Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Biotechnol J ; 14(2): e1700721, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30024096

RESUMO

Due to the growing interest in integrated continuous processing in the biopharmaceutical industry, productivity comparison of batch-based and continuous processes is considered a challenge. Integrated continuous manufacturing of biopharmaceuticals requires scientists and engineers to collaborate effectively. Differing definitions, for example, of volumetric productivity, may cause confusion in this interdisciplinary field. Therefore, the aim of this communication is to reiterate the standard definitions and their underlying assumptions. Applying them to an exemplary model scenario allows to demonstrate the differences and to develop recommendations for the comparison of productivity of different upstream processes.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Técnicas de Cultura Celular por Lotes/normas , Biofarmácia/normas , Reatores Biológicos/normas , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/química , Técnicas de Cultura Celular por Lotes/economia , Biofarmácia/economia , Reatores Biológicos/economia , Células CHO , Contagem de Células , Cricetulus , Meios de Cultura , Modelos Teóricos
2.
BioDrugs ; 32(5): 405-414, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30143982

RESUMO

Biologics are produced from living organisms in complex, multi-stage manufacturing processes and contain inherent variability, which must be understood and controlled during manufacturing to avoid unexpected changes in key quality attributes that may contribute to clinically meaningful differences. The process must also meet large commercial demand, while simultaneously being able to accommodate change without sacrificing product consistency. The four key components of successful biologics manufacturing are (1) a stable, well-defined proprietary cell line; (2) a good manufacturing practice (GMP)-compliant supply chain with a process control strategy defining acceptable levels of variability for target product/process attributes and capable of managing complex material flows; (3) a tightly controlled procedure for implementation of proposed process changes that ensures product consistency; and (4) built-in redundancy and flexibility providing the ability to adapt rapidly to unexpected developments. This report describes the requirements for the manufacturing and distribution of biologics, using Remicade® (infliximab, Janssen Biotech, Horsham, PA, USA) as an example of best practices. Since Remicade's first marketing approval in 1998, Janssen has manufactured > 150 million vials used to treat > 2.6 million patients around the world for a variety of inflammatory diseases. Remicade displays a highly consistent quality attribute profile and meets all product/process specifications across multiple manufacturing sites and process scales. Janssen's experience with Remicade demonstrates that deep product knowledge, extensive manufacturing experience, diligent product/process monitoring and a sustained commitment to compliance and research are required to ensure quality, consistency and uninterrupted patient supply for large-volume biologics over the long term.


Assuntos
Produtos Biológicos/provisão & distribuição , Indústria Farmacêutica/normas , Infliximab , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Técnicas de Cultura Celular por Lotes/métodos , Técnicas de Cultura Celular por Lotes/normas , Produtos Biológicos/normas , Linhagem Celular , Indústria Farmacêutica/métodos , Rotulagem de Medicamentos/normas , Armazenamento de Medicamentos , Liofilização , Controle de Qualidade
3.
PDA J Pharm Sci Technol ; 71(3): 189-205, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27974629

RESUMO

In order to operate profitably under different product demand scenarios, biopharmaceutical companies must design their facilities with mass output flexibility in mind. Traditional biologics manufacturing technologies pose operational challenges in this regard due to their high costs and slow equipment turnaround times, restricting the types of products and mass quantities that can be processed. Modern plant design, however, has facilitated the development of lean and efficient bioprocessing facilities through footprint reduction and adoption of disposable and continuous manufacturing technologies. These development efforts have proven to be crucial in seeking to drastically reduce the high costs typically associated with the manufacturing of recombinant proteins. In this work, mathematical modeling is used to optimize annual production schedules for a single-product commercial facility operating with a continuous upstream and discrete batch downstream platform. Utilizing cell culture duration and volumetric productivity as process variables in the model, and annual plant throughput as the optimization objective, 3-D surface plots are created to understand the effect of process and facility design on expected mass output. The model shows that once a plant has been fully debottlenecked it is capable of processing well over a metric ton of product per year. Moreover, the analysis helped to uncover a major limiting constraint on plant performance, the stability of the neutralized viral inactivated pool, which may indicate that this should be a focus of attention during future process development efforts.LAY ABSTRACT: Biopharmaceutical process modeling can be used to design and optimize manufacturing facilities and help companies achieve a predetermined set of goals. One way to perform optimization is by making the most efficient use of process equipment in order to minimize the expenditure of capital, labor and plant resources. To that end, this paper introduces a novel mathematical algorithm used to determine the most optimal equipment scheduling configuration that maximizes the mass output for a facility producing a single product. The paper also illustrates how different scheduling arrangements can have a profound impact on the availability of plant resources, and identifies limiting constraints on the plant design. In addition, simulation data is presented using visualization techniques that aid in the interpretation of the scientific concepts discussed.


Assuntos
Produtos Biológicos/química , Biofarmácia/normas , Indústria Farmacêutica/normas , Arquitetura de Instituições de Saúde , Preparações Farmacêuticas/química , Controle de Qualidade , Tecnologia Farmacêutica/normas , Fluxo de Trabalho , Técnicas de Cultura Celular por Lotes/normas , Biofarmácia/instrumentação , Biofarmácia/métodos , Reatores Biológicos/normas , Cromatografia/normas , Composição de Medicamentos , Indústria Farmacêutica/instrumentação , Indústria Farmacêutica/métodos , Filtração/normas , Modelos Teóricos , Tecnologia Farmacêutica/instrumentação , Tecnologia Farmacêutica/métodos , Fatores de Tempo
4.
Adv Biochem Eng Biotechnol ; 139: 93-121, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23828505

RESUMO

The principles of quality by design (QbD) have been applied in cell culture manufacturing process development and characterization in the biotech industry. Here we share our approach and practice in developing and characterizing a cell culture manufacturing process using QbD principles for establishing a process control strategy. Process development and characterization start with critical quality attribute identification, followed by process parameter and incoming raw material risk assessment, design of experiment, and process parameter classification, and conclude with a design space construction. Finally, a rational process control strategy is established and documented.


Assuntos
Algoritmos , Técnicas de Cultura Celular por Lotes/normas , Separação Celular/normas , Indústria Farmacêutica/normas , Desenho de Equipamento/normas , Análise de Falha de Equipamento/normas , Controle de Qualidade , Técnicas de Cultura Celular por Lotes/instrumentação , Separação Celular/instrumentação , Internacionalidade
5.
Adv Biochem Eng Biotechnol ; 139: 167-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23719711

RESUMO

Contamination by viral and microbial agents is a serious risk for biopharmaceuticals produced by mammalian cell culture processes. In order to effectively mitigate the risk and minimize the occurrence of such contamination events, a multi-tiered approach has been adopted to safeguard the manufacturing processes from A to Z. The multi-tiered approach consists of three separate, yet complementary, elements: (1) control and testing of raw materials in general, and animal sourced materials (ASM) in particular; (2) in-process and release testing for adventitious agents with emphasis on viruses based on risk assessment; and (3) demonstration of an adequate, robust, and consistent viral clearance capability by the downstream purification process. The implementations of these measures will be described in the context of regulatory compliance and GMP manufacturing.


Assuntos
Técnicas de Cultura Celular por Lotes/normas , Produtos Biológicos/normas , Qualidade de Produtos para o Consumidor/normas , Contaminação de Medicamentos/prevenção & controle , Indústria Farmacêutica/normas , Garantia da Qualidade dos Cuidados de Saúde/normas , Animais , Técnicas de Cultura Celular por Lotes/instrumentação , Produtos Biológicos/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Desenho de Equipamento/normas , Análise de Falha de Equipamento/normas , Humanos , Internacionalidade , Mamíferos
6.
Adv Biochem Eng Biotechnol ; 139: 123-66, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23748351

RESUMO

The manufacturing of a biologic drug from mammalian cells results in not a single substance, but an array of product isoforms, also known as variants. These isoforms arise due to intracellular or extracellular events as a result of biological or chemical modification. The most common examples related to biomanufacturing include amino acid modifications (glycosylation, isomerization, oxidation, adduct formation, pyroglutamate formation, phosphorylation, sulfation, amidation), amino acid sequence variants (genetic mutations, amino acid misincorporation, N- and C-terminal heterogeneity, clipping), and higher-order structure modifications (misfolding, aggregation, disulfide pairing). Process-related impurities (HCP, DNA, media components, viral particles) are also important quality attributes related to product safety. The observed ranges associated with each quality attribute define the product quality profile. A biologic drug must have a correct and consistent quality profile throughout clinical development and scale-up to commercial production to ensure product safety and efficacy. In general, the upstream process (cell culture) defines the quality of product-related substances, whereas the downstream process (purification) defines the residual level of process- and product-related impurities. The purpose of this chapter is to review the impact of the cell culture process on product quality. Emphasis is placed on studies with industrial significance and where the direct mechanism of product quality impact was determined. Where possible, recommendations for maintaining consistent or improved quality are provided.


Assuntos
Técnicas de Cultura Celular por Lotes/normas , Produtos Biológicos/normas , Indústria Farmacêutica/normas , Desenho de Equipamento/normas , Análise de Falha de Equipamento/normas , Controle de Qualidade , Algoritmos , Técnicas de Cultura Celular por Lotes/instrumentação , Produtos Biológicos/metabolismo , Separação Celular/instrumentação , Separação Celular/normas , Desenho de Fármacos , Internacionalidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA