Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Pharm Sci ; 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39098520

RESUMO

Protein aggregation is challenging for biopharmaceutical drug, because it affects the stability of protein formulations in real-time. However, current techniques for protein aggregate indication meet a number of limitations including limited aggregate size range, complex pre-treatments and lack of chromatographic approaches. Herein, a rapid, automatic, non-invasive and wide-scale coverage technique for aggregates indication is developed to overcome these challenges. Firstly, the response of low-field nuclear magnetic resonance (LF-NMR) to the aggregates is explored by making a comparison with certain established techniques. LF-NMR achieves a high sensitivity of water proton transverse relaxation rate (R2 of H2O, hereinafter referred as R2(H2O)) to protein aggregates from nanometer to micrometer. Then, the quantitative relationship between R2(H2O) and aggregates is investigated furtherly. R2(H2O) could serve as an all-size coverage protein aggregates indicator during development. As a non-invasive method, LF-NMR does not need any sample handling. It takes only 44 s for one test, and saves a lot of manpower, materials and costs. Compared with other established analytical techniques, the technique developed here could be a powerful tool for a rapid, automatic, non-invasive and wide-scale coverage technique for aggregates indication in biomacromolecule development.

2.
Artigo em Inglês | MEDLINE | ID: mdl-38942483

RESUMO

Vial and syringe filling by peristaltic pump has been widely implemented by contract manufacturing organizations and biopharmaceutical companies. Fill volume is commonly considered as critical quality attribute related in aseptic filling process and the variation needs to be well controlled to guarantee the safety, efficacy and consistency of drug products. However, the criteria for justifying the filling variation and underlying mechanisms that affect the variability are not fully revealed quantitatively in the literatures. This study selected filling accuracy, filling process capability and filling precision as three criteria for evaluating the filling process performance with four statistical indexes: Relative Error Mean, Critical Control Limit (Cpk ≥ 1.33), Relative Standard Deviation and Relative Moving Range Mean. The impact of liquid properties, pump tubing sizes and pump settings on above indexes were investigated using a bench-top system with a peristatic pump and a high-precision balance. The results showed that the viscosity, target fill volume, pump tubing size, pump speed, acceleration/deceleration rate and suck-back had statistical significance on the fill volume variability. Definitive Screening Design was further applied to clarify and visualize the priorities and interaction impact of above factors on fill volume variability. Stepwise approach for fill volume variability optimization and control based on predictive models was established and verified for drug product solution with viscosity between 1-23 cp and target fill volume between 0.2-2.0 mL.

3.
Antib Ther ; 7(1): 67-76, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38371955

RESUMO

Background: Lyophilized drug products with high protein concentration often perform long reconstitution time, which is inconvenient for clinical use. The objective of this work is to achieve short reconstitution time with multiple and combined strategies. Methods: Here, we describe the following approaches that lead to reduction of reconstitution time, including adding annealing step, decreasing headspace pressure, decreasing protein concentration with reducing diluent volume, increasing high surface-area-to-height ratio of the cakes, increasing frequency of swirling and diluent temperature. Results: Among these strategies, reducing diluent volume to achieve high protein concentration and reducing headspace pressure show markedly reduction of reconstitution time. Moreover, we propose combined strategies to mitigate the reconstitution time, at the same time, to achieve same target dose in clinics. Conclusions: Therefore, this paper provides insights on the application of multiple strategies to accelerate the reconstitution of lyophilized drug products with high concentration, and facilitates their widespread clinical application.

4.
Eur J Pharm Biopharm ; 197: 114221, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38378097

RESUMO

The development of PFS requires a detailed understanding of the forces occurring during the drug administration process and patient's capability. This research describes an advanced mathematic injection force model that consisting hydrodynamic force and friction force. The hydrodynamic force follows the basic law of Hagen-Poiseuille but refines the modeling approach by delving into specific properties of drug viscosity (Newtonian and Shear-thinning) and syringe shape constant, while the friction force was accounted from empty barrel injection force. Additionally, we take actual temperature of injection into consideration, providing more accurate predication. The results show that the derivation of the needle dimension constant and the rheological behavior of the protein solutions are critical parameters. Also, the counter pressure generated by the tissue has been considered in actual administration to address the issue of the inaccuracies of current injection force evaluation preformed in air, especially when the viscosity of the injected drug solution is below 9.0 cP (injecting with 1 mL L PFS staked with 29G ½ inch needle). Human factor studies on patients' capability against medication viscosity filled the gap in design space of PFS drug product and available viscosity data in very early phase.


Assuntos
Fenômenos Mecânicos , Seringas , Humanos , Viscosidade , Injeções , Preparações Farmacêuticas
5.
J Pharm Sci ; 113(6): 1478-1487, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38246363

RESUMO

Vaccine manufacturing is one of the most challenging and complex processes in pharmaceutical industry, and the process control strategy is critical for the safety, effectiveness, and consistency of a vaccine. The efficacy of aluminum salt adjuvant on vaccines strongly depends on its physicochemical properties, such as size, structure, surface charge, etc. However, stresses during the vaccine manufacturing may affect the stability of adjuvant. In this study, the impacts of cold/thermal stress, autoclaving, pumping, mixing, and filling shear stress on the physicochemical properties of aluminum hydroxide (AH) adjuvant were evaluated as part of the manufacturing process development. The results showed that the autoclaving process would slightly influence the structure and properties of the investigated AH adjuvant, but thermal incubation at 2-8 °C, 25 °C and 40 °C for 4 weeks did not. However, -20 °C freezing AH adjuvant led to the adjuvant agglomeration and rapid sedimentation. For the high shear stress study with mixing at 500 rpm in a 1-L mixing bag and pumping at 220 rpm for up to 24 h, the average particle dimension of the bulk AH adjuvant decreased, along with decreasing protein adsorption ratio. The studies indicate that various stresses during manufacturing process could affect the structure and physicochemical properties of AH adjuvant, which calls for more attention on the control of adjuvant process parameters during manufacturing.


Assuntos
Adjuvantes Imunológicos , Hidróxido de Alumínio , Vacinas , Hidróxido de Alumínio/química , Vacinas/química , Adjuvantes Imunológicos/química , Tamanho da Partícula , Estabilidade de Medicamentos
6.
J Pharm Sci ; 113(4): 866-879, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38160713

RESUMO

With the remarkably strong growth of the biopharmaceutical market, an increasing demand for self-administration and rising competitions attract substantial interest to the biologic-device combination products. The ease-of-use of biologic-device combination products can minimize dosing error, improve patient compliance and add value to the life-cycle management of biological products. As listed in the purple book issued by the U.S. Food and Drug Administration (FDA), a total of 98 brand biologic-device combination products have been approved with Biologic License Application from January 2000 to August 2023, where this review mainly focused on 63 products containing neither insulin nor vaccine. Prefilled syringes (PFS) and autoinjectors are the most widely adopted devices, whereas innovative modifications like needle safety guard and dual-chamber design and novel devices like on-body injector also emerged as promising presentations. All 16 insulin products employ pen injectors, while all 19 vaccine products are delivered by a PFS. This review provides a systematic summary of FDA-approved biologic-device combination products regarding their device configurations, routes of administration, formulations, instructions for use, etc. In addition, challenges and opportunities associated with biologic-device compatibility, regulatory complexity, and smart connected devices are also discussed. It is believed that evolving technologies will definitely move the boundaries of biologic-device combination product development even further.


Assuntos
Produtos Biológicos , Vacinas , Estados Unidos , Humanos , United States Food and Drug Administration , Autoadministração , Insulina , Seringas
7.
Mol Pharm ; 20(5): 2536-2544, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37036270

RESUMO

The interaction of protein drugs with the air-liquid interface plays a crucial role in the overall stability in aqueous formulations, particularly when the adsorbed proteins are subjected to the surface flow. Nonionic surfactants are usually added into the formulation solutions to address this issue. A diversity of studies have been focused on the usage of surfactants, the stability mechanism of surfactants, or seeking new pharmaceutical surfactants. However, the real protagonist, the basic properties of protein drugs, was neglected, which may play a vital role in the stability of protein drugs. Herein, we aim to clarify the correlation between the surface behavior of proteins and the interfacial stability. A force tensiometer is used to track the surface tension reduction and the competition between surfactants and proteins at the surface. We find that the surface behaviors of proteins vary with storage temperature and protein types including monoclonal antibodies (mAb), bispecific monoclonal antibodies (BsAb), and antibody-drug conjugates (ADCs). Especially for the protein stored at 5 °C, the surface activity of proteins is better than that of surfactants. It indicates that the ability of proteins to adsorb at the interface should not be ignored compared to surfactants. The significant difference in the interfacial stability of protein pharmaceuticals formulated in the same buffer and excipients as well as the surfactants with the same concentration further confirms the interfacial adsorption capacity of proteins that should not be ignored. These findings provide a new angle and valuable insights into the correlation between the surface activity of the proteins and interfacial stability, which may pave the way for future preformulation studies on therapeutic proteins and broaden the thoughts of formulation development.


Assuntos
Proteínas de Membrana , Tensoativos , Tensão Superficial , Excipientes , Adsorção
8.
J Pharm Sci ; 97(4): 1427-42, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17724660

RESUMO

A rapid solubility-screening assay was developed with a focus on Biopharmaceutic Classification Scheme (BCS) class II drug solubility in animal and simulated human gastrointestinal (GI) fluids. The assay enables biologically promising drug leads to be evaluated for solubility limitations earlier in the drug development process, minimizes GI fluid needs, and produces in vitro solubility information with potential in vivo implications. A number of BCS II drugs were dissolved in DMSO at approximately 40 mM, and robotically distributed to a 96-well plate. The DMSO was evaporated and drugs were equilibrated with selected GI fluids, both fed and fasted states. After equilibration, precipitated wells were subjected to HPLC analysis. A spreadsheet calculated solubility automatically from HPLC output. Intra-day, inter-day, and inter-plate reproducibility were within 15% RSTD for the tested drugs with the primary source of variability being injection precision of our injector system. The reported solubility from screening assays was well correlated with literature data (r(2) = 0.80) with a slope of 0.86 and (r(2) = 0.99) with a slope of 0.89. This screening assay converts conventional solubility measurements to a 96-well format for increased throughput (>12 samples/h), reduces fluid needs, and minimizes drug consumption.


Assuntos
Biofarmácia/métodos , Preparações Farmacêuticas/classificação , Solubilidade , Animais , Líquidos Corporais/metabolismo , Filtração , Trato Gastrointestinal/metabolismo , Humanos , Reprodutibilidade dos Testes
9.
J Pharm Sci ; 97(6): 2080-90, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17879292

RESUMO

A rapid-throughput screening assay was developed to estimate the salt solubility parameter, K(SP), with a minimal quantity of drug. This assay allows for early evaluation of salt limited solubility with a large number of counter-ions and biologically promising drug leads. Drugs dissolved (typically 10 mM) in DMSO are robotically distributed to a 96-well plate. DMSO is evaporated, and drugs are equilibrated with various acids at different concentrations (typically <1 M) to yield final total drug concentrations around 2.5 mM. The plate is checked for precipitation. Filtrates from only those precipitated wells were subjected to rapid gradient HPLC analysis. An iterative procedure is employed to calculate all species concentrations based on mass and charge balance equations. The apparent K(SP) values assuming 1:1 stoichiometry are determined from counter-ion and ionized drug activities. A correlation coefficient >0.975 for eight drugs totaling 16 salts is reported. Intra-day and inter-day reproducibility was <10%. Conventional apparent K(SP) measurements were translated to 96-well format for increased throughput and minimal drug consumption (typically 10 mg) to evaluate at least eight different counter-ions. Although the current protocol estimates K(SP) from 10(-3) to 10(-7) M, the dynamic range of the assay could be expanded by adjusting drug and counter-ion concentrations.


Assuntos
Microquímica , Preparações Farmacêuticas/química , Tecnologia Farmacêutica/métodos , Precipitação Química , Cromatografia Líquida de Alta Pressão , Dimetil Sulfóxido/química , Concentração de Íons de Hidrogênio , Modelos Químicos , Nefelometria e Turbidimetria , Reprodutibilidade dos Testes , Robótica , Solubilidade , Solventes/química
10.
PDA J Pharm Sci Technol ; 61(6): 441-51, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18410045

RESUMO

The process of freeze-thaw not only subjects bioproducts to potentially destabilizing stress, but also imposes challenges to retain container integrity. Shipment and storage of frozen products in glass vials and thawing of the vials prior to use at clinics is a common situation. Vial integrity failure during freeze-thaw results in product loss and safety issues. Formulations of biomolecules often include crystallizable excipients, which can cause glass vial breakage during freeze-thaw operations. In this study, mannitol formulations served as models for mechanistic investigation of root causes for vial breakage. Several parameters and their impacts on vial breakage were investigated, including mannitol concentration (5% and 15%), different freeze-thaw conditions (fast, slow, and staging), fill configurations (varying fill volume/vial size ratio), and vial tray materials (plastic, stainless steel, corrugated cardboard, aluminum, and polyurethane foam). The results in this study were subjected to a statistical proportion test. The data showed that large fill volumes strongly correlated with higher percentage of vial cracks. Furthermore, the 15% mannitol was found to cause more breakage than 5% mannitol, especially with fast temperature gradient. Significantly more thawing vial breakage occurred in the fast compared to slow freeze-thaw with all types of vial trays. The freezing breakage was substantially lower than the thawing breakage using the fast temperature gradient, and the trend was reversed with the slow temperature gradient. An intermediate hold at -30 degrees C prior to further decrease in temperature proved to be a practical approach to minimize mannitol-induced vial breakage. Thermal mechanical analysis (TMA) and strain gage techniques were employed to gain mechanistic insights, and it was found that the primary causes for mannitol-induced vial breakage were partial crystallization during freezing and "secondary" crystallization of non-crystallized fraction during thawing. The strain on the vial's axial direction was significantly higher than the hoop direction, typically resulting in bottom lens of the vial coming off. Without a -30 degrees C hold, rapid volume expansions due to initial crystallization and secondary crystallization of mannitol were observed in TMA profiles, and these expansions were more apparent in 15% mannitol compared to 5% mannitol. With the introduction of a -30 degrees C hold step, abrupt expansions diminished in TMA profiles, suggesting that most of the mannitol crystallization occurred concurrently with ice solidification during the -30 degrees C holding step and, thus, secondary crystallization during thawing was minimal and the sudden expansion event was eliminated. Therefore, vial breakage during both freezing and thawing was reduced.


Assuntos
Embalagem de Medicamentos , Manitol/química , Química Farmacêutica , Cristalização , Armazenamento de Medicamentos , Excipientes/química , Congelamento , Vidro , Termodinâmica
11.
PDA J Pharm Sci Technol ; 61(6): 452-60, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18410046

RESUMO

In an accompanying article we have described parameters that influence vial breakage in freeze-thaw operations when using crystalizable mannitol formulations, and further provided a practical approach to minimize the breakage in manufacturing settings. Using two diagnostic tools-thermal mechanical analysis (TMA) and strain gage, we investigated the mechanism of mannitol vial breakage and concluded that the breakage is related to sudden volume expansions in the frozen plug due to crystallization events. Glass vial breakage has also been observed with a number of frozen protein formulations consisting of only amorphous ingredients. Therefore, in this study, we applied the methodologies and learnings from the prior investigation to further explore the mechanism of vial breakage during freeze-thaw of amorphous protein products. It was found that temperature is a critical factor, as breakage typically occurred when the products were frozen to -70 degrees C, while freezing only to -30 degrees C resulted in negligible breakage. When freezing to -70 degrees C, increased protein concentration and higher fill volume induced more vial breakage, and the breakage occurred mostly during freezing. In contrast to the previous findings for crystallizable formulations, an intermediate staging step at -30 degrees C did not reduce breakage for amorphous protein formulations, and even slightly increased the breakage rate. The TMA profiles revealed substantially higher thermal contraction of frozen protein formulations when freezing below -30 degrees C, as compared to glass. Such thermal contraction of frozen protein formulations caused inward deformation of glass and subsequent rapid movement of glass when the frozen plug separates from the vial. Increasing protein concentration caused more significant inward glass deformation, and therefore a higher level of potential energy was released during the separation between the glass and frozen formulation, causing higher breakage rates. The thermal expansion during thawing generated moderate positive strain on glass and explained the thaw breakage occasionally observed. The mechanism of vial breakage during freeze-thaw of amorphous protein formulations is different compared to crystallizable formulations, and accordingly requires different approaches to reduce vial breakage in manufacturing. Storing and shipping at no lower than -30 degrees C effectively prevents breakage of amorphous protein solutions. If lower temperature such as -70 degrees C is unavoidable, the risk of breakage can be reduced by lowering fill volume.


Assuntos
Embalagem de Medicamentos , Proteínas/química , Química Farmacêutica , Cristalização , Armazenamento de Medicamentos , Excipientes/química , Congelamento , Vidro , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA