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
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4106-4106
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4106-4106
    Abstract: Triple-negative breast cancer (TNBC) is the most aggressive and challenging breast cancer subtype, which does not respond to traditional endocrine and anti-HER2-targeted therapies. PD-L1 is highly enriched in TNBC and has been considered a therapeutic target. Despite the excellent anti-cancer activity, the atezolizumab-based chimeric antigen receptor (CAR) T cells showed a robust off-target effect. In addition, the treatment of solid tumors with CAR-T is limited by abnormal glycosylation in malignant tumors. Targeting glycosylated PD-L1 (gPD-L1) provided tissue specificity against TNBC, implying it can prevent antigen escape and off-target effect. In this study, we generated gPD-L1 CAR-T cells using lentiviral vectors expressing the scFv regions of the anti-gPD-L1 antibody. The gPD-L1 CAR-T cells exhibited antigen-specific activation, cytokine production, and cytolytic activity against TNBCs in vitro and in the xenograft tumors model. CyTOF and single-cell RNA sequencing (scRNA-seq) showed distinct IFNγ-positive cell types. Mechanistically, IFNγ crosstalked with EGFR signaling through Src activation and, in turn, triggered B3GNT3-mediated PD-L1 glycosylation. Inhibition of Src resulted in reduced gPD-L1 expression in TNBC. CRISPR/Cas9 knockout of B3GNT3 in TNBC cells impaired gPD-L1 CAR-T response. As a result of nonautonomous gPDL1 amplification in TNBCs, gPD-L1 CAR-T cells continued to annihilate TNBCs. Additionally, since gPD-L1 CAR-T cells provided higher specificity on TNBC, they had lower normal tissue toxicity. Overall, gPD-L1 CAR-T exhibits excellent anti-tumor activity against TNBCs, and it could be a promising immunotherapy tool to treat TNBCs in clinic. Furthermore, targeting glycosylation moiety on the tumor antigen is a novel approach to lessen CAR-T toxicity in patients. Citation Format: Chia-Wei Li, Shih-Han Wang, Yun-Ju Lai, Jyun Wang, Chun-Tse Kuo, Shih-Duo Hsu Hung, Shou-Hou Liu. Non-autonomous enhancement of gPDL1 CAR-T annihilates TNBC development. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4106.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...