Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 373: 22-32, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36375637

ABSTRACT

Dasatinib, a second-generation BCR-ABL inhibitor, is currently used as first-line treatment for patients with chronic myeloid leukemia. However, dasatinib treatment increases the risk of severe cutaneous toxicity, which limits its long-term safe use in clinic. The underlying mechanism for dasatinib-induced cutaneous toxicity has not been clarified. In this study, we tested the toxicity of dasatinib on human immortal keratinocyte line (HaCaT) and normal human epidermal keratinocytes (NHEK). We found that dasatinib directly caused cytotoxicity on keratinocytes, which could be the explanation of the clinical characteristic of pathology. Mechanistically, dasatinib impaired mitophagy by downregulating HMGB1 protein level in keratinocytes, which led to the accumulation of dysfunctional mitochondria. Mitochondria-derived ROS caused DNA damage and cell apoptosis. More importantly, we confirmed that overexpression of HMGB1 could reverse dasatinib-induced keratinocyte apoptosis, and preliminarily explored the intervention effect of saikosaponin A, which could increase HMGB1 expression, on cutaneous toxicity caused by dasatinib. Collectively, our study revealed that dasatinib induced keratinocyte apoptosis via inhibiting HMGB1-mediated mitophagy and saikosaponin A could be a viable strategy for prevention of dasatinib-induced cutaneous toxicity.


Subject(s)
Apoptosis , Dasatinib , Humans , Apoptosis/drug effects , Dasatinib/toxicity , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/drug effects , HMGB1 Protein/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mitophagy/drug effects , Protein Kinase Inhibitors/pharmacology
2.
J Pharmacol Exp Ther ; 380(2): 114-125, 2022 02.
Article in English | MEDLINE | ID: mdl-34794962

ABSTRACT

Drug-induced liver injury (DILI) is the leading cause of acute liver failure and a major concern in drug development. Altered bile acid homeostasis via inhibition of the bile salt export pump (BSEP) is one mechanism of DILI. Dasatinib, pazopanib, and sorafenib are tyrosine kinase inhibitors (TKIs) that competitively inhibit BSEP and increase serum biomarkers for hepatotoxicity in ∼25-50% of patients. However, the mechanism(s) of hepatotoxicity beyond competitive inhibition of BSEP are poorly understood. This study examined mechanisms of TKI-mediated hepatotoxicity associated with altered bile acid homeostasis. Dasatinib, pazopanib, and sorafenib showed bile acid-dependent toxicity at clinically relevant concentrations, based on the C-DILI assay using sandwich-cultured human hepatocytes (SCHH). Among several bile acid-relevant genes, cytochrome P450 (CYP) 7A1 mRNA was specifically upregulated by 6.2- to 7.8-fold (dasatinib) and 5.7- to 9.3-fold (pazopanib), compared with control, within 8 hours. This was consistent with increased total bile acid concentrations in culture medium up to 2.3-fold, and in SCHH up to 1.4-fold, compared with control, within 24 hours. Additionally, protein abundance of sodium taurocholate co-transporting polypeptide (NTCP) was increased up to 2.0-fold by these three TKIs. The increase in NTCP protein abundance correlated with increased function; dasatinib and pazopanib increased hepatocyte uptake clearance (CLuptake) of taurocholic acid, a probe bile acid substrate, up to 1.4-fold. In conclusion, upregulation of CYP7A1 and NTCP in SCHH constitute novel mechanisms of TKI-associated hepatotoxicity. SIGNIFICANCE STATEMENT: Understanding the mechanisms of hepatotoxicity associated with tyrosine kinase inhibitors (TKIs) is fundamental to development of effective and safe intervention therapies for various cancers. Data generated in sandwich-cultured human hepatocytes, an in vitro model of drug-induced hepatotoxicity, revealed that TKIs upregulate bile acid synthesis and alter bile acid uptake and excretion. These findings provide novel insights into additional mechanisms of bile acid-mediated drug-induced liver injury, an adverse effect that limits the use and effectiveness of TKI treatment in some cancer patients.


Subject(s)
Antineoplastic Agents/toxicity , Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/drug effects , Protein Kinase Inhibitors/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , Cells, Cultured , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Dasatinib/toxicity , Hepatocytes/metabolism , Humans , Indazoles/toxicity , Organic Anion Transporters, Sodium-Dependent/metabolism , Pyrimidines/toxicity , Sorafenib/toxicity , Sulfonamides/toxicity , Symporters/metabolism
3.
Int J Pharm ; 596: 120226, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33484922

ABSTRACT

BACKGROUND AND AIMS: Ocular safety/biocompatibility is an essential element of ophthalmic drug delivery. We previously applied poly(ethylene glycol)-block-poly(ɛ-caprolactone) (PEG-b-PCL) micelles to deliver dasatinib for the management of proliferative vitreoretinopathy (PVR) in vitro. Herein, we seek to ascertain the ocular safety/compatibility of blank and dasatinib loaded PEG-b-PCL micelles, which will set the stage for the future in vivo efficacy evaluations and/or clinical translation for PVR or other eye diseases. METHODS: To access the safety of blank and dasatinib loaded micelles, in vitro cell based assays (LDH cell membrane damage test, SRB cytotoxicity, TEER and permeability of RPE tight junctions), in vivo slit lamp biomicroscopy and optical coherence tomography, Ex vivo histology (H&E staining, GFAP immunofluorescence staining and TUNEL assay) were undertaken. RESULTS: Both blank and dasatinib loaded micelles showed remarkable safety profiles at cellular levels. They also caused negligible ocular toxicity/abnormalities up to 28 days post-intravitreal injection in mice. The micelles did not insult the cornea, as demonstrated by slit-lamp biomicroscopy. Ex vivo histology and in vivo optical coherence tomography revealed a normal retinal structure with minimal apoptosis and stresses. CONCLUSION: Taken together, both blank and dasatinib loaded micelles appear to be safe and their applications in drug delivery for eye diseases should be explored.


Subject(s)
Drug Delivery Systems , Micelles , Animals , Cell Survival , Dasatinib/toxicity , Drug Carriers , Mice , Polyesters , Polyethylene Glycols , Polymers
4.
Biomed Pharmacother ; 134: 111134, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33341672

ABSTRACT

Dasatinib is a targeted cancer therapy, while programmed death ligand 1 (PD-L1) inhibitors are a form of immune checkpoint therapy used to treat various types of cancers. Several studies showed the potential efficacy of these drugs in the management of triple-negative breast cancer- an aggressive subtype of breast cancer, which can develop during pregnancy. Nevertheless, side effects of Dasatinib (DA) and PD-L1 drugs during pregnancy, especially in the early stages of embryogenesis are not explored yet. The aim of this study is to assess the individual and combined toxicity of DA and PD-L1 inhibitors during the early stages of embryogenesis and to evaluate their effect(s) on angiogenesis using the chorioallantoic membrane (CAM) model of the embryo. Our results show that embryos die at greater rates after exposure to DA and PD-L1 inhibitors as compared to their matched controls. Moreover, treatment with these drugs significantly inhibits angiogenesis of the CAM. To further elucidate key regulator genes of embryotoxicity induced by the actions of PD-L1 and DA, an RT-PCR analysis was performed for seven target genes that regulate cell proliferation, angiogenesis, and survival (ATF3, FOXA2, MAPRE2, RIPK1, INHBA, SERPINA4, and VEGFC). Our data revealed that these genes are significantly deregulated in the brain, heart, and liver tissues of exposed embryos, compared to matched control tissues. Nevertheless, further studies are necessary to evaluate the effects of these anti breast cancer drugs and elucidate their role during pregnancy.


Subject(s)
Acetamides/toxicity , Angiogenesis Inhibitors/toxicity , B7-H1 Antigen/antagonists & inhibitors , Chorioallantoic Membrane/blood supply , Dasatinib/toxicity , Immune Checkpoint Inhibitors/toxicity , Neovascularization, Physiologic/drug effects , Protein Kinase Inhibitors/toxicity , Pyridines/toxicity , Animals , B7-H1 Antigen/metabolism , Chick Embryo , Embryonic Development/drug effects , Gene Expression Regulation, Developmental , Neovascularization, Physiologic/genetics , Signal Transduction
5.
Cardiovasc Toxicol ; 20(4): 380-389, 2020 08.
Article in English | MEDLINE | ID: mdl-32124237

ABSTRACT

The anticancer drug dasatinib (Sprycel) is a BCR-ABL1-targeted tyrosine kinase inhibitor used in treating chronic myelogenous leukemia that has been shown in clinical trials to display cardiovascular toxicities. While dasatinib potently inhibits BCR-ABL1, it is not a highly selective kinase inhibitor and may have off-target effects. A neonatal rat cardiac myocyte model was used to investigate potential mechanisms by which dasatinib damaged myocytes. The anthracycline cardioprotective drug dexrazoxane was shown to be ineffective in preventing dasatinib-induced myocyte damage. Dasatinib treatment increased doxorubicin accumulation in myocytes and doxorubicin-induced myocyte damage, likely through its ability to bind to one or more ABC-type efflux transporters. Dasatinib induced myocyte damage either after a brief treatment that mimicked the clinical situation, or more potently after continuous treatment. Dasatinib slightly induced apoptosis in myocytes as evidenced by increases in caspase-3/7 activity. Dasatinib treatment reduced pERK levels in myocytes most likely through inhibition of RAF, which dasatinib strongly inhibits. Thus, inhibition of the RAF/MEK/ERK pro-survival pathway in the heart may be, in part, a mechanism by which dasatinib induces cardiovascular toxicity.


Subject(s)
Antineoplastic Agents/toxicity , Dasatinib/toxicity , Heart Diseases/chemically induced , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/toxicity , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cardiotoxicity , Caspase 7/metabolism , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart Diseases/metabolism , Heart Diseases/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , raf Kinases/metabolism
6.
Cardiovasc Toxicol ; 20(1): 58-70, 2020 02.
Article in English | MEDLINE | ID: mdl-31280457

ABSTRACT

Tyrosine kinase inhibitors are known to clinically induce various types of cardiovascular adverse events; however, it is still difficult to predict them at preclinical stage. In order to explore how to better predict such drug-induced cardiovascular adverse events, we tried to develop a new protocol by assessing acute electrophysiological, cardiohemodynamic, and cytotoxic effects of dasatinib in vivo and in vitro. Dasatinib at 0.03 and 0.3 mg/kg was intravenously administered to the halothane-anesthetized dogs for 10 min with an interval of 20 min between the dosing (n = 4). Meanwhile, that at 0.1, 0.3, and 1 µM was cumulatively applied to the human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) (n = 7). In the dogs, the low and high doses provided peak plasma concentrations of 40 ± 5 (0.08) and 615 ± 38 ng/mL (1.26 µM), respectively. The low dose decreased the heart rate, impaired the left ventricular mechanical function, and prolonged the ventricular effective refractory period. The high dose prolonged the repolarization period, induced hemorrhagic tendency, and increased plasma cardiac troponin I level in addition to enhancement of the changes observed after the low dose, whereas it neither affected the cardiac conduction nor induced ventricular arrhythmias. In the hiPSC-CMs, dasatinib prolonged the repolarization and refractory periods like in dogs, while it did not induce apoptotic or necrotic process, but that it increased the conduction speed. Clinically observed major cardiovascular adverse events of dasatinib were observed qualitatively by currently proposed assay protocol, which may become a useful guide for predicting the cardiotoxicity of new tyrosine kinase inhibitors.


Subject(s)
Antineoplastic Agents/toxicity , Arrhythmias, Cardiac/chemically induced , Dasatinib/toxicity , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/toxicity , Ventricular Dysfunction, Left/chemically induced , Ventricular Function, Left/drug effects , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cardiotoxicity , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Female , Heart Rate/drug effects , Humans , Induced Pluripotent Stem Cells/metabolism , Molecular Targeted Therapy/adverse effects , Myocytes, Cardiac/metabolism , Refractory Period, Electrophysiological , Risk Assessment , Time Factors , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
7.
Sci Rep ; 9(1): 15400, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659178

ABSTRACT

Finding novel targets in non-small cell lung cancer (NSCLC) is highly needed and identification of synthetic lethality between two genes is a new approach to target NSCLC. We previously found that TMPRSS4 promotes NSCLC growth and constitutes a prognostic biomarker. Here, through large-scale analyses across 5 public databases we identified consistent co-expression between TMPRSS4 and DDR1. Similar to TMPRSS4, DDR1 promoter was hypomethylated in NSCLC in 3 independent cohorts and hypomethylation was an independent prognostic factor of disease-free survival. Treatment with 5-azacitidine increased DDR1 levels in cell lines, suggesting an epigenetic regulation. Cells lacking TMPRSS4 were highly sensitive to the cytotoxic effect of the DDR1 inhibitor dasatinib. TMPRSS4/DDR1 double knock-down (KD) cells, but not single KD cells suffered a G0/G1 cell cycle arrest with loss of E2F1 and cyclins A and B, increased p21 levels and a larger number of cells in apoptosis. Moreover, double KD cells were highly sensitized to cisplatin, which caused massive apoptosis (~40%). In vivo studies demonstrated tumor regression in double KD-injected mice. In conclusion, we have identified a novel vulnerability in NSCLC resulting from a synthetic lethal interaction between DDR1 and TMPRSS4.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Discoidin Domain Receptor 1/genetics , Lung Neoplasms/genetics , Membrane Proteins/genetics , Serine Endopeptidases/genetics , Synthetic Lethal Mutations , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/toxicity , DNA Methylation , Dasatinib/toxicity , Drug Resistance, Neoplasm , Humans , Lung Neoplasms/pathology , Mice , Promoter Regions, Genetic
8.
Toxicol Lett ; 296: 39-47, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30086328

ABSTRACT

Dasatinib shows remarkable activity against imatinib-refractory chronic myelogenous leukemia (CML) and Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ALL). However, severe cardiovascular toxicity limits the clinical applications of dasatinib. Since the underlying mechanism of dasatinib-induced cardiotoxicity is still elusive, we aim to clarify this. Recent studies have shown that necroptosis and apoptosis participate in multiple toxicity development. Here, we first report that dasatinib could directly induce cardiomyocytes death, as analyzed by the Sulforhodamine B (SRB) assay. This type of cardiomyocytes death was mediated by the necrosis pathway rather than apoptosis, as determined by using flow cytometry to characterize the mode of dasatinib-induced cell death. Inhibition of receptor-interacting protein kinase 1 (RIP1)activity and knockdown of receptor-interacting protein kinase 3 (RIP3)expression can block dasatinib-evoked cardiotoxicity, which further confirmed the involvement of necroptosis. We next found that the classic substrates of RIP3, mixed lineage kinase domain-like protein (MLKL) and Ca2+-calmodulin-dependent protein kinase II (CaMKII) were not involved in dasatinib-induced cardiomyocytes necroptosis. What's more, unlike the inflammation-associated necroptosis, dasatinib-triggered necroptosis was dependent on intracellular instead of secreted High-mobility group box 1 (HMGB1) protein. Collectively, our study revealed that dasatinib-induced cardiotoxicity acted via leading cardiomyocytes to HMGB1-mediated necroptosis, indicating a viable strategy for prevention of dasatinib-induced cardiotoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Dasatinib/toxicity , HMGB1 Protein/metabolism , Heart Diseases/chemically induced , Necrosis/chemically induced , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiotoxicity , Cell Death/drug effects , Cell Line , Humans , Myocytes, Cardiac/drug effects , Necrosis/pathology , Nuclear Pore Complex Proteins/biosynthesis , Nuclear Pore Complex Proteins/genetics , Protein Kinases/biosynthesis , Protein Kinases/genetics , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/biosynthesis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
9.
Toxicol Mech Methods ; 28(9): 678-684, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29975149

ABSTRACT

Dasatinib is a new selective tyrosine kinase inhibitor that targets certain kinases involved in cellular growth and development. This drug belongs to a novel anticancer therapy aiming to increase the survival in patients with imatinib-resistant mutations. However, the dasatinib toxicity was reported as a side effect leading to arrhythmias and/or heart failure. Here, we investigated the possibility of dasatinib-induced toxicity in rat cardiomyocyte H9c2 cells. Our objectives were to investigate the ability of dasatinib to induce expression of cytochrome P450 (CYP1A1, CYP1B1) and cardiac hypertrophy markers (BNP, ß-MHC) genes in H9c2 cells. To test this hypothesis, H9c2 cells were incubated with dasatinib at two concentrations (20 and 40 µM). Thereafter, CYP1A1, CYP1B1, BNP, and ß-MHC were determined at gene expression level. Our findings showed that dasatinib induces the CYP1A1, CYP1B1, BNP, and ß-MHC mRNA. The involvement of AhR/CYP1A1 pathway in dasatinib toxicity was tested by resveratrol (RES), an AhR antagonist. Interestingly, the increase in mRNA of different genes by dasatinib was not affected by RES, which confirms that these effects are not mediated through AhR. In addition, this was accompanied by a significant inhibition of constitutive expression of these genes by RES. The current work provides the first evidence for the ability of dasatinib to induce hypertrophic markers in H9c2 cells through AhR-independent pathway.


Subject(s)
Antineoplastic Agents/toxicity , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1/genetics , Dasatinib/toxicity , Myocytes, Cardiac/drug effects , Myosin Heavy Chains/genetics , Natriuretic Peptide, Brain/genetics , Animals , Biomarkers/analysis , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiotoxicity , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Gene Expression/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats
10.
Toxicol Lett ; 295: 10-21, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-29803841

ABSTRACT

Dasatinib, a new tyrosine kinase inhibitor, is used clinically to kill chronic myelogenous leukemia and acute lymphoblastic leukemia through apoptosis. Obviously, anemia is developed in many patients receiving dasatinib for treatment. Until now, the mechanism for the cytotoxic effects of dasatinib in human erythrocytes is not fully understood. As many tyrosine kinases are found in human erythrocytes, it is therefore logical to hypothesize that dasatinib is able to induce apoptosis (or eryptosis) in human erythrocytes. True to our expectation, dasatinib inhibited tyrosine kinase and induced eryptosis in human erythrocytes with early denature of esterase, cell shrinkage, loss of membrane integrity with inside-out phosphatidylserine, increase in the cytosolic Ca2+ ion concentration ([Ca2+]i), caspase-3 activation and change in cellular redox state. Mechanistically, the rise of [Ca2+]i seems to be a key mediator in the dasatinib-mediated eryptosis because depletion of external Ca2+ could suppress the eryptotic effects. Also, dasatinib was able to reduce membrane fluidity in human RBCs. For the direct action on membrane, dasatinib permeabilized RBC ghosts in a way similar to digitonin. Taken together, we report here for the first time that dasatinib inhibited tyrosine kinase and induced eryptosis in human erythrocytes through Ca2+ loading and membrane permeabilization.


Subject(s)
Antineoplastic Agents/toxicity , Dasatinib/toxicity , Eryptosis/drug effects , Erythrocytes/drug effects , Protein Kinase Inhibitors/toxicity , Calcium/metabolism , Calcium Signaling/drug effects , Carboxylesterase/metabolism , Caspase 3/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/pathology , Erythrocytes/enzymology , Erythrocytes/pathology , HL-60 Cells , Hep G2 Cells , Humans , Membrane Fluidity/drug effects , Oxidation-Reduction , Phosphatidylserines/metabolism , Protein Denaturation
11.
Bioorg Med Chem Lett ; 28(4): 700-706, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29395973

ABSTRACT

We used the concept of bioisosteres to design and synthesize a novel series of dasatinib derivatives for the treatment of leukemia. Unfortunately, most of the dasatinib derivatives did not show appreciable inhibition against leukemia cell lines K562 and HL60. However, acrylamide compound 2c had comparable inhibitory activity with dasatinib against K562 cells (IC50 = 0.039 nM vs. 0.069 nM). And amide compound 2a and acrylamide compound 2c also had comparable inhibitory activity with dasatinib against the leukemia cell line HL60 (IC50 = 0.25 nM and 0.26 nM vs. 0.11 nM). Against the leukemia progenitor cell line KG1a, triazole compounds 15a and 15d-15f and oxadiazole compounds 24a-24d were more potent than dasatinib. In particular, the hydroxyl compounds 15a and 24a were about 64 and 180 fold more potent than dasatinib against KG1a cells (IC50 = 0.14 µM and 0.05 µM vs. 8.98 µM). Compounds 15a and 24a also inhibited colony formation in MCF-7 cells and inhibited cell migration in the cell wound scratch assay in B16BL6 cells. Moreover, hydroxyl compounds 15a and 24a had low toxicity in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Dasatinib/analogs & derivatives , Dasatinib/pharmacology , Leukemia/drug therapy , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Dasatinib/chemical synthesis , Dasatinib/toxicity , Drug Design , Drug Screening Assays, Antitumor , Humans , Mice , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacology , Oxadiazoles/toxicity , Triazoles/chemical synthesis , Triazoles/pharmacology , Triazoles/toxicity
12.
Cancer Chemother Pharmacol ; 79(4): 711-723, 2017 04.
Article in English | MEDLINE | ID: mdl-28283735

ABSTRACT

PURPOSE: Pulmonary arterial hypertension (PAH) results from occlusion or vasoconstriction of pulmonary vessels, leading to progressive right ventricular failure. Dasatinib, a BCR-ABL1 tyrosine kinase inhibitor (TKI) approved for the treatment of chronic myelogenous leukemia, has been associated with PAH. In contrast, the BCR-ABL1 TKI imatinib has demonstrated anti-vasoproliferative properties and has been investigated as a potential treatment for PAH. Here we describe studies evaluating the effects of dasatinib and imatinib on cardiovascular and pulmonary functions to understand the reported differential consequences of the two TKIs in a clinical setting. METHODS: The direct effects of dasatinib and imatinib were explored in vivo to investigate possible mechanisms of dasatinib-induced PAH. In addition, effects of dasatinib and imatinib on PAH-related mediators were evaluated in vitro. RESULTS: In rats, both TKIs increased plasma nitric oxide (NO), did not induce PAH-related structural or molecular changes in PA or lungs, and did not alter hemodynamic lung function compared with positive controls. Similarly, in the pulmonary artery endothelial cells and smooth muscle cells co-culture model, imatinib and dasatinib increased NO and decreased endothelin-1 protein and mRNA. CONCLUSIONS: The results of these studies indicated that dasatinib did not induce physiological changes or molecular signatures consistent with PAH when compared to positive controls. Instead, dasatinib induced changes consistent with imatinib. Both dasatinib and imatinib induced biochemical and structural changes consistent with a protective effect for PAH. These data suggest that other factors of unclear etiology contributed to the development of PAH in patients treated with dasatinib.


Subject(s)
Antineoplastic Agents/toxicity , Dasatinib/toxicity , Hypertension, Pulmonary/chemically induced , Imatinib Mesylate/toxicity , Protein Kinase Inhibitors/toxicity , Animals , Antineoplastic Agents/pharmacokinetics , Dasatinib/pharmacokinetics , Endothelin-1/blood , Gene Expression/drug effects , Hemodynamics/drug effects , Hypertension, Pulmonary/physiopathology , Imatinib Mesylate/pharmacokinetics , Lung/pathology , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Nitric Oxide/blood , Protein Kinase Inhibitors/pharmacokinetics , Pulmonary Artery/drug effects , Pulmonary Circulation/drug effects , RNA, Messenger/blood , Rats , Rats, Sprague-Dawley
13.
PLoS One ; 10(6): e0128542, 2015.
Article in English | MEDLINE | ID: mdl-26083411

ABSTRACT

Multivariate biomarkers that can predict the effectiveness of targeted therapy in individual patients are highly desired. Previous biomarker discovery studies have largely focused on the identification of single biomarker signatures, aimed at maximizing prediction accuracy. Here, we present a different approach that identifies multiple biomarkers by simultaneously optimizing their predictive power, number of features, and proximity to the drug target in a protein-protein interaction network. To this end, we incorporated NSGA-II, a fast and elitist multi-objective optimization algorithm that is based on the principle of Pareto optimality, into the biomarker discovery workflow. The method was applied to quantitative phosphoproteome data of 19 non-small cell lung cancer (NSCLC) cell lines from a previous biomarker study. The algorithm successfully identified a total of 77 candidate biomarker signatures predicting response to treatment with dasatinib. Through filtering and similarity clustering, this set was trimmed to four final biomarker signatures, which then were validated on an independent set of breast cancer cell lines. All four candidates reached the same good prediction accuracy (83%) as the originally published biomarker. Although the newly discovered signatures were diverse in their composition and in their size, the central protein of the originally published signature - integrin ß4 (ITGB4) - was also present in all four Pareto signatures, confirming its pivotal role in predicting dasatinib response in NSCLC cell lines. In summary, the method presented here allows for a robust and simultaneous identification of multiple multivariate biomarkers that are optimized for prediction performance, size, and relevance.


Subject(s)
Algorithms , Antineoplastic Agents/toxicity , Dasatinib/toxicity , Proteome/drug effects , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cluster Analysis , Dasatinib/therapeutic use , Humans , Integrin beta4/genetics , Integrin beta4/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phosphopeptides/metabolism , Phosphorylation/drug effects , Proteome/metabolism
14.
PLoS One ; 10(3): e0122555, 2015.
Article in English | MEDLINE | ID: mdl-25799134

ABSTRACT

Up to now, several clinical studies have been started investigating the relevance of receptor tyrosine kinase (RTK) inhibitors upon progression free survival in various pediatric brain tumors. However, single targeted kinase inhibition failed, possibly due to tumor resistance mechanisms. The present study will extend our previous observations that vascular endothelial growth factor receptor (VEGFR)-2, platelet derived growth factor receptor (PDGFR)ß, Src, the epidermal growth factor receptor (ErbB) family, and hepatocyte growth factor receptor (HGFR/cMet) are potentially drugable targets in pediatric low grade astrocytoma and ependymoma with investigations concerning growth-factor-driven rescue. This was investigated in pediatric low grade astrocytoma and ependymoma cell lines treated with receptor tyrosine kinase (RTK) inhibitors e.g. sorafenib, dasatinib, canertinib and crizotinib. Flow cytometry analyses showed high percentage of cells expressing VEGFR-1, fibroblast growth factor receptor (FGFR)-1, ErbB1/EGFR, HGFR and recepteur d'origine nantais (RON) (respectively 52-77%, 34-51%, 63-90%, 83-98%, 65-95%). Their respective inhibitors induced decrease of cell viability, measured with WST-1 cell viability assays. At least this was partially due to increased apoptotic levels measured by Annexin V/Propidium Iodide apoptosis assays. EGF, HGF and FGF, which are normally expressed in brain (tumor) tissue, showed to be effective rescue inducing growth factors resulting in increased cell survival especially during treatment with dasatinib (complete rescue) or sorafenib (partial rescue). Growth-factor-driven rescue was less prominent when canertinib or crizotinib were used. Rescue was underscored by significantly activating downstream Akt and/or Erk phosphorylation and increased tumor cell migration. Combination treatment showed to be able to overcome the growth-factor-driven rescue. In conclusion, our study highlights the extensive importance of environmentally present growth factors in developing tumor escape towards RTK inhibitors in pediatric low grade astrocytoma and ependymoma. It is of great interest to anticipate upon these results for the design of new therapeutic trials with RTK inhibitors in these pediatric brain tumors.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Astrocytoma/drug therapy , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Crizotinib , Dasatinib/therapeutic use , Dasatinib/toxicity , Ependymoma/drug therapy , Ependymoma/metabolism , Ependymoma/pathology , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Niacinamide/toxicity , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/toxicity , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrazoles/toxicity , Pyridines/therapeutic use , Pyridines/toxicity , Sorafenib
15.
PLoS One ; 10(2): e0117215, 2015.
Article in English | MEDLINE | ID: mdl-25658745

ABSTRACT

Cancer cells are characterized by a complex network of interrelated and compensatory signaling driven by multiple kinases that reduce their sensitivity to targeted therapy. Therefore, strategies directed at inhibiting two or more kinases are required to robustly block the growth of refractory tumour cells. Here we report on a novel strategy to promote sustained inhibition of two oncogenic kinases (Kin-1 and Kin-2) by designing a molecule K1-K2, termed "combi-molecule", to induce a tandem blockade of Kin-1 and Kin-2, as an intact structure and to be further hydrolyzed to two inhibitors K1 and K2 directed at Kin-1 and Kin-2, respectively. We chose to target EGFR (Kin-1) and c-Src (Kin-2), two tyrosine kinases known to synergize to promote tumour growth and progression. Variation of K1-K2 linkers led to AL776, our first optimized EGFR-c-Src targeting prototype. Here we showed that: (a) AL776 blocked EGFR and c-Src as an intact structure using an in vitro kinase assay (IC50 EGFR = 0.12 µM and IC50 c-Src = 3 nM), (b) it could release K1 (AL621, a nanomolar EGFR inhibitor) and K2 (dasatinib, a clinically approved Abl/c-Src inhibitor) by hydrolytic cleavage both in vitro and in vivo, (c) it could robustly inhibit phosphorylation of EGFR and c-Src (0.25-1 µM) in cells, (d) it induced 2-4 fold stronger growth inhibition than gefitinib or dasatinib and apoptosis at concentrations as low as 1 µM, and, (e) blocked motility and invasion at sub-micromolar doses in the highly invasive 4T1 and MDA-MB-231 cells. Despite its size (MW = 1032), AL776 blocked phosphorylation of EGFR and c-Src in 4T1 tumours in vivo. We now term this new targeting model consisting of designing a kinase inhibitor K1-K2 to target Kin-1 and Kin-2, and to further release two inhibitors K1 and K2 of the latter kinases, "type III combi-targeting".


Subject(s)
Apoptosis/drug effects , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/toxicity , Quinazolines/toxicity , Thiazoles/toxicity , src-Family Kinases/antagonists & inhibitors , Animals , Binding Sites , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dasatinib/toxicity , Drug Design , ErbB Receptors/metabolism , Female , Gefitinib , Humans , Mice , Mice, Inbred BALB C , Molecular Dynamics Simulation , NIH 3T3 Cells , Neoplasms/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Quinazolines/chemical synthesis , Quinazolines/therapeutic use , Thiazoles/chemical synthesis , Thiazoles/therapeutic use , Transplantation, Heterologous , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...