Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cancer Immunol Res ; 11(7): 946-961, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37257169

RESUMO

Despite the remarkable success of autologous chimeric antigen receptor (CAR) T cells, some patients relapse due to tumor antigen escape and low or uneven antigen expression, among other mechanisms. Therapeutic options after relapse are limited, emphasizing the need to optimize current approaches. In addition, there is a need to develop allogeneic "off-the-shelf" therapies from healthy donors that are readily available at the time of treatment decision and can overcome limitations of current autologous approaches. To address both challenges simultaneously, we generated a CD20xCD22 dual allogeneic CAR T cell. Herein, we demonstrate that allogeneic CD20x22 CAR T cells display robust, sustained and dose-dependent activity in vitro and in vivo, while efficiently targeting primary B-cell non-Hodgkin lymphoma (B-NHL) samples with heterogeneous levels of CD22 and CD20. Altogether, we provide preclinical proof-of-concept data for an allogeneic dual CAR T cell to overcome current mechanisms of resistance to CAR T-cell therapies in B-NHL, while providing a potential alternative to CD19 targeting.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Linfoma de Células B , Humanos , Receptores de Antígenos de Linfócitos T , Recidiva Local de Neoplasia , Linfócitos T , Linfócitos B , Imunoterapia Adotiva , Antígenos CD19
2.
Nat Commun ; 13(1): 3453, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35773273

RESUMO

Universal CAR T-cell therapies are poised to revolutionize cancer treatment and to improve patient outcomes. However, realizing these advantages in an allogeneic setting requires universal CAR T-cells that can kill target tumor cells, avoid depletion by the host immune system, and proliferate without attacking host tissues. Here, we describe the development of a novel immune-evasive universal CAR T-cells scaffold using precise TALEN-mediated gene editing and DNA matrices vectorized by recombinant adeno-associated virus 6. We simultaneously disrupt and repurpose the endogenous TRAC and B2M loci to generate TCRαß- and HLA-ABC-deficient T-cells expressing the CAR construct and the NK-inhibitor named HLA-E. This highly efficient gene editing process enables the engineered T-cells to evade NK cell and alloresponsive T-cell attacks and extend their persistence and antitumor activity in the presence of cytotoxic levels of NK cell in vivo and in vitro, respectively. This scaffold could enable the broad use of universal CAR T-cells in allogeneic settings and holds great promise for clinical applications.


Assuntos
Edição de Genes , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Humanos , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T
3.
Cancer Res ; 75(18): 3853-64, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26183927

RESUMO

Adoptive immunotherapy using autologous T cells endowed with chimeric antigen receptors (CAR) has emerged as a powerful means of treating cancer. However, a limitation of this approach is that autologous CAR T cells must be generated on a custom-made basis. Here we show that electroporation of transcription activator-like effector nuclease (TALEN) mRNA allows highly efficient multiplex gene editing in primary human T cells. We use this TALEN-mediated editing approach to develop a process for the large-scale manufacturing of T cells deficient in expression of both their αß T-cell receptor (TCR) and CD52, a protein targeted by alemtuzumab, a chemotherapeutic agent. Functionally, T cells manufactured with this process do not mediate graft-versus-host reactions and are rendered resistant to destruction by alemtuzumab. These characteristics enable the administration of alemtuzumab concurrently or prior to engineered T cells, supporting their engraftment. Furthermore, endowing the TALEN-engineered cells with a CD19 CAR led to efficient destruction of CD19(+) tumor targets even in the presence of the chemotherapeutic agent. These results demonstrate the applicability of TALEN-mediated genome editing to a scalable process, which enables the manufacturing of third-party CAR T-cell immunotherapies against arbitrary targets. As such, CAR T-cell immunotherapies can therefore be used in an "off-the-shelf" manner akin to other biologic immunopharmaceuticals


Assuntos
Técnicas de Inativação de Genes , Imunoterapia Adotiva , Linfócitos T/transplante , Alemtuzumab , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Antígenos CD/genética , Antígenos CD19/imunologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Sequência de Bases , Antígeno CD52 , Citotoxicidade Imunológica , Resistência a Medicamentos , Glicoproteínas/deficiência , Glicoproteínas/genética , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Ativação Linfocitária , Linfoma/terapia , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , RNA Mensageiro , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/deficiência , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nucleic Acids Res ; 40(13): 6367-79, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22467209

RESUMO

The ability to specifically engineer the genome of living cells at precise locations using rare-cutting designer endonucleases has broad implications for biotechnology and medicine, particularly for functional genomics, transgenics and gene therapy. However, the potential impact of chromosomal context and epigenetics on designer endonuclease-mediated genome editing is poorly understood. To address this question, we conducted a comprehensive analysis on the efficacy of 37 endonucleases derived from the quintessential I-CreI meganuclease that were specifically designed to cleave 39 different genomic targets. The analysis revealed that the efficiency of targeted mutagenesis at a given chromosomal locus is predictive of that of homologous gene targeting. Consequently, a strong genome-wide correlation was apparent between the efficiency of targeted mutagenesis (≤ 0.1% to ≈ 6%) with that of homologous gene targeting (≤ 0.1% to ≈ 15%). In contrast, the efficiency of targeted mutagenesis or homologous gene targeting at a given chromosomal locus does not correlate with the activity of individual endonucleases on transiently transfected substrates. Finally, we demonstrate that chromatin accessibility modulates the efficacy of rare-cutting endonucleases, accounting for strong position effects. Thus, chromosomal context and epigenetic mechanisms may play a major role in the efficiency rare-cutting endonuclease-induced genome engineering.


Assuntos
Efeitos da Posição Cromossômica , Enzimas de Restrição do DNA/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Enzimas de Restrição do DNA/química , Marcação de Genes , Engenharia Genética , Genoma Humano , Humanos , Mutagênese
5.
Mol Ther ; 19(4): 694-702, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21224832

RESUMO

Herpes simplex virus type 1 (HSV1) is a major health problem. As for most viral diseases, current antiviral treatments are based on the inhibition of viral replication once it has already started. As a consequence, they impair neither the viral cycle at its early stages nor the latent form of the virus, and thus cannot be considered as real preventive treatments. Latent HSV1 virus could be addressed by rare cutting endonucleases, such as meganucleases. With the aim of a proof of concept study, we generated several meganucleases recognizing HSV1 sequences, and assessed their antiviral activity in cultured cells. We demonstrate that expression of these proteins in African green monkey kidney fibroblast (COS-7) and BSR cells inhibits infection by HSV1, at low and moderate multiplicities of infection (MOIs), inducing a significant reduction of the viral load. Furthermore, the remaining viral genomes display a high rate of mutation (up to 16%) at the meganuclease cleavage site, consistent with a mechanism of action based on the cleavage of the viral genome. This specific mechanism of action qualifies meganucleases as an alternative class of antiviral agent, with the potential to address replicative as well as latent DNA viral forms.


Assuntos
Desoxirribonucleases/metabolismo , Infecções por Herpesviridae/prevenção & controle , Animais , Western Blotting , Células CHO , Células COS , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Desoxirribonucleases/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Humanos
6.
Nucleic Acids Res ; 39(2): 729-43, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20846960

RESUMO

Homing endonucleases recognize long target DNA sequences generating an accurate double-strand break that promotes gene targeting through homologous recombination. We have modified the homodimeric I-CreI endonuclease through protein engineering to target a specific DNA sequence within the human RAG1 gene. Mutations in RAG1 produce severe combined immunodeficiency (SCID), a monogenic disease leading to defective immune response in the individuals, leaving them vulnerable to infectious diseases. The structures of two engineered heterodimeric variants and one single-chain variant of I-CreI, in complex with a 24-bp oligonucleotide of the human RAG1 gene sequence, show how the DNA binding is achieved through interactions in the major groove. In addition, the introduction of the G19S mutation in the neighborhood of the catalytic site lowers the reaction energy barrier for DNA cleavage without compromising DNA recognition. Gene-targeting experiments in human cell lines show that the designed single-chain molecule preserves its in vivo activity with higher specificity, further enhanced by the G19S mutation. This is the first time that an engineered meganuclease variant targets the human RAG1 locus by stimulating homologous recombination in human cell lines up to 265 bp away from the cleavage site. Our analysis illustrates the key features for à la carte procedure in protein-DNA recognition design, opening new possibilities for SCID patients whose illness can be treated ex vivo.


Assuntos
Reparo do DNA , Enzimas de Restrição do DNA/química , Genes RAG-1 , Linhagem Celular , DNA/química , Clivagem do DNA , Enzimas de Restrição do DNA/genética , Enzimas de Restrição do DNA/metabolismo , Marcação de Genes , Loci Gênicos , Humanos , Modelos Moleculares , Mutação , Conformação de Ácido Nucleico , Ligação Proteica , Engenharia de Proteínas , Recombinação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA