GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (2)
  • Kim, Taeil  (2)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4438-4438
    Abstract: The success of immune checkpoint inhibitors CTLA4 and PD-1 monoclonal antibody, both FDA approved cancer therapies, specifically counteracts inhibitory pathways to activate antigen-specific T cells. However, their success is limited by the fact that not all patients respond to immunotherapy and a variety of adverse events due to non specific and systemic T cell activation limits their administration. A newly identified B7 family receptor, CD28H/TMIGD2, is constitutively expressed on T, pDCs and innate lymphoid cells including NK and ILCs. CD28H is also detected in TILs including the CD8+ tissue resident memory T cells (TRM), a T cell subset that correlates with better prognosis and response to immune checkpoint inhibitor therapy. Functionally, CD28H provides costimulatory function to T cells in the context of TCR signaling events during activation. We engineered a bispecific antibody (Bis mAb) targeting CD28H and PDL1 aimed to augment T cell costimulation and NK activation. The CD28H-PDL1 Bis mAb potentiates T cell proliferative and cytokine responses in antigen-specific human T cell assays and induces human NK -mediated redirected killing of PDL1+ tumor cells. In T cells, the mechanism of action of the Bis mAb requires intracellular signaling domain of the CD28H which downmodulates SHP phosphorylation upon CD3 activation. Intriguingly, we found that the Bis mAb increases induction of CD8 TRM cells in vitro. In human TILs, where CD28H is mostly expressed on CD8 TRM in tumors, the Bis mAb enables higher cytokine responses over PDL1 mAb alone. Given the recent relevance of TRM cells as an important pool of anti-tumor T cell immunity, the rationale for targeting this population via CD28H in the context of blocking PDL1 may prove to be a therapeutic tool for enhancing responses to checkpoint inhibitor therapy. Citation Format: Madhu Ramaswamy, Taeil Kim, Desmond Jones, Hormas Ghadially, Tamer Mahmoud, Andrew Garcia, Susan Wilson, Jeffrey Riggs, Darren Schofield, GIanluca Carlesso. A bispecific antibody targeting CD28H and PDL-1 is a novel and potent immunomodulator of T cell responses [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4438.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 10, No. 2 ( 2022-02-01), p. 200-214
    Abstract: Checkpoint blockade therapies targeting PD-1/PD-L1 and CTLA-4 are clinically successful but also evoke adverse events due to systemic T-cell activation. We engineered a bispecific, mAb targeting CD28 homolog (CD28H), a newly identified B7 family receptor that is constitutively expressed on T and natural killer (NK) cells, with a PD-L1 antibody to potentiate tumor-specific immune responses. The bispecific antibody led to T-cell costimulation, induced NK-cell cytotoxicity of PD-L1–expressing tumor cells, and activated tissue-resident memory CD8+ T cells. Mechanistically, the CD28H agonistic arm of the bispecific antibody reduced PD-L1/PD-1–induced SHP2 phosphorylation while simultaneously augmenting T-cell receptor signaling by activating the MAPK and AKT pathways. This bispecific approach could be used to target multiple immune cells, including CD8+ T cells, tissue-resident memory T cells, and NK cells, in a tumor-specific manner that may lead to induction of durable, therapeutic antitumor responses.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...