Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Chromatogr A ; 1241: 37-45, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22560704

ABSTRACT

In the production of a human therapeutic protein from inclusion bodies, product related impurities of very similar size and charge to the product are created as byproducts of the refold process. Their removal is usually challenging even when using chromatography with high performance resins and elution by shallow linear gradients. Additionally, performing this type of separation for commercial production adds increased complexity. To maximize productivity, columns are loaded so high that product elution profiles are not well separated from the impurities and pooling decisions are challenging. In this paper, conventional UV pooling based on fractionation or predefined absorbance based criteria will be compared to pooling based on fast on-line HPLC analytic. The development and implementation in a GMP process will be shown for a specific challenging separation by hydrophobic interaction chromatography. The different approaches have their unique complexities, timelines, uncertainties, and risks during development and implementation as well as during manufacturing. This study presents a probabilistic framework for quantitative comparison of two processes with unequal variability and uncertainty to evaluate the potential benefits of a PAT technology for its routine use in GMP Bioprocess manufacturing.


Subject(s)
Chemistry, Pharmaceutical/methods , Chemistry, Pharmaceutical/standards , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Drug Contamination/prevention & control , Humans , Models, Theoretical , Recombinant Proteins/isolation & purification , Research Design , Risk Assessment
2.
Biotechnol J ; 3(9-10): 1185-200, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18759247

ABSTRACT

Process advancements driven through innovations have been key factors that enabled successful commercialization of several human therapeutic antibodies in recent years. The production costs of these molecules are higher in comparison to traditional medicines. In order to lower the development and later manufacturing costs, recent advances in antibody production technologies target higher throughput processes with increased clinical and commercial economics. In this review, essential considerations and trends for commercial process development and optimization are described, followed by the challenges to obtain a high titer cell culture process and its subsequent impact on the purification process. One of these recent technical advances is the development and implementation of a disposable Q membrane adsorber as an alternative to a Q-packed-bed column in a flow-through mode. The scientific concept and principles underlining Q membrane technology and its application are also reviewed.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Protein Engineering/instrumentation , Protein Engineering/methods , Antibodies, Monoclonal/isolation & purification , Bioreactors , Cell Culture Techniques , Chromatography , Chromatography, Affinity
3.
Biotechnol Bioeng ; 100(3): 488-96, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18438883

ABSTRACT

Once highly selective protein A affinity is chosen for robust mAb downstream processing, the major role of polishing steps is to remove product related impurities, trace amounts of host cell proteins, DNA/RNA, and potential viral contaminants. Disposable systems can act as powerful options either to replace or in addition to polishing column chromatography to ensure product purity and excellent viral clearance power for patients' safety. In this presentation, the implementation of three disposable systems such as depth filtration, membrane chromatography, and nanometer filtration technology in a commercial process are introduced. The data set of viral clearance with these systems is presented. Application advantages and disadvantages including cost analysis are further discussed.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Biotechnology , Disposable Equipment/statistics & numerical data , Drug Contamination/prevention & control , Viruses/isolation & purification , Animals , Chromatography/instrumentation , Chromatography/methods , Costs and Cost Analysis , Disposable Equipment/economics , Filtration/instrumentation , Filtration/methods , Humans , Hydrogen-Ion Concentration , Pressure , Temperature
4.
J Chromatogr A ; 1175(1): 69-80, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17980374

ABSTRACT

The commercial production of recombinant human monoclonal antibody therapeutics demands robust processes. In this article we describe the development of a pH-conductivity hybrid gradient for a cation-exchange chromatography step to obtain high binding capacity and consistent purification resolution in scale process. Operational parameters and their ranges were characterized with DOE statistical method. Aggregate, DNA and leached protein A removal were examined during development. The advantages and disadvantages of hybrid gradient elution compared to sodium chloride gradient elution were explored. As this step was designed as a good fit for the compatibility of the feed and operating pH/conductivity conditions for next step, the effects of elution by either changing sodium chloride concentration or changing pH of elution buffers on overall separation efficiency were compared. The operation condition was further confirmed in six 2000 L scale runs. The thorough evaluation demonstrated process reliability of hybrid gradient cation-exchange chromatography with high step purity and yield.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Chromatography, Ion Exchange/methods , Animals , Buffers , CHO Cells , Cations , Chemical Fractionation , Chromatography, Gel , Cricetinae , Cricetulus , DNA/isolation & purification , Humans , Hydrogen-Ion Concentration , Polymers/chemistry , Resins, Synthetic/chemistry , Sodium Acetate/chemistry , Sodium Chloride/chemistry , Staphylococcal Protein A/isolation & purification , Temperature
5.
Article in English | MEDLINE | ID: mdl-17046339

ABSTRACT

This paper presents an overview of large-scale downstream processing of monoclonal antibodies and Fc fusion proteins (mAbs). This therapeutic modality has become increasingly important with the recent approval of several drugs from this product class for a range of critical illnesses. Taking advantage of the biochemical similarities in this product class, several templated purification schemes have emerged in the literature. In our experience, significant biochemical differences and the variety of challenges to downstream purification make the use of a completely generic downstream process impractical. Here, we describe the key elements of a flexible, generic downstream process platform for mAbs that we have adopted at Amgen. This platform consists of a well-defined sequence of unit operations with most operating parameters being pre-defined and a small subset of parameters requiring development effort. The platform hinges on the successful use of Protein A chromatography as a highly selective capture step for the process. Key elements of each type of unit operation are discussed along with data from 14 mAbs that have undergone process development. Aspects that can be readily templated as well as those that require focused development effort are identified for each unit operation. A brief description of process characterization and validation activities for these molecules is also provided. Finally, future directions in mAb processing are summarized.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/genetics , Chromatography, Affinity/methods , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/isolation & purification , Models, Molecular , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Staphylococcal Protein A/chemistry , Staphylococcal Protein A/genetics
6.
J Chromatogr A ; 1134(1-2): 66-73, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16965788

ABSTRACT

Process-scale antibody production requires polishing steps with extremely high product throughput and robust operation. In this communication, the Sartobind Q membrane adsorber for process-scale antibody production is evaluated as an alternative to Q column chromatography. Although the capacity seen with large-scale membrane adsorbers is competitive with column chromatography, the same throughput is not achieved with the current scale-down models. The operational issues currently found in membrane scale-down models, including backpressure, which significantly compromises the membrane's capacity, were examined. A new scale-down model was designed to mimic the liquid flow path found in the large-scale capsule, and a new process capacity equivalent at both small and large scale was successfully achieved. Results of a 4-model virus study with a redesigned Sartobind Q absorber scale-down model at the new process capacity are presented.


Subject(s)
Antibodies/isolation & purification , Membranes, Artificial , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Models, Biological , Reproducibility of Results , Virus Physiological Phenomena
7.
Biotechnol Prog ; 22(2): 341-9, 2006.
Article in English | MEDLINE | ID: mdl-16599545

ABSTRACT

The large-scale production of recombinant human monoclonal antibodies demands economical purification processes with high throughputs. In this article we briefly describe a common antibody process and evaluate the Q membrane adsorber for process-scale antibody production as an alternative to a Q-packed-bed column in a flow-through mode. The scientific concepts underlining Q membrane technology and its application are reviewed. The disadvantages and advantages of using Q membrane chromatography as a purification unit in large-scale production are discussed, including problems initially seen with the Q membrane scale-down model but solved with the invention of a new scale-down model. The new Q-membrane unit operation has a process capacity greater than 3,000 g/m(2) or 10.7 kg/L with a LRV over 5 for four model viruses. In this Review, a cost analysis illustrates that Q membrane chromatography is a viable alternative to Q column chromatography as a polishing step in a flow-through mode for process-scale antibody production.


Subject(s)
Antibodies/isolation & purification , Chromatography/methods , Animals , Chromatography/economics , Humans , Models, Biological , Temperature , Time Factors , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL
...