101
|
Descriptive and Functional Genomics in Acute Myeloid Leukemia (AML): Paving the Road for a Cure. Cancers (Basel) 2021; 13:cancers13040748. [PMID: 33670178 PMCID: PMC7916915 DOI: 10.3390/cancers13040748] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
Over the past decades, genetic advances have allowed a more precise molecular characterization of AML with the identification of novel oncogenes and tumor suppressors as part of a comprehensive AML molecular landscape. Recent advances in genetic sequencing tools also enabled a better understanding of AML leukemogenesis from the preleukemic state to posttherapy relapse. These advances resulted in direct clinical implications with the definition of molecular prognosis classifications, the development of treatment recommendations based on minimal residual disease (MRD) measurement and the discovery of novel targeted therapies, ultimately improving AML patients' overall survival. The more recent development of functional genomic studies, pushed by novel molecular biology technologies (short hairpin RNA (shRNA) and CRISPR-Cas9) and bioinformatics tools design on one hand, along with the engineering of humanized physiologically relevant animal models on the other hand, have opened a new genomics era resulting in a greater knowledge of AML physiopathology. Combining descriptive and functional genomics will undoubtedly open the road for an AML cure within the next decades.
Collapse
|
102
|
Wang H, Li XQ, Chu TT, Han SY, Qi JQ, Tang YQ, Qiu HY, Fu CC, Tang XW, Ruan CG, Wu DP, Han Y. Clinical significance of FLT3-ITD/CEBPA mutations and minimal residual disease in cytogenetically normal acute myeloid leukemia after hematopoietic stem cell transplantation. J Cancer Res Clin Oncol 2021; 147:2659-2670. [PMID: 33550446 DOI: 10.1007/s00432-021-03530-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/10/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Genetic changes have prognostic significance in cytogenetically normal acute myeloid leukemia (CN-AML). We set out to evaluate the prognostic of 6 gene mutations in CN-AML. METHODS We performed a mutational analysis and evaluated prognostic findings of six genes (NPM1, CEBPA, DNMT3A, FLT3-ITD, FLT3-TKD, and C-KIT) in 428 CN-AML patients at our center over 10 years. RESULTS A total of 282 patients (65.9%) had at least one gene mutation, and the mutation frequencies were as follows: 29.7% (NPM1), 24.1% (CEBPA), 20.1% (FLT3-ITD), 4.0% (FLT3-TKD), 11.9% (DNMT3A), and 4.7% (C-KIT). Multivariate analysis indicated that FLT3-ITDmut and CEBPAwt were independent risk factors correlated with poor overall survival (OS) and disease-free survival (DFS) of CN-AML. Compared with patients who received chemotherapy as consolidation, hematopoietic stem cell transplantation (HSCT) significantly improved OS of CN-AML patients. For standard/high risk patients, HSCT improved both OS and DFS. Combined analysis showed that patients with CEBPAmut/FLT3-ITDwt had the best prognosis, and patients with CEBPAwt/FLT3-ITDmut had the worst OS, with 3-year OS of only 44%. In 212 patients who received HSCT, FLT3-ITD/CEBPA mutations and minimal residual disease (MRD) were correlated with OS and DFS in univariate analysis. CONCLUSIONS We found that HSCT significantly improves the prognosis of standard/high risk CN-AML patients with superior OS and DFS. Molecular marker analyses, especially combined analysis of the FLT3-ITD and CEBPA status revealed a correlation with the prognosis of CN-AML. For patients who have received HSCT, MRD before transplantation was a strong prognostic marker predicting patient outcome.
Collapse
Affiliation(s)
- Hong Wang
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Xue-Qian Li
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Tian-Tian Chu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Shi-Yu Han
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Jia-Qian Qi
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Ya-Qiong Tang
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Hui-Ying Qiu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Cheng-Cheng Fu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Xiao-Wen Tang
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Chang-Geng Ruan
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China.,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - De-Pei Wu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China. .,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China. .,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China. .,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China.
| | - Yue Han
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215000, China. .,Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Suzhou, China. .,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China. .,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China.
| |
Collapse
|
103
|
FLT3-ITD DNA allelic burden, but not mRNA levels, influences the biological characteristics of AML patients. REV ROMANA MED LAB 2021. [DOI: 10.2478/rrlm-2021-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
FMS-like tyrosine kinase 3 gene internal tandem (FLT3-ITD) mutations represent one of the most frequent genetic lesions in acute myeloid leukemia (AML) and imparts a negative prognostic. For an optimal patient management, current clinical guidelines recommend the evaluation of the allelic ratio (AR), expressed as the DNA FLT3-ITD/WT mutational burden. We sought to evaluate the differences between the AR and FLT3-ITD/WT mRNA ratio (RR) and their respective impact on the biological characteristics of AML patients. A total of 32 DNA and mRNA samples from AML patients with FLT3-ITD were evaluated. There was a good correlation between the AR and RR (Spearman’s rho= 0.652, P <0.001). None of the biological characteristics were influenced by the RR values, whereas patients with high AR values (≥0.5) had higher WBC counts (Mann-Whitney, P= 0.01), LDH levels (Mann-Whitney, P= 0.037), and circulating blasts levels (Mann-Whitney, P= 0.023) than patients with low AR values (<0.5). Also, there was a good correlation between AR values and WBC count (Spearman’s correlation, P= 0.001), and LDH levels (Spearman’s correlation, P= 0.007). In our study population the AR, but not the RR, influenced the biological characteristic of patients suggesting a dose-independent effect of FLT3-ITD mutations.
Collapse
|
104
|
Alarbeed IF, Wafa A, Moassass F, Al-Halabi B, Al-Achkar W, Liehr T, Aboukhamis I. De novo adult acute myeloid leukemia with two new mutations in juxtatransmembrane domain of the FLT3 gene: a case report. J Med Case Rep 2021; 15:22. [PMID: 33494808 PMCID: PMC7836474 DOI: 10.1186/s13256-020-02587-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/16/2020] [Indexed: 11/29/2022] Open
Abstract
Background Approximately 30% of adult acute myeloid leukemia (AML) acquire within fms-like tyrosine kinase 3 gene (FLT3) internal tandem duplications (FLT3/ITDs) in their juxtamembrane domain (JMD). FLT3/ITDs range in size from three to hundreds of nucleotides, and confer an adverse prognosis. Studies on a possible relationship between of FLT3/ITDs length and clinical outcomes in those AML patients were inconclusive, yet. Case presentation Here we report a 54-year-old Arab male diagnosed with AML who had two FLT3-ITD mutations in addition to NPM1 mutation. Cytogenetic approaches (banding cytogenetics) and fluorescence in situ hybridization (FISH) using specific probes to detect translocations t(8;21), t(15;17), t(16;16), t(12;21), and deletion del(13q)) were applied to exclude chromosomal abnormalities. Molecular genetic approaches (polymerase chain reaction (PCR) and the Sanger sequencing) identified a yet unreported combination of two new mutations in FLT3-ITDs. The first mutation induced a frameshift in JMD, and the second led to a homozygous substitution of c.1836T>A (p.F612L) also in JMD. Additionally a NPM1 type A mutation was detected. The first chemotherapeutic treatment was successful, but 1 month after the initial diagnosis, the patient experienced a relapse and unfortunately died. Conclusions To the best of our knowledge, a combination of two FLT3-ITD mutations in JMD together with an NPM1 type A mutation were not previously reported in adult AML. Further studies are necessary to prove or rule out whether the size of these FLT3-ITDs mutations and potential other double mutations in FLT3-ITD are correlated with the observed adverse outcome.
Collapse
Affiliation(s)
- Ismael F Alarbeed
- Department of Microbiology, Hematology and Immunology, Faculty of Pharmacy, Damascus University, Ministry of High Education, Damascus, Syria.
| | - Abdulsamad Wafa
- Department of Molecular Biology and Biotechnology, Human Genetics Division, Atomic Energy Commission, Damascus, Syria
| | - Faten Moassass
- Department of Molecular Biology and Biotechnology, Human Genetics Division, Atomic Energy Commission, Damascus, Syria
| | - Bassel Al-Halabi
- Department of Molecular Biology and Biotechnology, Human Genetics Division, Atomic Energy Commission, Damascus, Syria
| | - Walid Al-Achkar
- Department of Molecular Biology and Biotechnology, Human Genetics Division, Atomic Energy Commission, Damascus, Syria
| | - Thomas Liehr
- Department of Molecular Biology and Biotechnology, Human Genetics Division, Atomic Energy Commission, Damascus, Syria.,Jena University Hospital, Institute of Human Genetics, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Imad Aboukhamis
- Department of Microbiology, Hematology and Immunology, Faculty of Pharmacy, Damascus University, Ministry of High Education, Damascus, Syria
| |
Collapse
|
105
|
Rodríguez-Arbolí E, Martínez-Cuadrón D, Rodríguez-Veiga R, Carrillo-Cruz E, Gil-Cortés C, Serrano-López J, Bernal Del Castillo T, Martínez-Sánchez MDP, Rodríguez-Medina C, Vidriales B, Bergua JM, Benavente C, García-Boyero R, Herrera-Puente P, Algarra L, Sayas-Lloris MJ, Fernández R, Labrador J, Lavilla-Rubira E, Barrios-García M, Tormo M, Serrano-Maestro A, Sossa-Melo CL, García-Belmonte D, Vives S, Rodríguez-Gutiérrez JI, Albo-López C, Garrastazul-Sánchez MP, Colorado-Araujo M, Mariz J, Sanz MÁ, Pérez-Simón JA, Montesinos P. Long-Term Outcomes After Autologous Versus Allogeneic Stem Cell Transplantation in Molecularly-Stratified Patients With Intermediate Cytogenetic Risk Acute Myeloid Leukemia: A PETHEMA Study. Transplant Cell Ther 2021; 27:311.e1-311.e10. [PMID: 33836871 DOI: 10.1016/j.jtct.2020.12.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022]
Abstract
Acute myeloid leukemia (AML) with intermediate risk cytogenetics (IRcyto) comprises a variety of biological entities with distinct mutational landscapes that translate into differential risks of relapse and prognosis. Optimal postremission therapy choice in this heterogeneous patient population is currently unsettled. In the current study, we compared outcomes in IRcyto AML recipients of autologous (autoSCT) (n = 312) or allogeneic stem cell transplantation (alloSCT) (n = 279) in first complete remission (CR1). Molecular risk was defined based on CEBPA, NPM1, and FLT3-ITD mutational status, per European LeukemiaNet 2017 criteria. Five-year overall survival (OS) in patients with favorable molecular risk (FRmol) was 62% (95% confidence interval [CI], 50-72) after autoSCT and 66% (95% CI, 41-83) after matched sibling donor (MSD) alloSCT (P = .68). For patients of intermediate molecular risk (IRmol), MSD alloSCT was associated with lower cumulative incidence of relapse (P < .001), as well as with increased nonrelapse mortality (P = .01), as compared to autoSCT. The 5-year OS was 47% (95% CI, 34-58) after autoSCT and 70% (95% CI, 59-79) after MSD alloSCT (P = .02) in this patient subgroup. In a propensity-score matched IRmol subcohort (n = 106), MSD alloSCT was associated with superior leukemia-free survival (hazard ratio [HR] 0.33, P = .004) and increased OS in patients alive 1 year after transplantation (HR 0.20, P = .004). These results indicate that, within IRcyto AML in CR1, autoSCT may be a valid option for FRmol patients, whereas MSD alloSCT should be the preferred postremission strategy in IRmol patients.
Collapse
Affiliation(s)
- Eduardo Rodríguez-Arbolí
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), University of Seville, Seville, Spain
| | | | | | - Estrella Carrillo-Cruz
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), University of Seville, Seville, Spain
| | - Cristina Gil-Cortés
- Department of Hematology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Josefina Serrano-López
- Department of Hematology, Reina Sofía University Hospital/Maimónides Biomedical Research Institute of Córdoba (IMIBIC)/University of Córdoba, Córdoba, Spain
| | | | | | - Carlos Rodríguez-Medina
- Department of Hematology, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Spain
| | - Belén Vidriales
- Department of Hematology, University Hospital of Salamanca (HUS/IBSAL), CIBERONC- CB16/12/00233 and Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Juan Miguel Bergua
- Department of Hematology, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - Celina Benavente
- Department of Hematology, Hospital Clínico San Carlos, Madrid, Spain
| | - Raimundo García-Boyero
- Department of Hematology, Hospital General Universitario de Castellón, Castellón de la Plana, Spain
| | | | - Lorenzo Algarra
- Department of Hematology, Hospital General de Albacete, Albacete, Spain
| | | | - Rosa Fernández
- Department of Hematology, Hospital Universitario Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Jorge Labrador
- Department of Hematology and Research Unit, Hospital Universitario de Burgos, Burgos, Spain
| | | | | | - Mar Tormo
- Deparment of Hematology, Hospital Clínico Universitario de Valencia, Instituto de Investigación INCLIVA, Valencia, Spain
| | | | | | | | - Susana Vives
- Department of Hematology - ICO Hospital Germans Trias i Pujol , Josep Carreras Leukemia Research Institute, Badalona , Spain
| | | | - Carmen Albo-López
- Department of Hematology, Complexo Hospitalario Universitario de Vigo, Vigo, Spain
| | | | | | - José Mariz
- Department of Hematology, Instituto Português de Oncologia do Porto FG, Porto, Portugal
| | - Miguel Ángel Sanz
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain
| | - José Antonio Pérez-Simón
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), University of Seville, Seville, Spain.
| | - Pau Montesinos
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain
| | | |
Collapse
|
106
|
Mori M, Hidaka K. [Pharmacological and clinical profile of gilteritinib (Xospata ® tablets 40 mg), a therapeutic agent for relapsed or refractory FLT3-mutated acute myeloid leukemia]. Nihon Yakurigaku Zasshi 2021; 156:37-46. [PMID: 33390479 DOI: 10.1254/fpj.20050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Gilteritinib fumarate (Xospata® tablets 40 mg) is a novel, highly selective, oral FMS-like tyrosine kinase 3 (FLT3) inhibitor used for the treatment of patients with relapsed or refractory FLT3-mutated acute myeloid leukemia (AML), and it was approved in Japan in September 2018. Preclinical studies demonstrated that gilteritinib inhibited FLT3 and showed antiproliferative activity against Ba/F3 cells expressing mutated FLT3. In addition, gilteritinib inhibited tumor growth, induced tumor regression, and prolonged survival in mice xenografted with MV4-11 cells endogenously expressing FLT3-internal tandem duplication. In clinical trials conducted in the United States, Europe, and Japan, plasma concentrations after administration of gilteritinib 20 to 450 mg/day were generally dose proportional, and gilteritinib was well tolerated. Multiple clinical trials, including a global Phase III study, in patients with relapsed or refractory FLT3-mutated AML treated with gilteritinib demonstrated higher response rates of complete remission or complete remission with partial hematologic recovery and longer overall survival compared with patients treated with salvage chemotherapy. Some clinical trials are ongoing in patients with FLT3-mutated AML at various treatment stages, such as induction therapy, maintenance therapy, and treatment after hematopoietic stem cell transplantation. In conclusion, in vitro, in vivo, and clinical data indicate that gilteritinib fumarate is an effective treatment option in adult patients with relapsed or refractory FLT3-mutated AML in Japan.
Collapse
Affiliation(s)
- Masamichi Mori
- Research Program Management, Drug Discovery Research, Astellas Pharma Inc
| | - Kazuyuki Hidaka
- Medical Science, Medical Affairs, Astellas Pharma Inc.,Current affiliation: Syneos Health Commercial K.K
| |
Collapse
|
107
|
Abstract
Mouse models of human myeloid malignancies support the detailed and focused investigation of selected driver mutations and represent powerful tools in the study of these diseases. Carefully developed murine models can closely recapitulate human myeloid malignancies in vivo, enabling the interrogation of a number of aspects of these diseases including their preclinical course, interactions with the microenvironment, effects of pharmacological agents, and the role of non-cell-autonomous factors, as well as the synergy between co-occurring mutations. Importantly, advances in gene-editing technologies, particularly CRISPR-Cas9, have opened new avenues for the development and study of genetically modified mice and also enable the direct modification of mouse and human hematopoietic cells. In this review we provide a concise overview of some of the important mouse models that have advanced our understanding of myeloid leukemogenesis with an emphasis on models relevant to clonal hematopoiesis, myelodysplastic syndromes, and acute myeloid leukemia with a normal karyotype.
Collapse
Affiliation(s)
- Faisal Basheer
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Department of Haematology, University of Cambridge, Cambridge CB2 0AW, United Kingdom
- Haematological Cancer Genetics, Wellcome Trust Sanger Institute, Cambridge CB10 1SA, United Kingdom
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - George Vassiliou
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Department of Haematology, University of Cambridge, Cambridge CB2 0AW, United Kingdom
- Haematological Cancer Genetics, Wellcome Trust Sanger Institute, Cambridge CB10 1SA, United Kingdom
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
108
|
Minetto P, Candoni A, Guolo F, Clavio M, Zannier ME, Miglino M, Dubbini MV, Carminati E, Sicuranza A, Ciofini S, Colombo N, Pugliese G, Marcolin R, Santoni A, Ballerini F, Lanino L, Cea M, Gobbi M, Bocchia M, Fanin R, Lemoli RM. Fludarabine, High-Dose Cytarabine and Idarubicin-Based Induction May Overcome the Negative Prognostic Impact of FLT3-ITD in NPM1 Mutated AML, Irrespectively of FLT3-ITD Allelic Burden. Cancers (Basel) 2020; 13:cancers13010034. [PMID: 33374216 PMCID: PMC7796342 DOI: 10.3390/cancers13010034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The prognostic relevance of molecular aberrations in acute myeloid leukemia (AML) has been prevalently tested in patients receiving conventional 3+7 induction. Recently, there has been a renewed interest in intensified inductions, but very few data are available on the impact of the most frequent genetic alterations with these alternative treatments. We analyzed a large multicentric cohort of younger AML patients harboring NPM1 and FLT3-ITD mutations receiving an intensified fludarabine-containing regimen (FLAI). Our data suggest that in NPM1 mut patients, FLAI may overcome the prognostic influence of co-mutated FLT3-ITD. The increased efficacy of this treatment seems to reduce the need for early consolidation with allogeneic transplant in double-mutated patients. Our data strongly support FLAI as an ideal backbone for combination with innovative targeted drugs, in order to further improve patients’ outcome. Abstract The mutations of NPM1 and FLT3-ITD represent the most frequent genetic aberration in acute myeloid leukemia. Indeed, the presence of an NPM1 mutation reduces the negative prognostic impact of FLT3-ITD in patients treated with conventional “3+7” induction. However, little information is available on their prognostic role with intensified regimens. Here, we investigated the efficacy of a fludarabine, high-dose cytarabine and idarubicin induction (FLAI) in 149 consecutive fit AML patients (median age 52) carrying the NPM1 and/or FLT3-ITD mutation, treated from 2008 to 2018. One-hundred-and-twenty-nine patients achieved CR (86.6%). After a median follow up of 68 months, 3-year overall survival was 58.6%. Multivariate analysis disclosed that both NPM1mut (p < 0.05) and ELN 2017 risk score (p < 0.05) were significant predictors of survival. NPM1-mutated patients had a favorable outcome, with no significant differences between patients with or without concomitant FLT3-ITD (p = 0.372), irrespective of FLT3-ITD allelic burden. Moreover, in landmark analysis, performing allogeneic transplantation (HSCT) in first CR proved to be beneficial only in ELN 2017 high-risk patients. Our data indicate that FLAI exerts a strong anti-leukemic effect in younger AML patients with NPM1mut and question the role of HSCT in 1st CR in NPM1mut patients with concomitant FLT3-ITD.
Collapse
Affiliation(s)
- Paola Minetto
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Correspondence: (P.M.); (F.G.); Tel.: +39-10-555-4329 (P.M.); +39-10-555-4491 (F.G.)
| | - Anna Candoni
- Division of Hematology and Bone Marrow Transplantation, Azienda Sanitaria Universitaria Integrata di Udine, 33100 Udine, Italy; (A.C.); (M.E.Z.); (M.V.D.); (R.F.)
| | - Fabio Guolo
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Correspondence: (P.M.); (F.G.); Tel.: +39-10-555-4329 (P.M.); +39-10-555-4491 (F.G.)
| | - Marino Clavio
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Maria Elena Zannier
- Division of Hematology and Bone Marrow Transplantation, Azienda Sanitaria Universitaria Integrata di Udine, 33100 Udine, Italy; (A.C.); (M.E.Z.); (M.V.D.); (R.F.)
| | - Maurizio Miglino
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Maria Vittoria Dubbini
- Division of Hematology and Bone Marrow Transplantation, Azienda Sanitaria Universitaria Integrata di Udine, 33100 Udine, Italy; (A.C.); (M.E.Z.); (M.V.D.); (R.F.)
| | - Enrico Carminati
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Anna Sicuranza
- Hematology Unit, University of Siena, Azienda Ospedaliera Universitaria, 53100 Siena, Italy; (A.S.); (S.C.); (A.S.); (M.B.)
| | - Sara Ciofini
- Hematology Unit, University of Siena, Azienda Ospedaliera Universitaria, 53100 Siena, Italy; (A.S.); (S.C.); (A.S.); (M.B.)
| | | | - Girolamo Pugliese
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Riccardo Marcolin
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
| | - Adele Santoni
- Hematology Unit, University of Siena, Azienda Ospedaliera Universitaria, 53100 Siena, Italy; (A.S.); (S.C.); (A.S.); (M.B.)
| | - Filippo Ballerini
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Luca Lanino
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
| | - Michele Cea
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Marco Gobbi
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
| | - Monica Bocchia
- Hematology Unit, University of Siena, Azienda Ospedaliera Universitaria, 53100 Siena, Italy; (A.S.); (S.C.); (A.S.); (M.B.)
| | - Renato Fanin
- Division of Hematology and Bone Marrow Transplantation, Azienda Sanitaria Universitaria Integrata di Udine, 33100 Udine, Italy; (A.C.); (M.E.Z.); (M.V.D.); (R.F.)
| | - Roberto Massimo Lemoli
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, 16132 Genova, Italy; (M.C.); (M.M.); (E.C.); (G.P.); (R.M.); (F.B.); (L.L.); (M.C.); (M.G.); (R.M.L.)
- IRCCS-Ospedale Policlinico San Martino, 16132 Genova, Italy;
| |
Collapse
|
109
|
Carter JL, Hege K, Yang J, Kalpage HA, Su Y, Edwards H, Hüttemann M, Taub JW, Ge Y. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther 2020; 5:288. [PMID: 33335095 PMCID: PMC7746731 DOI: 10.1038/s41392-020-00361-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults and the second most common form of acute leukemia in children. Despite this, very little improvement in survival rates has been achieved over the past few decades. This is partially due to the heterogeneity of AML and the need for more targeted therapeutics than the traditional cytotoxic chemotherapies that have been a mainstay in therapy for the past 50 years. In the past 20 years, research has been diversifying the approach to treating AML by investigating molecular pathways uniquely relevant to AML cell proliferation and survival. Here we review the development of novel therapeutics in targeting apoptosis, receptor tyrosine kinase (RTK) signaling, hedgehog (HH) pathway, mitochondrial function, DNA repair, and c-Myc signaling. There has been an impressive effort into better understanding the diversity of AML cell characteristics and here we highlight important preclinical studies that have supported therapeutic development and continue to promote new ways to target AML cells. In addition, we describe clinical investigations that have led to FDA approval of new targeted AML therapies and ongoing clinical trials of novel therapies targeting AML survival pathways. We also describe the complexity of targeting leukemia stem cells (LSCs) as an approach to addressing relapse and remission in AML and targetable pathways that are unique to LSC survival. This comprehensive review details what we currently understand about the signaling pathways that support AML cell survival and the exceptional ways in which we disrupt them.
Collapse
Affiliation(s)
- Jenna L Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA.,MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Katie Hege
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jay Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hasini A Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yongwei Su
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA. .,Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA. .,Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
110
|
Gholami M, Bayat S, Pashaiefar H, Pouriamanesh S, Manoochehrabadi S, Behjati F, Mirfakhraie R. Mutational screening of RTK-BRAF genes in de novo adult acute myeloid leukemia. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100904] [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]
|
111
|
NPM1-Mutated Myeloid Neoplasms with <20% Blasts: A Really Distinct Clinico-Pathologic Entity? Int J Mol Sci 2020; 21:ijms21238975. [PMID: 33255988 PMCID: PMC7730332 DOI: 10.3390/ijms21238975] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Nucleophosmin (NPM1) gene mutations rarely occur in non-acute myeloid neoplasms (MNs) with <20% blasts. Among nearly 10,000 patients investigated so far, molecular analyses documented NPM1 mutations in around 2% of myelodysplastic syndrome (MDS) cases, mainly belonging to MDS with excess of blasts, and 3% of myelodysplastic/myeloproliferative neoplasm (MDS/MPN) cases, prevalently classified as chronic myelomonocytic leukemia. These uncommon malignancies are associated with an aggressive clinical course, relatively rapid progression to overt acute myeloid leukemia (AML) and poor survival outcomes, raising controversies on their classification as distinct clinico-pathologic entities. Furthermore, fit patients with NPM1-mutated MNs with <20% blasts could benefit most from upfront intensive chemotherapy for AML rather than from moderate intensity MDS-directed therapies, although no firm conclusion can currently be drawn on best therapeutic approaches, due to the limited available data, obtained from small and mainly retrospective series. Caution is also suggested in definitely diagnosing NPM1-mutated MNs with blast count <20%, since NPM1-mutated AML cases frequently present dysplastic features and multilineage bone marrow cells showing abnormal cytoplasmic NPM1 protein delocalization by immunohistochemical staining, therefore belonging to NPM1-mutated clone regardless of blast morphology. Further prospective studies are warranted to definitely assess whether NPM1 mutations may become sufficient to diagnose AML, irrespective of blast percentage.
Collapse
|
112
|
Koebley SR, Mikheikin A, Leslie K, Guest D, McConnell-Wells W, Lehman JH, Al Juhaishi T, Zhang X, Roberts CH, Picco L, Toor A, Chesney A, Reed J. Digital Polymerase Chain Reaction Paired with High-Speed Atomic Force Microscopy for Quantitation and Length Analysis of DNA Length Polymorphisms. ACS NANO 2020; 14:15385-15393. [PMID: 33169971 DOI: 10.1021/acsnano.0c05897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
DNA length polymorphisms are found in many serious diseases, and assessment of their length and abundance is often critical for accurate diagnosis. However, measuring their length and frequency in a mostly wild-type background, as occurs in many situations, remains challenging due to their variable and repetitive nature. To overcome these hurdles, we combined two powerful techniques, digital polymerase chain reaction (dPCR) and high-speed atomic force microscopy (HSAFM), to create a simple, rapid, and flexible method for quantifying both the size and proportion of DNA length polymorphisms. In our approach, individual amplicons from each dPCR partition are imaged and sized directly. We focused on internal tandem duplications (ITDs) located within the FLT3 gene, which are associated with acute myeloid leukemia and often indicative of a poor prognosis. In an analysis of over 1.5 million HSAFM-imaged amplicons from cell line and clinical samples containing FLT3-ITDs, dPCR-HSAFM returned the expected variant length and variant allele frequency, down to 5% variant samples. As a high-throughput method with single-molecule resolution, dPCR-HSAFM thus represents an advance in HSAFM analysis and a powerful tool for the diagnosis of length polymorphisms.
Collapse
Affiliation(s)
- Sean R Koebley
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Andrey Mikheikin
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Kevin Leslie
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Daniel Guest
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Wendy McConnell-Wells
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Joshua H Lehman
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Taha Al Juhaishi
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Xiaojie Zhang
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Catherine H Roberts
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Loren Picco
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Amir Toor
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Alden Chesney
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Jason Reed
- Physics Department, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
113
|
Koenig K, Mims A, Levis MJ, Horowitz MM. The Changing Landscape of Treatment in Acute Myeloid Leukemia. Am Soc Clin Oncol Educ Book 2020; 40:1-12. [PMID: 32239961 DOI: 10.1200/edbk_279129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The treatment of acute myeloid leukemia is evolving, with increased understanding of molecular pathogenesis allowing better risk stratification and development of new therapies. Tests to identify and drugs to target specific molecular abnormalities are improving remission rates and prolonging survival in patients with high-risk disease. Allogeneic hematopoietic stem cell transplantation remains an important curative therapy, with advances in donor availability and approaches to reduce transplant-related mortality making it applicable in many more patients. Considerations in identifying appropriate patients for targeted therapy and transplantation are presented.
Collapse
Affiliation(s)
- Kristin Koenig
- Division of Hematology, Department of Medicine, The Ohio State University and The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Alice Mims
- Division of Hematology, Department of Medicine, The Ohio State University and The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Mark J Levis
- Hematologic Malignancies and Bone Marrow Transplant Program, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Mary M Horowitz
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
114
|
Xu J, Zhang Y, Hu J, Ren Y, Wang H. Clinical features and prognosis of normal karyotype acute myeloid leukemia pediatric patients with WT1 mutations: an analysis based on TCGA database. ACTA ACUST UNITED AC 2020; 25:79-84. [PMID: 32019476 DOI: 10.1080/16078454.2020.1720102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objectives: To explore the clinical features and prognosis of normal karyotype acute myeloid leukemia (NK-AML) pediatric patients with WT1 mutations.Methods: The clinical data and prognostic information of 220 NK-AML pediatric patients were selected from target-AML project of The Cancer Genome Atlas (TCGA) database. Survival analyses were performed for NK-AML pediatric patients with different combinations of mutations.Results: We found that 28(12.7%) NK-AML patients harbored WT1 mutations. The positive rate of FLT3-ITD in the WT1-mutated group was higher than that in the WT1 wild-type group (P = 0.002). In contrast, WT1 mutation and NPM1 mutation were mutually exclusive (P = 0.013). Furthermore, the WT1-mutated group suffered lower rates of complete remission (CR) (P < 0.001 and P < 0.001, respectively) but higher rates of minimal residual disease (MRD) (P = 0.003 and P = 0.021, respectively) after both one and two courses of induction chemotherapy. Patients with WT1 mutations had significantly worse overall survival (OS) and event-free survival (EFS) in both univariate (P < 0.001 and P = 0.007, respectively) and multivariate survival analyses (P < 0.001 and P < 0.001, respectively). The stratification analysis showed that for FLT3-ITD positive patients, WT1 mutations predicted shorter OS (P = 0.003) and EFS (P < 0.001).Conclusion: WT1 mutations conferred an independent poor prognosis for NK-AML pediatric patients.
Collapse
Affiliation(s)
- Jing Xu
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yaofang Zhang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Jinjun Hu
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yan Ren
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hongwei Wang
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, People's Republic of China.,Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
115
|
Ofran Y, Leiba R, Frisch A, Horesh N, Henig I, Yehudai-Ofir D, Moshe Y, Neaman M, Ganzel C, Gal-Rabinovich K, Hellmann I, Weinstein V, Berger T, Wolach O. Midostaurin in combination with chemotherapy is most effective in patients with acute myeloid leukemia presenting with high FLT3-ITD allelic ratio who proceed to allogeneic stem cell transplantation while in first complete remission. Eur J Haematol 2020; 106:64-71. [PMID: 32949053 DOI: 10.1111/ejh.13518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Midostaurin, a multikinase and FLT3 inhibitor, is the first non-chemotherapy agent approved and widely adopted for the treatment of FLT3-ITD acute myeloid leukemia (AML). Yet, its role in improving survival of patients referred to allogeneic stem cell transplantation (allo-SCT) in first complete remission (CR1) needs to be defined. METHODS This multicenter study retrospectively evaluated the outcome of 119 FLT3-ITD AML patients [59 (49.6%) males and 60 females] intensively treated between 2015 and 2019 at five Israeli centers. In our cohort, allo-SCT in CR1 was widely implemented (47%) and patient stratification was based on the current allelic ratio (AR) cutoff of 0.5. RESULTS Ninety-eight patients (82.3%) achieved CR1/CR with incomplete count recovery (CRi). Death during induction was reported in 7 (5.9%) patients. In multivariate analysis, midostaurin use and allo-SCT in CR1 were the most significant factors affecting overall survival (OS). Midostaurin incorporation in chemotherapy regimens significantly improved CR + CRi rates (P = .002), reduced relapse rates (P = .02), and was remarkably advantageous for high-AR patients (2-year OS 82%). In low-AR patients, the midostaurin effect was much less prominent. CONCLUSIONS Our results demonstrate benefits of midostaurin incorporation in intensive chemotherapy regimens, particularly for high-AR AML patients to whom it should be offered along with allo-SCT in CR1.
Collapse
Affiliation(s)
- Yishai Ofran
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ronit Leiba
- Department of Statistics, Rambam Health Care Campus, Haifa, Israel
| | - Avraham Frisch
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Nurit Horesh
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Israel Henig
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Dana Yehudai-Ofir
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yakir Moshe
- Department of Hematology and Bone Marrow Transplantation, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Miriam Neaman
- Department of Hematology and Bone Marrow Transplantation, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Chezi Ganzel
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Kinneret Gal-Rabinovich
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | | | | | - Tamar Berger
- Hematology Institute, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Ofir Wolach
- Hematology Institute, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
116
|
Aldoss I, Zhang J, Mei M, Al Malki MM, Arslan S, Ngo D, Aribi A, Ali H, Sandhu K, Salhotra A, Koller P, Khaled S, Artz A, Snyder D, Nakamura R, Forman SJ, Stein AS, Marcucci G, Pullarkat V. Venetoclax and hypomethylating agents in FLT3-mutated acute myeloid leukemia. Am J Hematol 2020; 95:1193-1199. [PMID: 32628327 DOI: 10.1002/ajh.25929] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 01/10/2023]
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations are prevalent in acute myeloid leukemia (AML), and their presence confers adverse risk. FLT3-mutated (FLT3m) AML is a challenging leukemia to manage, particularly in older and unfit patients as well as patients with relapsed/refractory (r/r) disease. We retrospectively analyzed the outcomes of 50 FLT3m AML patients (17 treatment-naïve, 33 r/r) treated with venetoclax (VEN) and hypomethylating agents (HMA). The overall CR/CRi rate with VEN-HMA was 60% (94% in treatment-naïve AML and 42% in r/r AML). Early (60-days) treatment related mortality was 2%. The r/r AML setting was an independent predictor of lower complete response (OR: 0.08; 95%CI: 0.00-0.60, P = .03). Cytogenetics-molecular risk, concurrent mutations, the type of FLT3 mutation (ITD vs TKD), the ITD allelic ratio, the type of HMA, age, prior exposure to HMA and receipt of prior allogeneic transplant did not independently impact response or leukemia-free survival (LFS). Concurrent IDH mutations were associated with lower CR/CRi (P = .01), while ASXL1 or TET2 mutations showed a non-significant association toward higher CR/CRi (P = .07, for both). However, none of the concurrent mutations were an independent predictor for response when adjusted to AML setting. In conclusion, VEN-HMA is associated with encouraging efficacy in FLT3m AML among both newly diagnosed unfit and r/r patients.
Collapse
Affiliation(s)
- Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Jianying Zhang
- Department of Information Sciences, City of Hope Medical Center, Duarte, California, USA
| | - Matthew Mei
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Shukaib Arslan
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Dat Ngo
- Department of Pharmacy, City of Hope Medical Center, Duarte, California, USA
| | - Ahmed Aribi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Haris Ali
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Karamjeet Sandhu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Amandeep Salhotra
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Paul Koller
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Samer Khaled
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Andrew Artz
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - David Snyder
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Ryotaro Nakamura
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Anthony S Stein
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Guido Marcucci
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Gehr Family Center for Leukemia Research, Duarte, California, USA
| |
Collapse
|
117
|
Travaglini S, Angelini DF, Alfonso V, Guerrera G, Lavorgna S, Divona M, Nardozza AM, Consalvo MI, Fabiani E, De Bardi M, Neri B, Forghieri F, Marchesi F, Paterno G, Cerretti R, Barragan E, Fiori V, Dominici S, Del Principe MI, Venditti A, Battistini L, Arcese W, Lo-Coco F, Voso MT, Ottone T. Characterization of FLT3-ITD mut acute myeloid leukemia: molecular profiling of leukemic precursor cells. Blood Cancer J 2020; 10:85. [PMID: 32843624 PMCID: PMC7447750 DOI: 10.1038/s41408-020-00352-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022] Open
Abstract
Acute myeloid leukemia (AML) with FLT3-ITD mutations (FLT3-ITDmut) remains a therapeutic challenge, with a still high relapse rate, despite targeted treatment with tyrosine kinase inhibitors. In this disease, the CD34/CD123/CD25/CD99+ leukemic precursor cells (LPCs) phenotype predicts for FLT3-ITD-positivity. The aim of this study was to characterize the distribution of FLT3-ITD mutation in different progenitor cell subsets to shed light on the subclonal architecture of FLT3-ITDmut AML. Using high-speed cell sorting, we sequentially purified LPCs and CD34+ progenitors in samples from patients with FLT3-ITDmut AML (n = 12). A higher FLT3-ITDmut load was observed within CD34/CD123/CD25/CD99+ LPCs, as compared to CD34+ progenitors (CD123+/-,CD25-,CD99low/-) (p = 0.0005) and mononuclear cells (MNCs) (p < 0.0001). This was associated with significantly increased CD99 mean fluorescence intensity in LPCs. Significantly higher FLT3-ITDmut burden was also observed in LPCs of AML patients with a small FLT3-ITDmut clones at diagnosis. On the contrary, the mutation burden of other myeloid genes was similar in MNCs, highly purified LPCs and/or CD34+ progenitors. Treatment with an anti-CD99 mAb was cytotoxic on LPCs in two patients, whereas there was no effect on CD34+ cells from healthy donors. Our study shows that FLT3-ITD mutations occur early in LPCs, which represent the leukemic reservoir. CD99 may represent a new therapeutic target in FLT3-ITDmut AML.
Collapse
Affiliation(s)
- Serena Travaglini
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | | | - Valentina Alfonso
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Gisella Guerrera
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| | - Serena Lavorgna
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Mariadomenica Divona
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Anna Maria Nardozza
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Maria Irno Consalvo
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Emiliano Fabiani
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Marco De Bardi
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| | - Benedetta Neri
- Ematologia, Ospedale S. Eugenio, Dipartimento di Biomedicina e Prevenzione, Rome, Italy
| | - Fabio Forghieri
- University of Modena and Reggio Emilia, Azienda Ospedaliera di Modena, Modena, Italy
| | - Francesco Marchesi
- Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Raffaella Cerretti
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Eva Barragan
- Hospital Universitari i Politècnic La Fe, Valencia, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Valentina Fiori
- Diatheva srl, via Sant'Anna 131, 61030, Cartoceto, (PU), Italy
| | | | | | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Luca Battistini
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| | - William Arcese
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Francesco Lo-Coco
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy.
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy.
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| |
Collapse
|
118
|
Roboz GJ, Strickland SA, Litzow MR, Dalovisio A, Perl AE, Bonifacio G, Haines K, Barbera A, Purkayastha D, Sweet K. Updated safety of midostaurin plus chemotherapy in newly diagnosed FLT3 mutation-positive acute myeloid leukemia: the RADIUS-X expanded access program. Leuk Lymphoma 2020; 61:3146-3153. [PMID: 32812818 DOI: 10.1080/10428194.2020.1805109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Approval of midostaurin, a multikinase inhibitor, in combination with chemotherapy for the treatment of adults with newly diagnosed FLT3 mutation-positive acute myeloid leukemia, was based on the phase 3 RATIFY trial results. RADIUS-X (NCT02624570) was an expanded access program providing access to midostaurin during regulatory review and extending the understanding of the safety and tolerability of midostaurin. Patients aged ≥18 years received midostaurin with 1-2 cycles of induction therapy (cytarabine plus daunorubicin or idarubicin) and ≤4 cycles of high-dose cytarabine consolidation chemotherapy or as single-agent maintenance therapy. The study enrolled 103 patients. No new safety events were observed; toxicities were not influenced by age, anthracycline choice, or coadministration of CYP3A4 inhibitors. The most common adverse events (AEs) were febrile neutropenia, nausea, and diarrhea. During maintenance, 46% of patients reported AEs. Midostaurin demonstrated a manageable safety profile and was associated with high transplant and low on-treatment relapse rates.
Collapse
Affiliation(s)
- Gail J Roboz
- Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
| | | | | | | | | | | | - Kelly Haines
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Alysha Barbera
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | |
Collapse
|
119
|
Transcriptome analysis offers a comprehensive illustration of the genetic background of pediatric acute myeloid leukemia. Blood Adv 2020; 3:3157-3169. [PMID: 31648321 DOI: 10.1182/bloodadvances.2019000404] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Recent advances in the genetic understanding of acute myeloid leukemia (AML) have improved clinical outcomes in pediatric patients. However, ∼40% of patients with pediatric AML relapse, resulting in a relatively low overall survival rate of ∼70%. The objective of this study was to reveal the comprehensive genetic background of pediatric AML. We performed transcriptome analysis (RNA sequencing [RNA-seq]) in 139 of the 369 patients with de novo pediatric AML who were enrolled in the Japanese Pediatric Leukemia/Lymphoma Study Group AML-05 trial and investigated correlations between genetic aberrations and clinical information. Using RNA-seq, we identified 54 in-frame gene fusions and 1 RUNX1 out-of-frame fusion in 53 of 139 patients. Moreover, we found at least 258 gene fusions in 369 patients (70%) through reverse transcription polymerase chain reaction and RNA-seq. Five gene rearrangements were newly identified, namely, NPM1-CCDC28A, TRIP12-NPM1, MLLT10-DNAJC1, TBL1XR1-RARB, and RUNX1-FNBP1. In addition, we found rare gene rearrangements, namely, MYB-GATA1, NPM1-MLF1, ETV6-NCOA2, ETV6-MECOM, ETV6-CTNNB1, RUNX1-PRDM16, RUNX1-CBFA2T2, and RUNX1-CBFA2T3. Among the remaining 111 patients, KMT2A-PTD, biallelic CEBPA, and NPM1 gene mutations were found in 11, 23, and 17 patients, respectively. These mutations were completely mutually exclusive with any gene fusions. RNA-seq unmasked the complexity of gene rearrangements and mutations in pediatric AML. We identified potentially disease-causing alterations in nearly all patients with AML, including novel gene fusions. Our results indicated that a subset of patients with pediatric AML represent a distinct entity that may be discriminated from their adult counterparts. Based on these results, risk stratification should be reconsidered.
Collapse
|
120
|
Ly BTK, Chi HT. Combined effect of (-)-epigallocatechin-3-gallate and all-trans retinoic acid in FLT3-mutated cell lines. Biomed Rep 2020; 13:25. [PMID: 32765864 DOI: 10.3892/br.2020.1332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 05/05/2020] [Indexed: 11/06/2022] Open
Abstract
Patents diagnosed with acute promyelocytic leukemia were treated with Vesanoid® [all-trans retinoic acid (ATRA)]. ATRA promotes the maturation and differentiation of leukemia cells and is therefore capable of reducing the symptoms of leukemia by preventing aggregation of myeloid cells. However, the clinical applications of ATRA are limited by its side effects, including acute retinoid resistance, hypertriglyceridemia, mucocutaneous dryness, nausea, brief recovery time relapse and drug resistance. Therefore, combinations of ATRA and other anticancer drugs are being investigated to overcome these limitations. In our previous study it was shown that in leukemia cells, (-)-epigallocatechin-3-gallate (EGCG) reduced cell proliferation and induced apoptotic cell death. In the present study, an in vitro evaluation of the effects of the combination of EGCG and ATRA on FLT3-mutated cell lines was performed using the isobologram method. The results showed that there was an additive effect in leukemic cells when treated with a combination of ATRA and EGCG. Thus, it was concluded that the cytotoxic effects of EGCG were improved by ATRA.
Collapse
Affiliation(s)
- Bui Thi Kim Ly
- Department of Food Technology, Ho Chi Minh City University of Technology Institute of Applied Sciences, Ho Chi Minh City University of Technology, Ho Chi Minh 72308, Vietnam.,Southern Key Laboratory of Biotechnology, Institute of Fungal Research and Biotechnology, Hanoi 12000, Vietnam
| | - Hoang Thanh Chi
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh 72915, Vietnam.,Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh 72915, Vietnam
| |
Collapse
|
121
|
Abstract
FMS-like tyrosine kinase 3 (FLT3) is mutated in ∼30% of patients that suffer from acute myeloid leukemia (AML). In about 25% of all AML patients, in-frame insertions are observed in the sequence. Most of those insertions are internal tandem duplications (ITDs) of a sequence from the protein. The characteristics of such mutations in terms of length, sequence, and location were hitherto studied in different populations, but not in a comprehensive mutation database. Here, in-frame insertions into the FLT3 gene were extracted from the Catalogue of Somatic Mutations in Cancer (COSMIC) database. These were analyzed with respect to the length, location, and sequence of the mutations. Furthermore, characteristic strings (sequences) of different lengths were identified. Mutations were shown to occur most often in the juxtamembrane zipper (JM-Z) domain of FLT3, followed by the hinge domain and first tyrosine kinase domain (TKD1), upstream of the phosphate-binding loop (P-loop). Interestingly, there are specific hot spot residues where insertions are more likely to occur. The insertions vary in length between one and 67 amino acids, with the largest insertions spanning the phosphate binding loop. Insertions that occur downstream of the P-loop are shorter. Our analysis further shows that acidic and aromatic residues are enriched in the insertions. Finally, molecular dynamics simulations were run for FLT3 with ITD insertions in the hinge and tyrosine kinase domains. On the basis of the findings, a mechanism is proposed for activation by ITDs, according to which there is no direct coupling between the length of the insertion and the activity of the mutated protein. The effect of insertions on the sensitivity of FLT3 to kinase inhibitors is discussed based on our findings.
Collapse
Affiliation(s)
- Guido Todde
- Department of Chemistry and Biomedical Sciences, Linnæus University, 391 82 Kalmar, Sweden
| | - Ran Friedman
- Department of Chemistry and Biomedical Sciences, Linnæus University, 391 82 Kalmar, Sweden
| |
Collapse
|
122
|
Jiang G, Capo-Chichi JM, Liu A, Atenafu EG, Kumar R, Minden MD, Chang H. Combination of FLT3-ITD Allelic Ratio, NPM1 Mutation, and Immunophenotypic Markers to Modulate Outcome Prediction in Patients with Normal Karyotype Acute Myelogenous Leukemia Undergoing Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2020; 26:1995-2000. [PMID: 32712325 DOI: 10.1016/j.bbmt.2020.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/01/2022]
Abstract
NPM1 mutation status and the allelic ratio (AR) of FLT3-internal tandem duplication (FLT3-ITD) are routinely tested for disease risk stratification in patients with normal karyotype (NK) acute myelogenous leukemia (AML); however, the predictive impact of immunophenotypic markers on different NPM1/FLT3 genotypes remains unclear. We performed a retrospective analysis of 423 patients with NK-AML subclassified into groups based on NPM1/FLT3 genotype. Allogeneic hematopoietic stem cell transplantation (HSCT) was performed in 124 of 423 patients (29%) and was significantly associated with longer event-free survival (EFS) and overall survival (OS), except for patients with the favorable genotype, defined as mutated NPM1 (NPM1mut) combined with normal FLT3 status (FLT3-ITDneg) or FLT3-ITD AR <.5 (FLT3-ITDlow). A subset of AML patients bearing the favorable NPM1mut/FLT3-ITDneg/low genotype share similar outcomes with AML patients who have the intermediate FLT3/NPM1 genotype defined by normal NPM1 (NPM1wt) and FLT3-ITDneg/low. In these individuals, the lack of CD13 expression (CD13neg) was associated with shorter EFS (P = .041) and OS (P = .017). CD13neg was an independent predictor for shorter OS (hazard ratio, 1.985; P = .028).
Collapse
Affiliation(s)
- Gina Jiang
- Department of Laboratory Hematology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Jose-Mario Capo-Chichi
- Department of Clinical Laboratory Genetics, Genome Diagnostics, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Aijun Liu
- Department of Hematology, Beijing Chaoyang Hospital, Capital University Beijing, Beijing, China
| | - Eshetu G Atenafu
- Department of Biostatistics, University Health Network, Toronto, Ontario, Canada
| | - Rajat Kumar
- Department of Hematology and Medical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mark D Minden
- Department of Hematology and Medical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hong Chang
- Department of Laboratory Hematology, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
123
|
Practical Considerations for Treatment of Relapsed/Refractory FLT3-ITD Acute Myeloid Leukaemia with Quizartinib: Illustrative Case Reports. Clin Drug Investig 2020; 40:227-235. [PMID: 31912423 PMCID: PMC7035240 DOI: 10.1007/s40261-019-00881-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Quizartinib is a tyrosine kinase inhibitor selectively targeting the FMS-like tyrosine kinase 3 (FLT3) receptor that has been developed for the treatment of acute myeloid leukaemia (AML). The Phase 3 QuANTUM-R study investigated the efficacy of quizartinib monotherapy in patients with relapsed/refractory FLT3-ITD mutation-positive AML. The clinical course of four QuANTUM-R participants exemplifies issues specific to quizartinib treatment and is described here. Patient 1 was FLT3-ITD mutation-negative at AML diagnosis, but became FLT3-ITD mutation-positive during treatment that included several lines of chemotherapy and was therefore a suitable candidate for quizartinib. Because of the clonal shifts of AML during treatment, retesting genetic alterations at each relapse or resistance may help to identify candidates for targeted treatment options. Patient 2 developed QTc prolongation during quizartinib treatment, but the QTc interval normalised after dose reduction, allowing the patient to continue treatment and eventually resume the recommended dose. Patient 3 responded to quizartinib and was scheduled for haematopoietic stem cell transplant (HSCT), but developed febrile neutropenia and invasive aspergillosis during conditioning and subsequently died (to avoid drug-drug interactions, no azole antifungal was administered concomitantly). Care is required when selecting concomitant medications, and if there is potential for interactions (e.g. if prophylactic azole antifungals are required) the quizartinib dose should be reduced to minimise the risk of QTc prolongation. Patient 4 was able to undergo HSCT after responding to quizartinib and experienced a durable response after HSCT while on quizartinib maintenance therapy. Together, these cases illustrate the main issues to be addressed when managing patients under quizartinib, allowing for adequate scheduling and tolerability, bridging to HSCT, and durable remission on maintenance therapy in some patients.
Collapse
|
124
|
Hou HA, Tien HF. Genomic landscape in acute myeloid leukemia and its implications in risk classification and targeted therapies. J Biomed Sci 2020; 27:81. [PMID: 32690020 PMCID: PMC7372828 DOI: 10.1186/s12929-020-00674-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy in terms of clinical features, underlying pathogenesis and treatment outcomes. Recent advances in genomic techniques have unraveled the molecular complexity of AML leukemogenesis, which in turn have led to refinement of risk stratification and personalized therapeutic strategies for patients with AML. Incorporation of prognostic and druggable genetic biomarkers into clinical practice to guide patient-specific treatment is going to be the mainstay in AML therapeutics. Since 2017 there has been an explosion of novel treatment options to tailor personalized therapy for AML patients. In the past 3 years, the U.S. Food and Drug Administration approved a total of eight drugs for the treatment of AML; most specifically target certain gene mutations, biological pathways, or surface antigen. These novel agents are especially beneficial for older patients or those with comorbidities, in whom the treatment choice is limited and the clinical outcome is very poor. How to balance efficacy and toxicity to further improve patient outcome is clinically relevant. In this review article, we give an overview of the most relevant genetic markers in AML with special focus on the therapeutic implications of these aberrations.
Collapse
Affiliation(s)
- Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
125
|
Miyamoto K, Minami Y. Cutting Edge Molecular Therapy for Acute Myeloid Leukemia. Int J Mol Sci 2020; 21:ijms21145114. [PMID: 32698349 PMCID: PMC7404220 DOI: 10.3390/ijms21145114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/31/2022] Open
Abstract
Recently, whole exome sequencing for acute myeloid leukemia (AML) has been performed by a next-generation sequencer in several studies. It has been revealed that a few gene mutations are identified per AML patient. Some of these mutations are actionable mutations that affect the response to an approved targeted treatment that is available for off-label treatment or that is available in clinical trials. The era of precision medicine for AML has arrived, and it is extremely important to detect actionable mutations relevant to treatment decision-making. However, the percentage of actionable mutations found in AML is about 50% at present, and therapeutic development is also needed for AML patients without actionable mutations. In contrast, the newly approved drugs are less toxic than conventional intensive chemotherapy and can be combined with low-intensity treatments. These combination therapies can contribute to the improvement of prognosis, especially in elderly AML patients who account for more than half of all AML patients. Thus, the treatment strategy for leukemia is changing drastically and showing rapid progress. In this review, we present the latest information regarding the recent development of treatment for AML.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Combined Modality Therapy/methods
- Drug Approval
- Epigenesis, Genetic/drug effects
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Molecular Targeted Therapy/methods
- Mutation/drug effects
- Precision Medicine/methods
- Signal Transduction/drug effects
- Small Molecule Libraries/pharmacology
- Small Molecule Libraries/therapeutic use
Collapse
Affiliation(s)
| | - Yosuke Minami
- Correspondence: ; Tel.: +81-4-7133-1111; Fax: +81-7133-6502
| |
Collapse
|
126
|
Kirtonia A, Pandya G, Sethi G, Pandey AK, Das BC, Garg M. A comprehensive review of genetic alterations and molecular targeted therapies for the implementation of personalized medicine in acute myeloid leukemia. J Mol Med (Berl) 2020; 98:1069-1091. [PMID: 32620999 DOI: 10.1007/s00109-020-01944-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/18/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is an extremely heterogeneous disease defined by the clonal growth of myeloblasts/promyelocytes not only in the bone marrow but also in peripheral blood and/or tissues. Gene mutations and chromosomal abnormalities are usually associated with aberrant proliferation and/or block in the normal differentiation of hematopoietic cells. So far, the combination of cytogenetic profiling and molecular and gene mutation analyses remains an essential tool for the classification, diagnosis, prognosis, and treatment for AML. This review gives an overview on how the development of novel innovative technologies has allowed us not only to detect the genetic alterations as early as possible but also to understand the molecular pathogenesis of AML to develop novel targeted therapies. We also discuss the remarkable advances made during the last decade to understand the AML genome both at primary and relapse diseases and how genetic alterations might influence the distinct biological groups as well as the clonal evolution of disease during the diagnosis and relapse. Also, the review focuses on how the persistence of epigenetic gene mutations during morphological remission is associated with relapse. It is suggested that along with the prognostic and therapeutic mutations, the novel molecular targeted therapies either approved by FDA or those under clinical trials including CART-cell therapy would be of immense importance in the effective management of AML.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology (AIB), Amity University, Gurgaon, Haryana, 122413, India
| | - Bhudev C Das
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
127
|
Tsai HK, Brackett DG, Szeto D, Frazier R, MacLeay A, Davineni P, Manning DK, Garcia E, Lindeman NI, Le LP, Lennerz JK, Gibson CJ, Lindsley RC, Kim AS, Nardi V. Targeted Informatics for Optimal Detection, Characterization, and Quantification of FLT3 Internal Tandem Duplications Across Multiple Next-Generation Sequencing Platforms. J Mol Diagn 2020; 22:1162-1178. [PMID: 32603763 PMCID: PMC7479488 DOI: 10.1016/j.jmoldx.2020.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/27/2020] [Accepted: 06/08/2020] [Indexed: 01/09/2023] Open
Abstract
Assessment of internal tandem duplications in FLT3 (FLT3-ITDs) and their allelic ratio (AR) is recommended by clinical guidelines for diagnostic workup of acute myeloid leukemia and traditionally performed through capillary electrophoresis (CE). Although significant progress has been made integrating FLT3-ITD detection within contemporary next-generation sequencing (NGS) panels, AR estimation is not routinely part of clinical NGS practice because of inherent biases and challenges. In this study, data from multiple NGS platforms—anchored multiplex PCR (AMP), amplicon [TruSeq Custom Amplicon (TSCA)], and hybrid-capture—were analyzed through a custom algorithm, including platform-specific measures of AR. Sensitivity and specificity of NGS for FLT3-ITD status relative to CE were 100% (42/42) and 99.4% (1076/1083), respectively, by AMP on an unselected cohort and 98.1% (53/54) and 100% (48/48), respectively, by TSCA on a selected cohort. Primer analysis identified criteria for ITDs to escape detection by TSCA, estimated to occur in approximately 9% of unselected ITDs. Allelic fractions under AMP or TSCA were highly correlated to CE, with linear regression slopes near 1 for ITDs not duplicating primers, and systematically underestimated for ITDs duplicating a primer. Bias was alleviated in AMP through simple adjustments. This article provides an approach for targeted computational FLT3-ITD analysis for NGS data from multiple platforms; AMP was found capable of near perfect sensitivity and specificity with relatively accurate estimates of ARs.
Collapse
Affiliation(s)
- Harrison K Tsai
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Diane G Brackett
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - David Szeto
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ryan Frazier
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Allison MacLeay
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Phani Davineni
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Danielle K Manning
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Elizabeth Garcia
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Long P Le
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jochen K Lennerz
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Christopher J Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - R Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
128
|
Impact of NPM1/FLT3-ITD genotypes defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood 2020; 135:371-380. [PMID: 31826241 DOI: 10.1182/blood.2019002697] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Patients with acute myeloid leukemia (AML) harboring FLT3 internal tandem duplications (ITDs) have poor outcomes, in particular AML with a high (≥0.5) mutant/wild-type allelic ratio (AR). The 2017 European LeukemiaNet (ELN) recommendations defined 4 distinct FLT3-ITD genotypes based on the ITD AR and the NPM1 mutational status. In this retrospective exploratory study, we investigated the prognostic and predictive impact of the NPM1/FLT3-ITD genotypes categorized according to the 2017 ELN risk groups in patients randomized within the RATIFY trial, which evaluated the addition of midostaurin to standard chemotherapy. The 4 NPM1/FLT3-ITD genotypes differed significantly with regard to clinical and concurrent genetic features. Complete ELN risk categorization could be done in 318 of 549 trial patients with FLT3-ITD AML. Significant factors for response after 1 or 2 induction cycles were ELN risk group and white blood cell (WBC) counts; treatment with midostaurin had no influence. Overall survival (OS) differed significantly among ELN risk groups, with estimated 5-year OS probabilities of 0.63, 0.43, and 0.33 for favorable-, intermediate-, and adverse-risk groups, respectively (P < .001). A multivariate Cox model for OS using allogeneic hematopoietic cell transplantation (HCT) in first complete remission as a time-dependent variable revealed treatment with midostaurin, allogeneic HCT, ELN favorable-risk group, and lower WBC counts as significant favorable factors. In this model, there was a consistent beneficial effect of midostaurin across ELN risk groups.
Collapse
|
129
|
Liang C, Peng CJ, Wang LN, Li Y, Zheng LM, Fan Z, Huang DP, Tang WY, Zhang XL, Huang LB, Tang YL, Luo XQ. Arsenic trioxide and all-trans retinoic acid suppress the expression of FLT3-ITD. Leuk Lymphoma 2020; 61:2692-2699. [PMID: 32536235 DOI: 10.1080/10428194.2020.1775212] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Cong Liang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chun-Jin Peng
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Na Wang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Li
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Min Zheng
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Fan
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan-Ping Huang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen-Yan Tang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Li Zhang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Bin Huang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Lai Tang
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xue-Qun Luo
- Department of Pediatrics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
130
|
Di Ciaccio P, Kearney A, Ling S. Comparing the area under the curve and peak height methods in the calculation of FLT3-ITD allelic ratio. Int J Lab Hematol 2020; 42:e234-e236. [PMID: 32533751 DOI: 10.1111/ijlh.13263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/23/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Pietro Di Ciaccio
- Department of Haematology, Liverpool Hospital, Sydney, NSW, Australia
| | - Anabel Kearney
- Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Silvia Ling
- Department of Haematology, Liverpool Hospital, Sydney, NSW, Australia.,Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| |
Collapse
|
131
|
Nucleophosmin 1 Mutations in Acute Myeloid Leukemia. Genes (Basel) 2020; 11:genes11060649. [PMID: 32545659 PMCID: PMC7348733 DOI: 10.3390/genes11060649] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022] Open
Abstract
Nucleophosmin (NPM1) is a ubiquitously expressed nucleolar protein involved in ribosome biogenesis, the maintenance of genomic integrity and the regulation of the ARF-p53 tumor-suppressor pathway among multiple other functions. Mutations in the corresponding gene cause a cytoplasmic dislocation of the NPM1 protein. These mutations are unique to acute myeloid leukemia (AML), a disease characterized by clonal expansion, impaired differentiation and the proliferation of myeloid cells in the bone marrow. Despite our improved understanding of NPM1 mutations and their consequences, the underlying leukemia pathogenesis is still unclear. Recent studies that focused on dysregulated gene expression in AML with mutated NPM1 have shed more light into these mechanisms. In this article, we review the current evidence on normal functions of NPM1 and aberrant functioning in AML, and highlight investigational strategies targeting these mutations.
Collapse
|
132
|
Bazarbachi A, Bug G, Baron F, Brissot E, Ciceri F, Dalle IA, Döhner H, Esteve J, Floisand Y, Giebel S, Gilleece M, Gorin NC, Jabbour E, Aljurf M, Kantarjian H, Kharfan-Dabaja M, Labopin M, Lanza F, Malard F, Peric Z, Prebet T, Ravandi F, Ruggeri A, Sanz J, Schmid C, Shouval R, Spyridonidis A, Versluis J, Vey N, Savani BN, Nagler A, Mohty M. Clinical practice recommendation on hematopoietic stem cell transplantation for acute myeloid leukemia patients with FLT3-internal tandem duplication: a position statement from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Haematologica 2020; 105:1507-1516. [PMID: 32241850 PMCID: PMC7271578 DOI: 10.3324/haematol.2019.243410] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
The FMS-like tyrosine kinase 3 (FLT3) gene is mutated in 25-30% of patients with acute myeloid leukemia (AML). Because of the poor prognosis associated with FLT3-internal tandem duplication mutated AML, allogeneic hematopoietic stem-cell transplantation (SCT) was commonly performed in first complete remission. Remarkable progress has been made in frontline treatments with the incorporation of FLT3 inhibitors and the development of highly sensitive minimal/measurable residual disease assays. Similarly, recent progress in allogeneic hematopoietic SCT includes improvement of transplant techniques, the use of haploidentical donors in patients lacking an HLA matched donor, and the introduction of FLT3 inhibitors as post-transplant maintenance therapy. Nevertheless, current transplant strategies vary between centers and differ in terms of transplant indications based on the internal tandem duplication allelic ratio and concomitant nucleophos-min-1 mutation, as well as in terms of post-transplant maintenance/consolidation. This review generated by international leukemia or transplant experts, mostly from the European Society for Blood and Marrow Transplantation, attempts to develop a position statement on best approaches for allogeneic hematopoietic SCT for AML with FLT3-internal tandem duplication including indications for and modalities of such transplants and on the potential optimization of post-transplant maintenance with FLT inhibitors.
Collapse
Affiliation(s)
- Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Gesine Bug
- Department of Medicine 2, Hematology and Oncology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Eolia Brissot
- Sorbonne Universités, UPMC University of Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Hematology Department, AP-HP, Saint Antoine Hospital, Paris, France
| | - Fabio Ciceri
- Vita-Salute San Raffaele University of Milan, Milan, ItalyHematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Iman Abou Dalle
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Jordi Esteve
- Hematology Department, Hospital Clínic of Barcelona, IDIBAPS, University of Barcelona, Barcellona, Spain
| | - Yngvar Floisand
- Department of Hematology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Oncohematology, Maria Sklodowska-Curie Institute - Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Maria Gilleece
- Department of Haematology, Leeds Teaching Hospitals Trust, Leeds, UK
| | - Norbert-Claude Gorin
- Department of Hematology and Cell Therapy, European Society for Blood and Marrow Transplantation, Paris Office, Hopital Saint-Antoine, Paris, France
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahmoud Aljurf
- Department of Hematology King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapies Program, Mayo Clinic, Jacksonville, FL, USA
| | - Myriam Labopin
- Acute Leukemia Working Party, Paris Study Office, European Society for Blood and Marrow Transplantation, Paris, France
- Sorbonne Universités, UPMC University of Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Hematology Department, AP-HP, Saint Antoine Hospital, Paris, France
| | | | - Florent Malard
- Sorbonne Universités, UPMC University of Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Hematology Department, AP-HP, Saint Antoine Hospital, Paris, France
| | - Zinaida Peric
- University Hospital Center Zagreb, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Thomas Prebet
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Annalisa Ruggeri
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Roma, ItalyEurocord, Hôpital Saint Louis, Paris, France
| | - Jaime Sanz
- Hematology Department, Hospital Universitari i Politecnic La Fe. Instituto de Investigación Sanitaria La Fe, Valencia, CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Christoph Schmid
- Department of Hematology and Oncology, Augsburg University Hospital, Augsburg, Germany
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jurjen Versluis
- Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Norbert Vey
- Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Bipin N Savani
- Department of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Arnon Nagler
- Hematology and Bone Marrow Transplantation Division, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Israel
| | - Mohamad Mohty
- Sorbonne Universités, UPMC University of Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Hematology Department, AP-HP, Saint Antoine Hospital, Paris, France
| |
Collapse
|
133
|
Soare DS, Radu E, Dumitru I, Popov VM, Bumbea H, Vlădăreanu AM. FLT3-ITD DNA and mRNA levels in AML do not correlate with CD7, CD33 and CD123 expression. J Cell Mol Med 2020; 24:7675-7679. [PMID: 32460405 PMCID: PMC7339186 DOI: 10.1111/jcmm.15255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/31/2022] Open
Abstract
Introduction FLT3 internal tandem duplication (ITD) mutations are found in around 25% of all acute myeloid leukaemia (AML) cases and is associated with shorter disease‐free and overall survival. Previous reports have shown that FLT3‐ITD induces a specific phenotype in leukemic blasts, which is characterized by high levels of CD33 and CD123, and that expression of CD33 and CD123 is directly influenced by the DNA FLT3‐ITD/wild‐type FLT3 allelic ratio (AR). Methods A total of 42 FLT3‐ITD and 104 FLT3‐ITD–negative AML patients were analysed. Immunophenotyping data were used to calculate antigen expression levels as the ratio between the geometric mean fluorescence intensities (MFIs) of leukemic blasts and MFIs of negative lymphocyte populations. FLT3‐ITD‐DNA and RNA analysis was performed, under the same conditions, by capillary electrophoresis. Results Compared with the control group, the FLT3‐ITD cohort presented significantly higher CD7, CD33 and CD123 levels. In order to assess the impact of FLT3‐ITD abundance on antigen expression, the patients were grouped for each parameter into two cohorts using the following threshold values: (a) 0.5 for the AR, according to current AML guidelines; (b) 0.7 for the FLT3‐ITD/FLT3‐WT mRNA ratio (RR); and (c) 1.3 for the FLT3‐ITD RR/AR ratio. We found higher values of CD33 for RR/AR ≥1.3, and no other statistical differences between CD7, CD33 and CD123 levels of the other FLT3‐ITD groups. In terms of correlations between MFI values and FLT3‐ITD parameters, we only observed a moderate interdependence between CD33 MFI and the RR/AR ratio, and a weak negative correlation between CD123 MFI and AR. Conclusion FLT3‐ITD mutations induce a specific antigen profile in AML blasts, and our data do not onfirm previous reports of FLT3‐ITD AR influencing both CD33 and CD123 expression.
Collapse
Affiliation(s)
- Dan-Sebastian Soare
- University Emergency Hospital Bucharest, Bucharest, Romania.,Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Eugen Radu
- University Emergency Hospital Bucharest, Bucharest, Romania.,Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ion Dumitru
- University Emergency Hospital Bucharest, Bucharest, Romania.,Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Viola Maria Popov
- Department of Hematology, Colentina Clinical Hospital, Bucharest, Romania
| | - Horia Bumbea
- University Emergency Hospital Bucharest, Bucharest, Romania.,Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ana Maria Vlădăreanu
- University Emergency Hospital Bucharest, Bucharest, Romania.,Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
134
|
Kawashima N, Ishikawa Y, Atsuta Y, Sawa M, Ozawa Y, Hayashi M, Kohno A, Tomita A, Maeda T, Sakaida E, Usuki K, Hagihara M, Kanamori H, Matsuoka H, Kobayashi M, Asou N, Ohtake S, Matsumura I, Miyazaki Y, Naoe T, Kiyoi H. Allogeneic hematopoietic stem cell transplantation at the first remission for younger adults with FLT3-internal tandem duplication AML: The JALSG AML209-FLT3-SCT study. Cancer Sci 2020; 111:2472-2481. [PMID: 32391628 PMCID: PMC7484840 DOI: 10.1111/cas.14448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/16/2020] [Accepted: 05/03/2020] [Indexed: 01/14/2023] Open
Abstract
In this phase II multicenter study (JALSG AML209-FLT3-SCT), we aimed to prospectively elucidate the role of allogeneic hematopoietic stem cell transplantation (allo-HSCT) at first complete remission (CR1) for FLT3-internal tandem duplication (ITD)-positive AML. Newly diagnosed de novo AML patients with FLT3-ITD were enrolled at the achievement of CR1 and received allo-HSCT as soon as possible after the first consolidation therapy. Mutations of 57 genes in AML cells at diagnosis were also analyzed. Among 48 eligible patients with a median age of 38.5 (17-49) years, 36 (75%) received allo-HSCT at a median of 108 days after CR1. The median follow-up was 1726 days. The primary end-point, 3-year disease-free survival (DFS) based on an intent to treat analysis, was 43.8% (95% confidence interval [CI], 30%-57%), suggesting the efficacy of this treatment because the lower limit of the 95% CI exceeded the threshold response rate of 20%. The 3-year overall survival, post-transplant DFS, and non-relapse mortality rates were 54.2% (95% CI, 39%-67%), 58.3% (95% CI, 41%-72%), and 25.0% (95% CI, 12%-40%), respectively. The median ITD allelic ratio (AR) was 0.344 (0.006-4.099). Neither FLT3-ITD AR nor cooccurring genetic alterations was associated with a poor DFS. This prospective study indicated the efficacy and safety of allo-HSCT for FLT3-ITD AML patients in CR1. This study was registered at: www.umin.ac.jp/ctr/ as #UMIN000003433.
Collapse
Affiliation(s)
- Naomi Kawashima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Anjo, Japan
| | - Yukiyasu Ozawa
- Department of Hematology, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Masaki Hayashi
- Department of Hematology, Nakagami Hospital, Okinawa, Japan
| | - Akio Kohno
- Department of Hematology and Oncology, JA Aichi Konan Kosei Hospital, Konan, Japan
| | - Akihiro Tomita
- Department of Hematology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tomoya Maeda
- Department of Hemato-Oncology, International Medical Center, Saitama Medical University, Hidaka, Japan
| | - Emiko Sakaida
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Kensuke Usuki
- Department of Hematology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Maki Hagihara
- Department of Hematology and Clinical Immunology, Yokohama City University Hospital, Yokohama, Japan
| | - Heiwa Kanamori
- Department of Hematology, Kanagawa Cancer Center, Yokohama, Japan
| | - Hiroshi Matsuoka
- The Division of Medical Oncology and Hematology, Department of Medicine, Kobe University Hospital, Kobe, Japan
| | - Miki Kobayashi
- Department of Hematology and Oncology, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Norio Asou
- Department of Hemato-Oncology, International Medical Center, Saitama Medical University, Hidaka, Japan
| | | | - Itaru Matsumura
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomoki Naoe
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | |
Collapse
|
135
|
Shouval R, Labopin M, Bomze D, Baerlocher GM, Capria S, Blaise D, Hänel M, Forcade E, Huynh A, Saccardi R, Milone G, Zuckerman T, Reményi P, Versluis J, Esteve J, Gorin NC, Mohty M, Nagler A. Risk stratification using FLT3 and NPM1 in acute myeloid leukemia patients autografted in first complete remission. Bone Marrow Transplant 2020; 55:2244-2253. [PMID: 32388535 DOI: 10.1038/s41409-020-0936-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 11/09/2022]
Abstract
FLT3-ITD and NPM1 mutation refine prognostic stratification in acute myeloid leukemia (AML) with intermediate-risk cytogenetics. However, data on their role in patients undergoing autologous stem cell transplantation (Auto-SCT) as post-remission therapy (PRT) are limited. We therefore sought to retrospectively evaluate the role of FLT3-ITD and NPM1 in a cohort of AML patients (n = 405) with intermediate-risk cytogenetics, autografted in first complete remission (CR1). Patients were transplanted between 2000 and 2014 and reported to the European Society for Blood and Marrow Transplantation (EBMT) registry. Leukemia-free survival (LFS) was the primary outcome. Median follow-up was 5.5 years. FLT3-ITDneg/NPM1WT was the leading molecular subtype (50%), followed by FLT3-ITDneg/NPM1mut (30%). In the univariate analysis, molecular subtype was associated with LFS, overall survival (OS), and relapse incidence (RI) (p < 0.001); 5-year LFS: FLT3-ITDneg/NPM1mut 62%, FLT3-ITDpos/NPM1mut 38%, FLT3-ITDneg/NPM1WT 32%, and FLT3-ITDpos/NPM1WT 21%. At 5 years, OS and RI in the FLT3-ITDneg/NPM1mut subtype were 74% and 35%, respectively. The corresponding OS and RI in other subtypes were below 48% and over 57%. In a Cox multivariable model, molecular subtype was the strongest predictor of LFS, OS, and relapse. In conclusion, AML patients with intermediate-risk cytogenetics and FLT3-ITDneg/NPM1mut experience favorable outcomes when autografted in CR1, suggesting that Auto-SCT is a valid PRT option.
Collapse
Affiliation(s)
- Roni Shouval
- Hematology and Bone Marrow Transplantation Division, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel. .,Dr. Pinchas Bornstein Talpiot Medical Leadership Program, Chaim Sheba Medical Center, Tel-Hashomer, Israel.
| | - Myriam Labopin
- Acute Leukemia Working Party of EBMT, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - David Bomze
- Hematology and Bone Marrow Transplantation Division, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriela M Baerlocher
- Department of Hematology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Saveria Capria
- Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University, Policlinico Umberto I, Rome, Italy
| | - Didier Blaise
- Department of Hematology, Institut Paoli Calmettes, Marseille, France
| | - Mathias Hänel
- Department of Hematology, Oncology, Stem Cell Transplantation, Hospital Chemnitz, Chemnitz, Germany
| | - Edouard Forcade
- Hématologie Clinique et Thérapie cellulaire, CHU Bordeaux, Pessac, France
| | - Anne Huynh
- CHU Toulouse, IUCT-Oncopole, Toulouse, France
| | | | | | - Tsila Zuckerman
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Péter Reményi
- St. István and St. László Hospital of Budapest, Budapest, Hungary
| | - Jurjen Versluis
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Jordi Esteve
- Hematology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Mohamad Mohty
- Hospital Saint-Antoine, APHP, Sorbonne University, INSERM U938, Paris, France
| | - Arnon Nagler
- Hematology and Bone Marrow Transplantation Division, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Acute Leukemia Working Party of EBMT, Paris, France
| |
Collapse
|
136
|
Abou Dalle I, Ghorab A, Patel K, Wang X, Hwang H, Cortes J, Issa GC, Yalniz F, Sasaki K, Chihara D, Price A, Kadia T, Pemmaraju N, Daver N, DiNardo C, Ravandi F, Kantarjian HM, Borthakur G. Impact of numerical variation, allele burden, mutation length and co-occurring mutations on the efficacy of tyrosine kinase inhibitors in newly diagnosed FLT3- mutant acute myeloid leukemia. Blood Cancer J 2020; 10:48. [PMID: 32366841 PMCID: PMC7198530 DOI: 10.1038/s41408-020-0318-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/07/2020] [Accepted: 02/17/2020] [Indexed: 11/09/2022] Open
Abstract
FLT3-ITD mutations in newly diagnosed acute myeloid leukemia (AML) are associated with worse overall survival (OS). FLT3-ITD diversity can further influence clinical outcomes. Addition of FLT3 inhibitors to standard chemotherapy has improved OS. The aim of this study is to evaluate the prognostic impact of FLT3 diversity and identify predictors of efficacy of FLT3 inhibitors. We reviewed prospectively collected data from 395 patients with newly diagnosed FLT3-ITD mutant AML. 156 (39%) patients received FLT3 inhibitors combined with either high or low intensity chemotherapy. There was no statistically significant difference in clinical outcomes among patients treated with FLT3 inhibitors based on FLT3 numerical variation (p = 0.85), mutation length (p = 0.67). Overall, the addition of FLT3 inhibitor to intensive chemotherapy was associated with an improved OS (HR = 0.35, 95% CI: 0.24-0.5, p = 0.0005), but not in combination with lower intensity chemotherapy (HR = 0.98, 95%CI: 0.7-1.36, p = 0.85). A differential effect of FLT3 inhibitor on OS was more pronounced in younger patients with FLT3 allelic ratio ≥0.5 (HR = 0.41, 95% CI: 0.25-0.66, p < 0.001), single ITD mutation (HR = 0.55, 95% CI: 0.34-0.88, p = 0.01), diploid cytogenetics (HR = 0.52, 95% CI: 0.35-0.76, p = 0.001), NPM1 co-mutation (HR = 0.35, 95% CI: 0.19-0.67, p = 0.001). Our analysis identifies predictors of survival among diverse FLT3 related variables in patients treated with FLT3 inhibitor.
Collapse
Affiliation(s)
- Iman Abou Dalle
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmad Ghorab
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur Patel
- Department of hemopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuemei Wang
- Department of biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hyunsoo Hwang
- Department of biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jorge Cortes
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ghayas C Issa
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fevzi Yalniz
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dai Chihara
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Allyson Price
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney DiNardo
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
137
|
Sakaguchi M, Nakajima N, Yamaguchi H, Najima Y, Shono K, Marumo A, Omori I, Fujiwara Y, Terada K, Yui S, Wakita S, Mitaya M, Arai K, Kitano T, Doki N, Ohashi K, Inokuchi K. The sensitivity of the FLT3-ITD detection method is an important consideration when diagnosing acute myeloid leukemia. Leuk Res Rep 2020; 13:100198. [PMID: 32368486 PMCID: PMC7184170 DOI: 10.1016/j.lrr.2020.100198] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 11/23/2022] Open
Abstract
Fms-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) is a key predictive factor for the prognosis of acute myeloid leukemia (AML). We compared the detection sensitivity of fragment analysis with that of PCR-electrophoresis using MV4-11 (FLT3-ITD) and NKM-1 (FLT3-wild type) cell lines. DNA of these cells was mixed at different ratios and subjected to PCR-electrophoresis or fragment analysis. PCR-electrophoresis was found to have an FLT3-ITD allelic ratio (AR) detection limit of 0.034-0.072. Visual inspection of the PCR-electrophoresis revealed a lower detection sensitivity than that of fragment analysis. Therefore, it is essential to conduct fragment analysis when screening for FLT3-ITD.
Collapse
Affiliation(s)
| | - Nana Nakajima
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | | | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Katsuhiro Shono
- Department of Hematology, Chiba Aoba Municipal Hospital, Chiba, Japan
| | - Atsushi Marumo
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Ikuko Omori
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Yusuke Fujiwara
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Kazuki Terada
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Shunsuke Yui
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Satoshi Wakita
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Miho Mitaya
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Kunihito Arai
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Tomoaki Kitano
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kazuteru Ohashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Koiti Inokuchi
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
138
|
Terminal deoxynucleotidyl transferase promotes acute myeloid leukemia by priming FLT3-ITD replication slippage. Blood 2020; 134:2281-2290. [PMID: 31650168 DOI: 10.1182/blood.2019001238] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 09/30/2019] [Indexed: 12/30/2022] Open
Abstract
FLT3-internal tandem duplications (FLT3-ITDs) are prognostic driver mutations found in acute myeloid leukemia (AML). Although these short duplications occur in 25% of AML patients, little is known about the molecular mechanism underlying their formation. Understanding the origin of FLT3-ITDs would advance our understanding of the genesis of AML. We analyzed the sequence and molecular anatomy of 300 FLT3-ITDs to address this issue, including 114 ITDs with additional nucleotides of unknown origin located between the 2 copies of the repeat. We observed anatomy consistent with replication slippage, but could only identify the germline microhomology (1-6 bp) anticipated to prime such slippage in one-third of FLT3-ITDs. We explain the paradox of the "missing" microhomology in the majority of FLT3-ITDs through occult microhomology: specifically, by priming through use of nontemplated nucleotides (N-nucleotides) added by terminal deoxynucleotidyl transferase (TdT). We suggest that TdT-mediated nucleotide addition in excess of that required for priming creates N-regions at the duplication junctions, explaining the additional nucleotides observed at this position. FLT3-ITD N-regions have a G/C content (66.9%), dinucleotide composition (P < .001), and length characteristics consistent with synthesis by TdT. AML types with high TdT show an increased incidence of FLT3-ITDs (M0; P = .0017). These results point to an unexpected role for the lymphoid enzyme TdT in priming FLT3-ITDs. Although the physiological role of TdT is to increase antigenic diversity through N-nucleotide addition during V(D)J recombination of IG/TCR genes, here we propose that illegitimate TdT activity makes a significant contribution to the genesis of AML.
Collapse
|
139
|
Tzogani K, Røshol H, Olsen HH, Aas IB, Dalhus ML, Håkonsen GD, Nilssen LS, Lindberg V, Økvist M, Bolstad B, Rogovska I, Karpova N, Enzmann H, Gisselbrecht C, Pignatti F. The European Medicines Agency Review of Gilteritinib (Xospata) for the Treatment of Adult Patients with Relapsed or Refractory Acute Myeloid Leukemia with an FLT3 Mutation. Oncologist 2020; 25:e1070-e1076. [PMID: 32154636 DOI: 10.1634/theoncologist.2019-0976] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/14/2020] [Indexed: 11/17/2022] Open
Abstract
On October 24, 2019, a marketing authorization valid through the European Union (EU) was issued for gilteritinib monotherapy for adult patients who have relapsed or refractory acute myeloid leukemia (AML) with an Fms-like tyrosine kinase 3 (FLT3) mutation. Gilteritinib inhibits FLT3 receptor signaling and proliferation in cells exogenously expressing FLT3 including FLT3 internal tandem duplication (ITD), FLT3 D835Y, and FLT3 ITD D835Y, and it induced apoptosis in leukemic cells expressing FLT3 ITD. The recommended starting dose of gilteritinib is 120 mg (three 40 mg tablets) once daily. Gilteritinib was evaluated in one, phase III, open-label, multicenter, randomized study of gilteritinib (n = 247, gilteritinib arm) versus salvage chemotherapy (n = 124, salvage chemotherapy arm) in patients with relapsed or refractory AML with FLT3 mutation. Overall survival (OS) was statistically significantly different between the two groups with a median OS of 9.3 months in the gilteritinib arm compared with 5.6 months for salvage chemotherapy (hazard ratio, 0.637; 95% confidence interval, 0.490-0.830; p = .0004 one-sided log-rank test). The most common adverse reactions with gilteritinib treatment were blood creatine phosphokinase increase, alanine aminotransferase increase, aspartate aminotransferase increase, blood alkaline phosphatase increase, diarrhea, fatigue, nausea, constipation, cough, peripheral edema, dyspnea, dizziness, hypotension, pain in extremity, asthenia, arthralgia, and myalgia. The objective of this article is to summarize the scientific review of the application leading to regulatory approval in the EU. IMPLICATIONS FOR PRACTICE: Xospata was approved in the European Union as monotherapy for the treatment of adult patients with relapsed or refractory acute myeloid leukemia (AML) with an Fms-like tyrosine kinase 3 (FLT3) mutation. Gilteritinib resulted in a clinically meaningful and statistically significant improvement of overall survival compared with salvage chemotherapy. At the time of the marketing authorization of gilteritinib, there were no approved standard therapies specifically for adult patients diagnosed with relapsed or refractory AML with FLT3 mutation. In terms of safety, the overall accepted safety profile was considered manageable.
Collapse
Affiliation(s)
| | - Hilde Røshol
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Helga Haugom Olsen
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Ida B Aas
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Marianne Løiten Dalhus
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Gro Dahlseng Håkonsen
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Laila Sortvik Nilssen
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Vibeke Lindberg
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Mats Økvist
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Bjørg Bolstad
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Statens legemiddelverk, Oslo, Norway
| | - Irēna Rogovska
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Zāļu valsts aǵentūra, Riga, Latvia
| | - Natalja Karpova
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Zāļu valsts aǵentūra, Riga, Latvia
| | - Harald Enzmann
- Committee for Medicinal Products for Human Use (CHMP), Amsterdam, The Netherlands
- Bundesinstitut für Arzneimittel und Medizinprodukte, Bonn, Germany
| | | | | |
Collapse
|
140
|
Arindrarto W, Borràs DM, de Groen RAL, van den Berg RR, Locher IJ, van Diessen SAME, van der Holst R, van der Meijden ED, Honders MW, de Leeuw RH, Verlaat W, Jedema I, Kroes WGM, Knijnenburg J, van Wezel T, Vermaat JSP, Valk PJM, Janssen B, de Knijff P, van Bergen CAM, van den Akker EB, Hoen PAC', Kiełbasa SM, Laros JFJ, Griffioen M, Veelken H. Comprehensive diagnostics of acute myeloid leukemia by whole transcriptome RNA sequencing. Leukemia 2020; 35:47-61. [PMID: 32127641 PMCID: PMC7787979 DOI: 10.1038/s41375-020-0762-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 01/17/2020] [Accepted: 02/12/2020] [Indexed: 01/12/2023]
Abstract
Acute myeloid leukemia (AML) is caused by genetic aberrations that also govern the prognosis of patients and guide risk-adapted and targeted therapy. Genetic aberrations in AML are structurally diverse and currently detected by different diagnostic assays. This study sought to establish whole transcriptome RNA sequencing as single, comprehensive, and flexible platform for AML diagnostics. We developed HAMLET (Human AML Expedited Transcriptomics) as bioinformatics pipeline for simultaneous detection of fusion genes, small variants, tandem duplications, and gene expression with all information assembled in an annotated, user-friendly output file. Whole transcriptome RNA sequencing was performed on 100 AML cases and HAMLET results were validated by reference assays and targeted resequencing. The data showed that HAMLET accurately detected all fusion genes and overexpression of EVI1 irrespective of 3q26 aberrations. In addition, small variants in 13 genes that are often mutated in AML were called with 99.2% sensitivity and 100% specificity, and tandem duplications in FLT3 and KMT2A were detected by a novel algorithm based on soft-clipped reads with 100% sensitivity and 97.1% specificity. In conclusion, HAMLET has the potential to provide accurate comprehensive diagnostic information relevant for AML classification, risk assessment and targeted therapy on a single technology platform.
Collapse
Affiliation(s)
- Wibowo Arindrarto
- Center for Computational Biology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Daniel M Borràs
- GenomeScan B.V, 2333 BZ, Leiden, The Netherlands.,Department of Chemical Cell Biology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Ruben A L de Groen
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Redmar R van den Berg
- Department of Human Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Irene J Locher
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | | | - Rosalie van der Holst
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | | | - M Willy Honders
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Rick H de Leeuw
- Forensic Laboratory for DNA Research, Department of Human Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Wina Verlaat
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Inge Jedema
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Wilma G M Kroes
- Department of Clinical Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Jeroen Knijnenburg
- Department of Clinical Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Joost S P Vermaat
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Peter J M Valk
- Department of Hematology, Erasmus University Medical Center, 3015CN, Rotterdam, The Netherlands
| | - Bart Janssen
- GenomeScan B.V, 2333 BZ, Leiden, The Netherlands
| | - Peter de Knijff
- Forensic Laboratory for DNA Research, Department of Human Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | | | - Erik B van den Akker
- Center for Computational Biology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands.,The Delft Bioinformatics Lab, Delft University of Technology, 2628CD, Delft, The Netherlands.,Section of Molecular Epidemiology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands.,The Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Szymon M Kiełbasa
- Center for Computational Biology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Jeroen F J Laros
- Department of Human Genetics, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands.
| | - Hendrik Veelken
- Department of Hematology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| |
Collapse
|
141
|
Linch DC, Hills RK, Burnett AK, Russell N, Gale RE. Analysis of the clinical impact of NPM1 mutant allele burden in a large cohort of younger adult patients with acute myeloid leukaemia. Br J Haematol 2020; 188:852-859. [PMID: 31595497 DOI: 10.1111/bjh.16239] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/14/2019] [Indexed: 02/02/2023]
Abstract
Although an NPM1 mutation is generally considered to be a good prognostic marker in acute myeloid leukaemia, it has recently been suggested that a higher level of NPM1 mutant (NPM1MUT ) alleles relative to wild-type alleles is associated with poor clinical outcome. We therefore sought to confirm this finding in a larger study of 876 NPM1MUT cases entered into UK national trials. In univariate analysis, the higher NPM1MUT allele burden was associated with a lower complete remission (CR) rate, higher relapse rate and reduced overall survival, but this was largely attributable to the association of the higher NPM1MUT allele burden with other known poor risk factors, particularly the presence of a concomitant FLT3 internal tandem duplication. In multivariate analysis, there was no significant impact of the NPM1MUT allele burden on CR rates, and the impact on relapse and overall survival, whilst still significant, was greatly reduced. This impact was similar in patients who did or did not receive an allogeneic transplant in first CR. We conclude that the binary presence or absence of an NPM1 mutation, combined with minimal residual disease levels following induction therapy, should continue to be used in therapeutic management rather than stratification according to the NPM1MUT level.
Collapse
Affiliation(s)
- David C Linch
- Department of Haematology, UCL Cancer Institute, London, UK
| | | | - Alan K Burnett
- Department of Haematology, School of Medicine, Cardiff University, Cardiff, UK
| | - Nigel Russell
- Department of Haematology, Nottingham University Hospital NHS Trust, Nottingham, UK
| | | |
Collapse
|
142
|
He R, Devine DJ, Tu ZJ, Mai M, Chen D, Nguyen PL, Oliveira JL, Hoyer JD, Reichard KK, Ollila PL, Al-Kali A, Tefferi A, Begna KH, Patnaik MM, Alkhateeb H, Viswanatha DS. Hybridization capture-based next generation sequencing reliably detects FLT3 mutations and classifies FLT3-internal tandem duplication allelic ratio in acute myeloid leukemia: a comparative study to standard fragment analysis. Mod Pathol 2020; 33:334-343. [PMID: 31471587 PMCID: PMC7051912 DOI: 10.1038/s41379-019-0359-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 01/12/2023]
Abstract
FLT3-internal tandem duplication occurs in 20-30% of acute myeloid leukemia and confers an adverse prognosis with its allelic ratio being a key risk stratifier. The US Food and Drug Administration recently approved FLT3 inhibitors midostaurin and gilteritinib in FLT3 mutation-positive acute myeloid leukemia. Historically, FLT3 was tested by fragment analysis, which has become the standard method endorsed by international guidelines. However, next generation sequencing is increasingly used at acute myeloid leukemia diagnosis given its ability to simultaneously evaluate multiple clinically informative markers. As FLT3-internal tandem duplication detection was known to be challenging by next generation sequencing and the results carry profound prognostic and therapeutic implications, it is important to thoroughly examine its performance in FLT3-internal tandem duplication detection and allelic ratio classification. In a comparative study with fragment analysis, we retrospectively reviewed our experience using a custom-designed, hybridization capture-based, targeted next generation sequencing panel. Among 7902 cases, FLT3-internal tandem duplication was detected in 335 with variable sizes (3-231 bp) and insertion sites. Fragment analysis was also performed in 402 cases, demonstrating 100% concordance in FLT3-internal tandem duplication detection. In 136 dual-tested, positive cases, 128/136 (94%) exhibited concordant high/low allelic ratio classifications. The remaining 6% showed borderline low allelic ratio by next generation sequencing. The two methods were concordant in FLT3-tyrosine kinase domain mutation detection at the hotspot D835/I836 targeted by fragment analysis. Furthermore, seven mutations which may benefit from FLT3 inhibitor therapy were detected by next generation sequencing, in regions not covered by fragment analysis. Our study demonstrates that using a hybridization capture-based chemistry and optimized bioinformatics pipeline, next generation sequencing can reliably detect FLT3-internal tandem duplication and classify its allelic ratio for acute myeloid leukemia risk stratification. Next generation sequencing also exhibits superior comprehensiveness in FLT3 mutation detection and may further improve personalized, targeted therapy in acute myeloid leukemia.
Collapse
Affiliation(s)
- Rong He
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | - Daniel J Devine
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Zheng Jin Tu
- Biomedical statistics and informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ming Mai
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dong Chen
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Phuong L Nguyen
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jennifer L Oliveira
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - James D Hoyer
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Kaaren K Reichard
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Paul L Ollila
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Aref Al-Kali
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Ayalew Tefferi
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Kebede H Begna
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mrinal M Patnaik
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Hassan Alkhateeb
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - David S Viswanatha
- Division of Hematopathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
143
|
Dillon R, Hills R, Freeman S, Potter N, Jovanovic J, Ivey A, Kanda AS, Runglall M, Foot N, Valganon M, Khwaja A, Cavenagh J, Smith M, Ommen HB, Overgaard UM, Dennis M, Knapper S, Kaur H, Taussig D, Mehta P, Raj K, Novitzky-Basso I, Nikolousis E, Danby R, Krishnamurthy P, Hill K, Finnegan D, Alimam S, Hurst E, Johnson P, Khan A, Salim R, Craddock C, Spearing R, Gilkes A, Gale R, Burnett A, Russell NH, Grimwade D. Molecular MRD status and outcome after transplantation in NPM1-mutated AML. Blood 2020; 135:680-688. [PMID: 31932839 PMCID: PMC7059484 DOI: 10.1182/blood.2019002959] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
Relapse remains the most common cause of treatment failure for patients with acute myeloid leukemia (AML) who undergo allogeneic stem cell transplantation (alloSCT), and carries a grave prognosis. Multiple studies have identified the presence of measurable residual disease (MRD) assessed by flow cytometry before alloSCT as a strong predictor of relapse, but it is not clear how these findings apply to patients who test positive in molecular MRD assays, which have far greater sensitivity. We analyzed pretransplant blood and bone marrow samples by reverse-transcription polymerase chain reaction in 107 patients with NPM1-mutant AML enrolled in the UK National Cancer Research Institute AML17 study. After a median follow-up of 4.9 years, patients with negative, low (<200 copies per 105ABL in the peripheral blood and <1000 copies in the bone marrow aspirate), and high levels of MRD had an estimated 2-year overall survival (2y-OS) of 83%, 63%, and 13%, respectively (P < .0001). Focusing on patients with low-level MRD before alloSCT, those with FLT3 internal tandem duplications(ITDs) had significantly poorer outcome (hazard ratio [HR], 6.14; P = .01). Combining these variables was highly prognostic, dividing patients into 2 groups with 2y-OS of 17% and 82% (HR, 13.2; P < .0001). T-depletion was associated with significantly reduced survival both in the entire cohort (2y-OS, 56% vs 96%; HR, 3.24; P = .0005) and in MRD-positive patients (2y-OS, 34% vs 100%; HR, 3.78; P = .003), but there was no significant effect of either conditioning regimen or donor source on outcome. Registered at ISRCTN (http://www.isrctn.com/ISRCTN55675535).
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Female
- Hematopoietic Stem Cell Transplantation/mortality
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/mortality
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/genetics
- Nuclear Proteins/genetics
- Nucleophosmin
- Recurrence
- Young Adult
Collapse
Affiliation(s)
- Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
- Department of Haematology, Guy's Hospital, London, United Kingdom
| | - Robert Hills
- Nuffield Department of Population Health, University of Oxford, United Kingdom
| | - Sylvie Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Nicola Potter
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
| | - Jelena Jovanovic
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Adam Ivey
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Anju Shankar Kanda
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Manohursingh Runglall
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| | - Nicola Foot
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
| | - Mikel Valganon
- Cancer Genetics Service, Viapath, Guy's Hospital, London, United Kingdom
| | - Asim Khwaja
- Department of Haematology, University College, London, United Kingdom
| | | | | | | | | | - Mike Dennis
- Christie Hospital, Manchester, United Kingdom
| | - Steven Knapper
- Department of Haematology, Cardiff University, Cardiff, United Kingdom
| | - Harpreet Kaur
- Royal Hallamshire Hospital, Sheffield, United Kingdom
| | | | - Priyanka Mehta
- Bristol Haematology and Oncology Centre, Bristol, United Kingdom
| | - Kavita Raj
- Department of Haematology, Guy's Hospital, London, United Kingdom
| | | | | | | | | | - Kate Hill
- University Hospital, Southampton, United Kingdom
| | | | - Samah Alimam
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Department of Haematology, Guy's Hospital, London, United Kingdom
| | - Erin Hurst
- Royal Victoria Infirmary, Newcastle, United Kingdom
| | | | - Anjum Khan
- St James' Hospital, Leeds, United Kingdom
| | - Rahuman Salim
- Clatterbridge Cancer Centre, Liverpool, United Kingdom
| | | | | | - Amanda Gilkes
- Department of Haematology, Cardiff University, Cardiff, United Kingdom
| | - Rosemary Gale
- Department of Haematology, University College, London, United Kingdom
| | - Alan Burnett
- Blackwaterfoot, Isle of Arran, United Kingdom; and
| | - Nigel H Russell
- Department of Haematology, Guy's Hospital, London, United Kingdom
- Nottingham University Hospital, Nottingham, United Kingdom
| | - David Grimwade
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Department of Haematology, Guy's Hospital, London, United Kingdom
| |
Collapse
|
144
|
Lam SS, Leung AY. Overcoming Resistance to FLT3 Inhibitors in the Treatment of FLT3-Mutated AML. Int J Mol Sci 2020; 21:E1537. [PMID: 32102366 PMCID: PMC7073218 DOI: 10.3390/ijms21041537] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/13/2023] Open
Abstract
Acute myeloid leukaemia (AML) carrying internal tandem duplication (ITD) of Fms-Like Tyrosine kinase 3 (FLT3) gene is associated with high risk of relapse and poor clinical outcome upon treatment with conventional chemotherapy. FLT3 inhibitors have been approved for the treatment of this AML subtype but leukaemia relapse remains to be a major cause of treatment failure. Mechanisms of drug resistance have been proposed, including evolution of resistant leukaemic clones; adaptive cellular mechanisms and a protective leukaemic microenvironment. These models have provided important leads that may inform design of clinical trials. Clinically, FLT3 inhibitors in combination with conventional chemotherapy as induction treatment for fit patients; with low-intensity treatment as salvage treatment or induction for unfit patients as well as maintenance treatment with FLT3 inhibitors post HSCT hold promise to improve survival in this AML subtype.
Collapse
Affiliation(s)
| | - Anskar Y.H. Leung
- Division of Haematology, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
145
|
Onecha E, Ruiz-Heredia Y, Martínez-Cuadrón D, Barragán E, Martinez-Sanchez P, Linares M, Rapado I, Perez-Oteyza J, Magro E, Herrera P, Rojas JL, Gorrochategui J, Villoria J, Boluda B, Sargas C, Ballesteros J, Montesinos P, Martínez-López J, Ayala R. Improving the prediction of acute myeloid leukaemia outcomes by complementing mutational profiling with ex vivo chemosensitivity. Br J Haematol 2020; 189:672-683. [PMID: 32068246 DOI: 10.1111/bjh.16432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
Abstract
Refractoriness to induction therapy and relapse after complete remission are the leading causes of death in patients with acute myeloid leukaemia (AML). This study focussed on the prediction of response to standard induction therapy and outcome of patients with AML using a combined strategy of mutational profiling by next-generation sequencing (NGS, n = 190) and ex vivo PharmaFlow testing (n = 74) for the 10 most widely used drugs for AML induction therapy, in a cohort of adult patients uniformly treated according to Spanish PETHEMA guidelines. We identified an adverse mutational profile (EZH2, KMT2A, U2AF1 and/or TP53 mutations) that carries a greater risk of death [hazard ratio (HR): 3·29, P < 0·0001]. A high correlation was found between the ex vivo PharmaFlow results and clinical induction response (69%). Clinical correlation analysis showed that the pattern of multiresistance revealed by ex vivo PharmaFlow identified patients with a high risk of death (HR: 2·58). Patients with mutation status also ran a high risk (HR 4·19), and the risk was increased further in patients with both adverse profiles (HR 4·82). We have developed a new score based on NGS and ex vivo drug testing for AML patients that improves upon current prognostic risk stratification and allows clinicians to tailor treatments to minimise drug resistance.
Collapse
Affiliation(s)
- Esther Onecha
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain
| | - Yanira Ruiz-Heredia
- Hematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,Vivia Biotech, Tres Cantos, Madrid, Spain
| | - David Martínez-Cuadrón
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Madrid, Spain
| | - Eva Barragán
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Madrid, Spain
| | - Pilar Martinez-Sanchez
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,Complutense University, Madrid, Spain
| | - María Linares
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,Complutense University, Madrid, Spain
| | - Inmaculada Rapado
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Jaime Perez-Oteyza
- Hematology Department, Hospital Universitario Sanchinarro, Madrid, Spain
| | - Elena Magro
- Hematology Department, Hospital Universitario Principe de Asturias, Madrid, Spain
| | - Pilar Herrera
- Hematology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | | | | | | | - Blanca Boluda
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Madrid, Spain
| | - Claudia Sargas
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Madrid, Spain
| | | | - Pau Montesinos
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Madrid, Spain
| | - Joaquín Martínez-López
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,Complutense University, Madrid, Spain.,CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Rosa Ayala
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Hematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain.,Complutense University, Madrid, Spain.,CIBERONC, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
146
|
Linch DC, Hills RK, Burnett AK, Gale RE. The clinical impact of mutant DNMT3A R882 variant allele frequency in acute myeloid leukaemia. Br J Haematol 2020; 189:e81-e86. [PMID: 32004382 DOI: 10.1111/bjh.16486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- David C Linch
- Department of Haematology, UCL Cancer Institute, London, UK
| | - Robert K Hills
- Nuffield Department of Population Health, Oxford University, Oxford, UK
| | - Alan K Burnett
- Department of Haematology, School of Medicine, Cardiff University, Cardiff, UK
| | | |
Collapse
|
147
|
Nucleophosmin mutations confer an independent favorable prognostic impact in 869 pediatric patients with acute myeloid leukemia. Blood Cancer J 2020; 10:1. [PMID: 31915364 PMCID: PMC6949268 DOI: 10.1038/s41408-019-0268-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/17/2019] [Accepted: 10/29/2019] [Indexed: 12/28/2022] Open
Abstract
Studies on the clinical significance of Nucleophosmin (NPM1) mutations in pediatric AML in a large cohort are lacking. Moreover, the prognosis of patients with co-occurring NPM1 and FLT3/ITD mutations is controversial. Here, we analyzed the impact of NPM1 mutations on prognoses of 869 pediatric AML patients from the TAGET dataset. The frequency of NPM1 mutations was 7.6%. NPM1 mutations were significantly associated with older age (P < 0.001), normal cytogenetics (P < 0.001), FLT3/ITD mutations (P < 0.001), and high complete remission induction rates (P < 0.05). Overall, NPM1-mutated patients had a significantly better 5-year EFS (P = 0.001) and OS (P = 0.016) compared to NPM1 wild-type patients, and this favorable impact was maintained even in the presence of FLT3/ITD mutations. Stem cell transplantation had no significant effect on the survival of patients with both NPM1 and FLT3/ITD mutations. Multivariate analysis revealed that NPM1 mutations were independent predictors of better outcome in terms of EFS (P = 0.004) and OS (P = 0.012). Our findings showed that NPM1 mutations confer an independent favorable prognostic impact in pediatric AML despite of FLT3/ITD mutations. In addition, pediatric AML patients with both NPM1 and FLT3/ITD mutations appear to have favorable prognoses and may not need hematopoietic stem cell transplantations.
Collapse
|
148
|
Acute Myeloid Neoplasms. Genomic Med 2020. [DOI: 10.1007/978-3-030-22922-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
149
|
Burnett AK, Hills RK, Russell N. Twenty five years of UK trials in acute myeloid leukaemia: what have we learned? Br J Haematol 2020; 188:86-100. [PMID: 31828788 DOI: 10.1111/bjh.16359] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Alan K Burnett
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, UK
| | - Robert K Hills
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Nigel Russell
- Department of Haematology, Centre for Clinical Haematology, Nottingham University Hospital (City Campus), Nottingham, UK
| |
Collapse
|
150
|
Alabdulwahab AS, Elsayed HG, Sherisher MA, Zeeneldin A, Elbjeirami WM. AML in Saudi Arabia: Analysis According to the European LeukaemiaNet 2017 Cytogenetic Classification. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 20:e212-e220. [PMID: 32127298 DOI: 10.1016/j.clml.2019.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 11/18/2022]
Abstract
INTRODUCTION To the best of our knowledge, few studies have addressed the prognosis of patients with acute myeloid leukemia (AML) in Saudi Arabia. The present study retrospectively analyzed the prognostic factors in patients with de novo AML at a single institution owing to the observation of some differences with the reported data from the Western world. PATIENTS AND METHODS Patients with de novo AML who had been referred to King Abdulla Medical City were included. All patients had undergone bone marrow aspiration, biopsy, flow cytometry, cytogenetics (conventional and fluorescence in situ hybridization panel performed at Mayo Clinic), molecular tests, and other routine tests. RESULTS The data from 170 patients were reviewed. Of the 170 patients, 26 had had acute promyelocytic leukemia, 16 with AML had received less intensive therapy, 119 had received intensive induction, and 8 had refused treatment. The present analysis was limited to the 119 patients who had received intensive induction therapy. For the major cytogenetic categories, 17 of 27 patients with core binding factor leukemia (62.9%) were reassigned to the intermediate (n = 10; 37%) or unfavorable (n = 7; 25.9%) risk group according to the FLT3-ITD and NPM results. Of the 50 cases of normal cytogenetic findings, 2 (4%) were considered unfavorable, 12 (24%), favorable, 30 intermediate (60%), and 6 (12%) unknown. The median leukemia-free survival was 21.5 months. The median overall survival was 16.4 ± 2.2 months, with a 3-year survival rate of 37.2%. Multivariate Cox regression analysis revealed that the cytogenetics results (P = .002) and the presence of FLT-3 (P = .03) were independent prognostic factors for relapse-free survival. Performance status, response, relapse, and cytogenetics findings were independent prognostic factors for survival. CONCLUSIONS The results from the present study revealed some differences in patient age and cytogenetic risk groups for patients with AML in our region and those in the Western world, including a younger median age, relevance of core binding factor leukemia, and a greater incidence of monosomies.
Collapse
Affiliation(s)
- Amal S Alabdulwahab
- Department of Hematology, King Abdullah Medical City, Holly Capital, Kingdom of Saudi Arabia
| | - Hussein G Elsayed
- Department of Hematology, King Abdullah Medical City, Holly Capital, Kingdom of Saudi Arabia.
| | - Mohamed A Sherisher
- Department of Hematology, King Abdullah Medical City, Holly Capital, Kingdom of Saudi Arabia; Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Ahmed Zeeneldin
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt; Department of Medical Oncology, King Abdullah Medical City, Holly Capital, Kingdom of Saudi Arabia
| | - Wafa M Elbjeirami
- Laboratory and Blood Bank Department, King Abdullah Medical City, Holly Capital, Kingdom of Saudi Arabia
| |
Collapse
|