Your browser doesn't support javascript.
loading
Harnessing the cytotoxic granule exocytosis to augment the efficacy of T-cell-engaging bispecific antibody therapy.
Casey, Mika; Lee, Carol; Hoyte, Sharon M; Johnston, Rebecca L; Kwok, Wing Yu; Law, Soi Cheng; Gandhi, Maher K; Harrison, Simon J; Nakamura, Kyohei.
Affiliation
  • Casey M; Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD.
  • Lee C; Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD.
  • Hoyte SM; Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD.
  • Johnston RL; Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD.
  • Kwok WY; Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD.
  • Law SC; Mater Research, University of Queensland, Brisbane, QLD.
  • Gandhi MK; Mater Research, University of Queensland, Brisbane, QLD.
  • Harrison SJ; Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital Melbourne VIC Australia; Sir Peter MacCallum, Department of Oncology, University of Melbourne, Parkville.
  • Nakamura K; Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD. kyohei.nakamura@qimrberghofer.edu.au.
Haematologica ; 109(7): 2131-2143, 2024 07 01.
Article in En | MEDLINE | ID: mdl-38268493
ABSTRACT
T-cell-engaging bispecific antibody (T-BsAb, also known as BiTE) therapy has emerged as a powerful therapeutic modality against multiple myeloma. Given that T-BsAb therapy redirects endogenous T cells to eliminate tumor cells, reinvigorating dysfunctional T cells may be a potential approach to improve the efficacy of T-BsAb. While various immunostimulatory cytokines can potentiate effector T-cell functions, the optimal cytokine treatment for T-BsAb therapy is yet to be established, partly due to a concern of cytokine release syndrome driven by aberrant interferon (IFN)-γ production. Here, we functionally screen immunostimulatory cytokines to determine an ideal combination partner for T-BsAb therapy. This approach reveals interleukin (IL)-21 as a potential immunostimulatory cytokine with the ability to augment T-BsAb-mediated release of granzyme B and perforin, without increasing IFN-γ release. Transcriptome profiling and functional characterization strongly support that IL-21 selectively targets the cytotoxic granule exocytosis pathway, but not pro-inflammatory responses. Notably, IL-21 modulates multiple steps of cytotoxic effector functions including upregulation of co-activating CD226 receptor, increasing cytotoxic granules, and promoting cytotoxic granule delivery at the immunological synapse. Indeed, T-BsAb-mediated myeloma killing is cytotoxic granule-dependent, and IL-21 priming significantly augments cytotoxic activities. Furthermore, in vivo IL-21 treatment induces cytotoxic effector reprogramming in bone marrow T cells, showing synergistic anti-myeloma effects in combination with T-BsAb therapy. Together, harnessing the cytotoxic granule exocytosis pathway by IL-21 may be a potential approach to achieve better responses by T-BsAb therapy.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Antibodies, Bispecific / Exocytosis / Multiple Myeloma Limits: Animals / Humans Language: En Journal: Haematologica Year: 2024 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Antibodies, Bispecific / Exocytosis / Multiple Myeloma Limits: Animals / Humans Language: En Journal: Haematologica Year: 2024 Document type: Article