scholarly journals Evaluation of Excipient Risk in BCS Class I and III Biowaivers

2022 ◽  
Vol 24 (1) ◽  
Author(s):  
Melissa Metry ◽  
James E. Polli

AbstractThe objective of this review article is to summarize literature data pertinent to potential excipient effects on intestinal drug permeability and transit. Despite the use of excipients in drug products for decades, considerable research efforts have been directed towards evaluating their potential effects on drug bioavailability. Potential excipient concerns stem from drug formulation changes (e.g., scale-up and post-approval changes, development of a new generic product). Regulatory agencies have established in vivo bioequivalence standards and, as a result, may waive the in vivo requirement, known as a biowaiver, for some oral products. Biowaiver acceptance criteria are based on the in vitro characterization of the drug substance and drug product using the Biopharmaceutics Classification System (BCS). Various regulatory guidance documents have been issued regarding BCS-based biowaivers, such that the current FDA guidance is more restrictive than prior guidance, specifically about excipient risk. In particular, sugar alcohols have been identified as potential absorption-modifying excipients. These biowaivers and excipient risks are discussed here.

Pharmaceutics ◽  
2019 ◽  
Vol 11 (5) ◽  
pp. 228 ◽  
Author(s):  
Yang Mai ◽  
Liu Dou ◽  
Christine M. Madla ◽  
Sudaxshina Murdan ◽  
Abdul W. Basit

It is known that males and females respond differently to medicines and that differences in drug behaviour are due to inter-individual variability and sex specificity. In this work, we have examined the influence of pharmaceutical excipients on drug bioavailability in males and females. Using a rat model, we report that a portfolio of polyoxyethylated solubilising excipients (polyethylene glycol 2000, Cremophor RH 40, Poloxamer 188 and Tween 80) increase ranitidine bioavailability in males but not in females. The in vivo sex and excipient effects were reflected in vitro in intestinal permeability experiments using an Ussing chamber system. The mechanism of such an effect on drug bioavailability is suggested to be due to the interaction between the excipients and the efflux membrane transporter P-glycoprotein (P-gp), whose expression in terms of gene and protein levels were inhibited by the solubilising agents in male but not in female rats. In contrast, the non-polyoxyethylated excipient, Span 20, significantly increased ranitidine bioavailability in both males and females in a non-sex-dependent manner. These findings have significant implications for the use of polyoxyethylated solubilising excipients in drug formulation in light of their sex-specific modulation on the bioavailability of drugs that are P-gp substrates. As such, pharmaceutical research is required to retract from a ‘one size fits all’ approach and to, instead, evaluate the potential impact of the interplay between excipients and sex on drug effect to ensure effective pharmacotherapy.


2020 ◽  
Author(s):  
Πάτρικ Ο' Ντουάιρ

Η βιοδιαθεσιμότητα ενός φαρμάκου μετά από per os χορήγηση επηρεάζεται από πολλές διαδικασίες. Η γαστρεντερική μεταφορά αποτελεί μια σημαντική φυσιολογική διαδικασία, που επηρεάζει της συγκεντρώσεις του φαρμάκου στο λεπτό έντερο, δηλαδή στην περιοχή από την οποία απορρόφουνται τις περισσότερες φορές τα per os χορηγούμενα φάρμακα. Στην παρούσα εργασία αναπτύχθηκαν δυο μικρής κλίμακας μέθοδοι για την εκτίμηση της σημασίας της γαστρεντερικής μεταφοράς στην απορρόφηση ενός φαρμάκου μετά από per os χορήγηση κατά τη διάρκεια της διαπεπτικής περιόδου. Οι μέθοδοι βασίστηκαν σε ένα διφασικό σύστημα διάλυσης δύο σταδίων και σε μια συσκευή διάλυσης-διαπέρασης δυο σταδίων. Η διφασική μέθοδος διάλυσης προσομοιώνει τη διαδικασία της μεταφοράς μέσω του εντερικού επιθηλίου με τη χρήση μιας στοιβάδας δεκανόλης. Η μέθοδος διάλυσης-διαπέρασης προσομοιώνει τη διαδικασία μεταφοράς μέσω του γαστρεντερικού επιθηλίου με τη χρήση μιας συνθετικής λιπιδικής μεμβράνης και ενός διαμερίσματος υποδοχής του φαρμάκου. Οι δυο μικρής κλίμακας μέθοδοι δυο σταδίων αναπτύχθηκαν και αξιολογήθηκαν με βάση δεδομένα δυο ασθενών βάσεων που ανήκουν στην Τάξη ΙΙ της Βιοφαρμακευτικής Ταξινόμησης των Φαρμάκων, της διπυριδαμόλης και της κετοκοναζόλης. Οι ρυθμοί καθίζησης, που υπολογίσθηκαν από τα δεδομένα των πειραμάτων με τις δυο μεθόδους, χρησιμοποιήθηκαν για την προσομοίωση των απεικονίσεων των συγκεντρώσεων στο πλάσμα μετά από χορήγηση σε ενήλικες με τη βοήθεια φυσιολογικών φαρμακοκινητικών μοντέλων. Οι τιμές των φαρμακοκινητικών παραμέτρων, που υπολογίσθηκαν από τις απεικονίσεις, συγκρίθηκαν με τις τιμές των φαρμακοκινητικών παραμέτρων, που είχαν υπολογισθεί από προηγούμενες κλινικές μελέτες των φαρμάκων σε υγιείς ενήλικες. Τα δεδομένα της διφασικής μεθόδου διάλυσης οδήγησαν σε καλύτερη εκτίμηση των φαρμακοκινητικών παραμέτρων των δυο ασθενών βάσεων. Τα δεδομένα της μεθόδου διάλυσης-διαπέρασης οδήγησαν σε υπερεκτίμηση της καθίζησης, πιθανότατα λόγω της αργής ροής μεταφοράς στο διαμέρισμα υποδοχής. Στη συνέχεια, οι δυο μικρής κλίμακας μέθοδοι δυο σταδίων αξιολογήθηκαν χρησιμοποιώντας φαρμακευτικά προϊόντα με αυξημένες δυνατότητες (enabling drug products) που περιλάμβαναν άμορφες στερεές διασπορές, ένα σύμπλοκο φαρμάκου με κυκλοδεξτρίνη και λιπιδικά προϊόντα. Για τη μελέτη των λιπιδικών προϊόντων η διφασική μέθοδος τροποποιήθηκε για να ενσωματωθεί και η διαδικασία της πέψης των εκδόχων. Συνολικά, η διφασική μέθοδος ήταν περισότερο χρήσιμη από τη μέθοδο-διάλυσης διαπέρασης και τα δεδομένα σχετίστηκαν καλύτερα με τα in vivo δεδομένα. Τέλος, η χρησιμότητα των δυο μικρής κλίμακας μεθόδων δυο σταδίων σε σχέση με δυο άλλες χρησιμοποιούμενες μεθόδους, τη διατάξη μικρού περιστρεφόμενου πτερυγίου Erweka και τη διάταξη ΒioGIT, μελετήθηκε με βάση δεδομένα δυο φαρμάκων που ανήκουν στην Τάξη ΙΙ της Βιοφαρμακευτικής Ταξινόμησης των Φαρμάκων, της δικλοφενάκης (ασθενές εξύ) και της ριτοναβίρης (ασθενής βάση). Με όλες τις μεθόδους, η δικλοφενάκη διαλύθηκε πολύ γρήγορα σε μέσα που προσομοιώνουν σε Επίπεδο ΙΙ τα περιεχόμενα του λεπτού εντέρου, τόσο μετά από μορφοποίηση σε σκόνη όσο και μετά από μορφοποίηση σε δισκίο. Φυσιολογική φαρμακοκινητική μοντελοποίηση των δεδομένων έδειξε, ότι μετά από χορήγηση προϊόντων άμεσης αποδέσμευσης δικλοφενάκης, η απορρόφηση εξαρτάται από τη γαστρική κένωση και τη μεταφορά μέσω του εντερικού επιθηλίου, ανεξάρτητα από τον τύπο του προϊόντος. Αντίθετα, η άμορφη στερεή διασπορά της ριτοναβίρης οδηγεί σε καθίζηση του φαρμάκου στον αυλό του λεπτού εντέρου. Όλες οι μέθοδοι που προσομοίωσαν την αλλαγή των συνθηκών από το στόμαχο στο λεπτό έντερο προσομοίωσαν επαρκώς την επίδραση του μειωμένου ρυθμού έκκρισης γαστρικού οξέος στη συμπεριφορά του προϊόντος in vivo. Με βάση τα δεδομένα συνολικά, κάθε μέθοδος παρουσιάζει ιδιαίτερα προτερήματα και αδυναμίες. Συνολικά, με βάση τα αποτελέσματα της παρούσας μελέτης η διφασική μέθοδος θα μπορούσε να διευκολύνει σημαντικά στα αρχικά στάδια της ανάπτυξης νέων φαρμάκων, ιδιαίτερα αν συνδυαστεί με φυσιολογικά φαρμακοκινητικά μοντέλα.


2021 ◽  
Vol 8 ◽  
Author(s):  
Bart Hens ◽  
Patrick Augustijns ◽  
Hans Lennernäs ◽  
Mark McAllister ◽  
Bertil Abrahamsson

A thorough understanding of the behavior of drug formulations in the human gastrointestinal (GI) tract is essential when working in the field of oral drug development in a pharmaceutical company. For orally administered drug products, various GI processes, including disintegration of the drug formulation, drugrelease, dissolution, precipitation, degradation, dosage form transit and permeation, dictate absorption into the systemic circulation. These processes are not always fully captured in predictive in vitro and in silico tools, as commonly applied in the pre-clinical stage of formulation drug development. A collaborative initiative focused on the science of oral biopharmaceutics was established in 2012 between academic institutions and industrial companies to innovate, optimize and validate these in vitro and in silico biopharmaceutical tools. From that perspective, the predictive power of these models can be revised and, if necessary, optimized to improve the accuracy toward predictions of the in vivo performance of orally administered drug products in patients. The IMI/EFPIA-funded “Oral Bioavailability Tools (OrBiTo)” project aimed to improve our fundamental understanding of the GI absorption process. The gathered information was integrated into the development of new (or already existing) laboratory tests and computer-based methods in order to deliver more accurate predictions of drug product behavior in a real-life setting. These methods were validated with the use of industrial data. Crucially, the ultimate goal of the project was to set up a scientific framework (i.e., decision trees) to guide the use of these new tools in drug development. The project aimed to facilitate and accelerate the formulation development process and to significantly reduce the need for animal experiments in this area as well as for human clinical studies in the future. With respect to the positive outcome for patients, high-quality oral medicines will be developed where the required dose is well-calculated and consistently provides an optimal clinical effect. In a first step, this manuscript summarizes the setup of the project and how data were collected across the different work packages. In a second step, case studies of how this project contributed to improved knowledge of oral drug delivery which can be used to develop improved products for patients will be illustrated.


Author(s):  
Bhikshapathi D. V. R. N. ◽  
Haarika B ◽  
Jyothi Sri S ◽  
K Abbulu

The purpose of present investigation was to develop floating matrix tablets of gemifloxacin mesylate, which after oral administration could prolong the gastric residence time, increase the drug bioavailability and diminish the side effects of irritating drugs. Tablets containing drug, various viscosity grades of hydroxypropyl methylcellulose such as HPMC K4M and HPMC K15M as matrix forming agent, Sodium bicarbonate as gas-forming agent and different additives were tested for their usefulness in formulating gastric floating tablets by direct compression method. The physical parameters, in vitro buoyancy, release characteristics and in vivo radiographic study were investigated in this study. The gemifloxacin mesylate floating tablets were prepared using HPMC K4M polymer giving more sustained drug release than the tablet containing HPMC K15M. All these formulations showed floating lag time of 30 to 47 sec and total floating time more than 12 h. The drug release was decreased when polymer concentration increases and gas generating agent decreases. Formulation that contains maximum concen-tration of both HPMC K15M and sodium bicarbonate (F9) showing sufficiently sustained with 99.2% of drug release at 12 h. The drug release from optimized formulation follows Higuchi model that indicates the diffusion controlled release. The best formulation (F9) was selected based on in vitro characteristics and used in vivo radiographic studies by incorporating barium sulphate as a radio-opaque agent and the tablet remained in the stomach for about 6 h.   


Pharmaceutics ◽  
2021 ◽  
Vol 13 (5) ◽  
pp. 710
Author(s):  
Tanja Ilić ◽  
Ivana Pantelić ◽  
Snežana Savić

Due to complex interdependent relationships affecting their microstructure, topical semisolid drug formulations face unique obstacles to the development of generics compared to other drug products. Traditionally, establishing bioequivalence is based on comparative clinical trials, which are expensive and often associated with high degrees of variability and low sensitivity in detecting formulation differences. To address this issue, leading regulatory agencies have aimed to advance guidelines relevant to topical generics, ultimately accepting different non-clinical, in vitro/in vivo surrogate methods for topical bioequivalence assessment. Unfortunately, according to both industry and academia stakeholders, these efforts are far from flawless, and often upsurge the potential for result variability and a number of other failure modes. This paper offers a comprehensive review of the literature focused on amending regulatory positions concerning the demonstration of (i) extended pharmaceutical equivalence and (ii) equivalence with respect to the efficacy of topical semisolids. The proposed corrective measures are disclosed and critically discussed, as they span from mere demands to widen the acceptance range (e.g., from ±10% to ±20%/±25% for rheology and in vitro release parameters highly prone to batch-to-batch variability) or reassess the optimal number of samples required to reach the desired statistical power, but also rely on specific data modeling or novel statistical approaches.


Drug Research ◽  
2018 ◽  
Vol 69 (03) ◽  
pp. 173-180 ◽  
Author(s):  
Bilal Houshaymi ◽  
Nadine Nasreddine ◽  
Mamdouh Kedees ◽  
Zeina Soayfane

AbstractThe bioavailability of ivermectin is modulated by lipid-based formulations and membrane efflux transporters such as Breast Cancer Resistance Protein and P-glycoprotein (BCRP and P-gp). We have investigated the effect of oleic acid on the uptake of ivermectin in vitro using Caco-2 cells and in vivo in the intestines of wild-type mice. Complex micelles (M) with oleic acid induced a significant increase (e. g. for M3 was 7-fold, p≤0.001) in the uptake of the drug in a time-dependent manner with no involvement of cholesterol in the mechanism. In vivo results showed a significant increase in the concentration of plasma and intestinal mucosa ivermectin (p≤0.01) in mice receiving oleic acid-based drug formulation. We also examined the expression of the drug efflux transporter, BCRP and P-gp in Caco-2 cells and found a significant decrease (p≤0.001) in their level in the presence of 5 mM oleic acid. Treatment of mice with oleic acid-based formulation showed a significant decrease in the activity of P-gp in the intestinal mucosa (p≤0.01). This study highlighted the effect of oleic acid in decreasing the expression and the activity of P-gp-mediated ivermectin efflux and in limiting the drug absorption by increasing its uptake and bioavailability in Caco-2 cells and intestine, respectively.


2021 ◽  
Author(s):  
◽  
Oliver Bayley

<p>Cancer is currently the second largest cause of death globally, leading to a high demand for new and effective chemotherapeutics. For years, natural products have been used as a source of new bioactive compounds; of particular interest in this context, as a source of new chemotherapeutics. One chemotherapeutic candidate which has attracted significant attention in synthetic and medicinal chemistry communities, is peloruside A. Peloruside A is a bioactive secondary metabolite isolated from the New Zealand marine sponge Mycale hentscheli. Since its discovery, peloruside A has shown great promise in cancer studies both in vivo and in vitro with effects observed even at nanomolar concentrations. These chemotherapeutic effects have been shown to occur by halting cell division at the G2/M checkpoint via microtubule stabilisation. Of particular interest is that this stabilisation occurs in a manner distinct from that of the already established taxane class of microtubule stabilising drugs. This means that peloruside A is able to offer both inhibition of cell division in Taxol® resistant cells and synergistic inhibition alongside the current taxane drugs. Since peloruside A is not abundantly available from its natural source, there is a strong incentive for the development of new synthetic strategies for peloruside A production. Unfortunately attempts at aquaculture and attempts at developing an industrial scale synthesis have both proven unsuccessful thus far. In an attempt to overcome some of the difficulties with the scale up of peloruside, analogues have been developed that are intended to have similar bioactivity to peloruside A but simpler, more concise, synthetic routes. These analogues will also enable further elucidation of the binding properties of peloruside A. This project focuses on the generation of a functionalised pyran fragment, starting from a simple carbohydrate, that may be incorporated into the proposed analogues.</p>


1997 ◽  
Vol 4 (4) ◽  
pp. 23-32 ◽  
Author(s):  
Henry Malinowski ◽  
Patrick Marroum ◽  
Venkata Ramana Uppoor ◽  
William Gillespie ◽  
Hae-Young Ahn ◽  
...  

2021 ◽  
Vol 3 (1) ◽  
Author(s):  
Ryan J Duchatel ◽  
Abdul Mannan ◽  
Ameha S Woldu ◽  
Tom Hawtrey ◽  
Phoebe A Hindley ◽  
...  

Abstract Background Diffuse intrinsic pontine glioma (DIPG) is a fatal childhood brainstem tumor for which radiation is the only treatment. Case studies report a clinical response to ONC201 for patients with H3K27M-mutant gliomas. Oncoceutics (ONC201) is only available in the United States and Japan; however, in Germany, DIPG patients can be prescribed and dispensed a locally produced compound—ONC201 German-sourced ONC201 (GsONC201). Pediatric oncologists face the dilemma of supporting the administration of GsONC201 as conjecture surrounds its authenticity. Therefore, we compared GsONC201 to original ONC201 manufactured by Oncoceutics Inc. Methods Authenticity of GsONC201 was determined by high-resolution mass spectrometry and nuclear magnetic resonance spectroscopy. Biological activity was shown via assessment of on-target effects, in vitro growth, proliferation, and apoptosis analysis. Patient-derived xenograft mouse models were used to assess plasma and brain tissue pharmacokinetics, pharmacodynamics, and overall survival (OS). The clinical experience of 28 H3K27M+ mutant DIPG patients who received GsONC201 (2017–2020) was analyzed. Results GsONC201 harbored the authentic structure, however, was formulated as a free base rather than the dihydrochloride salt used in clinical trials. GsONC201 in vitro and in vivo efficacy and drug bioavailability studies showed no difference compared to Oncoceutics ONC201. Patients treated with GsONC201 (n = 28) showed a median OS of 18 months (P = .0007). GsONC201 patients who underwent reirradiation showed a median OS of 22 months compared to 12 months for GsONC201 patients who did not (P = .012). Conclusions This study confirms the biological activity of GsONC201 and documents the OS of patients who received the drug; however, GsONC201 was never used as a monotherapy.


2007 ◽  
pp. 557-588 ◽  
Author(s):  
Kazuya Maeda ◽  
Yuichi Sugiyama
Keyword(s):  

Sign in / Sign up

Export Citation Format

Share Document