Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biotechnol Prog ; 40(1): e3399, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37874920

RESUMO

Monoclonal antibodies (mAbs) are effective therapeutic agents against many acute infectious diseases including COVID-19, Ebola, RSV, Clostridium difficile, and Anthrax. mAbs can therefore help combat a future pandemic. Unfortunately, mAb development typically takes years, limiting its potential to save lives during a pandemic. Therefore "pandemic mAb" timelines need to be shortened. One acceleration tool is "deferred cloning" and leverages new Chinese hamster ovary (CHO) technology based on targeted gene integration (TI). CHO pools, instead of CHO clones, can be used for Phase I/II clinical material production. A final CHO clone (producing the mAb with a similar product quality profile and preferably with a higher titer) can then be used for Phase III trials and commercial manufacturing. This substitution reduces timelines by ~3 months. We evaluated our novel CHO TI platform to enable deferred cloning. We created four unique CHO pools expressing three unique mAbs (mAb1, mAb2, and mAb3), and a bispecific mAb (BsAb1). We then performed single-cell cloning for mAb1 and mAb2, identifying three high-expressing clones from each pool. CHO pools and clones were inoculated side-by-side in ambr15 bioreactors. CHO pools yielded mAb titers as high as 10.4 g/L (mAb3) and 7.1 g/L (BsAb1). Subcloning yielded CHO clones expressing higher titers relative to the CHO pools while yielding similar product quality profiles. Finally, we showed that CHO TI pools were stable by performing a 3-month cell aging study. In summary, our CHO TI platform can increase the speed to clinic for a future "pandemic mAb."


Assuntos
Anticorpos Biespecíficos , Cricetinae , Animais , Cricetulus , Anticorpos Biespecíficos/genética , Células CHO , Anticorpos Monoclonais/genética , Células Clonais
2.
Biotechnol Bioeng ; 121(1): 395-402, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37902721

RESUMO

The gene therapy field has advanced in recent years with five recombinant adeno-associated virus (rAAV) based products winning Food and Drug Administration (FDA) approval. As the number of therapeutic applications and overall production demands for rAAV increase, it is valuable to evaluate rAAV production in different production cells. Chinese hamster ovary (CHO) cells have been a robust host for biomolecule manufacturing for more than 35 years. However, there is no report to our knowledge describing the use of CHO cells for rAAV production. In this study, we examined the ability of CHO cells to produce rAAV using a transient plasmid transfection approach. Our results demonstrated that CHO is capable of producing rAAV with detectable viral fundamental components including viral RNAs, proteins, and rAAV viral particles. We identified the expression of cap proteins as one of the limiting factors for rAAV production in CHO cells. We therefore added an additional cytomegalovirus (CMV)-Cap plasmid to the CHO transfection. After increasing cap protein expression, we detected rAAV titers as high as 3 × 108 viral genomes for every 2 × 109 capsids in CHO cells using a quintuple transfection method (standard AAV2 Rep/Cap, helper, gene of interest plasmids, plus CMV-E1, and CMV-Cap plasmids) with comparable full particle percent (average 15%) to that of human embryo kidney (HEK)-derived rAAV. Our study provides a foundation for potential rAAV production in CHO cells.


Assuntos
Infecções por Citomegalovirus , Vetores Genéticos , Animais , Cricetinae , Humanos , Cricetulus , Células CHO , Dependovirus/genética , Plasmídeos/genética
3.
Biotechnol Prog ; 36(6): e3047, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33411420

RESUMO

We developed a simple transient Chinese Hamster Ovary expression platform. Titers for a random panel of 20 clinical monoclonal antibodies (mAbs) ranged from 0.6 to 2.7 g/L after 7 days. Two factors were the key in obtaining these high titers. First, we utilized an extremely high starting cell density (20 million cells/ml), and then arrested further cell growth by employing mild hypothermic conditions (32°C). Second, we performed a 6-variable Design of Experiments to find optimal concentrations of plasmid DNA (coding DNA), boost DNA (DNA encoding the XBP1S transcription factor), transfection reagent (polyethylenimine [PEI]), and nutrient feed amounts. High coding DNA concentrations (12.5 mg/L) were found to be optimal. We therefore diluted expensive coding DNA with inexpensive inert filler DNA (herring sperm DNA). Reducing the coding DNA concentration by 70% from 12.5 to 3.75 mg/L did not meaningfully reduce mAb titers. Titers for the same panel of 20 clinical mAbs ranged from 0.7 to 2.2 g/L after reducing the coding DNA concentration to 3.75 mg/L. Finally, we found that titer and product quality attributes were similar for a clinical mAb (rituximab) expressed at very different scales (volumes ranging from 3 ml to 2 L).


Assuntos
Anticorpos Monoclonais/biossíntese , Biotecnologia/métodos , Técnicas de Cultura de Células/métodos , Plasmídeos/genética , Animais , Anticorpos Monoclonais/genética , Células CHO , Cricetinae , Cricetulus , Humanos , Transfecção
4.
Biotechnol J ; 13(10): e1700748, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29797786

RESUMO

Clonally derived cell lines (CDCL) from Chinese Hamster Ovary (CHO) host cell lines, remain the most popular method to manufacture therapeutic proteins. However, CHO cell pools are increasingly being used as an alternate method to produce therapeutic proteins for preclinical drug development in an effort to shorten the time required for new drug development. It is essential that these CHO pools exhibit the desired attributes of CHO CDCLs such as high protein titers and consistent product quality attributes (PQAs). In this study the authors evaluated the Leap-In Transposase®, for the expression of four different proteins (three mAbs and one Bispecific mAb). The resultant pool titers ranges from 2.0 to 5.0 g L-1 for the four proteins compared to 1.5-3.3 g L-1 from the respective control pools (generated by random gene integration). The resultant cell pools are a homogeneously expressing cell population. The average gene copy numbers are similar or lower in the evaluation pools relative to the control pools. The higher titers in the evaluation pools are attributed to higher levels of both IgG-LC and IgG-HC mRNA. In conclusion, the Leap-In transposase generates high titer, homogeneous CHO pools in a short time-period without introducing any undesired PQAs.


Assuntos
Anticorpos Biespecíficos , Anticorpos Monoclonais , Técnicas de Cultura de Células , Transposases , Animais , Anticorpos Biespecíficos/biossíntese , Anticorpos Monoclonais/biossíntese , Células CHO , Cricetulus , Plasmídeos
5.
Nat Med ; 23(10): 1215-1219, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28846098

RESUMO

Growth/differentiation factor 15 (GDF15), also known as MIC-1, is a distant member of the transforming growth factor-ß (TGF-ß) superfamily and has been implicated in various biological functions, including cancer cachexia, renal and heart failure, atherosclerosis and metabolism. A connection between GDF15 and body-weight regulation was initially suggested on the basis of an observation that increasing GDF15 levels in serum correlated with weight loss in individuals with advanced prostate cancer. In animal models, overexpression of GDF15 leads to a lean phenotype, hypophagia and other improvements in metabolic parameters, suggesting that recombinant GDF15 protein could potentially be used in the treatment of obesity and type 2 diabetes. However, the signaling and mechanism of action of GDF15 are poorly understood owing to the absence of a clearly identified cognate receptor. Here we report that GDNF-family receptor α-like (GFRAL), an orphan member of the GFR-α family, is a high-affinity receptor for GDF15. GFRAL binds to GDF15 in vitro and is required for the metabolic actions of GDF15 with respect to body weight and food intake in vivo in mice. Gfral-/- mice were refractory to the effects of recombinant human GDF15 on body-weight, food-intake and glucose parameters. Blocking the interaction between GDF15 and GFRAL with a monoclonal antibody prevented the metabolic effects of GDF15 in rats. Gfral mRNA is highly expressed in the area postrema of mouse, rat and monkey, in accordance with previous reports implicating this region of the brain in the metabolic actions of GDF15 (refs. 4,5,6). Together, our data demonstrate that GFRAL is a receptor for GDF15 that mediates the metabolic effects of GDF15.


Assuntos
Área Postrema/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator 15 de Diferenciação de Crescimento/farmacologia , Obesidade/metabolismo , Redução de Peso/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Ingestão de Alimentos/genética , Citometria de Fluxo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Células HEK293 , Humanos , Immunoblotting , Macaca fascicularis , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Ressonância de Plasmônio de Superfície , Redução de Peso/genética
6.
Biotechnol Prog ; 33(5): 1393-1400, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28722325

RESUMO

Most biopharmaceutical drugs, especially monoclonal antibodies (mAbs), bispecific antibodies (BsAbs) and Fc-fusion proteins, are expressed using Chinese Hamster Ovary (CHO) cell lines. CHO cells typically yield high product titers and high product quality. Unfortunately, CHO cell lines also generate high molecular weight (HMW) aggregates of the desired product during cell culture along with CHO host cell protein (HCP) and CHO DNA. These immunogenic species, co-purified during Protein A purification, must be removed in a multi-step purification process. Our colleagues have reported the use of a novel polymer-mediated flocculation step to simultaneously reduce HMW, HCP and DNA from stable CHO cell cultures prior to Protein A purification. The objective of this study was to evaluate this novel "smart polymer" (SmP) in a high throughput antibody discovery workflow using transiently transfected CHO cultures. SmP treatment of 19 different molecules from four distinct molecular categories (human mAbs, murine mAbs, BsAbs and Fabs) with 0.1% SmP and 25 mM stimulus resulted in minimal loss of monomeric protein. Treatment with SmP also demonstrated a variable, concentration-dependent removal of HMW aggregates after Protein A purification. SmP treatment also effectively reduced HCP levels at each step of mAb purification with final HCP levels being several fold lower than the untreated control. Interestingly, SmP treatment was able to significantly reduce high concentrations of artificially spiked levels of endotoxin in the cultures. In summary, adding a simple flocculation step to our existing transient CHO process reduced the downstream purification burden to remove impurities and improved final product quality. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:1393-1400, 2017.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Floculação , Polímeros/química , Proteínas Recombinantes/normas , Animais , Células CHO , Cricetinae , Cricetulus , Endotoxinas/análise , Endotoxinas/química , Endotoxinas/isolamento & purificação , Humanos , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Proteínas/análise , Proteínas/química , Proteínas/isolamento & purificação , Proteínas Recombinantes/isolamento & purificação
7.
Biotechnol Prog ; 33(6): 1436-1448, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28547769

RESUMO

Generating purified protein for GLP toxicology studies (GLP-Tox) represents an important and often rate limiting step in the biopharmaceutical drug development process. Toxicity testing requires large amounts of therapeutic protein (>100 g), typically produced in a single 500-2,500 L bioreactor, using the final CHO clonally derived cell line (CDCL). One approach currently used to save time is to manufacture GLP-Tox material using pools of high-producing CHO CDCLs instead of waiting for the final CDCL. Recently, we reported CHO pools producing mAb titers >7 g/L using piggyBac-mediated gene integration (PB CHO pools). In this study, we wanted to leverage high titer PB CHO pools to produce GLP-Tox material. A detailed product quality attribute (PQA) assessment was conducted comparing PB CHO pools to pooled Top4 CDCLs. Four mAbs were evaluated. First, we found that PB CHO pools expressed all four mAbs at high titers (2.8-4.4 g/L in shake flasks). Second, all four PB CHO pools were aged to 55 generations (Gen). All four PB CHO Pools were found to be suitable over 55 Gen. Finally, we performed bioreactor scale-up. PB CHO pool titers (3.7-4.8 g/L) were similar or higher than the pooled Top 4 CDCLs in 5 L bioreactors (2.4-4.1 g/L). The PQAs of protein derived from PB CHO pools were very similar to pooled Top 4 CHO CDCLs according to multiple orthogonal techniques including peptide mapping analysis. Taken together, these results demonstrate the technical feasibility of using PB CHO pools to manufacture protein for GLP-Tox. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:1436-1448, 2017.


Assuntos
Anticorpos Monoclonais/genética , Reatores Biológicos , Células CHO/efeitos dos fármacos , Proteínas Recombinantes/genética , Animais , Anticorpos Monoclonais/farmacologia , Células CHO/metabolismo , Cricetulus , Avaliação Pré-Clínica de Medicamentos , Humanos , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/isolamento & purificação
8.
Biotechnol Prog ; 33(2): 534-540, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28188692

RESUMO

Chinese hamster ovary (CHO) cells remain the most popular host for the production of biopharmaceutical drugs, particularly monoclonal antibodies (mAbs), bispecific antibodies, and Fc-fusion proteins. Creating and characterizing the stable CHO clonally-derived cell lines (CDCLs) needed to manufacture these therapeutic proteins is a lengthy and laborious process. Therefore, CHO pools have increasingly been used to rapidly produce protein to support and enable preclinical drug development. We recently described the generation of CHO pools yielding mAb titers as high as 7.6 g/L in a 16 day bioprocess using piggyBac transposon-mediated gene integration. In this study, we wanted to understand why the piggyBac pool titers were significantly higher (2-10 fold) than the control CHO pools. Higher titers were the result of a combination of increased average gene copy number, significantly higher messenger RNA levels and the homogeneity (i.e. less diverse population distribution) of the piggyBac pools, relative to the control pools. In order to validate the use of piggyBac pools to support preclinical drug development, we then performed an in-depth product quality analysis of purified protein. The product quality of protein obtained from the piggyBac pools was very similar to the product quality profile of protein obtained from the control pools. Finally, we demonstrated the scalability of these pools from shake flasks to 36L bioreactors. Overall, these results suggest that gram quantities of therapeutic protein can be rapidly obtained from piggyBac CHO pools without significantly changing product quality attributes. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:534-540, 2017.


Assuntos
Anticorpos Monoclonais/biossíntese , Reatores Biológicos , Proliferação de Células/fisiologia , Elementos de DNA Transponíveis/genética , Engenharia de Proteínas/métodos , Animais , Anticorpos Monoclonais/genética , Técnicas de Cultura Celular por Lotes/métodos , Células CHO , Cricetulus , Projetos Piloto , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Regulação para Cima
9.
Biotechnol Prog ; 33(2): 469-477, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27977915

RESUMO

IgG bispecific antibodies (BsAbs) represent one of the preferred formats for bispecific antibody therapeutics due to their native-like IgG properties and their monovalent binding to each target. Most reported studies utilized transient expression in HEK293 cells to produce BsAbs. However, the expression of biotherapeutic molecules using stable CHO cell lines is commonly used for biopharmaceutical manufacturing. Unfortunately, limited information is available in the scientific literature on the expression of BsAbs in CHO cell lines. In this study we describe an alternative approach to express the multiple components of IgG BsAbs using a single plasmid vector (quad vector). This single plasmid vector contains both heavy chain genes and both light chain genes required for the expression and assembly of the IgG BsAb, along with a selectable marker. We expressed, purified, and characterized four different IgG BsAbs or "hetero-mAbs" using transient CHO expression and stable CHO minipools. Transient CHO titers ranged from 90 to 160 mg/L. Stable CHO titers ranged from 0.4 to 2.3 g/L. Following a simple Protein A purification step, the percentage of correctly paired BsAbs ranged from 74% to 98% as determined by mass spectrometry. We also found that information generated from transient CHO expression was similar to information generated using stable CHO minipools. In conclusion, the quad vector approach represents a simple, but effective, alternative approach for the generation of IgG BsAbs in both transient CHO and stable CHO expression systems. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:469-477, 2017.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Proliferação de Células/fisiologia , Clonagem Molecular/métodos , Imunoglobulina G/imunologia , Engenharia de Proteínas/métodos , Transfecção/métodos , Animais , Anticorpos Monoclonais/isolamento & purificação , Células CHO , Cricetulus , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
10.
Biotechnol Prog ; 32(5): 1301-1307, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27254818

RESUMO

Chinese hamster ovary (CHO) cells remain the default production host for many biopharmaceutical drugs, particularly monoclonal antibodies (mAb). Production of gram and kilogram quantities of protein typically requires the generation of stable CHO clones. Unfortunately, this process takes several months, significantly slowing down the drug discovery and development process. Therefore, improved technologies are needed to accelerate biopharmaceutical drug discovery and final drug substance manufacturing. In this study, we describe the generation of stable CHO pools using the piggyBac transposon system. We evaluated the system using four model antibody molecules (3 mAbs and 1 bispecific Ab). Stable CHO pools were isolated in 7-12 days. Using a simple 16-day fed-batch process, we measured titers ranging from 2.3 to 7.6 g/L for the four model antibodies. This represented a 4- to 12-fold increase relative to the controls. Additionally, we isolated stable CHO clones. We found that the stable CHO clones isolated from the piggyBac transposon pools yielded titers two to threefold higher relative to the control clones. Taken together, these results suggest that stable CHO pool and clone generation can be significantly improved by using the piggyBac transposon system. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:1301-1307, 2016.


Assuntos
Anticorpos/análise , Elementos de DNA Transponíveis , Animais , Anticorpos/metabolismo , Células CHO , Células Cultivadas , Células Clonais , Cricetulus
11.
Biotechnol Lett ; 37(12): 2379-86, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26298077

RESUMO

OBJECTIVE: To develop a simple approach to increase titers of transient gene expression in CHO cells without relying on host cell line engineering as recent reports suggest that for PEI-mediated transfections, under optimized conditions, DNA delivery into cells and nuclei is not the limiting factor. RESULTS: N, N-Dimethyl acetamide (DMA) was utilized to enhance transcription. To target post-transcriptional events, we evaluated the co-expression of various genes involved in the unfolded protein response, namely XBP1S, ATF4, CHOP and HSPA5. XBP1S overexpression led to a 15-85 % increase in titer for multiple therapeutic proteins. Mechanistic studies confirmed that addition of 0.125 % DMA increased transgene mRNA levels as expected. However, overexpression of XBP1S had no effect on transgene mRNA levels, indicating that it influenced post-transcriptional events. Since DMA and XBP1S targeted different pathways, the combination of the two approaches led to an additive improvement in protein titer (150-250 % titer increase). CONCLUSION: Transcriptional and post-transcriptional pathways of transient gene expression can be targeted to increase titers without resorting to host cell line engineering in a simple, short, 7 day production process.


Assuntos
Expressão Gênica , Proteínas Recombinantes/biossíntese , Animais , Células CHO , Cricetulus , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Proteínas Recombinantes/genética , Transcrição Gênica/efeitos dos fármacos
12.
Biotechnol Prog ; 31(1): 239-47, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25403790

RESUMO

A high-cell-density transient transfection system was recently developed in our laboratory based on a CHO-GS-KO cell line. This method yields monoclonal antibody titers up to 350 mg/L from a simple 7-day process, in volumes ranging from 2 mL to 2 L. By performing transfections in 24-deep-well plates, a large number of mAbs can be expressed simultaneously. We coupled this new high-throughput transfection process to a semiautomated protein A purification process. Using a Biomek FX(p) liquid handling robot, up to 72 unique mAbs can be simultaneously purified. Our primary goal was to obtain >0.25 mg of purified mAb at a concentration of >0.5 mg/mL, without any concentration or buffer-exchange steps. We optimized both the batch-binding and the batch elution steps. The length of the batch-binding step was important to minimize mAb losses in the flowthrough fraction. The elution step proved to be challenging to simultaneously maximize protein recovery and protein concentration. We designed a variable volume elution strategy based on the average supernatant titer. Finally, we present two case studies. In the first study, we produced 56 affinity maturation mAb variants at an average yield of 0.33 ± 0.05 mg (average concentration of 0.65 ± 0.10 mg/mL). In a second study, we produced 42 unique mAbs, from an early-stage discovery effort, at an average yield of 0.79 ± 0.31 mg (average concentration of 1.59 ± 0.63 mg/mL). The combination of parallel high-yielding transient transfection and semiautomated high-throughput protein A purification represents a valuable mAb drug discovery tool.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Técnicas de Cultura de Células/métodos , Ensaios de Triagem em Larga Escala/métodos , Animais , Anticorpos Monoclonais/análise , Células CHO , Cromatografia Líquida de Alta Pressão , Cricetinae , Cricetulus , Eletroforese em Gel de Poliacrilamida , Proteína Estafilocócica A/química , Proteína Estafilocócica A/metabolismo
13.
Biotechnol Bioeng ; 112(5): 977-86, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25502369

RESUMO

Transient gene expression (TGE) is a rapid method for the production of recombinant proteins in mammalian cells. While the volumetric productivity of TGE has improved significantly over the past decade, most methods involve extensive cell line engineering and plasmid vector optimization in addition to long fed batch cultures lasting up to 21 days. Our colleagues have recently reported the development of a CHO K1SV GS-KO host cell line. By creating a bi-allelic glutamine synthetase knock out of the original CHOK1SV host cell line, they were able to improve the efficiency of generating high producing stable CHO lines for drug product manufacturing. We developed a TGE method using the same CHO K1SV GS-KO host cell line without any further cell line engineering. We also refrained from performing plasmid vector engineering. Our objective was to setup a TGE process to mimic protein quality attributes obtained from stable CHO cell line. Polyethyleneimine (PEI)-mediated transfections were performed at high cell density (4 × 10(6) cells/mL) followed by immediate growth arrest at 32 °C for 7 days. Optimizing DNA and PEI concentrations proved to be important. Interestingly, found the direct transfection method (where DNA and PEI were added sequentially) to be superior to the more common indirect method (where DNA and PEI are first pre-complexed). Moreover, the addition of a single feed solution and a polar solvent (N,N dimethylacetamide) significantly increased product titers. The scalability of process from 2 mL to 2 L was demonstrated using multiple proteins and multiple expression volumes. Using this simple, short, 7-day TGE process, we were able to successfully produce 54 unique proteins in a fraction of the time that would have been required to produce the respective stable CHO cell lines. The list of 54 unique proteins includes mAbs, bispecific antibodies, and Fc-fusion proteins. Antibody titers of up to 350 mg/L were achieved with the simple 7-day process. Titers were increased to 1 g/L by extending the culture to 16 days. We also present two case studies comparing product quality of material generated by transient HEK293, transient CHO K1SV GS-KO, and stable CHO K1SV KO pool. Protein from transient CHO was more representative of stable CHO protein compared to protein produced from HEK293.


Assuntos
Células CHO/metabolismo , Glutamato-Amônia Ligase/genética , Transfecção/instrumentação , Animais , Anticorpos Monoclonais/genética , Células CHO/citologia , Contagem de Células , Cricetulus , DNA/administração & dosagem , DNA/genética , Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Polietilenoimina/metabolismo , Proteínas Recombinantes/genética
14.
J Biotechnol ; 157(1): 198-206, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22100268

RESUMO

Pichia pastoris is a methylotropic yeast that has gained great importance as an organism for protein expression in recent years. Here, we report the expression of recombinant human erythropoietin (rhEPO) in glycoengineered P. pastoris. We show that glycosylation fidelity is maintained in fermentation volumes spanning six orders of magnitude and that the protein can be purified to high homogeneity. In order to increase the half-life of rhEPO, the purified protein was coupled to polyethylene glycol (PEG) and then compared to the currently marketed erythropoiesis stimulating agent, Aranesp(®) (darbepoetin). In in vitro cell proliferation assays the PEGylated protein was slightly, and the non-PEGylated protein was significantly more active than comparator. Pharmacodynamics as well as pharmacokinetic activity of PEGylated rhEPO in animals was comparable to that of Aranesp(®). Taken together, our results show that glycoengineered P. pastoris is a suitable production host for rhEPO, yielding an active biologic that is comparable to those produced in current mammalian host systems.


Assuntos
Eritropoetina/biossíntese , Pichia/metabolismo , Engenharia de Proteínas/métodos , Animais , Proliferação de Células/efeitos dos fármacos , Darbepoetina alfa , Eritropoetina/análogos & derivados , Eritropoetina/sangue , Eritropoetina/genética , Eritropoetina/farmacocinética , Eritropoetina/farmacologia , Feminino , Glicosilação , Humanos , Masculino , Camundongos , Pichia/genética , Polietilenoglicóis , Polissacarídeos/química , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
15.
J Ind Microbiol Biotechnol ; 37(9): 961-71, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20711797

RESUMO

The methylotrophic yeast Pichia pastoris has recently been engineered to express therapeutic glycoproteins with uniform human N-glycans at high titers. In contrast to the current art where producing therapeutic proteins in mammalian cell lines yields a final product with heterogeneous N-glycans, proteins expressed in glycoengineered P. pastoris can be designed to carry a specific, preselected glycoform. However, significant variability exists in fermentation performance between genotypically similar clones with respect to cell fitness, secreted protein titer, and glycan homogeneity. Here, we describe a novel, multidimensional screening process that combines high and medium throughput tools to identify cell lines producing monoclonal antibodies (mAbs). These cell lines must satisfy multiple selection criteria (high titer, uniform N-glycans and cell robustness) and be compatible with our large-scale production platform process. Using this selection process, we were able to isolate a mAb-expressing strain yielding a titer (after protein A purification) in excess of 1 g/l in 0.5-l bioreactors.


Assuntos
Anticorpos Monoclonais/biossíntese , Engenharia Genética , Glicoproteínas/biossíntese , Pichia/isolamento & purificação , Proteínas Recombinantes/biossíntese , Anticorpos Monoclonais/genética , Reatores Biológicos , Técnicas de Cultura de Células , Linhagem Celular , DNA Fúngico/genética , Fermentação , Expressão Gênica , Glicoproteínas/genética , Glicosilação , Humanos , Técnicas Microbiológicas , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes/genética , Seleção Genética , Transformação Genética
16.
Protein Expr Purif ; 46(2): 179-88, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16249097

RESUMO

Self-assembling peptides have emerged as an attractive scaffold material for tissue engineering, yet the expense associated with solid phase chemical synthesis has limited their broad use. In addition, the fidelity of chemical synthesis constrains the length of polypeptides that can be produced homogeneously by this method. Template-derived biosynthesis by recombinant DNA technology may overcome both of these problems. However, recovery of polypeptides from recombinant protein expression systems typically involves multi-step purification schemes. In this study, we report an integrated approach to recombinantly produce and purify self-assembling peptides from the recently developed expression host Ralstonia eutropha. The purification is based on the specific affinity of carbohydrate binding modules (CBMs) to cellulose. In a first step, we identified CBMs that express well in R. eutropha by assembling a fusion library of green fluorescent protein (GFP) and CBMs and determining the fluorescence of cell-free extracts. Three GFP::CBM fusions were found to express at levels similar to GFP alone, of which two CBMs were able to mediate cellulose binding of the GFP::CBM fusion. These two CBMs were then fused to multiple repeats of the self-assembling peptide RAD16-I::E (N-RADARADARADARADAE-C). The fusion protein CBM::E::(RAD16-I::E)4 was expressed in R. eutropha and purified using the CBM's affinity for cellulose. Subsequent proteolytic cleavage with endoproteinase GluC liberated RAD16-I::E peptide monomers with similar properties to the chemically synthesized counterpart RAD16-I.


Assuntos
Proteínas de Transporte/biossíntese , Cupriavidus necator/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Cupriavidus necator/crescimento & desenvolvimento , Expressão Gênica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Engenharia Tecidual/métodos
17.
Appl Environ Microbiol ; 71(10): 5735-42, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16204482

RESUMO

Protein purification of recombinant proteins constitutes a significant cost of biomanufacturing and various efforts have been directed at developing more efficient purification methods. We describe a protein purification scheme wherein Ralstonia eutropha is used to produce its own "affinity matrix," thereby eliminating the need for external chromatographic purification steps. This approach is based on the specific interaction of phasin proteins with granules of the intracellular polymer polyhydroxybutyrate (PHB). By creating in-frame fusions of phasins and green fluorescent protein (GFP) as a model protein, we demonstrated that GFP can be efficiently sequestered to the surface of PHB granules. In a second step, we generated a phasin-intein-GFP fusion, wherein the self-cleaving intein can be activated by the addition of thiols. This construct allowed for the controlled binding and release of essentially pure GFP in a single separation step. Finally, pure, active beta-galactosidase was obtained in a single step using the above described method.


Assuntos
Cupriavidus necator/metabolismo , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biotecnologia/métodos , Cupriavidus necator/genética , Cupriavidus necator/crescimento & desenvolvimento , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ditiotreitol , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hidroxibutiratos/metabolismo , Inteínas/genética , Poliésteres/metabolismo , Proteínas Recombinantes de Fusão/genética
18.
Protein Expr Purif ; 38(2): 264-71, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15555942

RESUMO

We report further development of a novel recombinant protein expression system based on the Gram-negative bacterium, Ralstonia eutropha. In this study, we were able to express soluble, active, organophosphohydrolase (OPH), a protein that is prone to inclusion body formation in Escherichia coli, at titers greater than 10 g/L in high cell density fermentation. This represents a titer that is approximately 100-fold greater than titers previously reported in E. coli for this enzyme. R. eutropha strains expressing OPH were generated in two cloning steps. First, the T7 RNA polymerase gene was placed under the control of the strong, inducible phaP promoter and integrated into the phaP locus of R. eutropha NCIMB 40124. Second, a single copy of the oph gene under control of the T7 promoter was randomly integrated into the chromosome using a transposon cloning vector.


Assuntos
Cupriavidus necator/enzimologia , RNA Polimerases Dirigidas por DNA/genética , Regulação Enzimológica da Expressão Gênica , Monoéster Fosfórico Hidrolases/genética , Clonagem Molecular , Cupriavidus necator/metabolismo , Ativação Enzimática , Escherichia coli/enzimologia , Fermentação , Vetores Genéticos/genética , Monoéster Fosfórico Hidrolases/metabolismo , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Proteínas Virais
19.
Biotechnol Bioeng ; 84(1): 114-20, 2003 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-12910550

RESUMO

We have previously reported the development of a novel protein expression system based on Ralstonia eutropha. In this study we report on the influence of gene copynumber on recombinant protein expression in R. eutropha. We compare recombinant gene stability and expression levels of chromosomal integration with a plasmid-based expression system. Single, double, and triple copies of a gene encoding organophosphohydrolase (OPH), an enzyme prone to inclusion-body formation in E. coli, were integrated into the R. eutropha chromosome. A linear increase between the concentration of soluble, active OPH and gene copynumber was found. Using a triple-copy integrant, we were able to produce approximately 4.3 g/L of OPH in a high-cell-density fermentation. This represents the highest titer reported to date for this enzyme, and is approximately 30 times greater than expression levels reported in E. coli.


Assuntos
Arildialquilfosfatase/biossíntese , Arildialquilfosfatase/genética , Cupriavidus necator/enzimologia , Cupriavidus necator/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Engenharia de Proteínas/métodos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Contagem de Células , Técnicas de Cultura de Células/métodos , Divisão Celular/fisiologia , Cupriavidus necator/citologia , Cupriavidus necator/crescimento & desenvolvimento , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Dosagem de Genes , Especificidade da Espécie
20.
Appl Environ Microbiol ; 68(12): 5925-32, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12450812

RESUMO

We describe the development of a novel protein expression system based on the industrial fermentation organism Ralstonia eutropha (formerly known as Alcaligenes eutrophus) NCIMB 40124. This new system overcomes some of the shortcomings of traditional Escherichia coli-based protein expression systems, particularly the propensity of such systems to form inclusion bodies during high-level expression. Using a proteomics approach, we identified promoters that can be induced by simple process parameters or medium compositions in high-density cell culture or shake flasks, respectively. By combining newly developed molecular biological tools with a high-cell-density fermentation process, we were able to produce high levels (>1 g/liter) of soluble, active organophosphohydrolase, a model enzyme prone to inclusion body formation in E. coli.


Assuntos
Cupriavidus necator/metabolismo , Fermentação , Proteínas Recombinantes/biossíntese , Microbiologia Industrial , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...