Your browser doesn't support javascript.
loading
Granzyme B-activated IL18 potentiates αß and γδ CAR T cell immunotherapy in a tumor-dependent manner.
Hull, Caroline M; Larcombe-Young, Daniel; Mazza, Roberta; George, Molly; Davies, David M; Schurich, Anna; Maher, John.
Affiliation
  • Hull CM; Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
  • Larcombe-Young D; King's College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, Guy's Cancer Centre, Great Maze Pond, London SE1 9RT, UK.
  • Mazza R; Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
  • George M; King's College London, Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
  • Davies DM; Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
  • Schurich A; King's College London, Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
  • Maher J; Leucid Bio Ltd, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; King's College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, Guy's Cancer Centre, Great Maze Pond, London SE1 9RT, UK; Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex B
Mol Ther ; 32(7): 2373-2392, 2024 Jul 03.
Article in En | MEDLINE | ID: mdl-38745414
ABSTRACT
Interleukin (IL)18 is a potent pro-inflammatory cytokine that is activated upon caspase 1 cleavage of the latent precursor, pro-IL18. Therapeuticcell armoring with IL18 promotes autocrine stimulation and positive modulation of the tumor microenvironment (TME). However, existing strategies are imperfect since they involve constitutive/poorly regulated activity or fail to modify the TME. Here, we have substituted the caspase 1 cleavage site within pro-IL18 with that preferred by granzyme B, yielding GzB-IL18. We demonstrate that GzB-IL18 is constitutively released but remains functionally latent unless chimeric antigen receptor (CAR) T cells are activated, owing to concomitant granzyme B release. Armoring with GzB-IL18 enhances cytolytic activity, proliferation, interferon (IFN)-γ release, and anti-tumor efficacy by a similar magnitude to constitutively active IL18. We also demonstrate that GzB-IL18 provides a highly effective armoring strategy for γδ CARcells, leading to enhanced metabolic fitness and significant potentiation of therapeutic activity. Finally, we show that constitutively active IL18 can unmask CARcell-mediated cytokine release syndrome in immunocompetent mice. By contrast, GzB-IL18 promotes anti-tumor activity and myeloid cell re-programming without inducing such toxicity. Using this stringent system, we have tightly coupled the biological activity of IL18 to the activation state of the host CARcell, favoring safer clinical implementation of this technology.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Immunotherapy, Adoptive / Interleukin-18 / Granzymes / Receptors, Chimeric Antigen Limits: Animals / Humans Language: En Journal: Mol Ther Journal subject: BIOLOGIA MOLECULAR / TERAPEUTICA Year: 2024 Document type: Article Country of publication:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Immunotherapy, Adoptive / Interleukin-18 / Granzymes / Receptors, Chimeric Antigen Limits: Animals / Humans Language: En Journal: Mol Ther Journal subject: BIOLOGIA MOLECULAR / TERAPEUTICA Year: 2024 Document type: Article Country of publication: