Your browser doesn't support javascript.
loading
Novel extragenic genomic safe harbors for precise therapeutic T-cell engineering.
Odak, Ashlesha; Yuan, Han; Feucht, Judith; Cantu, Vito Adrian; Mansilla-Soto, Jorge; Kogel, Friederike; Eyquem, Justin; Everett, John; Bushman, Frederic D; Leslie, Christina S; Sadelain, Michel.
Afiliación
  • Odak A; Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Yuan H; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY.
  • Feucht J; Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Cantu VA; Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Mansilla-Soto J; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Kogel F; Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Eyquem J; Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Everett J; Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.
  • Bushman FD; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Leslie CS; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Sadelain M; Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.
Blood ; 141(22): 2698-2712, 2023 06 01.
Article en En | MEDLINE | ID: mdl-36745870
ABSTRACT
Cell therapies that rely on engineered immune cells can be enhanced by achieving uniform and controlled transgene expression in order to maximize T-cell function and achieve predictable patient responses. Although they are effective, current genetic engineering strategies that use γ-retroviral, lentiviral, and transposon-based vectors to integrate transgenes, unavoidably produce variegated transgene expression in addition to posing a risk of insertional mutagenesis. In the setting of chimeric antigen receptor (CAR) therapy, inconsistent and random CAR expression may result in tonic signaling, T-cell exhaustion, and variable T-cell persistence. Here, we report and validate an algorithm for the identification of extragenic genomic safe harbors (GSH) that can be efficiently targeted for DNA integration and can support sustained and predictable CAR expression in human peripheral blood T cells. The algorithm is based on 7 criteria established to minimize genotoxicity by directing transgene integration away from functionally important genomic elements, maximize efficient CRISPR/Cas9-mediated targeting, and avert transgene silencing over time. T cells engineered to express a CD19 CAR at GSH6, which meets all 7 criteria, are curative at low cell dose in a mouse model of acute lymphoblastic leukemia, matching the potency of CAR T cells engineered at the TRAC locus and effectively resisting tumor rechallenge 100 days after their infusion. The identification of functional extragenic GSHs thus expands the human genome available for therapeutic precision engineering.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Receptores de Antígenos de Linfocitos T / Linfocitos T Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Revista: Blood Año: 2023 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Receptores de Antígenos de Linfocitos T / Linfocitos T Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Revista: Blood Año: 2023 Tipo del documento: Article