Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 717
Filter
1.
Int J Nanomedicine ; 19: 5681-5703, 2024.
Article in English | MEDLINE | ID: mdl-38882541

ABSTRACT

Introduction: Diabetes mellitus is frequently associated with foot ulcers, which pose significant health risks and complications. Impaired wound healing in diabetic patients is attributed to multiple factors, including hyperglycemia, neuropathy, chronic inflammation, oxidative damage, and decreased vascularization. Rationale: To address these challenges, this project aims to develop bioactive, fast-dissolving nanofiber dressings composed of polyvinylpyrrolidone loaded with a combination of an antibiotic (moxifloxacin or fusidic acid) and anti-inflammatory drug (pirfenidone) using electrospinning technique to prevent the bacterial growth, reduce inflammation, and expedite wound healing in diabetic wounds. Results: The fabricated drug-loaded fibers exhibited diameters of 443 ± 67 nm for moxifloxacin/pirfenidone nanofibers and 488 ± 92 nm for fusidic acid/pirfenidone nanofibers. The encapsulation efficiency, drug loading and drug release studies for the moxifloxacin/pirfenidone nanofibers were found to be 70 ± 3% and 20 ± 1 µg/mg, respectively, for moxifloxacin, and 96 ± 6% and 28 ± 2 µg/mg, respectively, for pirfenidone, with a complete release of both drugs within 24 hours, whereas the fusidic acid/pirfenidone nanofibers were found to be 95 ± 6% and 28 ± 2 µg/mg, respectively, for fusidic acid and 102 ± 5% and 30 ± 2 µg/mg, respectively, for pirfenidone, with a release rate of 66% for fusidic acid and 80%, for pirfenidone after 24 hours. The efficacy of the prepared nanofiber formulations in accelerating wound healing was evaluated using an induced diabetic rat model. All tested formulations showed an earlier complete closure of the wound compared to the controls, which was also supported by the histopathological assessment. Notably, the combination of fusidic acid and pirfenidone nanofibers demonstrated wound healing acceleration on day 8, earlier than all tested groups. Conclusion: These findings highlight the potential of the drug-loaded nanofibrous system as a promising medicated wound dressing for diabetic foot applications.


Subject(s)
Anti-Bacterial Agents , Bandages , Diabetic Foot , Drug Liberation , Fusidic Acid , Moxifloxacin , Nanofibers , Pyridones , Wound Healing , Diabetic Foot/drug therapy , Diabetic Foot/therapy , Nanofibers/chemistry , Animals , Moxifloxacin/administration & dosage , Moxifloxacin/pharmacology , Moxifloxacin/chemistry , Moxifloxacin/pharmacokinetics , Wound Healing/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Pyridones/chemistry , Pyridones/pharmacology , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Fusidic Acid/administration & dosage , Fusidic Acid/pharmacology , Fusidic Acid/chemistry , Fusidic Acid/pharmacokinetics , Rats , Male , Diabetes Mellitus, Experimental , Povidone/chemistry , Rats, Sprague-Dawley
2.
Bioorg Med Chem Lett ; 109: 129849, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38876177

ABSTRACT

Clinical studies have shown that inhibitors of bromodomain and extra-terminal domain (BET) proteins, particularly BRD4, have antitumor activity and efficacy. The BET protein has two domains, BD1 and BD2, and we previously focused on BD1 and reported orally bioavailable BD1-selective inhibitors. In this study, we obtained a BD1 inhibitor, a more potent and highly selective pyrazolopyridone derivative 13a, and confirmed its in vivo efficacy.


Subject(s)
Pyridones , Humans , Administration, Oral , Structure-Activity Relationship , Animals , Pyridones/chemistry , Pyridones/pharmacology , Pyridones/chemical synthesis , Pyridones/pharmacokinetics , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/chemical synthesis , Drug Discovery , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Molecular Structure , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Mice , Protein Domains , Dose-Response Relationship, Drug , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Rats , Bromodomain Containing Proteins
3.
Pharmacogenet Genomics ; 34(6): 184-190, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38728170

ABSTRACT

PURPOSE: This study was the first to evaluate the effect of CYP3A5*3 gene polymorphisms on plasma concentration of perampanel (PER) in Chinese pediatric patients with epilepsy. METHODS: We enrolled 98 patients for this investigation. Plasma PER concentrations were measured using liquid chromatography-tandem mass spectrometry. Leftover samples from standard therapeutic drug monitoring were allocated for genotyping analysis. The primary measure of efficacy was the rate of seizure reduction with PER treatment at the final checkup. RESULTS: The plasma concentration showed a linear correlation with the daily dose taken ( r  = 0.17; P  < 0.05). The ineffective group showed a significantly lower plasma concentration of PER (490.5 ±â€…297.1 vs. 633.8 ±â€…305.5 µg/ml; P  = 0.019). For the mean concentration-to-dose (C/D) ratio, the ineffective group showed a significantly lower C/D ratio of PER (3.2 ±â€…1.7 vs. 3.8 ±â€…2.0; P  = 0.040). The CYP3A5*3 CC genotype exhibited the highest average plasma concentration of PER at 562.8 ±â€…293.9 ng/ml, in contrast to the CT and TT genotypes at 421.1 ±â€…165.6 ng/ml and 260.0 ±â€…36.1 ng/ml. The mean plasma PER concentration was significantly higher in the adverse events group (540.8 ±â€…285.6 vs. 433.0 ±â€…227.2 ng/ml; P  = 0.042). CONCLUSION: The CYP3A5*3 gene's genetic polymorphisms influence plasma concentrations of PER in Chinese pediatric patients with epilepsy. Given that both efficacy and potential toxicity are closely tied to plasma PER levels, the CYP3A5*3 genetic genotype should be factored in when prescribing PER to patients with epilepsy.


Subject(s)
Anticonvulsants , Cytochrome P-450 CYP3A , Epilepsy , Nitriles , Pyridones , Humans , Cytochrome P-450 CYP3A/genetics , Child , Female , Male , Epilepsy/drug therapy , Epilepsy/genetics , Nitriles/pharmacokinetics , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Pyridones/adverse effects , Child, Preschool , Anticonvulsants/pharmacokinetics , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Polymorphism, Single Nucleotide/genetics , Genotype , Adolescent , Asian People/genetics , East Asian People
4.
Clin Transl Sci ; 17(5): e13819, 2024 May.
Article in English | MEDLINE | ID: mdl-38747478

ABSTRACT

The equivalence of absorption rates and extents between generic drugs and their reference formulations is crucial for ensuring therapeutic comparability. Bioequivalence (BE) studies are widely utilized and play a pivotal role in substantiating the approval and promotional efforts for generic drugs. Virtual BE simulation is a valuable tool for mitigating risks and guiding clinical BE studies, thereby minimizing redundant in vivo BE assessments. Herein, we successfully developed a physiologically based absorption model for virtual BE simulations, which precisely predicts the BE of the apixaban test and reference formulations. The modeling results confirm that the test and reference formulations were bioequivalent under both fasted and fed conditions, consistent with clinical studies. This highlights the efficacy of physiologically based absorption modeling as a powerful tool for formulation screening and can be adopted as a methodological and risk assessment strategy to detect potential clinical BE risks.


Subject(s)
Models, Biological , Pyrazoles , Pyridones , Therapeutic Equivalency , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Pyrazoles/pharmacokinetics , Pyrazoles/administration & dosage , Humans , Factor Xa Inhibitors/pharmacokinetics , Factor Xa Inhibitors/administration & dosage , Drugs, Generic/pharmacokinetics , Drugs, Generic/administration & dosage , Computer Simulation , Administration, Oral , Male
5.
Ther Drug Monit ; 46(3): 277-280, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38723113

ABSTRACT

BACKGROUND: Carbamazepine (CBZ) is an antiseizure medication known to induce the expression of cytochrome P4503A metabolic enzymes. Here, we describe a man living with HIV who underwent several changes in the daily dose of CBZ, which resulted in different induction effects on darunavir trough concentrations. METHODS: A 59-year-old man with HIV, successfully undergoing maintenance antiretroviral treatment with darunavir/cobicistat once daily (combined with raltegravir), was prescribed CBZ for recurrent trigeminal neuralgia. Over subsequent months, the patient underwent various changes in the doses (from 200 to 800 mg/d) and trough concentrations (from 3.6 to 18.0 mg/L) of CBZ, guided by clinical response to trigeminal neuralgia. RESULTS: A highly significant inverse association was observed between darunavir trough concentration and both CBZ dose or trough concentration (coefficient of determination >0.75, P < 0.0001). Ultimately, the darunavir dose was increased to 600 mg twice daily with ritonavir and dolutegravir to ensure optimal antiretroviral coverage, anticipating potential further uptitration of CBZ doses. CONCLUSIONS: The impact of CBZ on boosted darunavir exposure seemed to be dose- and concentration-dependent. The management of such drug-drug interactions in daily practice was facilitated through therapeutic drug monitoring. This case underscores the importance of a multidisciplinary approach that incorporates both antiretroviral and nonantiretroviral comedications contributing to the optimal management of polypharmacy in individuals living with HIV.


Subject(s)
Carbamazepine , Darunavir , Drug Interactions , HIV Infections , Humans , Darunavir/therapeutic use , Darunavir/pharmacokinetics , Male , Middle Aged , Carbamazepine/therapeutic use , Carbamazepine/pharmacokinetics , HIV Infections/drug therapy , Trigeminal Neuralgia/drug therapy , Ritonavir/therapeutic use , Ritonavir/administration & dosage , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Anticonvulsants/administration & dosage , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Pyridones/blood , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Heterocyclic Compounds, 3-Ring/administration & dosage , Piperazines/therapeutic use , Piperazines/pharmacokinetics , Oxazines/therapeutic use , Oxazines/pharmacokinetics , Dose-Response Relationship, Drug , Anti-HIV Agents/therapeutic use , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/administration & dosage , Drug Monitoring/methods
6.
AIDS Res Ther ; 21(1): 34, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773606

ABSTRACT

INTRODUCTION: Peritoneal dialysis (PD) is an effective renal replacement modality in people with HIV (PWH) with end-stage kidney disease (ESKD), particularly those with residual kidney function. Data on pharmacokinetics (PK) of antiretrovirals in patients on peritoneal dialysis are limited. METHODS: A single-participant study was performed on a 49-year-old gentleman with ESKD on PD and controlled HIV on once daily dolutegravir (DTG) 50 mg + tenofovir alafenamide (TAF) 25 mg / emtricitabine (FTC) 200 mg. He underwent serial blood plasma, peripheral blood mononuclear cell, and urine PK measurements over 24 h after an observed DTG + FTC/TAF dose. RESULTS: Plasma trough (Cmin) concentrations of TAF, tenofovir (TFV), FTC, and DTG were 0.05, 164, 1,006, and 718 ng/mL, respectively. Intracellular trough concentrations of TFV-DP and FTC-TP were 1142 and 11,201 fmol/million cells, respectively. Compared to published mean trough concentrations in PWH with normal kidney function, observed TFV and FTC trough concentrations were 15.5- and 20-fold higher, while intracellular trough concentrations of TFV-DP and FTC-TP were 2.2-fold and 5.4-fold higher, respectively. TFV and FTC urine levels were 20 times lower than in people with normal GFR. CONCLUSIONS: In a single ESKD PWH on PD, daily TAF was associated with plasma TFV and intracellular TFV-DP trough concentrations 15-fold and 2-fold higher than those of people with uncompromised kidney function, potentially contributing to nephrotoxicity. This suggests that TFV accumulates on PD; thus, daily TAF in PD patients may require dose adjustment or regimen change to optimize treatment, minimize toxicity, and preserve residual kidney function.


Subject(s)
Adenine , Alanine , Anti-HIV Agents , Emtricitabine , HIV Infections , Heterocyclic Compounds, 3-Ring , Kidney Failure, Chronic , Oxazines , Peritoneal Dialysis , Piperazines , Pyridones , Tenofovir , Humans , Male , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Heterocyclic Compounds, 3-Ring/adverse effects , Oxazines/pharmacokinetics , Pyridones/pharmacokinetics , Middle Aged , Tenofovir/pharmacokinetics , Tenofovir/therapeutic use , Tenofovir/analogs & derivatives , Emtricitabine/pharmacokinetics , Emtricitabine/therapeutic use , Piperazines/pharmacokinetics , HIV Infections/drug therapy , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Alanine/pharmacokinetics , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Adenine/therapeutic use , Kidney Failure, Chronic/therapy
7.
Epilepsia ; 65(6): 1687-1697, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38572689

ABSTRACT

OBJECTIVE: The purposes of this study were to explore the pharmacokinetics of perampanel (PER) in children with epilepsy, identify factors that contribute to pharmacokinetic variations among subjects, evaluate the connection between PER exposure and clinical outcome, and establish an evidence-based approach for tailoring individualized antiepileptic treatment in this specific population. METHODS: In this prospective study, PER plasma concentrations and genetic information on metabolic enzymes were obtained from 194 patients younger than 18 years. The disposition kinetics of PER in pediatric patients following oral dosing were characterized using nonlinear mixed effect models. The effective range for the plasma concentration of PER was determined by assessing the efficacy and safety of PER treatment and analyzing the relationship between drug exposure and clinical response. Monte Carlo simulations were then performed to evaluate and optimize the current dosing regimens. RESULTS: The pharmacokinetic profile of PER was adequately described by a one-compartment model with first-order absorption and elimination. Body weight, total bilirubin level, and concomitant oxcarbazepine were found to have significant influences on PER pharmacokinetics. Model estimates of apparent clearance and volume of distribution were .016 ± .009 L/h/kg and 1.47 ± .78 L/kg, respectively. The effective range predicted from plasma concentration data in responders was 215-862 µg/L. Dosing scenarios stratified according to essential covariates were proposed through simulation analysis. SIGNIFICANCE: In this study, we captured the pharmacokinetic/pharmacodynamic characteristics of PER in pediatric epilepsy patients through analysis of real-world data and adopted a pharmacometric approach to support an individualized dosing strategy for PER in this specific population.


Subject(s)
Anticonvulsants , Epilepsy , Nitriles , Pyridones , Humans , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Pyridones/therapeutic use , Pyridones/blood , Anticonvulsants/pharmacokinetics , Anticonvulsants/administration & dosage , Anticonvulsants/therapeutic use , Child , Nitriles/pharmacokinetics , Female , Male , Epilepsy/drug therapy , Child, Preschool , Adolescent , Prospective Studies , Dose-Response Relationship, Drug , Infant , Monte Carlo Method , Treatment Outcome
8.
Clin Pharmacokinet ; 63(5): 721-728, 2024 May.
Article in English | MEDLINE | ID: mdl-38573477

ABSTRACT

BACKGROUND AND OBJECTIVE: Within the UNIVERSAL project (RIA2019PD-2882) we aim to develop a paediatric dolutegravir (DTG)/emtricitabine (FTC or F)/tenofovir alafenamide (TAF) fixed-dose combination. To inform dosing of this study, we undertook a relative bioavailability (RBA) study in healthy volunteers to investigate a potential pharmacokinetic effect when paediatric formulations of DTG and F/TAF are taken together. METHODS: Participants received all of the following treatments as paediatric formulations in randomised order: a single dose of 180/22.5 mg F/TAF; a single dose of 30 mg DTG; a single dose of 180/22.5 mg F/TAF plus 30 mg DTG. Blood concentrations of DTG, FTC, TAF, and tenofovir (TFV) were measured over 48 h post-dose. If the 90% confidence intervals (CIs) of the geometric least squares mean (GLSM) ratios of area under the curve (AUC) and maximum concentration (Cmax) of each compound were within 0.70-1.43, we considered this as no clinically relevant PK interaction. RESULTS: A total of 15 healthy volunteers were included. We did not observe a clinically relevant PK interaction between the paediatric DTG and F/TAF formulations for the compounds DTG, FTC, and TFV. For TAF, the lower boundaries of the 90% CIs of the GLSM ratios of the AUC0-∞ and Cmax fell outside our acceptance criteria of 0.70-1.43. CONCLUSIONS: Although TAF AUC and Cmax 90% CIs fell outside the pre-defined criteria (0.62-1.11 and 0.65-1.01, respectively), no consistent effect on TAF PK was observed, likely due to high inter-subject variability. Moreover, there are several reasons to rely on TFV exposure as being more clinically relevant than TAF exposure. Therefore, we found no clinically relevant interactions in this study.


Subject(s)
Alanine , Biological Availability , Emtricitabine , Healthy Volunteers , Heterocyclic Compounds, 3-Ring , Oxazines , Piperazines , Pyridones , Tablets , Tenofovir , Humans , Pyridones/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/blood , Tenofovir/pharmacokinetics , Tenofovir/administration & dosage , Tenofovir/analogs & derivatives , Male , Emtricitabine/pharmacokinetics , Emtricitabine/administration & dosage , Piperazines/pharmacokinetics , Female , Adult , Oxazines/pharmacokinetics , Oxazines/administration & dosage , Alanine/pharmacokinetics , Alanine/administration & dosage , Drug Combinations , Young Adult , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Adenine/administration & dosage , Adenine/blood , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/blood , Area Under Curve , Middle Aged , Adolescent , Cross-Over Studies
9.
Int J Clin Pharmacol Ther ; 62(7): 319-325, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38660886

ABSTRACT

OBJECTIVE: The impact of pancreaticoduodenectomy on absorption of drugs in the duodenum remains largely unknown. We aim to characterize the pharmacokinetics of apixaban in patients who had previously undergone pancreaticoduodenectomy. MATERIALS AND METHODS: A single 10-mg dose of apixaban was administered to 4 volunteers who underwent pancreaticoduodenectomy at least 6 months prior. The maximum plasma apixaban concentration (Cmax) and area under the plasma concentration time-curve (AUC0-24, AUC0-inf) were compared against healthy historical control subjects (N = 12). Geometric mean ratios (GMR) with 90% confidence interval (CI) were calculated for determination of comparative bioequivalence. RESULTS: In pancreaticoduodenectomy patients, AUC0-24 and AUC0-inf were 1,861 and 2,080 ng×h/mL, respectively. The GMRs of AUC0-24 and AUC0-inf between study subjects and healthy controls were 1.27 (90% CI 0.88 - 1.83) and 1.18 (90% CI 0.82 - 1.72). The mean Cmax of apixaban was 201 ng/mL (SD 15.6) occurring at a median tmax of 3.25 hours (range 2.5 - 4 hours). The GMR of Cmax between study subjects and healthy controls was 1.12 (90% CI 0.77 - 1.63). CONCLUSION: The pharmacokinetic characteristics of apixaban in subjects who had undergone pancreaticoduodenectomy are not significantly different from those of healthy controls. Though the sample size of this study is small, results suggest that no change to apixaban dose regimen is needed in patients who have had a pancreaticoduodenectomy.


Subject(s)
Area Under Curve , Factor Xa Inhibitors , Pancreaticoduodenectomy , Pyrazoles , Pyridones , Humans , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Pancreaticoduodenectomy/adverse effects , Pyrazoles/pharmacokinetics , Pyrazoles/administration & dosage , Male , Middle Aged , Female , Factor Xa Inhibitors/pharmacokinetics , Factor Xa Inhibitors/administration & dosage , Aged , Adult , Therapeutic Equivalency
10.
Antimicrob Agents Chemother ; 68(5): e0150423, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38587380

ABSTRACT

A fixed-dose combination (FDC) of 50 mg dolutegravir and 300 mg lamivudine is indicated for the treatment of HIV-1 infection. This analysis aimed to characterize the population pharmacokinetics (PK) of dolutegravir and lamivudine based on data from a phase 3 study (TANGO) in virologically suppressed adults living with HIV-1 switching to dolutegravir/lamivudine FDC. These analyses included 362 participants who contributed 2,629 dolutegravir and 2,611 lamivudine samples collected over 48 weeks. A one-compartment model with first-order absorption and elimination parameterized by apparent oral clearance (CL/F), apparent volume of distribution (V/F), and absorption rate constant (Ka) described dolutegravir PK. Covariate search yielded body weight, bilirubin, and ethnicity as predictors of CL/F, and weight was predictive for V/F. The estimates of CL/F, V/F, and Ka were 0.858 L/h, 16.7 L, and 2.15 h-1, respectively. A two-compartment model with first-order absorption and elimination parameterized by CL/F, apparent intercompartmental clearance (Q/F), apparent central volume of distribution (V2/F), apparent peripheral volume of distribution (V3/F), and Ka described lamivudine PK. Covariate search yielded eGFR and race as predictors of CL/F, and weight was predictive for V2/F. The estimated parameter values were CL/F = 19.6 L/h, Q/F = 2.97 L/h, V2/F = V3/F = 105 L, and Ka = 2.30 h-1. The steady-state prediction suggested that the effect of covariates dolutegravir and lamivudine exposures was small (<20%) and not clinically relevant. Therefore, no dose adjustments are recommended based on these analyses. The results support the use of dolutegravir/lamivudine FDC in the treatment of HIV-1 infection in adults.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT03446573.


Subject(s)
Anti-HIV Agents , HIV Infections , HIV-1 , Heterocyclic Compounds, 3-Ring , Lamivudine , Oxazines , Piperazines , Pyridones , Humans , Lamivudine/pharmacokinetics , Lamivudine/therapeutic use , Lamivudine/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/therapeutic use , Oxazines/pharmacokinetics , HIV Infections/drug therapy , HIV Infections/virology , Piperazines/pharmacokinetics , Pyridones/pharmacokinetics , Adult , Male , Female , HIV-1/drug effects , Middle Aged , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Anti-HIV Agents/administration & dosage , Drug Combinations
11.
Antiviral Res ; 227: 105890, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657838

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a highly pathogenic bunyavirus with a fatality rate of up to 40%. Currently, there are no licensed antiviral drugs for the treatment of CCHF; thus, the World Health Organization (WHO) listed the disease as a priority. A unique viral transcription initiation mechanism called "cap-snatching" is shared by influenza viruses and bunyaviruses. Thus, we tested whether baloxavir (an FDA-approved anti-influenza drug that targets the "cap-snatching" mechanism) could inhibit CCHFV infection. In cell culture, baloxavir acid effectively inhibited CCHFV infection and targeted CCHFV RNA transcription/replication. However, it has weak oral bioavailability. Baloxavir marboxil (the oral prodrug of baloxavir) failed to protect mice against a lethal dose challenge of CCHFV. To solve this problem, baloxavir sodium was synthesized owing to its enhanced aqueous solubility and pharmacokinetic properties. It consistently and significantly improved survival rates and decreased tissue viral loads. This study identified baloxavir sodium as a novel scaffold structure and mechanism of anti-CCHF compound, providing a promising new strategy for clinical treatment of CCHF after further optimization.


Subject(s)
Antiviral Agents , Dibenzothiepins , Morpholines , Pyridines , Pyridones , Triazines , Virus Replication , Animals , Morpholines/pharmacology , Morpholines/pharmacokinetics , Morpholines/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/pharmacokinetics , Antiviral Agents/chemistry , Dibenzothiepins/pharmacology , Dibenzothiepins/pharmacokinetics , Mice , Pyridines/pharmacology , Pyridines/pharmacokinetics , Pyridines/chemistry , Virus Replication/drug effects , Triazines/pharmacology , Triazines/pharmacokinetics , Triazines/chemistry , Triazines/therapeutic use , Pyridones/pharmacology , Pyridones/pharmacokinetics , Pyridones/chemistry , Thiepins/pharmacology , Thiepins/therapeutic use , Thiepins/pharmacokinetics , Thiepins/chemistry , Viral Load/drug effects , Chlorocebus aethiops , Vero Cells , Female , Oxazines/pharmacology , Oxazines/pharmacokinetics , Oxazines/therapeutic use , Mice, Inbred BALB C , Humans , Thiazoles/pharmacology , Thiazoles/pharmacokinetics , Thiazoles/chemistry
12.
Contemp Clin Trials ; 142: 107540, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636725

ABSTRACT

BACKGROUND: There is increasing interest in utilising two-drug regimens for HIV treatment with the goal of reducing toxicity and improve acceptability. The D3 trial evaluates the efficacy and safety of DTG/3TC in children and adolescents and includes a nested pharmacokinetics(PK) substudy for paediatric drug licensing. METHODS: D3 is an ongoing open-label, phase III, 96-week non-inferiority randomised controlled trial(RCT) conducted in South Africa, Spain, Thailand, Uganda and the United Kingdom. D3 has enrolled 386 children aged 2- < 15 years, virologically suppressed for ≥6 months, with no prior treatment failure. Participants were randomised 1:1 to receive DTG/3TC or DTG plus two nucleoside reverse transcriptase inhibitors(NRTIs), stratified by region, age (2- < 6, 6- < 12, 12- < 15 years) and DTG use at enrolment (participants permitted to start DTG at enrolment). The primary outcome is confirmed HIV-1 RNA viral rebound ≥50 copies/mL by 96-weeks. The trial employs the Smooth Away From Expected(SAFE) non-inferiority frontier, which specifies the non-inferiority margin and significance level based on the observed event risk in the control arm. The nested PK substudy evaluates WHO weight-band-aligned dosing in the DTG/3TC arm. DISCUSSION: D3 is the first comparative trial evaluating DTG/3TC in children and adolescents. Implications of integrating a PK substudy and supplying data for prompt regulatory submission, were carefully considered to ensure the integrity of the ongoing trial. The trial uses an innovative non-inferiority frontier for the primary analysis to allow for a lower-than-expected confirmed viral rebound risk in the control arm, while ensuring interpretability of results and maintaining the planned sample size in an already funded trial. TRIAL REGISTRATION: International Standard Randomised Clinical Trial Number Register: ISRCTN17157458. European Clinical Trials Database: 2020-001426-57. CLINICALTRIALS: gov: NCT04337450.


Subject(s)
HIV Infections , HIV-1 , Heterocyclic Compounds, 3-Ring , Lamivudine , Oxazines , Piperazines , Pyridones , Humans , Adolescent , HIV Infections/drug therapy , Pyridones/administration & dosage , Pyridones/therapeutic use , Pyridones/pharmacokinetics , Child , Oxazines/administration & dosage , Oxazines/therapeutic use , Child, Preschool , Lamivudine/administration & dosage , Lamivudine/therapeutic use , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Piperazines/administration & dosage , Male , Female , HIV-1/drug effects , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Anti-HIV Agents/pharmacokinetics , Viral Load , Equivalence Trials as Topic , RNA, Viral , Drug Therapy, Combination , Drug Combinations , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/pharmacokinetics
13.
Clin Appl Thromb Hemost ; 30: 10760296241249167, 2024.
Article in English | MEDLINE | ID: mdl-38659339

ABSTRACT

Apixaban is a direct oral Xa inhibitor and is indicated for the treatment of venous thrombo-embolism (VTE) and prevention of stroke in atrial fibrillation (AF). Recently, a generic (ZyQuis, Zydus Lifesciences Limited, India) has received Food and Drug Administration approval. While bioequivalence has been demonstrated with Eliquis (Bristol-Myers Squibb/Pfizer, UK), it is necessary to monitor its effectiveness prior to acceptance in medical practice. This prospective study independently evaluated Apixaban (ZyQuis) at two accredited laboratories. Participants were converted from Warfarin or Rivaroxaban to Apixaban 5 mg bd for a duration of one month. Peak anti-Xa levels were measured 3-4 h post the morning dose. The samples were processed on the Atellica COAG 360 (Siemens Healthineers, Marburg, Germany) analyzers with a chromogenic anti-Xa assay (Innovance, reference interval 69-321 ng/mL). There were 26 participants; 5 men, 21 women; mean ± standard deviation age of 46 ± 12 years. Indications for anticoagulation included: VTE (88.5%) and AF (11.5%). 69.2% of the participants had at least one comorbidity. 96.2% of the anti-Xa levels were within the laboratory's 95% reference interval. Mean anti-Xa activity was 191 ± 69 ng/mL and 186 ± 68 ng/mL measured at respective laboratories. Mean differences in anti-Xa measurements represented by Bland-Altman statistics were small (bias of -2.6%, 95% confidence interval -1.11 to -4.09) and a strong correlation was observed on Deming regression analysis (0.995). Apixaban (ZyQuis) was effective for the management of VTE and AF as evidenced by anti-Xa activity.


Subject(s)
Atrial Fibrillation , Factor Xa Inhibitors , Pyrazoles , Pyridones , Venous Thromboembolism , Humans , Pyridones/therapeutic use , Pyridones/administration & dosage , Pyridones/pharmacology , Pyridones/pharmacokinetics , Pyrazoles/therapeutic use , Pyrazoles/pharmacokinetics , Pyrazoles/administration & dosage , Atrial Fibrillation/drug therapy , Atrial Fibrillation/complications , Atrial Fibrillation/blood , Male , Female , Middle Aged , Factor Xa Inhibitors/therapeutic use , Factor Xa Inhibitors/pharmacokinetics , Factor Xa Inhibitors/pharmacology , Venous Thromboembolism/drug therapy , Venous Thromboembolism/prevention & control , Prospective Studies , Adult , Drug Monitoring/methods
14.
Lancet HIV ; 11(5): e300-e308, 2024 May.
Article in English | MEDLINE | ID: mdl-38621393

ABSTRACT

BACKGROUND: Coformulated bictegravir, emtricitabine, and tenofovir alafenamide is a single-tablet regimen and was efficacious and well tolerated in children and adolescents with HIV (aged 6 years to <18 years) in a 48-week phase 2/3 trial. In this study, we report data from children aged at least 2 years and weighing 14 kg to less than 25 kg. METHODS: We conducted this open-label, multicentre, multicohort, single-arm study in South Africa, Thailand, Uganda, and the USA. Participants were virologically suppressed children with HIV, aged at least 2 years, weighing 14 kg to less than 25 kg. Participants received bictegravir (30 mg), emtricitabine (120 mg), and tenofovir alafenamide (15 mg) once daily, switching to bictegravir (50 mg), emtricitabine (200 mg), and tenofovir alafenamide (25 mg) upon attaining a bodyweight of at least 25 kg. The study included pharmacokinetic evaluation at week 2 to confirm the dose of coformulated bictegravir, emtricitabine, and tenofovir alafenamide for this weight band by comparing with previous adult data. Primary outcomes were bictegravir area under the curve over the dosing interval (AUCtau) and concentration at the end of the dosing interval (Ctau) at week 2, and incidence of treatment-emergent adverse events and laboratory abnormalities until the end of week 24 in all participants who received at least one dose of bictegravir, emtricitabine, and tenofovir alafenamide. This study is registered with ClinicalTrials.gov, NCT02881320. FINDINGS: Overall, 22 participants were screened (from Nov 14, 2018, to Jan 11, 2020), completed treatment with bictegravir, emtricitabine, and tenofovir alafenamide (until week 48), and entered an extension phase. The geometric least squares mean (GLSM) ratio for AUCtau for bictegravir was 7·6% higher than adults (GLSM ratio 107·6%, 90% CI 96·7-119·7); Ctau was 34·6% lower than adults (65·4%, 49·1-87·2). Both parameters were within the target exposure range previously found in adults, children, or both". Grade 3-4 laboratory abnormalities occurred in four (18%) participants by the end week 24 and six (27%) by the end of week 48. Drug-related adverse events occurred in three participants (14%) by the end of week 24 and week 48; none were severe. No Grade 3-4 adverse events, serious adverse events, or adverse events leading to discontinuation occurred by the end of week 24 and week 48. INTERPRETATION: Data support the use of single-tablet coformulated bictegravir (30 mg), emtricitabine (120 mg), and tenofovir alafenamide (15 mg) for treatment of HIV in children aged at least 2 years and weighing 14 kg to less than 25 kg. FUNDING: Gilead Sciences.


Subject(s)
Adenine , Alanine , Amides , Anti-HIV Agents , Emtricitabine , HIV Infections , Heterocyclic Compounds, 3-Ring , Heterocyclic Compounds, 4 or More Rings , Piperazines , Pyridones , Tenofovir , Tenofovir/analogs & derivatives , Humans , Emtricitabine/pharmacokinetics , Emtricitabine/administration & dosage , Emtricitabine/therapeutic use , Emtricitabine/adverse effects , HIV Infections/drug therapy , HIV Infections/virology , Tenofovir/pharmacokinetics , Tenofovir/administration & dosage , Tenofovir/adverse effects , Tenofovir/therapeutic use , Child , Male , Female , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/adverse effects , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Child, Preschool , Alanine/pharmacokinetics , Alanine/adverse effects , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/adverse effects , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Amides/pharmacokinetics , Adolescent , Pyridones/pharmacokinetics , Pyridones/adverse effects , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/adverse effects , Heterocyclic Compounds, 3-Ring/administration & dosage , Piperazines/adverse effects , Piperazines/pharmacokinetics , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Adenine/adverse effects , Adenine/administration & dosage , Adenine/therapeutic use , Thailand , United States , South Africa , Drug Combinations , Uganda , Viral Load/drug effects
15.
J Antimicrob Chemother ; 79(5): 1126-1132, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38530862

ABSTRACT

OBJECTIVE: Large inter-individual variability in the pharmacokinetics of rilpivirine and cabotegravir has been reported in the first weeks after starting long-acting injectable (LAI) therapy. Here, we assessed the distribution of rilpivirine and cabotegravir trough concentrations in people with HIV (PWH) on long-term LAI treatment. METHODS: Adult PWH treated with LAI for at least 32 weeks with an assessment of drug plasma trough concentrations were considered. The proportion of rilpivirine and cabotegravir plasma trough concentrations below four-times the protein-adjusted concentrations required for 90% inhibition of viral replication (4×PA-IC90) was estimated. RESULTS: Sixty-seven PWH were identified. LAI treatment duration was 216 ±â€Š80 weeks (range 32-320 weeks). Cabotegravir concentrations were associated with lower inter-individual variability compared with rilpivirine (45% versus 84%; P < 0.05). No differences were found in rilpivirine (160 ±â€Š118 versus 189 ±â€Š81 ng/mL; P = 0.430) and cabotegravir (1758 ±â€Š807 versus 1969 ±â€Š802 ng/mL; P = 0.416) trough concentrations in males (n = 55) versus females (n = 12). A non-significant trend for lower cabotegravir concentrations was found in PWH with a body mass index >30 kg/m2 (n = 9) versus non-obese participants (1916 ±â€Š905 versus 1606 ±â€Š576 ng/mL; P = 0.131). Three out of the 67 PWH had at least one drug concentration <4×PA-IC90: 100% of PWH had undetectable HIV viral load. CONCLUSIONS: At steady state, optimal systemic exposure of cabotegravir and rilpivirine was found in most PWH; cabotegravir trough concentrations were associated with lower inter-individual variability compared with rilpivirine. The study was not powered to assess the contribution of sex and/or body weight on LAI exposure due to the small number of females and obese PWH included.


Subject(s)
Anti-HIV Agents , Diketopiperazines , HIV Infections , Pyridones , Rilpivirine , Humans , Rilpivirine/pharmacokinetics , Rilpivirine/administration & dosage , Rilpivirine/therapeutic use , Rilpivirine/blood , Male , Female , HIV Infections/drug therapy , Middle Aged , Adult , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/blood , Anti-HIV Agents/therapeutic use , Aged , Injections , Viral Load/drug effects
17.
Clin Pharmacol Ther ; 115(6): 1450-1459, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38519844

ABSTRACT

Long-acting cabotegravir has been studied mainly in the stringent framework of clinical trials, which does not necessarily reflect the situation of people with HIV (PWH) in routine clinical settings. The present population pharmacokinetic analysis aims to build real-world reference percentile curves of cabotegravir concentrations, accounting for patient-related factors that may affect cabotegravir exposure. The second objective is to simulate whether dosing interval adjustments of cabotegravir could be considered in specific subpopulations. Overall, 238 PWH contributed to 1,038 cabotegravir levels (186 during the initial oral administration phase and 852 after intramuscular injection). Cabotegravir pharmacokinetics was best described using a one-compartment model with distinct first order-absorption for oral and intramuscular administrations, and identical volume and clearance. Our model showed almost 40% faster absorption and 30% higher clearance than previously reported, resulting in a time to steady-state of 8 months and an elimination half-life of 4.6 weeks for long-acting cabotegravir. Sex and body mass index significantly influenced absorption, and bodyweight affected clearance. Model-based simulations showed that cabotegravir trough concentrations in females were 25% lower 4 weeks after the intramuscular loading dose, but 42% higher during the late maintenance phase. Finally, simulations indicated that in females, despite significantly higher cabotegravir concentrations, longer intervals between injections may not consistently ensure levels above the 4-fold protein-adjusted 90% inhibitory target concentration.


Subject(s)
HIV Infections , Models, Biological , Pyridones , Humans , Injections, Intramuscular , Female , Male , HIV Infections/drug therapy , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Adult , Administration, Oral , Middle Aged , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/administration & dosage , Half-Life , Delayed-Action Preparations/pharmacokinetics , Young Adult , Aged , Diketopiperazines
19.
Cancer Chemother Pharmacol ; 93(5): 509-517, 2024 May.
Article in English | MEDLINE | ID: mdl-38520556

ABSTRACT

Tazemetostat, a novel oral selective inhibitor of enhancer of zeste homolog 2 (EZH2), was approved by the Food and Drug Administration (FDA) in 2020 for use in patients with advanced epithelioid sarcoma or relapsed/refractory (R/R) EZH2-mutated follicular lymphoma. These indications were approved by the FDA trough accelerated approval based on objective response rate and duration of response that resulted from phase 2 clinical trials. Tazemetostat competes with S-adenosylmethionine (SAM) cofactor to inhibit EZH2, reducing the levels of trimethylated lysine 27 of histone 3 (H3K27me3), considered as pharmacodynamic marker. Tazemetostat is orally bioavailable, characterized by rapid absorption and dose-proportional exposure, which is not influenced by coadministration with food or gastric acid reducing agents. It highly distributes in tissues, but with limited access to central nervous system. Tazemetostat is metabolized by CYP3A in the liver to 3 major inactive metabolites (M1, M3, and M5), has a short half-life and is mainly excreted in feces. Drug-drug interactions were shown with moderate CYP3A inhibitors as fluconazole, leading the FDA to recommend a 50% dose reduction, while studies investigating coadministration of tazemetostat with strong inhibitors/inducers are ongoing. No dosage modifications are recommended based on renal or hepatic dysfunctions. Overall, tazemetostat is the first-in-class EZH2 inhibitor approved by the FDA for cancer treatment. Current clinical studies are evaluating combination therapies in patients with several malignancies.


Subject(s)
Benzamides , Biphenyl Compounds , Drug Interactions , Morpholines , Humans , Morpholines/pharmacokinetics , Morpholines/pharmacology , Morpholines/administration & dosage , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Biphenyl Compounds/administration & dosage , Pyridones/pharmacokinetics , Pyridones/pharmacology , Pyridones/administration & dosage , Pyridones/therapeutic use , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Sulfonamides/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Animals , Organophosphates/pharmacokinetics , Organophosphates/pharmacology
20.
Antimicrob Agents Chemother ; 68(4): e0140023, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38415667

ABSTRACT

Contezolid is a novel oxazolidinone antibiotic with a promising safety profile. Oral contezolid and its intravenous (IV) prodrug contezolid acefosamil (CZA) are in development for treatment of diabetic foot and acute bacterial skin and skin structure infections (ABSSSI). The prodrug CZA is converted to active contezolid via intermediate MRX-1352. This study aimed to provide the pharmacokinetic rationale for safe, effective, and flexible dosage regimens with initial IV CZA followed by oral contezolid. We simultaneously modeled plasma concentrations from 110 healthy volunteers and 74 phase 2 patients with ABSSSI via population pharmacokinetics (using the importance sampling estimation algorithm), and optimized dosage regimens by Monte Carlo simulations. This included data on MRX-1352, contezolid, and its metabolite MRX-1320 from 66 healthy volunteers receiving intravenous CZA (150-2400 mg) for up to 28 days, and 74 patients receiving oral contezolid [800 mg every 12 h (q12h)] for 10 days. The apparent total clearance for 800 mg oral contezolid with food was 16.0 L/h (23.4% coefficient of variation) in healthy volunteers and 17.7 L/h (53.8%) in patients. CZA was rapidly converted to MRX-1352, which subsequently transformed to contezolid. The proposed dosage regimen used an IV CZA 2000 mg loading dose with 1000 mg IV CZA q12h as maintenance dose(s), followed by 800 mg oral contezolid q12h (with food). During each 24-h period, Monte Carlo simulations predicted this regimen to achieve consistent areas under the curve of 91.9 mg·h/L (range: 76.3-106 mg·h/L) under all scenarios. Thus, this regimen was predicted to reliably achieve efficacious contezolid exposures independent of timing of switch from IV CZA to oral contezolid.IMPORTANCEThis study provides the population pharmacokinetic rationale for the dosage regimen of the intravenous (IV) prodrug contezolid acefosamil (CZA) followed by oral contezolid. We developed the first integrated population model for the pharmacokinetics of the MRX-1352 intermediate prodrug, active contezolid, and its main metabolite MRX-1320 based on data from three clinical studies in healthy volunteers and phase 2 patients. The proposed regimen was predicted to reliably achieve efficacious contezolid exposures independent of timing of switch from IV CZA to oral contezolid.


Subject(s)
Oxazolidinones , Prodrugs , Humans , Anti-Bacterial Agents/pharmacokinetics , Oxazolidinones/pharmacokinetics , Pyridones/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...