Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61.611
Filter
1.
Chem Pharm Bull (Tokyo) ; 72(9): 776-780, 2024.
Article in English | MEDLINE | ID: mdl-39218701

ABSTRACT

Protein kinase CK2 type α (CK2α) inhibitors are expected to be a new anticancer drug and a treatment for nephritis. Virtual screening for CK2α inhibitors has been conducted and active compounds with various scaffolds have been obtained. Research on compound optimization is currently in progress for some of them with the aim of improving their activity. This process involves the combination of various computational chemistry methods and crystal analyses. In this review, case studies of structure-based compound designs that have efficiently improved the activity of screening hit compounds, including compounds with a thiadiazole ring and a purine scaffold, are introduced.


Subject(s)
Casein Kinase II , Drug Design , Protein Kinase Inhibitors , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Casein Kinase II/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Humans , Structure-Activity Relationship , Molecular Structure , Crystallography, X-Ray , Drug Evaluation, Preclinical , Computational Chemistry
2.
Sci Rep ; 14(1): 20302, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39218919

ABSTRACT

Secondary BRAF variations have been identified as a mechanism of resistance to tyrosine kinase inhibitors (TKIs) in patients with driver gene-positive NSCLC. Nevertheless, there is still a lack of consensus regarding the characteristics and subsequent treatment strategies for these patients. We retrospectively reviewed the medical records of patients with driver gene-positive NSCLC who received TKIs therapy at Zhejiang Cancer Hospital between May 2016 and December 2023. The clinical and genetic characteristics of these patients were assessed, along with the impact of various treatment strategies on survival. This study enrolled 27 patients with advanced NSCLC, in whom BRAF variations occurred at a median time of 28 months after the initiation of targeted therapy. The multivariate accelerated failure time (AFT) model revealed that, compared to chemotherapy-based regimens group, the combined targeted therapy group (p < 0.001) and the combined local treatment group for oligo-progression (p < 0.001) significantly extended patient survival. In contrast, continuing the original signaling pathway's targeted monotherapy was associated with shorter survival (p = 0.034). The median global OS for each treatment group was as follows: chemotherapy-based regimens group, 45 months; combined targeted therapy group, 59 months; combined local treatment group for patients with oligo-progression, 46 months; and targeted monotherapy group, 36 months. Study results indicate that the combination targeted therapy group (including TKIs, BRAF inhibitors, and/or MEK inhibitors) and the localized treatment group are more effective than traditional chemotherapy-based regimens in improving survival. Additionally, continuing targeted monotherapy along the original signaling pathway proves less effective than chemotherapy-based regimens.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug Resistance, Neoplasm , Lung Neoplasms , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Female , Male , Middle Aged , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Aged , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Molecular Targeted Therapy/methods , Adult , Mutation
3.
Mol Med Rep ; 30(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39219269

ABSTRACT

Tyrosine kinase inhibitors (TKIs) offer targeted therapy for cancers but can cause severe cardiotoxicities. Determining their dose­dependent impact on cardiac function is required to optimize therapy and minimize adverse effects. The dose­dependent cardiotoxic effects of two TKIs, imatinib and ponatinib, were assessed in vitro using H9c2 cardiomyoblasts and in vivo using zebrafish embryos. In vitro, H9c2 cardiomyocyte viability, apoptosis, size, and surface area were evaluated to assess the impact on cellular health. In vivo, zebrafish embryos were analyzed for heart rate, blood flow velocity, and morphological malformations to determine functional and structural changes. Additionally, reverse transcription­quantitative PCR (RT­qPCR) was employed to measure the gene expression of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), established markers of cardiac injury. This comprehensive approach, utilizing both in vitro and in vivo models alongside functional and molecular analyses, provides a robust assessment of the potential cardiotoxic effects. TKI exposure decreased viability and surface area in H9c2 cells in a dose­dependent manner. Similarly, zebrafish embryos exposed to TKIs exhibited dose­dependent heart malformation. Both TKIs upregulated ANP and BNP expression, indicating heart injury. The present study demonstrated dose­dependent cardiotoxic effects of imatinib and ponatinib in H9c2 cells and zebrafish models. These findings emphasize the importance of tailoring TKI dosage to minimize cardiac risks while maintaining therapeutic efficacy. Future research should explore the underlying mechanisms and potential mitigation strategies of TKI­induced cardiotoxicities.


Subject(s)
Cardiotoxicity , Imatinib Mesylate , Imidazoles , Myocytes, Cardiac , Pyridazines , Zebrafish , Animals , Zebrafish/embryology , Imidazoles/toxicity , Pyridazines/adverse effects , Pyridazines/pharmacology , Pyridazines/toxicity , Imatinib Mesylate/toxicity , Imatinib Mesylate/adverse effects , Imatinib Mesylate/pharmacology , Cardiotoxicity/etiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/toxicity , Protein Kinase Inhibitors/pharmacology , Cell Line , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, Brain/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Cell Survival/drug effects , Apoptosis/drug effects , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Rats
4.
Arch Pharm (Weinheim) ; 357(9): e2300562, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39219313

ABSTRACT

A novel group of indolyl-1,2,4-triazole-chalcone hybrids was designed, synthesized, and assessed for their anticancer activity. The synthesized compounds exhibited significant antiproliferative activity. Compounds 9a and 9e exhibited significant cancer inhibition with GI50 ranging from 3.69 to 20.40 µM and from 0.29 to >100 µM, respectively. Both compounds displayed a broad spectrum of anticancer activity with selectivity ratios ranging between 0.50-2.78 and 0.25-2.81 at the GI50 level, respectively. The synthesized compounds were also screened for their cytotoxicity by 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazol (MTT) assay and for inhibition of epidermal growth factor receptor (EGFR) and c-MET (mesenchymal-epithelial transition factor). Some of the tested compounds exhibited significant inhibition against EGFR and/or c-MET. Compound 9b showed the highest c-MET inhibition (IC50 = 4.70 nM) compared to foretinib (IC50 = 2.5 nM). Compound 9d showed equipotent activity compared with erlotinib against EGFR (IC50 = 0.052 µM) and displayed significant c-MET inhibition with an IC50 value of 4.90 nM.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Drug Design , Drug Screening Assays, Antitumor , ErbB Receptors , Indoles , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-met , Triazoles , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Structure-Activity Relationship , Cell Proliferation/drug effects , Triazoles/pharmacology , Triazoles/chemistry , Triazoles/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Indoles/pharmacology , Indoles/chemistry , Indoles/chemical synthesis , Dose-Response Relationship, Drug , Molecular Structure , Cell Line, Tumor , Chalcones/pharmacology , Chalcones/chemical synthesis , Chalcones/chemistry , Chalcone/pharmacology , Chalcone/chemistry , Chalcone/chemical synthesis
5.
PeerJ ; 12: e17797, 2024.
Article in English | MEDLINE | ID: mdl-39221276

ABSTRACT

Numerous aspects of cellular signaling are regulated by the kinome-the network of over 500 protein kinases that guides and modulates information transfer throughout the cell. The key role played by both individual kinases and assemblies of kinases organized into functional subnetworks leads to kinome dysregulation driving many diseases, particularly cancer. In the case of pancreatic ductal adenocarcinoma (PDAC), a variety of kinases and associated signaling pathways have been identified for their key role in the establishment of disease as well as its progression. However, the identification of additional relevant therapeutic targets has been slow and is further confounded by interactions between the tumor and the surrounding tumor microenvironment. In this work, we attempt to link the state of the human kinome, or kinotype, with cell viability in treated, patient-derived PDAC tumor and cancer-associated fibroblast cell lines. We applied classification models to independent kinome perturbation and kinase inhibitor cell screen data, and found that the inferred kinotype of a cell has a significant and predictive relationship with cell viability. We further find that models are able to identify a set of kinases whose behavior in response to perturbation drive the majority of viability responses in these cell lines, including the understudied kinases CSNK2A1/3, CAMKK2, and PIP4K2C. We next utilized these models to predict the response of new, clinical kinase inhibitors that were not present in the initial dataset for model devlopment and conducted a validation screen that confirmed the accuracy of the models. These results suggest that characterizing the perturbed state of the human protein kinome provides significant opportunity for better understanding of signaling behavior and downstream cell phenotypes, as well as providing insight into the broader design of potential therapeutic strategies for PDAC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Cell Survival , Pancreatic Neoplasms , Protein Kinases , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/enzymology , Cell Survival/drug effects , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/enzymology , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/enzymology , Protein Kinases/metabolism , Signal Transduction , Tumor Microenvironment , Protein Kinase Inhibitors/pharmacology
6.
Clin Transl Sci ; 17(9): e70010, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39222377

ABSTRACT

Tepotinib is approved for the treatment of patients with non-small-cell lung cancer harboring MET exon 14 skipping alterations. While edema is the most prevalent adverse event (AE) and a known class effect of MET inhibitors including tepotinib, there is still limited understanding about the factors contributing to its occurrence. Herein, we apply machine learning (ML)-based approaches to predict the likelihood of occurrence of edema in patients undergoing tepotinib treatment, and to identify factors influencing its development over time. Data from 612 patients receiving tepotinib in five Phase I/II studies were modeled with two ML algorithms, Random Forest, and Gradient Boosting Trees, to predict edema AE incidence and severity. Probability calibration was applied to give a realistic estimation of the likelihood of edema AE. Best model was tested on follow-up data and on data from clinical studies unused while training. Results showed high performances across all the tested settings, with F1 scores up to 0.961 when retraining the model with the most relevant covariates. The use of ML explainability methods identified serum albumin as the most informative longitudinal covariate, and higher age as associated with higher probabilities of more severe edema. The developed methodological framework enables the use of ML algorithms for analyzing clinical safety data and exploiting longitudinal information through various covariate engineering approaches. Probability calibration ensures the accurate estimation of the likelihood of the AE occurrence, while explainability tools can identify factors contributing to model predictions, hence supporting population and individual patient-level interpretation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Edema , Machine Learning , Humans , Edema/chemically induced , Female , Male , Middle Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Aged , Lung Neoplasms/drug therapy , Clinical Trials, Phase II as Topic , Pyrimidines/adverse effects , Pyrimidines/administration & dosage , Clinical Trials, Phase I as Topic , Adult , Antineoplastic Agents/adverse effects , Protein Kinase Inhibitors/adverse effects , Piperidines , Pyridazines
8.
Front Immunol ; 15: 1393404, 2024.
Article in English | MEDLINE | ID: mdl-39206183

ABSTRACT

Objective: To systematically evaluate the efficacy and safety of anlotinib targeted therapy for the treatment of patients with advanced digestive system neoplasms (DSNs). Methods: Clinical trials were extracted from PubMed, the Cochrane Library, Web of Science, Embase, China National Knowledge Infrastructure (CNKI) and the Wanfang database up to October 2023. Outcome measures, including therapeutic efficacy, quality of life (QOL) and adverse events, were extracted and evaluated. Results: Twenty trials, including 1,613 advanced DSNs patients, were included. The results indicated that, compared with conventional treatment alone, the combination of anlotinib targeted therapy with conventional treatment significantly improved the patients' 6-months overall survival (OS, OR=1.76, CI=1.53 to 2.02, P<0.00001), overall response (ORR, OR=1.76, CI=1.53 to 2.02, P<0.00001) and disease control rate (DCR, OR=1.51, 95% CI=1.25 to 1.84, P<0.0001). Moreover, the group that received the combined therapy had higher rates of hypertension (P<0.00001), proteinuria (P<0.00001), fatigue (P<0.00001), diarrhea (P<0.00001), hypertriglyceridemia (P=0.02), alanine aminotransfease (ALT)increased (P=0.004), aspartate transaminase (AST) increased (P=0.006), anorexia (P<0.00001), weight loss (P=0.002), abdominal pain (P=0.0006), hypothyroidism (P=0.02), prolonged QT interval (P=0.04). Analyses of other adverse events, such as gastrointestinal reaction, leukopenia, and neutropenia, did not reveal significant differences (P>0.05). Conclusion: The combination of anlotinib targeted therapy and conventional treatment is more effective for DSNs treatment than conventional treatment alone. However, this combined treatment could lead to greater rates of hypertension, albuminuria and hand-foot syndrome. Therefore, the benefits and risks should be considered before treatment.


Subject(s)
Digestive System Neoplasms , Indoles , Protein Kinase Inhibitors , Quinolines , Humans , Quinolines/therapeutic use , Quinolines/adverse effects , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/administration & dosage , Indoles/therapeutic use , Indoles/adverse effects , Indoles/administration & dosage , Digestive System Neoplasms/drug therapy , Treatment Outcome , Quality of Life , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use
9.
Nat Commun ; 15(1): 7596, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39217147

ABSTRACT

Machine learning provides efficient ways to map compound-kinase interactions. However, diverse bioactivity data types, including single-dose and multi-dose-response assay results, present challenges. Traditional models utilize only multi-dose data, overlooking information contained in single-dose measurements. Here, we propose a machine learning methodology for compound-kinase activity prediction that leverages both single-dose and dose-response data. We demonstrate that our two-stage approach yields accurate activity predictions and significantly improves model performance compared to training solely on dose-response labels. This superior performance is consistent across five diverse machine learning methods. Using the best performing model, we carried out extensive experimental profiling on a total of 347 selected compound-kinase pairs, achieving a high hit rate of 40% and a negative predictive value of 78%. We show that these rates can be improved further by incorporating model uncertainty estimates into the compound selection process. By integrating multiple activity data types, we demonstrate that our approach holds promise for facilitating the development of training activity datasets in a more efficient and cost-effective way.


Subject(s)
Machine Learning , Humans , Protein Kinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Phosphotransferases/metabolism , Algorithms , Drug Discovery/methods
10.
Nat Commun ; 15(1): 7388, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39191755

ABSTRACT

Functional blockade of the transforming growth factor-beta (TGFß) signalling pathway improves the efficacy of cytotoxic and immunotherapies. Here, we conducted a phase 1b study (ClinicalTrials.gov., NCT03143985) to determine the primary endpoints of safety, tolerability, and maximal tolerated dose (200 mg twice daily) for the orally-available TGFß type I receptor kinase inhibitor vactosertib in combination with pomalidomide in relapsed and/or refractory multiple myeloma (RRMM) patients who had received ≥2 lines of chemoimmunotherapy. Secondary endpoints demonstrated sustained clinical responses, favorable pharmacokinetic parameters and a 6-month progression-free survival of 82%. Vactosertib combined with pomalidomide was well-tolerated at all dose levels and displayed a manageable adverse event profile. Exploratory analysis indicated that vactosertib co-treatment with pomalidomide also reduced TGFß levels in patient bone marrow as well as the level of CD8+ T-cells that expressed the immunoinhibitory marker PD-1. In vitro experiments indicated that vactosertib+pomalidomide co-treatment decreased the viability of MM cell lines and patient tumor cells, and increased CD8+ T-cell cytotoxic activity. Vactosertib is a safe therapeutic that demonstrates tumor-intrinsic activity and can overcome immunosuppressive challenges within the tumor microenvironment to reinvigorate T-cell fitness. Vactosertib offers promise to improve immunotherapeutic responses in heavily-pretreated MM patients refractory to conventional agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Multiple Myeloma , Receptor, Transforming Growth Factor-beta Type I , Thalidomide , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Thalidomide/analogs & derivatives , Thalidomide/administration & dosage , Thalidomide/therapeutic use , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Middle Aged , Female , Male , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Transforming Growth Factor beta/metabolism , Neoplasm Recurrence, Local/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Adult , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Aniline Compounds , Triazoles
11.
Nat Cardiovasc Res ; 3(7): 799-818, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39196173

ABSTRACT

Pulmonary veno-occlusive disease (PVOD) is a rare form of pulmonary hypertension arising from EIF2AK4 gene mutations or mitomycin C (MMC) administration. The lack of effective PVOD therapies is compounded by a limited understanding of the mechanisms driving vascular remodeling in PVOD. Here we show that administration of MMC in rats mediates activation of protein kinase R (PKR) and the integrated stress response (ISR), which leads to the release of the endothelial adhesion molecule vascular endothelial (VE) cadherin (VE-Cad) in complex with RAD51 to the circulation, disruption of endothelial barrier and vascular remodeling. Pharmacological inhibition of PKR or ISR attenuates VE-Cad depletion, elevation of vascular permeability and vascular remodeling instigated by MMC, suggesting potential clinical intervention for PVOD. Finally, the severity of PVOD phenotypes was increased by a heterozygous BMPR2 mutation that truncates the carboxyl tail of the receptor BMPR2, underscoring the role of deregulated bone morphogenetic protein signaling in the development of PVOD.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II , Disease Models, Animal , Phenotype , Pulmonary Veno-Occlusive Disease , Animals , Pulmonary Veno-Occlusive Disease/genetics , Pulmonary Veno-Occlusive Disease/drug therapy , Pulmonary Veno-Occlusive Disease/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Vascular Remodeling/drug effects , Cadherins/genetics , Cadherins/metabolism , Humans , Male , Antigens, CD/metabolism , Antigens, CD/genetics , Rats, Sprague-Dawley , Signal Transduction/drug effects , Mutation , Capillary Permeability/drug effects , Rats , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
12.
Sci Rep ; 14(1): 19999, 2024 08 28.
Article in English | MEDLINE | ID: mdl-39198533

ABSTRACT

DNA-dependent protein kinase catalytic subunit (DNA-PKcs) plays a vital role in DNA damage repair and lymphocyte function, presenting a significant target in cancer and immune diseases. Current DNA-PKcs inhibitors are undergoing Phase I/II trials as adjuncts to radiotherapy and chemotherapy in cancer. Nevertheless, clinical utility is limited by suboptimal bioavailability. This study introduces DNA-PKcs inhibitors designed to enhance bioavailability. We demonstrate that a novel DNA-PKcs inhibitor, DA-143, surpasses NU7441 in aqueous solubility as well as other available inhibitors. In addition, DA-143 displayed an improvement in DNA-PKcs inhibition relative to NU7441 achieving an IC50 of 2.5 nM. Consistent with current inhibitors, inhibition of DNA-PKcs by DA-143 resulted in increased tumor cell sensitivity to DNA-damage from chemotherapy and inhibition of human T cell function. The improved solubility of DA-143 is critical for enhanced efficacy at reduced doses and facilitates more effective evaluation of DNA-PKcs inhibition in both preclinical and clinical development.


Subject(s)
Chromones , DNA-Activated Protein Kinase , Morpholines , Protein Kinase Inhibitors , Solubility , Humans , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Morpholines/chemistry , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Chromones/chemistry , Chromones/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
13.
Sci Rep ; 14(1): 19980, 2024 08 28.
Article in English | MEDLINE | ID: mdl-39198539

ABSTRACT

Most cancer-related deaths are due to the spread of tumour cells throughout the body-a process known as metastasis. While in the vasculature, these cells are referred to as circulating tumour cells (CTCs) and can be found as either single cells or clusters of cells (often including platelets), with the latter having the highest metastatic potential. However, the biology of CTC clusters is poorly understood, and there are no therapies that specifically target them. We previously developed an in vitro model system for CTC clusters and proposed a new extravasation model that involves cluster dissociation, adherence, and single-cell invasion in response to TGF-ß1 released by platelets. Here, we investigated TGF-ß1-induced gene expression changes in this model, focusing on genes for which targeted drugs are available. In addition to the upregulation of the TGF-ß1 signalling pathway, we found that (i) genes in the EGF/EGFR pathway, including those coding for EGFR and several EGFR ligands, were also induced, and (ii) Erlotinib and Osimertinib, two therapeutic EGFR/tyrosine kinase inhibitors, decreased the TGF-ß1-induced adherence and invasion of the CTC cluster-like line despite the line expressing wild-type EGFR. Overall, we suggest that EGFR inhibitors have the potential to decrease the dispersal of CTC clusters that respond to TGF-ß1 and overexpress EGFR (irrespective of its status) and thus could improve patient survival.


Subject(s)
ErbB Receptors , Neoplastic Cells, Circulating , Transforming Growth Factor beta1 , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/pathology , Transforming Growth Factor beta1/metabolism , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Erlotinib Hydrochloride/pharmacology , Signal Transduction/drug effects , Acrylamides , Aniline Compounds , Indoles , Pyrimidines
14.
Int J Mol Sci ; 25(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39201276

ABSTRACT

Checkpoint kinases 1 and 2 (CHK1 and CHK2) are enzymes that are involved in the control of DNA damage. At the present time, these enzymes are some of the most important targets in the fight against cancer since their inhibition produces cytotoxic effects in carcinogenic cells. This paper proposes the use of spirostans (Sp), natural compounds, as possible inhibitors of the enzymes CHK1 and CHK2 from an in silico analysis of a database of 155 molecules (S5). Bioinformatics studies of molecular docking were able to discriminate between 13 possible CHK1 inhibitors, 13 CHK2 inhibitors and 1 dual inhibitor for both enzymes. The administration, distribution, metabolism, excretion and toxicity (ADMETx) studies allowed a prediction of the distribution and metabolism of the potential inhibitors in the body, as well as determining the excretion routes and the appropriate administration route. The best inhibition candidates were discriminated by comparing the enzyme-substrate interactions from 2D diagrams and molecular docking. Specific inhibition candidates were obtained, in addition to studying the dual inhibitor candidate and observing their stability in dynamic molecular studies. In addition, Highest Occupied Molecular Orbital-Lowest Unoccupied Molecular Orbital (HOMO-LUMO) interactions were analyzed to study the stability of interactions between the selected enzymes and spirostans resulting in the predominant gaps from HOMOCHKs to LUMOSp (Highest Occupied Molecular Orbital of CHKs-Lowest Unoccupied Molecular Orbital of spirostan). In brief, this study presents the selection inhibitors of CHK1 and CHK2 as a potential treatment for cancer using a combination of molecular docking and dynamics, ADMETx predictons, and HOMO-LUMO calculation for selection.


Subject(s)
Checkpoint Kinase 1 , Checkpoint Kinase 2 , Molecular Docking Simulation , Protein Kinase Inhibitors , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/metabolism , Checkpoint Kinase 2/chemistry , Humans , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Computer Simulation , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Molecular Dynamics Simulation
15.
Genes (Basel) ; 15(8)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39202410

ABSTRACT

Histiocytic sarcoma (HS) is a rare and highly aggressive cancer in humans and dogs. In dogs, it has a high prevalence in certain breeds, such as Bernese mountain dogs (BMDs) and flat-coated retrievers. Hemophagocytic histiocytic sarcoma (HHS) is a unique form of HS that presents with erythrophagocytosis. Due to its rareness, the study of HHS is very limited, and mutations in canine HHS patients have not been studied to date. In previous work, our research group identified two major PTPN11/SHP2 driver mutations, E76K and G503V, in HS in dogs. Here, we report additional mutations located in exon 3 of PTPN11/SHP2 in both HS and HHS cases, further supporting that this area is a mutational hotspot in dogs and that mutations in tumors and liquid biopsies should be evaluated utilizing comprehensive methods such as Sanger and NextGen sequencing. The overall prevalence of PTPN11/SHP2 mutations was 55.8% in HS and 46.2% in HHS. In addition, we identified mutations in KRAS, in about 3% of HS and 4% of HHS cases. These findings point to the shared molecular pathology of activation of the MAPK pathway in HS and HHS cases. We evaluated the efficacy of the highly specific MEK inhibitor, cobimetinib, in canine HS and HHS cell lines. We found that the IC50 values ranged from 74 to 372 nM, which are within the achievable and tolerable ranges for cobimetinib. This finding positions cobimetinib as a promising potential candidate for future canine clinical trials and enhances our understanding of the molecular defects in these challenging cancers.


Subject(s)
Azetidines , Histiocytic Sarcoma , Mutation , Piperidines , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Proto-Oncogene Proteins p21(ras) , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Piperidines/pharmacology , Dogs , Animals , Histiocytic Sarcoma/drug therapy , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/veterinary , Histiocytic Sarcoma/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Azetidines/pharmacology , Dog Diseases/genetics , Dog Diseases/drug therapy , Dog Diseases/pathology , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor
16.
Eur J Drug Metab Pharmacokinet ; 49(5): 631-644, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39105991

ABSTRACT

BACKGROUND AND OBJECTIVES: Numerous clinical concerns have been expressed regarding the potential worsening of cyclin-dependent kinase 4/6 inhibitor effects in breast cancer patients because of co-administration of proton pump inhibitors. Hence, this study evaluated the effects of proton pump inhibitors on the pharmacokinetics of palbociclib and ribociclib in terms of  cytochrome P450 (CYP) 3A4 and P-glycoprotein involvement. METHODS: The effects of omeprazole and rabeprazole on drug metabolism and efflux of these drugs were investigated using molecular docking, metabolic stability assay in rat liver microsomes, human recombinant CYP3A4 (rCYP3A4) enzymes, and Caco-2 cell monolayers, and in vivo pharmacokinetics with omeprazole and rabeprazole in (5 and 10 mg/kg) 30 min and 7 days before orally dosing palbociclib and ribociclib (10 mg/kg). RESULTS: Omeprazole and rabeprazole inhibited CYP3A4 enzyme activity in rCYP3A4 baculosomes with a 50-60% inhibition at 30 µM. Additionally, both omeprazole and rabeprazole (10 µm) significantly reduced the P-glycoprotein-mediated drug efflux of palbociclib and ribociclib. The 7-day pretreatment of omeprazole at a dose of 10 mg/kg resulted in 24% and 26% reductions in palbociclib's mean maximum plasma concentration) Cmax and area under the plasma concentration-time curve (AUC0-24 h), respectively. Palbociclib's pharmacokinetics were not significantly altered by the pretreatment with rabeprazole; however, ribociclib pharmacokinetics exhibited an 83.94% increase in AUC0-24 h. CONCLUSION: The findings indicate that long-term treatment with therapeutic doses of both omeprazole and rabeprazole can alter the pharmacokinetics of palbociclib and ribociclib. The co-administration of rabeprazole may alter the pharmacokinetics of palbociclib and ribociclib via CYP enzyme and P-glycoprotein inhibition.


Subject(s)
Aminopyridines , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cytochrome P-450 CYP3A , Drug Interactions , Microsomes, Liver , Omeprazole , Piperazines , Proton Pump Inhibitors , Purines , Pyridines , Rabeprazole , Animals , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/pharmacokinetics , Piperazines/pharmacokinetics , Piperazines/pharmacology , Piperazines/administration & dosage , Humans , Purines/pharmacokinetics , Purines/pharmacology , Rats , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/administration & dosage , Rabeprazole/pharmacology , Rabeprazole/administration & dosage , Rabeprazole/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Omeprazole/pharmacology , Omeprazole/pharmacokinetics , Omeprazole/administration & dosage , Male , Caco-2 Cells , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Aminopyridines/administration & dosage , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Rats, Sprague-Dawley , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/administration & dosage , Liver/metabolism , Liver/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
18.
Eur J Endocrinol ; 191(2): 251-261, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39158090

ABSTRACT

OBJECTIVE: Targeted therapy (TT) with BRAF/MEK inhibitors has emerged as a potential treatment in papillary craniopharyngiomas (PCPs). However, standardized data on large cohorts are lacking. Our study aimed to assess real-life efficacy and safety of BRAF/MEK inhibition in patients with PCPs. DESIGN: Retrospective French multicenter study involving BRAF V600E-mutated PCP patients, treated with BRAF/MEK inhibitor combination dabrafenib and trametinib, from April 2019 to July 2023. METHODS: Objective response and clinical and safety outcomes were assessed after 3 months and at the last available follow-up during TT. RESULTS: Sixteen patients (8 females, mean age 50.5 ± 15.75 years), receiving either neoadjuvant therapy (NEO) for non-resectable tumors (n = 6), post-surgical adjuvant therapy (ADJ; n = 8), or palliative therapy (PAL) following failure of multimodal treatment (n = 2), were included.At the last follow-up (mean 7.6 ± 5.3 months), 12 patients showed subtotal response, 3 exhibited partial response, and 1 maintained stable disease. Mean volume reduction was 88.9 ± 4.4%, 73.3 ± 23.4%, and 91.8 ± 4.3% in the NEO, ADJ, and PAL groups, respectively.Targeted therapy resolved headaches in 5/5 patients and visual impairment in 6/9; 2/3 patients had improved neurological symptoms, 1/4 presented weight loss, and 2/14 recovered endocrine function.Targeted therapy was well-tolerated in 62.5% of cases; adverse events led to treatment discontinuation in 5 patients and definitive discontinuation in 3 cases. CONCLUSIONS: In this study, 94% of patients showed partial response or better to TT. Adverse events were acceptable. Further research is needed to establish standardized protocols; however, these results advocate for a NEO approach in invasive PCPs.


Subject(s)
Craniopharyngioma , Oximes , Pituitary Neoplasms , Proto-Oncogene Proteins B-raf , Pyridones , Pyrimidinones , Humans , Female , Male , Middle Aged , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Adult , Retrospective Studies , Craniopharyngioma/drug therapy , Pyridones/therapeutic use , Pyridones/administration & dosage , Pyridones/adverse effects , Aged , Pituitary Neoplasms/drug therapy , Pyrimidinones/therapeutic use , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects , Oximes/therapeutic use , Oximes/administration & dosage , Oximes/adverse effects , Cohort Studies , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Molecular Targeted Therapy/methods , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Imidazoles/adverse effects , Treatment Outcome , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
19.
Med ; 5(8): 856-858, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39127034

ABSTRACT

The standard of care for chronic myeloid leukemia (CML) involves tyrosine kinase inhibitors (TKIs), which suppress tyrosine kinase activity of BCR::ABL1. Hochhaus et al. reported that asciminib, a BCR::ABL1 inhibitor specifically targeting the ABL myristoyl pocket, showed superior efficacy and favorable safety compared with TKIs in the phase 3 ASC4FIRST trial in patients with newly diagnosed chronic-phase CML.1.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Protein Kinase Inhibitors , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/adverse effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/adverse effects , Niacinamide/analogs & derivatives , Pyrazoles
20.
J Clin Lab Anal ; 38(13-14): e25089, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39129486

ABSTRACT

BACKGROUND: Alectinib is a second-generation anaplastic lymphoma kinase (ALK) inhibitor indicated for ALK-mutated non-small-cell lung cancer. Recently, the association between alectinib and red cell morphological abnormalities has been reported in a few case series. This retrospective observational study aims to determine the frequency of occurrence of acanthocytosis in patients taking alectinib and to evaluate the red cell indices, biochemical markers of haemolysis and eosin-5-maleimide (EMA) binding assay results in patients receiving alectinib. METHODS: Patients who were on alectinib and had a complete blood count test performed in Queen Elizabeth Hospital Haematology Laboratory between 1 May 2021 and 31 August 2021 were included in the study. Haematological investigations that had been performed before and after the commencement of alectinib were reviewed. RESULTS: Fifty patients receiving alectinib were evaluated in this analysis. One hundred per cent of patients showed 3+ acanthocytes on the peripheral blood smears. Compared with the test results before starting alectinib, the post-alectinib blood tests showed a significantly lower haemoglobin concentration, red blood cell count and haematocrit; and a significantly higher mean corpuscular haemoglobin, mean corpuscular haemoglobin concentration and red cell distribution width. All the tested patients showed a marked reduction in EMA mean channel fluorescence compared with normal control. CONCLUSION: Our cohort revealed that alectinib caused significant acanthocytosis in all patients. Alectinib was also associated with changes in red cell indices and biochemical markers of haemolysis, compatible with a spherocytic and anisopoikilocytic morphology with haemolysis. Patients on alectinib had reduced EMA binding.


Subject(s)
Carbazoles , Erythrocytes , Piperidines , Humans , Piperidines/therapeutic use , Piperidines/pharmacology , Carbazoles/pharmacology , Male , Female , Middle Aged , Retrospective Studies , Aged , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocyte Indices/drug effects , Adult , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/blood , Lung Neoplasms/pathology , Aged, 80 and over , Hematologic Tests
SELECTION OF CITATIONS
SEARCH DETAIL