Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Front Immunol ; 10: 2630, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31798579

RESUMO

Immune cell therapies based on the integration of synthetic antigen receptors comprise a powerful strategy for the treatment of diverse diseases, most notably T cells engineered to express chimeric antigen receptors (CAR) for targeted cancer therapy. In addition to T lymphocytes, B lymphocytes may also represent valuable immune cells that can be engineered for therapeutic purposes such as protein replacement therapy or recombinant antibody production. In this article, we report a promising concept for the molecular design, optimization, and genomic integration of a novel class of synthetic antigen receptors, chimeric B cell receptors (CBCR). We initially optimized CBCR expression and detection by modifying the extracellular surface tag, the transmembrane regions and intracellular signaling domains. For this purpose, we stably integrated a series of CBCR variants using CRISPR-Cas9 into immortalized B cell hybridomas. Subsequently, we developed a reliable and consistent pipeline to precisely introduce cassettes of several kb size into the genome of primary murine B cells also using CRISPR-Cas9 induced HDR. Finally, we were able to show the robust surface expression and antigen recognition of a synthetic CBCR in primary B cells. We anticipate CBCRs and our approach for engineering primary B cells will be a valuable tool for the advancement of future B cell- based immune cell therapies.


Assuntos
Linfócitos B , Edição de Genes/métodos , Engenharia de Proteínas/métodos , Receptores de Antígenos de Linfócitos B/genética , Receptores Artificiais/genética , Animais , Sistemas CRISPR-Cas , Camundongos , Receptores de Antígenos de Linfócitos B/imunologia , Receptores Artificiais/imunologia
2.
MAbs ; 11(8): 1367-1380, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31478465

RESUMO

Antibody engineering in mammalian cells offers the important advantage of expression and screening of libraries in their native conformation, increasing the likelihood of generating candidates with more favorable molecular properties. Major advances in cellular engineering enabled by CRISPR-Cas9 genome editing have made it possible to expand the use of mammalian cells in biotechnological applications. Here, we describe an antibody engineering and screening approach where complete variable light (VL) and heavy (VH) chain cassette libraries are stably integrated into the genome of hybridoma cells by enhanced Cas9-driven homology-directed repair (HDR), resulting in their surface display and secretion. By developing an improved HDR donor format that utilizes in situ linearization, we are able to achieve >15-fold improvement of genomic integration, resulting in a screening workflow that only requires a simple plasmid electroporation. This proved suitable for different applications in antibody discovery and engineering. By integrating and screening an immune library obtained from the variable gene repertoire of an immunized mouse, we could isolate a diverse panel of >40 unique antigen-binding variants. Additionally, we successfully performed affinity maturation by directed evolution screening of an antibody library based on random mutagenesis, leading to the isolation of several clones with affinities in the picomolar range.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes , Biblioteca Gênica , Hibridomas/metabolismo , Anticorpos de Cadeia Única , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo
3.
Methods Mol Biol ; 1850: 79-111, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30242682

RESUMO

From the perspective of academic and small research laboratories, the most common and practical strategy for recombinant expression of full-length monoclonal antibodies is to perform transient plasmid transfection of mammalian cells, resulting in small-scale and limited protein production. The generation of stable antibody producing mammalian cell lines enables larger-scale and consistent expression, however this approach is rarely pursued due to the time-consuming and expensive process of single colony screening and characterization. In order to bridge the gap between the simplicity of transient transfection and consistent production by stable cell lines, we describe a method to stably integrate antibody genes into the endogenous immunogenomic loci of hybridoma cells using CRISPR/Cas9 genome editing. Initially, the antibody variable light (VL) chain is deleted by multiplexed Cas9 cleavage; subsequently, the variable heavy (VH) chain is replaced by a fluorescent reporter gene (mRuby) by Cas9-assisted homology-directed repair (HDR). This cell line is customized by replacing mRuby with a synthetic antibody (consisting of a VL, light constant region and VH) by once again using Cas9-assisted HDR. Due to hybridomas' inherent ability to surface display and secrete antibodies, they provide a suitable host for both the selection and the production process, substantially streamlining the process for stable cell line generation, and thus we refer to this platform as plug-and-(dis)play (PnP) hybridomas.


Assuntos
Edição de Genes/métodos , Hibridomas/metabolismo , Animais , Anticorpos Monoclonais , Sistemas CRISPR-Cas/genética , Linhagem Celular , Humanos
4.
Nucleic Acids Res ; 46(14): 7436-7449, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-29931269

RESUMO

Antibody engineering is often performed to improve therapeutic properties by directed evolution, usually by high-throughput screening of phage or yeast display libraries. Engineering antibodies in mammalian cells offer advantages associated with expression in their final therapeutic format (full-length glycosylated IgG); however, the inability to express large and diverse libraries severely limits their potential throughput. To address this limitation, we have developed homology-directed mutagenesis (HDM), a novel method which extends the concept of CRISPR/Cas9-mediated homology-directed repair (HDR). HDM leverages oligonucleotides with degenerate codons to generate site-directed mutagenesis libraries in mammalian cells. By improving HDR to a robust efficiency of 15-35% and combining mammalian display screening with next-generation sequencing, we validated this approach can be used for key applications in antibody engineering at high-throughput: rational library construction, novel variant discovery, affinity maturation and deep mutational scanning (DMS). We anticipate that HDM will be a valuable tool for engineering and optimizing antibodies in mammalian cells, and eventually enable directed evolution of other complex proteins and cellular therapeutics.


Assuntos
Anticorpos/imunologia , Sistemas CRISPR-Cas , Mutagênese Sítio-Dirigida , Engenharia de Proteínas/métodos , Sequência de Aminoácidos , Animais , Anticorpos/genética , Anticorpos/metabolismo , Afinidade de Anticorpos/genética , Afinidade de Anticorpos/imunologia , Sequência de Bases , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Hibridomas , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Reparo de DNA por Recombinação
5.
Immunology ; 153(1): 31-41, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28898398

RESUMO

Monoclonal antibody discovery and engineering is a field that has traditionally been dominated by high-throughput screening platforms (e.g. hybridomas and surface display). In recent years the emergence of high-throughput sequencing has made it possible to obtain large-scale information on antibody repertoire diversity. Additionally, it has now become more routine to perform high-throughput sequencing on antibody repertoires to also directly discover antibodies. In this review, we provide an overview of the progress in this field to date and show how high-throughput screening and sequencing are converging to deliver powerful new workflows for monoclonal antibody discovery and engineering.


Assuntos
Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Descoberta de Drogas , Engenharia Genética , Sequenciamento de Nucleotídeos em Larga Escala , Ensaios de Triagem em Larga Escala , Animais , Anticorpos Monoclonais/farmacologia , Descoberta de Drogas/métodos , Engenharia Genética/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Ensaios de Triagem em Larga Escala/métodos , Humanos
6.
Sci Rep ; 7: 45775, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28374766

RESUMO

The development of programmable nucleases has enabled the application of new genome engineering strategies for cellular immunotherapy. While targeted nucleases have mostly been used to knock-out or knock-in genes in immune cells, the scarless exchange of entire immunogenomic alleles would be of great interest. In particular, reprogramming the polymorphic MHC locus could enable the creation of matched donors for allogeneic cellular transplantation. Here we show a proof-of-concept for reprogramming MHC-specificity by performing CRISPR-Cas9-assisted cassette exchange. Using murine antigen presenting cell lines (RAW264.7 macrophages), we demonstrate that the generation of Cas9-induced double-stranded breaks flanking the native MHC-I H2-Kd locus led to exchange of an orthogonal H2-Kb allele. MHC surface expression allowed for easy selection of reprogrammed cells by flow cytometry, thus obviating the need for additional selection markers. MHC-reprogrammed cells were fully functional as they could present H2-Kd-restricted peptide and activate cognate T cells. Finally, we investigated the role of various donor template formats on exchange efficiency, discovering that templates that underwent in situ linearization resulted in the highest MHC-reprogramming efficiency. These findings highlight a potential new approach for the correcting of MHC mismatches in cellular transplantation.


Assuntos
Sistemas CRISPR-Cas , Complexo Principal de Histocompatibilidade/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Fibroblastos , Citometria de Fluxo , Antígenos H-2/genética , Antígenos H-2/imunologia , Macrófagos/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Camundongos , Camundongos Endogâmicos BALB C
7.
Nat Commun ; 7: 12535, 2016 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-27531490

RESUMO

Hybridomas, fusions of primary mouse B cells and myelomas, are stable, rapidly-proliferating cell lines widely utilized for antibody screening and production. Antibody specificity of a hybridoma clone is determined by the immunoglobulin sequence of the primary B cell. Here we report a platform for rapid reprogramming of hybridoma antibody specificity by immunogenomic engineering. Here we use CRISPR-Cas9 to generate double-stranded breaks in immunoglobulin loci, enabling deletion of the native variable light chain and replacement of the endogenous variable heavy chain with a fluorescent reporter protein (mRuby). New antibody genes are introduced by Cas9-targeting of mRuby for replacement with a donor construct encoding a light chain and a variable heavy chain, resulting in full-length antibody expression. Since hybridomas surface express and secrete antibodies, reprogrammed cells are isolated using flow cytometry and cell culture supernatant is used for antibody production. Plug-and-(dis)play hybridomas can be reprogrammed with only a single transfection and screening step.


Assuntos
Engenharia Genética/métodos , Hibridomas/imunologia , Animais , Anticorpos/metabolismo , Especificidade de Anticorpos , Antígenos/metabolismo , Linhagem Celular , Edição de Genes , Camundongos , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA