Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 886429, 2022.
Article in English | MEDLINE | ID: mdl-35720306

ABSTRACT

In recent years, immunotherapy for cancer has become mainstream with several products now authorized for therapeutic use in the clinic and are becoming the standard of care for some malignancies. Chimeric antigen receptor (CAR)-T cell therapies have demonstrated substantial efficacy for the treatment of hematological malignancies; however, they are complex and currently expensive to manufacture, and they can generate life-threatening adverse events such as cytokine release syndrome (CRS). The limitations of current CAR-T cells therapies have spurred an interest in alternative immunotherapy approaches with safer risk profiles and with less restrictive manufacturing constraints. Natural killer (NK) cells are a population of immune effector cells with potent anti-viral and anti-tumor activity; they have the capacity to swiftly recognize and kill cancer cells without the need of prior stimulation. Although NK cells are naturally equipped with cytotoxic potential, a growing body of evidence shows the added benefit of engineering them to better target tumor cells, persist longer in the host, and be fitter to resist the hostile tumor microenvironment (TME). NK-cell-based immunotherapies allow for the development of allogeneic off-the-shelf products, which have the potential to be less expensive and readily available for patients in need. In this review, we will focus on the advances in the development of engineering of NK cells for cancer immunotherapy. We will discuss the sourcing of NK cells, the technologies available to engineer NK cells, current clinical trials utilizing engineered NK cells, advances on the engineering of receptors adapted for NK cells, and stealth approaches to avoid recipient immune responses. We will conclude with comments regarding the next generation of NK cell products, i.e., armored NK cells with enhanced functionality, fitness, tumor-infiltration potential, and with the ability to overcome tumor heterogeneity and immune evasion.


Subject(s)
Hematologic Neoplasms , Neoplasms , Hematologic Neoplasms/etiology , Humans , Immunotherapy , Immunotherapy, Adoptive/adverse effects , Killer Cells, Natural , Tumor Microenvironment
2.
J Biotechnol ; 306S: 100006, 2019.
Article in English | MEDLINE | ID: mdl-34112369

ABSTRACT

Drug manufacturing processes must consistently deliver safe and effective product. A key part of achieving this is process validation utilizing Quality by Design (QbD) principles. To meet process validation requirements, process characterization (PC) studies are often performed to expand process understanding and establish an appropriate control strategy that enables the manufacturing process to consistently deliver a target product profile. Two key elements of the control strategy resulting from PC work are a list of critical process parameters (CPPs) and defined operating ranges (ORs). These are frequently derived based on mathematical models describing the relationship between process parameters and critical quality attributes (CQAs). Risk assessment and design of experiments (DOE) techniques are effectively deployed in the industry to identify parameters to study and build process understanding. However, traditional data analysis techniques do not fully utilize the data produced by these studies. In particular, stepwise regression algorithms based on p-values are prone to generate false positives and overfit data, potentially leading to unnecessarily complex control strategies. Many of the deficiencies of traditional stepwise regression can be alleviated by applying cross validation to stepwise regression algorithms, as well as Monte Carlo simulations to estimate model accuracy and predict CQA distributions. These methods can greatly enhance process understanding and assist in the selection of CPPs. A series of PC studies were performed in bioreactors to evaluate a process to produce a recombinant monoclonal antibody. The studies examined process parameters such as dissolved oxygen, pH, temperature, inoculation density, as well as cell density at two key process steps. The resulting data were analyzed using several Monte Carlo based methods. First, cross validation was used to determine model size and select parameters to be included in the model. Next, Monte Carlo cross validation was used to compare the accuracy of different models. Finally, simulated CQA profiles were generated to validate proposed ORs. This workflow provides greater process understanding based on a given PC data set and provides higher statistical confidence in both CPP selection and establishment of a control strategy.

3.
Biotechnol Prog ; 29(3): 727-37, 2013.
Article in English | MEDLINE | ID: mdl-23596153

ABSTRACT

Cell death due to apoptosis is frequently observed in large-scale manufacturing of therapeutic proteins, and can reduce product accumulation in bioreactors. Several different strategies that involve overexpression of antiapoptotic or downregulation of proapoptotic proteins have been designed in attempt to curb this problem in Chinese hamster ovary (CHO) cell culture. However, each of these designs has their own shortcomings and limits, rendering them ineffective for large-scale protein production. Recently, we have reported generation of a Bax and Bak deficient dhfr(-/-) CHO cell line using zinc-finger nucleases. Here we demonstrate that puromycin, but not methotrexate, selection can be used to generate antibody-expressing Bax and Bak deficient clones that are not only resistant to apoptosis, but that can also achieve higher titers relative to parental CHO cells due to higher cell density. Additionally, we show that Bax and Bak deficient cells have more mitochondria with healthy membrane potential, an attribute that perhaps contributes to their more potent growth compared to parental cells. Bax and Bak deficient cells do not readily apoptose, as shown by the ability to withstand high concentrations of apoptosis inducing agents, such as sodium butyrate, without a reduction in viability, growth, or titer. These traits render Bax and Bak deficient cells a potentially attractive host for production of therapeutic proteins at industrial scale.


Subject(s)
Antibodies/metabolism , Recombinant Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics , Animals , Antibodies/analysis , Antibodies/genetics , Apoptosis , Butyric Acid , CHO Cells , Cricetinae , Cricetulus , Gene Knockout Techniques , Membrane Potential, Mitochondrial/genetics , Methotrexate , Mitochondria/genetics , Puromycin , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Tetrahydrofolate Dehydrogenase/genetics
4.
Biotechnol Bioeng ; 106(5): 774-83, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20564614

ABSTRACT

IgG1 antibodies produced in Chinese hamster ovary (CHO) cells are heavily alpha1,6-fucosylated, a modification that reduces antibody-dependent cellular cytotoxicity (ADCC) and can inhibit therapeutic antibody function in vivo. Addition of fucose is catalyzed by Fut8, a alpha1,6-fucosyltransferase. FUT8(-/-) CHO cell lines produce completely nonfucosylated antibodies, but the difficulty of recapitulating the knockout in protein-production cell lines has prevented the widespread adoption of FUT8(-/-) cells as hosts for antibody production. We have created zinc-finger nucleases (ZFNs) that cleave the FUT8 gene in a region encoding the catalytic core of the enzyme, allowing the functional disruption of FUT8 in any CHO cell line. These reagents produce FUT8(-/-) CHO cells in 3 weeks at a frequency of 5% in the absence of any selection. Alternately, populations of ZFN-treated cells can be directly selected to give FUT8(-/-) cell pools in as few as 3 days. To demonstrate the utility of this method in bioprocess, FUT8 was disrupted in a CHO cell line used for stable protein production. ZFN-derived FUT8(-/-) cell lines were as transfectable as wild-type, had similar or better growth profiles, and produced equivalent amounts of antibody during transient transfection. Antibodies made in these lines completely lacked core fucosylation but had an otherwise normal glycosylation pattern. Cell lines stably expressing a model antibody were made from wild-type and ZFN-generated FUT8(-/-) cells. Clones from both lines had equivalent titer, specific productivity distributions, and integrated viable cell counts. Antibody titer in the best ZFN-generated FUT8(-/-) cell lines was fourfold higher than in the best-producing clones of FUT8(-/-) cells made by standard homologous recombination in a different CHO subtype. These data demonstrate the straightforward, ZFN-mediated transfer of the Fut8- phenotype to a production CHO cell line without adverse phenotypic effects. This process will speed the production of highly active, completely nonfucosylated therapeutic antibodies.


Subject(s)
DNA/metabolism , Deoxyribonucleases/metabolism , Fucosyltransferases/genetics , Gene Deletion , Genetic Techniques , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Biotechnology/methods , CHO Cells , Cell Culture Techniques , Cricetinae , Cricetulus , Zinc Fingers
5.
Biotechnol Bioeng ; 105(2): 330-40, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19777580

ABSTRACT

Anoxic and metabolic stresses in large-scale cell culture during biopharmaceutical production can induce apoptosis. Strategies designed to ameliorate the problem of apoptosis in cell culture have focused on mRNA knockdown of pro-apoptotic proteins and over-expression of anti-apoptotic ones. Apoptosis in cell culture involves mitochondrial permeabilization by the pro-apoptotic Bak and Bax proteins; activity of either protein is sufficient to permit apoptosis. We demonstrate here the complete and permanent elimination of both the Bak and Bax proteins in combination in Chinese hamster ovary (CHO) cells using zinc-finger nuclease-mediated gene disruption. Zinc-finger nuclease cleavage of BAX and BAK followed by inaccurate DNA repair resulted in knockout of both genes. Cells lacking Bax and Bak grow normally but fail to activate caspases in response to apoptotic stimuli. When grown using scale-down systems under conditions that mimic growth in large-scale bioreactors they are significantly more resistant to apoptosis induced by starvation, staurosporine, and sodium butyrate. When grown under starvation conditions, BAX- and BAK-deleted cells produce two- to fivefold more IgG than wild-type CHO cells. Under normal growth conditions in suspension culture in shake flasks, double-knockout cultures achieve equal or higher cell densities than unmodified wild-type cultures and reach viable cell densities relevant for large-scale industrial protein production.


Subject(s)
Apoptosis , Biotechnology/methods , Cricetulus/genetics , bcl-2-Associated X Protein/genetics , Animals , Base Sequence , CHO Cells , Cloning, Molecular , Cricetinae , Female , Gene Deletion , Immunoglobulin G/biosynthesis , Molecular Sequence Data , Ovary/cytology , Zinc Fingers
6.
Cell ; 118(5): 545-53, 2004 Sep 03.
Article in English | MEDLINE | ID: mdl-15339660

ABSTRACT

Methylation of arginine residues within histone H3 has been linked to active transcription. This modification appears on the estrogen-regulated pS2 promoter when the CARM1 methyltransferase is recruited during transcriptional activation. Here we describe a process, deimination, that converts histone arginine to citrulline and antagonizes arginine methylation. We show that peptidyl arginine deiminase 4 (PADI4) specifically deiminates, arginine residues R2, R8, R17, and R26 in the H3 tail. Deimination by PADI4 prevents arginine methylation by CARM1. Dimethylation of arginines prevents deimination by PADI4 although monomethylation still allows deimination to take place. In vivo targeting experiments on an endogenous promoter demonstrate that PADI4 can repress hormone receptor-mediated gene induction. Consistent with a repressive role for PADI4, this enzyme is recruited to the pS2 promoter following hormone induction when the gene is transcriptionally downregulated. The recruitment of PADI4 coincides with deimination of the histone H3 N-terminal tail. These results define deimination as a novel mechanism for antagonizing the transcriptional induction mediated by arginine methylation.


Subject(s)
Arginine/metabolism , Citrulline/metabolism , Histones/metabolism , Imines/metabolism , Transcriptional Activation/genetics , Cell Line, Tumor , Gene Expression Regulation/genetics , Gene Expression Regulation, Enzymologic/genetics , Histones/genetics , Humans , Hydrolases/metabolism , Methylation , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary/genetics , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Protein-Arginine N-Methyltransferases/metabolism , Proteins/metabolism , Trefoil Factor-1 , Tumor Suppressor Proteins
7.
Cancer Res ; 63(24): 8968-76, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14695215

ABSTRACT

Angiogenic factors are necessary for tumor proliferation and thus are attractive therapeutic targets. In this study, we have used engineered zinc finger protein (ZFP) transcription factors (TFs) to repress expression of vascular endothelial growth factor (VEGF)-A in human cancer cell lines. We create potent transcriptional repressors by fusing a designed ZFP targeted to the VEGF-A promoter with either the ligand-binding domain of thyroid hormone receptor alpha or its viral relative, vErbA. Moreover, this ZFP-vErbA repressor binds its intended target site in vivo and mediates the specific deacetylation of histones H3 and H4 at the targeted promoter, a result that emulates the natural repression mechanism of these domains. The potential therapeutic relevance of ZFP-mediated VEGF-A repression was addressed using the highly tumorigenic glioblastoma cell line U87MG. Despite the aberrant overexpression of VEGF-A in this cell line, engineered ZFP TFs were able to repress the expression of VEGF-A by >20-fold. The VEGF-A levels observed after ZFP TF-mediated repression were comparable to those of a nonangiogenic cancer line (U251MG), suggesting that the degree of repression obtained with the ZFP TF would be sufficient to suppress tumor angiogenesis. Thus, engineered ZFP TFs are shown to be potent regulators of gene expression with therapeutic promise in the treatment of disease.


Subject(s)
Glioblastoma/metabolism , Glioblastoma/therapy , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Zinc Fingers/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/blood supply , Glioblastoma/genetics , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oncogene Proteins v-erbA/genetics , Oncogene Proteins v-erbA/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Transfection , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
8.
Proc Natl Acad Sci U S A ; 100(21): 11997-2002, 2003 Oct 14.
Article in English | MEDLINE | ID: mdl-14514889

ABSTRACT

Zinc-finger protein transcription factors (ZFP TFs) can be designed to control the expression of any desired target gene, and thus provide potential therapeutic tools for the study and treatment of disease. Here we report that a ZFP TF can repress target gene expression with single-gene specificity within the human genome. A ZFP TF repressor that binds an 18-bp recognition sequence within the promoter of the endogenous CHK2 gene gives a >10-fold reduction in CHK2 mRNA and protein. This level of repression was sufficient to generate a functional phenotype, as demonstrated by the loss of DNA damage-induced CHK2-dependent p53 phosphorylation. We determined the specificity of repression by using DNA microarrays and found that the ZFP TF repressed a single gene (CHK2) within the monitored genome in two different cell types. These data demonstrate the utility of ZFP TFs as precise tools for target validation, and highlight their potential as clinical therapeutics.


Subject(s)
Gene Expression Regulation , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers/genetics , Base Sequence , Binding Sites/genetics , Cell Line , Checkpoint Kinase 2 , DNA/genetics , DNA/metabolism , DNA Damage , Gene Expression Regulation, Enzymologic , Genome, Human , Humans , Promoter Regions, Genetic , Protein Engineering , Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
9.
Mol Cell ; 11(4): 1043-54, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12718889

ABSTRACT

p300 and CREB binding protein can both activate and repress transcription. Here, we locate the CRD1 transcriptional repression domain between residues 1017 and 1029 of p300. This region contains two copies of the sequence psiKxE that are modified by the ubiquitin-like protein SUMO-1. Mutations that reduce SUMO modification increase p300-mediated transcriptional activity and expression of a SUMO-specific protease or catalytically inactive Ubc9 relieved repression, demonstrating that p300 repression was mediated by SUMO conjugation. SUMO-modified CRD1 domain bound HDAC6 in vitro, and p300 repression was relieved by histone deacetylase inhibition and siRNA-mediated ablation of HDAC6 expression. These results reveal a mechanism controlling p300 function and suggest that SUMO-dependent repression is mediated by recruitment of HDAC6.


Subject(s)
Eukaryotic Cells/metabolism , Genes, Regulator/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , SUMO-1 Protein/metabolism , Trans-Activators/metabolism , Ubiquitin-Conjugating Enzymes , Amino Acid Sequence/genetics , Binding Sites/genetics , Cell Nucleus Structures/genetics , Cell Nucleus Structures/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , HeLa Cells , Histone Deacetylase 6 , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Ligases/genetics , Ligases/metabolism , Mutation/genetics , Nuclear Proteins/genetics , Protein Structure, Tertiary/genetics , RNA, Small Interfering , Repressor Proteins/genetics , SUMO-1 Protein/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Curr Biol ; 12(24): 2159-66, 2002 Dec 23.
Article in English | MEDLINE | ID: mdl-12498693

ABSTRACT

Covalent modifications of chromatin have emerged as key determinants of the genome's transcriptional competence. Histone H3 lysine 9 (H3K9) methylation is an epigenetic signal that is recognized by HP1 and correlates with gene silencing in a variety of organisms. Discovery of the enzymes that catalyze H3K9 methylation has identified a second gene-specific function for this modification in transcriptional repression. Whether H3K9 methylation is causative in the initiation and establishment of gene repression or is a byproduct of the process leading to the repressed state remains unknown. To investigate the role of HMTs and specifically H3K9 methylation in gene repression, we have employed engineered zinc-finger transcription factors (ZFPs) to target HMT activity to a specific endogenous gene. By utilizing ZFPs that recognize the promoter of the endogenous VEGF-A gene, and thus employing this chromosomal locus as an in vivo reporter, we show that ZFPs linked to a minimal catalytic HMT domain affect local methylation of histone H3K9 and the consequent repression of target gene expression. Furthermore, amino acid substitutions within the HMT that ablate its catalytic activity effectively eliminate the ability of the ZFP fusions to repress transcription. Thus, H3K9 methylation is a primary signal that is sufficient for initiating a gene repression pathway in vivo.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Repressor Proteins/metabolism , Cells, Cultured , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Lysine/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A , Zinc Fingers
11.
Cell Cycle ; 1(5): 343-50, 2002.
Article in English | MEDLINE | ID: mdl-12461297

ABSTRACT

The tumor suppressors p300 and CREB-binding protein (CBP) are both multifunctional transcriptional coactivators. We have previously found that the cyclin dependent kinase (CDK) inhibitor p21(WAF1/CIP1) can stimulate transactivation by p300 and CBP through inhibiting transcriptional repression by a discrete domain within these proteins termed CRD1. Given the large number of p300/CBP associated functions, it is unlikely that p21 regulates the expression of every gene under their control, however. Here we have investigated the factors that help determine this specificity. We have discovered that while CRD1 can repress the activity of p300 at multiple promoters, induction of transcription by p21 though this motif is highly variable. Analysis of this effect revealed that p21 inducibility is determined by sequences flanking the TATA box. Significantly, p21 regulation of CRD1 domain function is independent of Cyclin /CDK inhibition suggesting a novel function of this protein. p21 does not interact directly with the CRD1 motif, however. These results give further insight into how regulators of cell growth and tumorigenesis, such as p21, can specifically target and induce the expression of select groups of genes.


Subject(s)
Cyclin-Dependent Kinases/physiology , Cyclins/physiology , Nuclear Proteins/chemistry , Promoter Regions, Genetic , Trans-Activators/chemistry , Transcriptional Activation , Amino Acid Motifs , Base Sequence , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Molecular Sequence Data , Nuclear Proteins/metabolism , Sequence Alignment , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...