Oops, you're using an old version of your browser so some of the features on this page may not be displaying properly.

MINIMAL Requirements: Google Chrome 24+Mozilla Firefox 20+Internet Explorer 11Opera 15–18Apple Safari 7SeaMonkey 2.15-2.23

ePoster Display

484P - CD300c blockade promotes anti-cancer immunity and synergizes with immune checkpoint inhibitor in colon cancer

Date

16 Sep 2021

Session

ePoster Display

Topics

Tumour Immunology;  Immunotherapy

Tumour Site

Colon and Rectal Cancer

Presenters

HyeRim Shin

Citation

Annals of Oncology (2021) 32 (suppl_5): S530-S582. 10.1016/annonc/annonc698

Authors

H. Shin1, W.S. LEE1, S.I. Lee2, J.H. Kim1, J. Yeom2, B. Kang1, J. Seo2, J. Cheon1, J. Jung2, J. Jeon2, C. Kim1, H.J. Chon1

Author affiliations

  • 1 Laboratory Of Translational Immuno-oncology, Bundang Cha Medical Center, 13496 - Seongnam/KR
  • 2 Immunotherapeutics, CentricsBio, 05836 - Seoul/KR

Resources

Login to get immediate access to this content.

If you do not have an ESMO account, please create one for free.

Abstract 484P

Background

CD300 glycoproteins are members of B7 family and are known to be involved in various T cell immune responses. Recently, CD300c was suggested as a negative regulator of T cell immunity. However, its role and therapeutic potential in cancer immunity is yet to be elucidated. Here, we developed a CD300c-targeted immunotherapy which triggers a potent anti-tumor immune responses and enhance PD-1 immune checkpoint inhibitor activity in colon cancer.

Methods

CL7, a monoclonal antibody against CD300c was generated and systemically administered to CT26 tumor-bearing mice either with or without PD-1 blockade. Tumor growth was monitored and tumor tissues were examined with flow cytometry, multiplex immunohistochemistry, and Nanostring.

Results

CL7 potently inhibited tumor growth in CT26 mice in a dose-dependent manner. Nanostring immune profiling analysis revealed extensive immunologic reprogramming within the tumor microenvironment, following CL7 treatment. Especially, gene expressions related to dendritic cell activation, Th1 response, and T cell activation were significantly upregulated after CL7 treatment. Consistently, flow cytometric and histologic analyses revealed that CL7 activated intratumoral CD8+ T cells, while decreasing CD4+Foxp3+ regulatory T cells, thereby promoting anti-tumor adaptive immunity within the tumor. Moreover, CL7 repolarized tumor-associated macrophages toward anti-tumoral M1 phenotype. Since CL7 treatment upregulated tumoral Pd-1 and Ctla-4 expressions as a negative feedback, we performed combination immunotherapy of CL7 treatment and PD-1 blockade to further enhance anti-tumor immune response. Combination treatment resulted in a stronger suppression of tumor growth and longer overall survival compared to monotherapy, supporting the further clinical development of this combination.

Conclusions

Overall, our study demonstrated that CD300c blockade is a novel strategy to induce a strong anti-cancer immunity and strengthen the efficacy of immune checkpoint inhibitor in cancer.

Clinical trial identification

Editorial acknowledgement

Legal entity responsible for the study

Jae-Won Jeon, Chan Kim, Hong Jae Chon.

Funding

CentricsBio.

Disclosure

S.I. Lee: Financial Interests, Institutional, Member: CentricsBio, Inc. J. Yeom: Financial Interests, Institutional, Member: CentricsBio, Inc. J. Seo: Financial Interests, Institutional, Member: CentricsBio, Inc. J. Jung: Financial Interests, Institutional, Member: CentricsBio, Inc. J. Jeon: Financial Interests, Institutional, Advisory Role: CentricsBio, Inc. All other authors have declared no conflicts of interest.

This site uses cookies. Some of these cookies are essential, while others help us improve your experience by providing insights into how the site is being used.

For more detailed information on the cookies we use, please check our Privacy Policy.

Customise settings
  • Necessary cookies enable core functionality. The website cannot function properly without these cookies, and you can only disable them by changing your browser preferences.