Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 904
Filter
1.
J Med Chem ; 67(11): 9355-9373, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38805667

ABSTRACT

Development of more efficacious medications with improved safety profiles to manage and treat multiple forms of pain is a critical element of healthcare. To this end, we have designed and synthesized a novel class of tetracyclic pyridopyrroloquinoxalinone derivatives with analgesic properties. The receptor binding profiles and analgesic properties of these tetracyclic compounds were studied. Systematic optimizations of this novel scaffold culminated in the discovery of the clinical candidate, (6bR,10aS)-8-[3-(4-fluorophenoxy)propyl]-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3',4':4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one (compound 5, ITI-333), which exhibited potent binding affinity to serotonin 5-HT2A (Ki = 8.3 nM) and µ-opioid receptors (MOR, Ki = 11 nM) and moderate affinity to adrenergic α1A (Ki = 28 nM) and dopamine D1 (Ki = 50 nM) receptors. ITI-333 acts as a 5-HT2A receptor antagonist, a MOR partial agonist, and an adrenergic α1A receptor antagonist. ITI-333 exhibited dose-dependent analgesic effects in rodent models of acute pain. Currently, this investigational new drug is in phase I clinical development.


Subject(s)
Analgesics , Pain , Animals , Humans , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/chemical synthesis , Analgesics/therapeutic use , Structure-Activity Relationship , Administration, Oral , Pain/drug therapy , Mice , Male , Rats , Drug Discovery , Rats, Sprague-Dawley , Biological Availability , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/agonists , Pyridines/chemistry , Pyridines/pharmacology , Pyridines/chemical synthesis , Pyridines/therapeutic use , Pyridines/pharmacokinetics , Pyrroles/chemistry , Pyrroles/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacokinetics
2.
Biomed Chromatogr ; 38(6): e5860, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38558021

ABSTRACT

A liquid chromatography-tandem mass spectrometry method with vonoprazan fumarate-d4 as a stable isotope-labeled internal standard was developed and validated aiming at quantification of vonoprazan fumarate in human plasma for a bioequivalence study. Chromatographic separation was achieved by acetonitrile one-step protein precipitation using a gradient elution of 0.1% formic acid aqueous solution and acetonitrile with a run time of 3.65 min. Detection was carried out on a tandem mass spectrometer in multiple reaction monitoring mode via a positive electrospray ionization interface. The multiple reaction monitoring mode of precursor-product ion transitions for vonoprazan fumarate and vonoprazan fumarate-d4 were m/z 346.0 → 315.1 and 350.0 → 316.0, respectively. The linear range was 0.150-60.000 ng/ml. This method was fully validated with acceptable results in terms of selectivity, carryover, lower limit of quantification, calibration curve, accuracy, precision, dilution effect, matrix effect, stability, recovery and incurred sample reanalysis. A successful application of this method was realized in the bioequivalence study of vonoprazan fumarate tablet (20 mg) among healthy Chinese volunteers.


Subject(s)
Pyrroles , Sulfonamides , Tandem Mass Spectrometry , Therapeutic Equivalency , Humans , Tandem Mass Spectrometry/methods , Sulfonamides/blood , Sulfonamides/pharmacokinetics , Sulfonamides/chemistry , Pyrroles/pharmacokinetics , Pyrroles/blood , Pyrroles/chemistry , Reproducibility of Results , Linear Models , Chromatography, Liquid/methods , Limit of Detection , Male , Adult , Liquid Chromatography-Mass Spectrometry
3.
Curr Med Chem ; 31(22): 3426-3435, 2024.
Article in English | MEDLINE | ID: mdl-38310397

ABSTRACT

BACKGROUND: Tofacitinib is an oral JAK inhibitor for the treatment of rheumatoid arthritis (RA). The clinical efficacy and safety of an administered tofacitinib, either monotherapy or in combination with conventional synthetic disease-modifying anti-rheumatic drugs, mainly methotrexate (MTX), have been evaluated. The high plasma concentration with delayed medicine clearance may affect the liver and/or kidney functions. In this study, an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC- MS/MS) method for the quantitative analysis of methotrexate, tofacitinib, and metabolite M9 in plasma of Sprague Dawley (SD) rats was developed, and its effectiveness was validated as well. METHODS: Methotrexate, tofacitinib, M9 and fedratinib (internal standard, IS) were separated by gradient elution. The chromatography was performed on an Acquity BEH C18 (2.1 mm × 50 mm, 1.7 µm) column with the mobile phases of acetonitrile and 0.1% formic acid aqueous solution with different proportions at the flow rate of 0.30 mL/min. In the positive ionization mode, the analyzes were detected using a Xevo TQ-S triple quadrupole tandem mass spectrometer, with the following mass transition pairs: m/z 313.12 → 148.97 for tofacitinib, m/z 329.10 → 165.00 for M9 and m/z 455.12 → 308.05 for methotrexate. RESULTS: The obtained results manifested good calibration linearity over the ranges of tofacitinib at 0.1-100 ng/mL, M9 at 0.05-100 ng/mL, and methotrexate at 0.05-100 ng/mL. The lower limit of quantifications (LLOQs) of methotrexate, tofacitinib and M9 were 0.05 ng/mL, 0.1 ng/mL and 0.05 ng/mL, respectively. Intra-day and inter-day accuracy values were confirmed with a range of -6.3% to 12.7%, while intra-day and inter-- day precision values were ≤14.4%. Additionally, recoveries were greater than 86.5% for each compound without significant matrix effects. CONCLUSION: The currently established analytical method exhibited great potential for the evaluation of plasma concentrations of methotrexate, tofacitinib and M9 simultaneously, greatly reducing the detection time, which would serve as a supplementary role in formulating dose decisions to achieve personalized treatment, identify drugs that cause adverse reactions and finally, to assess drug-drug interactions on clinical studies.


Subject(s)
Antirheumatic Agents , Methotrexate , Piperidines , Pyrimidines , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Animals , Methotrexate/pharmacokinetics , Pyrimidines/pharmacokinetics , Pyrimidines/chemistry , Pyrimidines/blood , Tandem Mass Spectrometry/methods , Antirheumatic Agents/therapeutic use , Antirheumatic Agents/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Rats , Male , Pyrroles/pharmacokinetics , Pyrroles/blood , Pyrroles/chemistry , Liquid Chromatography-Mass Spectrometry
4.
J Cyst Fibros ; 23(3): 549-553, 2024 May.
Article in English | MEDLINE | ID: mdl-38290918

ABSTRACT

BACKGROUND: The use of elexacaftor/tezacaftor/ivacaftor (ETI) in people with cystic fibrosis (pwCF) after solid organ transplantation is controversial because of potential drug-drug interactions (DDI) with tacrolimus. We aimed to improve insight into the safety and clinical benefits of co-administration of ETI and tacrolimus in liver or kidney transplanted adult pwCF. METHODS: In 5 pwCF, tacrolimus concentrations were monitored during 2 weeks before and 4 weeks after starting ETI treatment. Trough levels, area under the curve (AUC) and clinical effect of ETI were investigated. During the study (6 weeks in total) adverse events were monitored. RESULTS: The DDI between tacrolimus and ETI resulted in an increased exposure of tacrolimus in all subjects, the dose adjusted AUC0-24h was 1.79 (median) times higher at the end of the study. Five dose adjustments were performed in 4 subjects in order to attain tacrolimus target range. No adverse events were reported and all subjects showed clinical improvement during ETI treatment. CONCLUSION: The clinical value of ETI treatment in kidney and liver transplanted pwCF is clear. The use of ETI may increase tacrolimus levels moderately. Therefore, we recommend close monitoring of tacrolimus trough levels in patients who start ETI.


Subject(s)
Aminophenols , Benzodioxoles , Cystic Fibrosis , Drug Interactions , Immunosuppressive Agents , Indoles , Kidney Transplantation , Liver Transplantation , Quinolones , Tacrolimus , Humans , Cystic Fibrosis/surgery , Cystic Fibrosis/drug therapy , Tacrolimus/administration & dosage , Tacrolimus/pharmacokinetics , Tacrolimus/adverse effects , Male , Female , Adult , Benzodioxoles/adverse effects , Benzodioxoles/administration & dosage , Benzodioxoles/therapeutic use , Quinolones/administration & dosage , Quinolones/adverse effects , Quinolones/pharmacokinetics , Liver Transplantation/methods , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacokinetics , Kidney Transplantation/adverse effects , Aminophenols/administration & dosage , Aminophenols/adverse effects , Aminophenols/pharmacokinetics , Aminophenols/therapeutic use , Indoles/administration & dosage , Indoles/adverse effects , Indoles/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Drug Combinations , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Pyridines/adverse effects , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Pyrroles/adverse effects , Quinolines/administration & dosage , Quinolines/adverse effects , Quinolines/pharmacokinetics , Young Adult , Drug Monitoring/methods , Pyrrolidines
5.
Clin Pharmacol Drug Dev ; 13(6): 688-695, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38261231

ABSTRACT

Ubrogepant is a calcitonin gene-related peptide receptor antagonist indicated for the acute treatment of migraine with or without aura in adults. The objectives of this study were to evaluate (1) single-dose pharmacokinetics (PK) and dose proportionality of ubrogepant in Japanese participants, (2) the safety and tolerability of ubrogepant in healthy Japanese and White participants, and (3) to compare the PK of ubrogepant in Japanese versus White participants. A total of 48 participants were enrolled into 4 cohorts (N = 12 [9 active + 3 placebo] per cohort). A single dose was administered on Day 1 following an overnight fast to assess the PK of ubrogepant at 3 dose levels and test dose proportionality between 25 and 100 mg in Japanese participants. White participants were randomly assigned to ubrogepant (100 mg) or placebo. Dose proportionality was observed in the dose range of 25-100 mg in Japanese participants. Systemic exposure was 20% lower in Japanese participants as compared with White participants, but this difference is unlikely to be clinically relevant. Single doses of ubrogepant (25-100 mg) had a safety profile similar to placebo, and no differences in the safety profile of ubrogepant 100 mg were observed between Japanese versus White participants.


Subject(s)
Asian People , Calcitonin Gene-Related Peptide Receptor Antagonists , Dose-Response Relationship, Drug , Pyridines , White People , Humans , Male , Adult , Female , Pyridines/pharmacokinetics , Pyridines/adverse effects , Pyridines/administration & dosage , Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacokinetics , Calcitonin Gene-Related Peptide Receptor Antagonists/adverse effects , Calcitonin Gene-Related Peptide Receptor Antagonists/administration & dosage , Double-Blind Method , Young Adult , Pyrroles/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/adverse effects , Middle Aged , Healthy Volunteers , Japan , East Asian People
6.
Clin Pharmacol Drug Dev ; 12(12): 1142-1155, 2023 12.
Article in English | MEDLINE | ID: mdl-37909052

ABSTRACT

Saroglitazar magnesium, a dual peroxisome proliferator-activated receptor agonist, is under evaluation for treating various liver conditions. While the pharmacokinetics (PK) of saroglitazar have been extensively studied in diverse preclinical models and healthy subjects, a comprehensive assessment of its PK behavior under conditions of hepatic impairment is lacking. In this Phase 1, open-label, parallel-group study, the PK of a single dose of 4-mg saroglitazar magnesium was investigated in subjects having varying degrees of hepatic impairment with and without portal hypertension compared with appropriately matched individuals having normal hepatic function. Treatment-emergent adverse events for safety were also evaluated. Thirty-two subjects were enrolled in the hepatic-impaired groups and 23 subjects in the normal hepatic function group. Mild and moderate hepatic impairment did not significantly affect the PK of saroglitazar, compared with normal hepatic function. Although severe hepatic impairment did not alter maximum observed plasma concentration and half-life; saroglitazar exposure (area under the plasma concentration-time curve from time 0 to infinity) increased 3-fold, while the clearance was 61% lower compared to the subjects with normal hepatic function. This may require close monitoring or dose adjustments in individuals with severe hepatic impairment. A single oral dose of saroglitazar magnesium 4 mg was found to be safe and well tolerated in subjects with varying degrees of hepatic function.


Subject(s)
Liver Diseases , Phenylpropionates , Humans , Area Under Curve , Liver Diseases/drug therapy , Phenylpropionates/pharmacokinetics , Pyrroles/pharmacokinetics
7.
Molecules ; 28(14)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37513351

ABSTRACT

Secure and efficient treatment of diverse pain and inflammatory disorders is continually challenging. Although NSAIDs and other painkillers are well-known and commonly available, they are sometimes insufficient and can cause dangerous adverse effects. As yet reported, derivatives of pyrrolo[3,4-d]pyridazinone are potent COX-2 inhibitors with a COX-2/COX-1 selectivity index better than meloxicam. Considering that N-acylhydrazone (NAH) moiety is a privileged structure occurring in many promising drug candidates, we decided to introduce this pharmacophore into new series of pyrrolo[3,4-d]pyridazinone derivatives. The current paper presents the synthesis and in vitro, spectroscopic, and in silico studies evaluating the biological and physicochemical properties of NAH derivatives of pyrrolo[3,4-d]pyridazinone. Novel compounds 5a-c-7a-c were received with high purity and good yields and did not show cytotoxicity in the MTT assay. Their COX-1, COX-2, and 15-LOX inhibitory activities were estimated using enzymatic tests and molecular docking studies. The title N-acylhydrazones appeared to be promising dual COX/LOX inhibitors. Moreover, spectroscopic and computational methods revealed that new compounds form stable complexes with the most abundant plasma proteins-AAG and HSA, but do not destabilize their secondary structure. Additionally, predicted pharmacokinetic and drug-likeness properties of investigated molecules suggest their potentially good membrane permeability and satisfactory bioavailability.


Subject(s)
Cyclooxygenase Inhibitors , Hydrazones , Lipoxygenase Inhibitors , Pyridazines , Pyrroles , Hydrazones/chemical synthesis , Hydrazones/chemistry , Hydrazones/pharmacokinetics , Hydrazones/pharmacology , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacokinetics , Cyclooxygenase Inhibitors/pharmacology , Pyridazines/chemical synthesis , Pyridazines/chemistry , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Humans , Fibroblasts , Computer Simulation , Cell Membrane Permeability , Cell Line
8.
Article in English | MEDLINE | ID: mdl-37196527

ABSTRACT

Metarrestin is a first-in-class small molecule inhibitor targeting the perinucleolar compartment, a subnuclear body associated with metastatic capacity. Promising preclinical results led to the clinical translation of the compound into a first-in-human phase I trial (NCT04222413). To characterize metarrestin's pharmacokinetic profile in humans, a uHPLC-MS/MS assay was developed and validated to determine the disposition of the drug in human plasma. Efficient sample preparation was accomplished through one-step protein precipitation paired with elution through a phospholipid filtration plate. Chromatographic separation was achieved with gradient elution through an Acuity UPLC® BEH C18 column (50 × 2.1 mm, 1.7 µm). Tandem mass spectrometry facilitated the detection of metarrestin and tolbutamide, the internal standard. The effective calibration range spanned 1-5000 ng/mL and was both accurate (range -5.9 % to 4.9 % deviation) and precise (≤9.0 %CV). Metarrestin proved stable (≤4.9 % degradation) under various assay-imposed conditions. Matrix effects, extraction efficiency, and process efficiency were assessed. Further, the assay was successfully able to determine the disposition of orally administered metarrestin in patients from the lowest dose cohort (1 mg) for 48 h post-administration. Thus, the validated analytical method detailed in this work is simple, sensitive, and clinically applicable.


Subject(s)
Pyrimidines , Tandem Mass Spectrometry , Humans , Tandem Mass Spectrometry/methods , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Reproducibility of Results
9.
Ther Drug Monit ; 45(6): 817-822, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37074815

ABSTRACT

BACKGROUND: Sunitinib (SUN) malate is an oral, multitargeted, tyrosine kinase inhibitor approved for the treatment of metastatic renal cell carcinoma, imatinib-resistant or imatinib-intolerant gastrointestinal stromal tumors, and pancreatic neuroendocrine tumors. SUN has a narrow therapeutic window and high variability in interpatient pharmacokinetic parameters. Clinical detection methods for SUN and N -desethyl SUN limit the application of SUN to therapeutic drug monitoring. All published methods for quantifying SUN in human plasma require strict light protection to avoid light-induced isomerism or the use of additional quantitative software. To avoid these difficult processes in clinical routines, the authors propose a novel method that merges the peaks of the E -isomer and Z -isomer of SUN or N -desethyl SUN into a single peak. METHODS: The E -isomer and Z -isomer peaks of SUN or N -desethyl SUN were merged into a single peak by optimizing the mobile phases to decrease the resolution of the isomers. A suitable chromatographic column was selected to obtain a good peak shape. Thereafter, the conventional and single-peak methods (SPM) were simultaneously validated and compared according to the guidelines published by the Food and Drug Administration in 2018 and the Chinese Pharmacopoeia in 2020. RESULTS: The verification results showed that the SPM was superior to the conventional method in the matrix effect and met the requirements for biological sample analysis. SPM was then applied to detect the total steady-state concentration of SUN and N -desethyl SUN in tumor patients who received SUN malate. CONCLUSIONS: The established SPM makes the detection of SUN and N -desethyl SUN easier and faster without light protection or extra quantitative software, making it more appropriate for routine clinical use. The clinical application results showed that 12 patients took 37.5 mg per day, with a median total trough steady-state concentration of 75.0 ng/mL.


Subject(s)
Antineoplastic Agents , Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Sunitinib/therapeutic use , Carcinoma, Renal Cell/drug therapy , Chromatography, Liquid/methods , Liquid Chromatography-Mass Spectrometry , Imatinib Mesylate/therapeutic use , Drug Monitoring , Malates/therapeutic use , Kidney Neoplasms/drug therapy , Tandem Mass Spectrometry/methods , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use , Antineoplastic Agents/therapeutic use
10.
Cancer Chemother Pharmacol ; 90(5): 409-415, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36107220

ABSTRACT

BACKGROUND: Famitinib is an oral, small-molecule, multi-targeted tyrosine kinase inhibitor under clinical investigation for the treatment of solid tumors. As famitinib is metabolized mainly by cytochrome P450 3A4 (CYP3A4), the study was conducted to investigate the effect of potent CYP3A4 inducer rifampin on the pharmacokinetics of famitinb. METHODS: This single-center, single-arm and fixed-sequence drug-drug interaction study enrolled 21healthy Chinese male subjects. Subjects received a single oral dose of famitinib 25 mg on days 1 and 16 and repeated administration of oral rifampin 600 mg once daily on days 10-23. Blood samples were collected and plasma concentrations of famitinib were measured by validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. Pharmacokinetic parameters were calculated using noncompartmental analysis and safety was assessed. RESULTS: In the presence of rifampin, the famitinib geometric mean maximum plasma concentration (Cmax) and area under the plasma concentration-time curve from time zero to infinity (AUC0-∞) decreased by 48% and 69%, respectively, and the mean elimination half-life was shortened from 33.9 to 18.2 h. The geometric mean ratio (GMR) of famitinib Cmax and AUC0-∞ and their 90% CI were 0.52 (0.50, 0.54) and 0.31 (0.29, 0.33). Single dose of famitinib 25 mg was well tolerated and eight subjects (38.1%) reported treatment emergent adverse events, which were all grade 1-2 in severity. CONCLUSION: Co-administration of rifampin considerably reduces plasma concentration of famitinb due to CYP3A4 induction. Concomitant administration of famitinib and strong CYP3A4 inducers should be avoided, whereas when simultaneous use with inducers of CYP3A4, dose adjustment of famitinb is recommended. CLINICAL TRIAL REGISTRATION NUMBER: NCT04494659 (July 31, 2020).


Subject(s)
Indoles , Pyrroles , Rifampin , Chromatography, Liquid , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inducers/pharmacology , Drug Interactions , Healthy Volunteers , Humans , Indoles/pharmacokinetics , Male , Pyrroles/pharmacokinetics , Rifampin/pharmacology , Tandem Mass Spectrometry
11.
Molecules ; 27(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35011540

ABSTRACT

Pexidartinib is the first drug approved by the U.S. Food and Drug Administration specifically to treat the rare joint tumor tenosynovial giant cell tumor. In the current study, a validated, selective, and sensitive UPLC-MS/MS assay was developed for the quantitative determination of pexidartinib in plasma samples using gifitinib as an internal standard (IS). Pexidartinib and IS were extracted by liquid-liquid extraction using methyl tert-butyl ether and separated on an acquity BEH C18 column kept at 40 °C using a mobile phase of 0.1% formic acid in acetonitrile: 0.1% formic acid in de-ionized water (70:30). The flow rate was 0.25 mL/min. Multiple reaction monitoring (MRM) was operated in electrospray (ESI)-positive mode at the ion transition of 418.06 > 165.0 for the analyte and 447.09 > 128.0 for the IS. FDA guidance for bioanalytical method validation was followed in method validation. The linearity of the established UPLC-MS/MS assay ranged from 0.5 to 1000 ng/mL with r > 0.999 with a limit of quantitation of 0.5 ng/mL. Moreover, the metabolic stability of pexidartinib in liver microsomes was estimated.


Subject(s)
Aminopyridines/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacokinetics , Chromatography, High Pressure Liquid , Protein Kinase Inhibitors/pharmacokinetics , Pyrroles/pharmacokinetics , Tandem Mass Spectrometry , Aminopyridines/chemistry , Antineoplastic Agents, Immunological/chemistry , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Drug Monitoring/methods , Drug Monitoring/standards , Drug Stability , Molecular Structure , Protein Kinase Inhibitors/chemistry , Pyrroles/chemistry , Reproducibility of Results , Sensitivity and Specificity , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/standards
12.
Clin Pharmacol Drug Dev ; 11(4): 454-466, 2022 04.
Article in English | MEDLINE | ID: mdl-35092702

ABSTRACT

Pemigatinib is a fibroblast growth factor receptor 1-3 inhibitor used to treat cholangiocarcinoma. A compartmental population pharmacokinetics model was developed using data from 318 patients with cancer enrolled in a phase 1 dose-escalation/dose-expansion study, a phase 1 Japanese PK bridging study, and a phase 2 cholangiocarcinoma study. The final model for pemigatinib was a 2-compartment disposition model with first-order absorption and linear elimination. All fixed- and random-effect parameters were estimated with good precision, and no apparent biases in the overall model fit were observed. For females, the estimated typical pemigatinib absorption rate constant (ka ) and oral clearance (CL/F) were estimated (1.49 L/h and 10.3 L/h, respectively). For males, the typical apparent clearance and ka are 19.0% higher and 56.5% lower, respectively, compared with females. Typical apparent volume of distribution of the central compartment (Vc /F) and peripheral compartment for a 73.3-kg patient was estimated to be 122.0 L and 80.1 L, respectively; both increased with body weight. Phosphate binder coadministration decreases typical pemigatinib CL/F by 14.1%. Proton pump inhibitor coadministration increases typical pemigatinib apparent Vc/F by 24.4%. Phosphate binders and sex contribute a <20% change to CL/F. The impact of the investigated covariates on pemigatinib pharmacokinetics are not clinically significant.


Subject(s)
Neoplasms , Pyrimidines , Clinical Trials, Phase I as Topic , Female , Humans , Male , Morpholines/pharmacokinetics , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics
13.
J Infect Dis ; 226(1): 109-118, 2022 08 12.
Article in English | MEDLINE | ID: mdl-32604406

ABSTRACT

BACKGROUND: Both the elderly and individuals with comorbidities are at increased risk of developing influenza-related complications. Novel influenza antivirals are required, given limitations of current drugs (eg, resistance emergence and poor efficacy). Pimodivir is a first-in-class antiviral for influenza A under development for these patients. METHODS: Hospitalized patients with influenza A infection were randomized 2:1 to receive pimodivir 600 mg plus oseltamivir 75 mg or placebo plus oseltamivir 75 mg twice daily for 7 days in this phase 2b study. The primary objective was to compare pimodivir pharmacokinetics in elderly (aged 65-85 years) versus nonelderly adults (aged 18-64 years). Secondary end points included time to patient-reported symptom resolution. RESULTS: Pimodivir pharmacokinetic parameters in nonelderly and elderly patients were similar. Time to influenza symptom resolution was numerically shorter with pimodivir (72.45 hours) than placebo (94.15 hours). There was a lower incidence of influenza-related complications in the pimodivir group (7.9%) versus placebo group (15.6%). Treatment was generally well tolerated. CONCLUSIONS: No apparent relationship was observed between pimodivir pharmacokinetics and age. Our data demonstrate the need for a larger study of pimodivir in addition to oseltamivir to test whether it results in a clinically significant decrease in time-to-influenza-symptom alleviation and/or the frequency of influenza complications. CLINICAL TRIALS REGISTRATION: NCT02532283.


Subject(s)
Influenza, Human , Oseltamivir , Adult , Aged , Humans , Antiviral Agents , Influenza, Human/drug therapy , Oseltamivir/therapeutic use , Pyridines/therapeutic use , Pyrroles/pharmacokinetics , Treatment Outcome , Adolescent , Young Adult , Middle Aged , Aged, 80 and over
14.
Eur J Clin Pharmacol ; 78(1): 65-73, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34415382

ABSTRACT

PURPOSE: Esaxerenone is a novel, oral, nonsteroidal treatment for hypertension. Physiologically based pharmacokinetic (PBPK) modelling was performed to predict the drug-drug interaction (DDI) effect of cytochrome P450 (CYP)3A modulators on esaxerenone pharmacokinetics in healthy subjects and subjects with hepatic impairment. METHODS: In our PBPK model, the fraction of esaxerenone metabolised by CYP3A was estimated from mass-balance data and verified and optimised by clinical DDI study results with strong CYP3A modulators. The model was also verified by the observed pharmacokinetics after multiple oral dosing and by the effect of hepatic impairment on esaxerenone pharmacokinetics. The model was applied to predict the DDI effects on esaxerenone pharmacokinetics with untested CYP3A modulators in healthy subjects and with strong CYP3A modulators in subjects with hepatic impairment. RESULTS: The PBPK model well described esaxerenone pharmacokinetics after multiple oral dosing. The predicted fold changes in esaxerenone plasma exposure after coadministration with strong CYP3A modulators were comparable with the observed data (1.53-fold with itraconazole and 0.31-fold with rifampicin). Predicted DDIs with untested moderate CYP3A modulators were less than the observed DDI with strong CYP3A modulators. The PBPK model also described the effect of hepatic impairment on esaxerenone plasma exposure. The predicted DDI results with strong CYP3A modulators in subjects with hepatic impairment indicate that, for concomitant use of CYP3A modulators, caution is advised for subjects with hepatic impairment, as is for healthy subjects. CONCLUSION: The PBPK model developed predicted esaxerenone pharmacokinetics and DDIs and informed concurrent use of esaxerenone with CYP3A modulators.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Liver Failure/metabolism , Pyrroles/pharmacokinetics , Sulfones/pharmacokinetics , Area Under Curve , Computer Simulation , Drug Interactions , Healthy Volunteers , Humans , Itraconazole/pharmacology , Japan , Metabolic Clearance Rate , Models, Biological , Rifampin/pharmacology
15.
Pharmacol Res Perspect ; 10(1): e00906, 2022 02.
Article in English | MEDLINE | ID: mdl-34951522

ABSTRACT

Pemigatinib is a potent inhibitor of the fibroblast growth factor receptor (FGFR) family of receptors that is approved for the treatment of cholangiocarcinoma with FGFR2 fusion or other rearrangements. Data from a first-in-human clinical study were used to assess the potential for pemigatinib to produce clinically significant effects on heart rate (HR) and cardiac repolarization (QTc). A central tendency analysis for electrocardiogram (ECG) outliers and a plasma concentration-QTc analysis were conducted to assess cardiac safety in the first-in-human pemigatinib study (FIGHT-101; NCT02393248). The study included 113 participants who received at least one dose of pemigatinib as monotherapy and had at least one pair of plasma pharmacokinetic (PK) and ECG data points collected. Timed 12-lead ECGs were performed within 15 min of PK blood draws. The ECG parameters for each dose group in the study varied within expectations for patients with advanced malignancies. Categorical analysis of QT interval corrected for HR by Fridericia's method did not reveal dose dependence in the incidence of outliers, and the results of the central tendency and concentration-QTc analyses did not suggest a dose- or concentration-dependent drug effect. Least squares mean change from baseline in HR was small and did not indicate a clinically relevant effect on HR, and no effect was observed on cardiac conduction as assessed by PR and QRS intervals. In conclusion, pemigatinib does not exhibit any clinically significant prolongation of QTc or dose-dependent changes in HR. Clinical trial registration: ClinicalTrials.gov NCT02393248.


Subject(s)
Morpholines/adverse effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , Adult , Aged , Dose-Response Relationship, Drug , Electrocardiography , Female , Humans , Male , Middle Aged , Morpholines/administration & dosage , Morpholines/pharmacokinetics , Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics
16.
Clin Pharmacol Drug Dev ; 11(2): 270-277, 2022 02.
Article in English | MEDLINE | ID: mdl-34811955

ABSTRACT

Ubrogepant is a calcitonin gene-related peptide receptor antagonist for the acute treatment of migraine. Esomeprazole magnesium increases intragastric pH, which may affect oral ubrogepant absorption. This open-label, nonrandomized, crossover trial evaluated esomeprazole magnesium's impact on the pharmacokinetics and safety of coadministered ubrogepant in healthy adults. Participants received ubrogepant 100 mg on day 1, esomeprazole magnesium 40 mg on days 9 to 13, and ubrogepant 100 mg with esomeprazole magnesium 40 mg on day 14. No effect on ubrogepant pharmacokinetics was concluded if 90% confidence intervals of geometric mean ratios were within 80% to 125% for comparison of pharmacokinetic parameters between ubrogepant + esomeprazole magnesium versus ubrogepant alone. Thirty participants enrolled (mean age, 31.7 years; 53.3% males). Ubrogepant peak plasma concentration (Cmax ) decreased 23%, time to Cmax increased by 1.5 hours, and area under the plasma concentration-time curve was reduced by ≈10% when coadministered with esomeprazole magnesium versus ubrogepant alone. The 90% confidence interval of the geometric mean ratio for Cmax did not fall within the 80% to 125% equivalence range, but the decrease was not considered clinically meaningful. Esomeprazole magnesium coadministered with ubrogepant did not increase the incidence rate of treatment-emergent adverse events, and interactions between the medications are likely to have limited clinical relevance.


Subject(s)
Esomeprazole , Migraine Disorders , Adult , Esomeprazole/administration & dosage , Esomeprazole/adverse effects , Esomeprazole/pharmacokinetics , Female , Humans , Male , Migraine Disorders/drug therapy , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/adverse effects , Pyrroles/pharmacokinetics
17.
Nat Med ; 27(12): 2183-2191, 2021 12.
Article in English | MEDLINE | ID: mdl-34873347

ABSTRACT

Advanced systemic mastocytosis (AdvSM) is a rare hematologic neoplasm driven by the KIT D816V mutation and associated with poor survival. This phase 1 study ( NCT02561988 ) evaluated avapritinib (BLU-285), a selective KIT D816V inhibitor, in patients with AdvSM. The primary endpoints were the maximum tolerated dose, recommended phase 2 dose and safety of avapritinib. Secondary endpoints included overall response rate and changes in measures of mast cell burden. Avapritinib was evaluated at doses of 30-400 mg once daily in 86 patients, 69 with centrally confirmed AdvSM. Maximum tolerated dose was not reached, and 200 mg and 300 mg daily were studied in dose-expansion cohorts. The most frequent adverse events observed were periorbital edema (69%), anemia (55%), diarrhea (45%), thrombocytopenia (44%) and nausea (44%). Intracranial bleeding occurred in 13% overall, but in only 1% of patients without severe thrombocytopenia (platelets <50 × 109/l). In 53 response-evaluable patients, the overall response rate was 75%. The complete remission rate was 36%. Avapritinib elicited ≥50% reductions in marrow mast cells and serum tryptase in 92% and 99% of patients, respectively. Avapritinib induced deep and durable responses, including molecular remission of KIT D816V in patients with AdvSM, and was well tolerated at the recommended phase 2 dose of 200 mg daily.


Subject(s)
Mastocytosis, Systemic/drug therapy , Pyrazoles/therapeutic use , Pyrroles/therapeutic use , Triazines/therapeutic use , Adult , Aged , Aged, 80 and over , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Triazines/administration & dosage , Triazines/adverse effects , Triazines/pharmacokinetics
18.
Bioorg Med Chem Lett ; 49: 128314, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34391891

ABSTRACT

A series of IDO1 inhibitors containing a decahydroquinoline, decahydro-1,6-naphthyridine, or octahydro-1H-pyrrolo[3,2-c]pyridine scaffold were identified with good cellular and human whole blood activity against IDO1. These inhibitors contain multiple chiral centers and all diastereomers were separated. The absolute stereochemistry of each isomers were not determined. Compounds 15 and 27 stood out as leads due to their good cellular as well as human whole blood IDO1 inhibition activity, low unbound clearance, and reasonable mean residence time in rat cassette PK studies.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Naphthyridines/pharmacology , Pyrroles/pharmacology , Quinolines/pharmacology , Animals , Catalytic Domain , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , HeLa Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Molecular Docking Simulation , Naphthyridines/chemical synthesis , Naphthyridines/metabolism , Naphthyridines/pharmacokinetics , Pyrroles/chemical synthesis , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Quinolines/chemical synthesis , Quinolines/metabolism , Quinolines/pharmacokinetics , Rats , Stereoisomerism , Structure-Activity Relationship
19.
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
20.
Eur J Med Chem ; 223: 113627, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34171657

ABSTRACT

The tropomyosin receptor kinases TRKs are responsible for different tumor types which caused by NTRK gene fusion, and have been identified as a successful target for anticancer therapeutics. Herein, we report a potent and selectivity TRKs inhibitor 19m through rational drug design strategy from a micromolar potency hit 17a. Compound 19m significantly inhibits the proliferation of TRK-dependent cell lines (Km-12), while it has no inhibitory effect on TRK-independent cell lines (A549 and THLE-2). Furthermore, kinases selectivity profiling showed that in addition to TRKs, compound 19m only displayed relatively strong inhibitory activity on ALK. These data may indicate that compound 19m has a good drug safety. Partial ADME properties were evaluated in vitro and in vivo. Compound 19m exhibited a good AUC values and volume of distribution and low clearance in the pharmacokinetics experiment of rats. Finally, a pharmacophore model guided by experimental results is proposed. We hope this theoretical model can help researchers find type I TRK inhibitors more efficiently.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrroles/pharmacology , Receptor, trkA/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Design , Drug Stability , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrroles/chemical synthesis , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Rats, Sprague-Dawley , Receptor, trkA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...