101
|
Wang J, Pan X, Song Y, Liu J, Ma F, Wang P, Liu Y, Zhao L, Kang D, Hu L. Discovery of a Potent and Selective FLT3 Inhibitor ( Z)- N-(5-((5-Fluoro-2-oxoindolin-3-ylidene)methyl)-4-methyl-1 H-pyrrol-3-yl)-3-(pyrrolidin-1-yl)propanamide with Improved Drug-like Properties and Superior Efficacy in FLT3-ITD-Positive Acute Myeloid Leukemia. J Med Chem 2021; 64:4870-4890. [PMID: 33797247 DOI: 10.1021/acs.jmedchem.0c02247] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overcoming the FLT3-ITD mutant has been a promising drug design strategy for treating acute myeloid leukemia (AML). Herein, we discovered a novel FLT3 inhibitor 17, which displayed potent inhibitory activity against the FLT3-ITD mutant (IC50 = 0.8 nM) and achieved good selectivity over c-KIT kinase (over 500-fold). Compound 17 selectively inhibited the proliferation of FLT3-ITD-positive AML cell lines MV4-11 (IC50 = 23.5 nM) and MOLM-13 (IC50 = 35.5 nM) and exhibited potent inhibitory effects against associated acquired resistance mutations. In cellular mechanism studies, compound 17 strongly inhibited FLT3-mediated signaling pathways and induced apoptosis by arresting the cell cycle in the sub-G1 phase. In in vivo studies, compound 17 demonstrated a good bioavailability (73.6%) and significantly suppressed tumor growth in MV4-11 (10 mg/kg, TGI 93.4%) and MOLM-13 (20 mg/kg, TGI 98.0%) xenograft models without exhibiting obvious toxicity. These results suggested that compound 17 may be a promising drug candidate for treating FLT3-ITD-positive AML.
Collapse
Affiliation(s)
- Junwei Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Xiang Pan
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Yi Song
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Jian Liu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Fei Ma
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, P.R. China
| | - Ping Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Yan Liu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Lin Zhao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Di Kang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, P.R. China
| |
Collapse
|
102
|
Singh Mali R, Zhang Q, DeFilippis RA, Cavazos A, Kuruvilla VM, Raman J, Mody V, Choo EF, Dail M, Shah NP, Konopleva M, Sampath D, Lasater EA. Venetoclax combines synergistically with FLT3 inhibition to effectively target leukemic cells in FLT3-ITD+ acute myeloid leukemia models. Haematologica 2021; 106:1034-1046. [PMID: 32414851 PMCID: PMC8017817 DOI: 10.3324/haematol.2019.244020] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
FLT3 internal tandem duplication (FLT3-ITD) mutations account for ~25% of adult acute myeloid leukemia cases and are associated with poor prognosis. Venetoclax, a selective BCL-2 inhibitor, has limited monotherapy activity in relapsed/refractory acute myeloid leukemia with no responses observed in a small subset of FLT3-ITD+ patients. Further, FLT3-ITD mutations emerged at relapse following venetoclax monotherapy and combination therapy suggesting a potential mechanism of resistance. Therefore, we investigated the convergence of FLT3-ITD signaling on the BCL-2 family proteins and determined combination activity of venetoclax and FLT3-ITD inhibition in preclinical models. In vivo, venetoclax combined with quizartinib, a potent FLT3 inhibitor, showed greater anti-tumor efficacy and prolonged survival compared to monotherapies. In a patient-derived FLT3-ITD+ xenograft model, cotreatment with venetoclax and quizartinib at clinically relevant doses had greater anti-tumor activity in the tumor microenvironment compared to quizartinib or venetoclax alone. Use of selective BCL-2 family inhibitors further identified a role for BCL-2, BCL-XL and MCL-1 in mediating survival in FLT3-ITD+ cells in vivo and highlighted the need to target all three proteins for greatest anti-tumor activity. Assessment of these combinations in vitro revealed synergistic combination activity for quizartinib and venetoclax but not for quizartinib combined with BCL-XL or MCL-1 inhibition. FLT3-ITD inhibition was shown to indirectly target both BCL-XL and MCL-1 through modulation of protein expression, thereby priming cells toward BCL-2 dependence for survival. These data demonstrate that FLT3-ITD inhibition combined with venetoclax has impressive anti-tumor activity in FLT3-ITD+ acute myeloid leukemia preclinical models and provides strong mechanistic rational for clinical studies.
Collapse
Affiliation(s)
- Raghuveer Singh Mali
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Qi Zhang
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Rosa Anna DeFilippis
- Division of Hematology and Oncology, University of California at San Francisco, San Francisco, USA
| | - Antonio Cavazos
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Vinitha Mary Kuruvilla
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jayant Raman
- Division of Hematology and Oncology, University of California at San Francisco, San Francisco, USA
| | - Vidhi Mody
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Edna F Choo
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Monique Dail
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Neil P Shah
- Helen Diller Comprehensive Cancer Center, University of California at San Francisco, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Deepak Sampath
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Elisabeth A Lasater
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
103
|
Wang Z, Cai J, Cheng J, Yang W, Zhu Y, Li H, Lu T, Chen Y, Lu S. FLT3 Inhibitors in Acute Myeloid Leukemia: Challenges and Recent Developments in Overcoming Resistance. J Med Chem 2021; 64:2878-2900. [PMID: 33719439 DOI: 10.1021/acs.jmedchem.0c01851] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene are often present in newly diagnosed acute myeloid leukemia (AML) patients with an incidence rate of approximately 30%. Recently, many FLT3 inhibitors have been developed and exhibit positive preclinical and clinical effects against AML. However, patients develop resistance soon after undergoing FLT3 inhibitor treatment, resulting in short durable responses and poor clinical effects. This review will discuss the main mechanisms of resistance to clinical FLT3 inhibitors and summarize the emerging strategies that are utilized to overcome drug resistance. Basically, medicinal chemistry efforts to develop new small-molecule FLT3 inhibitors offer a direct solution to this problem. Other potential strategies include the combination of FLT3 inhibitors with other therapies and the development of multitarget inhibitors. It is hoped that this review will provide inspiring insights into the discovery of new AML therapies that can eventually overcome the resistance to current FLT3 inhibitors.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jiongheng Cai
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Wenqianzi Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Yifan Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
104
|
Abstract
Aberrant FLT3 receptor signaling is common in acute myeloid leukemia (AML) and has important implications for the biology and clinical management of the disease. Patients with FLT3-mutated AML frequently present with critical illness, are more likely to relapse after treatment, and have worse clinical outcomes than their FLT3 wild type counterparts. The clinical management of FLT3-mutated AML has been transformed by the development of FLT3 inhibitors, which are now in use in the frontline and relapsed/refractory settings. However, many questions regarding the optimal approach to the treatment of these patients remain. In this paper, we will review the rationale for targeting the FLT3 receptor in AML, the impact of FLT3 mutation on patient prognosis, the current standard of care approaches to FLT3-mutated AML management, and the diverse array of FLT3 inhibitors in use and under investigation. We will also explore new opportunities and strategies for targeting the FLT3 receptor. These include targeting the receptor in patients with non-canonical FLT3 mutations or wild type FLT3, pairing FLT3 inhibitors with other novel therapies, using minimal residual disease (MRD) testing to guide the targeting of FLT3, and novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alexander J Ambinder
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark Levis
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
105
|
Sorafenib or placebo in patients with newly diagnosed acute myeloid leukaemia: long-term follow-up of the randomized controlled SORAML trial. Leukemia 2021; 35:2517-2525. [PMID: 33603142 PMCID: PMC8410595 DOI: 10.1038/s41375-021-01148-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/11/2020] [Accepted: 01/21/2021] [Indexed: 01/07/2023]
Abstract
Early results of the randomized placebo-controlled SORAML trial showed that, in patients with newly diagnosed acute myeloid leukaemia (AML), sorafenib led to a significant improvement in event-free (EFS) and relapse-free survival (RFS). In order to describe second-line treatments and their implications on overall survival (OS), we performed a study after a median follow-up time of 78 months. Newly diagnosed fit AML patients aged ≤60 years received sorafenib (n = 134) or placebo (n = 133) in addition to standard chemotherapy and as maintenance treatment. The 5-year EFS was 41 versus 27% (HR 0.68; p = 0.011) and 5-year RFS was 53 versus 36% (HR 0.64; p = 0.035). Allogeneic stem cell transplantation (allo SCT) was performed in 88% of the relapsed patients. Four years after salvage allo SCT, the cumulative incidence of relapse was 54 versus 35%, and OS was 32 versus 50%. The 5-year OS from randomization in all study patients was 61 versus 53% (HR 0.82; p = 0.282). In conclusion, the addition of sorafenib to chemotherapy led to a significant prolongation of EFS and RFS. Although the OS benefit did not reach statistical significance, these results confirm the antileukaemic activity of sorafenib.
Collapse
|
106
|
K. Bhanumathy K, Balagopal A, Vizeacoumar FS, Vizeacoumar FJ, Freywald A, Giambra V. Protein Tyrosine Kinases: Their Roles and Their Targeting in Leukemia. Cancers (Basel) 2021; 13:cancers13020184. [PMID: 33430292 PMCID: PMC7825731 DOI: 10.3390/cancers13020184] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Protein phosphorylation is a key regulatory mechanism that controls a wide variety of cellular responses. This process is catalysed by the members of the protein kinase superfamily that are classified into two main families based on their ability to phosphorylate either tyrosine or serine and threonine residues in their substrates. Massive research efforts have been invested in dissecting the functions of tyrosine kinases, revealing their importance in the initiation and progression of human malignancies. Based on these investigations, numerous tyrosine kinase inhibitors have been included in clinical protocols and proved to be effective in targeted therapies for various haematological malignancies. In this review, we provide insights into the role of tyrosine kinases in leukaemia and discuss their targeting for therapeutic purposes with the currently available inhibitory compounds. Abstract Protein kinases constitute a large group of enzymes catalysing protein phosphorylation and controlling multiple signalling events. The human protein kinase superfamily consists of 518 members and represents a complicated system with intricate internal and external interactions. Protein kinases are classified into two main families based on the ability to phosphorylate either tyrosine or serine and threonine residues. Among the 90 tyrosine kinase genes, 58 are receptor types classified into 20 groups and 32 are of the nonreceptor types distributed into 10 groups. Tyrosine kinases execute their biological functions by controlling a variety of cellular responses, such as cell division, metabolism, migration, cell–cell and cell matrix adhesion, cell survival and apoptosis. Over the last 30 years, a major focus of research has been directed towards cancer-associated tyrosine kinases owing to their critical contributions to the development and aggressiveness of human malignancies through the pathological effects on cell behaviour. Leukaemia represents a heterogeneous group of haematological malignancies, characterised by an uncontrolled proliferation of undifferentiated hematopoietic cells or leukaemia blasts, mostly derived from bone marrow. They are usually classified as chronic or acute, depending on the rates of their progression, as well as myeloid or lymphoblastic, according to the type of blood cells involved. Overall, these malignancies are relatively common amongst both children and adults. In malignant haematopoiesis, multiple tyrosine kinases of both receptor and nonreceptor types, including AXL receptor tyrosine kinase (AXL), Discoidin domain receptor 1 (DDR1), Vascular endothelial growth factor receptor (VEGFR), Fibroblast growth factor receptor (FGFR), Mesenchymal–epithelial transition factor (MET), proto-oncogene c-Src (SRC), Spleen tyrosine kinase (SYK) and pro-oncogenic Abelson tyrosine-protein kinase 1 (ABL1) mutants, are implicated in the pathogenesis and drug resistance of practically all types of leukaemia. The role of ABL1 kinase mutants and their therapeutic inhibitors have been extensively analysed in scientific literature, and therefore, in this review, we provide insights into the impact and mechanism of action of other tyrosine kinases involved in the development and progression of human leukaemia and discuss the currently available and emerging treatment options based on targeting these molecules.
Collapse
Affiliation(s)
- Kalpana K. Bhanumathy
- Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (A.B.); (F.J.V.)
- Correspondence: (K.K.B.); (V.G.); Tel.: +1-(306)-716-7456 (K.K.B.); +39-0882-416574 (V.G.)
| | - Amrutha Balagopal
- Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (A.B.); (F.J.V.)
| | - Frederick S. Vizeacoumar
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (F.S.V.); (A.F.)
| | - Franco J. Vizeacoumar
- Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (A.B.); (F.J.V.)
- Cancer Research Department, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (F.S.V.); (A.F.)
| | - Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy
- Correspondence: (K.K.B.); (V.G.); Tel.: +1-(306)-716-7456 (K.K.B.); +39-0882-416574 (V.G.)
| |
Collapse
|
107
|
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
|
108
|
Damnernsawad A, Bottomly D, Kurtz SE, Eide CA, McWeeney SK, Tyner JW, Nechiporuk T. Genome-wide CRISPR screen identifies regulators of MAPK and MTOR pathways mediating sorafenib resistance in acute myeloid leukemia. Haematologica 2020; 107:77-85. [PMID: 33375770 PMCID: PMC8719098 DOI: 10.3324/haematol.2020.257964] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Indexed: 11/11/2022] Open
Abstract
Drug resistance impedes the long-term effect of targeted therapies in acute myeloid leukemia (AML), necessitating the identification of mechanisms underlying resistance. Approximately 25% of AML patients carry FLT3 mutations and develop post-treatment insensitivity to FLT3 inhibitors, including sorafenib. Using a genomewide CRISPR screen, we identified LZTR1, NF1, TSC1 and TSC2, negative regulators of the MAPK and MTOR pathways, as mediators of resistance to sorafenib. Analyses of ex vivo drug sensitivity assays in samples from patients with FLT3-ITD AML revealed that lower expression of LZTR1, NF1, and TSC2 correlated with sensitivity to sorafenib. Importantly, MAPK and/or MTOR complex 1 (MTORC1) activity was upregulated in AML cells made resistant to several FLT3 inhibitors, including crenolanib, quizartinib, and sorafenib. These cells were sensitive to MEK inhibitors, and the combination of FLT3 and MEK inhibitors showed enhanced efficacy, suggesting the effectiveness of such treatment in AML patients with FLT3 mutations and those with resistance to FLT3 inhibitors.
Collapse
Affiliation(s)
- Alisa Damnernsawad
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA; Department of Biology, Faculty of Science, Mahidol University, Bangkok
| | - Daniel Bottomly
- Division of Bioinformatics and Computational Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Stephen E Kurtz
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Christopher A Eide
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Shannon K McWeeney
- Division of Bioinformatics and Computational Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Jeffrey W Tyner
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA; Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR.
| | - Tamilla Nechiporuk
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR.
| |
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: 131] [Impact Index Per Article: 26.2] [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
|
Zhao X, Liu HQ, Wang LN, Yang L, Liu XL. Current and emerging molecular and epigenetic disease entities in acute myeloid leukemia and a critical assessment of their therapeutic modalities. Semin Cancer Biol 2020; 83:121-135. [PMID: 33242577 DOI: 10.1016/j.semcancer.2020.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
Acute myeloid leukemia (AML) is the most frequently diagnosed acute leukemia, and its incidence increases with age. Although the etiology of AML remains unknown, exposure to genotoxic agents or some prior hematologic disorders could lead to the development of this condition. The pathogenesis of AML involves the development of malignant transformation of hematopoietic stem cells that undergo successive genomic alterations, ultimately giving rise to a full-blown disease. From the disease biology perspective, AML is considered to be extremely complex with significant genetic, epigenetic, and phenotypic variations. Molecular and cytogenetic alterations in AML include mutations in those subsets of genes that are involved in normal cell proliferation, maturation and survival, thus posing significant challenge to targeting these pathways without attendant toxicity. In addition, multiple malignant cells co-exist in the majority of AML patients. Individual subclones are characterized by unique genetic and epigenetic abnormalities, which contribute to the differences in their response to treatment. As a result, despite a dramatic progress in our understanding of the pathobiology of AML, not much has changed in therapeutic approaches to treat AML in the past four decades. Dose and regimen modifications with improved supportive care have contributed to improved outcomes by reducing toxicity-related side effects. Several drug candidates are currently being developed, including targeted small-molecule inhibitors, cytotoxic chemotherapies, monoclonal antibodies and epigenetic drugs. This review summarizes the current state of affairs in the pathobiological and therapeutic aspects of AML.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Paediatrics, The First Hospital of Jilin University, Changchun, China
| | - Huan-Qiu Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Li-Na Wang
- Department of Paediatrics, The First Hospital of Jilin University, Changchun, China
| | - Le Yang
- Department of Endocrinology, The People's Hospital of Jilin Province, Changchun, China.
| | - Xiao-Liang Liu
- Department of Hematology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
112
|
Marensi V, Keeshan KR, MacEwan DJ. Pharmacological impact of FLT3 mutations on receptor activity and responsiveness to tyrosine kinase inhibitors. Biochem Pharmacol 2020; 183:114348. [PMID: 33242449 DOI: 10.1016/j.bcp.2020.114348] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023]
Abstract
Acute myelogenous leukaemia (AML) is an aggressive blood cancer characterized by the rapid proliferation of immature myeloid blast cells, resulting in a high mortality rate. The 5-year overall survival rate for AML patients is approximately 25%. Circa 35% of all patients carry a mutation in the FLT3 gene which have a poor prognosis. Targeting FLT3 receptor tyrosine kinase has become a treatment strategy in AML patients possessing FLT3 mutations. The most common mutations are internal tandem duplications (ITD) within exon 14 and a single nucleotide polymorphism (SNP) that leads to a point mutation in the D835 of the tyrosine kinase domain (TKD). Variations in the ITD sequence and the occurrence of other point mutations that lead to ligand-independent FLT3 receptor activation create difficulties in developing personalized therapeutic strategies to overcome observed mutation-driven drug resistance. Midostaurin and quizartinib are tyrosine kinase inhibitors (TKIs) with inhibitory efficacy against FLT3-ITD, but exhibit limited clinical impact. In this review, we focus on the structural aspects of the FLT3 receptor and correlate those mutations with receptor activation and the consequences for molecular and clinical responsiveness towards therapies targeting FLT3-ITD positive AML.
Collapse
Affiliation(s)
- Vanessa Marensi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Karen R Keeshan
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David J MacEwan
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
113
|
Molecular Mechanisms of Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia: Ongoing Challenges and Future Treatments. Cells 2020; 9:cells9112493. [PMID: 33212779 PMCID: PMC7697863 DOI: 10.3390/cells9112493] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/07/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022] Open
Abstract
Treatment of FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD)-positive acute myeloid leukemia (AML) remains a challenge despite the development of novel FLT3-directed tyrosine kinase inhibitors (TKI); the relapse rate is still high even after allogeneic stem cell transplantation. In the era of next-generation FLT3-inhibitors, such as midostaurin and gilteritinib, we still observe primary and secondary resistance to TKI both in monotherapy and in combination with chemotherapy. Moreover, remissions are frequently short-lived even in the presence of continuous treatment with next-generation FLT3 inhibitors. In this comprehensive review, we focus on molecular mechanisms underlying the development of resistance to relevant FLT3 inhibitors and elucidate how this knowledge might help to develop new concepts for improving the response to FLT3-inhibitors and reducing the development of resistance in AML. Tailored treatment approaches that address additional molecular targets beyond FLT3 could overcome resistance and facilitate molecular responses in AML.
Collapse
|
114
|
Stanchina M, Soong D, Zheng-Lin B, Watts JM, Taylor J. Advances in Acute Myeloid Leukemia: Recently Approved Therapies and Drugs in Development. Cancers (Basel) 2020; 12:E3225. [PMID: 33139625 PMCID: PMC7692236 DOI: 10.3390/cancers12113225] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous malignancy comprised of various cytogenetic and molecular abnormalities that has notoriously been difficult to treat with an overall poor prognosis. For decades, treatment options were limited to either intensive chemotherapy with anthracycline and cytarabine-based regimens (7 + 3) or lower intensity regimens including hypomethylating agents or low dose cytarabine, followed by either allogeneic stem cell transplant or consolidation chemotherapy. Fortunately, with the influx of rapidly evolving molecular technologies and new genetic understanding, the treatment landscape for AML has dramatically changed. Advances in the formulation and delivery of 7 + 3 with liposomal cytarabine and daunorubicin (Vyxeos) have improved overall survival in secondary AML. Increased understanding of the genetic underpinnings of AML has led to targeting actionable mutations such as FLT3, IDH1/2 and TP53, and BCL2 or hedgehog pathways in more frail populations. Antibody drug conjugates have resurfaced in the AML landscape and there have been numerous advances utilizing immunotherapies including immune checkpoint inhibitors, antibody-drug conjugates, bispecific T cell engager antibodies, chimeric antigen receptor (CAR)-T therapy and the development of AML vaccines. While there are dozens of ongoing studies and new drugs in the pipeline, this paper serves as a review of the advances achieved in the treatment of AML in the last several years and the most promising future avenues of advancement.
Collapse
Affiliation(s)
- Michele Stanchina
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (D.S.)
| | - Deborah Soong
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (D.S.)
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Binbin Zheng-Lin
- Department of Medicine, Icahn School of Medicine Mount Sinai West-Morningside, New York, NY 10025, USA;
| | - Justin M. Watts
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Justin Taylor
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| |
Collapse
|
115
|
Malik HS, Bilal A, Ullah R, Iqbal M, Khan S, Ahmed I, Krohn K, Saleem RSZ, Hussain H, Faisal A. Natural and Semisynthetic Chalcones as Dual FLT3 and Microtubule Polymerization Inhibitors. JOURNAL OF NATURAL PRODUCTS 2020; 83:3111-3121. [PMID: 32975953 DOI: 10.1021/acs.jnatprod.0c00699] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Activating mutations in FLT3 receptor tyrosine kinase are found in a third of acute myeloid leukemia (AML) patients and are associated with disease relapse and a poor prognosis. The majority of these mutations are internal tandem duplications (ITDs) in the juxtamembrane domain of FLT3, which have been validated as a therapeutic target. The clinical success of selective inhibitors targeting oncogenic FLT3, however, has been limited due to the acquisition of drug resistance. Herein the identification of a dual FLT3/microtubule polymerization inhibitor, chalcone 4 (2'-allyloxy-4,4'-dimethoxychalcone), is reported through screening of 15 related chalcones for differential antiproliferative activity in leukemia cell lines dependent on FLT3-ITD (MV-4-11) or BCR-ABL (K562) oncogenes and by subsequent screening for mitotic inducers in the HCT116 cell line. Three natural chalcones (1-3) were found to be differentially more potent toward the MV-4-11 (FLT3-ITD) cell line compared to the K562 (BCR-ABL) cell line. Notably, the new semisynthetic chalcone 4, which is a 2'-O-allyl analogue of the natural chalcone 3, was found to be more potent toward the FLT3-ITD+ cell line and inhibited FLT3 signaling in FLT3-dependent cells. An in vitro kinase assay confirmed that chalcone 4 directly inhibited FLT3. Moreover, chalcone 4 induced mitotic arrest in these cells and inhibited tubulin polymerization in both cellular and biochemical assays. Treatment of MV-4-11 cells with this inhibitor for 24 and 48 h resulted in apoptotic cell death. Finally, chalcone 4 was able to overcome TKD mutation-mediated acquired resistance to FLT3 inhibitors in a MOLM-13 cell line expressing FLT3-ITD with the D835Y mutation. Chalcone 4 is, therefore, a promising lead for the discovery of dual-target FLT3 inhibitors.
Collapse
Affiliation(s)
- Haleema Sadia Malik
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Aishah Bilal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rahim Ullah
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Maheen Iqbal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Sardraz Khan
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Ishtiaq Ahmed
- Department of Chemistry, University of Paderborn, Warburger Straße 100, 33098 Paderborn, Germany
| | - Karsten Krohn
- Department of Chemistry, University of Paderborn, Warburger Straße 100, 33098 Paderborn, Germany
| | - Rahman Shah Zaib Saleem
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Hidayat Hussain
- Department of Chemistry, University of Paderborn, Warburger Straße 100, 33098 Paderborn, Germany
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120 Halle (Saale), Germany
| | - Amir Faisal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
116
|
Novel Approaches to Target Mutant FLT3 Leukaemia. Cancers (Basel) 2020; 12:cancers12102806. [PMID: 33003568 PMCID: PMC7600363 DOI: 10.3390/cancers12102806] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is a haematologic disease in which oncogenic mutations in the receptor tyrosine kinase FLT3 frequently lead to leukaemic development. Potent treatment of AML patients is still hampered by inefficient targeting of leukemic stem cells expressing constitutive active FLT3 mutants. This review summarizes the current knowledge about the regulation of FLT3 activity at cellular level and discusses therapeutical options to affect the tumor cells and the microenvironment to impair the haematological aberrations. Abstract Fms-like tyrosine kinase 3 (FLT3) is a member of the class III receptor tyrosine kinases (RTK) and is involved in cell survival, proliferation, and differentiation of haematopoietic progenitors of lymphoid and myeloid lineages. Oncogenic mutations in the FLT3 gene resulting in constitutively active FLT3 variants are frequently found in acute myeloid leukaemia (AML) patients and correlate with patient’s poor survival. Targeting FLT3 mutant leukaemic stem cells (LSC) is a key to efficient treatment of patients with relapsed/refractory AML. It is therefore essential to understand how LSC escape current therapies in order to develop novel therapeutic strategies. Here, we summarize the current knowledge on mechanisms of FLT3 activity regulation and its cellular consequences. Furthermore, we discuss how aberrant FLT3 signalling cooperates with other oncogenic lesions and the microenvironment to drive haematopoietic malignancies and how this can be harnessed for therapeutical purposes.
Collapse
|
117
|
Mosquera Orgueira A, Bao Pérez L, Mosquera Torre A, Peleteiro Raíndo A, Cid López M, Díaz Arias JÁ, Ferreiro Ferro R, Antelo Rodríguez B, González Pérez MS, Albors Ferreiro M, Alonso Vence N, Pérez Encinas MM, Bello López JL, Martinelli G, Cerchione C. FLT3 inhibitors in the treatment of acute myeloid leukemia: current status and future perspectives. Minerva Med 2020; 111:427-442. [PMID: 32955823 DOI: 10.23736/s0026-4806.20.06989-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene arise in 25-30% of all acute myeloid leukemia (AML) patients. These mutations lead to constitutive activation of the protein product and are divided in two broad types: internal tandem duplication (ITD) of the juxtamembrane domain (25% of cases) and point mutations in the tyrosine kinase domain (TKD). Patients with FLT3 ITD mutations have a high relapse risk and inferior cure rates, whereas the role of FLT3 TKD mutations still remains to be clarified. Additionally, growing research indicates that FLT3 status evolves through a disease continuum (clonal evolution), where AML cases can acquire FLT3 mutations at relapse - not present in the moment of diagnosis. Several FLT3 inhibitors have been tested in patients with FLT3-mutated AML. These drugs exhibit different kinase inhibitory profiles, pharmacokinetics and adverse events. First-generation multi-kinase inhibitors (sorafenib, midostaurin, lestaurtinib) are characterized by a broad-spectrum of drug targets, whereas second-generation inhibitors (quizartinib, crenolanib, gilteritinib) show more potent and specific FLT3 inhibition, and are thereby accompanied by less toxic effects. Notwithstanding, all FLT3 inhibitors face primary and acquired mechanisms of resistance, and therefore the combinations with other drugs (standard chemotherapy, hypomethylating agents, checkpoint inhibitors) and its application in different clinical settings (upfront therapy, maintenance, relapsed or refractory disease) are under study in a myriad of clinical trials. This review focuses on the role of FLT3 mutations in AML, pharmacological features of FLT3 inhibitors, known mechanisms of drug resistance and accumulated evidence for the use of FLT3 inhibitors in different clinical settings.
Collapse
Affiliation(s)
- Adrián Mosquera Orgueira
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain - .,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain - .,University of Santiago de Compostela, Santiago de Compostela, Spain -
| | - Laura Bao Pérez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain
| | - Alicia Mosquera Torre
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain
| | - Andrés Peleteiro Raíndo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain.,University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Cid López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain.,University of Santiago de Compostela, Santiago de Compostela, Spain
| | - José Á Díaz Arias
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain
| | - Roi Ferreiro Ferro
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Beatriz Antelo Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain.,University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marta S González Pérez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain
| | - Manuel Albors Ferreiro
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain
| | - Natalia Alonso Vence
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain
| | - Manuel M Pérez Encinas
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain.,University of Santiago de Compostela, Santiago de Compostela, Spain
| | - José L Bello López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS - SERGAS), Santiago de Compostela, Spain.,University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Giovanni Martinelli
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Claudio Cerchione
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| |
Collapse
|
118
|
Molina Garay C, Carrillo Sánchez K, Flores Lagunes LL, Jiménez Olivares M, Muñoz Rivas A, Villegas Torres BE, Flores Aguilar H, Núñez Enríquez JC, Jiménez Hernández E, Bekker Méndez VC, Torres Nava JR, Flores Lujano J, Martín Trejo JA, Mata Rocha M, Medina Sansón A, Espinoza Hernández LE, Peñaloza Gonzalez JG, Espinosa Elizondo RM, Flores Villegas LV, Amador Sanchez R, Pérez Saldívar ML, Sepúlveda Robles OA, Rosas Vargas H, Rangel López A, Domínguez López ML, García Latorre EA, Reyes Maldonado E, Galindo Delgado P, Mejía Aranguré JM, Alaez Verson C. Profiling FLT3 Mutations in Mexican Acute Myeloid Leukemia Pediatric Patients: Impact on Overall Survival. Front Pediatr 2020; 8:586. [PMID: 33042924 PMCID: PMC7525023 DOI: 10.3389/fped.2020.00586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/07/2020] [Indexed: 01/09/2023] Open
Abstract
Background: Acute myeloid leukemia (AML) is the second most frequent leukemia in childhood. The FLT3 gene participates in hematopoietic stem cell proliferation. FLT3 mutations are recurrent in AML and influence prognosis. In Mexican pediatric AML patients, FLT3 mutational profile, and their clinical impact have not been evaluated. Aim of the study: This study aimed to identify the profile of FLT3 mutations in pediatric patients with de novo AML and to assess their possible influence on overall survival (OS) and other clinical features. Methods: Massive parallel target sequencing of FLT3 was performed in 80 patients. Results: FLT3 mutations [internal tandem duplication (ITD) or tyrosine kinase domain (TKD)] were identified in 24% of them. OS was significantly lower in FLT3 POS cases than in FLT3 NEG (p = 0.03). The average OS for FLT3 POS was 1.2 vs. 2.2 years in FLT3 NEG. There were no significant differences in the children's sex, age, percentage of blasts in bone marrow aspirate, or white blood cell count in peripheral blood at diagnosis between both groups. No differences were identified stratifying by the mutational load (high > 0.4) or type of mutation. The negative effect of FLT3 mutations was also observed in patients with acute promyelocytic leukemia (APL). Conclusions: FLT3 mutational profile is described in Mexican pediatric AML patients for the first time. Mutated FLT3 negatively impacts the outcome of AML patients, even considering the APL group. The clinical benefit from treatment with tyrosine kinase inhibitors in the FLT3 POS pediatric patients needs to be assessed in clinical trials. FLT3 testing may contribute to better risk stratification in our pediatric AML patients.
Collapse
Affiliation(s)
- Carolina Molina Garay
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (Inmegen), Mexico City, Mexico
| | - Karol Carrillo Sánchez
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (Inmegen), Mexico City, Mexico
| | | | - Marco Jiménez Olivares
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (Inmegen), Mexico City, Mexico
| | - Anallely Muñoz Rivas
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (Inmegen), Mexico City, Mexico
| | | | | | - Juan Carlos Núñez Enríquez
- Unidad de Investigación Médica en Epidemiología Clínica, UMAE Hospital de Pediatría, Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Elva Jiménez Hernández
- Servicio de Hematología Pediátrica, Hospital General “Gaudencio González Garza”, Centro Médico Nacional (CMN) “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Vilma Carolina Bekker Méndez
- Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología “Dr. Daniel Méndez Hernández”, “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - José Refugio Torres Nava
- Servicio de Oncología, Hospital Pediátrico de Moctezuma, Secretaria de Salud del D.F., Mexico City, Mexico
| | - Janet Flores Lujano
- Unidad de Investigación Médica en Epidemiología Clínica, UMAE Hospital de Pediatría, Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Jorge Alfonso Martín Trejo
- Servicio de Hematología Pediátrica, UMAE Hospital de Pediatría, Centro Médico Nacional (CMN) “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Minerva Mata Rocha
- Unidad de Investigación Médica en Epidemiología Clínica, UMAE Hospital de Pediatría, Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Aurora Medina Sansón
- Servicio de Hemato-Oncología, Hospital Infantil de México Federico Gómez, Secretaria de Salud (SSa), Mexico City, Mexico
| | - Laura Eugenia Espinoza Hernández
- Servicio de Hematología Pediátrica, Hospital General “Gaudencio González Garza”, Centro Médico Nacional (CMN) “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | | | - Luz Victoria Flores Villegas
- Servicio de Hematología Pediátrica, Centro Médico Nacional (CMN) “20 de Noviembre”, Instituto de Seguridad Social al Servicio de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - Raquel Amador Sanchez
- Hospital General Regional No. 1 “Carlos McGregor Sánchez Navarro”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Maria Luisa Pérez Saldívar
- Unidad de Investigación Médica en Epidemiología Clínica, UMAE Hospital de Pediatría, Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Omar Alejandro Sepúlveda Robles
- Unidad de Investigación Médica en Genética Humana, Unidad Médica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Haydeé Rosas Vargas
- Unidad de Investigación Médica en Genética Humana, Unidad Médica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Angélica Rangel López
- Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | | | - Elba Reyes Maldonado
- Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | | | - Juan Manuel Mejía Aranguré
- Unidad de Investigación Médica en Epidemiología Clínica, UMAE Hospital de Pediatría, Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
- Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Carmen Alaez Verson
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (Inmegen), Mexico City, Mexico
| |
Collapse
|
119
|
Zhou X, Yang C, Zou Z, Lang X, Chen P, Chen S, Wang K, Chen Y, Hao Y, Chen N, Ding J, Li Y, Shen Y, Xiao S. ZBTB44-FLT3 fusion in a patient with a myeloproliferative neoplasm. Br J Haematol 2020; 191:297-301. [PMID: 32866288 DOI: 10.1111/bjh.16995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/02/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Xin Zhou
- Department of Hematology, Wu Xi People's Hospital, Wuxi, China
| | | | - Zhijian Zou
- Department of Hematology, Wu Xi People's Hospital, Wuxi, China
| | | | - Ping Chen
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Si Chen
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Kai Wang
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Yongyan Chen
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Yang Hao
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Nan Chen
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Jingwen Ding
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Yan Li
- Suzhou Ninth People's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yufeng Shen
- Department of Hematology, Wu Xi People's Hospital, Wuxi, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
120
|
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
|
121
|
Elmeliegy M, Den Haese J, Talati C, Wetzler M, Jusko WJ. Towards better combination regimens of cytarabine and FLT3 inhibitors in acute myeloid leukemia. Cancer Chemother Pharmacol 2020; 86:325-337. [PMID: 32748108 DOI: 10.1007/s00280-020-04114-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AML patients with FLT3/ITD mutations have poor response to cytarabine-based chemotherapy. FLT3 inhibitors (FLT3i) may resensitize cells to cytarabine (CYT). Improving treatment outcome of this combination may benefit from a mechanistic extrapolation approach from in vitro data. METHODS The effects of CYT and several FLT3i on cell proliferation and cell cycle kinetics were examined in AML cell lines. The effect of FLT3i (quizartinib, midostaurin, sorafenib) on cell proliferation and cell cycle kinetics was assessed in AML cell lines with differing FLT3 status; HEL (negligible expression of wild-type FLT3), EOL1 (wild-type FLT3), MV4-11 (FLT3-ITD resulting in constitutively active isoform). Semi-mechanistic cell cycle models for CYT and FLT3i were developed. Clinical CYT and quizartinib pharmacokinetic dosage regimens were modeled. Survival of AML patients was described via a hazard model. Simulations exploring different CYT/quizartinib regimens were conducted with the goal of improving treatment outcome. RESULTS FLT3 status was associated with sensitivity to CYT (HEL cells most sensitive > EOL1 > MV4-11 cells). This order of sensitivity is reversed for FLT3i. Cytarabine induced apoptosis in the S-phase while all FLT3i induced apoptosis and cell cycle arrest at G1 phase. Simulations of candidate clinical regimens predict better cell kill upon adding quizartinib simultaneously with or immediately after CYT exposure. Overall survival was predicted to be significantly better with quizartinib 200 mg administered every 48 h vs every 24 h in patients with FLT3 aberrations. CONCLUSION Simultaneous administration of quizartinib and CYT every other day is a promising combination regimen for AML patients with FLT3 mutations.
Collapse
Affiliation(s)
- Mohamed Elmeliegy
- Pfizer, Inc., 10555 Science Center Dr., San Diego, CA, 92121, USA. .,Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| | - Jason Den Haese
- Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA.,Department of Biology and Mathematics, D'Youville College, Buffalo, NY, USA
| | - Chetasi Talati
- Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA.,Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Meir Wetzler
- Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| |
Collapse
|
122
|
Eguchi M, Minami Y, Kuzume A, Chi S. Mechanisms Underlying Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia. Biomedicines 2020; 8:biomedicines8080245. [PMID: 32722298 PMCID: PMC7459983 DOI: 10.3390/biomedicines8080245] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 01/03/2023] Open
Abstract
FLT3-ITD and FLT3-TKD mutations were observed in approximately 20 and 10% of acute myeloid leukemia (AML) cases, respectively. FLT3 inhibitors such as midostaurin, gilteritinib and quizartinib show excellent response rates in patients with FLT3-mutated AML, but its duration of response may not be sufficient yet. The majority of cases gain secondary resistance either by on-target and off-target abnormalities. On-target mutations (i.e., FLT3-TKD) such as D835Y keep the TK domain in its active form, abrogating pharmacodynamics of type II FLT3 inhibitors (e.g., midostaurin and quizartinib). Second generation type I inhibitors such as gilteritinib are consistently active against FLT3-TKD as well as FLT3-ITD. However, a “gatekeeper” mutation F691L shows universal resistance to all currently available FLT3 inhibitors. Off-target abnormalities are consisted with a variety of somatic mutations such as NRAS, AXL and PIM1 that bypass or reinforce FLT3 signaling. Off-target mutations can occur just in the primary FLT3-mutated clone or be gained by the evolution of other clones. A small number of cases show primary resistance by an FL-dependent, FGF2-dependent, and stromal CYP3A4-mediated manner. To overcome these mechanisms, the development of novel agents such as covalently-coupling FLT3 inhibitor FF-10101 and the investigation of combination therapy with different class agents are now ongoing. Along with novel agents, gene sequencing may improve clinical approaches by detecting additional targetable mutations and determining individual patterns of clonal evolution.
Collapse
Affiliation(s)
- Motoki Eguchi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
- Correspondence: ; Tel.: +81-4-7133-1111; Fax: +81-7133-6502
| | - Ayumi Kuzume
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa 296-8602, Japan
| | - SungGi Chi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (M.E.); (A.K.); (S.C.)
| |
Collapse
|
123
|
Zhong Y, Qiu RZ, Sun SL, Zhao C, Fan TY, Chen M, Li NG, Shi ZH. Small-Molecule Fms-like Tyrosine Kinase 3 Inhibitors: An Attractive and Efficient Method for the Treatment of Acute Myeloid Leukemia. J Med Chem 2020; 63:12403-12428. [PMID: 32659083 DOI: 10.1021/acs.jmedchem.0c00696] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fms-like tyrosine kinase 3 (FLT3) is an important member of the class III receptor tyrosine kinase (RTK) family, which is involved in the proliferation of hematopoietic cells and lymphocytes. In recent years, increasing evidence have demonstrated that the activation and mutation of FLT3 is closely implicated in the occurrence and development of acute myeloid leukemia (AML). The exploration of small-molecule inhibitors targeting FLT3 has aroused wide interest of pharmaceutical chemists and is expected to bring new hope for AML therapy. In this review, we specifically highlighted FLT3 mediated JAK/STAT, RAS/MAPK, and PI3K/AKT/mTOR signaling. The structural properties and biological activities of representative FLT3 inhibitors reported from 2014 to the present were also summarized. In addition, the major challenges in the current advance of novel FLT3 inhibitors were further analyzed, with the aim to guide future drug discovery.
Collapse
Affiliation(s)
- Yue Zhong
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Run-Ze Qiu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chao Zhao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tian-Yuan Fan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Min Chen
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhi-Hao Shi
- Department of Organic Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
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: 46] [Impact Index Per Article: 9.2] [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
|
Schmidt-Arras D, Böhmer FD. Mislocalisation of Activated Receptor Tyrosine Kinases - Challenges for Cancer Therapy. Trends Mol Med 2020; 26:833-847. [PMID: 32593582 DOI: 10.1016/j.molmed.2020.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/20/2022]
Abstract
Activating mutations in genes encoding receptor tyrosine kinases (RTKs) mediate proliferation, cell migration, and cell survival, and are therefore important drivers of oncogenesis. Numerous targeted cancer therapies are directed against activated RTKs, including small compound inhibitors, and immunotherapies. It has recently been discovered that not only certain RTK fusion proteins, but also many full-length RTKs harbouring activating mutations, notably RTKs of the class III family, are to a large extent mislocalised in intracellular membranes. Active kinases in these locations cause aberrant activation of signalling pathways. Moreover, low levels of activated RTKs at the cell surface present an obstacle for immunotherapy. We outline here why understanding of the mechanisms underlying mislocalisation will help in improving existing and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Dirk Schmidt-Arras
- Christian-Albrechts-University Kiel, Institute of Biochemistry, 24118 Kiel, Germany.
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| |
Collapse
|
126
|
Hu X, Cai J, Zhu J, Lang W, Zhong J, Zhong H, Chen F. Arsenic trioxide potentiates Gilteritinib-induced apoptosis in FLT3-ITD positive leukemic cells via IRE1a-JNK-mediated endoplasmic reticulum stress. Cancer Cell Int 2020; 20:250. [PMID: 32565734 PMCID: PMC7298957 DOI: 10.1186/s12935-020-01341-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Acute myeloid leukemia (AML) patients with FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) have a high relapse rate and poor prognosis. This study aims to explore the underlying mechanism of combining Gilteritinib with ATO at low concentration in the treatment of FLT3-ITD positive leukemias. Methods We used both in vitro and in vivo studies to investigate the effects of combination of Gilteritinib with ATO at low concentration on FLT3-ITD positive leukemias, together with the underlying molecular mechanisms of these processes. Results Combination of Gilteritinib with ATO showed synergistic effects on inhibiting proliferation, increasing apoptosis and attenuating invasive ability in FLT3-ITD-mutated cells and reducing tumor growth in nude mice. Results of western blot indicated that Gilteritinib increased a 160KD form of FLT3 protein on the surface of cell membrane. Detection of endoplasmic reticulum stress marker protein revealed that IRE1a and its downstream signal phosphorylated JNK were suppressed in Gilteritinib-treated FLT3-ITD positive cells. The downregulation of IRE1a induced by Gilteritinib was reversed with addition of ATO. Knockdown of IRE1a diminished the combinatorial effects of Gilteritinib plus ATO treatment and combination of tunicamycin (an endoplasmic reticulum pathway activator) with Gilteritinib achieved the similar effect as treatment with Gilteritinib plus ATO. Conclusions Thus, ATO at low concentration potentiates Gilteritinib-induced apoptosis in FLT3-ITD positive leukemic cells via IRE1a-JNK signal pathway, targeting IRE1a to cooperate with Gilteritinib may serve as a new theoretical basis on FLT3-ITD mutant AML treatment.
Collapse
Affiliation(s)
- Xiaoli Hu
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Jiayi Cai
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Jianyi Zhu
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Wenjing Lang
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Jihua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Hua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Fangyuan Chen
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| |
Collapse
|
127
|
Stemler J, Koehler P, Maurer C, Müller C, Cornely OA. Antifungal prophylaxis and novel drugs in acute myeloid leukemia: the midostaurin and posaconazole dilemma. Ann Hematol 2020; 99:1429-1440. [PMID: 32514626 PMCID: PMC7316674 DOI: 10.1007/s00277-020-04107-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022]
Abstract
With the advent of new targeted drugs in hematology and oncology patient prognosis is improved. Combination with antifungal prophylaxis challenges clinicians due to pharmacological profiles prone to drug–drug interactions (DDI). Midostaurin is a novel agent for FLT3-TKD/-ITDmut-acute myeloid leukemia (AML) and metabolized via cytochrome P450 3A4 (CYP3A4). Posaconazole is a standard of care antifungal agent used for prophylaxis during induction treatment of AML and a strong CYP3A4 inhibitor. Concomitant administration of both drugs leads to elevated midostaurin exposure. Both drugs improve overall survival at low numbers needed to treat. The impact of CYP3A4-related DDI remains to be determined. Severe adverse events have been observed; however, it remains unclear if they can be directly linked to DDI. The lack of prospective clinical studies assessing incidence of invasive fungal infections and clinical impact of DDI contributes to neglecting live-saving antifungal prophylaxis. Management strategies to combine both drugs have been proposed, but evidence on which approach to use is scarce. In this review, we discuss several approaches in the specific clinical setting of concomitant administration of midostaurin and posaconazole and give examples from everyday clinical practice. Therapeutic drug monitoring will become increasingly important to individualize and personalize antineoplastic concomitant and antifungal treatment in the context of DDI. Pharmaceutical companies addressing the issue in clinical trials may take a pioneer role in this field. Other recently developed and approved drugs for the treatment of AML likely inhere potential of DDI marking a foreseeable issue in future treatment of this life-threatening disease.
Collapse
Affiliation(s)
- Jannik Stemler
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Excellence Center for Medical Mycology (ECMM), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - Philipp Koehler
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Excellence Center for Medical Mycology (ECMM), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Christian Maurer
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Excellence Center for Medical Mycology (ECMM), University of Cologne, Cologne, Germany
| | - Carsten Müller
- Centre of Pharmacology, Therapeutic Drug Monitoring, Faculty of Medicine, Cologne, University Hospital of Cologne, Cologne, Germany
| | - Oliver A Cornely
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Excellence Center for Medical Mycology (ECMM), University of Cologne, Cologne, Germany. .,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany. .,German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany. .,Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany.
| |
Collapse
|
128
|
Chew S, Mackey MC, Jabbour E. Gilteritinib in the treatment of relapsed and refractory acute myeloid leukemia with a FLT3 mutation. Ther Adv Hematol 2020; 11:2040620720930614. [PMID: 32547718 PMCID: PMC7271272 DOI: 10.1177/2040620720930614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is a malignancy of uncontrolled proliferation of immature myeloid blasts characterized by clonal evolution and genetic heterogeneity. FMS-like tyrosine kinase 3 (FLT3) mutations occur in up to a third of AML cases and are associated with highly proliferative disease, shorter duration of remission, and increased rates of disease relapse. The known impact of activating mutations in FLT3 in AML on disease pathogenesis, prognosis, and response to therapy has led to the development of tyrosine kinase inhibitors targeting FLT3. Gilteritinib is a potent, second generation inhibitor of both FLT3 and AXL, designed to address the limitations of other FLT3 inhibitors, particularly in targeting mechanisms of resistance to other drugs. In this review, we present comprehensive data on recent and ongoing studies evaluating the role of gilteritinib in the relapsed and refractory FLT3 mutated AML setting.
Collapse
Affiliation(s)
- Serena Chew
- University of Texas, MD Anderson Cancer Center,
Houston, TX, USA
| | | | - Elias Jabbour
- Department of Leukemia, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 428, Houston, TX
77030, USA
| |
Collapse
|
129
|
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: 15.8] [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
|
130
|
Yu J, Jiang PYZ, Sun H, Zhang X, Jiang Z, Li Y, Song Y. Advances in targeted therapy for acute myeloid leukemia. Biomark Res 2020; 8:17. [PMID: 32477567 PMCID: PMC7238648 DOI: 10.1186/s40364-020-00196-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal malignancy characterized by genetic heterogeneity due to recurrent gene mutations. Treatment with cytotoxic chemotherapy has been the standard of care for more than half of a century. Although much progress has been made toward improving treatment related mortality rate in the past few decades, long term overall survival has stagnated. Exciting developments of gene mutation-targeted therapeutic agents are now changing the landscape in AML treatment. New agents offer more clinical options for patients and also confer a more promising outcome. Since Midostaurin, a FLT3 inhibitor, was first approved by US FDA in 2017 as the first gene mutation-targeted therapeutic agent, an array of new gene mutation-targeted agents are now available for AML treatment. In this review, we will summarize the recent advances in gene mutation-targeted therapies for patients with AML.
Collapse
Affiliation(s)
- Jifeng Yu
- The First Affiliated Hospital of Zhengzhou University, #1 East Jianshe Road, Zhengzhou, 450052 China
- Academy of Medical and Pharmaceutical Sciences of Zhengzhou University, #1 East Jianshe Road, Zhengzhou, 450052 China
| | - Peter Y. Z. Jiang
- Department of Hematology and Oncology, The Everett Clinic and Providence Regional Cancer Partnership, 1717 13th Street, Everett, WA 98201 USA
| | - Hao Sun
- The First Affiliated Hospital of Zhengzhou University, #1 East Jianshe Road, Zhengzhou, 450052 China
| | - Xia Zhang
- The First Affiliated Hospital of Zhengzhou University, #1 East Jianshe Road, Zhengzhou, 450052 China
| | - Zhongxing Jiang
- The First Affiliated Hospital of Zhengzhou University, #1 East Jianshe Road, Zhengzhou, 450052 China
| | - Yingmei Li
- The First Affiliated Hospital of Zhengzhou University, #1 East Jianshe Road, Zhengzhou, 450052 China
| | - Yongping Song
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| |
Collapse
|
131
|
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: 0.4] [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
|
132
|
van Alphen C, Cloos J, Beekhof R, Cucchi DGJ, Piersma SR, Knol JC, Henneman AA, Pham TV, van Meerloo J, Ossenkoppele GJ, Verheul HMW, Janssen JJWM, Jimenez CR. Phosphotyrosine-based Phosphoproteomics for Target Identification and Drug Response Prediction in AML Cell Lines. Mol Cell Proteomics 2020; 19:884-899. [PMID: 32102969 PMCID: PMC7196578 DOI: 10.1074/mcp.ra119.001504] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 02/05/2020] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder arising from hematopoietic myeloid progenitors. Aberrantly activated tyrosine kinases (TK) are involved in leukemogenesis and are associated with poor treatment outcome. Kinase inhibitor (KI) treatment has shown promise in improving patient outcome in AML. However, inhibitor selection for patients is suboptimal.In a preclinical effort to address KI selection, we analyzed a panel of 16 AML cell lines using phosphotyrosine (pY) enrichment-based, label-free phosphoproteomics. The Integrative Inferred Kinase Activity (INKA) algorithm was used to identify hyperphosphorylated, active kinases as candidates for KI treatment, and efficacy of selected KIs was tested.Heterogeneous signaling was observed with between 241 and 2764 phosphopeptides detected per cell line. Of 4853 identified phosphopeptides with 4229 phosphosites, 4459 phosphopeptides (4430 pY) were linked to 3605 class I sites (3525 pY). INKA analysis in single cell lines successfully pinpointed driver kinases (PDGFRA, JAK2, KIT and FLT3) corresponding with activating mutations present in these cell lines. Furthermore, potential receptor tyrosine kinase (RTK) drivers, undetected by standard molecular analyses, were identified in four cell lines (FGFR1 in KG-1 and KG-1a, PDGFRA in Kasumi-3, and FLT3 in MM6). These cell lines proved highly sensitive to specific KIs. Six AML cell lines without a clear RTK driver showed evidence of MAPK1/3 activation, indicative of the presence of activating upstream RAS mutations. Importantly, FLT3 phosphorylation was demonstrated in two clinical AML samples with a FLT3 internal tandem duplication (ITD) mutation.Our data show the potential of pY-phosphoproteomics and INKA analysis to provide insight in AML TK signaling and identify hyperactive kinases as potential targets for treatment in AML cell lines. These results warrant future investigation of clinical samples to further our understanding of TK phosphorylation in relation to clinical response in the individual patient.
Collapse
Affiliation(s)
- Carolien van Alphen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, OncoProteomics Laboratory, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam Department of Hematology, De Boelelaan 1117, Amsterdam, Netherlands
| | - Jacqueline Cloos
- Amsterdam UMC, Vrije Universiteit Amsterdam Department of Hematology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pediatric Oncology, De Boelelaan 1117, Amsterdam, Netherlands
| | - Robin Beekhof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, OncoProteomics Laboratory, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - David G J Cucchi
- Amsterdam UMC, Vrije Universiteit Amsterdam Department of Hematology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pediatric Oncology, De Boelelaan 1117, Amsterdam, Netherlands
| | - Sander R Piersma
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, OncoProteomics Laboratory, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Jaco C Knol
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, OncoProteomics Laboratory, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Alex A Henneman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, OncoProteomics Laboratory, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Thang V Pham
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, OncoProteomics Laboratory, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Johan van Meerloo
- Amsterdam UMC, Vrije Universiteit Amsterdam Department of Hematology, De Boelelaan 1117, Amsterdam, Netherlands
| | - Gert J Ossenkoppele
- Amsterdam UMC, Vrije Universiteit Amsterdam Department of Hematology, De Boelelaan 1117, Amsterdam, Netherlands
| | - Henk M W Verheul
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands
| | - Jeroen J W M Janssen
- Amsterdam UMC, Vrije Universiteit Amsterdam Department of Hematology, De Boelelaan 1117, Amsterdam, Netherlands
| | - Connie R Jimenez
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center; Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, OncoProteomics Laboratory, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, Netherlands.
| |
Collapse
|
133
|
Elmongy EI. Thieno[2,3‐
d
]pyrimidine derivatives: Synthetic approaches and their FLT3 kinase inhibition. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.3934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Elshaymaa I. Elmongy
- Department of Pharmaceutical Chemistry, Faculty of PharmacyHelwan University Cairo Egypt
- Department of Pharmaceutical Sciences, Faculty of PharmacyPrincess Nourah bint Abdulrahman University Riyadh Saudi Arabia
| |
Collapse
|
134
|
Kattner AS, Holler E, Herr W, Reichle A, Wolff D, Heudobler D. Successful Treatment of Early Relapsed High-Risk AML After Allogeneic Hematopoietic Stem Cell Transplantation With Biomodulatory Therapy. Front Oncol 2020; 10:443. [PMID: 32391254 PMCID: PMC7190808 DOI: 10.3389/fonc.2020.00443] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/12/2020] [Indexed: 11/13/2022] Open
Abstract
Early relapse of acute myeloid leukemia (AML) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an often unsuccessful therapeutic challenge. Since treatment options are few and efficacy is low, new approaches such as de novo allo-HSCT, targeted therapies and biomodulatory drugs have been developed, albeit prognosis is very poor. In this manuscript we present an unusual case of a patient with high-risk AML with an unbalanced jumping translocation and FLT3-TKD (low) mutation who presented with early relapse (FLT3 negative) after allo-HSCT, refractory to one cycle of azacytidine and discontinuation of immunosuppression (IS). As salvage therapy, the patient received a biomodulatory therapy consisting of low-dose azacytidine 75 mg/day (given s.c. d1-7 of 28), pioglitazone 45 mg/day orally, and all-trans-retinoic acid (ATRA) 45 mg/m2/day orally achieving a complete remission after two cycles of therapy. Even after cessation of treatment after 5 cycles, the patient remained in complete remission with full chimerism in peripheral blood and bone marrow for another 7 months. In conclusion, we report about an unusual case of long-lasting complete remission of early relapsed high-risk AML after allo-HSCT treated with azacytidine, pioglitazone and ATRA after standard of care treatment with HMA and discontinuation of IS failed.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Internal Oncology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
135
|
Abstract
Objective: To summarize the abnormal location of FLT3 caused by different glycosylation status which further leads to the distinguishing signaling pathways and discuss targeting on FLT3 glycosylation by drugs reported in recent literatures. Methods: We review FLT3 glycosylation in endoplasmic reticulum. The abnormal signal of mutant FLT3 with different glycosylation status is discussed. We also address potential FLT3 glycosylation-targeting strategies for the treatment. Results: Inhibition of FLT3 mutant cells by drugs reported in recent literatures involves the influence of glycosylation of FLT3: 2-deoxy-D-glucose, Tunicamycin and Fluvastatin are reported to inhibit N-glycosylation of FLT3; Pim-1 inhibitors are proved to block the inhibition of Pim-1 on FLT3 Oglycosylation; HSP90 inhibitors and Tyrosine Kinase Inhibitors are shown to increase fully glycosylated form of FLT3. Discussion: The FMS-like tyrosine kinase 3 (FLT3) gene expressed only in CD34+ progenitor cells in bone marrow is located on chromosome 13q12 encoding FLT3 protein. FLT3 is initially synthesized as a 110 KD protein, which glycosylated in the endoplasmic reticulum to a 130 KD immature protein rich in mannose, and further processed into a mature 160 KD protein in the Golgi apparatus, which could be transferred to the cell surface. Therapy targeting on FLT3 glycosylation is a promising direction for AML treatment. Conclusions: The abnormal location of FLT3 caused by different glycosylation status leads to the distinguishing signaling pathways. Targeting on FLT3 glycosylation may provide a new perspective for therapeutic strategies. Abbreviations: ABCG2: ATP-binding cassette transporter breast cancer resistance protein; ATF: activating transcription factor; AML: acute myeloid leukemia; CHOP: CCAAT-enhancer-binding protein homologous protein; 2-DG: 2-deoxy-D-glucose; EFS: event free survival; EPO: erythropoietin; EPOR: erythropoietin receptor; ERS: endoplasmic reticulum stress; FLT3: FMS-like tyrosine kinase 3; GPI: glycosylphosphatidylinositol; HSP: heat shock protein; ITD: internal tandem duplication; IRE1a: inositol-requiring enzyme 1 alpha; JNK: c-Jun N-terminal kinase; JMD: juxtamembrane domain; JAK: janus kinase; MAPK/ERK: mitogen activated protein kinase/extracellular signal-regulated protein kinase; OS: overall survival; PI3K/AKT: phosphatidylinositide 3-kinases/protein kinase B; PERK: RNA-activated protein kinase-like endoplasmic reticulum kinase; Pgp: P-glycoprotein; PTX3: human pentraxin-3; STAT: signal transducer and activator of transcriptions; TKD: tyrosine-kinase domain; TKI: tyrosine kinase inhibitor; TM: Tunicamycin; UPR: unfolded protein reaction.
Collapse
Affiliation(s)
- Xiaoli Hu
- Department of Hematology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , People's Republic of China
| | - Fangyuan Chen
- Department of Hematology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , People's Republic of China
| |
Collapse
|
136
|
Role of CYP3A4 in bone marrow microenvironment-mediated protection of FLT3/ITD AML from tyrosine kinase inhibitors. Blood Adv 2020; 3:908-916. [PMID: 30898762 DOI: 10.1182/bloodadvances.2018022921] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/29/2019] [Indexed: 11/20/2022] Open
Abstract
An intriguing aspect of the clinical activity of FMS-like tyrosine kinase 3 inhibitors (FLT3 TKIs) is their apparent higher activity against peripheral blasts from FLT3/internal tandem duplication (ITD) acute myeloid leukemia than marrow disease in the same patients. Accordingly, studies showed that the bone marrow microenvironment plays a role in FLT3 TKI resistance, although the underlying mechanisms are unclear. We recently identified a previously undescribed mechanism by which the bone marrow microenvironment can contribute to drug resistance: expression of cytochrome P450 enzymes (CYPs). In fact, bone marrow stromal cells (BMSCs) expressed most CYPs, including CYP3A4. Because hepatic CYP3A4 plays a role in the inactivation of several FLT3 TKIs, we explored the potential role of CYP3A4 in bone marrow microenvironment-mediated FLT3 TKI resistance. We found that CYP3A4 plays a major role in BMSC-mediated inhibition in the activity of 3 different FLT3 TKIs (sorafenib, quizartinib, and gilteritinib) against FLT3/ITD acute myeloid leukemia (AML). Furthermore, clarithromycin, a clinically active CYP3A4 inhibitor, significantly reversed the protective effects of BMSCs. We show, for the first time, that bone marrow stromal CYP3A4 contributes to FLT3 TKI resistance in the bone marrow. These results suggest that combining FLT3 TKIs with CYP3A4 inhibitors could be a promising strategy toward improving the activity of FLT3 TKIs.
Collapse
|
137
|
Sellmer A, Pilsl B, Beyer M, Pongratz H, Wirth L, Elz S, Dove S, Henninger SJ, Spiekermann K, Polzer H, Klaeger S, Kuster B, Böhmer FD, Fiebig HH, Krämer OH, Mahboobi S. A series of novel aryl-methanone derivatives as inhibitors of FMS-like tyrosine kinase 3 (FLT3) in FLT3-ITD-positive acute myeloid leukemia. Eur J Med Chem 2020; 193:112232. [PMID: 32199135 DOI: 10.1016/j.ejmech.2020.112232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
Mutants of the FLT3 receptor tyrosine kinase (RTK) with duplications in the juxtamembrane domain (FLT3-ITD) act as drivers of acute myeloid leukemia (AML). Potent tyrosine kinase inhibitors (TKi) of FLT3-ITD entered clinical trials and showed a promising, but transient success due to the occurrence of secondary drug-resistant AML clones. A further caveat of drugs targeting FLT3-ITD is the co-targeting of other RTKs which are required for normal hematopoiesis. This is observed quite frequently. Therefore, novel drugs are necessary to treat AML effectively and safely. Recently bis(1H-indol-2-yl)methanones were found to inhibit FLT3 and PDGFR kinases. In order to optimize these agents we synthesized novel derivatives of these methanones with various substituents. Methanone 16 and its carbamate derivative 17b inhibit FLT3-ITD at least as potently as the TKi AC220 (quizartinib). Models indicate corresponding interactions of 16 and quizartinib with FLT3. The activity of 16 is accompanied by a high selectivity for FLT3-ITD.
Collapse
Affiliation(s)
- Andreas Sellmer
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| | - Bernadette Pilsl
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| | - Mandy Beyer
- Department of Toxicology, University Medical Center, Mainz, Germany
| | - Herwig Pongratz
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| | - Lukas Wirth
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| | - Sigurd Elz
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| | - Stefan Dove
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| | | | | | - Harald Polzer
- Department of Medicine III, University Hospital, LMU Munich, Germany
| | - Susan Klaeger
- Technische Universität München, Wissenschaftszentrum Weihenstephan für Ernährung, Landnutzung und Umwelt, Germany
| | - Bernhard Kuster
- Technische Universität München, Wissenschaftszentrum Weihenstephan für Ernährung, Landnutzung und Umwelt, Germany
| | - Frank D Böhmer
- Universitätsklinikum Jena - Bachstrasse 18 - D-07743 Jena, Germany
| | | | - Oliver H Krämer
- Department of Toxicology, University Medical Center, Mainz, Germany
| | - Siavosh Mahboobi
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany.
| |
Collapse
|
138
|
Jeong P, Moon Y, Lee JH, Lee SD, Park J, Lee J, Kim J, Lee HJ, Kim NY, Choi J, Heo JD, Shin JE, Park HW, Kim YG, Han SY, Kim YC. Discovery of orally active indirubin-3'-oxime derivatives as potent type 1 FLT3 inhibitors for acute myeloid leukemia. Eur J Med Chem 2020; 195:112205. [PMID: 32272419 DOI: 10.1016/j.ejmech.2020.112205] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 01/13/2023]
Abstract
FMS-like receptor tyrosine kinase-3 (FLT3) is expressed on acute leukemia cells and is implicated in the survival, proliferation and differentiation of hematopoietic cells in most acute myeloid leukemia (AML) patients. Despite recent achievements in the development of FLT3-targeted small-molecule drugs, there are still unmet medical needs related to kinase selectivity and the progression of some mutant forms of FLT3. Herein, we describe the discovery of novel orally available type 1 FLT3 inhibitors from structure-activity relationship (SAR) studies for the optimization of indirubin derivatives with biological and pharmacokinetic profiles as potential therapeutic agents for AML. The SAR exploration provided important structural insights into the key substituents for potent inhibitory activities of FLT3 and in MV4-11 cells. The profile of the most optimized inhibitor (36) showed IC50 values of 0.87 and 0.32 nM against FLT3 and FLT3/D835Y, respectively, along with potent inhibition against MV4-11 and FLT3/D835Y expressed MOLM14 cells with a GI50 value of 1.0 and 1.87 nM, respectively. With the high oral bioavailability of 42.6%, compound 36 displayed significant in vivo antitumor activity by oral administration of 20 mg/kg once daily dosing schedule for 21 days in a mouse xenograft model. The molecular docking study of 36 in the homology model of the DFG-in conformation of FLT3 resulted in a reasonable binding mode in type 1 kinases similar to the reported type 1 FLT3 inhibitors Crenolanib and Gilteritinib.
Collapse
Affiliation(s)
- Pyeonghwa Jeong
- Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yeongyu Moon
- Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, Gyeongsangnam-do, 52834, Republic of Korea
| | - Je-Heon Lee
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - So-Deok Lee
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jiyeon Park
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jungeun Lee
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jiheon Kim
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Hyo Jeong Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, 52828, Republic of Korea
| | - Na Yoon Kim
- College of Pharmacy, Dankook University, Cheonan, 330-714, Republic of Korea
| | - Jungil Choi
- Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, Gyeongsangnam-do, 52834, Republic of Korea
| | - Jeong Doo Heo
- Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, Gyeongsangnam-do, 52834, Republic of Korea
| | - Ji Eun Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon-Gyoon Kim
- College of Pharmacy, Dankook University, Cheonan, 330-714, Republic of Korea
| | - Sun-Young Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, 52828, Republic of Korea.
| | - Yong-Chul Kim
- Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea; School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| |
Collapse
|
139
|
Borowczyk M, Szczepanek-Parulska E, Dębicki S, Budny B, Janicka-Jedyńska M, Gil L, Verburg FA, Filipowicz D, Wrotkowska E, Majchrzycka B, Marszałek A, Ziemnicka K, Ruchała M. High incidence of FLT3 mutations in follicular thyroid cancer: potential therapeutic target in patients with advanced disease stage. Ther Adv Med Oncol 2020; 12:1758835920907534. [PMID: 32180839 PMCID: PMC7057406 DOI: 10.1177/1758835920907534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 01/22/2020] [Indexed: 11/30/2022] Open
Abstract
Background: Conventional treatments for follicular thyroid cancer (FTC) can be ineffective, leading to poor prognosis. The aim of this study was to identify mutations associated with FTC that would serve as novel molecular markers of the disease and its outcome and could potentially identify new therapeutic targets. Methods: FLT3 mutations were first detected in a 29-year-old White female diagnosed with metastasized, treatment-refractory FTC. Analyses of FLT3 mutational status through next-generation sequencing of formalin-fixed, paraffin-embedded FTC specimens were subsequently performed in 35 randomly selected patients diagnosed with FTC. Results: FLT3 mutations were found in 69% of patients. FLT3 mutation-positive patients were significantly older than those that were FLT3 mutation-negative [median age at diagnosis 54 (36–82) versus 45 (27–58) (p = 0.023)]. Patients over 60 years were 23 times more likely to be FLT3 mutation-positive (p = 0.006). However, the number of FLT3 mutations did not correlate with age (r-Pearson: –0.244, p-value: 0.25). A total of 26 mutations were identified in the FLT3 gene with 2–16 FLT3 mutations in each FLT3 mutation-positive patient (mean: 5.6 mutations/patient). Tyrosine kinase domain (TKD) mutations in the FLT3 gene were detected in 58% of FLT3 mutation-positive patients. All FLT3 mutation-positive patients with a disease stage of pT2N1 or worse harbored at least one mutation in the TKD of FLT3. Conclusions: There is a wide spectrum and high frequency of FLT3 mutations in FTC. The precise role of FLT3 mutations in the genesis of FTC, as well as its potential role as a therapeutic target, requires further investigation.
Collapse
Affiliation(s)
- Martyna Borowczyk
- Department of Endocrinology, Metabolism and Internal Diseases, Poznań University of Medical Sciences, Przybyszewskiego Street, 49, Poznan, 60-355, Poland
| | - Ewelina Szczepanek-Parulska
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Szymon Dębicki
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Lidia Gil
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznan, Poland
| | - Frederik A Verburg
- Department of Nuclear Medicine, University Hospital Marburg, Marburg, Germany
| | - Dorota Filipowicz
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Elżbieta Wrotkowska
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Blanka Majchrzycka
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Marszałek
- Department of Oncologic Pathology and Prophylaxis, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
140
|
Garcia JS, Bhatt S, Fell G, Sperling AS, Burgess M, Keshishian H, Yilma B, Brunner A, Neuberg D, Carr SA, Ebert BL, Ballen K, Stone RM, DeAngelo DJ, Medeiros BC, Letai A. Increased mitochondrial apoptotic priming with targeted therapy predicts clinical response to re-induction chemotherapy. Am J Hematol 2020; 95:245-250. [PMID: 31804723 PMCID: PMC10683501 DOI: 10.1002/ajh.25692] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/30/2019] [Indexed: 02/06/2023]
Abstract
Most patients with relapsed or refractory (R/R) acute myeloid leukemia (AML) do not benefit from current re-induction or approved targeted therapies. In the absence of targetable genetic mutations, there is minimal guidance on optimal treatment selection particularly in the R/R setting highlighting an unmet need for clinically useful functional biomarkers. Blood and bone marrow samples from patients treated on two clinical trials were used to test the combination of lenalidomide (LEN) and MEC (mitoxantrone, etoposide, and cytarabine) chemotherapy in R/R AML patients. The bone marrow samples were available to test the clinical utility of the mitochondrial apoptotic BH3 and dynamic BH3 profiling (DBP) assays in predicting response, as there was no clear genetic biomarker identifying responders. To test whether LEN-induced mitochondrial priming predicted clinical response to LEN-MEC therapy, we performed DBP on patient myeloblasts. We found that short-term ex vivo treatment with lenalidomide discriminated clinical responders from non-responders based on drug-induced change in priming (delta priming). Using paired patient samples collected before and after clinical LEN treatment (prior to MEC dosing), we confirmed LEN-induced increased apoptotic priming in vivo, suggesting LEN enhanced vulnerability of myeloblasts to cytotoxic MEC chemotherapy. This is the first study demonstrating the potential role of DBP in predicting clinical response to a combination regimen. Our findings demonstrate that functional properties of relapsed AML can identify active therapies.
Collapse
Affiliation(s)
| | - Shruti Bhatt
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Michael Burgess
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - Binyam Yilma
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Steven A. Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - Karen Ballen
- University of Virginia Health System, Charlottesville, Virginia
| | | | | | | | | |
Collapse
|
141
|
Bair SM, Brandstadter JD, Ayers EC, Stadtmauer EA. Hematopoietic stem cell transplantation for blood cancers in the era of precision medicine and immunotherapy. Cancer 2020; 126:1837-1855. [PMID: 32073653 DOI: 10.1002/cncr.32659] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/08/2019] [Accepted: 09/19/2019] [Indexed: 01/12/2023]
Abstract
Hematopoietic stem cell transplantation (HCT) has been an integral component in the treatment of many hematologic malignancies. Since the development of HCT nearly 50 years ago, the role of this modality has evolved as newer treatment approaches have been developed and integrated into the standard of care. In the last decade, novel and highly active targeted therapies and immunotherapies have been approved for many hematologic malignancies, raising the question of whether HCT continues to retain its prominent role in the treatment paradigms of various hematologic malignancies. In this review, the authors have described the current role of autologous and allogeneic HCT in the treatment of patients with acute leukemias, aggressive B-cell lymphomas, and multiple myeloma and discussed how novel targeted therapies and immunotherapies have changed the potential need, timing, and goal of HCT in patients with these diseases.
Collapse
Affiliation(s)
- Steven M Bair
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua D Brandstadter
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily C Ayers
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward A Stadtmauer
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
142
|
Gurnari C, Voso MT, Maciejewski JP, Visconte V. From Bench to Bedside and Beyond: Therapeutic Scenario in Acute Myeloid Leukemia. Cancers (Basel) 2020; 12:cancers12020357. [PMID: 32033196 PMCID: PMC7072629 DOI: 10.3390/cancers12020357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of clonal disorders characterized by abnormal proliferation of undifferentiated myeloid progenitors, impaired hematopoiesis, and variable response to therapy. To date, only about 30% of adult patients with AML become long-term survivors and relapse and/or disease refractoriness are the major cause of treatment failure. Thus, this is an urgent unmet clinical need and new drugs are envisaged in order to ameliorate disease survival outcomes. Here, we review the latest therapeutic approaches (investigational and approved agents) for AML treatment. A specific focus will be given to molecularly targeted therapies for AML as a representation of possible agents for precision medicine. We will discuss experimental and preclinical data for FLT3, IDH1, BCL-2, Hedgehog pathway inhibitors, and epitherapy.
Collapse
Affiliation(s)
- Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (J.P.M.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
- Neuro-Oncohematology Unit, Fondazione Santa Lucia, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), 00143 Rome, Italy
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (J.P.M.)
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (J.P.M.)
- Correspondence:
| |
Collapse
|
143
|
Conneely SE, Stevens AM. Advances in Pediatric Acute Promyelocytic Leukemia. CHILDREN-BASEL 2020; 7:children7020011. [PMID: 32024232 PMCID: PMC7072343 DOI: 10.3390/children7020011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/25/2020] [Accepted: 01/27/2020] [Indexed: 02/07/2023]
Abstract
Acute promyelocytic leukemia (APL) is a rare disease accounting for only 5%-10% of pediatric acute myeloid leukemia (AML) and fewer than 1000 cases occur annually in the United States across all age groups. Characterized by t (15; 17), with a resultant PML-RARA gene fusion driving leukemia development, advances in therapy have improved outcomes for APL significantly in the past several decades, now making APL the most curable form of AML in both children and adults. Cure rates in APL are now comparable to pediatric B-lymphoid leukemias. The success of APL treatment is due, in part, to the breadth of understanding of the driver PML-RARA mutation as well as collaborative efforts to quickly introduce and maximize the benefit of new therapies. Here, we review the presentation, clinical features, pathogenesis, and treatment advances in pediatric APL.
Collapse
|
144
|
Georgoulia PS, Bjelic S, Friedman R. Deciphering the molecular mechanism of FLT3 resistance mutations. FEBS J 2020; 287:3200-3220. [PMID: 31943770 DOI: 10.1111/febs.15209] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/13/2019] [Accepted: 01/09/2020] [Indexed: 12/18/2022]
Abstract
FMS-like tyrosine kinase 3 (FLT3) has been found to be mutated in ~ 30% of acute myeloid leukaemia patients. Small-molecule inhibitors targeting FLT3 that are currently approved or still undergoing clinical trials are subject to drug resistance due to FLT3 mutations. How these mutations lead to drug resistance is hitherto poorly understood. Herein, we studied the molecular mechanism of the drug resistance mutations D835N, Y842S and M664I, which confer resistance against the most advanced inhibitors, quizartinib and PLX3397 (pexidartinib), using enzyme kinetics and computer simulations. In vitro kinase assays were performed to measure the comparative catalytic activity of the native protein and the mutants, using a bacterial expression system developed to this aim. Our results reveal that the differential drug sensitivity profiles can be rationalised by the dynamics of the protein-drug interactions and perturbation of the intraprotein contacts upon mutations. Drug binding induced a single conformation in the native protein, whereas multiple conformations were observed otherwise (in the mutants or in the absence of drugs). The end-point kinetics measurements indicated that the three resistant mutants conferred catalytic activity that is at least as high as that of the reference without such mutations. Overall, our calculations and measurements suggest that the structural dynamics of the drug-resistant mutants that affect the active state and the increased conformational freedom of the remaining inactive drug-bound population are the two major factors that contribute to drug resistance in FLT3 harbouring cancer cells. Our results explain the mechanism of drug resistance mutations and can aid to the design of more effective tyrosine kinase inhibitors.
Collapse
Affiliation(s)
| | - Sinisa Bjelic
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Ran Friedman
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
145
|
Aguilera-Diaz A, Vazquez I, Ariceta B, Mañú A, Blasco-Iturri Z, Palomino-Echeverría S, Larrayoz MJ, García-Sanz R, Prieto-Conde MI, del Carmen Chillón M, Alfonso-Pierola A, Prosper F, Fernandez-Mercado M, Calasanz MJ. Assessment of the clinical utility of four NGS panels in myeloid malignancies. Suggestions for NGS panel choice or design. PLoS One 2020; 15:e0227986. [PMID: 31978184 PMCID: PMC6980571 DOI: 10.1371/journal.pone.0227986] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/04/2020] [Indexed: 12/17/2022] Open
Abstract
The diagnosis of myeloid neoplasms (MN) has significantly evolved through the last few decades. Next Generation Sequencing (NGS) is gradually becoming an essential tool to help clinicians with disease management. To this end, most specialized genetic laboratories have implemented NGS panels targeting a number of different genes relevant to MN. The aim of the present study is to evaluate the performance of four different targeted NGS gene panels based on their technical features and clinical utility. A total of 32 patient bone marrow samples were accrued and sequenced with 3 commercially available panels and 1 custom panel. Variants were classified by two geneticists based on their clinical relevance in MN. There was a difference in panel’s depth of coverage. We found 11 discordant clinically relevant variants between panels, with a trend to miss long insertions. Our data show that there is a high risk of finding different mutations depending on the panel of choice, due both to the panel design and the data analysis method. Of note, CEBPA, CALR and FLT3 genes, remains challenging the use of NGS for diagnosis of MN in compliance with current guidelines. Therefore, conventional molecular testing might need to be kept in place for the correct diagnosis of MN for now.
Collapse
Affiliation(s)
- Almudena Aguilera-Diaz
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Iria Vazquez
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Beñat Ariceta
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Amagoia Mañú
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Zuriñe Blasco-Iturri
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | | | - María José Larrayoz
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca, IBSAL and CIBERONC, Salamanca, Spain
| | | | | | - Ana Alfonso-Pierola
- Hematology Department, Clinica Universidad de Navarra (CUN), Pamplona, Spain
| | - Felipe Prosper
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematology Department, Clinica Universidad de Navarra (CUN), Pamplona, Spain
| | - Marta Fernandez-Mercado
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
- Biomedical Engineering Department, School of Engineering, University of Navarra, San Sebastian, Spain
- * E-mail: ,
| | - María José Calasanz
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
- Scientific Co-Director of CIMA LAB Diagnostics, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| |
Collapse
|
146
|
Antar AI, Otrock ZK, Jabbour E, Mohty M, Bazarbachi A. FLT3 inhibitors in acute myeloid leukemia: ten frequently asked questions. Leukemia 2020; 34:682-696. [PMID: 31919472 DOI: 10.1038/s41375-019-0694-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/22/2019] [Accepted: 12/06/2019] [Indexed: 12/18/2022]
Abstract
The FMS-like tyrosine kinase 3 (FLT3) gene is mutated in approximately one third of patients with acute myeloid leukemia (AML), either by internal tandem duplications (FLT3-ITD), or by a point mutation mainly involving the tyrosine kinase domain (FLT3-TKD). Patients with FLT3-ITD have a high risk of relapse and low cure rates. Several FLT3 tyrosine kinase inhibitors have been developed in the last few years with variable kinase inhibitory properties, pharmacokinetics, and toxicity profiles. FLT3 inhibitors are divided into first generation multi-kinase inhibitors (such as sorafenib, lestaurtinib, midostaurin) and next generation inhibitors (such as quizartinib, crenolanib, gilteritinib) based on their potency and specificity of FLT3 inhibition. These diverse FLT3 inhibitors have been evaluated in myriad clinical trials as monotherapy or in combination with conventional chemotherapy or hypomethylating agents and in various settings, including front-line, relapsed or refractory disease, and maintenance therapy after consolidation chemotherapy or allogeneic stem cell transplantation. In this practical question-and-answer-based review, the main issues faced by the leukemia specialists on the use of FLT3 inhibitors in AML are addressed.
Collapse
Affiliation(s)
- Ahmad I Antar
- Department of Hematology and Oncology, Hammoud Hospital University Medical Center, Saida, Lebanon
| | - Zaher K Otrock
- Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Wayne State University, Detroit, MI, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamad Mohty
- Service d'hématologie clinique et thérapie cellulaire, Hôpital Saint-Antoine, INSERM UMRs 938 and université Sorbonne, Paris, France
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
147
|
Duan C, Fukuda T, Isaji T, Qi F, Yang J, Wang Y, Takahashi S, Gu J. Deficiency of core fucosylation activates cellular signaling dependent on FLT3 expression in a Ba/F3 cell system. FASEB J 2020; 34:3239-3252. [DOI: 10.1096/fj.201902313rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Chengwei Duan
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Feng Qi
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Jie Yang
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Yuqin Wang
- Department of Pharmacology Pharmacy College Nantong University Nantong China
| | - Shinichiro Takahashi
- Division of Laboratory Medicine Faculty of Medicine Tohoku Medical and Pharmaceutical University Sendai Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology Institute of Molecular Biomembrane and Glycobiology Tohoku Medical and Pharmaceutical University Sendai Japan
| |
Collapse
|
148
|
Palmieri R, Paterno G, De Bellis E, Mercante L, Buzzatti E, Esposito F, Del Principe MI, Maurillo L, Buccisano F, Venditti A. Therapeutic Choice in Older Patients with Acute Myeloid Leukemia: A Matter of Fitness. Cancers (Basel) 2020; 12:cancers12010120. [PMID: 31906489 PMCID: PMC7016986 DOI: 10.3390/cancers12010120] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/19/2019] [Accepted: 12/31/2019] [Indexed: 01/17/2023] Open
Abstract
Acute myeloid leukemia (AML), with an incidence increasing with age, is the most common acute leukemia in adults. Concurrent comorbidities, mild to severe organ dysfunctions, and low performance status (PS) are frequently found in older patients at the onset, conditioning treatment choice and crucially influencing the outcome. Although anthracyclines plus cytarabine-based chemotherapy, also called “7 + 3” regimen, remains the standard of care in young adults, its use in patients older than 65 years should be reserved to selected cases because of higher incidence of toxicity. These adverse features of AML in the elderly underline the importance of a careful patient assessment at diagnosis as a critical tool in the decision-making process of treatment choice. In this review, we will describe selected recently approved drugs as well as examine prognostic algorithms that may be helpful to assign treatment in elderly patients properly.
Collapse
Affiliation(s)
- Raffaele Palmieri
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Giovangiacinto Paterno
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Eleonora De Bellis
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Lisa Mercante
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Elisa Buzzatti
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Fabiana Esposito
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Maria Ilaria Del Principe
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Luca Maurillo
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Francesco Buccisano
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy; (R.P.); (G.P.); (E.D.B.); (L.M.); (E.B.); (F.E.); (M.I.D.P.); (F.B.)
- Fondazione Policlinico Tor Vergata, 00133 Rome, Italy;
- Correspondence: ; Tel.: +39-0620903226
| |
Collapse
|
149
|
Kiyoi H, Kawashima N, Ishikawa Y. FLT3 mutations in acute myeloid leukemia: Therapeutic paradigm beyond inhibitor development. Cancer Sci 2019; 111:312-322. [PMID: 31821677 PMCID: PMC7004512 DOI: 10.1111/cas.14274] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is a type III receptor tyrosine kinase that plays an important role in hematopoietic cell survival, proliferation and differentiation. The most clinically important point is that mutation of the FLT3 gene is the most frequent genetic alteration and a poor prognostic factor in acute myeloid leukemia (AML) patients. There are two major types of FLT3 mutations: internal tandem duplication mutations in the juxtamembrane domain (FLT3-ITD) and point mutations or deletion in the tyrosine kinase domain (FLT3-TKD). Both mutant FLT3 molecules are activated through ligand-independent dimerization and trans-phosphorylation. Mutant FLT3 induces the activation of multiple intracellular signaling pathways, mainly STAT5, MAPK and AKT signals, leading to cell proliferation and anti-apoptosis. Because high-dose chemotherapy and allogeneic hematopoietic stem cell transplantation cannot sufficiently improve the prognosis, clinical development of FLT3 kinase inhibitors expected. Although several FLT3 inhibitors have been developed, it takes more than 20 years from the first identification of FLT3 mutations until FLT3 inhibitors become clinically available for AML patients with FLT3 mutations. To date, three FLT3 inhibitors have been clinically approved as monotherapy or combination therapy with conventional chemotherapeutic agents in Japan and/or Europe and United states. However, several mechanisms of resistance to FLT3 inhibitors have already become apparent during their clinical trials. The resistance mechanisms are complex and emerging resistant clones are heterogenous. Further basic and clinical studies are required to establish the best therapeutic strategy for AML patients with FLT3 mutations.
Collapse
Affiliation(s)
- Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - 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
| |
Collapse
|
150
|
Sun X, Gao H, Yang Y, He M, Wu Y, Song Y, Tong Y, Rao Y. PROTACs: great opportunities for academia and industry. Signal Transduct Target Ther 2019; 4:64. [PMID: 31885879 PMCID: PMC6927964 DOI: 10.1038/s41392-019-0101-6] [Citation(s) in RCA: 401] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/17/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
Although many kinds of therapies are applied in the clinic, drug-resistance is a major and unavoidable problem. Another disturbing statistic is the limited number of drug targets, which are presently only 20-25% of all protein targets that are currently being studied. Moreover, the focus of current explorations of targets are their enzymatic functions, which ignores the functions from their scaffold moiety. As a promising and appealing technology, PROteolysis TArgeting Chimeras (PROTACs) have attracted great attention both from academia and industry for finding available approaches to solve the above problems. PROTACs regulate protein function by degrading target proteins instead of inhibiting them, providing more sensitivity to drug-resistant targets and a greater chance to affect the nonenzymatic functions. PROTACs have been proven to show better selectivity compared to classic inhibitors. PROTACs can be described as a chemical knockdown approach with rapidity and reversibility, which presents new and different biology compared to other gene editing tools by avoiding misinterpretations that arise from potential genetic compensation and/or spontaneous mutations. PRTOACs have been widely explored throughout the world and have outperformed not only in cancer diseases, but also in immune disorders, viral infections and neurodegenerative diseases. Although PROTACs present a very promising and powerful approach for crossing the hurdles of present drug discovery and tool development in biology, more efforts are needed to gain to get deeper insight into the efficacy and safety of PROTACs in the clinic. More target binders and more E3 ligases applicable for developing PROTACs are waiting for exploration.
Collapse
Affiliation(s)
- Xiuyun Sun
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084 P. R. China
| | - Hongying Gao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084 P. R. China
| | - Yiqing Yang
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084 P. R. China
| | - Ming He
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
| | - Yue Wu
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
| | - Yugang Song
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
| | - Yan Tong
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
| | - Yu Rao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084 P. R. China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| |
Collapse
|