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

E-Poster Display

576P - The synergistic anticancer effect of PLAG on the PD-1 immune checkpoint inhibitor in the LLC-1 murine lung carcinoma syngeneic model

Date

17 Sep 2020

Session

E-Poster Display

Topics

Clinical Research

Tumour Site

Thoracic Malignancies

Presenters

Guen Tae Kim

Citation

Annals of Oncology (2020) 31 (suppl_4): S462-S504. 10.1016/annonc/annonc271

Authors

G.T. Kim1, S.Y. Yoon2, K. Park2, K. Sohn2, M. Kim3, J.W. Kim1

Author affiliations

  • 1 Cell Factory Research Center, KRIBB-Korea Research Institute of Bioscience and Biotechnology, 34141 - Daejeon/KR
  • 2 Development Of Global New Drug, ENZYCHEM lifesciences, 06774 - Seoul/KR
  • 3 Department Of Gastroenterology, Asan Medical Center, 05505 - 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 576P

Background

Although Immune checkpoint inhibitor (ICI) therapy usage has been increasing for various indications, some patients of various types of cancer were shown to not respond to ICI. To improve ICI response rate, a combination therapy targeting additional mechanisms to prevent tumour immune evasion by modulating the tumour microenvironment may be needed.

Methods

To investigate the enhanced anti-tumour effect of the anti-PD-1 antibody (aPD-1) with the addition of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG), the syngeneic model was used (n=6/group), LLC-1 lung carcinoma was implanted into the C57BL/6 mice subcutaneously. PLAG was daily administrated for 4 weeks with or without aPD-1 (RMP1-14). aPD-1 was delivered via IP injection once a week. The degree of infiltrated lymphocyte population and neutrophils in the tumour and blood on the sacrifice day were analyzed.

Results

In PLAG treated (50 or 100 mpk) mice group, the tumour burden was significantly reduced compared to a positive control (p < 0.05). In the group treated with aPD-1 alone, the tumour growth decreased by about 65% compared to the positive control. However, in mice co-treated with PLAG, the tumour was significantly reduced (18%) compared to the aPD-1 alone. The neutrophil-to-lymphocyte ratio levels in the group co-treated with PLAG were decreased remarkably compared to the aPD-1 alone. In particular, the degree of neutrophil infiltration in the tumour was effectively reduced upon PLAG treatment. Besides, the activity and infiltration of cytotoxic T-lymphocytes (CTLs) in the tumour were effectively increased in the group co-treated with PLAG compared to the aPD-1 alone. Such improvement was caused by a significant reduction of the population of Th17 which induced massive neutrophil infiltration in the tumour, compared to the positive control.

Conclusions

PLAG enhanced the anti-cancer effect of aPD-1 synergistically on the regression of tumour burden via decreasing the tumour-infiltrating neutrophils and Th17 population while increasing the CTLs. Therefore, combining aPD-1 with PLAG, which has excellent safety profiles, may contribute to enhancing the antitumor response of aPD-1 while lowering immune-related toxicities by reducing the dose of ICI.

Clinical trial identification

Editorial acknowledgement

Legal entity responsible for the study

Jae Wha Kim.

Funding

ENZYCHEM lifesciences.

Disclosure

All 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.