Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
AAPS PharmSciTech ; 25(1): 12, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38182862

RESUMO

This study outlines a practical approach for assessing chemical instability by heating the drug-excipient binary mixtures or multi-excipient formulations at 75°C for 3 days before characterization. Differentiating itself from other excipient compatibility methods, our methodology necessitates a saturated aqueous slurry rather than arbitrarily fixed water content. This allows bulk and surface water in the excipient to contribute to drug degradation. The synergistic impact of surface water and elevated temperature expedites degradation kinetics, resulting in accelerated data generation. Among excipient compatibility methods available, our method is quantitative and merges with traditionally used methodologies. The devised nomograph enables extrapolation of shelf life at 20°C from experimental data obtained at 75°C. This methodology also helped identify stabilizers for the drug NVS-1 where traditional excipient compatibility programs had failed. Incorporation of monovalent salts, such as sodium/potassium chloride and sodium bicarbonate at 5% w/w, significantly enhanced the chemical stability of NVS-1, ensuring stable tablet formulations. Our hypothesis posits that stabilization is due to increased ionic strength in the slurry, which stabilizes an induced dipole within the polar NVS-1 drug. Additionally, the presence of ions in the moisture layer is anticipated to stabilize π-π stacking of two planar aromatic NVS-1 molecules. The expedited generation of experimental data allowed the identification of inorganic salts to supplement a standard excipient compatibility screening panel. Considering the economic implications of stability testing methodologies in effort, cost, and duration, a faster turnaround in chemical stability data enhances formulation selection. This ultimately facilitates the development of drug formulations with greater efficiency without delays.


Assuntos
Excipientes , Sais , Suplementos Nutricionais , Calefação , Água
2.
AAPS PharmSciTech ; 24(6): 149, 2023 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-37420118

RESUMO

This technical note investigated the loss of dissolution rate during accelerated stability studies with a dry blend capsule formulation containing an amorphous salt of drug NVS-1 (Tg 76°C). After 6 m at 40°C/75%RH, dissolution of NVS-1 was ≤40% of initial value. Scanning electron microscope characterization of the undissolved capsule contents from samples stored at 50°C/75%RH for 3 weeks showed agglomeration with a distinct "melt and fuse" morphology of particles. At elevated temperature and humidity conditions, undesired sintering among the amorphous drug particles was observed. Humidity plasticizes the drug as the stability temperature (T) gets closer to the glass transition temperature (Tg) of the amorphous salt (i.e., smaller Tg-T); a decreased viscosity favors viscoplastic deformation and sintering of drug particles. When moisture is adsorbed onto agglomerated drug particles, partial dissolution of the drug forms a viscous surface layer, further reducing the rate of dissolution media penetration into the bulk solid, hence the slower dissolution rate. Formulation intervention focused on the use of L-HPC and fumed silica as disintegrant and glidant and the removal of the hygroscopic crospovidone. Reformulation improved dissolution performance at short-term accelerated stability conditions of 50°C (± 75%RH); however, sintering to a lesser extent was still observed at high humidity, impacting the dissolution rate. We infer reducing the impact of moisture at high humidity conditions in a formulation with a 34% drug load is challenging. Future formulation efforts will focus on the addition of water scavengers, reducing drug load by ~50% to physically separate drug particles by water-insoluble excipients, and optimizing disintegrant levels.


Assuntos
Água , Solubilidade , Cristalização , Estabilidade de Medicamentos , Temperatura de Transição
3.
AAPS J ; 25(1): 25, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36788163

RESUMO

Physiologically based pharmacokinetic and absorption modeling has increasingly been implemented for biopharmaceutics applications to define the safe space for drug product quality attributes such as dissolution. For fevipiprant/QAW039, simulations were performed to assess the impact of in vitro dissolution on the in vivo performance of immediate-release film-coated tablets during development and scaling up to commercial scale. A fevipiprant dissolution safe space was established using observed clinical intravenous and oral PK data from bioequivalent and non-bioequivalent formulations. Quality control dissolution profiles with tablets were used as GastroPlus™ model inputs to estimate the in vivo dissolution in the gastrointestinal tract and to simulate human exposure. The model was used to evaluate the intraluminal performance of the dosage forms and to predict the absorption rate limits for the 450 mg dose. The predictive model performance was demonstrated for various oral dosage forms (150‒500 mg), including the non-bioequivalent batches in fasted healthy adults. To define the safe space at 450 mg, simulations were performed using theoretical dissolution profiles. A specification of Q = 80% dissolved in 60 min or less for an immediate-release oral solid dosage form reflected the boundaries of the safe space. The dissolution profile of the 450 mg commercial scale batch was within a dissolution region where bioequivalence is anticipated, not near an edge of failure for dissolution, providing additional confidence to the proposed acceptance criteria. Thus, the safe space allowed for a wider than 10% dissolution difference for bioequivalent batches, superseding f2 similarity analyses.


Assuntos
Biofarmácia , Modelos Biológicos , Adulto , Humanos , Solubilidade , Equivalência Terapêutica , Comprimidos , Administração Oral
4.
Drug Dev Ind Pharm ; 48(5): 211-225, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35861393

RESUMO

OBJECTIVE: This work summarizes select methodology of twin-screw melt granulation (TSMG) and process analytical technology that were used in the successful scaling-up and commercial transfer of high drug load (80.5% w/w) immediate release fevipiprant tablets. SIGNIFICANCE: The unique and compelling learnings from this industry work are (1) insights into Novartis AG's commercial scale transfer using TSMG and (2) rapid, nondestructive NIR methodology as a PAT tool for RTR testing. No prior literature combines these two aspects at the level of detail we present/disclose. METHODS: Scaling up of TSMG was guided by specific energy values obtained for the 27 mm (pilot scale) and 50 mm (commercial scale) twin-screw extruders (TSE). Proven acceptable ranges (PARs) were confirmed by varying the critical process parameters (CPPs) for granulation (screw speed) and tableting (dwell time and crushing strength) at three process levels (upper, target, and lower). An at-line NIR method was developed and validated for real-time release testing (RTRT). RESULTS: The combination of CPPs were selected to have the same effect on critical quality attributes (CQAs), that is, lower (-) and upper (+) process level challenged tablet aspect/appearance and dissolution, respectively. TSMG was performed using a 50 mm extruder at constant feed rate. Compression of the six final blends (∼300 kg) showed no impact of varied granulation and compression process conditions on both CQAs. A near-infrared spectroscopy method was validated to determine content uniformity, assay, identity, and to predict CQAs on uncoated tablets in preparation for a real RTRT of future batches.


Assuntos
Excipientes , Piridinas , Composição de Medicamentos/métodos , Excipientes/química , Ácidos Indolacéticos , Comprimidos/química , Tecnologia Farmacêutica/métodos
5.
J Pharm Sci ; 106(2): 579-588, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27938895

RESUMO

Process-induced inadvertent phase change of an active pharmaceutical ingredient in a drug product could impact chemical stability, physical stability, shelf life, and bioperformance. In this study, dispersive Raman spectroscopy is presented as an alternative method for the nondestructive, high-throughput, at-line quantification of amorphous conversion. A quantitative Raman method was developed using a multivariate partial least squares (PLS) regression calibration technique with solid-state nuclear magnetic resonance (ssNMR) spectroscopy as the reference method. Compositionally identical calibration tablets containing 20% w/w total MK-A drug in varying weight proportions (0%-50% w/w based on total MK-A) of amorphous and crystalline MK-A were compressed at 10-45 kN force. PLS predictions of amorphous content of tablets using Raman spectroscopy correlated well with ssNMR quantification. The predictive accuracy of this model led to a strong correlation (R2 = 0.987) with a root mean-squared error of prediction of 1.5% w/w amorphous MK-A in tablets up to 50% w/w amorphous conversion in compressive stress range of 60-320 MPa. Overall, these results suggest that dispersive Raman spectroscopy offers fast, sensitive, and high-throughput (<5 min/tablet) method for quantitating amorphous conversion.


Assuntos
Preparações Farmacêuticas/química , Análise Espectral Raman/métodos , Cristalização , Análise dos Mínimos Quadrados , Espectroscopia de Ressonância Magnética , Análise Multivariada , Comprimidos
6.
J Pharm Sci ; 105(9): 2498-2508, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26886316

RESUMO

Maximizing oral bioavailability of drug candidates represents a challenge in the pharmaceutical industry. In recent years, there has been an increase in the use of amorphous solid dispersions (ASDs) to address this issue, where a growing number of solid dispersion formulations have been introduced to the market. However, an increase in solubility or dissolution rate through ASD does not always result in sufficient improvement of oral absorption because solubility limitations may still exist at high doses. Chemical modification in the form of a prodrug may offer an alternative approach for these cases. Although prodrugs have been primarily used to improve membrane permeability, examples are available in which prodrugs have been used to increase drug solubility beyond what can be achieved via formulation approaches. In this mini review, the role of ASDs and prodrugs as 2 complementary approaches in improving oral bioavailability of drug candidates is discussed. We discuss the fundamental principles of absorption and bioavailability, and review available literature on both solid dispersions and prodrugs, providing a summary of their use and examples of successful applications, and cover some of the biopharmaceutics evaluation aspects for these approaches.


Assuntos
Pró-Fármacos/química , Tecnologia Farmacêutica/métodos , Administração Oral , Disponibilidade Biológica , Portadores de Fármacos/química , Excipientes/química , Humanos , Absorção pela Mucosa Oral , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Solubilidade
7.
J Pharm Sci ; 99(6): 2711-23, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20020525

RESUMO

The C-phosphoryloxymethyl (POM) and phosphoryloxymethyloxymethyl (POMOM) prodrugs resulting from derivatization at the reactive alpha-carbon of beta-dicarbonyl carbon acid drugs represent a unique approach for improving their chemical stability and aqueous solubility. This work evaluates the physicochemical and in vitro enzymatic bioconversion lability of selected prodrugs of phenylbutazone and phenindione. The POM and POMOM prodrug derivatives of phenylbutazone are highly water soluble (>or=250 mg/mL), chemically stable with projected shelf-lives of 4.5 years (pH 3.5, 25 degrees C) and 1.1 years (pH 6.0, 25 degrees C), respectively. Interestingly, both prodrug derivatives do not display a pH-dependency typical of many phosphate monoesters, although the similarities of their apparent thermodynamic activation parameters indicate a hydrolysis mechanism similar to other phosphates. These prodrugs undergo alkaline phosphatases catalyzed bioconversion to their respective carbon acids with an expected faster rate exhibited by the POMOM derivatives. Additionally, in marked contrast to the oxidative instability of phenindione, its POM prodrug is stable. The results from these studies reaffirm the rationale of transiently "masking" the reactive alpha-carbon/proton bond by covalently incorporating a POM or POMOM promoiety. This prodrug strategy presents a twofold advantage, enhancement of aqueous solubility and prevention of oxidative instability, two intrinsic formulation limitations found for beta-dicarbonyl carbon acid drugs.


Assuntos
Pró-Fármacos/química , Pró-Fármacos/síntese química , Ácidos , Fosfatase Alcalina/química , Ácido Carbônico , Catálise , Química Farmacêutica , Humanos , Hidrólise , Fenilbutazona , Prótons , Solubilidade
8.
J Pharm Sci ; 98(5): 1804-12, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18781645

RESUMO

The kinetics of conversion of C-hydroxymethyl derivatives of pharmaceutically relevant beta-dicarbonyl carbon acids of two series, pyrazolidin-3,5-diones and inden-1,3-diones, and a model carbon acid back to the respective carbon acids were studied as a function of pH at 25 degrees C and an ionic strength of 0.15 M. This is a somewhat surprising reaction since it involves the facile breakdown of a carbon-carbon bond. The slopes of the pH-rate profiles for the dehydroxymethylation were approximately unity, which along with the lack of buffer catalysis, indicates a specific-base mechanism involving spontaneous breakdown of the oxymethyl anion. This breakdown generates the conjugate base of the respective carbon acids. Thus within a series, there exists a correlation between the second-order rate constant for dehydroxymethylation and the pK(a) of the corresponding carbon acid with a shorter conversion/dehydroxymethylation half-life (at all given pH values) with decreasing pK(a) of the parent carbon acid. The increasing acidity of the carbon acid affords an increase in the leaving group ability of the carbanion, and therefore facilitation of the rate-determining unimolecular carbon-carbon bond cleavage. Since the hydroxymethyl derivative is an intermediate in the bioconversion of C-phosphoryloxymethyl prodrugs of carbon acids, also under study, the relationship allows one to reasonably predict how facile the dehydroxymethylation would be for any new beta-dicarbonyl carbon acid.


Assuntos
Cetonas/química , Pró-Fármacos/química , Pirazóis/química , Fosfatase Alcalina/química , Soluções Tampão , Meia-Vida , Concentração de Íons de Hidrogênio , Indicadores e Reagentes , Cinética , Espectrofotometria Ultravioleta
9.
J Pharm Sci ; 97(10): 4184-93, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18288723

RESUMO

The title of this commentary contains a frequently asked question whenever someone presents or proposes a prodrug strategy that releases formaldehyde as a result of bioconversion of a prodrug to parent drug. Formaldehyde, a highly water-soluble one-carbon molecule, is endogenous to cells, tissues, and body fluids. Although formaldehyde is generated and incorporated into essential metabolic processes by the human body, exposure to large amounts of formaldehyde vapor can irritate the nasal mucosa and may potentially be carcinogenic. It also gives a positive Ames test. Metabolism of both endogenous and exogenous formaldehyde involves rapid oxidation to formic acid catalyzed by glutathione dependent and independent dehydrogenases in the liver and erythrocytes. Balancing this rapid detoxification pathway is endogenous formation from normal metabolic processes and exogenous formaldehyde input, resulting in approximately 0.1 mM systemic levels. The possibility that formaldehyde released upon bioconversion of prodrugs might induce toxicity has been repeatedly stated, but no convincing evidence for this perceived toxicity has been documented in experimental studies. Therefore, as pharmaceutical chemists and not as toxicologists, we present our perspective on the apparent concern with release of formaldehyde as a by-product of in vivo bioconversion of selective prodrugs, and suggest that in comparison to the total amount of daily endogenous formaldehyde production from metabolism, and exogenous exposure from food and the environment, the amount generated by prodrugs is minute and is unlikely to cause any systemic toxicity in humans. Such an argument does not preclude formaldehyde-based toxicity assessment of a prodrug. Instead, it reduces the risk that in vivo liberation of formaldehyde will cause undue toxicity.


Assuntos
Formaldeído/farmacocinética , Pró-Fármacos/farmacocinética , Adulto , Humanos , Exposição por Inalação , Metanol/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...