Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Mol Pharm ; 17(1): 219-228, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31809062

RESUMEN

Amorphous phases are frequently employed to overcome the solubility limitation that is nowadays commonplace in developmental small-molecule drugs intended for oral administration. However, since the solubility enhancement has finite longevity (it is a "kinetic solubility" effect), characterizing its duration (i.e., the so-called "parachute" effect) can be important for optimizing a formulation with regard to its in vivo exposure. Two semiempirical models, based on dispersive kinetics theory, are evaluated for their ability to precisely describe experimental transients depicting a loss in supersaturation (initially generated by the dissolution of the amorphous phase) over time, as the solubilized drug recrystallizes. It is found that in cases where the drug solubility significantly exceeds that of the crystal at longer times, the mechanism has substantial "denucleation" (dissolution) character. On the other hand, "nucleation and growth" (recrystallization) kinetics best describe systems in which the recrystallization goes to completion within the experimental time frame. Kinetic solubility profiles taken from the recent literature are modeled for the following drugs: glibenclamide, indomethacin, loratadine, and terfenadine. In the last case, a combination of three different kinetic models, two classical ones plus the dispersive model, are used together in describing the entire dissolution-recrystallization transient of the drug, obtaining a fit of R2 = 0.993. By precisely characterizing the duration of the "parachute" in vitro (e.g., under biorelevant conditions), the proposed models can be useful in predicting trends and thereby guiding formulation development and optimization.


Asunto(s)
Preparaciones Farmacéuticas/química , Química Farmacéutica , Cristalización , Gliburida/química , Indometacina/química , Cinética , Loratadina/química , Modelos Químicos , Solubilidad , Terfenadina/química
2.
Phys Chem Chem Phys ; 19(31): 20523-20532, 2017 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-28730199

RESUMEN

Predicting the glass transition temperature (Tg) of mixtures has applications that span across industries and scientific disciplines. By plotting experimentally determined Tg values as a function of the glass composition, one can usually apply the Gordon-Taylor (G-T) equation to determine the slope, k, which subsequently can be used in Tg predictions. Traditionally viewed as a phenomenological/empirical model, this work proposes a physical basis for the G-T equation. The proposed equations allow for the calculation of k directly and, hence, they determine/predict the Tg values of mixtures algebraically. Two derivations for k are provided, one for strong glass-formers and the other for fragile mixtures, with the modeled trehalose-water and naproxen-indomethacin systems serving as examples of each. Separately, a new equation is described for the first time that allows for the direct determination of the crossover temperature, Tx, for fragile glass-formers. Lastly, the so-called "Rule of 2/3", which is commonly used to estimate the Tg of a pure amorphous phase based solely on the fusion/melting temperature, Tf, of the corresponding crystalline phase, is shown to be underpinned by the heat capacity ratio of the two phases referenced to a common temperature, as evidenced by the calculations put forth for indomethacin and felodipine.

3.
Anal Chem ; 84(10): 4357-72, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22497462

RESUMEN

The characterization of orally-delivered peptide pharmaceuticals presents several challenges to analytical methods in comparison to characterization of conventional small-molecule drugs. These challenges include the analysis and characterization of difficult-to-separate impurities, secondary structure, the amorphous solid-state form, and the integrity of enteric-coated drug delivery systems. This work presents the multidisciplinary analytical characterization of a parathyroid hormone (PTH) peptide active pharmaceutical ingredient (API) and an oral formulation of this API within enteric-coated sucrose spheres. The analysis of impurities and degradation products in API and formulated drug product was facilitated by the development of an ultrahigh-performance liquid chromatography (UHPLC) method for analysis by high-resolution mass spectrometry (MS). The use of UHPLC allowed for additional resolution needed to detect impurities and degradation products of interest. The secondary structure was probed using a combination of solution-state NMR, infrared, and circular dichroism spectroscopic methods. Solid-state NMR is used to detect amorphous API in a nondestructive manner directly within the coated sucrose sphere formulation. Fluorescence and Raman microscopy were used in conjunction with Raman mapping to show enteric coating integrity and observe the distribution of API beneath the enteric-coating on the sucrose spheres. The methods are combined in a multidisciplinary approach to characterize the quality of the enteric-coated peptide product.


Asunto(s)
Cromatografía Líquida de Alta Presión , Composición de Medicamentos , Espectrometría de Masas , Péptidos/análisis , Administración Oral , Secuencia de Aminoácidos , Humanos , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Hormona Paratiroidea/análisis , Hormona Paratiroidea/genética , Hormona Paratiroidea/metabolismo , Estructura Secundaria de Proteína , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría Raman , Sacarosa/química
4.
J Pharm Sci ; 109(4): 1627-1629, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31930977

RESUMEN

The solubility enhancement generated by an amorphous phase over its crystalline counterpart is unaffected by the pH of the solution (at fixed ionic strength and temperature), even for a drug containing ionizable moieties, provided that both the ionized and neutral species generated through dissolution of the solids remain dissolved.


Asunto(s)
Preparaciones Farmacéuticas , Cristalización , Concentración de Iones de Hidrógeno , Concentración Osmolar , Solubilidad
5.
Int J Pharm ; 567: 118465, 2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31279056

RESUMEN

The accurate prediction of the solubility enhancement offered by neat amorphous drugs and amorphous solid dispersions, over their crystalline (API) counterparts, has been discussed in several landmark works dating back at least two decades. Against this backdrop, an assessment of the current state-of-the-art for rigorously, yet simply (circumventing computational methods), determining the amorphous:crystalline solubility ratio based on thermo-analytical quantities is presented herein. Included in this work is a brief survey of the literature together with a discussion of the advantages and shortcomings of some of the most popular approaches, to-date. While the focus is on neat amorphous drugs, both before and after moisture sorption, the methodology presented is readily extended to more complex (e.g. ternary) systems that form a single, homogeneous phase. Six key questions are addressed in the context of how to most accurately determine the amorphous:crystalline solubility ratio: (1) How is the lattice energy of the crystalline phase assessed? (2) What is the role of heat capacity? (3) How does the pKa impact the solubility ratio prediction (for ionizable drugs)? (4) How does one incorporate the effects of moisture sorption on the amorphous phase? (5) How might one characterize (predict) the rate of drug recrystallization under various conditions (since the duration of the solubility enhancement is a kinetic phenomenon)? (6) What is the best approach for linking the (loss in) solubility enhancement to the Tg-lowering of the amorphous drug (by water) and vice-versa? In addressing these questions, this work aims to put forth a standardized methodology for determining the amorphous solubility enhancement with improved accuracy.


Asunto(s)
Modelos Teóricos , Química Farmacéutica , Cristalización , Cinética , Preparaciones Farmacéuticas/química , Solubilidad , Termodinámica
6.
Int J Pharm ; 555: 100-108, 2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-30448307

RESUMEN

Water is often readily absorbed by amorphous compounds, lowering their glass transition temperature (Tg) and facilitating their recrystallization (via nucleation-and-growth). At the same time, the increase in moisture content translates to a decrease in both the thermodynamic solubility and intrinsic dissolution rate, as compared to the corresponding dry (pure) amorphous phase, e.g. see [Murdande SB, Pikal MJ, Shanker RM, Bogner RH. 2010. Solubility advantage of amorphous pharmaceuticals: I. A thermodynamic analysis. J Pharm Sci 99:1254-1264.]. In the case of pure indomethacin and felodipine, the solubility advantage of each amorphous phase over its crystalline counterpart were previously determined to be 7.6 and 4.7, respectively, using a new methodology together with basic calorimetric data taken from the literature. Herein, we demonstrate that, theoretically, following the uptake of just ∼0.5% w/w water, the solubility ratios decrease to 6.9 and 4.5, in the same order. Moreover, as the predicted intrinsic dissolution rate (based on the Noyes-Whitney equation) is directly proportional to the solubility advantage of a given amorphous-crystalline pair, it decreases proportionately upon moisture uptake. Applying the methodology presented herein, one can directly predict the extent of Tg-lowering observed at any moisture content, for a given amorphous phase. Knowing that value, it is possible to estimate the relative decrease in the solubility and/or intrinsic dissolution rate of the plasticized phase compared to the pure glass, and vice-versa.


Asunto(s)
Química Farmacéutica/métodos , Felodipino/administración & dosificación , Indometacina/administración & dosificación , Agua/química , Calorimetría/métodos , Cristalización , Liberación de Fármacos , Felodipino/química , Indometacina/química , Solubilidad , Termodinámica , Temperatura de Transición , Vitrificación
7.
J Pharm Sci ; 105(9): 2625-2630, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27372548

RESUMEN

Use of amorphous phases can mitigate the low in vivo exposures of poorly soluble, crystalline active pharmaceutical ingredients. However, it remains challenging to accurately predict the solubility enhancement offered even by a pure amorphous phase relative to the crystalline form. In this work, a methodology is presented that allows estimation of the amorphous:crystalline solubility ratio, α, using only measured thermodynamic quantities for each of the pure phases. With this approach, α values of 7.6 and 4.7 were calculated for indomethacin and felodipine, respectively, correlating more closely than previous predictions with the experimentally measured values of 4.9 and 4.7 reported in the literature. There are 3 key benefits to this approach. First, it uses simple mathematical functions to more precisely relate the temperature variations in the heat capacity (Cp) to allow a more accurate estimation of the configurational energy difference between the 2 phases, whereas traditional models typically assume that Cp of both phases are constant(s). Second, the Hoffman equation is leveraged in translating the free energy of crystal lattice formation to the actual temperature of interest (selected to be 25°C/298K in this work), again, for better accuracy. Finally, as only 2 modulated differential scanning calorimetry scans are required (one for each phase), it is attractive from an experimental simplicity standpoint.


Asunto(s)
Felodipino/química , Indometacina/química , Modelos Químicos , Química Farmacéutica , Cristalización , Solubilidad , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA