1
|
Yang B, Cheng C, Zhou J, Ni H, Liu H, Fu Y, Li R. AI-powered genomic mutation signature for predicting immune checkpoint inhibitor therapy outcomes in gastroesophageal cancer: a multi-cohort analysis. Discov Oncol 2024; 15:507. [PMID: 39342515 PMCID: PMC11439860 DOI: 10.1007/s12672-024-01400-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have significantly transformed the treatment of gastroesophageal cancer (GEC). However, the lack of reliable prognostic biomarkers hinders the ability to predict patient response to ICI therapy. METHODS In this study, we engineered and validated a genomic mutation signature (GMS) utilizing an innovative artificial intelligence (AI) algorithm to forecast ICI therapy outcomes in GEC patients. We further explored immune profiles across subtypes through comprehensive multiomics analysis. Our investigation of drug sensitivity data from the Genomics of Drug Sensitivity in Cancer (GDSC) database led to the identification of trametinib as a potential therapeutic agent. We subsequently evaluated trametinib's efficacy in AGS and MKN45 cell lines using Cell Counting Kit-8 (CCK8) assays and clonogenic experiments. RESULTS We developed a GMS by integrating 297 algorithms, enabling autonomous prognosis prediction for GEC patients. The GMS demonstrated consistent performance across three public cohorts, exhibiting high sensitivity and specificity for overall survival (OS) at 6, 12, and 18 months, as shown by Receiver Operator Characteristic Curve (ROC) analysis. Notably, the GMS surpassed traditional clinical and molecular features, including tumor mutational burden (TMB), programmed death-ligand 1 (PD-L1) expression, and microsatellite instability (MSI), in predictive accuracy. Low-risk samples exhibited elevated levels of cytolytic immune cells and heightened immunogenic potential compared to high-risk samples. Our investigation identified trametinib as a potential therapeutic agent. An inverse correlation was observed between GMS and trametinib IC50. Moreover, the high-risk-derived AGS cell line showed increased sensitivity to trametinib compared to the low-risk-derived MKN45 cell line. CONCLUSION The GMS utilized in this study successfully demonstrated the ability to reliably predict the survival advantage for patients with GECs undergoing ICI therapy.
Collapse
Affiliation(s)
- Bingyin Yang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Gastroenterology, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Cuie Cheng
- Department of Gastroenterology, Affiliated Changshu Hospital of Nantong University, Suzhou, Jiangsu, China
| | - Jingfang Zhou
- Department of Gastroenterology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Haoxiang Ni
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haoran Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yiwei Fu
- Department of Gastroenterology, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China.
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
2
|
Fridland S, Kim HS, Chae YK. Differential impact of intratumor heterogeneity (ITH) on survival outcomes in early-stage lung squamous and adenocarcinoma based on tumor mutational burden (TMB). Transl Lung Cancer Res 2024; 13:1481-1494. [PMID: 39118891 PMCID: PMC11304137 DOI: 10.21037/tlcr-24-226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/06/2024] [Indexed: 08/10/2024]
Abstract
Background Molecular biomarkers are reshaping patient stratification and treatment decisions, yet their precise use and best implementation remain uncertain. Intratumor heterogeneity (ITH), an area of increasing research interest with prognostic value across various conditions, lacks defined clinical relevance in certain non-small cell lung cancer (NSCLC) subtypes. Exploring the relationship between ITH and tumor mutational burden (TMB) is crucial, as their interplay might reveal distinct patient subgroups. This study evaluates how the ITH-TMB dynamic affects prognosis across the two main histological subtypes of NSCLC, squamous cell and adenocarcinoma, with a specific focus on early-stage cases to address their highly unmet clinical needs. Methods We stratify a cohort of 741 early-stage NSCLC patients from The Cancer Genome Atlas (TCGA) based on ITH and TMB and evaluate differences in clinical outcomes. Additionally, we compare driver mutations and the tumor microenvironment (TME) between high and low ITH groups. Results In lung squamous cell carcinoma (LUSC), high ITH predicts an extended progression-free survival (PFS) (median: 21 vs. 14 months, P=0.01), while in lung adenocarcinoma (LUAD), high ITH predicts a reduced PFS (median: 15 vs. 20 months, P=0.04). This relationship is driven by the low TMB subset of patients. Additionally, we found that CD8 T cells were enriched in better-performing subgroups, regardless of histologic subtype or ITH status. Conclusions There are significant differences in clinical outcomes, driver mutations, and the TME between high and low ITH groups among early-stage NSCLC patients. These differences may have treatment implications, necessitating further validation in other NSCLC datasets.
Collapse
Affiliation(s)
- Stanislav Fridland
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hye Sung Kim
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Young Kwang Chae
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
3
|
Ye B, Li Z, Wang Q. A novel artificial intelligence network to assess the prognosis of gastrointestinal cancer to immunotherapy based on genetic mutation features. Front Immunol 2024; 15:1428529. [PMID: 38994371 PMCID: PMC11236566 DOI: 10.3389/fimmu.2024.1428529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have revolutionized gastrointestinal cancer treatment, yet the absence of reliable biomarkers hampers precise patient response prediction. Methods We developed and validated a genomic mutation signature (GMS) employing a novel artificial intelligence network to forecast the prognosis of gastrointestinal cancer patients undergoing ICIs therapy. Subsequently, we explored the underlying immune landscapes across different subtypes using multiomics data. Finally, UMI-77 was pinpointed through the analysis of drug sensitization data from the Genomics of Drug Sensitivity in Cancer (GDSC) database. The sensitivity of UMI-77 to the AGS and MKN45 cell lines was evaluated using the cell counting kit-8 (CCK8) assay and the plate clone formation assay. Results Using the artificial intelligence network, we developed the GMS that independently predicts the prognosis of gastrointestinal cancer patients. The GMS demonstrated consistent performance across three public cohorts and exhibited high sensitivity and specificity for 6, 12, and 24-month overall survival (OS) in receiver operating characteristic (ROC) curve analysis. It outperformed conventional clinical and molecular features. Low-risk samples showed a higher presence of cytolytic immune cells and enhanced immunogenic potential compared to high-risk samples. Additionally, we identified the small molecule compound UMI-77. The half-maximal inhibitory concentration (IC50) of UMI-77 was inversely related to the GMS. Notably, the AGS cell line, classified as high-risk, displayed greater sensitivity to UMI-77, whereas the MKN45 cell line, classified as low-risk, showed less sensitivity. Conclusion The GMS developed here can reliably predict survival benefit for gastrointestinal cancer patients on ICIs therapy.
Collapse
Affiliation(s)
- Bicheng Ye
- School of Clinical Medicine, Yangzhou Polytechnic College, Yangzhou, China
| | - Zhongyan Li
- Department of Geriatric Medicine, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, China
| | - Qiqi Wang
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
- Department of Gastroenterology, The Second Afliated Hospital of Shanghai University, Wenzhou, China
| |
Collapse
|
4
|
Kim DH, Lim ST, Kim HR, Kang EJ, Ahn HK, Lee YG, Sun DS, Kwon JH, Lee SC, Lee HW, Kim MK, Keam B, Park KU, Shin SH, Yun HJ. Impact of PIK3CA and cell cycle pathway genetic alterations on durvalumab efficacy in patients with head and neck squamous cell carcinoma: Post hoc analysis of TRIUMPH study. Oral Oncol 2024; 151:106739. [PMID: 38458039 DOI: 10.1016/j.oraloncology.2024.106739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/10/2024]
Abstract
OBJECTIVES This study aimed to investigate whether genetic alterations in PI3KCA and the cell cycle pathways influence the efficacy of durvalumab, an immune checkpoint inhibitor, in patients with head and neck squamous cell carcinoma (HNSCC) who had previously failed platinum-based treatment. MATERIALS AND METHODS We obtained data from a phase II umbrella trial of patients with HNSCC who failed platinum-based treatment (TRIUMPH, NCT03292250). Patients receiving durvalumab treatment comprised those with PIK3CA alterations (Group A), those with cell cycle pathway alterations such as CDKN2A (Group B), and those with no druggable genetic alterations (Group C). We analyzed the overall response rate (ORR), progression-free survival (PFS), and overall survival (OS) in each group and evaluated the potential predictive factors for durvalumab. RESULTS We analyzed the data of 87 patients: 18, 12, and 57 in groups A, B, and C, respectively. The ORRs were 27.8 %, 8.3 %, and 15.8 % in Groups A, B, and C, respectively (P = 0.329), and the median PFS for each group was 2.3, 1.6, and 1.7 months, respectively, with no significant differences between the groups (P = 0.24). Notably, patients with lower neutrophil-lymphocyte ratio (NLR) (≤5.8) had longer PFS (median, 2.8 vs 1.6 months, P < 0.001), while those with lower platelet-lymphocyte ratio (PLR) (≤491.2) exhibited longer PFS (median, 1.8 vs 1.2 months, P < 0.001). CONCLUSION Durvalumab's efficacy was similar, irrespective of the presence of PIK3CA or cell cycle pathway genetic alterations in patients with platinum-resistant HNSCC. The NLR and PLR may be promising predictive biomarkers.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Taek Lim
- Hematology and Medical Oncology, Wonju Severance Christianity Hospital, Wonju, Republic of Korea
| | - Hye Ryun Kim
- Divison of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Seoul, Republic of Korea
| | - Eun Joo Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Kyung Ahn
- Department of Medical Oncology, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Yun-Gyoo Lee
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Der Sheng Sun
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hye Kwon
- Division of Hematology-Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Sang-Cheol Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Min Kyoung Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Yeungnam University Hospital, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Keon-Uk Park
- Department of Internal Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Seong-Hoon Shin
- Department of Internal Medicine, Kosin University Gospel Hospital, Busan, Republic of Korea.
| | - Hwan Jung Yun
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| |
Collapse
|
5
|
Yu B, Zhang N, Feng Y, Xu W, Zhang T, Wang L. A gene mutation-based risk model for prognostic prediction in liver metastases. BMC Genomics 2023; 24:489. [PMID: 37633919 PMCID: PMC10463705 DOI: 10.1186/s12864-023-09595-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023] Open
Abstract
BACKGROUND Liver metastasis is the major challenge in the treatment for malignant tumors. Genomic profiling is increasingly used in the diagnosis, treatment and prediction of prognosis in malignancies. In this study, we constructed a gene mutation-based risk model to predict the survival of liver metastases. METHOD We identified the gene mutations associated with survival and constructed the risk model in the training cohort including 800 patients with liver metastases from Memorial Sloan-Kettering Cancer Center (MSKCC) dataset. Other 794 patients with liver metastases were collected from 4 cohorts for validation. Furthermore, the analyses of tumor microenvironment (TME) and somatic mutations were performed on 51 patients with breast cancer liver metastases (BCLM) who had both somatic mutation data and RNA-sequencing data. RESULTS A gene mutation-based risk model involved 10 genes was constructed to divide patients with liver metastases into the high- and low-risk groups. Patients in the low-risk group had a longer survival time compared to those in the high-risk group, which was observed in both training and validation cohorts. The analyses of TME in BCLM showed that the low-risk group exhibited more immune infiltration than the high-risk group. Furthermore, the mutation signatures of the high-risk group were completely different from those of the low-risk group in patients with BCLM. CONCLUSIONS The gene mutation-based risk model constructed in our study exhibited the reliable ability of predicting the prognosis in liver metastases. The difference of TME and somatic mutations among BCLM patients with different risk score can guide the further research and treatment decisions for liver metastases.
Collapse
Affiliation(s)
- Bingran Yu
- Department of Hepatic Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Ning Zhang
- Department of Hepatic Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Yun Feng
- Department of Hepatic Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Weiqi Xu
- Department of Hepatic Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Ti Zhang
- Department of Hepatic Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China
| | - Lu Wang
- Department of Hepatic Surgery, Shanghai Cancer Center, Shanghai Medical College, Fudan University, No. 270 Dongan Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
6
|
Wang M, Zhu L, Yang X, Li J, Liu Y, Tang Y. Targeting immune cell types of tumor microenvironment to overcome resistance to PD-1/PD-L1 blockade in lung cancer. Front Pharmacol 2023; 14:1132158. [PMID: 36874015 PMCID: PMC9974851 DOI: 10.3389/fphar.2023.1132158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Lung cancer is the common malignant tumor with the highest mortality rate. Lung cancer patients have achieved benefits from immunotherapy, including immune checkpoint inhibitors (ICIs) therapy. Unfortunately, cancer patients acquire adaptive immune resistance, leading to poor prognosis. Tumor microenvironment (TME) has been demonstrated to play a critical role in participating in acquired adaptive immune resistance. TME is associated with molecular heterogeneity of immunotherapy efficacy in lung cancer. In this article, we discuss how immune cell types of TME are correlated with immunotherapy in lung cancer. Moreover, we describe the efficacy of immunotherapy in driven gene mutations in lung cancer, including KRAS, TP53, EGFR, ALK, ROS1, KEAP1, ZFHX3, PTCH1, PAK7, UBE3A, TNF-α, NOTCH, LRP1B, FBXW7, and STK11. We also emphasize that modulation of immune cell types of TME could be a promising strategy for improving adaptive immune resistance in lung cancer.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lijie Zhu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoxu Yang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiahui Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Ying Tang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|