Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.590
Filtrer
1.
Sci Rep ; 14(1): 19963, 2024 08 28.
Article de Anglais | MEDLINE | ID: mdl-39198495

RÉSUMÉ

Clarithromycin extended-release (CLA-ER) was used as companion drug to rifampicin (RIF) for Mycobacterium ulcerans infection in the intervention arm of a WHO drug trial. RIF enhances CYP3A4 metabolism, thereby reducing CLA serum concentrations, and RIF concentrations might be increased by CLA co-administration. We studied the pharmacokinetics of CLA-ER at a daily dose of 15 mg/kg combined with RIF at a dose of 10 mg/kg in a subset of trial participants, and compared these to previously obtained pharmacokinetic data. Serial dried blood spot samples were obtained over a period of ten hours, and analyzed by LC-MS/MS in 30 study participants-20 in the RIF-CLA study arm, and 10 in the RIF-streptomycin study arm. Median CLA Cmax was 0.4 mg/L-and median AUC 3.9 mg*h/L, following 15 mg/kg CLA-ER. Compared to standard CLA dosed at 7.5 mg/kg previously, CLA-ER resulted in a non-significant 58% decrease in Cmax and a non-significant 30% increase in AUC. CLA co-administration did not alter RIF Cmax or AUC. Treatment was successful in all study participants. No effect of CLA co-administration on RIF pharmacokinetics was observed. Based on our serum concentration studies, the benefits CLA-ER over CLA immediate release are unclear.


Sujet(s)
Ulcère de Buruli , Clarithromycine , Préparations à action retardée , Mycobacterium ulcerans , Rifampicine , Humains , Clarithromycine/pharmacocinétique , Clarithromycine/administration et posologie , Mâle , Femelle , Adulte , Rifampicine/pharmacocinétique , Rifampicine/administration et posologie , Rifampicine/usage thérapeutique , Adulte d'âge moyen , Ulcère de Buruli/traitement médicamenteux , Ulcère de Buruli/microbiologie , Mycobacterium ulcerans/effets des médicaments et des substances chimiques , Préparations à action retardée/pharmacocinétique , Antibactériens/pharmacocinétique , Antibactériens/administration et posologie , Sujet âgé , Jeune adulte , Aire sous la courbe , Spectrométrie de masse en tandem
2.
PLoS One ; 19(8): e0307166, 2024.
Article de Anglais | MEDLINE | ID: mdl-39133725

RÉSUMÉ

Metformin, the primary therapy for type 2 diabetes mellitus (T2DM), showed limitations such as varying absorption, rapid system clearance, required large amount, resistance, longstanding side effects. Use of Nano formulations for pharmaceuticals is emerging as a viable technique to reduce negative consequences of drug, while simultaneously attaining precise release and targeted distribution. This study developed a Polyethylene Glycol conjugated Graphene Oxide Quantum dots (GOQD-PEG) nanocomposite for the sustained release of metformin. Herein, we evaluated the effectiveness of metformin-loaded nanoconjugate in in vitro insulin resistance model. Results demonstrated drug loaded nanoconjugate successfully restored glucose uptake and reversed insulin resistance in in vitro conditions at reduced dosage compared to free metformin.


Sujet(s)
Préparations à action retardée , Graphite , Insulinorésistance , Metformine , Nanoconjugués , Polyéthylène glycols , Boîtes quantiques , Graphite/composition chimique , Boîtes quantiques/composition chimique , Metformine/administration et posologie , Metformine/pharmacologie , Metformine/pharmacocinétique , Metformine/composition chimique , Polyéthylène glycols/composition chimique , Nanoconjugués/composition chimique , Préparations à action retardée/composition chimique , Préparations à action retardée/pharmacocinétique , Humains , Hypoglycémiants/administration et posologie , Hypoglycémiants/pharmacologie , Hypoglycémiants/composition chimique , Systèmes de délivrance de médicaments , Diabète de type 2/traitement médicamenteux , Glucose/métabolisme , Glucose/composition chimique
3.
AAPS PharmSciTech ; 25(6): 182, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138709

RÉSUMÉ

Local anesthesia is essential in dental practices, particularly for managing pain in tooth socket wounds, yet improving drug delivery systems remains a significant challenge. This study explored the physicochemical characteristics of lidocaine hydrochloride (LH) incorporated into a polyelectrolyte complex and poloxamer thermosensitivity hydrogel, assessing its local anesthetic efficacy in mouse models and its onset and duration of action as topical anesthetics in clinical trials. The thermoresponsive hydrogel exhibited a rapid phase transition within 1-3 minutes and demonstrated pseudo-plastic flow behavior. Its release kinetics followed Korsmeyer-Peppas, with 50% of biodegradation occurring over 48 h. In mouse models, certain thermogels showed superior anesthetic effects, with rapid onset and prolonged action, as evidenced by heat tolerance in tail-flick and hot plate models. In clinical trials, the LH-loaded thermoresponsive hydrogel provided rapid numbness onset, with anesthesia (Ton) beginning at an average of 46.5 ± 22.5 seconds and lasting effectively (Teff) for 202.5 ± 41.0 seconds, ranging from 120 to 240 seconds, indicating sustained release. These results highlight the promising properties of these formulations: rapid onset, prolonged duration, mucoadhesion, biodegradability, and high anesthesia effectiveness. This study demonstrates the potential for advancing local anesthesia across various medical fields, emphasizing the synergy between material science and clinical applications to improve patient care and safety.


Sujet(s)
Anesthésiques locaux , Systèmes de délivrance de médicaments , Hydrogels , Lidocaïne , Poloxamère , Lidocaïne/administration et posologie , Lidocaïne/composition chimique , Animaux , Hydrogels/composition chimique , Anesthésiques locaux/administration et posologie , Anesthésiques locaux/composition chimique , Souris , Poloxamère/composition chimique , Systèmes de délivrance de médicaments/méthodes , Polyélectrolytes/composition chimique , Mâle , Libération de médicament , Humains , Préparations à action retardée/administration et posologie , Préparations à action retardée/pharmacocinétique
4.
AAPS PharmSciTech ; 25(7): 198, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39192157

RÉSUMÉ

(-)-Phenserine ("phenserine") and (+)-phenserine (posiphen; buntanetap) are longer-acting enantiomeric analogs of physostigmine with demonstrated promise in the treatment of Alzheimer's and Parkinson's diseases. Both enantiomers have short plasma half-lives, and their pharmacokinetics might be improved through the use of either once or twice-daily administration of an extended-release dosage form. Phenserine was observed to form a colored degradation product in near-neutral and alkaline pH environments, and at pH 7, the half-life of posiphen was determined to be ~ 9 h (40 °C). To limit luminal degradation which would reduce bioavailability, a gastroretentive tablet composed of a polyethylene oxide-xanthan gum matrix was developed. When placed in simulated gastric fluid (pH 1.2), approximately 70% of the phenserine was released over a 12 h period, and no degradants were detected in the release medium. In comparison, a traditional hydrophilic-matrix, extended-release tablet showed measurable amounts of phenserine degradation in a pH 7.2 medium over an 8 h release interval. These results confirm that a gastroretentive tablet can reduce the luminal degradation of phenserine or posiphen by limiting exposure to neutral pH conditions while providing sustained release of the drug over at least 12 h. Additional advantages of the gastroretentive tablet include reduced gastric and intestinal concentrations of the drug resulting from the slower release from the gastroretentive tablet which may also limit the occurrence of the dose-limiting GI side effects previously observed with immediate-release phenserine capsules.


Sujet(s)
Préparations à action retardée , Comprimés , Concentration en ions d'hydrogène , Préparations à action retardée/pharmacocinétique , Physostigmine/administration et posologie , Physostigmine/pharmacocinétique , Physostigmine/analogues et dérivés , Physostigmine/composition chimique , Stéréoisomérie , Systèmes de délivrance de médicaments/méthodes , Biodisponibilité , Période , Libération de médicament
5.
J Biomed Mater Res B Appl Biomater ; 112(9): e35455, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39177322

RÉSUMÉ

Battlefield wounds are at high risk of infection due to gross contamination and delays in evacuation from forward-deployed locations. The aim of this study was to formulate an antibiotic wound gel for application by a field medic in austere environments to protect traumatic wounds from infection during transport. Formulation development was conducted over multiple phases to meet temperature, handling, in vitro elution, and in vivo tissue response requirements. Thermal properties were evaluated by vial inversion, DSC, and syringe expression force in a temperature range of 4-49°C. Handling was evaluated by spreading onto blood-contaminated tissue and irrigation resistance. Controlled antibiotic release was evaluated by a modified USP immersion cell dissolution method. Local tissue effects were evaluated in vivo by subcutaneous implantation in rats for 7 and 28 days. An oleogel composition of cholesterol, hydrogenated castor oil, soybean oil, and glyceryl monocaprylocaprate met the target performance criteria. Peak expression force from a 5 mL syringe at 4°C was 48.3 N, the dropping point temperature was 68°C, and the oleogel formulation could be spread onto blood-contaminated tissue and resisted aqueous irrigation. The formulation demonstrated sustained release of tobramycin in PBS at 32°C for 5 days. Implantation in a rat dorsal pocket demonstrated a slight tissue reaction after 7 days with minimal to no reaction after 28 days, comparable to a commercial hemostat control. Material resorption was evident after 28 days. The formulation met target characteristics and is appropriate for further evaluation in a large animal contaminated blast wound model.


Sujet(s)
Antibactériens , Préparations à action retardée , Gels , Animaux , Rats , Antibactériens/composition chimique , Antibactériens/pharmacologie , Gels/composition chimique , Préparations à action retardée/pharmacologie , Préparations à action retardée/composition chimique , Préparations à action retardée/pharmacocinétique , Mâle , Rat Sprague-Dawley , Mileux défavorisés
6.
AAPS PharmSciTech ; 25(7): 193, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39168956

RÉSUMÉ

Physiologically based pharmacokinetic (PBPK) or physiologically based biopharmaceutics models (PBBM) demonstrated plethora of applications in both new drugs and generic product development. Justification of dissolution specifications and establishment of dissolution safe space is an important application of such modeling approaches. In case of molecules exhibiting saturable absorption behavior, justification of dissolution specifications requires development of a model that incorporates effects of transporters is critical to simulate in vivo scenario. In the present case, we have developed a semi-mechanistic PBBM to describe the non-linearity of BCS class III molecule metformin for justification of dissolution specifications of extended release formulation at strengths 500 mg and 1000 mg. Semi-mechanistic PBBM was built using physicochemical properties, dissolution and non-linearity was accounted through incorporation of multiple transporter kinetics at absorption level. The model was extensively validated using literature reported intravenous, oral (immediate & extended release) formulations and further validated using in-house bioequivalence data in fasting and fed conditions. Virtual dissolution profiles at lower and upper specifications were generated to justify the dissolution specifications. The model predicted literature as well as in-house clinical study data with acceptable prediction errors. Further, virtual bioequivalence trials predicted the bioequivalence outcome that matched with clinical study data. The model predicted bioequivalence when lower and upper specifications were compared against pivotal test formulations thereby justifying dissolution specifications. Overall, complex and saturable absorption pathway of metformin was successfully simulated and this work resulted in regulatory acceptance of dissolution specifications which has ability to reduce multiple dissolution testing.


Sujet(s)
Biopharmacie , Préparations à action retardée , Metformine , Modèles biologiques , Solubilité , Équivalence thérapeutique , Metformine/pharmacocinétique , Metformine/administration et posologie , Metformine/composition chimique , Préparations à action retardée/pharmacocinétique , Humains , Biopharmacie/méthodes , Libération de médicament , Chimie pharmaceutique/méthodes , Hypoglycémiants/pharmacocinétique , Hypoglycémiants/administration et posologie , Hypoglycémiants/composition chimique , Administration par voie orale , Absorption intestinale
7.
J Pharm Sci ; 113(9): 2786-2794, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38986870

RÉSUMÉ

Topical ocular sustained-release drug delivery systems represent an effective strategy for the treatment of ocular diseases, for which a suitable carrier has yet to be sufficiently developed. Herein, an eye-compatible sodium polystyrene sulfonate resin (SPSR) was synthesized with a uniform particle size of about 3 µm. Ligustrazine phosphate (LP) was adsorbed to SPSR by cation exchange to form LP@SPSR. LP@SPSR suspension eye drops were further developed using the combination of Carbopol 934P and xanthan gum as suspending agents. The LP@SPSR suspension showed a sustained release in vitro, which was consistent with the observed porcine corneal penetration ex vivo. Pharmacokinetics in tear fluid of rabits indicated that LP@SPSR suspension led to prolonged ocular retention of LP and a 2-fold improved the area under the drug concentration-time curve (AUC0-t). Pharmacokinetics in the aqueous humor of rabbits showed 2.8-fold enhancement in the AUC0-t compared to LP solution. The LP@SPSR suspension exhibited no cytotoxicity to human corneal epithelial cells, nor irritation was observed in rabbit eyes. Thus, the LP@SPSR suspension has been validated as a safe and sustained release system leading to enhanced ophthalmic bioavailability for treating ocular diseases.


Sujet(s)
Biodisponibilité , Préparations à action retardée , Vecteurs de médicaments , Polystyrènes , Pyrazines , Animaux , Lapins , Pyrazines/pharmacocinétique , Pyrazines/administration et posologie , Pyrazines/composition chimique , Préparations à action retardée/pharmacocinétique , Polystyrènes/composition chimique , Polystyrènes/pharmacocinétique , Humains , Vecteurs de médicaments/composition chimique , Solutions ophtalmiques/pharmacocinétique , Solutions ophtalmiques/administration et posologie , Suidae , Mâle , Administration par voie ophtalmique , Systèmes de délivrance de médicaments/méthodes , Libération de médicament , Cornée/métabolisme , Cornée/effets des médicaments et des substances chimiques
8.
Int J Biol Macromol ; 276(Pt 2): 133841, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39032888

RÉSUMÉ

This study aims to design and optimize ondansetron (OND) gastro-retentive floating minitablets for better and prolonged control of postoperative nausea and vomiting (PONV) with improved patient compliance. Minitablets were directly compressed and encapsulated in a size 2 capsule shell with an overall dose of 24 mg. Central composite design (CCD) was applied keeping one cellulose ether derivative HPMC K15M and Carbopol 971 as variable and used as swelling and rate retarding agents. The other cellulose derivative i.e. sodium carboxymethyl cellulose, along with mannitol, sodium bicarbonate, and talc, were used in fixed quantities. The floating lag time, total floating time, swelling index, in-vitro drug release, and zero-order (RSQ value), were critical quality parameters. The optimized formulation (Fpred) was evaluated for all critical parameters, along with surface morphology, thermal stability, chemical interaction, and accelerated stability. The in silico PBPK modeling was applied to compare the bioavailability of Fpred with reference OND immediate-release tablets. The numerical optimization model predicted >90 % drug release with zero-order at 12 h. In silico PBPK modeling revealed comparable relative bioavailability of Fpred with the reference formulation. The gastroretentive floating minitablets of OND were successfully designed for prolonged emesis control in patients receiving chemotherapeutic agents.


Sujet(s)
Cellulose , Préparations à action retardée , Libération de médicament , Ondansétron , Comprimés , Ondansétron/pharmacocinétique , Ondansétron/composition chimique , Ondansétron/administration et posologie , Préparations à action retardée/pharmacocinétique , Cellulose/analogues et dérivés , Cellulose/composition chimique , Cellulose/pharmacocinétique , Humains , Acrylates/composition chimique , Acrylates/pharmacocinétique , Chimie pharmaceutique/méthodes , Biodisponibilité , Préparation de médicament , Résines acryliques/composition chimique , Modèles biologiques
9.
ACS Biomater Sci Eng ; 10(8): 5014-5026, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-38982893

RÉSUMÉ

Achieving rapid clotting and clot stability are important unmet goals of clinical management of noncompressible hemorrhage. This study reports the development of a spatiotemporally controlled release system of an antihemorrhagic drug, etamsylate, in the management of internal hemorrhage. Gly-Arg-Gly-Asp-Ser (GRGDS) peptide-functionalized chitosan nanoparticles, with high affinity to bind with the GPIIa/IIIb receptor of activated platelets, were loaded with the drug etamsylate (etamsylate-loaded GRGDS peptide-functionalized chitosan nanoparticles; EGCSNP). Peptide conjugation was confirmed by LCMS, and the delivery system was characterized by DLS, SEM, XRD, and FTIR. In vitro study exhibited 90% drug release till 48 h fitting into the Weibull model. Plasma recalcification time and prothrombin time tests of GRGDS-functionalized nanoparticles proved that clot formation was 1.5 times faster than nonfunctionalized chitosan nanoparticles. The whole blood clotting time was increased by 2.5 times over clot formed under nonfunctionalized chitosan nanoparticles. Furthermore, the application of rheometric analysis revealed a 1.2 times stiffer clot over chitosan nanoparticles. In an in vivo liver laceration rabbit model, EGCSNP spatially localized at the internal injury site within 5 min of intravenous administration, and no rebleeding was recorded up to 3 h. The animals survived for 3 weeks after the injury, indicating the strong potential of the system for the management of noncompressible hemorrhage.


Sujet(s)
Coagulation sanguine , Chitosane , Modèles animaux de maladie humaine , Hémorragie , Nanoparticules , Animaux , Lapins , Nanoparticules/composition chimique , Chitosane/composition chimique , Hémorragie/traitement médicamenteux , Coagulation sanguine/effets des médicaments et des substances chimiques , Préparations à action retardée/pharmacocinétique , Préparations à action retardée/composition chimique , Mâle , Peptides/composition chimique , Peptides/pharmacologie , Peptides/usage thérapeutique
10.
AAPS PharmSciTech ; 25(6): 160, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38992299

RÉSUMÉ

In part I, we reported Hansen solubility parameters (HSP, HSPiP program), experimental solubility at varied temperatures for TOTA delivery. Here, we studied dose volume selection, stability, pH, osmolality, dispersion, clarity, and viscosity of the explored combinations (I-VI). Ex vivo permeation and deposition studies were performed to observe relative diffusion rate from the injected site in rat skin. Confocal laser scanning microscopy (CLSM) study was conducted to support ex vivo findings. Moreover, GastroPlus predicted in vivo parameters in humans and the impact of various critical factors on pharmacokinetic parameters (PK). Immediate release product (IR) contained 60% of PEG400 whereas controlled release formulation (CR) contained PEG400 (60%), water (10%) and d-limonene (30%) to deliver 2 mg of TOTA. GastroPlus predicted the plasma drug concentration of weakly basic TOTA as function of pH (from pH 2.0 to 9). The cumulative drug permeation and drug deposition were found to be in the order as B-VI˃ C-VI˃A-VI across rat skin. This finding was further supported with CLSM. Moreover, IR and CR were predicted to achieve Cmax of 0.0038 µg/ mL and 0.00023 µg/mL, respectively, after sub-Q delivery. Added limonene in CR extended the plasma drug concentration over period of 12 h as predicted in GastroPlus. Parameters sensitivity analysis (PSA) assessment predicted that sub-Q blood flow rate is the only factor affecting PK parameters in IR formulation whereas this was insignificant for CR. Thus, sub-Q delivery CR would be promising alternative with ease of delivery to children and aged patient.


Sujet(s)
Absorption cutanée , Solubilité , Tartrate de toltérodine , Animaux , Rats , Humains , Absorption cutanée/effets des médicaments et des substances chimiques , Absorption cutanée/physiologie , Tartrate de toltérodine/administration et posologie , Tartrate de toltérodine/pharmacocinétique , Thermodynamique , Solvants/composition chimique , Peau/métabolisme , Concentration en ions d'hydrogène , Préparations à action retardée/pharmacocinétique , Préparations à action retardée/administration et posologie , Terpènes/composition chimique , Terpènes/administration et posologie , Terpènes/pharmacocinétique , Administration par voie cutanée , Limonène/administration et posologie , Limonène/pharmacocinétique , Limonène/composition chimique , Mâle , Polyéthylène glycols/composition chimique , Systèmes de délivrance de médicaments/méthodes , Chimie pharmaceutique/méthodes , Cyclohexènes/composition chimique , Cyclohexènes/pharmacocinétique , Cyclohexènes/administration et posologie , Rat Sprague-Dawley
11.
Int J Nanomedicine ; 19: 7547-7566, 2024.
Article de Anglais | MEDLINE | ID: mdl-39071505

RÉSUMÉ

Cancer poses a significant threat to human life and health. Chemotherapy is currently one of the effective cancer treatments, but many chemotherapy drugs have cell toxicity, low solubility, poor stability, a narrow therapeutic window, and unfavorable pharmacokinetic properties. To solve the above problems, target drug delivery to tumor cells, and reduce the side effects of drugs, an anti-tumor drug delivery system based on tumor microenvironment has become a focus of research in recent years. The construction of a reduction-sensitive nanomedicine delivery system based on disulfide bonds has attracted much attention. Disulfide bonds have good reductive responsiveness and can effectively target the high glutathione (GSH) levels in the tumor environment, enabling precise drug delivery. To further enhance targeting and accelerate drug release, disulfide bonds are often combined with pH-responsive nanocarriers and highly expressed ligands in tumor cells to construct drug delivery systems. Disulfide bonds can connect drug molecules and polymer molecules in the drug delivery system, as well as between different drug molecules and carrier molecules. This article summarized the drug delivery systems (DDS) that researchers have constructed in recent years based on disulfide bond drug delivery systems targeting the tumor microenvironment, disulfide bond cleavage-triggering conditions, various drug loading strategies, and carrier design. In this review, we also discuss the controlled release mechanisms and effects of these DDS and further discuss the clinical applicability of delivery systems based on disulfide bonds and the challenges faced in clinical translation.


Sujet(s)
Antinéoplasiques , Disulfures , Systèmes de délivrance de médicaments , Tumeurs , Microenvironnement tumoral , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Disulfures/composition chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/pharmacocinétique , Tumeurs/traitement médicamenteux , Systèmes de délivrance de médicaments/méthodes , Nanomédecine/méthodes , Vecteurs de médicaments/composition chimique , Vecteurs de médicaments/pharmacocinétique , Animaux , Nanoparticules/composition chimique , Glutathion/composition chimique , Concentration en ions d'hydrogène , Préparations à action retardée/composition chimique , Préparations à action retardée/pharmacocinétique , Libération de médicament
12.
AAPS PharmSciTech ; 25(6): 167, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39043962

RÉSUMÉ

The aim was to employ site-dependent absorption of mirabegron (MB) as a guide for fabrication of oral disintegrating controlled release tablet (ODCRT) which undergoes instantaneous release of loading fraction followed by delayed release of the rest of MB. The goal was to release MB in a manner consistent with the chronobiology of overactive bladder (OAB) syndrome. In situ rabbit intestinal permeability of MB was adopted to assess absorption sites. MB was subjected to dry co-grinding with citric acid to develop the fast-dissolving fraction in the mouth. Delayed release fraction was formulated by ethanol-assisted co-processing with increasing proportions of Eudragit polymer (S100) as pH responsive polymer. The developed dry mixtures underwent thermal (DSC) and physical (X-ray diffraction) characterization, in addition to in vitro release behavior. Optimized fast dissolving and delayed release formulations were mixed with tablet excipient before compression in ODCRT which was assessed for release profile using continuous pH variation. MB underwent preferential permeation through ileum and colon. Co-grinding with citric acid provided co-amorphous powder with fast dissolution. Co-amorphization of MB with Eudragit S100 (1:5) showed pH-dependent release to release most of the dose at pH 7.4. The developed ODCRT released 43.5% of MB in the buccal environment and retained MB at acidic pH to start release at pH 7.4. The study successfully fabricated ODCRT guided by site-dependent absorption. The ODCRT instantaneously released loading fraction to support the patient after administration with delayed fraction to sustain the effect.


Sujet(s)
Acétanilides , Préparations à action retardée , Excipients , Absorption intestinale , Solubilité , Comprimés , Thiazoles , Préparations à action retardée/pharmacocinétique , Animaux , Thiazoles/administration et posologie , Thiazoles/pharmacocinétique , Thiazoles/composition chimique , Acétanilides/composition chimique , Acétanilides/administration et posologie , Acétanilides/pharmacocinétique , Lapins , Administration par voie orale , Excipients/composition chimique , Chimie pharmaceutique/méthodes , Libération de médicament , Concentration en ions d'hydrogène , Perméabilité , Poly(acides méthacryliques)
13.
Drugs R D ; 24(2): 275-283, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39042293

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Venlafaxine hydrochloride extended-release (ER) capsules are commonly used to treat depression and anxiety disorders. Evaluation of the bioequivalence of generic formulations with reference products is essential to ensure therapeutic equivalence. The objective of this study was to evaluate the bioequivalence, safety, and tolerability of Chinese-manufactured venlafaxine hydrochloride extended-release capsules compared with USA-manufactured EFFEXOR® XR in healthy Chinese volunteers under fed conditions. METHODS: A randomized, open-label, single-dose, crossover study was conducted. Subjects were randomly assigned to receive the test formulation (one 150-mg ER capsule manufactured in China) or the reference formulation (one 150-mg ER capsule manufactured in the USA). The bioequivalence of the two drugs was assessed using the area under the plasma concentration-time curve from time zero to the last sampling time (AUC0-t) and the maximum observed concentration (Cmax). RESULTS: A total of 28 subjects were enrolled and randomly assigned to receive a single dose of either the test or reference capsule. All the subjects completed the study and were included in the pharmacokinetic (PK) and safety analyses. The mean AUC0-t and Cmax of venlafaxine and its active metabolite O-desmethylvenlafaxine were comparable between the test and reference products with both parameters close to 100% and the corresponding 90% confidence intervals within the specified 80-125% bioequivalence boundary. Safety was also assessed between the two products and all adverse events (AEs) in this study were mild in severity. CONCLUSIONS: Both the test and reference venlafaxine hydrochloride ER capsules were bioequivalent and showed a similar safety and tolerability profile in the population studied. CLINICAL TRIALS REGISTRATION: This study was registered at the Drug Clinical Trial Registration and Information Publicity Platform ( http://www.chinadrugtrials.org.cn/index.html ) with registration number CTR20211243, date: June 1, 2021.


Sujet(s)
Capsules , Études croisées , Préparations à action retardée , Volontaires sains , Équivalence thérapeutique , Chlorhydrate de venlafaxine , Humains , Chlorhydrate de venlafaxine/pharmacocinétique , Chlorhydrate de venlafaxine/administration et posologie , Chlorhydrate de venlafaxine/effets indésirables , Mâle , Adulte , Préparations à action retardée/pharmacocinétique , Femelle , Jeune adulte , Aire sous la courbe , Médicaments génériques/pharmacocinétique , Médicaments génériques/administration et posologie , Médicaments génériques/effets indésirables , Asiatiques , Chine , Adulte d'âge moyen , Peuples d'Asie de l'Est
14.
Biomacromolecules ; 25(8): 4946-4955, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-38949062

RÉSUMÉ

Negatively charged poly(l-Thr-co-l-Thr succinate) (PTTs) was developed as a new thermogel. Aqueous PTT solutions underwent thermogelation over a concentration range of 6.0-8.3 wt %. Dynamic light scattering, FTIR, 1H NMR, and COSY spectra revealed the partial strengthening of the ß-sheet conformation and the dehydration of PTTs during the transition. Extendin-4 was released from the PTTs thermogel with a large initial burst release, whereas positively charged lixisenatide significantly reduced its initial burst release to 25%, and up to 77% of the dose was released from the gel over 14 days. In vivo study revealed a high plasma concentration of lixisenatide over 5 days and hypoglycemic efficacy was observed for type II diabetic rats over 7-10 days. The biocompatible PTTs were degraded by subcutaneous enzymes. This study thus demonstrates an effective strategy for reducing the initial burst release of protein drugs from thermogels with the introduction of electrostatic interactions between the drug and the thermogel.


Sujet(s)
Gels , Peptides , Animaux , Peptides/composition chimique , Peptides/pharmacologie , Rats , Gels/composition chimique , Diabète expérimental/traitement médicamenteux , Préparations à action retardée/composition chimique , Préparations à action retardée/pharmacocinétique , Mâle , Hypoglycémiants/composition chimique , Hypoglycémiants/pharmacologie , Hypoglycémiants/administration et posologie , Rat Sprague-Dawley , Libération de médicament , Diabète de type 2/traitement médicamenteux , Récepteur du peptide-2 similaire au glucagon
15.
Br J Clin Pharmacol ; 90(9): 2256-2270, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38866397

RÉSUMÉ

AIMS: The aims of this study were to develop a population pharmacokinetic (PK) model for risperidone ISM® and to investigate the relationships between active moiety exposure, as described by apparent clearance (CL40), and several covariates using all data from five clinical studies. METHODS: A population PK model was developed using active moiety concentrations from a study in healthy volunteers and two studies in patients with schizophrenia. Data from a comparative bioavailability study in medically stable patients and a Phase III study in patients with acute exacerbation of schizophrenia were then incorporated, using empirical Bayesian feedback and model refinement in NONMEM. Finally, covariate analysis was performed on CL40. RESULTS: The final model adequately described the pharmacokinetics of 6288 active moiety concentrations in 17 healthy volunteers and 430 patients with schizophrenia. This one-compartment disposition model had a complex absorption process, combining a small amount immediately entering the central active moiety compartment, two first-order absorption processes and a combined zero-order and first order process, with first-order elimination from the central compartment. Significant covariates on CL40 were BMI and sex. Goodness-of-fit (GOF) plots and visual predictive checks (VPC) confirmed acceptable description of the data. CONCLUSIONS: The population PK model adequately described active moiety concentrations from five clinical studies after risperidone ISM® administration. Relationships between active moiety exposure and covariates were defined in order to facilitate simulations for future studies. The model showed that risperidone ISM® rapidly achieves therapeutic plasma levels within the first hours after the first injection that are maintained sustainedly throughout the whole dosing interval following once-monthly gluteal injections of 100 mg and 75 mg.


Sujet(s)
Neuroleptiques , Préparations à action retardée , Modèles biologiques , Rispéridone , Schizophrénie , Humains , Rispéridone/pharmacocinétique , Rispéridone/administration et posologie , Neuroleptiques/pharmacocinétique , Neuroleptiques/administration et posologie , Mâle , Femelle , Schizophrénie/traitement médicamenteux , Adulte , Préparations à action retardée/pharmacocinétique , Adulte d'âge moyen , Jeune adulte , Théorème de Bayes , Biodisponibilité , Adolescent , Volontaires sains
16.
Int J Nanomedicine ; 19: 5441-5458, 2024.
Article de Anglais | MEDLINE | ID: mdl-38868593

RÉSUMÉ

Introduction: Quercetin (QUER), a flavonoid abundant in fruits and vegetables, is emerging as a promising alternative therapeutic agent for obesity treatment due to its antioxidant and anti-adipogenic properties. However, the clinical application of QUER is limited by its poor solubility, low bioavailability, and potential toxicity at high doses. To address these challenges, this study aims to develop an advanced drug delivery system using fluorescent mesoporous silica nanoparticles (FMSNs) coated with polydopamine (PDA) for the efficient and sustained delivery of QUER to inhibit adipogenesis. Methods: The research included the synthesis of PDA-coated FMSNs for encapsulation of QUER, characterization of their mesoporous structures, and systematic investigation of the release behavior of QUER. The DPPH assay was used to evaluate the sustained radical scavenging potential. Concentration-dependent effects on 3T3-L1 cell proliferation, cellular uptake and adipogenesis inhibition were investigated. Results: PDA-coated FMSNs exhibited well-aligned mesoporous structures. The DPPH assay confirmed the sustained radical scavenging potential, with FMSNs-QUER@PDA showing 53.92 ± 3.48% inhibition at 72 h, which was higher than FMSNs-QUER (44.66 ± 0.57%) and free QUER (43.37 ± 5.04%). Concentration-dependent effects on 3T3-L1 cells highlighted the enhanced efficacy of PDA-coated FMSNs for cellular uptake, with a 1.5-fold increase compared to uncoated FMSNs. Adipogenesis inhibition was also improved, with relative lipid accumulation of 44.6 ± 4.6%, 37.3 ± 4.6%, and 36.5 ± 7.3% at 2.5, 5, and 10 µM QUER concentrations, respectively. Conclusion: The study successfully developed a tailored drug delivery system, emphasizing sustained QUER release and enhanced therapeutic effects. FMSNs, especially when coated with PDA, exhibit promising properties for efficient QUER delivery, providing a comprehensive approach that integrates advanced drug delivery technology and therapeutic efficacy.


Sujet(s)
Cellules 3T3-L1 , Adipogenèse , Préparations à action retardée , Vecteurs de médicaments , Indoles , Nanoparticules , Polymères , Quercétine , Silice , Quercétine/composition chimique , Quercétine/pharmacologie , Quercétine/pharmacocinétique , Quercétine/administration et posologie , Animaux , Souris , Adipogenèse/effets des médicaments et des substances chimiques , Silice/composition chimique , Indoles/composition chimique , Indoles/pharmacologie , Indoles/pharmacocinétique , Indoles/administration et posologie , Nanoparticules/composition chimique , Préparations à action retardée/composition chimique , Préparations à action retardée/pharmacologie , Préparations à action retardée/pharmacocinétique , Vecteurs de médicaments/composition chimique , Polymères/composition chimique , Porosité , Libération de médicament , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antioxydants/composition chimique , Antioxydants/pharmacologie , Antioxydants/pharmacocinétique , Antioxydants/administration et posologie
17.
Acta Biomater ; 183: 101-110, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38834149

RÉSUMÉ

Controlled release of low molecular weight hydrophilic drugs, administered locally, allows maintenance of high concentrations at the target site, reduces systemic side effects, and improves patient compliance. Injectable hydrogels are commonly used as a vehicle. However, slow release of low molecular weight hydrophilic drugs is very difficult to achieve, mainly due to a rapid diffusion of the drug out of the drug delivery system. Here we present an injectable and self-healing hydrogel based entirely on the self-assembly of liposomes. Gelation of liposomes, without damaging their structural integrity, was induced by modifying the cholesterol content and surface charge. The small hydrophilic molecule, sodium fluorescein, was loaded either within the extra-liposomal space or encapsulated into the aqueous cores of the liposomes. This encapsulation strategy enabled the achievement of controlled and adjustable release profiles, dependent on the mechanical strength of the gel. The hydrogel had a high mechanical strength, minimal swelling, and slow degradation. The liposome-based hydrogel had prolonged mechanical stability in vivo with benign tissue reaction. This work presents a new class of injectable hydrogel that holds promise as a versatile drug delivery system. STATEMENT OF SIGNIFICANCE: The porous nature of hydrogels poses a challenge for delivering small hydrophilic drug, often resulting in initial burst release and shorten duration of release. This issue is particularly pronounced with physically crosslinked hydrogels, since their matrix can swell and dissipate rapidly, but even in cases where the polymers in the hydrogel are covalently cross-linked, small molecules can be rapidly released through its porous mesh. Here we present an injectable self-healing hydrogel based entirely on the self-assembly of liposomes. Small hydrophilic molecules were entrapped inside the extra-liposomal space or loaded into the aqueous cores of the liposomes, allowing controlled and tunable release profiles.


Sujet(s)
Préparations à action retardée , Hydrogels , Interactions hydrophobes et hydrophiles , Liposomes , Liposomes/composition chimique , Préparations à action retardée/composition chimique , Préparations à action retardée/pharmacocinétique , Hydrogels/composition chimique , Injections , Animaux , Fluorescéine/composition chimique
18.
Int J Pharm ; 660: 124294, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-38823467

RÉSUMÉ

Atropine sulfate (ATS) eye drops at low concentrations constitute a limited selection for myopia treatment, with challenges such as low ophthalmic bioavailability and inadequate stability. This study proposes a novel strategy by synthesizing ophthalmic sodium polystyrene sulfonate resin (SPSR) characterized by a spherical shape and uniform size for cationic exchange with ATS. The formulation of ATS@SPSR suspension eye drops incorporates xanthan gum and hydroxypropyl methylcellulose (HPMC) as suspending agents. In vitro studies demonstrated that ATS@SPSR suspension eye drops exhibited sustained release characteristics, and tropic acid, its degradation product, remained undetected for 30 days at 40 °C. The ATS levels in the tear fluids and aqueous humor of New Zealand rabbits indicated a significant increase in mean residence time (MRT) and area under the drug concentration-time curve (AUC0-12h) for ATS@SPSR suspension eye drops compared to conventional ATS eye drops. Moreover, safety assessment confirmed the non-irritating nature of ATS@SPSR suspension eye drops in rabbit eyes. In conclusion, the cation-responsive sustained-release ATS@SPSR suspension eye drops enhanced the bioavailability and stability of ATS, offering a promising avenue for myopia treatment.


Sujet(s)
Atropine , Biodisponibilité , Préparations à action retardée , Stabilité de médicament , Solutions ophtalmiques , Polystyrènes , Animaux , Lapins , Préparations à action retardée/pharmacocinétique , Polystyrènes/composition chimique , Polystyrènes/pharmacocinétique , Solutions ophtalmiques/pharmacocinétique , Solutions ophtalmiques/administration et posologie , Atropine/pharmacocinétique , Atropine/administration et posologie , Atropine/composition chimique , Mâle , Dérivés de l'hypromellose/composition chimique , Larmes/métabolisme , Libération de médicament , Humeur aqueuse/métabolisme , Polyosides bactériens/composition chimique , Administration par voie ophtalmique
19.
Eur J Drug Metab Pharmacokinet ; 49(4): 507-516, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38874900

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Model-based bioequivalence (MBBE) encompasses the use of nonlinear mixed effect models supporting the estimation of pharmacokinetic endpoints to assess the relative bioavailability between multi-source drug products. This application emerges as a valuable alternative to the standard non-compartmental analysis (NCA) in bioequivalence (BE) studies in which dense sampling is not possible. In this work, we aimed to assess the application of MBBE compared to traditional methods in evaluating the relative bioavailability of two formulations with different drug release properties. Additionally, we sought to predict the performance of a modified-release formulation in a multiple-dose scenario, leveraging data from a single-dose study. METHODS: MBBE analysis was implemented to estimate the BE endpoints (90% CI for the Test/Reference geometric mean ratio, T/R GMR) in area under the concentration-time curve (AUC) and maximum concentration (Cmax) using data from a single-dose, 2-period, 2-sequence BE study performed in 14 healthy subjects between a locally developed valproic acid extended-release formulation (Test) and the brand-name delayed-release formulation (Reference). RESULTS: Results were compared with the standard approach, revealing that MBBE analysis achieved higher discrimination between formulations for Cmax, addressing limitations of the experimental sampling design and highlighting an advantage for this model-based analysis even when rich data are available. Additionally, the bioequivalence outcome under the multiple-dose scenario was predicted through a simulation-based study for both total and unbound valproic acid concentrations, considering the impact of valproic acid saturable binding on BE conclusions. CONCLUSIONS: The MBBE analysis was superior to the NCA approach in detecting product-related differences, overcoming limitations in the study experimental design. Predictions for the multiple-dose scenario preclude that the extended-release properties of the Test formulation would persist at steady state, resulting in lower peak-to-trough fluctuation and bioequivalent performance in terms of the extent of drug absorption. Overall, these results should discourage unnecessary experimentation in healthy subjects.


Sujet(s)
Aire sous la courbe , Biodisponibilité , Préparations à action retardée , Modèles biologiques , Équivalence thérapeutique , Acide valproïque , Acide valproïque/pharmacocinétique , Acide valproïque/administration et posologie , Humains , Préparations à action retardée/pharmacocinétique , Mâle , Adulte , Jeune adulte , Anticonvulsivants/pharmacocinétique , Anticonvulsivants/administration et posologie , Femelle , Volontaires sains , Études croisées
20.
Eur J Pharm Sci ; 200: 106828, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38862047

RÉSUMÉ

The potential of micron-sized amorphous mesoporous silica particles as a novel controlled release drug delivery system for pulmonary administration has been investigated. Mesoporous silica formulations were demonstrated to provide a narrower particle size distribution and (spherical) shape uniformity compared to commercial micronized formulations, which is critical for repeatable and targeted aerosol delivery to the lungs. The release profiles of a well-known pulmonary drug loaded into mesoporous particles of different mean particle diameters (2.4, 3.9 and 6.3 µm) were analysed after aerosolization in a modified Andersen Cascade Impactor. Systematic control of the release rate of drug loaded into the particles was demonstrated in simulated lung fluid by variation of the mean particle diameter, as well as an enhanced release compared to a commercial micronized formulation. The mesoporous silica formulations all demonstrated an increased release rate of the loaded drug and moreover, under aerosolization from a commercial, low-cost dry powder inhaler (DPI) device, the formulations showed excellent performance, with low retainment and commercially viable fine particle fractions (FPFs). In addition, the measured median mass aerodynamic diameter (MMAD) of the different formulations (2.8, 4.1 and 6.2 µm) was shown to be tuneable with particle size, which can be helpful for targeting different regions in the lung. Together these results demonstrate that mesoporous silica formulations offer a promising novel alternative to current dry powder formulations for pulmonary drug delivery.


Sujet(s)
Aérosols , Budésonide , Libération de médicament , Inhalateurs à poudre sèche , Taille de particule , Silice , Silice/composition chimique , Silice/administration et posologie , Budésonide/composition chimique , Budésonide/administration et posologie , Budésonide/pharmacocinétique , Porosité , Inhalateurs à poudre sèche/méthodes , Administration par inhalation , Systèmes de délivrance de médicaments/méthodes , Préparations à action retardée/composition chimique , Préparations à action retardée/pharmacocinétique , Bronchodilatateurs/administration et posologie , Bronchodilatateurs/composition chimique , Bronchodilatateurs/pharmacocinétique , Vecteurs de médicaments/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE