Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Pharmacol Res Perspect ; 12(4): e1241, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992911

ABSTRACT

Lenvatinib (LEN), a multitarget tyrosine kinase inhibitor used in various cancer treatments, is mainly metabolized by cytochrome P450 3A (CYP3A) enzymes. The importance of therapeutic drug monitoring (TDM) in patients administered LEN has been proposed. Although some biomarkers of endogenous CYP3A activity have been reported, their utility in dosage adjustments has not been well evaluated. This study investigated the correlation between plasma LEN concentrations and endogenous urinary CYP3A biomarkers in clinical practice. Concentrations of plasma LEN (N = 225) and CYP3A biomarkers (cortisol, 6ß-hydroxycortisol, deoxycholic acid, and 1ß-hydroxydeoxycholic acid) in urine (N = 214) from 20 patients (hepatocellular carcinoma, N = 6; thyroid cancer, N = 3; endometrial cancer, N = 8; and renal cell carcinoma, N = 3) collected for consultation for up to 1 year were evaluated using liquid chromatography-tandem mass spectrometry. Moreover, plasma trough LEN concentrations were predicted using a three-compartment model with linear elimination for outpatients administered LEN before sample collection. Moderate correlations were observed between the quantified actual concentrations and the predicted trough concentrations of LEN, whereas there was no correlation with endogenous urinary CYP3A biomarkers. The utility of endogenous urinary CYP3A biomarkers could not be determined. However, TDM for outpatients administered orally available medicines may be predicted using a nonlinear mixed effect model (NONMEM). This study investigated the utility of endogenous urinary CYP3A biomarkers for personalized medicine and NONMEM for predicting plasma trough drug concentrations. These findings will provide important information for further clinical investigation and detailed TDM.


Subject(s)
Biomarkers , Cytochrome P-450 CYP3A , Drug Monitoring , Phenylurea Compounds , Quinolines , Humans , Phenylurea Compounds/urine , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/blood , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/administration & dosage , Female , Quinolines/urine , Quinolines/therapeutic use , Quinolines/blood , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Aged , Middle Aged , Male , Biomarkers/urine , Biomarkers/blood , Drug Monitoring/methods , Adult , Aged, 80 and over , Antineoplastic Agents/urine , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Protein Kinase Inhibitors/urine , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Neoplasms/drug therapy , Neoplasms/blood , Neoplasms/urine , Tandem Mass Spectrometry/methods , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/urine , Endometrial Neoplasms/blood , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/urine , Chromatography, Liquid/methods , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/urine , Thyroid Neoplasms/blood , Liver Neoplasms/drug therapy , Liver Neoplasms/blood , Liver Neoplasms/urine , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/urine , Carcinoma, Renal Cell/blood
2.
Article in English | MEDLINE | ID: mdl-38761468

ABSTRACT

In clinical practice, the determination of unbound drug concentration is very important for dose adjustment and toxicity prediction because only the unbound fraction can achieve a pharmacological effect. A fast, sensitive and accurate analytical method of centrifugal ultrafiltration coupled with high performance liquid chromatography-tandem mass spectrometry method was developed and applied to allow the quantification of unbound lenvatinib concentration. The application of linear regression analysis was used to examine the effects of centrifugal force, centrifugal time, and protein content on ultrafiltrate volume (Vu). The results indicated that the centrifugal force and centrifugal time have an influence on Vu that is significantly positive (P < 0.05). This developed method with good linearity (r2 = 0.9996), good accuracy (bias % ≤ 2.24 %), good precision (CV % ≤ 7.10 %), and good recovery (95.46 %-106.46 %) was suitable for routine clinical practice and studies. Particularly, the ultrafiltration membrane had no non-specific binding to lenvatinib. The unbound fractions can be separated in just 15 min. This method was applied to quantify clinical samples and to determine the plasma protein binding and unbound fraction of lenvatinib. This study provides a more effective and promising method for determination of unbound lenvatinib. It could be beneficial to measure the unbound concentration of lenvatinib in personalized medicine and therapeutic drug monitoring in routine clinical practice.


Subject(s)
Phenylurea Compounds , Quinolines , Tandem Mass Spectrometry , Ultrafiltration , Humans , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/chemistry , Phenylurea Compounds/analysis , Quinolines/blood , Quinolines/chemistry , Quinolines/pharmacokinetics , Tandem Mass Spectrometry/methods , Chromatography, High Pressure Liquid/methods , Ultrafiltration/methods , Linear Models , Reproducibility of Results , Protein Binding , Limit of Detection
3.
CPT Pharmacometrics Syst Pharmacol ; 13(6): 954-969, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38528813

ABSTRACT

Lenvatinib is a receptor tyrosine kinase (RTK) inhibitor targeting vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor (FGF) receptors 1-4, platelet-derived growth factor receptor-α (PDGFRα), KIT, and RET that have been implicated in pathogenic angiogenesis, tumor growth, and cancer. The primary objective of this work was to evaluate, by establishing quantitative relationships, whether lenvatinib exposure and longitudinal serum biomarker data (VEGF, Ang-2, Tie-2, and FGF-23) are predictors for change in longitudinal tumor size which was assessed based on data from 558 patients with radioiodine-refractory differentiated thyroid cancer (RR-DTC) receiving either lenvatinib or placebo treatment. Lenvatinib PK was best described by a 3-compartment model with simultaneous first- and zero-order absorption and linear elimination from the central compartment with significant covariates (body weight, albumin <30 g/dL, ALP>ULN, RR-DTC, RCC, HCC subjects, and concomitant CYP3A inhibitors). Except for body weight, none of the covariates have any clinically meaningful effect on exposure to lenvatinib. Longitudinal biomarker measurements over time were reasonably well defined by a PK/PD model with common EC50, Emax, and a slope for disease progression for all biomarkers. Longitudinal tumor measurements over time were reasonably well defined by a tumor growth inhibition Emax model, which in addition to lenvatinib exposure, included model-predicted relative changes from baseline over time for Tie-2 and Ang-2 as having significant association with tumor response. The developed PK/PD models pave the way for dose optimization and potential prediction of clinical response.


Subject(s)
Iodine Radioisotopes , Phenylurea Compounds , Quinolines , Thyroid Neoplasms , Humans , Quinolines/pharmacokinetics , Quinolines/administration & dosage , Quinolines/therapeutic use , Quinolines/blood , Quinolines/pharmacology , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/blood , Thyroid Neoplasms/pathology , Male , Female , Middle Aged , Adult , Iodine Radioisotopes/pharmacokinetics , Iodine Radioisotopes/therapeutic use , Aged , Biomarkers, Tumor/blood , Models, Biological , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/blood , Vascular Endothelial Growth Factor A/blood , Receptor, TIE-2/blood , Young Adult , Angiopoietin-2/blood
4.
Invest New Drugs ; 42(3): 252-260, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38517650

ABSTRACT

The aim of the present study was to determine whether the trough plasma concentrations (C0) of regorafenib and its metabolites, the N-oxide metabolite (M-2) and the desmethyl N-oxide metabolite (M-5), in 21 patients receiving regorafenib therapy were affected by albumin-bilirubin (ALBI) grade. Regorafenib was administered at dosages ranging from 40 to 160 mg once daily on a 3-week-on, 1-week-off cycle. C0 values of regorafenib and its major metabolites were measured by high-performance liquid chromatography on day 8 after treatment initiation. The C0 values of regorafenib and metabolites M-2 and M-5 were significantly lower in patients with ALBI grade 2 as compared with grade 1 (P = 0.023, 0.003 and 0.017, respectively). The total C0 of regorafenib and its metabolites was significantly higher in ALBI grade 1 patients relative to grade 2 (3.489 µg/mL vs. 1.48 µg/mL; P = 0.009). The median relative dose intensity (RDI) of patients categorized as ALBI grade 2 was significantly lower than that of grade 1 patients (21.9% vs. 62.9%; P = 0.006). In 15 colorectal cancer patients among the total 21 patients, patients with ALBI grade 2 (n = 9) had a significantly shorter median overall survival time than patients with grade 1 (n = 6; P = 0.013). Administering a low dose of regorafenib to patients with ALBI grade 2 reduces the RDI of regorafenib and lowers treatment efficacy, as an appropriate C0 of regorafenib is not maintained. Monitoring the C0 of regorafenib regularly is necessary to guide dose adjustment.


Subject(s)
Bilirubin , Phenylurea Compounds , Pyridines , Humans , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/blood , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/administration & dosage , Pyridines/pharmacokinetics , Pyridines/blood , Pyridines/therapeutic use , Pyridines/administration & dosage , Female , Male , Aged , Middle Aged , Bilirubin/blood , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/blood , Aged, 80 and over , Adult , Japan , Asian People , Serum Albumin/metabolism , East Asian People
5.
Bioanalysis ; 16(10): 461-473, 2024.
Article in English | MEDLINE | ID: mdl-38530220

ABSTRACT

Aim: Investigation of the pharmacokinetics of sorafenib (SRF) in rats with hepatocellular carcinoma (HCC). Methods: A reproducible ultra-HPLC-MS method for simultaneous determination of serum SRF, N-hydroxymethyl sorafenib and N-demethylation sorafenib. Results: Both the maximum serum concentrations (2.5-times) and the area under the serum concentration-time curve from 0 h to infinity (4.5-times) of SRF were observed to be significantly higher, with a greater than 3.0-fold decrease in the clearance rate in the HCC-bearing rats compared with these values in healthy animals. Further study revealed approximately 3.8- and 3.2-times increases in the apparent Michaelis constant for N-hydroxymethyl sorafenib and N-demethylation sorafenib conversions in the HCC-bearing rats. Conclusion: The low efficiency for the SRF conversions was a key contributor to the increased serum concentrations of SRF.


[Box: see text].


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Niacinamide , Phenylurea Compounds , Sorafenib , Sorafenib/pharmacokinetics , Sorafenib/blood , Sorafenib/therapeutic use , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Niacinamide/analogs & derivatives , Niacinamide/blood , Niacinamide/pharmacokinetics , Rats , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Male , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/blood , Phenylurea Compounds/therapeutic use , Chromatography, High Pressure Liquid/methods , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/blood , Rats, Sprague-Dawley , Mass Spectrometry
6.
PLoS One ; 16(10): e0259137, 2021.
Article in English | MEDLINE | ID: mdl-34699578

ABSTRACT

Lenvatinib (LENVA) is an oral antineoplastic drug used for the treatment of hepatocellular carcinoma and thyroid carcinoma. LENVA therapeutic drug monitoring (TDM) should be mandatory for a precision medicine to optimize the drug dosage. To this end, the development of a sensitive and robust quantification method to be applied in the clinical setting is essential. The aim of this work was to develop and validate a sensitive, rapid, and cost-effective LC-MS/MS method for the quantification of LENVA in human plasma. On this premise, sample preparation was based on a protein precipitation and the chromatographic separation was achieved on a Synergi Fusion RP C18 column in 4 min. The method was completely and successfully validated according to European Medicines Agency (EMA) and Food and Drug Administration (FDA) guidelines, with good linearity in the range of 0.50-2000 ng/mL (R≥0.9968). Coefficient of variation (CV) for intra- and inter-day precision was ≤11.3% and accuracy ranged from 96.3 to 109.0%, internal standard normalized matrix effect CV% was ≤2.8% and recovery was ≥95.6%. Successful results were obtained for sensitivity (signal to noise (S/N) ratio >21) and selectivity, dilution integrity (CV% ≤ 4.0% and accuracy 99.9-102%), and analyte stability under various handling and storage conditions both in matrix and solvents. This method was applied to quantify LENVA in patient's plasma samples and covered the concentration range achievable in patients. In conclusion, a sensitive and robust quantification method was developed and validated to be applied in the clinical setting.


Subject(s)
Antineoplastic Agents/blood , Chromatography, Liquid/methods , Drug Monitoring/methods , Phenylurea Compounds/blood , Quinolines/blood , Tandem Mass Spectrometry/methods , Humans , Limit of Detection , Reproducibility of Results
7.
Cancer Chemother Pharmacol ; 87(6): 767-777, 2021 06.
Article in English | MEDLINE | ID: mdl-33635392

ABSTRACT

PURPOSE: The association between the pharmacokinetics and pharmacodynamics of regorafenib, a multiple tyrosine kinase inhibitor, remains unclear. This study assessed the trough plasma concentrations (Ctrough) of regorafenib and its N-oxide (M2) and N-oxide/desmethyl (M5) metabolites, and evaluated the associations among these levels, adverse events, and pharmacokinetic-related genetic polymorphisms in patients with metastatic colorectal cancer. METHODS: The Ctrough levels of regorafenib and its metabolites were assessed in a single-center, prospective, observational study, 7 days after the initial treatment. The correlation between those values and adverse events was then examined. In addition, the genetic polymorphisms of ABCG2, SLCO1B1, and UGT1A9 were determined and evaluated for associations with the levels of regorafenib, M2, and M5. RESULTS: We analyzed 43 patients who received regorafenib 40-120 mg/day; among them, 35 patients started at 120 mg/day. With regard to bilirubin increase, the Ctrough values of regorafenib were significantly higher in the group with grade ≥ 2 than in groups with grades 0 and 1 (p = 0.010). The M5 Ctrough levels were significantly associated with the severity of hypertension or rash (p < 0.05). In a multivariate analysis, the M5 Ctrough values and age were significant predictors of severe rash. Lastly, significant differences were noted in the M5 concentration-to-dose ratio values between the patients with ABCG2 421A/A and ABCG2 421C/A or C/C polymorphisms (p = 0.035). CONCLUSION: This study showed that the Ctrough of regorafenib was associated with bilirubin increase, and also clarified for the first time that the Ctrough of M5 was significantly correlated with hypertension and severe rash.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Neoplasm Proteins/genetics , Phenylurea Compounds/blood , Polymorphism, Genetic/genetics , Pyridines/blood , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/blood , Female , Humans , Male , Middle Aged , Phenylurea Compounds/adverse effects , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/therapeutic use , Prospective Studies , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyridines/therapeutic use
8.
Ann Hematol ; 99(9): 1989-2007, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32683457

ABSTRACT

Several small molecule inhibitors (SMIs) have been recently approved for AML patients. These targeted therapies could be more tolerable than classical antineoplastics, but potential drug-drug interactions (DDI) are relatively frequent. Underestimation or lack of appropriate awareness and management of DDIs with SMIs can jeopardize therapeutic success in AML patients, which often require multiple concomitant medications in the context of prior comorbidities or for the prevention and treatment of infectious and other complications. In this systematic review, we analyze DDIs of glasdegib, venetoclax, midostaurin, quizartinib, gilteritinib, enasidenib, and ivosidenib. CYP3A4 is the main enzyme responsible for SMIs metabolism, and strong CYP3A4 inhibitors, such azoles, could increase drug exposure and toxicity; therefore dose adjustments (venetoclax, quizartinib, and ivosidenib) or alternative therapies or close monitoring (glasdegib, midostaurin, and gilteritinib) are recommended. Besides, coadministration of strong CYP3A4 inducers with SMIs should be avoided due to potential decrease of efficacy. Regarding tolerability, QTc prolongation is frequently observed for most of approved SMIs, and drugs with a potential to prolong the QTc interval and CYP3A4 inhibitors should be avoided and replaced by alternative treatments. In this study, we critically assess the DDIs of SMIs, and we summarize best management options for these new drugs and concomitant medications.


Subject(s)
Antineoplastic Agents/blood , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Approval , Drug Interactions/physiology , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/drug therapy , Antineoplastic Agents/adverse effects , Benzimidazoles/adverse effects , Benzimidazoles/blood , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/blood , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Drugs, Investigational/adverse effects , Drugs, Investigational/metabolism , Humans , Long QT Syndrome/blood , Long QT Syndrome/chemically induced , Phenylurea Compounds/adverse effects , Phenylurea Compounds/blood , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Staurosporine/adverse effects , Staurosporine/analogs & derivatives , Staurosporine/blood , Sulfonamides/adverse effects , Sulfonamides/blood
9.
Eur J Clin Pharmacol ; 76(8): 1125-1133, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32382947

ABSTRACT

PURPOSE: To investigate whether the CYP3A4/5 and ABC transporter genetic polymorphisms could affect the pharmacokinetics of lenvatinib in Chinese healthy subjects. METHODS: Thirty-two healthy Chinese volunteers were enrolled and took oral administration of 8 mg lenvatinib. Plasma concentration of lenvatinib was determined by UPLC-MS/MS, the CYP3A4*1G, CYP3A5*3, ABCB1 (3435 C>T, 1236 C>T, 2677 G>T/A), ABCG2 (421 C>A, 34 G>A), and ABCC2-24 C>T genotypes were determined by SnapShot Technique. RESULTS: In ABCB1 3435T carriers (n = 19), AUC0-120h (815.7 (701.9-923.9) ng·h/mL) and AUC0-∞ (843.3 (722.2-977.7) ng·h/mL) were significantly higher than ABCB1 3435CC homozygous subjects (n = 13, 575.3 (513.7-756.9) ng·h/mL and 590.0 (540.5-782.0) ng·h/mL, respectively); on the contrary, the clearance (CL/F) of ABCB1 3435T carriers was significantly lower (9.5 (8.2-11.1) L/h vs. 13.6 (10.4-14.8) L/h). And the Cmax in CYP3A4*1G/*1G allele carrier subjects was higher than *1 carrier (73.4 ng/mL vs. 53.5 (46.1-60.6) ng/mL), but did not reach the level of significantly statistical difference. Genetic polymorphisms of ABCC2, ABCG2, and CYP3A5 could not influence pharmacokinetic parameters of lenvatinib. CONCLUSIONS: This work presented an evidence that the ABCB1 3435 C>T polymorphism could significantly affect the exposure and clearance of lenvatinib. These findings may explain the reasons for the huge inter-individual differences in lenvatinib, and should contribute to clinical individualized treatment.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A/genetics , Phenylurea Compounds/pharmacokinetics , Quinolines/pharmacokinetics , Adult , Antineoplastic Agents/blood , Asian People/genetics , Diet, High-Fat , Fasting/metabolism , Female , Genotype , Healthy Volunteers , Humans , Male , Multidrug Resistance-Associated Protein 2 , Phenylurea Compounds/blood , Polymorphism, Single Nucleotide , Quinolines/blood , Young Adult
10.
Ther Drug Monit ; 42(4): 554-558, 2020 08.
Article in English | MEDLINE | ID: mdl-32398418

ABSTRACT

BACKGROUND: Therapeutic drug monitoring (TDM) and dose adjustment of lenvatinib may be beneficial in the treatment of radioiodine-refractory thyroid cancer, by maximizing antitumor effects and minimizing adverse drug reactions. The aim of this study was, therefore, to develop and validate a high-performance liquid chromatography method using an ultraviolet detection system for routine serum lenvatinib detection in patients with thyroid cancer. METHODS: Serum specimens, spiked with an internal standard, were treated by a solid-phase extraction through an octadecylsilyl silica cartridge. Lenvatinib and internal standard were concomitantly separated from serum using a conventional octadecylsilyl silica column through isocratic elution, using a mobile phase consisting of 0.02 mol/L sodium phosphate (pH 6.7) and acetonitrile (50/50, vol/vol) at a flow rate of 1.0 mL/min. The detection wavelength was set at 244 nm. Serum samples from 5 patients were used for clinical validation of the method. RESULTS: The calibration curve for lenvatinib was linear (Pearson correlation coefficient, r = 0.9998) over the concentration range of 6.25-400 ng/mL, with a lower limit of quantification of 6.25 ng/mL. Extraction recoveries for lenvatinib were 97% or more, with coefficients of variation less than 2.2%. The coefficients of variation for intraday and interday assays were less than 4.7% and 6.0%, respectively. CONCLUSIONS: This sensitive high-performance liquid chromatography method can be used for lenvatinib therapeutic drug monitoring when liquid chromatography-tandem mass spectrometry facilities are unavailable.


Subject(s)
Chromatography, High Pressure Liquid/methods , Drug Monitoring/methods , Phenylurea Compounds/blood , Quinolines/blood , Acetonitriles/blood , Aged , Calibration , Female , Humans , Iodine Radioisotopes/blood , Limit of Detection , Male , Middle Aged , Reproducibility of Results , Solid Phase Extraction/methods
11.
Clin Pharmacol Ther ; 108(3): 586-595, 2020 09.
Article in English | MEDLINE | ID: mdl-32034953

ABSTRACT

Regorafenib treatment improves survival of patients with metastatic colorectal cancer, but it is also characterized by detrimental side effects that may require modified dosing or interval schedules. Regorafenib is metabolized by cytochrome P450 3A4 in the liver to its active metabolites, M-2 and M-5. We examined area under the unbound plasma concentration-time curve (AUCu) to these compounds to establish pharmacokinetic bases for individualized dosing strategies. The plasma protein binding of M-2 and M-5 was approximately 10-fold lower than that of regorafenib, whereas AUCu values for active metabolites on both days 1 and 15 were significantly higher than that of regorafenib. Patients with higher AUCu values of M-2 or M-5 on day 1 showed significantly shorter progression-free survival than others, likely due, at least in part, to treatment discontinuation as a result of adverse events, especially occurred during first cycle.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Colorectal Neoplasms/drug therapy , Phenylurea Compounds/pharmacokinetics , Pyridines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Activation, Metabolic , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/mortality , Cytochrome P-450 CYP3A/metabolism , Drug Dosage Calculations , Female , Humans , Liver/enzymology , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pharmacogenomic Variants , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/adverse effects , Phenylurea Compounds/blood , Polymorphism, Genetic , Progression-Free Survival , Prospective Studies , Protein Binding , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/blood
12.
Clin Pharmacol Drug Dev ; 9(2): 277-286, 2020 02.
Article in English | MEDLINE | ID: mdl-31916418

ABSTRACT

Quizartinib is an oral, highly potent, and selective type II FMS-like tyrosine kinase 3 inhibitor in development for acute myeloid leukemia. This parallel-group study evaluated potential food effects on quizartinib absorption in healthy subjects who received a single 30-mg dose after overnight fasting (n = 34) or a high-fat, high-calorie meal (n = 30). Blood samples were collected through 504 hours after dosing, and pharmacokinetic parameters calculated were maximum observed concentration (Cmax ) and area under plasma concentration-time curve from time 0 to last quantifiable concentration (AUClast ) and from time 0 to infinity (AUCinf ). Mean quizartinib pharmacokinetic profiles were similar under fasted and fed conditions. The geometric least squares means ratios (%) for fed/fasted and associated 90% confidence intervals (CIs) for Cmax , AUClast , and AUCinf were 91.58 (82.15-102.08), 105.39 (90.79-122.35), and 108.39 (91.54-128.34), respectively. The 90%CI for the ratio fell within the 80% to 125% limits for Cmax and AUClast , with 90%CI for AUCinf slightly outside the limits (ie, 128%). Food delayed quizartinib time to Cmax by 2 hours. All adverse events were either mild or moderate; no discontinuations due to adverse events occurred. Based on these results, quizartinib can be administered without regard to food.


Subject(s)
Benzothiazoles/pharmacokinetics , Food/adverse effects , Leukemia, Myeloid, Acute/drug therapy , Phenylurea Compounds/pharmacokinetics , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Administration, Oral , Adult , Area Under Curve , Benzothiazoles/administration & dosage , Benzothiazoles/adverse effects , Benzothiazoles/blood , Biological Availability , Body Mass Index , Fasting/blood , Fasting/metabolism , Female , Food-Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/adverse effects , Phenylurea Compounds/blood , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Safety
13.
Int J Clin Oncol ; 25(4): 531-540, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31807967

ABSTRACT

BACKGROUND: Regorafenib is a multiple tyrosine kinase inhibitor, and the use of this drug is approved for the treatment of cancers that are resistant to chemotherapy, which include advanced colorectal cancer, gastrointestinal stromal tumor, and hepatocellular carcinoma. However, the drug causes adverse events, including skin toxicities that require dose modification in approximately 75% of cases. At present, the blood concentration of regorafenib is not assessed in clinical settings; thus, we recently developed a method that can assess the blood concentration of the drug using high-performance liquid chromatography. METHODS: We measured the trough blood concentrations (Ctrough) of regorafenib and its metabolites (M2 and M5) in 14 and 4 patients with advanced colorectal cancer and gastrointestinal stromal tumor, respectively, using high-performance liquid chromatography. Then, the correlation between the Ctrough and therapeutic outcomes of regorafenib was analyzed. RESULTS: In patients who were receiving regorafenib 40-160 mg/day, the Ctrough values of regorafenib, M2, and M5 were 318-9467, 34-3594, and 38-3796 ng/mL, respectively. The difference in the values was significant. Although the specific factors influencing this difference were not elucidated, the Ctrough of regorafenib was extremely lower in some patients, even though the drug was administered at a standard dose, which may explain the lower response rate. Moreover, the Ctrough value of M5 was significantly correlated to the incidence of skin toxicities, which is the most frequent cause of dose modification. CONCLUSIONS: The use of regorafenib may not be suitable in patients with a low Ctrough value. To prevent skin toxicities, the Ctrough value of M5 should be monitored.


Subject(s)
Antineoplastic Agents/adverse effects , Colorectal Neoplasms/drug therapy , Gastrointestinal Stromal Tumors/drug therapy , Phenylurea Compounds/adverse effects , Pyridines/adverse effects , Skin Diseases/chemically induced , Skin/drug effects , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Aged , Aged, 80 and over , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/pathology , Cytochrome P-450 CYP3A/genetics , Drug Monitoring/methods , Female , Gastrointestinal Stromal Tumors/pathology , Glucuronosyltransferase/genetics , Humans , Incidence , Male , Middle Aged , Neoplasm Proteins/genetics , Phenylurea Compounds/blood , Phenylurea Compounds/metabolism , Phenylurea Compounds/therapeutic use , Polymorphism, Single Nucleotide , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/therapeutic use , Pyridines/blood , Pyridines/metabolism , Pyridines/therapeutic use , Treatment Outcome
14.
Cancer Med ; 8(13): 5891-5902, 2019 10.
Article in English | MEDLINE | ID: mdl-31429205

ABSTRACT

This open-label, phase I dose-finding study evaluated the gonadotropin-releasing hormone antagonist, TAK-385, in Japanese patients with nonmetastatic prostate cancer. In a two-part design, patients received daily oral TAK-385 at doses of 320 (loading, day 1)/80 (maintenance, day 2 and thereafter), 320/120, 320/160, or 360/120 mg for 28 days in a dose-escalation phase (part A, n = 13), and at 320/80 or 320/120 mg for up to 96 weeks in a randomized expansion phase (part B, n = 30). Primary endpoint in both parts was safety, including dose-limiting toxicity in part A. Secondary endpoints included pharmacokinetics, pharmacodynamics, and prostate-specific antigen concentration. Ten (77%) patients in part A and all patients in part B experienced an adverse event; hot flush (part A, n = 4; part B, n = 15), viral upper respiratory tract infection (part A, n = 1; part B, n = 10), and diarrhea (part B, n = 8) were most frequent. No dose-limiting toxicities were observed (part A). In 12 evaluable patients (part A), TAK-385 was rapidly absorbed after a single loading dose; on day 28 (maintenance dose), median steady-state Tmax was ~1-2 hours and mean t1/2z was 67-79 hours. All doses rapidly reduced testosterone concentrations to castration levels within 1 week. Durable reductions in prostate-specific antigen of >90% from baseline were observed through 96 weeks. TAK-385 appeared tolerable and resulted in sustained reductions in testosterone to castration levels at all doses. The lowest loading/maintenance dose required for a clinical effect was 320/80 mg. ClinicalTrials.gov: NCT02141659.


Subject(s)
Antineoplastic Agents/administration & dosage , Phenylurea Compounds/administration & dosage , Prostatic Neoplasms/blood , Pyrimidinones/administration & dosage , Receptors, LHRH/antagonists & inhibitors , Administration, Oral , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Asian People , Humans , Kallikreins/blood , Male , Phenylurea Compounds/adverse effects , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacokinetics , Prostate-Specific Antigen/blood , Prostatic Neoplasms/drug therapy , Pyrimidinones/adverse effects , Pyrimidinones/blood , Pyrimidinones/pharmacokinetics , Testosterone/blood
15.
Article in English | MEDLINE | ID: mdl-31352204

ABSTRACT

The first bioanalytical assay for tivozanib in human and mouse plasma, mouse tissue homogenates and culture medium was developed and validated over a linear dynamic range from 0.5 to 5000 ng/mL. The extended concentration range will cover the quantification of tivozanib in the majority of study samples, reducing the need for reanalysis which is often not possible due to limited amount of sample in preclinical studies. A simple and fast pretreatment method was used consisting of protein precipitation with acetonitrile followed by dilution of the supernatant. The final extract was injected onto an Ultra-Performance Liquid Chromatography (UPLC) BEH C18 column with gradient elution of formic acid in water and formic acid in acetonitrile mobile phase. Chromatographic separation was followed by detection with a triple-quadrupole mass spectrometer operating in the positive ion-mode. By simultaneously monitoring the sensitive conventional [M + H]+ isotopologue- product transition for quantification of low concentrations and a less abundant [M + H]++1 isotopologue- product transition to reduce the sensitivity for quantification of high concentrations, we were able to extend the overall linear dynamic range up to 0.5-5000 ng/mL. A full validation was performed in human plasma and a partial validation was executed for the other matrices. All results were within the acceptance criteria of the European Medicines Agency (EMA) guidelines and the US Food and Drug Administration (FDA) guidance, except for the carry-over. This was solved by the analysis of extra matrix blanks and by grouping study samples containing a high tivozanib concentration in the sample sequence. In this way carry-over did not impact the data integrity. We demonstrated that by measuring two multiple reaction monitoring (MRM) transitions for tivozanib, the linear dynamic range could be extended from two to four decades. The assay was successfully applied in pharmacokinetic studies in mice and a transport assay.


Subject(s)
Chromatography, High Pressure Liquid/methods , Culture Media/chemistry , Phenylurea Compounds/chemistry , Quinolines/chemistry , Tandem Mass Spectrometry/methods , Animals , Humans , Kidney/chemistry , Liver/chemistry , Lung/chemistry , Mice , Phenylurea Compounds/blood , Plasma/chemistry , Quinolines/blood , Spleen/chemistry
16.
Sci Rep ; 9(1): 5404, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30931962

ABSTRACT

Lenvatinib is a substrate of cytochrome P450 (CYP) 3A and ATP-binding cassette (ABC) transporters. In this study, we aimed to evaluate how CYP3A4/5 and ABC transporter polymorphisms affected the mean steady-state dose-adjusted plasma trough concentrations (C0) of lenvatinib in a cohort of 40 Japanese patients with thyroid cancer. CYP3A4 20230G > A (*1G), CYP3A5 6986A > G (*3), ABCB1 1236C > T, ABCB1 2677G > T/A, ABCB1 3435C > T, ABCC2 -24C > T, and ABCG2 421C > A genotypes were determined using polymerase chain reaction-restriction fragment length polymorphism. In univariate analysis, there were no significant differences in the mean dose-adjusted C0 values of lenvatinib between the ABCB1, ABCG2, and CYP3A5 genotypes. However, the mean dose-adjusted C0 values of lenvatinib in patients with the CYP3A4*1/*1 genotype and ABCC2 -24T allele were significantly higher than those in patients with the CYP3A4*1G allele and -24C/C genotype, respectively (P = 0.018 and 0.036, respectively). In multivariate analysis, CYP3A4 genotype and total bilirubin were independent factors influencing the dose-adjusted C0 of lenvatinib (P = 0.010 and 0.046, respectively). No significant differences were found in the incidence rates of hypertension, proteinuria, and hand-foot syndrome following treatment with lenvatinib between the genotypes of CYP3A4/5 and ABC transporters. Lenvatinib pharmacokinetics were significantly influenced by the CYP3A4*1G polymorphism. If the target plasma concentration of lenvatinib for efficacy or toxicity is determined, elucidation of the details of the CYP3A4*1G genotype may facilitate decision-making related to the appropriate initial lenvatinib dosage to achieve optimal plasma concentrations.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cytochrome P-450 CYP3A/genetics , Phenylurea Compounds/therapeutic use , Polymorphism, Single Nucleotide , Quinolines/therapeutic use , Thyroid Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Aged , Alleles , Asian People/genetics , Female , Gene Frequency , Genotype , Humans , Japan , Male , Middle Aged , Multidrug Resistance-Associated Protein 2 , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacokinetics , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Quinolines/blood , Quinolines/pharmacokinetics , Regression Analysis , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/ethnology
17.
Med Oncol ; 36(5): 39, 2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30919115

ABSTRACT

The aim of this study was to examine the association of lenvatinib-induced adverse events with the trough plasma concentration (C0) in Japanese patients with thyroid cancer. Patients received lenvatinib 24 mg as an initial dose, and sequential dose reductions were conducted based on the grade of each side effect. Assessment of adverse events, assay of lenvatinib C0, and analysis of clinical laboratory tests were performed at the same time of day and were retrospectively analyzed. There were no significant differences in lenvatinib C0 among grades of hypertension, proteinuria, hand-foot syndrome, and diarrhea. However, levels of aspartate transaminase, alanine transaminase, and total bilirubin were significantly higher in lenvatinib C0 quartile (Q) 4 (≥ 88 ng/mL) than in Q1 (< 42 ng/mL) and Q2-3 (42-88 ng/mL). Additionally, platelet counts were highest in the lowest Q1 group. The median dose of lenvatinib in patients with UGT1A1*6/*6 or *6/*28 (poor metabolizers [PMs]) was significantly lower than that in patients with UGT1A1*1/*1 (10 and 14 mg, respectively), whereas the median bilirubin levels were significant higher in UGT1A1 PMs (0.9 and 0.5 mg/dL, respectively). There were no significant differences in median lenvatinib C0 values between patients with UGT1A1*1/*1 and PMs (58.0 and 50.0 ng/mL, respectively). The threshold between the C0 and toxicity of lenvatinib may be more than 88 ng/mL. Therefore, the dose of lenvatinib could be controlled to maintain a lower C0 of less than 88 ng/mL. The target C0 for lenvatinib as the threshold between the C0 and optimal response may be in the range from 42 to 88 ng/mL; however, further studies are necessary.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/blood , Phenylurea Compounds/adverse effects , Protein Kinase Inhibitors/adverse effects , Quinolines/adverse effects , Thyroid Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Drug Monitoring , Drug-Related Side Effects and Adverse Reactions/pathology , Drug-Related Side Effects and Adverse Reactions/physiopathology , Female , Genotype , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Japan , Male , Middle Aged , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Quinolines/administration & dosage , Quinolines/blood , Quinolines/pharmacokinetics , Retrospective Studies , Thyroid Neoplasms/blood , Thyroid Neoplasms/genetics
18.
Cancer Chemother Pharmacol ; 83(1): 107-113, 2019 01.
Article in English | MEDLINE | ID: mdl-30368586

ABSTRACT

PURPOSE: The ability of predicting severe adverse reactions caused by regorafenib is important. We evaluated regorafenib concentrations for adverse reaction risks and assessed the relevance of laboratory values and gene polymorphisms. METHODS: A total of 28 Japanese cancer patients who were treated with regorafenib were evaluated for the steady state of serum regorafenib concentrations and adverse reactions for 28 days. In addition, we determined the association of regorafenib concentrations with ABCG2 and OATP1B1 polymorphisms, which are regorafenib transporters. RESULTS: Regorafenib concentrations were significantly higher in the group with Grade 2 or higher total bilirubin elevation and thrombocytopenia compared with the group with grades 0 or 1 [3.45 (2.18-7.31) vs. 1.76 (0.26-2.77) µg/mL, P = 0.01 and 3.45 (2.12-7.31) vs. 1.76 (0.26-2.77) µg/mL, P = 0.02, respectively]. A strong association was noted between serum regorafenib concentrations and total bilirubin levels, but the physical and genetic factors predicting regorafenib pharmacokinetics could not be clarified. CONCLUSIONS: Regorafenib concentrations were associated with total bilirubin elevation and thrombocytopenia. Total serum bilirubin could be a useful marker when estimating regorafenib pharmacokinetics.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Colonic Neoplasms/drug therapy , Gastrointestinal Stromal Tumors/drug therapy , Liver-Specific Organic Anion Transporter 1/genetics , Neoplasm Proteins/genetics , Phenylurea Compounds/adverse effects , Phenylurea Compounds/blood , Polymorphism, Genetic , Pyridines/adverse effects , Pyridines/blood , Adult , Aged , Aged, 80 and over , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Follow-Up Studies , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Genotype , Humans , Male , Middle Aged , Prognosis , Prospective Studies
19.
Clin Pharmacol Ther ; 105(6): 1456-1461, 2019 06.
Article in English | MEDLINE | ID: mdl-30570132

ABSTRACT

Regorafenib exposure could potentially be influenced by an interaction with acid-reducing drugs. In this crossover trial, patients were randomized into two sequence groups consisting of three phases: regorafenib intake alone, regorafenib with concomitant esomeprazole, and regorafenib with esomeprazole 3 hours prior. The primary end point was the relative difference (RD) in geometric means for regorafenib 0-24-hour area under the concentration-time curve (AUC0-24h ) and was analyzed by a linear mixed model in 14 patients. AUC0-24h for regorafenib alone was 55.9 µg·hour/mL (coefficient of variance (CV): 40%), and for regorafenib with concomitant esomeprazole or with esomeprazole 3 hours prior AUC0-24h was 53.7 µg·hour/mL (CV: 34%) and 53.6 µg·hour/mL (CV: 43%), respectively. No significant differences were identified when regorafenib alone was compared with regorafenib with concomitant esomeprazole (RD: -3.9%; 95% confidence interval (CI): -20.5 to 16.1%; P = 1.0) or regorafenib with esomeprazole 3 hours prior (RD: -4.1%; 95% CI: -22.8 to 19.2%; P = 1.0). These findings indicate that regorafenib and esomeprazole can be safely combined in clinical practice.


Subject(s)
Colorectal Neoplasms/blood , Drug Interactions/physiology , Esomeprazole/blood , Phenylurea Compounds/blood , Proton Pump Inhibitors/blood , Pyridines/blood , Aged , Biological Availability , Colorectal Neoplasms/drug therapy , Cross-Over Studies , Esomeprazole/therapeutic use , Female , Humans , Male , Middle Aged , Phenylurea Compounds/therapeutic use , Proton Pump Inhibitors/therapeutic use , Pyridines/therapeutic use
20.
Mol Imaging ; 17: 1536012118795952, 2018.
Article in English | MEDLINE | ID: mdl-30251592

ABSTRACT

CEP-32496, also known as RXDX-105 or Agerafenib, is a new orally active inhibitor for the mutated v-raf murine sarcoma viral oncogene homolog B1 (BRAFV600E), which has attracted considerable attention in clinical trials for the treatment of human cancers. Here, we used carbon-11-labeled CEP-32496 ([11C]CEP-32496) as a positron emission tomography (PET) radiotracer to evaluate its pharmacokinetic properties and explore its potential for in vivo imaging. Following radiotracer synthesis, we performed in vitro binding assays and autoradiography of [11C]CEP-32496 in the A375 melanoma cell line and on tumor tissue sections from mice harboring the BRAFV600E mutation. These were followed by PET scans and biodistribution studies on nude mice bearing subcutaneous A375 cell-induced melanoma. [11C]CEP-32496 showed high binding affinity for BRAFV600E-positive A375 melanoma cells and densely accumulated in the respective tissue sections; this could be blocked by the BRAFV600E selective antagonist sorafenib and by unlabeled CEP-32496. The PET and biodistribution results revealed that [11C]CEP-32496 accumulated continuously but slowly into the tumor within a period of 0 to 60 minutes postinjection in A375-melanoma-bearing nude mice. Metabolite analysis showed high in vivo stability of [11C]CEP-32496 in plasma. Our results indicate that [11C]CEP-32496 has excellent specificity and affinity for the BRAFV600E mutation in vitro, while its noninvasive personalized diagnostic role needs to be studied further.


Subject(s)
Melanoma/genetics , Mutation/genetics , Phenylurea Compounds/pharmacokinetics , Proto-Oncogene Proteins B-raf/genetics , Quinazolines/pharmacokinetics , Animals , Autoradiography , Cell Line, Tumor , Humans , Lipids/chemistry , Melanoma/blood , Melanoma/urine , Mice, Nude , Phenylurea Compounds/blood , Phenylurea Compounds/chemistry , Phenylurea Compounds/urine , Quinazolines/blood , Quinazolines/chemistry , Quinazolines/urine , Tissue Distribution , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL