1
|
Indermun S, Kumar P, Govender M, Choonara YE. Can Nanomedicinal Approaches Provide an Edge to the Efficacy of Tyrosine Kinase Inhibitors? Curr Med Chem 2023; 30:1482-1501. [PMID: 35726410 DOI: 10.2174/0929867329666220618162303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/10/2022] [Accepted: 04/04/2022] [Indexed: 11/22/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are effective drug molecules for the treatment of various cancers. Nanomedicinal interventions and approaches may not only provide carrying capacities for TKIs but also potentially target tumor-specific environments and even cellular compartments. Nano-inspired drug delivery systems may hence enhance the efficacy of the drugs through enhanced tumour-availability resulting in greater efficacy and decreased side effects. A variety of nanosystems have been developed for the delivery of TKIs for the enhanced treatment of cancers, each with their own preparation methods and physicochemical properties. This review will therefore discuss the applicability of nano-interventions towards combination therapies, dose reduction, and greater potential treatment outcomes. The individual nanosystems have been highlighted with emphasis on the developed systems and their efficacy against various cancer cell lines and models.
Collapse
Affiliation(s)
- Sunaina Indermun
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Mershen Govender
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Yahya E Choonara
- Department of Pharmacy and Pharmacology, Wits Advanced Drug Delivery Platform Research Unit, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| |
Collapse
|
2
|
Huang K, Huo B, Li D, Xue J, Chen J. Enhanced Efficacy of Gefitinib in Drug-Sensitive and Drug-Resistant Cancer Cell Lines after Arming with a Singlet Oxygen Releasing Moiety. ChemMedChem 2020; 15:794-798. [PMID: 32162481 DOI: 10.1002/cmdc.202000036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/05/2020] [Indexed: 12/24/2022]
Abstract
Attractive results have been achieved with small-molecule target-based drugs in the anticancer field; however, enhancing their treatment effect and solving the problem of drug resistance remain key concerns worldwide. Inspired by the specific affinity of gefitinib for tumour cells and the strong oxidation capacity of singlet oxygen, we combined a chemically generated singlet oxygen moiety with the small-molecule targeted drug gefitinib to improve its anticancer effect. We designed and synthesised a novel compound (Y5-1), in which a small-molecule targeted therapy agent (gefitinib) and a singlet oxygen (provided by an in vitro photodynamic reaction) thermally controlled releasing moiety are covalently conjugated. We demonstrated that the introduction of the singlet oxygen thermally controlled releasing moiety enhanced the anticancer activities of gefitinib. The results of this study are expected to provide a novel strategy to enhance the effect of chemotherapy drugs on drug-resistant cell lines.
Collapse
Affiliation(s)
- Kunshan Huang
- National and Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, P. R. China
| | - Beibei Huo
- National and Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, P. R. China
| | - Dongyao Li
- National and Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, P. R. China
| | - Jinping Xue
- National and Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, P. R. China
| | - Juanjuan Chen
- National and Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, P. R. China
| |
Collapse
|
3
|
Ciftci HI, Can M, Ellakwa DE, Suner SC, Ibrahim MA, Oral A, Sekeroglu N, Özalp B, Otsuka M, Fujita M, Alparslan M, Radwan MO. Anticancer activity of Turkish marine extracts: a purple sponge extract induces apoptosis with multitarget kinase inhibition activity. Invest New Drugs 2020; 38:1326-1333. [PMID: 32062733 DOI: 10.1007/s10637-020-00911-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/10/2020] [Indexed: 11/29/2022]
Abstract
Marine natural products have drawn a great deal of attention as a vital source of new drugs for the last five decades. However, marine organisms in the seas surrounding Turkey (the Black Sea, the Aegean Sea and the Mediterranean Sea) haven't been yet extensively explored. In the present study, three marine organisms (Dysidea avara, Microcosmus sabatieri and Echinaster sepositus) were sampled from the Dardanelles (Turkish Straits System, Western Turkey) by scientific divers, transferred to the laboratory and then were extracted with 70% ethanol. The extracts were tested for their cytotoxic effect against K562, KMS-12PE, A549, and A375 cancer cell lines. The sponge extract elicited the most promising cytotoxic activity, thus it was further evaluated against H929, MCF-7, HeLa, and HCT116 cancer cells. Most of the designated cells showed a considerable sensitivity for the sponge extract particularly H929, K562, KMS-12PE and HeLa cells with IC50 less than 10 μg/mL. On the contrary, the other two extracts exhibited no cytotoxic activity on all cells at 100 μg/mL concentration. The sponge extract was tested for its capacity to induce apoptosis in cancer cells and to inhibit a panel of tyrosine kinases showing remarkable results. The outcome of this study represents a platform for discovery of new chemotherapeutic agents of marine natural origin.
Collapse
Affiliation(s)
- Halil I Ciftci
- Department of Drug Discovery, Science Farm Ltd., 1-7-30-805 Kuhonji, Chuo-ku, Kumamoto, 862-0976, Japan.,Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan
| | - Mustafa Can
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan.,Department of Engineering Sciences, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Havaalani Sosesi Caddesi No:25, 35620, Cigli/Izmir, Turkey
| | - Doha E Ellakwa
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan.,Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Greater Cairo, Nasr City, 11651, Egypt
| | - Salih C Suner
- Chemical and Chemical Processing, Vocational School of Lapseki, Canakkale Onsekiz Mart University, 17800, Canakkale, Turkey
| | - Mohamed A Ibrahim
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Ayhan Oral
- Department of Chemistry, Faculty of Science, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey
| | - Nazim Sekeroglu
- Department of Horticulture, Faculty of Agriculture, Kilis 7 Aralik University, 79000, Kilis, Turkey
| | - Barış Özalp
- Section of Underwater Technology, Vocational School of Ocean Engineering, Canakkale Onsekiz Mart University, 17100, Canakkale, Turkey
| | - Masami Otsuka
- Department of Drug Discovery, Science Farm Ltd., 1-7-30-805 Kuhonji, Chuo-ku, Kumamoto, 862-0976, Japan.,Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan.
| | - Mustafa Alparslan
- Faculty of Fisheries and Aquaculture, Izmir Katip Celebi University, 35620, Izmir, Turkey.
| | - Mohamed O Radwan
- Department of Drug Discovery, Science Farm Ltd., 1-7-30-805 Kuhonji, Chuo-ku, Kumamoto, 862-0976, Japan. .,Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-Ku, Kumamoto, 8620973, Japan. .,Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki, Cairo, 12622, Egypt.
| |
Collapse
|
4
|
Antonenko SV, Kravchuk IV, Telegeev GD. Interaction of Bcl-Abl Oncoprotein with the Glg1 Protein in K562 Cells: its Role in the Pathogenesis of Chronic Myeloid Leukemia. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720010028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
5
|
Siveen KS, Prabhu KS, Achkar IW, Kuttikrishnan S, Shyam S, Khan AQ, Merhi M, Dermime S, Uddin S. Role of Non Receptor Tyrosine Kinases in Hematological Malignances and its Targeting by Natural Products. Mol Cancer 2018; 17:31. [PMID: 29455667 PMCID: PMC5817858 DOI: 10.1186/s12943-018-0788-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Tyrosine kinases belong to a family of enzymes that mediate the movement of the phosphate group to tyrosine residues of target protein, thus transmitting signals from the cell surface to cytoplasmic proteins and the nucleus to regulate physiological processes. Non-receptor tyrosine kinases (NRTK) are a sub-group of tyrosine kinases, which can relay intracellular signals originating from extracellular receptor. NRTKs can regulate a huge array of cellular functions such as cell survival, division/propagation and adhesion, gene expression, immune response, etc. NRTKs exhibit considerable variability in their structural make up, having a shared kinase domain and commonly possessing many other domains such as SH2, SH3 which are protein-protein interacting domains. Recent studies show that NRTKs are mutated in several hematological malignancies, including lymphomas, leukemias and myelomas, leading to aberrant activation. It can be due to point mutations which are intragenic changes or by fusion of genes leading to chromosome translocation. Mutations that lead to constitutive kinase activity result in the formation of oncogenes, such as Abl, Fes, Src, etc. Therefore, specific kinase inhibitors have been sought after to target mutated kinases. A number of compounds have since been discovered, which have shown to inhibit the activity of NRTKs, which are remarkably well tolerated. This review covers the role of various NRTKs in the development of hematological cancers, including their deregulation, genetic alterations, aberrant activation and associated mutations. In addition, it also looks at the recent advances in the development of novel natural compounds that can target NRTKs and perhaps in combination with other forms of therapy can show great promise for the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Kodappully S Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Iman W Achkar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Sunitha Shyam
- Medical Research Center, Hamad Medical Corporation, Doha, State of Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, State of Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, State of Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar.
| |
Collapse
|
6
|
Parker LL, Kron SJ. Kinase activation in circulating cells: opportunities for biomarkers for diagnosis and therapeutic monitoring. ACTA ACUST UNITED AC 2013; 2:33-46. [PMID: 23485115 DOI: 10.1517/17530059.2.1.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A clinically useful tool to assay phosphorylation-dependent signaling in circulating cells has the potential to provide a wealth of information about a patient's health, including information unavailable by any other method. Patterns of kinase activation, such as the abnormal signaling characteristic of myeloproliferative disorders, may offer highly specific biomarkers for diagnosis or monitoring the efficacy of therapeutics. For assays of kinase activity in circulating leukocytes to be standardized, let alone made practical for the clinic, numerous technical hurdles must be overcome. In this review the current status of analysis of kinase signaling in circulating cells and recent progress in biomarker discovery and validation is discussed. Looking forward, the potential value of signaling patterns as complex biomarkers and the resulting need for future development of robust, multiplexed assays of kinase activation is addressed.
Collapse
Affiliation(s)
- Laurie L Parker
- University of Chicago, Ludwig Center for Metastasis Research, Knapp R322, 924 E. 57th Street, Chicago, IL 6063, USA
| | | |
Collapse
|
7
|
Yui MA, Feng N, Zhang JA, Liaw CY, Rothenberg EV, Longmate JA. Loss of T cell progenitor checkpoint control underlies leukemia initiation in Rag1-deficient nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2013; 190:3276-88. [PMID: 23440410 DOI: 10.4049/jimmunol.1202970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NOD mice exhibit major defects in the earliest stages of T cell development in the thymus. Genome-wide genetic and transcriptome analyses were used to investigate the origins and consequences of an early T cell developmental checkpoint breakthrough in Rag1-deficient NOD mice. Quantitative trait locus analysis mapped the presence of checkpoint breakthrough cells to several known NOD diabetes susceptibility regions, particularly insulin-dependent diabetes susceptibility genes (Idd)9/11 on chromosome 4, suggesting common genetic origins for T cell defects affecting this trait and autoimmunity. Genome-wide RNA deep-sequencing of NOD and B6 Rag1-deficient thymocytes revealed the effects of genetic background prior to breakthrough, as well as the cellular consequences of the breakthrough. Transcriptome comparison between the two strains showed enrichment in differentially expressed signal transduction genes, prominently tyrosine kinase and actin-binding genes, in accord with their divergent sensitivities to activating signals. Emerging NOD breakthrough cells aberrantly expressed both stem cell-associated proto-oncogenes, such as Lmo2, Hhex, Lyl1, and Kit, which are normally repressed at the commitment checkpoint, and post-β-selection checkpoint genes, including Cd2 and Cd5. Coexpression of genes characteristic of multipotent progenitors and more mature T cells persists in the expanding population of thymocytes and in the thymic leukemias that emerge with age in these mice. These results show that Rag1-deficient NOD thymocytes have T cell defects that can collapse regulatory boundaries at two early T cell checkpoints, which may predispose them to both leukemia and autoimmunity.
Collapse
Affiliation(s)
- Mary A Yui
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA.
| | | | | | | | | | | |
Collapse
|
8
|
Knapper S. The clinical development of FLT3 inhibitors in acute myeloid leukemia. Expert Opin Investig Drugs 2011; 20:1377-95. [DOI: 10.1517/13543784.2011.611802] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
9
|
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
|
10
|
Kanaujiya JK, Lochab S, Pal P, Christopeit M, Singh SM, Sanyal S, Behre G, Trivedi AK. Proteomic approaches in myeloid leukemia. Electrophoresis 2011; 32:357-67. [DOI: 10.1002/elps.201000428] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 10/29/2010] [Accepted: 11/25/2010] [Indexed: 01/17/2023]
|
11
|
Schwartz LB, Sheikh J, Singh A. Current strategies in the management of hypereosinophilic syndrome, including mepolizumab. Curr Med Res Opin 2010; 26:1933-46. [PMID: 20565230 DOI: 10.1185/03007995.2010.493132] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Patients with hypereosinophilic syndrome (HES) vary considerably in their clinical presentation with regard to the severity and pattern of end-organ involvement. Clinical manifestations range from nonspecific symptoms to life-threatening, multisystem damage caused by eosinophil infiltration and local release of proinflammatory mediators and toxic granule products from these invading cells. The primary objective of treatment is to reduce blood and tissue eosinophilia and prevent eosinophil-mediated tissue damage as safely as possible. Systemic corticosteroids, such as prednisone, are first-line therapy for the management of patients with symptomatic HES who lack the Fip1-like 1-platelet-derived growth factor receptor-alpha (FIP1L1-PDGFRA) gene fusion mutation. The tyrosine kinase inhibitor, imatinib, is first-line treatment for FIP1L1-PDGFRA-positive patients). Because of the toxicity and serious side-effects that can occur with oral corticosteroids, alternative therapies may need to be introduced to reduce the cumulative corticosteroid exposure while maintaining disease control. SCOPE Among corticosteroid-sparing agents are cytotoxic drugs and interferon-alpha; anti-interleukin-5 (IL-5) monoclonal antibodies are also currently under investigation for the treatment of HES. This manuscript reviews the available treatments for HES and the range of side-effects associated with long-term corticosteroid use, and then focuses on the anti-IL-5 monoclonal antibodies, mepolizumab and reslizumab. Of these, only mepolizumab has been studied in a randomized, placebo-controlled trial. Literature search methodology utilized www.pubmed.gov and www.clinicaltrials.gov with search terms including hypereosinophilic syndrome and corticosteroid side-effects coupled with search terms including eosinophils, mepolizumab and reslizumab through March 2010. FINDINGS Three case studies are presented that demonstrate the limitations of corticosteroid therapy in terms of tolerability and quality of life, and the subsequent use of mepolizumab as a corticosteroid-sparing agent in these individuals. CONCLUSION Targeted eosinophil-directed therapy with an anti-IL-5 neutralizing monoclonal antibody reduced the need for corticosteroids in these three HES patients without disease exacerbations.
Collapse
Affiliation(s)
- Lawrence B Schwartz
- Division of Rheumatology, Allergy & Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | |
Collapse
|
12
|
Messina M, Chiaretti S, Tavolaro S, Peragine N, Vitale A, Elia L, Sica S, Levis A, Guarini A, Foà R. Protein kinase gene expression profiling and in vitro functional experiments identify novel potential therapeutic targets in adult acute lymphoblastic leukemia. Cancer 2010; 116:3426-37. [DOI: 10.1002/cncr.25113] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
13
|
Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY, O'Brien S, Estrov Z, Borthakur G, Thomas D, Pierce SR, Brandt M, Byrd A, Bekele BN, Pratz K, Luthra R, Levis M, Andreeff M, Kantarjian HM. Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol 2010; 28:1856-62. [PMID: 20212254 PMCID: PMC2930809 DOI: 10.1200/jco.2009.25.4888] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To determine the efficacy and toxicity of the combination of sorafenib, cytarabine, and idarubicin in patients with acute myeloid leukemia (AML) younger than age 65 years. PATIENTS AND METHODS In the phase I part of the study, 10 patients with relapsed AML were treated with escalating doses of sorafenib with chemotherapy to establish the feasibility of the combination. We then treated 51 patients (median age, 53 years; range, 18 to 65 years) who had previously untreated AML with cytarabine at 1.5 g/m(2) by continuous intravenous (IV) infusion daily for 4 days (3 days if > 60 years of age), idarubicin at 12 mg/m(2) IV daily for 3 days, and sorafenib at 400 mg orally twice daily for 7 days. RESULTS Overall, 38 (75%) patients have achieved a complete remission (CR), including 14 (93%) of 15 patients with mutated FMS-like tyrosine kinase-3 (FLT3; the 15th patient had complete remission with incomplete platelet recovery [CRp]) and 24 (66%) of 36 patients with FLT3 wild-type (WT) disease (three additional FLT3-WT patients had CRp). FLT3-mutated patients were more likely to achieve a CR than FLT3-WT patients (P = .033). With a median follow-up of 54 weeks (range, 8 to 87 weeks), the probability of survival at 1 year is 74%. Among the FLT3-mutated patients, 10 have relapsed and five remain in CR with a median follow-up of 62 weeks (range, 10 to 76 weeks). Plasma inhibitory assay demonstrated an on-target effect on FLT3 kinase activity. CONCLUSION Sorafenib can be safely combined with chemotherapy, produces a high CR rate in FLT3-mutated patients, and inhibits FLT3 signaling.
Collapse
Affiliation(s)
- Farhad Ravandi
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD.,Corresponding author: Farhad Ravandi, MD, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX 77030; e-mail:
| | - Jorge E. Cortes
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Daniel Jones
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Stefan Faderl
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Guillermo Garcia-Manero
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Marina Y. Konopleva
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Susan O'Brien
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Zeev Estrov
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Gautam Borthakur
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Deborah Thomas
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Sherry R. Pierce
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Mark Brandt
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Anna Byrd
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - B. Nebiyou Bekele
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Keith Pratz
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Rajyalakshmi Luthra
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Mark Levis
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Michael Andreeff
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Hagop M. Kantarjian
- From the Departments of Leukemia, Hematopathology, Stem Cell Transplantation and Cellular Therapy, and Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX; and Division of Hematological Malignancies, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| |
Collapse
|
14
|
Paradis FW, Simard R, Gaudet D. Quantitative assay for the detection of the V617F variant in the Janus kinase 2 (JAK2) gene using the Luminex xMAP technology. BMC MEDICAL GENETICS 2010; 11:54. [PMID: 20359349 PMCID: PMC2861026 DOI: 10.1186/1471-2350-11-54] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 04/01/2010] [Indexed: 11/25/2022]
Abstract
Background The availability of clinically valid biomarkers contribute to improve the diagnosis and clinical management of diseases. A valine-to-phenylalanine substitution at position 617 (V617F) in the Janus kinase 2 (JAK2) gene has been recently associated with key signaling abnormalities in the transduction of haemopoietic growth-factor receptors and is now considered as a useful clinical marker of myeloproliferative neoplasms. Several methods have recently been reported to detect the JAK2 V617F point mutation and show variable sensitivity. Methods Using the Luminex xMAP technology, we developed a quantitative assay to detect the JAK2V617F variant. The method was based on polymerase chain reaction (PCR) followed by hybridization to specific probes coupled with internally dyed microspheres. The assay comprises 3 steps: genomic DNA extraction, end point PCR reaction, direct hybridization of PCR fragments and quantification. It has been tested with different sources of nucleic acid. Results Applied to whole blood samples, this quantitative assay showed a limit of detection of 2%. A highly sensitive allele-specific primer extension reaction performed in parallel allowed to validate the results and to identify the specimens with values below 2%. Conclusion Direct hybridization assay using the Luminex xMAP technology allows sensitive quantification of JAK2V617F from blood spots. It is simple and can be easily performed in a clinical setting.
Collapse
Affiliation(s)
- François W Paradis
- ECOGENE-21, department of médecine, Université de Montréal, Chicoutimi Hospital, 225 rue St-Vallier, Saguenay, Province of Québec, G7H 7P2, Canada
| | | | | |
Collapse
|
15
|
Odgerel T, Kikuchi J, Wada T, Shimizu R, Kano Y, Furukawa Y. MSK1 activation in acute myeloid leukemia cells with FLT3 mutations. Leukemia 2010; 24:1087-90. [PMID: 20357825 DOI: 10.1038/leu.2010.48] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
16
|
Guerrouahen BS, Futami M, Vaklavas C, Kanerva J, Whichard ZL, Nwawka K, Blanchard EG, Lee FY, Robinson LJ, Arceci R, Kornblau SM, Wieder E, Cayre YE, Corey SJ. Dasatinib inhibits the growth of molecularly heterogeneous myeloid leukemias. Clin Cancer Res 2010; 16:1149-58. [PMID: 20145167 DOI: 10.1158/1078-0432.ccr-09-2416] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Dasatinib is a dual Src/Abl inhibitor recently approved for Bcr-Abl+ leukemias with resistance or intolerance to prior therapy. Because Src kinases contribute to multiple blood cell functions by triggering a variety of signaling pathways, we hypothesized that their molecular targeting might lead to growth inhibition in acute myeloid leukemia (AML). EXPERIMENTAL DESIGN We studied growth factor-dependent and growth factor-independent leukemic cell lines, including three cell lines expressing mutants of receptor tyrosine kinases (Flt3 or c-Kit) as well as primary AML blasts for responsiveness to dasatinib. RESULTS Dasatinib resulted in the inhibition of Src family kinases in all cell lines and blast cells at approximately 1 x 10(-9) mol/L. It also inhibited mutant Flt3 or Kit tyrosine phosphorylation at approximately 1 x 10(-6) mol/L. Mo7e cells expressing the activating mutation (codon 816) of c-Kit were most sensitive to growth inhibition with a GI(50) of 5 x 10(-9) mol/L. Primary AML blast cells exhibited a growth inhibition of <1 x 10(-6) mol/L. Cell lines that showed growth inhibition at approximately 1 x 10(-6) mol/L showed a G(1) cell cycle arrest and correlated with accumulation of p21 and p27 protein. The addition of rapamycin or cytotoxic agents enhanced growth inhibition. Dasatinib also caused the apoptosis of Mo7e cells expressing oncogenic Kit. CONCLUSIONS Although all of the precise targets for dasatinib are not known, this multikinase inhibitor causes either growth arrest or apoptosis in molecularly heterogeneous AML. The addition of cytotoxic or targeted agents can enhance its effects.
Collapse
Affiliation(s)
- Bella S Guerrouahen
- Division of Pediatrics, Departments of Leukemia and Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Apostolidou E, Kantarjian HM, Verstovsek S. JAK2 inhibitors: A reality? A hope? CLINICAL LYMPHOMA & MYELOMA 2009; 9 Suppl 3:S340-5. [PMID: 19778862 DOI: 10.3816/clm.2009.s.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Myelofibrosis (MF; primary or post-polycythemia vera/essential thrombocythemia) carries the worst prognosis among BCR-ABL-negative myeloproliferative neoplasms (MPNs). Stem cell transplantation is the only curative approach but is hampered by significant nonrelapse mortality. Thus, effective, targeted therapies are needed. A mutated Janus kinase 2 (JAK2) gene (JAK2(V617F)), found in a significant portion of patients with MPN, results in increased JAK2 tyrosine kinase activity, leading to clonal proliferation; several small molecules inhibit the growth of hematopoietic colonies harboring JAK2(V617). Several JAK2 inhibitors have reached the clinical trial stage and are reviewed here. The most developed among them is INCB018424, which has demonstrated noteworthy clinical activity, with a rapid and profound reduction in splenomegaly and associated improvement in constitutional symptoms in MF patients receiving 10-25 mg orally twice daily, continuously. Thrombocytopenia (reversible) was the most common adverse event, seen in 30% of patients treated with 25 mg twice daily but not with 10 mg twice daily. Interestingly, INCB018424 was equally active in patients with and without JAK2 mutation. Other JAK2 inhibitors are less developed but show a similar type of clinical benefit. Conclusively, JAK2 inhibitors, particularly INCB018424, are clinically active in MF and are well tolerated. Whether they have an effect on the natural course of MF in treated patients remains to be elucidated.
Collapse
|
18
|
Chu SH, Small D. Mechanisms of resistance to FLT3 inhibitors. Drug Resist Updat 2009; 12:8-16. [PMID: 19162530 PMCID: PMC4891941 DOI: 10.1016/j.drup.2008.12.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 12/02/2008] [Indexed: 11/21/2022]
Abstract
The success of the small molecule tyrosine kinase receptor inhibitor (TKI) imatinib mesylate (Gleevec) in the treatment of chronic myeloid leukemia (CML) constitutes an eminent paradigm shift advocating the rational design of cancer therapeutics specifically targeting the transformation events that drive tumorigenicity. In acute myeloid leukemias (AMLs), the most frequent identified transforming events are activating mutations in the FLT3 receptor tyrosine kinase that constitutively activate survival and proliferation pathways. FLT3 TKIs that are in various phases of clinical trials are showing some initial promise. However, primary and secondary acquired resistance stands to severely compromise long-term and durable efficacy of these inhibitors as a therapeutic strategy. Here, we discuss the mechanisms of resistance to FLT3 inhibitors and possible strategies to overcome resistance through closer examination of the events of leukemogenesis and design of combination therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/enzymology
- Protein Kinase Inhibitors/pharmacokinetics
- Protein Kinase Inhibitors/therapeutic use
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- S. Haihua Chu
- Department of Oncology, Johns Hopkins University School of Medicine, CRB1-251, 1650 Orleans St., Baltimore, MD 21231-1000, United States
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, CRB1-251, 1650 Orleans St., Baltimore, MD 21231-1000, United States
| | - Donald Small
- Department of Oncology, Johns Hopkins University School of Medicine, CRB1-251, 1650 Orleans St., Baltimore, MD 21231-1000, United States
- Department of Pediatrics, Johns Hopkins University School of Medicine, CRB1-251, 1650 Orleans St., Baltimore, MD 21231-1000, United States
| |
Collapse
|
19
|
Kasper S, Breitenbuecher F, Hoehn Y, Heidel F, Lipka DB, Markova B, Huber C, Kindler T, Fischer T. The kinase inhibitor LS104 induces apoptosis, enhances cytotoxic effects of chemotherapeutic drugs and is targeting the receptor tyrosine kinase FLT3 in acute myeloid leukemia. Leuk Res 2008; 32:1698-708. [DOI: 10.1016/j.leukres.2008.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 04/29/2008] [Accepted: 05/03/2008] [Indexed: 02/01/2023]
|
20
|
Trowe T, Boukouvala S, Calkins K, Cutler RE, Fong R, Funke R, Gendreau SB, Kim YD, Miller N, Woolfrey JR, Vysotskaia V, Yang JP, Gerritsen ME, Matthews DJ, Lamb P, Heuer TS. EXEL-7647 inhibits mutant forms of ErbB2 associated with lapatinib resistance and neoplastic transformation. Clin Cancer Res 2008; 14:2465-75. [PMID: 18413839 DOI: 10.1158/1078-0432.ccr-07-4367] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Mutations associated with resistance to kinase inhibition are an important mechanism of intrinsic or acquired loss of clinical efficacy for kinase-targeted therapeutics. We report the prospective discovery of ErbB2 mutations that confer resistance to the small-molecule inhibitor lapatinib. EXPERIMENTAL DESIGN We did in vitro screening using a randomly mutagenized ErbB2 expression library in Ba/F3 cells, which were dependent on ErbB2 activity for survival and growth. RESULTS Lapatinib resistance screens identified mutations at 16 different ErbB2 amino acid residues, with 12 mutated amino acids mapping to the kinase domain. Mutations conferring the greatest lapatinib resistance cluster in the NH2-terminal kinase lobe and hinge region. Structural computer modeling studies suggest that lapatinib resistance is caused by multiple mechanisms; including direct steric interference and restriction of conformational flexibility (the inactive state required for lapatinib binding is energetically unfavorable). ErbB2 T798I imparts the strongest lapatinib resistance effect and is analogous to the epidermal growth factor receptor T790M, ABL T315I, and cKIT T670I gatekeeper mutations that are associated with clinical drug resistance. ErbB2 mutants associated with lapatinib resistance transformed NIH-3T3 cells, including L755S and T733I mutations known to occur in human breast and gastric carcinomas, supporting a direct mechanism for lapatinib resistance in ErbB2-driven human cancers. The epidermal growth factor receptor/ErbB2/vascular endothelial growth factor receptor inhibitor EXEL-7647 was found to inhibit almost all lapatinib resistance-associated mutations. Furthermore, no ErbB2 mutations were found to be associated with EXEL-7647 resistance and lapatinib sensitivity. CONCLUSIONS Taken together, these data suggest potential target-based mechanisms of resistance to lapatinib and suggest that EXEL-7647 may be able to circumvent these effects.
Collapse
|
21
|
Abstract
The biological picture of cancer is rapidly advancing from models built from phenomenological descriptions to network models derived from systems biology, which can capture the evolving pathophysiology of the disease at the molecular level. The translation of this (still academic) picture into a clinically relevant framework can be enabling for the war on cancer, but it is a scientific and technological challenge. In this review, we discuss emerging in vitro diagnostic technologies and therapeutic approaches that are being developed to handle this challenge. Our discussion of in vitro diagnostics is guided by the theme of making large numbers of measurements accurately, sensitively, and at very low cost. We discuss diagnostic approaches based on microfluidics and nanotechnology. We then review the current state of the art of nanoparticle-based therapeutics that have reached the clinic. The goal of the presentation is to identify nanotherapeutic strategies that are designed to increase efficacy while simultaneously minimizing the toxic side effects commonly associated with cancer chemotherapies.
Collapse
Affiliation(s)
- James R Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA.
| | | |
Collapse
|
22
|
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of neoplastic disorders with great variability in clinical course and response to therapy, as well as in the genetic and molecular basis of the pathology. Major advances in the understanding of leukemogenesis have been made by the characterization and the study of acquired cytogenetic abnormalities, particularly reciprocal translocations observed in AML. Besides these major cytogenetic abnormalities, gene mutations also constitute key events in AML pathogenesis. In this review, we describe the contribution of known gene mutations to the understanding of AML pathogenesis and their clinical significance. To gain more insight in this understanding, we clustered these alterations in three groups: (1) mutations affecting genes that contribute to cell proliferation (FLT3, c-KIT, RAS, protein tyrosine standard phosphatase nonreceptor 11); (2) mutations affecting genes involved in myeloid differentiation (AML1 and CEBPA) and (3) mutations affecting genes implicated in cell cycle regulation or apoptosis (P53, NPM1). This nonexhaustive review aims to show how gene mutations interact with each other, how they contribute to refine prognosis and how they can be useful for risk-adapted therapeutic management of AML patients.
Collapse
|
23
|
Abstract
Abnormalities of cytokine and growth factor signaling pathways are characteristic of all forms of leukemia: lymphoid and myeloid, acute and chronic. In normal hematopoietic cells, cytokines provide the stimulus for proliferation, survival, self-renewal, differentiation and functional activation. In leukemic cells, these pathways are usurped to subserve critical parts of the malignant program. In this review, our current knowledge of leukemic cell cytokine signaling will be summarized, and some speculations on the significance and implications of these insights will be advanced. A better understanding of aberrant cytokine signaling in leukemia should provide additional targets for the rational therapy of these diseases.
Collapse
Affiliation(s)
- R A Van Etten
- Molecular Oncology Research Institute and Division of Hematology/Oncology, Tufts-New England Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
24
|
Gaikwad A, Prchal JT. Study of two tyrosine kinase inhibitors on growth and signal transduction in polycythemia vera. Exp Hematol 2007; 35:1647-56. [PMID: 17976517 PMCID: PMC2206678 DOI: 10.1016/j.exphem.2007.08.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 07/20/2007] [Accepted: 08/07/2007] [Indexed: 01/23/2023]
Abstract
OBJECTIVE An activating somatic mutation of Janus kinase 2 V617F (JAK2V617F) is present in most polycythemia vera (PV) patients. We studied efficacy of two potent tyrosine kinase inhibitors (TKI), AEE788 and AMN107, in vitro on cells bearing this mutation. MATERIALS AND METHODS We employed reporter cells expressing wild-type JAK2 and mutant JAK2V617F, human erythroleukemic cells (HEL) carrying JAK2V617F)to study the efficacy of these TKIs by cell proliferation assay, Annexin-V/propidium iodide staining, and on relevant cell-signaling and apoptotic events. These data were compared to ex vivo expanded native human erythroid PV progenitor cells grown in liquid cultures. RESULTS AEE788 showed a time- and dose-dependent growth inhibitory effect that was greater in FDCP cells expressing JAK2V617F and HEL cells than in cells expressing wild-type JAK2. AEE788 caused dephosphorylation of Akt(S243) and signal transducer and activator of transcription 5(Y694) proteins, increase in Annexin-V binding and caspase-3 cleavage, suggesting induction of apoptosis. We also observed AEE788-mediated decrease in heat shock protein 70 and 90 antiapoptotic proteins. Similarly, native PV erythroid progenitors showed more sensitivity to AEE788 than normal erythroid progenitors. AEE788 also exerted dose-dependent inhibition of PV-specific erythroid colonies. Nilotinib (AMN107) however, lacked specificity and required high (>8 microM) concentrations to inhibit growth of JAK2V617F-carrying cells. CONCLUSION Our data suggest that AEE788 exerts its apoptotic activity via downregulation of proliferative and antiapoptotic regulatory proteins. To our knowledge, this is the first report demonstrating the effect of AEE788 on PV erythroid progenitors. Differential effects on PV and normal progenitor cells suggest AEE788 has potential in the treatment of PV and other JAK2V617F positive hematologic malignancies.
Collapse
Affiliation(s)
- Amos Gaikwad
- Department of Pediatric-Hematology-Oncology, Texas Children’s Cancer Center, Baylor College of Medicine
| | - Josef T Prchal
- School of Medicine, Hematology Division, University of Utah
| |
Collapse
|
25
|
Lee BH, Tothova Z, Levine RL, Anderson K, Buza-Vidas N, Cullen DE, McDowell EP, Adelsperger J, Fröhling S, Huntly BJ, Beran M, Jacobsen SE, Gilliland DG. FLT3 mutations confer enhanced proliferation and survival properties to multipotent progenitors in a murine model of chronic myelomonocytic leukemia. Cancer Cell 2007; 12:367-80. [PMID: 17936561 PMCID: PMC2104473 DOI: 10.1016/j.ccr.2007.08.031] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Revised: 07/09/2007] [Accepted: 08/29/2007] [Indexed: 11/27/2022]
Abstract
Despite their known transforming properties, the effects of leukemogenic FLT3-ITD mutations on hematopoietic stem and multipotent progenitor cells and on hematopoietic differentiation are not well understood. We report a mouse model harboring an ITD in the murine Flt3 locus that develops myeloproliferative disease resembling CMML and further identified FLT3-ITD mutations in a subset of human CMML. These findings correlated with an increase in number, cell cycling, and survival of multipotent stem and progenitor cells in an ITD dose-dependent manner in animals that exhibited alterations within their myeloid progenitor compartments and a block in normal B cell development. This model provides insights into the consequences of constitutive signaling by an oncogenic tyrosine kinase on hematopoietic progenitor quiescence, function, and cell fate.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Proliferation
- Cell Survival
- Cells, Cultured
- Exons
- Gene Expression Regulation, Neoplastic
- Genotype
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Kaplan-Meier Estimate
- Leukemia, Experimental/metabolism
- Leukemia, Experimental/pathology
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/metabolism
- Leukemia, Myelomonocytic, Chronic/mortality
- Leukemia, Myelomonocytic, Chronic/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Multipotent Stem Cells/metabolism
- Multipotent Stem Cells/pathology
- Mutation
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/metabolism
- Myeloproliferative Disorders/pathology
- Phenotype
- Signal Transduction
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- Benjamin H. Lee
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: D. Gary Gilliland, MD, PhD, Karp Family Research Building, 1 Blackfan Circle; 5th Floor, Boston, MA 02115 USA, 617-355-9092 (tel), 617-355-9093 (fax), or Benjamin H. Lee, MD, PhD, Karp Family Research Building, 1 Blackfan Circle; 5th Floor, Boston, MA 02115 USA, 617-355-9085 (tel), 617-355-9093 (fax),
| | - Zuzana Tothova
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ross L. Levine
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kristina Anderson
- Hematopoietic Stem Cell Laboratory, Lund Stem Cell Center, Lund University, Biomedical Center, B10, Klinikgatan 26, 221 84 Lund, Sweden
| | - Natalija Buza-Vidas
- Hematopoietic Stem Cell Laboratory, Lund Stem Cell Center, Lund University, Biomedical Center, B10, Klinikgatan 26, 221 84 Lund, Sweden
| | - Dana E. Cullen
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth P. McDowell
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer Adelsperger
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stefan Fröhling
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Brian J.P. Huntly
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 OXY, UK
| | - Miloslav Beran
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sten Eirik Jacobsen
- Hematopoietic Stem Cell Laboratory, Lund Stem Cell Center, Lund University, Biomedical Center, B10, Klinikgatan 26, 221 84 Lund, Sweden
| | - D. Gary Gilliland
- Division of Hematology, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: D. Gary Gilliland, MD, PhD, Karp Family Research Building, 1 Blackfan Circle; 5th Floor, Boston, MA 02115 USA, 617-355-9092 (tel), 617-355-9093 (fax), or Benjamin H. Lee, MD, PhD, Karp Family Research Building, 1 Blackfan Circle; 5th Floor, Boston, MA 02115 USA, 617-355-9085 (tel), 617-355-9093 (fax),
| |
Collapse
|
26
|
Mrózek K, Paschka P, Marcussi G, Whitman S, Bloomfield C. E01 Biology, cytogenetics and molecular studies in acute myeloid leukemia. Leuk Res 2007; 31 Suppl 2:S3-117. [DOI: 10.1016/s0145-2126(07)70266-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
27
|
Abstract
FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase that appears to play a significant role in leukaemogenesis. Activating mutations of FLT3 are present in approximately one-third of acute myeloid leukaemia patients and are associated with adverse clinical outcome, while many non-mutated cases also show evidence of FLT3 activation. FLT3 thus represents a potentially exciting molecular therapeutic target. A number of small-molecule tyrosine kinase inhibitors with anti-FLT3 activity have been developed and several of these compounds have entered early phase clinical trials where clinical anti-leukaemic activity has been demonstrated. The depth and duration of clinical responses to FLT3 inhibitor monotherapy have been modest, however, and a number of mechanisms by which blasts may acquire resistance have been proposed. Based on preclinical evidence of synergy with conventional chemotherapy, several combination trials are now underway. FLT3 inhibition may also be effective used in combination with other molecularly targeted agents, in postchemotherapy stem-cell-directed maintenance therapy and in MLL-rearranged infant acute lymphoblastic leukaemia.
Collapse
Affiliation(s)
- Steven Knapper
- Department of Haematology, Cardiff University, Heath Park, Cardiff CF14 4XW, UK.
| |
Collapse
|
28
|
Gaikwad A, Verstovsek S, Yoon D, Chang KT, Manshouri T, Nussenzveig R, Cortes J, Vainchenker W, Prchal JT. Imatinib effect on growth and signal transduction in polycythemia vera. Exp Hematol 2007; 35:931-8. [PMID: 17533047 DOI: 10.1016/j.exphem.2007.03.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2006] [Revised: 02/20/2007] [Accepted: 03/12/2007] [Indexed: 11/28/2022]
Abstract
OBJECTIVE An activating mutation of Janus kinase 2 (JAK2) in majority of polycythemia vera (PV) and other myeloproliferative disorders was reported. As imatinib inhibits several tyrosine kinases, we studied its effect in PV. PATIENTS AND METHODS We employed FDCP reporter cells expressing wild-type JAK2 and mutant JAK2(V617F) to study the efficacy of imatinib by cell proliferation assay and its effect on several cell-signaling events. Imatinib's efficacy was also examined on in vitro expanded native human erythroid progenitors. In addition, analysis of the percent JAK2 T-allele and phospho-signal transducer and activator of transcription-5 (STAT5) in granulocytes of PV patients following imatinib therapy was assessed. RESULTS Imatinib showed a specific time- and dose-dependent growth inhibitory effect on FDCP cells expressing JAK2(V617F), wherein we observed imatinib's inactivation of JAK2, STAT5 and cKIT proteins. In vitro expanded human PV erythroid progenitors were more sensitive to imatinib than normal erythroid progenitors and FDCP cells expressing JAK2(V617F), with growth inhibition at concentrations attainable in vivo. In an ongoing clinical study, a PV patient showed strong correlation between the percent JAK2 T-allele and his responsiveness to imatinib therapy. CONCLUSION Our data elucidate the therapeutic benefit of imatinib seen in some PV patients. Our data suggest that JAK2/STAT5 and cKIT activation may be integrated. To our knowledge, this is the first report demonstrating imatinib's effect on PV erythroid progenitors. These studies underscore the limitation of experiments using cell lines expressing the gene of interest.
Collapse
Affiliation(s)
- Amos Gaikwad
- Department of Pediatric-Hematology-Oncology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
A highly sensitive method for the detection of PKC412 (CGP41251) and its metabolites by high-performance liquid chromatography. J Pharmacol Toxicol Methods 2007; 56:23-7. [DOI: 10.1016/j.vascn.2006.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2006] [Accepted: 11/20/2006] [Indexed: 11/23/2022]
|
30
|
Gaikwad A, Nussenzveig R, Liu E, Gottshalk S, Chang K, Prchal JT. In vitro expansion of erythroid progenitors from polycythemia vera patients leads to decrease in JAK2 V617F allele. Exp Hematol 2007; 35:587-95. [PMID: 17379069 PMCID: PMC1931508 DOI: 10.1016/j.exphem.2006.12.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Revised: 12/20/2006] [Accepted: 12/20/2006] [Indexed: 11/16/2022]
Abstract
OBJECTIVES A G>T transversion in a tyrosine kinase JAK2 (V617F) was reported in over 80% of patients with polycythemia vera (PV). Current evidence suggests that JAK2(V617F) somatic mutation is involved in the pathogenesis of PV, as it confers erythropoietin-independent proliferation to erythroid progenitor cells. However, several unanswered questions regarding the essential role of JAK2(V617F) arose as 1) it is not a dominant mutation, 2) it is not PV-specific as it is found in several myeloproliferative disorders, and 3) some ( approximately 20%) PV patients lack the JAK2(V617F) mutation. We investigated the relative frequency of JAK2(V617F) in in vitro-expanded PV progenitors. METHODS In vitro expansion of erythroid progenitors from mononuclear cells was optimized. Frequency of JAK2(V617F) allele was measured by using allele-specific real-time polymerase chain reaction. Clonality was performed using established procedure. RESULTS In vitro expansion of PV erythroid progenitors and differentiated dendritic cells resulted in a decrease of the frequency of JAK2(V617F) allele compared with granulocytes or CD235(+) erythroid progenitors. Clonality analysis demonstrated that although granulocytes of these PV patients were clonal, expanded erythroid cells were polyclonal. However, in vitro-expanded PV erythroid progenitors still had approximately a twofold increased proliferative capacity in comparison with erythroid progenitors from healthy individuals. Erythropoietin favors the cells without JAK2(V617F) allele. Dendritic cells in one out of three patients remained clonal. CONCLUSION JAK2(V617F) mutation does not provide a proliferative/survival advantage to the PV clone during in vitro expansion. These data suggest that the JAK2(V617F) mutation plays an important role in the biology of PV, yet it may not be the PV-initiating event.
Collapse
Affiliation(s)
- Amos Gaikwad
- Department of Pediatric-Hematology Oncology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | - Enli Liu
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Stephen Gottshalk
- Department of Pediatric-Hematology Oncology, Baylor College of Medicine, Houston, Texas
- Department of Immunology, Baylor College of Medicine, Houston, Texas
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, Texas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - KoTung Chang
- Division of Hematology, University of Utah, Salt Lake City, Utah
| | - Josef T. Prchal
- Institute of Pathological Physiology, Charles University School of Medicine, Prague, Czech Republic
- Division of Hematology, University of Utah, Salt Lake City, Utah
- *Address correspondence and reprint requests to: Josef T. Prchal MD, School of Medicine, Hematology, University of Utah, Salt Lake City, UT 84124, , Phone: 801 581- 4220 (Office)
| |
Collapse
|
31
|
Prindull G. Hemangioblasts representing a functional endothelio-hematopoietic entity in ontogeny, postnatal life, and CML neovasculogenesis. ACTA ACUST UNITED AC 2007; 1:277-84. [PMID: 17142866 DOI: 10.1385/scr:1:3:277] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The life-long interdependencies/interactions between hemato- and endotheliopoiesis suggest that they form a supplementary functional entity. This view is compatible with the concept of stem cell plasticity as a reversible continuum and is substantiated by the common hematopoietic-endothelial stem cell, i.e., hemangioblasts, with bidirectional, reversible gene transcription and persistence in postnatal life. Indeed, embryonal stem cells/hemangioblasts appear to form a reservior in the adult with the possibility of dedifferentiation of more differentiated progenitor cells back to hemangioblasts. The recent detection of BCR/ABL fusion proteins in endothelial cells during vascular neoangiogenesis in CML suggests that endothelial cells are part of the neoplastic clone, and extends the concept of a functional entity to include CML angiogenesis. Thus, hemangioblasts rather than committed hematopoietic stem cells appear to be target cells for the first oncogenic hit in CML, which could occur as early as during the first steps of embryonal stem cell differentiation towards hemato-endotheliopoiesis and/or in hemangioblasts persisting in adults. The relation of the other leukemias to hemangioblasts is not known.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Embryonic Stem Cells/metabolism
- Embryonic Stem Cells/pathology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Fusion Proteins, bcr-abl
- Gene Expression Regulation, Leukemic
- Hematopoiesis
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
Collapse
Affiliation(s)
- Gregor Prindull
- Pediatric Hematology/Oncology, University of Göttingen, Germany.
| |
Collapse
|
32
|
Mrózek K, Marcucci G, Paschka P, Whitman SP, Bloomfield CD. Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification? Blood 2007; 109:431-48. [PMID: 16960150 PMCID: PMC1785102 DOI: 10.1182/blood-2006-06-001149] [Citation(s) in RCA: 408] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Accepted: 08/20/2006] [Indexed: 12/17/2022] Open
Abstract
Recent molecular analyses of leukemic blasts from pretreatment marrow or blood of patients with acute myeloid leukemia (AML) and a normal karyotype, the largest cytogenetic subset (ie, 40%-49%) of AML, have revealed a striking heterogeneity with regard to the presence of acquired gene mutations and changes in gene expression. Multiple submicroscopic genetic alterations with prognostic significance have been discovered, including internal tandem duplication of the FLT3 gene, mutations in the NPM1 gene, partial tandem duplication of the MLL gene, high expression of the BAALC gene, and mutations in the CEBPA gene. Application of gene-expression profiling has also identified a gene-expression signature that appears to separate cytogenetically normal AML patients into prognostic subgroups, although gene-expression signature-based classifiers predicting outcome for individual patients with greater accuracy are needed. These and similar future findings are likely to have a major impact on the clinical management of cytogenetically normal AML not only in prognostication but also in selection of appropriate treatment, since many of the identified genetic alterations already constitute or will potentially become targets for specific therapeutic intervention. In this report, we review prognostic genetic findings in karyotypically normal AML and discuss their clinical implications.
Collapse
Affiliation(s)
- Krzysztof Mrózek
- Department of Internal Medicine, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Room 1248B, The Ohio State University, 300 West Tenth Ave, Columbus, OH 43210-1228, USA.
| | | | | | | | | |
Collapse
|
33
|
Ndassa YM, Orsi C, Marto JA, Chen S, Ross MM. Improved immobilized metal affinity chromatography for large-scale phosphoproteomics applications. J Proteome Res 2006; 5:2789-99. [PMID: 17022650 DOI: 10.1021/pr0602803] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dysregulated protein phosphorylation is a primary culprit in multiple physiopathological states. Hence, although analysis of signaling cascades on a proteome-wide scale would provide significant insight into both normal and aberrant cellular function, such studies are simultaneously limited by sheer biological complexity and concentration dynamic range. In principle, immobilized metal affinity chromatography (IMAC) represents an ideal enrichment method for phosphoproteomics. However, anecdotal evidence suggests that this technique is not widely and successfully applied beyond analysis of simple standards, gel bands, and targeted protein immunoprecipitations. Here, we report significant improvements in IMAC-based methodology for enrichment of phosphopeptides from complex biological mixtures. Moreover, we provide detailed explanation for key variables that in our hands most influenced the outcome of these experiments. Our results indicate 5- to 10-fold improvement in recovery of singly- and multiply phosphorylated peptide standards in addition to significant improvement in the number of high-confidence phosphopeptide sequence assignments from global analysis of cellular lysate. In addition, we quantitatively track phosphopeptide recovery as a function of phosphorylation state, and provide guidance for impedance-matching IMAC column capacity with anticipated phosphopeptide content of complex mixtures. Finally, we demonstrate that our improved methodology provides for identification of phosphopeptide distributions that closely mimic physiological conditions.
Collapse
Affiliation(s)
- Yasmine M Ndassa
- Biophysics Program, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
34
|
Knapper S, Mills KI, Gilkes AF, Austin SJ, Walsh V, Burnett AK. The effects of lestaurtinib (CEP701) and PKC412 on primary AML blasts: the induction of cytotoxicity varies with dependence on FLT3 signaling in both FLT3-mutated and wild-type cases. Blood 2006; 108:3494-503. [PMID: 16868253 DOI: 10.1182/blood-2006-04-015487] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe receptor tyrosine kinase FLT3 is a promising molecular therapeutic target in acute myeloid leukemia (AML). Activating mutations of FLT3 are present in approximately one-third of patients, while many nonmutants show evidence of FLT3 activation, which appears to play a significant role in leukemogenesis. We studied the effects of lestaurtinib (CEP701) and PKC412, 2 small molecule inhibitors of FLT3, on 65 diagnostic AML blast samples. Both agents induced concentration-dependent cytotoxicity in most cases, although responses to PKC412 required higher drug concentrations. Cytotoxic responses were highly heterogeneous and were only weakly associated with FLT3 mutation status and FLT3 expression. Importantly, lestaurtinib induced cytotoxicity in a synergistic fashion with cytarabine, particularly in FLT3 mutant samples. Both lestaurtinib and PKC412 caused inhibition of FLT3 phosphorylation in all samples. Translation of FLT3 inhibition into cytotoxicity was influenced by the degree of residual FLT3 phosphorylation remaining and correlated with deactivation of STAT5 and MAP kinase. FLT3 mutant and wild-type cases both varied considerably in their dependence on FLT3 signaling for survival. These findings support the continued clinical assessment of FLT3 inhibitors in combination with cytotoxic chemotherapy: Entry to future clinical trials should include FLT3 wild-type patients and should remain unrestricted by FLT3 expression level.
Collapse
Affiliation(s)
- Steven Knapper
- Department of Haematology, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XW, United Kingdom.
| | | | | | | | | | | |
Collapse
|
35
|
Remacha AF, Puget G, Nomdedéu JF, Estivill C, Sardà MP, Canals C. [Evaluation of V617F mutation of JAK2 in negative chromosome Philadelphia chronic myeloproliferative disorders]. Med Clin (Barc) 2006; 127:601-4. [PMID: 17145024 DOI: 10.1157/13094416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Polycythemia vera (PV) and essential thrombocytemia (ET) are chronic myeloproliferative diseases (MPD) characterized by overactive hemopoiesis. A single point mutation of JAK2 (Val617Phe) has been detected in PV, ET and myelofibrosis (MF). The aim of this work was to investigate the JAK2 mutation in patients with MPD and to compare the results to those of the endogenous formation of BFU-E erythroid colonies (EEC). Finally, different sources of hematopoietic cells to obtain DNA were evaluated. PATIENTS AND METHOD In this work 146 patents were studied (81 MPD: 27 PV, 28 ET, 11 MF and 15 with myeloid chronic leukemia). Moreover, 28 patients showed secondary polycythemias or reactive thrombocytosis, 8 MPD/myelodysplastic syndromes and 29 other disorders. In 54 patients, EEC were also evaluated. Peripheral blood cells were used as source of DNA in 122 patients, bone marrow in 33, cells from BFU-E in 14 and cells from EEC in 24 patients. Their DNA samples were analyzed using an allele-specific polimerase chain reaction methodology. RESULTS The JAK2 mutation was present in 96% of PV patients, 59% of ET and 63.6% of MF. None of the remaining patients showed this mutation. Diagnostic agreement was excellent between EEC and the mutation (kappa index = 0.93; 97% positive agreement and 95% negative agreement). DNA was obtained in 119 out of 122 samples from peripheral blood, in all patients with bone marrow, and in 50% of patients with BFU-E or EEC. In 7 cases, samples from different cell sources were studied. Their results were identical. CONCLUSIONS The V617F mutation of JAK2 is present in most of PV patients and half of those with MF or ET. There is an excellent concordance with the EEC results.
Collapse
Affiliation(s)
- Angel F Remacha
- Departamento de Hematología, Hospital de Sant Pau, Barcelona, España.
| | | | | | | | | | | |
Collapse
|
36
|
Schnittger S, Kohl TM, Haferlach T, Kern W, Hiddemann W, Spiekermann K, Schoch C. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood 2006; 107:1791-9. [PMID: 16254134 DOI: 10.1182/blood-2005-04-1466] [Citation(s) in RCA: 267] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mutations in codon D816 of the KIT gene represent a recurrent genetic alteration in acute myeloid leukemia (AML). To clarify the biologic implication of activation loop mutations of the KIT gene, 1940 randomly selected AML patients were analyzed. In total, 33 (1.7%) of 1940 patients were positive for D816 mutations. Of these 33 patients, 8 (24.2%) had a t(8;21), which was significantly higher compared with the subgroup without D816 mutations. Analyses of genetic subgroups showed that KIT-D816 mutations were associated with t(8;21)/AML1-ETO and other rare AML1 translocations. In contrast, other activating mutations like FLT3 and NRAS mutations were very rarely detected in AML1-rearranged leukemia. KIT mutations had an independent negative impact on overall (median 304 vs 1836 days; P = .006) and event-free survival (median 244 vs 744 days; P = .003) in patients with t(8;21) but not in patients with a normal karyotype. The KIT-D816V receptor expressed in Ba/F3 cells was resistant to growth inhibition by the selective PTK inhibitors imatinib and SU5614 but fully sensitive to PKC412. Our findings clearly indicate that activating mutations of receptor tyrosine kinases are associated with distinct genetic subtypes in AML. The KIT-D816 mutations confer a poor prognosis to AML1-ETO-positive AML and should therefore be included in the diagnostic workup. Patients with KIT-D816-positive/AML1-ETO-positive AML might benefit from early intensification of treatment or combination of conventional chemotherapy with KIT PTK inhibitors.
Collapse
MESH Headings
- Adult
- Aged
- Amino Acid Substitution
- Cell Line
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 8/genetics
- Codon/genetics
- Core Binding Factor Alpha 2 Subunit/genetics
- Disease-Free Survival
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression/genetics
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Male
- Middle Aged
- Oncogene Proteins, Fusion/genetics
- Point Mutation
- Prognosis
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-kit/genetics
- RUNX1 Translocation Partner 1 Protein
- Retrospective Studies
- Translocation, Genetic/genetics
Collapse
Affiliation(s)
- Susanne Schnittger
- Laboratory of Leukemia Diagnostics and Clinical Cooperative Group Leukemia, Department of Medicine III, University Hospital Grosshadern, Ludwig-Maximilians University, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Advances in molecular genetics have revolutionized our understanding of acute myeloid and lymphoblastic leukemia. Structural and numerical chromosomal aberrations are common, and their detection is vital for leukemia diagnosis, risk stratification, and monitoring of response to therapy. Fusion proteins resulting from chromosomal translocations are necessary but not sufficient for leukemogenesis, and there is intense research activity to elucidate the cooperating molecular abnormalities that may be suitable targets for novel therapeutic approaches. Candidate gene approaches have identified mutations in kinases and transcription factors in a proportion of patients, but more comprehensive genomic approaches are required. Gene expression profiling accurately classifies known subtypes of acute leukemia and has highlighted potentially leukemogenic abnormalities in gene expression. Newer techniques, such as single-nucleotide polymorphism arrays to analyze changes in gene copy number and zygosity, cancer genome sequencing, and RNA interference, are promising tools to identify mutations, although at present, data from these approaches are limited. This review provides an overview of these techniques in clinical practice and as research tools to develop new therapeutic approaches in pediatric leukemia.
Collapse
Affiliation(s)
- Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | |
Collapse
|
38
|
Song KW, Lipton J. Is it appropriate to offer allogeneic hematopoietic stem cell transplantation to patients with primary refractory acute myeloid leukemia? Bone Marrow Transplant 2005; 36:183-91. [PMID: 15937497 DOI: 10.1038/sj.bmt.1705038] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although continued advances have been made in the treatment of acute myeloid leukemia (AML), approximately 20-30% of patients will never achieve a remission. For these patients with primary refractory AML, the only curative option remains an allogeneic stem cell transplant. Allogeneic transplantation provides the ability to administer myeloablative doses of chemotherapy or chemoradiotherapy, as well as the advantage of a possible graft-versus-leukemia effect. Difficulty in interpreting the literature is due to selection bias, in particular, the varying definitions of primary refractory disease with respect to the morphological criteria and the number of induction regimen required before being defined as being refractory. Regardless, it is a procedure with high treatment-related mortality and risk of relapse. Most studies demonstrate an event-free survival of 10-20% at 5 years. Predictive factors of outcome include blast cell count in the marrow, karyotype, the number of prior regimen, age, performance status and availability of a related donor. These prognostic factors should be considered prior to offering allogeneic transplantation for primary refractory AML. Those patients with many favorable prognostic factors and an HLA-matched related donor available would be the best candidate for the procedure. Those with many poor prognostic factors and only an unrelated donor available may be better served by being offered palliation or being enrolled in investigational studies.
Collapse
Affiliation(s)
- K W Song
- The Leukemia/Bone Marrow Transplantation Program of British Columbia, Vancouver Hospital and Health Sciences Center, British Columbia Cancer Agency, University of British Columbia, Vancouver, British Columbia, Canada.
| | | |
Collapse
|
39
|
Haas M, Lonial S. Targeted therapy for haematological malignancies: clinical update from the American Society of Hematology, 2004. Expert Opin Investig Drugs 2005; 14:1161-9. [PMID: 16144500 DOI: 10.1517/13543784.14.9.1161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Recent advances in the treatment and management of haematological malignancies are due in large part to an improved understanding of the basic biology that drives tumour cell growth and survival. This improved understanding has led to the clinical study and approval of a number of different targeted agents across a number of different haematological tumours. This review of clinical data covers some of the exciting clinical advances that were reported at the recent American Society of Hematology meeting in San Diego, USA. This paper focuses on three important areas of biological research that has yielded clinical trials that have affected clinical outcomes. The areas covered include proteasome inhibition and myeloma, tyrosine kinase inhibitors that are directed at the BCR-ABL fusion protein and chronic myeloid leukaemia/acute lymphoblastic leukaemia, and FLT3 inhibitors and acute myeloid leukaemia acute lymphoblastic leukaemia therapy.
Collapse
Affiliation(s)
- Maureen Haas
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
40
|
Abstract
Antibodies represent a unique class of therapeutics because of their high specificity toward a defined target antigen. Recent clinical success with antibody-based cancer therapeutics has led to an upsurge in the development of these agents. Antibodies directed against FLT3 represent a promising approach for the treatment of human leukemia. We discuss some basic aspects of antibody-based cancer therapeutics, including their mechanisms of action, with a focus on recent progress in the generation and development of anti-FLT3 antibodies as well as their therapeutic potentials in the treatment of human hematologic malignancies.
Collapse
Affiliation(s)
- Yiwen Li
- ImClone Systems Incorporated, New York, NY 10014, USA.
| | | |
Collapse
|
41
|
Carter TA, Wodicka LM, Shah NP, Velasco AM, Fabian MA, Treiber DK, Milanov ZV, Atteridge CE, Biggs WH, Edeen PT, Floyd M, Ford JM, Grotzfeld RM, Herrgard S, Insko DE, Mehta SA, Patel HK, Pao W, Sawyers CL, Varmus H, Zarrinkar PP, Lockhart DJ. Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases. Proc Natl Acad Sci U S A 2005; 102:11011-6. [PMID: 16046538 PMCID: PMC1180625 DOI: 10.1073/pnas.0504952102] [Citation(s) in RCA: 412] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To realize the full potential of targeted protein kinase inhibitors for the treatment of cancer, it is important to address the emergence of drug resistance in treated patients. Mutant forms of BCR-ABL, KIT, and the EGF receptor (EGFR) have been found that confer resistance to the drugs imatinib, gefitinib, and erlotinib. The mutations weaken or prevent drug binding, and interestingly, one of the most common sites of mutation in all three kinases is a highly conserved "gatekeeper" threonine residue near the kinase active site. We have identified existing clinical compounds that bind and inhibit drug-resistant mutant variants of ABL, KIT, and EGFR. We found that the Aurora kinase inhibitor VX-680 and the p38 inhibitor BIRB-796 inhibit the imatinib- and BMS-354825-resistant ABL(T315I) kinase. The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. EKB-569 and CI-1033 are already in clinical trials, and our results suggest that they should be considered for testing in the treatment of gefitinib/erlotinib-resistant non-small cell lung cancer. The results highlight the strategy of screening existing clinical compounds against newly identified drug-resistant mutant variants to find compounds that may serve as starting points for the development of next-generation drugs, or that could be used directly to treat patients that have acquired resistance to first-generation targeted therapy.
Collapse
Affiliation(s)
- Todd A Carter
- Ambit, Inc., 4215 Sorrento Valley Boulevard, San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Affiliation(s)
- Daniela S Krause
- Molecular Oncology Research Institute, Division of Hematology-Oncology, Tufts-New England Medical Center, Boston, MA 02111, USA
| | | |
Collapse
|
43
|
Ugo V, James C, Vainchenker W. Une mutation unique de la protéine kinase JAK2 dans la polyglobulie de Vaquez et les syndromes myéloprolifératifs non-LMC. Med Sci (Paris) 2005; 21:669-70. [PMID: 15985214 DOI: 10.1051/medsci/2005216-7669] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Valérie Ugo
- Inserm U.362, PR1, Institut Gustave Roussy, Villejuif, France.
| | | | | |
Collapse
|
44
|
Meyer-Monard S, Mühlematter D, Streit A, Chase AJ, Gratwohl A, Cross NCP, Jotterand M, Tichelli A. Broad molecular screening of an unclassifiable myeloproliferative disorder reveals an unexpected ETV6/ABL1 fusion transcript. Leukemia 2005; 19:1096-9. [PMID: 15789067 DOI: 10.1038/sj.leu.2403697] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Accepted: 01/07/2005] [Indexed: 11/08/2022]
|
45
|
Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, Scott MA, Erber WN, Green AR. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005; 365:1054-61. [PMID: 15781101 DOI: 10.1016/s0140-6736(05)71142-9] [Citation(s) in RCA: 2021] [Impact Index Per Article: 101.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Human myeloproliferative disorders form a range of clonal haematological malignant diseases, the main members of which are polycythaemia vera, essential thrombocythaemia, and idiopathic myelofibrosis. The molecular pathogenesis of these disorders is unknown, but tyrosine kinases have been implicated in several related disorders. We investigated the role of the cytoplasmic tyrosine kinase JAK2 in patients with a myeloproliferative disorder. METHODS We obtained DNA samples from patients with polycythaemia vera, essential thrombocythaemia, or idiopathic myelofibrosis. The coding exons of JAK2 were bidirectionally sequenced from peripheral-blood granulocytes, T cells, or both. Allele-specific PCR, molecular cytogenetic studies, microsatellite PCR, Affymetrix single nucleotide polymorphism array analyses, and colony assays were undertaken on subgroups of patients. FINDINGS A single point mutation (Val617Phe) was identified in JAK2 in 71 (97%) of 73 patients with polycythaemia vera, 29 (57%) of 51 with essential thrombocythaemia, and eight (50%) of 16 with idiopathic myelofibrosis. The mutation is acquired, is present in a variable proportion of granulocytes, alters a highly conserved valine present in the negative regulatory JH2 domain, and is predicted to dysregulate kinase activity. It was heterozygous in most patients, homozygous in a subset as a result of mitotic recombination, and arose in a multipotent progenitor capable of giving rise to erythroid and myeloid cells. The mutation was present in all erythropoietin-independent erythroid colonies. INTERPRETATION A single acquired mutation of JAK2 was noted in more than half of patients with a myeloproliferative disorder. Its presence in all erythropoietin-independent erythroid colonies demonstrates a link with growth factor hypersensitivity, a key biological feature of these disorders. RELEVANCE TO PRACTICE Identification of the Val617Phe JAK2 mutation lays the foundation for new approaches to the diagnosis, classification, and treatment of myeloproliferative disorders.
Collapse
Affiliation(s)
- E Joanna Baxter
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Licht JD, Sternberg DW. The molecular pathology of acute myeloid leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2005:137-42. [PMID: 16304371 DOI: 10.1182/asheducation-2005.1.137] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The pathogenesis of acute myelogenous leukemia (AML) involves an array of molecular alterations that disrupt almost every facet of cell transformation. These processes include the regulation of cell proliferation, differentiation, self-renewal, survival, cell cycle checkpoint control, DNA repair and chromatin stability, and cell dissemination. Normal regulatory networks are disrupted or usurped by these leukemogenic insults, and the understanding of these alterations is guiding the design of new therapeutic strategies. This overview describes some of the critical molecular alterations and implicates the rogue leukemogenic proteins in the onset and progression of AML.
Collapse
Affiliation(s)
- Jonathan D Licht
- Mt. Sinai School of Medicine, Division of Hematology/Oncology, One Gustave L. Levy Pl., Box 1079, New York, NY 10029-6504, USA.
| | | |
Collapse
|
47
|
Stone RM, O'Donnell MR, Sekeres MA. Acute myeloid leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2004; 2004:98-117. [PMID: 15561679 DOI: 10.1182/asheducation-2004.1.98] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Advances in our understanding of the pathophysiology of acute myeloid leukemia (AML) have not yet led to major improvements in disease-free and overall survival of adults with this disease. Only about one-third of those between ages 18-60 who are diagnosed with AML can be cured; disease-free survival is rare and current therapy devastating in older adults. In this chapter, challenges in the management of the adult with AML are discussed, including ongoing questions concerning the optimal choice of induction and postremission therapy such as the rationale for and role of allogeneic and autologous stem cell transplantation in a variety of settings, the special considerations pertaining to the older patient, and the development of new, so-called targeted therapies. In Section I, Dr. Richard Stone reviews state-of the-art therapy in AML in the era of change from a morphological to a genetically based classification system. Questions being addressed in ongoing randomized cooperative group trials include anthracycline dose during induction, the efficacy of drug-resistance modulators, and the utility of pro-apoptotic agents such as the anti-bcl-2 antisense oligonucloetide. Developmental therapeutics in AML include drug resistance modulation, anti-angiogenic strategies, immunotherapy, and signal transduction-active agents, particularly the farnesyl transferase inhibitors as well as those molecules that inhibit the FLT3 tyrosine kinase, activated via mutation in 30% of patients. In Section II Dr. Margaret O'Donnell discusses the role of stem cell transplantation in AML. Several advances including expanded donor pools, the movement toward peripheral blood stem cell collection, newer immunosuppressive drugs and antifungals, and particularly the advent of nonmyeloablative transplant have made the allogeneic option more viable. The subset-specific role for high-dose chemotherapy with autologous stem cell support and/or for allogeneic transplant in AML patients in first remission is outlined. Although preconceived notions about the role of transplant abound, the clinical data supporting a risk-adapted approach are covered. Finally, guidance concerning the use of nonmyeloablative or reduced-intensity allogeneic transplantation is provided. In Section III Dr. Mikkael Sekeres reviews the approach to the older patient with AML. Unique biological and therapeutic considerations make AML in this age group a vastly different disease than that in younger adults. The outcome data, including the role of specific anthracylines, hematopoietic growth factors, and drug-resistance modulators, are summarized. Communicating with older adults with AML and their families regarding selection of the optimal treatment strategy, often a stark choice between induction chemotherapy and palliative care, is covered.
Collapse
|