Cargando…

Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures

The continued need to improve therapeutic recombinant protein productivity has led to ongoing assessment of appropriate strategies in the biopharmaceutical industry to establish robust processes with optimized critical variables, that is, viable cell density (VCD) and specific productivity (product...

Descripción completa

Detalles Bibliográficos
Autores principales: Du, Zhimei, Treiber, David, McCarter, John D, Fomina-Yadlin, Dina, Saleem, Ramsey A, McCoy, Rebecca E, Zhang, Yuling, Tharmalingam, Tharmala, Leith, Matthew, Follstad, Brian D, Dell, Brad, Grisim, Brent, Zupke, Craig, Heath, Carole, Morris, Arvia E, Reddy, Pranhitha
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BlackWell Publishing Ltd 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4282109/
https://www.ncbi.nlm.nih.gov/pubmed/25042542
http://dx.doi.org/10.1002/bit.25332
_version_ 1782351074112307200
author Du, Zhimei
Treiber, David
McCarter, John D
Fomina-Yadlin, Dina
Saleem, Ramsey A
McCoy, Rebecca E
Zhang, Yuling
Tharmalingam, Tharmala
Leith, Matthew
Follstad, Brian D
Dell, Brad
Grisim, Brent
Zupke, Craig
Heath, Carole
Morris, Arvia E
Reddy, Pranhitha
author_facet Du, Zhimei
Treiber, David
McCarter, John D
Fomina-Yadlin, Dina
Saleem, Ramsey A
McCoy, Rebecca E
Zhang, Yuling
Tharmalingam, Tharmala
Leith, Matthew
Follstad, Brian D
Dell, Brad
Grisim, Brent
Zupke, Craig
Heath, Carole
Morris, Arvia E
Reddy, Pranhitha
author_sort Du, Zhimei
collection PubMed
description The continued need to improve therapeutic recombinant protein productivity has led to ongoing assessment of appropriate strategies in the biopharmaceutical industry to establish robust processes with optimized critical variables, that is, viable cell density (VCD) and specific productivity (product per cell, qP). Even though high VCD is a positive factor for titer, uncontrolled proliferation beyond a certain cell mass is also undesirable. To enable efficient process development to achieve consistent and predictable growth arrest while maintaining VCD, as well as improving qP, without negative impacts on product quality from clone to clone, we identified an approach that directly targets the cell cycle G1-checkpoint by selectively inhibiting the function of cyclin dependent kinases (CDK) 4/6 with a small molecule compound. Results from studies on multiple recombinant Chinese hamster ovary (CHO) cell lines demonstrate that the selective inhibitor can mediate a complete and sustained G0/G1 arrest without impacting G2/M phase. Cell proliferation is consistently and rapidly controlled in all recombinant cell lines at one concentration of this inhibitor throughout the production processes with specific productivities increased up to 110 pg/cell/day. Additionally, the product quality attributes of the mAb, with regard to high molecular weight (HMW) and glycan profile, are not negatively impacted. In fact, high mannose is decreased after treatment, which is in contrast to other established growth control methods such as reducing culture temperature. Microarray analysis showed major differences in expression of regulatory genes of the glycosylation and cell cycle signaling pathways between these different growth control methods. Overall, our observations showed that cell cycle arrest by directly targeting CDK4/6 using selective inhibitor compound can be utilized consistently and rapidly to optimize process parameters, such as cell growth, qP, and glycosylation profile in recombinant antibody production cultures.
format Online
Article
Text
id pubmed-4282109
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BlackWell Publishing Ltd
record_format MEDLINE/PubMed
spelling pubmed-42821092015-01-15 Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures Du, Zhimei Treiber, David McCarter, John D Fomina-Yadlin, Dina Saleem, Ramsey A McCoy, Rebecca E Zhang, Yuling Tharmalingam, Tharmala Leith, Matthew Follstad, Brian D Dell, Brad Grisim, Brent Zupke, Craig Heath, Carole Morris, Arvia E Reddy, Pranhitha Biotechnol Bioeng Articles The continued need to improve therapeutic recombinant protein productivity has led to ongoing assessment of appropriate strategies in the biopharmaceutical industry to establish robust processes with optimized critical variables, that is, viable cell density (VCD) and specific productivity (product per cell, qP). Even though high VCD is a positive factor for titer, uncontrolled proliferation beyond a certain cell mass is also undesirable. To enable efficient process development to achieve consistent and predictable growth arrest while maintaining VCD, as well as improving qP, without negative impacts on product quality from clone to clone, we identified an approach that directly targets the cell cycle G1-checkpoint by selectively inhibiting the function of cyclin dependent kinases (CDK) 4/6 with a small molecule compound. Results from studies on multiple recombinant Chinese hamster ovary (CHO) cell lines demonstrate that the selective inhibitor can mediate a complete and sustained G0/G1 arrest without impacting G2/M phase. Cell proliferation is consistently and rapidly controlled in all recombinant cell lines at one concentration of this inhibitor throughout the production processes with specific productivities increased up to 110 pg/cell/day. Additionally, the product quality attributes of the mAb, with regard to high molecular weight (HMW) and glycan profile, are not negatively impacted. In fact, high mannose is decreased after treatment, which is in contrast to other established growth control methods such as reducing culture temperature. Microarray analysis showed major differences in expression of regulatory genes of the glycosylation and cell cycle signaling pathways between these different growth control methods. Overall, our observations showed that cell cycle arrest by directly targeting CDK4/6 using selective inhibitor compound can be utilized consistently and rapidly to optimize process parameters, such as cell growth, qP, and glycosylation profile in recombinant antibody production cultures. BlackWell Publishing Ltd 2015-01 2014-09-02 /pmc/articles/PMC4282109/ /pubmed/25042542 http://dx.doi.org/10.1002/bit.25332 Text en © 2014 The Authors. Biotechnology and Bioengineering Published by Wiley Periodicals, Inc. http://creativecommons.org/licenses/by-nc/3.0/ This is an open access article under the terms of the Creative Commons Attribution-NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
spellingShingle Articles
Du, Zhimei
Treiber, David
McCarter, John D
Fomina-Yadlin, Dina
Saleem, Ramsey A
McCoy, Rebecca E
Zhang, Yuling
Tharmalingam, Tharmala
Leith, Matthew
Follstad, Brian D
Dell, Brad
Grisim, Brent
Zupke, Craig
Heath, Carole
Morris, Arvia E
Reddy, Pranhitha
Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures
title Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures
title_full Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures
title_fullStr Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures
title_full_unstemmed Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures
title_short Use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in CHO cell cultures
title_sort use of a small molecule cell cycle inhibitor to control cell growth and improve specific productivity and product quality of recombinant proteins in cho cell cultures
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4282109/
https://www.ncbi.nlm.nih.gov/pubmed/25042542
http://dx.doi.org/10.1002/bit.25332
work_keys_str_mv AT duzhimei useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT treiberdavid useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT mccarterjohnd useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT fominayadlindina useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT saleemramseya useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT mccoyrebeccae useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT zhangyuling useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT tharmalingamtharmala useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT leithmatthew useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT follstadbriand useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT dellbrad useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT grisimbrent useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT zupkecraig useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT heathcarole useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT morrisarviae useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures
AT reddypranhitha useofasmallmoleculecellcycleinhibitortocontrolcellgrowthandimprovespecificproductivityandproductqualityofrecombinantproteinsinchocellcultures