Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 473
Filter
1.
Toxicol Appl Pharmacol ; 486: 116944, 2024 May.
Article in English | MEDLINE | ID: mdl-38677603

ABSTRACT

Despite significant success, targeted therapeutics such as kinase inhibitors (KIs) still pose adverse events such as the cardiotoxicity. There is a lot of variation in the type and intensity of cardiotoxicity caused by different KIs and current pre-clinical models are inadequate to predict it. Thus, there is a need to develop more simple and rapid models for screening of novel KIs at the pre-clinical step itself. We thus aimed to establish a rapid and robust pre-clinical animal model for predicting cardiotoxicity of KIs and identify comparative cardiotoxicity profiles of a panel of FDA-approved KIs. Heart rate measurement and survival analysis of Daphnia was performed at regular intervals following treatment with ten KIs that were approved for the treatment of various cancers. The heart rates of Daphnia as well as the survival varied between KIs in a dose and time dependent manner suggesting differential cardiotoxicity profiles of various KIs. Further, the correlation between the cardiotoxicity and survival also varied among the ten KIs. Importantly, sorafenib and vemurafenib displayed maximum and least cardiotoxicity, respectively. The comparative cardiotoxicity profiles also are in conformity with the previous studies indicating the utility of Daphnia as a valuable and relevant animal model to rapidly predict the cardiotoxicity of novel KIs at a pre-clinical stage.


Subject(s)
Cardiotoxicity , Daphnia , Protein Kinase Inhibitors , Animals , Protein Kinase Inhibitors/toxicity , Daphnia/drug effects , Heart Rate/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Antineoplastic Agents/toxicity
2.
Circ Res ; 134(5): 482-501, 2024 03.
Article in English | MEDLINE | ID: mdl-38323474

ABSTRACT

BACKGROUND: Mitochondrial dysfunction is a primary driver of cardiac contractile failure; yet, the cross talk between mitochondrial energetics and signaling regulation remains obscure. Ponatinib, a tyrosine kinase inhibitor used to treat chronic myeloid leukemia, is among the most cardiotoxic tyrosine kinase inhibitors and causes mitochondrial dysfunction. Whether ponatinib-induced mitochondrial dysfunction triggers the integrated stress response (ISR) to induce ponatinib-induced cardiotoxicity remains to be determined. METHODS: Using human induced pluripotent stem cells-derived cardiomyocytes and a recently developed mouse model of ponatinib-induced cardiotoxicity, we performed proteomic analysis, molecular and biochemical assays to investigate the relationship between ponatinib-induced mitochondrial stress and ISR and their role in promoting ponatinib-induced cardiotoxicity. RESULTS: Proteomic analysis revealed that ponatinib activated the ISR in cardiac cells. We identified GCN2 (general control nonderepressible 2) as the eIF2α (eukaryotic translation initiation factor 2α) kinase responsible for relaying mitochondrial stress signals to trigger the primary ISR effector-ATF4 (activating transcription factor 4), upon ponatinib exposure. Mechanistically, ponatinib treatment exerted inhibitory effects on ATP synthase activity and reduced its expression levels resulting in ATP deficits. Perturbed mitochondrial function resulting in ATP deficits then acts as a trigger of GCN2-mediated ISR activation, effects that were negated by nicotinamide mononucleotide, an NAD+ precursor, supplementation. Genetic inhibition of ATP synthase also activated GCN2. Interestingly, we showed that the decreased abundance of ATP also facilitated direct binding of ponatinib to GCN2, unexpectedly causing its activation most likely because of a conformational change in its structure. Importantly, administering an ISR inhibitor protected human induced pluripotent stem cell-derived cardiomyocytes against ponatinib. Ponatinib-treated mice also exhibited reduced cardiac function, effects that were attenuated upon systemic ISRIB administration. Importantly, ISRIB does not affect the antitumor effects of ponatinib in vitro. CONCLUSIONS: Neutralizing ISR hyperactivation could prevent or reverse ponatinib-induced cardiotoxicity. The findings that compromised ATP production potentiates GCN2-mediated ISR activation have broad implications across various cardiac diseases. Our results also highlight an unanticipated role of ponatinib in causing direct activation of a kinase target despite its role as an ATP-competitive kinase inhibitor.


Subject(s)
Imidazoles , Induced Pluripotent Stem Cells , Mitochondrial Diseases , Pyridazines , Humans , Animals , Mice , Protein Serine-Threonine Kinases/metabolism , Cardiotoxicity/pathology , Proteomics , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase Inhibitors/toxicity , Mitochondrial Diseases/pathology , Adenosine Triphosphate
3.
PLoS One ; 18(11): e0294438, 2023.
Article in English | MEDLINE | ID: mdl-37983208

ABSTRACT

BCR-ABL tyrosine kinase inhibitors (TKIs) have dramatically improved survival in Philadelphia chromosome-positive leukemias. Newer BCR-ABL TKIs provide superior cancer outcomes but with increased risk of acute arterial thrombosis, which further increases in patients with cardiovascular comorbidities and mitigates survival benefits compared to imatinib. Recent studies implicate endothelial cell (EC) damage in this toxicity by unknown mechanisms with few side-by-side comparisons of multiple TKIs and with no available data on endothelial impact of recently approved TKIs or novels TKIs being tested in clinical trials. To characterize BCR-ABL TKI induced EC dysfunction we exposed primary human umbilical vein ECs in 2D and 3D culture to clinically relevant concentrations of seven BCR-ABL TKIs and quantified their impact on EC scratch-wound healing, viability, inflammation, and permeability mechanisms. Dasatinib, ponatinib, and nilotinib, the TKIs associated with thrombosis in patients, all significantly impaired EC wound healing, survival, and proliferation compared to imatinib, but only dasatinib and ponatinib impaired cell migration and only nilotinib enhanced EC necrosis. Dasatinib and ponatinib increased leukocyte adhesion to ECs with upregulation of adhesion molecule expression in ECs (ICAM1, VCAM1, and P-selectin) and leukocytes (PSGL1). Dasatinib increased permeability and impaired cell junctional integrity in human engineered microvessels, consistent with its unique association with pleural effusions. Of the new agents, bafetinib decreased EC viability and increased microvessel permeability while asciminib and radotinib did not impact any EC function tested. In summary, the vasculotoxic TKIs (dasatinib, ponatinib, nilotinib) cause EC toxicity but with mechanistic differences, supporting the potential need for drug-specific vasculoprotective strategies. Asciminib and radotinib do not induce EC toxicity at clinically relevant concentrations suggesting a better safety profile.


Subject(s)
Antineoplastic Agents , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Thrombosis , Humans , Imatinib Mesylate/adverse effects , Dasatinib/adverse effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/toxicity , Endothelial Cells , Thrombosis/drug therapy , Fusion Proteins, bcr-abl , Antineoplastic Agents/therapeutic use
4.
Int J Toxicol ; 42(6): 515-534, 2023 12.
Article in English | MEDLINE | ID: mdl-37667445

ABSTRACT

Roginolisib (IOA-244) is a novel, non-ATP competitive phosphoinositide-3-kinase (PI3K) delta inhibitor that regulates Akt/mTOR signaling. Roginolisib was administered once daily to rats and dogs in dose-range finding (DRF) and 4-week GLP toxicology studies. Free plasma levels of roginolisib exceeded the cellular target engagement IC90 for PI3Kδ for ≥12 hours at doses of 5 mg/kg, the IC90 for PI3Kß for ≥2 hours at doses ≥15 mg/kg, and the IC50 for PI3Kα for ≥2 hours at dose levels ≥45 mg/kg. Toxicity in rats occurred at doses ≥100 mg/kg. In dogs, we observed dose-dependent skin and gastrointestinal toxicity and doses ≥30 mg/kg had a greater incidence of mortality. Lymphoid tissue toxicity occurred in both species. Toxicities in dogs observed at the ≥15 mg/kg dose, affecting the digestive mucosa, liver, and skin, cleared after treatment cessation. Doses ≤75 mg/kg were tolerated in rats and the no-observed-adverse-effect-level (NOAEL) in rats was 15 mg/kg. Due to mainly epithelial lesions of the skin at 5 mg/kg and necrotizing damage of the intestinal epithelia at ≥15 mg/kg, no NOAEL was determined in dogs. However, the adverse effects observed in dogs at 5 mg/kg were considered monitorable and reversible in patients with advanced malignancies. Furthermore, the PK profile subsequently proved to be a decisive factor for achieving selective PI3Kδ inhibition without the toxicities observed in dogs. As the result of the unique PK profile of roginolisib, patients were able to take daily roginolisib without dose modification and showed pharmacodynamic PI3Kδ inhibition over several months without gastrointestinal or dermatologic toxicities.


Subject(s)
Antineoplastic Agents , Drug-Related Side Effects and Adverse Reactions , Humans , Animals , Dogs , Rats , Phosphatidylinositol 3-Kinases , Protein Kinase Inhibitors/toxicity , Phosphoinositide-3 Kinase Inhibitors/toxicity
5.
Toxicol In Vitro ; 93: 105691, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37660997

ABSTRACT

Severe diarrhea is a common side effect of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs). We aimed to evaluate the risk of EGFR-TKI-induced diarrhea using spheroids of human and monkey crypt-derived intestinal stem cells. Intestinal spheroids exhibited higher toxic susceptibility to EGFR-TKIs than Caco-2 cells. As concentration of EGFR-TKIs increased, cellular ATP first decreased relative to the control condition, followed by an increase in LDH release, in contrast with their simultaneous changes with traditional cytotoxic anticancer drugs. The toxic sensitivity of spheroids to various EGFR-TKIs corresponded to clinical diarrhea incidence. Afatinib, a second-generation EGFR-TKI, exhibited higher toxic sensitivity compared with the first-generation ones, corresponding to the clinical evidence that afatinib-induced diarrhea is almost inevitable and severe. By contrast, the third-generation osimertinib, which reduces the risk of diarrhea, showed mitigated cytotoxicity compared with afatinib. For irreversible EGFR-TKIs, the decreased ATP level persisted or its recovery was delayed even after drug removal compared with reversible ones. Furthermore, the highest drug accumulation in spheroids (TKIspheroids) and inhibition potency against EGFR (TKIspheroids/Ki) were observed for afatinib. This system would be useful for predicting the risk of EGFR-TKI-induced diarrhea; moreover, on-target cytotoxicity against intestinal stem cells might contribute to clinically observed diarrhea.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Afatinib/toxicity , Afatinib/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/toxicity , Haplorhini/metabolism , Caco-2 Cells , ErbB Receptors/metabolism , Mutation , Antineoplastic Agents/pharmacology , Diarrhea/chemically induced , Adenosine Triphosphate
6.
Biochem Pharmacol ; 215: 115636, 2023 09.
Article in English | MEDLINE | ID: mdl-37290598

ABSTRACT

Crizotinib is the first-line drug for advanced non-small cell lung cancer with the abnormal expression of anaplastic lymphoma kinase gene. Severe, life-threatening, or fatal interstitial lung disease/pneumonia has been reported in patients treated with crizotinib. The clinical benefit of crizotinib is limited by its pulmonary toxicity, but the underlying mechanisms have not been adequately studied, and protective strategies are relatively scarce. Here, we established an in vivo mouse model in which crizotinib was continuously administered to C57BL/6 at 100 mg/kg/day for 6 weeks and verified that crizotinib induced interstitial lung disease in vivo, which was consistent with the clinical observations. We further treated BEAS-2B and TC-1 cells, the alveolar epithelial cell lines, with crizotinib and found the increased apoptosis rate. We proved that crizotinib-blocked autophagic flux caused apoptosis of the alveolar epithelial cells and then promoted the recruitment of immune cells, suggesting that limited autophagy activity was the key reason for pulmonary injury and inflammation caused by crizotinib. Subsequently, we found that metformin could reduce the macrophage recruitment and pulmonary fibrosis by recovering the autophagy flux, thereby ameliorating impaired lung function caused by crizotinib. In conclusion, our study revealed the mechanism of crizotinib-induced apoptosis of alveolar epithelial cells and activation of inflammation during the onset of pulmonary toxicity and provided a promising therapeutic strategy for the treatment of crizotinib-induced pulmonary toxicity.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Diseases, Interstitial , Lung Neoplasms , Mice , Animals , Crizotinib/toxicity , Alveolar Epithelial Cells , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mice, Inbred C57BL , Lung Diseases, Interstitial/drug therapy , Autophagy , Inflammation/metabolism , Protein Kinase Inhibitors/toxicity
7.
Toxicol Appl Pharmacol ; 465: 116469, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36918129

ABSTRACT

Promiscuity of therapeutics has important implications in treatment and toxicity. So far, a comprehensive understanding of promiscuity related to kinase inhibitors is lacking and such an analysis may offer potential opportunities for drug repurposing. In the present study, profiling of inhibitor-specific kinases based on the available biochemical IC50s was performed, fold-change of IC50 values for additional targets were calculated by taking the primary target as the reference kinase, and finally the promiscuity degree (PD) for FDA-approved kinase inhibitors was calculated. Surprisingly, class II inhibitors showed more PD than that of the class I inhibitors. We further identified cancer types and sub-types in which additional kinase targets or off-targets of inhibitors were overexpressed for potential drug repurposing. In addition, the expression of these kinases in normal human tissues were also profiled to predict toxicity following drug repositioning. Taken together, the study offers opportunities for cancer treatment in a kinase-specific manner.


Subject(s)
Drug Repositioning , Neoplasms , Humans , Neoplasms/drug therapy , Protein Kinase Inhibitors/toxicity
8.
Tumori ; 109(2): 186-196, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35311404

ABSTRACT

BACKGROUND: Anlotinib, a multitargeted tyrosine kinase inhibitor, has been shown to have encouraging activity against many tumors, but its cardiovascular toxicity has not been investigated specifically. We reviewed anlotinib-associated cardiovascular adverse events in patients and explored its cardiotoxicity in vitro. METHODS: We retrospectively reviewed all cardiovascular events in 62 patients with unresectable tumors who had taken anlotinib and mainly examined anlotinib's effects on left ventricular ejection fraction (LVEF) and blood pressure. Besides, we investigated its cardiotoxicity in Neonatal Rat Ventricular Myocytes (NRVMs). RESULTS: All-grade hypertension was seen in 60 patients (97%), and 25 individuals (40%) developed grade 3 hypertension. Significant univariate associations for predictors of post-treatment hypertension were age (P<0.001), BMI (P=0.003), ECOG PS(P<0.001), diabetes mellitus (P=0.035), dose of anlotinib (P=0.025). Multivariate analysis suggested that age [odds ratio (OR) 1.079, 95% confidence interval (CI): 1.029-1.130, P= 0.001] and BMI [OR 3.448, 95% CI: 1.410-8.433, P= 0.007] were the only significant independent predictors. No grade 3/4 left ventricular systolic dysfunction was reported. One patient (2%) had acute myocardial infarction, leading to cardiac death. In vitro, western blotting results showed that the levels of ANP, BNP, c-Myc and Cleaved Caspase3 were notably increased and cardiomyocyte apoptosis was strikingly increased in anlotinib group, as detected by TUNEL staining and Annexin V-FITC/PI flow cytometry. CONCLUSIONS: Our study results showed that anlotinib could induce rat cardiomyocytes apoptosis. Nonetheless, anlotinib-associated cardiovascular toxicity was acceptable and manageable for patients with unresectable tumors.


Subject(s)
Hypertension , Neoplasms , Quinolines , Humans , Rats , Animals , Cardiotoxicity/etiology , Cardiotoxicity/drug therapy , Stroke Volume , Retrospective Studies , Ventricular Function, Left , Protein Kinase Inhibitors/toxicity , Quinolines/toxicity , Neoplasms/drug therapy , Hypertension/chemically induced , Hypertension/drug therapy
9.
Lung Cancer ; 173: 116-123, 2022 11.
Article in English | MEDLINE | ID: mdl-36198244

ABSTRACT

CONTEXTE: The Epidermal Growth Factor Receptor (EGFR) is mutated in 10-15% of patients with lung adenocarcinoma. At metastatic stage EGFR tyrosine kinase inhibitors (TKIs) are used front line for patients harboring targetable mutations. Novel anti-EGFR therapies are being developed. Amivantamab is a bispecific anti-EGFR and anti-MET antibody with expected skin toxicities. OBJECTIVE: We developed here guidelines for prevention and treatment of cutaneous toxicities under amivantamab according to our experience at Institut Curie. MATERIEL & METHOD: The first patients with metastatic lung cancer harboring EGFR Exon20ins mutation, included in the phase 1 CHRYSALIS trial and cured at Institute Curie from November 1st 2019 until December 31st 2021 were selected for this work. Retrospectively, all cutaneous adverse events were registered and classified according to the CTCAE 6.0 classification, and actions we implemented to minimize and treat these adverse events were collected. We then developed guidelines based on these datas. RESULTS: A total of seven patients started amivantamab as monotherapy. The two most frequent dermatological adverse events were: acneiform rash and paronychia (100 % of patients). Other adverse events presented by the patients were reported: modification of hair growth with hypertrichosis in 50 % of men (n = 1/2) and hirsutism in 80 % of women (n = 4/5); skin abrasion of the scalp in 71 % (n = 5/7); and skin fissure in 57 % (n = 4/7). We recommend first a rigorous inspection of the skin and teguments to determine the risk rate to have dryer skin under treatment; second a prevention of paronychia/acneiform rash/and skin fissures with prophylactic tetracycline, skin moisturizing, and hygienic measures starting at least 14 days before treatment initiation; third a particular attention to the psychological impact of skin toxicities with access to psychological support. CONCLUSION: We propose here guidelines for the management of dermatological toxicities under amivantamab with a multidisciplinary approach for the proactive management of cutaneous toxicities with a focus on preventive actions.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Protein Kinase Inhibitors , Skin Diseases , Female , Humans , Male , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Exanthema/chemically induced , Exanthema/prevention & control , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Paronychia/chemically induced , Paronychia/prevention & control , Protein Kinase Inhibitors/toxicity , Skin Diseases/chemically induced , Skin Diseases/prevention & control
10.
Blood Cancer J ; 12(8): 123, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35999205

ABSTRACT

Targeted therapies against phosphatidylinositol 3-kinase (PI3K), Bruton's tyrosine kinase (BTK), and B-cell lymphoma-2 (BCL-2) are approved for chronic lymphocytic leukemia (CLL). Since approval of the first-in-class drugs, next-generation agents have become available and are continuously under development. While these therapies act on well-characterized molecular targets, this knowledge is only to some extent taken into consideration when determining their dose in phase I trials. For example, BTK occupancy has been assessed in dose-finding studies of various BTK inhibitors, but the minimum doses that result in full BTK occupancy were not determined. Although targeted agents have a different dose-response relationship than cytotoxic agents, which are more effective near the maximum tolerated dose, the traditional 3 + 3 toxicity-driven trial design remains heavily used in the era of targeted therapies. If pharmacodynamic biomarkers were more stringently used to guide dose selection, the recommended phase II dose would likely be lower as compared to the toxicity-driven selection. Reduced drug doses may lower toxicity, which in some cases is severe for these agents, and are supported by retrospective studies demonstrating non-inferior outcomes for patients with clinically indicated dose reductions. Here, we review strategies that were used for dose selection in phase I studies of currently approved and select investigational targeted therapies in CLL, and discuss how our initial clinical experience with targeted therapies have pointed to dose reductions, intermittent dosing, and drug combinations as strategies to overcome treatment intolerance and resistance.


Subject(s)
Antineoplastic Agents , Leukemia, Lymphocytic, Chronic, B-Cell , Protein Kinase Inhibitors , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/toxicity , Clinical Trials as Topic , Dose-Response Relationship, Drug , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Phosphoinositide-3 Kinase Inhibitors/toxicity , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Retrospective Studies
11.
Pharmacol Ther ; 237: 108256, 2022 09.
Article in English | MEDLINE | ID: mdl-35901905

ABSTRACT

Small molecule tyrosine kinase inhibitors (TKIs) are widely used as anticancer drugs approved by U.S. FDA. However, the toxicities of TKIs to multiple organs have greatly limited their clinical applications. The metabolism of TKIs generates several potentially toxic metabolites in vivo, that can disturb the endogenous metabolism as well as cellular function, leading to organ damage. Therefore, it is essential to identify the toxic metabolites and elucidate the underlying mechanism of TKI-induced toxicity. Metabolomics is a powerful tool for the identification of the xenobiotic metabolites and metabolic derangement associated with xenobiotic exposure, that is helpful to understand the toxicity of TKIs. The study using metabolomics approach has revealed that the reactive metabolites/intermediates (e.g., N-oxide metabolite, primary amine metabolite, 1,4-benzoquinone intermediate) and adducts with glutathione, cysteine and mercapturic acid can be derived from TKIs. Fourteen metabolic pathways could be affected following the TKI treatment, including lipid metabolism, bile acid metabolism, and gut microbiota-related pathway. Modulation of xenobiotic receptor signaling, inhibition of xenobiotic metabolism, and supplementation of endogenous metabolites are potential strategies to protect against TKI-induced toxicity. In this review, studies on the metabolism of TKIs and the alterations of endogenous metabolism are discussed, and the potential preventions against TKI-induced toxicity are summarized.


Subject(s)
Antineoplastic Agents , Protein Kinase Inhibitors , Antineoplastic Agents/toxicity , Humans , Protein Kinase Inhibitors/toxicity , Xenobiotics/toxicity
12.
Toxicol Appl Pharmacol ; 450: 116162, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35830948

ABSTRACT

Pathological angiogenesis is fundamental to progression of cancerous tumors and blinding eye diseases. Anti-angiogenic receptor tyrosine kinase inhibitors (TKIs) are in broad use for the treatment of these diseases. With more and more TKIs available, it is a challenge to make an optimal choice. It remains unclear whether TKIs demonstrate similar anti-angiogenesis activities in different tissues. Many TKIs have shown varying degrees of toxic effects that should also be considered in clinical use. This study investigates the anti-angiogenic effects of 13 FDA-approved TKIs on the intersegmental vessels (ISVs), subintestinal vessels (SIVs) and retinal vasculature in zebrafish embryos. The results show that vascular endothelial growth factor receptor TKIs (VEGFR-TKIs) exhibit anti-angiogenic abilities similarly on ISVs and SIVs, and their efficacy is consistent with their IC50 values against VEGFR2. In addition, VEGFR-TKIs selectively induces the apoptosis of endothelial cells in immature vessels. Among all TKIs tested, axitinib demonstrates a strong inhibition on retinal neovascularization at a low dose that do not strongly affect ISVs and SIVs, supporting its potential application for retinal diseases. Zebrafish embryos demonstrate cardiotoxicity after VEGFR-TKIs treatment, and ponatinib and sorafenib show a narrow therapeutic window, suggesting that these two drugs may need to be dosed more carefully in patients. We propose that zebrafish is an ideal model for studying in vivo antiangiogenic efficacy and cardiotoxicity of TKIs.


Subject(s)
Neoplasms , Zebrafish , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/toxicity , Animals , Cardiotoxicity/drug therapy , Endothelial Cells/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/toxicity , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/metabolism
13.
Toxicol Lett ; 365: 11-23, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35680041

ABSTRACT

Cardiotoxicity by tyrosine kinase inhibitors remains an important concern. Nilotinib and vandetanib clinically carry high proarrhythmic risk and the exact mechanism underlying arrhythmogenesis is not fully understood. In this study, we investigated the effects of nilotinib and vandetanib on the abundance of human ether-á-go-go-related gene (hERG) K+ channel and assessed the potential role of acute hERG blockage versus chronic effects in arrhythmogenesis. We found that both nilotinib and vandetanib prolonged the field potential duration reflecting the repolarisation process and induced cellrythmias of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in a time-and concentration-dependent manner after, after chronic exposure. Patch-clamp recordings revealed significant reductions of hERG current densities by nilotinib or vandetanib after chronic incubation with hERG-HEK293 cells in addition to the acute inhibition. Western blot analysis showed that nilotinib and vandetanib decreased mature hERG protein (155-kDa) expression, in a greater extent than that of the immature form (135-kDa). A serum and glucocorticoid kinase 1 (SGK1) activator, C4-ceramide, prevented the nilotinib-and vandetanib-induced hERG protein downregulation and thus the incidence of cellrrhythmias. Taken together, our data demonstrated that the downregulation of hERG channel abundance on the cellular membrane predominantly contributed to the proarrhythmic effect of nilotinib and vandetanib.


Subject(s)
Ether-A-Go-Go Potassium Channels , Induced Pluripotent Stem Cells , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/metabolism , Down-Regulation , ERG1 Potassium Channel/metabolism , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Myocytes, Cardiac , Piperidines , Protein Kinase Inhibitors/toxicity , Pyrimidines , Quinazolines
14.
J Med Chem ; 65(4): 3229-3248, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35138851

ABSTRACT

Herein, we report two promising compounds 30 and 36 possessing nanomolar FLT3 inhibitory activities (IC50 = 1.5-7.2 nM), high selectivity over c-KIT (>1000-fold), and excellent anti-AML activity (MV4-11 IC50 = 0.8-3.2 nM). Furthermore, these two compounds efficiently inhibited the growth of multiple mutant BaF3 cells expressing FLT3-ITD, FLT3-D835V/F, FLT3-F691L, FLT3-ITD-F691L, and FLT3-ITD-D835Y. Oral administration of 30 and 36 at 6 mg/kg/d could significantly suppress tumor growth in the MV4-11 cell-inoculated xenograft model, exhibiting tumor growth inhibitory rates of 83.5% and 95.1%, respectively. Importantly, 36 could prolong the mouse survival time in the FLT3-ITD-TKD dual mutation syngeneic mouse model (BaF3-FLT3-ITD-D835Y) at a dose of 6 mg/kg p.o. bid/4W. No clear myelosuppression was observed in the treated group of 36 in the MPO strain of zebrafish, even at 10 µM. In summary, our data demonstrated that 36 may represent a promising candidate for the treatment of FLT3 mutant AML.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Cell Line, Tumor , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Dose-Response Relationship, Drug , Humans , Leukemia, Myeloid, Acute/drug therapy , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/toxicity , Pyrimidines/toxicity , Signal Transduction/drug effects , Substrate Specificity , Xenograft Model Antitumor Assays , Zebrafish
15.
J Med Chem ; 65(4): 3420-3433, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35114084

ABSTRACT

Senexins are potent and selective quinazoline inhibitors of CDK8/19 Mediator kinases. To improve their potency and metabolic stability, quinoline-based derivatives were designed through a structure-guided strategy based on the simulated drug-target docking model of Senexin A and Senexin B. A library of quinoline-Senexin derivatives was synthesized to explore the structure-activity relationship (SAR). An optimized compound 20a (Senexin C) exhibits potent CDK8/19 inhibitory activity with high selectivity. Senexin C is more metabolically stable and provides a more sustained inhibition of CDK8/19-dependent cellular gene expression when compared with the prototype inhibitor Senexin B. In vivo pharmacokinetic (PK) and pharmacodynamic (PD) evaluation using a novel tumor-based PD assay showed good oral bioavailability of Senexin C with a strong tumor-enrichment PK profile and tumor-PD marker responses. Senexin C inhibits MV4-11 leukemia growth in a systemic in vivo model with good tolerability.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Colonic Neoplasms/drug therapy , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/therapeutic use , Biological Availability , Cell Line, Tumor , Humans , Leukemia/drug therapy , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/toxicity , Quinolines , Structure-Activity Relationship , Substrate Specificity , Xenograft Model Antitumor Assays
16.
Biochem Pharmacol ; 197: 114914, 2022 03.
Article in English | MEDLINE | ID: mdl-35041812

ABSTRACT

Tyrosine kinase inhibitors (TKIs) are molecular-targeted anticancer drugs. Their benefits are limited by dermal toxicities, including hand-foot skin reaction (HFSR), which is commonly found in skin areas subjected to friction. The present study aimed to explain the incidence of HFSR in patients treated with TKIs by focusing on keratinocyte toxicity and inhibition of vascular endothelial growth factor receptor (VEGFR), which plays an essential role in angiogenesis. Mice with gene knockout for the immunosuppressive cytokine interleukin-10 exhibited HFSR-like phenotypes, such as cytotoxicity in keratinocytes and increased number and size of blood vessels after repeated doses of regorafenib, sorafenib, and pazopanib, all of which cause high incidence of HFSR, in combination with tape-stripping mimicking skin damage at the friction site. Comprehensive examination of the direct cytotoxic effects of 21 TKIs on primary cultured human keratinocytes revealed that 18 of them reduced the cell viability dose-dependently. Importantly, the ratio of the trough concentration in patients (Ctrough) to the LC50 values of cell viability reduction was higher than unity for four HFSR-inducing TKIs, suggesting that these TKIs cause keratinocyte toxicity at clinically relevant concentrations. In addition, eight HFSR-inducing TKIs caused inhibition of VEGFR-2 kinase activity, which was validated by their ratios of Ctrough to the obtained IC50,VEGFR-2 of more than unity. All 12 TKIs with no reported incidence of HFSR exhibited less than unity values for both Ctrough/LC50,keratinocytes and Ctrough/IC50,VEGFR-2. These results suggested that a combination of keratinocyte toxicity and VEGFR-2 inhibition may explain the incidence of HFSR upon TKI usage in humans.


Subject(s)
Exanthema/chemically induced , Keratinocytes/drug effects , Protein Kinase Inhibitors/toxicity , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Animals, Newborn , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Exanthema/metabolism , Exanthema/pathology , Foot/pathology , Hand/pathology , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Phenylurea Compounds/toxicity , Pyridines/toxicity , Sorafenib/toxicity , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
J Med Chem ; 65(3): 1867-1882, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34985276

ABSTRACT

Hybrid compounds containing structural fragments of the Rho kinase inhibitor fasudil and the NRF2 inducers caffeic and ferulic acids were designed with the aid of docking and molecular mechanics studies. Following the synthesis of the compounds using a peptide-coupling methodology, they were characterized for their ROCK2 inhibition, radical scavenging, effects on cell viability (MTT assay), and NRF2 induction (luciferase assay). One of the compounds (1d) was selected in view of its good multitarget profile and good tolerability. It was able to induce the NRF2 signature, promoting the expression of the antioxidant response enzymes HO-1 and NQO1, via a KEAP1-dependent mechanism. Analysis of mRNA and protein levels of the NRF2 pathway showed that 1d induced the NRF2 signature in control and SOD1-ALS lymphoblasts but not in sALS, where it was already increased in the basal state. These results show the therapeutic potential of this compound, especially for ALS patients with a SOD1 mutation.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Amyotrophic Lateral Sclerosis/drug therapy , Coumaric Acids/therapeutic use , Free Radical Scavengers/therapeutic use , Protein Kinase Inhibitors/therapeutic use , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/chemical synthesis , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/toxicity , Aged , Cell Line, Tumor , Cell Survival/drug effects , Coumaric Acids/chemical synthesis , Coumaric Acids/toxicity , Female , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/toxicity , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Lymphocytes/drug effects , Male , Middle Aged , NF-E2-Related Factor 2/agonists , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/toxicity , rho-Associated Kinases/antagonists & inhibitors
18.
Toxicol Appl Pharmacol ; 435: 115852, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34973290

ABSTRACT

Imatinib Mesylate (IMA) has been widely used to treat with chronic myeloid leukemia (CML). However, cardiotoxicity associated with IMA is included among the therapeutic strategies. The present study was aimed to discover whether ferroptosis, a programmed iron-dependent cell death, is involved in IMA-induced cardiotoxicity. In vivo, mouse model was established after treated with 25 mg/kg, 50 mg/kg and 100 mg/kg IMA. Serum CK, LDH, AST activities were determined. Cardiac tissues were examined by H&E and Oil Red O staining. MDA was measured to assess production of lipid peroxide. Tissue iron and GSH content were measured. In vitro, cell viability, mitochondria membrane potential, generation of reactive oxygen species (ROS) and cellular iron levels were performed to explore the mechanism of IMA. The in vivo results revealed that IMA treatment significantly increased serum CK, LDH and AST. H&E staining showed that IMA caused cardiac structural injuries. The dose-dependent decrease of GSH and increase of tissue iron and MDA were observed in IMA-treated groups. Oil Red O staining suggested obvious cardiac lipid accumulation after treated with IMA. In H9c2 cardiomyocytes, IMA significantly inhibited cell proliferation in a dose-dependent manner. Mitochondria membrane potential assay showed that IMA destroyed the mitochondrial function. Additionally, IMA increased the cellular ROS and iron levels. Furthermore, IMA down-regulated the expression of Nrf2 and up-regulated the expression of P53 and TfR. These results provided compelling evidence that ferroptosis participates in IMA-induced cardiotoxicity. Ferroptosis could be regarded as a target to protect against cardiotoxicity in IMA-exposed patients.


Subject(s)
Antineoplastic Agents/toxicity , Cardiotoxicity , Ferroptosis/drug effects , Imatinib Mesylate/toxicity , NF-E2-Related Factor 2/biosynthesis , Protein Kinase Inhibitors/toxicity , Signal Transduction/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Glutathione/metabolism , Iron/metabolism , Lipid Metabolism/drug effects , Lipid Peroxides/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism
19.
Arch Toxicol ; 96(3): 845-857, 2022 03.
Article in English | MEDLINE | ID: mdl-35098321

ABSTRACT

Tyrosine kinase inhibitors (TKIs), which have been developed and approved for cancer treatment in the last few years, are involved in synaptic plasticity of learning and memory. Epigenetic modifications also play crucial roles in the process of learning and memory, but its relationship with TKI-induced learning and memory impairment has not been investigated. We hypothesized that LPM4870108, an effective anti-cancer Trk inhibitor, might affect the learning and memory via epigenetic modifications. In this study, rats were orally administered with LPM4870108 (0, 1.25, 2.5, or 5.0 mg/kg) twice daily for 28 days, after which animals were subjected to a Morris water maze test. LPM4870108 exposure caused learning and memory impairments in this test in a dose-dependent manner and reduced the spine densities. Whole-genome transcriptomic analysis revealed significant differences in the patterns of hippocampal gene expression in LPM4870108-treated rats. These transcriptomic data were combined with next-generation bisulfite sequencing analysis, after which RT-PCR and pyrosequencing were conducted, revealing epigenetic alterations associated with genes (Snx8, Fgfr1, Dusp4, Vav2, and Satb2) known to regulate learning and memory. Increased mRNA and protein expression levels of hippocampal Dnmt1 and Dnmt3a were also observed in these rats. Overall, these data suggest that gene-specific alterations in patterns of DNA methylation can potentially contribute to the incidence of learning and memory deficits associated with exposure to LPM4870108.


Subject(s)
DNA Methylation , Maze Learning , Memory Disorders , Protein Kinase Inhibitors , Animals , Female , Male , Rats , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Maze Learning/drug effects , Memory Disorders/chemically induced , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/toxicity , Rats, Sprague-Dawley , Transcriptome
20.
Biochim Biophys Acta Mol Cell Res ; 1869(1): 119168, 2022 01.
Article in English | MEDLINE | ID: mdl-34728235

ABSTRACT

The induction of DNA damage together with the interference with DNA repair represents a promising strategy in cancer treatment. Here we show that the PARP-1/2/3 inhibitor AZD2461 in combination with the CHK1 inhibitor UCN-01 altered the DNA damage response and reduced cell proliferation in PEL cells, an aggressive B cell lymphoma highly resistant to chemotherapies. AZD2461/UCN-01 combination activated p53/p21 and downregulated c-Myc in these cells, leading to a reduced expression level of RAD51, molecule involved in DNA repair. The effect of AZD2461/UCN-01 on c-Myc and p53/p21 was inter-dependent and, besides impairing cell proliferation, contributed to the activation of the replicative cycle of KSHV, carried in a latent state in PEL cells. Finally, we found that the pharmacological or genetic inhibition of p21 counteracted the viral lytic cycle activation and further reduced PEL cell proliferation, suggesting that it could induce a double beneficial effect in this setting. This study unveils that, therapeutic approaches, based on the induction of DNA damage and the reduction of DNA repair, could be used to successfully treat this malignant lymphoma.


Subject(s)
Cell Proliferation , DNA Damage , Lymphoma, Primary Effusion/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism , Virus Replication , Cell Line , Cells, Cultured , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/metabolism , Herpesvirus 8, Human/physiology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , Phthalazines/pharmacology , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/toxicity , Protein Kinase Inhibitors/toxicity , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...