Abstract
Recently, there has been a growing demand for therapeutic monoclonal antibodies (MAbs) on the global market. Because therapeutic MAbs are more expensive than low-molecular-weight drugs, there have been strong demands to lower their production costs. Therefore, efficient methods to minimize the cost of goods are currently active areas of research. We have screened several enhancers of specific MAb production rate (SPR) using a YB2/0 cell line and found that coenzyme-Q10 (CoQ10) is a promising enhancer candidate. CoQ10 is well known as a strong antioxidant in the respiratory chain and is used for healthcare and other applications. Because CoQ10 is negligibly water soluble, most studies are limited by low concentrations. We added CoQ10 to a culture medium as dispersed nanoparticles at several concentrations (Q-Media) and conducted a fed-batch culture. Although the Q-Media had no effect on cumulative viable cell density, it enhanced SPR by 29%. In addition, the Q-Media had no effect on the binding or cytotoxic activity of MAbs. Q-Media also enhanced SPR with CHO and NS0 cell lines by 30%. These observations suggest that CoQ10 serves as a powerful aid in the production of MAbs by enhancing SPR without changing the characteristics of cell growth, or adversely affecting the quality or biological activity of MAbs.
Keywords: Antioxidant, Coenzyme-Q10, Chinese hamster ovary (CHO), 8-hydroxy-2′-deoxyguanosine, Monoclonal antibodies, NS0, Specific production rate, YB2/0, Enhancer, Productivity
Introduction
Recently, more than twenty therapeutic monoclonal antibodies (MAbs) and related proteins have been launched in the market (Shukla and Thömmes 2010; Walsh 2006). This success is a double-edged sword, however, because it leads to pressure on the pharmaceutical economy. Minimizing the cost of goods and maximizing antibody activity are therefore active areas of research in the development of MAbs for therapeutic use (Shields et al. 2002; Shinkawa et al. 2003; Wurm 2004). There is also great interest in establishing a stable global supply of MAbs.
Several approaches have been explored to improve MAb production in cell cultures. Over the past decade, various approaches such as genetic engineering, clone selection, medium optimization, and reactor engineering have dramatically improved production (Takenouchi and Sugahara 2003). The addition of an enhancer has also proven effective. In the development of biotherapeutics, when an appropriate cell bank is established, we want to maintain use of the same cell bank to avoid risks derived from shifting characterization. The advantage of adding enhancers is the flexibility that can be applied at any development phase. Such enhancers should meet the following requirements: (a) be safe for operators and patients, (b) maintain the quality of the final product, (c) be cost-effective, and (d) be easy to handle. With these requirements in mind, the following enhancers have been suggested: butyrate (De Leon Gatti et al. 2007; Jiang and Sharfstein 2008; Mimura et al. 2001; Oh et al. 1993, 2005), caspase inhibitor (Arden et al. 2007), carnosine (Yegorov et al. 2007), glycerol (Liu and Chen 2007), nucleosides (Carvalhal et al. 2003), linoleic acid (Butler et al. 1999), lipopolysaccharide (Martin-Lopez et al. 2007), dimethyl sulfoxide (Ling et al. 2003; Tsao et al. 2001), rapamycin (Balcarcel and Stephanopoulos 2001), retinoic acid (Inoue et al. 2000), sericin (Terada et al. 2002), spermine (Miyazaki et al. 1998), various sugars (Coroadinha et al. 2006), valproic acid (Backliwal et al. 2008; Konno et al. 2006), and vitamin A (Inoue et al. 1999) (summarized in Table 1).
Table 1.
Reported enhancers of protein production in animal cell lines
| Enhancer | Range/optimal | Product | Effecta | Cell line | Reference |
|---|---|---|---|---|---|
| AMP/nucleotides, nucleosides, and bases | 1 mM | Human placental alkaline phosphatase enzyme (SEAP) | 3-fold in SPR | CHO-K1 | Carvalhal et al. (2003) |
| 1 mM | Factor VII | 2-fold in SPR | CHO-K1 | ||
| 0.3 mM | Factor VII | 2.5-fold in SPR | BHK | ||
| Carnosine | 20 mM | Telomerized cells | 2.5-fold in total number cells | Telomerized cells | Yegorov et al. (2007) |
| Chemical caspase inhibitors | 5–50 μM/5 μM | MAb IgG | 1.2-fold in MAb titer | CHO-K1, HEK-293 | Arden et al. (2007) |
| Dimethyl sulfoxide (DMSO) | 0–1.5%/0.2% (v/v) | MAb | 2-fold in SPR | Hybridoma clone 19 | Ling et al. (2003) |
| Dimethyl sulfoxide (DMSO) | 0–5 mM/1 mM | Adenoviral vector | 3-fold in Virus production | HEK293 | Tsao et al. (2001) |
| Sodium butyrate, | 0–5 mM/0.5 mM | 1.5-fold | |||
| Ethyl alcohol, | 0–200 mM/100 mM | 1.8-fold | |||
| N-acetyl-l-cysteine | 0–5 mM/2.5 mM | 1.8-fold | |||
| Glycerol | 0–2.0%/1% | Macrophage-colony stimulating factor (M-CSF) | 1.4-fold in titer | CHO | Liu and Chen (2007) |
| Lactate dehydrogenase (LDH) | 320 μg/mL | IgM | 12.4-fold | Human–human hybridoma HB4C5 | Takenouchi and Sugahara (2003) |
| Linoleic acid | 25–50 μM/25 μM | MAb (IgG1) | – | Hybridoma (CC9C10) | Butler et al. (1999) |
| Long™ R3 IGF-I | – | MAb | 1.2-fold in titer | CHO | Kim et al. (2005) |
| Triiodothyronine (T3) | – | 1.2-fold in titer | |||
| Lipopolysaccharide (LPS) | 60 pg cell−1 | IgG2a to human immunodeficiency virus (HIV) glycoprotein 120 (gp120). | 3-fold in SPR | Mouse-mouse B cell hybridoma line (CD40 and CD19-deficient expression) | Martin-Lopez et al. (2007) |
| Pyruvate | 0.02 M | Anti-ribonuclease A antibody | 1.4-fold in SPR | Hybridoma 3A21 | Omasa et al. (2010) |
| Malate | 0.005 M | 1.1-fold in SPR | |||
| Citrate | 0.05 M | 1.1-fold in SPR | |||
| Rapamycin | 100 nM | MAb | 0.25 to 0.56 g/L,1.24-fold | Hybridomas CRL 1606 | Balcarcel and Stephanopoulos (2001) |
| Retinoic acid | 10−7 M | IgG | 8-fold | Human–human hybridoma BD9 | Inoue et al. (2000) |
| Small molecule enhancers | 0.5 mM | MAb | 1.6-fold | CHO | Allen et al. (2008) |
| Sodium butyrate (NaBu) + N-acetylcystein (NAC) | 1 mM NaBu and 8 mM NAC | IFN-beta | 2-fold | CHO | Oh et al. (2005) |
| Sodium butyrate | 0–5 mM/2 mM | Mouse/human chimeric IgG3 | 2- to 4-fold | CHO-K1 | Mimura et al. (2001) |
| Sodium butyrate | 5 mM | Humanized IgG | 1- to 4-fold | CHO 9 clones | Jiang and Sharfstein (2008) |
| Spermine | 7.3 mM | IgM, IgG | 6-fold | Human–human hybridoma, HB4C5 | Miyazaki et al. (1998) |
| Sugar sources: glucose, galactose, sorbitol, and fructose | Glc 25–140 mM Frc 83–140 mM | MoMLV-derived recombinant retroviral vectors | 14-fold | Retroviral producer cell line | Coroadinha et al. (2006) |
| Valproic Acid | 100 μM-20 mM/500 μM | MAb | 4-fold, 1.5-fold | CHO-DG44 | Backliwal et al. (2008) |
| Valproic Acid | 100 μM | MAb | 3-fold | CHO | Konno et al. (2006) |
| Vitamin A acetate | 1 μg/L | Human MAb AE6F4 | 0.9- to 2.9-fold | Hybridoma | Inoue et al. (1999) |
aRecalculated % to fold: 20% to 1.2-fold
Coenzyme-Q is a biosynthesized quinone that occurs widely in living organisms such as yeasts, plants, and animals and as such is also known as ubiquinone (ubiquitously occurring quinone). In higher organisms, including humans, this compound has 10 isoprenoid units in its side chain and is therefore named Coenzyme-Q10 (CoQ10; Hathcock and Shao 2006). CoQ10, also known as Ubiquinone-Q10, is widely used as a therapeutic agent, a dietary supplement, an ingredient in cosmetic products, and various other products (Kitano et al. 2004; Müller et al. 2007; Ochiai et al. 2007). CoQ10 is a lipid-soluble compound located in the inner mitochondrial membrane where it functions as a part of the electron transport chain as well as a strong endogenous lipophilic antioxidant (Turunen et al. 2004). CoQ10 is commonly used as a dietary supplement, with the rationale that increasing intake of the nutrient boosts cellular metabolism, particularly in cells with energy deficiencies (Mancuso et al. 2010). The effect of CoQ10 has been tested on various diseases such as diabetes (Hodgson et al. 2002), myocardial infarction (Singh et al. 1998; Soja and Mortensen 1997), angina (Kogan et al. 1999), and Parkinson’s disease (Shults et al. 2004), as well as in anti-breast cancer clinical trials (Portakal et al. 2000). It is also used as adjunct therapy in patients following cardiac surgery (Chello et al. 1996; Hathcock and Shao 2006).
CoQ10 was found to inhibit cell growth and induce apoptosis in HeLa cells (Gorelick et al. 2004), but to decrease apoptosis in human T-acute lymphatic leukemia cells by inhibiting the activity of caspase-3 (Navas et al. 2002). Therefore, the effects of CoQ10 on apoptosis remain controversial. CoQ10 was also shown to promote the growth of many cell lines, including HeLa, HL-60, mouse fibroblast, and bovine embryo cells (Stojkovic et al. 1999; Sun et al. 1992a, 1995). Although CoQ10 as an ingredient in various media appears to stimulate growth, there have been no reports that have directly analyzed this effect on SPR. In this paper, we report the effects of CoQ10 on YB2/0, CHO, and NS0 cell lines.
Materials and methods
Materials
CoQ10 (Kyowa Hakko Bio Co., Ltd., Tokyo, Japan) and Tween-80 (Tokyo Chemical Industry Co., Ltd., Tokyo, Japan) at a 1:2 weight ratio were dissolved completely in ethanol. The solution was heated to remove ethanol, diluted with water, and filtered with a 0.2-μm filter to produce a CoQ10/Tween-80 stock solution. SANOMIT™ Q10 (MSE, Hamburg, Germany) was also used as a possible enhancer of SPR because of its easy handling compared with various other commercial CoQ10 preparations. SANOMIT™ Q10 is a dispersion of nanoparticles (diameter <50 nm) of CoQ10 in water and is useful for preparing stable liquid–liquid dispersions of various CoQ10 concentrations. Hereafter, we refer to the media supplemented with SANOMIT™ Q10 as Q-Media, irrespective of the original composition of the media (Konno et al. 2001). We also tested other enhancer candidates including l-glutathione, alpha-tocopherol, l-ascorbic acid, 2-mercaptoethanol, and 2,3-dimethoxy-5-methyl-p-benzoquinone (Sigma Chemical. Co., Ltd., MO, USA).
Cell culture
In this study, the anchorage-dependent NS0 cell lines (RCB0213; Riken, Ibaraki, Japan) were cultivated in 225 cm2 T-flasks (Asahi Glass, Tokyo, Japan). CHO (Urlaub and Chasin 1985) and YB2/0 cell lines (ATCC 1662) were cultivated in 250 mL shaker flasks (431406; Corning, NY, USA), or in 1 L bioreactors (ABLE, Tokyo, Japan; Ogawa et al. 1999). CHO, YB2/0, and NS0 cell lines expressing proprietary recombinant mouse/human chimeric IgG1 antibody were cultured in Q-Media. Different Q-Media were prepared from ExCell™ 302 (SAFC-Bioscience, St. Louis, MO, USA), RPMI-1640 Medium, Hybridoma-SFM (serum-free medium, with added BSA and transferrin), or CD-Hybridoma (animal-derived, protein-free; Life Technologies, Carlsbad, CA, USA) depending on the cell line used. The cultures were inoculated at a density of 2 × 105 cells mL−1 and cultivated at 37 °C with occasional feeding with serum-free Iscove’s Modified Dulbecco’s Medium until the decline phase. In the culture of 1 L bioreactors, the culture was maintained at pH 7.1 by CO2 gas overlay and 1 mM Na2CO3 alkaline solution at 50% DO by sparger O2 gas.
Cell culture monitoring
The number of viable and dead cells was determined using a CEDEX™ counter (Innovatis AG, Bielefeld, Germany) by the trypan blue dye exclusion method. Off-line measurements of culture glucose and lactate were carried out using an YSI 2700 bioanalyzer (Yellow Springs Inc., OH, USA). The concentration of MAbs in the culture supernatant was determined by HPLC using a Protein A column. The 8-OHdG concentration was quantified using an ELISA kit (8-OHdG check; Japan Institute for the Control of Aging, Shizuoka, Japan).
ADCC activity
Antibody-dependent cell-mediated cytotoxicity (ADCC) was determined by 51Cr release assay, as previously reported (Nakamura et al. 1999).
Equations
![]() |
![]() |
![]() |
Results and discussion
CoQ10 enhanced specific MAb production rate
When an enhancer is used as an additive to culture media for the production of commercial MAbs, consideration must be given to various factors including safety, cost, stability, stable supply, simple usage, patents, and regulations. With these restrictions in mind, we screened various compounds, which can be classified into groups related to the following: (1) nutrition, (2) anti-oxidization, (3) electron transport, (4) growth inhibition or cell cycle, and (5) stress response. In preliminary experiments using YB2/0 cells in 250-mL flasks, media supplemented with 50 μM CoQ10 (dissolved with the aid of Tween-80) exhibited marked enhancement of SPR compared with media supplemented with only Tween-80. Such enhancement (1.26-fold) was also observed for the YB2/0 culture in a 1 L bioreactor (Fig. 1B). Although CoQ10 dissolved with the aid of Tween-80 exhibited enhanced SPR, CoQ10 concentration was limited (<100 μM), and dissolution of CoQ10 required tedious procedures. These problems were circumvented by the use of a stable dispersion of CoQ10 nanoparticles (SANOMIT™ Q10); CoQ10 concentration was easily elevated to as high as 500 μM, and a 1.28-fold increase in SPR could be obtained (Fig. 2). Although the promotion of cell growth by CoQ10 (typically at 30 μM) has been reported in several cell lines (Stojkovic et al. 1999; Sun et al. 1992b, 1995), our results are the first to show the enhancement of SPR by CoQ10.
Fig. 1.
Effects of CoQ10 on SPR. The YB2/0 cell line was cultured in medium supplemented with CoQ10 (dissolved with Tween-80; closed circle) and with Tween-80 alone (control; open circle) in a 1 L reactor (ABLE, Tokyo, Japan). A Comparison of cell growth, B comparison of SPR. Two other independent experiments with similar conditions exhibited similar results
Fig. 2.
Effects of Q-Media (containing 500 μM CoQ10 in Hybridoma-SFM) on SPR. The YB2/0 cell line (expressing recombinant mouse/human chimeric IgG1 antibody) was cultured in Q-Media (closed circle) or in regular medium (open circle) in a 1 L bioreactor (ABLE, Tokyo, Japan). A Time course of 8-OHdG concentration, B comparison of cell growth, and C amount of MAb vs. cumulative cell density (its slope corresponds to SPR). Six other independent experiments with similar conditions exhibited similar results
CoQ10 did not affect oxidative response in fed-batch cultures
CoQ10 is known as a strong antioxidant, and 8-OHdG is known as a marker for oxidative DNA damage (Takane et al. 2002). We evaluated the change in 8-OHdG concentrations in fed-batch cultures to examine the antioxidant effects of Q-Media (containing 500 μM CoQ10). Although SPR was enhanced by the Q-Media, the 8-OHdG concentrations (and cumulative cell density) did not change (Fig. 2). To confirm that the enhancement of SPR by CoQ10 was not ascribed to the antioxidant effect of CoQ10, we analyzed the effects of other antioxidants. None of the following compounds, namely, glutathione (≤1,000 μM), alpha-tocopherol (≤5,000 μM), proanthocyanidin B2 (≤1,000 μM), uric acid (≤1,000 μM), ascorbic acid (≤1,000 μM), or 2-mercaptoethanol (≤5,000 μM), enhanced SPR as expected (data not shown).
Q-Media enhanced SPR for CHO and NS0 cell lines
To examine whether the enhancement of SPR by Q-Media is a general phenomenon, we analyzed the effects of Q-Media on two additional host cell lines commonly used in MAb production—CHO and NS0. SPR was also enhanced in CHO and NS0 cell lines by 28.8 and 31.9%, respectively, and in YB2/0 by 29.0%, which suggests that Q-Media is a versatile agent for enhancing SPR. Although a 20% increase in SPR may not seem notable, this degree of enhancement is most welcome. To illustrate the potential effectiveness of such enhancement, a 20% increase in SPR could reduce the annual cost ($65 million) of obtaining 250 kg of MAbs with a yield of 1,000 mg/L in a 10,000 L plant (Werner 2004) by $6 million. In fact, increases of 10%–20% in SPR of various MAbs have already been reported (Arden et al. 2007; Kim et al. 2005; Omasa et al. 2010).
Activities of MAbs were not changed by culturing in Q-Media
Because strict comparability to original MAbs is required when production conditions are changed, and because MAbs produced in YB2/0 cells have been known to exhibit high ADCC activities, we analyzed antigen binding and ADCC activities of MAbs expressed in YB2/0 cells that were cultured in Q-Media and regular media. MAbs produced in both media exhibited indistinguishable results for both activities (Fig. 3), which warrants the safe use of Q-Media for enhancing SPR.
Fig. 3.
Comparison of MAbs produced in Q-Media and regular medium. MAbs expressed by YB2/0 cells cultured in Q-Media and regular medium were separately purified and their activities to bind to their antigen (panel A) and kill their target cells (ADCC, panel B) were analyzed by ELISA and 51Cr-release assays, respectively
Conclusion
In this study, we found that supplementing culture media with CoQ10 widely enhances the production rate of MAbs in YB2/0, CHO, and NS0 cells without affecting cell growth or MAb activities such as antigen binding and ADCC. Although the mechanisms of enhancement are unclear, CoQ10 will be useful for the efficient production of pharmaceutically useful MAbs.
Acknowledgments
We would like to thank Dr. Kazuyasu Nakamura, Ms. Masako Wakitani, and Mr. Noriyuki Takahashi for their expert analysis, and Mr. Hiroshi Takasugi, Dr. Kazuhisa Uchida, Dr. Jun Yamaya, and Dr. Mitsuo Sato for helpful discussions and encouragement.
Abbreviations
- MAbs
Monoclonal antibodies
- CoQ10
Coenzyme-Q10
- Q-Media
Culture media supplemented with dispersed nanoparticles of Q10
- 8OHdG
8-hydroxy-2′-deoxyguanosine
- SPR
Specific MAb production rate (pg cell−1 d−1)
References
- Allen MJ, Boyce JP, Trentalange MT, et al. Identification of novel small molecule enhancers of protein production by cultured mammalian cells. Biotechnol Bioeng. 2008;100:1193–1204. doi: 10.1002/bit.21839. [DOI] [PubMed] [Google Scholar]
- Arden N, Ahn S-h, Vaz W, et al. Chemical caspase inhibitors enhance cell culture viabilities and protein titer. Biotechnol Prog. 2007;23:506–511. doi: 10.1021/bp060222m. [DOI] [PubMed] [Google Scholar]
- Backliwal G, Hildinger M, Kuettel I, et al. Valproic acid: a viable alternative to sodium butyrate for enhancing protein expression in mammalian cell cultures. Biotechnol Bioeng. 2008;101:182–189. doi: 10.1002/bit.21882. [DOI] [PubMed] [Google Scholar]
- Balcarcel RR, Stephanopoulos G. Rapamycin reduces hybridoma cell death and enhances monoclonal antibody production. Biotechnol Bioeng. 2001;76:1–10. doi: 10.1002/bit.1020. [DOI] [PubMed] [Google Scholar]
- Butler M, Huzel N, Barnabé N, et al. Linoleic acid improves the robustness of cells in agitated cultures. Cytotechnology. 1999;30:27–36. doi: 10.1023/A:1008048126055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Urlaub GCA, Chasin LA. Efficient cloning of single-copy genes using specialized cosmid vectors: isolation of mutant dihydrofolate reductase genes. Proc Natl Acad Sci USA. 1985;82:1189–1193. doi: 10.1073/pnas.82.4.1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carvalhal AV, Santos SS, Calado J, et al. Cell growth arrest by nucleotides, nucleosides and bases as a tool for improved production of recombinant proteins. Biotechnol Prog. 2003;19:69–83. doi: 10.1021/bp0255917. [DOI] [PubMed] [Google Scholar]
- Chello M, Mastroroberto P, Romano R, et al. Protection by coenzyme Q10 of tissue reperfusion injury during abdominal aortic cross-clamping. J Cardiovasc Surg (Torino) 1996;37:229–235. [PubMed] [Google Scholar]
- Coroadinha AS, Ribeiro J, Roldão A, et al. Effect of medium sugar source on the production of retroviral vectors for gene therapy. Biotechnol Bioeng. 2006;94:24–36. doi: 10.1002/bit.20778. [DOI] [PubMed] [Google Scholar]
- Leon Gatti M, Wlaschin KF, Nissom PM, et al. Comparative transcriptional analysis of mouse hybridoma and recombinant Chinese hamster ovary cells undergoing butyrate treatment. J Biosci Bioeng. 2007;103:82–91. doi: 10.1263/jbb.103.82. [DOI] [PubMed] [Google Scholar]
- Gorelick C, Lopez-Jones M, Goldberg GL, et al. Coenzyme Q10 and lipid-related gene induction in HeLa cells. Am J Obstet Gynecol. 2004;190:1432–1434. doi: 10.1016/j.ajog.2004.01.076. [DOI] [PubMed] [Google Scholar]
- Hathcock JN, Shao A. Risk assessment for coenzyme Q10 (Ubiquinone) Regul Toxicol Pharmacol. 2006;45:282–288. doi: 10.1016/j.yrtph.2006.05.006. [DOI] [PubMed] [Google Scholar]
- Hodgson JM, Watts GF, Playford DA, et al. Coenzyme Q10 improves blood pressure and glycaemic control: a controlled trial in subjects with type 2 diabetes. Eur J Clin Nutr. 2002;56:1137–1142. doi: 10.1038/sj.ejcn.1601464. [DOI] [PubMed] [Google Scholar]
- Inoue Y, Fujisawa M, Kawamoto S, et al. Effectiveness of vitamin A acetate for enhancing the production of lung cancer specific monoclonal antibodies. Cytotechnology. 1999;31:77–83. doi: 10.1023/A:1008016020785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inoue Y, Fujisawa M, Shoji M, et al. Enhanced antibody production of human-human hybridomas by retinoic acid. Cytotechnology. 2000;33:83–88. doi: 10.1023/A:1008155609072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang Z, Sharfstein ST. Sodium butyrate stimulates monoclonal antibody over-expression in CHO cells by improving gene accessibility. Biotechnol Bioeng. 2008;100:189–194. doi: 10.1002/bit.21726. [DOI] [PubMed] [Google Scholar]
- Kim D, Lee J, Chang H, et al. Effects of supplementation of various medium components on chinese hamster ovary cell cultures producing recombinant antibody. Cytotechnology. 2005;47:37–49. doi: 10.1007/s10616-005-3775-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitano M, Hosoe K, Fukutomi N, et al. 28-Day repeated dose toxicity study of dried microorganism in rats. Food Chem Toxicol. 2004;42:1817–1824. doi: 10.1016/j.fct.2004.06.013. [DOI] [PubMed] [Google Scholar]
- Kogan A, Syrkin AL, Drinitsina SV et al. (1999) The antioxidant protection of the heart by coenzyme Q10 in stable stenocardia of effort. Patol Fiziol Eksp Ter:16–19 [PubMed]
- Konno Y, Aoki M, Takasugi H et al. (2001) Process for producing substance. WO/2003/046174
- Konno Y, Sakai N, Sakai K et al. (2006) Method for production of substance. WO/2007/049567
- Ling WLW, Deng L, Lepore J, et al. Improvement of monoclonal antibody production in hybridoma cells by dimethyl sulfoxide. Biotechnol Prog. 2003;19:158–162. doi: 10.1021/bp020068d. [DOI] [PubMed] [Google Scholar]
- Liu C-H, Chen L-H. Enhanced recombinant M-CSF production in CHO cells by glycerol addition: model and validation. Cytotechnology. 2007;54:89–96. doi: 10.1007/s10616-007-9078-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mancuso MOD, Volpi L, Calsolaro V, Siciliano G. (2010) Coenzyme Q10 in neuromuscular and neurodegenerative disorders. Curr Drug Targets 11:111–121 [DOI] [PubMed]
- Martin-Lopez A, Garcia-Camacho F, Contreras-Gomez A, et al. Enhanced monoclonal antibody production in hybridoma cells by LPS and Anti-mIgG. Biotechnol Prog. 2007;23:1447–1453. doi: 10.1021/bp070191a. [DOI] [PubMed] [Google Scholar]
- Mimura Y, Lund J, Church S, et al. Butyrate increases production of human chimeric IgG in CHO-K1 cells whilst maintaining function and glycoform profile. J Immunol Methods. 2001;247:205–216. doi: 10.1016/S0022-1759(00)00308-2. [DOI] [PubMed] [Google Scholar]
- Miyazaki Y, Nishimoto S, Sasaki T, et al. Spermine enhances IgM productivity of human-human hybridoma HB4C5 cells and human peripheral blood lymphocytes. Cytotechnology. 1998;26:111–118. doi: 10.1023/A:1007939213564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Müller RH, Petersen RD, Hommoss A, et al. Nanostructured lipid carriers (NLC) in cosmetic dermal products. Adv Drug Deliv Rev. 2007;59:522–530. doi: 10.1016/j.addr.2007.04.012. [DOI] [PubMed] [Google Scholar]
- Nakamura K, Hanibuchi M, Yano S, et al. Apoptosis induction of human lung cancer cell line in multicellular heterospheroids with humanized antiganglioside GM2 monoclonal antibody. Cancer Res. 1999;59:5323–5330. [PubMed] [Google Scholar]
- Navas P, Fernandez-Ayala DM, Martin SF, et al. Ceramide-dependent caspase 3 activation is prevented by coenzyme Q from plasma membrane in serum-deprived cells. Free Radic Res. 2002;36:369–374. doi: 10.1080/10715760290021207. [DOI] [PubMed] [Google Scholar]
- Ochiai A, Itagaki S, Kurokawa T, et al. Improvement in intestinal coenzyme Q10 absorption by food intake. Yakugaku Zasshi. 2007;127:1251–1254. doi: 10.1248/yakushi.127.1251. [DOI] [PubMed] [Google Scholar]
- Ogawa T, Konno Y, Akashi N et al. (1999) Process for producing polypeptide. WO/2001/029246
- Oh SK, Vig P, Chua F, et al. Substantial overproduction of antibodies by applying osmotic pressure and sodium butyrate. Biotechnol Bioeng. 1993;42:601–610. doi: 10.1002/bit.260420508. [DOI] [PubMed] [Google Scholar]
- Oh HK, So MK, Yang J, et al. Effect of N-Acetylcystein on butyrate-treated chinese hamster ovary cells to improve the production of recombinant human interferon-beta-1a. Biotechnol Prog. 2005;21:1154–1164. doi: 10.1021/bp050057v. [DOI] [PubMed] [Google Scholar]
- 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;33:117–125. doi: 10.1007/s00449-009-0351-8. [DOI] [PubMed] [Google Scholar]
- Portakal O, Özkaya Ö, Erdeninal M, et al. Coenzyme Q10 concentrations and antioxidant status in tissues of breast cancer patients. Clin Biochem. 2000;33:279–284. doi: 10.1016/S0009-9120(00)00067-9. [DOI] [PubMed] [Google Scholar]
- Shields RL, Lai J, Keck R, et al. Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J Biol Chem. 2002;277:26733–26740. doi: 10.1074/jbc.M202069200. [DOI] [PubMed] [Google Scholar]
- Shinkawa T, Nakamura K, Yamane N, et al. The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J Biol Chem. 2003;278:3466–3473. doi: 10.1074/jbc.M210665200. [DOI] [PubMed] [Google Scholar]
- Shukla AA, Thömmes J. Recent advances in large-scale production of monoclonal antibodies and related proteins. Trends Biotechnol. 2010;28:253–261. doi: 10.1016/j.tibtech.2010.02.001. [DOI] [PubMed] [Google Scholar]
- Shults CW, Flint Beal M, Song D, et al. Pilot trial of high dosages of coenzyme Q10 in patients with Parkinson’s disease. Exp Neurol. 2004;188:491–494. doi: 10.1016/j.expneurol.2004.05.003. [DOI] [PubMed] [Google Scholar]
- Singh RB, Wander GS, Rastogi A, et al. Randomized, double-blind placebo-controlled trial of coenzyme Q10 in patients with acute myocardial infarction. Cardiovasc Drugs Ther. 1998;12:347–353. doi: 10.1023/A:1007764616025. [DOI] [PubMed] [Google Scholar]
- Soja AM, Mortensen SA. Treatment of congestive heart failure with coenzyme Q10 Illuminated by meta-analyses of clinical trials. Mol Aspects Med. 1997;18:159–168. doi: 10.1016/S0098-2997(97)00042-3. [DOI] [PubMed] [Google Scholar]
- Stojkovic M, Westesen K, Zakhartchenko V, et al. Coenzyme Q10 in submicron-sized dispersion improves development, hatching, cell proliferation, and adenosine triphosphate content of in vitro-produced bovine embryos. Biol Reprod. 1999;61:541–547. doi: 10.1095/biolreprod61.2.541. [DOI] [PubMed] [Google Scholar]
- Sun IL, Sun EE, Crane FL. Stimulation of serum-free cell proliferation by coenzyme Q. Biochem Biophys Res Commun. 1992;189:8–13. doi: 10.1016/0006-291X(92)91517-T. [DOI] [PubMed] [Google Scholar]
- Sun IL, Sun EE, Crane FL, et al. Requirement for coenzyme Q in plasma membrane electron transport. Proc Natl Acad Sci USA. 1992;89:11126–11130. doi: 10.1073/pnas.89.23.11126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun IL, Sun EE, Crane FL. Comparison of growth stimulation of HeLa cells, HL-60 cells, and mouse fibroblasts by coenzyme Q10. Protoplasma. 1995;184:214–219. doi: 10.1007/BF01276923. [DOI] [Google Scholar]
- Takane M, Sugano N, Iwasaki H, et al. New biomarker evidence of oxidative DNA damage in whole saliva from clinically healthy and periodontally diseased individuals. J Periodontol. 2002;73:551–554. doi: 10.1902/jop.2002.73.5.551. [DOI] [PubMed] [Google Scholar]
- Takenouchi S, Sugahara T. Lactate dehydrogenase enhances immunoglobulin production by human hybridoma and human peripheral blood lymphocytes. Cytotechnology. 2003;42:133–143. doi: 10.1023/B:CYTO.0000015838.06536.de. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terada S, Nishimura T, Sasaki M, et al. Sericin, a protein derived from silkworms, accelerates the proliferation of several mammalian cell lines including a hybridoma. Cytotechnology. 2002;40:3–12. doi: 10.1023/A:1023993400608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsao Y-S, Condon R, Schaefer E, et al. Development and improvement of a serum-free suspension process for the production of recombinant adenoviral vectors using HEK293 cells. Cytotechnology. 2001;37:189–198. doi: 10.1023/A:1020555310558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turunen M, Olsson J, Dallner G. Metabolism and function of coenzyme Q. Biochimica et Biophysica Acta (BBA) Biomemb. 2004;1660:171–199. doi: 10.1016/j.bbamem.2003.11.012. [DOI] [PubMed] [Google Scholar]
- Walsh G. Biopharmaceutical benchmarks 2006. Nat Biotech. 2006;24:769–776. doi: 10.1038/nbt0706-769. [DOI] [PubMed] [Google Scholar]
- Werner RG. Economic aspects of commercial manufacture of biopharmaceuticals. J Biotechnol. 2004;113:171–182. doi: 10.1016/j.jbiotec.2004.04.036. [DOI] [PubMed] [Google Scholar]
- Wurm FM. Production of recombinant protein therapeutics in cultivated mammalian cells. Nat Biotechnol. 2004;22:1393–1398. doi: 10.1038/nbt1026. [DOI] [PubMed] [Google Scholar]
- Yegorov Y, Moldaver M, Vishnyakova K, et al. Enhanced control of proliferation in telomerized cells. Russ J Develop Biol. 2007;38:76–89. doi: 10.1134/S106236040702004X. [DOI] [PMC free article] [PubMed] [Google Scholar]






