Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 9(11)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34835266

ABSTRACT

The ongoing COVID-19 pandemic drew global attention to infectious diseases, attracting numerous resources for development of pandemic preparedness plans and vaccine platforms-technologies with robust manufacturing processes that can quickly be pivoted to target emerging diseases. Newcastle Disease Virus (NDV) has been studied as a viral vector for human and veterinary vaccines, but its production relies heavily on embryonated chicken eggs, with very few studies producing NDV in cell culture. Here, NDV is produced in suspension Vero cells, and analytical assays (TCID50 and ddPCR) are developed to quantify infectious and total viral titer. NDV-GFP and NDV-FLS (SARS-CoV-2 full-length spike protein) constructs were adapted to replicate in Vero and HEK293 suspension cultures using serum-free media, while fine-tuning parameters such as MOI, temperature, and trypsin concentration. Shake flask productions with Vero cells resulted in infectious titers of 1.07 × 108 TCID50/mL for NDV-GFP and 1.33 × 108 TCID50/mL for NDV-FLS. Production in 1 L batch bioreactors also resulted in high titers in culture supernatants, reaching 2.37 × 108 TCID50/mL for NDV-GFP and 3.16 × 107 TCID50/mL for NDV-FLS. This shows effective NDV production in cell culture, building the basis for a scalable vectored-vaccine manufacturing process that can be applied to different targets.

2.
NPJ Vaccines ; 6(1): 106, 2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34417462

ABSTRACT

The Vero cell line is the most used continuous cell line for viral vaccine manufacturing with more than 40 years of accumulated experience in the vaccine industry. Additionally, the Vero cell line has shown a high affinity for infection by MERS-CoV, SARS-CoV, and recently SARS-CoV-2, emerging as an important discovery and screening tool to support the global research and development efforts in this COVID-19 pandemic. However, the lack of a reference genome for the Vero cell line has limited our understanding of host-virus interactions underlying such affinity of the Vero cell towards key emerging pathogens, and more importantly our ability to redesign high-yield vaccine production processes using Vero genome editing. In this paper, we present an annotated highly contiguous 2.9 Gb assembly of the Vero cell genome. In addition, several viral genome insertions, including Adeno-associated virus serotypes 3, 4, 7, and 8, have been identified, giving valuable insights into quality control considerations for cell-based vaccine production systems. Variant calling revealed that, in addition to interferon, chemokines, and caspases-related genes lost their functions. Surprisingly, the ACE2 gene, which was previously identified as the host cell entry receptor for SARS-CoV and SARS-CoV-2, also lost function in the Vero genome due to structural variations.

3.
Biotechnol Bioeng ; 118(7): 2649-2659, 2021 07.
Article in English | MEDLINE | ID: mdl-33837958

ABSTRACT

The Vero cell line is the most used continuous cell line in viral vaccine manufacturing. This adherent cell culture platform requires the use of surfaces to support cell growth, typically roller bottles, or microcarriers. We have recently compared the production of rVSV-ZEBOV on Vero cells between microcarrier and fixed-bed bioreactors. However, suspension cultures are considered superior with regard to process scalability. Therefore, we further explore the Vero suspension system for recombinant vesicular stomatitis virus (rVSV)-vectored vaccine production. Previously, this suspension cell line was only able to be cultivated in a proprietary medium. Here, we expand the adaptation and bioreactor cultivation to a serum-free commercial medium. Following small-scale optimization and screening studies, we demonstrate bioreactor productions of highly relevant vaccines and vaccine candidates against Ebola virus disease, HIV, and coronavirus disease 2019 in the Vero suspension system. rVSV-ZEBOV, rVSV-HIV, and rVSVInd -msp-SF -Gtc can replicate to high titers in the bioreactor, reaching 3.87 × 107 TCID50 /ml, 2.12 × 107 TCID50 /ml, and 3.59 × 109 TCID50 /ml, respectively. Furthermore, we compare cell-specific productivities, and the quality of the produced viruses by determining the ratio of total viral particles to infectious viral particles.


Subject(s)
Bioreactors/virology , Cell Culture Techniques/methods , Ebola Vaccines , Vesiculovirus/genetics , Animals , COVID-19 Vaccines , Chlorocebus aethiops , Culture Media, Serum-Free , Vero Cells , Viral Vaccines
4.
Vaccine ; 38(50): 7949-7955, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33139138

ABSTRACT

Acquired Immune Deficiency Syndrome (AIDS) in humans is a result of the destruction of the immune system caused by Human Immunodeficiency Virus (HIV) infection. This serious epidemic is still progressing world-wide. Despite advances in treatment, a safe and effective preventive HIV vaccine is desired to combat this disease, and to save millions of lives. However, such a vaccine is not available yet although extensive amounts of resources in research and development have been invested over three decades. In light of the recently approved Ebola virus disease vaccine based on a recombinant vesicular stomatitis virus (rVSV-ZEBOV), we present the results of our work on three novel VSV-vectored HIV vaccine candidates. We describe the design, rescue, production and purification method and evaluate their immunogenicity in mice prior to preclinical studies that will be performed in non-human primates. The production of each of the three candidate vaccines (rVSV-B6-NL4.3Env/SIVtm, rVSV-B6-NL4.3Env/Ebtm and rVSV-B6-A74Env(PN6)/SIVtm) was evaluated in small scale in Vero cells and it was found that production kinetics on Vero cells vary depending on the HIV gp surface protein used. Purified virus preparations complied with the WHO restrictions for the residual DNA and host cell protein contents. Finally, when administered to mice, all three rVSV-HIV vaccine candidates induced an HIV gp140-specific antibody response.


Subject(s)
AIDS Vaccines , Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola , Vesicular Stomatitis , Animals , Cell Culture Techniques , Chlorocebus aethiops , Genetic Vectors , Mice , Vaccines, Synthetic/genetics , Vero Cells
5.
Biotechnol Adv ; 44: 107608, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32768520

ABSTRACT

The Vero cell line is considered the most used continuous cell line for the production of viral vectors and vaccines. Historically, it is the first cell line that was approved by the WHO for the production of human vaccines. Comprehensive experimental data on the production of many viruses using the Vero cell line can be found in the literature. However, the vast majority of these processes is relying on the microcarrier technology. While this system is established for the large-scale manufacturing of viral vaccine, it is still quite complex and labor intensive. Moreover, scale-up remains difficult and is limited by the surface area given by the carriers. To overcome these and other drawbacks and to establish more efficient manufacturing processes, it is a priority to further develop the Vero cell platform by applying novel bioprocess technologies. Especially in times like the current COVID-19 pandemic, advanced and scalable platform technologies could provide more efficient and cost-effective solutions to meet the global vaccine demand. Herein, we review the prevailing literature on Vero cell bioprocess development for the production of viral vectors and vaccines with the aim to assess the recent advances in bioprocess development. We critically underline the need for further research activities and describe bottlenecks to improve the Vero cell platform by taking advantage of recent developments in the cell culture engineering field.


Subject(s)
Coronavirus Infections , Pandemics , Pneumonia, Viral , Viral Vaccines , Animals , Betacoronavirus , Bioreactors , COVID-19 , Chlorocebus aethiops , Humans , SARS-CoV-2 , Vero Cells
6.
MethodsX ; 7: 100806, 2020.
Article in English | MEDLINE | ID: mdl-32195130

ABSTRACT

The recombinant Vesicular Stomatitis Virus (rVSV) is an emerging platform for viral vector-based vaccines. Promising results have been reported in clinical trials for the rVSV-ZEBOV vaccine for Ebola virus disease prevention. In this study, we describe the titration tools elaborated to assess the titre of rVSV-ZEBOV productions. • A streamlined Median Tissue Culture Infectious Dose (TCID50) assay to determine the infectious titer of this vaccine was established. • A digital polymerase chain reaction (dPCR) assay to assess the total number of viral particles present in cell-free culture supernatants of rVSV productions was developed. • These assays are used to titre rVSV-ZEBOV samples and characterize the ratio of total particles to infectious units for monitoring process robustness and product quality attributes and can be used to titre samples generated in the production of further rVSV vectors.

7.
J Biotechnol ; 310: 32-39, 2020 Feb 20.
Article in English | MEDLINE | ID: mdl-32006630

ABSTRACT

Ebola virus disease outbreaks have repeatedly occurred on the African continent over the last decades, with more serious outbreaks in recent years. Being highly transmissible and associated to high fatality rates, it constitutes a serious threat to public health. Vaccination, however, may allow for efficient control of its propagation. The most promising Ebola vaccine candidate to date, rVSV-ZEBOV, relies on a recombinant vesicular stomatitis virus construct, in which the native viral glycoprotein is replaced by the glycoprotein of Ebola virus (Zaire). However, its cell-based manufacturing process is still lengthy and cumbersome, thus urging the implementation of a new and more efficient bioprocess. To address these issues, serum-free production of rVSV-ZEBOV in Vero cells has been studied with the aim to test an alternative upstream process. Until viable options of suspension cell culture are available, Vero cell cultures still rely on adherent bioprocesses and have thus been developed in this work. Particularly, a bioprocess developed with standard microcarrier bioreactor technology was successfully transferred to the novel single-use scale-X™ hydro fixed-bed.


Subject(s)
Bioreactors , Ebola Vaccines/biosynthesis , Vesiculovirus , Animals , Chlorocebus aethiops , Culture Media, Serum-Free/pharmacology , Ebola Vaccines/genetics , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics , Vero Cells
8.
Vaccine ; 37(44): 6624-6632, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31548015

ABSTRACT

Ebola virus disease is an urgent international priority. Promising results for several vaccine candidates have been reported in non-human primate studies and clinical trials with the most promising being the rVSV-ZEBOV vaccine. In this study, we sought to produce rVSV-ZEBOV in HEK 293SF cells in suspension and serum-free media. The purpose of this study was to establish a process using the HEK 293SF production platform, optimise the production titre, demonstrate scalability and the efficiency of the generated material to elicit an immune reaction in an animal model. Critical process parameters were evaluated to maximize production yield and process robustness and the following operating conditions: 1-2 × 106 cells/mL grown in HyClone HyCell TransFx-H media infected at an MOI of 0.001 with a temperature shift to 34 °C during the production phase and a harvest of the product after 48 h. Using these conditions, scalability in a 3.5 L controlled bioreactor was shown reaching a titre of 1.19 × 108 TCID50/mL at the peak of production, the equivalent of 4165 doses of vaccine per litre. The produced virus was shown to be thermostable in the culture media and, when concentrated, purified and administered to mice, demonstrated the ability to induce a ZEBOV-specific immune response.


Subject(s)
Batch Cell Culture Techniques , Ebola Vaccines/biosynthesis , Ebola Vaccines/immunology , Ebolavirus/immunology , Vaccines, DNA/biosynthesis , Vaccines, DNA/immunology , Vesiculovirus , Animals , Antibodies, Viral/immunology , Bioreactors , Disease Models, Animal , Ebola Vaccines/administration & dosage , Ebola Vaccines/genetics , Ebolavirus/genetics , Female , HEK293 Cells , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Humans , Immunization , Mice , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vesiculovirus/genetics
9.
World J Microbiol Biotechnol ; 32(6): 103, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27116969

ABSTRACT

Economic realities for the rising industrial biofuel production have changed substantially during the low oil price period starting in the mid 2010's. Increased competition requires the sector to increase productivity through the reduction of low-value by-products and full utilization of all value and energy stored in their respective feedstock. Biodiesel is produced commercially from substrates such as animal fat and vegetable oil, generating approximately 10 wt% crude glycerol as its main, currently underutilized, by-product. This crude glycerol is contaminated with catalyst, soap, free fatty acids, glycerides and methyl esters; hence only a small fraction enters the existing glycerol markets, while the purification costs for the majority of crude glycerol are simply too high. However, this presents a unique opportunity to generate additional value. One technical possibility is to use crude glycerol as a carbon source for butanol production, a compound of higher value and energy, a potential additive for gasoline and diesel fuels and bulk chemical commodity. Conversion facilities could be co-located with biodiesel plants, utilizing established infrastructure and adding significant value and productivity to the existing biodiesel industry. This review focuses on the current activities geared towards the bioconversion of crude glycerol to butanol.


Subject(s)
Bacteria/metabolism , Biofuels , Butanols/metabolism , Glycerol/metabolism , Bacteria/genetics , Carbon/metabolism , Fermentation , Industrial Microbiology , Industry , Metabolic Engineering , Metabolic Networks and Pathways , Mutagenesis
10.
Biotechnol Bioeng ; 112(9): 1822-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25850946

ABSTRACT

The ease of use and versatility of the Baculovirus Expression Vector System (BEVS) has made it one of the most widely used systems for recombinant protein production However, co-expression systems currently in use mainly make use of the very strong very late p10 and polyhedron (polh) promoters to drive expression of foreign genes, which does not provide much scope for tailoring expression ratios within the cell. This work demonstrates the use of different Autographa californica multicapsid nucleopolyhedrovirus (AcMNPV) promoters to control the timing and expression of two easily traceable fluorescent proteins, the enhanced green fluorescent protein (eGFP), and a red fluorescent protein (DsRed2) in a BEVS co-expression system. Our results show that gene expression levels can easily be controlled using this strategy, and also that modulating the expression level of one protein can influence the level of expression of the other protein within the system, thus confirming the concept of genes "competing" for limited cellular resources. Plots of "expression ratios" of the two model genes over time were obtained, and may be used in future work to tightly control timing and levels of foreign gene expression in an insect cell co-expression system.


Subject(s)
Baculoviridae/genetics , Biotechnology/methods , Green Fluorescent Proteins/metabolism , Luminescent Proteins/metabolism , Promoter Regions, Genetic/genetics , Recombinant Proteins/metabolism , Cell Proliferation , Genes, Reporter/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Sf9 Cells , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...