Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 105
1.
Int Immunopharmacol ; 133: 112090, 2024 May 30.
Article En | MEDLINE | ID: mdl-38640718

BACKGROUND: Diminished bioavailability of imatinib in leukemic cells contributes to poor clinical response. We examined the impact of genetic polymorphisms of imatinib on the pharmacokinetics and clinical response in 190 patients with chronic myeloid leukaemia (CML). METHODS: Single nucleotide polymorphisms were genotyped using pyrophosphate sequencing. Plasma trough levels of imatinib were measured using liquid chromatography-tandem mass spectrometry. RESULTS: Patients carrying the TT genotype for ABCB1 (rs1045642, rs2032582, and rs1128503), GG genotype for CYP3A5-rs776746 and AA genotype for ABCG2-rs2231142 polymorphisms showed higher concentration of imatinib. Patients with T allele for ABCB1 (rs1045642, rs2032582, and rs1128503), A allele for ABCG2-rs2231142, and G allele for CYP3A5-rs776746 polymorphisms showed better cytogenetic response and molecular response. In multivariate analysis, carriers of the CYP3A5-rs776746 G allele exhibited higher rates of complete cytogenetic response (CCyR) and major molecular response (MMR). Similarly, patients with the T allele of ABCB1-rs1045642 and rs1128503 demonstrated significantly increased CCyR rates. Patients with the A allele of ABCG2-rs2231142 were associated with higher MMR rates. The AA genotype for CYP3A5-rs776746, and the CC genotype for ABCB1-rs104562, and rs1128503 polymorphisms were associated with a higher risk of imatinib failure. Patients with the G allele for CYP3A5-rs776746 exhibited a higher incidence of anemia, and T allele for ABCB1-rs2032582 demonstrated an increased incidence of diarrhea. CONCLUSIONS: Genotyping of ABCB1, ABCG2, and CYP3A5 genes may be considered in the management of patients with CML to tailor therapy and optimize clinical outcomes.


ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Antineoplastic Agents , Cytochrome P-450 CYP3A , Imatinib Mesylate , Neoplasm Proteins , Polymorphism, Single Nucleotide , Humans , Imatinib Mesylate/therapeutic use , Imatinib Mesylate/pharmacokinetics , Male , Female , Middle Aged , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Adult , ATP Binding Cassette Transporter, Subfamily B/genetics , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/blood , Cytochrome P-450 CYP3A/genetics , Neoplasm Proteins/genetics , Genotype , Young Adult , Leukemia, Myeloid, Chronic-Phase/drug therapy , Leukemia, Myeloid, Chronic-Phase/genetics , Adolescent , Treatment Outcome , Aged, 80 and over , Protein Kinase Inhibitors/therapeutic use
2.
Eur J Clin Pharmacol ; 80(7): 1061-1068, 2024 Jul.
Article En | MEDLINE | ID: mdl-38536418

BACKGROUND: Imatinib treatment for certain cancers can lead to elevated creatine kinase (CK) levels, potentially indicating muscle injury, and ongoing research aims to understand the correlation between imatinib levels and creatine kinase to assess its impact on treatment response. METHODS: This single-center observational study involved 76 chronic myeloid leukemia (CML) patients receiving imatinib treatment, focusing on evaluating drug and metabolite levels using liquid chromatography-mass spectrometry (LC-MS-MS) instrumentation. Serum CK and creatine kinase-MB (CK-MB) levels were assessed using Colorimetric kits. RESULTS: CK and CK-MB levels were measured, CK showed a median value of 211.5 IU/l and CK-MB showed a median value of 4.4 IU/l. Comparing low and high CK groups, significant differences were found in peak and trough plasma concentrations of imatinib and its metabolites. Correlations between CK levels and pharmacokinetic parameters were explored, with notable associations identified. Binary logistic regression revealed predictors influencing the therapeutic response to imatinib and categorized expected CK levels into high or low, with peak levels of imatinib emerging as a significant predictor for CK level categorization. CONCLUSION: The study highlights the link between imatinib's pharmacokinetics and elevated CK levels, indicating a possible correlation between specific metabolites and improved treatment response. Individualized monitoring of CK levels and imatinib pharmacokinetics could enhance care for CML patients.


Antineoplastic Agents , Creatine Kinase , Drug Monitoring , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Imatinib Mesylate/pharmacokinetics , Imatinib Mesylate/therapeutic use , Imatinib Mesylate/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Female , Male , Middle Aged , Adult , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/blood , Creatine Kinase/blood , Aged , Drug Monitoring/methods , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/blood , Young Adult , Treatment Outcome , Creatine Kinase, MB Form/blood , Tandem Mass Spectrometry , Aged, 80 and over , Chromatography, Liquid
3.
Ther Drug Monit ; 46(3): 344-350, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38176855

BACKGROUND: Imatinib is the first-line treatment for gastrointestinal stromal tumors; however, the clinical prognosis and adverse reactions of patients vary owing to individualized discrepancies in plasma exposure. METHODS: To determine the safe interval for steady-state plasma trough concentrations (C min ) of imatinib and its active metabolite, N-demethyl imatinib (NDI), 328 plasma samples from 273 patients treated with imatinib were retrospectively analyzed. Imatinib C min and NDI C min were tested, and adverse reactions were recorded. The association between imatinib C min , NDI C min , and serious adverse reactions was evaluated. RESULTS: The C min range of imatinib was 209.5-4950.0 ng/mL, with the mean value and SD of 1491.8 ± 731.4 ng/mL. The C min range of NDI was 80.0-2390.0 ng/mL with the mean value and SD of 610.8 ± 281.5 ng/mL. NDI C min was positively correlated with imatinib C min , whereas the ratio of NDI C min to imatinib C min (NDI C min /imatinib C min ) was negatively correlated with imatinib C min . Univariate logistic regression analysis demonstrated that the treatment objective, daily dose, imatinib C min , NDI C min , and imatinib C min + NDI C min were significantly associated with serious adverse reactions. Multivariate logistic regression analysis showed that NDI C min was an independent risk factor for serious adverse reactions, with a threshold of 665 ng/mL. CONCLUSIONS: NDI C min was an independent risk factor for serious adverse reactions, with a threshold of 665 ng/mL. Monitoring NDI C min was beneficial for the rational application of imatinib and individualized treatment of patients with gastrointestinal stromal tumors.


Antineoplastic Agents , Gastrointestinal Neoplasms , Gastrointestinal Stromal Tumors , Imatinib Mesylate , Humans , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/blood , Imatinib Mesylate/therapeutic use , Imatinib Mesylate/pharmacokinetics , Imatinib Mesylate/adverse effects , Imatinib Mesylate/blood , Retrospective Studies , Male , Female , Middle Aged , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Adult , Gastrointestinal Neoplasms/drug therapy , Aged, 80 and over , Cohort Studies , Drug Monitoring/methods , Young Adult
4.
Pharmacol Res Perspect ; 11(4): e01082, 2023 Aug.
Article En | MEDLINE | ID: mdl-37417254

We aimed to use physiologically based pharmacokinetic (PBPK) modeling and simulation to predict imatinib steady-state plasma exposure in patients with chronic myeloid leukemia (CML) to investigate variability in outcomes. A validated imatinib PBPK model (Simcyp Simulator) was used to predict imatinib AUCss , Css,min and Css,max for patients with CML (n = 68) from a real-world retrospective observational study. Differences in imatinib exposure were evaluated based on clinical outcomes, (a) Early Molecular Response (EMR) achievement and (b) occurrence of grade ≥3 adverse drug reactions (ADRs), using the Kruskal-Wallis rank sum test. Sensitivity analyses explored the influence of patient characteristics and drug interactions on imatinib exposure. Simulated imatinib exposure was significantly higher in patients who achieved EMR compared to patients who did not (geometric mean AUC0-24,ss 51.2 vs. 42.7 µg h mL-1 , p < 0.05; Css,min 1.1 vs. 0.9 µg mL-1 , p < 0.05; Css,max 3.4 vs. 2.8 µg mL-1 , p < 0.05). Patients who experienced grade ≥3 ADRs had a significantly higher simulated imatinib exposure compared to patients who did not (AUC0-24,ss 56.1 vs. 45.9 µg h mL-1 , p < 0.05; Css,min 1.2 vs. 1.0 µg mL-1 , p < 0.05; Css,max 3.7 vs. 3.0 µg mL-1 , p < 0.05). Simulations identified a range of patient (sex, age, weight, abundance of hepatic CYP2C8 and CYP3A4, α1 -acid glycoprotein concentrations, liver and kidney function) and medication-related factors (dose, concomitant CYP2C8 modulators) contributing to the inter-individual variability in imatinib exposure. Relationships between imatinib plasma exposure, EMR achievement and ADRs support the rationale for therapeutic drug monitoring to guide imatinib dosing to achieve optimal outcomes in CML.


Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Imatinib Mesylate/adverse effects , Imatinib Mesylate/pharmacokinetics , Cytochrome P-450 CYP2C8 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Cytochrome P-450 CYP3A
5.
Br J Clin Pharmacol ; 89(3): 1089-1098, 2023 03.
Article En | MEDLINE | ID: mdl-36178950

AIMS: Patients on treatment with oral fixed dose imatinib are frequently under- or overexposed to the drug. We investigated the association between the gene activity score (GAS) of imatinib-metabolizing cytochromes (CYP3A4, CYP3A5, CYP2D6, CYP2C9, CYP2C19, CYP2C8) and imatinib and nor-imatinib exposure. We also investigated the impact of concurrent drug-drug-interactions (DDIs) on the association between GAS and imatinib exposure. METHODS: Serial plasma samples were collected from 33 GIST patients treated with imatinib 400 mg daily within a prospective clinical trial. Imatinib and nor-imatinib Ctrough were quantified by liquid chromatography with tandem mass spectrometry (LC-MS/MS). Genetic polymorphisms with a functional impact on imatinib-metabolizing cytochromes were identified and a GAS was calculated for each gene. A DDI-adjusted GAS was also generated. RESULTS: Imatinib and nor-imatinib Ctrough were measured in 161 plasma samples. CYP2D6 GAS and metabolizer status based on genotype were associated with imatinib and (imatinib + nor-imatinib) Ctrough . CYP2D6 poor and intermediate metabolizers were predicted to have a lower nor-imatinib/imatinib metabolic ratio than normal metabolizers (0.197 and 0.193 vs. 0.247, P = .0205), whereas CYP2C8*3 carriers had a higher ratio than CYP2C8*1/*1 patients (0.263 vs. 0.201, P = .0220). CYP2C9 metabolizer status was inversely related to the metabolic ratio with an effect probably driven by the linkage disequilibrium between CYP2C9*2 and CYP2C8*3. The CYP2D6 DDI-adjusted GAS was still predictive of imatinib exposure. CONCLUSIONS: These findings highlight that CYP2D6 plays a major role in imatinib pharmacokinetics, but other players (i.e., CYP2C8) may influence imatinib exposure. These findings could drive the selection of patients more susceptible to imatinib under- or overexposure who could be candidates for personalized treatment and intensified monitoring strategies.


Cytochrome P-450 CYP2D6 , Gastrointestinal Stromal Tumors , Humans , Cytochrome P-450 CYP2D6/genetics , Imatinib Mesylate/adverse effects , Imatinib Mesylate/pharmacokinetics , Cytochrome P-450 CYP2C8/genetics , Pharmacogenetics , Cytochrome P-450 CYP2C9/genetics , Prospective Studies , Chromatography, Liquid , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Tandem Mass Spectrometry , Cytochromes/genetics , Genotype , Cytochrome P-450 CYP2C19/genetics
6.
Eur J Clin Pharmacol ; 78(8): 1321-1330, 2022 Aug.
Article En | MEDLINE | ID: mdl-35652931

PURPOSE: Imatinib is a substrate of CYP3A4, ABCB1 and ABCG2, and is known to have wide variability in pharmacokinetics (PK). At the same time, a clear relationship between drug levels and response also exists for imatinib in chronic myeloid leukaemia (CML). Therefore, pharmacogenetic-based dosing of imatinib is an attractive proposition. This study aims to characterize the population pharmacokinetics of imatinib in order to identify significant covariates including pharmacogenetic variants. METHODS: Forty-nine patients with CML were enrolled in the study after being on imatinib for at least 4 consecutive weeks. Steady-state pharmacokinetic sampling was performed either in a sparse (4 samples each, n = 44) or intensive manner (9 samples each, n = 5). An additional pharmacogenetic sample was also collected from all patients. Plasma imatinib levels were estimated using a validated HPLC method. Pharmacogenetic variants were identified using the PharmacoScan array platform. Population pharmacokinetic analysis was carried out using NONMEM v7.2. Seven SNPs within CYP3A4, ABCB1 and ABCG2 genes were evaluated for covariate effect on the clearance of imatinib. RESULTS: Imatinib PK was well characterized using a one-compartment model with zero-order absorption. The clearance and volume of distribution were found to be 10.2 L/h and 389 L respectively. Only SNP rs1128503 of the ABCB1 gene had a small but insignificant effect on imatinib clearance, with a 25% reduction in clearance observed in patients carrying the polymorphism. Twenty-three out of forty-nine patients (47%) carried the polymorphic allele, of whom 17 were heterozygous and six were homozygous. CONCLUSION: Our study conclusively proves that genetic polymorphisms in the CYP3A4 and ABC family of transporters do not have any role in the personalized dosing of imatinib in CML.


Antineoplastic Agents , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A/genetics , Humans , Imatinib Mesylate/pharmacokinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Pharmacogenetics , Polymorphism, Single Nucleotide
7.
J Clin Pharmacol ; 62(10): 1285-1296, 2022 10.
Article En | MEDLINE | ID: mdl-35460539

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, which causes coronavirus disease 2019 (COVID-19), manifests as mild respiratory symptoms to severe respiratory failure and is associated with inflammation and other physiological changes. Of note, substantial increases in plasma concentrations of α1 -acid-glycoprotein and interleukin-6 have been observed among patients admitted to the hospital with advanced SARS-CoV-2 infection. A physiologically based pharmacokinetic (PBPK) approach is a useful tool to evaluate and predict disease-related changes on drug pharmacokinetics. A PBPK model of imatinib has previously been developed and verified in healthy people and patients with cancer. In this study, the PBPK model of imatinib was successfully extrapolated to patients with SARS-CoV-2 infection by accounting for disease-related changes in plasma α1 -acid-glycoprotein concentrations and the potential drug interaction between imatinib and dexamethasone. The model demonstrated a good predictive performance in describing total and unbound imatinib concentrations in patients with SARS-CoV-2 infection. PBPK simulations highlight that an equivalent dose of imatinib may lead to substantially higher total drug concentrations in patients with SARS-CoV-2 infection compared to that in patients with cancer, while the unbound concentrations remain comparable between the 2 patient populations. This supports the notion that unbound trough concentration is a better exposure metric for dose adjustment of imatinib in patients with SARS-CoV-2 infection, compared to the corresponding total drug concentration. Potential strategies for refinement and generalization of the PBPK modeling approach in the patient population with SARS-CoV-2 are also provided in this article, which could be used to guide study design and inform dose adjustment in the future.


COVID-19 Drug Treatment , Imatinib Mesylate , Glycoproteins , Humans , Imatinib Mesylate/pharmacokinetics , Imatinib Mesylate/therapeutic use , Models, Biological , SARS-CoV-2
8.
Curr Drug Metab ; 22(10): 802-810, 2021.
Article En | MEDLINE | ID: mdl-34473615

BACKGROUND: Imatinib, sunitinib, and gefitinib are the three most common tyrosine kinase inhibitors (TKIs). However, their quantitative drug-drug interaction potentials In vivo and the relationship between their structure and inhibitory activity remain unknown. OBJECTIVE: This study aimed to investigate the potential drug-drug interaction risk of three TKIs based on CYP3A. METHODS: 6ß-Hydroxylated testosterone formation was selected to probe the CYP3A activity in human liver microsomes. A molecular docking simulation was performed to explore the potential structural alerts. RESULTS: Imatinib exhibited the strongest inhibitory effect towards CYP3A, while the inhibitory potential of gefitinib and sunitinib were comparable to each other but weaker than imatinib. IC50 shift assays demonstrated that the inhibitory potential of all three TKIs was significantly increased after a 30-min preincubation with NADPH. The KI and Kinact values of imatinib, sunitinib, and gefitinib were 3.75 µM and 0.055 min-1, 1.96 µM and 0.037 min-1, and 9.94 µM and 0.031 min-1, respectively. IVIVE results showed that there was a 1.3- to 43.1-fold increase in the AUC of CYP3A-metabolizing drugs in the presence of the TKIs. CONCLUSION: All three TKIs exhibited a typical irreversible inhibitory effect towards CYP3A. The presence of more N-heterocycles and the resulting better binding confirmation of imatinib may have been responsible for its stronger inhibitory effect than sunitinib and gefitinib. Therefore, caution should be taken when CYP3A-metabolizing drugs are co-administrated with imatinib, sunitinib, or gefitinib.


Cytochrome P-450 CYP3A/metabolism , Gefitinib/pharmacokinetics , Hydroxytestosterones/pharmacokinetics , Imatinib Mesylate/pharmacokinetics , Microsomes, Liver/metabolism , Sunitinib/pharmacokinetics , Drug Interactions , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacokinetics , Structure-Activity Relationship
9.
AAPS PharmSciTech ; 22(5): 192, 2021 Jun 28.
Article En | MEDLINE | ID: mdl-34184160

Neurodegenerative diseases like Alzheimer's disease require treatment where it is essential for drug to reach brain. Nose to brain delivery of drugs enables direct transport to brain bypassing blood brain barrier. Imatinib mesylate, an anti-cancer agent, was found to have potential anti-Alzheimer's activity and thus repurposed for the same. However, the drug has severe side effects, poor brain bioavailability which may hinder effective treatment of Alzheimer's disease. In the current work, imatinib mesylate-loaded liposomes were prepared with particle size below 150 nm with sustained drug release up to 96 h. The liposomal drug formulation was compared with plain drug solution for cytotoxicity on N2a cells and did not show any kind of toxicity at concentrations up to 25 µg/mL. The nanocarrier formulation was then evaluated for brain deposition by nose to brain administration in comparison with drug solution in rats. The liposomes effectively improved the brain deposition of drug in brain from formulation compared to pure drug solution as indicated by AUC from in vivo experiments. These results indicate that the nose to brain delivery of liposomal imatinib mesylate improved the drug deposition and residence time in brain compared to drug solution administered through oral and intranasal routes.


Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Delivery Systems/methods , Drug Development/methods , Imatinib Mesylate/administration & dosage , Imatinib Mesylate/pharmacokinetics , Administration, Intranasal , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Drug Evaluation, Preclinical/methods , Imatinib Mesylate/chemical synthesis , Liposomes , Male , Mice , Rats , Rats, Sprague-Dawley
10.
Int J Nanomedicine ; 16: 1221-1229, 2021.
Article En | MEDLINE | ID: mdl-33628019

BACKGROUND: Pulmonary arterial hypertension (PAH) is characterized by abnormal proliferation of vascular endothelial and smooth muscle cells and causes occlusion of pulmonary arterioles that eventually results in right heart failure and death. The platelet-derived growth factor (PDGF) plays a prominent role in abnormal remodeling of pulmonary resistance vessels. Imatinib mesylate (IM), a PDGF-receptor tyrosine kinase inhibitor, was able to ameliorate PAH by reversing pulmonary vascular remodeling. METHODS: In the present study, IM-loaded liposomes (IM-LPs) were developed and administered via the pulmonary route to delay the drug release and improve patient compliance for the treatment of PAH. The IM-LPs were prepared by the transmembrane gradient method with the spherical vesicles. The compatibility of the IM-LPs was studied by determining the viability of pulmonary arterial smooth muscle cells (PASMCs). Particle uptake by rat PASMCs was evaluated by incubating the particles with rat PASMCs. Pharmacokinetic studies were performed in male SD rats. RESULTS: The IM-LPs showed an average size of 101.6 ± 50.80 nm with a zeta potential value of 19.66 ± 0.55 mV, a PDI of 0.250 and 81.96% ± 0.98% drug entrapment efficiency, meanwhile displayed a sustained release profile. Liposomes obviously increased intracellular accumulation of Rhodamine B by PASMCs using the fluorescence microscopic. Following intratracheal administration to rats, IM-LPs not only extended the half-life of IM, but also prolonged retention of IM compared with plain IM solution after intratracheal and intravenous administration. CONCLUSION: The study show potential applications of the LPs for pulmonary delivery of IM and the method for the development of LPs in sustained release of IM for better therapeutic outcomes. Conclusively, the prepared IM-LPs were well designed in nanosized ranges and may be a promising formulation for pulmonary delivery of IM.


Drug Delivery Systems , Imatinib Mesylate/pharmacokinetics , Lung/drug effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Liberation , Imatinib Mesylate/blood , Imatinib Mesylate/pharmacology , Liposomes , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Rats, Sprague-Dawley
11.
BMC Pharmacol Toxicol ; 21(1): 77, 2020 11 16.
Article En | MEDLINE | ID: mdl-33198812

BACKGROUND: Imatinib is mainly metabolized by CYP3A4 and to a lesser extent by other isoenzymes, with N-desmethyl imatinib being its major equipotent metabolite. Being a CYP3A4 substrate, imatinib co-administration with CYP3A4 modulators would change its pharmacokinetic profile. The cancer chemoprevention potential and anticancer efficacy of many herbal products such as grape seed (GS) and green tea (GT) extracts had led to an increase in their concomitant use with anticancer agents. GS and GT extracts were demonstrated to be potent inhibitors of CYP3A4. The aim of this study is to investigate the effect of standardized GS and/or GT extracts at two different doses on the pharmacokinetics of imatinib and its metabolite, N-desmethyl imatinib, in SD-rats. METHODS: Standardized GS and/or GT extracts were administered orally once daily for 21 days, at low (l) and high (h) doses, 50 and 100 mg/kg, respectively, before the administration of a single intragastric dose of imatinib. Plasma samples were collected and analyzed for imatinib and N-desmethyl imatinib concentrations using LC-MS/MS method, then their non-compartmental pharmacokinetic parameters were determined. RESULTS: h-GS dose significantly decreased imatinib's Cmax and the [Formula: see text] by 61.1 and 72.2%, respectively. Similar effects on N-desmethyl imatinib's exposure were observed as well, in addition to a significant increase in its clearance by 3.7-fold. l-GT caused a significant decrease in imatinib's Cmax and [Formula: see text] by 53.6 and 63.5%, respectively, with more significant effects on N-desmethyl imatinib's exposure, which exhibited a significant decrease by 79.2 and 81.1%, respectively. h-GT showed similar effects as those of l-GT on the kinetics of imatinib and its metabolite. However, when these extracts were co-administered at low doses, no significant effects were shown on the pharmacokinetics of imatinib and its metabolite. Nevertheless, increasing the dose caused a significant decrease in Cmax of N-desmethyl imatinib by 71.5%. CONCLUSIONS: These results demonstrated that the pharmacokinetics of imatinib and N-desmethyl imatinib had been significantly affected by GS and/or GT extracts, which could be partially explained by the inhibition of CYP3A-mediated metabolism. However, the involvement of other kinetic pathways such as other isoenzymes, efflux and uptake transporters could be involved and should be characterized.


Grape Seed Extract/administration & dosage , Herb-Drug Interactions/physiology , Imatinib Mesylate/pharmacokinetics , Plant Extracts/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Tea , Administration, Oral , Animals , Imatinib Mesylate/administration & dosage , Injections, Intraperitoneal , Male , Protein Kinase Inhibitors/administration & dosage , Rats , Rats, Sprague-Dawley , Vitis
12.
Article En | MEDLINE | ID: mdl-32764164

Objectives Among Chronic Myeloid Leukemia (CML) patients treated with Tyrosine Kinase Inhibitor (TKI-imatinib-nilotinib), some showed a suboptimal response. Based on pharmacokinetic studies, TKI trough level ( C m i n ∞ ${C}_{min}\hat{\infty }$ ) is associated with clinical outcomes, reflected by the BCR-ABL ratio. However, the interindividual pharmacokinetic variability of imatinib and nilotinib is found to be moderate-high. This study aims to analyze the relationship between TKI C m i n ∞   ${C}_{min}\hat{\infty }$ and BCL-ABL ratio in chronic-phase CML patients. Methods Cross-sectional study to CML chronic-phase patients treated with imatinib 400 mg daily or nilotinib 400 or 800 mg daily for ≥12 months. The exclusion criteria were therapy discontinuation within 29 days (imatinib) or 8 days (nilotinib) before the sampling day. Blood samples were drawn 1 h before the next dose. Imatinib-nilotinib C m i n ∞ ${C}_{min}\hat{\infty }$ and BCR-ABL ratio were measured using HPLC and RT-qPCR. The relationship was analyzed using bivariate correlation Spearman's rho test. Results Twenty-three imatinib and 11 nilotinib patients met the inclusion criteria. The mean imatinib and nilotinib C m i n ∞ ${C}_{min}\hat{\infty }$ were 1,065.46 ± 765.71 and 1,445 ± 1,010.35 ng/mL respectively. There were large interindividual variations in both groups (71.87% vs. 69.88%). Half of the patients in each group were found to reach C m i n ∞ ${C}_{min}\hat{\infty }$ target (≥1.000 ng/mL, imatinib; ≥800 ng/mL nilotinib), but only 12 (35,29%) of them result in BCR-ABL ratio ≤0.1%. C m i n ∞   ${C}_{min}\hat{\infty }$ imatinib was found to be significantly associated with BCR-ABL ratio. But, not with the nilotinib group. Conclusions There were high interindividual variations of imatinib and nilotinib correlated with BCR-ABL ratio, but no correlation in nilotinib.


Fusion Proteins, bcr-abl/blood , Imatinib Mesylate/administration & dosage , Leukemia, Myeloid, Chronic-Phase/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Adolescent , Adult , Aged , Cross-Sectional Studies , Dose-Response Relationship, Drug , Female , Humans , Imatinib Mesylate/pharmacokinetics , Imatinib Mesylate/pharmacology , Indonesia , Leukemia, Myeloid, Chronic-Phase/pathology , Male , Middle Aged , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Young Adult
13.
Eur J Cancer ; 136: 140-148, 2020 09.
Article En | MEDLINE | ID: mdl-32688207

AIM: Higher imatinib exposure is correlated with longer time to progression, while the variability in exposure is high. This provides a strong rationale for therapeutic drug monitoring, which has therefore been implemented in routine clinical practice in our institute. The aim of this study is to evaluate whether pharmacokinetically (PK)-guided dose increases are feasible in daily clinical practice and result in an improved exposure (Cmin≥1100 ng/mL) and longer progression-free survival (PFS). METHODS: This retrospective study included all patients with a gastrointestinal stromal tumour (GIST) in the Netherlands Cancer Institute who started imatinib treatment at a dose of 400 mg and of whom PK plasma samples were available. Of these patients, minimum plasma concentrations (Cmin) of imatinib, frequency and successfulness of PK-guided dose increases and PFS in the palliative treatment setting were analysed. RESULTS: In total, 169 consecutive patients were included, of whom 1402 PK samples were collected. In 126 patients (75%), Cmin was below the efficacy threshold of 1100 ng/mL. In 78 of these patients (62%), a PK-guided dose increase was performed, which was successful in 49 patients (63%). PFS was similar in patients with and without imatinib dose increase. However, due to the small number of patients with progressive disease, no definite conclusions on the effect on PFS could yet be drawn. CONCLUSION: This is the largest cohort evaluating PK-guided dose increases of imatinib in patients with GIST in routine clinical practice and demonstrating its feasibility. PK-guided dose increases should be applied to optimise exposure in the significant subset of patients with a low Cmin.


Drug Monitoring , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Stromal Tumors/drug therapy , Imatinib Mesylate/pharmacokinetics , Imatinib Mesylate/therapeutic use , Aged , Cohort Studies , Dose-Response Relationship, Drug , Drug Monitoring/methods , Drug Monitoring/statistics & numerical data , Drug-Related Side Effects and Adverse Reactions/epidemiology , Female , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/epidemiology , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Humans , Male , Middle Aged , Netherlands/epidemiology , Practice Patterns, Physicians'/statistics & numerical data , Progression-Free Survival , Retrospective Studies
14.
Pharm Res ; 37(6): 109, 2020 May 31.
Article En | MEDLINE | ID: mdl-32476052

PURPOSE: Cancer-Immunity Cycle is a cascade of anticancer immune responses in the body that continues and fights against the cancer expansion. The Cancer-Immunity Cycle is halted by tumor cell immunosuppression of host T cell through programmed cell death receptor 1 (PD-1) and programmed cell death ligand 1 (PD-L1) interactions that induce the functional suppression of tumor-reactive cytotoxic T cells and actively promotes the tumorigenesis via the mTOR signaling pathway. METHODS: Here, we demonstrated that this Cycle could be enhanced by the synergistic knock down of PD-L1 through co-delivery of siRNA-PD-L1 (siPD-L1) and imatinib (IMT) in a liposomal nanoparticle. RESULTS: The siPDIN effectively downregulated the protein expressions of PD-L1 and significantly knocked down the expression of p-S6k protein at in vitro and in vivo conditions which inhibited tumorigenic mTOR pathway. The combination-based siPDIN exhibited a significantly higher cytotoxic effect compared to that of individual anticancer agents. B16F10 cells treated with siPDIN exhibited a significantly higher cancer cell apoptosis (~60%) compared to cocktail combination of siRNA+IMT (~35%) analyzed by flow cytometer. Importantly, siPDIN significantly delayed the tumor growth with significantly lower tumor-specific growth rate than the animals treated with individual free IMT or siRNA. siPDIN produced a 3-fold higher IFN-γ compared to control in DLNs and 4-fold higher IFN-γ in spleens. CONCLUSION: Overall, results revealed that the tumors treated with siPDIN restored the immunity of CTLs by potentially inhibiting the immune checkpoint interactions, suppressed the mTOR signaling pathway and exhibited an enhanced anticancer efficacy in melanoma.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Imatinib Mesylate/administration & dosage , Melanoma, Experimental/drug therapy , RNA, Small Interfering/administration & dosage , Skin Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Line, Tumor/transplantation , Drug Liberation , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Imatinib Mesylate/pharmacokinetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Liposomes , Melanoma, Experimental/immunology , Mice , Nanoparticles , RNA, Small Interfering/pharmacokinetics , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
15.
Eur J Clin Pharmacol ; 76(8): 1075-1082, 2020 Aug.
Article En | MEDLINE | ID: mdl-32430518

PURPOSE: The aim of this study was to ascertain whether the absolute bioavailability of oral imatinib (Glivec®) during steady state plasma pharmacokinetics in cancer patients could be determined through a concomitant intravenous administration of a single 100 µg microdose of deuterium labeled imatinib (imatinib-d8). Secondly, the usefulness of liquid chromatography-tandem mass spectrometry (LC-MS/MS) was investigated for simultaneous analysis of orally and intravenously administered imatinib. METHODS: Included patients were on a stable daily dose of 400 mg oral imatinib prior to study participation. On day 1, patients received a 100 µg intravenous imatinib-d8 microdose 2.5 h after intake of the oral dose. Plasma samples were collected for 48 h. Imatinib and imatinib-d8 concentrations were simultaneously quantified using a validated LC-MS/MS assay. The absolute bioavailability was calculated by comparing the dose-normalized exposure with unlabeled and stable isotopically labeled imatinib in plasma. RESULTS: A total of six patients were enrolled. All patients had a history of gastrointestinal stromal tumors (GIST). The median absolute bioavailability of oral imatinib at steady state was 76% (range 44-106%). Imatinib and imatinib-d8 plasma concentrations were quantified in all collected plasma samples, with no samples below the limit of quantification for imatinib-d8. CONCLUSION: The absolute bioavailability of imatinib was successfully estimated at steady state plasma pharmacokinetics using the stable isotopically labeled microdose trial design. This study exhibits the use of a stable isotopically labeled intravenous microdose to determine the absolute bioavailability of an oral anticancer agent in patients with LC-MS/MS as the analytical tool.


Antineoplastic Agents/administration & dosage , Imatinib Mesylate/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Administration, Intravenous , Administration, Oral , Aged , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Biological Availability , Chromatography, Liquid , Deuterium , Female , Gastrointestinal Stromal Tumors/blood , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/metabolism , Humans , Imatinib Mesylate/blood , Imatinib Mesylate/pharmacokinetics , Isotope Labeling , Male , Middle Aged , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Tandem Mass Spectrometry
16.
Clin Drug Investig ; 40(7): 617-628, 2020 Jul.
Article En | MEDLINE | ID: mdl-32415468

BACKGROUND: Validated genomic biomarkers for oncological drugs are expanding to improve targeted therapies. Pharmacogenetics research focusing on the mechanisms underlying imatinib suboptimal response might help to explain the different treatment outcomes and drug safety profiles. OBJECTIVE: To investigate whether polymorphisms in genes encoding cytochrome P450 (CYP) enzymes and ABCB1 transporter affect imatinib pharmacokinetic parameters. METHODS: A prospective, multicenter, pharmacogenetic pilot study was performed in the context of two separate oral imatinib bioequivalence clinical trials, which included 26 healthy volunteers. DNA was extracted in order to analyze polymorphisms in genes CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP3A5 and ABCB1. Imatinib plasma concentrations were measured by HPLC-MS/MS. Pharmacokinetic parameters were calculated by non-compartmental methods using WinNonlin software. RESULTS: Volunteers (n = 26; aged 24 ± 3 years; 69% male) presented regular pharmacokinetic imatinib data (concentration at 24 h, 436 ± 140 ng/mL and at 72 h, 40 ± 26 ng/mL; AUC0-72 32,868 ± 10,713 ng/mL⋅h; and Cmax 2074 ± 604 ng/mL). CYP2B6 516GT carriers showed a significant reduction of imatinib concentration at 24 h (23%, 391 ng/dL vs 511 ng/dL in 516GG carriers, p = 0.005) and elimination half-life (11%, 12.6 h vs 14.1 h in 516GG carriers, p = 0.041). Carriers for CYP3A4 (*22/*22, *1/*20 and *1/*22 variants) showed a reduced frequency of adverse events compared to *1/*1 carriers (0 vs 64%, p = 0.033). The other polymorphisms analyzed did not influence pharmacokinetics or drug toxicity. CONCLUSION: CYP2B6 G516T and CYP3A4 *20,*22 polymorphisms could influence imatinib plasma concentrations and safety profile, after single-dose administration to healthy subjects. This finding needs to be confirmed before it is implemented in clinical practice in oncological patients under treatment with imatinib.


Cytochrome P-450 Enzyme System/genetics , Imatinib Mesylate/pharmacokinetics , Polymorphism, Genetic , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Female , Healthy Volunteers , Humans , Imatinib Mesylate/administration & dosage , Male , Pharmacogenetics , Pilot Projects , Prospective Studies , Young Adult
17.
Pharmazie ; 75(4): 136-141, 2020 04 06.
Article En | MEDLINE | ID: mdl-32295689

Here we describe the development and validation of an LC-MS/MS method for the quantification of imatinib and imatinib-d8 in plasma for the support of a clinical absolute bioavailability microdosing trial. The focus lies on the technical aspects to analyse high concentrations of imatinib and low concentrations of imatinib-d8 that are present simultaneously in study samples, using a single sample processing and analytical method. With the validated assay, imatinib and imatinib-d8 can be quantified simultaneously in ranges from 25.0 - 5,000 ng/mL and 0.01 - 2.0 ng/mL, respectively. The method was successfully applied in an imatinib-d8 absolute bioavailability microdosing trial, where a 100 µg imatinib-d8 microdose was intravenously administered to a patient on oral imatinib treatment 400 mg once daily.


Imatinib Mesylate/blood , Imatinib Mesylate/pharmacokinetics , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Biological Availability , Chromatography, High Pressure Liquid , Humans , Imatinib Mesylate/administration & dosage , Limit of Detection , Mass Spectrometry , Protein Kinase Inhibitors/administration & dosage , Reproducibility of Results , Sensitivity and Specificity
18.
Anal Bioanal Chem ; 412(24): 5925-5933, 2020 Sep.
Article En | MEDLINE | ID: mdl-32166445

Therapeutic drug monitoring is used in the clinical setting in the optimisation of dosages to overcome inter-patient pharmacokinetic variability, increasing efficacy whilst reducing toxicity. Imatinib is a tyrosine kinase inhibitor, displaying large variations in plasma concentrations that impact therapeutic success. As a result, imatinib has been the focus in the development of innovative techniques, aimed at its quantification in plasma. Liquid chromatography coupled with tandem mass spectrometry is currently the gold standard; however, cost and availability of the equipment limit its wider application in clinical settings. Recent advances in the field have shown Raman spectroscopy and electrochemistry to be key techniques for the development of promising analytical tools. This article reviews the latest advances towards less costly, more portable solutions that can be used at the point of care. Graphical abstract.


Antineoplastic Agents/blood , Drug Monitoring/instrumentation , Imatinib Mesylate/blood , Point-of-Care Systems , Protein Kinase Inhibitors/blood , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Chromatography, Liquid/methods , Electrochemical Techniques/methods , Enzyme-Linked Immunosorbent Assay/methods , Humans , Imatinib Mesylate/administration & dosage , Imatinib Mesylate/pharmacokinetics , Limit of Detection , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Reproducibility of Results , Spectrophotometry, Ultraviolet/methods , Spectrum Analysis, Raman/methods , Tandem Mass Spectrometry/methods
19.
J Pharm Pharm Sci ; 23(1): 1-9, 2020.
Article En | MEDLINE | ID: mdl-32027818

PURPOSE: We investigated the relationship between imatinib trough concentrations and genetic polymorphisms with efficacy of imatinib in Chinese patients with chronic myeloid leukemia (CML). METHODS: There were 171 eligible patients. Peripheral blood samples were collected from 171 eligible patients between 21 and 27 hours after the last imatinib administration. Complete cytogenetic response (CCyR), major molecular response (MMR) and complete molecular response (CMR) were used as metrics for efficacy. Nine single nucleotide polymorphisms in 5 genes, SLC22A4 (917 T>C, -248 C>G and -538 C>G), SLC22A5 (-945 T>G and -1889 T>C), SLCO1A2 (-361 G>A), SLCO1B3 (334 T>G and 699 G>A) and ABCG2 (421C>A) were selected for genotyping. RESULTS: Patients with CCyR achieve higher trough concentrations than those without CCyR (1478.18±659.83 vs 984.89±454.06 ng mL-1, p<0.001). Patients with MMR and CMR achieve higher trough concentrations than those without MMR and CMR, respectively (1486.40±703.38 vs 1121.17±527.14 ng mL-1, p=0.007; 1528.00±709.98 vs 1112.67±518.35 ng mL-1, p=0.003, respectively). Carriers of A allele in SLCO1A2 -361G>A achieve higher CCyR and MMR rates (p=0.047, OR=4.320, 95% CI: 0.924-20.206; p=0.042, OR=2.825, 95% CI: 1.016-7.853, respectively). Both trough concentrations and SLCO1A2 -361G>A genotypes are independent factors affecting imatinib efficacy. The positive and negative predictive values for CCyR are 71.01% and 68.75%, respectively. The positive and negative predictive values for MMR are 62.86% and 69.70%, respectively. CONCLUSION: Imatinib trough concentrations and SLCO1A2 -361G>A genotypes are associated with imatinib efficacy in Chinese patients with CML.


Antineoplastic Agents , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Membrane Transport Proteins/genetics , Neoplasm Proteins/genetics , Protein Kinase Inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Asian People/genetics , Female , Genotype , Humans , Imatinib Mesylate/blood , Imatinib Mesylate/pharmacokinetics , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Treatment Outcome , Young Adult
...