Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.548
Filter
1.
Eur J Pharm Biopharm ; 199: 114313, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718842

ABSTRACT

The aim of the present study was to investigate the gastroretentive capacity of different formulation principles. This was indirectly determined by the absorption behavior of caffeine from the dosage forms. A slow and continuous appearance of caffeine in the saliva of healthy volunteers was used as a parameter for a prolonged gastric retention time. For this purpose, a four-way study was conducted with twelve healthy volunteers using the following test procedures: (1) Effervescent granules with 240 mL of still water administered in fed state, (2) effervescent granules with 20 mL of still water in fed state, (3) extended release (ER) tablet with 240 mL of still water in fed state, and (4) effervescent granules with 240 mL of still water in fasted state. The initial rise of the caffeine concentrations was more pronounced after the intake of the effervescent granules in the fed state compared to that of the ER tablets. However, tmax tended to be shorter in the fed study arms following administration of the ER tablet compared to the granules. Overall, the application of active pharmaceutical ingredients formulated as effervescent granules seems to be a promising approach to increase their gastric residence time after intake in fed state.


Subject(s)
Caffeine , Delayed-Action Preparations , Tablets , Humans , Caffeine/administration & dosage , Caffeine/pharmacokinetics , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Male , Adult , Young Adult , Female , Fasting , Administration, Oral , Saliva/metabolism , Saliva/chemistry , Healthy Volunteers , Gastric Mucosa/metabolism , Cross-Over Studies , Stomach/drug effects
2.
Acta Biomater ; 180: 423-435, 2024 May.
Article in English | MEDLINE | ID: mdl-38641183

ABSTRACT

Communication between tumors and lymph nodes carries substantial significance for antitumor immunotherapy. Remodeling the immune microenvironment of tumor-draining lymph nodes (TdLN) plays a key role in enhancing the anti-tumor ability of immunotherapy. In this study, we constructed a biomimetic artificial lymph node structure composed of F127 hydrogel loading effector memory T (TEM) cells and PD-1 inhibitors (aPD-1). The biomimetic lymph nodes facilitate the delivery of TEM cells and aPD-1 to the TdLN and the tumor immune microenvironment, thus realizing effective and sustained anti-tumor immunotherapy. Exploiting their unique gel-forming and degradation properties, the cold tumors were speedily transformed into hot tumors via TEM cell supplementation. Meanwhile, the efficacy of aPD-1 was markedly elevated compared with conventional drug delivery methods. Our finding suggested that the development of F127@TEM@aPD-1 holds promising potential as a future novel clinical drug delivery technique. STATEMENT OF SIGNIFICANCE: F127@TEM@aPD-1 show unique advantages in cancer treatment. When injected subcutaneously, F127@TEM@aPD-1 can continuously supplement TEM cells and aPD-1 to tumor draining lymph nodes (TdLN) and the tumor microenvironment, not only improving the efficacy of ICB therapy through slow release, but also exhibiting dual regulatory effects on the tumor and TdLN.


Subject(s)
Delayed-Action Preparations , Hydrogels , Lymph Nodes , Memory T Cells , Programmed Cell Death 1 Receptor , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/pathology , Lymph Nodes/immunology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Memory T Cells/drug effects , Memory T Cells/immunology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/pharmacokinetics , Tumor Microenvironment/drug effects , Cell Line, Tumor , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Female , Mice, Inbred C57BL , Humans
3.
Mol Pharm ; 21(5): 2394-2405, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38647653

ABSTRACT

Doxorubicin (DOX) is one of the most commonly used anticancer drugs; however, its clinical application is greatly limited due to its toxicity and chemotherapy resistance. The delivery of DOX by liposomes (Lipos) can improve the blood circulation time in vivo and reduce toxic side effects, but the drug's accumulation in the tumor is often insufficient for effective treatment. In this study, we present a calcium cross-linked liposome gel for the encapsulation of DOX, demonstrating its superior long-term release capabilities compared to conventional Lipos. By leveraging this enhanced long-term release, we can enhance drug accumulation within tumors, ultimately leading to improved antitumor efficacy. Lipos were prepared using the thin-film dispersion method in this study. We utilized the ion-responsiveness of glutathione-gelatin (GSH-GG) to form the gel outside the Lipos and named the nanoparticles coated with GSH-GG on the outside of Lipos as Lipos@GSH-GG. The average size of Lipos@GSH-GG was around 342.9 nm, with a negative charge of -25.6 mV. The in vitro experiments revealed that Lipos@GSH-GG exhibited excellent biocompatibility and slower drug release compared to conventional Lipos. Further analysis of cellular uptake and cytotoxicity demonstrated that Lipos@GSH-GG loading DOX (DOX&Lipos@GSH-GG) exhibited superior long-term release effects and lower toxic side effects compared to Lipos loading DOX (DOX&Lipos). Additionally, the findings regarding the long-term release effect in vivo and the tumor accumulation within tumor-bearing mice of Lipos@GSH-GG suggested that, compared to Lipos, it demonstrated superior long-term release capabilities and achieved greater drug accumulation within tumors. In vivo antitumor efficacy experiments showed that DOX&Lipos@GSH-GG demonstrated superior antitumor efficacy to DOX&Lipos. Our study highlights Lipos@GSH-GG as a promising nanocarrier with the potential to enhance efficacy and safety by means of long-term release effects and may offer an alternative approach for effective antitumor therapy in the future.


Subject(s)
Calcium , Doxorubicin , Drug Liberation , Glutathione , Liposomes , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Animals , Mice , Liposomes/chemistry , Humans , Calcium/chemistry , Calcium/metabolism , Glutathione/chemistry , Female , Gels/chemistry , Gelatin/chemistry , Mice, Nude , Nanoparticles/chemistry , Mice, Inbred BALB C , Cell Line, Tumor , Xenograft Model Antitumor Assays , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Cross-Linking Reagents/chemistry , Drug Delivery Systems/methods
4.
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
5.
CPT Pharmacometrics Syst Pharmacol ; 13(5): 781-794, 2024 May.
Article in English | MEDLINE | ID: mdl-38429889

ABSTRACT

There is growing interest in the use of long-acting (LA) injectable drugs to improve treatment adherence. However, their long elimination half-life complicates the conduct of clinical trials. Physiologically-based pharmacokinetic (PBPK) modeling is a mathematical tool that allows to simulate unknown clinical scenarios for LA formulations. Thus, this work aimed to develop and verify a mechanistic intramuscular PBPK model. The framework describing the release of a LA drug from the depot was developed by including both the physiology of the injection site and the physicochemical properties of the drug. The framework was coded in Matlab® 2020a and implemented in our existing PBPK model for the verification step using clinical data for LA cabotegravir, rilpivirine, and paliperidone. The model was considered verified when the simulations were within twofold of observed data. Furthermore, a local sensitivity analysis was conducted to assess the impact of various factors relevant for the drug release from the depot on pharmacokinetics. The PBPK model was successfully verified since all predictions were within twofold of observed clinical data. Peak concentration, area under the concentration-time curve, and trough concentration were sensitive to media viscosity, drug solubility, drug density, and diffusion layer thickness. Additionally, inflammation was shown to impact the drug release from the depot. The developed framework correctly described the release and the drug disposition of LA formulations upon intramuscular administration. It can be implemented in PBPK models to address pharmacological questions related to the use of LA formulations.


Subject(s)
Computer Simulation , Models, Biological , Rilpivirine , Humans , Injections, Intramuscular , Rilpivirine/pharmacokinetics , Rilpivirine/administration & dosage , Paliperidone Palmitate/pharmacokinetics , Paliperidone Palmitate/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Male , Adult , Anti-Retroviral Agents/pharmacokinetics , Anti-Retroviral Agents/administration & dosage , Drug Liberation , Middle Aged , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/administration & dosage , Female , Pyridones , Diketopiperazines
6.
Epilepsy Res ; 202: 107350, 2024 May.
Article in English | MEDLINE | ID: mdl-38513537

ABSTRACT

OBJECTIVES: Assess the bioequivalence of lacosamide extended-release (XR) capsules and immediate-release (IR) tablets and answer real-world clinical questions regarding the use of lacosamide XR. METHODS: An open-label, randomized, two-treatment, two-sequence, oral comparative bioavailability study was conducted to assess the bioequivalence of two lacosamide formulations. Participants were randomized 1:1 to receive lacosamide XR capsules (400 mg once-daily) or IR tablets (200 mg twice-daily) in 1 of 2 sequences over 7-day periods. Primary outcome was the area under the lacosamide concentration-time curve over 24 h at steady-state (AUC0-τ,ss). Secondary outcomes were maximum (Cmax,ss) and minimum concentrations at steady-state (Cmin,ss). Bioequivalence was established when 90% confidence intervals (CIs) for geometric least square means ratios (GLSMs) were between 80% and 125%. Adverse events (AEs) and other safety outcomes were also assessed. Pharmacokinetic simulations, including adherent and partially adherent dosing scenarios with XR and IR formulations, modeled the clinical use of lacosamide XR. RESULTS: Thirty-five healthy adult males were enrolled in the bioequivalence study. After 7 days of study drug, mean AUC0-τ,ss, Cmax,ss, and Cmin,ss values were similar between XR and IR formulations; all 90% CIs for GLSMs were between 80% and 125%. AEs were mild and no serious AEs or other clinically significant safety findings were observed. Pharmacokinetic simulations suggested that partial adherence affected formulations similarly; and the best strategy for switching formulations was to take the morning lacosamide IR dose followed by the evening lacosamide XR dose, as this resulted in the most consistent lacosamide plasma concentrations. CONCLUSIONS: Once-daily lacosamide XR capsules were bioequivalent to twice-daily lacosamide IR tablets. Pharmacokinetic simulations indicated lacosamide XR and IR formulations were similarly affected by partial adherence, though once-daily dosing with lacosamide XR may offer clinical advantages, and formulations can be easily switched. These results support the use of lacosamide XR capsules as a once-daily alternative to lacosamide IR tablets.


Subject(s)
Anticonvulsants , Capsules , Delayed-Action Preparations , Lacosamide , Tablets , Therapeutic Equivalency , Humans , Lacosamide/pharmacokinetics , Lacosamide/administration & dosage , Male , Adult , Anticonvulsants/pharmacokinetics , Anticonvulsants/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Young Adult , Female , Middle Aged , Biological Availability , Area Under Curve , Adolescent , Computer Simulation , Administration, Oral
7.
J Pharm Sci ; 113(6): 1653-1663, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38382809

ABSTRACT

Drug-Combination Nanoparticles (DcNP) are a novel drug delivery system designed for synchronized delivery of multiple drugs in a single, long-acting, and targeted dose. Unlike depot formulations, slowly releasing drug at the injection site into the blood, DcNP allows multiple-drug-in-combination to collectively distribute from the injection site into the lymphatic system. Two distinct classes of long-acting injectables products are proposed based on pharmacokinetic mechanisms. Class I involves sustained release at the injection site. Class II involves a drug-carrier complex composed of lopinavir, ritonavir, and tenofovir uptake and retention in the lymphatic system before systemic access as a part of the PBPK model validation. For clinical development, Class II long-acting drug-combination products, we leverage data from 3 nonhuman primate studies consisting of nine PK datasets: Study 1, varying fixed-dose ratios; Study 2, short multiple dosing with kinetic tails; Study 3, long multiple dosing (chronic). PBPK validation criteria were established to validate each scenario for all drugs. The models passed validation in 8 of 9 cases, specifically to predict Study 1 and 2, including PK tails, with ritonavir and tenofovir, fully passing Study 3 as well. PBPK model for lopinavir in Study 3 did not pass the validation due to an observable time-varying and delayed drug accumulation, which likely was due to ritonavir's CYP3A inhibitory effect building up during multiple dosing that triggered a mechanism-based drug-drug interaction (DDI). Subsequently, the final model enables us to account for this DDI scenario.


Subject(s)
Anti-HIV Agents , Drug Combinations , Lopinavir , Models, Biological , Nanoparticles , Ritonavir , Tenofovir , Ritonavir/pharmacokinetics , Ritonavir/administration & dosage , Lopinavir/pharmacokinetics , Lopinavir/administration & dosage , Tenofovir/pharmacokinetics , Tenofovir/administration & dosage , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Male , Drug Delivery Systems/methods , Humans
8.
Adv Healthc Mater ; 13(12): e2303134, 2024 May.
Article in English | MEDLINE | ID: mdl-38348511

ABSTRACT

The effective repair of large bone defects remains a major challenge due to its limited self-healing capacity. Inspired by the structure and function of the natural periosteum, an electrospun biomimetic periosteum is constructed to programmatically promote bone regeneration using natural bone healing mechanisms. The biomimetic periosteum is composed of a bilayer with an asymmetric structure in which an aligned electrospun poly(ε-caprolactone)/gelatin/deferoxamine (PCL/GEL/DFO) layer mimics the outer fibrous layer of the periosteum, while a random coaxial electrospun PCL/GEL/aspirin (ASP) shell and PCL/silicon nanoparticles (SiNPs) core layer mimics the inner cambial layer. The bilayer controls the release of ASP, DFO, and SiNPs to precisely regulate the inflammatory, angiogenic, and osteogenic phases of bone repair. The random coaxial inner layer can effectively antioxidize, promoting cell recruitment, proliferation, differentiation, and mineralization, while the aligned outer layer can promote angiogenesis and prevent fibroblast infiltration. In particular, different stages of bone repair are modulated in a rat skull defect model to achieve faster and better bone regeneration. The proposed biomimetic periosteum is expected to be a promising candidate for bone defect healing.


Subject(s)
Biomimetic Materials , Bone Regeneration , Periosteum , Polyesters , Bone Regeneration/drug effects , Animals , Periosteum/drug effects , Rats , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Polyesters/chemistry , Rats, Sprague-Dawley , Deferoxamine/pharmacology , Deferoxamine/chemistry , Gelatin/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/pharmacokinetics , Osteogenesis/drug effects , Skull/drug effects , Skull/injuries , Male , Nanoparticles/chemistry , Tissue Engineering/methods , Cell Differentiation/drug effects , Tissue Scaffolds/chemistry
9.
Ther Drug Monit ; 46(3): 309-320, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38176856

ABSTRACT

PURPOSE: Torasemide is a potassium-sparing loop diuretic used to treat fluid retention associated with congestive heart failure and kidney and hepatic diseases. This systematic review was conducted to combine all accessible data on the pharmacokinetics (PK) of torasemide in healthy and diseased populations, which may help clinicians avert adverse drug reactions and determine the correct dosage regimen. METHODS: Four databases were systematically searched to screen for studies associated with the PK of torasemide, and 21 studies met the eligibility criteria. The review protocol was registered in the PROSPERO database (CRD42023390178). RESULTS: A decrease in maximum plasma concentration (C max ) was observed for torasemide after administration of the prolonged-release formulation in comparison to that after administration of the immediate-release formulation, that is, 1.12 ± 0.17 versus 1.6 ± 0.2 mcg/mL. After administering an oral dose of torasemide, a 2-fold increase in the area under the concentration-time curve (AUC) was reported in patients with congestive heart failure compared with the healthy population. Moreover, the patients with renal failure (clearance < 30 mL/min) showed an increase in value of AUC 0-∞ that is, 42.9 versus 8.091 mcg.h -1 .mL -1 compared with healthy subjects. In addition, some studies have reported interactions with different drugs, in which irbesartan showed a slight increase in the AUC 0-∞ of torasemide, whereas losartan and empagliflozin did not. CONCLUSIONS: The current review summarizes all available PK parameters of torasemide that may be beneficial for avoiding drug-drug interactions in subjects with renal and hepatic dysfunction and for predicting doses in patients with different diseases.


Subject(s)
Torsemide , Humans , Torsemide/pharmacokinetics , Heart Failure/drug therapy , Diuretics/pharmacokinetics , Area Under Curve , Delayed-Action Preparations/pharmacokinetics
10.
Neuropsychopharmacology ; 49(6): 1050-1057, 2024 May.
Article in English | MEDLINE | ID: mdl-38200140

ABSTRACT

Buprenorphine is used to treat opioid use disorder (OUD). Weekly and monthly subcutaneous long-acting buprenorphine injections (CAM2038) provide more stable buprenorphine plasma levels and reduce the treatment burden, misuse, and diversion associated with sublingual transmucosal buprenorphine formulations. To characterize the pharmacokinetic/pharmacodynamic (PK/PD) relationship, a maximum inhibition (Imax) model was developed relating CAM2038 buprenorphine plasma concentration to drug liking maximum effect (Emax) visual analog scale (VAS; bipolar) score after intramuscular hydromorphone administration. Data included time-matched observations of buprenorphine plasma concentration and drug liking Emax VAS score after hydromorphone 18 mg administration in 47 non-treatment-seeking adults with moderate to severe OUD in a phase 2 study. Analysis used non-|linear mixed-effects modeling (NONMEM®). The final Imax model adequately described the PK/PD relationship between buprenorphine plasma concentration and drug liking Emax VAS score. Simulations showed drug liking was effectively blocked at low buprenorphine plasma concentrations (0.4 ng/mL) where the upper 95% confidence interval of the drug liking Emax VAS score was below the pre-defined 11-point complete blockade threshold. The buprenorphine plasma concentration required to achieve 90% of the maximal effect (IC90) of drug liking was 0.675 ng/mL. Interindividual variability in responses to buprenorphine was observed; some participants experienced fluctuating responses, and a few did not achieve drug liking blockade even with higher buprenorphine plasma concentrations. This affirms the need to individualize treatment and titrate doses for optimal treatment outcomes. PK/PD models were also developed for desire to use VAS and Clinical Opiate Withdrawal Scale (COWS) scores, with results aligned to those for drug liking.


Subject(s)
Buprenorphine , Opioid-Related Disorders , Adult , Female , Humans , Male , Middle Aged , Young Adult , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Buprenorphine/pharmacokinetics , Buprenorphine/administration & dosage , Buprenorphine/pharmacology , Delayed-Action Preparations/pharmacokinetics , Hydromorphone/pharmacokinetics , Hydromorphone/administration & dosage , Hydromorphone/pharmacology , Injections, Subcutaneous , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy
11.
Int J Pharm ; 652: 123869, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38296171

ABSTRACT

Food and formulation characteristics are crucial factors affecting the gastrointestinal release and absorption kinetics of oral solid preparations. In the present study, the dynamic continuous release and bioaccessibility of metformin hydrochloride immediate-release (IR) and sustained-release (SR) tablets were investigated in the dynamic human stomach-intestine (DHSI-IV) system simulating fasted and fed states in healthy adults. Both tablet formulations (particularly IR tablet) exhibited a postponed release in the fed state compared to the fasted state. Correspondingly, the bioaccessible fraction of metformin from IR tablets in the presence of high-fat meal was significantly reduced to 76.2 % of the fasted state. However, the in vitro bioaccessibility was less impaired by food for SR tablets with a fed/fasted ratio of 95.5 %. A convolution-based approach was used to convert in vitro bioaccessibility results to plasma concentration data. The predicted plasma concentration curve showed good agreement with human data in terms of pharmacokinetic (PK) parameters. In the fasted state, the predicted Cmax, Tmax and AUC0-24h of IR tablets were 943.9 ± 25.7 ng/mL, 2.0 ± 0.4 h and 7090.7 ± 112.0 ng.h/mL, respectively, mirroring values observed in healthy subjects. Overall, the DHSI-IV system has demonstrated potential to assess and predict the impact of meal intake on the in vivo release and absorption behaviors of oral solid preparations.


Subject(s)
Fasting , Metformin , Adult , Humans , Administration, Oral , Gastrointestinal Tract , Tablets , Delayed-Action Preparations/pharmacokinetics , Cross-Over Studies , Biological Availability , Area Under Curve
12.
J Clin Pharmacol ; 64(4): 428-436, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38084781

ABSTRACT

Data on the pharmacokinetics of tacrolimus during pregnancy are limited. Therefore, the aim of this retrospective study was to characterize the whole-blood pharmacokinetics of tacrolimus throughout pregnancy. In this single-center retrospective cohort study, whole-blood tacrolimus trough concentrations corrected for the dose (concentration-to-dose [C/D] ratios) were compared before, monthly during, and after pregnancy in kidney, liver, and lung transplant recipients who became pregnant and gave birth between 2000 and 2022. Descriptive statistics and linear mixed models were used to characterize changes in tacrolimus C/D ratios before, during, and after pregnancy. The total study population included 46 pregnancies (31 pregnant women). Nineteen, 21, and 6 pregnancies were following kidney, liver, and lung transplantation, respectively. Immediate-release or extended-release formulations were used in 54.5% and 45.5% of the women, respectively. Tacrolimus C/D ratios significantly (P < .001) decreased (-48%) compared to the prepregnancy state at 7 months of pregnancy. These ratios recovered within 3 months postpartum (P = .002). C/D ratios tended to be lower during treatment with an extended-release formulation than with an immediate-release formulation (P = .071). Transplantation type did not significantly affect C/D ratios during pregnancy (P = .873). In conclusion, we found that tacrolimus whole-blood pharmacokinetics change throughout pregnancy, with the lowest C/D ratios (48% decrease) in the 7th month of pregnancy. In general, the decrease in C/D ratios seems to stabilize from month 4 onward compared to prepregnancy.


Subject(s)
Kidney Transplantation , Tacrolimus , Pregnancy , Humans , Female , Tacrolimus/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Retrospective Studies , Transplant Recipients , Drug Administration Schedule , Delayed-Action Preparations/pharmacokinetics
13.
Clin Pharmacol Drug Dev ; 13(2): 122-127, 2024 02.
Article in English | MEDLINE | ID: mdl-37548105

ABSTRACT

Vildagliptin is one of the dipeptidyl peptidase-4 inhibitors. This study aimed to compare vildagliptin exposure between 50-mg immediate-release (IR) and 100-mg new sustained-release (SR) tablets, and evaluate the food effect on the pharmacokinetics (PKs) of vildagliptin. A randomized, open-label, 3-period, 3-treatment, 6-sequence crossover study was conducted on healthy subjects. During each period, subjects received the SR tablet either in the fasted (T1) or high-fat fed (T2) state once a day, or IR tablets administered twice a day in the fasted state (R). Blood samples for PK analysis were obtained serially up to 24 hours after dosing. Thirty-four subjects completed the study. The geometric mean ratios for the Cmax and AUC0-24h of T1 to R were 1.15 and 0.89, respectively. The corresponding values of T2 to T1 were 0.94 and 1.07, respectively. Vildagliptin exposure over 24 hours was similar between the SR and IR tablets. In addition, the PK profiles of the SR tablets were not altered by food. The SR tablets can be administered without a food effect and be an alternative option to IR tablets.


Subject(s)
Vildagliptin , Humans , Healthy Volunteers , Cross-Over Studies , Delayed-Action Preparations/pharmacokinetics , Tablets
14.
CPT Pharmacometrics Syst Pharmacol ; 13(1): 132-142, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37864318

ABSTRACT

Delayed-release and extended-release methylphenidate hydrochloride (JORNAY PM®) is a novel capsule formulation of methylphenidate hydrochloride, used to treat attention deficit hyperactivity disorder in patients 6 years and older. In this paper, we develop a Level A in vitro-in vivo correlation (IVIVC) model for extended-release methylphenidate hydrochloride to support post-approval manufacturing changes by evaluating a point-to-point correlation between the fraction of drug dissolved in vitro and the fraction of drug absorbed in vivo. Dissolution data from an in vitro study of three different release formulations: fast, medium, and slow, and pharmacokinetic data from two in vivo studies were used to develop an IVIVC model using a convolution-based approach. The time-course of the drug concentration resulting from an arbitrary dose was considered as a function of the in vivo drug absorption and the disposition and elimination processes defined by the unit impulse response function using the convolution integral. An IVIVC was incorporated in the model due to the temporal difference seen in the scatterplots of the estimated fraction of drug absorbed in vivo and the fraction of drug dissolved in vitro and Levy plots. Finally, the IVIVC model was subjected to evaluation of internal predictability. This IVIVC model can be used to predict in vivo profiles for different in vitro profiles of extended-release methylphenidate hydrochloride.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Methylphenidate , Humans , Delayed-Action Preparations/pharmacokinetics , Attention Deficit Disorder with Hyperactivity/drug therapy , Area Under Curve
15.
Eur J Drug Metab Pharmacokinet ; 49(2): 149-170, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38127227

ABSTRACT

BACKGROUND AND OBJECTIVE: Attention deficit hyperactivity disorder is one of the most common neuropsychiatric conditions in children, and methylphenidate (MPH) is one of the first-line therapies. MPH is available in a variety of extended-release (ER) formulations worldwide, and most formulations are not considered bioequivalent due to differences in pharmacokinetics. It is hypothesized that the current bioequivalence guidelines from the different regulatory bodies may generate inconsistent findings or recommendations when assessing the bioequivalence of ER MPH formulations. This manuscript aims to conduct a comprehensive and narrative critical literature review to analyze pharmacokinetic data pertaining to ER formulations of MPH in order to assess bioequivalence, differences in regulatory guidelines, and additional pharmacokinetic-pharmacodynamic parameters that may help define interchangeability. METHODS: A literature search was conducted in EMBASE, Medline, and Cochrane Library with no time limits. Study characteristics, non-compartmental pharmacokinetic parameters, and bioequivalence data were extracted for analysis. RESULTS: Thirty-three studies were identified with primary pharmacokinetic data after the administration of ER MPH, of which 10 were direct comparative studies (i.e., at least 2 formulations tested within a single setting) and 23 were indirect comparisons (i.e., different experimental settings). Two formulations were consistently reported as bioequivalent across the regulatory bodies using criteria from their guidance documents, although inconsistencies have been observed. However, when additional kinetic criteria (discussed in this manuscript) were imposed, only one study met the more stringent definition of bioequivalence. Various clinical factors also had inconsistent effects on the pharmacokinetics and interchangeability of the different formulations, which were associated with a lack of standardization for assessing covariates across the regulatory agencies. CONCLUSION: Additional pharmacokinetic parameters and consistency in guidelines across the regulatory bodies may improve bioequivalence assessments. Based on our findings, more research is also required to understand whether bioequivalence is an appropriate measure for determining MPH interchangeability. This critical review is suitable for formulation scientists, clinical pharmacologists, and clinicians.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Central Nervous System Stimulants , Methylphenidate , Child , Humans , Methylphenidate/pharmacokinetics , Methylphenidate/therapeutic use , Central Nervous System Stimulants/pharmacokinetics , Therapeutic Equivalency , Attention Deficit Disorder with Hyperactivity/drug therapy , Delayed-Action Preparations/pharmacokinetics , Cross-Over Studies
16.
Drug Des Devel Ther ; 17: 3551-3561, 2023.
Article in English | MEDLINE | ID: mdl-38046282

ABSTRACT

Purpose: To develop an UPLC-MS/MS method for the quantitative analysis of pentoxifylline in beagle dog plasma and apply it to a pharmacokinetic study of food effect. Methods: Sample separation was achieved using a Kinetex Phenyl-Hexyl column (50 × 2.1 mm, 1.7 µm) with a gradient elution program in 5.5 min after a simple protein precipitation with methanol. Using the mobile phase that made up by 0.2% formic acid and 5mM ammonium formate water (A) and methanol (B). Quantitation was carried out using the positive ionization mode with multiple reaction monitoring (MRM). A randomized, single-dose, two-period crossover study was conducted in six fasted or fed beagles that received 400 mg pentoxifylline sustained-release tablets (Brand name: Shuanling™, CSPC Pharmaceutical Group). WinNonlin® software was used to calculate pharmacokinetic parameters. Results: The linear calibration range was 2-1000 ng/mL (r2> 0.99). Both intra- and inter-batch precision were less than 6.27%, and the accuracy ranged from 88.65% to 97.18%. Pentoxifylline was readily absorbed in fasted and fed dogs administered a dose of 400 mg (tmax:1.54h vs 1.83h). Compared to the fasted group, the AUC0→t and Cmax in the fed group increased by 1.71-fold and 1.30-fold, respectively. In the fasted group, the AUC0→t and Cmax values were 4684.08 h•ng/mL and 2402.33 ng/mL, respectively. In the fed group, these values were 8027.75 h•ng/mL and 3119.67 ng/mL. The difference in AUC0-t between the fed and fasted group was statistically significant. Conclusion: The novel optimized UPLC-MS/MS assay is an effective tool for the determination of pentoxifylline and has been successfully applied in pharmacokinetic studies of pentoxifylline in beagle dogs. The administration of pentoxifylline sustained-release tablets with food significantly increased the area under the time curve, and it is recommended that they should be administered during or shortly after feeding.


Subject(s)
Pentoxifylline , Tandem Mass Spectrometry , Animals , Dogs , Chromatography, High Pressure Liquid/methods , Chromatography, Liquid/methods , Cross-Over Studies , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/analysis , Delayed-Action Preparations/pharmacokinetics , Methanol , Pentoxifylline/administration & dosage , Pentoxifylline/blood , Pentoxifylline/pharmacokinetics , Reproducibility of Results , Tandem Mass Spectrometry/methods
17.
Drug Des Devel Ther ; 17: 2369-2381, 2023.
Article in English | MEDLINE | ID: mdl-37600497

ABSTRACT

Background: Pramipexole dihydrochloride extended-release tablet is a novel long-acting form of non-ergot dopamine agonist indicated as one of main therapeutic approaches for Parkinson's disease. However, pharmacokinetic properties of extended-release pramipexole in healthy Chinese subjects remain unclear. Methods: A single-center, randomized, open-label, two-period crossover, single-dose study was performed to investigate comparative pharmacokinetics and evaluate bioequivalence of 0.375 mg test (Yangtze River Pharmaceutical Group Co., Ltd.) and reference (Trade name: Sifrol®, Boehringer Ingelheim Pharma GmbH & Co. KG) formulations of pramipexole dihydrochloride extended-release tablets in healthy Chinese subjects under fasted and fed states. Results: A total of 56 subjects (28 in each dietary trial) were enrolled and randomized. After single dose of 0.375 mg test and reference formulations under fasted condition, main pharmacokinetics of pramipexole were as follows: peak concentration (Cmax) were 409.33±95.93 and 413.77±132.03 pg/mL; plasma area under concentration-time curve from time 0 to last measurable concentration (AUC0-t) were 8801.95±1966.83 and 8646.37±2600.49 h*pg/mL; AUC from time 0 to infinity (AUC0-∞) were 9469.03±1991.61 and 9082.95±2666.26 h*pg/mL; elimination half-life (t1/2) were 11.98±3.91 and 9.85±2.63 h; both time to reach Cmax (Tmax) were about 4.50 h, respectively, for test and reference formulations. The 90% confidence intervals of geometric mean ratios (test/reference) of Cmax, AUC0-t and AUC0-∞ under fasted and fed conditions were all within 80-125%. Following administration under fed condition, Cmax and Tmax for both test and reference formulations slightly increased and prolonged to 5.0 h, respectively, but AUC approximately remained unchanged compared with dosing under fasted condition. Test and reference formulations showed similar bioequivalence and favorable safety under fasted and fed states. Conclusion: Test and reference formulations of pramipexole dihydrochloride extended-release tablets (0.375 mg) showed similar bioequivalence and well safety and tolerability in healthy Chinese subjects under fasted and fed states, which supports further investigations of test formulation in patients with Parkinson's disease.


Subject(s)
Dopamine Agonists , East Asian People , Pramipexole , Therapeutic Equivalency , Humans , Fasting , Parkinson Disease/drug therapy , Pramipexole/pharmacokinetics , Cross-Over Studies , Delayed-Action Preparations/pharmacokinetics , Healthy Volunteers , Eating , Dopamine Agonists/pharmacokinetics
18.
Drug Des Devel Ther ; 17: 2421-2429, 2023.
Article in English | MEDLINE | ID: mdl-37614377

ABSTRACT

Purpose: To study and compare the pharmacokinetic characteristics of enteric-coated sustained-release (EcSr) aspirin tablets with enteric-coated (Ec) aspirin tablets (Bayer S.p.A) in healthy Chinese participants. Patients and Methods: In this open, randomized, single-dose, three-way, crossover study, 18 healthy participants randomly received 100 mg EcSr tablets pre-prandially (a.c.), EcSr tablets post-prandially (p.c.), or Ec tablets a.c. in each period. The concentrations of acetylsalicylic acid (ASA) and salicylic acid (SA) in plasma were determined by the LC-MS/MS method, and the pharmacokinetic parameters were calculated using WinNonlin (version 8.1). Results: The essential PK parameters under the three treatment conditions (ie Ec a.c., EcSr a.c. and EcSr p.c.) were as follows: Cmax, ASA: 758.38±455.34, 222.77±98.04 and 194.54±61.19 ng, Tmax, ASA: 6.75(2,16), 4.5(2,11) and 8.25(5,11) h, T1/2, ASA: 0.43±0.08, 1.44±0.59 and 4.32±10.04 h, AUC0-t, ASA: 1008.88±452.27, 918.04±238.40 and 845.55±183.25 h·ng/mL; Cmax, SA: 6409.38±2098.52, 2863.53±679.73 and 2913.75±853.27ng/mL, Tmax, SA: 7.25(2,24), 10(3.5-14) and 10(7,14) h, T1/2, SA: 2.21±0.46, 2.69±0.72 and 3.51±2.06h, AUC0-t, SA: 29,131.41±9376.23, 27,243.97±7465.16, 27,240.25±7444.67 h·ng/mL. When taking EcSr aspirin tablets, the 90% confidence intervals of the geometric mean ratios (pre-prandial/post-prandial) of AUC0-t, ASA and AUC0-∞, ASA, Cmax, SA, AUC0-t, SA and AUC0-∞, SA were within the range of 80.00%-125.00%. Conclusion: EcSr aspirin tablets showed less inter-individual variation in release and absorption than Ec aspirin tablets, which was well reflected by comparing essential PK parameters. Furthermore, meals had no significant effect on the pharmacokinetics of EcSr aspirin tablets.


Subject(s)
Aspirin , Delayed-Action Preparations , East Asian People , Humans , Aspirin/pharmacokinetics , Chromatography, Liquid , Cross-Over Studies , Delayed-Action Preparations/pharmacokinetics , Healthy Volunteers , Salicylic Acid , Tandem Mass Spectrometry , Tablets, Enteric-Coated/pharmacokinetics
19.
Int J Pharm ; 644: 123301, 2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37572859

ABSTRACT

To prolong the absorption of the drug and achieve the effect of gastric retention, new brivaracetam tablets together with the characteristics of rapid swelling and sustained floating have been developed here. The tablets were optimized and prepared by direct compression techniques using Kollidon® SR and cross-linked polyvinylpyrrolidone (PVPP) XL as the matrix and disintegrant respectively, and carbomer 71G NF and polyethylene oxide (PEO) N60K as the gel materials to achieve sustained release effect. The characteristics of static expansion, floating time, drug release and dynamic swelling performance in vitro of the tablets were evaluated. The optimized formulations (F5 and F10) exhibited satisfactory swelling and floating properties, mechanical strength, and in vitro sustained-release characteristic with diffusion and matrix erosion mechanisms. X-ray images of beagle dogs showed that the tablet F5 could be retained in the stomach for more than 6 h. Furthermore, the pharmacokinetic studies in volunteers exhibited that the bioavailability of F5 and F10 was 95.70% (90% CI, 83.80%-109.28%) and 103.39% (90% CI, 87.61%-122.01%), respectively, relative to commercial tablets, with Tmax prolonged, demonstrating an excellent sustained-release effect. Therefore, the present system can reduce dosing frequency and improve patient compliance, which is expected to be a promising treatment option for epilepsy patients.


Subject(s)
Povidone , Stomach , Animals , Dogs , Delayed-Action Preparations/pharmacokinetics , Administration, Oral , Tablets , Edema
20.
J Med Primatol ; 52(6): 369-373, 2023 12.
Article in English | MEDLINE | ID: mdl-37432036

ABSTRACT

BACKGROUND: A novel buprenorphine (BUP) extended-release formulation (BUP-XR) produced as a lipid-encapsulated, low viscosity BUP suspension for subcutaneous (SC) injection to control pain was evaluated for pharmacokinetics and safety in four adult male cynomolgus monkeys. METHODS: Each animal was given 0.2 mg/kg reformulated BUP-XR SC. Clinical observations were made during the course of the study. Blood samples were obtained from each animal before BUP-XR administration, 6, 24, 48, 72, and 96 h post-BUP-XR injection. Plasma levels of buprenorphine were analyzed using HPLC-MS/MS. The PK values calculated included peak plasma concentration of the BUP analyte, time to peak plasma concentration, plasma half-life, area under the plasma concentration-time curve, clearance, apparent volume of distribution, and elimination rate constant (Cmax , Tmax , T½ , AUC0-t , CL, Vd, and Ke, respectively). RESULTS: Observable adverse clinical signs were not detected. BUP concentration peaked from 6 to 48 h, then declined in a linear fashion. Quantifiable plasma BUP was measured in all monkeys at all time points. Results indicate that a single BUP-XR dose at 0.2 mg/kg can reliably provide plasma levels of BUP reported in the literature to be therapeutically relevant for up to 96 h. CONCLUSIONS: Because of the lack of any clinical observations or adverse effects at the injection site or absence of observable abnormal behaviors, it may be concluded that the use of BUP-XR is safe and efficacious in this species of non-human primate at the dose regimen described in this study for up to 96 h post-administration.


Subject(s)
Buprenorphine , Male , Animals , Buprenorphine/adverse effects , Macaca fascicularis , Tandem Mass Spectrometry , Delayed-Action Preparations/adverse effects , Delayed-Action Preparations/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...