1
|
Sambo T, Mathe E, Shai L, Mapfumari S, Gololo S. Inhibition of Kinase Activity and In Vitro Downregulation of the Protein Kinases in Lung Cancer and Cervical Cancer Cell Lines and the Identified Known Anticancer Compounds of Ziziphus mucronata. PLANTS (BASEL, SWITZERLAND) 2025; 14:395. [PMID: 39942957 PMCID: PMC11820244 DOI: 10.3390/plants14030395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025]
Abstract
Plants have long been used as sources of natural compounds with therapeutic benefits, providing molecules capable of inhibiting multiple kinases. Many medicinal plants are recognized for their anticancer properties and may offer ways to mitigate the adverse effects of conventional cancer treatments. In this study, the potential of Ziziphus mucronata methanol extract as a kinase inhibitor was assessed using the MTT assay, a universal kinase assay, and a human phosphokinase antibody array, along with a GC-MS analysis of volatile anticancer compounds. The MTT assay revealed strong cytotoxicity in A549 cells, with an IC50 of 31.25 µg/mL, while HeLa cells showed weaker cytotoxicity with an IC50 of 125 µg/mL. In comparison, paclitaxel exhibited potent inhibitory effects on A549 cells (IC50 of 31.25 µg/mL) and moderate inhibition on HeLa cells (IC50 of 65 µg/mL). Enzyme activity, measured by ADP production in the ADP-Glo assay, indicated that the extract inhibited protein kinase activity in both A549 and HeLa cells after 24 h of treatment. Additionally, the human phosphokinase antibody array, which includes 44 pre-spotted kinases, showed that the extract downregulated multiple phosphorylated kinases in both cell lines. Some of the affected kinases, such as TOR, Fyn, HcK, Fgr, STAT5b, PLC-γ1, p38α, ERK1/2, AMPKA, Akt1/2, GSK-3α/β, MSK1/2, CREB, RSK1/2/3, PLC-γ1, and STAT5a are critical regulators of various cellular processes, including apoptosis, differentiation, and proliferation. The findings of this study suggest that extract from Z. mucronata may have the capacity to regulate protein kinase activity, highlighting their significant potential as growth inhibitors for cancer cells.
Collapse
Affiliation(s)
- Themba Sambo
- Department of Biochemistry & Biotechnology, School of Science and Technology, Sefako Makgatho Health Sciences University, Ga-Rankuwa 0208, South Africa; (E.M.); (S.G.)
| | - Emelinah Mathe
- Department of Biochemistry & Biotechnology, School of Science and Technology, Sefako Makgatho Health Sciences University, Ga-Rankuwa 0208, South Africa; (E.M.); (S.G.)
| | - Leswheni Shai
- Department of Biochemical Sciences, Tshwane University of Technology, Pretoria 0183, South Africa;
| | - Sipho Mapfumari
- Department of Physiology, School of Medicine, Sefako Makgatho Health Sciences University, Ga-Rankuwa 0208, South Africa
| | - Stanley Gololo
- Department of Biochemistry & Biotechnology, School of Science and Technology, Sefako Makgatho Health Sciences University, Ga-Rankuwa 0208, South Africa; (E.M.); (S.G.)
| |
Collapse
|
2
|
Nascimento M, Moura S, Parra L, Vasconcellos V, Costa G, Leite D, Dias M, Fernandes TVA, Hoelz L, Pimentel L, Bastos M, Boechat N. Ponatinib: A Review of the History of Medicinal Chemistry behind Its Development. Pharmaceuticals (Basel) 2024; 17:1361. [PMID: 39459001 PMCID: PMC11510555 DOI: 10.3390/ph17101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The primary treatment for chronic myeloid leukemia (CML) involves first- and second-generation tyrosine kinase inhibitors (TKIs), such as imatinib, nilotinib, bosutinib, and dasatinib. However, these medications are ineffective against mutations in the kinase domain of the ABL1 protein, particularly in the protein with the T315I mutation. To address this, ponatinib (PNT), a third-generation inhibitor, was developed. Despite its efficacy in treating the BCR-ABL1T315I mutation, the use of PNT was briefly suspended in 2013 due to serious adverse effects but was subsequently reintroduced to the market. During the drug discovery and development process, it is rare to consolidate all information into a single article, as is the case with ponatinib. This review aims to compile and chronologically organize the research on the discovery of ponatinib using medicinal chemistry tools and computational methods. It includes in silico calculations, such as the octanol/water partition coefficient (cLogP) via SwissAdme, and 2D maps of intermolecular interactions through molecular docking. This approach enhances understanding for both specialists and those interested in medicinal chemistry and pharmacology, while also contextualizing future directions for further optimizations of ponatinib, facilitating the development of new analogs of this crucial inhibitor for the treatment of CML and Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL).
Collapse
Affiliation(s)
- Mayara Nascimento
- Programa de Pós-Graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, RJ, Brazil; (M.N.); (S.M.)
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Stefany Moura
- Programa de Pós-Graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, RJ, Brazil; (M.N.); (S.M.)
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Lidia Parra
- Programa de Pós-Graduação Acadêmico em Pesquisa Translacional em Fármacos e Medicamentos–Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil;
| | - Valeska Vasconcellos
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
- Programa de Pós-Graduação Acadêmico em Pesquisa Translacional em Fármacos e Medicamentos–Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil;
| | - Gabriela Costa
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Debora Leite
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Maria Dias
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Tácio Vinício Amorim Fernandes
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Lucas Hoelz
- Laboratório Computacional de Química Medicinal—LCQM, Instituto Federal do Rio de Janeiro—IFRJ, Campus Pinheiral, Pinheiral 27197-000, RJ, Brazil;
| | - Luiz Pimentel
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Monica Bastos
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Nubia Boechat
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| |
Collapse
|
3
|
Kuo YH, Wei SH, Jiang JH, Chang YS, Liu MY, Fu SL, Huang CYF, Lin WJ. Perturbation of p38α MAPK as a Novel Strategy to Effectively Sensitize Chronic Myeloid Leukemia Cells to Therapeutic BCR-ABL Inhibitors. Int J Mol Sci 2021; 22:ijms222212573. [PMID: 34830455 PMCID: PMC8623086 DOI: 10.3390/ijms222212573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a hematopoietic malignancy characterized by the presence of the BCR-ABL oncogene. Therapeutic regimens with tyrosine kinase inhibitors (TKIs) specifically targeting BCR-ABL have greatly improved overall survival of CML. However, drug intolerance and related toxicity remain. Combined therapy is effective in reducing drug magnitude while increasing therapeutic efficacy and, thus, lowers undesired adverse side effects. The p38 MAPK activity is critically linked to the pathogenesis of a number of diseases including hematopoietic diseases; however, the role of each isozyme in CML and TKI-mediated effects is still elusive. In this study, we used specific gene knockdown to clearly demonstrate that the deficiency of p38α greatly enhanced the therapeutic efficacy in growth suppression and cytotoxicity of TKIs, first-generation imatinib, and second generation dasatinib by approximately 2.5–3.0-fold in BCR-ABL-positive CML-derived leukemia K562 and KMB5 cells. Knockdown of p38β, which displays the most sequence similarity to p38α, exerted distinct and opposite effects on the TKI-mediated therapeutic efficacy. These results show the importance of isotype-specific intervention in enhancing the therapeutic efficacy of TKI. A highly specific p38α inhibitor, TAK715, also significantly enhanced the imatinib- and dasatinib-mediated therapeutic efficacy, supporting the feasibility of p38α deficiency in future clinic application. Taken together, our results demonstrated that p38α is a promising target for combined therapy with BCR-ABL-targeting tyrosine kinase inhibitors for future application to increase therapeutic efficacy.
Collapse
MESH Headings
- Cell Proliferation/drug effects
- Combined Modality Therapy
- Dasatinib/pharmacology
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Gene Knockdown Techniques
- Genetic Therapy
- Humans
- Imatinib Mesylate/pharmacology
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mitogen-Activated Protein Kinase 14/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 14/deficiency
- Mitogen-Activated Protein Kinase 14/genetics
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Yi-Hue Kuo
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.K.); (S.-H.W.); (J.-H.J.); (M.-Y.L.); (C.-Y.F.H.)
| | - Shih-Hsiang Wei
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.K.); (S.-H.W.); (J.-H.J.); (M.-Y.L.); (C.-Y.F.H.)
| | - Jie-Hau Jiang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.K.); (S.-H.W.); (J.-H.J.); (M.-Y.L.); (C.-Y.F.H.)
| | - Yueh-Shih Chang
- Hemato-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of Medicine, Keelung & Chang Gung University, Taoyuan City 33302, Taiwan;
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Mei-Yin Liu
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.K.); (S.-H.W.); (J.-H.J.); (M.-Y.L.); (C.-Y.F.H.)
| | - Shu-Ling Fu
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.K.); (S.-H.W.); (J.-H.J.); (M.-Y.L.); (C.-Y.F.H.)
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wey-Jinq Lin
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.K.); (S.-H.W.); (J.-H.J.); (M.-Y.L.); (C.-Y.F.H.)
- Correspondence: ; Tel.: +886-2-2826-7257
| |
Collapse
|
4
|
Shinde A, Panchal K, Katke S, Paliwal R, Chaurasiya A. Tyrosine kinase inhibitors as next generation oncological therapeutics: Current strategies, limitations and future perspectives. Therapie 2021; 77:425-443. [PMID: 34823895 DOI: 10.1016/j.therap.2021.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/04/2021] [Accepted: 10/19/2021] [Indexed: 02/09/2023]
Abstract
Protein kinases, a class of enzymes that govern various biological phenomena at a cellular level, are responsible for signal transduction in cells that regulate cellular proliferation, differentiation, and growth. Protein kinase enzyme mutation results in abnormal cell division leading to a pathological condition like cancer. Tyrosine kinase (TK) inhibitors, which helps as a potential drug candidate for the treatment of cancer, are continuously being developed. Majority of these drug candidates are being administered as conventional oral dosage form, which provides limited safety and efficacy due to non-specific delivery and uncontrolled biodistribution resulting into the adverse effects. A controlled drug delivery approach for the delivery of TK inhibitors may be a potential strategy with significant safety and efficacy profile. Novel drug delivery strategies provide target-specific drug delivery, improved pharmacokinetic behaviour, and sustained release leading to lower doses and dosing frequency with significantly reduced side effects. Along with basic aspects of tyrosine kinase, this review discusses various aspects related to the application of tyrosine kinase inhibitors in clinical oncological setting. Furthermore, the limitations/challenges and formulation advancements related to this class of candidates particularly for cancer management have been reviewed. It is expected that innovations in drug delivery approaches for TK inhibitors using novel techniques will surely provide a new insights for improved cancer treatment and patients' life quality.
Collapse
Affiliation(s)
- Aishwarya Shinde
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana 500078, India
| | - Kanan Panchal
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana 500078, India
| | - Sumeet Katke
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana 500078, India
| | - Rishi Paliwal
- Nanomedicine and Bioengineering Research Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak 484886, India
| | - Akash Chaurasiya
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana 500078, India.
| |
Collapse
|
5
|
Gupta SK, Singh P, Chhabra R, Verma M. Novel pharmacological approach for the prevention of multidrug resistance (MDR) in a human leukemia cell line. Leuk Res 2021; 109:106641. [PMID: 34144313 DOI: 10.1016/j.leukres.2021.106641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Drug resistance mechanisms are the regulatory factors associated with drug metabolism and drug transport to inward and outward of the target cells. Maybridge fragment (MBF) library is a collection of pharmacophore rich compounds having affinity with membrane transporters. This study has been designed to evaluate the efficacy of MBFs in overcoming the leukemic cells' resistance to imatinib. METHODS Imatinib resistant cells (K562-R) were prepared using myelogenous leukemia cell line (K562) by titration method. The four MBFs were prioritized for determining their effect on imatinib resistance. The cells were treated with imatinib and MBFs and the MTT assay was performed to evaluate the efficacy of MBFs in enhancing the imatinib mediated cell death. The transcript levels of Bcr-Abl1 gene and efflux transporter genes were determined by RT-qPCR analysis. RESULTS The MBFs enhanced the imatinib mediated cell death of K562-R cells. There was also a significant decrease in the mRNA levels of the major drug efflux genes (ABCB1, ABCB10, ABCC1 and ABCG2) when treated with a combination of imatinib and MBF in comparison to imatinib treatment alone. CONCLUSION The drug efflux is one of the mechanisms of multidrug resistance in cancer cells and the MBFs used in this study were all found to significantly overcome the imatinib resistance by limiting the expression of efflux genes. This study, therefore, highlights the potential of Maybridge compounds in treating the drug resistant leukemia.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis
- Biomarkers, Tumor/genetics
- Cell Proliferation
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Sonu Kumar Gupta
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Priyanka Singh
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Ravindresh Chhabra
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Malkhey Verma
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
6
|
|
7
|
Walker VE, Degner A, Carter EW, Nicklas JA, Walker DM, Tretyakova N, Albertini RJ. 1,3-Butadiene metabolite 1,2,3,4 diepoxybutane induces DNA adducts and micronuclei but not t(9;22) translocations in human cells. Chem Biol Interact 2019; 312:108797. [PMID: 31422076 DOI: 10.1016/j.cbi.2019.108797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/26/2019] [Accepted: 08/14/2019] [Indexed: 11/26/2022]
Abstract
Epidemiological studies of 1,3-butadiene (BD) exposures have reported a possible association with chronic myelogenous leukemia (CML), which is defined by the presence of the t(9;22) translocation (Philadelphia chromosome) creating an oncogenic BCR-ABL fusion gene. Butadiene diepoxide (DEB), the most mutagenic of three epoxides resulting from BD, forms DNA-DNA crosslink adducts that can lead to DNA double-strand breaks (DSBs). Thus, a study was designed to determine if (±)-DEB exposure of HL60 cells, a promyelocytic leukemia cell line lacking the Philadelphia chromosome, can produce t(9;22) translocations. In HL60 cells exposed for 3 h to 0-10 μM DEB, overlapping dose-response curves suggested a direct relationship between 1,4-bis-(guan-7-yl)-2,3-butanediol crosslink adduct formation (R = 0.977, P = 0.03) and cytotoxicity (R = 0.961, P = 0.002). Experiments to define the relationships between cytotoxicity and the induction of micronuclei (MN), a dosimeter of DNA DSBs, showed that 24 h exposures of HL60 cells to 0-5.0 μM DEB caused significant positive correlations between the concentration and (i) the degree of cytotoxicity (R = 0.998, p = 0.002) and (ii) the frequency of MN (R = 0.984, p = 0.016) at 48 h post exposure. To determine the relative induction of MN and t(9;22) translocations following exposures to DEB, or x-rays as a positive control for formation of t(9;22) translocations, HL60 cells were exposed for 24 h to 0, 1, 2.5, or 5 μM DEB or to 0, 2.0, 3.5, or 5.0 Gy x-rays, or treatments demonstrated to yield 0, 20%, 50%, or 80% cytotoxicity. Treatments between 0 and 3.5 Gy x-rays caused significant dose-related increases in both MN (p < 0.001) and t(9;22) translocations (p = 0.01), whereas DEB exposures causing similar cytotoxicity levels did not increase translocations over background. These data indicate that, while DEB induces DNA DSBs required for formation of MN and translocations, acute DEB exposures of HL60 cells did not produce the Philadelphia chromosome obligatory for CML.
Collapse
Affiliation(s)
- Vernon E Walker
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States.
| | - Amanda Degner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, United States.
| | - Elizabeth W Carter
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States.
| | - Janice A Nicklas
- Department of Pediatrics, University of Vermont, Burlington, VT, United States.
| | - Dale M Walker
- The Burlington HC Research Group, Inc., Jericho, VT, United States.
| | - Natalia Tretyakova
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, United States.
| | - Richard J Albertini
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States.
| |
Collapse
|
8
|
Schoene J, Gazzi T, Lindemann P, Christmann M, Volkamer A, Nazaré M. Probing 2
H
‐Indazoles as Templates for SGK1, Tie2, and SRC Kinase Inhibitors. ChemMedChem 2019; 14:1514-1527. [DOI: 10.1002/cmdc.201900328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/26/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Jens Schoene
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Thais Gazzi
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Peter Lindemann
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Mathias Christmann
- Organische ChemieInstitut für Chemie und BiochemieFreie Universität Berlin Takustrasse. 3 14195 Berlin Germany
| | - Andrea Volkamer
- In silico Toxicology and Structural Bioinformatics Group, Institute of PhysiologyCharité—Universitätsmedizin Berlin Charitéplatz 1 10117 Berlin Germany
| | - Marc Nazaré
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
- Anna-Louisa-Karsch-Str. 2 10178 Berlin Germany
| |
Collapse
|
9
|
Jia X, Zheng Y, Guo Y, Chen K. Sodium butyrate and panobinostat induce apoptosis of chronic myeloid leukemia cells via multiple pathways. Mol Genet Genomic Med 2019; 7:e613. [PMID: 30891950 PMCID: PMC6503025 DOI: 10.1002/mgg3.613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 01/05/2019] [Accepted: 02/10/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose Histone deacetylase inhibitor (HDACI) is a novel therapeutic option for cancer. However, the effects of HDACIs on chronic myeloid leukemia (CML) and the underlying mechanisms are still unknown. The aim of this study was to investigate the effect and the mechanism‐of‐action of two HDACI members, sodium butyrate (NaBu) and panobinostat (LBH589) in K562 and the adriamycin–resistant cell line K562/ADR. Methods Cell viability was assessed using MTT assay. Cell apoptosis was detected with flow cytometry. Cell cycle analysis and western blot were performed to explore the possible molecules related to HDACIs effects. Results The effect of NaBu was more powerful on K562/ADR than on K562 cells. LBH589 triggered apoptosis and inhibited the growth of K562 cells. Both HDACIs inhibited K562 and K562/ADR cells via activation of intrinsic/extrinsic apoptotic pathways and inhibition of AKT‐mTOR pathway while NaBu also activated endoplasmic reticulum stress (ERS) mediated apoptotic pathway in K562/ADR cells. LBH589 reduced the expression of drug–resistant related proteins in K562 cells. However, neither NaBu nor LBH589 could significantly influence the expression of the drug–resistant related proteins in K562/ADR cells. Conclusion The combination of HDACI and other therapeutic strategies are likely required to overcome drug resistance in CML therapy.
Collapse
Affiliation(s)
- Xiaoyuan Jia
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yinsuo Zheng
- Department of Hematology, Baoji Central Hospital, Baoji, China
| | - Yanzi Guo
- The Second Affiliated Hospital of Shaanxi Traditional University, Xianyang, China
| | - Kan Chen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
10
|
Costa FB, Cortez AP, de Ávila RI, de Carvalho FS, Andrade WM, da Cruz AF, Reis KB, Menegatti R, Lião LM, Romeiro LAS, Noël F, Fraga CAM, Barreiro EJ, Sanz G, Rodrigues MF, Vaz BG, Valadares MC. The novel piperazine-containing compound LQFM018: Necroptosis cell death mechanisms, dopamine D 4 receptor binding and toxicological assessment. Biomed Pharmacother 2018; 102:481-493. [PMID: 29579709 DOI: 10.1016/j.biopha.2018.02.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/21/2018] [Accepted: 02/23/2018] [Indexed: 01/12/2023] Open
Abstract
Piperazine is a promising scaffold for drug development due to its broad spectrum of biological activities. Based on this, the new piperazine-containing compound LQFM018 (2) [ethyl 4-((1-(4-chlorophenyl)-1H-pyrazol-4-yl)methyl)piperazine-1-carboxylate] was synthetized and some biological activities investigated. In this work, we described its ability to bind aminergic receptors, antiproliferative effects as well as the LQFM018 (2)-triggered cell death mechanisms, in K562 leukemic cells, by flow cytometric analyses. Furthermore, acute oral systemic toxicity and potential myelotoxicity assessments of LQFM018 (2) were carried out. LQFM018 (2) was originally obtained by molecular simplification from LASSBio579 (1), an analogue compound of clozapine, with 33% of global yield. Binding profile assay to aminergic receptors showed that LQFM018 (2) has affinity for the dopamine D4 receptor (Ki = 0.26 μM). Moreover, it showed cytotoxicity in K562 cells, in a concentration and time-dependent manner; IC50 values obtained were 399, 242 and 119 μM for trypan blue assay and 427, 259 and 50 μM for MTT method at 24, 48 or 72 h, respectively. This compound (427 μM) also promoted increase in LDH release and cell cycle arrest in G2/M phase. Furthermore, it triggered necrotic morphologies in K562 cells associated with intense cell membrane rupture as confirmed by Annexin V/propidium iodide double-staining. LQFM018 (2) also triggered mitochondrial disturb through loss of ΔΨm associated with increase of ROS production. No significant accumulation of cytosolic cytochrome c was verified in treated cells. Furthermore, it was verified an increase of expression of TNF-R1 and mRNA levels of CYLD with no involviment in caspase-3 and -8 activation and NF-κB in K562 cells. LQFM018 (2) showed in vitro myelotoxicity potential, but it was orally well tolerated and classified as UN GHS category 5 (LD50 > 2000-5000 mg/Kg). Thus, LQFM018 (2) seems to have a non-selective action considering hematopoietic cells. In conclusion, it is suggested LQFM018 (2) promotes cell death in K562 cells via necroptotic signaling, probably with involvement of dopamine D4 receptor. These findings open new perspectives in cancer therapy by use of necroptosis inducing agents as a strategy of reverse cancer cell chemoresistance.
Collapse
Affiliation(s)
- Fabiana Bettanin Costa
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Alane P Cortez
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Renato Ivan de Ávila
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Flávio S de Carvalho
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Wanessa M Andrade
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Andrezza F da Cruz
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Karinna B Reis
- Laboratório de Química Farmacêutica Medicinal (LQFM), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Ricardo Menegatti
- Laboratório de Química Farmacêutica Medicinal (LQFM), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Luciano M Lião
- Laboratório de Ressonância Magnética Nuclear, Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Luiz Antônio S Romeiro
- Universidade Católica de Brasília, Brasília, DF, Brazil; Universidade de Brasília, Brasília, DF, Brazil
| | - François Noël
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biológicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carlos Alberto M Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Eliezer J Barreiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Germán Sanz
- Laboratório de Cromatografia e Espectrometria de Massas (LaCEM), Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Marcella F Rodrigues
- Laboratório de Cromatografia e Espectrometria de Massas (LaCEM), Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Boniek G Vaz
- Laboratório de Cromatografia e Espectrometria de Massas (LaCEM), Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Marize Campos Valadares
- Laboratório de Farmacologia e Toxicologia Celular - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
11
|
Lin X, Lu T, Yan F, Li R, Huang X. Mean residual life regression with functional principal component analysis on longitudinal data for dynamic prediction. Biometrics 2018; 74:1482-1491. [DOI: 10.1111/biom.12876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 01/01/2018] [Accepted: 02/01/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Xiao Lin
- Research Center of Biostatistics and Computational PharmacyChina Pharmaceutical University Nanjing 210009 P.R. China
- Department of BiostatisticsThe University of Texas MD Anderson Cancer Center Houston, Texas 77030 U.S.A
| | - Tao Lu
- Research Center of Biostatistics and Computational PharmacyChina Pharmaceutical University Nanjing 210009 P.R. China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational PharmacyChina Pharmaceutical University Nanjing 210009 P.R. China
| | - Ruosha Li
- Department of BiostatisticsThe University of Texas Health Science Center at Houston Houston, Texas 77030 U.S.A
| | - Xuelin Huang
- Department of BiostatisticsThe University of Texas MD Anderson Cancer Center Houston, Texas 77030 U.S.A
| |
Collapse
|
12
|
Peixoto-da-Silva J, Calgarotto AK, Rocha KR, Palmeira-dos-Santos C, Smaili SS, Pereira GJ, Pericole FV, da Silva S. Duarte A, Saad ST, Bincoletto C. Lithium, a classic drug in psychiatry, improves nilotinib-mediated antileukemic effects. Biomed Pharmacother 2018; 99:237-244. [DOI: 10.1016/j.biopha.2018.01.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/21/2017] [Accepted: 01/03/2018] [Indexed: 12/11/2022] Open
|
13
|
Rajendran V, Gopalakrishnan C, Sethumadhavan R. Pathological role of a point mutation (T315I) in BCR‐ABL1 protein—A computational insight. J Cell Biochem 2017; 119:918-925. [DOI: 10.1002/jcb.26257] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/05/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Vidya Rajendran
- Computational Biology LabDepartment of BiotechnologyVellore Institute of Technology UniversityVelloreTamil NaduIndia
| | | | - Rao Sethumadhavan
- Computational Biology LabDepartment of BiotechnologyVellore Institute of Technology UniversityVelloreTamil NaduIndia
| |
Collapse
|
14
|
Mlejnek P, Dolezel P, Ruzickova E. Drug resistance of cancer cells is crucially affected by expression levels of ABC-transporters. BIODISCOVERY 2017. [DOI: 10.3897/biodiscovery.20.e11211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
15
|
Matsuda Y, Yamauchi T, Hosono N, Uzui K, Negoro E, Morinaga K, Nishi R, Yoshida A, Kimura S, Maekawa T, Ueda T. Combination of panobinostat with ponatinib synergistically overcomes imatinib-resistant CML cells. Cancer Sci 2016; 107:1029-38. [PMID: 27166836 PMCID: PMC4946706 DOI: 10.1111/cas.12965] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/28/2016] [Accepted: 05/02/2016] [Indexed: 12/21/2022] Open
Abstract
The major mechanism of imatinib (IM) resistance of CML is the reactivation of ABL kinase either through BCR-ABL gene amplification or mutation. We investigated the cytotoxicity of a pan-ABL tyrosine kinase inhibitor, ponatinib, and a pan-histone deacetylase inhibitor, panobinostat, against IM-resistant CML cells in vitro. Two different IM-resistant cell lines, K562/IM-R1 and Ba/F3/T315I were evaluated in comparison with their respective, parental cell lines, K562 and Ba/F3. K562/IM-R1 overexpressed BCR-ABL due to gene amplification. Ba/F3/T315I was transfected with a BCR-ABL gene encoding T315I-mutated BCR-ABL. Ponatinib inhibited the growth of both K562/IM-R1 and Ba/F3/T315I as potently as it inhibited their parental cells with an IC50 of 2-30 nM. Panobinostat also similarly inhibited the growth of all of the cell lines with an IC50 of 40-51 nM. This was accompanied by reduced histone deacetylase activity, induced histone H3 acetylation, and an increased protein level of heat shock protein 70, which suggested disruption of heat shock protein 90 chaperone function for BCR-ABL and its degradation. Importantly, the combination of ponatinib with panobinostat showed synergistic growth inhibition and induced a higher level of apoptosis than the sum of the apoptosis induced by each agent alone in all of the cell lines. Ponatinib inhibited phosphorylation not only of BCR-ABL but also of downstream signal transducer and activator of transcription 5, protein kinase B, and ERK1/2 in both K562/IM-R1 and Ba/F3/T315I, and the addition of panobinostat to ponatinib further inhibited these phosphorylations. In conclusion, panobinostat enhanced the cytotoxicity of ponatinib towards IM-resistant CML cells including those with T315I-mutated BCR-ABL.
Collapse
Affiliation(s)
- Yasufumi Matsuda
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Takahiro Yamauchi
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Naoko Hosono
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kanako Uzui
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Eiju Negoro
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Koji Morinaga
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Rie Nishi
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Akira Yoshida
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University, Saga, Japan
| | - Taira Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Takanori Ueda
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
16
|
|
17
|
1,3-Butadiene, CML and the t(9:22) translocation: A reality check. Chem Biol Interact 2015; 241:32-9. [DOI: 10.1016/j.cbi.2015.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/28/2015] [Accepted: 05/12/2015] [Indexed: 11/23/2022]
|
18
|
Clapp GD, Lepoutre T, El Cheikh R, Bernard S, Ruby J, Labussière-Wallet H, Nicolini FE, Levy D. Implication of the Autologous Immune System in BCR–ABL Transcript Variations in Chronic Myelogenous Leukemia Patients Treated with Imatinib. Cancer Res 2015; 75:4053-62. [DOI: 10.1158/0008-5472.can-15-0611] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/29/2015] [Indexed: 11/16/2022]
|
19
|
Barr LA, Makarewich CA, Berretta RM, Gao H, Troupes CD, Woitek F, Recchia F, Kubo H, Force T, Houser SR. Imatinib activates pathological hypertrophy by altering myocyte calcium regulation. Clin Transl Sci 2014; 7:360-7. [PMID: 24931551 DOI: 10.1111/cts.12173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Imatinib mesylate is a selective tyrosine-kinase inhibitor used in the treatment of multiple cancers, most notably chronic myelogenous leukemia. There is evidence that imatinib can induce cardiotoxicity in cancer patients. Our hypothesis is that imatinib alters calcium regulatory mechanisms and can contribute to development of pathological cardiac hypertrophy. METHODS AND RESULTS Neonatal rat ventricular myocytes (NRVMs) were treated with clinical doses (low: 2 μM; high: 5 μM) of imatinib and assessed for molecular changes. Imatinib increased peak systolic Ca(2+) and Ca(2+) transient decay rates and Western analysis revealed significant increases in phosphorylation of phospholamban (Thr-17) and the ryanodine receptor (Ser-2814), signifying activation of calcium/calmodulin-dependent kinase II (CaMKII). Imatinib significantly increased NRVM volume as assessed by Coulter counter, myocyte surface area, and atrial natriuretic peptide abundance seen by Western. Imatinib induced cell death, but did not activate the classical apoptotic program as assessed by caspase-3 cleavage, indicating a necrotic mechanism of death in myocytes. We expressed AdNFATc3-green fluorescent protein in NRVMs and showed imatinib treatment significantly increased nuclear factor of activated T cells translocation that was inhibited by the calcineurin inhibitor FK506 or CaMKII inhibitors. CONCLUSION These data show that imatinib can activate pathological hypertrophic signaling pathways by altering intracellular Ca(2+) dynamics. This is likely a contributing mechanism for the adverse cardiac effects of imatinib.
Collapse
Affiliation(s)
- Larry A Barr
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Xu HL, Wang ZJ, Liang XM, Li X, Shi Z, Zhou N, Bao JK. In silico identification of novel kinase inhibitors targeting wild-type and T315I mutant ABL1 from FDA-approved drugs. MOLECULAR BIOSYSTEMS 2014; 10:1524-37. [PMID: 24691568 DOI: 10.1039/c3mb70577c] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The constitutively active fusion protein BCR-ABL1 is the major cause of chronic myeloid leukemia (CML), and selective inhibition of ABL1 is a promising approach for the treatment of CML. Reported drugs worked well in clinical practice, such as imatinib, dasatinib, nilotinib and bosutinib. However, resistance arises due to ABL1 mutation in patients, especially the T315I gate-keeper mutation. Thus, wide spectrum drugs targeting ABL1 are urgently needed. In order to screen potential drugs targeting wild-type ABL1 and T315I mutant ABL1, 1408 FDA approved small molecule drugs were subjected to molecular docking. With subsequent molecular dynamic (MD) simulation and MM/GBSA binding free energy calculation and energy decomposition, we identified chlorhexidine and sorafenib as potential "new use" drugs targeting wild-type ABL1, while nicergoline and plerixafor targeted T315I ABL1. Meanwhile, we also found that residues located in the ATP-binding site and A-loop motif played key roles in drug discovery towards ABL1. These findings may not only serve as a paradigm for the repositioning of existing approved drugs, but also instill new vitality to ABL1-targeted anti-CML therapeutics.
Collapse
Affiliation(s)
- Huai-long Xu
- School of Life Sciences and Key laboratory of Bio-resources, Ministry of Education, Sichuan University, Chengdu 610064, China.
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The chronic leukemias include chronic myeloid leukemia (CML) and chronic lymphocytic leukemia (CLL). CML is a clonal myeloproliferative hematopoietic stem-cell disorder, and CLL is a monoclonal B-cell disorder. CML is Philadelphia chromosome positive. There are 3 phases of CML: the chronic phase, the accelerated phase, and the blast phase. The primary treatment of CML consists of tyrosine kinase inhibitors. CLL can present as indolent or fulminant disease. Early disease is managed with observation. Fulminant disease is currently treated with alkylating agents, purine analogues, and monoclonal antibodies, but new biotarged therapies are being developed.
Collapse
Affiliation(s)
- Edythe M Lyn Greenberg
- Department of Leukemia, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | | |
Collapse
|
22
|
Bitencourt R, Zalcberg I, Louro ID. Imatinib resistance: a review of alternative inhibitors in chronic myeloid leukemia. Rev Bras Hematol Hemoter 2013; 33:470-5. [PMID: 23049365 PMCID: PMC3459369 DOI: 10.5581/1516-8484.20110124] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 10/21/2011] [Indexed: 12/15/2022] Open
Abstract
The development of point mutations in the BCR-ABL kinase domain is the main reason for imatinib resistance in chronic myeloid leukemia. Different detection methods are used in chronic myeloid leukemia monitoring, such as direct sequencing, denaturing high performance liquid chromatography and allele specific polymerase chain reaction. Mutation analysis has become mandatory during patient workup of chronic myeloid leukemia in order for the physician to choose the most suitable tyrosine kinase inhibitor. This article, a review of possible therapies used to overcome imatinib resistance, investigates the current position by searching the PubMed electronic database using the following keywords: imatinib, dasatinib, nilotinib, aurora kinase, SRC kinase, mutation, treatment, drugs and resistance. New tyrosine kinase inhibitors include BCR-ABL kinase selective inhibitors, dual ABL/SRC kinase inhibitors and aurora kinase inhibitors. Awareness of the spectrum of new drugs against mutations, in particular the T315I mutation, makes it possible to properly select the best therapy for each patient.
Collapse
Affiliation(s)
- Roberta Bitencourt
- Master Program in Biotechnology, Universidade Federal do Espírito Santo - UFES, Vitória, ES, Brazil
| | | | | |
Collapse
|
23
|
Rezende VM, Rivellis AJ, Gomes MM, Dörr FA, Novaes MMY, Nardinelli L, Costa ALDL, Chamone DDAF, Bendit I. Determination of serum levels of imatinib mesylate in patients with chronic myeloid leukemia: validation and application of a new analytical method to monitor treatment compliance. Rev Bras Hematol Hemoter 2013; 35:103-8. [PMID: 23741187 PMCID: PMC3672119 DOI: 10.5581/1516-8484.20130030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/30/2012] [Indexed: 11/27/2022] Open
Abstract
Objective The goal of this study was to monitor imatinib mesylate therapeutically in the
Tumor Biology Laboratory, Department of Hematology and Hemotherapy, Hospital das
Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP). A
simple and sensitive method to quantify imatinib and its metabolite (CGP74588) in
human serum was developed and fully validated in order to monitor treatment
compliance. Methods The method used to quantify these compounds in serum included protein
precipitation extraction followed by instrumental analysis using high performance
liquid chromatography coupled with mass spectrometry. The method was validated for
several parameters, including selectivity, precision, accuracy, recovery and
linearity. Results The parameters evaluated during the validation stage exhibited satisfactory
results based on the Food and Drug Administration and the Brazilian Health
Surveillance Agency (ANVISA) guidelines for validating bioanalytical methods.
These parameters also showed a linear correlation greater than 0.99 for the
concentration range between 0.500 µg/mL and 10.0 µg/mL and a total
analysis time of 13 minutes per sample. This study includes results (imatinib
serum concentrations) for 308 samples from patients being treated with imatinib
mesylate. Conclusion The method developed in this study was successfully validated and is being
efficiently used to measure imatinib concentrations in samples from chronic
myeloid leukemia patients to check treatment compliance. The imatinib serum levels
of patients achieving a major molecular response were significantly higher than
those of patients who did not achieve this result. These results are thus
consistent with published reports concerning other populations.
Collapse
|
24
|
Design and synthesis of novel 4-benzothiazole amino quinazolines Dasatinib derivatives as potential anti-tumor agents. Eur J Med Chem 2013; 63:702-12. [PMID: 23567960 DOI: 10.1016/j.ejmech.2013.03.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 03/04/2013] [Accepted: 03/06/2013] [Indexed: 02/04/2023]
Abstract
Three series of novel 4-benzothiazole amino quinazolines Dasatinib derivatives have been designed and synthesized. The entire target compounds were investigated for their in vitro cytotoxic activity by the MTT-based assay against 6 human cancer cell lines. Compared with the parental Dasatinib, most of the new compounds, especially 2, 4, 6-trimethylaniline series (3), demonstrated significant inhibitory activities against six cell lines. Furthermore, the target compounds were screened for Src and Abl kinase inhibitory activity. Among them, 1a, 1f and 3a-3f are more potential dual Src/Abl kinase inhibitors. Thus they may be promising lead compounds to be developed as an alternative for current Dasatinib therapy or for Imatinib-resistant patients, potentially via simultaneously blocking multiple RTK signaling pathways.
Collapse
|
25
|
Pan X, Wang F, Zhang Y, Gao H, Hu Z, Wang S, Zhang J. Design, synthesis and biological activities of Nilotinib derivates as antitumor agents. Bioorg Med Chem 2013; 21:2527-34. [PMID: 23538233 DOI: 10.1016/j.bmc.2013.02.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 02/23/2013] [Accepted: 02/25/2013] [Indexed: 01/03/2023]
Abstract
A novel class of Nilotinib derivatives, B1-B20, were synthesized in high yields using various substituted anilines. All the title compounds were evaluated for their inhibitory activities against Bcr-Abl and antiproliferative effects on human leukemia cell (K562). The pharmacological results indicated that some compounds exhibited promising anticancer activity. In particular, compound B14 containing tertiary amine side chain exhibited Bcr-Abl inhibitory activity similar to that of Nilotinib. It was suggested that the introduction of the tertiary amine moiety could improve Bcr-Abl inhibitory activity and antitumor effects.
Collapse
Affiliation(s)
- Xiaoyan Pan
- School of Medicine, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, Shaanxi Province 710061, PR China
| | | | | | | | | | | | | |
Collapse
|
26
|
Baran Y, Saydam G. Cumulative clinical experience from a decade of use: imatinib as first-line treatment of chronic myeloid leukemia. J Blood Med 2012. [PMID: 23180974 PMCID: PMC3503471 DOI: 10.2147/jbm.s29132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a malignant disease that originates in the bone marrow and is designated by the presence of the Philadelphia (Ph+) chromosome, a translocation between chromosomes 9 and 22. Targeted therapy against CML commenced with the development of small-molecule tyrosine kinase inhibitors (TKIs) exerting their effect against the oncogenic breakpoint cluster region (BCR)-ABL fusion protein. Imatinib emerged as the first successful example of a TKI used for the treatment of chronic-phase CML patients and resulted in significant improvements in response rate and overall survival compared with previous treatments. However, a significant portion of patients failed to respond to the therapy and developed resistance against imatinib. Second-generation TKIs nilotinib and dasatinib were to have higher efficiency in clinical trials in imatinib- resistant or intolerant CML patients compared with imatinib. Identification of novel strategies such as dose escalation, drug combination therapy, and use of novel BCR-ABL inhibitors may eventually overcome resistance against BCR-ABL TKIs. This article reviews the history of CML, including the treatment strategies used prediscovery of TKIs and the preclinical and clinical data obtained after the use of imatinib, and the second-generation TKIs developed for the treatment of CML.
Collapse
Affiliation(s)
- Yusuf Baran
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | | |
Collapse
|
27
|
Retnakumari AP, Hanumanthu PL, Malarvizhi GL, Prabhu R, Sidharthan N, Thampi MV, Menon D, Mony U, Menon K, Keechilat P, Nair S, Koyakutty M. Rationally designed aberrant kinase-targeted endogenous protein nanomedicine against oncogene mutated/amplified refractory chronic myeloid leukemia. Mol Pharm 2012; 9:3062-78. [PMID: 22971013 DOI: 10.1021/mp300172e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Deregulated protein kinases play a very critical role in tumorigenesis, metastasis, and drug resistance of cancer. Although molecularly targeted small molecule kinase inhibitors (SMI) are effective against many types of cancer, point mutations in the kinase domain impart drug resistance, a major challenge in the clinic. A classic example is chronic myeloid leukemia (CML) caused by BCR-ABL fusion protein, wherein a BCR-ABL kinase inhibitor, imatinib (IM), was highly successful in the early chronic phase of the disease, but failed in the advanced stages due to amplification of oncogene or point mutations in the drug-binding site of kinase domain. Here, by identifying critical molecular pathways responsible for the drug-resistance in refractory CML patient samples and a model cell line, we have rationally designed an endogenous protein nanomedicine targeted to both cell surface receptors and aberrantly activated secondary kinase in the oncogenic network. Molecular diagnosis revealed that, in addition to point mutations and amplification of oncogenic BCR-ABL kinase, relapsed/refractory patients exhibited significant activation of STAT5 signaling with correlative overexpression of transferrin receptors (TfR) on the cell membrane. Accordingly, we have developed a human serum albumin (HSA) based nanomedicine, loaded with STAT5 inhibitor (sorafenib), and surface conjugated the same with holo-transferrin (Tf) ligands for TfR specific delivery. This dual-targeted "transferrin conjugated albumin bound sorafenib" nanomedicine (Tf-nAlb-Soraf), prepared using aqueous nanoprecipitation method, displayed uniform spherical morphology with average size of ∼150 nm and drug encapsulation efficiency of ∼74%. TfR specific uptake and enhanced antileukemic activity of the nanomedicine was found maximum in the most drug resistant patient sample having the highest level of STAT5 and TfR expression, thereby confirming the accuracy of our rational design and potential of dual-targeting approach. The nanomedicine induced downregulation of key survival pathways such as pSTAT5 and antiapoptotic protein MCL-1 was demonstrated using immunoblotting. This study reveals that, by implementing molecular diagnosis, personalized nanomedicines can be rationally designed and nanoengineered by imparting therapeutic functionality to endogenous proteins to overcome clinically important challenges like molecular drug resistance.
Collapse
Affiliation(s)
- Archana P Retnakumari
- Amrita Center for Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham University, Cochin, India
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
A Phase I clinical trial of the combination of imatinib and paclitaxel in patients with advanced or metastatic solid tumors refractory to standard therapy. Cancer Chemother Pharmacol 2012; 70:843-53. [PMID: 23014737 DOI: 10.1007/s00280-012-1969-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 09/02/2012] [Indexed: 12/12/2022]
Abstract
PURPOSE Pre-clinical data suggest that combining imatinib with traditional cytotoxic chemotherapy may improve imatinib efficacy. We conducted a Phase I study of imatinib in combination with paclitaxel in patients with advanced or metastatic solid tumors. METHODS Patients were accrued to the study in a standard 3 + 3 design. Patients were restaged every two cycles, and those with stable disease (SD), or better, continued study treatment without interruption. Maximally tolerated doses (MTDs) and pharmacokinetic profiles of combination imatinib and paclitaxel were assessed. RESULTS Fifty-eight patients were enrolled, including 40 in the Phase I dose escalation portion. Alternating dose escalation of imatinib and paclitaxel on a 28-day cycle resulted in MTDs of 800 mg imatinib daily, on days 1-4, 8-11, 15-18, and 22-25, and 100 mg/m(2) paclitaxel weekly, on days 3, 10, and 17. Two expansion cohorts, comprising 10 breast cancer patients and 8 patients with soft-tissue sarcomas, were enrolled at the MTDs. The most common adverse events were flu-like symptoms (64 %) and nausea/vomiting (71 %). The most common Grade 3/4 toxicities were neutropenia (26 %), flu-like symptoms (12 %), and pain (12 %). There were no relevant differences in the pharmacokinetic profiles of either drug when given in combination compared with alone. Thirty-eight subjects were evaluable for response, 18 (47.4 %) of whom experienced clinical benefit. Five patients (13.2 %) had a partial response (PR) and 13 patients (34.2 %) had SD; the average time to progression in those with clinical benefit was 17 weeks (range: 7-28 weeks). CONCLUSIONS This combination of imatinib and paclitaxel was reasonably safe and tolerable, and demonstrated evidence of anti-tumor activity. Further exploration in disease-specific Phase II trials is warranted.
Collapse
|
29
|
Chen YJ, Chang LS. Gallic acid downregulates matrix metalloproteinase-2 (MMP-2) and MMP-9 in human leukemia cells with expressed Bcr/Abl. Mol Nutr Food Res 2012; 56:1398-412. [DOI: 10.1002/mnfr.201200167] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Revised: 05/31/2012] [Accepted: 06/21/2012] [Indexed: 02/06/2023]
Affiliation(s)
- Ying-Jung Chen
- Institute of Biomedical Sciences; National Sun Yat-Sen University; Kaohsiung Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences; National Sun Yat-Sen University; Kaohsiung Taiwan
- Department of Biotechnology; Kaohsiung Medical University; Kaohsiung Taiwan
| |
Collapse
|
30
|
Dos Santos SC, Mira NP, Moreira AS, Sá-Correia I. Quantitative- and phospho-proteomic analysis of the yeast response to the tyrosine kinase inhibitor imatinib to pharmacoproteomics-guided drug line extension. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2012; 16:537-51. [PMID: 22775238 DOI: 10.1089/omi.2012.0012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Imatinib mesylate (IM) is a potent tyrosine kinase inhibitor used as front-line therapy in chronic myeloid leukemia, a disease caused by the oncogenic kinase Bcr-Abl. Although the clinical success of IM set a new paradigm in molecular-targeted therapy, the emergence of IM resistance is a clinically significant problem. In an effort to obtain new insights into the mechanisms of adaptation and tolerance to IM, as well as the signaling pathways potentially affected by this drug, we performed a two-dimensional electrophoresis-based quantitative- and phospho-proteomic analysis in the eukaryotic model Saccharomyces cerevisiae. We singled out proteins that were either differentially expressed or differentially phosphorylated in response to IM, using the phosphoselective dye Pro-Q(®) Diamond, and identified 18 proteins in total. Ten were altered only at the content level (mostly decreased), while the remaining 8 possessed IM-repressed phosphorylation. These 18 proteins are mainly involved in cellular carbohydrate processes (glycolysis/gluconeogenesis), translation, protein folding, ion homeostasis, and nucleotide and amino acid metabolism. Remarkably, all 18 proteins have human functional homologs. A role for HSP70 proteins in the response to IM, as well as decreased glycolysis as a metabolic marker of IM action are suggested, consistent with findings from studies in human cell lines. The previously-proposed effect of IM as an inhibitor of vacuolar H(+)-ATPase function was supported by the identification of an underexpressed protein subunit of this complex. Taken together, these findings reinforce the role of yeast as a valuable eukaryotic model for pharmacological studies and identification of new drug targets, with potential clinical implications in drug reassignment or line extension under a personalized medicine perspective.
Collapse
Affiliation(s)
- Sandra C Dos Santos
- Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Technical University of Lisbon, Lisbon, Portugal
| | | | | | | |
Collapse
|
31
|
SKLB1028, a novel oral multikinase inhibitor of EGFR, FLT3 and Abl, displays exceptional activity in models of FLT3-driven AML and considerable potency in models of CML harboring Abl mutants. Leukemia 2012; 26:1892-5. [DOI: 10.1038/leu.2012.67] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Borriello A, Caldarelli I, Basile MA, Bencivenga D, Tramontano A, Perrotta S, Ragione FD, Oliva A. The tyrosine kinase inhibitor dasatinib induces a marked adipogenic differentiation of human multipotent mesenchymal stromal cells. PLoS One 2011; 6:e28555. [PMID: 22164306 PMCID: PMC3229607 DOI: 10.1371/journal.pone.0028555] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/10/2011] [Indexed: 12/22/2022] Open
Abstract
Background The introduction of specific BCR-ABL inhibitors in chronic myelogenous leukemia therapy has entirely mutated the prognosis of this hematologic cancer from being a fatal disorder to becoming a chronic disease. Due to the probable long lasting treatment with tyrosine-kinase inhibitors (TKIs), the knowledge of their effects on normal cells is of pivotal importance. Design and Methods We investigated the effects of dasatinib treatment on human bone marrow-derived mesenchymal stromal cells (MSCs). Results Our findings demonstrate, for the first time, that dasatinib induces MSCs adipocytic differentiation. Particularly, when the TKI is added to the medium inducing osteogenic differentiation, a high MSCs percentage acquires adipocytic morphology and overexpresses adipocytic specific genes, including PPARγ, CEBPα, LPL and SREBP1c. Dasatinib also inhibits the activity of alkaline phosphatase, an osteogenic marker, and remarkably reduces matrix mineralization. The increase of PPARγ is also confirmed at protein level. The component of osteogenic medium required for dasatinib-induced adipogenesis is dexamethasone. Intriguingly, the increase of adipocytic markers is also observed in MSCs treated with dasatinib alone. The TKI effect is phenotype-specific, since fibroblasts do not undergo adipocytic differentiation or PPARγ increase. Conclusions Our data demonstrate that dasatinib treatment affects bone marrow MSCs commitment and suggest that TKIs therapy might modify normal phenotypes with potential significant negative consequences.
Collapse
Affiliation(s)
- Adriana Borriello
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Ilaria Caldarelli
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Maria Assunta Basile
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Debora Bencivenga
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Annunziata Tramontano
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| | - Silverio Perrotta
- Department of Pediatrics, Second University of Naples, Naples, Italy
| | - Fulvio Della Ragione
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
- * E-mail:
| | - Adriana Oliva
- Department of Biochemistry and Biophysics, Second University of Naples, Naples, Italy
| |
Collapse
|
33
|
Adaphostin promotes caffeine-evoked autocrine Fas-mediated death pathway activation in Bcr/Abl-positive leukaemia cells. Biochem J 2011; 439:453-67. [DOI: 10.1042/bj20110725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The present study was conducted to verify whether caffeine is beneficial for improving leukaemia therapy. Co-treatment with adaphostin (a Bcr/Abl inhibitor) was found to potentiate caffeine-induced Fas/FasL up-regulation. Although adaphostin did not elicit ASK1 (apoptosis signal-regulating kinase 1)-mediated phosphorylation of p38 MAPK (mitogen-activated protein kinase) and JNK (c-Jun N-terminal kinase), co-treatment with adaphostin notably increased p38 MAPK/JNK activation in caffeine-treated cells. Suppression of p38 MAPK and JNK abrogated Fas/FasL up-regulation in caffeine- and caffeine/adaphostin-treated cells. Compared with caffeine, adaphostin markedly suppressed Akt/ERK (extracellular-signal-regulated kinase)-mediated MKP-1 (MAPK phosphatase 1) protein expression in K562 cells. MKP-1 down-regulation eventually elucidated the enhanced effect of adaphostin on p38 MAPK/JNK activation and subsequent Fas/FasL up-regulation in caffeine-treated cells. Knockdown of p38α MAPK and JNK1, ATF-2 (activating transcription factor 2) and c-Jun by siRNA (small interfering RNA) proved that p38α MAPK/ATF-2 and JNK1/c-Jun pathways were responsible for caffeine-evoked Fas/FasL up-regulation. Moreover, Ca2+ and ROS (reactive oxygen species) were demonstrated to be responsible for ASK1 activation and Akt/ERK inactivation respectively in caffeine- and caffeine/adaphostin-treated cells. Likewise, adaphostin functionally enhanced caffeine-induced Fas/FasL up-regulation in leukaemia cells that expressed Bcr/Abl. Taken together, the results of the present study suggest a therapeutic strategy in improving the efficacy of adaphostin via Fas-mediated death pathway activation in Bcr/Abl-positive leukaemia.
Collapse
|
34
|
Abu-Ghanem S, Rabinovich A, Delgado B, Benharroch D, Ariad S, Levi I, Geffen DB. Chronic myelogenous leukemia diagnosed in two patients with breast cancer. Leuk Lymphoma 2011; 52:2399-401. [DOI: 10.3109/10428194.2011.600483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Podo F, Canevari S, Canese R, Pisanu ME, Ricci A, Iorio E. MR evaluation of response to targeted treatment in cancer cells. NMR IN BIOMEDICINE 2011; 24:648-672. [PMID: 21387442 DOI: 10.1002/nbm.1658] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 11/22/2010] [Accepted: 11/29/2010] [Indexed: 05/30/2023]
Abstract
The development of molecular technologies, together with progressive sophistication of molecular imaging methods, has allowed the further elucidation of the multiple mutations and dysregulatory effects of pathways leading to oncogenesis. Acting against these pathways by specifically targeted agents represents a major challenge for current research efforts in oncology. As conventional anatomically based pharmacological endpoints may be inadequate to monitor the tumor response to these targeted treatments, the identification and use of more appropriate, noninvasive pharmacodynamic biomarkers appear to be crucial to optimize the design, dosage and schedule of these novel therapeutic approaches. An aberrant choline phospholipid metabolism and enhanced flux of glucose derivatives through glycolysis, which sustain the redirection of mitochondrial ATP to glucose phosphorylation, are two major hallmarks of cancer cells. This review focuses on the changes detected in these pathways by MRS in response to targeted treatments. The progress and limitations of our present understanding of the mechanisms underlying MRS-detected phosphocholine accumulation in cancer cells are discussed in the light of gene and protein expression and the activation of different enzymes involved in phosphatidylcholine biosynthesis and catabolism. Examples of alterations induced in the MRS choline profile of cells exposed to different agents or to tumor environmental factors are presented. Current studies aimed at the identification in cancer cells of MRS-detected pharmacodynamic markers of therapies targeted against specific conditional or constitutive cell receptor stimulation are then reviewed. Finally, the perspectives of present efforts addressed to identify enzymes of the phosphatidylcholine cycle as possible novel targets for anticancer therapy are summarized.
Collapse
Affiliation(s)
- Franca Podo
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
36
|
Cloughesy TF, Mischel PS. New strategies in the molecular targeting of glioblastoma: how do you hit a moving target? Clin Cancer Res 2011; 17:6-11. [PMID: 21208902 PMCID: PMC3075730 DOI: 10.1158/1078-0432.ccr-09-2268] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer is a molecularly complex, genomically unstable disease. Selection for drug-resistant mutations, activation of feedback loops, and upregulation of cross-talk pathways provide escape routes by which cancer cells maintain signal flux through critical downstream effectors to promote therapeutic resistance. Attempts to target signal transduction pathways in cancer may therefore require investigators to aim at a moving target. We need to anticipate the routes of resistance to guide the selection of drugs that will lead to durable therapeutic response. In this New Strategies article, we discuss the challenges imposed by the complexity and adaptive capacity of cancer and suggest potential new diagnostic strategies to more effectively guide targeted cancer therapy. We focus on glioblastoma, the most common malignant primary brain tumor of adults. Glioblastoma is a model for a pathway-driven, molecularly heterogeneous cancer for which new genomic insights obtained through The Cancer Genome Atlas are ripe for integration with functional biology and incorporation into new molecular diagnostic assays.
Collapse
Affiliation(s)
- Timothy F Cloughesy
- Department of Neurology, David Geffen UCLA School of Medicine, Los Angeles, California 90095-1732, USA
| | | |
Collapse
|
37
|
Simões BP, Braga Junior JWR, Rego MADC, de Souza CA. Importance of monitoring and early switch to second generation tyrosine kinase inhibitors for the prognosis of patients with chronic myeloid leukemia with imatinib resistance or intolerance. Rev Bras Hematol Hemoter 2011; 33:65-72. [PMID: 23284246 PMCID: PMC3521438 DOI: 10.5581/1516-8484.20110017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 09/24/2010] [Indexed: 11/27/2022] Open
Abstract
Imatinib mesylate was the first BCR-ABL-target agent approved for the treatment of
chronic myeloid leukemia. Although most patients respond well to imatinib therapy,
the literature shows that one third develops resistance or intolerance. The timing of
second-line treatment after failure of initial treatment may have a significant
impact on long-term outcome. Thus, appropriate monitoring to identify resistance
and/or intolerance is crucial to early intervention with second generation tyrosine
kinase inhibitors and attainment of better results
Collapse
Affiliation(s)
- Belinda Pinto Simões
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo USP, Ribeirão Preto (SP), Brazil
| | | | | | | |
Collapse
|
38
|
|
39
|
Bayraktar S, Morency B, Escalón MP. Successful pregnancy in a patient with chronic myeloid leukaemia exposed to dasatinib during the first trimester. BMJ Case Rep 2010; 2010:2010/oct18_2/bcr0520102975. [PMID: 22791487 DOI: 10.1136/bcr.05.2010.2975] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
We report a case of a 25-year-old woman with chronic myeloid leukaemia who was randomised to dasatinib 100 mg orally daily in a clinical trial. The patient was advised to avoid pregnancy while on treatment due to its teratogenic potential. Five months later, the patient was found to be 6 weeks pregnant. Once pregnancy was confirmed, dasatinib was immediately stopped and the patient was taken off the trial. She was managed conservatively during the remainder of her pregnancy with close observation only. She delivered a healthy infant at 37 weeks' gestation without any documented birth defects. Dasatinib has been shown in animal studies to cause fetal toxicities, but the effect of exposure during conception and pregnancy in humans is not known. We provide a full report of a successful pregnancy in a patient who was exposed to dasatinib during the first trimester; we also give a brief literature review.
Collapse
Affiliation(s)
- Soley Bayraktar
- Department of Hematology/Oncology, MD Anderson Cancer Center, Houston, Texas, USA.
| | | | | |
Collapse
|
40
|
An X, Tiwari AK, Sun Y, Ding PR, Ashby CR, Chen ZS. BCR-ABL tyrosine kinase inhibitors in the treatment of Philadelphia chromosome positive chronic myeloid leukemia: a review. Leuk Res 2010; 34:1255-68. [PMID: 20537386 DOI: 10.1016/j.leukres.2010.04.016] [Citation(s) in RCA: 213] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 04/25/2010] [Accepted: 04/25/2010] [Indexed: 01/09/2023]
Abstract
Chronic Myeloid Leukemia (CML) is a clonal disease characterized by the presence of the Philadelphia (Ph+) chromosome and its oncogenic product, BCR-ABL, a constitutively active tyrosine kinase, that is present in >90% of the patients. Epidemiologic data indicates that almost 5000 new cases are reported every year and 10% of these patients eventually succumb to the disease. The treatment of CML was revolutionized by the introduction of imatinib mesylate (IM, Gleevec), a BCR-ABL tyrosine kinase inhibitor (TKI). The clinical use of specific BCR-ABL inhibitors has resulted in a significantly improved prognosis, response rate, overall survival, and patient outcome in CML patients compared to previous therapeutic regimens. However, the complete eradication of CML in patients receiving imatinib was limited by the emergence of resistance mostly due to mutations in the ABL kinase domain and to a lesser extent by molecular residual disease after treatment. The second-generation BCR-ABL TKIs nilotinib (Tasigna) and dasatinib (Sprycel), showed significant activity in clinical trials in patients intolerant or resistant to imatinib therapy, except in those patients with the T315I BCR-ABL mutation. Identifying key components involved in the CML pathogenesis may lead to the exploration of new approaches that might eventually overcome resistance mediated to the BCR-ABL TKIs. Here, we present an overview about the current treatment of Ph+ CML patients with the TKIs and the obstacles to successful treatment with these drugs.
Collapse
Affiliation(s)
- Xin An
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA
| | | | | | | | | | | |
Collapse
|
41
|
Rocha GDG, Simões M, Oliveira RR, Kaplan MAC, Gattass CR. 3β-acetyl tormentic acid induces apoptosis of resistant leukemia cells independently of P-gp/ABCB1 activity or expression. Invest New Drugs 2010; 30:105-13. [DOI: 10.1007/s10637-010-9524-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 08/16/2010] [Indexed: 01/09/2023]
|
42
|
Razmkhah F, Razavi M, Zaker F, Kazemi A, Negari S, Rasighaemi P, Kalantarmotamedi M, Zarei M, Pazhakh V. Hematologic and Molecular Responses to Generic Imatinib in Patients With Chronic Myeloid Leukemia. Lab Med 2010. [DOI: 10.1309/lmr1ka61jykuxrdx] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
43
|
The proximal signaling network of the BCR-ABL1 oncogene shows a modular organization. Oncogene 2010; 29:5895-910. [PMID: 20697350 DOI: 10.1038/onc.2010.331] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BCR-ABL1 is a fusion tyrosine kinase, which causes multiple types of leukemia. We used an integrated proteomic approach that includes label-free quantitative protein complex and phosphorylation profiling by mass spectrometry to systematically characterize the proximal signaling network of this oncogenic kinase. The proximal BCR-ABL1 signaling network shows a modular and layered organization with an inner core of three leukemia transformation-relevant adaptor protein complexes (Grb2/Gab2/Shc1 complex, CrkI complex and Dok1/Dok2 complex). We introduced an 'interaction directionality' analysis, which annotates static protein networks with information on the directionality of phosphorylation-dependent interactions. In this analysis, the observed network structure was consistent with a step-wise phosphorylation-dependent assembly of the Grb2/Gab2/Shc1 and the Dok1/Dok2 complexes on the BCR-ABL1 core. The CrkI complex demonstrated a different directionality, which supports a candidate assembly on the Nedd9 (Hef1, CasL) scaffold. As adaptor protein family members can compensate for each other in leukemic transformation, we compared members of the Dok and Crk protein families and found both overlapping and differential binding patterns. We identified an additional level of regulation for the CrkII protein via binding to 14-3-3 proteins, which was independent from its inhibitory phosphorylation. We also identified novel components of the inner core complexes, including the kinases Pragmin (Sgk223) and Lrrk1 (Lrrk2 paralog). Pragmin was found as a component of the CrkI complex and is a potential link between BCR-ABL1/CrkI and RhoA signaling. Lrrk1 is an unusual kinase with a GTPase domain. We detected Lrrk1 as a component of the Grb2/Gab2/Shc1 complex and found that it functionally interacts with the regulator of small GTPases Arap1 (Centd2) and possibly participates in the mitogen-activated protein kinase response to cellular stresses. This modular and phosphorylation-driven interaction network provides a framework for the integration of pleiotropic signaling effects of BCR-ABL1 toward leukemic transformation.
Collapse
|
44
|
Harmon B, Campbell N, Ratner L. Role of Abl kinase and the Wave2 signaling complex in HIV-1 entry at a post-hemifusion step. PLoS Pathog 2010; 6:e1000956. [PMID: 20585556 PMCID: PMC2887473 DOI: 10.1371/journal.ppat.1000956] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 05/19/2010] [Indexed: 11/18/2022] Open
Abstract
Entry of human immunodeficiency virus type 1 (HIV-1) commences with binding of the envelope glycoprotein (Env) to the receptor CD4, and one of two coreceptors, CXCR4 or CCR5. Env-mediated signaling through coreceptor results in Galphaq-mediated Rac activation and actin cytoskeleton rearrangements necessary for fusion. Guanine nucleotide exchange factors (GEFs) activate Rac and regulate its downstream protein effectors. In this study we show that Env-induced Rac activation is mediated by the Rac GEF Tiam-1, which associates with the adaptor protein IRSp53 to link Rac to the Wave2 complex. Rac and the tyrosine kinase Abl then activate the Wave2 complex and promote Arp2/3-dependent actin polymerization. Env-mediated cell-cell fusion, virus-cell fusion and HIV-1 infection are dependent on Tiam-1, Abl, IRSp53, Wave2, and Arp3 as shown by attenuation of fusion and infection in cells expressing siRNA targeted to these signaling components. HIV-1 Env-dependent cell-cell fusion, virus-cell fusion and infection were also inhibited by Abl kinase inhibitors, imatinib, nilotinib, and dasatinib. Treatment of cells with Abl kinase inhibitors did not affect cell viability or surface expression of CD4 and CCR5. Similar results with inhibitors and siRNAs were obtained when Env-dependent cell-cell fusion, virus-cell fusion or infection was measured, and when cell lines or primary cells were the target. Using membrane curving agents and fluorescence microscopy, we showed that inhibition of Abl kinase activity arrests fusion at the hemifusion (lipid mixing) step, suggesting a role for Abl-mediated actin remodeling in pore formation and expansion. These results suggest a potential utility of Abl kinase inhibitors to treat HIV-1 infected patients.
Collapse
Affiliation(s)
- Brooke Harmon
- Division of Molecular Oncology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | | | | |
Collapse
|
45
|
Kantarjian HM, Cortes J, La Rosée P, Hochhaus A. Optimizing therapy for patients with chronic myelogenous leukemia in chronic phase. Cancer 2010; 116:1419-30. [PMID: 20120030 DOI: 10.1002/cncr.24928] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Identification of BCR-ABL as the defining leukemogenic event in chronic myelogenous leukemia (CML) revolutionized the treatment of the disease. Imatinib, a potent BCR-ABL inhibitor, is the standard of care for the first-line treatment of patients with chronic-phase CML because of its high long-term response rates and favorable tolerability profile compared with previous standard therapies. However, resistance to imatinib develops in 2% to 4% of patients annually. For patients with acquired cytogenetic resistance to standard-dose imatinib (400 mg daily), imatinib dose escalation (600-800 mg daily) is an excellent first option for managing patients and achieving cytogenetic responses. However, for patients with primary resistance to imatinib, hematologic disease recurrence, or emergent BCR-ABL kinase domain mutations, imatinib dose escalation may not be sufficient to control the disease. For these patients, the potent second-generation tyrosine kinase inhibitors dasatinib and nilotinib are available. Both agents provide effective therapeutic options for patients with imatinib resistance or intolerance. For the current overview, the authors reviewed the data supporting the use of both dasatinib and nilotinib in imatinib-resistant or imatinib-intolerant patients, and they have highlighted the future of CML therapy. Overall, the article is intended to offer physicians a comprehensive review of the current literature and to provide data supporting various treatment options for patients with CML throughout the course of imatinib therapy and beyond.
Collapse
Affiliation(s)
- Hagop M Kantarjian
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | |
Collapse
|
46
|
Kanojia D, Garg M, Saini S, Agarwal S, Kumar R, Suri A. Sperm associated antigen 9 expression and humoral response in chronic myeloid leukemia. Leuk Res 2010; 34:858-63. [PMID: 20138665 DOI: 10.1016/j.leukres.2010.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 12/12/2009] [Accepted: 01/15/2010] [Indexed: 02/08/2023]
Abstract
Early diagnosis and cure for patients with chronic myeloid leukemia (CML) exhibit significant clinical challenges because of the disease progression from chronic phase (CP) into a rapidly fatal blast crisis. Our earlier studies suggested an association of sperm associated antigen 9 (SPAG9) with various human malignancies. The present investigation revealed that SPAG9 mRNA and protein are expressed in CML patients (88%), in K562 and KCL-22 cells. Further, SPAG9 protein expression was also detected on cell surface suggesting that this molecule may be a suitable target for immunotherapy. Interestingly, 90% CML-CP patients showed humoral response against SPAG9, suggesting its important role in early diagnostic of CML-CP. Further investigation is warranted in establishing the potential of SPAG9 as a biomarker and immunotherapeutic target for early treatment of CML-CP patients.
Collapse
Affiliation(s)
- Deepika Kanojia
- Cancer Microarray, Genes and Proteins Laboratory, National Institute of Immunology, New Delhi, India
| | | | | | | | | | | |
Collapse
|
47
|
Breccia M, Palandri F, Iori AP, Colaci E, Latagliata R, Castagnetti F, Torelli GF, Usai S, Valle V, Martinelli G, Rosti G, Foà R, Baccarani M, Alimena G. Second-generation tyrosine kinase inhibitors before allogeneic stem cell transplantation in patients with chronic myeloid leukemia resistant to imatinib. Leuk Res 2010; 34:143-7. [PMID: 19481800 DOI: 10.1016/j.leukres.2009.04.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 04/25/2009] [Accepted: 04/28/2009] [Indexed: 12/11/2022]
|
48
|
Modulation of Akt and ERK1/2 pathways by resveratrol in chronic myelogenous leukemia (CML) cells results in the downregulation of Hsp70. PLoS One 2010; 5:e8719. [PMID: 20090934 PMCID: PMC2806839 DOI: 10.1371/journal.pone.0008719] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2009] [Accepted: 12/15/2009] [Indexed: 02/03/2023] Open
Abstract
Background Resveratrol is known to downregulate the high endogenous level of Heat shock protein 70 (Hsp70) in Chronic Myelogenous Leukemia (CML) K562 cells and induce apoptosis. Since Heat Shock Factor 1 (HSF1) controls transcription of Hsp70, we wanted to probe the signaling pathways responsible for transcriptional activation of HSF1. Methodology/Principal Findings Cells exposed to 40µM Resveratrol rapidly abolished serine473 phosphorylation of Akt and significantly reduced its kinase activity. Inactivation of Akt pathway by Resveratrol subsequently blocked serine9 phosphorylation of Gsk3β. Active non-phosphorylated Gsk3β rendered HSF1 transcriptionally inactive and reduced Hsp70 production. Blocking PI3K/Akt activity also demonstrated similar effects on Hsp70 comparable to Resveratrol. Inactivation of Gsk3β activity by inhibitors SB261763 or LiCl upregulated Hsp70. Resveratrol significantly modulated ERK1/2 activity as evident from hyper phosphorylation at T302/Y304 residues and simultaneous upregulation in kinase activity. Blocking ERK1/2 activation resulted in induction of Hsp70. Therefore, increase in ERK1/2 activity by Resveratrol provided another negative influence on Hsp70 levels through negative regulation of HSF1 activity. 17-allylamino-17-demethoxygeldanamycin (17AAG), a drug that inhibits Hsp90 chaperone and degrades its client protein Akt concomitantly elevated Hsp70 levels by promoting nuclear translocation of HSF1 from the cytosol. This effect is predominantly due to inhibition of both Akt and ERK1/2 activation by 17AAG. Simultaneously treating K562 with Resveratrol and 17AAG maintained phosho-ERK1/2 levels close to untreated controls demonstrating their opposite effects on ERK1/2 pathway. Resveratrol was found not to interfere with Bcr-Abl activation in K562 cells. Conclusion/Significance Thus our study comprehensively illustrates that Resveratrol acts downstream of Bcr-Abl and inhibits Akt activity but stimulates ERK1/2 activity. This brings down the transcriptional activity of HSF1 and Hsp70 production in K562 cells. Additionally, Resveratrol can be used in combination with chemotherapeutic agents such as 17AAG, an Hsp90 inhibitor reported to induce Hsp70 and hence compromise its chemotherapeutic potential.
Collapse
|
49
|
Huang WS, Zhu X, Wang Y, Azam M, Wen D, Sundaramoorthi R, Thomas RM, Liu S, Banda G, Lentini SP, Das S, Xu Q, Keats J, Wang F, Wardwell S, Ning Y, Snodgrass JT, Broudy MI, Russian K, Daley GQ, Iuliucci J, Dalgarno DC, Clackson T, Sawyer TK, Shakespeare WC. 9-(Arenethenyl)purines as dual Src/Abl kinase inhibitors targeting the inactive conformation: design, synthesis, and biological evaluation. J Med Chem 2009; 52:4743-56. [PMID: 19572547 DOI: 10.1021/jm900166t] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A novel series of potent dual Src/Abl kinase inhibitors based on a 9-(arenethenyl)purine core has been identified. Unlike traditional dual Src/Abl inhibitors targeting the active enzyme conformation, these inhibitors bind to the inactive, DFG-out conformation of both kinases. Extensive SAR studies led to the discovery of potent and orally bioavailable inhibitors, some of which demonstrated in vivo efficacy. Once-daily oral administration of inhibitor 9i (AP24226) significantly prolonged the survival of mice injected intravenously with wild type Bcr-Abl expressing Ba/F3 cells at a dose of 10 mg/kg. In a separate model, oral administration of 9i to mice bearing subcutaneous xenografts of Src Y527F expressing NIH 3T3 cells elicited dose-dependent tumor shrinkage with complete tumor regression observed at the highest dose. Notably, several inhibitors (e.g., 14a, AP24163) exhibited modest cellular potency (IC50 = 300-400 nM) against the Bcr-Abl mutant T315I, a variant resistant to all currently marketed therapies for chronic myeloid leukemia.
Collapse
Affiliation(s)
- Wei-Sheng Huang
- ARIAD Pharmaceuticals, Inc., 26 Landsdowne Street, Cambridge, Massachusetts 02139, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fujii Y, Amano M, Seriu T. Pharmacological properties and clinical efficacy of dasatinib hydrate (Sprycel), an anticancer drug for chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. Nihon Yakurigaku Zasshi 2009; 134:159-67. [PMID: 19749489 DOI: 10.1254/fpj.134.159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|