Please wait a minute...

中国生物工程杂志

China Biotechnology
China Biotechnology  2013, Vol. 33 Issue (7): 103-111    DOI:
    
The Lasted Development of Large Scale Cell Culture Technology for Commercial Antibody Manufacture
LIU Bo-ning
New Drug Reaserch and Development Center, North China Pharmaceutical Group Corporation and State Key Laboratory of Antibody drug Reaserch and Development, Shijiazhuang 050015, China
Download: HTML   PDF(1010KB) HTML
Export: BibTeX | EndNote (RIS)      

Abstract  Large scale animal cell culture technology has leaped forward in the lasted years. The state-of-art of cell culture process development for commercial monoclonal antibody production, which is involved with medium optimization methodology and process development of fed batch/perfusion mode, was discussed in detail. Some methods and compositions used for improving recombinant yield and impact of cell culture process on critical quality attribute of recombinant antibody (such as: aggregate、degradation、glycosylation, charge heterogeneity)were summarized and illustrated by cases. Additional, some topic involved with cGMP manufacturing was addressed, including process characterization and validation. It can been forecasted that the omics research of animal cell and PAT (process analysis technology) will accelerate process development of large scale cell culture.

Key wordsRecombinant antibody      Large scale cell culture      Medium optimization      Process development     
Received: 07 April 2013      Published: 25 July 2013
ZTFLH:  Q511  
Cite this article:

LIU Bo-ning. The Lasted Development of Large Scale Cell Culture Technology for Commercial Antibody Manufacture. China Biotechnology, 2013, 33(7): 103-111.

URL:

https://manu60.magtech.com.cn/biotech/     OR     https://manu60.magtech.com.cn/biotech/Y2013/V33/I7/103

[1] Chartrain M, Chu L. Development and production of commercial therapeutic monoclonal antibodies in mammalian cell expression systems: An overview of the current upstream technologies. Current Pharmaceutical Biotechnology, 2008, 9(6): 447-467.
[2] Huang Y M, Hu W, Rustandi E, et al. Maximizing productivity of cho cell-based fed-batch culture using chemically defined media conditions and typical manufacturing equipment. Biotechnology Progress, 2010, 26(5): 1400-1410.
[3] 刘伯宁. 治疗性抗体研究进展与抗体产业关键技术. 中国生物工程杂志, 2013,33(5):132-138. Liu B N. The progress of therapeutic antibody drug and the industrial key-technology of antibody production. China Biotechnoloy, 2013,33(5):132-138 .
[4] DePalma A. Enhancement of cell culture techniques. Genetic Engineering & Biotechnology News, 2009, 29(18).
[5] 刘伯宁. 用于重组抗体生产的细胞构建技术研究进展. 中国生物工程杂志, 2013,33(6):111-116. Liu B N. The technology progress of antibody-producing cell line development. China Biotechnoloy, 2013,33 (6):111-116.
[6] Xing Z, Li Z, Chow V, et al. Identifying inhibitory threshold values of repressing metabolites in cho cell culture using multivariate analysis methods. Biotechnology Progress, 2008, 24(3): 675-683.
[7] Li F, Vijayasankaran N, Shen A Y, et al. Cell culture processes for monoclonal antibody production. MAbs, 2010, 2(5): 466-479.
[8] Whitford CJaW. Bioreactor chapter two: Bioreactor control. Bioprocess Int Supplement, 2007.
[9] Hermes P A, Castro C D. A fully defined, fed-batch, recombinant NS0 culture process for monoclonal antibody production. Biotechnology Progress, 2010, 26(5): 1411-1416.
[10] Burky J E, Wesson M C, Young A, et al. Protein-free fed-batch culture of non-gs NS0 cell lines for production of recombinant antibodies. Biotechnology and Bioengineering, 2007, 96(2): 281-293.
[11] Chee F W D, Tin K W K, Tang G L, et al. Impact of dynamic online fed-batch strategies on metabolism, productivity and n-glycosylation quality in cho cell cultures. Biotechnology and Bioengineering, 2005, 89(2): 164-177.
[12] Khattak S F, Xing Z, Kenty B, et al. Feed development for fed-batch cho production process by semisteady state analysis. Biotechnology Progress, 2010, 26(3): 797-804.
[13] De Alwis D M, Dutton R L, Scharer J, et al. Statistical methods in media optimization for batch and fed-batch animal cell culture. Bioprocess Biosyst Eng, 2007, 30(2): 107-113.
[14] Jardin B A, Montes J, Lanthier S, et al. High cell density fed batch and perfusion processes for stable non-viral expression of secreted alkaline phosphatase (seap) using insect cells: Comparison to a batch sf-9-bev system. Biotechnology and Bioengineering, 2007, 97(2): 332-345.
[15] Wang L, Hu H, Yang J, et al. High yield of human monoclonal antibody produced by stably transfected drosophila schneider 2 cells in perfusion culture using wave bioreactor. Molecular Biotechnology, 2011,52(2):170-179.
[16] Vermasvuori R, Hurme M. Economic comparison of diagnostic antibody production in perfusion stirred tank and in hollow fiber bioreactor processes. Biotechnology Progress, 2011, 27(6): 1588-1598.
[17] Pollock J, Ho S V, Farid S S. Fed-batch and perfusion culture processes: Economic, environmental, and operational feasibility under uncertainty. Biotechnology and Bioengineering, 2012,110(1):206-219.
[18] Konstantinov K, Goudar C, Ng M, et al. The "push-to-low"approach for optimization of high-density perfusion cultures of animal cells. Adv Biochem Eng Biotechnol, 2006, 101:75-98.
[19] Yang J D, Angelillo Y, Chaudhry M, et al. Achievement of high cell density and high antibody productivity by a controlled-fed perfusion bioreactor process. Biotechnology and Bioengineering, 2000, 69(1): 74-82.
[20] Kim B J, Diao J, Shuler M L. Mini-scale bioprocessing systems for highly parallel animal cell cultures. Biotechnology Progress, 2012, 28(3): 595-607.
[21] Legmann R, Schreyer H B, Combs R G, et al. A predictive high-throughput scale-down model of monoclonal antibody production in cho cells. Biotechnology and Bioengineering, 2009, 104(6): 1107-1120.
[22] Chen A, Chitta R, Chang D, et al. Twenty-four well plate miniature bioreactor system as a scale-down model for cell culture process development. Biotechnology and Bioengineering, 2009, 102(1): 148-160.
[23] Vijayasankaran N, Li J, Shawley R, et al. Animal cell culture media. Encyclopedia of Industrial Biotechnology, 2009.1-15.
[24] Paula Decaria A S, William W. Many considerations in selecting bioproduction culture media. Bioprocess Int, 2009(11):44-51.
[25] Mosser M, Chevalot I, Olmos E, et al. Combination of yeast hydrolysates to improve cho cell growth and igg production. Cytotechnology, 2012,14(12).
[26] Jordan M, Voisard D, Berthoud A, et al. Cell culture medium improvement by rigorous shuffling of components using media blending. Cytotechnology, 2013,65(1):31-40.
[27] Gonzalez-Leal I J, Carrillo-Cocom L M, Ramirez-Medrano A, et al. Use of a plackett-burman statistical design to determine the effect of selected amino acids on monoclonal antibody production in cho cells. Biotechnology Progress, 2011, 27(6): 1709-1717.
[28] Fan L, Zhao L, Sun Y, et al. A high-yielding, generic fed-batch process for recombinant antibody production of gs-engineered cell lines. J Microbiol Biotechnol, 2009, 19(12): 1695-1702.
[29] Xie L, Wang D I. Stoichiometric analysis of animal cell growth and its application in medium design. Biotechnology and Bioengineering, 1994, 43(11): 1164-1174.
[30] Zhou W, Rehm J, Hu W S. High viable cell concentration fed-batch cultures of hybridoma cells through on-line nutrient feeding. Biotechnology and Bioengineering, 1995, 46(6): 579-587.
[31] Fletcher T. Designing culture media for recombinant protein production:a rational approach. Bioprocess Int, 2005,3(1):30-36.
[32] Zhou Jiang K D, Zhaohui Geng, Susan Casnocha, Zhihua Xiao, Steve Gorfien, and Scott J. Jacobia. Fed-batch cell culture process optimization: A rationally integrated approach. Bioprocess Int, 2012, 10(3): 40-45.
[33] Min Zhang A L, Kerry K, Terrell J, et al. Rapid development and optimization of cell culture media. BioPharm International, 2008, 21(1):60-68.
[34] Hammett K, Kuchibhatla J, Hunt C, et al. Developing chemically defined media through doe: Complete optimization with increased protein production in less than 8 months cell technology for cell products. 2007, 683-691.
[35] Efren Pacis N V, Jincai L i, Martin G, et al. Systematic approaches to develop chemically defined cell culture feed media: It is important to ensure that the transition from peptone-containing to cd media doesn't affect product quality. BioPharm International Supplements, 2010, 23(11):22-32.
[36] Ningning Ma J E, Centy O, Paul H, et al. A single nutrient feed supports both chemically defined ns0 and cho fed-batch processes: Improved productivity and lactate metabolism. Biotechnology Progress, 2009, 25(5):1353-1363.
[37] Maranga L, Goochee C F. Metabolism of per.C6 cells cultivated under fed-batch conditions at low glucose and glutamine levels. Biotechnology and Bioengineering, 2006, 94(1): 139-150.
[38] Kuwae S, Ohda T, Tamashima H, et al. Development of a fed-batch culture process for enhanced production of recombinant human antithrombin by chinese hamster ovary cells. Journal of Bioscience and Bioengineering, 2005, 100(5): 502-510.
[39] Luo W, Sun X, Yi X, et al. Enhancement of hepatitis b surface antigen production by adenosine 5'-monophosphate in culture of recombinant chinese hamster ovary cells. Journal of Bioscience and Bioengineering, 2005, 100(4): 475-477.
[40] Omasa T, Furuichi K, Iemura T, et al. Enhanced antibody production following intermediate addition based on flux analysis in mammalian cell continuous culture. Bioprocess Biosyst Eng, 2010, 33(1): 117-125.
[41] Bai Y, Wu C, Zhao J, et al. Role of iron and sodium citrate in animal protein-free cho cell culture medium on cell growth and monoclonal antibody production. Biotechnology Progress, 2011, 27(1): 209-219.
[42] Konno Y, Aoki M, Takagishi M, et al. Enhancement of antibody production by the addition of coenzyme-q(10). Cytotechnology, 2011, 63(2): 163-170.
[43] Zhang J, Robinson D, Salmon P. A novel function for selenium in biological system: Selenite as a highly effective iron carrier for chinese hamster ovary cell growth and monoclonal antibody production. Biotechnology and Bioengineering, 2006, 95(6): 1188-1197.
[44] deZengotita V M, Miller W M, Aunins J G, et al. Phosphate feeding improves high-cell-concentration ns0 myeloma culture performance for monoclonal antibody production. Biotechnology and Bioengineering, 2000, 69(5): 566-576.
[45] Thombre S, Gadgil M. Increase in efficiency of media utilization for recombinant protein production in chinese hamster ovary culture through dilution. Biotechnology and Applied Biochemistry, 2011, 58(1): 25-31.
[46] Chaderjian W B, Chin E T, Harris R J, et al. Effect of copper sulfate on performance of a serum-free cho cell culture process and the level of free thiol in the recombinant antibody expressed. Biotechnol Prog, 2005, 21(2): 550-553.
[47] Qian Y, Khattak S F, Xing Z, et al. Cell culture and gene transcription effects of copper sulfate on chinese hamster ovary cells. Biotechnology Progress, 2011,27(4):1190-1194.
[48] Schatz S M, Kerschbaumer R J, Gerstenbauer G, et al. Higher expression of fab antibody fragments in a cho cell line at reduced temperature. Biotechnology and Bioengineering, 2003, 84(4): 433-438.
[49] Yoon S K, Song J Y, Lee G M. Effect of low culture temperature on specific productivity, transcription level, and heterogeneity of erythropoietin in chinese hamster ovary cells. Biotechnology and Bioengineering, 2003, 82(3): 289-298.
[50] Yoon S K, Choi S L, Song J Y, et al. Effect of culture ph on erythropoietin production by chinese hamster ovary cells grown in suspension at 32.5 and 37.0 degrees c. Biotechnology and Bioengineering, 2005, 89(3): 345-356.
[51] Sauer P W, Burky J E, Wesson M C, et al. A high-yielding, generic fed-batch cell culture process for production of recombinant antibodies. Biotechnology and Bioengineering, 2000, 67(5): 585-597.
[52] Wu M H, Dimopoulos G, Mantalaris A, et al. The effect of hyperosmotic pressure on antibody production and gene expression in the gs-ns0 cell line. Biotechnology and Applied Biochemistry, 2004, 40(Pt 1): 41-46.
[53] Shen D, Kiehl T R, Khattak S F, et al. Transcriptomic responses to sodium chloride-induced osmotic stress: A study of industrial fed-batch cho cell cultures. Biotechnology Progress, 2010, 26(4): 1104-1115.
[54] Jiang Z, Sharfstein S T. Sodium butyrate stimulates monoclonal antibody over-expression in cho cells by improving gene accessibility. Biotechnology and Bioengineering, 2008, 100(1): 189-194.
[55] Liu C, Chu I, Hwang S. Pentanoic acid, a novel protein synthesis stimulant for chinese hamster ovary (cho) cells. Journal of Bioscience and Bioengineering, 2001, 91(1): 71-75.
[56] Backliwal G, Hildinger M, Kuettel I, et al. Valproic acid: A viable alternative to sodium butyrate for enhancing protein expression in mammalian cell cultures. Biotechnology and Bioengineering, 2008, 101(1): 182-189.
[57] Chun B H, Park S Y, Chung N, et al. Enhanced production of recombinant b-domain deleted factor viii from chinese hamster ovary cells by propionic and butyric acids. Biotechnology Letters, 2003, 25(4): 315-319.
[58] Ling W L, Deng L, Lepore J, et al. Improvement of monoclonal antibody production in hybridoma cells by dimethyl sulfoxide. Biotechnology Progress, 2003, 19(1): 158-162.
[59] Balcarcel R R, Stephanopoulos G. Rapamycin reduces hybridoma cell death and enhances monoclonal antibody production. Biotechnology and Bioengineering, 2001, 76(1): 1-10.
[60] Qian Y, Jing Y, Li Z J. Glucocorticoid receptor-mediated reduction of igg-fusion protein aggregation in chinese hamster ovary cells. Biotechnology Progress, 2010, 26(5): 1417-1423.
[61] Jing Y, Qian Y, Li Z J. Sialylation enhancement of ctla4-ig fusion protein in chinese hamster ovary cells by dexamethasone. Biotechnology and Bioengineering, 2010, 107(3): 488-496.
[62] Rouiller Y, Perilleux A, Marsaut M, et al. Effect of hydrocortisone on the production and glycosylation of an fc-fusion protein in cho cell cultures. Biotechnology Progress, 2012, 28(3): 803-813.
[63] Allen M J, Boyce J P, Trentalange M T, et al. Identification of novel small molecule enhancers of protein production by cultured mammalian cells. Biotechnology and Bioengineering, 2008, 100(6): 1193-1204.
[64] Eon-Duval A, Broly H, Gleixner R. Quality attributes of recombinant therapeutic proteins: An assessment of impact on safety and efficacy as part of a quality by design development approach. Biotechnology Progress, 2012, 28(3): 608-622.
[65] Walsh G, Jefferis R. Post-translational modifications in the context of therapeutic proteins. Nature Biotechnology, 2006, 24(10): 1241-1252.
[66] Swann P G, Tolnay M, Muthukkumar S, et al. Considerations for the development of therapeutic monoclonal antibodies. Curr Opin Immunol, 2008, 20(4): 493-499.
[67] Schiestl M, Stangler T, Torella C, et al. Acceptable changes in quality attributes of glycosylated biopharmaceuticals. Nature Biotechnology, 2011, 29(4): 310-312.
[68] Vazquez-Rey M, Lang D A. Aggregates in monoclonal antibody manufacturing processes. Biotechnology and Bioengineering, 2011, 108(7): 1494-1508.
[69] Gomez N, Subramanian J, Ouyang J, et al. Culture temperature modulates aggregation of recombinant antibody in cho cells. Biotechnology and Bioengineering, 2012, 109(1): 125-136.
[70] Ying Jinga M B, Samiksha Nayakb, Susan Egana, Yueming Qiana, Shih-Hsie Pana, Zheng Jian Lia. Identification of cell culture conditions to control protein aggregation of igg fusion proteins expressed in chinese hamster ovary cells. Process Biochemistry, 2012, 47(1): 69-75.
[71] Jing Y, Qian Y, Ghandi M, et al. A mechanistic study on the effect of dexamethasone in moderating cell death in chinese hamster ovary cell cultures. Biotechnology Progress, 2012, 28(2): 490-496.
[72] Trexler-Schmidt M, Sargis S, Chiu J, et al. Identification and prevention of antibody disulfide bond reduction during cell culture manufacturing. Biotechnology and Bioengineering, 2010, 106(3): 452-461.
[73] Kao Y H, Hewitt D P, Trexler-Schmidt M, et al. Mechanism of antibody reduction in cell culture production processes. Biotechnol Bioeng, 2010, 107(4): 622-632.
[74] Robert F, Bierau H, Rossi M, et al. Degradation of an fc-fusion recombinant protein by host cell proteases: Identification of a cho cathepsin d protease. Biotechnology and Bioengineering, 2009, 104(6): 1132-1141.
[75] Kshirsagar R, McElearney K, Gilbert A, et al. Controlling trisulfide modification in recombinant monoclonal antibody produced in fed-batch cell culture. Biotechnology and Bioengineering, 2012, 109(10): 2523-2532.
[76] Raju T S, Jordan R E. Galactosylation variations in marketed therapeutic antibodies. MAbs, 2012, 4(3):385-391.
[77] Read E K, Park J T, Brorson K A. Industry and regulatory experience of the glycosylation of monoclonal antibodies. Biotechnology and Applied Biochemistry, 2011, 58(4): 213-219.
[78] Hossler P, Khattak S F, Li Z J. Optimal and consistent protein glycosylation in mammalian cell culture. Glycobiology, 2009, 19(9): 936-949.
[79] Yuk I H, Zhang B, Yang Y, et al. Controlling glycation of recombinant antibody in fed-batch cell cultures. Biotechnology and Bioengineering, 2011, 108(11): 2600-2610.
[80] Pacis E, Yu M, Autsen J, et al. Effects of cell culture conditions on antibody n-linked glycosylation-what affects high mannose 5 glycoform. Biotechnology and Bioengineering, 2011,108(10):2348-2358.
[81] Malphettes L, Freyvert Y, Chang J, et al. Highly efficient deletion of fut8 in cho cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies. Biotechnology and Bioengineering, 2010, 106(5): 774-783.
[82] Zhou Q, Shankara S, Roy A, et al. Development of a simple and rapid method for producing non-fucosylated oligomannose containing antibodies with increased effector function. Biotechnology and Bioengineering, 2008, 99(3): 652-665.
[83] Borys M C, Dalal N G, Abu-Absi N R, et al. Effects of culture conditions on n-glycolylneuraminic acid (neu5gc) content of a recombinant fusion protein produced in cho cells. Biotechnology and Bioengineering, 2010, 105(6): 1048-1057.
[84] Gramer M J, Eckblad J J, Donahue R, et al. Modulation of antibody galactosylation through feeding of uridine, manganese chloride, and galactose. Biotechnology and Bioengineering, 2011, 108(7): 1591-1602.
[85] Khawli L A, Goswami S, Hutchinson R, et al. Charge variants in igg1: Isolation, characterization, in vitro binding properties and pharmacokinetics in rats. MAbs, 2010, 2(6): 613-624.
[86] Kaschak T, Boyd D, Lu F, et al. Characterization of the basic charge variants of a human igg1: Effect of copper concentration in cell culture media. MAbs, 2011, 3(6): 577-583.
[87] Luo J, Zhang J, Ren D, et al. Probing of c-terminal lysine variation in a recombinant monoclonal antibody production using chinese hamster ovary cells with chemically defined media. Biotechnology and Bioengineering, 2012, 109(9): 2306-2315.
[88] Khetan A, Huang Y M, Dolnikova J, et al. Control of misincorporation of serine for asparagine during antibody production using cho cells. Biotechnology and Bioengineering, 2010, 107(1): 116-123.
[89] Wen D, Vecchi M M, Gu S, et al. Discovery and investigation of misincorporation of serine at asparagine positions in recombinant proteins expressed in chinese hamster ovary cells. J Biol Chem, 2009, 284(47): 32686-32694.
[90] Seamans T C, Beck A, Wurch T, et al. Cell cultivation process transfer and scale-up:in support of production of early clinical supplies of an anti igf-1r antibody, part 1. BioProcess International, 2008, 26-36.
[91] Seamans T C, Beck A, Wurch T, et al. Cell cultivation processtransfer and scale-up:in support of production of early clinical supplies of an anti igf-1r antibody, part 2. BioProcess International, 2008, 34-42.
[92] Xing Z, Kenty B M, Li Z J, et al. Scale-up analysis for a cho cell culture process in large-scale bioreactors. Biotechnology and Bioengineering, 2009, 103(4): 733-746.
[93] Yang J D, Lu C, Stasny B, et al. Fed-batch bioreactor process scale-up from 3-l to 2,500-l scale for monoclonal antibody production from cell culture. Biotechnology and Bioengineering, 2007, 98(1): 141-154.
[94] Abu-Absi S F, Yang L, Thompson P, et al. Defining process design space for monoclonal antibody cell culture. Biotechnology and Bioengineering, 2010, 106(6): 894-905.
[95] Looby M, Ibarra N, Pierce J J, et al. Application of quality by design principles to the development and technology transfer of a major process improvement for the manufacture of a recombinant protein. Biotechnology Progress, 2011, 27(6): 1718-1729.
[96] Rathore A S, Winkle H. Quality by design for biopharmaceuticals. Nat Biotechnol, 2009, 27(1): 26-34.
[97] Dietmair S, Nielsen L K, Timmins N E. Mammalian cells as biopharmaceutical production hosts in the age of omics. Biotechnology Journal, 2012, 7(1): 75-89.
[98] Read E K, Park J T, Shah R B, et al. Process analytical technology (pat) for biopharmaceutical products: Part i. Concepts and applications. Biotechnology and Bioengineering, 2010, 105(2): 276-284.
[99] Teixeira A P, Oliveira R, Alves P M, et al. Advances in on-line monitoring and control of mammalian cell cultures: Supporting the pat initiative. Biotechnology Advances, 2009, 27(6): 726-732.
[1] WANG Xiao-jie,MENG Fan-qiang,ZHOU Li-bang,LV Feng-xia,BIE Xiao-mei,ZHAO Hai-zhen,LU Zhao-xin. Breeding of Brevibacillin Producing Strain by Genome Shuffling and Optimization of Culture Conditions[J]. China Biotechnology, 2021, 41(8): 42-51.
[2] Hai-jiao JI,Wen-lei LI,Rui-jing Huang,Jian LI,Han-mei XU. Anti-CD20rh MAb Quality Evaluation and Monoclonal Cell Line Screening[J]. China Biotechnology, 2018, 38(8): 34-40.
[3] Yu-lei GUO,Liang TANG,Rui-qiang SUN,You LI,Yi-jun CHEN. High-Throughput Micro Bioreactor Development for Biopharmaceuticals[J]. China Biotechnology, 2018, 38(8): 69-75.
[4] LI Guo-kun, GAO Xiang-dong, XU Chen. Advances on Mammalian Cell Expression System[J]. China Biotechnology, 2014, 34(1): 95-100.
[5] LIU Bo-ning. The Technology Progress of Antibody-producing Cell Line Development[J]. China Biotechnology, 2013, 33(6): 111-116.
[6] LIU Bo-ning. The Progress of Therapeutic Antibody Drug and the Industrial Key-technology of Antibody Production[J]. China Biotechnology, 2013, 33(5): 132-138.
[7] ZHOU Guang-qi, MA Peng-bo, LIU Qiao, QUAN Chun-shan, FAN Sheng-di. Optimization of Culture Medium and Prediction of Antibacterial Activity by Bacillus Amyloliquefaciens Q-426 Fermentation[J]. China Biotechnology, 2013, 33(11): 21-26.
[8] CHEN Jie, WEI Hong-gang, LUO Yuan-chan, ZHANG Dao-jing, LI Shu-lan, TIAN Li, LI Yuan-guang. Medium Optimization for the Production of New Antifungl Cyclic Lipopeptide Marinhysin A by Bacillus Marinus B-9987[J]. China Biotechnology, 2013, 33(1): 84-89.
[9] RUAN Wen-Bing, CHEN Bi-Qin, CHEN Su-Hua, CHEN Bing-Mei, HU Xiao-Beng. Optimization of (R)-Mandelic acid Fermentation Medium by Using Response Surface Methodology[J]. China Biotechnology, 2010, 30(08): 112-117.
[10] ZHANG Ying, HE Jin-Sheng, HONG Chao. Advances and Applications of Recombinant Antibody Drugs[J]. China Biotechnology, 2009, 29(08): 102-106.
[11] . Optimization of fermentation medium with lactose as a inducer for high molecular weight recombinant spider silk protein[J]. China Biotechnology, 2008, 28(12): 41-46.