Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Opin Biotechnol ; 76: 102719, 2022 08.
Article in English | MEDLINE | ID: mdl-35569340

ABSTRACT

Cell-free protein synthesis (CFPS) technologies have grown from lab-scale research tools to biopharmaceutical production at the Good Manufacturing Practice manufacturing scale. Multiple human clinical trials are in progress with CFPS-based products. In addition, applications of CFPS in research have continued to expand over the years and play an important role in biopharmaceutical product discovery and development. The unique, open nature of CFPS has enabled efficient non-natural amino acid (nnAA) incorporation into protein products, which expands the range of biotherapeutics that can be considered for novel treatments. The flexibility and speed of CFPS combined with novel nnAA capabilities are poised to open a new chapter in the continuing evolution of biotherapies.


Subject(s)
Biological Products , Amino Acids/chemistry , Cell-Free System/chemistry , Humans , Protein Biosynthesis , Proteins/chemistry
2.
Biotechnol Prog ; 31(3): 823-31, 2015.
Article in English | MEDLINE | ID: mdl-25826247

ABSTRACT

Cell-free protein synthesis (CFPS) systems allow for robust protein expression with easy manipulation of conditions to improve protein yield and folding. Recent technological developments have significantly increased the productivity and reduced the operating costs of CFPS systems, such that they can compete with conventional in vivo protein production platforms, while also offering new routes for the discovery and production of biotherapeutics. As cell-free systems have evolved, productivity increases have commonly been obtained by addition of components to previously designed reaction mixtures without careful re-examination of the essentiality of reagents from previous generations. Here we present a systematic sensitivity analysis of the components in a conventional Escherichia coli CFPS reaction mixture to evaluate their optimal concentrations for production of the immunoglobulin G trastuzumab. We identify eight changes to the system, which result in optimal expression of trastuzumab. We find that doubling the potassium glutamate concentration, while entirely eliminating pyruvate, coenzyme A, NAD, total tRNA, folinic acid, putrescine and ammonium glutamate, results in a highly productive cell-free system with a 95% reduction in reagent costs (excluding cell-extract, plasmid, and T7 RNA polymerase made in-house). A larger panel of other proteins was also tested and all show equivalent or improved yields with our simplified system. Furthermore, we demonstrate that all of the reagents for CFPS can be combined in a single freeze-thaw stable master mix to improve reliability and ease of use. These improvements are important for the application of the CFPS system in fields such as protein engineering, high-throughput screening, and biotherapeutics.


Subject(s)
Escherichia coli/metabolism , Immunoglobulin G/biosynthesis , Protein Biosynthesis , Protein Engineering/methods , Trastuzumab/biosynthesis , Coenzyme A/chemistry , DNA-Directed RNA Polymerases/chemistry , Escherichia coli/genetics , Gene Expression , Glutamic Acid/chemistry , Immunoglobulin G/genetics , Leucovorin/chemistry , NAD/chemistry , Polyamines/chemistry , Protein Folding , Putrescine/chemistry , Pyruvic Acid/chemistry , RNA, Transfer/chemistry , Reproducibility of Results , Trastuzumab/genetics , Viral Proteins/chemistry
3.
Methods Mol Biol ; 805: 31-41, 2012.
Article in English | MEDLINE | ID: mdl-22094798

ABSTRACT

Crude cell-free extracts are useful tools for investigating biochemical phenomena and exploiting complex enzymatic processes such as protein synthesis. Extracts derived from E. coli have been used for over 50 years to study the mechanism of protein synthesis. In addition, these S30 extracts are commonly used as a laboratory tool for protein production. The preparation of S30 extract has been streamlined over the years and now it is a relatively simple process. The procedure described here includes some suggestions for extracts to be used for ribosome display.


Subject(s)
Cell Extracts , Escherichia coli Proteins/biosynthesis , Escherichia coli/metabolism , Protein Biosynthesis , Transcription, Genetic , Escherichia coli/cytology , Escherichia coli Proteins/isolation & purification , Ribosomes/metabolism
4.
Biotechnol Bioeng ; 108(7): 1570-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21337337

ABSTRACT

Engineering robust protein production and purification of correctly folded biotherapeutic proteins in cell-based systems is often challenging due to the requirements for maintaining complex cellular networks for cell viability and the need to develop associated downstream processes that reproducibly yield biopharmaceutical products with high product quality. Here, we present an alternative Escherichia coli-based open cell-free synthesis (OCFS) system that is optimized for predictable high-yield protein synthesis and folding at any scale with straightforward downstream purification processes. We describe how the linear scalability of OCFS allows rapid process optimization of parameters affecting extract activation, gene sequence optimization, and redox folding conditions for disulfide bond formation at microliter scales. Efficient and predictable high-level protein production can then be achieved using batch processes in standard bioreactors. We show how a fully bioactive protein produced by OCFS from optimized frozen extract can be purified directly using a streamlined purification process that yields a biologically active cytokine, human granulocyte-macrophage colony-stimulating factor, produced at titers of 700 mg/L in 10 h. These results represent a milestone for in vitro protein synthesis, with potential for the cGMP production of disulfide-bonded biotherapeutic proteins.


Subject(s)
Biotechnology/methods , Escherichia coli/enzymology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Technology, Pharmaceutical/methods , Bioreactors , Cell-Free System , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
5.
Biotechnol Prog ; 21(2): 460-5, 2005.
Article in English | MEDLINE | ID: mdl-15801786

ABSTRACT

Escherichia coli extracts activate cell-free protein synthesis systems by providing the catalysts for translation and other supporting reactions. Recent results suggest that high-density fermentations can be used to provide the source cells, but the subsequent cell extract preparation procedure requires multiple centrifugation and dialysis steps as well as an expensive runoff reaction. In the work reported here, the extract preparation protocol duration was reduced by nearly 50% by significantly shortening several steps. In addition, by optimizing the runoff incubation, overall reagent costs were reduced by 70%. Nonetheless, extracts produced from the shorter, less expensive procedure were equally active. Crucial steps were further examined to indicate minimal ribosome loss during the standard 30,000g centrifugations. Furthermore, sucrose density centrifugation analysis indicated that although an incubation step significantly activates the extract, ribosome/polysome dissociation is not required. These insights suggest that consistent cell extract can be produced more quickly and with considerably less expense for large-scale cell-free protein production, especially when combined with high-density fermentation protocols.


Subject(s)
Escherichia coli/genetics , Recombinant Proteins/biosynthesis , Cell-Free System , Centrifugation , Electrophoresis, Polyacrylamide Gel , Fermentation
SELECTION OF CITATIONS
SEARCH DETAIL
...