Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Pharm ; 618: 121670, 2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35304242

ABSTRACT

For over 15 years, US and EU regulations ensure that medicines developed for children are explicitly authorised for such use with age-appropriate forms and formulations, implying dedicated research. To shed light on how these regulations have been adopted by pharmaceutical companies and how various aspects of paediatric oral drug formulation development are currently handled, an exploratory survey was conducted. Topics included: general company policy, regulatory aspects, dosage form selection, in-vitro, in-silico and (non-)clinical in-vivo methods, and food effects assessment. The survey results clearly underline the positive impact of the paediatric regulations and their overall uptake across the pharmaceutical industry. Even though significant improvements have been made in paediatric product development, major challenges remain. In this respect, dosage form selection faces a discrepancy between the youngest age groups (liquid products preference) and older subpopulations (adult formulation preference). Additionally, concerted research is needed in the development and validation of in-vitro tools and physiology based pharmacokinetic models tailored to the paediatric population, and in estimating the effect of non-standard and paediatric relevant foods. The current momentum in paediatric drug development and research should allow for an evolution in standardised methodology and guidance to develop paediatric formulations, which would benefit pharmaceutical industry and regulators.


Subject(s)
Drug Development , Drug Industry , Adult , Child , Drug Compounding , Food , Humans , Pharmaceutical Preparations , Surveys and Questionnaires
2.
J Pharm Sci ; 110(2): 584-593, 2021 02.
Article in English | MEDLINE | ID: mdl-33058891

ABSTRACT

This workshop report summarizes the proceedings of Day 2 of a three-day workshop on "Current State and Future Expectations of Translational Modeling Strategies toSupportDrug Product Development, Manufacturing Changes and Controls". From a drug product quality perspective, physiologically based biopharmaceutics modeling (PBBM) is a tool to link variations in the drug product quality attributes to in vivo outcomes enabling the establishment of clinically relevant drug product specifications (CRDPS). Day 2 of the workshop focused on best practices in developing, verifying and validating PBBM. This manuscript gives an overview of podium presentations and summarizes breakout (BO) session discussions related to (1) challenges and opportunities for using PBBM to assess the clinical impact of formulation and manufacturing changes on the in vivo performance of a drug product, (2) best practices to account for parameter uncertainty and variability during model development, (3) best practices in the development, verification and validation of PBBM and (4) opportunities and knowledge gaps related to leveraging PBBM for virtual bioequivalence simulations.


Subject(s)
Biopharmaceutics , Research Report , Models, Biological , Solubility , Therapeutic Equivalency
3.
Eur J Pharm Biopharm ; 156: 50-63, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32805361

ABSTRACT

Oral drug absorption is a complex process depending on many factors, including the physicochemical properties of the drug, formulation characteristics and their interplay with gastrointestinal physiology and biology. Physiological-based pharmacokinetic (PBPK) models integrate all available information on gastro-intestinal system with drug and formulation data to predict oral drug absorption. The latter together with in vitro-in vivo extrapolation and other preclinical data on drug disposition can be used to predict plasma concentration-time profiles in silico. Despite recent successes of PBPK in many areas of drug development, an improvement in their utility for evaluating oral absorption is much needed. Current status of predictive performance, within the confinement of commonly available in vitro data on drugs and formulations alongside systems information, were tested using 3 PBPK software packages (GI-Sim (ver.4.1), Simcyp® Simulator (ver.15.0.86.0), and GastroPlus™ (ver.9.0.00xx)). This was part of the Innovative Medicines Initiative (IMI) Oral Biopharmaceutics Tools (OrBiTo) project. Fifty eight active pharmaceutical ingredients (APIs) were qualified from the OrBiTo database to be part of the investigation based on a priori set criteria on availability of minimum necessary information to allow modelling exercise. The set entailed over 200 human clinical studies with over 700 study arms. These were simulated using input parameters which had been harmonised by a panel of experts across different software packages prior to conduct of any simulation. Overall prediction performance and software packages comparison were evaluated based on performance indicators (Fold error (FE), Average fold error (AFE) and absolute average fold error (AAFE)) of pharmacokinetic (PK) parameters. On average, PK parameters (Area Under the Concentration-time curve (AUC0-tlast), Maximal concentration (Cmax), half-life (t1/2)) were predicted with AFE values between 1.11 and 1.97. Variability in FEs of these PK parameters was relatively high with AAFE values ranging from 2.08 to 2.74. Around half of the simulations were within the 2-fold error for AUC0-tlast and around 90% of the simulations were within 10-fold error for AUC0-tlast. Oral bioavailability (Foral) predictions, which were limited to 19 APIs having intravenous (i.v.) human data, showed AFE and AAFE of values 1.37 and 1.75 respectively. Across different APIs, AFE of AUC0-tlast predictions were between 0.22 and 22.76 with 70% of the APIs showing an AFE > 1. When compared across different formulations and routes of administration, AUC0-tlast for oral controlled release and i.v. administration were better predicted than that for oral immediate release formulations. Average predictive performance did not clearly differ between software packages but some APIs showed a high level of variability in predictive performance across different software packages. This variability could be related to several factors such as compound specific properties, the quality and availability of information, and errors in scaling from in vitro and preclinical in vivo data to human in vivo behaviour which will be explored further. Results were compared with previous similar exercise when the input data selection was carried by the modeller rather than a panel of experts on each in vitro test. Overall, average predictive performance was increased as reflected in smaller AAFE value of 2.8 as compared to AAFE value of 3.8 in case of previous exercise.


Subject(s)
Biopharmaceutics/standards , Data Analysis , Intestinal Absorption/drug effects , Models, Biological , Pharmaceutical Preparations/metabolism , Software/standards , Administration, Oral , Biopharmaceutics/methods , Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , Databases, Factual/standards , Forecasting , Humans , Intestinal Absorption/physiology , Pharmaceutical Preparations/administration & dosage
4.
Mol Pharm ; 17(4): 1310-1323, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32176503

ABSTRACT

In vivo studies have shown cyclic bile salt (BS) outputs during fasting whereas higher amounts have been observed in fed states. This leads to fluctuations of intestinal BS concentrations ([BS]) that can affect the rate and extent of absorption of lipophilic drugs in particular. However, most PBPK models use fixed values of [BS] in fasted and fed states albeit with different values in different regions of the GI tract. During fasting, there is a relationship between gallbladder volume (GBV) and the phase of the Interdigestive Migrating Motor Complex cycle (IMMCc), showing cyclic GBV changes with periodic filling and emptying. This relationship is also affected by the origin of the IMMCc (antral or duodenal). In fed states, meta-analysis indicated that GB residual volume (% of fasting maximum) was 46.4 ± 27%CV and 30.7 ± 48%CV for low- and high-fat meals, respectively. The corresponding values for the duration of the emptying phase were for low fat meals 0.72h ± 1%CV and for high fat meals 1.17h ± 37%CV. The model, the Advanced Dynamic Bile Salt Model (ADBSM), was built bottom-up and its parameters were not fitted against in vivo measurements of [BS]. It involved update of the dynamic luminal fluid volumes model based on meta-analysis of available imaging data. The ADBSM is incorporated into the Simcyp (v18r2) PBPK simulator. The model predictivity was good (within 1.25-fold error for 11/20 of the clinical studies) and was assessed against clinical studies of luminal [BS] that provide only the type of meal (i.e., low- or high-fat), the timing of the meal and/or water intake events, and the number and age range of the study participants. The model is also an important component of models capturing enterohepatic recirculation of drug and metabolite. Further work is required to validate the current model and compare to simpler models with respect to drug absorption, especially of the lipophilic compounds.


Subject(s)
Bile Acids and Salts/metabolism , Body Fluids/metabolism , Gastrointestinal Tract/metabolism , Pharmaceutical Preparations/metabolism , Animals , Diet, High-Fat/methods , Digestion/physiology , Gallbladder/metabolism , Humans
5.
Drug Metab Dispos ; 47(8): 854-864, 2019 08.
Article in English | MEDLINE | ID: mdl-31076413

ABSTRACT

The aim of this study was to derive region-specific transporter expression data suitable for in vitro-to-in vivo extrapolation (IVIVE) within a physiologically based pharmacokinetic (PBPK) modeling framework. A meta-analysis was performed whereby literary sources reporting region-specific transporter expression obtained via absolute and relative quantification approaches were considered in healthy adult Caucasian individuals. Furthermore, intestinal total membrane protein yield was calculated to enable mechanistic IVIVE via absolute transporter abundances. Where required, authors were contacted for additional information. A refined database was constructed where samples were excluded based on quantification in, non-Caucasian subjects, disease tissue, subjects <18 years old, duplicated samples, non-total membrane matrix, pooled matrices, or cDNA. Demographic data were collected where available. The weighted and geometric mean, coefficient of variation, and between-study homogeneity was calculated in each of eight gut segments (duodenum, two jejunum, four ileum, and colon) for 16 transporters. Expression data were normalized to that in the proximal jejunum. From a total of 47 articles, the final database consisted of 2238 measurements for 16 transporters. The solute carrier peptide transporter 1 (PepT1) showed the highest jejunal abundance, while multidrug resistance-associated protein (MRP) 2 was the highest abundance ATP-binding cassette transporter. Transporters displaying significant region-specific expression included the ileal bile acid transporter, which showed 18-fold greater terminal ileum expression compared with the proximal jejunum, while MRP3, organic cation transporter type 1 (OCTN1), and OCT1 showed >2-fold higher expression in other regions compared with the proximal jejunum. This is the first systematic analysis incorporating absolute quantification methodology to determine region-specific intestinal transporter expression. It is expected to be beneficial for mechanistic transporter IVIVE in healthy adult Caucasians. SIGNIFICANCE STATEMENT: Given the burgeoning reports of absolute transporter abundances in the human intestine, the incorporation of such information into mechanistic IVIVE-PBPK models could offer a distinct advantage in facilitating the robust assessment of the impact of gut transporters on drug disposition. The systematic and formal assessment via a literature meta-analysis described herein, enables assignment of the regional-specific expression, absolute transporter abundances, interindividual variability, and other associated scaling factors to healthy Caucasian populations within PBPK models. The resulting values are available to incorporate into PBPK models, and offer a verifiable account describing intestinal transporter expression within PBPK models for persons wishing to utilize them. Furthermore, these data facilitate the development of appropriate IVIVE scaling strategies using absolute transporter abundances.


Subject(s)
Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Jejunum/metabolism , Membrane Transport Proteins/metabolism , Models, Biological , Adult , Humans , Membrane Transport Proteins/analysis , Multidrug Resistance-Associated Protein 2 , Proteomics , White People
6.
J Pharm Sci ; 108(4): 1604-1618, 2019 04.
Article in English | MEDLINE | ID: mdl-30476508

ABSTRACT

The physiological relevance of single-phase (aqueous only) and 2-phase (aqueous and organic phase) in vitro dissolution experiments was compared by mechanistic modeling. For orally dosed dipyridamole, stepwise, sequential estimation/confirmation of biopharmaceutical parameters from in vitro solubility-dissolution data was followed, before applying them within a physiologically based pharmacokinetic (PBPK) model. The PBPK model predicted clinical dipyridamole luminal and plasma concentration profiles reasonably well for a range of doses only where the precipitation rate constant was derived from the 2-phase experiment. The population model predicted a distribution of maximal precipitated fractions from 0% to 45% of the 90 mg dose (mean 7.6%). Such population information cannot be obtained directly from a few in vitro experiments; however well they may represent an "average" and several extreme subjects (those with low-high luminal fluid volumes, pH, etc.) because there is no indication of outcome likelihood. For this purpose, direct input of in vitro dissolution/precipitation profiles to a PBPK model is insufficient-mechanistic modeling is required. Biopharmaceutical in vitro-in vivo extrapolation tools can also simulate the effect of key experimental parameters (dissolution volumes, pH, paddle speed, etc.) on dissolution/precipitation behavior, thereby helping to identify critical variables, which may impact the number or design of in vitro experiments.


Subject(s)
Biopharmaceutics/methods , Drug Development/methods , Models, Biological , Administration, Oral , Computer Simulation , Dipyridamole/administration & dosage , Dipyridamole/pharmacokinetics , Drug Liberation , Duodenum/metabolism , Humans , Intestinal Absorption , Intestinal Mucosa/metabolism , Solubility , Workflow
7.
Mol Pharm ; 14(12): 4305-4320, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28771009

ABSTRACT

Mechanistic modeling of in vitro data generated from metabolic enzyme systems (viz., liver microsomes, hepatocytes, rCYP enzymes, etc.) facilitates in vitro-in vivo extrapolation (IVIV_E) of metabolic clearance which plays a key role in the successful prediction of clearance in vivo within physiologically-based pharmacokinetic (PBPK) modeling. A similar concept can be applied to solubility and dissolution experiments whereby mechanistic modeling can be used to estimate intrinsic parameters required for mechanistic oral absorption simulation in vivo. However, this approach has not widely been applied within an integrated workflow. We present a stepwise modeling approach where relevant biopharmaceutics parameters for ketoconazole (KTZ) are determined and/or confirmed from the modeling of in vitro experiments before being directly used within a PBPK model. Modeling was applied to various in vitro experiments, namely: (a) aqueous solubility profiles to determine intrinsic solubility, salt limiting solubility factors and to verify pKa; (b) biorelevant solubility measurements to estimate bile-micelle partition coefficients; (c) fasted state simulated gastric fluid (FaSSGF) dissolution for formulation disintegration profiling; and (d) transfer experiments to estimate supersaturation and precipitation parameters. These parameters were then used within a PBPK model to predict the dissolved and total (i.e., including the precipitated fraction) concentrations of KTZ in the duodenum of a virtual population and compared against observed clinical data. The developed model well characterized the intraluminal dissolution, supersaturation, and precipitation behavior of KTZ. The mean simulated AUC0-t of the total and dissolved concentrations of KTZ were comparable to (within 2-fold of) the corresponding observed profile. Moreover, the developed PBPK model of KTZ successfully described the impact of supersaturation and precipitation on the systemic plasma concentration profiles of KTZ for 200, 300, and 400 mg doses. These results demonstrate that IVIV_E applied to biopharmaceutical experiments can be used to understand and build confidence in the quality of the input parameters and mechanistic models used for mechanistic oral absorption simulations in vivo, thereby improving the prediction performance of PBPK models. Moreover, this approach can inform the selection and design of in vitro experiments, potentially eliminating redundant experiments and thus helping to reduce the cost and time of drug product development.


Subject(s)
Drug Liberation , Intestinal Absorption/physiology , Ketoconazole/pharmacokinetics , Models, Biological , Absorption, Physiological , Administration, Oral , Biopharmaceutics/methods , Chemistry, Pharmaceutical , Computer Simulation , Humans , Models, Chemical , Permeability , Solubility
8.
Mol Pharm ; 14(12): 4321-4333, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28817288

ABSTRACT

The aim of this study was to evaluate gastrointestinal (GI) dissolution, supersaturation, and precipitation of posaconazole, formulated as an acidified (pH 1.6) and neutral (pH 7.1) suspension. A physiologically based pharmacokinetic (PBPK) modeling and simulation tool was applied to simulate GI and systemic concentration-time profiles of posaconazole, which were directly compared with intraluminal and systemic data measured in humans. The Advanced Dissolution Absorption and Metabolism (ADAM) model of the Simcyp Simulator correctly simulated incomplete gastric dissolution and saturated duodenal concentrations of posaconazole in the duodenal fluids following administration of the neutral suspension. In contrast, gastric dissolution was approximately 2-fold higher after administration of the acidified suspension, which resulted in supersaturated concentrations of posaconazole upon transfer to the upper small intestine. The precipitation kinetics of posaconazole were described by two precipitation rate constants, extracted by semimechanistic modeling of a two-stage medium change in vitro dissolution test. The 2-fold difference in exposure in the duodenal compartment for the two formulations corresponded with a 2-fold difference in systemic exposure. This study demonstrated for the first time predictive in silico simulations of GI dissolution, supersaturation, and precipitation for a weakly basic compound in part informed by modeling of in vitro dissolution experiments and validated via clinical measurements in both GI fluids and plasma. Sensitivity analysis with the PBPK model indicated that the critical supersaturation ratio (CSR) and second precipitation rate constant (sPRC) are important parameters of the model. Due to the limitations of the two-stage medium change experiment the CSR was extracted directly from the clinical data. However, in vitro experiments with the BioGIT transfer system performed after completion of the in silico modeling provided an almost identical CSR to the clinical study value; this had no significant impact on the PBPK model predictions.


Subject(s)
Computer Simulation , Drug Liberation , Gastrointestinal Tract/physiology , Models, Biological , Triazoles/pharmacokinetics , Administration, Oral , Biopharmaceutics/methods , Chemistry, Pharmaceutical , Humans , Hydrogen-Ion Concentration , Intestinal Absorption/physiology , Models, Chemical , Solubility
10.
Eur J Pharm Sci ; 96: 626-642, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27693299

ABSTRACT

Three Physiologically Based Pharmacokinetic software packages (GI-Sim, Simcyp® Simulator, and GastroPlus™) were evaluated as part of the Innovative Medicine Initiative Oral Biopharmaceutics Tools project (OrBiTo) during a blinded "bottom-up" anticipation of human pharmacokinetics. After data analysis of the predicted vs. measured pharmacokinetics parameters, it was found that oral bioavailability (Foral) was underpredicted for compounds with low permeability, suggesting improper estimates of intestinal surface area, colonic absorption and/or lack of intestinal transporter information. Foral was also underpredicted for acidic compounds, suggesting overestimation of impact of ionisation on permeation, lack of information on intestinal transporters, or underestimation of solubilisation of weak acids due to less than optimal intestinal model pH settings or underestimation of bile micelle contribution. Foral was overpredicted for weak bases, suggesting inadequate models for precipitation or lack of in vitro precipitation information to build informed models. Relative bioavailability was underpredicted for both high logP compounds as well as poorly water-soluble compounds, suggesting inadequate models for solubility/dissolution, underperforming bile enhancement models and/or lack of biorelevant solubility measurements. These results indicate areas for improvement in model software, modelling approaches, and generation of applicable input data. However, caution is required when interpreting the impact of drug-specific properties in this exercise, as the availability of input parameters was heterogeneous and highly variable, and the modellers generally used the data "as is" in this blinded bottom-up prediction approach.


Subject(s)
Biopharmaceutics/methods , Computer Simulation , Models, Biological , Pharmaceutical Preparations/classification , Pharmaceutical Preparations/metabolism , Administration, Oral , Drug Evaluation, Preclinical/methods , Forecasting , Humans , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Pharmaceutical Preparations/administration & dosage
11.
Eur J Pharm Sci ; 96: 610-625, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27816631

ABSTRACT

Orally administered drugs are subject to a number of barriers impacting bioavailability (Foral), causing challenges during drug and formulation development. Physiologically-based pharmacokinetic (PBPK) modelling can help during drug and formulation development by providing quantitative predictions through a systems approach. The performance of three available PBPK software packages (GI-Sim, Simcyp®, and GastroPlus™) were evaluated by comparing simulated and observed pharmacokinetic (PK) parameters. Since the availability of input parameters was heterogeneous and highly variable, caution is required when interpreting the results of this exercise. Additionally, this prospective simulation exercise may not be representative of prospective modelling in industry, as API information was limited to sparse details. 43 active pharmaceutical ingredients (APIs) from the OrBiTo database were selected for the exercise. Over 4000 simulation output files were generated, representing over 2550 study arm-institution-software combinations and approximately 600 human clinical study arms simulated with overlap. 84% of the simulated study arms represented administration of immediate release formulations, 11% prolonged or delayed release, and 5% intravenous (i.v.). Higher percentages of i.v. predicted area under the curve (AUC) were within two-fold of observed (52.9%) compared to per oral (p.o.) (37.2%), however, Foral and relative AUC (Frel) between p.o. formulations and solutions were generally well predicted (64.7% and 75.0%). Predictive performance declined progressing from i.v. to solution and immediate release tablet, indicating the compounding error with each layer of complexity. Overall performance was comparable to previous large-scale evaluations. A general overprediction of AUC was observed with average fold error (AFE) of 1.56 over all simulations. AFE ranged from 0.0361 to 64.0 across the 43 APIs, with 25 showing overpredictions. Discrepancies between software packages were observed for a few APIs, the largest being 606, 171, and 81.7-fold differences in AFE between SimCYP and GI-Sim, however average performance was relatively consistent across the three software platforms.


Subject(s)
Biopharmaceutics/methods , Computer Simulation , Models, Biological , Pharmaceutical Preparations/metabolism , Administration, Oral , Drug Evaluation, Preclinical/methods , Forecasting , Humans , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Pharmaceutical Preparations/administration & dosage
12.
Eur J Pharm Sci ; 96: 598-609, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27671970

ABSTRACT

Predicting oral bioavailability (Foral) is of importance for estimating systemic exposure of orally administered drugs. Physiologically-based pharmacokinetic (PBPK) modelling and simulation have been applied extensively in biopharmaceutics recently. The Oral Biopharmaceutical Tools (OrBiTo) project (Innovative Medicines Initiative) aims to develop and improve upon biopharmaceutical tools, including PBPK absorption models. A large-scale evaluation of PBPK models may be considered the first step. Here we characterise the OrBiTo active pharmaceutical ingredient (API) database for use in a large-scale simulation study. The OrBiTo database comprised 83 APIs and 1475 study arms. The database displayed a median logP of 3.60 (2.40-4.58), human blood-to-plasma ratio of 0.62 (0.57-0.71), and fraction unbound in plasma of 0.05 (0.01-0.17). The database mainly consisted of basic compounds (48.19%) and Biopharmaceutics Classification System class II compounds (55.81%). Median human intravenous clearance was 16.9L/h (interquartile range: 11.6-43.6L/h; n=23), volume of distribution was 80.8L (54.5-239L; n=23). The majority of oral formulations were immediate release (IR: 87.6%). Human Foral displayed a median of 0.415 (0.203-0.724; n=22) for IR formulations. The OrBiTo database was found to be largely representative of previously published datasets. 43 of the APIs were found to satisfy the minimum inclusion criteria for the simulation exercise, and many of these have significant gaps of other key parameters, which could potentially impact the interpretability of the simulation outcome. However, the OrBiTo simulation exercise represents a unique opportunity to perform a large-scale evaluation of the PBPK approach to predicting oral biopharmaceutics.


Subject(s)
Biopharmaceutics/methods , Databases, Factual , Models, Biological , Pharmaceutical Preparations/metabolism , Administration, Oral , Drug Evaluation, Preclinical/methods , Forecasting , Humans , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Pharmaceutical Preparations/administration & dosage
13.
Drug Dev Ind Pharm ; 40(11): 1421-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24467407

ABSTRACT

Bioequivalence testing of transdermal drug delivery systems (TDDS) has always been a subject of high concern for generic companies due to the formulation complexity and the fact that they are subtle to even minor manufacturing differences and hence should be clearly qualified in terms of quality, safety and efficacy. In recent times bioequivalence testing of transdermal patches has gained a global attention and many regulatory authorities worldwide have issued recommendations to set specific framework for demonstrating equivalence between two products. These current regulatory procedures demand a complete characterization of the generic formulation in terms of its physicochemical sameness, pharmacokinetics disposition, residual content and/or skin irritation/sensitization testing with respect to the reference formulation. This paper intends to highlight critical in vitro tests in assessing the therapeutic equivalence of products and also outlines their valuable applications in generic product success. Understanding these critical in vitro parameters can probably help to decode the complex bioequivalence outcomes, directing the generic companies to optimize the formulation design in reduced time intervals. It is difficult to summarize a common platform which covers all possible transdermal products; hence few case studies based on this approach has been presented in this review.


Subject(s)
Drug Delivery Systems/standards , Drugs, Generic/standards , Transdermal Patch/standards , Chemistry, Pharmaceutical , Drug Approval , Humans , In Vitro Techniques , Skin Absorption , Therapeutic Equivalency , United States , United States Food and Drug Administration/standards
14.
Pharm Dev Technol ; 19(4): 401-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23607272

ABSTRACT

In vivo equivalence of highly variable drugs (HVD) has always been a subject of great concern, in terms of both safety and efficacy, for regulatory agencies. Successful demonstration of their bioequivalence thus presents the most crucial component of a generic application, significantly contributing toward the cost and time of development. For poorly soluble drugs, such as telmisartan, dissolution represents the rate-limiting step in the gastric region and in many cases may not be complete, thereby contributing to low and highly variable bioavailability. Consequently, simulation of gastrointestinal conditions is essential to adequately predict the in vivo behavior of drug formulations. In this study, we evaluated usefulness of physiologically relevant dissolution method over commonly used acidic media to forecast comparable in vivo performance of telmisartan formulation to that of reference samples. In the present study, telmisartan was classified as a HVD and a partial replicate design with repeating the reference product and scaling the bioequivalence for the reference variability has been presented. The design has effectively decreased sample size, without increasing patient risk. Results from this project suggest that scaled average bioequivalence (SABE) provides a good approach for evaluating the bioequivalence of HVD, meeting the need for international guidelines for bioequivalence.


Subject(s)
Drugs, Generic/pharmacology , Drugs, Generic/pharmacokinetics , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Benzoates/pharmacokinetics , Benzoates/pharmacology , Biological Availability , Chemistry, Pharmaceutical/methods , Cross-Over Studies , Humans , Solubility , Tablets/pharmacology , Telmisartan , Therapeutic Equivalency
15.
Arzneimittelforschung ; 61(1): 23-31, 2011.
Article in English | MEDLINE | ID: mdl-21355443

ABSTRACT

The need for a reliable bioanalytical method is of primary importance during preclinical studies. The aim of the present study was simultaneous determination of pioglitazone (CAS 111025-46-8) (PIO) and glimepiride (CAS 93479-97-1) (GLM) in plasma of rats. A high-performance liquid chromatographic method has been developed and validated using C18 column and UV detector. A mobile phase composed of acetonitrile and ammonium acetate buffer pH 4.5 in the ratio of 55:45%. The plasma samples clean-up was carried out using solid phase cartridges. The method was in the linear range of 50-8000 ng/mL for PIO and 50-2000 ng/mL for GLM. The coefficient of regression was found to be > or = 0.99. Precision and accuracy were within the acceptable limits, as indicated by relative standard deviation varying from 1.5 to 6.1% for PIO and 3.1 to 7.0% for GLM whereas the accuracy ranged from 97.0 to 106.4% for PIO and 96.5 to 106.4% for GLM. The mean extraction recovery was found to be 90.2 +/- 4.5, 76.8 +/- 2.8 and 85.2 +/- 5.2% for PIO, GLM and internal standard, respectively. Moreover, PIO and GLM were stable in plasma, up to 30 days of storage at -70 degrees C and after being subjected to bench top, auto-sampler, and three freeze-thaw cycles. The developed method was applied for preclinical pharmacokinetic studies.


Subject(s)
Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Sulfonylurea Compounds/blood , Sulfonylurea Compounds/pharmacokinetics , Thiazolidinediones/blood , Thiazolidinediones/pharmacokinetics , Animals , Area Under Curve , Calibration , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical/methods , Drug Interactions , Half-Life , Male , Pioglitazone , Rats , Rats, Wistar , Reference Standards , Reproducibility of Results , Solid Phase Extraction , Spectrophotometry, Ultraviolet
16.
Bioanalysis ; 2(1): 95-104, 2010 Jan.
Article in English | MEDLINE | ID: mdl-21083123

ABSTRACT

BACKGROUND: A simple HPLC-UV method with a high reproducibility and sensitivity for the determination of talinolol in rat plasma was developed in this study. METHOD: After liquid-liquid extraction, the compounds were separated on a Vydac(®) C18 monomeric column (250 × 4.6 mm inner diameter × 5-µm particle size) using a mobile phase composed of acetonitrile and potassium dihydrogen phosphate buffer (34:66 v/v), delivered isocratically at a flow rate of 1.0 ml min(-1). Escitalopram was used as an internal standard. The chromatographic peak-area ratio, based on UV absorbency at 245 nm, was used for quantitative analysis. RESULTS: Calibration standards with concentrations over the range of 10-1000 ng ml(-1) were validated for routine sample analysis to support pharmacokinetic studies with talinolol in rats. The intra- and inter-day precision studies showed good reproducibility with coefficients of variation of less than 11.49%. The developed method is simpler and more sensitive than previously reported methods. DISCUSSION: The analytical sensitivity and accuracy of this assay were adequate for characterization of talinolol in rat plasma and the assay has been applied successfully to the in vivo kinetic study of talinolol in rats. After talinolol (10 mg kg(-1)) was given orally, the maximum concentration and the AUC(0-∞) were 341.8 ± 99.4 ng ml(-1) and 976.26 ± 173.37 ng h ml(-1), respectively. The oral bioavailability was approximately 52.14 ± 9.26%. CONCLUSION: The advantages of our method are a small sample volume (200 µl), short analysis time (13.5 min) and a simple sample extraction and clean-up compared with multiple extraction and washing steps and a longer analysis time in previously published methods.


Subject(s)
Adrenergic beta-Antagonists/blood , Adrenergic beta-Antagonists/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Propanolamines/blood , Propanolamines/pharmacokinetics , Animals , Least-Squares Analysis , Male , Rats , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity , Spectrophotometry, Ultraviolet/methods
17.
Eur J Pharm Sci ; 41(3-4): 440-51, 2010 Nov 20.
Article in English | MEDLINE | ID: mdl-20656025

ABSTRACT

Saquinavir (SQV) is a weak base compound, whose solubility is strongly influenced by pH variations. Thus, in the present work, we thought it worthy of interest to investigate in-depth the combined effect of pH control and cyclodextrin (CyD) complexation on SQV solubilization. Phase-solubility studies were performed by adding excess drug to buffered (pH from 1.1 to 7.4) aqueous solutions containing increasing concentrations of Methyl-Beta-CyD (M-ß-CyD) in order to evaluate the role of the unionized species of SQV in improving solubility by CyD complexation and to be able to select the most suitable conditions for optimizing drug solubilization. Our study reveals that the integrated approach of pH adjustment and CyD complexation can be successfully used for improving the CyD solubilizing power towards an ionizable drug such as SQV, thus allowing a smaller quantity of CyD to solubilize a given amount of drug, offering clear economic and technologic advantages as well. When biopharmaceutics of the optimized cyclodextrin-based formulation of SQV was studied in Wistar rats after intravenous and oral administrations, we found that inclusion of SQV into M-ß-CyD could dramatically improve its oral bioavailability and decrease the variation of its oral pharmacokinetics. Compared to the control, the presence of M-ß-CyD significantly increased the area under the plasma concentration-time curve (439.7±161.35 to 2312.03±159.53, p<0.01) and the peak plasma concentration (117.24±35.77 to 1347.88±276.76, p<0.01) of orally administered SQV. The modulating effect of M-ß-CyD on the bidirectional transport of SQV was also investigated using a modified Ussing chamber system. The results demonstrated that the enhancing effect of M-ß-CyD on the oral bioavailability of SQV is due not only to its solubilizing effect on SQV but also, at least in part, to the inhibitory effect of M-ß-CyD on the P-glycoprotein (P-gp) mediated efflux of SQV in the gastrointestinal tract. The present results suggest that M-ß-CyD is particularly useful in designing oral preparations of SQV with an enhanced bioavailability and a reduced variability in absorption.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , Saquinavir/pharmacokinetics , beta-Cyclodextrins/chemistry , Absorption , Administration, Oral , Animals , Chemistry, Pharmaceutical , Computer Simulation , HIV Protease Inhibitors/chemistry , Hydrogen-Ion Concentration , Molecular Structure , Rats , Rats, Wistar , Saquinavir/chemistry , Solubility
18.
Biopharm Drug Dispos ; 31(2-3): 202-14, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20238375

ABSTRACT

Most known interactions between herbal extracts and drugs involve the inhibition of drug-metabolizing enzymes, but little is yet known about the possible role of transporters in these interactions. In order to evaluate the effect of one of such prominent flavonoids, morin, on P-glycoprotein related efflux carriers, measurements of transport characteristics through Ussing chambers, in situ perfusion and in vivo drug absorption studies were performed with the transported, yet not metabolized model compound talinolol.This study investigated the effects of orally administered morin (1.0, 2.5 and 5.0 mg kg(-1)), on the pharmacokinetics of orally (10 mg kg(-1)) and intravenously (1.0 mg kg(-1)) administered talinolol in rats. In the presence of morin, the pharmacokinetic parameters of talinolol were significantly altered in the oral group but not in the intravenous group. The presence of 2.5 and 5.0 mg kg(-1) of morin significantly increased (1.8-2.0 fold, p<0.01) the area under the plasma concentration-time curve and the peak plasma concentration (2.3-3.0 fold, p<0.01) of orally administered talinolol. The absolute bioavailability (F %) of talinolol in the rats pretreated with morin was significantly higher (89.09-98.29%, p<0.01) than the control (52.14%). Talinolol demonstrated asymmetric transport across rat ileum with significantly greater basolateral-to-apical (B-A) permeability than that in the apical-to-basolateral (A-B) direction. The addition of morin resulted in a concentration dependent effect, especially on the secretory transport of talinolol.The present study demonstrates that morin bears the ability to interfere with secretory intestinal transport processes. This might be due to an interaction with P-glycoprotein.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antihypertensive Agents/pharmacokinetics , Biological Availability , Flavonoids/pharmacokinetics , Propanolamines/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Caco-2 Cells , Calcium Channel Blockers/pharmacokinetics , Drug Interactions/physiology , Humans , Intestinal Absorption/physiology , Rats , Rats, Wistar
19.
Talanta ; 76(2): 338-46, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18585287

ABSTRACT

A sensitive high performance liquid chromatographic (HPLC) method involving fluorescence detection was developed for the determination of fexofenadine (FEX), known to have low oral bioavailability, in rat plasma. In order to understand the effect of various chromatographic factors on the separation of analytes and to simultaneously optimize the resolution and analysis run time, a response surface method was used. The chromatographic separation was achieved using a Supelco C(18)-DB (250 mm x 4.6mm I.D./5 microm particle size) column with mobile phase comprising of ammonium acetate buffer and acetonitrile (63:37, v/v), delivered isocratically at a flow rate of 1.0 mL min(-1). Diphenhydramine was used as an internal standard (I.S.). The statistical evaluation of the method was examined and the method was found to be precise and accurate with a linearity range of 1-500 ng mL(-1) (r>0.9980). The intra- and inter-day precision studies showed good reproducibility with coefficients of variation (C.V.) less than 12.26%. The advantages of our method are small sample volume (100 microL), short time of analysis (13 min) and a simple sample extraction and clean-up as compared to the previously published methods. The established method provides a reliable bioanalytical methodology to carry out FEX pharmacokinetics in rat plasma.


Subject(s)
Chromatography, High Pressure Liquid/methods , Terfenadine/analogs & derivatives , Animals , Chromatography, High Pressure Liquid/standards , Fluorescence , Rats , Reproducibility of Results , Terfenadine/blood , Terfenadine/pharmacokinetics
20.
Anal Chim Acta ; 594(2): 248-56, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17586122

ABSTRACT

This research work aims to exploit the high selectivity and sensitivity of fluorescence detector to develop and validate a high performance liquid chromatography (HPLC) method having very small sampling volume, much better mass-sensitive detection limit and lower operating cost for the determination of Saquinavir (SQV), known to have low oral bioavailability, in rat plasma. Unlike the traditional methods that require at least 0.25 mL of plasma for each measurement, the present method requires only a 0.1 mL sample volume. This is very useful in reducing the blood collection from study rats, offering the possibility to make sufficient number of samples for pharmacokinetic study and minimizing the amount of blood-derived biological waste. After liquid-liquid extraction, the compounds were separated on a Vydac C18 monomeric column (250 mm x 4.6 mm i.d. x 5 microm particle size) using a mobile phase composed of acetonitrile and potassium dihydrogen phosphate buffer (45:55, v/v). Fluorescence detection was performed at 237 nm (excitation) and 380 nm (emission). Validity of the method was studied and the method was found to be precise and accurate with a linearity range from 0.005 to 1.000 microg mL(-1) (r>0.9980). The limit of detection (LOD) was found to be 0.001 microg mL(-1). The intra-day and inter-day precision studies showed good reproducibility with coefficients of variation (C.V.) less than 11.4%. The developed method was applied successfully to monitor the pharmacokinetic profile following oral administration of SQV to rats.


Subject(s)
HIV Protease Inhibitors/blood , Saquinavir/blood , Animals , Chromatography, High Pressure Liquid/methods , Fluorescence , HIV Protease Inhibitors/pharmacokinetics , Male , Rats , Rats, Wistar , Reproducibility of Results , Saquinavir/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...