Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(22)2023 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-37998348

RESUMO

STIM1 has been identified as a new warm sensor, but the exact molecular mechanism remains unclear. In this study, a variety of mutants of STIM1, Orai1 and Orai3 were generated. The single-cell calcium imaging and confocal analysis were used to evaluate the thermal sensitivity of the resulting STIM mutants and the interaction between STIM1 and Orai mutants in response to temperature. Our results suggested that the CC1-SOAR of STIM1 was a direct activation domain of temperature, leading to subsequent STIM1 activation, and the transmembrane (TM) region and K domain but not EF-SAM were needed for this process. Furthermore, both the TM and SOAR domains exhibited similarities and differences between STIM1-mediated thermal sensation and store-operated calcium entry (SOCE), and the key sites of Orai1 showed similar roles in these two responses. Additionally, the TM23 (comprising TM2, loop2, and TM3) region of Orai1 was identified as the key domain determining the STIM1/Orai1 thermal response pattern, while the temperature reactive mode of STIM1/Orai3 seemed to result from a combined effect of Orai3. These findings provide important support for the specific molecular mechanism of STIM1-induced thermal response, as well as the interaction mechanism of STIM1 with Orai1 and Orai3 after being activated by temperature.


Assuntos
Canais de Cálcio , Cálcio , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Domínios Proteicos , Sensação
2.
Bot Stud ; 64(1): 11, 2023 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-37079162

RESUMO

BACKGROUND: Bakanae disease caused by Fusarium fujikuroi is an increasing threat to rice production. The infected plants show symptoms such as elongation, slenderness, chlorosis, a large leaf angle, and even death. Bakanae disease is traditionally managed by seed treatment. However, fungicide-resistant F. fujikuroi isolates have emerged in several Asian areas, including Taiwan. This study aimed to identify new bakanae resistance quantitative trait loci (QTLs) and provide molecular markers to assist future breeding. RESULTS: A population of F2:9 recombinant inbred lines (RILs) was derived from the cross between an elite japonica Taiwanese cultivar 'Taikeng 16 (TK16)' and an indica variety 'Budda'. 'Budda' was found highly resistant to all 24 representative isolates of the F. fujikuroi population in Taiwan. For the RIL population, 6,492 polymorphic single nucleotide polymorphisms (SNPs) spanning the rice genome were obtained by genotyping-by-sequencing (GBS) technique, and the disease severity index (DSI) was evaluated by inoculation with a highly virulent F. fujikuroi isolate Ff266. Trait-marker association analysis of 166 RILs identified two QTLs in 'Budda'. qBK2.1 (21.97-30.15 Mb) is a novel and first bakanae resistance QTL identified on chromosome 2. qBK1.8 (5.24-8.66 Mb) partially overlaps with the previously reported qBK1.3 (4.65-8.41 Mb) on chromosome 1. The log of odds (LOD) scores of qBK1.8 and qBK2.1 were 4.75 and 6.13, accounting for 4.9% and 8.1% of the total phenotypic variation, respectively. 64 RILs carrying both qBK1.8 and qBK2.1 showed lower DSI (7%) than the lines carrying only qBK1.8 (15%), only qBK2.1 (13%), or none of the two QTLs (21%). For the future application of identified QTLs, 11 KBioscience competitive allele-specific PCR (KASP) markers and 3 insertion-deletion (InDel) markers were developed. CONCLUSIONS: Compared to other important rice diseases, knowledge of bakanae resistance has been insufficient, which limited the development and deployment of resistant cultivars. The discovery of qBK2.1 has provided a new source of bakanae resistance. The resistant RILs inheriting good plant type, good taste, and high yield characteristics from 'TK16' can be used as good resistance donors. Our newly developed markers targeting qBK2.1 and qBK1.8 can also serve as an important basis for future fine-mapping and resistance breeding.

3.
Mol Pharm ; 20(1): 409-418, 2023 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-36529939

RESUMO

Inhomogeneity is a key factor that significantly influences the dissolution behavior of amorphous solid dispersion (ASD). However, the underlying mechanisms of the effects of inhomogeneous phase on the dissolution characteristics as well as the bioavailability of ASDs are still unclear. In this study, two types of felodipine/PVPVA based ASDs with 30 wt % drug loading but different homogeneity were prepared: homogeneous "30 wt % ASD" prepared by spray drying, as well as inhomogeneous "30 wt % PM" prepared by physically mixing the sprayed dried 70 wt % ASD with PVPVA. We aimed to investigate (1) drug-polymer interaction mechanism and "apparent" interaction strength within the two ASDs and (2) dissolution mechanism as well as in vivo performance of the two ASDs. DSC thermogram revealing a single Tg in 30 wt % ASD confirmed its homogeneous phase. 1H NMR, FT-IR, and DVS studies collectively proved that strong hydrogen bonding interactions formed between felodipine and PVPVA in ASDs. Moreover, homogeneous "30 wt % ASD" has more numbers of interacting drug-polymer pairs, and thus exhibits stronger "apparent" interaction strength comparing with that of inhomogeneous "30 wt % PM". Unexpectedly,in the in vitro dissolution studies, inhomogeneous "30 wt % PM" showed much faster dissolution and also generated drug concentration ∼4.4 times higher than that of homogeneous "30 wt % ASD". However, drug precipitate recrystallized much slower in homogeneous "30 wt % ASD", presumably because much more polymer coprecipitated with amorphous drug in this system, which helps inhibiting drug crystallization. Surprisingly, homogeneous "30 wt % ASD" showed a significantly higher bioavailability in the in vivo pharmacokinetic studies, with the maximum plasma concentrations (Cmax) and the area under the curve (AUC) values of about 2.7 and 2.3 times higher than those of inhomogeneous "30 wt % PM". The above findings indicated that the amorphous state of drug precipitate contributes significantly to increase bioavailability of ASDs, while traditional in vitro dissolution studies, for instance, if we only compare the dissolved drug in solution or the capability of an ASD to generate supersaturation, are inadequate to predict in vivo performance of ASDs. In conclusion, the phase behavior of ASDs directly impact the formation of drug-polymer interaction, which controls not only drug supersaturation in solution but also drug crystallization in precipitate, and ultimately affect the in vivo performance of ASDs.


Assuntos
Felodipino , Polímeros , Felodipino/química , Solubilidade , Disponibilidade Biológica , Espectroscopia de Infravermelho com Transformada de Fourier , Polímeros/química , Liberação Controlada de Fármacos
5.
Pharm Res ; 36(7): 105, 2019 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-31102031

RESUMO

PURPOSE: To reveal the underlying mechanism inducing the opposite trends of surface composition enrichment of spray dried amorphous solid dispersions (ASD) of sorafenib and regorafenib, two compounds only differ in hydrogen to fluorine substitution. METHODS: Sorafenib/PVP and regorafenib/PVP ASDs were prepared by spray drying. Morphology of ASDs was visually inspected and examined by SEM. The surface compositions of ASDs were analyzed by XPS. Glass transition temperature (Tg) of ASDs was determined by DSC. Water vapor sorption isotherms of ASDs were studied by moisture sorption analyzer. Molecular interaction between the drug and the polymer was analyzed by solution NMR. RESULTS: In 10% and 20% drug loading sorafenib/PVP ASDs, short time moisture exposure induced PVP enrichment on the surface, and the appearance of initial ASDs powder became gel-like after water uptake. While in 30% sorafenib/PVP and any regorafenib/PVP ASDs regardless of drug loading, moisture exposure induced surface drug enrichment, while their powder-like appearance and average particle size remained unchanged. Meanwhile, sorafenib/PVP had similar water vapor sorption isotherms as regorafenib/PVP, before and after moisture induced phase separation. NMR study demonstrated a hex atomic ring H-bonding interaction between the drug and PVP, with a 1:1 drug: monomer stoichiometry molar ratio, which persisted in sorafenib/PVP but not regorafenib/PVP system under 95%RH moisture. CONCLUSIONS: Moisture exposure could lead to drug or polymer enrichment on the surface of ASDs, while the viability of drug-polymer interaction persisting in water environment contributed to such surface composition enrichment.


Assuntos
Flúor/química , Hidrogênio/química , Sorafenibe/química , Umidade , Transição de Fase , Compostos de Fenilureia/química , Polímeros/química , Povidona/química , Piridinas/química , Solubilidade , Vapor , Propriedades de Superfície
7.
Mol Pharm ; 16(1): 205-213, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30452278

RESUMO

We investigated the drug-polymer interactions in nonaqueous and aqueous environments between a poorly water-soluble drug, BAY1161909 (909), and two commonly used polymers in amorphous solid dispersions, i.e., PVP and HPMC-AS. In an nonaqueous state, with a drug-polymer Flory-Huggins interaction parameter, solution NMR and FT-IR results revealed that strong specific interactions existed between 909 and PVP, while not between 909 and HPMC-AS. After prolonged moisture exposure under 95% RH, 909/PVP intermolecular interaction no longer existed, while hydrophobic interaction between 909 and HPMC-AS occurred and persisted. In an aqueous supersaturation study of 909, codissolved PVP significantly outperformed predissolved PVP in maintaining 909 supersaturation. We hypothesized that the codissolved PVP formed a specific interaction with 909, and thus, it was able to prolong 909 supersaturation before disruption of the interaction in aqueous medium, while predissolved PVP formed hydrogen bonds with water, and thus, it was no longer able to form specific interactions with 909 to prolong its supersaturation. In contrast, HPMC-AS effectively mediated 909 supersaturation through hydrophobic interaction, which became pronounced in an aqueous environment and was independent of how HPMC-AS was added. This hypothesis was supported by dynamic light scattering analysis, wherein the formation of nanosized drug/polymer aggregations was found to be correlating with the supersaturation of 909. In summary, we concluded that polymer-mediated drug supersaturation was controlled by drug-polymer interactions persisting in an aqueous environment. Therefore, the physical nature of the drug-polymer interaction as well as the dissolution kinetic of the drug and polymer are all critically important to achieve an optimal ASD formulation design.


Assuntos
Polímeros/química , Varredura Diferencial de Calorimetria , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Espectroscopia de Ressonância Magnética , Solubilidade , Espectroscopia de Infravermelho com Transformada de Fourier , Água
8.
Mol Pharm ; 15(10): 4643-4653, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30130968

RESUMO

Hydroxypropyl methylcellulose acetate succinate (HPMC-AS) is one of the commonly selected polymers used in amorphous solid dispersions (ASD) with excellent capabilities to maintain aqueous supersaturation of poorly water-soluble drugs and inhibit their crystallization, but the underlying mechanisms remain elusive. In this study, posaconazole was chosen as the model drug to study the supersaturation maintaining and crystallization inhibition capabilities of different types of HPMC-AS under pH 5.5-7.5. We analyzed the HPMC-AS aggregation status in solution using combination of static and dynamic light scattering and observed higher polymer aggregation number when higher grade HPMC-AS or lower pH was used, which correlates well with prolonged drug supersaturation or crystallization inhibition. The amount of HPMC-AS coprecipitated with PSZ, a direct indicator of drug/HPMC-AS affinity, also showed positive correlation with the drug supersaturation and crystallization inhibition in the dissolution process. Therefore, we conclude that the aggregation behavior of HPMC-AS and the drug/HPMC-AS affinity are the key mechanisms that lead to posaconazole supersaturation and crystallization inhibition when HPMC-AS was applied.


Assuntos
Metilcelulose/análogos & derivados , Cristalização , Concentração de Íons de Hidrogênio , Luz , Metilcelulose/química
9.
Eur J Pharm Sci ; 122: 303-310, 2018 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-30006181

RESUMO

Hydroxypropyl methylcellulose acetate succinate (HPMC-AS) is one of the most widely used polymers used in amorphous solid dispersions (ASD) for solubility and bioavailability enhancement of poorly water-soluble drugs. Once released from ASDs, HPMC-AS was often found to be highly effective in maintaining drug supersaturation, and this capability is dependent on the concentration and substitution types of this pH-dependent polymer. Therefore, accurate quantification of different grades of HPMC-AS allows us to better understand the release and supersaturation mechanisms of HPMC-AS based ASDs. Since previously reported analytical methods were unable to quantify HPMC-AS in a complex medium with enough sensitivity, we hereby developed a high-sensitivity HPLC-ELSD (evaporative light scattering detector) method with satisfactory specificity, linearity, accuracy and precision, to quantify HPMC-AS down to 20 µg/mL in dissolution media, with the presence of various commonly used pharmaceutical excipients. With the assistance of this method, we compared the intrinsic dissolution rates (IDR) of both the drug and the polymer of posaconazole ASDs based on different types of HPMC-AS. We observed that: 1) For ASDs that were spray dried and uniformly mixed, drug and polymer released simultaneously into the medium with practically identical IDRs slower than the IDR of pure HPMC-AS; 2) For ASDs that were heterogeneously mixed, IDRs of the drug and polymer were significantly slower or faster than the IDRs of the drug and polymer of the uniform ASDs, respectively. In summary, the high sensitivity HPLC-ELSD method established here can be readily applied to quantify HPMC-AS in various dissolution media, thus helps to reveal the release kinetics and mechanisms of different HPMC-AS based ASDs.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Metilcelulose/análogos & derivados , Dessecação , Liberação Controlada de Fármacos , Cinética , Metilcelulose/análise , Metilcelulose/química
10.
Mol Pharm ; 15(7): 2754-2763, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29782805

RESUMO

Amorphous solid dispersion (ASD) is one of the most versatile supersaturating drug delivery systems to improve the dissolution rate and oral bioavailability of poorly water-soluble drugs. PVP based ASD formulation of nimodipine (NMD) has been marketed and effectively used in clinic for nearly 30 years, yet the mechanism by which PVP maintains the supersaturation and subsequently improves the bioavailability of NMD was rarely investigated. In this research, we first studied the molecular interactions between NMD and PVP by solution NMR, using CDCl3 as the solvent, and the drug-polymer Flory-Huggins interaction parameter. No strong specific interaction between PVP and NMD was detected in the nonaqueous state. However, we observed that aqueous supersaturation of NMD could be significantly maintained by PVP, presumably due to the hydrophobic interactions between the hydrophobic moieties of PVP and NMD in aqueous medium. This hypothesis was supported by dynamic light scattering (DLS) and supersaturation experiments in the presence of different surfactants. DLS revealed the formation of NMD/PVP aggregates when NMD was supersaturated, suggesting the formation of hydrophobic interactions between the drug and polymer. The addition of surfactants, sodium lauryl sulfate (SLS) or sodium taurocholate (NaTC), into PVP maintained that NMD supersaturation demonstrated different effects: SLS could only improve NMD supersaturation with concentration above its critical aggregation concentration (CAC) value while not with lower concentration. Nevertheless, NaTC could prolong NMD supersaturation independent of concentration, with lower concentration outperformed higher concentration. We attribute these observations to PVP-surfactant interactions and the formation of PVP/surfactant complexes. In summary, despite the lack of specific interactions in the nonaqueous state, NMD aqueous supersaturation in the presence of PVP was attained by hydrophobic interactions between the hydrophobic moieties of NMD and PVP. This hydrophobic interaction could be disrupted by surfactants, which interact with PVP competitively, thus hindering the capability of PVP to maintain NMD supersaturation. Therefore, caution is needed when evaluating such ASDs in vitro and in vivo when various surfactants are present either in the formulation or in the surrounding medium.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Excipientes/química , Nimodipina/química , Tensoativos/química , Disponibilidade Biológica , Liberação Controlada de Fármacos , Interações Hidrofóbicas e Hidrofílicas , Espectroscopia de Ressonância Magnética , Nimodipina/administração & dosagem , Nimodipina/farmacocinética , Polivinil/química , Pirrolidinas/química , Dodecilsulfato de Sódio/química , Solubilidade , Ácido Taurocólico/química
11.
Eur J Pharm Biopharm ; 124: 73-81, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29305142

RESUMO

The aim of this paper was to compare the in vitro dissolution and in vivo bioavailability of three solubility enhancement technologies for ß-lapachone (LPC), a poorly water soluble compound with extremely high crystallization propensity. LPC cocrystal was prepared by co-grinding LPC with resorcinol. LPC crystalline and amorphous solid dispersions (CSD and ASD) were obtained by spray drying with Poloxamer 188 and HPMC-AS, respectively. The cocrystal structure was solved by single crystal x-ray diffraction. All formulations were characterized by WAXRD, DSC, POM and SEM. USP II and intrinsic dissolution studies were used to compare the in vitro dissolution of these formulations, and a crossover dog pharmacokinetic study was used to compare their in vivo bioavailability. An 1:1 LPC-resorcinol cocrystal with higher solubility and faster dissolution rate was obtained, yet it converted to LPC crystal rapidly in solution. LPC/HPMC-AS ASD was confirmed to be amorphous and uniform, while the crystal and crystallite sizes of LPC in CSD were found to be ∼1-3 µm and around 40 nm, respectively. These formulations performed similarly during USP II dissolution, while demonstrated dramatically different oral bioavailability of ∼32%, ∼5%, and ∼1% in dogs, for CSD, co-crystal, and ASD, respectively. CSD showed the fastest intrinsic dissolution rate among the three. The three formulations showed poor IVIVC which could be due to rapid and unpredictable crystallization kinetics. Considering all the reasons, we conclude that for molecules with extremely high crystallization tendency that cannot be inhibited by any pharmaceutical excipients, size-reduction technologies such as CSD could be advantageous for oral bioavailability enhancement in vivo than technologies only generating transient but not sustained supersaturation.


Assuntos
Naftoquinonas/administração & dosagem , Naftoquinonas/farmacocinética , Administração Oral , Animais , Disponibilidade Biológica , Estudos Cross-Over , Cristalização , Cristalografia por Raios X , Cães , Formas de Dosagem , Composição de Medicamentos , Liberação Controlada de Fármacos , Metilcelulose/análogos & derivados , Metilcelulose/química , Naftoquinonas/química , Tamanho da Partícula , Poloxâmero/química , Resorcinóis/administração & dosagem , Resorcinóis/farmacocinética , Solubilidade , Tecnologia Farmacêutica/métodos
12.
Pharm Res ; 33(10): 2445-58, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27283830

RESUMO

PURPOSE: To identify the key formulation factors controlling the initial drug and polymer dissolution rates from an amorphous solid dispersion (ASD). METHODS: Ketoconazole (KTZ) ASDs using PVP, PVP-VA, HMPC, or HPMC-AS as polymeric matrix were prepared. For each drug-polymer system, two types of formulations with the same composition were prepared: 1. Spray dried dispersion (SDD) that is homogenous at molecular level, 2. Physical blend of SDD (80% drug loading) and pure polymer (SDD-PB) that is homogenous only at powder level. Flory-Huggins interaction parameters (χ) between KTZ and the four polymers were obtained by Flory-Huggins model fitting. Solution (13)C NMR and FT-IR were conducted to investigate the specific drug-polymer interaction in the solution and solid state, respectively. Intrinsic dissolution of both the drug and the polymer from ASDs were studied using a Higuchi style intrinsic dissolution apparatus. PXRD and confocal Raman microscopy were used to confirm the absence of drug crystallinity on the tablet surface before and after dissolution study. RESULTS: In solid state, KTZ is completely miscible with PVP, PVP-VA, or HPMC-AS, demonstrated by the negative χ values of -0.36, -0.46, -1.68, respectively; while is poorly miscible with HPMC shown by a positive χ value of 0.23. According to solution (13)C NMR and FT-IR studies, KTZ interacts with HPMC-AS strongly through H-bonding and dipole induced interaction; with PVPs and PVP-VA moderately through dipole-induced interactions; and with HPMC weakly without detectable attractive interaction. Furthermore, the "apparent" strength of drug-polymer interaction, measured by the extent of peak shift on NMR or FT-IR spectra, increases with the increasing number of interacting drug-polymer pairs. For ASDs with the presence of considerable drug-polymer interactions, such as KTZ/PVPs, KTZ/PVP-VA, or KTZ /HPMC-AS systems, drug released at the same rate as the polymer when intimate drug-polymer mixing was ensured (i.e., the SDD systems); while drug released much slower than the polymer when molecular level mixing or drug-polymer interaction was absent (SDD-PB systems). For ASDs without drug-polymer interaction (i.e., KTZ/HPMC systems), the mixing homogeneity had little impact on the release rate of either the drug or the polymer thus SDD and SDD-PB demonstrated the same drug or polymer release rate, while the drug released slowly and independently of polymer release. CONCLUSIONS: The initial drug release from an ASD was controlled by 1) the polymer release rate; 2) the strength of drug-polymer interaction, including the intrinsic interaction caused by the chemistry of the drug and the polymer (measured by the χ value), as well as that the apparent interaction caused by the drug-polymer ratio (measure by the extent of peak shift on spectroscopic analysis); and 3) the level of mixing homogeneity between the drug and polymer. In summary, the selection of polymer, drug-polymer ratio, and ASD processing conditions have profound impacts on the dissolution behavior of ASDs. Graphical Abstract Relationship between initial drug and polymer dissolution rates from amorphous solid dispersions with different mixing uniformity and drug-polymer interactions.


Assuntos
Liberação Controlada de Fármacos , Preparações Farmacêuticas/metabolismo , Polímeros/metabolismo , Interações Medicamentosas/fisiologia , Liberação Controlada de Fármacos/fisiologia , Preparações Farmacêuticas/química , Polímeros/química , Solubilidade , Difração de Raios X/métodos
13.
Mol Pharm ; 13(8): 2787-95, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27337060

RESUMO

Sodium lauryl sulfate (SLS), as an effective surfactant, is often used as a solubilizer and/or wetting agent in various dosage forms for the purpose of improving the solubility and dissolution of lipophilic, poorly water-soluble drugs. This study aims to understand the impact of SLS on the solution behavior and bioavailability of hypromellose acetate succinate (HPMC-AS)-based posaconazole (PSZ) ASDs, and to identify the underlying mechanisms governing the optimal oral bioavailability of ASDs when surfactants such as SLS are used in combination. Fluorescence spectroscopy and optical microscopy showed that "oil-out" or "liquid-liquid phase separation (LLPS)" occurred in the supersaturated PSZ solution once drug concentration surpassed ∼12 µg/mL, which caused the formation of drug-rich oily droplets with initial size of ∼300-400 nm. Although FT-IR study demonstrated the existence of specific interactions between PSZ and HPMC-AS in the solid state, predissolved HPMC-AS was unable to delay LLPS of the supersaturated PSZ solution and PSZ-rich amorphous precipitates with ∼16-18% HPMC-AS were formed within 10 min. The coprecipitated HPMC-AS was found to be able to significantly delay the crystallization of PSZ in the PSZ-rich amorphous phase from less than 10 min to more than 4 h, yet coexistent SLS was able to negate this crystallization inhibition effect of HPMC-AS in the PSZ-rich amorphous precipitates and cause fast PSZ crystallization within 30 min. 2D-NOESY and the CMC/CAC results demonstrated that SLS could assemble around HPMC-AS and competitively interact with HPMC-AS in the solution, thus prevent HPMC-AS from acting as an effective crystallization inhibitor. In a crossover dog PK study, this finding was found to be correlating well with the in vivo bioavailability of PSZ ASDs formulated with or without SLS. The SLS containing PSZ ASD formulation demonstrated an in vivo bioavailability ∼30% of that without SLS, despite the apparently better in vitro dissolution, which only compared the dissolved drug in solution, a small fraction of the total PSZ dose. We conclude that the bioavailability of ASDs is highly dependent on the molecular interactions between drug, surfactant, and polymer, not only in the solution phase but also in the drug-rich "oily" phase caused by supersaturation.


Assuntos
Derivados da Hipromelose/química , Dodecilsulfato de Sódio/química , Triazóis/química , Liberação Controlada de Fármacos , Espectroscopia de Ressonância Magnética , Soluções Farmacêuticas/química , Solubilidade , Espectrometria de Fluorescência , Espectroscopia de Infravermelho com Transformada de Fourier
14.
Mol Pharm ; 13(2): 599-608, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26709621

RESUMO

Sorafenib is a clinically important oral tyrosine kinase inhibitor for the treatment of various cancers. However, the oral bioavailability of sorafenib tablet (Nexavar) is merely 38-49% relative to the oral solution, due to the low aqueous solubility of sorafenib and its relatively high daily dose. It is desirable to improve the oral bioavailability of sorafenib to expand the therapeutic window, reduce the drug resistance, and enhance patient compliance. In this study, we observed that the solubility of sorafenib could be increased ∼50-fold in the coexistence of poly(vinylpyrrolidone-vinyl acetate) (PVP-VA) and sodium lauryl sulfate (SLS), due to the formation of PVP-VA/SLS complexes at a lower critical aggregation concentration. The enhanced solubility provided a faster initial sorafenib dissolution rate, analogous to a forceful "spring" to release drug into solution, from tablets containing both PVP-VA and SLS. However, SLS appears to impair the ability of PVP-VA to act as an efficient "parachute" to keep the drug in solution and maintain drug supersaturation. Using 2D (1)H NMR, (13)C NMR, and FT-IR analysis, we concluded that the solubility enhancement and supersaturation of sorafenib were achieved by PVP-VA/SLS complexes and PVP-VA/sorafenib interaction, respectively, both through molecular interactions hinged on the PVP-VA VA groups. Therefore, a balance between "spring" and "parachute" must be carefully considered in formulation design. To confirm the in vivo relevance of these molecular interaction mechanisms, we prepared three tablet formulations containing PVP-VA alone, SLS alone, and PVP-VA/SLS in combination. The USP II in vitro dissolution and dog pharmacokinetic in vivo evaluation showed clear differentiation between these three formulations, and also good in vitro-in vivo correlation. The formulation containing PVP-VA alone demonstrated the best bioavailability with 1.85-fold and 1.79-fold increases in Cmax and AUC, respectively, compared with the formulation containing SLS only, the poorest performing one. Despite its forceful "spring", the formulation containing both PVP-VA and SLS showed a moderate bioavailability enhancement, due to the lack of an efficient "parachute".


Assuntos
Antineoplásicos/administração & dosagem , Niacinamida/análogos & derivados , Compostos de Fenilureia/administração & dosagem , Polímeros/química , Comprimidos/química , Administração Oral , Animais , Antineoplásicos/farmacocinética , Disponibilidade Biológica , Química Farmacêutica , Cães , Masculino , Niacinamida/administração & dosagem , Niacinamida/farmacocinética , Compostos de Fenilureia/farmacocinética , Solubilidade , Sorafenibe , Espectroscopia de Infravermelho com Transformada de Fourier , Distribuição Tecidual
15.
Mol Pharm ; 12(2): 576-89, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25535667

RESUMO

The in vitro dissolution mechanism of an amorphous solid dispersion (ASD) remains elusive and highly individualized, yet rational design of ASDs with optimal performance and prediction of their in vitro/in vivo performance are very much desirable in the pharmaceutical industry. To this end, we carried out comprehensive investigation of various ASD systems of griseofulvin, felodipine, and ketoconazole, in PVP-VA or HPMC-AS at different drug loading. Physiochemical properties and processes related to drug-polymer-water interaction, including the drug crystallization tendency in aqueous medium, drug-polymer interaction before and after moisture exposure, supersaturation of drug in the presence of polymer, polymer dissolution kinetics, etc., were characterized and correlated with the dissolution performance of ASDs at different dose and different drug/polymer ratio. It was observed that ketoconazole/HPMC-AS ASD outperformed all other ASDs in various dissolution conditions, which was attributed to the drug's low crystallization tendency, the strong ketoconazole/HPMC-AS interaction and the robustness of this interaction against water disruption, the dissolution rate and the availability of HPMC-AS in solution, and the ability of HPMC-AS in maintaining ketoconazole supersaturation. It was demonstrated that all these properties have implications for the dissolution performance of various ASD systems, and further quantification of them could be used as potential predictors for in vitro dissolution of ASDs. For all ASDs investigated, HPMC-AS systems performed better than, or at least comparably with, their PVP-VA counterparts, regardless of the drug loading or dose. This observation cannot be solely attributed to the ability of HPMC-AS in maintaining drug supersaturation. We also conclude that, for fast crystallizers without strong drug-polymer interaction, the only feasible option to improve dissolution might be to lower the dose and the drug loading in the ASD. In this study, we implemented an ASD/water Flory-Huggins parameter plot, which might assist in revealing the physical nature of the drug-polymer interaction. We also introduced supersaturation parameter and dissolution performance parameter as two quantitative measurements to compare the abilities of polymers in maintaining drug supersaturation, and the dissolution performance of various solid dispersions, respectively.


Assuntos
Polímeros/química , Água/química , Cristalização , Felodipino/química , Griseofulvina/química , Cetoconazol/química , Espectroscopia de Ressonância Magnética , Solubilidade , Espectroscopia de Infravermelho com Transformada de Fourier
16.
Angew Chem Int Ed Engl ; 51(28): 6984-8, 2012 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-22674838

RESUMO

Let's swap: a scalable, atom-economic, enantio-, and diastereoselective synthetic route to trisubstituted γ-butyrolactones based on a Wagner-Meerwein-type dyotropic rearrangement of cis-ß-lactones is described. This methodology was applied in efficient and protecting-group-free formal syntheses and total syntheses of various xanthanolide natural products.


Assuntos
4-Butirolactona/química , Produtos Biológicos/síntese química , Lactonas/química , Sesquiterpenos/síntese química , Xanthium/química , Estrutura Molecular , Estereoisomerismo
17.
Coron Artery Dis ; 21(6): 369-77, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20613497

RESUMO

OBJECTIVE: Cardiac fibroblasts (CFs) regulate myocardial fibrosis and remodeling through proliferation and differentiation. Transforming growth factor-beta1 (TGF-beta1) plays a critical role in the development of myocardial fibrosis after myocardial infarction (MI). The aim of this study was to investigate the effects of inhibiting TGF-beta1 action on myofibroblast differentiation and cardiac function after MI. METHODS: CFs were cultured and treated, respectively with PBS, TGF-beta1, soluble TGF-beta1 receptor II (sTbetaRII), and TGF-beta1 plus sTbetaRII. Proliferation CFs were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Myofibroblast differentiation was examined by alpha-smooth muscle actin immunostaining. Expression of P-Smad2 and Smad2/3 was determined by immunostaining and western blot analysis. Four days after ligation of left anterior descending coronary artery, sTbetaRII was injected into injured heart. Two weeks after sTbetaRII administration, myofibroblast differentiation was measured with alpha-smooth muscle actin immunostaining. Four weeks after sTbetaRII administration, cardiac function was evaluated by hemodynamic measurements. Weight parameters, infarct size, and collagen fiber were detected with an earlier experimental method. RESULTS: Compared with TGF-beta1, TGF-beta1 plus sTbetaRII significantly decreased cell proliferation, myofibroblast differentiation, and expression of P-Smad2 in CFs (P<0.05). Two weeks after sTbetaRII administration, myofibroblast differentiation in MI rats treated with sTbetaRII was reduced compared with MI group (P<0.05). Four weeks after sTbetaRII administration, MI rats that received sTbetaRII showed significantly higher cardiac function and lower in weight parameters, infarct size, and collagen fiber than that of MI group (P<0.05). CONCLUSION: sTbetaRII could inhibit TGF-beta1-induced myofibroblast differentiation, alleviate myocardial fibrosis and remodeling, and improve ischemic cardiac function after MI.


Assuntos
Fármacos Cardiovasculares/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Miofibroblastos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/administração & dosagem , Receptores de Fatores de Crescimento Transformadores beta/administração & dosagem , Fator de Crescimento Transformador beta1/metabolismo , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Fibrose , Imuno-Histoquímica , Microinjeções , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo II , Recuperação de Função Fisiológica , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fatores de Tempo , Função Ventricular Esquerda , Pressão Ventricular , Remodelação Ventricular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...