1
|
Kazemi M, Naghdi Sadeh R, Shekari Khaniani M, Rezazadeh M, Derakhshan SM, Ghafouri-Fard S. Identification of RN7SK LncRNA as a novel biomarker in Alzheimer's disease using bioinformatics and expression analysis. Sci Rep 2024; 14:31192. [PMID: 39732800 DOI: 10.1038/s41598-024-82490-9] [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: 08/06/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Alzheimer's disease (AD) is a degenerative illness that accounts for the common type of dementia among adults over the age of 65. Despite extensive studies on the pathogenesis of the disease, early diagnosis of AD is still debatable. In this research, we performed bioinformatics approaches on the AD-related E-MTAB 6094 dataset to uncover new potential biomarkers for AD diagnosis. To achieve this, we performed in-depth in silico assays, including differentially expressed genes analysis, weighted gene co-expression network analyses, module-trait association analyses, gene ontology and pathway enrichment analyses, and hub genes network analyses. Finally, the expression of the identified candidate genes was evaluated in AD patients PBMC samples by qRT-PCR. Through computational analyses, we found that RN7SK LncRNA and its co-expressed genes of TNF, TNFAIP3, CCLT3, and FLT3 are from key genes in AD development that are associated with inflammatory responses. Our experimental validation revealed that RN7SK LncRNA and TNF were substantially up-regulated in AD samples (P = 0.006 and P = 0.023, respectively). Whereas, TNFAIP3 expression was significantly decreased (P = 0.016). However, the expression of CCL3 and FLT3 did not differ significantly between two groups (P = 0.396 and P = 0.521, respectively). In conclusion, in this study a novel LncRNA associated with AD pathogenesis were identified, which may provide new diagnostic biomarker for AD.
Collapse
Affiliation(s)
- Masoumeh Kazemi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Naghdi Sadeh
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahmoud Shekari Khaniani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Maryam Rezazadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Sima Mansoori Derakhshan
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Zhai J, Li C, Sun B, Wang S, Cui Y, Gao Q, Sang F. Sunitinib-based Proteolysis Targeting Chimeras (PROTACs) reduced the protein levels of FLT-3 and c-KIT in leukemia cell lines. Bioorg Med Chem Lett 2022; 78:129041. [DOI: 10.1016/j.bmcl.2022.129041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/15/2022] [Accepted: 10/27/2022] [Indexed: 11/08/2022]
|
3
|
Chen X, Huang Y, Xu W, Cai Y, Yang Y. 4-Aminopyrazolopyrimidine scaffold and its deformation in the design of tyrosine and serine/threonine kinase inhibitors in medicinal chemistry. RSC Med Chem 2022; 13:1008-1028. [DOI: 10.1039/d2md00139j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 11/21/2022] Open
Abstract
Deformation of the 4-aminopyrazolopyrimidine scaffold in designing small-molecule inhibitors.
Collapse
Affiliation(s)
- Xiaolu Chen
- Department of Pharmacy, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei 434020, China
| | - Yajiao Huang
- Department of Pharmacy, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei 434020, China
| | - Wanghan Xu
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311202, Zhejiang, P. R. China
| | - Yuepiao Cai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuanrong Yang
- Department of Pharmacy, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei 434020, China
| |
Collapse
|
4
|
Rehman A, Akram AM, Chaudhary A, Sheikh N, Hussain Z, Alsanie WF, Rehman RA, Hameed N, Saleem T, Zafar A, Absar M, Iqbal Z, Alhazmi A, Baeshen HA, Mohammedsaleh ZM, Qamer S, Sayed S, Gaber A. RUNX1 mutation and elevated FLT3 gene expression cooperates to induce inferior prognosis in cytogenetically normal acute myeloid leukemia patients. Saudi J Biol Sci 2021; 28:4845-4851. [PMID: 34466057 PMCID: PMC8381075 DOI: 10.1016/j.sjbs.2021.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 11/25/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a bone marrow malignancy having multiple molecular pathways driving its progress. In recent years, the main causes of AML considered all over the world are genetic variations in cancerous cells. The RUNX1 and FLT3 genes are necessary for the normal hematopoiesis and differentiation process of hematopoietic stem cells into mature blood cells, therefore they are the most common targets for point mutations resulting in AML. Methods We screened 32 CN-AML patients for FLT3-ITD (by Allele-specific PCR) and RUNX1 mutations (by Sanger sequencing). The FLT3 mRNA expression was assessed in all AML patients and its subgroups. Results Eight patients (25%) carried RUNX1 mutation (K83E) while three patients (9.37%) were found to have internal tandem duplications in FLT3 gene. The RUNX1 mutation data were correlated with clinical parameters and FLT3 gene expression profile. The RUNX1 mutations were observed to be significantly prevalent in older males. Moreover, RUNX1 and FLT3-mutated patients had lower complete remission rate, event-free survival rate, and lower overall survival rate than patients with wild-type RUNX1 and FLT3 gene. The RUNX1 and FLT3 mutant patients with up-regulated FLT3 gene expression showed even worse prognosis. Bradford Assay showed that protein concentration was down-regulated in RUNX1 and FLT3 mutants in comparison to RUNX1 and FLT3 wild-type groups. Conclusion This study constitutes the first report from Pakistan reporting significant molecular mutation analysis of RUNX1 and FLT3 genes including FLT3 expression evaluation with follow-up. This provides an insight that aforementioned mutations are markers of poor prognosis but the study with a large AML cohort will be useful to further investigate their role in disease biology of AML.
Collapse
Affiliation(s)
- Atia Rehman
- Cell and Molecular Biology Lab (CMBL), Department of Zoology, University of the Punjab, Lahore, Pakistan.,Molecular Biology Lab, University of Education, Township Campus, College Road, Lahore, Pakistan
| | - Afia Muhammad Akram
- Molecular Biology Lab, University of Education, Township Campus, College Road, Lahore, Pakistan
| | - Asma Chaudhary
- Molecular Biology Lab, University of Education, Township Campus, College Road, Lahore, Pakistan
| | - Nadeem Sheikh
- Cell and Molecular Biology Lab (CMBL), Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Zawar Hussain
- Molecular Biology Lab, University of Education, Township Campus, College Road, Lahore, Pakistan
| | - Walaa F Alsanie
- Center of Biomedical Sciences Research (CBSR), Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.,Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Rahat Abdul Rehman
- Department of Forensic Science, University of Health Sciences, Lahore, Pakistan
| | - Naila Hameed
- Cell and Molecular Biology Lab (CMBL), Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Tayyaba Saleem
- Cell and Molecular Biology Lab (CMBL), Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Amjad Zafar
- Department of Oncology, Mayo Hospital, Anarkali Bazar, Lahore, Pakistan
| | - Muhammad Absar
- Microbiology, Pathology & Laboratory Medicine, King Abdulaziz Hospital, Ministry of National Guard-Health Affairs, Saudi Arabia
| | - Zafar Iqbal
- Cancer and Medical Genetics, CAMS-A, King Saud Bin Abdulaziz University for Health Sciences & King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Al Ahsa, Saudi Arabia
| | - Alaa Alhazmi
- Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia.,SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Hosam Ali Baeshen
- Department of Orthodontics, Faculty of Dentistry, King Abdulaziz University, P.O. Box 80209, Jeddah 21589, Saudi Arabia
| | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Samina Qamer
- Department of Zoology, Government College University, Faisalabad, Pakistan
| | - Samy Sayed
- Department of Science and Technology, University College-Ranyah, Taif University, B.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmed Gaber
- Center of Biomedical Sciences Research (CBSR), Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.,Department of Biology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
5
|
Almatani MF, Ali A, Onyemaechi S, Zhao Y, Gutierrez L, Vaikari VP, Alachkar H. Strategies targeting FLT3 beyond the kinase inhibitors. Pharmacol Ther 2021; 225:107844. [PMID: 33811956 PMCID: PMC11490306 DOI: 10.1016/j.pharmthera.2021.107844] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022]
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by clonal expansion and differentiation arrest of the myeloid progenitor cells, which leads to the accumulation of immature cells called blasts in the bone marrow and peripheral blood. Mutations in the receptor tyrosine kinase FLT3 occur in 30% of normal karyotype patients with AML and are associated with a higher incidence of relapse and worse survival. Targeted therapies against FLT3 mutations using small-molecule FLT3 tyrosine kinase inhibitors (TKIs) have long been investigated, with some showing favorable clinical outcomes. However, major setbacks such as limited clinical efficacy and the high risk of acquired resistance remain unresolved. FLT3 signaling, mutations, and FLT3 inhibitors are topics that have been extensively reviewed in recent years. Strategies to target FLT3 beyond the small molecule kinase inhibitors are expanding, nevertheless they are not receiving enough attention. These modalities include antibody-based FLT3 targeted therapies, immune cells mediated targeting strategies, and approaches targeting downstream signaling pathways and FLT3 translation. Here, we review the most recent advances and the challenges associated with the development of therapeutic modalities targeting FLT3 beyond the kinase inhibitors.
Collapse
Affiliation(s)
- Mohammed F Almatani
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Atham Ali
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Sandra Onyemaechi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Yang Zhao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Lucas Gutierrez
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Vijaya Pooja Vaikari
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Houda Alachkar
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States; USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, United States.
| |
Collapse
|
6
|
Reddy JS, Chen CM, Coumar MS, Sun HY, Sun N, Hsieh HP. Development of a Robust Scale-Up Synthetic Route for BPR1K871: A Clinical Candidate for the Treatment of Acute Myeloid Leukemia and Solid Tumors. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.0c00515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Julakanti Satyanarayana Reddy
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
| | - Chih-Ming Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
| | - Mohane Selvaraj Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India
| | - Hsu-Yi Sun
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
| | - Na Sun
- China Gateway Pharmaceutical Development Co, Shanghai 201417, China
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
- Biomedical Translation Research Centre, Academia Sinica, Taipei 11520, Taiwan, ROC
| |
Collapse
|
7
|
Wang A, Hu C, Chen C, Liang X, Wang B, Zou F, Yu K, Li F, Liu Q, Qi Z, Wang J, Wang W, Wang L, Weisberg EL, Wang W, Li L, Ge J, Xia R, Liu J, Liu Q. Selectively targeting FLT3-ITD mutants over FLT3-wt by a novel inhibitor for acute myeloid leukemia. Haematologica 2021; 106:605-609. [PMID: 32299905 PMCID: PMC7849575 DOI: 10.3324/haematol.2019.244186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/09/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lili Li
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University
| | - Jian Ge
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University
| | - Ruixiang Xia
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University
| | | | | |
Collapse
|
8
|
Takahashi S. Mutations of FLT3 receptor affect its surface glycosylation, intracellular localization, and downstream signaling. Leuk Res Rep 2019; 13:100187. [PMID: 31853441 PMCID: PMC6911968 DOI: 10.1016/j.lrr.2019.100187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/29/2019] [Accepted: 11/23/2019] [Indexed: 11/29/2022] Open
Abstract
This review describes the effects of FLT3 mutations that alter its intracellular localization and modify its glycosylation, leading to differences in downstream signaling pathways. The most common type of FLT3 mutation, internal tandem duplication (FLT3-ITD), leads to localization in the endoplasmic reticulum and constitutive strong activation of STAT5. In contrast, the ligand-activated FLT3-wild type is mainly expressed on the cell surface and activates MAP kinases. Based on these backgrounds, several reports have demonstrated that glycosylation inhibitors are effective for inhibition of FLT3-ITD expression and intracellular localization. The general subcellular localization regulatory mechanisms for receptor tyrosine kinases are also discussed.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- Division of Laboratory Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| |
Collapse
|
9
|
Mat Yusoff Y, Abu Seman Z, Othman N, Kamaluddin NR, Esa E, Zulkiply NA, Abdullah J, Zakaria Z. Identification of FLT3 and NPM1 Mutations in Patients with Acute Myeloid Leukaemia. Asian Pac J Cancer Prev 2019; 20:1749-1755. [PMID: 31244296 PMCID: PMC7021611 DOI: 10.31557/apjcp.2019.20.6.1749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Indexed: 11/25/2022] Open
Abstract
Objective: The most frequent acquired molecular abnormalities and important prognostic indicators in patients with Acute Myeloid Leukaemia (AML) are fms-like tyrosine kinase-3 gene (FLT3) and nucleophosmin-1 (NPM1) mutations. Our study aims to develop a cost effective and comprehensive in-house conventional PCR method for detection of FLT3-ITD, FLT3-D835 and NPM1 mutations and to evaluate the frequency of these mutations in patients with cytogenetically normal (CN) AML in our population. Methods: A total of 199 samples from AML patients (95 women, 104 men) were included in the study. Mutation analyses were performed using polymerase chain reaction (PCR) and gene sequencing. Result: Sixty-eight patients were positive for the mutations. FLT3-ITD mutations were detected in 32 patients (16.1%), followed by FLT3-D835 in 5 (2.5%) and NPM1 in 54 (27.1%). Double mutations of NPM1 and FLT3-ITD were detected in 23 cases (11.6%). Assays validation were performed using Sanger sequencing and showed 100% concordance with in house method. Conclusion: The optimized in-house PCR assays for the detection of FLT3-ITD, FLT3-D835 and NPM1 mutations in AML patients were robust, less labour intensive and cost effective. These assays can be used as diagnostic tools for mutation detection in AML patients since identification of these mutations are important for prognostication and optimization of patient care.
Collapse
Affiliation(s)
- Yuslina Mat Yusoff
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Zahidah Abu Seman
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Norodiyah Othman
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Nor Rizan Kamaluddin
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Ezalia Esa
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Nor Amalina Zulkiply
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Julia Abdullah
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Zubaidah Zakaria
- Haematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| |
Collapse
|
10
|
Gokhale P, Chauhan APS, Arora A, Khandekar N, Nayarisseri A, Singh SK. FLT3 inhibitor design using molecular docking based virtual screening for acute myeloid leukemia. Bioinformation 2019; 15:104-115. [PMID: 31435156 PMCID: PMC6677903 DOI: 10.6026/97320630015104] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/10/2019] [Accepted: 02/19/2019] [Indexed: 02/08/2023] Open
Abstract
Acute Myeloid Leukaemia (AML) is a blood cancer, which affects the red blood cells in the bone marrow. Of the possible proteins that are affected in AML, fms-like tyrosine kinase 3 (FLT3) has long been recognized as a potential therapeutic target as it affects the other signaling pathways and leads to a cascade of events. First-generation inhibitors sorafenib and midostaurin, as well as secondgeneration agents such as quizartinib and crenolanib are known. It is of interest to identify new compounds against FLT3 with improved activity using molecular docking and virtual screening. Molecular docking of existing inhibitors selected a top scoring bestestablished candidate Quizartinib having PubChem CID: 24889392. Similarity searching resulted in compound XGIQBUNWFCCMASUHFFFAOYSA-NPubChemCID: 44598530 which shows higher affinity scores. A comparative study of both the compounds using a drug-drug comparison, ADMET studies, boiled egg plot and pharmacophore parameters and properties confirmed the result and predicted the ligand to be an efficient inhibitor of FLT3.
Collapse
Affiliation(s)
- Padmini Gokhale
- In silico Research Laboratory,Eminent Biosciences,Mahalakshmi Nagar,Indore-452010,Madhya Pradesh,India
| | | | - Anushka Arora
- In silico Research Laboratory,Eminent Biosciences,Mahalakshmi Nagar,Indore-452010,Madhya Pradesh,India
| | - Natasha Khandekar
- In silico Research Laboratory,Eminent Biosciences,Mahalakshmi Nagar,Indore-452010,Madhya Pradesh,India
| | - Anuraj Nayarisseri
- In silico Research Laboratory,Eminent Biosciences,Mahalakshmi Nagar,Indore-452010,Madhya Pradesh,India
- Bioinformatics Research Laboratory,LeGene Biosciences Pvt Ltd.,Mahalakshmi Nagar,Indore-452010,Madhya Pradesh,India
- Computer Aided Drug Designing and Molecular Modeling Lab,Department of Bioinformatics,Alagappa University,Karaikudi-630 003,Tamil Nadu,India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Designing and Molecular Modeling Lab,Department of Bioinformatics,Alagappa University,Karaikudi-630 003,Tamil Nadu,India
| |
Collapse
|
11
|
FLT3 receptor/CD135 expression by flow cytometry in acute myeloid leukemia: Relation to FLT3 gene mutations and mRNA transcripts. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2018. [DOI: 10.1016/j.ejmhg.2018.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
12
|
Hsu YC, Coumar MS, Wang WC, Shiao HY, Ke YY, Lin WH, Kuo CC, Chang CW, Kuo FM, Chen PY, Wang SY, Li AS, Chen CH, Kuo PC, Chen CP, Wu MH, Huang CL, Yen KJ, Chang YI, Hsu JTA, Chen CT, Yeh TK, Song JS, Shih C, Hsieh HP. Discovery of BPR1K871, a quinazoline based, multi-kinase inhibitor for the treatment of AML and solid tumors: Rational design, synthesis, in vitro and in vivo evaluation. Oncotarget 2018; 7:86239-86256. [PMID: 27863392 PMCID: PMC5349910 DOI: 10.18632/oncotarget.13369] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/07/2016] [Indexed: 12/15/2022] Open
Abstract
The design and synthesis of a quinazoline-based, multi-kinase inhibitor for the treatment of acute myeloid leukemia (AML) and other malignancies is reported. Based on the previously reported furanopyrimidine 3, quinazoline core containing lead 4 was synthesized and found to impart dual FLT3/AURKA inhibition (IC50 = 127/5 nM), as well as improved physicochemical properties. A detailed structure-activity relationship study of the lead 4 allowed FLT3 and AURKA inhibition to be finely tuned, resulting in AURKA selective (5 and 7; 100-fold selective over FLT3), FLT3 selective (13; 30-fold selective over AURKA) and dual FLT3/AURKA selective (BPR1K871; IC50 = 19/22 nM) agents. BPR1K871 showed potent anti-proliferative activities in MOLM-13 and MV4-11 AML cells (EC50 ∼ 5 nM). Moreover, kinase profiling and cell-line profiling revealed BPR1K871 to be a potential multi-kinase inhibitor. Functional studies using western blot and DNA content analysis in MV4-11 and HCT-116 cell lines revealed FLT3 and AURKA/B target modulation inside the cells. In vivo efficacy in AML xenograft models (MOLM-13 and MV4-11), as well as in solid tumor models (COLO205 and Mia-PaCa2), led to the selection of BPR1K871 as a preclinical development candidate for anti-cancer therapy. Further detailed studies could help to investigate the full potential of BPR1K871 as a multi-kinase inhibitor.
Collapse
Affiliation(s)
- Yung Chang Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Mohane Selvaraj Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry, India
| | - Wen-Chieh Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Hui-Yi Shiao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Yi-Yu Ke
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Wen-Hsing Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chun-Wei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Fu-Ming Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Pei-Yi Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Sing-Yi Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - An-Siou Li
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chun-Hwa Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Po-Chu Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Ming-Hsine Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Kuei-Jung Yen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Yun-I Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - John T-A Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC.,Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan, ROC
| |
Collapse
|
13
|
Dual inhibition of AKT/FLT3-ITD by A674563 overcomes FLT3 ligand-induced drug resistance in FLT3-ITD positive AML. Oncotarget 2018; 7:29131-42. [PMID: 27074558 PMCID: PMC5045383 DOI: 10.18632/oncotarget.8675] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/28/2016] [Indexed: 11/25/2022] Open
Abstract
The FLT3-ITD mutation is one of the most prevalent oncogenic mutations in AML. Several FLT3 kinase inhibitors have shown impressive activity in clinical evaluation, however clinical responses are usually transient and clinical effects are rapidly lost due to drug resistance. One of the resistance mechanisms in the AML refractory patients involves FLT3-ligand induced reactivation of AKT and/or ERK signaling via FLT3 wt kinase. Via a screen of numerous AKT kinase inhibitors, we identified the well-established orally available AKT inhibitor, A674563, as a dual suppressor of AKT and FLT3-ITD. A674563 suppressed FLT3-ITD positive AML both in vitro and in vivo. More importantly, compared to other FLT3 inhibitors, A674563 is able to overcome FLT3 ligand-induced drug resistance through simultaneous inhibition of FLT3-ITD- and AKT-mediated signaling. Our findings suggest that A674563 might be a potential drug candidate for overcoming FLT3 ligand-mediated drug resistance in FLT3-ITD positive AML.
Collapse
|
14
|
|
15
|
Li X, Wang A, Yu K, Qi Z, Chen C, Wang W, Hu C, Wu H, Wu J, Zhao Z, Liu J, Zou F, Wang L, Wang B, Wang W, Zhang S, Liu J, Liu Q. Discovery of (R)-1-(3-(4-Amino-3-(4-phenoxyphenyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)piperidin-1-yl)-2-(dimethylamino)ethanone (CHMFL-FLT3-122) as a Potent and Orally Available FLT3 Kinase Inhibitor for FLT3-ITD Positive Acute Myeloid Leukemia. J Med Chem 2015; 58:9625-38. [PMID: 26630553 DOI: 10.1021/acs.jmedchem.5b01611] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
FLT3-ITD mutant has been observed in about 30% of AML patients and extensively studied as a drug discovery target. On the basis of the structure of PCI-32765 (ibrutinib), a BTK kinase inhibitor that was recently reported to bear FLT3 kinase activity through a structure-guided drug design approach, we have discovered compound 18 (CHMFL-FLT3-122), which displayed an IC50 of 40 nM against FLT3 kinase and achieved selectivity over BTK kinase (over 10-fold). It significantly inhibited the proliferation of FLT3-ITD positive AML cancer cell lines MV4-11 (GI50 = 22 nM), MOLM13/14 (GI50 = 21 nM/42 nM). More importantly, 18 demonstrated 170-fold selectivity between FLT3 kinase and c-KIT kinase (GI50 = 11 nM versus 1900 nM) in the TEL-fusion isogenic BaF3 cells indicating a potential to avoid the FLT3/c-KIT dual inhibition induced myelosuppression toxicity. In the cellular context it strongly affected FLT3-ITD mediated signaling pathways and induced apoptosis by arresting the cell cycle into the G0/G1 phase. In the in vivo studies 18 demonstrated a good bioavailability (30%) and significantly suppressed the tumor growth in MV4-11 cell inoculated xenograft model (50 mg/kg) without exhibiting obvious toxicity. Compound 18 might be a potential drug candidate for FLT3-ITD positive AML.
Collapse
Affiliation(s)
- Xixiang Li
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Aoli Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,University of Science and Technology of China , Hefei 230036, Anhui, P. R. China
| | - Kailin Yu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,University of Science and Technology of China , Hefei 230036, Anhui, P. R. China
| | - Ziping Qi
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Cheng Chen
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Wenchao Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Chen Hu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Hong Wu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,University of Science and Technology of China , Hefei 230036, Anhui, P. R. China
| | - Jiaxin Wu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,University of Science and Technology of China , Hefei 230036, Anhui, P. R. China
| | - Zheng Zhao
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Juan Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Fengming Zou
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Li Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Beilei Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Wei Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Shanchun Zhang
- CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,Hefei Cosource Medicine Technology Co. Ltd., 358 Ganquan Road, Hefei 230031, Anhui, P. R. China
| | - Jing Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| | - Qingsong Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,CHMFL-HCMTC Target Therapy Joint Laboratory , 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China.,University of Science and Technology of China , Hefei 230036, Anhui, P. R. China.,Hefei Science Center, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei 230031, Anhui, P. R. China
| |
Collapse
|
16
|
Chiarini F, Lonetti A, Evangelisti C, Buontempo F, Orsini E, Evangelisti C, Cappellini A, Neri LM, McCubrey JA, Martelli AM. Advances in understanding the acute lymphoblastic leukemia bone marrow microenvironment: From biology to therapeutic targeting. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:449-463. [PMID: 26334291 DOI: 10.1016/j.bbamcr.2015.08.015] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/26/2015] [Accepted: 08/27/2015] [Indexed: 02/07/2023]
Abstract
The bone marrow (BM) microenvironment regulates the properties of healthy hematopoietic stem cells (HSCs) localized in specific niches. Two distinct microenvironmental niches have been identified in the BM, the "osteoblastic (endosteal)" and "vascular" niches. Nevertheless, these niches provide sanctuaries where subsets of leukemic cells escape chemotherapy-induced death and acquire a drug-resistant phenotype. Moreover, it is emerging that leukemia cells are able to remodel the BM niches into malignant niches which better support neoplastic cell survival and proliferation. This review focuses on the cellular and molecular biology of microenvironment/leukemia interactions in acute lymphoblastic leukemia (ALL) of both B- and T-cell lineage. We shall also highlight the emerging role of exosomes/microvesicles as efficient messengers for cell-to-cell communication in leukemia settings. Studies on the interactions between the BM microenvironment and ALL cells have led to the discovery of potential therapeutic targets which include cytokines/chemokines and their receptors, adhesion molecules, signal transduction pathways, and hypoxia-related proteins. The complex interplays between leukemic cells and BM microenvironment components provide a rationale for innovative, molecularly targeted therapies, designed to improve ALL patient outcome. A better understanding of the contribution of the BM microenvironment to the process of leukemogenesis and leukemia persistence after initial remission, may provide new targets that will allow destruction of leukemia cells without adversely affecting healthy HSCs. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis,Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
Affiliation(s)
- Francesca Chiarini
- Institute of Molecular Genetics, National Research Council, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Annalisa Lonetti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Camilla Evangelisti
- Institute of Molecular Genetics, National Research Council, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Francesca Buontempo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Ester Orsini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Cecilia Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Cappellini
- Department of Human Social and Health Sciences, University of Cassino, Cassino, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
17
|
Bertacchini J, Heidari N, Mediani L, Capitani S, Shahjahani M, Ahmadzadeh A, Saki N. Targeting PI3K/AKT/mTOR network for treatment of leukemia. Cell Mol Life Sci 2015; 72:2337-47. [PMID: 25712020 PMCID: PMC11113278 DOI: 10.1007/s00018-015-1867-5] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 02/10/2015] [Accepted: 02/16/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Increased activity of PI3K/AKT/mTOR pathway has been observed in a huge number of malignancies. This pathway can function as a prosurvival factor in leukemia stem cells and early committed leukemic precursors and its inhibition is regarded as a therapeutic approach. Accordingly, the aim of this review is to evaluate the PI3K/Akt/mTOR inhibitors used in leukemia models. DISCUSSION Inhibition of the PI3K/AKT/mTOR pathway has been reported to have beneficial therapeutic effects in leukemias, both in vitro in leukemia cell lines and in vivo in animal models. Overall, the use of dual PI3K/mTOR inhibitor, dual Akt/RTK inhibitor, Akt inhibitor, selective inhibitor of PI3K, mTOR inhibitor and dual PI3K/PDK1 inhibitor in CML, AML, APL, CLL, B-ALL and T-ALL has a better therapeutic effect than conventional treatments. CONCLUSIONS Targeting the PI3K/Akt/mTOR pathway may have pro-apoptotic and antiproliferative effects on hematological malignancies. Furthermore, modulation of miRNA can be used as a novel therapeutic approach to regulate the PI3K/Akt/mTOR pathway. However, both aspects require further clinical studies.
Collapse
Affiliation(s)
- Jessika Bertacchini
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Nazanin Heidari
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Laura Mediani
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- LTTA Center, University of Ferrara, Ferrara, Italy
| | - Mohammad Shahjahani
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Ahmadzadeh
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
18
|
Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, Kolb M. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45:1434-45. [PMID: 25745043 PMCID: PMC4416110 DOI: 10.1183/09031936.00174914] [Citation(s) in RCA: 667] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/05/2015] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disease characterised by fibrosis of the lung parenchyma and loss of lung function. Although the pathogenic pathways involved in IPF have not been fully elucidated, IPF is believed to be caused by repetitive alveolar epithelial cell injury and dysregulated repair, in which there is uncontrolled proliferation of lung fibroblasts and differentiation of fibroblasts into myofibroblasts, which excessively deposit extracellular matrix (ECM) proteins in the interstitial space. A number of profibrotic mediators including platelet-derived growth factor (PDGF), fibroblast growth factor (FGF) and transforming growth factor-β are believed to play important roles in the pathogenesis of IPF. Nintedanib is a potent small molecule inhibitor of the receptor tyrosine kinases PDGF receptor, FGF receptor and vascular endothelial growth factor receptor. Data from in vitro studies have shown that nintedanib interferes with processes active in fibrosis such as fibroblast proliferation, migration and differentiation, and the secretion of ECM. In addition, nintedanib has shown consistent anti-fibrotic and anti-inflammatory activity in animal models of lung fibrosis. These data provide a strong rationale for the clinical efficacy of nintedanib in patients with IPF, which has recently been demonstrated in phase III clinical trials. Nintedanib interferes with processes active in fibrosis, e.g. fibroblast proliferation, migration anddifferentiationhttp://ow.ly/Iae9z
Collapse
Affiliation(s)
- Lutz Wollin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Eva Wex
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Gisela Schnapp
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Susanne Stowasser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim am Rhein, Germany
| | | |
Collapse
|
19
|
Hirohata S, Yanagida T, Tomita T, Yoshikawa H. Enhanced expression of mRNA for FLT3 in bone marrow CD34+ cells in rheumatoid arthritis. Inflamm Regen 2015. [DOI: 10.2492/inflammregen.35.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Shunsei Hirohata
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tamiko Yanagida
- Department of Internal Medicine, Teikyo University School of Medicine, Itabashi, Tokyo, Japan
| | - Tetsuya Tomita
- Department of Orthopedic Surgery, Osaka University Medical School, Suita, Osaka, Japan
| | - Hideki Yoshikawa
- Department of Orthopedic Surgery, Osaka University Medical School, Suita, Osaka, Japan
| |
Collapse
|
20
|
Abstract
INTRODUCTION Approximately 23% of acute myeloid leukemia (AML) patients younger than 60 years of age carry a mutation in the transmembrane domain of the FMS-like tyrosine kinase-3 (FLT3) gene (FLT3/internal tandem duplications [ITD]). In normal karyotype AML, the presence of a FLT3/ITD mutation is associated with poor prognosis, as mirrored by a high risk of relapse even after allogeneic stem cell transplantation. The poor prognostic impact along with the observation that FLT3 is frequently overexpressed in the majority of AML cases has formed the platform for the development of FLT3-targeted strategies. To date, several FLT3 kinase inhibitors have been investigated in preclinical and clinical studies. However, as of yet, none of the studied FLT3 inhibitors has received FDA approval for routine clinical use in AML. This is in part due to the 'off target' effects observed with most inhibitors when administered at concentrations needed to achieve sustained levels of FLT3 inhibition, which are required to exhibit substantial cytotoxic effects against leukemic blasts. Furthermore, the development of resistance mutations has emerged as a clinical issue posing a threat to successful FLT3 inhibitor therapy. AREAS COVERED In this review, the authors provide a brief summary of FLT3 inhibitors investigated thus far, and discuss current treatment approaches and strategies how to best incorporate FLT3 tyrosine kinase inhibitors (TKIs) into therapy. EXPERT OPINION The combination of a FLT3 inhibitor with conventional chemotherapeutic regimens, epigenetic modifiers or inhibitors of FLT3 downstream and collateral effectors has emerged as a promising strategy to improve treatment outcome. The future of a tailored, molecular-based treatment approach for FLT3-mutated AML demands novel clinical trial concepts based on harmonized and aligned research goals between clinical and research centers and industry.
Collapse
Affiliation(s)
- Heiko Konig
- Johns Hopkins University, Medical Oncology , 1650 Orleans Street, Baltimore, MD , USA
| | | |
Collapse
|
21
|
FLT3 Internal Tandem Duplication and D835 Mutations in Patients with Acute Lymphoblastic Leukemia and its Clinical Significance. Mediterr J Hematol Infect Dis 2014; 6:e2014038. [PMID: 24959335 PMCID: PMC4063605 DOI: 10.4084/mjhid.2014.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 05/09/2014] [Indexed: 12/26/2022] Open
Abstract
The fms-like tyrosine kinase 3 (FLT3) gene is a member of the class III receptor tyrosine kinase family. Mutations of FLT3 were first described in 1997 and account for the most frequent molecular mutations in acute myeloid leukemia. Currently, there is no published data on FLT3 mutations in Saudi acute lymphoblastic leukemia (ALL) patients. In this retrospective study, we have examined a cohort of 77 ALL patients to determine the prevalence of FLT3 mutations and the possible prognostic relevance of these mutations in ALL patients. Correlations to other biologic factors such as karyotype, molecular mutations, and leukocyte count were also considered. FLT3 internal tandem duplication (ITD) mutations and point mutation in tyrosine kinase domain (D835) were analyzed in ALL patients, at diagnosis, by polymerase chain reaction (PCR). Two cases (2.6%, 2/77) were positive for FLT3 mutations; one was found to have FLT3/ITD and the other FLT3/D835. Our findings suggest that FLT3 mutations are not common in Saudi ALL and do not affect clinical outcome.
Collapse
|
22
|
Duran JM, Makarewich CA, Trappanese D, Gross P, Husain S, Dunn J, Lal H, Sharp TE, Starosta T, Vagnozzi RJ, Berretta RM, Barbe M, Yu D, Gao E, Kubo H, Force T, Houser SR. Sorafenib cardiotoxicity increases mortality after myocardial infarction. Circ Res 2014; 114:1700-1712. [PMID: 24718482 DOI: 10.1161/circresaha.114.303200] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
RATIONALE Sorafenib is an effective treatment for renal cell carcinoma, but recent clinical reports have documented its cardiotoxicity through an unknown mechanism. OBJECTIVE Determining the mechanism of sorafenib-mediated cardiotoxicity. METHODS AND RESULTS Mice treated with sorafenib or vehicle for 3 weeks underwent induced myocardial infarction (MI) after 1 week of treatment. Sorafenib markedly decreased 2-week survival relative to vehicle-treated controls, but echocardiography at 1 and 2 weeks post MI detected no differences in cardiac function. Sorafenib-treated hearts had significantly smaller diastolic and systolic volumes and reduced heart weights. High doses of sorafenib induced necrotic death of isolated myocytes in vitro, but lower doses did not induce myocyte death or affect inotropy. Histological analysis documented increased myocyte cross-sectional area despite smaller heart sizes after sorafenib treatment, further suggesting myocyte loss. Sorafenib caused apoptotic cell death of cardiac- and bone-derived c-kit+ stem cells in vitro and decreased the number of BrdU+ (5-bromo-2'-deoxyuridine+) myocytes detected at the infarct border zone in fixed tissues. Sorafenib had no effect on infarct size, fibrosis, or post-MI neovascularization. When sorafenib-treated animals received metoprolol treatment post MI, the sorafenib-induced increase in post-MI mortality was eliminated, cardiac function was improved, and myocyte loss was ameliorated. CONCLUSIONS Sorafenib cardiotoxicity results from myocyte necrosis rather than from any direct effect on myocyte function. Surviving myocytes undergo pathological hypertrophy. Inhibition of c-kit+ stem cell proliferation by inducing apoptosis exacerbates damage by decreasing endogenous cardiac repair. In the setting of MI, which also causes large-scale cell loss, sorafenib cardiotoxicity dramatically increases mortality.
Collapse
Affiliation(s)
- Jason M Duran
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | | | - Danielle Trappanese
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Polina Gross
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Sharmeen Husain
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Jonathan Dunn
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Hind Lal
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Thomas E Sharp
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Timothy Starosta
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Ronald J Vagnozzi
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Remus M Berretta
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Mary Barbe
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA
| | - Daohai Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Hajime Kubo
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Thomas Force
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Steven R Houser
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| |
Collapse
|
23
|
Chang Hsu Y, Ke YY, Shiao HY, Lee CC, Lin WH, Chen CH, Yen KJ, Hsu JTA, Chang C, Hsieh HP. Facile identification of dual FLT3-Aurora A inhibitors: a computer-guided drug design approach. ChemMedChem 2014; 9:953-61. [PMID: 24665000 DOI: 10.1002/cmdc.201300571] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Indexed: 12/22/2022]
Abstract
Computer-guided drug design is a powerful tool for drug discovery. Herein we disclose the use of this approach for the discovery of dual FMS-like receptor tyrosine kinase-3 (FLT3)-Aurora A inhibitors against cancer. An Aurora hit compound was selected as a starting point, from which 288 virtual molecules were screened. Subsequently, some of these were synthesized and evaluated for their capacity to inhibit FLT3 and Aurora kinase A. To further enhance FLT3 inhibition, structure-activity relationship studies of the lead compound were conducted through a simplification strategy and bioisosteric replacement, followed by the use of computer-guided drug design to prioritize molecules bearing a variety of different terminal groups in terms of favorable binding energy. Selected compounds were then synthesized, and their bioactivity was evaluated. Of these, one novel inhibitor was found to exhibit excellent inhibition of FLT3 and Aurora kinase A and exert a dramatic antiproliferative effect on MOLM-13 and MV4-11 cells, with an IC50 value of 7 nM. Accordingly, it is considered a highly promising candidate for further development.
Collapse
Affiliation(s)
- Yung Chang Hsu
- Institute of Biotechnology & Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 350, Taiwan (ROC)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Frequency and Prognostic Relevance of FLT3 Mutations in Saudi Acute Myeloid Leukemia Patients. Adv Hematol 2014; 2014:141360. [PMID: 24696688 PMCID: PMC3950551 DOI: 10.1155/2014/141360] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 12/21/2013] [Accepted: 01/12/2014] [Indexed: 01/30/2023] Open
Abstract
The Fms-like tyrosine kinase-3 (FLT3) is a receptor tyrosine kinase that plays a key role in cell survival, proliferation, and differentiation of hematopoietic stem cells. Mutations of FLT3 were first described in 1997 and account for the most frequent molecular mutations in acute myeloid leukemia (AML). AML patients with FLT3 internal tandem duplication (ITD) mutations have poor cure rates the prognostic significance of point mutations; tyrosine kinase domain (TKD) is still unclear. We analyzed the frequency of FLT3 mutations (ITD and D835) in patients with AML at diagnosis; no sufficient data currently exist regarding FLT3 mutations in Saudi AML patients. This study was aimed at evaluating the frequency of FLT3 mutations in patients with AML and its significance for prognosis. The frequency of FLT3 mutations in our study (18.56%) was lower than many of the reported studies, FLT3-ITD mutations were observed in 14.4%, and FLT3-TKD in 4.1%, of 97 newly diagnosed AML patients (82 adult and 15 pediatric). Our data show significant increase of FLT3 mutations in male more than female (13 male, 5 female). Our results support the view that FLT3-ITD mutation has strong prognostic factor in AML patients and is associated with high rate of relapse, and high leucocytes and blast count at diagnosis and relapse.
Collapse
|
25
|
Lin WH, Yeh TK, Jiaang WT, Yen KJ, Chen CH, Huang CT, Yen SC, Hsieh SY, Chou LH, Chen CP, Chiu CH, Kao LC, Chao YS, Chen CT, Hsu JTA. Evaluation of the antitumor effects of BPR1J-340, a potent and selective FLT3 inhibitor, alone or in combination with an HDAC inhibitor, vorinostat, in AML cancer. PLoS One 2014; 9:e83160. [PMID: 24416160 PMCID: PMC3885398 DOI: 10.1371/journal.pone.0083160] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/31/2013] [Indexed: 01/09/2023] Open
Abstract
Overexpression or/and activating mutation of FLT3 kinase play a major driving role in the pathogenesis of acute myeloid leukemia (AML). Hence, pharmacologic inhibitors of FLT3 are of therapeutic potential for AML treatment. In this study, BPR1J-340 was identified as a novel potent FLT3 inhibitor by biochemical kinase activity (IC50 approximately 25 nM) and cellular proliferation (GC50 approximately 5 nM) assays. BPR1J-340 inhibited the phosphorylation of FLT3 and STAT5 and triggered apoptosis in FLT3-ITD+ AML cells. The pharmacokinetic parameters of BPR1J-340 in rats were determined. BPR1J-340 also demonstrated pronounced tumor growth inhibition and regression in FLT3-ITD+ AML murine xenograft models. The combination treatment of the HDAC inhibitor vorinostat (SAHA) with BPR1J-340 synergistically induced apoptosis via Mcl-1 down-regulation in MOLM-13 AML cells, indicating that the combination of selective FLT3 kinase inhibitors and HDAC inhibitors could exhibit clinical benefit in AML therapy. Our results suggest that BPR1J-340 may be further developed in the preclinical and clinical studies as therapeutics in AML treatments.
Collapse
Affiliation(s)
- Wen-Hsing Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Weir-Torn Jiaang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kuei-Jung Yen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chun-Hwa Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chin-Ting Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Chieh Yen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shu-Yi Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Ling-Hui Chou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chun-Hsien Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Li-Chun Kao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Sheng Chao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
- * E-mail: (CTC); (JT-AH)
| | - John T.-A. Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- * E-mail: (CTC); (JT-AH)
| |
Collapse
|
26
|
Evangelisti C, Evangelisti C, Bressanin D, Buontempo F, Chiarini F, Lonetti A, Soncin M, Spartà A, McCubrey JA, Martelli AM. Targeting phosphatidylinositol 3-kinase signaling in acute myelogenous leukemia. Expert Opin Ther Targets 2013; 17:921-36. [DOI: 10.1517/14728222.2013.808333] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Martelli AM, Chiarini F, Evangelisti C, Cappellini A, Buontempo F, Bressanin D, Fini M, McCubrey JA. Two hits are better than one: targeting both phosphatidylinositol 3-kinase and mammalian target of rapamycin as a therapeutic strategy for acute leukemia treatment. Oncotarget 2012; 3:371-94. [PMID: 22564882 PMCID: PMC3380573 DOI: 10.18632/oncotarget.477] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) are two key components of the PI3K/Akt/mTOR signaling pathway. This signal transduction cascade regulates a wide range of physiological cell processes, that include differentiation, proliferation, apoptosis, autophagy, metabolism, motility, and exocytosis. However, constitutively active PI3K/Akt/mTOR signaling characterizes many types of tumors where it negatively influences response to therapeutic treatments. Hence, targeting PI3K/Akt/mTOR signaling with small molecule inhibitors may improve cancer patient outcome. The PI3K/Akt/mTOR signaling cascade is overactive in acute leukemias, where it correlates with enhanced drug-resistance and poor prognosis. The catalytic sites of PI3K and mTOR share a high degree of sequence homology. This feature has allowed the synthesis of ATP-competitive compounds targeting the catalytic site of both kinases. In preclinical models, dual PI3K/mTOR inhibitors displayed a much stronger cytotoxicity against acute leukemia cells than either PI3K inhibitors or allosteric mTOR inhibitors, such as rapamycin. At variance with rapamycin, dual PI3K/mTOR inhibitors targeted both mTOR complex 1 and mTOR complex 2, and inhibited the rapamycin-resistant phosphorylation of eukaryotic initiation factor 4E-binding protein 1, resulting in a marked inhibition of oncogenic protein translation. Therefore, they strongly reduced cell proliferation and induced an important apoptotic response. Here, we reviewed the evidence documenting that dual PI3K/mTOR inhibitors may represent a promising option for future targeted therapies of acute leukemia patients.
Collapse
Affiliation(s)
- Alberto M Martelli
- Department of Human Anatomy, University of Bologna, Cellular Signalling Laboratory, Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Pietschmann K, Bolck HA, Buchwald M, Spielberg S, Polzer H, Spiekermann K, Bug G, Heinzel T, Böhmer FD, Krämer OH. Breakdown of the FLT3-ITD/STAT5 axis and synergistic apoptosis induction by the histone deacetylase inhibitor panobinostat and FLT3-specific inhibitors. Mol Cancer Ther 2012; 11:2373-83. [PMID: 22942377 DOI: 10.1158/1535-7163.mct-12-0129] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activating mutations of the class III receptor tyrosine kinase FLT3 are the most frequent molecular aberration in acute myeloid leukemia (AML). Mutant FLT3 accelerates proliferation, suppresses apoptosis, and correlates with poor prognosis. Therefore, it is a promising therapeutic target. Here, we show that RNA interference against FLT3 with an internal tandem duplication (FLT3-ITD) potentiates the efficacy of the histone deacetylase inhibitor (HDACi) panobinostat (LBH589) against AML cells expressing FLT3-ITD. Similar to RNA interference, tyrosine kinase inhibitors (TKI; AC220/cpd.102/PKC412) in combination with LBH589 exhibit superior activity against AML cells. Median dose-effect analyses of drug-induced apoptosis rates of AML cells (MV4-11 and MOLM-13) revealed combination index (CI) values indicating strong synergism. AC220, the most potent and FLT3-specific TKI, shows highest synergism with LBH589 in the low nanomolar range. A 4-hour exposure to LBH589 + AC220 already generates more than 50% apoptosis after 24 hours. Different cell lines lacking FLT3-ITD as well as normal peripheral blood mononuclear cells are not significantly affected by LBH589 + TKI, showing the specificity of this treatment regimen. Immunoblot analyses show that LBH589 + TKI induce apoptosis via degradation of FLT3-ITD and its prosurvival target STAT5. Previously, we showed the LBH589-induced proteasomal degradation of FLT3-ITD. Here, we show that activated caspase-3 also contributes to the degradation of FLT3-ITD and that STAT5 is a direct target of this protease. Our data strongly emphasize HDACi/TKI drug combinations as promising modality for the treatment of FLT3-ITD-positive AMLs.
Collapse
Affiliation(s)
- Kristin Pietschmann
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich-Schiller University, Hans-Knöll-Str. 2, Jena 07745, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Grafone T, Palmisano M, Nicci C, Storti S. An overview on the role of FLT3-tyrosine kinase receptor in acute myeloid leukemia: biology and treatment. Oncol Rev 2012; 6:e8. [PMID: 25992210 PMCID: PMC4419636 DOI: 10.4081/oncol.2012.e8] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 03/06/2012] [Accepted: 04/13/2012] [Indexed: 01/10/2023] Open
Abstract
Hematopoiesis, the process by which the hematopoietic stem cells and progenitors differentiate into blood cells of various lineages, involves complex interactions of transcription factors that modulate the expression of downstream genes and mediate proliferation and differentiation signals. Despite the many controls that regulate hematopoiesis, mutations in the regulatory genes capable of promoting leukemogenesis may occur. The FLT3 gene encodes a tyrosine kinase receptor that plays a key role in controlling survival, proliferation and differentiation of hematopoietic cells. Mutations in this gene are critical in causing a deregulation of the delicate balance between cell proliferation and differentiation. In this review, we provide an update on the structure, synthesis and activation of the FLT3 receptor and the subsequent activation of multiple downstream signaling pathways. We also review activating FLT3 mutations that are frequently identified in acute myeloid leukemia, cause activation of more complex downstream signaling pathways and promote leukemogenesis. Finally, FLT3 has emerged as an important target for molecular therapy. We, therefore, report on some recent therapies directed against it.
Collapse
Affiliation(s)
- Tiziana Grafone
- Department of Onco-Hematology, Fondazione di Ricerca e Cura Giovanni Paolo II, Campobasso
| | - Michela Palmisano
- San Raffaele Vita-Salute University, School of Molecular Medicine, Milano, Italy
| | - Chiara Nicci
- Department of Onco-Hematology, Fondazione di Ricerca e Cura Giovanni Paolo II, Campobasso
| | - Sergio Storti
- Department of Onco-Hematology, Fondazione di Ricerca e Cura Giovanni Paolo II, Campobasso
| |
Collapse
|
30
|
Lin WH, Jiaang WT, Chen CW, Yen KJ, Hsieh SY, Yen SC, Chen CP, Chang KY, Chang CY, Chang TY, Huang YL, Yeh TK, Chao YS, Chen CT, Hsu JTA. BPR1J-097, a novel FLT3 kinase inhibitor, exerts potent inhibitory activity against AML. Br J Cancer 2011; 106:475-81. [PMID: 22187040 PMCID: PMC3273346 DOI: 10.1038/bjc.2011.564] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background: Activating mutations of Fms-like tyrosine kinase 3 (FLT3) constitute a major driver in the pathogenesis of acute myeloid leukaemia (AML). Hence, pharmacological inhibitors of FLT3 are of therapeutic interest for AML. Methods: The effects of inhibition of FLT3 activity by a novel potent FLT3 inhibitor, BPR1J-097, were investigated using in vitro and in vivo assays. Results: The 50% inhibitory concentration (IC50) of BPR1J-097 required to inhibit FLT3 kinase activity ranged from 1 to 10 nM, and the 50% growth inhibition concentrations (GC50s) were 21±7 and 46±14 nM for MOLM-13 and MV4-11 cells, respectively. BPR1J-097 inhibited FLT3/signal transducer and activator of transcription 5 phosphorylation and triggered apoptosis in FLT3-driven AML cells. BPR1J-097 also showed favourable pharmacokinetic property and pronounced dose-dependent tumour growth inhibition and regression in FLT3-driven AML murine xenograft models. Conclusion: These results indicate that BPR1J-097 is a novel small molecule FLT-3 inhibitor with promising in vivo anti-tumour activities and suggest that BPR1J-097 may be further developed in preclinical and clinical studies as therapeutics in AML treatments.
Collapse
Affiliation(s)
- W-H Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli County 350, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Shih KC, Lin CY, Chi HC, Hwang CS, Chen TS, Tang CY, Hsiao NW. Design of novel FLT-3 inhibitors based on dual-layer 3D-QSAR model and fragment-based compounds in silico. J Chem Inf Model 2011; 52:146-55. [PMID: 22142286 DOI: 10.1021/ci200434f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
FMS-like tyrosine kinase 3 (FLT-3) is strongly correlated with acute myeloid leukemia, but no FLT-3-inhibitor cocomplex structure is available to assist the design of therapeutic inhibitors. Hence, we propose a dual-layer 3D-QSAR model for FLT-3 that integrates the pharmacophore, CoMFA, and CoMSIA. We then coupled the model with the fragment-based design strategy to identify novel FLT-3 inhibitors. In the first layer, the previously established model, Hypo02, was evaluated in terms of its correlation coefficient (r), RMS, cost difference, and configuration cost, with values of 0.930, 1.24, 106.45, and 16.44, respectively. Moreover, Fischer's cross-validation test of data generated by Hypo02 yielded a 98% confidence level, and the validation of the testing set yielded a best r value of 0.87. The features of Hypo02 were separated into two parts and then used to screen the MiniMaybridge fragment compound database. Nine novel FLT-3 inhibitors were generated in this layer. In the second layer, Hypo02 was subjected to an alignment rule to generate CoMFA- and CoMSIA-based models, for which the partial least-squares validation method was utilized. The values of q(2), r(2), and predictive r(2) were 0.58, 0.98, and 0.76, respectively, derived from the CoMFA model with steric and electrostatic fields. The CoMSIA model with five different fields yielded values of 0.54, 0.97, and 0.76 for q(2), r(2), and predictive r(2), respectively. The CoMFA and CoMSIA models were used to constrain 3D structures of the nine novel FLT-3 inhibitors. This dual-layer 3D-QSAR model constitutes a valuable tool to easily and quickly screen and optimize novel potential FLT-3 inhibitors for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Kuei-Chung Shih
- Department of Computer Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | | | | | | | |
Collapse
|
32
|
Markovic A, MacKenzie KL, Lock RB. Induction of vascular endothelial growth factor secretion by childhood acute lymphoblastic leukemia cells via the FLT-3 signaling pathway. Mol Cancer Ther 2011; 11:183-93. [PMID: 22084166 DOI: 10.1158/1535-7163.mct-11-0503] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human leukemia cells secrete VEGF, which can act in a paracrine manner within the bone marrow microenvironment to promote leukemia cell survival and proliferation. The FLT-3 receptor tyrosine kinase plays an essential role in regulating normal hematopoiesis, but its constitutive activation via mutation in acute leukemias is generally associated with poor outcome. The aim of this study was to investigate interactions between the FLT-3 and VEGF signaling pathways in acute leukemia using cell lines and ex vivo cultures of pediatric acute lymphoblastic leukemia cells following expansion of direct patient explants in immunodeficient mice. Different xenograft lines exhibited variable cell surface FLT-3 expression, as well as basal and FLT-3 ligand-induced VEGF secretion, whereas the MV4;11 cell line, which expresses constitutively active FLT-3, secreted high levels of VEGF. The FLT-3 inhibitor, SU11657, significantly reduced VEGF secretion in three of six xenograft lines and MV4;11 cells, in conjunction with inhibition of FLT-3 tyrosine phosphorylation. Moreover, exposure of xenograft cells to the FLT-3-blocking antibody, D43, also reduced VEGF secretion to basal levels and decreased FLT-3 tyrosine phosphorylation. In terms of downstream signaling, SU11657 and D43 both caused dephosphorylation of extracellular signal-regulated kinase 1/2, with no changes in AKT or STAT5 phosphorylation. Finally, partial knockdown of FLT-3 expression by short interfering RNA also resulted in inhibition of VEGF secretion. These results indicate that FLT-3 signaling plays a central role in the regulation of VEGF secretion and that inhibition of the FLT-3/VEGF pathway may disrupt paracrine signaling between leukemia cells and the bone marrow microenvironment.
Collapse
Affiliation(s)
- Ana Markovic
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | | | | |
Collapse
|
33
|
Takahashi S. Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: biology and therapeutic implications. J Hematol Oncol 2011; 4:13. [PMID: 21453545 PMCID: PMC3076284 DOI: 10.1186/1756-8722-4-13] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 04/01/2011] [Indexed: 01/13/2023] Open
Abstract
FLT3 is a type III receptor tyrosine kinase. Mutations of FLT3 comprise one of the most frequently identified types of genetic alterations in acute myeloid leukemia. One-third of acute myeloid leukemia patients have mutations of this gene, and the majority of these mutations involve an internal tandem duplication in the juxtamembrane region of FLT3, leading to constitutive activation of downstream signaling pathways and aberrant cell growth. This review summarizes the current understanding of the effects of the downstream molecular signaling pathways after FLT3 activation, with a particular focus on the effects on transcription factors. Moreover, this review describes novel FLT3-targeted therapies, as well as efficient combination therapies for FLT3-mutated leukemia cells.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- The Division of Molecular Hematology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Japan.
| |
Collapse
|
34
|
Chen C, Wei X, Rao X, Wu J, Yang S, Chen F, Ma D, Zhou J, Dackor RT, Zeldin DC, Wang DW. Cytochrome P450 2J2 is highly expressed in hematologic malignant diseases and promotes tumor cell growth. J Pharmacol Exp Ther 2011; 336:344-55. [PMID: 21030485 PMCID: PMC3033713 DOI: 10.1124/jpet.110.174805] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 10/27/2010] [Indexed: 01/09/2023] Open
Abstract
Cytochrome P450 2J2 (CYP2J2) epoxygenase converts arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs) that exert multiple biological effects in the cardiovascular system and in various human solid cancers. However, it is unknown whether this enzyme is expressed or plays any role in malignant hematological diseases. In this study, we found strong and highly selective CYP2J2 expression in five human-derived malignant hematological cell lines and in leukemia cells from peripheral blood and bone marrow in 36 of 42 patients (86%) with malignant hematologic diseases. Furthermore, increased levels of EETs were detected in urine and blood samples from these patients. Addition of exogenous EET or CYP2J2 overexpression in cultured human-derived malignant hematologic cell lines markedly accelerated proliferation and attenuated apoptosis. Addition of the selective CYP2J2 inhibitor compound 26 (C26; 1-[4-(vinyl) phenyl]-4-[4-(diphenyl-hydroxymethyl)-piperidinyl]-butanone hydrochloride) inhibited cell proliferation and increased apoptosis, an effect that was significantly reversed by EET. CYP2J2 overexpression and exogenous EET activated AMP-activated protein kinase, c-Jun NH(2)-terminal kinase, and phosphatidylinositol 3-kinase/Akt signaling pathways, and increased epidermal growth factor receptor phosphorylation levels. CYP2J2 overexpression also enhanced malignant xenograft growth, which was efficiently inhibited by oral administration of C26 in Tie2-CYP2J2 transgenic mice and in severe combined immunodeficiency (SCID) xenograft mice. Together, these results suggest that CYP2J2 plays a key role in the pathogenesis of human hematologic malignant diseases. Selective inhibition of CYP2J2 may be a promising therapeutic strategy for these conditions.
Collapse
Affiliation(s)
- Chen Chen
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Novel azulene-based derivatives as potent multi-receptor tyrosine kinase inhibitors. Bioorg Med Chem Lett 2010; 20:6129-32. [DOI: 10.1016/j.bmcl.2010.08.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 08/04/2010] [Accepted: 08/05/2010] [Indexed: 11/17/2022]
|
36
|
Cells expressing FLT3/ITD mutations exhibit elevated repair errors generated through alternative NHEJ pathways: implications for genomic instability and therapy. Blood 2010; 116:5298-305. [PMID: 20807885 DOI: 10.1182/blood-2010-03-272591] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The internal tandem duplication (ITD) mutations of the FMS-like tyrosine kinase-3 (FLT3) receptor found in acute myeloid leukemia patients are associated with poor prognosis. Although DNA double-strand breaks (DSBs) are mainly repaired by the DNA-PK-dependent nonhomologous end-joining (NHEJ) pathway in normal mammalian cells, an alternative and less well-defined NHEJ pathway, characterized by microhomology at the repair junctions, play a role in the generation of deletions and translocations leading to cancer progression. Here we report that in FLT3/ITD-expressing cell lines and bone marrow mononuclear cells from FLT3/ITD knock-in mice, end-joining of DSBs occurs at microhomologous sequences resulting in a high frequency of DNA deletions. Strikingly, levels of Ku proteins, key components of the main NHEJ pathway, are decreased in FLT3/ITD(+) cell lines and murine FLT3/ITD bone marrow mononuclear cells. Concomitantly, levels of DNA ligase IIIα, a component of ALT NHEJ, are increased in FLT3/ITD-expressing cells. Cells treated with a FLT3 inhibitor demonstrate decreased DNA ligase IIIα and a reduction in DNA deletions, suggesting that FLT3 signaling regulates the pathways by which DSBs are repaired. Thus, therapy to inhibit FLT3/ITD signaling and/or DNA ligase IIIα may lead to repair that reduces repair errors and genomic instability.
Collapse
|
37
|
Rogers BB. Advances in the management of acute myeloid leukemia in older adult patients. Oncol Nurs Forum 2010; 37:E168-79. [PMID: 20439202 DOI: 10.1188/10.onf.e168-e179] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE/OBJECTIVES To update oncology nurses on new developments in the care of older adult patients with acute myeloid leukemia (AML). DATA SOURCES Clinical trial data, published guidelines, review articles, and conference proceedings. DATA SYNTHESIS Therapies for older adult patients with AML include cytarabine-based intensive-induction chemotherapy, investigational therapy, and supportive care. Nonmyeloablative allogeneic hematopoietic stem cell transplantation may provide the best opportunity for cure following remission if a human leukocyte antigen-matched hematopoietic stem cell donor is available. Careful assessment of patient- and disease-related factors is critical in determining the appropriate treatment modality for individuals. Assessment tools and algorithms may identify patients likely to be intolerant of intensive therapies. Supportive care may improve treatment tolerance and quality of life. CONCLUSIONS Survival outcomes have not improved significantly for older adult patients with AML. Novel therapies, coupled with better understanding of prognostic factors, may allow more highly individualized care. Performance status or comorbidity score may provide insights regarding outcome, particularly when combined with age, cytogenetic risk, or molecular markers. Efforts to improve transplantation safety may increase use in the older adult patient population. IMPLICATIONS FOR NURSING Nurses provide disease and treatment information, manage adverse effects, and offer emotional support. Knowledge of the key management issues for older adult patients with AML is critical in fulfilling this role.
Collapse
|
38
|
DeBoy CA, Rus H, Tegla C, Cudrici C, Jones MV, Pardo CA, Small D, Whartenby KA, Calabresi PA. FLT-3 expression and function on microglia in multiple sclerosis. Exp Mol Pathol 2010; 89:109-16. [PMID: 20566414 DOI: 10.1016/j.yexmp.2010.05.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 05/20/2010] [Indexed: 01/27/2023]
Abstract
Inflammatory cell infiltration and resident microglial activation within the central nervous system (CNS) are pathological events in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). While MS therapies target the peripheral immune system, no treatment is currently known to also modulate microglia. FMS-like tyrosine-3 (FLT-3) is expressed on hematopoietic and dendritic cells. We reported that FLT-3 inhibition ameliorates early actively induced EAE by predominantly modulating dendritic cell function as compared to microglia. We demonstrate in this report that FLT-3 is expressed in perivascular cuffs, brain parenchyma and in non-lesioned gray and white matter within MS brain but not in these regions within control brain. Furthermore, we demonstrate that FLT-3 is expressed on two populations of cells within MS brain; one which expresses the dendritic cell marker CD209, and the other which does not, suggesting that FLT-3 within MS brain is expressed on infiltrating dendritic cells and a non-dendritic cell such as microglia. Additionally, we report that FLT-3 inhibition in murine microglia blocks, in a dose-dependent manner, IFN-γ-induced expression of MHC class II and CD86, and LPS-induced secretion of IL-6. These data suggest that FLT-3 is involved in microglial cells' capacity to respond to environmental cues to function as antigen presenting cells and mediate CNS inflammation. Furthermore these data suggest that FLT-3 may be a therapeutic target on microglia to mitigate CNS inflammation.
Collapse
Affiliation(s)
- Cynthia A DeBoy
- Neurology, Johns Hopkins University, Pathology 627, 600 N. Wolfe Street, Baltimore, MD 21287,USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ubiquitin conjugase UBCH8 targets active FMS-like tyrosine kinase 3 for proteasomal degradation. Leukemia 2010; 24:1412-21. [PMID: 20508617 DOI: 10.1038/leu.2010.114] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The class III receptor tyrosine kinase FMS-like tyrosine kinase 3 (FLT3) regulates normal hematopoiesis and immunological functions. Nonetheless, constitutively active mutant FLT3 (FLT3-ITD) causally contributes to transformation and is associated with poor prognosis of acute myeloid leukemia (AML) patients. Histone deacetylase inhibitors (HDACi) can counteract deregulated gene expression profiles and decrease oncoprotein stability, which renders them candidate drugs for AML treatment. However, these drugs have pleiotropic effects and it is often unclear how they correct oncogenic transcriptomes and proteomes. We report here that treatment of AML cells with the HDACi LBH589 induces the ubiquitin-conjugating enzyme UBCH8 and degradation of FLT3-ITD. Gain- and loss-of-function approaches show that UBCH8 and the ubiquitin-ligase SIAH1 physically interact with and target FLT3-ITD for proteasomal degradation. These ubiquitinylating enzymes though have a significantly lesser effect on wild-type FLT3. Furthermore, physiological and pharmacological stimulation of FLT3 phosphorylation, inhibition of FLT3-ITD autophosphorylation and analysis of kinase-inactive FLT3-ITD revealed that tyrosine phosphorylation determines degradation of FLT3 and FLT3-ITD by the proteasome. These results provide novel insights into antileukemic activities of HDACi and position UBCH8, which have been implicated primarily in processes in the nucleus, as a previously unrecognized important modulator of FLT3-ITD stability and leukemic cell survival.
Collapse
|
40
|
Abstract
Acute myeloid leukaemia (AML) is a complicated, heterogeneous disease and the prognostic factors may be useful in deciding upon appropriate therapy. Cytogenetic testing at diagnosis yields critical prognostic information, but more refinement in prognosis is required. Within cytogenetic subgroups, further important subgroup definitions are possible based on the mutation status and expression analysis of genes such as C-KIT, FLT3, NPM1 and CEBPA, although defining therapies based on such mutations remains controversial.
Collapse
Affiliation(s)
- Richard M Stone
- Harvard Medical School, Dana-Farber Cancer Institute, Boston MA 02115-6084, USA.
| |
Collapse
|
41
|
Molecular characterization and prognostic significance of FLT3 in CML progression. Leuk Res 2009; 34:995-1001. [PMID: 20031210 DOI: 10.1016/j.leukres.2009.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 11/09/2009] [Accepted: 11/10/2009] [Indexed: 11/23/2022]
Abstract
To characterize the molecular mechanisms involved in the transition from the chronic phase to blast crisis in chronic myelogenous leukemia (CML), gene expression profiles of leukemic cells from patients in the chronic and blast crisis phases were analyzed using an 8.7K cDNA chip and real-time PCR. A transient transfection analysis was conducted to evaluate the role of FLT3, which was significantly upregulated in the blast crisis patients. Abl and c-Kit induction was detected in K562 cells transfected with FLT3 cDNA (K562/FLT3), and Abl and c-Kit levels were reduced in K562/FLT3 cells transfected with FLT3-siRNA (K562/FLT3-siRNA). The induction of FLT3 in CML cells attenuated imatinib-induced apoptosis. The opposite effect was observed in K562/FLT3-siRNA cells. An increased level of cleaved PARP and decreased level of pro-caspase 3 were noted when K562/FLT3-siRNA cells were treated with imatinib. These findings indicate that FLT3 is associated with disease progression, despite imatinib therapy. These results may help in the prediction of disease progression in CML patients and the development of more appropriate therapeutic modalities.
Collapse
|
42
|
Gocek E, Kiełbiński M, Baurska H, Haus O, Kutner A, Marcinkowska E. Different susceptibilities to 1,25-dihydroxyvitamin D3-induced differentiation of AML cells carrying various mutations. Leuk Res 2009; 34:649-57. [PMID: 19880182 DOI: 10.1016/j.leukres.2009.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 09/12/2009] [Accepted: 10/04/2009] [Indexed: 11/16/2022]
Abstract
This study was designed to compare the differentiation-inducing potential of 1,25-dihydroxyvitamin D(3) (1,25D) with some analogs (VDAs) in a panel of acute myeloid leukemia (AML) cell lines and in blast cells isolated from patients with AML. Of the cell lines studied, HL60 proved to be the most sensitive to each of the differentiation-inducing agents when compared to THP-1, NB-4 and U-937 cell lines. Three of the VDAs tested (PRI-1906, PRI-2191 and PRI-2201) were similarly effective as 1,25D in all the cell lines tested. However, blast cells from AML showed a varying sensitivity towards 1,25D. For example, blast cells isolated from patients in which the whole or part of chromosome 7 was deleted were extremely sensitive to 1,25D and its analogs. In contrast, 1,25D failed to increase the expression of differentiation markers in blast cells isolated from patients carrying activating mutations in Flt3 gene. Since, the expression of vitamin D receptor (VDR) in cells with Flt3 mutations was increased to the same extent as in other AML cells this suggests that failure of these cells to differentiate lies downstream of the receptor. That blast cells with different cytogenetic abnormalities have dissimilar responses to 1,25D and its analogs, may have implications in the use of 1,25D as a 'differentiation therapy' for myeloid leukemias. The analog PRI-2191 (tacalcitol) was found to be the most potent in inducing patient's cells differentiation.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Department of Biotechnology, University of Wrocław, Tamka 2, 50-137 Wrocław, Poland
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Angiogenesis is a hallmark of tumor development and metastasis and is now a validated target for cancer treatment. However, the survival benefits of antiangiogenic drugs have thus far been rather modest, stimulating interest in developing more effective ways to combine antiangiogenic drugs with established chemotherapies. This review discusses recent progress and emerging challenges in this field; interactions between antiangiogenic drugs and conventional chemotherapeutic agents are examined, and strategies for the optimization of combination therapies are discussed. Antiangiogenic drugs such as the anti-vascular endothelial growth factor antibody bevacizumab can induce a functional normalization of the tumor vasculature that is transient and can potentiate the activity of coadministered chemoradiotherapies. However, chronic angiogenesis inhibition typically reduces tumor uptake of coadministered chemotherapeutics, indicating a need to explore new approaches, including intermittent treatment schedules and provascular strategies to increase chemotherapeutic drug exposure. In cases where antiangiogenesis-induced tumor cell starvation augments the intrinsic cytotoxic effects of a conventional chemotherapeutic drug, combination therapy may increase antitumor activity despite a decrease in cytotoxic drug exposure. As new angiogenesis inhibitors enter the clinic, reliable surrogate markers are needed to monitor the progress of antiangiogenic therapies and to identify responsive patients. New targets for antiangiogenesis continue to be discovered, increasing the opportunities to interdict tumor angiogenesis and circumvent resistance mechanisms that may emerge with chronic use of these drugs.
Collapse
Affiliation(s)
- Jie Ma
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215
| | - David J. Waxman
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215
| |
Collapse
|
44
|
Internal tandem duplication of FLT3 (FLT3/ITD) induces increased ROS production, DNA damage, and misrepair: implications for poor prognosis in AML. Blood 2008; 111:3173-82. [PMID: 18192505 DOI: 10.1182/blood-2007-05-092510] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Activating mutations of the FMS-like tyrosine kinase-3 (FLT3) receptor occur in approximately 30% of acute myeloid leukemia (AML) patients and, at least for internal tandem duplication (ITD) mutations, are associated with poor prognosis. FLT3 mutations trigger downstream signaling pathways including RAS-MAP/AKT kinases and signal transducer and activator of transcription-5 (STAT5). We find that FLT3/ITD mutations start a cycle of genomic instability whereby increased reactive oxygen species (ROS) production leads to increased DNA double-strand breaks (DSBs) and repair errors that may explain aggressive AML in FLT3/ITD patients. Cell lines transfected with FLT3/ITD and FLT3/ITD-positive AML cell lines and primary cells demonstrate increased ROS. Increased ROS levels appear to be produced via STAT5 signaling and activation of RAC1, an essential component of ROS-producing NADPH oxidases. A direct association of RAC1-GTP binding to phosphorylated STAT5 (pSTAT5) provides a possible mechanism for ROS generation. A FLT3 inhibitor blocked increased ROS in FLT3/ITD cells resulting in decreased DSB and increased repair efficiency and fidelity. Our study suggests that the aggressiveness of the disease and poor prognosis of AML patients with FLT3/ITD mutations could be the result of increased genomic instability that is driven by higher endogenous ROS, increased DNA damage, and decreased end-joining fidelity.
Collapse
|
45
|
Lee EM, Bachmann PS, Lock RB. Xenograft models for the preclinical evaluation of new therapies in acute leukemia. Leuk Lymphoma 2007; 48:659-68. [PMID: 17454623 DOI: 10.1080/10428190601113584] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Major advances in understanding the pathophysiology of acute leukemia have resulted in a dramatic increase in the availability of novel compounds for clinical trials. However, since the number of new drugs far exceeds the number of clinical trials that can be conducted because of the availability of eligible patients, there is an urgent need to utilize reliable preclinical models for the prioritization of the most promising potential therapies for those clinical trials. The most widely used preclinical models for the acute leukemias are human tumor xenografts established in immune-deficient mice, and genetically engineered mouse strains. This review summarizes the recent developments and considerations in the use of xenograft models of acute lymphoblastic leukemia, acute myeloid leukemia, and acute promyelocytic leukemia for the preclinical testing of new therapies.
Collapse
Affiliation(s)
- Erwin M Lee
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | | | | |
Collapse
|
46
|
McCubrey JA, Steelman LS, Franklin RA, Abrams SL, Chappell WH, Wong EWT, Lehmann BD, Terrian DM, Basecke J, Stivala F, Libra M, Evangelisti C, Martelli AM. Targeting the RAF/MEK/ERK, PI3K/AKT and p53 pathways in hematopoietic drug resistance. ADVANCES IN ENZYME REGULATION 2007; 47:64-103. [PMID: 17382374 PMCID: PMC2696319 DOI: 10.1016/j.advenzreg.2006.12.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Shankar DB, Li J, Tapang P, Owen McCall J, Pease LJ, Dai Y, Wei RQ, Albert DH, Bouska JJ, Osterling DJ, Guo J, Marcotte PA, Johnson EF, Soni N, Hartandi K, Michaelides MR, Davidsen SK, Priceman SJ, Chang JC, Rhodes K, Shah N, Moore TB, Sakamoto KM, Glaser KB. ABT-869, a multitargeted receptor tyrosine kinase inhibitor: inhibition of FLT3 phosphorylation and signaling in acute myeloid leukemia. Blood 2007; 109:3400-8. [PMID: 17209055 PMCID: PMC1852258 DOI: 10.1182/blood-2006-06-029579] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In 15% to 30% of patients with acute myeloid leukemia (AML), aberrant proliferation is a consequence of a juxtamembrane mutation in the FLT3 gene (FMS-like tyrosine kinase 3-internal tandem duplication [FLT3-ITD]), causing constitutive kinase activity. ABT-869 (a multitargeted receptor tyrosine kinase inhibitor) inhibited the phosphorylation of FLT3, STAT5, and ERK, as well as Pim-1 expression in MV-4-11 and MOLM-13 cells (IC(50) approximately 1-10 nM) harboring the FLT3-ITD. ABT-869 inhibited the proliferation of these cells (IC(50) = 4 and 6 nM, respectively) through the induction of apoptosis (increased sub-G(0)/G(1) phase, caspase activation, and PARP cleavage), whereas cells harboring wild-type (wt)-FLT3 were less sensitive. In normal human blood spiked with AML cells, ABT-869 inhibited phosphorylation of FLT3 (IC(50) approximately 100 nM), STAT5, and ERK, and decreased Pim-1 expression. In methylcellulose-based colony-forming assays, ABT-869 had no significant effect up to 1000 nM on normal hematopoietic progenitor cells, whereas in AML patient samples harboring both FLT3-ITD and wt-FLT3, ABT-869 inhibited colony formation (IC(50) = 100 and 1000 nM, respectively). ABT-869 dose-dependently inhibited MV-4-11 and MOLM-13 flank tumor growth, prevented tumor formation, regressed established MV-4-11 xenografts, and increased survival by 20 weeks in an MV-4-11 engraftment model. In tumors, ABT-869 inhibited FLT3 phosphorylation, induced apoptosis (transferase-mediated dUTP nick-end labeling [TUNEL]) and decreased proliferation (Ki67). ABT-869 is under clinical development for AML.
Collapse
Affiliation(s)
- Deepa B Shankar
- Division of Hematology/Oncology, Department of Pediatrics, Gwynne Hazen Cherry Memorial Laboratories, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, Schwartz B, Simantov R, Kelley S. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov 2006; 5:835-44. [PMID: 17016424 DOI: 10.1038/nrd2130] [Citation(s) in RCA: 1354] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Since the molecular revolution of the 1980s, knowledge of the aetiology of cancer has increased considerably, which has led to the discovery and development of targeted therapies tailored to inhibit cancer-specific pathways. The introduction and refinement of rapid, high-throughput screening technologies over the past decade has greatly facilitated this targeted discovery and development process. Here, we describe the discovery and continuing development of sorafenib (previously known as BAY 43-9006), the first oral multikinase inhibitor that targets Raf and affects tumour signalling and the tumour vasculature. The discovery cycle of sorafenib (Nexavar; Bayer Pharmaceuticals) - from initial screening for a lead compound to FDA approval for the treatment of advanced renal cell carcinoma in December 2005 - was completed in just 11 years, with approval being received approximately 5 years after the initiation of the first Phase I trial.
Collapse
Affiliation(s)
- Scott Wilhelm
- Department of Cancer Research, Bayer Pharmaceuticals Corp., West Haven, Connecticut 06516, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mikalsen T, Gerits N, Moens U. Inhibitors of signal transduction protein kinases as targets for cancer therapy. BIOTECHNOLOGY ANNUAL REVIEW 2006; 12:153-223. [PMID: 17045195 DOI: 10.1016/s1387-2656(06)12006-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer development requires that tumour cells attain several capabilities, including increased replicative potentials, anchorage and growth-factor independency, evasion of apoptosis, angiogenesis and metastasis. Many of these processes involve the actions of protein kinases, which have emerged as key regulators of all aspects of neoplasia. Perturbed protein kinase activity is repeatedly found to be associated with human malignancies, making these proteins attractive targets for anti-cancer therapy. The last decade has witnessed an exponential increase in the development of specific small protein kinase inhibitors. Many of them are in clinical trials in patients with different types of cancer and some are successfully used in clinic. This review describes different approaches that are currently applied to develop such specific protein kinase inhibitors and provides an overview of protein kinase inhibitors that are currently in clinical trials or are administered in the clinic. Focus is directed on inhibitors against receptor tyrosine kinases and protein kinases participating in the signalling cascades.
Collapse
Affiliation(s)
- Theresa Mikalsen
- Department of Microbiology and Virology, Institute of Medical Biology, University of Tromsø, N-9037 Tromsø, Norway
| | | | | |
Collapse
|