Identification of novel immune checkpoint targets and metabolic biomarkers for enhancing cancer immunotherapy efficacy
- Abstract
- Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, yielding durable responses in various tumors. However, a major challenge remains, as only a subset of patients shows strong responses, while many patients exhibit resistance. To overcome these limitations, the discovery of novel immune checkpoint (ICP) targets and predictive biomarkers has emerged as a promising approach to enhance ICI efficacy. In the first part of the study, we identified contactin-4 (CNTN4) as a promising ICP that impairs T cell functions. CNTN4 was highly expressed in tumor tissues, and its interaction with amyloid precursor protein (APP) on T cells diminished TCR-related signaling and impaired antitumor immune responses. To counteract this, we developed antibodies targeting CNTN4 (GENA-104A16) and APP (5A7), both of which effectively disrupted the CNTN4-APP interaction. These antibodies restored T cell functions, increased tumor-infiltrating lymphocytes, and promoted antitumor responses in a syngeneic mouse model. These results suggest that CNTN4-APP represents a potential ICP axis that can be targeted to boost immune responses in cancer therapy. In the second part of the study, we investigated the role of metabolic pathways—particularly amino acid and bile acid metabolism—in shaping ICI efficacy in non-small cell lung cancer (NSCLC) patients. Through metabolomic profiling and machine learning analysis, we identified histidine (His) as a positive predictor of ICI response, while homocysteine (HCys), phenylalanine (Phe), and sarcosine (Sar) were associated with poor outcomes. The His/HCys+Phe+Sar ratio emerged as a robust biomarker for predicting therapeutic outcomes. Additionally, bile acids such as glycochenodeoxycholic acid (GCDCA) and taurolithocholic acid (TLCA) were linked to improved survival probability and response to ICIs, with TLCA enhancing T cell proliferation and anti-tumor immunity. We also uncovered a relationship between the gut microbiota and TLCA levels, with the Eubacterium genus influencing TLCA-related metabolism. In summary, this research provides novel insights into overcoming the limitations of ICIs through the discovery of a novel ICP target and the identification of metabolic biomarkers that influence ICI response. Our findings suggest potential strategies to enhance the efficacy of immunotherapy and pave the way for more personalized treatment approaches in cancer immunotherapy.
- Author(s)
- 김수정
- Issued Date
- 2025
- Type
- Thesis
- URI
- https://scholar.gist.ac.kr/handle/local/19367
- Alternative Author(s)
- Sujeong Kim
- Department
- 대학원 의생명공학과
- Advisor
- Park, Han Soo
- Table Of Contents
- Abstract
Contents
List of Figures
Part 1. Therapeutic potential of targeting the CNTN4-APP axis in enhancing T cell mediated anti-cancer
response 1
1. Introduction 1
2. Materials and Methods 2
2.1. Cells 2
2.2. Animals 2
2.3. Cancer cell characterization (proliferation, migration, and invasion) 3
2.4. Tumor graft 3
2.5. Whole transcriptomic analysis of tumor tissues 4
2.6. In vitro plate-bound T cell proliferation or neutralization assay 4
2.7. Measurement of cytokine secretion 4
2.8. Affinity of GENA-104A16 against recombinant proteins or overexpressing HEK293FT cells 5
2.9. Biolayer interferometry 5
2.10. In vitro cytotoxicity assay 6
2.11. In vitro conjugation assay 6
2.12. Western blotting 6
2.13. Subcellular fractionation 7
2.14. Immune cell profiling 7
2.15. scRNA analysis for tumor-infiltrating immune cells in mouse tumor tissues 8
2.16. Statistical analyses 9
3. Results 10
3.1. Suppression of T cell activity by CNTN4 10
3.2. Neutralization of CNTN4-induced T cell suppression by GENA-104A16 14
3.3. In vivo effectiveness of GENA-104A16 17
3.4. Impact of GENA-104A16 on tumor immune landscape 19
3.5. Interaction between CNTN4 and APP 22
3.6. Role of APP in CNTN4-mediated T cell inhibition 24
3.7. In vivo effectiveness of anti-APP antibody (5A7) 26
3.8. Clinical implications of CNTN4 expression 28
3.9. Clinical implications of APP expression 33
4. Discussion 35
Part 2. Exploring the immunometabolic landscape: amino acid and bile acid metabolism as biomarkers
for NSCLC immunotherapy efficacy 38
1. Introduction 38
2. Materials and Methods 39
2.1. Patient cohort and characteristics 39
2.2. Targeted metabolic profiling using Biocrates kit 40
2.3. Analysis of bile acids in plasma samples 40
2.4. T cell assay 41
2.5. Mice 42
2.6. Immune cell profiling 43
2.7. Metagenomic 16S rRNA sequencing 43
2.8. CatBoost classification analysis 44
2.9. Random forest model analysis 44
2.10. RNA sequencing analysis 45
2.11. Single-cell RNA sequencing analysis 45
2.12. Statistical analysis 46
3. Results 47
3.1. Patients characteristics 47
3.2. The role of amino acid metabolism in predicting cancer immunotherapy outcomes 48
3.3. Clinical and immune significance of the His/HCys+Phe+Sar ratio in NSCLC patients 58
3.4. Identification of key bile acids modulating T cell function and immunotherapy response 61
3.5. Pivotal role of TLCA in boosting T cell activation and enhancing anti-tumor immune response 68
3.6. Association between TLCA levels and gut microbiota in NSCLC patients 72
4. Discussion 75
IV. Summary 78
V. References 78
VI. Acknowledgements 82
VII. Curriculum Vitae 83
- Degree
- Doctor
-
Appears in Collections:
- Department of Biomedical Science and Engineering > 4. Theses(Ph.D)
- 공개 및 라이선스
-
- 파일 목록
-
Items in Repository are protected by copyright, with all rights reserved, unless otherwise indicated.