51
|
Hogan FL, Williams V, Knapper S. FLT3 Inhibition in Acute Myeloid Leukaemia - Current Knowledge and Future Prospects. Curr Cancer Drug Targets 2021; 20:513-531. [PMID: 32418523 DOI: 10.2174/1570163817666200518075820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/12/2020] [Accepted: 03/29/2020] [Indexed: 12/20/2022]
Abstract
Activating mutations of FMS-like tyrosine kinase 3 (FLT3) are present in 30% of acute myeloid leukaemia (AML) patients at diagnosis and confer an adverse clinical prognosis. Mutated FLT3 has emerged as a viable therapeutic target and a number of FLT3-directed tyrosine kinase inhibitors have progressed through clinical development over the last 10-15 years. The last two years have seen United States Food and Drug Administration (US FDA) approvals of the multi-kinase inhibitor midostaurin for newly-diagnosed FLT3-mutated patients, when used in combination with intensive chemotherapy, and of the more FLT3-selective agent gilteritinib, used as monotherapy, for patients with relapsed or treatment-refractory FLT3-mutated AML. The 'second generation' agents, quizartinib and crenolanib, are also at advanced stages of clinical development. Significant challenges remain in negotiating a variety of potential acquired drug resistance mechanisms and in optimizing sequencing of FLT3 inhibitory drugs with existing and novel treatment approaches in different clinical settings, including frontline therapy, relapsed/refractory disease, and maintenance treatment. In this review, the biology of FLT3, the clinical challenge posed by FLT3-mutated AML, the developmental history of the key FLT3-inhibitory compounds, mechanisms of disease resistance, and the future outlook for this group of agents, including current and planned clinical trials, is discussed.
Collapse
Affiliation(s)
- Francesca L Hogan
- Department of Haematology, University Hospital of Wales, Cardiff, United Kingdom
| | - Victoria Williams
- Department of Haematology, University Hospital of Wales, Cardiff, United Kingdom
| | - Steven Knapper
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
52
|
RSK Isoforms in Acute Myeloid Leukemia. Biomedicines 2021; 9:biomedicines9070726. [PMID: 34202904 PMCID: PMC8301392 DOI: 10.3390/biomedicines9070726] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022] Open
Abstract
Ribosomal S6 Kinases (RSKs) are a group of serine/threonine kinases that function downstream of the Ras/Raf/MEK/ERK signaling pathway. Four RSK isoforms are directly activated by ERK1/2 in response to extracellular stimuli including growth factors, hormones, and chemokines. RSKs phosphorylate many cytosolic and nuclear targets resulting in the regulation of diverse cellular processes such as cell proliferation, survival, and motility. In hematological malignancies such as acute myeloid leukemia (AML), RSK isoforms are highly expressed and aberrantly activated resulting in poor outcomes and resistance to chemotherapy. Therefore, understanding RSK function in leukemia could lead to promising therapeutic strategies. This review summarizes the current information on human RSK isoforms and discusses their potential roles in the pathogenesis of AML and mechanism of pharmacological inhibitors.
Collapse
|
53
|
Qiao X, Ma J, Knight T, Su Y, Edwards H, Polin L, Li J, Kushner J, Dzinic SH, White K, Wang J, Lin H, Wang Y, Wang L, Wang G, Taub JW, Ge Y. The combination of CUDC-907 and gilteritinib shows promising in vitro and in vivo antileukemic activity against FLT3-ITD AML. Blood Cancer J 2021; 11:111. [PMID: 34099621 PMCID: PMC8184771 DOI: 10.1038/s41408-021-00502-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 12/31/2022] Open
Abstract
About 25% of patients with acute myeloid leukemia (AML) harbor FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations and their prognosis remains poor. Gilteritinib is a FLT3 inhibitor approved by the US FDA for use in adult FLT3-mutated relapsed or refractory AML patients. Monotherapy, while efficacious, shows short-lived responses, highlighting the need for combination therapies. Here we show that gilteritinib and CUDC-907, a dual inhibitor of PI3K and histone deacetylases, synergistically induce apoptosis in FLT3-ITD AML cell lines and primary patient samples and have striking in vivo efficacy. Upregulation of FLT3 and activation of ERK are mechanisms of resistance to gilteritinib, while activation of JAK2/STAT5 is a mechanism of resistance to CUDC-907. Gilteritinib and CUDC-907 reciprocally overcome these mechanisms of resistance. In addition, the combined treatment results in cooperative downregulation of cellular metabolites and persisting antileukemic effects. CUDC-907 plus gilteritinib shows synergistic antileukemic activity against FLT3-ITD AML in vitro and in vivo, demonstrating strong translational therapeutic potential.
Collapse
Affiliation(s)
- Xinan Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jun Ma
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tristan Knight
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| | - Yongwei Su
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lisa Polin
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jing Li
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Juiwanna Kushner
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sijana H Dzinic
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kathryn White
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jian Wang
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hai Lin
- Department of Hematology and Oncology, The First Hospital of Jilin University, Changchun, P.R. China
| | - Yue Wang
- Department of Pediatric Hematology and Oncology, The First Hospital of Jilin University, Changchun, P.R. China
| | - Liping Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.
| | - Jeffrey W Taub
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA.
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
54
|
Deletions in FLT-3 juxtamembrane domain define a new class of pathogenic mutations: case report and systematic analysis. Blood Adv 2021; 5:2285-2293. [PMID: 33914060 DOI: 10.1182/bloodadvances.2020002876] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 03/17/2021] [Indexed: 11/20/2022] Open
Abstract
The FMS-like tyrosine kinase 3 (FLT-3) is the most frequently mutated gene in acute myeloid leukemia (AML), a high-risk feature, and now the target of tyrosine kinase inhibitors (TKIs), which are approved and in development. The most common mutation is the internal tandem duplication (ITD). We present a novel mutation, FLT-3/Q575Δ, identified in a patient with AML through next-generation sequencing (NGS). This mutation is activating, drives downstream signaling comparable to FLT-3/ITD, and can be targeted using available FLT-3 TKIs. We present the results of a systematic analysis that identified Y572Δ, E573Δ, and S574Δ as similarly activating and targetable deletions located in the FLT-3 juxtamembrane domain (JMD). These mutations target key residues in the JMD involved in the interactions within FLT-3 that regulate its activation. Our results suggest a new class of FLT-3 mutations that may have an impact on patient care and highlight the increasing importance of a systematic understanding of FLT-3 mutations other than ITD. It is likely that, as NGS becomes more commonly used in the diagnosis of patients with AML, these and other activating mutations will be discovered with increasing frequency.
Collapse
|
55
|
Lai SH, Li YC, Zhang S, Deng R, Deng Y, Fan FY. Whole genome, exon mutation and transcriptomic profiling of acute myeloid leukemia: A case report. Oncol Lett 2021; 22:559. [PMID: 34084226 PMCID: PMC8161460 DOI: 10.3892/ol.2021.12820] [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: 03/19/2020] [Accepted: 12/14/2020] [Indexed: 11/28/2022] Open
Abstract
The present study aimed to observe previously unidentified gene mutation and expression profiles associated with acute myeloid leukemia (AML) at the individual level, based on the blood samples of a father-son pair. Genomic DNA and RNA samples from blood serum were collected. Whole-genome sequencing (WGS) and whole-exome sequencing (WES), as well as mRNA sequencing of the son, were performed. For the father's sample, a total of 3,897,164 single nucleotide polymorphisms (SNPs) and 780,834 insertion and deletions (indels) were identified. Regarding amino acid translation, there were 11,316 non-synonymous, 12 stop-loss, 12,033 synonymous, 92 stop-gain SNPs, 63 frameshift insertions, 73 frameshift deletions, 242 non-frameshift insertions, 248 non-frameshift deletions, four stop-gains and two stop-loss for indel variants. Among the AML-related genes that had been previously identified, 14 genes were found in the father's exon region. For WES of the son's DNA, 96,639 SNPs were identified, including 10,504 non-synonymous SNPs. Seven mutant genes were found in sons' exon region compared with 121 AML-related genes. Based on the transcriptomic sequencing, there were 54 differentially expressed mRNAs, including 31 upregulated and 23 downregulated mRNAs. In the exon region, 10,072 SNPs were detected, and different types of alternative splicing in the son's sample were observed. Overall, whole genome, exon mutation and transcriptomic profiling of the present two patients with AML may provide a new insight into the molecular events governing the development of AML.
Collapse
Affiliation(s)
- Si-Han Lai
- Hematology Department and Hematopoietic Stem Cell Transplantation Center, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Ye-Cheng Li
- Hematology Department and Hematopoietic Stem Cell Transplantation Center, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Shan Zhang
- Hematology Department and Hematopoietic Stem Cell Transplantation Center, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Rui Deng
- Hematology Department and Hematopoietic Stem Cell Transplantation Center, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Yan Deng
- Hematology Department and Hematopoietic Stem Cell Transplantation Center, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Fang-Yi Fan
- Hematology Department and Hematopoietic Stem Cell Transplantation Center, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
56
|
Arries CD, Yohe SL. Monocytic Maturation Induced by FLT3 Inhibitor Therapy of Acute Myeloid Leukemia: Morphologic and Immunophenotypic Characteristics. Lab Med 2021; 51:478-483. [PMID: 31872224 DOI: 10.1093/labmed/lmz094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND FMS-like tyrosine kinase-3 (FLT3-ITD) mutations are some of the most common mutations in acute myeloid leukemia (AML), and patient outcomes have improved since the advent of tyrosine kinase inhibitors. First, granulocytic differentiation was described in FLT3-positive AML treated with FLT3 inhibitors, and more recently, monocytic differentiation was reported. METHODS Two patients with myelomonocytic cells in their bone marrow were identified during routine follow-up after AML treatment that included FLT3 inhibitors. The bone marrow study was done as standard of care. RESULTS Both patients had FLT3-ITD+ AML and showed an atypical maturing monocytic cell population and a decrease in the leukemic blast cell population after FLT3 inhibitor therapy. Concurrent genetic testing revealed persistent genetic abnormalities. CONCLUSIONS These cases illustrate monocytic maturation in FLT3+ AML after FLT3 inhibitor treatment. It is critical for pathologists and clinicians to be aware of the differentiation phenomenon, as these patients have persistent molecular abnormalities despite response to treatment and normalization of blast counts.
Collapse
|
57
|
Gilteritinib: potent targeting of FLT3 mutations in AML. Blood Adv 2021; 4:1178-1191. [PMID: 32208491 DOI: 10.1182/bloodadvances.2019000174] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/26/2020] [Indexed: 01/13/2023] Open
Abstract
Since the discovery of FMS-like tyrosine kinase-3 (FLT3)-activating mutations as genetic drivers in acute myeloid leukemia (AML), investigators have tried to develop tyrosine kinase inhibitors that could effectively target FLT3 and alter the disease trajectory. Giltertinib (formerly known as ASP2215) is a novel compound that entered the field late, but moved through the developmental process with remarkable speed. In many ways, this drug's rapid development was facilitated by the large body of knowledge gained over the years from efforts to develop other FLT3 inhibitors. Single-agent gilteritinib, a potent and selective oral FLT3 inhibitor, improved the survival of patients with relapsed or refractory FLT3-mutated AML compared with standard chemotherapy. This continues to validate the approach of targeting FLT3 itself and establishes a new backbone for testing combination regimens. This review will frame the preclinical and clinical development of gilteritinib in the context of the lessons learned from its predecessors.
Collapse
|
58
|
Yuan D, He X, Han X, Yang C, Liu F, Zhang S, Luan H, Li R, He J, Duan X, Wang D, Zhou Q, Gao S, Niu B. Comprehensive review and evaluation of computational methods for identifying FLT3-internal tandem duplication in acute myeloid leukaemia. Brief Bioinform 2021; 22:6225087. [PMID: 33851200 DOI: 10.1093/bib/bbab099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/15/2021] [Accepted: 03/06/2021] [Indexed: 12/25/2022] Open
Abstract
Internal tandem duplication (ITD) of FMS-like tyrosine kinase 3 (FLT3-ITD) constitutes an independent indicator of poor prognosis in acute myeloid leukaemia (AML). AML with FLT3-ITD usually presents with poor treatment outcomes, high recurrence rate and short overall survival. Currently, polymerase chain reaction and capillary electrophoresis are widely adopted for the clinical detection of FLT3-ITD, whereas the length and mutation frequency of ITD are evaluated using fragment analysis. With the development of sequencing technology and the high incidence of FLT3-ITD mutations, a multitude of bioinformatics tools and pipelines have been developed to detect FLT3-ITD using next-generation sequencing data. However, systematic comparison and evaluation of the methods or software have not been performed. In this study, we provided a comprehensive review of the principles, functionality and limitations of the existing methods for detecting FLT3-ITD. We further compared the qualitative and quantitative detection capabilities of six representative tools using simulated and biological data. Our results will provide practical guidance for researchers and clinicians to select the appropriate FLT3-ITD detection tools and highlight the direction of future developments in this field. Availability: A Docker image with several programs pre-installed is available at https://github.com/niu-lab/docker-flt3-itd to facilitate the application of FLT3-ITD detection tools.
Collapse
Affiliation(s)
- Danyang Yuan
- Computer Network Information Center, Chinese Academy of Sciences. She is mainly engaged in leukaemia-related bioinformatics and cancer genomics research. Her affiliation is with Computer Network Information Center, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoyu He
- Computer Network Information Center, Chinese Academy of Sciences. She is mainly engaged in research related to the cancer genome and construction of the Chinese Cancer Genome Database. Her affiliation is with Computer Network Information Center, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Xinyin Han
- Computer Network Information Center, Chinese Academy of Sciences. He is mainly engaged in cancer genomics research focusing on the precise detection of tumour immunotherapy biomarkers. His affiliation is with Computer Network Information Center, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Chunyan Yang
- Vice Director of the Laboratory of ChosenMed Technology (Beijing) Co., Ltd. She is mainly engaged in research regarding solid tumours and haematologic malignancy using multiple approaches, including next-generation sequencing. Her affiliation is with ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| | - Fei Liu
- bioinformatics analysis engineer of ChosenMed Technology (Beijing) Co., Ltd. She is mainly engaged in the collection of biological information and analysis of genomic and cancer data or other biological information. Her affiliation is with ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| | - Shuying Zhang
- Computer Network Information Center, Chinese Academy of Sciences. Her research mainly focuses on the cancer genome and bioinformatics. Her affiliation is with Computer Network Information Center, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Haijing Luan
- Computer Network Information Center, Chinese Academy of Sciences. She is mainly engaged in researching cancers of unknown primary sites (CUP) based on deep learning. Her affiliation is with Computer Network Information Center, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Ruilin Li
- Computer Network Information Center, Chinese Academy of Sciences. Her research interests include high-performance computing and bioinformatics. Her affiliation is with Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
| | - Jiayin He
- George Washington University. She is currently researching at the Computer Network Information Center, Chinese Academy of Sciences. Her research interests include biostatistics and computational statistics. Her affiliation is with Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaohong Duan
- Laboratory of ChosenMed Technology (Beijing) Co., Ltd. She is mainly engaged in the research of solid tumours and haematologic malignancies using multiple approaches, including next-generation sequencing. Her affiliation is with ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| | - Dongliang Wang
- Harbin Medical University. He is now the Chief Medical Officer of ChosenMed Technology (Beijing). His research mainly focuses on the mining and verification of molecular markers for tumour therapy. His affiliation is with ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| | - Qiming Zhou
- CTO of ChosenMed Technology (Beijing) Co., Ltd. He is mainly engaged in the development of new molecular diagnostics technologies in genetic testing. His affiliation is with ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| | - Sujun Gao
- Department of Haematology, The First Hospital of Jilin University. Her research mainly focuses on the experimental and clinical research of malignant haematological disorders and haematopoietic stem cell transplantation. Her affiliation is with Department of Haematology, The First Hospital of Jilin University, Changchun 130021, China
| | - Beifang Niu
- Computer Network Information Center, Chinese Academy of Sciences. His research interests include cancer genomics, metagenomics, and the development of computational tools for working with data from next-generation sequencing technologies. His affiliation is with Computer Network Information Center, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing 100190, China, ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| |
Collapse
|
59
|
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
|
60
|
Luo Y, Lu Y, Long B, Lin Y, Yang Y, Xu Y, Zhang X, Zhang J. Blocking DNA Damage Repair May Be Involved in Stattic (STAT3 Inhibitor)-Induced FLT3-ITD AML Cell Apoptosis. Front Cell Dev Biol 2021; 9:637064. [PMID: 33796529 PMCID: PMC8007876 DOI: 10.3389/fcell.2021.637064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The FMS-like tyrosine kinase 3 (FLT3)- internal tandem duplication (ITD) mutation can be found in approximately 25% of all acute myeloid leukemia (AML) cases and is associated with a poor prognosis. The main treatment for FLT3-ITD-positive AML patients includes genotoxic therapy and FLT3 inhibitors, which are rarely curative. Inhibiting STAT3 activity can improve the sensitivity of solid tumor cells to radiotherapy and chemotherapy. This study aimed to explore whether Stattic (a STAT3 inhibitor) affects FLT3-ITD AML cells and the underlying mechanism. Stattic can inhibit the proliferation, promote apoptosis, arrest cell cycle at G0/G1, and suppress DNA damage repair in MV4-11cells. During the process, through mRNA sequencing, we found that DNA damage repair-related mRNA are also altered during the process. In summary, the mechanism by which Stattic induces apoptosis in MV4-11cells may involve blocking DNA damage repair machineries.
Collapse
Affiliation(s)
- Yuxuan Luo
- Department of Pediatric, Guangzhou Women and Children's Medical Center, Guangzhou, China.,Department of Hematology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Lu
- Department of Hematology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Blood Transfusion, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bing Long
- Department of Hematology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Sen Yat-sen Institute of Hematology, Guangzhou, China
| | - Yansi Lin
- Department of General Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanling Yang
- Department of Hematology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yichuang Xu
- Department of Hematology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiangzhong Zhang
- Department of Hematology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Sen Yat-sen Institute of Hematology, Guangzhou, China
| | - Jingwen Zhang
- Department of Hematology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Sen Yat-sen Institute of Hematology, Guangzhou, China
| |
Collapse
|
61
|
OTS167 blocks FLT3 translation and synergizes with FLT3 inhibitors in FLT3 mutant acute myeloid leukemia. Blood Cancer J 2021; 11:48. [PMID: 33658483 PMCID: PMC7930094 DOI: 10.1038/s41408-021-00433-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/22/2021] [Accepted: 02/03/2021] [Indexed: 12/30/2022] Open
Abstract
Internal tandem duplication (-ITD) mutations of Fms-like tyrosine kinase 3 (FLT3) provide growth and pro-survival signals in the context of established driver mutations in FLT3 mutant acute myeloid leukemia (AML). Maternal embryonic leucine zipper kinase (MELK) is an aberrantly expressed gene identified as a target in AML. The MELK inhibitor OTS167 induces cell death in AML including cells with FLT3 mutations, yet the role of MELK and mechanisms of OTS167 function are not understood. OTS167 alone or in combination with tyrosine kinase inhibitors (TKIs) were used to investigate the effect of OTS167 on FLT3 signaling and expression in human FLT3 mutant AML cell lines and primary cells. We describe a mechanism whereby OTS167 blocks FLT3 expression by blocking FLT3 translation and inhibiting phosphorylation of eukaryotic initiation factor 4E–binding protein 1 (4E-BP1) and eukaryotic translation initiation factor 4B (eIF4B). OTS167 in combination with TKIs results in synergistic induction of FLT3 mutant cell death in FLT3 mutant cell lines and prolonged survival in a FLT3 mutant AML xenograft mouse model. Our findings suggest signaling through MELK is necessary for the translation and expression of FLT3-ITD, and blocking MELK with OTS167 represents a viable therapeutic strategy for patients with FLT3 mutant AML.
Collapse
|
62
|
Mosquera Orgueira A, Peleteiro Raíndo A, Cid López M, Antelo Rodríguez B, Díaz Arias JÁ, Ferreiro Ferro R, Alonso Vence N, Bendaña López Á, Abuín Blanco A, Bao Pérez L, Melero Valentín P, González Pérez MS, Cerchione C, Martinelli G, Montesinos Fernández P, Pérez Encinas MM, Bello López JL. Gene expression profiling identifies FLT3 mutation-like cases in wild-type FLT3 acute myeloid leukemia. PLoS One 2021; 16:e0247093. [PMID: 33592069 PMCID: PMC7886212 DOI: 10.1371/journal.pone.0247093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/01/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND FLT3 mutation is present in 25-30% of all acute myeloid leukemias (AML), and it is associated with adverse outcome. FLT3 inhibitors have shown improved survival results in AML both as upfront treatment and in relapsed/refractory disease. Curiously, a variable proportion of wild-type FLT3 patients also responded to these drugs. METHODS We analyzed 6 different transcriptomic datasets of AML cases. Differential expression between mutated and wild-type FLT3 AMLs was performed with the Wilcoxon-rank sum test. Hierarchical clustering was used to identify FLT3-mutation like AMLs. Finally, enrichment in recurrent mutations was performed with the Fisher's test. RESULTS A FLT3 mutation-like gene expression pattern was identified among wild-type FLT3 AMLs. This pattern was highly enriched in NPM1 and DNMT3A mutants, and particularly in combined NPM1/DNMT3A mutants. CONCLUSIONS We identified a FLT3 mutation-like gene expression pattern in AML which was highly enriched in NPM1 and DNMT3A mutations. Future analysis about the predictive role of this biomarker among wild-type FLT3 patients treated with FLT3 inhibitors is envisaged.
Collapse
Affiliation(s)
- Adrián Mosquera Orgueira
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
- University of Santiago de Compostela, Santiago, Spain
- * E-mail:
| | - Andrés Peleteiro Raíndo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
- University of Santiago de Compostela, Santiago, Spain
| | - Miguel Cid López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
- University of Santiago de Compostela, Santiago, Spain
| | - Beatriz Antelo Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
- University of Santiago de Compostela, Santiago, Spain
| | - José Ángel Díaz Arias
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | - Roi Ferreiro Ferro
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | - Natalia Alonso Vence
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | - Ángeles Bendaña López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | - Aitor Abuín Blanco
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | - Laura Bao Pérez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | | | - Marta Sonia González Pérez
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
| | | | - Giovanni Martinelli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy
| | | | - Manuel Mateo Pérez Encinas
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
- University of Santiago de Compostela, Santiago, Spain
| | - José Luis Bello López
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
- Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Santiago, Spain
- University of Santiago de Compostela, Santiago, Spain
| |
Collapse
|
63
|
Song IC, Jo DY, Kim HJ, Min YH, Hong DS, Lee WS, Shin HJ, Lee JH, Park J, Kim HJ. Clinical features and outcomes of hypocellular acute myeloid leukemia in adults: A Korean AML registry data. Medicine (Baltimore) 2021; 100:e24185. [PMID: 33429807 PMCID: PMC7793401 DOI: 10.1097/md.0000000000024185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/28/2020] [Accepted: 12/12/2020] [Indexed: 01/05/2023] Open
Abstract
The hypocellular variant of acute myeloid leukemia (AML) is defined as bone marrow cellularity of <20% in a biopsy specimen at presentation. We performed a retrospective analysis of the clinical features and survival outcomes of hypocellular AML in a Korean population. We reviewed the medical records of all patients diagnosed with AML at nine hospitals participating in the Korean AML registry from 2006 to 2012. Overall survival (OS) and event-free survival (EFS) rates were calculated from the time of diagnosis until death or an event, respectively. In total, 2110 patients were enrolled and 102 (4.8%) were identified as having hypocellular AML. Patients with hypocellular AML were older than those with non-hypocellular AML (median age: 59 vs 49 years; P < .001) and presented with leukopenia more frequently (mean white blood cell count: 5810/μL vs 40549/μL; P < .001). There was no difference between patients with and without hypocellular AML in terms of the presence of antecedent hematologic disorders (5.9% vs 5.3%; P = .809). FLT3-ITD and NPM1 mutations were less common in hypocellular than non-hypocellular AML (FLT3-ITD mutations: 1.2% vs 14.3%, P < .001; NPM1 mutations: 0% vs 9.5%, P = .019). No differences were seen between the hypocellular and non-hypocellular AML groups in the complete remission rate (53.9% vs 61.3%, P = .139) or early death rate (defined as any death before 8 weeks; 14.7% vs 13.0%, P = .629). The OS and EFS did not differ between the hypocellular and non-hypocellular AML groups (median OS: 16 vs 23 months, P = .169; median EFS: 6 vs 9 months, P = .215). Hypocellular AML is more frequently observed in older-aged patients and have fewer FLT3-ITD and NPM1 mutation, but the clinical outcomes of hypocellular AML do not differ from those of non-hypocellular AML.
Collapse
Affiliation(s)
- Ik-Chan Song
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon
| | - Deog-Yeon Jo
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon
| | - Hyeoung-Joon Kim
- Division of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do
| | - Yoo-Hong Min
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul
| | - Dae Sik Hong
- Department of Hemato-Oncolgy, Soon Chun Hyang University Hospital, Bucheon
| | - Won-Sik Lee
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan
| | - Ho-Jin Shin
- Division of Hematology-Oncology, Department of Internal Medicine, School of Medicine, Pusan National University Hospital, Pusan
| | - Je-Hwan Lee
- Department of Hematology, Asan Medical Center, University of Ulsan College of Medicine, Seoul
| | - Jinny Park
- Division of Hematology-Oncology, Department of Internal Medicine, Gachon University Gil Hospital, Incheon
| | - Hee-Je Kim
- Department of Hematology, Leukemia Research Institute, Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, South Korea
| |
Collapse
|
64
|
Pandya BJ, Yang H, Schmeichel C, Qi CZ, Shah MV. A budget impact analysis of gilteritinib for the treatment of relapsed or refractory FLT3 mut+ acute myeloid leukemia in a US health plan. J Med Econ 2021; 24:19-28. [PMID: 33280474 DOI: 10.1080/13696998.2020.1851698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIMS To estimate the economic impact of the introduction of gilteritinib for the treatment of relapsed/refractory (R/R) FLT3 mutation-positive (FLT3 mut+) acute myeloid leukemia (AML) from a US payer's perspective. METHODS A budget impact model (BIM) was developed to evaluate the 3-year total budgetary impact of treating adults with R/R FLT3 mut+ AML eligible for gilteritinib in a hypothetical US health plan. Total costs (drugs/administration, hospitalization, monitoring, adverse events, transfusions, subsequent hematopoietic stem cell transplantation, post-progression, and FLT3 testing) were estimated before and after gilteritinib entry. The budget impact was the total cost difference between the two scenarios. The target population size and cost inputs were based on public data or published literature, drug market share was informed by market research data, and the model included recommended treatments for R/R FLT3 mut+ AML per clinical guidelines. Deterministic sensitivity analyses (DSAs) and scenario analyses varying key model inputs and assumptions were conducted to test for robustness. RESULTS In a hypothetical health plan with 1 million members, 20.9 adults with R/R FLT3 mut+ AML were estimated to be eligible for gilteritinib. Of these, it was assumed 30.0% would be treated with gilteritinib in Year 1 following gilteritinib entry, increasing the total plan budget by $663,795 and the per-member-per-month (PMPM) cost by $0.055. In Years 2-3, the market share of gilteritinib increased to 45.0%, increasing the total plan budget impact by $1,078,371 and $1,087,230, and the PMPM cost by $0.090 and $0.091, respectively. The model results remained robust in DSAs and scenario analyses, with the largest impact observed when the projected uptake of gilteritinib was changed. LIMITATIONS The results of this BIM are contingent upon the model's assumptions and inputs. CONCLUSIONS Adding gilteritinib to the formulary for the treatment of adults with R/R FLT3 mut+ AML had a minimal budget impact from a US payer's perspective.
Collapse
Affiliation(s)
- Bhavik J Pandya
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | | | | | | | - Manasee V Shah
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| |
Collapse
|
65
|
Jones LM, Melgar K, Bolanos L, Hueneman K, Walker MM, Jiang JK, Wilson KM, Zhang X, Shen J, Jiang F, Sutter P, Wang A, Xu X, Tawa GJ, Hoyt SB, Wunderlich M, O'Brien E, Perentesis JP, Starczynowski DT, Thomas CJ. Targeting AML-associated FLT3 mutations with a type I kinase inhibitor. J Clin Invest 2020; 130:2017-2023. [PMID: 32149729 DOI: 10.1172/jci127907] [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: 02/02/2019] [Accepted: 01/08/2020] [Indexed: 12/31/2022] Open
Abstract
Tyrosine kinase domain (TKD) mutations contribute to acquired resistance to FMS-like tyrosine kinase 3 (FLT3) inhibitors used to treat FLT3-mutant acute myeloid leukemia (AML). We report a cocrystal structure of FLT3 with a type I inhibitor, NCGC1481, that retained potent binding and activity against FLT3 TKD and gatekeeper mutations. Relative to the current generation of advanced FLT3 inhibitors, NCGC1481 exhibited superior antileukemic activity against the common, clinically relevant FLT3-mutant AML cells in vitro and in vivo.
Collapse
Affiliation(s)
| | - Katelyn Melgar
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lyndsey Bolanos
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathleen Hueneman
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Morgan M Walker
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Jian-Kang Jiang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Kelli M Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | | | | | - Patrick Sutter
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Amy Wang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Xin Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Gregory J Tawa
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Scott B Hoyt
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA.,Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland
| |
Collapse
|
66
|
Suknuntha K, Choi YJ, Jung HS, Majumder A, Shah S, Slukvin I, Ranheim EA. Megakaryocytic Expansion in Gilteritinib-Treated Acute Myeloid Leukemia Patients Is Associated With AXL Inhibition. Front Oncol 2020; 10:585151. [PMID: 33363015 PMCID: PMC7756118 DOI: 10.3389/fonc.2020.585151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 01/09/2023] Open
Abstract
Numerous recurrent genetic mutations are known to occur in acute myeloid leukemia (AML). Among these common mutations, Fms-like tyrosine kinase 3 remains as one of the most frequently mutated genes in AML. We observed apparent marrow expansion of megakaryocytes in three out of six patients with Flt3-mutated AML following treatment with a recently FDA-approved Flt3 inhibitor, gilteritinib which possesses activity against internal tandem duplication and tyrosine kinase domain Flt3 mutations and also inhibits tyrosine kinase AXL. To assess whether biopsy findings can be attributed to promotion of megakaryocytic (Mk) differentiation with gilteritinib, we devised a cellular assay by overexpressing double mutated Flt3-ITDY591F/Y919F in chronic myeloid leukemia cell line K562 to study Mk differentiation in the presence of Flt3 and AXL inhibitors with non-mutually exclusive mechanisms. These experiments demonstrated the lack of direct effect Flt3 inhibitors gilteritinib and quizartinib on megakaryocytic differentiation at either transcriptional or phenotypic levels, and highlighted antileukemic effects of AXL receptor tyrosine kinase inhibitor and its potential role in megakaryocytic development.
Collapse
Affiliation(s)
- Kran Suknuntha
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States.,Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Yoon Jung Choi
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States
| | - Ho Sun Jung
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States
| | - Aditi Majumder
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States
| | - Sujal Shah
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| | - Igor Slukvin
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States
| | - Erik A Ranheim
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
67
|
Shen J, Lu Z, Wang J, Zhang T, Yang J, Li Y, Liu G, Zhang X. Advances of Nanoparticles for Leukemia Treatment. ACS Biomater Sci Eng 2020; 6:6478-6489. [PMID: 33320613 DOI: 10.1021/acsbiomaterials.0c01040] [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] [Indexed: 02/06/2023]
Abstract
Leukemia is a liquid tumor caused by a hematopoietic stem cell malignant clone, which seriously affects the normal function of the hematopoietic system. Conventional drugs have poor therapeutic effects due to their poor specificity and stability. With the development of nanotechnology, nonviral nanoparticles bring hope for the efficient treatment of leukemia. Nanoparticles are easily modified. They can be designed to target lesion sites and control drug release. Thereby, nanoparticles can improve the effects of drugs and reduce side effects. This review mainly focuses on and summarizes the current research progress of nanoparticles to deliver different leukemia therapeutic drugs, as to demonstrate the potential of nanoparticles in leukemia treatment.
Collapse
Affiliation(s)
- Jie Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China.,School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China.,School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jianze Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Tianlu Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yan Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Guiying Liu
- Department of Pediatrics, Capital Medical University Affiliated Beijing Anzhen Hospital, Beijing, 100029, PR China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
| |
Collapse
|
68
|
Abstract
Acute myeloid leukemia (AML) is a clinically, morphologically, and genetically heterogeneous disorder. Like many malignancies, the genomic landscape of pediatric AML has been mapped recently through sequencing of large cohorts of patients. Much has been learned about the biology of AML through studies of specific recurrent genetic lesions. Further, genetic lesions have been linked to specific clinical features, response to therapy, and outcome, leading to improvements in risk stratification. Lastly, targeted therapeutic approaches have been developed for the treatment of specific genetic lesions, some of which are already having a positive impact on outcomes. While the advances made based on the discoveries of sequencing studies are significant, much work is left. The biologic, clinical, and prognostic impact of a number of genetic lesions, including several seemingly unique to pediatric patients, remains undefined. While targeted approaches are being explored, for most, the efficacy and tolerability when incorporated into standard therapy is yet to be determined. Furthermore, the challenge of how to study small subpopulations with rare genetic lesions in an already rare disease will have to be considered. In all, while questions and challenges remain, precisely defining the genomic landscape of AML, holds great promise for ultimately leading to improved outcomes for affected patients.
Collapse
Affiliation(s)
- Shannon E Conneely
- Division of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates Avenue, Feigin Tower, Suite 1025, Houston, TX, 77030, USA
| | - Rachel E Rau
- Division of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates Avenue, Feigin Tower, Suite 1025, Houston, TX, 77030, USA.
| |
Collapse
|
69
|
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
|
70
|
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
|
71
|
Liu M, Ju X, Zou J, Shi J, Jia G. Recent researches for dual Aurora target inhibitors in antitumor field. Eur J Med Chem 2020; 203:112498. [PMID: 32693295 DOI: 10.1016/j.ejmech.2020.112498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/28/2020] [Indexed: 11/17/2022]
Abstract
Non-infectious and chronic diseases such as malignant tumors are now one of the main causes of human death. Its occurrence is a multi-factor, multi-step complex process with biological characteristics such as cell differentiation, abnormal proliferation, uncontrolled growth, and metastasis. It has been found that a variety of human malignant tumors are accompanied by over-expression and proliferation of Aurora kinase, which causes abnormalities in the mitotic process and is related to the instability of the genome that causes tumors. Therefore, the use of Aurora kinase inhibitors to target tumors is becoming a research hotspot. However, in cancer, because of the complexity of signal transduction system and the participation of different proteins and enzymes, the anticancer effect of selective single-target drugs is limited. After inhibiting one pathway, signal molecules can be conducted through other pathways, resulting in poor therapeutic effect of single-target drug treatment. Multi-target drugs can solve this problem very well. It can regulate the various links that cause disease at the same time without completely eliminating the relationship between the signal transmission systems, and it is not easy to cause drug resistance. Currently, studies have shown that Aurora dual-target inhibitors generated with the co-inhibition of Aurora and another target (such as CDK, PLK, JAK2, etc.) have better therapeutic effects on tumors. In this paper, we reviewed the studies of dual Aurora inhibitors that have been discovered in recent years.
Collapse
Affiliation(s)
- Maoyu Liu
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xueming Ju
- Department of Ultrasound, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jing Zou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
72
|
Khongsti S, Shunyu BN, Ghosh S. Promoter-associated DNA methylation & expression profiling of genes ( FLT 3, EPB41L3 & SFN) in patients with oral squamous cell carcinoma in the Khasi & Jaintia population of Meghalaya, India. Indian J Med Res 2020; 150:584-591. [PMID: 32048621 PMCID: PMC7038811 DOI: 10.4103/ijmr.ijmr_620_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background & objectives: Oral squamous cell carcinoma is one of the most lethal forms of cancer, and its aetiology has been attributed to both genetic and epigenetic factors working in liaison to contribute to the disease. Epigenetic changes especially DNA methylation is involved in the activation or repression of gene functions. The aim of this study was to investigate the DNA methylation pattern and expression profiling of the promoter regions of FMS-related tyrosine kinase 3 (FLT3), erythrocyte membrane protein band 4.1-like 3 (EPB41L3) and stratifin (SFN) genes in oral cancer within the Khasi and Jaintia tribal population of Meghalaya in North East India. Methods: Quantitative methylation analyses of the selected genes were carried out by MassARRAY platform System, and the relative expression profiling was carried out by real-time polymerase chain reaction. Results: Quantitative methylation results indicated that the level of methylation was significantly higher (hypermethylated) for FLT3 and EPB41L3 and significantly lower (hypomethylated) for SFN in tumour tissues as compared to the adjacent paired normal tissue. Expression profiling was in concurrence with the methylation data whereby hypermethylated genes showed low mRNA level and vice versa for the hypomethylated gene. Interpretation & conclusions: The findings show that hyper- and hypomethylation of the selected genes play a potential role in oral carcinogenesis in the selected Khasi and Jaintia tribal population of Meghalaya. The methylation status of these genes has not been reported in oral cancer, so these genes may serve as promising biomarkers for oral cancer diagnosis as well as in disease monitoring.
Collapse
Affiliation(s)
- Shngainlang Khongsti
- Department of Zoology, North Eastern Hill University, Shillong, Meghalaya, India
| | - Brian Neizekhotuo Shunyu
- Department of Otorhinolaryngology, North East Indira Gandhi Regional Institute for Health & Medical Sciences, Shillong, Meghalaya, India
| | - Srimoyee Ghosh
- Department of Zoology, North Eastern Hill University, Shillong, Meghalaya, India
| |
Collapse
|
73
|
Li X, Jiang Y, Peterson YK, Xu T, Himes RA, Luo X, Yin G, Inks ES, Dolloff N, Halene S, Chan SSL, Chou CJ. Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity. J Med Chem 2020; 63:5501-5525. [PMID: 32321249 DOI: 10.1021/acs.jmedchem.0c00442] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Here, we present a new series of hydrazide-bearing class I selective HDAC inhibitors designed based on panobinostat. The cap, linker, and zinc-binding group were derivatized to improve HDAC affinity and antileukemia efficacy. Lead inhibitor 13a shows picomolar or low nanomolar IC50 values against HDAC1 and HDAC3 and exhibits differential toxicity profiles toward multiple cancer cells with different FLT3 and p53 statuses. 13a indirectly inhibits the FLT3 signaling pathway and down-regulates master antiapoptotic proteins, resulting in the activation of pro-caspase3 in wt-p53 FLT3-ITD MV4-11 cells. While in the wt-FLT3 and p53-null cells, 13a is incapable of causing apoptosis at a therapeutic concentration. The MDM2 antagonist and the proteasome inhibitor promote 13a-triggered apoptosis by preventing p53 degradation. Furthermore, we demonstrate that apoptosis rather than autophagy is the key contributing factor for 13a-triggered cell death. When compared to panobinostat, 13a is not mutagenic and displays superior in vivo bioavailability and a higher AUC0-inf value.
Collapse
Affiliation(s)
- Xiaoyang Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China.,Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Yuqi Jiang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Tongqiang Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| | - Richard A Himes
- Department of Chemistry and Biochemistry, College of Charleston, 66 George Street, Charleston, South Carolina 29424, United States
| | - Xin Luo
- Technology Center of Qingdao Customs, Qingdao, Shandong 266002, China
| | - Guilin Yin
- Technology Center of Qingdao Customs, Qingdao, Shandong 266002, China
| | - Elizabeth S Inks
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Nathan Dolloff
- Department of Cellular and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston SC29425, United States
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06511, United States
| | - Sherine S L Chan
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - C James Chou
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
74
|
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
|
75
|
Aikawa T, Togashi N, Iwanaga K, Okada H, Nishiya Y, Inoue S, Levis MJ, Isoyama T. Quizartinib, a selective FLT3 inhibitor, maintains antileukemic activity in preclinical models of RAS-mediated midostaurin-resistant acute myeloid leukemia cells. Oncotarget 2020; 11:943-955. [PMID: 32215183 PMCID: PMC7082118 DOI: 10.18632/oncotarget.27489] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/29/2020] [Indexed: 12/29/2022] Open
Abstract
FLT3 internal tandem duplication (ITD) mutations are associated with poor prognosis in patients with acute myeloid leukemia (AML). In this preclinical study, we characterized the binding affinity and selectivity of quizartinib, a small-molecule inhibitor of FLT3, and AC886, the active metabolite of quizartinib, compared with those of other FLT3 inhibitors. Selectivity profiling against >400 kinases showed that quizartinib and AC886 were highly selective against FLT3. Quizartinib and AC886 inhibited FLT3 signaling pathways in FLT3-ITD–mutated AML cells, leading to potent growth inhibition with IC50 values of <1 nM. When quizartinib was administered to mice bearing FLT3-ITD mutated tumors, AC886 was rapidly detected and tumor regression was observed at doses of ≥1 mg/kg without severe body weight loss. In addition, quizartinib inhibited the viability of midostaurin-resistant MOLM-14 cells and exerted potent antitumor activity in mouse xenograft models without severe body weight loss, while midostaurin and gilteritinib did not show significant antitumor effects. This is the first detailed characterization of quizartinib and AC886 in comparison with other FLT3 inhibitors under the same experimental conditions. Preclinical antileukemic activity in midostaurin-resistant FLT3-ITD–mutated AML cells suggests the potential value of quizartinib following midostaurin failure in patients with FLT3-ITD mutated AML.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mark J Levis
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, United States of America
| | | |
Collapse
|
76
|
circMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood 2020; 134:1533-1546. [PMID: 31387917 DOI: 10.1182/blood.2019000802] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/03/2019] [Indexed: 01/16/2023] Open
Abstract
Internal tandem duplication (ITD) mutations within FMS-like tyrosine kinase-3 (FLT3) occur in up to 30% of acute myeloid leukemia (AML) patients and confer a very poor prognosis. The oncogenic form of FLT3 is an important therapeutic target, and inhibitors specifically targeting FLT3 kinase can induce complete remission; however, relapse after remission has been observed due to acquired resistance with secondary mutations in FLT3, highlighting the need for new strategies to target FLT3-ITD mutations. Recent studies have reported that the aberrant formations of circular RNAs (circRNAs) are biological tumorigenesis-relevant mechanisms and potential therapeutic targets. Herein, we discovered a circRNA, circMYBL2, derived from the cell-cycle checkpoint gene MYBL2. circMYBL2 is more highly expressed in AML patients with FLT3-ITD mutations than in those without the FLT3-ITD mutation. We found that circMYBL2 knockdown specifically inhibits proliferation and promotes the differentiation of FLT3-ITD AML cells in vitro and in vivo. Interestingly, we found that circMYBL2 significantly influences the protein level of mutant FLT3 kinase, which contributes to the activation of FLT3-ITD-dependent signaling pathways. Mechanistically, circMYBL2 enhanced the translational efficiency of FLT3 kinase by increasing the binding of polypyrimidine tract-binding protein 1 (PTBP1) to FLT3 messenger RNA. Moreover, circMYBL2 knockdown impaired the cytoactivity of inhibitor-resistant FLT3-ITD+ cells, with a significant decrease in FLT3 kinase expression, followed by the inactivation of its downstream pathways. In summary, we are the first to reveal a circRNA that specifically influences FLT3-ITD AML and regulates FLT3 kinase levels through translational regulation, suggesting that circMYBL2 may be a potential therapeutic target for FLT3-ITD AML.
Collapse
|
77
|
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
|
78
|
All-trans retinoic acid exerts selective anti-FLT3-ITD acute myeloid leukemia efficacy through downregulating Chk1 kinase. Cancer Lett 2020; 473:130-138. [PMID: 31904486 DOI: 10.1016/j.canlet.2019.12.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/27/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
All-trans retinoic acid (ATRA) is known to be a potent inhibitor of FLT3-ITD acute myeloid leukemia (AML) cells, although the exact mechanism remains unclear. In this work, we report that ATRA causes fatal mitotic catastrophe in FLT3-ITD AML cells by degrading Chk1 kinase, and therefore preventing DNA damage repair. In order to explore a further enhancement in the inhibitory effect of ATRA on FLT3-ITD AML cells, we investigated the suitability of a combination of ATRA and DNA damage drug SN38. In vitro experiments showed that this combinatorial approach effectively inhibited the proliferation of FLT3-ITD cells and induced cell apoptosis in AML. In vivo experiments confirmed that the combination could substantially improve the anti-tumor effect of SN38. Taken together, our results indicate that ATRA down-regulates Chk1 in FLT3-ITD AML cells, and the combination of ATRA and SN38 significantly improves the anti-tumor effect of either ATRA or SN38 when used alone.
Collapse
|
79
|
Zhang L, Bu Z, Shen J, Shang L, Chen Y, Wang Y. A novel circular RNA (hsa_circ_0000370) increases cell viability and inhibits apoptosis of FLT3-ITD-positive acute myeloid leukemia cells by regulating miR-1299 and S100A7A. Biomed Pharmacother 2019; 122:109619. [PMID: 31919040 DOI: 10.1016/j.biopha.2019.109619] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/18/2019] [Accepted: 10/26/2019] [Indexed: 01/16/2023] Open
Abstract
FLT3-ITD+ acute myeloid leukemia (AML) is an important subtype of AML, accounting for approximately 25 % of all AML cases in the world. Recently, increasing evidence has shown that circular RNAs (circRNAs) can act as effective biomarkers of various human cancers. However, the roles of circRNAs in AML remain largely unclear. In the present study, circ_0000370 was found to be significantly increased in FLT3-ITD+ AML and was demonstrated to act as an oncogenic circRNA of AML in vitro. TargetScan results showed that miR-1299, miR-370-3p, miR-502-5p, miR-1281 and miR-640 were potential targets of circ_0000370, and miR-1299 had the broadest range of interactome compared with other microRNAs of interest. Moreover, we demonstrated that S100A7A was a target gene of miR-1299, and circ_0000370 could regulate S100A7A expression by sponging miR-1299 in AML cell lines. Therefore, we suggest that the promoting effects of circ_0000370 on the progression of FLT3-ITD+ AML might be relevant to the inhibition of miR-1299 and the upregulation of S100A7A.
Collapse
Affiliation(s)
- Lingyan Zhang
- Division of Hematology-oncology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, PR China.
| | - Zibin Bu
- Division of Hematology-oncology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Juan Shen
- Division of Hematology-oncology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Liping Shang
- Division of Hematology-oncology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Yuanyuan Chen
- Division of Hematology-oncology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| | - Yan Wang
- Division of Hematology-oncology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, PR China
| |
Collapse
|
80
|
Reinecke M, Heinzlmeir S, Wilhelm M, Médard G, Klaeger S, Kuster B. Kinobeads: A Chemical Proteomic Approach for Kinase Inhibitor Selectivity Profiling and Target Discovery. ACTA ACUST UNITED AC 2019. [DOI: 10.1002/9783527818242.ch4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
81
|
Azrakhsh NA, Mensah-Glanowska P, Sand K, Kittang AO. Targeting Immune Signaling Pathways in Clonal Hematopoiesis. Curr Med Chem 2019; 26:5262-5277. [PMID: 30907306 DOI: 10.2174/0929867326666190325100636] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/05/2019] [Accepted: 03/12/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myeloid neoplasms are a diverse group of malignant diseases with different entities and numerous patho-clinical features. They arise from mutated clones of hematopoietic stem- and progenitor cells which expand by outperforming their normal counterparts. The intracellular signaling profile of cancer cells is the sum of genetic, epigenetic and microenvironmental influences, and the multiple interconnections between different signaling pathways make pharmacological targeting complicated. OBJECTIVE To present an overview of known somatic mutations in myeloproliferative neoplasms (MPN), myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) and the inflammatory signaling pathways affected by them, as well as current efforts to therapeutically modulate this aberrant inflammatory signaling. METHODS In this review, we extensively reviewed and compiled salient information with ClinicalTrials.gov as our source on ongoing studies, and PubMed as our authentic bibliographic source, using a focused review question. RESULTS Mutations affecting immune signal transduction are present to varying extents in clonal myeloid diseases. While MPN are dominated by a few common mutations, a multitude of different genes can be mutated in MDS and AML. Mutations can also occur in asymptomatic persons, a finding called clonal hematopoiesis of indeterminate potential (CHIP). Mutations in FLT3, JAK, STAT, CBL and RAS can lead to aberrant immune signaling. Protein kinase inhibitors are entering the clinic and are extensively investigated in clinical trials in MPN, MDS and AML. CONCLUSION In summary, this article summarizes recent research on aberrant inflammatory signaling in clonal myeloid diseases and the clinical therapeutic potential of modulation of signal transduction and effector proteins in the affected pathways.
Collapse
Affiliation(s)
| | - Patrycja Mensah-Glanowska
- Department of Hematology, Jagiellonian University Medical College / University Hospital, Krakow, Poland
| | - Kristoffer Sand
- Clinic of Medicine and Rehabilitation, More and Romsdal Hospital Trust, Alesund, Norway
| | - Astrid Olsnes Kittang
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, Section for Hematology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
82
|
Baska F, Sipos A, Őrfi Z, Nemes Z, Dobos J, Szántai-Kis C, Szabó E, Szénási G, Dézsi L, Hamar P, Cserepes MT, Tóvári J, Garamvölgyi R, Krekó M, Őrfi L. Discovery and development of extreme selective inhibitors of the ITD and D835Y mutant FLT3 kinases. Eur J Med Chem 2019; 184:111710. [DOI: 10.1016/j.ejmech.2019.111710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/02/2019] [Accepted: 09/16/2019] [Indexed: 10/25/2022]
|
83
|
Wood FL, Shepherd S, Hayes A, Liu M, Grira K, Mok Y, Atrash B, Faisal A, Bavetsias V, Linardopoulos S, Blagg J, Raynaud FI. Metabolism of the dual FLT-3/Aurora kinase inhibitor CCT241736 in preclinical and human in vitro models: Implication for the choice of toxicology species. Eur J Pharm Sci 2019; 139:104899. [PMID: 30953752 PMCID: PMC6892276 DOI: 10.1016/j.ejps.2019.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/26/2019] [Accepted: 04/02/2019] [Indexed: 02/02/2023]
Abstract
CCT241736 is a dual fms-like tyrosine kinase 3 (FLT3)/Aurora kinase inhibitor in development for the treatment of acute myeloid leukaemia. The successful development of any new drug relies on adequate safety testing including preclinical toxicology studies. Selection of an appropriate preclinical species requires a thorough understanding of the compound's metabolic clearance and pathways, as well as other pharmacokinetic and pharmacodynamic considerations. In addition, elucidation of the metabolising enzymes in human facilitates improved clinical prediction based on population pharmacokinetics and can inform drug-drug interaction studies. Intrinsic clearance (CLint) determination and metabolite profiling of CCT241736 in human and four preclinical species (dog, minipig, rat and mouse) was undertaken in cryopreserved hepatocytes and liver microsomes. Recombinant human cytochrome P450 bactosomes (rCYP) were utilised to provide reaction phenotyping data and support prediction of metabolic pathways. CCT241736 exhibited low CLint in both hepatocytes and liver microsomes of human, dog, minipig and rat, but considerably higher CLint in mouse. CYP3A4 and CYP3A5 were identified as the major enzymes responsible for biotransformation of CCT241736 in human, exclusively forming five out of seven metabolites. Minipig showed greatest similarity to human with regard to both overall metabolic profile and abundance of specific metabolites relative to parent compound, and is therefore proposed as the most appropriate toxicological species. The greatest disparity was observed between human and dog. Based on metabolic profile, either mouse or rat is a suitable rodent species for toxicology studies.
Collapse
Affiliation(s)
- Francesca L Wood
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Sam Shepherd
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Angela Hayes
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Manjuan Liu
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Katia Grira
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Yi Mok
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Butrus Atrash
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Amir Faisal
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Vassilios Bavetsias
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Spiros Linardopoulos
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Florence I Raynaud
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
84
|
Bazarbachi AH, Al Hamed R, Malard F, Mohty M, Bazarbachi A. Allogeneic transplant for FLT3- ITD mutated AML: a focus on FLT3 inhibitors before, during, and after transplant. Ther Adv Hematol 2019; 10:2040620719882666. [PMID: 31700594 PMCID: PMC6826920 DOI: 10.1177/2040620719882666] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/25/2019] [Indexed: 01/17/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations are one of the most frequently encountered genetic alterations in acute myeloid leukemia (AML), and are generally associated with unfavorable outcomes. Several tools are currently available to provide an accurate prognosis for patients with these mutations, including FLT3 mutation type (internal tandem duplication versus tyrosine kinase domain), mutation allelic ratio (high versus low), and concurrent nucleophosmin-1 (NPM1) mutation, to help decide on optimal treatment. Recent advances in targeted therapies have paved the way for modern treatment strategies, such as the development of FLT3 kinase inhibitors. These novel drugs can be incorporated into any treatment component, including induction and consolidation, the relapse/refractory setting, bridging for transplant, salvage post-transplant, and as prophylactic long-term post-transplant maintenance. Many challenges remain though, such as their intolerability with high-dose chemotherapy in frail patients; whether their optimal use involves watchful waiting for molecular or hematologic relapse compared with prophylactic use as maintenance; and the exact role and indication for allogeneic stem cell transplantation, which arguably remains the only curative option for these high-risk patients.
Collapse
Affiliation(s)
- Abdul Hamid Bazarbachi
- Department of Haematology, Saint Antoine Hospital, Paris, France, INSERM UMR 938, Paris, France
- Sorbonne University, Paris, Île-de-France, France
| | - Rama Al Hamed
- Department of Haematology, Saint Antoine Hospital, Paris, France, INSERM UMR 938, Paris, France
- Sorbonne University, Paris, Île-de-France, France
| | - Florent Malard
- Department of Haematology, Saint Antoine Hospital, Paris, France, INSERM UMR 938, Paris, France
- Sorbonne University, Paris, Île-de-France, France
| | - Mohamad Mohty
- Department of Haematology, Saint Antoine Hospital, Paris, France, INSERM UMR 938, Paris, France
- Sorbonne University, Paris, Île-de-France, France
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, PO Box 113-6044, Beirut, Lebanon
| |
Collapse
|
85
|
Han Y, Sengupta S, Lee BJ, Cho H, Kim J, Choi HG, Dash U, Kim JH, Kim ND, Kim JH, Sim T. Identification of a Unique Resorcylic Acid Lactone Derivative That Targets Both Lymphangiogenesis and Angiogenesis. J Med Chem 2019; 62:9141-9160. [PMID: 31513411 DOI: 10.1021/acs.jmedchem.9b01025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We synthesized 11 novel L-783277 derivatives, in which a structure rigidifying phenyl ring is incorporated into the 14-membered chiral resorcylic acid lactone system. The SAR study with these substances demonstrated that 17 possesses excellent kinase selectivity against a panel of 335 kinases in contrast to L-783277 and inhibits VEGFR3, VEGFR2, and FLT3 with single-digit nanomolar IC50 values. Also, we found that 21, a stereoisomer of 17, has excellent potency (IC50 = 9 nM) against VEGFR3 and selectivity over VEGFR2 and FLT3. 17, a potent dual VEGFR3 and VEGFR2 inhibitor, effectively suppresses both lymphangiogenesis and angiogenesis in a 3D-microfluidic tumor lymphangiogenesis assay and in vivo corneal assay while SAR131675 blocks only lymphangiogenesis. In addition, 17 blocks the endothelial tube formation and suppresses proliferation of PHE tumor vascular model. 17 will be a valuable templatefor developing therapeutically active and selective substances that target both lymphangiogenesis and angiogenesis.
Collapse
Affiliation(s)
- Youngsun Han
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro, Seongbuk-gu , Seoul 02841 , Republic of Korea
| | - Sandip Sengupta
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Byung Joo Lee
- Fight Against Angiogenesis-Related Blindness Laboratory, Clinical Research Institute , Seoul National University Hospital , 101 Daehak-ro, Jongno-gu , Seoul 110-744 , Republic of Korea.,Department of Biomedical Sciences, College of Medicine , Seoul National University , 103 Daehakro, Jongro-gu , Seoul 110-744 , Republic of Korea
| | - Hanna Cho
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro, Seongbuk-gu , Seoul 02841 , Republic of Korea
| | - Jiknyeo Kim
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Hwan Geun Choi
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Uttam Dash
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Jin Hyoung Kim
- Fight Against Angiogenesis-Related Blindness Laboratory, Clinical Research Institute , Seoul National University Hospital , 101 Daehak-ro, Jongno-gu , Seoul 110-744 , Republic of Korea
| | - Nam Doo Kim
- NDBio Therapeutics Inc. , 32 Songdogwahak-ro, Yeonsu-gu , Incheon 21984 , Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness Laboratory, Clinical Research Institute , Seoul National University Hospital , 101 Daehak-ro, Jongno-gu , Seoul 110-744 , Republic of Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro, Seongbuk-gu , Seoul 02841 , Republic of Korea.,Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| |
Collapse
|
86
|
Abbas HA, Alfayez M, Kadia T, Ravandi-Kashani F, Daver N. Midostaurin In Acute Myeloid Leukemia: An Evidence-Based Review And Patient Selection. Cancer Manag Res 2019; 11:8817-8828. [PMID: 31632141 PMCID: PMC6782026 DOI: 10.2147/cmar.s177894] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/12/2019] [Indexed: 01/08/2023] Open
Abstract
Fms-related-tyrosine kinase 3 (FLT3) mutations occur in approximately a third of acute myeloid leukemia (AML) patients and confer an adverse prognosis. Numerous studies have evaluated FLT3 targeting as single agent and in combination approaches in frontline and relapsed AML. At this time, midostaurin, a multikinase inhibitor, is the only FLT3-inhibitor that is US FDA approved to be used in combination with induction therapy in the frontline FLT3-mutated AML setting based on improved overall survival noted in the RATIFY Phase III trial. The utility of midostaurin in maintenance post stem cell transplantation has shown promising results and further studies are still ongoing. In this review, we discuss the studies that led to the inception of midostaurin as a targeted kinase inhibitor, its evaluation in AML, the early clinical trials and the large Phase III clinical trial that led to its eventual US FDA-approval in FLT3-mutated AML. Our review also discusses data on midostaurin adverse effects, mechanisms of resistance and limitations of its utility. We further discuss emerging second-generation FLT3 inhibitors, with a focus on quizartinib and gilteritinib and future directions to enhance FLT3-inhibitor efficacy and overcome mechanisms of resistance.
Collapse
Affiliation(s)
- Hussein A Abbas
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Mansour Alfayez
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi-Kashani
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
87
|
Pratz KW, Rudek MA, Smith BD, Karp J, Gojo I, Dezern A, Jones RJ, Greer J, Gocke C, Baer MR, Duong VH, Rosner G, Zahurak M, Wright JJ, Emadi A, Levis M. A Prospective Study of Peritransplant Sorafenib for Patients with FLT3-ITD Acute Myeloid Leukemia Undergoing Allogeneic Transplantation. Biol Blood Marrow Transplant 2019; 26:300-306. [PMID: 31550496 DOI: 10.1016/j.bbmt.2019.09.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 12/23/2022]
Abstract
FLT3-ITD-mutated acute myeloid leukemia (AML) remains a therapeutic challenge. FLT3 inhibition in the setting of minimal residual disease and a new immune system via allogeneic transplantation offers a promise of improved survival for these patients. We performed a prospective study of patients with FLT3-ITD AML undergoing allogeneic transplant that was conducted to evaluate the safety, tolerability, and outcome of sorafenib administered peritransplant. Sorafenib dosing was individualized, starting at 200 mg twice a day (BID), and titrated based on tolerability or toxicities until a tolerable dose was identified. Forty-four patients, with a median age of 52 years, undergoing allogeneic transplant were started on sorafenib in the peritransplant period (21 pretransplant). The median duration of post-transplant follow-up was 27.6 months (range, 5.2 to 60.4). Overall survival was 76% at both 24 and 36 months. Event-free survival at 24 and 36 months was 74% and 64%, respectively. Ten patients died in the post-transplant period, with 6 deaths due to relapsed leukemia and 4 from transplant-associated toxicity. Tolerable doses ranged from 200 mg every other day to 400 mg BID with similar exposure. Correlative studies evaluating FLT3 inhibition via a plasma inhibitory activity assay showed consistent inhibition of FLT3 at all tolerability-determined dosing levels. Sorafenib is well tolerated in the peritransplant setting irrespective of the conditioning intensity or the donor source. Our findings indicate that sorafenib dosing can be individualized in the post-transplantation setting according to patient tolerability. This approach results in effective in vivo FLT3 inhibition and yields encouraging survival results.
Collapse
Affiliation(s)
- Keith W Pratz
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.
| | - Michelle A Rudek
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - B Douglas Smith
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Judith Karp
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Ivana Gojo
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Amy Dezern
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Richard J Jones
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Jackie Greer
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Christopher Gocke
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Maria R Baer
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Vu H Duong
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Gary Rosner
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Marianna Zahurak
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - John J Wright
- IDB/CTEP/NCI, National Cancer Institute, Rockville, Maryland
| | - Ashkan Emadi
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Mark Levis
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | | |
Collapse
|
88
|
Yalniz F, Abou Dalle I, Kantarjian H, Borthakur G, Kadia T, Patel K, Loghavi S, Garcia‐Manero G, Sasaki K, Daver N, DiNardo C, Pemmaraju N, Short NJ, Yilmaz M, Bose P, Naqvi K, Pierce S, Nogueras González GM, Konopleva M, Andreeff M, Cortes J, Ravandi F. Prognostic significance of baseline FLT3-ITD mutant allele level in acute myeloid leukemia treated with intensive chemotherapy with/without sorafenib. Am J Hematol 2019; 94:984-991. [PMID: 31237017 DOI: 10.1002/ajh.25553] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/01/2019] [Accepted: 06/06/2019] [Indexed: 11/11/2022]
Abstract
Internal tandem duplication (ITD) of the fms-related tyrosine kinase-3 gene (FLT3) confer a poor prognosis in adult AML. Studies have reported that a higher mutant allelic burden is associated with a worse prognosis. Adult patients with FLT3-ITD mutated AML treated at our institution were identified. Patients were assigned into 2 groups; patients who received idarubicin and cytarabine (IA, group one) containing induction, and who received sorafenib in addition to IA containing regimens at induction (group two). The optimal FLT3-ITD mutant allele cut-off was defined as the cut-off to divide the whole cohort with the highest statistical significance. A total of 183 patients including 104 (57%) in group one and 79 (43%) in group two were identified. The complete remission (CR)/CR with incomplete hematologic recovery (CRi) for group one and group two were 85% and 99%, respectively (P = .004). The median relapse free survival (RFS) for group one and two were 12 and 45 months, respectively (P = .02). The median overall survival (mOS) was 17 months in group one, and has not been reached in group two (P = .008). The optimal FLT3-ITD mutant allele cut-off for OS was 6.9% in group one, there was no optimal cut-off in group two. On multivariate analysis, poor performance status (PS) (P = .003), sorafenib (P = .01), and presenting white blood cells (WBC) (P < .001) were independent predictors of OS. Higher FLT3-ITD allele burden is associated with a worse outcome in patients treated with IA-based chemotherapy. Addition of sorafenib to chemotherapy not only nullifies the negative prognostic impact of higher allele burden, but also improves outcome of FLT3-ITD mutated AML patients regardless of the allele burden.
Collapse
Affiliation(s)
- Fevzi Yalniz
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Iman Abou Dalle
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Hagop Kantarjian
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Gautam Borthakur
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Tapan Kadia
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Keyur Patel
- Department of PathologyThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Sanam Loghavi
- Department of PathologyThe University of Texas MD Anderson Cancer Center Houston Texas
| | | | - Koji Sasaki
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Naval Daver
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Courtney DiNardo
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Naveen Pemmaraju
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Nicholas J. Short
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Musa Yilmaz
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Prithviraj Bose
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Kiran Naqvi
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Sherry Pierce
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | | | - Marina Konopleva
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Michael Andreeff
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Jorge Cortes
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| | - Farhad Ravandi
- Department of LeukemiaThe University of Texas MD Anderson Cancer Center Houston Texas
| |
Collapse
|
89
|
Ma J, Zhao S, Qiao X, Knight T, Edwards H, Polin L, Kushner J, Dzinic SH, White K, Wang G, Zhao L, Lin H, Wang Y, Taub JW, Ge Y. Inhibition of Bcl-2 Synergistically Enhances the Antileukemic Activity of Midostaurin and Gilteritinib in Preclinical Models of FLT3-Mutated Acute Myeloid Leukemia. Clin Cancer Res 2019; 25:6815-6826. [PMID: 31320594 DOI: 10.1158/1078-0432.ccr-19-0832] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/24/2019] [Accepted: 07/09/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the efficacy of the combination of the FLT3 inhibitors midostaurin or gilteritinib with the Bcl-2 inhibitor venetoclax in FLT3-internal tandem duplication (ITD) acute myeloid leukemia (AML) and the underlying molecular mechanism. EXPERIMENTAL DESIGN Using both FLT3-ITD cell lines and primary patient samples, Annexin V-FITC/propidium iodide staining and flow cytometry analysis were used to quantify cell death induced by midostaurin or gilteritinib, alone or in combination with venetoclax. Western blot analysis was performed to assess changes in protein expression levels of members of the JAK/STAT, MAPK/ERK, and PI3K/AKT pathways, and members of the Bcl-2 family of proteins. The MV4-11-derived xenograft mouse model was used to assess in vivo efficacy of the combination of gilteritinib and venetoclax. Lentiviral overexpression of Mcl-1 was used to confirm its role in cell death induced by midostaurin or gilteritinib with venetoclax. Changes of Mcl-1 transcript levels were assessed by RT-PCR. RESULTS The combination of midostaurin or gilteritinib with venetoclax potently and synergistically induces apoptosis in FLT3-ITD AML cell lines and primary patient samples. The FLT3 inhibitors induced downregulation of Mcl-1, enhancing venetoclax activity. Phosphorylated-ERK expression is induced by venetoclax but abolished by the combination of venetoclax with midostaurin or gilteritinib. Simultaneous downregulation of Mcl-1 by midostaurin or gilteritinib and inhibition of Bcl-2 by venetoclax results in "free" Bim, leading to synergistic induction of apoptosis. In vivo results show that gilteritinib in combination with venetoclax has therapeutic potential. CONCLUSIONS Inhibition of Bcl-2 via venetoclax synergistically enhances the efficacy of midostaurin and gilteritinib in FLT3-mutated AML.See related commentary by Perl, p. 6567.
Collapse
Affiliation(s)
- Jun Ma
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Shoujing Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xinan Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tristan Knight
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, Michigan.,Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Lisa Polin
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Juiwanna Kushner
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Sijana H Dzinic
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Kathryn White
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, P.R.China
| | - Hai Lin
- Department of Hematology and Oncology, The First Hospital of Jilin University, Changchun, P.R. China
| | - Yue Wang
- Department of Pediatric Hematology and Oncology, The First Hospital of Jilin University, Changchun, P.R. China
| | - Jeffrey W Taub
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, Michigan.,Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan
| | - Yubin Ge
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan. .,Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
90
|
Papayannidis C, Sartor C, Marconi G, Fontana MC, Nanni J, Cristiano G, Parisi S, Paolini S, Curti A. Acute Myeloid Leukemia Mutations: Therapeutic Implications. Int J Mol Sci 2019; 20:ijms20112721. [PMID: 31163594 PMCID: PMC6600275 DOI: 10.3390/ijms20112721] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 01/25/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is an extremely heterogeneous group of hematological neoplasms, for which allogeneic stem cell transplantation (HSCT) still represents the only potentially curative option in the majority of cases. However, elderly age and clinically severe comorbidities may often exclude a wide amount of patients from this therapeutic approach, underlying the urgent need for alternative strategies. Thanks to the introduction of advanced high-throughput techniques, light is being shed on the pathogenesis of AML, identifying molecular recurrent mutations as responsible for the onset, as well as progression, of disease. As a consequence, and in parallel, many new compounds, including targeted therapies (FMS-like tyrosine kinase 3 (FLT3) and Isocitrate dehydrogenase 1-2 (IDH1-2) inhibitors), have found a wide room of application in this setting, and are now available in daily practice, or in late phases of clinical development. Moreover, several further innovative molecules are currently under investigation, and promising results for many of them have already been reported. In this review, we will present an update on the most relevant molecular alterations of AML, focusing on the most frequent genomic mutations of the disease, for which compounds have been approved or are still currently under investigation.
Collapse
Affiliation(s)
- Cristina Papayannidis
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Chiara Sartor
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Giovanni Marconi
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Maria Chiara Fontana
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Jacopo Nanni
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Gianluca Cristiano
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Sarah Parisi
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Stefania Paolini
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Antonio Curti
- Istituto di Ematologia e Oncologia Medica "L. e A. Seràgnoli", S.Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| |
Collapse
|
91
|
Affiliation(s)
- Kiran Naqvi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
92
|
Wu M, Li L, Hamaker M, Small D, Duffield AS. FLT3-ITD cooperates with Rac1 to modulate the sensitivity of leukemic cells to chemotherapeutic agents via regulation of DNA repair pathways. Haematologica 2019; 104:2418-2428. [PMID: 30975911 PMCID: PMC6959181 DOI: 10.3324/haematol.2018.208843] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 04/09/2019] [Indexed: 01/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic neoplasm, and patients with an internal tandem duplication (ITD) mutation of the FMS-like tyrosine kinase-3 (FLT3) receptor gene have a poor prognosis. FLT3-ITD interacts with DOCK2, a G effector protein that activates Rac1/2. Previously, we showed that knockdown of DOCK2 leads to decreased survival of FLT3-ITD leukemic cells. We further investigated the mechanisms by which Rac1/DOCK2 activity affects cell survival and chemotherapeutic response in FLT3-ITD leukemic cells. Exogenous expression of FLT3-ITD led to increased Rac1 activity, reactive oxygen species, phosphorylated STAT5, DNA damage response factors and cytarabine resistance. Conversely, DOCK2 knockdown resulted in a decrease in these factors. Consistent with the reduction in DNA damage response factors, FLT3-ITD cells with DOCK2 knockdown exhibited significantly increased sensitivity to DNA damage response inhibitors. Moreover, in a mouse model of FLT3-ITD AML, animals treated with the CHK1 inhibitor MK8776 + cytarabine survived longer than those treated with cytarabine alone. These findings suggest that FLT3-ITD and Rac1 activity cooperatively modulate DNA repair activity, the addition of DNA damage response inhibitors to conventional chemotherapy may be useful in the treatment of FLT3-ITD AML, and inhibition of the Rac signaling pathways via DOCK2 may provide a novel and promising therapeutic target for FLT3-ITD AML.
Collapse
Affiliation(s)
| | - Li Li
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | | - Donald Small
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | | |
Collapse
|
93
|
Zhou F, Ge Z, Chen B. Quizartinib (AC220): a promising option for acute myeloid leukemia. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1117-1125. [PMID: 31114157 PMCID: PMC6497874 DOI: 10.2147/dddt.s198950] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/04/2019] [Indexed: 12/27/2022]
Abstract
Quizartinib is an effective therapy for patients with FLT3-ITD acute myeloid leukemia (AML) by continuing to inhibit the activity of FLT3 gene, leading to apoptosis of tumor cells. Multiple clinical trials have proved that it is effective in relapsed or refractory AML with an FLT3-ITD mutation. In this review, we focus on the characteristics of FLT3/ITD mutations, the mechanism and pharmacokinetics of quizartinib, and the mechanisms of resistance to quizartinib. We also summarize clinical experiences and adverse effects with quizartinib and recommend crucial approaches of quizartinib in the therapy of patients with newly diagnosed AML and patients with relapsed/refractory AML, particularly those with FLT3-ITD mutation. Quizartinib presents its advantages as a very promising agent in the treatment of AML, especially in patients with FLT3-ITD mutations. FLT3/ITD mutation can lead to constitutive autophosphorylation of FLT3 and activation of its downstream effectors including RAS/RAF/MEK, MAPK/ERK, PI3K/AKT/mTOR and JAK/STAT5 signal pathways, while Quizartinib can inhibit these downstream pathways through specific FLT3 inhibition. Quizartinib has received US Food and Drug Administration breakthrough therapy designation in patients with relapsed/refractory FLT3-ITD AML based on clinical trials. A larger sample of clinical trials are needed to verify its safety and efficacy, and the efficacy of quizartinib combined with chemotherapy or allogeneic hematopoietic cell transplantation should also be estimated in clinical trials. Meanwhile, for the side effects of quizartinib, further studies are needed to find a way to reduce its toxicity.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Zheng Ge
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| |
Collapse
|
94
|
Bazarbachi A, Labopin M, Battipaglia G, Djabali A, Forcade E, Arcese W, Socié G, Blaise D, Halter J, Gerull S, Cornelissen JJ, Chevallier P, Maertens J, Schaap N, El-Cheikh J, Esteve J, Nagler A, Mohty M. Allogeneic Stem Cell Transplantation for FLT3-Mutated Acute Myeloid Leukemia: In vivo T-Cell Depletion and Posttransplant Sorafenib Maintenance Improve Survival. A Retrospective Acute Leukemia Working Party-European Society for Blood and Marrow Transplant Study. Clin Hematol Int 2019; 1:58-74. [PMID: 34595412 PMCID: PMC8432385 DOI: 10.2991/chi.d.190310.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 02/20/2019] [Indexed: 12/23/2022] Open
Abstract
Acute myeloid leukemia (AML) with FLT3-mutation carries a poor prognosis, and allogeneic stem cell transplantation (allo-SCT) is recommended at first complete remission (CR1). We assessed 462 adults (median age 50 years) with FLT3-mutated AML allografted between 2010 and 2015 from a matched related (40%), unrelated (49%), or haploidentical donor (11%). The median follow-up of alive patients was 39 months. Day-100 acute graft versus host disease (GVHD) grades II–IV and III–IV were encountered in 26% and 9%, whereas the 2-year incidence of chronic and extensive chronic GVHD were 34% and 16%, respectively. The 2-year incidences of relapse and nonrelapse mortality were 34% and 15%, respectively. The 2-year leukemia-free survival, overall survival (OS), and GVHD relapse-free survival (GRFS) were 51%, 59%, and 38%, respectively. In multivariate analysis, NPM1-mutation, transplantation in CR1, in vivo T-cell depletion, and posttransplant sorafenib improved OS, whereas more than one induction (late CR1) negatively affected OS. Similarly, NPM1-mutation, a haploidentical donor, T-cell depletion, and sorafenib maintenance improved GRFS, whereas late CR1 or persistent disease negatively affected it. In conclusion, FLT3-mutated AML remains a challenge even following allo-SCT. In vivo T-cell depletion and posttransplant sorafenib significantly improve OS and GRFS, and may be considered as standard of care.
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
| | | | - Giorgia Battipaglia
- Department of hematology and cellular therapy Hopital Saint Antoine, Paris, France.,Department of hematology and cellular therapy, Hopital Saint Antoine, Université Pierre & Marie Curie, INSERM, UMRs 938, Paris, France
| | | | - Edouard Forcade
- Department of Hematology, CHU Bordeaux Hôpital Haut-leveque, Pessac, France
| | - William Arcese
- Department of Stem cell transplant, Tor Vergata University of Rome, Rome, Italy
| | - Gerard Socié
- Department of Hematology-bone marrow transplant, Hopital Saint Louis, Paris, France
| | - Didier Blaise
- Department of Hematology, programme de Transplantation & Therapie Cellulaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, Marseille, France
| | - Joerg Halter
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | - Sabine Gerull
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | - Jan J Cornelissen
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - Johan Maertens
- Department of Hematology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Nicolaas Schaap
- Department of Hematology, Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Jean El-Cheikh
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jordi Esteve
- Hematology Department, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - Arnon Nagler
- Department of Hematology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Mohamad Mohty
- Department of hematology and cellular therapy Hopital Saint Antoine, Paris, France.,Department of hematology and cellular therapy, Hopital Saint Antoine, Université Pierre & Marie Curie, INSERM, UMRs 938, Paris, France
| |
Collapse
|
95
|
Windisch R, Pirschtat N, Kellner C, Chen-Wichmann L, Lausen J, Humpe A, Krause DS, Wichmann C. Oncogenic Deregulation of Cell Adhesion Molecules in Leukemia. Cancers (Basel) 2019; 11:E311. [PMID: 30841639 PMCID: PMC6468598 DOI: 10.3390/cancers11030311] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 01/01/2023] Open
Abstract
Numerous cell⁻cell and cell⁻matrix interactions within the bone marrow microenvironment enable the controlled lifelong self-renewal and progeny of hematopoietic stem and progenitor cells (HSPCs). On the cellular level, this highly mutual interaction is granted by cell adhesion molecules (CAMs) integrating differentiation, proliferation, and pro-survival signals from the surrounding microenvironment to the inner cell. However, cell⁻cell and cell⁻matrix interactions are also critically involved during malignant transformation of hematopoietic stem/progenitor cells. It has become increasingly apparent that leukemia-associated gene products, such as activated tyrosine kinases and fusion proteins resulting from chromosomal translocations, directly regulate the activation status of adhesion molecules, thereby directing the leukemic phenotype. These observations imply that interference with adhesion molecule function represents a promising treatment strategy to target pre-leukemic and leukemic lesions within the bone marrow niche. Focusing on myeloid leukemia, we provide a current overview of the mechanisms by which leukemogenic gene products hijack control of cellular adhesion to subsequently disturb normal hematopoiesis and promote leukemia development.
Collapse
Affiliation(s)
- Roland Windisch
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Nina Pirschtat
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Christian Kellner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Linping Chen-Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Jörn Lausen
- Institute for Transfusion Medicine and Immunohematology, Johann-Wolfgang-Goethe University and German Red Cross Blood Service, 60528 Frankfurt am Main, Germany.
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Daniela S Krause
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt am Main, Germany.
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
96
|
Ali EMH, Abdel-Maksoud MS, Oh CH. Thieno[2,3-d]pyrimidine as a promising scaffold in medicinal chemistry: Recent advances. Bioorg Med Chem 2019; 27:1159-1194. [PMID: 30826188 DOI: 10.1016/j.bmc.2019.02.044] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 02/16/2019] [Accepted: 02/20/2019] [Indexed: 12/20/2022]
Abstract
Thienopyrimidine scaffold is a fused heterocyclic ring system that structurally can be considered as adenine, the purine base that is found in both DNA and RNA-bioisosteres. Thienopyrimidines exist in three distinct isomeric forms. The current review discusses thieno[2,3-d]pyrimidine as a one of the opulent heterocycles in drug discovery. Its broad range of medical applications such as anticancer, anti-inflammatory, anti-microbial, and CNS protective agents has inspired us to study its structure-activity relationship (SAR), along with its relevant synthetic strategies. The present review briefly summarizes synthetic approaches for the preparation of thieno[2,3-d]pyrimidine derivatives. In addition, the promising biological activities of this scaffold are also illustrated with explanatory diagrams for their SAR studies.
Collapse
Affiliation(s)
- Eslam M H Ali
- Center for Biomaterials, Korea Institute of Science & Technology (KIST), Seoul, Seongbuk-gu 02792, Republic of Korea; Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Yuseong-gu 34113, Republic of Korea
| | - Mohammed S Abdel-Maksoud
- Medicinal & Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), Dokki, Giza 12622, Egypt
| | - Chang-Hyun Oh
- Center for Biomaterials, Korea Institute of Science & Technology (KIST), Seoul, Seongbuk-gu 02792, Republic of Korea; Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Yuseong-gu 34113, Republic of Korea.
| |
Collapse
|
97
|
Hofmann S, Schubert ML, Wang L, He B, Neuber B, Dreger P, Müller-Tidow C, Schmitt M. Chimeric Antigen Receptor (CAR) T Cell Therapy in Acute Myeloid Leukemia (AML). J Clin Med 2019; 8:jcm8020200. [PMID: 30736352 PMCID: PMC6406805 DOI: 10.3390/jcm8020200] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/02/2019] [Accepted: 02/03/2019] [Indexed: 02/07/2023] Open
Abstract
Despite high response rates after initial chemotherapy in patients with acute myeloid leukemia (AML), relapses occur frequently, resulting in a five-year-survival by <30% of the patients. Hitherto, allogeneic hemotopoietic stem cell transplantation (allo-HSCT) is the best curative treatment option in intermediate and high risk AML. It is the proof-of-concept for T cell-based immunotherapies in AML based on the graft-versus-leukemia (GvL)-effect, but it also bears the risk of graft-versus-host disease. CD19-targeting therapies employing chimeric antigen receptor (CAR) T cells are a breakthrough in cancer therapy. A similar approach for myeloid malignancies is highly desirable. This article gives an overview on the state-of-the art of preclinical and clinical studies on suitable target antigens for CAR T cell therapy in AML patients.
Collapse
Affiliation(s)
- Susanne Hofmann
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Maria-Luisa Schubert
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Lei Wang
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Bailin He
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Brigitte Neuber
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Peter Dreger
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany.
| | - Carsten Müller-Tidow
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany.
| | - Michael Schmitt
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), University Hospital Heidelberg, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany.
| |
Collapse
|
98
|
Zhang H, Savage S, Schultz AR, Bottomly D, White L, Segerdell E, Wilmot B, McWeeney SK, Eide CA, Nechiporuk T, Carlos A, Henson R, Lin C, Searles R, Ho H, Lam YL, Sweat R, Follit C, Jain V, Lind E, Borthakur G, Garcia-Manero G, Ravandi F, Kantarjian HM, Cortes J, Collins R, Buelow DR, Baker SD, Druker BJ, Tyner JW. Clinical resistance to crenolanib in acute myeloid leukemia due to diverse molecular mechanisms. Nat Commun 2019; 10:244. [PMID: 30651561 PMCID: PMC6335421 DOI: 10.1038/s41467-018-08263-x] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
FLT3 mutations are prevalent in AML patients and confer poor prognosis. Crenolanib, a potent type I pan-FLT3 inhibitor, is effective against both internal tandem duplications and resistance-conferring tyrosine kinase domain mutations. While crenolanib monotherapy has demonstrated clinical benefit in heavily pretreated relapsed/refractory AML patients, responses are transient and relapse eventually occurs. Here, to investigate the mechanisms of crenolanib resistance, we perform whole exome sequencing of AML patient samples before and after crenolanib treatment. Unlike other FLT3 inhibitors, crenolanib does not induce FLT3 secondary mutations, and mutations of the FLT3 gatekeeper residue are infrequent. Instead, mutations of NRAS and IDH2 arise, mostly as FLT3-independent subclones, while TET2 and IDH1 predominantly co-occur with FLT3-mutant clones and are enriched in crenolanib poor-responders. The remaining patients exhibit post-crenolanib expansion of mutations associated with epigenetic regulators, transcription factors, and cohesion factors, suggesting diverse genetic/epigenetic mechanisms of crenolanib resistance. Drug combinations in experimental models restore crenolanib sensitivity.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Benzimidazoles/pharmacology
- Benzimidazoles/therapeutic use
- Cell Line, Tumor
- Drug Resistance, Neoplasm/genetics
- Drug Screening Assays, Antitumor
- Epigenesis, Genetic/drug effects
- Female
- GTP Phosphohydrolases/genetics
- HEK293 Cells
- Humans
- Inhibitory Concentration 50
- Isocitrate Dehydrogenase/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Male
- Membrane Proteins/genetics
- Mice
- Middle Aged
- Mutation/drug effects
- Mutation/genetics
- Piperidines/pharmacology
- Piperidines/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Pyridones/pharmacology
- Pyridones/therapeutic use
- Pyrimidinones/pharmacology
- Pyrimidinones/therapeutic use
- Tandem Repeat Sequences/genetics
- Treatment Outcome
- Exome Sequencing
- fms-Like Tyrosine Kinase 3/genetics
Collapse
Affiliation(s)
- Haijiao Zhang
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Samantha Savage
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Anna Reister Schultz
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Daniel Bottomly
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Libbey White
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Erik Segerdell
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Beth Wilmot
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Shannon K McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Christopher A Eide
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
- Howard Hughes Medical Institute, Portland, 97239, OR, USA
| | - Tamilla Nechiporuk
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Amy Carlos
- Integrated Genomics, Knight Cancer Institute, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Rachel Henson
- Integrated Genomics, Knight Cancer Institute, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Chenwei Lin
- Integrated Genomics, Knight Cancer Institute, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Robert Searles
- Integrated Genomics, Knight Cancer Institute, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Hoang Ho
- AROG Pharmaceuticals, Dallas, 75240, TX, USA
| | | | | | | | - Vinay Jain
- AROG Pharmaceuticals, Dallas, 75240, TX, USA
| | - Evan Lind
- Molecular Microbiology & Immunology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
| | - Gautam Borthakur
- The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | | | - Farhad Ravandi
- The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | - Hagop M Kantarjian
- The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | - Jorge Cortes
- The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | - Robert Collins
- University of Texas Southwestern Medical Center, Dallas, 75390, TX, USA
| | - Daelynn R Buelow
- The Ohio State University College of Pharmacy and Comprehensive Cancer Center, Columbus, 43210, OH, USA
| | - Sharyn D Baker
- The Ohio State University College of Pharmacy and Comprehensive Cancer Center, Columbus, 43210, OH, USA
| | - Brian J Druker
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA
- Howard Hughes Medical Institute, Portland, 97239, OR, USA
| | - Jeffrey W Tyner
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA.
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, 97239, OR, USA.
| |
Collapse
|
99
|
Novel Agents for Acute Myeloid Leukemia. Cancers (Basel) 2018; 10:cancers10110429. [PMID: 30423907 PMCID: PMC6267447 DOI: 10.3390/cancers10110429] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/05/2018] [Accepted: 11/06/2018] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematological disease characterized by genetic and clinical heterogeneity. Recent advances in the understanding of AML pathogenesis have paved the way for the development of new agents targeting specific molecules or mechanisms that contribute to finally move beyond the current standard of care, which is "3 + 7" regimen. In particular, new therapeutic options such as targeted therapies (midostaurin and enasidenib), monoclonal antibodies (gemtuzumab ozogamicin), and a novel liposomal formulation of cytarabine and daunorubicin (CPX-351) have been recently approved, and will be soon available for the treatment of adult patients with AML. In this review, we will present and describe these recently approved drugs as well as selected novel agents against AML that are currently under investigation, and show the most promising results as monotherapy or in combination with chemotherapy. The selection of these emerging treatments is based on the authors' opinion.
Collapse
|
100
|
Lee-Sherick AB, Jacobsen KM, Henry CJ, Huey MG, Parker RE, Page LS, Hill AA, Wang X, Frye SV, Earp HS, Jordan CT, DeRyckere D, Graham DK. MERTK inhibition alters the PD-1 axis and promotes anti-leukemia immunity. JCI Insight 2018; 3:97941. [PMID: 30385715 PMCID: PMC6238750 DOI: 10.1172/jci.insight.97941] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 09/19/2018] [Indexed: 01/22/2023] Open
Abstract
MERTK is ectopically expressed and promotes survival in acute lymphoblastic leukemia (ALL) cells and is thus a potential therapeutic target. Here we demonstrate both direct therapeutic effects of MERTK inhibition on leukemia cells and induction of anti-leukemia immunity via suppression of the coinhibitory PD-1 axis. A MERTK-selective tyrosine kinase inhibitor, MRX-2843, mediated therapeutic anti-leukemia effects in immunocompromised mice bearing a MERTK-expressing human leukemia xenograft. In addition, inhibition of host MERTK by genetic deletion (Mertk-/- mice) or treatment with MRX-2843 significantly decreased tumor burden and prolonged survival in immune-competent mice inoculated with a MERTK-negative ALL, suggesting immune-mediated therapeutic activity. In this context, MERTK inhibition led to significant decreases in expression of the coinhibitory ligands PD-L1 and PD-L2 on CD11b+ monocytes/macrophages in the leukemia microenvironment. Furthermore, although T cells do not express MERTK, inhibition of MERTK indirectly decreased PD-1 expression on CD4+ and CD8+ T cells and decreased the incidence of splenic FOXP3+ Tregs at sites of leukemic infiltration, leading to increased T cell activation. These data demonstrate direct and immune-mediated therapeutic activities in response to MERTK inhibition in ALL models and provide validation of a translational agent targeting MERTK for modulation of tumor immunity.
Collapse
Affiliation(s)
| | - Kristen M. Jacobsen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Curtis J. Henry
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Madeline G. Huey
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Rebecca E. Parker
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | | | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy
| | - Stephen V. Frye
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy
- Lineberger Comprehensive Cancer Center, and
| | - H. Shelton Earp
- Lineberger Comprehensive Cancer Center, and
- Departments of Medicine and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Craig T. Jordan
- Division of Hematology, University of Colorado, Aurora, Colorado, USA
| | - Deborah DeRyckere
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Douglas K. Graham
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|