Your browser doesn't support javascript.
loading
An engineered IL-2 partial agonist promotes CD8+ T cell stemness.
Mo, Fei; Yu, Zhiya; Li, Peng; Oh, Jangsuk; Spolski, Rosanne; Zhao, Liang; Glassman, Caleb R; Yamamoto, Tori N; Chen, Yun; Golebiowski, Filip M; Hermans, Dalton; Majri-Morrison, Sonia; Picton, Lora K; Liao, Wei; Ren, Min; Zhuang, Xiaoxuan; Mitra, Suman; Lin, Jian-Xin; Gattinoni, Luca; Powell, Jonathan D; Restifo, Nicholas P; Garcia, K Christopher; Leonard, Warren J.
Afiliação
  • Mo F; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Yu Z; Surgery Branch, National Cancer Institute, Bethesda, MD, USA.
  • Li P; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Oh J; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Spolski R; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Zhao L; Johns Hopkins University School of Medicine, Baltimore, MD, USA.
  • Glassman CR; Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
  • Yamamoto TN; Surgery Branch, National Cancer Institute, Bethesda, MD, USA.
  • Chen Y; National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
  • Golebiowski FM; National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
  • Hermans D; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Majri-Morrison S; Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
  • Picton LK; Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
  • Liao W; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
  • Ren M; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Zhuang X; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Mitra S; Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
  • Lin JX; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Gattinoni L; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
  • Powell JD; Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
  • Restifo NP; Department of Functional Immune Cell Modulation, Regensburg Center for Interventional Immunology, Regensburg, Germany.
  • Garcia KC; University of Regensburg, Regenburg, Germany.
  • Leonard WJ; Johns Hopkins University School of Medicine, Baltimore, MD, USA.
Nature ; 597(7877): 544-548, 2021 09.
Article em En | MEDLINE | ID: mdl-34526724
ABSTRACT
Adoptive transfer of antigen-specific T cells represents a major advance in cancer immunotherapy, with robust clinical outcomes in some patients1. Both the number of transferred T cells and their differentiation state are critical determinants of effective responses2,3. T cells can be expanded with T cell receptor (TCR)-mediated stimulation and interleukin-2, but this can lead to differentiation into effector T cells4,5 and lower therapeutic efficacy6, whereas maintenance of a more stem-cell-like state before adoptive transfer is beneficial7. Here we show that H9T, an engineered interleukin-2 partial agonist, promotes the expansion of CD8+ T cells without driving terminal differentiation. H9T led to altered STAT5 signalling and mediated distinctive downstream transcriptional, epigenetic and metabolic programs. In addition, H9T treatment sustained the expression of T cell transcription factor 1 (TCF-1) and promoted mitochondrial fitness, thereby facilitating the maintenance of a stem-cell-like state. Moreover, TCR-transgenic and chimeric antigen receptor-modified CD8+ T cells that were expanded with H9T showed robust anti-tumour activity in vivo in mouse models of melanoma and acute lymphoblastic leukaemia. Thus, engineering cytokine variants with distinctive properties is a promising strategy for creating new molecules with translational potential.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco / Diferenciação Celular / Interleucina-2 / Linfócitos T CD8-Positivos / Proteínas Mutantes / Agonismo Parcial de Drogas Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Nature Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco / Diferenciação Celular / Interleucina-2 / Linfócitos T CD8-Positivos / Proteínas Mutantes / Agonismo Parcial de Drogas Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Nature Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Estados Unidos