Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.083
Filter
1.
Pharmacol Res Perspect ; 12(3): e1202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38764241

ABSTRACT

The primary aim was to demonstrate bioequivalence between the 10/20 mg fixed-dose combination (FDC) of macitentan/tadalafil in a single tablet and the free combination of both drugs, and to evaluate the food effect on the 10/20 mg FDC in healthy participants. In this single-center, randomized, open-label, 3-way crossover, single-dose Phase 1 study in healthy adult participants, macitentan/tadalafil was administered as a 10/20 mg FDC formulation and compared with the free combination of macitentan and tadalafil. The food effect on the FDC was also evaluated. Pharmacokinetic sampling (216 h) was conducted. The 90% confidence intervals (CIs) for the geometric mean ratios of maximum observed plasma analyte concentration (Cmax) and area under the plasma analyte concentration-time curves (AUCs) for Treatment A (FDC, fasted) versus C (free combination, fasted) were within bioequivalence limits demonstrating that the FDC formulation can be considered bioequivalent to the free combination. The 90% CIs for the geometric mean ratios of Cmax and AUC for Treatment B (FDC, fed) versus A (FDC, fasted) were contained within bioequivalence limits demonstrating that there was no food effect. The administration of the 10/20 mg FDC was generally safe and well tolerated in healthy participants. This study demonstrated bioequivalence between the FDC of macitentan/tadalafil (10/20 mg) in a single tablet and the free combination of both drugs in healthy participants, and that the FDC can be taken without regard to food, similarly to the individual components. The FDC was generally safe and well tolerated.


Subject(s)
Area Under Curve , Cross-Over Studies , Drug Combinations , Food-Drug Interactions , Healthy Volunteers , Pyrimidines , Sulfonamides , Tablets , Tadalafil , Therapeutic Equivalency , Humans , Male , Adult , Pyrimidines/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/blood , Tadalafil/pharmacokinetics , Tadalafil/administration & dosage , Tadalafil/blood , Young Adult , Female , Sulfonamides/pharmacokinetics , Sulfonamides/administration & dosage , Sulfonamides/blood , Middle Aged , Administration, Oral , Fasting , Adolescent
2.
Pharmacol Res Perspect ; 9(5): e00846, 2021 10.
Article in English | MEDLINE | ID: mdl-34624174

ABSTRACT

The COVID-19 pandemic has forced clinical studies to accommodate imposed limitations. In this study, the bioequivalence part could not be conducted as planned. Thus, the aim was to demonstrate bioequivalence, using an adaptive study design, of tadalafil in fixed-dose combination (FDC) tablets of macitentan/tadalafil with single macitentan and tadalafil (Canadian-sourced) tablets and assess the effect of food on FDC tablets in healthy subjects. This Phase 1, single-center, open-label, single-dose, two-part, two-period, randomized, crossover study enrolled 62 subjects. Tadalafil bioequivalence as part of FDC of macitentan/tadalafil (10/40 mg) with single-component tablets of macitentan (10 mg) and tadalafil (40 mg) was determined by pharmacokinetic (PK) assessment under fasted conditions. The effect of food on FDC was evaluated under fed and fasted conditions. Fasted 90% confidence intervals (CIs) for geometric mean ratios (GMRs) were within bioequivalence limits for tadalafil and macitentan. Fed and fasted 90% CIs for area under the curve (AUC) GMR were within bioequivalence limits. However, 90% CIs for maximum plasma concentration (Cmax ) GMR for macitentan and tadalafil were outside bioequivalence limits. One FDC-treated subject experienced a serious adverse event of transient ischemic attack (bioequivalence part). To address pandemic-imposed limitations, an adaptive study design was implemented to demonstrate that the FDC tablet was bioequivalent to the free combination of macitentan and tadalafil (Canadian-sourced). No clinically significant differences in PK were determined between fed and fasted conditions; the FDC formulation could be taken irrespective of meals. The FDC formulation under fasted and fed conditions was well tolerated with no clinically relevant differences in safety profiles between the treatment groups. NCT Number: NCT04235270.


Subject(s)
COVID-19/epidemiology , Fasting/blood , Food-Drug Interactions/physiology , Pyrimidines/blood , Research Design , Sulfonamides/blood , Tadalafil/blood , Adult , COVID-19/prevention & control , Cross-Over Studies , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Pyrimidines/administration & dosage , Research Design/trends , Sulfonamides/administration & dosage , Tadalafil/administration & dosage , Therapeutic Equivalency , Young Adult
3.
J Pharm Pharm Sci ; 24: 127-136, 2021.
Article in English | MEDLINE | ID: mdl-34559626

ABSTRACT

PURPOSE: Therapeutic drug monitoring (TDM) is widely used in clinical practice to maximize drug efficacy and minimize toxicities. Currently, it is also practiced in the use of oral molecular targeted drugs. The objective of this study was to assess the clinical importance of measuring the systemic concentration of oral molecular targeted drugs used to treat renal cell carcinoma (RCC). METHODS: The systemic concentrations of the oral molecular targeted drugs sorafenib, sunitinib, axitinib, pazopanib, and everolimus used for RCC were useful for therapeutic interventions, and clinical outcomes were evaluated retrospectively. RESULTS: The interventional use of systemic drug concentration was confirmed in 26 of 87, and their categories are presented. The systemic concentration of sunitinib was useful in dose reduction and/or discontinuation (n = 10), dose escalation (n = 3), and adherence monitoring (n = 2). Nine of the 10 patients whose dose was reduced showed reduced adverse event. Two patients who were intervened in adherence monitor showed improved adherence. For axitinib, dose reduction and/or discontinuation (n = 1) and dose escalation (n = 6) were confirmed. For pazopanib, dose reduction and/or discontinuation (n = 1) and drug interaction detection (n = 1) were confirmed, both of them were confirmed to have reduced adverse events. For everolimus, dose reduction and/or discontinuation (n = 1) and drug interaction detection (n = 1) were confirmed, a patient with reduced dose recovered from adverse events. Interventions for sorafenib were not identified. CONCLUSIONS: This study demonstrated that systemic concentrations of oral molecular targeted drugs for RCC were considered to be clinically useful for dose adjustment, monitoring of treatment adherence, and the detection of drug interactions. Moreover, this information could be successfully used to guide individualized therapy to maximize the antitumor effects of these drugs.


Subject(s)
Antineoplastic Agents/blood , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Axitinib/administration & dosage , Axitinib/blood , Axitinib/therapeutic use , Everolimus/administration & dosage , Everolimus/blood , Everolimus/therapeutic use , Female , Humans , Indazoles/administration & dosage , Indazoles/blood , Indazoles/therapeutic use , Male , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/blood , Pyrimidines/therapeutic use , Sorafenib/administration & dosage , Sorafenib/blood , Sorafenib/therapeutic use , Sulfonamides/administration & dosage , Sulfonamides/blood , Sulfonamides/therapeutic use , Sunitinib/administration & dosage , Sunitinib/blood , Sunitinib/therapeutic use
4.
Cancer Chemother Pharmacol ; 88(6): 973-983, 2021 12.
Article in English | MEDLINE | ID: mdl-34505930

ABSTRACT

PURPOSE: Knowledge on Ruxolitinib exposure in patients with graft versus host disease (GvHD) is scarce. The purpose of this prospective study was to analyze Ruxolitinib concentrations of GvHD patients and to investigate effects of CYP3A4 and CYP2C9 inhibitors and other covariates as well as concentration-dependent effects. METHODS: 262 blood samples of 29 patients with acute or chronic GvHD who were administered Ruxolitinib during clinical routine were analyzed. A population pharmacokinetic model obtained from myelofibrosis patients was adapted to our population and was used to identify relevant pharmacokinetic properties and covariates on drug exposure. Relationships between Ruxolitinib exposure and adverse events were assessed. RESULTS: Median of individual mean trough serum concentrations was 39.9 ng/mL at 10 mg twice daily (IQR 27.1 ng/mL, range 5.6-99.8 ng/mL). Applying a population pharmacokinetic model revealed that concentrations in our cohort were significantly higher compared to myelofibrosis patients receiving the same daily dose (p < 0.001). Increased Ruxolitinib exposure was caused by a significant reduction in Ruxolitinib clearance by approximately 50%. Additional comedication with at least one strong CYP3A4 or CYP2C9 inhibitor led to a further reduction by 15% (p < 0.05). No other covariate affected pharmacokinetics significantly. Mean trough concentrations of patients requiring dose reduction related to adverse events were significantly elevated (p < 0.05). CONCLUSION: Ruxolitinib exposure is increased in GvHD patients in comparison to myelofibrosis patients due to reduced clearance and comedication with CYP3A4 or CYP2C9 inhibitors. Elevated Ruxolitinib trough concentrations might be a surrogate for toxicity.


Subject(s)
Cytochrome P-450 CYP2C9 Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Graft vs Host Disease/drug therapy , Nitriles/pharmacokinetics , Practice Patterns, Physicians'/statistics & numerical data , Primary Myelofibrosis/drug therapy , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Acute Disease , Adult , Aged , Chronic Disease , Cytochrome P-450 CYP2C9/chemistry , Cytochrome P-450 CYP3A/chemistry , Female , Follow-Up Studies , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Humans , Male , Metabolic Clearance Rate , Middle Aged , Nitriles/administration & dosage , Nitriles/blood , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Prognosis , Prospective Studies , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrimidines/administration & dosage , Pyrimidines/blood , Tissue Distribution , Young Adult
5.
Pharmacol Res ; 172: 105850, 2021 10.
Article in English | MEDLINE | ID: mdl-34450308

ABSTRACT

BACKGROUND AND PURPOSE: Pralsetinib is an FDA-approved oral small-molecule inhibitor for treatment of rearranged during transfection (RET) proto-oncogene fusion-positive non-small cell lung cancer. We investigated how the efflux transporters ABCB1 and ABCG2, the SLCO1A/1B uptake transporters and the drug-metabolizing enzyme CYP3A influence pralsetinib pharmacokinetics. EXPERIMENTAL APPROACH: In vitro, transepithelial pralsetinib transport was assessed. In vivo, pralsetinib (10 mg/kg) was administered orally to relevant genetically modified mouse models. Pralsetinib concentrations in cell medium, plasma samples and organ homogenates were measured using liquid chromatography-tandem mass spectrometry. KEY RESULTS: Pralsetinib was efficiently transported by human (h)ABCB1 and mouse (m)Abcg2, but not hACBG2. In vivo, mAbcb1a/1b markedly and mAbcg2 slightly limited pralsetinib brain penetration (6.3-and 1.8-fold, respectively). Testis distribution showed similar results. Abcb1a/1b;Abcg2-/- mice showed 1.5-fold higher plasma exposure, 23-fold increased brain penetration, and 4-fold reduced recovery of pralsetinib in the small intestinal content. mSlco1a/1b deficiency did not affect pralsetinib oral availability or tissue exposure. Oral coadministration of the ABCB1/ABCG2 inhibitor elacridar boosted pralsetinib plasma exposure (1.3-fold) and brain penetration (19.6-fold) in wild-type mice. Additionally, pralsetinib was a modest substrate of mCYP3A, but not of hCYP3A4, which did not noticeably restrict the oral availability or tissue distribution of pralsetinib. CONCLUSIONS AND IMPLICATIONS: SLCO1A/1B and CYP3A4 are unlikely to affect the pharmacokinetics of pralsetinib, but ABCG2 and especially ABCB1 markedly limit its brain and testis penetration, as well as oral availability. These effects are mostly reversed by oral coadministration of the ABCB1/ABCG2 inhibitor elacridar. These insights may be useful in the further clinical development of pralsetinib.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Organic Anion Transporters/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Pyrazoles/pharmacokinetics , Pyridines/pharmacokinetics , Pyrimidines/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/blood , Biological Availability , Brain/metabolism , Cytochrome P-450 CYP3A/genetics , Female , Male , Mice, Knockout , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Protein Kinase Inhibitors/blood , Pyrazoles/blood , Pyridines/blood , Pyrimidines/blood , Testis/metabolism
6.
Eur J Clin Pharmacol ; 77(12): 1887-1897, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34282472

ABSTRACT

PURPOSE: Pemigatinib (INCB054828), a potent and selective oral fibroblast growth factor receptor 1-3 inhibitor, is a Biopharmaceutical Classification System class II compound with good permeability and pH-dependent solubility that is predominantly metabolized by cytochrome P450 (CYP) 3A. Two drug-drug interaction studies, one with acid-reducing agents, esomeprazole (proton pump inhibitor [PPI]) and ranitidine (histamine-2 [H2] antagonist), and the other with potent CYP3A-modulating agents, itraconazole (CYP3A inhibitor) and rifampin (CYP3A inducer), were performed. METHODS: Both were open-label, fixed-sequence studies conducted in up to 36 healthy participants each, enrolled into two cohorts (n = 18 each). Pemigatinib plasma concentration was measured, and pharmacokinetic parameters were derived by non-compartmental analysis. RESULTS: There was an 88% and 17% increase in pemigatinib area under the plasma drug concentration-time curve (AUC) and maximum plasma drug concentration (Cmax), respectively, with itraconazole, and an 85% and 62% decrease in pemigatinib AUC and Cmax with rifampin coadministration. There was a 35% and 8% decrease in pemigatinib AUC and Cmax, respectively, with esomeprazole, and a 2% decrease in Cmax and 3% increase in AUC with ranitidine coadministration. In both studies, all adverse events reported were grade ≤ 2. CONCLUSION: Coadministration with itraconazole or rifampin resulted in a clinically significant change in pemigatinib exposure. Therefore, coadministration of strong CYP3A inducers with pemigatinib should be avoided, and the dose of pemigatinib should be reduced if coadministration with strong CYP3A inhibitors cannot be avoided. The effect of PPIs/H2 antagonists on pemigatinib exposure was modest, and pemigatinib can be administered without regard to coadministration of PPIs/H2 antagonists.


Subject(s)
Anti-Ulcer Agents/pharmacology , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Morpholines/pharmacokinetics , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Area Under Curve , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Healthy Volunteers , Humans , Metabolic Clearance Rate , Morpholines/adverse effects , Morpholines/blood , Pyrimidines/adverse effects , Pyrimidines/blood , Pyrroles/adverse effects , Pyrroles/blood
7.
Article in English | MEDLINE | ID: mdl-33957355

ABSTRACT

Sotorasib is a KRAS inhibitor with promising anticancer activity in phase I clinical studies. This compound is currently under further clinical evaluation as monotherapy and combination therapy against solid tumors. In this study, a liquid chromatography-tandem mass spectrometric method to quantify sotorasib in mouse plasma and eight tissue-related matrices (brain, liver, spleen, kidney, small intestine, small intestine content, lung, and testis homogenates) was developed and validated. Protein precipitation using acetonitrile was utilized in 96-well format to extract sotorasib and erlotinib (internal standard) from mouse plasma and tissue homogenates. Separation of the analytes was performed on an Acquity UPLC® BEH C18 column by gradient elution of methanol and 0.1% formic acid in water at a flow rate of 0.6 ml/min. Sotorasib was detected by a triple quadrupole mass spectrometer with positive electrospray ionization in selected reaction monitoring mode. A linear calibration range of 2-2,000 ng/ml of sotorasib was achieved during the validation. Accuracy values were in the range of 90.7-111.4%, and precision values (intra- and interday) were between 1.7% and 9.2% for all tested levels in all investigated matrices. The method was successfully applied to investigate the plasma pharmacokinetics and tissue accumulation of sotorasib in female wild-type mice.


Subject(s)
Chromatography, High Pressure Liquid/methods , Piperazines/blood , Pyridines/blood , Pyrimidines/blood , Tandem Mass Spectrometry/methods , Animals , Female , Linear Models , Mice , Piperazines/analysis , Piperazines/chemistry , Piperazines/pharmacokinetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Pyridines/analysis , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyrimidines/analysis , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
8.
Biopharm Drug Dispos ; 42(6): 263-284, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33904202

ABSTRACT

Renal (RIP) and hepatic (HIP) impairments are prevalent conditions in cancer patients. They can cause changes in gastric emptying time, albumin levels, hematocrit, glomerular filtration rate, hepatic functional volume, blood flow rates, and metabolic activity that can modify drug pharmacokinetics. Performing clinical studies in such populations has ethical and practical issues. Using predictive physiologically-based pharmacokinetic (PBPK) models in the evaluation of the PK of alectinib, ruxolitinib, and panobinostat exposures in the presence of cancer, RIP, and HIP can help in using optimal doses with lower toxicity in these populations. Verified PBPK models were customized under scrutiny to account for the pathophysiological changes induced in these diseases. The PBPK model-predicted plasma exposures in patients with different health conditions within average 2-fold error. The PBPK model predicted an area under the curve ratio (AUCR) of 1, and 1.8, for ruxolitinib and panobinostat, respectively, in the presence of severe RIP. On the other hand, the severe HIP was associated with AUCR of 1.4, 2.9, and 1.8 for alectinib, ruxolitinib, and panobinostat, respectively, in agreement with the observed AUCR. Moreover, the PBPK model predicted that alectinib therapeutic cerebrospinal fluid levels are achieved in patients with non-small cell lung cancer, moderate HIP, and severe HIP at 1-, 1.5-, and 1.8-fold that of healthy subjects. The customized PBPK models showed promising ethical alternatives for simulating clinical studies in patients with cancer, RIP, and HIP. More work is needed to quantify other pathophysiological changes induced by simultaneous affliction by cancer and RIP or HIP.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carbazoles/pharmacokinetics , Liver Diseases/blood , Models, Biological , Neoplasms/blood , Nitriles/pharmacokinetics , Panobinostat/pharmacokinetics , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Renal Insufficiency/blood , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/blood , Area Under Curve , Carbazoles/blood , Fasting/metabolism , Female , Humans , Liver Diseases/metabolism , Male , Middle Aged , Neoplasms/metabolism , Nitriles/blood , Panobinostat/blood , Piperidines/blood , Protein Kinase Inhibitors/blood , Pyrazoles/blood , Pyrimidines/blood , Renal Insufficiency/metabolism
9.
Biomed Chromatogr ; 35(8): e5124, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33772839

ABSTRACT

Recent studies have shown that therapeutic drug monitoring of tyrosine kinase inhibitors (TKIs) could improve treatment efficacy and safety. A simple analytical method using high-performance LC/electrospray ionization-tandem mass spectrometry has been developed and validated for simultaneous quantification of BCR-ABL and Bruton's TKIs used for chronic leukemia (imatinib, dasatinib, bosutinib, nilotinib, and ibrutinib) in human plasma. Although these structures and physical properties are similar, owing to their different linear ranges, simultaneously determining the plasma levels of these five TKIs by applying optimal MS parameters remains difficult. A quantitative range exceeding 60,000-fold was required, and the linear dynamic ranges of imatinib, bosutinib, and nilotinib were limited because of the presence of a saturated detection signal. In this study, we applied the in-source collision-induced dissociation technique to control the ion amounts in mass spectrometry. This new method allowed rapid determination within 5 min with simple pretreatment. The method was validated according to the US Food and Drug Administration guidelines. Moreover, all samples of patients with chronic leukemia were successfully measured and their values were within the linear range of measurement. Therefore, our high-throughput analytical system is useful to measure the plasma concentrations of imatinib, dasatinib, bosutinib, nilotinib, and ibrutinib in clinical practice.


Subject(s)
Chromatography, Liquid/methods , Drug Monitoring/methods , Protein Kinase Inhibitors/blood , Spectrometry, Mass, Electrospray Ionization/methods , Adenine/analogs & derivatives , Adenine/blood , Adenine/therapeutic use , Aniline Compounds/blood , Aniline Compounds/therapeutic use , Dasatinib/blood , Dasatinib/therapeutic use , Female , High-Throughput Screening Assays , Humans , Imatinib Mesylate/blood , Imatinib Mesylate/therapeutic use , Leukemia/drug therapy , Male , Middle Aged , Nitriles/blood , Nitriles/therapeutic use , Piperidines/blood , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/blood , Pyrimidines/therapeutic use , Quinolines/blood , Quinolines/therapeutic use , Tandem Mass Spectrometry/methods
10.
Invest New Drugs ; 39(5): 1306-1314, 2021 10.
Article in English | MEDLINE | ID: mdl-33742299

ABSTRACT

Background Brigatinib, a next-generation anaplastic lymphoma kinase (ALK) inhibitor, targets activated, mutant forms of ALK and overcomes mechanisms of resistance to the ALK inhibitors crizotinib, ceritinib, and alectinib. Brigatinib is approved in multiple countries for treatment of patients with ALK-positive non-small cell lung cancer. Based on population pharmacokinetic (PK) analyses, no dosage adjustment is required for patients with mild or moderate renal impairment. Methods An open-label, single-dose study was conducted to evaluate the PK of brigatinib (90 mg) in patients with severe renal impairment (estimated glomerular filtration rate < 30 mL/min/1.73 m2; n = 8) and matched healthy volunteers with normal renal function (estimated glomerular filtration rate ≥ 90 mL/min/1.73 m2; n = 8). Plasma and urine were collected for the determination of plasma protein binding and estimation of plasma and urine PK parameters. Results Plasma protein binding of brigatinib was similar between patients with severe renal impairment (92 % bound) and matched healthy volunteers with normal renal function (91 % bound). Unbound brigatinib exposure (area under the plasma concentration-time curve from time zero to infinity) was approximately 92 % higher in patients with severe renal impairment compared with healthy volunteers with normal renal function. The renal clearance of brigatinib in patients with severe renal impairment was approximately 20 % of that observed in volunteers with normal renal function. Conclusions These findings support a brigatinib dosage reduction of approximately 50 % in patients with severe renal impairment.Trial registry: Not applicable.


Subject(s)
Organophosphorus Compounds/pharmacokinetics , Pyrimidines/pharmacokinetics , Renal Insufficiency/metabolism , Aged , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Area Under Curve , Female , Glomerular Filtration Rate , Half-Life , Humans , Male , Metabolic Clearance Rate , Middle Aged , Organophosphorus Compounds/blood , Organophosphorus Compounds/urine , Patient Acuity , Protein Binding/physiology , Pyrimidines/blood , Pyrimidines/urine
11.
Curr Drug Metab ; 22(5): 399-410, 2021.
Article in English | MEDLINE | ID: mdl-33563190

ABSTRACT

BACKGROUND: Aprocitentan is an orally active, dual endothelin receptor antagonist that may offer a new therapeutic option for the treatment of difficult-to-control hypertension. OBJECTIVE: To investigate safety, tolerability, mass balance, absorption, distribution, metabolism, and excretion of aprocitentan. METHODS: In this single-center, open-label study, a single oral dose of 25 mg containing 3.7 MBq of 14C-radiolabeled aprocitentan was administered to 6 healthy male subjects. Metabolites were identified using mass spectrometry and, where possible, confirmed and quantified with reference compounds. RESULTS: Aprocitentan was well tolerated and there were no clinically significant findings for any safety variable. The geometric mean cumulative recovery of radioactivity from urine and feces over 14 days was 77% of the administered radioactive dose, with 52.1% cumulative recovery from urine and 24.8% from feces. Concentrations of total radioactivity in whole blood were markedly lower compared to plasma. In plasma, 94.3% of total radioactivity was aprocitentan. In urine and feces, 5 and 2, respectively (in feces one being aprocitentan) main products were identified. Metabolism data of aprocitentan identified two main elimination pathways, glucosidation to M3 and hydrolysis to M1, representing approximately 25% and 32% of the radioactive dose, respectively. CONCLUSIONS: Based on these metabolism data, aprocitentan can be concomitantly administered without dose adjustment with drugs that are inhibitors or inducers of any metabolizing enzyme, specifically cytochrome P450 enzymes.


Subject(s)
Endothelin Receptor Antagonists/metabolism , Endothelin Receptor Antagonists/pharmacology , Hypertension/drug therapy , Pyrimidines/metabolism , Pyrimidines/pharmacology , Sulfonamides/metabolism , Sulfonamides/pharmacology , Administration, Oral , Aged , Endothelin Receptor Antagonists/blood , Endothelin Receptor Antagonists/urine , Humans , Male , Middle Aged , Pyrimidines/blood , Pyrimidines/urine , Sulfonamides/blood , Sulfonamides/urine
12.
Biomed Chromatogr ; 35(4): e5043, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33283304

ABSTRACT

AMG 510 is the first-in-class KRASG12C inhibitor, currently in phase 2 clinical trials as an orphan drug to treat non-small cell lung cancer patients. We developed and validated a sensitive, selective, and high-throughput HPLC-MS/MS method for the quantitation of AMG 510 in mouse plasma per the regulatory guideline of the US Food and Drug and Administration. AMG 510 and the IS (MRTX-1257) were extracted from mouse plasma using tert-butyl methyl ether and chromatographed using an isocratic mobile phase (0.2% formic acid:acetonitrile; 25:75, v/v) at a flow rate of 0.65 mL/min on an Atlantis dC18 column. AMG 510 and the IS eluted at ~0.95 and 0.73 min, respectively. AMG 510 and the IS were detected by positive electrospray ionization in multiple reaction monitoring using transition pair (Q1 → Q3) m/z 561.1 → 134.1 and m/z 566.5 → 98.2, respectively. Excellent linearity was achieved in the concentration range of 1.08-5040 ng/mL (r > 0.0996). No matrix effect and carryover were observed. Intra- and inter-day accuracies and precisions were within the acceptance range. AMG 510 was demonstrated to be stable under the tested storage conditions. This novel method has been applied to a pharmacokinetic study in mice.


Subject(s)
Chromatography, High Pressure Liquid/methods , Piperazines/blood , Piperazines/pharmacokinetics , Pyridines/blood , Pyridines/pharmacokinetics , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Tandem Mass Spectrometry/methods , Animals , Linear Models , Male , Mice , Mice, Inbred BALB C , Piperazines/chemistry , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Pyridines/chemistry , Pyrimidines/chemistry , Reproducibility of Results , Sensitivity and Specificity
13.
Comb Chem High Throughput Screen ; 24(9): 1410-1416, 2021.
Article in English | MEDLINE | ID: mdl-33155888

ABSTRACT

AIM AND OBJECTIVE: An analytical method for the determination of mobocertinib, an investigational tyrosine kinase inhibitor, was developed and optimized by high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) in rat plasma. MATERIALS AND METHODS: Plasma samples were pretreated by the protein precipitation method with a methanol solution of osimertinib as the internal standard (IS). Chromatographic separation was performed using an Inertsil ODS-3 column (50 mm × 4.6 mm, I.D. 5 µm) with the temperature maintained at 40°C. The mobile phase consisted of water (containing 0.1% formic acid) and methanol in a gradient mode at a flow rate of 0.5 mL/min. Mass spectrometric detection was carried out in the selected reaction monitoring (SRM) mode with positive electrospray ionization, and the mass transitions of mobocertinib and osimertinib were m/z 587.01 → 71.88 and m/z 499.80 → 71.94, respectively. The method was validated in terms of selectivity, linearity, accuracy and precision, extraction recovery and matrix effect, stability and carryover as per the guidelines for bioanalytical method validation (FDA, 2018). The method was applied to the pharmacokinetic study of mobocertinib in rats by oral gavage at the doses of 2, 6, and 18 mg/kg. A total of 216 plasma samples from 18 rats were analyzed. RESULTS: The results showed good linearity over the range of 1-1000 ng/mL (R2 = 0.9957). The intra-batch accuracy was within 94.65-102.59% and the precision was within 5.49-10.46%. The inter-batch accuracy was within 97.08-102.25% with a precision of 7.54-10.13%. The extraction recovery and matrix factor were acceptable for the bioanalysis of mobocertinib. Additionally, mobocertinib was found to be stable under the detected conditions. Mobocertinib showed linear pharmacokinetic characteristics following oral administration to rats at 2.0-18.0 mg/kg. CONCLUSION: The developed and validated method was successfully employed in the pharmacokinetic study in rats following oral administration of mobocertinib at the doses of 2, 6, and 18 mg/kg.


Subject(s)
Aniline Compounds/blood , Aniline Compounds/pharmacokinetics , Indoles/blood , Indoles/pharmacokinetics , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Aniline Compounds/chemistry , Animals , Chromatography, Liquid , Indoles/chemistry , Male , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
14.
Biomed Chromatogr ; 35(2): e4981, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32895916

ABSTRACT

An LC-tandem mass spectrometry method was developed and validated for the simultaneous quantitation of fimasartan and sacubitrilat using positive ion mode. The protein precipitation method was employed for the extraction of fimasartan, sacubitrilat and alprazolam (internal standard) from rat heparinized plasma. Baseline separation of the analytes was accomplished using an ACE-5, C18 (4.6 × 50 mm) column and gradient elution of mobile phase A (5 mm ammonium formate and 0.1% formic acid in purified water) and B (acetonitrile:methanol, 80:20; v/v). All peaks of interest were eluted within a 5-min runtime. The quantitation was achieved in the selected reaction monitoring mode. The developed method was validated as per US Food and Drug Administration guidelines and met the pre-defined acceptance criteria. The method showed linearity from 5 to 10,000 ng/mL. The accuracy/precision of intra- and inter-batch assays was 96.64%/2.05% to 109.17%/13.70% and 100.74%/3.76% to 106.39%/9.75% for fimasartan and 100.02%/1.49% to 113.80%/9.38% and 100.75%/2.31% to 108.40%/7.74% for sacubitrilat, respectively, in rat plasma. Fimasartan and sacubitrilat remained stable in rat plasma at different experimental conditions up to 21 days. The developed method was sensitive, selective and applied successfully to monitor plasma concentrations of fimasartan and sacubitrilat in an oral rat pharmacokinetic study.


Subject(s)
Aminobutyrates/blood , Biphenyl Compounds/blood , Chromatography, Liquid/methods , Pyrimidines/blood , Tandem Mass Spectrometry/methods , Tetrazoles/blood , Aminobutyrates/chemistry , Aminobutyrates/pharmacokinetics , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Linear Models , Male , Prodrugs , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/methods , Tetrazoles/chemistry , Tetrazoles/pharmacokinetics
15.
J Acquir Immune Defic Syndr ; 86(4): e90-e96, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33278181

ABSTRACT

BACKGROUND: There continues to be a need for HIV prevention options that women can initiate and use autonomously. The dapivirine vaginal ring (VR) has been shown to have a favorable safety profile and reduce the risk of HIV-1 acquisition. We report on women's experiences with VR adherence during the MTN-025/HIV Open-label Prevention Extension (HOPE) study and responses to Residual Drug Level (RDL) results. SETTING: Ten women at each of the 6 HOPE research sites in Lilongwe, Malawi; Durban (2 sites) and Johannesburg, South Africa; Kampala, Uganda; and Chitungwiza, Zimbabwe, were randomly selected (n = 60). METHODS: After confirmation of eligibility criteria, in-depth interviews were conducted where available RDL results were presented. RESULTS: Many women with low RDL release measurements deflected blame onto other factors (the ring, the drug, and faulty testing machines) and distrust of the testing method. The disclosure of RDL results enabled some users to discuss their challenges experienced (fear of partner objections, perceived side effects, and removals during menses). Consistent users reported important motivators (support from others, protection from HIV, and enhanced sexual experiences from the VR). CONCLUSION: The VR provided a sense of security for some women; however, adherence was still challenging for others regardless of it being a female controlled, long-acting HIV prevention technology. Adherence measurements may not be sustainable in the real-world implementation of the VR, although they can be seen as a benefit as they provide a better understanding of actual product use and provide women with a platform to discuss their experiences.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1 , Medication Adherence , Pyrimidines/therapeutic use , Administration, Intravaginal , Adult , Africa South of the Sahara/epidemiology , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/blood , Contraceptive Devices, Female , HIV Infections/epidemiology , Humans , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/blood
16.
Drug Metab Dispos ; 49(1): 31-38, 2021 01.
Article in English | MEDLINE | ID: mdl-33144331

ABSTRACT

Lemborexant is a novel dual orexin receptor antagonist recently approved for the treatment of insomnia in the United States and Japan. Here, disposition and metabolic profiles were investigated in healthy human subjects. After single oral administration of 10 mg [14C]lemborexant (100 µCi), plasma concentrations of lemborexant and radioactivity peaked at 1 hour postdose and decreased biphasically. Cumulative recovery of the administered radioactivity within 480 hours was 86.5% of the dose, with 29.1% in urine and 57.4% in feces. Unchanged lemborexant was not detected in urine but accounted for 13.0% of the dose in feces, suggesting that the main elimination pathway of lemborexant was metabolism. Metabolite analyses revealed that the major metabolic pathways of lemborexant are oxidation of the dimethylpyrimidine moiety and subsequent further oxidation and/or glucuronidation. In plasma, lemborexant was the dominant component, accounting for 26.5% of total drug-related exposure. M4, M9, M10, and M18 were detected as the major radioactive components; M10 was the only metabolite exceeding 10% of total drug-related exposure. Although M4, M9, and M10 showed binding affinity for orexin receptors comparable to that of lemborexant, their contributions to the sleep-promoting effects of lemborexant are likely low because of the limited brain penetration by P-glycoprotein. Exposure comparison between humans and nonclinical toxicology species confirmed that plasma exposure of M10 was higher in at least one animal species compared with that in humans, indicating that there is no disproportionate metabolite in humans, as defined by International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use M3(R2) and U.S. Food and Drug Administration Metabolite in Safety Testing guidance; therefore, no additional toxicology studies are needed. SIGNIFICANCE STATEMENT: This study provides detailed data of the disposition and metabolism of lemborexant, a novel therapeutic drug for insomnia, in humans, as well as a characterization of the circulating metabolites and assessment of their contributions to efficacy and safety. The information presented herein furthers our understanding of the pharmacokinetic profiles of lemborexant and its metabolites and will promote the safe and effective use of lemborexant in the clinic.


Subject(s)
Drug Monitoring/methods , Pyridines , Pyrimidines , Sleep Initiation and Maintenance Disorders/drug therapy , Administration, Oral , Healthy Volunteers , Humans , Metabolic Networks and Pathways , Orexin Receptor Antagonists/blood , Orexin Receptor Antagonists/pharmacokinetics , Pharmacokinetics , Pyridines/blood , Pyridines/pharmacokinetics , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Radioactivity
17.
APMIS ; 129(3): 128-137, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33155332

ABSTRACT

We aimed to use serum metabolomics to discriminate infants with severe respiratory syncytial virus (RSV) bronchiolitis who later developed subsequent recurrent wheezing from those who did not and to investigate the relationship between serum metabolome and host immune responses with regard to the subsequent development of recurrent wheezing. Fifty-one infants who were hospitalized during an initial episode of severe RSV bronchiolitis at 6 months of age or less were included and followed for up to the age of 3 years. Of them, 24 developed subsequent recurrent wheezing and 27 did not. Untargeted serum metabolomics was performed by ultraperformance liquid chromatography coupled with high-resolution mass spectrometry (UPLC-MS/MS). Cytokines were measured by multiplex immunoassay. Difference in serum metabolomic profiles was observed between infants who developed recurrent wheezing and those who did not. L-lactic acid level was significantly higher in infants with recurrent wheezing than those without. Pyrimidine metabolism, glycerophospholipid metabolism, and arginine biosynthesis were identified as the most significant changed pathways between the two groups. Moreover, L-lactic acid level was positively associated with serum CXCL8 level. This exploratory study showed that differential serum metabolic signatures during severe RSV bronchiolitis in early infancy were associated with the development of subsequent recurrent wheezing in later childhood.


Subject(s)
Bronchiolitis/complications , Respiratory Sounds/etiology , Respiratory Syncytial Virus Infections/blood , Respiratory Syncytial Virus Infections/complications , Serum/chemistry , Bronchiolitis/blood , Bronchiolitis/virology , Child, Preschool , Chromatography, High Pressure Liquid , Cytokines/blood , Female , Follow-Up Studies , Humans , Infant , Interleukin-8/blood , Lactic Acid/blood , Male , Metabolomics , Pyrimidines/blood , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Tandem Mass Spectrometry
18.
Ther Drug Monit ; 43(3): 416-421, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33009287

ABSTRACT

BACKGROUND: Pazopanib is widely used to treat renal cell carcinomas and soft tissue tumors in Japan. Pazopanib has significant therapeutic efficacy but it is associated with frequent severe adverse effects. Therapeutic drug monitoring (TDM) may help to prevent adverse effects. A more convenient and rapid pazopanib assay is desirable for the application of TDM in clinical settings. In this study, the authors developed a high-throughput method for quantifying pazopanib in human plasma using ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). METHODS: After a simple solid-phase extraction step using a 96-well plate, pazopanib was analyzed by UHPLC-MS/MS in the positive electrospray ionization mode. RESULTS: The novel method fulfilled the requirements of the US Food and Drug Administration and the European Medicines Agency guidelines for assay validation, and the lower limit of quantification was 0.5 mcg/mL. The calibration curves were linear over the concentration range of 0.5-100 mcg/mL. The average recovery rate was 102.0% ± 3.9% (mean ± SD). The precision was below 5.0%, and the accuracy was within 12.0% for all quality control levels. Matrix effect varied between 90.9% and 97.1%. This assay was successfully applied to TDM of pazopanib trough concentrations in 3 patients treated with the drug for soft tissue tumors. CONCLUSIONS: The authors succeeded in developing a novel high-throughput UHPLC-MS/MS method for quantifying pazopanib in human plasma. This method can be applied to TDM of patients receiving pazopanib in clinical settings.


Subject(s)
Drug Monitoring , Indazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Soft Tissue Neoplasms , Sulfonamides/pharmacokinetics , Chromatography, High Pressure Liquid , Humans , Indazoles/blood , Pyrimidines/blood , Reproducibility of Results , Soft Tissue Neoplasms/drug therapy , Sulfonamides/blood , Tandem Mass Spectrometry
19.
CPT Pharmacometrics Syst Pharmacol ; 10(1): 40-47, 2021 01.
Article in English | MEDLINE | ID: mdl-33200566

ABSTRACT

Selection of a personalized dose for an individual patient can be informed by the patient's preferences, translated as weights on each of the clinically relevant safety and efficacy drug attributes, based on results from a brief patient preference elicitation questionnaire. In this analysis, the weighted attributes were simulated to represent various endometriosis patient profiles. Exposure-response simulations were performed for elagolix, a drug approved for management of moderate to severe pain associated with endometriosis, across a range of plasma exposures corresponding to a range of doses. The results were combined to calculate a personalized clinical utility index. An interactive user-friendly online application was developed and envisioned as a physician's desk tool to personalize the dose selection process based on individual patient preferences. This demonstration should serve as an example of how patient/physician conversation can be facilitated with quantitative tools for personalizing the dose.


Subject(s)
Analgesics/administration & dosage , Endometriosis/drug therapy , Hydrocarbons, Fluorinated/administration & dosage , Models, Biological , Pain/drug therapy , Patient Preference , Pyrimidines/administration & dosage , Adolescent , Adult , Analgesics/blood , Analgesics/pharmacokinetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase III as Topic , Computer Simulation , Dose-Response Relationship, Drug , Endometriosis/complications , Endometriosis/metabolism , Female , Humans , Hydrocarbons, Fluorinated/blood , Hydrocarbons, Fluorinated/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/genetics , Middle Aged , Pain/blood , Pain/etiology , Precision Medicine , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Surveys and Questionnaires , Young Adult
20.
Clin Pharmacol Ther ; 109(5): 1334-1341, 2021 05.
Article in English | MEDLINE | ID: mdl-33141923

ABSTRACT

Firsocostat (FIR: previously GS-0976), a highly sensitive OATP substrate, reduces hepatic de novo lipogenesis (DNL) by inhibiting acetyl-CoA carboxylases (ACC). Measuring the pharmacodynamic (PD) efficacy of FIR on DNL provides a unique opportunity to determine optimal dosing strategies for liver-targeted OATP substrates in settings of altered OATP function. A randomized, four-way crossover drug-drug interaction study was conducted. Hepatic DNL, a marker for ACC activity, was measured in 28 healthy volunteers after reference, single dose FIR 10 mg, FIR 10 mg plus the OATP inhibitor rifampin (RIF) 300 mg i.v., or RIF 300 mg i.v. (control for DNL effect of RIF), each separated by a 7-day washout. Samples were collected for pharmacokinetic (PK) and PD assessments through 24 hours after each treatment. Hepatic DNL and its inhibition by FIR were assessed. Twenty-four subjects completed the study. All adverse events were mild. RIF alone increased hepatic DNL area under the effect curve from time of administration up to the time of the last quantifiable concentration (AUEClast ; 35.7%). Despite a 5.2-fold increase in FIR plasma exposure (area under the concentration-time curve from zero to infinity (AUCinf )) when administered with RIF, FIR alone, and FIR + RIF had the same hepatic PD effect, 37.1% and 34.9% reduction in DNL AUEClast , respectively, compared with their respective controls. These findings indicate that large decreases in OATP activity do not alter hepatic intracellular exposure (as inferred by no change in PD) for drugs that are primarily eliminated hepatically and permeability rate-limited, such as FIR. These results support PK theory that has been difficult to test and provide practical guidance on administration of liver-targeted drugs in settings of reduced OATP function.


Subject(s)
Isobutyrates/pharmacokinetics , Liver/drug effects , Organic Anion Transporters/antagonists & inhibitors , Oxazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Adult , Drug Interactions , Female , Humans , Isobutyrates/administration & dosage , Isobutyrates/adverse effects , Isobutyrates/blood , Liver/metabolism , Male , Middle Aged , Oxazoles/administration & dosage , Oxazoles/adverse effects , Oxazoles/blood , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/blood , Rifampin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...