Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
BMC Biotechnol ; 11: 84, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21884612

RESUMEN

BACKGROUND: Cell culture-based production of influenza vaccine remains an attractive alternative to egg-based production. Short response time and high production yields are the key success factors for the broader adoption of cell culture technology for industrial manufacturing of pandemic and seasonal influenza vaccines. Recently, HEK293SF cells have been successfully used to produce influenza viruses, achieving hemagglutinin (HA) and infectious viral particle (IVP) titers in the highest ranges reported to date. In the same study, it was suggested that beyond 4 × 10(6) cells/mL, viral production was limited by a lack of nutrients or an accumulation of toxic products. RESULTS: To further improve viral titers at high cell densities, perfusion culture mode was evaluated. Productivities of both perfusion and batch culture modes were compared at an infection cell density of 6 × 10(6) cells/mL. The metabolism, including glycolysis, glutaminolysis and amino acids utilization as well as physiological indicators such as viability and apoptosis were extensively documented for the two modes of culture before and after viral infection to identify potential metabolic limitations. A 3 L bioreactor with a perfusion rate of 0.5 vol/day allowed us to reach maximal titers of 3.3 × 10(11) IVP/mL and 4.0 logHA units/mL, corresponding to a total production of 1.0 × 10(15) IVP and 7.8 logHA units after 3 days post-infection. Overall, perfusion mode titers were higher by almost one order of magnitude over the batch culture mode of production. This improvement was associated with an activation of the cell metabolism as seen by a 1.5-fold and 4-fold higher consumption rates of glucose and glutamine respectively. A shift in the viral production kinetics was also observed leading to an accumulation of more viable cells with a higher specific production and causing an increase in the total volumetric production of infectious influenza particles. CONCLUSIONS: These results confirm that the HEK293SF cell is an excellent substrate for high yield production of influenza virus. Furthermore, there is great potential in further improving the production yields through better control of the cell culture environment and viral production kinetics. Once accomplished, this cell line can be promoted as an industrial platform for cost-effective manufacturing of the influenza seasonal vaccine as well as for periods of peak demand during pandemics.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células HEK293/virología , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Virión/metabolismo , Cultivo de Virus/métodos , Aminoácidos/metabolismo , Animales , Apoptosis , Reactores Biológicos/virología , Línea Celular , Proliferación Celular , Supervivencia Celular , Perros , Glucólisis , Células HEK293/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A/química , Cinética , Perfusión/métodos , Temperatura , Virión/química
2.
J Virol Methods ; 148(1-2): 106-14, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18079009

RESUMEN

Helper-dependent adenovirus (HDAd), deleted in all viral protein-coding sequences has been designed to reduce immune response and favor long-term expression of therapeutic genes in clinical programs. Its production requires co-infection of E1-complementing cells with helper adenovirus (HAd). Significant progresses have been made in the molecular design of HDAd, but large scale production remains a challenge. In this work, a scalable system for HDAd production is designed and evaluated focusing on the co-infection step. A human embryo kidney 293 (293) derived cell line, the 293SF/FLPe was generated to produce efficiently HDAd while restricting the packaging of HAd. This cell line was adapted to grow in suspension and in serum-free medium. Multiplicity of infection (MOI) of HDAd ranging from 0.1 to 50 was evaluated in presence of HAd at a MOI of 5. Optimal MOIs for HDAd amplification were found in the range of 5-10. HAd contamination was only 1%. These results were validated in a 3 L bioreactor under controlled operating conditions where a higher HDAd yield of 2.6 x 10(9) viral particles (VP)/mL or 3.5 x 10(8) infectious units (IU)/mL of HDAd was obtained.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Cultivo de Virus/métodos , Proteínas E1 de Adenovirus/genética , Recuento de Células , Línea Celular , Supervivencia Celular , Medio de Cultivo Libre de Suero , Vectores Genéticos , Virus Helper/fisiología , Humanos , Transducción Genética
3.
J Virol Methods ; 142(1-2): 21-8, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17306891

RESUMEN

Recombinant baculoviruses carrying mammalian expression cassettes or "BacMam" are promising gene delivery vehicles shown to transduce mammalian cells efficiently both in vitro and in vivo. These viruses are vectors of choice because they are non-pathogenic; able to accommodate large foreign DNA inserts and can be produced at high titers. Hence, the demand for pure and functional baculovirus vectors for gene delivery experiments is anticipated in the future. The main goal of this work is to develop a simple and efficient process to purify recombinant baculovirus derived from Autographa californica multiple nucleopolyhedrovirus from a culture supernatant by size exclusion chromatography. The final yields obtained for total and infectious particles were 1.39 x 10(11) and 1.02 x 10(10) and recoveries of 25% and 24%, respectively. The virus was purified from the majority of the protein contaminants as shown by sodium dodecyl sulfate polyacrylamide gel electrophoresis. Negative stain electron microscopy demonstrated that >95% of the purified virus was intact particles with shape like rod and average diameter and length of 60 and 266 nm, respectively. Transduction of 293 human embryonic kidney cells by a purified GFP-expressing BacMam at a multiplicity of transduction of 200 resulted in 36% positive cell population.


Asunto(s)
Baculoviridae/aislamiento & purificación , Cromatografía Liquida/métodos , Vectores Genéticos/aislamiento & purificación , Filtros Microporos , Animales , Baculoviridae/genética , Baculoviridae/crecimiento & desarrollo , Línea Celular , Células Cultivadas , Filtración/métodos , Regulación Viral de la Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Mariposas Nocturnas/virología , Nucleopoliedrovirus/genética , Nucleopoliedrovirus/crecimiento & desarrollo , Nucleopoliedrovirus/aislamiento & purificación , Recombinación Genética , Spodoptera , Transducción Genética
4.
J Virol Methods ; 144(1-2): 32-40, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17467815

RESUMEN

Recombinant adeno-associated virus (rAAV) has emerged in recent years as a promising gene therapy vector that may be used in the treatment of diverse human diseases. The major obstacle to broadening the usage of rAAV vectors remains the limited capacity of available production systems to provide sufficient rAAV quantities for preclinical and clinical trials. The impracticality of expanding commonly used adherent cell lines represents a limitation to large-scale production. This paper describes successful productions of rAAV type 2 using suspension-growing human embryonic kidney (HEK293) cells in serum-free medium. The developed process, based on triple transfection employing polyethylenimine (PEI) as DNA transporter, allowed for a serum-free production of AAV, yielding viral vector titer up to 4.5x10(11) infectious viral particles (IVP) in a 3.5-L bioreactor. A maximum ratio of VG:IVP in the order of 200:1 was obtained, indicating the efficient encapsidation of viral vectors in HEK293 cells. The effect of varying the ratio of three plasmids and the influence of cell density at transfection were studied. The conditioned medium did not limit or inhibit the rAAV production; therefore, the elimination of the medium exchange step before or after transfection greatly simplified the scale-up of rAAV production. The cell-specific viral titers obtained in bioreactor suspension cultures were similar or higher than those obtained with control adherent cell cultures which further supported the scalability of the process. From multiple aspects including process simplicity, scalability, and low operating costs, this transfection method appears to be the most promising technology for large-scale production of rAAV.


Asunto(s)
Dependovirus/crecimiento & desarrollo , Transfección , Cultivo de Virus/métodos , Reactores Biológicos , Técnicas de Cultivo de Célula , Línea Celular , Dependovirus/genética , Vectores Genéticos , Humanos
5.
Methods Mol Biol ; 388: 281-96, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17951776

RESUMEN

The ability to make a large variety of virus-like particles (VLPs) has been successfully achieved in the baculovirus expression vector system (BEVS)/insect cell system. The production and scale-up of these particles, which are mostly sought as candidate vaccines, are currently being addressed. Furthermore, these VLPs are being investigated as delivery agents for use as therapeutics. Recently, adeno-associated viral (AAV) vectors, which can be potentially used for human gene therapy, have been produced in insect cells using three baculovirus vectors to supply the required genes. The use of host insect cells allows mass production of VLPs in a proven scaleable system. This chapter focuses on the methodology, based on the work done in our lab, for the production of AAV-like particles and vectors in a BEVS/insect cell system.


Asunto(s)
Baculoviridae/genética , Dependovirus/genética , Vectores Genéticos/genética , Insectos/virología , Animales , Baculoviridae/metabolismo , Baculoviridae/patogenicidad , Western Blotting , Línea Celular , Dependovirus/metabolismo , Dependovirus/patogenicidad , Electroforesis en Gel de Poliacrilamida , Humanos , Insectos/citología
6.
Vaccine ; 34(29): 3381-7, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27154390

RESUMEN

Tuberculosis (TB) is the second leading cause of death by infectious disease worldwide. The only available TB vaccine is the Bacille Calmette-Guerin (BCG). However, parenterally administered Mycobacterium bovis BCG vaccine confers only limited immune protection from pulmonary tuberculosis in humans. There is a need for developing effective boosting vaccination strategies. AdAg85A, an adenoviral vector expressing the mycobacterial protein Ag85A, is a new tuberculosis vaccine candidate, and has shown promising results in pre-clinical studies and phase I trial. This adenovirus vectored vaccine is produced using HEK 293 cell culture. Here we report on the optimization of cell culture conditions, scale-up of production and purification of the AdAg85A at different scales. Four commercial serum-free media were evaluated under various conditions for supporting the growth of HEK293 cell and production of AdAg85A. A culturing strategy was employed to take advantages of two culture media with respective strengths in supporting the cell growth and virus production, which enabled to maintain virus productivity at higher cell densities and resulted in more than two folds of increases in culture titer. The production of AdAg85A was successfully scaled up and validated at 60L bioreactor under the optimal conditions. The AdAg85A generated from the 3L and 60L bioreactor runs was purified through several purification steps. More than 98% of total cellular proteins was removed, over 60% of viral particles was recovered after the purification process, and purity of AdAg85A was similar to that of the ATCC VR-1516 Ad5 standard. Vaccination of mice with the purified AdAg85A demonstrated a very good level of Ag85A-specific antibody responses. The optimized production and purification conditions were transferred to a GMP facility for manufacturing of AdAg85A for generation of clinical grade material to support clinical trials.


Asunto(s)
Adenoviridae , Técnicas de Cultivo de Célula , Vacunas contra la Tuberculosis/biosíntesis , Aciltransferasas/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Formación de Anticuerpos , Antígenos Bacterianos/inmunología , Reactores Biológicos , Medio de Cultivo Libre de Suero , Femenino , Células HEK293 , Humanos , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos BALB C , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/prevención & control
7.
Biotechnol Prog ; 21(1): 154-60, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15903253

RESUMEN

Production of recombinant adeno-associated viral vectors using a baculovirus/insect cell system at various scales is presented. Shake flask studies were conducted to assess conditions to be used in bioreactors. Two insect cell lines, Trichoplusia ni (H5) and Spodoptera frugiperda (Sf9), were compared for their ability to produce rAAV-2 after infection with recombinant baculoviruses coding for the essential components of the vector. The effect of varying the ratio between individual baculoviruses and the effect of the overall multiplicity of infection (MOI), as well as the cell density at infection, were also examined. Infectious rAAV-2 particles were proportionally produced when increasing the individual MOI of BacRep virus up to 1.6. When equal amounts of each virus were used, a leveling effect occurred beyond an overall MOI of 5 and a maximum titer was obtained. Increasing the cell density at infection resulted in higher yields when infecting the cells in fresh medium; however, for the production of bioactive particles, an optimal peak cell density of approximately 1 x 10(6) cells/mL was observed without medium exchange. Infection in 3- and 20-L bioreactors was done at an overall MOI of 5 with a ratio of the three baculoviruses equal to 1:1:1. Under these conditions and infecting the cells in fresh medium, a total of approximately 2.2 x 10(12) infectious viral particles (bioactive particles) or 2.6 x 10(15) viral particles were produced in a 3-L bioreactor. Without replacing the medium at infection, similar titers were produced in 20 L. Our data demonstrates the feasibility of rAAV-2 production by BEVS at various scales in bioreactors and indicates that further optimization is required for production at high cell densities.


Asunto(s)
Baculoviridae/metabolismo , Reactores Biológicos , Dependovirus/genética , Dependovirus/metabolismo , Vectores Genéticos/biosíntesis , Vectores Genéticos/genética , Animales , Baculoviridae/genética , Recuento de Células , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Medio de Cultivo Libre de Suero , Insectos , Proteínas Recombinantes/biosíntesis , Suspensiones , Factores de Tiempo , Transfección
8.
Biotechnol Prog ; 20(3): 858-63, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15176892

RESUMEN

Human 293S cells culture for recombinant adenovirus production is traditionally carried out in batch at a maximum of 6 x 10(5) cells/mL. A previous report demonstrated that fed-batch, applied to the adenovirus/293S cells system, improves the volumetric production of viral proteins by increasing the cell density at which cells can be infected, up to 2 x 10(6) cells/mL, without reducing the per-cell yield of product. To increase this cell density limit, the adenovirus production was performed in a perfusion system where the cells were separated by means of a tangential flow filtration device. 293S cell growth to 14 x 10(6) cells/mL was achieved in 10 days, at a medium renewal rate of 1 volume of medium per reactor volume and day (VVD). For adenovirus production, three 293S cell cultures were perfused at 1 VVD in parallel and infected at an average density of 8 x 10(6) cells/mL. One of the cultures was set at 37 degrees C and the two others at 35 degrees C. After a rapid initial cell loss, the average cell density stabilized at 5.75 x 10(6) cells/mL, 12 h postinfection, which was 8 times higher than the cell density in the batch control. This allowed the production of 3.2 x 10(9) infectious viral particles/mL (IVP/mL) at 37 degrees C and 7.8 x 10(9) IVP/mL at 35 degrees C, this last result being 5.5 times higher than the control. To our knowledge, this nonconcentrated titer is the highest value that has ever been published for adenovirus vector production. These observations lead to the conclusion that perfusion is an efficient tool to maintain, at high cell density, a specific production rate level sufficient to increase significantly the adenovirus volumetric production. Furthermore, it shows that perfusion at 35 degrees C can improve viral titer by 2.4-fold compared to 37 degrees C, in accordance with a previous study on adenovirus batch production.


Asunto(s)
Adenoviridae/crecimiento & desarrollo , Adenoviridae/aislamiento & purificación , Reactores Biológicos/microbiología , Técnicas de Cultivo de Célula/métodos , Fibroblastos/fisiología , Fibroblastos/virología , Cultivo de Virus/métodos , Adenoviridae/genética , Línea Celular , Proliferación Celular , Supervivencia Celular , Fibroblastos/citología , Vectores Genéticos/genética , Humanos
9.
Vaccine ; 30(2): 300-6, 2012 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-22079076

RESUMEN

Rabies virus is an important causative agent of disease resulting in an acute infection of the nervous system and death. Although curable if treated in a timely manner, rabies remains a serious public health issue in developing countries, and the indigenous threat of rabies continues in developed countries because of wildlife reservoirs. Control of rabies in wildlife is still an important challenge for governmental authorities. There are a number of rabies vaccines commercially available for control of wildlife rabies infection. However, the vaccines currently distributed to wildlife do not effectively immunize all at-risk species, particularly skunks. A replication competent recombinant adenovirus expressing rabies glycoprotein (AdRG1.3) has shown the most promising results in laboratory trials. The adenovirus vectored vaccine is manufactured using HEK 293 cells. This study describes the successful scale-up of AdRG1.3 adenovirus production from 1 to 500 L and the manufacturing of large quantities of bulk material required for field trials to demonstrate efficacy of this new candidate vaccine. The production process was streamlined by eliminating a medium replacement step prior to infection and the culture titer was increased by over 2 fold through optimization of cell culture medium. These improvements produced a more robust and cost-effective process that facilitates industrialization and commercialization. Over 17,000 L of AdRG1.3 adenovirus cultures were manufactured to support extensive field trials. AdRG1.3 adenovirus is formulated and packaged into baits by Artemis Technologies Inc. using proprietary technology. Field trials of AdRG1.3 rabies vaccine baits have been conducted in several Canadian provinces including Ontario, Quebec and New Brunswick. The results from field trials over the period 2006-2009 demonstrated superiority of the new vaccine over other licensed vaccines in immunizing wild animals that were previously difficult to vaccinate.


Asunto(s)
Adenoviridae/aislamiento & purificación , Portadores de Fármacos/aislamiento & purificación , Vectores Genéticos/aislamiento & purificación , Vacunas Antirrábicas/aislamiento & purificación , Virus de la Rabia/genética , Tecnología Farmacéutica/métodos , Adenoviridae/genética , Adenoviridae/crecimiento & desarrollo , Animales , Técnicas de Cultivo de Célula/métodos , Línea Celular , Humanos , Vacunas Antirrábicas/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/aislamiento & purificación
10.
Vaccine ; 28(21): 3661-71, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20347632

RESUMEN

Cell culture processes offer an attractive alternative to conventional chicken egg-based influenza vaccine production methods. However, most protocols still rely on the use of adherent cells, which makes process scale-up a challenging issue. In this study, it is demonstrated that the HEK-293 human cell line is able to efficiently replicate influenza virus. Production in serum-free suspension of HEK-293 cultures resulted in high titers of infectious influenza viruses for different subtypes and variants including A/H1, A/H3 and B strains. After virus adaptation and optimization of infection conditions, production in 3-L bioreactor resulted in titers of up to 10(9)IVP/mL demonstrating the scale-up potential of the process.


Asunto(s)
Alphainfluenzavirus/crecimiento & desarrollo , Cultivo de Virus/métodos , Animales , Reactores Biológicos , Recuento de Células , Línea Celular , Perros , Pruebas de Hemaglutinación , Humanos , Alphainfluenzavirus/aislamiento & purificación , Betainfluenzavirus/crecimiento & desarrollo , Betainfluenzavirus/aislamiento & purificación , Receptores Virales/metabolismo , Tripsina/química
11.
Vaccine ; 28(2): 309-16, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19879996

RESUMEN

A robust and reliable GMP-compatible fed-batch process was successfully developed for the production of recombinant hemagglutinin (rHA) proteins by expresSF cells. The feeding solution, feeding strategy as well as the cell density at infection were optimized to maximize the final rHA production yields without affecting the existing rHA recovery protocol and downstream process. A simple and stable feeding solution was formulated and a rational feeding regimen designed to yield, depending on the rHA baculovirus used, between 2- and 3-fold enhancements in volumetric rHA production with increased specific productivity compared to the batch culture. Recombinant HA from fed-batch cultures could be simply recovered following cell lysis and purified through chromatographic steps. Overall, the increased rHA yield was maintained throughout the whole process. The performance, reproducibility and scalability of the fed-batch process was successfully demonstrated in 12 bioreactor runs of 2- and 10-L working volume using five different rHA encoding baculoviruses.


Asunto(s)
Vacunas contra la Influenza/biosíntesis , Vacunas contra la Influenza/metabolismo , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/inmunología , Animales , Línea Celular , Hemaglutininas/genética , Hemaglutininas/metabolismo , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Spodoptera
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA