Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
J Pharm Sci ; 112(8): 2069-2078, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36638959

ABSTRACT

These proceedings contain presentation summaries and discussion highlights from the University of Maryland Center of Excellence in Regulatory Science and Innovation (M-CERSI) Workshop on Co-processed API, held on July 13 and 14, 2022. This workshop examined recent advances in the use of co-processed active pharmaceutical ingredients as a technology to improve drug substance physicochemical properties and drug product manufacturing process robustness, and explored proposals for enabling commercialization of these transformative technologies. Regulatory considerations were discussed with a focus on the classification, CMC strategies, and CMC documentation supporting the use of this class of materials from clinical studies through commercialization. The workshop format was split between presentations from industry, academia and the FDA, followed by breakout sessions structured to facilitate discussion. Given co-processed API is a relatively new concept, the authors felt it prudent to compile these proceedings to gain further visibility to topics discussed and perspectives raised during the workshop, particularly during breakout discussions. Disclaimer: This paper reflects discussions that occurred among stakeholder groups, including FDA, on various topics. The topics covered in the paper, including recommendations, therefore, are intended to capture key discussion points. The paper should not be interpreted to reflect alignment on the different topics by the participants, and the recommendations provided should not be used in lieu of FDA published guidance or direct conversations with the Agency about a specific development program. This paper should not be construed to represent FDA's views or policies.

3.
J Pharm Sci ; 112(8): 2046-2056, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36462708

ABSTRACT

Continuous direct compression (CDC) of solid oral dosage forms requires materials exhibiting acceptable flow and compression properties. The desired active pharmaceutical ingredient (API) powder properties can be difficult to achieve through conventional particle engineering approaches, such as particle size and habit modification during crystallization. Co-processing of API with excipients can significantly improve the powder properties to overcome these difficulties. In this manuscript, performance of a co-processed API was evaluated in a continuous feeding and blending process using GEA ConsiGma® Continuous Dosing and Blending Unit (CDB1). The co-processed theophylline was generated via a methodology in which polymer was precipitated and coated the crystalline theophylline particles resulting in nearly spherical agglomerates. A range of drug loads (1-25% w/w), flow rates (15-40 kg/h) and blender speeds (220-400 rpm) were studied. The results demonstrated that the co-processed API can be successfully fed through a loss-in-weight feeder and blended with other excipients in a high shear blender to generate tablets with acceptable content uniformity at 1-25% w/w drug loads. This study supports that using co-processed API with enhanced powder properties is a promising approach to enable continuous manufacturing for APIs with challenging properties.


Subject(s)
Chemistry, Pharmaceutical , Pharmacy , Chemistry, Pharmaceutical/methods , Excipients/chemistry , Powders/chemistry , Theophylline , Tablets/chemistry , Particle Size , Drug Compounding/methods , Technology, Pharmaceutical/methods
4.
Mol Pharm ; 17(7): 2232-2244, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32392068

ABSTRACT

Optimized physical properties (e.g., bulk, surface/interfacial, and mechanical properties) of active pharmaceutical ingredients (APIs) are key to the successful integration of drug substance and drug product manufacturing, robust drug product manufacturing operations, and ultimately to attaining consistent drug product critical quality attributes. However, an appreciable number of APIs have physical properties that cannot be managed via routes such as form selection, adjustments to the crystallization process parameters, or milling. Approaches to control physical properties in innovative ways offer the possibility of providing additional and unique opportunities to control API physical properties for both batch and continuous drug product manufacturing, ultimately resulting in simplified and more robust pharmaceutical manufacturing processes. Specifically, diverse opportunities to significantly enhance API physical properties are created if allowances are made for generating co-processed APIs by introducing nonactive components (e.g., excipients, additives, carriers) during drug substance manufacturing. The addition of a nonactive coformer during drug substance manufacturing is currently an accepted approach for cocrystals, and it would be beneficial if a similar allowance could be made for other nonactive components with the ability to modify the physical properties of the API. In many cases, co-processed APIs could enable continuous direct compression for small molecules, and longer term, this approach could be leveraged to simplify continuous end-to-end drug substance to drug product manufacturing processes for both small and large molecules. As with any novel technology, the regulatory expectations for co-processed APIs are not yet clearly defined, and this creates challenges for commercial implementation of these technologies by the pharmaceutical industry. The intent of this paper is to highlight the opportunities and growing interest in realizing the benefits of co-processed APIs, exemplified by a body of academic research and industrial examples. This work will highlight reasons why co-processed APIs would best be considered as drug substances from a regulatory perspective and emphasize the areas where regulatory strategies need to be established to allow for commercialization of innovative approaches in this area.


Subject(s)
Drug Compounding/methods , Drug Industry/methods , Pharmaceutical Preparations/chemistry , Chemical Precipitation , Chemistry, Pharmaceutical/methods , Crystallization , Drug Carriers/chemistry , Excipients/chemistry , Flavoring Agents/chemistry , Particle Size , Quality Control
5.
Drug Dev Ind Pharm ; 44(10): 1606-1612, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29916275

ABSTRACT

A novel process for generating agglomerates of active pharmaceutical ingredient (API) and polymer by swelling the polymer in a water/organic mixture has been developed to address formulation issues resulting from a water sensitive, high drug load API with poor powder properties. Initially, the API is dissolved in water, following which hydroxypropyl methylcellulose (HPMC) is added, resulting in the imbibing of water, along with the dissolved API, into the HPMC matrix. The addition of acetone and isopropyl acetate (anti-solvents) then causes the API to crystallize inside and on the surface of HPMC agglomerates. The process was scaled up to 20 kg scale. The agglomerates of API and HPMC generated by this process are ∼350 µm diameter, robust, and have significantly better flow than the API as measured by Erweka flow testing. These agglomerates exhibit improved bulk density, acceptable chemical stability, and high compressibility. The agglomerates process well through roller compaction and tableting, with no flow or sticking issues. This process is potentially adaptable to other APIs with similar attributes.


Subject(s)
Chemistry, Pharmaceutical/methods , Hypromellose Derivatives/chemical synthesis , Hypromellose Derivatives/pharmacokinetics , Crystallization , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/pharmacokinetics , Drug Compounding , Drug Liberation
6.
Annu Rev Chem Biomol Eng ; 2: 259-80, 2011.
Article in English | MEDLINE | ID: mdl-22432619

ABSTRACT

Polymorphism in molecular crystals is a prevalent phenomenon and is of great interest to the pharmaceutical community. The solid-state form is a key quality attribute of a crystalline product. Inconsistencies in the solid phase produced during the manufacturing and storage of drug substances and drug products may have severe consequences. It is essential to understand the solid-state behavior of the drug and to judiciously select the optimal solid form for development. This review highlights the pervasiveness and relevance of polymorphism and describes solid form screening and selection processes. Moreover, case studies on controlling polymorphs from a chemical development perspective are provided.


Subject(s)
Crystallization , Molecular Structure , Pharmaceutical Preparations/chemistry , Chemistry, Pharmaceutical/methods , Drug Design , Technology, Pharmaceutical/methods
7.
Acc Chem Res ; 42(5): 621-9, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19402623

ABSTRACT

Crystallization is vital to many processes occurring in nature and in the chemical, pharmaceutical, and food industries. Notably, crystallization is an attractive isolation step for manufacturing because this single process combines both particle formation and purification. Almost all of the products based on fine chemicals, such as dyes, explosives, and photographic materials, require crystallization in their manufacture, and more than 90% of all pharmaceutical products contain bioactive drug substances and excipients in the crystalline solid state. Hence control over the crystallization process allows manufacturers to obtain products with desired and reproducible properties. We judge the quality of a crystalline product based on four main properties: size, purity, morphology, and crystal structure. The pharmaceutical industry in particular requires production of the desired crystal form (polymorph) to assure the bioavailability and stability of the drug substance. In solution crystallization, nucleation plays a decisive role in determining the crystal structure and size distribution. Therefore, understanding the fundamentals of nucleation is crucial to achieve control over these properties. Because of its analytical simplicity, researchers have widely applied classical nucleation theory to solution crystallization. However, a number of differences between theoretical predictions and experimental results suggest that nucleation of solids from solution does not proceed via the classical pathway but follows more complex routes. In this Account, we discuss the shortcomings of classical nucleation theory and review studies contributing to the development of the modern two-step model. In the two-step model that was initially proposed for protein crystallization, a sufficient-sized cluster of solute molecules forms first, followed by reorganization of that cluster into an ordered structure. In recent experimental and theoretical studies, we and other researchers have demonstrated the applicability of the two-step mechanism to both macromolecules and small organic molecules, suggesting that this mechanism may underlie most crystallization processes from solutions. Because we have observed an increase in the organization time of appropriate lattice structures with greater molecular complexity, we propose that organization is the rate-determining step. Further development of a clearer picture of nucleation may help determine the optimum conditions necessary for the effective organization within the clusters. In addition, greater understanding of these processes may lead to the design of auxiliaries that can increase the rate of nucleation and avoid the formation of undesired solid forms, allowing researchers to obtain the final product in a timely and reproducible manner.


Subject(s)
Liquid Crystals/chemistry , Models, Chemical , Solutions , Thermodynamics
8.
Curr Opin Drug Discov Devel ; 10(6): 746-55, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17987526

ABSTRACT

Solution crystallization is an important separation and purification process used in the chemical, pharmaceutical and food industries. The quality of a crystalline product is generally judged by four main criteria: purity, crystal habit, particle size and solid form. Consistent production of the desired polymorph is crucial as the unanticipated emergence of a different crystal form may have severe consequences. Thus, the selection of a solid-state form for a crystalline product is vital and is ultimately based on knowledge of the properties of the other polymorphs. This review discusses the role of nucleation, crystal growth and molecular modeling on polymorphism in molecular crystals. Examples are presented demonstrating how the first two factors can govern the appearance of a particular crystalline form, and how the latter factor can be used as a tool for understanding polymorphism.


Subject(s)
Models, Molecular , Pharmaceutical Preparations/chemistry , Technology, Pharmaceutical/methods , Chemical Engineering/methods , Crystallization/methods , Drug Design , Molecular Conformation , Particle Size
9.
Phys Rev Lett ; 99(11): 115702, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17930450

ABSTRACT

Small angle x-ray scattering is utilized to directly examine the formation of clusters in supersaturated solutions of glycine, in an attempt to understand their role in the nucleation process. The results suggest that the majority of glycine molecules exist as dimers in aqueous solutions, and monomers in 13% (upsilon/upsilon) acetic acid-water mixtures. As the water and acetic acid-water solutions crystallize into alpha and gamma forms, respectively, the findings indicate a direct correlation between molecular self-association in solution and the polymorphic outcome.


Subject(s)
Glycine/chemistry , Acetic Acid , Biophysical Phenomena , Biophysics , Crystallization , Dimerization , Scattering, Small Angle , Solutions , Water , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...