Functional and Structural Genomic Platform, Lille University, Lille, France
| | - Karine Celli-Lebras
- Department of Hematology, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | | | - Jean-Baptiste Micol
- Department of Hematology, Gustave Roussy Institute, INSERM UMR 1170, Villejuif, France; Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | | | - Norbert Ifrah
- Department of Hematology, CHU Angers, Angers, France; and
| | - Hervé Dombret
- Department of Hematology, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Guy Leverger
- Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Eric Jourdan
- Department of Hematology, CHU Nîmes, Nîmes, France
| | - Claude Preudhomme
- Biology and Pathology Center, Laboratory of Hematology, Centre Hospitalier Universitaire (CHU) Lille, Lille, France; Cancer Research Institute, INSERM Unité Mixte de Recherche (UMR)-S 1172, Lille, France
| |
Collapse
|
7
|
Rustagi N, Hampton OA, Li J, Xi L, Gibbs RA, Plon SE, Kimmel M, Wheeler DA. ITD assembler: an algorithm for internal tandem duplication discovery from short-read sequencing data. BMC Bioinformatics 2016; 17:188. [PMID: 27121965 PMCID: PMC4847212 DOI: 10.1186/s12859-016-1031-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 04/12/2016] [Indexed: 11/13/2022] Open
Abstract
Background Detection of tandem duplication within coding exons, referred to as internal tandem duplication (ITD), remains challenging due to inefficiencies in alignment of ITD-containing reads to the reference genome. There is a critical need to develop efficient methods to recover these important mutational events. Results In this paper we introduce ITD Assembler, a novel approach that rapidly evaluates all unmapped and partially mapped reads from whole exome NGS data using a De Bruijn graphs approach to select reads that harbor cycles of appropriate length, followed by assembly using overlap-layout-consensus. We tested ITD Assembler on The Cancer Genome Atlas AML dataset as a truth set. ITD Assembler identified the highest percentage of reported FLT3-ITDs when compared to other ITD detection algorithms, and discovered additional ITDs in FLT3, KIT, CEBPA, WT1 and other genes. Evidence of polymorphic ITDs in 54 genes were also found. Novel ITDs were validated by analyzing the corresponding RNA sequencing data. Conclusions ITD Assembler is a very sensitive tool which can detect partial, large and complex tandem duplications. This study highlights the need to more effectively look for ITD’s in other cancers and Mendelian diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1031-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Navin Rustagi
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA. .,Department of Statistics, Rice University, Houston, TX, USA.
| | - Oliver A Hampton
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jie Li
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Dermatology, Xiangya Hospital, Central South University, Hunan, China
| | - Liu Xi
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sharon E Plon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Department of Pediatrics/Hematology-Oncology, Texas Children's Hospital, Houston, TX, USA
| | - Marek Kimmel
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - David A Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
8
|
Badar T, Kantarjian HM, Nogueras-Gonzalez GM, Borthakur G, Garcia Manero G, Andreeff M, Konopleva M, Kadia TM, Daver N, Wierda WG, Luthra R, Patel K, Oran B, Champlin R, Ravandi F, Cortes JE. Improvement in clinical outcome of FLT3 ITD mutated acute myeloid leukemia patients over the last one and a half decade. Am J Hematol 2015; 90:1065-70. [PMID: 26299958 DOI: 10.1002/ajh.24140] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/17/2015] [Accepted: 07/31/2015] [Indexed: 12/30/2022]
Abstract
AML with FLT3 ITD mutations are associated with poor outcome. We reviewed outcomes of patients with FLT3 ITD mutated AML to investigate trends over time. We analyzed 224 AML patients (excluding patients with core binding factor and acute promyelocytic leukemia) referred to our institution between 2000 and 2014. Patients were divided into five cohorts by era: 2000-2002 (Era 1, n = 19), 2003-2005 (Era 2, n = 41), 2006-2008 (Era 3, n = 53), 2009-2011 (Era 4, n = 55), and 2012-2014 (Era 5, n = 56) to analyze differences in outcome. The baseline characteristics were not statistically different across Eras. The response rate (CR/CRp) from Era 1-5 was 68%, 49%, 72%, 73%, and 75%, respectively. The overall response rate (all Eras) with chemotherapy alone versus chemotherapy plus FLT3 inhibitor was 67% and 72.5%, respectively (P = 0.4). The median time to relapse was 6, 3.6, 7.9, 8.1 months and not reached from Eras 1 through 5, respectively (P = 0.001). The median OS has improved: 9.6, 7.6, 14.4, 15.7, and 17.8 month from Eras 1-5, respectively (P = <0.001). Stem cell transplant as a time-dependent variable, showed better OS in the univariate analysis (HR: 0.57, 95% CI: 0.39-0.84, P = 0.004) but did not retained its significance in multivariate analysis (HR: 0.75, 95% CI: 0.50-1.13, P = 0.16). Our data suggest improvement in outcome of FLT3 ITD mutated AML patients over the last 15 years. This is probably due to improvement in treatment strategies, including but not limited to integration of FLT3 inhibitors and increased use of SCT.
Collapse
Affiliation(s)
- Talha Badar
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Hagop M. Kantarjian
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Gautam Borthakur
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Michael Andreeff
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Marina Konopleva
- Department of Leukemia, Section of Molecular Hematology and Therapy; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Tapan M. Kadia
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Naval Daver
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | - William G. Wierda
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Raja Luthra
- Department of Leukemia, Section of Molecular Hematology and Therapy; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Keyur Patel
- Department of Leukemia, Section of Molecular Hematology and Therapy; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Betul Oran
- Department of Stem Cell Transplantation and Cellular Therapy; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Farhad Ravandi
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| | - Jorge E. Cortes
- Department of Leukemia; University of Texas MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
9
|
Molecular characterization and testing in acute myeloid leukemia. J Hematop 2015. [DOI: 10.1007/s12308-015-0242-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
10
|
Shahab S, Nisa KU, Nadeem M, Zahid D, Ansari S, Farzana T, Taj M, Borhany M, Ahmed N, Shamsi TS. Survival Outcome of AML Patients with and without TKD Mutations. Asian Pac J Cancer Prev 2015; 15:10995. [DOI: 10.7314/apjcp.2014.15.24.10995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
11
|
Duployez N, Willekens C, Marceau-Renaut A, Boudry-Labis E, Preudhomme C. Prognosis and monitoring of core-binding factor acute myeloid leukemia: current and emerging factors. Expert Rev Hematol 2014; 8:43-56. [PMID: 25348871 DOI: 10.1586/17474086.2014.976551] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Core-binding factor acute myeloid leukemia (CBF-AML) - including AML with t(8;21) and AML with inv(16) - accounts for about 15% of adult AML and is associated with a relatively favorable prognosis. Nonetheless, relapse incidence may reach 40% in these patients. In this context, identification of prognostic markers is considered of great interest. Due to similarities between their molecular and prognostic features, t(8;21) and inv(16)-AML are usually grouped and reported together in clinical studies. However, considerable experimental evidences have highlighted that they represent two distinct entities and should be considered separately for further studies. This review summarizes recent laboratory and clinical findings in this particular subset of AML and how they could be used to improve management of patients in routine practice.
Collapse
Affiliation(s)
- Nicolas Duployez
- Hematology Laboratory, Biology and Pathology Center, Lille University Hospital, Lille, France
| | | | | | | | | |
Collapse
|
12
|
The prognostic relevance of flt3 and npm1 mutations on older patients treated intensively or non-intensively: a study of 1312 patients in the UK NCRI AML16 trial. Leukemia 2014; 28:1953-9. [PMID: 24573385 DOI: 10.1038/leu.2014.90] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/29/2014] [Accepted: 02/11/2014] [Indexed: 01/17/2023]
Abstract
Although the prognostic impact of mutations of FLT3 and NPM1 have been extensively studied in younger patients with acute myeloid leukaemia, less is known in older patients whether treated intensively or non-intensively, or in the context of existing prognostic scores. In 1312 patients 16 and 21%, respectively had an FLT3 and NPM1 mutation. An FLT3 mutation did not affect remission rate in intensively or non-intensively treated patients but was associated with an inferior survival. All patients with an NPM1c mutation had a significantly higher remission rate irrespective of treatment approach but survival was not improved, overall, or in any genotype except as in younger patients, in the FLT3 WT NPM1c mutant subgroup. When incorporated into an established multi-parameter prognostic risk score, the molecular information provided additional prognostic definition in 11% of patients.
Collapse
|
13
|
Jabbour E, Cortes J, Ravandi F, O'Brien S, Kantarjian H. Targeted therapies in hematology and their impact on patient care: chronic and acute myeloid leukemia. Semin Hematol 2013; 50:271-83. [PMID: 24246694 DOI: 10.1053/j.seminhematol.2013.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Advances in the genetic and molecular characterizations of leukemias have enhanced our capabilities to develop targeted therapies. The most dramatic example of targeted therapy in cancer to date is the use of targeted BCR-ABL protein tyrosine kinase inhibitors (TKI), which has revolutionized the treatment of chronic myeloid leukemia (CML). Inhibition of the signaling activity of this kinase has proved to be a highly successful treatment target, transforming the prognosis of patients with CML. In contrast, acute myeloid leukemia (AML) is an extremely heterogeneous disease with outcomes that vary widely according to subtype of the disease. Targeted therapy with monoclonal antibodies and small molecule kinase inhibitors are promising strategies to help improve the cure rates in AML. In this review, we will highlight the results of recent clinical trials in which outcomes of CML and AML have been influenced significantly. Also, novel approaches to sequencing and combining available therapies will be covered.
Collapse
Affiliation(s)
- Elias Jabbour
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX.
| | | | | | | | | |
Collapse
|
14
|
Allen C, Hills RK, Lamb K, Evans C, Tinsley S, Sellar R, O'Brien M, Yin JL, Burnett AK, Linch DC, Gale RE. The importance of relative mutant level for evaluating impact on outcome of KIT, FLT3 and CBL mutations in core-binding factor acute myeloid leukemia. Leukemia 2013; 27:1891-901. [DOI: 10.1038/leu.2013.186] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/24/2013] [Accepted: 06/14/2013] [Indexed: 12/25/2022]
|
15
|
Yang J, Schiffer CA. Genetic biomarkers in acute myeloid leukemia: will the promise of improving treatment outcomes be realized? Expert Rev Hematol 2013; 5:395-407. [PMID: 22992234 DOI: 10.1586/ehm.12.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Recent progress in the molecular genetics of acute myeloid leukemia (AML) has shown this disease to be more heterogeneous than previously realized. Recurrent cytogenetic and mutational changes in leukemic blasts have been confirmed to have high prognostic significance. High-throughput techniques to analyze the AML genome in greater depth have revealed novel mutations with putative roles in leukemogenesis. The use of prognostic biomarkers has allowed for a more detailed categorization of AML based on risk. Despite this tremendous progress, the understanding of the mechanisms by which these changes influence leukemia growth and response to treatment is still limited, which in turn has hindered the development of rationally targeted therapies for AML. The integration of clinical, cytogenetic and molecular data will be essential to translate the current research momentum into better outcomes for patients with AML.
Collapse
Affiliation(s)
- Jay Yang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| | | |
Collapse
|
16
|
Kok CH, Brown AL, Perugini M, Iarossi DG, Lewis ID, D'Andrea RJ. The preferential occurrence of FLT3-TKD mutations in inv(16) AML and impact on survival outcome: a combined analysis of 1053 core-binding factor AML patients. Br J Haematol 2012. [PMID: 23190472 DOI: 10.1111/bjh.12131] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
17
|
Wertheim GBW, Daber R, Bagg A. Molecular diagnostics of acute myeloid leukemia: it's a (next) generational thing. J Mol Diagn 2012; 15:27-30. [PMID: 23159532 DOI: 10.1016/j.jmoldx.2012.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 08/14/2012] [Indexed: 12/28/2022] Open
Abstract
This commentary highlights the article by Spencer et al that outlines a novel next-generation sequencing-based method for the detection of FLT3 mutations.
Collapse
Affiliation(s)
- Gerald B W Wertheim
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19014, USA.
| | | | | |
Collapse
|
18
|
Spencer DH, Abel HJ, Lockwood CM, Payton JE, Szankasi P, Kelley TW, Kulkarni S, Pfeifer JD, Duncavage EJ. Detection of FLT3 internal tandem duplication in targeted, short-read-length, next-generation sequencing data. J Mol Diagn 2012; 15:81-93. [PMID: 23159595 DOI: 10.1016/j.jmoldx.2012.08.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 08/07/2012] [Accepted: 08/24/2012] [Indexed: 12/25/2022] Open
Abstract
A recurrent somatic mutation frequently found in cytogenetically normal acute myeloid leukemia (AML) is internal tandem duplication (ITD) in the fms-related tyrosine kinase 3 gene (FLT3). This mutation is generally detected in the clinical laboratory by PCR and electrophoresis-based product sizing. As the number of clinically relevant somatic mutations in AML increases, it becomes increasingly attractive to incorporate FLT3 ITD testing into multiplex assays for many somatic mutations simultaneously, using next-generation sequencing (NGS). However, the performance of most NGS analysis tools for identifying medium-size insertions such as FLT3 ITD mutations is largely unknown. We used a multigene, targeted NGS assay to obtain deep sequence coverage (>1000-fold) of FLT3 and 26 other genes from 22 FLT3 ITD-positive and 29 ITD-negative specimens to examine the performance of several commonly used NGS analysis tools for identifying FLT3 ITD mutations. ITD mutations were present in hybridization-capture sequencing data, and Pindel was the only tool out of the seven tested that reliably detected these insertions. Pindel had 100% sensitivity (95% CI = 83% to 100%) and 100% specificity (95% CI = 88% to 100%) in our samples; Pindel provided accurate ITD insertion sizes and was able to detect ITD alleles present at estimated frequencies as low as 1%. These data demonstrate that FLT3 ITDs can be reliably detected in panel-based, next-generation sequencing assays.
Collapse
Affiliation(s)
- David H Spencer
- Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The prognostic factors in acute leukemia have undergone a major change over the past decade and are likely to be further refined in the coming years. While age is the single most important prognostic factor in both AML and in ALL, recurring cytogenetic abnormalities and molecular markers have become crucial for the prognosis of patients and for new directions in the development of targeted therapies. No less important is the development of a personalized approach for therapy as determined by the response to therapy using increasingly sensitive technologies. The assessment of MRD is rapidly superseding other prognostic factors in ALL and, somewhat lacking behind, coming into its own in AML. The next decade should see further refinement of response-driven prognostication, to include epigenetics as well as pharmacogenetics and pharmacodynamics of individual drugs used and the responses to them. It is hoped that these refinements and better predictors of response will also lead to a significantly improved overall outcome of patients with both AML and ALL.
Collapse
Affiliation(s)
- Chezi Ganzel
- Department of Hematology, Shaare Zedek Medical Center, PO Box 3235, Jerusalem 91031, Israel.
| | | |
Collapse
|
20
|
Miglino M, Colombo N, Pica G, Grasso R, Clavio M, Bergamaschi M, Ballerini F, Ghiso A, Ghiggi C, Mitscheunig L, Beltrami G, Cagnetta A, Vignolo L, Lucchetti MV, Aquino S, Pierri I, Sessarego M, Carella AM, Gobbi M. WT1 overexpression at diagnosis may predict favorable outcome in patients with de novo non-M3 acute myeloid leukemia. Leuk Lymphoma 2012; 52:1961-9. [PMID: 21942328 DOI: 10.3109/10428194.2011.585673] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We reviewed the frequency and prognostic significance of FLT3 (fms-like tyrosine kinase receptor-3) and NPM (nucleophosmin) gene mutations and WT1 (Wilms' tumor) and BAALC (brain and acute leukemia, cytoplasmic) gene expression in 100 consecutive patients with intermediate and poor cytogenetic risk de novo acute myeloid leukemia (AML) receiving conventional anthracycline-AraC based therapy. We observed a strict relationship between unfavorable karyotype and BAALC >1000 (p = 0.0001). Multivariate analysis of 81 patients with intermediate karyotype revealed that younger age (p = 0.00009), NPM gene mutation (p = 0.002), and WT1 >75th percentile (>2365) (p = 0.003) were independent, positive factors for complete remission (CR). WT1 expression above 2365 was correlated also to longer event-free survival (EFS) and overall survival (OS) in the same subset of patients (p = 0.003 and p = 0.02, respectively); the same finding occurred in younger patients with AML with intermediate karyotype (p = 0.008 and p = 0.01, respectively). In patients with intermediate karyotype, FLT3 internal tandem duplication (ITD) negatively affected EFS (EFS at 30 months: 30% vs. 6% in FLT3-ITD negative and FLT3 positive patients, respectively; p = 0.01) and OS (OS at 30 months: 38% vs. 20%, p = 0.03). The positive prognostic value of high WT1 expression does not have a clear explanation; it may be implicated either with WT1 anti-oncogenic function, or with the stimulating effect of WT1 oncogene on the leukemic cellular cycle, possibly associated with an enhanced response to chemotherapy.
Collapse
Affiliation(s)
- Maurizio Miglino
- Department of Hematology and Oncology, Azienda Ospedaliera Universitaria San Martino, Genova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Li W, Zhang L, Huang L, Mi Y, Wang J. Meta-analysis for the potential application of FLT3-TKD mutations as prognostic indicator in non-promyelocytic AML. Leuk Res 2012; 36:186-91. [DOI: 10.1016/j.leukres.2011.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 07/28/2011] [Accepted: 08/16/2011] [Indexed: 10/17/2022]
|
22
|
Tan AYC, Wong SQ, Nyvold CG, Carney DA, Wei A, Seymour JF, Hokland P, Dobrovic A. Rapid detection of FLT3 exon 20 tyrosine kinase domain mutations in patients with acute myeloid leukemia by high-resolution melting analysis. Leuk Lymphoma 2012; 53:1225-9. [PMID: 22132874 DOI: 10.3109/10428194.2011.645817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
23
|
Al-Kali A, Cortes J, Faderl S, Jones D, Abril C, Pierce S, Brandt M, Kantarjian H, Ravandi F. Patterns of molecular response to and relapse after combination of sorafenib, idarubicin, and cytarabine in patients with FLT3 mutant acute myeloid leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2011; 11:361-6. [PMID: 21816375 DOI: 10.1016/j.clml.2011.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 01/03/2011] [Accepted: 01/20/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND FMS-like tyrosine kinase 3 (FLT3) is a class III receptor tyrosine kinase involved in hematopoietic progenitor cell development. Mutations of FLT3 have been reported in about a third of patients with acute myeloid leukemia (AML), and inhibitors of FLT3 are of clinical interest. Sorafenib is an orally active multikinase inhibitor with potent activity against FLT3 and the Raf/ERK/MEK kinase pathway. METHODS We studied the patterns of molecular response and relapse in 18 patients with mutated FLT3 treated with the combination of sorafenib, idarubicin, and cytarabine. RESULTS The median follow-up was 9 months. Sixteen patients achieved complete remission (CR), and the other 2 patients achieved CR but lacked platelet recovery for an overall response rate of 100%. Ten patients had their FLT3-mutated clone eradicated, with 6 patients who showed some residual FLT3-mutated cells, and 2 patients who showed persistent FLT3-mutated cells. The elimination of FLT3-mutated population at the time of morphologic CR, however, was not predictive of relapse. After a median follow-up of 9 months (range, 1-16 months), 10 (55%) patients had relapsed, with a median CR duration of 8.8 months (range, 1-9.5 months). By DNA sequencing, there was no evidence of an acquired FLT3 point mutation at the time of relapse in 7 patients tested, which suggested the presence of other mechanisms of sorafenib resistance. CONCLUSION Sorafenib, combined with chemotherapy, is effective in attaining CR, but relapses still occur.
Collapse
Affiliation(s)
- Aref Al-Kali
- Department of Leukemia, The University of Texas--M D Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Park SH, Chi HS, Min SK, Cho YU, Jang S, Park CJ, Lee JH, Lee JH, Lee KH, Im HJ, Seo JJ. Prognostic significance of the FLT3 ITD mutation in patients with normal-karyotype acute myeloid leukemia in relapse. THE KOREAN JOURNAL OF HEMATOLOGY 2011; 46:88-95. [PMID: 21747880 PMCID: PMC3128906 DOI: 10.5045/kjh.2011.46.2.88] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 04/11/2011] [Accepted: 05/17/2011] [Indexed: 11/17/2022]
Abstract
Background Fms-like tyrosine kinase 3 internal tandem duplication (FLT3 ITD) mutation is related to poor prognosis in normal-karyotype acute myeloid leukemia (AML). However, the prognostic significance of the mutation at relapse has not been adequately investigated. We investigated the prognostic significance of the FLT3 ITD mutation at relapse in normal-karyotype AML patients. Methods We analyzed 69 normal-karyotype AML patients, in whom paired bone marrow samples taken at initial diagnosis and subsequent relapse were analyzed for the FLT3 ITD mutation at the Asan Medical Center between 1995 and 2009. Results Forty patients showed a persistent wild-type genotype, 11 showed the FLT3 ITD mutation at diagnosis and relapse, and 9 lost and another 9 acquired the mutation at relapse. The mutation status at relapse affected the overall survival (OS), with the mutation group showing shorter OS and survival after relapse than the wild-type group did (P<0.001 and P<0.001, respectively), despite having received more frequent stem cell transplantation after relapse than the wild-type group did. However, no difference was detected in the OS and survival after relapse with regard to the mutation status at diagnosis. Conclusion The patients with FLT3 ITD mutation at relapse showed poorer prognoses than those without the mutation. However, mutation status at diagnosis did not affect the outcome. These results suggest that, in normal-karyotype AML patients with relapse, the prognostic significance of FLT3 ITD mutation at relapse is greater than that of the mutation status at diagnosis.
Collapse
Affiliation(s)
- Sang Hyuk Park
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Warshawsky I, Mularo F. Locked nucleic acid probes for enhanced detection of FLT3 D835/I836, JAK2 V617F and NPM1 mutations. J Clin Pathol 2011; 64:905-10. [PMID: 21666141 DOI: 10.1136/jclinpath-2011-200086] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIMS Detecting low-level clinically significant cancer-relevant somatic mutations can be difficult. Several technologies exist for detecting minority mutations. One method is locked nucleic acid (LNA) PCR. In this study, LNA probes were used to enhance the sensitivity for detecting FLT3 D835/I836 tyrosine kinase domain (TKD) mutations, the JAK2 V617F mutation and insertion mutations in the nucleophosmin 1 gene. METHODS PCR was performed with and without LNA probes using DNA known to contain FLT3 D835/I836 TKD, JAK2 V617F and NPM1 mutations. FLT3 D835/I836 TKD mutations were detected following EcoRV restriction enzyme digestion and capillary electrophoresis. The JAK2 V617F mutation was detected by melt-curve analysis. NPM1 insertions were detected by capillary electrophoresis. RESULTS The detection of FLT3 D835/I836, JAK2 V617F and NPM1 mutations was enhanced approximately 10-50-fold using LNA probes. Rare JAK2 double mutants gave abnormal blocking patterns with the LNA probe. CONCLUSIONS Adding LNA probes to existing assays is a simple way to enhance and confirm the detection of mutations, especially those at low levels.
Collapse
Affiliation(s)
- Ilka Warshawsky
- Department of Molecular Pathology, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | |
Collapse
|
26
|
Abstract
Abstract
Acute myeloid leukemia (AML) is a disease with marked heterogeneity in both response to therapy and survival. Cytogenetics, age, and performance status have long determined prognosis and therapy. The advent of molecular diagnostics has heralded an explosion in new prognostic factors, including gene mutations in KIT, FLT3 (Fms-like tyrosine kinase 3), NPM1 (nucleophosmin 1), and CEBPA (CCAAT enhancer-binding protein-α). Microarray technology can now identify unique gene expression signatures associated with prognosis. Similarly microRNA expression, single nucleotide polymorphism arrays, and DNA methylation signatures have recently described important new prognostic subgroups of AML, and are contributing to our understanding of AML disease biology. Combined with proteomic profiling, these technologies have helped identify new targets and signaling pathways, and may soon help to identify individual patients likely to benefit from specific therapies, including allogeneic hematopoietic cell transplantation. In summary, new clinical and molecular prognostic markers have begun to significantly improve our understanding of AML biology. We are now close to a time when we will be able to use these prognostic factors and technologies to identify new targets for therapy and to determine who may benefit from that therapy, and ultimately change how we treat individual patients with AML.
Collapse
|
27
|
Ubiquitin conjugase UBCH8 targets active FMS-like tyrosine kinase 3 for proteasomal degradation. Leukemia 2010; 24:1412-21. [PMID: 20508617 DOI: 10.1038/leu.2010.114] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The class III receptor tyrosine kinase FMS-like tyrosine kinase 3 (FLT3) regulates normal hematopoiesis and immunological functions. Nonetheless, constitutively active mutant FLT3 (FLT3-ITD) causally contributes to transformation and is associated with poor prognosis of acute myeloid leukemia (AML) patients. Histone deacetylase inhibitors (HDACi) can counteract deregulated gene expression profiles and decrease oncoprotein stability, which renders them candidate drugs for AML treatment. However, these drugs have pleiotropic effects and it is often unclear how they correct oncogenic transcriptomes and proteomes. We report here that treatment of AML cells with the HDACi LBH589 induces the ubiquitin-conjugating enzyme UBCH8 and degradation of FLT3-ITD. Gain- and loss-of-function approaches show that UBCH8 and the ubiquitin-ligase SIAH1 physically interact with and target FLT3-ITD for proteasomal degradation. These ubiquitinylating enzymes though have a significantly lesser effect on wild-type FLT3. Furthermore, physiological and pharmacological stimulation of FLT3 phosphorylation, inhibition of FLT3-ITD autophosphorylation and analysis of kinase-inactive FLT3-ITD revealed that tyrosine phosphorylation determines degradation of FLT3 and FLT3-ITD by the proteasome. These results provide novel insights into antileukemic activities of HDACi and position UBCH8, which have been implicated primarily in processes in the nucleus, as a previously unrecognized important modulator of FLT3-ITD stability and leukemic cell survival.
Collapse
|
28
|
Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY, O'Brien S, Estrov Z, Borthakur G, Thomas D, Pierce SR, Brandt M, Byrd A, Bekele BN, Pratz K, Luthra R, Levis M, Andreeff M, Kantarjian HM. Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol 2010; 28:1856-62. [PMID: 20212254 PMCID: PMC2930809 DOI: 10.1200/jco.2009.25.4888] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To determine the efficacy and toxicity of the combination of sorafenib, cytarabine, and idarubicin in patients with acute myeloid leukemia (AML) younger than age 65 years. PATIENTS AND METHODS In the phase I part of the study, 10 patients with relapsed AML were treated with escalating doses of sorafenib with chemotherapy to establish the feasibility of the combination. We then treated 51 patients (median age, 53 years; range, 18 to 65 years) who had previously untreated AML with cytarabine at 1.5 g/m(2) by continuous intravenous (IV) infusion daily for 4 days (3 days if > 60 years of age), idarubicin at 12 mg/m(2) IV daily for 3 days, and sorafenib at 400 mg orally twice daily for 7 days. RESULTS Overall, 38 (75%) patients have achieved a complete remission (CR), including 14 (93%) of 15 patients with mutated FMS-like tyrosine kinase-3 (FLT3; the 15th patient had complete remission with incomplete platelet recovery [CRp]) and 24 (66%) of 36 patients with FLT3 wild-type (WT) disease (three additional FLT3-WT patients had CRp). FLT3-mutated patients were more likely to achieve a CR than FLT3-WT patients (P = .033). With a median follow-up of 54 weeks (range, 8 to 87 weeks), the probability of survival at 1 year is 74%. Among the FLT3-mutated patients, 10 have relapsed and five remain in CR with a median follow-up of 62 weeks (range, 10 to 76 weeks). Plasma inhibitory assay demonstrated an on-target effect on FLT3 kinase activity. CONCLUSION Sorafenib can be safely combined with chemotherapy, produces a high CR rate in FLT3-mutated patients, and inhibits FLT3 signaling.
Collapse
Affiliation(s)
- Farhad Ravandi
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD.,Corresponding author: Farhad Ravandi, MD, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX 77030; e-mail:
| | - Jorge E. Cortes
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Daniel Jones
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Stefan Faderl
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Guillermo Garcia-Manero
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Marina Y. Konopleva
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Susan O'Brien
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Zeev Estrov
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Gautam Borthakur
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Deborah Thomas
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Sherry R. Pierce
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Mark Brandt
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Anna Byrd
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - B. Nebiyou Bekele
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Keith Pratz
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Rajyalakshmi Luthra
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Mark Levis
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Michael Andreeff
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Hagop M. Kantarjian
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| |
Collapse
|
29
|
Borel C, Huynh A, Chaufour X, Lousteau O, Demur C, Delabesse E, Caverivière P, Attal M, Récher C. Uterine chloroma, aortic thrombus and CALM/AF10 acute myeloid leukemia. Leuk Res 2010; 34:e88-90. [DOI: 10.1016/j.leukres.2009.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 09/12/2009] [Accepted: 09/13/2009] [Indexed: 01/29/2023]
|
30
|
Ravandi F, Kantarjian H, Faderl S, Garcia-Manero G, O'Brien S, Koller C, Pierce S, Brandt M, Kennedy D, Cortes J, Beran M. Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. Leuk Res 2009; 34:752-6. [PMID: 19878996 DOI: 10.1016/j.leukres.2009.10.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 10/01/2009] [Accepted: 10/01/2009] [Indexed: 11/19/2022]
Abstract
Mutations of Fms-like tyrosine kinase-3 (FLT3) have been described in about 30% of patients with acute myeloid leukemia (AML) and are associated with a shorter disease-free and overall survival after initial therapy. We sought to examine whether the presence of these mutations in relapsed disease was also associated with a poor response to salvage chemotherapy by comparing the outcome of 34 patients with diploid cytogenetics and mutated FLT3 (internal tandem duplication mutation, ITD) to 69 patients with normal karyotype and wild-type FLT3 (FLT3-WT) in first relapse. On univariate analysis, patients with mutated FLT3 were less likely to achieve a CR to first salvage compared to FLT3-WT patients (24% vs. 41%; P=0.09). Furthermore, survival was longer for the FLT3-WT patients achieving a second CR after salvage compared to FLT3-mutated patients (P=0.017). Overall, patients with mutated FLT3 had a shorter survival from the time of relapse compared to those with FLT3-WT (P<0.001). The adverse prognostic impact of FLT3 mutations appears to persist beyond the initial treatment.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cohort Studies
- Follow-Up Studies
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Middle Aged
- Mutation/physiology
- Prognosis
- Recurrence
- Salvage Therapy
- Survival Analysis
- Treatment Outcome
- Young Adult
- fms-Like Tyrosine Kinase 3/genetics
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|