Acessibilidade / Reportar erro

Production of Lentiviral Vectors Encoding Recombinant Factor VIII Expression in Serum-Free Suspension Cultures

ABSTRACT

Lentiviral vector-mediated gene transfer offers several advantages over other gene delivery vectors when considering gene and cell therapy applications. However, using these therapies in clinical applications involves large-scale vector production in an efficient and cost-effective manner. Here we describe a high yield production of a lentivirus encoding recombinant factor VIII in a scalable and GMP-compliant culture system, based on serum free suspension cultures and transient transfection with an inexpensive reagent, polyethylenimine (PEI), reaching a total viral yield of 2.48x108 particles.

Key words:
Factor VIII; Hek293 cells; lentiviral vectors; polyethylenimine; serum-free suspension culture; transient transfection

INTRODUCTION

Hemophilia A is a bleeding disorder caused by coagulation factor VIII (FVIII) deficiency. Currently, the treatment of choice for hemophilia A is replacement therapy with plasma-derived or recombinant FVIII. Although this is an efficient treatment, there are some factors that make it an inconvenient therapy, such as the short period of FVIII activity in plasma. The treatment is a large burden for patients that require a frequent number of infusions. Furthermore, Hemophilia A is an ideal candidate for gene therapy since the defect is attributable to the lack of a single gene product (FVIII) that normally circulates in low amounts in plasma (200 ng/mL). In addition, the blood concentration of factor VIII obtained is not critical, once a small increase in plasma concentration can convert a severe case of hemophilia into a moderate case, improving the quality of life.

The most promising vectors for gene therapy concerning hemophilia A are the lentiviral and adeno- associated virus (AAV) vectors (Chuah et al. 2013Chuah MK, Evens H, VandenDriessche T. Gene therapy for hemophilia. J Thromb Haemost. 2013; 11 Suppl 1:99-110.). Lentivirus vectors (LV) have gained most attention over the last 20 years due to their ability to transduce non-dividing cells and large packaging capacity (Quinonez and Sutton 2002Quinonez R, Sutton RE. Lentiviral vectors for gene delivery into cells. DNA Cell Biol. 2002; 21(12):937-951.). Nevertheless, recent studies have shown that they have much lower oncogenic potential than other retroviruses because LV do not integrate with high frequency near promoters of proto-oncogenes and genes that control cell proliferation. Lentiviral gene therapy for hemophilia A has been successfully used in several pre-clinical studies (Liras et al. 2012Liras A, Segovia C, Gabán AS. Advanced therapies for the treatment of hemophilia: future perspectives. Orphanet J Rare Dis. 2012; 7(1):97.; Chuah et al. 2013Chuah MK, Evens H, VandenDriessche T. Gene therapy for hemophilia. J Thromb Haemost. 2013; 11 Suppl 1:99-110.; High et al. 2014High KH, Nathwani A, Spencer T, Lillicrap D. Current status of haemophilia gene therapy. Haemophilia. 2014; 20 Suppl 4:43-49.) and is close to clinical approval. Furthermore, it has been favorably reviewed by the US Recombinant DNA Advisory Committee, as well as by the US Food and Drug Administration (FDA) (High et al. 2014High KH, Nathwani A, Spencer T, Lillicrap D. Current status of haemophilia gene therapy. Haemophilia. 2014; 20 Suppl 4:43-49.).

To enable the clinical implementation of these encouraging studies, it has become necessary to develop efficient and scalable production processes for such vectors. Most studies focus on the generation of an optimized lentiviral vector encoding a high-expression of factor VIII. To the best of our knowledge, there is no study reporting the development of the production process for lentiviral vectors for hemophilia A, which is of paramount importance for the success of this therapy.

Traditionally, lentiviral vector productions for research and clinical purposes are performed by transfection of adherently growing cells, especially 293 cells, in staggered production systems. To maximize the cell culture surface, equipment such as Cell Factories (Thermo Scientific) or HYPERFlasks (Corning) are favored. The scale-up of this type of production is costly, labor-intensive and limited by the largest available scale (Schweizer and Merten 2010Schweizer M, Merten O-W. Large-scale production means for the manufacturing of lentiviral vectors. Curr Gene Ther. 2010; 10(6):474-486.). One of the main obstacles for LV production is its low-titer, specially encoding a large size protein such as FVIII, and low half-life. Average titers of crude LV stocks range between 105 to 107 transducing units (tu)/mL (Mochizuki et al. 1998Mochizuki H, Schwartz JP, Tanaka K, Brady RO, Reiser J. High-titer human immunodeficiency virus type 1- based vector systems for gene delivery into nondividing cells. J Virol. 1998; 72(11):8873-8883.; Ansorge et al. 2009Ansorge S, Lanthier S, Transfiguracion J, Durocher Y, Henry O, Kamen A. Development of a scalable process for high-yield lentiviral vector production by transient transfection of HEK293 suspension cultures. J Gene Med. 2009; 11(10):868-876.). These titers can be increased by centrifugation to generate 109 tu/mL. However, these concentrations remain too low for gene therapy clinical trials, where the number of functional vector particles needed is at least 1011-1012particles per patient (MacGregor 2001MacGregor RR. Clinical protocol. A phase 1 open-label clinical trial of the safety and tolerability of single escalating doses of autologous CD4 T cells transduced with VRX496 in HIV-positive subjects. Hum Gene Ther. 2001; 12(16):2028-9.).

Currently, the most successful alternative for large scale LV production is based on transient transfection in serum-free suspension cultures using polyethylenimine (PEI) (Ansorge et al. 2009Ansorge S, Lanthier S, Transfiguracion J, Durocher Y, Henry O, Kamen A. Development of a scalable process for high-yield lentiviral vector production by transient transfection of HEK293 suspension cultures. J Gene Med. 2009; 11(10):868-876.; Schweizer and Merten 2010Schweizer M, Merten O-W. Large-scale production means for the manufacturing of lentiviral vectors. Curr Gene Ther. 2010; 10(6):474-486.). PEI-mediated transfections have been reported to be more consistent, reproducible and productive (Pham et al. 2006Pham PL, Kamen A, Durocher Y. Large-scale transfection of mammalian cells for the fast production of recombinant protein. Mol Biotechnol. 2006; 34(2):225-237.; Kuroda et al. 2009Kuroda H, Kutner RH, Bazan NG, Reiser J. Simplified lentivirus vector production in protein-free media using polyethylenimine-mediated transfection. J Virol Methods. 2009; 157(2):113-121.; Toledo et al. 2009Toledo JR, Prieto Y, Oramas N, Sánchez O. Polyethylenimine-based transfection method as a simple and effective way to produce recombinant lentiviral vectors. Appl Biochem Biotechnol. 2009; 157(3):538-544.). Moreover, PEI is chemically stable, inexpensive and has a broad pH range for transfection (Boussif et al. 1995Boussif O, Lezoualc'h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci U S A. 1995; 92(16):7297-7301.). According to Ansorge et al. (2009), PEI-mediated transient transfection allows for the production of LV in suspension culture without the need of medium exchange prior to or after transfection, making the process scalable (Segura et al. 2007Segura MM, Garnier A, Durocher Y, Coelho H, Kamen A. Production of lentiviral vectors by large-scale transient transfection of suspension cultures and affinity chromatography purification. Biotechnol Bioeng. 2007; 98(4):789-799.).

Here we report on the production of a third generation LV encoding recombinant factor VIII expression by PEI-mediated transfection in a serum free suspension culture using HEK293SF-3F6 cells. We show that our production process can generate a total of 2.48 x 108 functional virus particles (2.48 x 108 tu in a total volume of 100 mL) on a scalable and GMP-compliant platform.

The cultures were performed using the Hek293SF-3F6 cell line (developed by the National Research Council, Canada). This cell line was adapted to a serum-free condition with high specific growth rate and cell density (Côté et al. 1998Côté J, Garnier A, Massie B, Kamen A. Serum-free production of recombinant proteins and adenoviral vectors by 293SF-3F6 cells. Biotechnol Bioeng. 1998; 59(5):567-575.). The cells were cultured at 37°C and 5% CO2 in the serum free commercial medium HyQSFM4TransFx293 (Hyclone, Logan, UT), specially developed for the transfection of Hek293SF-3F6 cells, in 125 mL Erlenmeyer flasks (Corning) at 120 rpm. Medium supplementation was evaluated with 5% of Cell Boost 5 (CB5 5%, Hyclone, Logan, UT, USA), which contains nutrients such as lipids, amino acids, vitamins, and growth factors. Cell growth and viability were determined by the dye exclusion test with erythrosine B in hemocytometer.

To produce lentiviral particles, containing the FVIII gene, we used a third generation vector cPPT- C(FVIIIdelB)IGWS (p1054) encoding a B-domain deleted FVIII and an EGFP separated by IRES element, under control of the CMV promoter (Picanço et al. 2007Picanço V, Heinz S, Bott D, Behrmann M, Covas DT, Seifried E, et al. Recombinant expression of coagulation factor VIII in hepatic and non-hepatic cell lines stably transduced with third generation lentiviral vectors comprising the minimal factor VIII promoter. Cytotherapy. 2007; 9(8):785-794.). We also used the packing plasmid pCMVΔ8.91 (encoding gag-pol, rev and tat genes from HIV-1) and the vector pMD2.VSVG encoding the vesicular stomatitis virus G (VSV-G). Plasmids were produced with an anion exchange purification method or with the commercially available kits QIAGEN Plasmid Giga Kit (Qiagen, Mississauga, ON).

In the transient transfection protocol, we used 250 mL Erlenmeyer flasks (Corning) with 50 mL of culture medium and PEI reagent (25-kDa linear PEI, Polysciences), as previously described by Ansorge et al. (2009Ansorge S, Lanthier S, Transfiguracion J, Durocher Y, Henry O, Kamen A. Development of a scalable process for high-yield lentiviral vector production by transient transfection of HEK293 suspension cultures. J Gene Med. 2009; 11(10):868-876.). The experiments for the production of lentiviral vector particles encoding FVIII expression (cPPT-C(FVIIIdelB)IGWS) were performed in Hek293SF-3F6 cells at the concentration of 5 x 106 cells/mL and DNA:PEI ratio of 1:2. A plasmid mass ratio of 1:1:2 and 1:2:4 (pMD2.VSVG:pCMVΔ8.91:p1054) was used to prepare the DNA-PEI complexes in a volume corresponding to 10% of the total culture volume. The mixture was incubated for 15 min at room temperature prior to adding the cell culture. A few hours before transfection, the cell suspension was centrifuged (300 x g for 5 min) and resuspended at 5 x 106 cells/mL. Sodium butyrate 5 mM, dissolved in 1M n-butyric acid (Sigma B-2503) and neutralized with 10 MNaOH, was added to the medium after the transfection in predetermined times. The medium was totally exchanged every day in order to simulate a perfusion culture and enhance the volumetric production. Every 24 h, the cell suspension was centrifuged (300 xg, 5min) and resuspended in 20 mL of fresh medium. A volume of 5 mL of metabolized supernatant was filtered in 0.45 um HT Tuffryn (Pall, Ann Arbor, MI) membranes, to remove cell debris, and stocked at - 80°C for lentiviral quantification. The culture was maintained for 4 days.

The viral titer was determined by flow cytometry-based methodology. We used Hek293E (clone 6E) cultivated in Freestyle 17 fresh medium (Invitrogen). The transduction and virus titer followed the methodology proposed by Segura et al. (2007Segura MM, Garnier A, Durocher Y, Coelho H, Kamen A. Production of lentiviral vectors by large-scale transient transfection of suspension cultures and affinity chromatography purification. Biotechnol Bioeng. 2007; 98(4):789-799.).

Effect of medium supplementation on FVIII lentiviral vector production

Transfection efficiency is dependent on the medium formulation, the presence of nutrients and additives (Pham et al. 2003Pham PL, Perret S, Doan HC, Cass B, St-Laurent G, Kamen A, et al. Large-scale transient transfection of serum-free suspension-growing HEK293 EBNA1 cells: peptone additives improve cell growth and transfection efficiency. Biotechnol Bioeng. 2003; 84(3):332-342.; Pham et al. 2005; Pham et al. 2006). Thus, we analyzed the lentiviral production profile using HYQSFM4 (HyQ) culture medium supplemented or not with Cell Boost 5 (CB5). Although there was no difference in cell viability, cell growth was higher in the supplemented medium during the lentiviral production, with a maximum cell density (Xmax) of 7.95 x 106 cells/mL (Fig. 1A). Therefore, the FVIII-LV production was also higher; reaching 5.94 x 106 tu/mL in CB5 supplemented cultures compared to control condition 3.58 x 106 tu/mL at 48 h post transfection (Fig. 1B). The effect of adding CB5 to the FVIII-LV production is better observed when considering the cumulative production rather than the volumetric daily production (Fig. 1B). In the CB5 supplemented medium, there was a total of 1.528 x 108 tu after 96 h.

Figure 1
Effect of CB5 supplementation in cell density (A), FVIII-LV volumetric (Bars) and cumulative (Curves) production (B).

Effect of DNA concentration on FVIII lentiviral vector production

In order to enhance lentiviral production, we performed a second experiment, evaluating different DNA concentrations: 0.4 µg/106 cells and 0.6 µg/106 cells. The use of 0.6 µg per 106 cells resulted in smaller cell densities (Fig. 2A) and cumulative LV-productivity of less than 5 x 107tu (Fig. 2B).

Although cell concentration also declined after 48 h in the 0.4 μg per 106cells protocol (Fig. 2A), the volumetric LV-production was significantly higher at 48 and 72 h and the cumulative production reached 1.1 x 108 transducing units after 96 h (Fig. 2B).

Figure 2
Effect of DNA concentration in cell density (A), FVIII-LV volumetric (Bars) and cumulative (Curves) production (B).

A number of studies have reported optimal efficiency of transfection using DNA concentration in the range of 0.4-0.6 μg per 106 cells in PEI-mediated protocols (Sun et al. 2008Sun X, Hia HC, Goh PE, Yap MGS. High-density transient gene expression in suspension-adapted 293 EBNA1 cells. Biotechnol Bioeng. 2008; 99(1):108-116; Kuroda et al. 2009Kuroda H, Kutner RH, Bazan NG, Reiser J. Simplified lentivirus vector production in protein-free media using polyethylenimine-mediated transfection. J Virol Methods. 2009; 157(2):113-121.). Besides, PEI is known to have a toxic effect (Godbey and Mikos 2001Godbey WT, Mikos AG. Recent progress in gene delivery using non-viral transfer complexes. J Control Release. 2001; 72(1-3):115-125.;Kunath et al. 2003Kunath K, von Harpe A, Fischer D, Petersen H, Bickel U, Voigt K, et al. Low-molecular-weight polyethylenimine as a non-viral vector for DNA delivery: comparison of physicochemical properties, transfection efficiency and in vivo distribution with high-molecular-weight polyethylenimine. J Control Release. 2003; 89(1):113-125.; Sun et al. 2008Sun X, Hia HC, Goh PE, Yap MGS. High-density transient gene expression in suspension-adapted 293 EBNA1 cells. Biotechnol Bioeng. 2008; 99(1):108-116) and the concentration of PEI: DNA complex (polyplex) may influence the efficiency of the transient transfection. In the study described here, the highest polyplex amount resulted in higher cell death and significantly lower LV production.

Effect of sodium butyrate on FVIII lentiviral vector production

Our results showed that NaBu addition inhibits cell growth. When NaBu was added 3 h post- tranfection (hpt), Xmax was 9 x 106cells/mL, declining after 48 h. However, when the addition of NaBu occurred after 16 h, Xmax was 7 x 106cells/mL, declining only after 24 h (Fig. 3A). Although a variety of studies use sodium butyrate after 16 h post-transfection (Karolewski et al. 2003Karolewski BA, Watson DJ, Parente MK, Wolfe JH. Comparison of transfection conditions for a lentivirus vector produced in large volumes. Hum Gene Ther. 2003; 14(14):1287-1296.), in our study the cumulative productivity was lower in comparison with the control experiment. However, adding NaBu after 3 h resulted in significantly higher titers than both the control and the 16-h protocol. At 48 hpt, we obtained a volumetric production of 8.23 x 106tu/mL (Fig. 3B). Moreover, the cumulative production reached 2.48 x 108tu, compared to 1.1 x 108 tu obtained with the 16-hpt protocol (Fig. 3B).

Figure 3
Effect of sodium butyrate on cell density (A), FVIII-LV volumetric (Bars) and cumulative (Curves) production (B).

Sodium butyrate (NaBu) was successfully used to enhance recombinant protein (Damiani et al. 2013Damiani R, Almeida BE, Oliveira JE, Bartolini P, Ribela MTCP. Enhancement of human thyrotropin synthesis by sodium butyrate addition to serum-free CHO cell culture. Appl Biochem Biotechnol. 2013; 171(7):1658-1672.; Lee et al. 2014Lee SM, Kim Y-G, Lee EG, Lee GM. Digital mRNA profiling of N-glycosylation gene expression in recombinant Chinese hamster ovary cells treated with sodium butyrate. J Biotechnol. 2014; 171:56-60.) and lentivirus and retrovirus production (Karolewski et al. 2003Karolewski BA, Watson DJ, Parente MK, Wolfe JH. Comparison of transfection conditions for a lentivirus vector produced in large volumes. Hum Gene Ther. 2003; 14(14):1287-1296.; Merten 2004Merten O-W. State-of-the-art of the production of retroviral vectors. J Gene Med. 2004; 6 Suppl 1:S105-24.). Transcriptional silencing of the LV-related transfected genes was demonstrated as a drawback in LV production ( Kafri et al. 1999Kafri T, van Praag H, Ouyang L, Gage FH, Verma IM. A packaging cell line for lentivirus vectors. J Virol. 1999; 73(1):576-584.; Jaalouk et al. 2000Jaalouk DE, Eliopoulos N, Couture C, Mader S, Galipeau J. Glucocorticoid-inducible retrovector for regulated transgene expression in genetically engineered bone marrow stromal cells. Hum Gene Ther. 2000; 11(13):1837-1849.; Ni et al. 2005Ni Y, Sun S, Oparaocha I, Humeau L, Davis B, Cohen R, et al. Generation of a packaging cell line for prolonged large-scale production of high-titer HIV-1-based lentiviral vector. J Gene Med. 2005; 7(6):818-834.). Therefore, the addition of sodium butyrate, at a concentration range of 2-20 mM, is reported to increase LV productivity (Soneoka et al. 1995Soneoka Y, Cannon PM, Ramsdale EE, Griffiths JC, Romano G, Kingsman SM, et al. A transient three-plasmid expression system for the production of high titer retroviral vectors. Nucleic Acids Res. 1995; 23(4):628--633.; Sakoda et al. 1999Sakoda T, Kasahara N, Hamamori Y, Kedes L. A high-titer lentiviral production system mediates efficient transduction of differentiated cells including beating cardiac myocytes. J Mol Cell Cardiol. 1999; 31(11):2037-2047.; Jaalouk et al. 2000; Karolewski et al. 2003; Merten 2004; Sena-Esteves et al. 2004Sena-Esteves M, Tebbets JC, Steffens S, Crombleholme T, Flake AW. Optimized large-scale production of high titer lentivirus vector pseudotypes. J Virol Methods. 2004; 122(2):131-139.) by enhancing transfected gene expression through histone hyperacetylation (Altenburg et al. 1976Altenburg BC, Via DP, Steiner SH. Modification of the phenotype of murine sarcoma virus-transformed cells by sodium butyrate. Effects on morphology and cytoskeletal elements. Exp Cell Res. 1976;102(2):223-231.; Kruh 1982Kruh J. Effects of sodium butyrate, a new pharmacological agent, on cells in culture. Mol Cell Biochem. 1982; 42(2):65-82.; Gloger et al. 1985Gloger I, Arad G, Panet A. Regulation of Moloney murine leukemia virus replication in chronically infected cells arrested at the G0/G1 phase. J Virol. 1985; 54(3):844-850.).

By investigating the effect of medium supplementation, total DNA mass and different times for sodium butyrate addition, we were able to obtain high vector titers up to 8.23 x 106tu/mL. This production level is significantly higher than the one obtained in other studies reporting LV carrying FVIII cDNA. Radcliffe and colleagues (2008Radcliffe PA, Sion CJM, Wilkes FJ, Custard EJ, Beard GL, Kingsman SM, et al. Analysis of factor VIII mediated suppression of lentiviral vector titres. Gene Ther. 2008; 15(4):289-297.), using 293T cells in static culture to produce LV carrying a B-domain deleted FVIII cDNA, were able to reach a titer of approximately1 x 106tu/mL (Radcliffe et al. 2008). The levels reported here can also be considered relevant considering the size of the FVIII insertion (9.3 kb). Yacoub and colleagues (2007) showed that the production of lentivirus with 7.2-7.5kb insert size resulted in titers of 1-5.105tu/mL.

In this study, we successfully described a scalable and GMP-compliant protocol for the production of lentiviral vectors encoding the coagulation factor VIII cDNA by PEI-mediated transient transfection in serum-free suspension culture. This protocol can be easily scaled-up in perfusion bioreactors to produce vectors at sufficient quantities for clinical studies. Moreover, it is cost-effective and less laborious than the traditional production platform based on adherent cells growing in cell factories.

This protocol can also be used to produce sufficient LV to transduce suspension serum-free adapted mammalian cells to enable the generation of a recombinant cell line producing FVIII. This methodology eliminates the time-consuming and laborious steps of adapting a FVIII producing cell line, which is normally generated with adherent cells growing in bovine fetal serum, and reduces the risk of decreasing or losing protein expression levels during the adaptation protocol.

ACKNOWLEDGEMENTS

The authors acknowledge the financial support from FAPESP (Process number 2012/04629-8), CTC- Center of Cell Therapy no 2013/08135-2 and a scholarship from CAPES.

REFERENCES

  • Altenburg BC, Via DP, Steiner SH. Modification of the phenotype of murine sarcoma virus-transformed cells by sodium butyrate. Effects on morphology and cytoskeletal elements. Exp Cell Res. 1976;102(2):223-231.
  • Ansorge S, Lanthier S, Transfiguracion J, Durocher Y, Henry O, Kamen A. Development of a scalable process for high-yield lentiviral vector production by transient transfection of HEK293 suspension cultures. J Gene Med. 2009; 11(10):868-876.
  • Boussif O, Lezoualc'h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci U S A. 1995; 92(16):7297-7301.
  • Chuah MK, Evens H, VandenDriessche T. Gene therapy for hemophilia. J Thromb Haemost. 2013; 11 Suppl 1:99-110.
  • Côté J, Garnier A, Massie B, Kamen A. Serum-free production of recombinant proteins and adenoviral vectors by 293SF-3F6 cells. Biotechnol Bioeng. 1998; 59(5):567-575.
  • Damiani R, Almeida BE, Oliveira JE, Bartolini P, Ribela MTCP. Enhancement of human thyrotropin synthesis by sodium butyrate addition to serum-free CHO cell culture. Appl Biochem Biotechnol. 2013; 171(7):1658-1672.
  • Gloger I, Arad G, Panet A. Regulation of Moloney murine leukemia virus replication in chronically infected cells arrested at the G0/G1 phase. J Virol. 1985; 54(3):844-850.
  • Godbey WT, Mikos AG. Recent progress in gene delivery using non-viral transfer complexes. J Control Release. 2001; 72(1-3):115-125.
  • High KH, Nathwani A, Spencer T, Lillicrap D. Current status of haemophilia gene therapy. Haemophilia. 2014; 20 Suppl 4:43-49.
  • Jaalouk DE, Eliopoulos N, Couture C, Mader S, Galipeau J. Glucocorticoid-inducible retrovector for regulated transgene expression in genetically engineered bone marrow stromal cells. Hum Gene Ther. 2000; 11(13):1837-1849.
  • Kafri T, van Praag H, Ouyang L, Gage FH, Verma IM. A packaging cell line for lentivirus vectors. J Virol. 1999; 73(1):576-584.
  • Karolewski BA, Watson DJ, Parente MK, Wolfe JH. Comparison of transfection conditions for a lentivirus vector produced in large volumes. Hum Gene Ther. 2003; 14(14):1287-1296.
  • Kruh J. Effects of sodium butyrate, a new pharmacological agent, on cells in culture. Mol Cell Biochem. 1982; 42(2):65-82.
  • Kunath K, von Harpe A, Fischer D, Petersen H, Bickel U, Voigt K, et al. Low-molecular-weight polyethylenimine as a non-viral vector for DNA delivery: comparison of physicochemical properties, transfection efficiency and in vivo distribution with high-molecular-weight polyethylenimine. J Control Release. 2003; 89(1):113-125.
  • Kuroda H, Kutner RH, Bazan NG, Reiser J. Simplified lentivirus vector production in protein-free media using polyethylenimine-mediated transfection. J Virol Methods. 2009; 157(2):113-121.
  • Lee SM, Kim Y-G, Lee EG, Lee GM. Digital mRNA profiling of N-glycosylation gene expression in recombinant Chinese hamster ovary cells treated with sodium butyrate. J Biotechnol. 2014; 171:56-60.
  • Liras A, Segovia C, Gabán AS. Advanced therapies for the treatment of hemophilia: future perspectives. Orphanet J Rare Dis. 2012; 7(1):97.
  • MacGregor RR. Clinical protocol. A phase 1 open-label clinical trial of the safety and tolerability of single escalating doses of autologous CD4 T cells transduced with VRX496 in HIV-positive subjects. Hum Gene Ther. 2001; 12(16):2028-9.
  • Merten O-W. State-of-the-art of the production of retroviral vectors. J Gene Med. 2004; 6 Suppl 1:S105-24.
  • Mochizuki H, Schwartz JP, Tanaka K, Brady RO, Reiser J. High-titer human immunodeficiency virus type 1- based vector systems for gene delivery into nondividing cells. J Virol. 1998; 72(11):8873-8883.
  • Ni Y, Sun S, Oparaocha I, Humeau L, Davis B, Cohen R, et al. Generation of a packaging cell line for prolonged large-scale production of high-titer HIV-1-based lentiviral vector. J Gene Med. 2005; 7(6):818-834.
  • Pham PL, Kamen A, Durocher Y. Large-scale transfection of mammalian cells for the fast production of recombinant protein. Mol Biotechnol. 2006; 34(2):225-237.
  • Pham PL, Perret S, Cass B, Carpentier E, St-Laurent G, Bisson L, et al. Transient gene expression in HEK293 cells: peptone addition posttransfection improves recombinant protein synthesis. Biotechnol Bioeng. 2005; 90(3):332-344.
  • Pham PL, Perret S, Doan HC, Cass B, St-Laurent G, Kamen A, et al. Large-scale transient transfection of serum-free suspension-growing HEK293 EBNA1 cells: peptone additives improve cell growth and transfection efficiency. Biotechnol Bioeng. 2003; 84(3):332-342.
  • Picanço V, Heinz S, Bott D, Behrmann M, Covas DT, Seifried E, et al. Recombinant expression of coagulation factor VIII in hepatic and non-hepatic cell lines stably transduced with third generation lentiviral vectors comprising the minimal factor VIII promoter. Cytotherapy. 2007; 9(8):785-794.
  • Quinonez R, Sutton RE. Lentiviral vectors for gene delivery into cells. DNA Cell Biol. 2002; 21(12):937-951.
  • Radcliffe PA, Sion CJM, Wilkes FJ, Custard EJ, Beard GL, Kingsman SM, et al. Analysis of factor VIII mediated suppression of lentiviral vector titres. Gene Ther. 2008; 15(4):289-297.
  • Sakoda T, Kasahara N, Hamamori Y, Kedes L. A high-titer lentiviral production system mediates efficient transduction of differentiated cells including beating cardiac myocytes. J Mol Cell Cardiol. 1999; 31(11):2037-2047.
  • Schweizer M, Merten O-W. Large-scale production means for the manufacturing of lentiviral vectors. Curr Gene Ther. 2010; 10(6):474-486.
  • Segura MM, Garnier A, Durocher Y, Coelho H, Kamen A. Production of lentiviral vectors by large-scale transient transfection of suspension cultures and affinity chromatography purification. Biotechnol Bioeng. 2007; 98(4):789-799.
  • Sena-Esteves M, Tebbets JC, Steffens S, Crombleholme T, Flake AW. Optimized large-scale production of high titer lentivirus vector pseudotypes. J Virol Methods. 2004; 122(2):131-139.
  • Soneoka Y, Cannon PM, Ramsdale EE, Griffiths JC, Romano G, Kingsman SM, et al. A transient three-plasmid expression system for the production of high titer retroviral vectors. Nucleic Acids Res. 1995; 23(4):628--633.
  • Sun X, Hia HC, Goh PE, Yap MGS. High-density transient gene expression in suspension-adapted 293 EBNA1 cells. Biotechnol Bioeng. 2008; 99(1):108-116
  • Toledo JR, Prieto Y, Oramas N, Sánchez O. Polyethylenimine-based transfection method as a simple and effective way to produce recombinant lentiviral vectors. Appl Biochem Biotechnol. 2009; 157(3):538-544.

Publication Dates

  • Publication in this collection
    Dec 2015

History

  • Received
    26 June 2015
  • Accepted
    23 Aug 2015
Instituto de Tecnologia do Paraná - Tecpar Rua Prof. Algacyr Munhoz Mader, 3775 - CIC, 81350-010 Curitiba PR Brazil, Tel.: +55 41 3316-3052/3054, Fax: +55 41 3346-2872 - Curitiba - PR - Brazil
E-mail: babt@tecpar.br