1
|
Sun Y, Li Y, Zhang A, Hu T, Li M. Prognostic model identification of ribosome biogenesis-related genes in pancreatic cancer based on multiple machine learning analyses. Discov Oncol 2025; 16:905. [PMID: 40411705 PMCID: PMC12103412 DOI: 10.1007/s12672-025-02733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 05/16/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive cancer characterized by low survival rate. Enhanced ribosome biogenesis may be associated with tumor drug resistance and malignant phenotypes, representing a potential therapeutic target in pancreatic cancer. Therefore, exploring the molecular mechanisms of ribosome biogenesis in pancreatic cancer may uncover new biomarkers and potential therapeutic targets, facilitating the development of personalized treatment strategies. METHODS Ribosome biogenesis-related gene signatures were acquired from TCGA and Gene Cards databases. Prognostic gene sets were screened using machine learning algorithms to construct a risk model, which was externally validated via GEO database. Single-cell RNA sequencing analysis (GSE155698 dataset) was performed to assess gene expression patterns and module scores. RESULTS Sixty ribosome biogenesis-related prognostic genes were identified in pancreatic cancer. Cox regression and machine learning algorithms selected nine pivotal biomarkers (ECT2; CKB; HMGA2; TPX2; ERBB3; SLC2A1; KRT13; PRSS3; CRABP2) with high diagnostic and prognostic specificity for PAAD. The machine learning-derived risk score correlated strongly with tumor proliferation pathways and immunosuppression, suggesting dual roles in tumor promotion and immunosuppressive microenvironment remodeling. Single-cell analysis highlighted predominant expression of CKB, SLC2A1, ERBB3, CRABP2, and PRSS3 in pancreatic ductal epithelial cells. CONCLUSIONS Our results shed light on the potential connections between ribosome biogenesis-related molecular characteristics and clinical features, the tumor microenvironment, and clinical drug responses. The research underscores the critical role of ribosome biogenesis in the progression and treatment resistance of pancreatic cancer, offering valuable new perspectives for prognostic evaluation and therapeutic response prediction in pancreatic cancer.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Anlan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Uzel K, Bilir F, Tosun M, Topbas Selcuki NF, Eren Keskin S, Gokbayrak M, Demir G, Cine N, Ulug P, Iyibozkurt AC, Savlı H. The Role of Ultrasonography in Predicting Genetic Characteristics of Endometrial Cancers. J Clin Med 2025; 14:3216. [PMID: 40364249 PMCID: PMC12072879 DOI: 10.3390/jcm14093216] [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: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: To evaluate the association between endometrial tissue stiffness, as measured by shear wave elastography (SWE), and the presence of specific gene mutations in patients diagnosed with endometrial cancer. Methods: Peripheral blood samples were collected for DNA extraction and next-generation sequencing (NGS) to identify gene mutations. Preoperative SWE was performed to measure endometrial stiffness, with values expressed in kilopascals (kPa). Statistical analyses were conducted to assess the correlation between SWE measurements and genetic findings. Results: Genetic mutations were detected in 66% (n = 31) of cases, with TTN, PLEC, and PRSS1 being the most frequently mutated genes. The median SWE measurement was 36.5 kPa (range: 19.1-70.4 kPa). No statistically significant correlation was found between SWE values and the presence of gene mutations (p > 0.05). Cases with metastasis exhibited higher median SWE values (40.1 kPa) compared to non-metastatic cases (34.7 kPa), though this difference was not statistically significant. Conclusions: While no significant association was observed between endometrial stiffness and specific gene mutations, higher SWE values in metastatic cases suggest that increased tissue stiffness may be linked to tumor aggressiveness. Further large-scale studies are warranted to validate these findings and explore the potential of SWE as a non-invasive tool in assessing endometrial cancer characteristics.
Collapse
Affiliation(s)
- Kemine Uzel
- Department of Obstetrics and Gynecology, Demiroglu Science University, 41100 Istanbul, Turkey;
| | - Filiz Bilir
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Turkey;
| | - Mesude Tosun
- Department of Radiology, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Turkey;
| | - Nura Fitnat Topbas Selcuki
- Department of Obstetrics and Gynecology, Istanbul Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences Turkey, 34000 Istanbul, Turkey;
| | - Seda Eren Keskin
- Department of Genetics, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Turkey; (S.E.K.); (M.G.); (G.D.); (N.C.); (H.S.)
| | - Merve Gokbayrak
- Department of Genetics, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Turkey; (S.E.K.); (M.G.); (G.D.); (N.C.); (H.S.)
| | - Gulhan Demir
- Department of Genetics, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Turkey; (S.E.K.); (M.G.); (G.D.); (N.C.); (H.S.)
| | - Naci Cine
- Department of Genetics, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Turkey; (S.E.K.); (M.G.); (G.D.); (N.C.); (H.S.)
| | - Pasa Ulug
- Department of Obstetrics and Gynecology, Istanbul Haseki Training and Research Hospital, 34096 Istanbul, Turkey;
| | - Ahmet Cem Iyibozkurt
- Department of Obstetrics and Gynecology, Demiroglu Science University, 41100 Istanbul, Turkey;
| | - Hakan Savlı
- Department of Genetics, Faculty of Medicine, Kocaeli University, 41001 Kocaeli, Turkey; (S.E.K.); (M.G.); (G.D.); (N.C.); (H.S.)
| |
Collapse
|
3
|
Liu B, Tao W, Zhou X, Xu LD, Luo Y, Yang X, Min Q, Huang M, Zhu Y, Cui X, Wang Y, Gong T, Zhang E, Huang YS, Chen W, Yan S, Wu N. Multi‑omics analysis identifies different molecular subtypes with unique outcomes in early-stage poorly differentiated lung adenocarcinoma. Mol Cancer 2025; 24:129. [PMID: 40312720 PMCID: PMC12044723 DOI: 10.1186/s12943-025-02333-7] [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: 02/22/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025] Open
Abstract
INTRODUCTION Early-stage poorly differentiated lung adenocarcinoma (LUAD) is plagued by a high risk of postoperative recurrence, and its prognostic heterogeneity complicates treatment and surveillance planning. We conducted this integrative multi-omics study to identify those patients with a truly high risk of adverse outcomes. METHODS Whole-exome, RNA and whole methylome sequencing were carried out on 101 treatment-naïve early-stage poorly differentiated LUADs. Integrated analyses were conducted to disclose molecular characteristics and explore molecular subtyping. Functional validation of key molecules was carried out through in vitro and in vivo experiments. RESULTS Recurrent tumors exhibited significantly higher ploidy (p = 0.024), the fraction of the genome altered (FGA, p = 0.042), and aneuploidy (p < 0.05) compared to non-recurrent tumors, as well as a higher frequency of CNVs. Additionally, recurrent tumors showed hypomethylation at both the global level and in CpG island regions. Integrative transcriptomic and methylation analyses identified three molecular subtypes (C1, C2, and C3), with the C1 subtype presenting the worst prognosis (p = 0.024). Although frequently mutated genes showed similar mutation frequencies across the three subtypes, the C1 subtype exhibited the highest tumor mutation burden (TMB), mutant-allele tumor heterogeneity (MATH), aneuploidy, and HLA loss of heterozygosity (HLA-LOH), along with relatively lower immune cell infiltration. Furthermore, GINS1 and CPT1C were found to promote LUAD progression, and their high expression correlated with a poor prognosis. CONCLUSIONS This multi-omics study identified three integrative subtypes with distinct prognostic implications, paving the way for more precise management and postoperative monitoring of early-stage poorly differentiated LUAD.
Collapse
Affiliation(s)
- Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wei Tao
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xuantong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Li-Di Xu
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yanrui Luo
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Miao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xinrun Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yaqi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tongyang Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Enli Zhang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yu S Huang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Weizhi Chen
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Nan Wu
- State Key Laboratory of Molecular Oncology, Frontiers Science Center for Cancer Integrative Omics, Department of Thoracic Surgery II, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan, China.
| |
Collapse
|
4
|
Dai H, Ren J, Wang C, Huang J, Wang X. Prognostic molecular subtype reveals the heterogeneity of tumor immune microenvironment in gastric cancer. Sci Rep 2025; 15:14453. [PMID: 40281016 PMCID: PMC12032113 DOI: 10.1038/s41598-025-96686-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric cancer (GC) remains a leading cause of cancer-related deaths and exhibits considerable heterogeneity among patients. Thus, accurate classifications are essential for predicting prognosis and developing personalized therapeutic strategies. To address this, we retrospectively analyzed multi-omics data from 359 GC samples, incorporating transcriptomic RNA (mRNA), DNA methylation, mutation data, and clinical parameters. Using ten clustering algorithms, we integrated these datasets to classify GC into molecular subtypes. The robustness of our clustering approach was externally validated using an independent cohort generated from different sequencing technologies, and we characterized the heterogeneity of each subtype. Our analysis identified three distinct molecular subtypes of GC, designated CS1, CS2, and CS3. These subtypes exhibited significant differences in survival outcomes, activation of cancer-related pathways, immune microenvironment composition, genomic alterations, and responses to immunotherapy and chemotherapy. Notably, Cathepsin V (CTSV) was significantly downregulated in the immunologically active and highly responsive CS3 subtype, while it was upregulated in the immunologically exhausted CS2 subtype. These findings suggest that CTSV could serve as both a prognostic marker and a molecular classifier. Furthermore, this study provides the first evidence of CTSV's high expression in GC and its potential role in tumor progression. The novel clustering approach, based on ten clustering algorithms and comprehensive analysis of multi-omics data in gastric cancer, can guide prognosis, characterize different clinical and biological features, and elucidate the tumor immune microenvironment, providing insights into the intratumor heterogeneity of GC and potential novel therapeutic strategies. Additionally, CTSV emerges as a prognostic marker linked to tumor immunity and disease progression, which lays the foundation for improved stratification strategies and the development of targeted therapeutic approaches in GC.
Collapse
Affiliation(s)
- Hui Dai
- Medical School, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jing Ren
- Medical School, Nantong University, Nantong, 226001, Jiangsu, China
| | - Chun Wang
- Medical School, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jianfei Huang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xudong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
5
|
Cheng JN, Jin Z, Su C, Jiang T, Zheng X, Guo J, Li X, Chu H, Jia J, Zhou Q, Ding X, Zhang Y, Xu S, Dong F, Zhang Q, Yang X, Yang T, Cheng X, Zha H, Chen D, Wan YY, Liu X, Ye L, Tang H, Symonds ALJ, Li QJ, Jia Q, Zhu B. Bone metastases diminish extraosseous response to checkpoint blockade immunotherapy through osteopontin-producing osteoclasts. Cancer Cell 2025:S1535-6108(25)00137-0. [PMID: 40280123 DOI: 10.1016/j.ccell.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/23/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Bone metastatic lesions typically associate with suboptimal responses to immune checkpoint blockade (ICB) therapies. In this study, we observed that across multiple clinical cohorts and a variety of mouse models, the presence of osseous metastases induces ICB resistance in extraosseous tumors. Mechanistically, this long-distance communication is mediated by osseous tumor-conditioned osteoclasts producing osteopontin (OPN). Through circulation, OPN reprograms the extraosseous tumor microenvironment and impairs T cell recruitment and differentiation of CD8+TCF1+ precursor cells, an essential population for ICB efficacy. In mice, ICB responsiveness is restored by αRANKL blockade of osteoclastogenesis, neutralization of OPN in circulation, or tissue-specific depletion of OPN in osteoclasts. Both the mode of action and therapeutic benefit were validated in clinical cohorts with the αRANKL-ICB combinatory regimen. These findings establish bone as a specific immunoregulatory organ exploited by tumor metastasis and suggest osteoclastogenesis as a promising target to improve ICB prognosis in patients with bone metastasis.
Collapse
Affiliation(s)
- Jia-Nan Cheng
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Zheng Jin
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China; Research Institute, GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai 201318, China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No.507, Zhengmin Road, Shanghai 200433, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No.507, Zhengmin Road, Shanghai 200433, China
| | - Xiaobin Zheng
- Department of Thoracic Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Jinming Guo
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xingyi Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China; Department of Oncology, The General Hospital of Western Theater Command, Chengdu, Sichuan Province 610083, China
| | - Han Chu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Jia Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Qin Zhou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Xiaofang Ding
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Yiwen Zhang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Shouxia Xu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China; School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Fancong Dong
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China; Department of Oncology, The Affiliated Dongnan Hospital of Xiamen University, Zhangzhou 363000, China
| | - Qiao Zhang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China; The 988th Hospital of Joint Logistic Support Force of PLA, Zhengzhou, Henan 450042, China
| | - Xinxin Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Tao Yang
- Department of Oncology, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan 572013, China
| | - Xiaoming Cheng
- Department of Respiratory Diseases, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Haoran Zha
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China; Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xindong Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Haidong Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Alistair L J Symonds
- Blizard Institute, Barts and London School of Medicine and Dentistry, University of London, London E12AT, UK
| | - Qi-Jing Li
- Institute of Molecular and Cell Biology (IMCB) & Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138668, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 138668, Singapore.
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Key Laboratory of Immunotherapy, Chongqing 400037, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; Chongqing Advanced Pathology Research Institute, Jinfeng Laboratory, Chongqing 401329, China; Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
6
|
Qiu H, Deng X, Zha J, Wu L, Liu H, Lu Y, Zhang X. Machine learning-based characterization of stemness features and construction of a stemness subtype classifier for bladder cancer. BMC Cancer 2025; 25:717. [PMID: 40247223 PMCID: PMC12004775 DOI: 10.1186/s12885-025-14109-9] [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: 12/08/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Bladder cancer (BLCA) is a highly heterogeneous disease that presents challenges in predicting prognosis and treatment response. Cancer stem cells are key drivers of tumor development, progression, metastasis, and treatment resistance. The features and prognostic significance of stemness in bladder cancer need further investigation. METHODS We used bladder cancer datasets from the TCGA and GEO databases, based on stemness gene sets from the StemChecker database, to identify stemness subtypes using the consensus clustering algorithm. We calculated the mRNA expression-based stemness index (mRNAsi) using the OCLR algorithm. We compared the differences in overall survival, genomic characteristics, tumor microenvironment, and treatment response between the stemness subtypes. We constructed the stemness subtype classifier using machine learning algorithms such as LASSO regression, random forest, and multivariate logistic regression. The function of the classifier gene was validated through experiments. RESULTS We divided bladder cancer patients into two subtypes based on the enrichment scores of stemness gene sets. Patients within subtype 1 have a higher mRNAsi score, a better survival rate, an antitumor microenvironment, and higher sensitivity to immunotherapy, while patients within subtype 2 show higher aneuploidy scores, greater homologous recombination defects, an elevated tumor mutation burden, and increased chemotherapy sensitivity. We constructed a stemness subtype classifier based on six differentially expressed genes between the two subtypes. The classifier demonstrated good performance in predicting prognosis on three additional datasets from the GEO database and two non-muscle invasive bladder cancer datasets. Through tumor sphere formation experiments and western blotting, we found that TNFAIP6, out of the six classifier genes, maintains stemness. TNFAIP6 silencing also facilitated the chemotherapy response of cisplatin, docetaxel, and paclitaxel on bladder cancer cells. Furthermore, decreasing TNFAIP6 expression caused the immune checkpoint gene PD-L1 to downregulate. CONCLUSION This study provided valuable insights into the heterogeneity of BLCA stemness, and the stemness subtype classifier may facilitate molecular classification and personalized treatment selection for BLCA patients. Besides, TNFAIP6 may serve as a potential future stemness target guiding bladder cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Heping Qiu
- Department of Urology, the Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Xiaolin Deng
- Department of Urology, Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Jing Zha
- No 908th hospital of Chinese PLA Joint Logistic Support Force, Nanchang, 330000, China
| | - Lihua Wu
- Physical Examination Center, the Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Haonan Liu
- Department of Urology, the Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Yichen Lu
- Queen Mary School of Nanchang University, Nanchang University, Nanchang, 330000, China.
| | - Xinji Zhang
- Department of Urology, the Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
| |
Collapse
|
7
|
Yan X, Gao X, Dong J, Wang F, Jiang X, Hu X, Zhang J, Wang N, Xu L, Liu Z, Hu S, Zhao H. Integration of Single-Cell and Bulk RNA-seq Data to Identify the Cancer-Associated Fibroblast Subtypes and Risk Model in Glioma. Biochem Genet 2025; 63:1275-1297. [PMID: 38536568 DOI: 10.1007/s10528-024-10751-3] [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: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the stroma. Studies showed that CAFs were pivotally in glioma progression which have long been considered a promising therapeutic target. Therefore, the identification of prognostic CAF markers might facilitate the development of novel diagnostic and therapeutic approaches. A total of 1333 glioma samples were obtained from the TCGA and CGGA datasets. The EPIC, MCP-counter, and xCell algorithms were used to evaluate the relative proportion of CAFs in glioma. CAF markers were identified by the single-cell RNA-seq datasets (GSE141383) from the Tumor Immune Single-Cell Hub database. Unsupervised consensus clustering was used to divide the glioma patients into different distinct subgroups. The least absolute shrinkage and selection operator regression model was utilized to establish a CAF-related signature (CRS). Finally, the prognostic CAF markers were further validated in clinical specimens by RT‒qPCR. Combined single-cell RNA-seq analysis and differential expression analysis of samples with high and low proportions of CAFs revealed 23 prognostic CAF markers. By using unsupervised consensus clustering, glioma patients were divided into two distinct subtypes. Subsequently, based on 18 differentially expressed prognostic CAF markers between the two CAF subtypes, we developed and validated a new CRS model (including PCOLCE, TIMP1, and CLIC1). The nomogram and calibration curves indicated that the CRS was an accurate prognostic marker for glioma. In addition, patients in the high-CRS score group had higher immune infiltration and tumor mutation burden levels. Moreover, the CRS score had the potential to predict the response to immune checkpoint blockade (ICB) therapy and chemotherapy. Finally, the expression profiles of three CAF markers were verified by RT‒qPCR. In general, our study classified glioma patients into distinct subgroups based on CAF markers, which will facilitate the development of individualized therapy. We also provided insights into the role of the CRS in predicting the response to ICB and chemotherapy in glioma patients.
Collapse
Affiliation(s)
- Xiuwei Yan
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Gao
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Dong
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fang Wang
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyan Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xueyan Hu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
| | - Jiheng Zhang
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nan Wang
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Xu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
| | - Zhihui Liu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China.
| | - Shaoshan Hu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Hongtao Zhao
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Liu G, Yu G, Yin D, Ma J. Discovery of a new mitophagy-related gene signature for predicting the outlook and immunotherapy in triple-negative breast cancer. Sci Rep 2025; 15:6794. [PMID: 40000809 PMCID: PMC11861672 DOI: 10.1038/s41598-025-91613-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 02/21/2025] [Indexed: 02/27/2025] Open
Abstract
Mitophagy is an essential cellular process that is conserved and crucial for maintaining cellular balance by selectively eliminating malfunctioning mitochondria. However, there is still limited knowledge regarding the influence of mitophagy-related genes (MRGs) on the prognosis and response to treatment of triple-negative breast cancer (TNBC). In here, the TCGA and GEO databases were used to acquire the transcriptomic and clinical information of patients with TNBC, correspondingly. Using LASSO and multivariable Cox regression analyses, a risk signature related to mitophagy was established based on the prognostic MRGs. The prognostic signature associated with mitophagy consisted of five genes (BSG, JMJD6, DNAJA3, DISC1, and SQSTM1) and independently predicted the prognosis of patients with TNBC, regardless of clinical factors (p < 0.05). Patients classified within the high-risk group demonstrated significantly lower overall survival rates when contrasted with those in the low-risk group. The model exhibited excellent performance in predicting survival and risk stratification, as evidenced by the receiver operating characteristic and C-index. The findings stayed unchanged following external validation. Moreover, we observed a notable variation in the tumor immune microenvironment among the different risk categories. Patients with a low risk of TNBC demonstrated a more favorable response to immunotherapy compared to patients with a high risk. In conclusion, our study uncovered the possible impacts of MRGs on the tumor microenvironment, clinical and pathological characteristics, and outlook of TNBC. The CRG-related signature was strongly linked to the immune response against TNBC and has the potential to serve as a valuable tool in predicting the prognosis and immunotherapy response of patients.
Collapse
Affiliation(s)
- Gang Liu
- Department of Thyroid and Breast Surgery, The People's Hospital of Suzhou New District, Suzhou, China
| | - Guozheng Yu
- Department of Breast Surgery, Thyroid Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, China
| | - Dongzhi Yin
- Department of Gastrointestinal Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Jianying Ma
- Department of Breast Surgery, Thyroid Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, China.
| |
Collapse
|
9
|
Heo YJ, Ahn S, Kang SY, Kim H, Min BH, Kim KM. Distinct genomic, transcriptomic, and immune profiles for tumor and non-tumor mucosal regions in early gastric cancer. Pathol Res Pract 2025; 266:155768. [PMID: 39719794 DOI: 10.1016/j.prp.2024.155768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 12/26/2024]
Abstract
In early gastric cancer, local recurrence develops after endoscopic resection by field cancerization. Understanding the nature of cancer-prone environments is important to establish effective strategies to prevent recurrence. We hypothesized that the molecular/immune profiles in non-tumor (cancer-prone) tissue differ according to the relative distance from the gastric tumor. For this purpose, we performed whole-exome and transcriptome sequencing of 16 early gastric cancer samples with paired non-tumor mucosa 1 cm (N1) and 3 cm (N3) away from the tumor. The whole exome sequencing revealed mutations in both the tumor and non-tumor mucosa. TTN was the most frequently altered gene in tumors (31 %) and was the second most frequently altered gene in N1 (25 %) samples; however, the mutation rate was significantly lower in N3 (12 %) samples (P = 0.0046). Moreover, the expression levels of TTN mRNA were higher in tumors than in the N1 and N3 samples and were significantly associated with TTN mutations (P = 0.04). TP53 mutations were mainly observed in tumors (50 %) and in 6.3 % of N1, with no mutation detected in N3 samples. Transcriptome sequencing revealed that the expression of the epithelial-mesenchymal transition signature, mesenchymal signature, and proliferation signature was increased in tumors, whereas programmed death-ligand 1 expression was decreased in the non-tumor mucosa. In the tumor, although the numbers of M0/M1 macrophages, neutrophils, and eosinophils increased, plasma cell numbers were markedly decreased compared to non-tumor mucosa. In conclusion, non-tumor mucosa at 1 cm and 3 cm from the tumor harbored different genomic, transcriptomic, and immune cell profiles. The non-tumor mucosa closer to the tumor (1 cm) exhibited similar genomic and transcriptomic features. These findings can offer clinical guidance for acquiring a safe horizontal margin in endoscopic resection for early gastric cancer.
Collapse
Affiliation(s)
- You Jeong Heo
- The Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Samsung Medical Center, Sungkyunkwan University School of Medicine and Neocella Inc., Seoul, Republic of Korea
| | - Soomin Ahn
- Department of Pathology & Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - So Young Kang
- Department of Pathology & Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyunjin Kim
- Department of Pathology & Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Seegene Medical Foundation, Seoul, Republic of Korea
| | - Byung-Hoon Min
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Kyoung-Mee Kim
- Department of Pathology & Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Center of Companion Diagnostics, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Xu Q, Kong L, Han Z, Jin X, Ding M, Piao Z, Zhang S. RNA modification writer-based immunological profile and genomic landscape of tumor microenvironment in lung adenocarcinoma. Discov Oncol 2025; 16:45. [PMID: 39812762 PMCID: PMC11735815 DOI: 10.1007/s12672-025-01791-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Recent studies have highlighted the role of RNA modification, that is, the dysregulation of epitranscriptomics, in tumorigenesis and progression. The potential for undoing epigenetic changes may develop novel therapeutic and prognostic approaches. However, the roles of these RNA modifications in the tumor microenvironment (TME) are still unknown. METHODS We assessed the expression properties and genetic alterations of 26 RNA modification writers, including adenosine-to-inosine RNA editing, alternative polyadenylation, m1A, and m6A in 502 lung adenocarcinoma (LUAD) samples from the Cancer Genome Atlas (TCGA) datasets. Then, we used differentially expressed gene (DEGs) to develop a signature for predicting patient outcomes, which was dubbed the "writer score" for RNA-modified writers. In addition, we analyzed the association between TME features, molecular subtypes, treatment sensitivity, and immunotherapy efficacy. RESULTS We comprehensively evaluated the changes in multilayer RNA modification writers and identified the role of RNA modification writer expression imbalances in LUAD emergence and progression. Additionally, we constructed a risk-score model based on six LUAD prognosis-associated differentially expressed RNA modification writer genes. Kaplan-Meier (K-M) analyses revealed that the low risk-score signature had high overall patient survival. The predictive significance of the risk-score model was demonstrated using both univariate and multivariate Cox analyses. The risk-score model was positively correlated with the immune- and proliferation-related pathways. In response to anti-cancer treatment, high-risk score is related with high TMB, which has been discovered to correlate with immunotherapy effectiveness. CONCLUSION This study showed a strong correlation between the TME variety, level of complexity, and the four types of RNA modification writers. In addition, this scoring system could potentially predict effective immunotherapy and deepens our understanding of TME characteristics.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Oncology, Yanbian University Hospital, Yanji, 133000, China
| | - Lingyu Kong
- Department of Oncology, Yanbian University Hospital, Yanji, 133000, China
| | - Zhezhu Han
- Department of Oncology, Yanbian University Hospital, Yanji, 133000, China
| | - Xiuying Jin
- Department of Oncology, Yanbian University Hospital, Yanji, 133000, China
| | - Mingyan Ding
- Department of Oncology, Yanbian University Hospital, Yanji, 133000, China
| | - Zhengri Piao
- Department of Radiation Oncology, Yanbian University Hospital, Yanji, 133000, China
| | - Songnan Zhang
- Department of Oncology, Yanbian University Hospital, Yanji, 133000, China.
| |
Collapse
|
11
|
Kounatidou NE, Vitkos E, Palioura S. Ocular surface squamous neoplasia: Update on genetics, epigenetics and opportunities for targeted therapy. Ocul Surf 2025; 35:1-14. [PMID: 39608452 DOI: 10.1016/j.jtos.2024.11.006] [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: 08/02/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE The purpose of this review is to explore the molecular foundations of ocular surface squamous neoplasia (OSSN), focusing on the genetic and epigenetic aspects. While current management strategies include surgical excision and medical therapies, the understanding of OSSN's molecular basis remains limited, hindering the development of targeted treatments. METHODS A comprehensive MEDLINE search was conducted for literature published between January 1993 and October 2023. Only studies with original data on molecular, genetic, or epigenetic mechanisms, such as mutations, gene expression, and genetic predispositions were included. Articles were excluded if they focused solely on clinical management without addressing these factors, or if they were reviews, editorials, or opinion pieces. RESULTS The search yielded a total of 108 articles, out of which 39 articles met the criteria for further analysis. Investigations into OSSN have identified key DNA mutations in the TP53, HGF, EGFR, TERT, and CDKN2A genes, indicating common oncogenic pathways shared with other squamous cell carcinomas (SCCs). Significant epigenetic changes were identified, including DNA methylation, histone modifications, and altered miRNA expression patterns. Epigenetic dysregulation of critical tumor suppressors and oncoproteins, further highlight the complex genetic landscape of OSSN. CONCLUSION The molecular alterations identified in OSSN not only enhance our understanding of its biology but also have potential as novel biomarkers for early detection, prognostic evaluation, and as therapeutic targets. The identification of genetic and epigenetic markers in OSSN signifies progress towards personalized medicine approaches. Further studies and collaborative efforts are essential to validate these molecular markers and translate them into clinical practice, potentially revolutionizing OSSN management and improving patient outcomes.
Collapse
Affiliation(s)
| | - Evangelos Vitkos
- Department of Oral and Maxillofacial Surgery, Klinikum Dortmund, Dortmund, Germany
| | - Sotiria Palioura
- Department of Ophthalmology, University of Cyprus Medical School, Nicosia, Cyprus.
| |
Collapse
|
12
|
Liu H, Liu J, Guan X, Zhao Z, Cheng P, Chen H, Jiang Z, Wang X. Titin gene mutations enhance radiotherapy efficacy via modulation of tumour immune microenvironment in rectum adenocarcinoma. Clin Transl Med 2025; 15:e70123. [PMID: 39748197 PMCID: PMC11695211 DOI: 10.1002/ctm2.70123] [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: 06/23/2024] [Revised: 10/11/2024] [Accepted: 11/24/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE This study investigates the impact of Titin (TTN) gene mutations on radiotherapy sensitivity in rectum adenocarcinoma (READ) by examining changes in the tumour immune microenvironment. METHODS Data on gene expression and mutations in READ were obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Bioinformatics analysis explored the correlation between TTN mutations and immune cell infiltration. In vitro, lentiviral vectors were used to assess TTN mutations' effects on ANKRD1 expression in two READ cell lines. ANKRD1 was overexpressed, and clonogenic assays evaluated radiotherapy sensitivity. Flow cytometry, immunofluorescence, and comet assays examined mutations' impact on cell cycle, apoptosis, and DNA damage response (DDR). An in vivo mouse model and formalin-fixed paraffin-embedded samples from locally advanced rectal cancer (LARC) patients before and after radiotherapy were analyzed, followed by prognostic evaluation. RESULTS Bioinformatics revealed that TTN mutations increase radiation sensitivity in LARC by slowing cell proliferation, promoting apoptosis, and reducing DDR. TTN mutations also inhibit ANKRD1 expression via JUN disruption and enhance CD4/CD8 T-cell infiltration, improving anti-tumour immunity and outcomes. Observations from the clinical study showed a substantial decline in ANKRD1 expression levels alongside a notable surge in the counts of CD4+ and CD8+ T cells after undergoing radiotherapy. Patients with TTN mutations, low ANKRD1 expression, and high densities of CD4+ and CD8+ T cells had longer 3-year disease-free survival in READ. CONCLUSION Our findings reveal that TTN mutations can serve as biomarkers for enhanced radiotherapy sensitivity in READ. By altering the tumour's immune microenvironment, these mutations may provide a novel target for personalized radiotherapy strategies, potentially improving therapeutic outcomes in patients with READ. HIGHLIGHTS The association between TTN mutations and tumour mutation burden, as well as immune cell infiltration in READ, is examined. TTN mutations enhance the radiation sensitivity of READ cells and weaken DNA damage repair in response to radiation. TTN mutations increase the radiation sensitivity of READ cells by inhibiting ANKRD1. The infiltration of CD8+ and CD4+ T cells induced by TTN mutations is essential for anti-tumour immunity. TTN mutations serve as a biomarker for the pathological response to preoperative radiotherapy in READ.
Collapse
Affiliation(s)
- Hengchang Liu
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jialiang Liu
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xu Guan
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhixun Zhao
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pu Cheng
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haipeng Chen
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zheng Jiang
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xishan Wang
- Department of Colorectal SurgeryNational Cancer Center/National Clinical Research Center of Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
13
|
He J, Fu Z, Zou B, Lei X, Lei L, Yang Q, Li G. Identification the Cellular Senescence Associated lncRNA LINC01579 in Gastric Cancer. J Cell Mol Med 2025; 29:e70360. [PMID: 39855898 PMCID: PMC11760997 DOI: 10.1111/jcmm.70360] [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: 09/29/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Cellular senescence is a key promoter of tumorigenesis and malignant progression. This study aimed to develop a predictive model for assessing cellular senescence in gastric cancer (GC) outcomes. We identified senescence-related genes and lncRNAs from 375 stomach adenocarcinoma (STAD) patients and established a prognostic senescence score using multivariate Cox regression, validated in testing, TCGA-STAD and the combined TCGA-COAD and READ cohorts. The model's predictive efficacy was evaluated across clinical subgroups, tumour microenvironments and immune cell infiltration. A total of 116 senescence-related lncRNAs were filtered, and patients were clustered into two senescent subtypes. The lncRNA signature identified LINC01579 as an independent prognostic factor for GC. Low-risk groups showed prolonged overall survival, increased immune cell infiltration and reduced mutation load. Downregulation of LINC01579 using antisense oligonucleotides (ASOs) on normal human fibroblasts decreased cellular proliferation and migration in GC. Collectively, this study established and validated a promising prognostic model connecting senescence-related lncRNAs and clinical outcome in GC and provided potential senescence-related biomarkers for GC prognosis prediction.
Collapse
Affiliation(s)
- Jiayong He
- Department of General Surgery, Nanfang HospitalSouthern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive SurgeryGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorGuangzhouPeople's Republic of China
| | - Ziyi Fu
- Department of General Surgery, Nanfang HospitalSouthern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive SurgeryGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorGuangzhouPeople's Republic of China
- The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Boya Zou
- Department of General Surgery, Nanfang HospitalSouthern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive SurgeryGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorGuangzhouPeople's Republic of China
- The First School of Clinical MedicineSouthern Medical UniversityGuangzhouPeople's Republic of China
- Department of DermatologySeoul National University College of MedicineSeoulRepublic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research InstituteSeoul National University HospitalSeoulRepublic of Korea
- Institute of Human‐Environment Interface BiologySeoul National UniversitySeoulRepublic of Korea
| | - Xuetao Lei
- Department of General Surgery, Nanfang HospitalSouthern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive SurgeryGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorGuangzhouPeople's Republic of China
| | - Linhan Lei
- Department of General Surgery, Nanfang HospitalSouthern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive SurgeryGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorGuangzhouPeople's Republic of China
| | - Qingbin Yang
- Department of General Surgery, Nanfang HospitalSouthern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive SurgeryGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorGuangzhouPeople's Republic of China
| | - Guoxin Li
- Department of General Surgery, Nanfang HospitalSouthern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive SurgeryGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorGuangzhouPeople's Republic of China
- Cancer Center of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine,Tsinghua MedicineTsinghua UniversityBeijingPeople's Republic of China
| |
Collapse
|
14
|
Xiao H, He Q, Hu Y, Li C, Tian H, Chen F, Song W. A novel DNA damage-related gene index for predicting prognosis in gastric cancer. 3 Biotech 2025; 15:32. [PMID: 39763491 PMCID: PMC11700079 DOI: 10.1007/s13205-024-04166-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/18/2024] [Indexed: 01/18/2025] Open
Abstract
Gastric cancer is one of the major cancers with high cancer mortality and shows significant heterogeneity. The development of precise prognostic models is crucial for advancing treatment strategies. Recognizing the pivotal role of DNA damage in tumor progression, we conducted a consensus clustering analysis of DNA damage-related genes to categorize gastric cancer patients from the TCGA clinical cohort into distinct subtypes. Prognostic models were then constructed utilizing machine learning algorithms following Cox regression with differentially expressed genes. Validation was performed using the GSE gastric cancer cohort. Additionally, we investigated other characteristic responses of patients through gene mapping and drug sensitivity analysis. This study 12 differentially prognostic signature genes between the 2 DNA damage subtypes identified were used to calculate risk scores for the patients. This score predicts the prognosis of patients with gastric cancer and their overall survival time. Higher risk scores mean less drug sensitivity, lower survival, and possibly a poorer response to immunotherapy. Our findings provide the basis for future studies targeting DNA damage and its immune microenvironment to improve prognosis and response to immunotherapy.
Collapse
Affiliation(s)
- Haipeng Xiao
- Department of General Surgery, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Qianjin He
- Department of Hepatobiliary Surgery/Hernia Surgical Ward, Huanggang Central Hospital of Yangtze University, No.6 Qi ‘an Avenue, Huangzhou District, Huanggang, 438000 Hubei China
| | - Yang Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000 China
| | - Chang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Han Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Chen
- Department of Hepatobiliary Surgery/Hernia Surgical Ward, Huanggang Central Hospital of Yangtze University, No.6 Qi ‘an Avenue, Huangzhou District, Huanggang, 438000 Hubei China
| | - Wenchong Song
- Department of Gastroenterology, Huanggang Central Hospital of Yangtze University, No.6 Qi ‘an Avenue, Huangzhou District, Huanggang, 438000 Hubei China
| |
Collapse
|
15
|
Li S, Yuan T, Yuan J, Zhu B, Chen D. Opportunities and challenges of using circulating tumor DNA to predict lung cancer immunotherapy efficacy. J Cancer Res Clin Oncol 2024; 150:501. [PMID: 39545998 PMCID: PMC11568038 DOI: 10.1007/s00432-024-06030-8] [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: 02/27/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Immune checkpoint inhibitors (ICIs), particularly anti-programmed death 1 (PD-1)/ programmed death ligand 1 (PD-L1) antibodies, have led to significant progress in lung cancer treatment. However, only a minority of patients have responses to these therapies. Detecting peripheral blood of circulating tumor DNA (ctDNA) allows minimally invasive diagnosis, characterization, and monitoring of lung cancer. ctDNA has potential to be a prognostic biomarker and a predictor of the response to ICI therapy since it can indicate the genomic status and tumor burden. Recent studies on lung cancer have shown that pretreatment ctDNA analysis can detect residual proliferative disease in the adjuvant immunotherapy setting and evaluate tumor burden in patients with metastatic disease. Early ctDNA dynamics can not only predict the clinical outcome of ICI therapy but also help distinguish between pseudoprogression and real progression. Furthermore, in addition to quantitative assessment, ctDNA can also detect genetic predictors of response to ICI therapy. However, barriers still exist in the application of ctDNA analysis in clinical lung cancer treatment. The predictive value of ctDNA in lung cancer immunotherapy requires further identification and resolution of these challenges. This review aims to summarize the existing data of ctDNA analysis in patients receiving immunotherapy for lung cancer, understand the limitations of clinical treatment, and discuss future research directions.
Collapse
Affiliation(s)
- Shanshan Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ting Yuan
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jing Yuan
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
16
|
Quan S, Li N, Lian S, Wang Y, Liu Y, Liu J, Zhang Z, Gao D, Li Y. SLC4A4 as a novel biomarker involved in immune system response and lung adenocarcinoma progression. Int Immunopharmacol 2024; 140:112756. [PMID: 39083932 DOI: 10.1016/j.intimp.2024.112756] [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: 05/07/2024] [Revised: 07/14/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Altered expression and activity of solute carrier family 4 member 4 (SLC4A4) could affect the growth, survival and metastasis of tumor cells. Currently, the role of SLC4A4 in lung adenocarcinoma (LUAD) immunotherapy and prognosis was not entirely clear. METHODS We analyzed SLC4A4 expression in LUAD tissues and cell lines using quantitative reverse transcription-polymerase chain reaction, Western blotting, and immunohistochemistry. The effects of SLC4A4 overexpression on angiogenesis, cell migration, invasion, and epithelial-mesenchymal transition were examined. Public databases helped construct a risk model evaluating SLC4A4's expression on LUAD prognosis and immunotherapy response. Additionally, a xenograft model, flow cytometry, and enzyme-linked immunosorbent assay further explored SLC4A4's role in tumor immune microenvironment infiltration. RESULTS Upregulation of SLC4A4 promoted apoptosis in the LUAD cell line and significantly inhibited the migration and invasive ability of cancer cells (P<0.01). A total of 10 key genes (including SIGLEC6, RHOV, PIR, MOB3B, MIR3135B, LPAR6, KRT8, ITGA2, CPS1, and C6) were screened according to SLC4A4 expression, immune score and stromal score, and a prognostic model with good outcome was constructed (AUC values of which in the training cohort at 1,3, and 5 years reached 0.73, 0.73, and 0.72, respectively). Importantly, we demonstrated that high expression of SLC4A4 was able to increase the proliferation level and cytokine secretion of CD8+ T cells for the purpose of promoting the immune system response to LUAD. CONCLUSION Our study revealed that SLC4A4 can serve as a prognostic indicator for LUAD, providing new insights into the treatment and diagnosis of LUAD.
Collapse
Affiliation(s)
- Siyu Quan
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Na Li
- Nephrology Department, Jinan Zhangqiu District People's Hospital, Jinan 250200, China
| | - Shihai Lian
- Out-patient Department, Zaozhuang Municipal Hospital, Zaozhuang 277102, China
| | - Yuanyuan Wang
- The Department of Hospital Infection, Jinan Fifth People's Hospital, Jinan 250022, China
| | - Yang Liu
- Thoracic Surgery, PLA 80th Group Army Hospital, Weifang 261011, China
| | - Jianbo Liu
- Department of Thoracic Surgery, The Fourth People's Hospital of Heze, Heze 274100, China
| | - Zewei Zhang
- Department of Thoracic Surgery, Gaoqing County People's Hospital, Gaoqing 256399, China
| | - Dejun Gao
- Department of Thoracic Surgery, The Second People's Hospital of Liaocheng, Liaocheng 252600, China
| | - Yun Li
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China.
| |
Collapse
|
17
|
Wu Z, Yang X, Yuan Z, Guo Y, Wang X, Qu L. Identification of a novel histone acetylation-related long non-coding RNA model combined with qRT-PCR experiments for prognosis and therapy in gastric cancer. Heliyon 2024; 10:e36615. [PMID: 39263162 PMCID: PMC11387370 DOI: 10.1016/j.heliyon.2024.e36615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Gastric cancer (GC) is considered a global health crisis due to the scarcity of early diagnostic methods. Numerous studies have substantiated the involvement of histone acetylation imbalance in the progression of diverse tumor types. The potential roles of long non-coding RNA (lncRNA) in improving prognostic, predictive as well as therapeutic approaches in cancers have made it a major hotspot in recent years. Nevertheless, existent studies have never concerned the prognostic and clinical value of histone acetylation-related lncRNAs (HARlncs) in GC. Based on the aforementioned rationale, we developed a prognostic model incorporating four HARlncs-AC114730.1, AL445250.1, LINC01778, and AL163953.1-which demonstrated potential as an independent predictor of prognosis. Subsequently, GC patients were stratified into high-risk and low-risk groups. The low-risk group exhibited significantly higher overall survival (OS) compared to the high-risk group. Based on the analyses of the tumor microenvironment (TME) and immune responses, significant differences were observed between the two risk groups in terms of immune cell infiltration, immune checkpoint (ICP) expression, and other TME alterations. Furthermore, the sensitivity of GC patients to some chemotherapeutic drugs and the discrepant biological behaviors of three tumor clusters were studied in this model. In summary, we developed an effective HARlncs model with the objective of offering novel prognostic prediction methods and identifying potential therapeutic targets for GC patients.
Collapse
Affiliation(s)
- Zhixuan Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Xuejia Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ziwei Yuan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yangyang Guo
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| | - Xiaowu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, 325200, China
- Department of Thyroid and Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Liangchen Qu
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 318000, China
| |
Collapse
|
18
|
Xie Y, Chen H, Tian M, Wang Z, Wang L, Zhang J, Wang X, Lian C. Integrating multi-omics and machine learning survival frameworks to build a prognostic model based on immune function and cell death patterns in a lung adenocarcinoma cohort. Front Immunol 2024; 15:1460547. [PMID: 39346927 PMCID: PMC11427295 DOI: 10.3389/fimmu.2024.1460547] [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: 07/06/2024] [Accepted: 08/23/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction The programmed cell death (PCD) plays a key role in the development and progression of lung adenocarcinoma. In addition, immune-related genes also play a crucial role in cancer progression and patient prognosis. However, further studies are needed to investigate the prognostic significance of the interaction between immune-related genes and cell death in LUAD. Methods In this study, 10 clustering algorithms were applied to perform molecular typing based on cell death-related genes, immune-related genes, methylation data and somatic mutation data. And a powerful computational framework was used to investigate the relationship between immune genes and cell death patterns in LUAD patients. A total of 10 commonly used machine learning algorithms were collected and subsequently combined into 101 unique combinations, and we constructed an immune-associated programmed cell death model (PIGRS) using the machine learning model that exhibited the best performance. Finally, based on a series of in vitro experiments used to explore the role of PSME3 in LUAD. Results We used 10 clustering algorithms and multi-omics data to categorize TCGA-LUAD patients into three subtypes. patients with the CS3 subtype had the best prognosis, whereas patients with the CS1 and CS2 subtypes had a poorer prognosis. PIGRS, a combination of 15 high-impact genes, showed strong prognostic performance for LUAD patients. PIGRS has a very strong prognostic efficacy compared to our collection. In conclusion, we found that PSME3 has been little studied in lung adenocarcinoma and may be a novel prognostic factor in lung adenocarcinoma. Discussion Three LUAD subtypes with different molecular features and clinical significance were successfully identified by bioinformatic analysis, and PIGRS was constructed using a powerful machine learning framework. and investigated PSME3, which may affect apoptosis in lung adenocarcinoma cells through the PI3K/AKT/Bcl-2 signaling pathway.
Collapse
Affiliation(s)
- Yiluo Xie
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, MolecularDiagnosis Center, Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Mei Tian
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, MolecularDiagnosis Center, Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Ziqang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Luyao Wang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, MolecularDiagnosis Center, Joint Research Center for Regional Diseases of Institute of Health and Medicine (IHM), First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| |
Collapse
|
19
|
Mai Z, Kongjia L, Wang X, Xie X, Pang L, Yang H, Wen J, Fu J. Impaired TGF-β signaling via AHNAK family mutations elicits an esophageal cancer subtype with sensitivities to genotoxic therapy and immunotherapy. Cancer Immunol Immunother 2024; 73:225. [PMID: 39235488 PMCID: PMC11377381 DOI: 10.1007/s00262-024-03798-z] [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: 03/22/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Genome instability (GI) is a hallmark of esophageal squamous cell carcinoma (ESCC) while factors affecting GI remain unclear. METHODS Here, we aimed to characterize genomic events representing specific mechanisms of GI based on 201 ESCC samples and validated our findings at the patient, single-cell and cancer cell-line levels, including a newly generated multi-omics dataset of the trial NCT04006041. RESULTS A two-gene (AHNAK and AHNAK2) mutation signature was identified to define the "AHNAK1/2-mutant" cancer subtype. Single-cell-assisted multi-omics analysis showed that this subtype had a higher neoantigen load, active antigen presentation, and proficient CD8 + T cell infiltrations, which were validated at pan-cancer levels. Mechanistically, AHNAK1/2-mutant ESCC was characterized by impaired response of TGF-β and the inefficient alternative end-join repair (Alt-EJ) that might promote GI. Knockdown of AHNAK in ESCC cell lines resulted in more Alt-EJ events and increased sensitivities to cisplatin. Furthermore, this two-gene signature accurately predicted better responses to DNA-damaging therapy in various clinical settings (HR ≈ 0.25). The two-gene signature predicted higher pCR rates in ESCCs receiving neoadjuvant immunotherapy-involved treatment. Finally, a molecular classification scheme was built and outperformed established molecular typing models in the prognosis stratification of ESCC patients. CONCLUSION Our study extended our understanding of the AHNAK family in promoting GI and selecting treatment responders of ESCC.
Collapse
Affiliation(s)
- Zihang Mai
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, Guangdong Province, China
| | - Luo Kongjia
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, Guangdong Province, China
| | - Xinye Wang
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, Guangdong Province, China
| | - Xiuying Xie
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, Guangdong Province, China
| | - Lanlan Pang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China
| | - Hong Yang
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, Guangdong Province, China.
| | - Jing Wen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, Guangdong Province, China.
| | - Jianhua Fu
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, Guangdong Province, China.
| |
Collapse
|
20
|
Hu W, Li D, Yang Y, Zheng Y, Zeng J, Sai K. TIM-3/CD68 double-high expression in Glioma: Prognostic characteristics and potential therapeutic approaches. Int Immunopharmacol 2024; 139:112665. [DOI: 39002523 10.1016/j.intimp.2024.112665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
|
21
|
Kodali N, Alomary S, Bhattaru A, Eldaboush A, Schwartz RA, Lipner SR. Gender and melanoma subtype-based prognostic implications of MUC16 and TTN co-occurrent mutations in melanoma: A retrospective multi-study analysis. Cancer Med 2024; 13:e70199. [PMID: 39240165 PMCID: PMC11378355 DOI: 10.1002/cam4.70199] [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: 02/09/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Most primary cutaneous melanomas have pathogenesis driven by ultraviolet exposure and genetic mutations, whereas acral lentiginous melanoma (ALM) and metastatic melanoma are much less, if at all, linked with the former. Thus, we evaluated both ultraviolet related and non-ultraviolet related melanomas. Mutations in the MUC16 and TTN genes commonly occur concurrently in these melanoma patients, but their combined prognostic significance stratified by gender and cancer subtype remains unclear. METHODS The cBioPortal database was queried for melanoma studies and returned 16 independent studies. Data from 2447 melanoma patients were utilized including those with ALM, cutaneous melanoma (CM), and melanoma of unknown primary (MUP). Patients were grouped based on the presence or absence of MUC16 and TTN mutations. Univariate Cox regression and Student's t-tests were used to analyze hazard ratios and total mutation count comparisons, respectively. RESULTS TTN mutations, either alone or concurrently with MUC16 mutations, significantly correlated with worse prognosis overall, in both genders, and in CM patients. ALM patients with both mutations had better prognoses than CM patients, while ALM patients with neither mutation had worse prognosis than CM patients. For MUP patients, only MUC16 mutations correlated with worse prognosis. ALM patients with neither MUC16 nor TTN mutations had significantly more total mutations than MUP patients, followed by CM patients. CONCLUSION TTN mutations are a potential marker of poor prognosis in melanoma, which is amplified in the presence of concurrent MUC16 mutations. ALM patients with neither gene mutations had worse prognosis, suggesting a protective effect of having both MUC16 and TTN mutations. Only MUC16 mutations conferred a worse prognosis for MUP patients. Comprehensive genetic profiling in melanoma patients may facilitate personalized treatment strategies to optimize patient outcomes.
Collapse
Affiliation(s)
- Nilesh Kodali
- Rutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | | | | | - Ahmed Eldaboush
- Department of DermatologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Shari R. Lipner
- Department of DermatologyWeill Cornell MedicineNew YorkNew YorkUSA
| |
Collapse
|
22
|
Su DG, Dhiman A, Bansal VV, Zha Y, Shergill A, Polite B, Alpert L, Turaga KK, Eng OS. Mutational Features and Tumor Microenvironment Alterations in High-Grade Appendiceal Cancers Treated With Iterative Hyperthermic Intraperitoneal Chemotherapy. JCO Precis Oncol 2024; 8:e2400149. [PMID: 39259912 PMCID: PMC11432692 DOI: 10.1200/po.24.00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/23/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024] Open
Abstract
PURPOSE High-grade appendiceal adenocarcinomas (HGAA) with peritoneal metastases (PMs) are associated with poor survival. Hyperthermic intraperitoneal chemotherapy (HIPEC) is a novel treatment approach for unresectable HGAA-PM. However, its influence on immunogenomic profiles has not yet been fully explored. MATERIALS AND METHODS We obtained 79 samples of metastatic peritoneal tumor deposits from patients diagnosed with HGAA and performed whole-exome sequencing, RNA sequencing, and immunoprofiling before and after HIPEC. Tumor biopsies were subjected to immunogenomic profiling to detect mutational signatures and immune populations associated with oncologic outcomes. RESULTS Fifteen patients with HGAA-PMs were included in the study. The median progression-free survival (PFS) was 6.7 months (2.7-25.3) and the median overall survival was 11.4 months (4.7-42). Mucin-associated genes (MUC16, MUC3A, and MUC5AC) and titin (TTN) had the highest mutation frequencies. Mutational signatures such as single-base substitution 29 and doublet-base substitution 11 were present in >50% of single-base and double-base mutations. Higher PD-L1 coexpression on CD8+ T cells demonstrated a higher PFS both intratumorally (P = .019) and at the margin (P = .025). CONCLUSION HIPEC-associated mutational signatures were identified in HGAA-PMs. Elevated PD-L1+ cytotoxic T-cell populations after HIPEC had better PFS, offering valuable insights for prognostication in the context of HIPEC treatment.
Collapse
Affiliation(s)
- David G Su
- Division of Surgical Oncology, Yale School of Medicine, New Haven, CT
| | - Ankit Dhiman
- Department of Surgery, Medical College of Georgia, Augusta, GA
| | - Varun V Bansal
- Division of Surgical Oncology, Yale School of Medicine, New Haven, CT
| | - Yuanyuan Zha
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Ardaman Shergill
- Department of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL
| | - Blasé Polite
- Department of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL
| | - Lindsay Alpert
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Kiran K Turaga
- Division of Surgical Oncology, Yale School of Medicine, New Haven, CT
| | - Oliver S Eng
- Department of Surgery, University of California, Irvine, Orange, CA
| |
Collapse
|
23
|
Thakor P, Siddiqui MQ, Patel TR. Analysis of the interlink between glucose-6-phosphate dehydrogenase (G6PD) and lung cancer through multi-omics databases. Heliyon 2024; 10:e35158. [PMID: 39165939 PMCID: PMC11334843 DOI: 10.1016/j.heliyon.2024.e35158] [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: 05/13/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Glucose-6-Phosphate Dehydrogenase (G6PD) is a crucial enzyme that executes the pentose phosphate pathway. Due to its critical nodal position in the metabolic network, it is associated with different forms of cancer tumorigeneses and progression. Nonetheless, its functional role and molecular mechanism in lung cancer remain unknown. The present study provides intricate information associated with G6PD and Lung Cancer. Varieties of public datasets were retrieved by us, including UALCAN, TCGA, cBioPortal, and the UCSC Xena browser. The data obtained were used to assess the expression of G6PD, its clinical features, epigenetic regulation, relationship with tumour infiltration, tumour mutation burden, microsatellite instability, tumour microenvironment, immune checkpoint genes, genomic alteration, and patient's overall survival rate. The present study revealed that the G6PD expression was correlated with the clinical features of lung cancer including disease stage, race, sex, age, smoking habits, and lymph node metastasis. Moreover, the expression profile of G6PD also imparts epigenetic changes by modulating the DNA promoter methylation activity. Methylation of promoters changes the expression of various transcription factors, genes leading to an influence on the immune system. These events linked with G6PD-related mutational gene alterations (FAM3A, LAG3, p53, KRAS). The entire circumstance influences the patient's overall survival rate and poor prognosis. Functional investigation using STRING, GO, and KEGG found that G6PD primarily engages in hallmark functions (metabolism, immunological responses, proliferation, apoptosis, p53, HIF-1, FOXO, PI3K-AKT signaling). This work provides a wide knowledge of G6PD's function in lung cancer, as well as a theoretical foundation for possible prognostic therapeutic markers.
Collapse
Affiliation(s)
- Parth Thakor
- Bapubhai Desaibhai Patel Institute of Paramedical Sciences, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, India
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, AB, T1K 2E1, Canada
| | - M. Quadir Siddiqui
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, AB, T1K 2E1, Canada
| | - Trushar R. Patel
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, AB, T1K 2E1, Canada
- Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| |
Collapse
|
24
|
Wei C, Ma Y, Wang M, Wang S, Yu W, Dong S, Deng W, Bie L, Zhang C, Shen W, Xia Q, Luo S, Li N. Tumor-associated macrophage clusters linked to immunotherapy in a pan-cancer census. NPJ Precis Oncol 2024; 8:176. [PMID: 39117688 PMCID: PMC11310399 DOI: 10.1038/s41698-024-00660-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Transcriptional heterogeneity of tumor-associated macrophages (TAMs) has been investigated in individual cancers, but the extent to which these states transcend tumor types and represent a general feature of cancer remains unclear. We performed pan-cancer single-cell RNA sequencing analysis across nine cancer types and identified distinct monocyte/TAM composition patterns. Using spatial analysis from clinical study tissues, we assessed TAM functions in shaping the tumor microenvironment (TME) and influencing immunotherapy. Two specific TAM clusters (pro-inflammatory and pro-tumor) and four TME subtypes showed distinct immunological features, genomic profiles, immunotherapy responses, and cancer prognosis. Pro-inflammatory TAMs resided in immune-enriched niches with exhausted CD8+ T cells, while pro-tumor TAMs were restricted to niches associated with a T-cell-excluded phenotype and hypoxia. We developed a machine learning model to predict immune checkpoint blockade response by integrating TAMs and clinical data. Our study comprehensively characterizes the common features of TAMs and highlights their interaction with the TME.
Collapse
Affiliation(s)
- Chen Wei
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yijie Ma
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Mengyu Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Siyi Wang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wenyue Yu
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Shuailei Dong
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Wenying Deng
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Liangyu Bie
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Chi Zhang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Wei Shen
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Qingxin Xia
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| | - Suxia Luo
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| | - Ning Li
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
25
|
Hu W, Li D, Yang Y, Zheng Y, Zeng J, Sai K. TIM-3/CD68 double-high expression in Glioma: Prognostic characteristics and potential therapeutic approaches. Int Immunopharmacol 2024; 139:112665. [PMID: 39002523 DOI: 10.1016/j.intimp.2024.112665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Immunotherapy has revolutionized the treatment of various types of tumors, but there has been no breakthrough in the treatment of gliomas. The aim of this study is to discover valuable immunotherapy target in glioma, analyze its expression in glioma and the related microenvironment, explore potential immunotherapy strategies, and propose new possibilities for the treatment of gliomas. METHODS Immunohistochemistry (IHC) and multiplex fluorescence immunohistochemistry (mIHC) were used to analyze the expression of common immune markers and checkpoints in 187 glioma patients from Sun Yat-sen University Caner Center (SYSUCC). Bioinformatics analysis was used to examine the expression of TIM-3 in different macrophages using the Chinese Glioma Genome Atlas (CGGA) single-cell sequencing database. The Kaplan-Meier curve was used to predict the prognostic value of samples with high TIM-3 and CD68 expression. The R package was used to analyze the somatic mutation status and the sensitivity of small molecule inhibitors in TIM-3/CD68 double-high expression samples. RESULTS TIM-3 is a relatively highly expressed immune checkpoint in glioma. Unlike other tumors, TIM-3 is mainly expressed on macrophages in the glioma microenvironment. TIM-3/CD68 double-high expression suggests poor survival in glioma and may be a new upgrade marker in both IDH-mutant glioma and IDH-wildtype low-grade glioma (LGG) glioma (P < 0.01). Exploring the combination of TIM-3 inhibitors and p38 MAPK inhibitor may be a potential treatment direction for TIM-3/CD68 double high expression gliomas in the future. CONCLUSIONS The combination of TIM-3 and CD68 holds significant importance as a potential target for both prognosis and therapeutic intervention in glioma.
Collapse
Affiliation(s)
- Wanming Hu
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, PR China
| | - Depei Li
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, PR China
| | - Yuanzhong Yang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, PR China
| | - Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, PR China
| | - Jing Zeng
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, PR China.
| | - Ke Sai
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Oncology in South China, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, PR China.
| |
Collapse
|
26
|
Pan Y, Zeng W, Nie X, Chen H, Xie C, Guo S, Xu D, Chen Y. Immunotherapy-relevance of a candidate prognostic score for Acute Myeloid Leukemia. Heliyon 2024; 10:e32154. [PMID: 38961904 PMCID: PMC11219318 DOI: 10.1016/j.heliyon.2024.e32154] [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: 07/26/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
Background Acute Myeloid Leukemia (AML) exhibits a wide array of phenotypic manifestations, progression patterns, and heterogeneous responses to immunotherapies, suggesting involvement of complex immunobiological mechanisms. This investigation aimed to develop an integrated prognostic model for AML by incorporating cancer driver genes, along with clinical and phenotypic characteristics of the disease, and to assess its implications for immunotherapy responsiveness. Methods Critical oncogenic driver genes linked to survival were identified by screening primary effector and corresponding gene pairs using data from The Cancer Genome Atlas (TCGA), through univariate Cox proportional hazard regression analysis. This was independently verified using dataset GSE37642. Primary effector genes were further refined using LASSO regression. Transcriptomic profiling was quantified using multivariate Cox regression, and the derived prognostic score was subsequently validated. Finally, a multivariate Cox regression model was developed, incorporating the transcriptomic score along with clinical parameters such as age, gender, and French-American-British (FAB) classification subtype. The 'Accurate Prediction Model of AML Overall Survival Score' (APMAO) was developed and subsequently validated. Investigations were conducted into functional pathway enrichment, alterations in the gene mutational landscape, and the extent of immune cell infiltration associated with varying APMAO scores. To further investigate the potential of APMAO scores as a predictive biomarker for responsiveness to cancer immunotherapy, we conducted a series of analyses. These included examining the expression profiles of genes related to immune checkpoints, the interferon-gamma signaling pathway, and m6A regulation. Additionally, we explored the relationship between these gene expression patterns and the Tumor Immune Dysfunction and Exclusion (TIDE) dysfunction scores. Results Through the screening of 95 cancer genes associated with survival and 313 interacting gene pairs, seven genes (ACSL6, MAP3K1, CHIC2, HIP1, PTPN6, TFEB, and DAXX) were identified, leading to the derivation of a transcriptional score. Age and the transcriptional score were significant predictors in Cox regression analysis and were integral to the development of the final APMAO model, which exhibited an AUC greater than 0.75 and was successfully validated. Notable differences were observed in the distribution of the transcriptional score, age, cytogenetic risk categories, and French-American-British (FAB) classification between high and low APMAO groups. Samples with high APMAO scores demonstrated significantly higher mutation rates and pathway enrichments in NFKB, TNF, JAK-STAT, and NOTCH signaling. Additionally, variations in immune cell infiltration and immune checkpoint expression, activation of the interferon-γ pathway, and expression of m6A regulators were noted, including a negative correlation between CD160, m6A expression, and APMAO scores. Conclusion The combined APMAO score integrating transcriptional and clinical parameters demonstrated robust prognostic performance in predicting AML survival outcomes. It was linked to unique phenotypic characteristics, distinctive immune and mutational profiles, and patterns of expression for markers related to immunotherapy sensitivity. These observations suggest the potential for facilitating precision immunotherapy and advocate for its exploration in upcoming clinical trials.
Collapse
Affiliation(s)
- Yiyun Pan
- Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
- Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Wen Zeng
- Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Xiaoming Nie
- Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Hailong Chen
- Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Chuanhua Xie
- Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Shouju Guo
- Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Dechang Xu
- Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Yijian Chen
- Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
27
|
Zeng L, Zhang L, Yin C, Chen X, Chen X, Sun L, Sun J. Characterization of zinc finger protein 536, a neuroendocrine regulator, using pan-cancer analysis. Eur J Med Res 2024; 29:273. [PMID: 38720348 PMCID: PMC11077744 DOI: 10.1186/s40001-024-01792-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/12/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Previous studies suggested that zinc finger protein 536 (ZNF536) was abundant in the central brain and regulated neuronal differentiation. However, the role of ZNF536 in cancer has remained unclear. METHODS ZNF536 mutation, copy number alteration, DNA methylation, and RNA expression were explored using public portals. Data from The Cancer Genome Atlas (TCGA) were utilized to analyze pathways and tumor microenvironment (TME), with a focus on prognosis in both TCGA and immunotherapy pan-cancer cohorts. Methylated ZNF536 from small cell lung cancer (SCLC) cell lines were utilized to train with probes for conducting enrichment analysis. Single-cell RNA profile demonstrated the sublocalization and co-expression of ZNF536, and validated its targets by qPCR. RESULTS Genetic alterations in ZNF536 were found to be high-frequency and a single sample could harbor different variations. ZNF536 at chromosome 19q12 exerted a bypass effect on CCNE1, supported by CRISPR data. For lung cancer, ZNF536 mutation was associated with longer survival in primary lung adenocarcinoma (LUAD), but its prognosis was poor in metastatic LUAD and SCLC. Importantly, ZNF536 mutation and amplification had opposite prognoses in Stand Up To Cancer-Mark Foundation (SU2C-MARK) LUAD cohort. ZNF536 mutation altered the patterns of genomic alterations in tumors, and had distinct impacts on the signaling pathways and TME compared to ZNF536 amplification. Additionally, ZNF536 expression was predominantly in endocrine tumors and brain tissues. High-dimensional analysis supported this finding and further revealed regulators of ZNF536. Considering that the methylation of ZNF536 was involved in the synaptic pathway associated with neuroendocrine neoplasms, demonstrating both diagnostic and prognostic value. Moreover, we experimentally verified ZNF536 upregulated neuroendocrine markers. CONCLUSIONS Our results showed that ZNF536 alterations in cancer, including variations in copy number, mutation, and methylation. We proved the involvement of ZNF536 in neuroendocrine regulation, and identified highly altered ZNF536 as a potential biomarker for immunotherapy.
Collapse
Affiliation(s)
- Longjin Zeng
- Department of Basic Medicine, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Longyao Zhang
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Chenrui Yin
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Xu Chen
- Department of Medical Affairs, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Xiewan Chen
- Department of Basic Medicine, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Lingyou Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Jianguo Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
28
|
Li X, Tang Z, Li Z, Li Z, Zhao P, Song Y, Yang K, Xia Z, Wang Y, Guo D. Somatic mutations that affect early genetic progression and immune microenvironment in gastric carcinoma. Pathol Res Pract 2024; 257:155310. [PMID: 38663178 DOI: 10.1016/j.prp.2024.155310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/24/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024]
Abstract
Gastric carcinoma (GC) is a high heterogeneity and malignant tumor with a poor prognosis. The current implementation of immunotherapy in GC is limited due to the insufficient exploration of immune-related mutations and speculated early mutation events. Therefore, we performed whole-exome sequencing on 40 patients with GC to explore their genetic characteristics, shedding light on the order of genetic events, somatic mutations impacting the immune microenvironment, and potential biomarkers for immunotherapy. Regarding genetic events, TP53 disruptions were identified as frequent and early events in GC progression, often occurring alongside other gene mutations. The mutations occurring in GANS, SMAD4, and POLE were early independent events. Patients harboring CSMD3, FAT4, FLG, KMT2C, LRP1B, MUC5B, MUC16, PLEC, RNF43, SYNE1, TP53, TTN, XIRP2, and ZFHX4 mutations tended to have decreased B cells, T cells, macrophage, neutrophil, and dendritic cells infiltration, except for the ARID1A gene mutations. We also found patients with microsatellite instability-high tumors had higher homologous recombination deficiency (HRD) scores. HRD showed a positive correlation with tumor mutational burden, which might serve as indirect evidence supporting the potential of HRD as a biomarker for GC. These findings highlighted GC's high heterogeneity and complexity and provided valuable insights into the somatic mutations that affect early genetic progression and immune microenvironment.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Center for GI Cancer Diagnosis and Treatment, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Zirui Tang
- School of Software Engineering, Northeastern University, Shenyang, Liaoning 110169, China; Shenzhen Byoryn Technology Co. Ltd, Shenzhen, China
| | - Zhaopeng Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Zhao Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Ping Zhao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Yi Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Kexin Yang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zihan Xia
- The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yinan Wang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| | - Dong Guo
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China.
| |
Collapse
|
29
|
Privitera GF, Alaimo S, Caruso A, Ferro A, Forte S, Pulvirenti A. TMBcalc: a computational pipeline for identifying pan-cancer Tumor Mutational Burden gene signatures. Front Genet 2024; 15:1285305. [PMID: 38645485 PMCID: PMC11026579 DOI: 10.3389/fgene.2024.1285305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 03/11/2024] [Indexed: 04/23/2024] Open
Abstract
Background In the precision medicine era, identifying predictive factors to select patients most likely to benefit from treatment with immunological agents is a crucial and open challenge in oncology. Methods This paper presents a pan-cancer analysis of Tumor Mutational Burden (TMB). We developed a novel computational pipeline, TMBcalc, to calculate the TMB. Our methodology can identify small and reliable gene signatures to estimate TMB from custom targeted-sequencing panels. For this purpose, our pipeline has been trained on top of 17 cancer types data obtained from TCGA. Results Our results show that TMB, computed through the identified signature, strongly correlates with TMB obtained from whole-exome sequencing (WES). Conclusion We have rigorously analyzed the effectiveness of our methodology on top of several independent datasets. In particular we conducted a comprehensive testing on: (i) 126 samples sourced from the TCGA database; few independent whole-exome sequencing (WES) datasets linked to colon, breast, and liver cancers, all acquired from the EGA and the ICGC Data Portal. This rigorous evaluation clearly highlights the robustness and practicality of our approach, positioning it as a promising avenue for driving substantial progress within the realm of clinical practice.
Collapse
Affiliation(s)
- Grete Francesca Privitera
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Salvatore Alaimo
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Anna Caruso
- Department of Physics and Astronomy, University of Catania, Catania, Italy
| | - Alfredo Ferro
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Stefano Forte
- Istituto Oncologico del Mediterraneo (IOM) Ricerca, Viagrande, Italy
| | - Alfredo Pulvirenti
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| |
Collapse
|
30
|
Gao M, Wang M, Chen Y, Wu J, Zhou S, He W, Shu Y, Wang X. Identification and validation of tryptophan metabolism-related lncRNAs in lung adenocarcinoma prognosis and immune response. J Cancer Res Clin Oncol 2024; 150:171. [PMID: 38558328 PMCID: PMC10984901 DOI: 10.1007/s00432-024-05665-x] [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: 12/07/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Tryptophan (Trp) is an essential amino acid. Increasing evidence suggests that tryptophan metabolism plays a complex role in immune escape from Lung adenocarcinoma (LUAD). However, the role of long non-coding RNAs (lncRNAs) in tryptophan metabolism remains to be investigated. METHODS This study uses The Cancer Genome Atlas (TCGA)-LUAD dataset as the training cohort, and several datasets from the Gene Expression Omnibus (GEO) database are merged into the validation cohort. Genes related to tryptophan metabolism were identified from the Molecular Signatures Database (MSigDB) database and further screened for lncRNAs with Trp-related expression. Subsequently, a prognostic signature of lncRNAs related to tryptophan metabolism was constructed using Cox regression analysis, (Least absolute shrinkage and selection operator regression) and LASSO analysis. The predictive performance of this risk score was validated by Kaplan-Meier (KM) survival analysis, (receiver operating characteristic) ROC curves, and nomograms. We also explored the differences in immune cell infiltration, immune cell function, tumor mutational load (TMB), tumor immune dysfunction and exclusion (TIDE), and anticancer drug sensitivity between high- and low-risk groups. Finally, we used real-time fluorescence quantitative PCR, CCK-8, colony formation, wound healing, transwell, flow cytometry, and nude mouse xenotransplantation models to elucidate the role of ZNF8-ERVK3-1 in LUAD. RESULTS We constructed 16 tryptophan metabolism-associated lncRNA prognostic models in LUAD patients. The risk score could be used as an independent prognostic indicator for the prognosis of LUAD patients. Kaplan-Meier survival analysis, ROC curves, and risk maps validated the prognostic value of the risk score. The high-risk and low-risk groups showed significant differences in phenotypes, such as the percentage of immune cell infiltration, immune cell function, gene mutation frequency, and anticancer drug sensitivity. In addition, patients with high-risk scores had higher TMB and TIDE scores compared to patients with low-risk scores. Finally, we found that ZNF8-ERVK3-1 was highly expressed in LUAD tissues and cell lines. A series of in vitro experiments showed that knockdown of ZNF8-ERVK3-1 inhibited cell proliferation, migration, and invasion, leading to cell cycle arrest in the G0/G1 phase and increased apoptosis. In vivo experiments with xenografts have shown that knocking down ZNF8-ERVK3-1 can significantly inhibit tumor size and tumor proliferation. CONCLUSION We constructed a new prognostic model for tryptophan metabolism-related lncRNA. The risk score was closely associated with common clinical features such as immune cell infiltration, immune-related function, TMB, and anticancer drug sensitivity. Knockdown of ZNF8-ERVK3-1 inhibited LUAD cell proliferation, migration, invasion, and G0/G1 phase blockade and promoted apoptosis.
Collapse
Affiliation(s)
- Mingjun Gao
- Dalian Medical University, Dalian, 116000, China
| | | | - Yong Chen
- Dalian Medical University, Dalian, 116000, China
| | - Jun Wu
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Siding Zhou
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Wenbo He
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Yusheng Shu
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, No. 98 Nantong West Road, Yangzhou, 225000, Jiangsu, China.
| | - Xiaolin Wang
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, No. 98 Nantong West Road, Yangzhou, 225000, Jiangsu, China.
| |
Collapse
|
31
|
Li L, Yue P, Zhu J, Li L, Wang K, Yuan G, Song Y. TTN Mutation in Endometrial Endometrioid Carcinoma Is Associated with Poor Clinical Outcomes and High Tumor Mutation Burden. Cancer Invest 2024; 42:297-308. [PMID: 38666471 DOI: 10.1080/07357907.2024.2334249] [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: 02/12/2024] [Accepted: 03/20/2024] [Indexed: 05/28/2024]
Abstract
Endometrioid endometrial carcinoma (EEC) stands as a prevalent gynecologic malignancy in developed regions. However, predicting relapse cases remains challenging, necessitating the identification of a novel biomarker for EEC relapse. The assessment of tumor mutational burden (TMB) is pivotal for immunotherapy in EEC patients. However, both whole-exome sequencing (WES) and targeted sequencing encountered application-related difficulties. In light of this, standardized and simplified techniques for TMB measurement are imperative. In this study, we employed WES on 25 EEC patients (12 relapsed cases and 13 non-relapsed cases) who accepted hysterectomy surgery (CHCAMS cohort). We additionally obtained a total of 391 tumor samples with clinicopathological features from TCGA website to broaden the study cohort. In the CHCAMS cohort, the TTN mutant group showed shorter progression-free survival (p < 0.001) and overall survival (p < 0.001) than TTN wild-type group. Additionally, we discovered that the number of TTN mutations per sample was significantly linked with TMB-WES in CHCAMS cohort and TCGA cohort (p < 0.05). And the number of TTN mutations per sample in POLE mutant group was greater than in the POLE wild-type group (p < 0.0001). In conclusion, TTN mutation may serve as a biomarker for EEC prognosis. TTN mutation is also associated with WES-TMB, and could be a simplified TMB measurement technique.
Collapse
Affiliation(s)
- Lihong Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pinli Yue
- State Key Lab of Molecular Oncology, Laboratory of Cell and Molecular Biology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiarun Zhu
- State Key Lab of Molecular Oncology, Laboratory of Cell and Molecular Biology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luyuan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaipeng Wang
- Record Room, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangwen Yuan
- Record Room, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Gynecology Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Song
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
32
|
Jia Q, Li B, Wang X, Ma Y, Li G. Comprehensive analysis of peroxisome proliferator-activated receptors to predict the drug resistance, immune microenvironment, and prognosis in stomach adenocarcinomas. PeerJ 2024; 12:e17082. [PMID: 38529307 PMCID: PMC10962337 DOI: 10.7717/peerj.17082] [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: 11/16/2023] [Accepted: 02/19/2024] [Indexed: 03/27/2024] Open
Abstract
Background Peroxisome proliferator-activated receptors (PPARs) exert multiple functions in the initiation and progression of stomach adenocarcinomas (STAD). This study analyzed the relationship between PPARs and the immune status, molecular mutations, and drug therapy in STAD. Methods The expression profiles of three PPAR genes (PPARA, PPARD and PPARG) were downloaded from The Cancer Genome Atlas (TCGA) dataset to analyze their expression patterns across pan-cancer. The associations between PPARs and clinicopathologic features, prognosis, tumor microenvironment, genome mutation and drug sensitivity were also explored. Co-expression between two PPAR genes was calculated using Pearson analysis. Regulatory pathways of PPARs were scored using gene set variation analysis (GSVA) package. Quantitative real-time polymerase chain reaction (qRT-PCR), Western blot, Cell Counting Kit-8 (CCK-8) assay and transwell assay were conducted to analyze the expression and function of the PPAR genes in STAD cell lines (AGS and SGC7901 cells). Results PPARA, PPARD and PPARG were more abnormally expressed in STAD samples and cell lines when compared to most of 32 type cancers in TCGA. In STAD, the expression of PPARD was higher in Grade 3+4 and male patients, while that of PPARG was higher in patient with Grade 3+4 and age > 60. Patients in high-PPARA expression group tended to have longer survival time. Co-expression analysis revealed 6 genes significantly correlated with the three PPAR genes in STAD. Single-sample GSEA (ssGSEA) showed that the three PPAR genes were enriched in 23 pathways, including MITOTIC_SPINDLE, MYC_TARGETS_V1, E2F_TARGETS and were closely correlated with immune cells, including NK_cells_resting, T_cells_CD4_memory_resting, and macrophages_M0. Immune checkpoint genes (CD274, SIGLEC15) were abnormally expressed between high-PPAR expression and low-PPAR expression groups. TTN, MUC16, FAT2 and ANK3 genes had a high mutation frequency in both high-PPARA/PPARG and low-PPARA/PPARG expression group. Fourteen and two PPARA/PPARD drugs were identified to be able to effectively treat patients in high-PPARA/PPARG and low-PPARA/PPARG expression groups, respectively. We also found that the chemotherapy drug Vinorelbine was positively correlated with the three PPAR genes, showing the potential of Vinorelbine to serve as a treatment drug for STAD. Furthermore, cell experiments demonstrated that PPARG had higher expression in AGS and SGC7901 cells, and that inhibiting PPARG suppressed the viability, migration and invasion of AGS and SGC7901 cells. Conclusions The current results confirmed that the three PPAR genes (PPARA, PPARD and PPARG) affected STAD development through mediating immune microenvironment and genome mutation.
Collapse
Affiliation(s)
- Qing Jia
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Baozhen Li
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Xiulian Wang
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Yongfen Ma
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Gaozhong Li
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| |
Collapse
|
33
|
Li G, Ping M, Zhang W, Wang Y, Zhang Z, Su Z. Establishment of the molecular subtypes and a risk model for stomach adenocarcinoma based on genes related to reactive oxygen species. Heliyon 2024; 10:e27079. [PMID: 38463816 PMCID: PMC10923688 DOI: 10.1016/j.heliyon.2024.e27079] [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: 11/08/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024] Open
Abstract
Background Oxidative stress promotes the development of stomach adenocarcinoma (STAD) and resistance of STAD patients to chemotherapy. This study developed a risk classification and prognostic model for STAD based on genes related to oxidative stress. Methods Univariate Cox regression and least absolute shrinkage and selection operator (Lasso) regression analysis were performed using transcriptome data of STAD from The Cancer Genome Atlas (TCGA) and reactive oxygen species (ROS)-related genes from Gene Set Enrichment Analysis (GSEA) website to develop a risk model. Genetic landscape, pathway characteristics and immune characteristics between the two risk groups were assessed to evaluate patients' response to anti-tumor therapy. Further, a nomogram was created to evaluate the clinical outcomes of STAD patients. The mRNA levels of genes were detected by reverse transcription quantitative PCR (RT-qPCR). Results Two ROS-related molecular subtypes (subtype C1 and C2) were classified, with subtype C2 having unfavorable prognosis, higher immune score, and greater infiltration of macrophages, myeloid-derived suppressor cells (MDSCs), mast cells, regulatory T cells, and C-C chemokine receptor (CCR). Five ROS-related genes (ASCL2, COMP, NOX1, PEG10, and VPREB3) were screened to develop a prognostic model, the robustness of which was validated in TCGA and external cohorts. RT-qPCR analysis showed that ASCL2, COMP, NOX1, and PEG10 were upregulated, while the mRNA level of VPREB3 was downregulated in gastric cancer cells. The risk score showed a negative relation to tumor mutation burden (TMB). Low-risk patients exhibited higher mutation frequencies of TTN, SYNE1, and ARID1A, higher response rate to immunotherapy and were more sensitive to 32 traditional chemotherapeutic drugs, while high-risk patients were sensitive to 13 drugs. Calibration curve and DCA confirmed the accuracy and reliability of the nomogram. Conclusion These findings provided novel understanding on the mechanism of ROS in STAD. The current study developed a ROS-related signature to help predict the prognosis of patients suffering from STAD and to guide personalized treatment.
Collapse
Affiliation(s)
- Guangyao Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Wuhu, Wuhu, 241000, China
| | - Miaomiao Ping
- Department of Pathophysiology, College of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Weiwei Zhang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Wuhu, Wuhu, 241000, China
| | - Yandong Wang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Wuhu, Wuhu, 241000, China
| | - Zhengjun Zhang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Wuhu, Wuhu, 241000, China
| | - Zhaoran Su
- Department of Gastrointestinal Surgery, People's Hospital of Tongling City, Tongling, 244000, China
| |
Collapse
|
34
|
Shatalov P, Falaleeva N, Bykova E, Korostin D, Belova V, Zabolotneva A, Shinkarkina A, Gorbachev AY, Potievskiy M, Surkova V, Khailova ZV, Kulemin N, Baranovskii D, Kostin A, Kaprin A, Shegai P. Genetic and therapeutic landscapes in cohort of pancreatic adenocarcinomas: next-generation sequencing and machine learning for full tumor exome analysis. Oncotarget 2024; 15:91-103. [PMID: 38329726 PMCID: PMC10852064 DOI: 10.18632/oncotarget.28512] [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: 06/04/2023] [Accepted: 09/04/2023] [Indexed: 02/09/2024] Open
Abstract
About 7% of all cancer deaths are caused by pancreatic cancer (PCa). PCa is known for its lowest survival rates among all oncological diseases and heterogenic molecular profile. Enormous amount of genetic changes, including somatic mutations, exceeds the limits of routine clinical genetic laboratory tests and further stagnates the development of personalized treatments. We aimed to build a mutational landscape of PCa in the Russian population based on full exome next-generation sequencing (NGS) of the limited group of patients. Applying a machine learning model on full exome individual data we received personalized recommendations for targeted treatment options for each clinical case and summarized them in the unique therapeutic landscape.
Collapse
Affiliation(s)
- P.A. Shatalov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - N.A. Falaleeva
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - E.A. Bykova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - D.O. Korostin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - V.A. Belova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - A.A. Zabolotneva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - A.P. Shinkarkina
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - A. Yu Gorbachev
- FSBI “Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine” FMBA, Moscow 119435, Russia
| | - M.B. Potievskiy
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - V.S. Surkova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - Zh V. Khailova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - N.A. Kulemin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| | - Denis Baranovskii
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - A.A. Kostin
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - A.D. Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - P.V. Shegai
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk 249036, Russia
| |
Collapse
|
35
|
Wang K, Shi J, Tong X, Qu N, Kong X, Ni S, Xing J, Li X, Zheng M. TG468: a text graph convolutional network for predicting clinical response to immune checkpoint inhibitor therapy. Brief Bioinform 2024; 25:bbae017. [PMID: 38390990 PMCID: PMC10886443 DOI: 10.1093/bib/bbae017] [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: 11/10/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/24/2024] Open
Abstract
Enhancing cancer treatment efficacy remains a significant challenge in human health. Immunotherapy has witnessed considerable success in recent years as a treatment for tumors. However, due to the heterogeneity of diseases, only a fraction of patients exhibit a positive response to immune checkpoint inhibitor (ICI) therapy. Various single-gene-based biomarkers and tumor mutational burden (TMB) have been proposed for predicting clinical responses to ICI; however, their predictive ability is limited. We propose the utilization of the Text Graph Convolutional Network (GCN) method to comprehensively assess the impact of multiple genes, aiming to improve the predictive capability for ICI response. We developed TG468, a Text GCN model framing drug response prediction as a text classification task. By combining natural language processing (NLP) and graph neural network techniques, TG468 effectively handles sparse and high-dimensional exome sequencing data. As a result, TG468 can distinguish survival time for patients who received ICI therapy and outperforms single gene biomarkers, TMB and some classical machine learning models. Additionally, TG468's prediction results facilitate the identification of immune status differences among specific patient types in the Cancer Genome Atlas dataset, providing a rationale for the model's predictions. Our approach represents a pioneering use of a GCN model to analyze exome data in patients undergoing ICI therapy and offers inspiration for future research using NLP technology to analyze exome sequencing data.
Collapse
Affiliation(s)
- Kun Wang
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jiangshan Shi
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences; 555 Zuchongzhi Road, Shanghai 201203, China
| | - Xiaochu Tong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences; 555 Zuchongzhi Road, Shanghai 201203, China
| | - Ning Qu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences; 555 Zuchongzhi Road, Shanghai 201203, China
| | - Xiangtai Kong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences; 555 Zuchongzhi Road, Shanghai 201203, China
| | - Shengkun Ni
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences; 555 Zuchongzhi Road, Shanghai 201203, China
| | - Jing Xing
- Lingang Laboratory, Shanghai 200031, China
| | - Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Mingyue Zheng
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
36
|
Andruszko A, Szydłowski J, Grabarek BO, Mazur K, Sirek T, Ossowski P, Kozikowski M, Kaminiów K, Zybek-Kocik A, Banaszewski J. Impact of Nutritional Status of Patients with Head and Neck Squamous Cell Carcinoma on the Expression Profile of Ghrelin, Irisin, and Titin. Cancers (Basel) 2024; 16:437. [PMID: 38275878 PMCID: PMC10814803 DOI: 10.3390/cancers16020437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
The goal of this paper was the evaluation of the changes in the expression profile of irisin, ghrelin, and titin in the carcinoma tissue and in the blood of patients with head and neck squamous cell carcinoma (HNSCC), including determining the profile of their expression in relation to patient nutrition. The study included 56 patients with diagnosed squamous cell carcinoma of HNSCC in the T3 and T4 stages of the disease. Healthy control tissue specimens were collected from an area 10 mm outside the histologically negative margin. In turn, the blood and serum from the control group came from healthy volunteers treated for non-oncologic reasons (n = 70). The molecular analysis allowed us to determine the profile of irisin, ghrelin, and titin methylation, evaluate their expression on the level of mRNA (quantitative Reverse Transcription Polymerase Chain Reaction; qRT-PCR) and protein (Enzyme-Linked Immunosorbent Assay Reaction; ELISA) in the carcinoma tissue and the margin of healthy tissue, as well as in serum of patients in the study and control groups. At the start of our observations, a Body Mass Index (BMI) < 18.5 was noted in 42 of the patients, while six months after the treatment a BMI < 18.5 was noted in 29 patients. We also noted a decrease in the expression of irisin, ghrelin, and titin both on the level of mRNA and protein, as well as a potential regulation of their expression via DNA methylation. There is no convincing evidence that the proteins assayed in the present work are specific with regard to HNSSC.
Collapse
Affiliation(s)
- Agata Andruszko
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Jarosław Szydłowski
- Department of Pediatric Otolaryngology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Beniamin Oskar Grabarek
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dąbrowa Górnicza, Poland; (B.O.G.); (P.O.); (K.K.)
- Gyncentrum, Laboratory of Molecular Biology and Virology, 40-851 Katowice, Poland
| | - Katarzyna Mazur
- Faculty of Health Sciences, The Higher School of Strategic Planning in Dąbrowa Górnicza, 41-300 Dabrowa Gornicza, Poland;
| | - Tomasz Sirek
- Department of Plastic Surgery, Faculty of Medicine, Academia of Silesia, 40-555 Katowice, Poland;
- Department of Plastic and Reconstructive Surgery, Hospital for Minimally Invasive and Reconstructive Surgery, 43-316 Bielsko-Biała, Poland
| | - Piotr Ossowski
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dąbrowa Górnicza, Poland; (B.O.G.); (P.O.); (K.K.)
| | - Mieszko Kozikowski
- Faculty of Medicine, Uczelnia Medyczna im. Marii Skłodowskiej-Curie, 00-136 Warszawa, Poland;
| | - Konrad Kaminiów
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dąbrowa Górnicza, Poland; (B.O.G.); (P.O.); (K.K.)
| | - Ariadna Zybek-Kocik
- Department of Metabolism Endocrinology and Internal Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Jacek Banaszewski
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| |
Collapse
|
37
|
Li J, Chen G, Luo Y, Xu J, He J. The molecular subtypes and clinical prognosis characteristic of tertiary lymphoid structures-related gene of cutaneous melanoma. Sci Rep 2023; 13:23097. [PMID: 38155221 PMCID: PMC10754817 DOI: 10.1038/s41598-023-50327-6] [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: 08/28/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023] Open
Abstract
Despite the remarkable efficacy of PD-1-associated immune checkpoint inhibitors in treating cutaneous melanoma (CM), the inconsistency in the expression of PD-1 and its ligand PD-L1, and resulting variability in the effectiveness of immunotherapy, present significant challenges for clinical application. Therefore, further research is necessary to identify tumor-related biomarkers that can predict the prognosis of immunotherapy. Tertiary lymphoid structures (TLSs) have been recognized as a crucial factor in predicting the response of immune checkpoint inhibitors in solid tumors, including CM. However, the study of TLSs in CM is not yet comprehensive. Gene expression profiles have been shown to correlate with CM risk stratification and patient outcomes. In this study, we identified TLS-related genes that can be used for prognostic purposes and developed a corresponding risk model. The impact of TLS-related genes on clinicopathological characteristics, immune infiltration and drug susceptibility was also explored. Our biological function enrichment analysis provided preliminary evidence of related signaling pathways. Our findings provide a new perspective on risk stratification and individualized precision therapy for CM.
Collapse
Affiliation(s)
- Juan Li
- Chongqing Academy of Chinese Materia Medica, Chongqing, People's Republic of China
- Chonging College of Traditional Chinese Medicine, Bishan District, 61 Puguoba Road, Bicheng Street, Chongqing, 402760, People's Republic of China
| | - Gang Chen
- Chongqing Academy of Chinese Materia Medica, Chongqing, People's Republic of China
| | - Yang Luo
- Chongqing Academy of Chinese Materia Medica, Chongqing, People's Republic of China
| | - Jin Xu
- Chongqing Academy of Chinese Materia Medica, Chongqing, People's Republic of China
| | - Jun He
- Chonging College of Traditional Chinese Medicine, Bishan District, 61 Puguoba Road, Bicheng Street, Chongqing, 402760, People's Republic of China.
| |
Collapse
|
38
|
Huang H, Pan Y, Huang J, Zhang C, Liao Y, Du Q, Qin S, Chen Y, Tan H, Chen M, Xu M, Xia M, Liu Y, Li J, Liu T, Zou Q, Zhou Y, Yuan L, Wang W, Liang Y, Pan CY, Liu J, Yao S. Patient-derived organoids as personalized avatars and a potential immunotherapy model in cervical cancer. iScience 2023; 26:108198. [PMID: 38026204 PMCID: PMC10679865 DOI: 10.1016/j.isci.2023.108198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/05/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Cervical cancer remains a significant health issue in developing countries. However, finding a preclinical model that accurately reproduces tumor characteristics is challenging. Therefore, we established a patient-derived organoids (PDOs) biobank containing 67 cases of heterogeneous cervical cancer that mimic the histopathological and genomic characteristics of parental tumors. The in vitro response of the organoids indicated their ability to capture the radiological heterogeneity of the patients. To model individual responses to adoptive T cell therapy (ACT), we expanded tumor-infiltrating lymphocytes (TILs) ex vivo and co-cultured them with paired organoids. The PDOs-TILs co-culture system demonstrates clear responses that correspond to established immunotherapy efficiency markers like the proportion of CTLs. This study supports the potential of the PDOs platform to guide treatment in prospective interventional trials in cervical cancer.
Collapse
Affiliation(s)
- Hua Huang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yuwen Pan
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Jiaming Huang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Chunyu Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yuandong Liao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Qiqiao Du
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Shuhang Qin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yili Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Hao Tan
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Ming Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Manman Xu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Meng Xia
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yunyun Liu
- Sun Yat-Sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Jie Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Tianyu Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Qiaojian Zou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yijia Zhou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Li Yuan
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Wei Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yanchun Liang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Chao yun Pan
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Junxiu Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
39
|
Zeng W, Xie F, Pan Y, Chen Z, Chen H, Liu X, Tian K, Xu D. A comprehensive prognostic score for head and neck squamous cancer driver genes and phenotype traits. Discov Oncol 2023; 14:193. [PMID: 37897503 PMCID: PMC10613197 DOI: 10.1007/s12672-023-00796-y] [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: 04/04/2023] [Accepted: 09/19/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Head and neck squamous cancer (HNSCC) presents variable phenotype and progression features. Clinically applicable, high-accuracy multifactorial prognostic models for HNSCC survival outcomes are warranted and an active area of research. This study aimed to construct a comprehensive prognostic tool for HNSCC overall survival by integrating cancer driver genes with tumor clinical and phenotype information. METHODS Key overall survival-related cancer driver genes were screened from among main effector and reciprocal gene pairs using TCGA data using univariate Cox proportional hazard regression analysis. Independent validation was performed using the GSE41613 dataset. The main effector genes among these were selected using LASSO regression and transcriptome score modeling was performed using multivariate Cox regression followed by validation analysis of the prognostic score. Next, multivariate Cox regression analysis was performed using the transcriptome score combined with age, grade, gender, and stage. An 'Accurate Prediction Model of HNSCC Overall Survival Score' (APMHO) was computed and validated. Enriched functional pathways, gene mutational landscape, immune cell infiltration, and immunotherapy sensitivity markers associated with high and low APMHO scores were analyzed. RESULTS Screening 107 overall survival-related cancer genes and 402 interacting gene pairs, 6 genes: CRLF2, HSP90AA1, MAP2K1, PAFAH1B2, MYCL and SET genes, were identified and a transcriptional score was obtained. Age, stage and transcriptional score were found to be significant predictors in Cox regression analysis and used to construct a final APMHO model showing an AUC > 0.65 and validated. Transcriptional score, age, pathologic_N, pathologic_T, stage, and TCGA_subtype were significantly different in distribution between high and low APMHO groups. High APMHO samples showed significantly higher mutation rate, enriched tumor-related pathways including Hypoxia, unfold_protein_response, Glycolysis, and mTORC1 signaling, along with differences in immune cell infiltration and immune checkpoint, interferon-γ pathway and m6A regulator expression patterns. CONCLUSION The APMHO score combining transcriptional and clinical variables showed good prognostic ability for HNSCC overall survival outcomes and was associated with different patterns of phenotypical features, immune and mutational landscape, and immunotherapy sensitivity marker expression. Future studies should validate this score in independent clinical cohorts.
Collapse
Affiliation(s)
- Wen Zeng
- Ganzhou Cancer Hospital, Gannan Medical College Affiliated Cancer Hospital, No.19, Huayuan Road, Zhanggong Avenue, Ganzhou, Jiangxi, People's Republic of China
| | - Fangfang Xie
- Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Yiyun Pan
- Ganzhou Cancer Hospital, Gannan Medical College Affiliated Cancer Hospital, No.19, Huayuan Road, Zhanggong Avenue, Ganzhou, Jiangxi, People's Republic of China
| | - Zhengcong Chen
- Ganzhou Cancer Hospital, Gannan Medical College Affiliated Cancer Hospital, No.19, Huayuan Road, Zhanggong Avenue, Ganzhou, Jiangxi, People's Republic of China
| | - Hailong Chen
- Ganzhou Cancer Hospital, Gannan Medical College Affiliated Cancer Hospital, No.19, Huayuan Road, Zhanggong Avenue, Ganzhou, Jiangxi, People's Republic of China
| | - Xiaomei Liu
- Ganzhou Cancer Hospital, Gannan Medical College Affiliated Cancer Hospital, No.19, Huayuan Road, Zhanggong Avenue, Ganzhou, Jiangxi, People's Republic of China
| | - Keqiang Tian
- Ganzhou Cancer Hospital, Gannan Medical College Affiliated Cancer Hospital, No.19, Huayuan Road, Zhanggong Avenue, Ganzhou, Jiangxi, People's Republic of China.
| | - Dechang Xu
- Ganzhou Cancer Hospital, Gannan Medical College Affiliated Cancer Hospital, No.19, Huayuan Road, Zhanggong Avenue, Ganzhou, Jiangxi, People's Republic of China.
| |
Collapse
|
40
|
Yang L, Shi W, Li Y, Tong J, Xue X, Zhao Z, Zhang N, Wang D, Fatim I, Liao M, Shi J. SCM is potential resource for non-invasive preimplantation genetic testing based on human embryos single-cell sequencing. Gene 2023; 882:147647. [PMID: 37473972 DOI: 10.1016/j.gene.2023.147647] [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: 03/27/2023] [Revised: 06/09/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
The ongoing development of assisted reproductive technologies has provided hope to individuals struggling with infertility, promising the potential for a healthy pregnancy. One significant innovation in field of pre-implantation genetic screening (PGS) requires the biopsy of embryos or oocytes, which has potential implications for the health and development of the resultant offspring. Therefore, a non-invasive approach to preimplantation genetic screening is highly sought after. The clinical application of non-invasive preimplantation genetic testing (ni-PGT) is currently limited, with its sensitivity and specificity requiring further investigation. In this study, we used 218 human embryos for single-cell whole genome amplification (WGA), along with ni-PGT of blastocoele fluid (BF) and spent culture medium (SCM). Whole blastocyst (WB), trophectoderm biopsy (TB), and inner cell mass (ICM) from embryo biopsies were used as controls to track genomic signal alterations. Our results showed that the overall genome similarity between SCM and ICM was higher than that of BF. Apart from the Y chromosome, both SCM and ICM demonstrated numerous variant sites across other chromosomes.Further categorization of gene variants in these two sample types revealed that missense variants were the most prevalent, single nucleotide polymorphisms were more common than insertions or deletions, and C > T was the dominant single nucleotide variants in both ICM and SCM. Lastly, we found that the mutant genes in SCM and ICM had different biological functions and pathways. This study indicates that SCM provides a more effective source of embryonic DNA for preimplantation genetic screening, offering a novel reference point for genetic screening research.
Collapse
Affiliation(s)
- Luyu Yang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Wenhao Shi
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Yayu Li
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiapei Tong
- College of Information Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Xia Xue
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Zhenghao Zhao
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Ning Zhang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Dongyang Wang
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Israr Fatim
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingzhi Liao
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China.
| | - Juanzi Shi
- The Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China.
| |
Collapse
|
41
|
Wei H, Ren K, Zhang Q, Jin Y, Cao B, Tian Z, Mao T, Ren L. Titin as a potential novel therapeutic target in colorectal cancer. J Cell Mol Med 2023; 27:2937-2944. [PMID: 37499109 PMCID: PMC10538261 DOI: 10.1111/jcmm.17866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/27/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023] Open
Abstract
Colorectal cancer (CRC) is identified as a primary cause of death around the world. The current chemotherapies are not cost-effective. Therefore, finding novel potential therapeutic target is urgent. Titin (TTN) is a muscle protein that is critical in hypertrophic cardiomyopathy. However, its role in CRC is not well understood. The study focused on exploring the possible role of TTN in CRC carcinogenesis. TTN mRNA and protein expression levels presented an obvious downregulation in CRC tissue samples, relative to normal control (p < 0.05). TTN expression significantly correlated with the clinical stage (normal vs. Stage 1, p < 0.05; normal vs. Stage 4, p < 0.05), node metastasis (normal vs. N1, p < 0.05; N1 vs. N2, p < 0.05), histological type (normal vs. adenocarcinoma, p < 0.05), race (Caucasian vs. Asian, p < 0.05; African-American vs. Asian, p < 0.05) and TP53 mutation (normal vs. TP53 mutation, p < 0.05), considering The Cancer Genome Atlas database. However, for patients who had higher TTN expression, the overall survival was remarkably shorter than patients who had low TTN expression. Furthermore, TTN was lowly expressed in four CRC cell lines. TTN overexpression facilitated CRC cells in terms of the proliferation, metastasis and invasion. Based on gene set enrichment analysis, the ERB pathway might be responsible for TTN-related CRC. Besides, TTN was involved in the response to azacitidine. Overall, TTN might serve as a potential novel therapeutic target for treating and overcoming chemotherapy resistance in CRC.
Collapse
Affiliation(s)
- Hongyun Wei
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Keyu Ren
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Qian Zhang
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yanchun Jin
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Bin Cao
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Zibin Tian
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Tao Mao
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Linlin Ren
- Department of GastroenterologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
42
|
Jiang H, Awuti G, Guo X. Construction of an Immunophenoscore-Related Signature for Evaluating Prognosis and Immunotherapy Sensitivity in Ovarian Cancer. ACS OMEGA 2023; 8:33017-33031. [PMID: 37720747 PMCID: PMC10500650 DOI: 10.1021/acsomega.3c04856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023]
Abstract
Ovarian cancer (OC) is the deadliest gynecological malignancy in the world, and immunotherapy is emerging as a promising treatment. Immunophenoscore (IPS) is a robust biomarker distinguishing sensitive responders from immunotherapy. In this study, we aimed to construct a prognostic model for predicting overall survival (OS) and identifying patients who would benefit from immunotherapy. First, we combined The Cancer Genome Atlas (TCGA) and The Cancer Immune Atlas (TCIA) data sets and incorporated 229 OC samples into a training cohort. The validation cohort included 240 OC samples from the Gene Expression Omnibus (GEO) cohort. The training cohort was divided into high- and low-IPS subgroups to obtain differentially expressed genes (DEGs). DEGs with OS were identified by Univariate Cox regression analysis. The least absolute shrinkage and selection operator (LASSO) Cox regression was used to construct the prognostic model. Then, immune and mutation analyses were performed to explore the relationship between the model and the tumor microenvironment (TME) and tumor mutation burden (TMB). Eighty-three DEGs were obtained between the high-and low-IPS subgroups, where 17 DEGs were associated with OS. The five essential genes were selected to establish the prognostic model, which showed high accuracy for predicting OS and could be an independent survival indicator. OC samples that were divided by risk scores showed distinguished immune status, TME, TMB, immunotherapy response, and chemotherapy sensitivity. Similar results were validated in the GEO cohort. We developed an immunophenoscore-related signature associated with the TME to predict OS and response to immunotherapy in OC.
Collapse
Affiliation(s)
- Haonan Jiang
- Shanghai
Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal
Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity
and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guzhanuer Awuti
- Shanghai
Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal
Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity
and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xiaoqing Guo
- Department
of Gynecological Oncology, Shanghai First Maternity and Infant Hospital,
School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
43
|
Li M, Zhao Z, Mak TK, Wang X, Chen J, Ren H, Yu Z, Zhang C. Neutrophil extracellular traps-related signature predicts the prognosis and immune infiltration in gastric cancer. Front Med (Lausanne) 2023; 10:1174764. [PMID: 37636564 PMCID: PMC10447905 DOI: 10.3389/fmed.2023.1174764] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Gastric cancer (GC) is the fifth most prevalent cancer globally, with the third highest case fatality rate. Neutrophil extracellular traps (NETs) are a reticulated structure of DNA, histones, and antimicrobial peptides produced by active neutrophils that trap pathogens. Even though NETs are associated with poorer recurrence-free survival (RFS) and overall survival (OS), the specifics of this interaction between NETs and cancer cells are yet unknown. Methods The keywords "neutrophil extracellular traps and gastric cancer" were used in the GEO database for retrieval, and the GSE188741 dataset was selected to obtain the NETs-related gene. 27 NETs-related genes were screened by univariate Cox regression analysis (p < 0.05). 27 NETs-related genes were employed to identify and categorize NETs-subgroups of GC patients under the Consensus clustering analysis. 808 GC patients in TCGA-STAD combined with GES84437 were randomly divided into a training group (n = 403) and a test group (n = 403) at a ratio of 1:1 to validate the NETs-related signature. Results Based on Multivariate Cox regression and LASSO regression analysis to develop a NETs-related prognosis model. We developed a very specific nomogram to improve the NETs-clinical score's usefulness. Similarly, we also performed a great result in pan-cancer study with NETs-score. Low NETs scores were linked to higher MSI-H (microsatellite instability-high), mutation load, and immune activity. The cancer stem cell (CSC) index and chemotherapeutic treatment sensitivity were also connected to the NET score. Our comprehensive analysis of NETs in GC suggests that NETs have a role in the tumor microenvironment, clinicopathological features, and prognosis. Discussion The NETs-score risk model provides a basis for better prognosis and therapy outcomes in GC patients.
Collapse
Affiliation(s)
- Mingzhe Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zidan Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tsz Kin Mak
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaoqun Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jingyao Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Hui Ren
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhiwei Yu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
44
|
Li H, Sha X, Wang W, Huang Z, Zhang P, Liu L, Wang S, Zhou Y, He S, Shi J. Identification of lysosomal genes associated with prognosis in lung adenocarcinoma. Transl Lung Cancer Res 2023; 12:1477-1495. [PMID: 37577321 PMCID: PMC10413022 DOI: 10.21037/tlcr-23-14] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 07/04/2023] [Indexed: 08/15/2023]
Abstract
Background Lung adenocarcinoma (LUAD) is the most common subtype of lung cancer, representing 40% of all cases of this tumor. Despite immense improvements in understanding the molecular basis, diagnosis, and treatment of LUAD, its recurrence rate is still high. Methods RNA-seq data from The Cancer Genome Atlas (TCGA) LUAD cohort were download from Genomic Data Commons Portal. The GSE13213 dataset from Gene Expression Omnibus (GEO) was used for external validation. Differential prognostic lysosome-related genes (LRGs) were identified by overlapping survival-related genes obtained via univariate Cox regression analysis with differentially expressed genes (DEGs). The prognostic model was built using Kaplan-Meier curves and least absolute shrinkage and selection operator (LASSO) analyses. In addition, univariate and multivariate Cox analyses were employed to identify independent prognostic factors. The responses of patients to immune checkpoint inhibitors (ICIs) were further predicted. The pRRophetic package and rank-sum test were used to compute the half maximal inhibitory concentrations (IC50) of 56 chemotherapeutic drugs and their differential effects in the low- and high-risk groups. Moreover, quantitative real-time polymerase chain reaction, Western blot, and human protein atlas (HPA) database were used to verify the expression of the four prognostic biomarkers in LUAD. Results Of the nine candidate differential prognostic LRGs, GATA2, TFAP2A, LMBRD1, and KRT8 were selected as prognostic biomarkers. The prediction of the risk model was validated to be reliable. Cox independent prognostic analysis revealed that risk score and stage were independent prognostic factors in LUAD. Furthermore, the nomogram and calibration curves of the independent prognostic factors performed well. Differential analysis of ICIs revealed CD276, ICOS, PDCD1LG2, CD27, TNFRSF18, TNFSF9, ENTPD1, and NT5E to be expressed differently in the low- and high-risk groups. The IC50 values of 12 chemotherapeutic drugs, including epothilone.B, JNK.inhibitor.VIII, and AKT.inhibitor.VIII, significantly differed between the two risk groups. KRT8 and TFAP2A were highly expressed, while GATA2 and LMBRD1 were poorly expressed in LUAD cell lines. In addition, KRT8 and TFAP2A were highly expressed, while GATA2 and LMBRD1 were poorly expressed in tumor tissues. Conclusions Four key prognostic biomarkers-GATA2, TFAP2A, LMBRD1, and KRT8-were used to construct a significant prognostic model for LUAD patients.
Collapse
Affiliation(s)
- Houqiang Li
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Xinyu Sha
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Wenmiao Wang
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Zhanghao Huang
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Peng Zhang
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Lei Liu
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Silin Wang
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Youlang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Shuai He
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
| | - Jiahai Shi
- Department of Thoracic Surgery, Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Nantong, China
- Graduate School, Dalian Medical University, Dalian, China
- School of Public Health, Nantong University, Nantong, China
| |
Collapse
|
45
|
Mao Y, Xie H, Lv M, Yang Q, Shuang Z, Gao F, Li S, Zhu L, Wang W. The landscape of objective response rate of anti-PD-1/L1 monotherapy across 31 types of cancer: a system review and novel biomarker investigating. Cancer Immunol Immunother 2023; 72:2483-2498. [PMID: 37022474 PMCID: PMC10992474 DOI: 10.1007/s00262-023-03441-3] [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: 01/10/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have dramatically changed the landscape of cancer treatment. However, only a few patients respond to ICI treatment. Thus, uncovering clinically accessible ICI biomarkers would help identify which patients will respond well to ICI treatment. A comprehensive objective response rate (ORR) data of anti-PD-1/PD-L1 monotherapy in pan-cancer would offer the original data to explore the new biomarkers for ICIs. METHODS We systematically searched PubMed, Cochrane, and Embase for clinical trials on July 1, 2021, limited to the years 2017-2021, from which we obtained studies centering around anti-PD-1/PD-L1 monotherapy. Finally, 121 out of 3099 publications and 143 ORR data were included. All of the 31 tumor types/subtypes can be found in the TCGA database. The gene expression profiles and mutation data were downloaded from TCGA. A comprehensive genome-wide screening of ORR highly correlated mutations among 31 cancers was conducted by Pearson correlation analysis based on the TCGA database. RESULTS According to the ORR, we classified 31 types of cancer into high, medium, and low response types. Further analysis uncovered that "high response" cancers had more T cell infiltration, more neoantigens, and less M2 macrophage infiltration. A panel of 28 biomarkers reviewed from recent articles were investigated with ORR. We also found the TMB as a traditional biomarker had a high correlation coefficient with ORR in pan-cancer, however, the correlation between ITH and ORR was low across pan-cancer. Moreover, we primarily identified 1044 ORR highly correlated mutations through a comprehensive screening of TCGA data, among which USH2A, ZFHX4 and PLCO mutations were found to be highly correlated to strengthened tumor immunogenicity and inflamed antitumor immunity, as well as improved outcomes for ICIs treatment among multiple immunotherapy cohorts. CONCLUSION Our study provides comprehensive data on ORR of anti-PD-1/PD-L1 monotherapy across 31 tumor types/subtypes and an essential reference of ORR to explore new biomarkers. We also screened out a list of 1044 immune response related genes and we showed that USH2A, ZFHX4 and PLCO mutations may act as good biomarkers for predicting patient response to anti-PD-1/PD-L1 ICIs.
Collapse
Affiliation(s)
- Yize Mao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Pancreatobiliary Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Hui Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Imaging Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Minyi Lv
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Institute of Gastroenterology, Supported By National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong Province, China
| | - Qiuxia Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Imaging Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zeyu Shuang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Feng Gao
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Institute of Gastroenterology, Supported By National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong Province, China
| | - Shengping Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Pancreatobiliary Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Lina Zhu
- National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wei Wang
- Department of Clinical Laboratory, Haining People's Hospital, Jiaxing, China.
| |
Collapse
|
46
|
Liu Z, Wan R, Bai H, Wang J. Damage-associated molecular patterns and sensing receptors based molecular subtypes in malignant pleural mesothelioma and implications for immunotherapy. Front Immunol 2023; 14:1104560. [PMID: 37033966 PMCID: PMC10079989 DOI: 10.3389/fimmu.2023.1104560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
OBJECTIVES Malignant pleural mesothelioma (MPM) is characterized as an incredibly aggressive form of cancer with a dismal diagnosis and a dearth of specific biomarkers and therapeutic options. For MPM patients, the effectiveness of immunotherapy may be influenced by damage-associated molecular pattern (DAMP)-induced immunogenic cell death (ICD).The objective of this work is to create a molecular profile associated with DAMPs to categorize MPM patients and predict their prognosis and response to immunotherapy. METHODS The RNA-seq of 397 patients (263 patients with clinical data, 57.2% male, 73.0% over 60 yrs.) were gathered from eight public datasets as a training cohort to identify the DAMPs-associated subgroups of MPMs using K-means analysis. Three validation cohorts of patients or murine were established from TCGA and GEO databases. Comparisons were made across each subtype's immune status, gene mutations, survival prognosis, and predicted response to therapy. RESULTS Based on the DAMPs gene expression, MPMs were categorized into two subtypes: the nuclear DAMPs subtype, which is classified by the upregulation of immune-suppressed pathways, and the inflammatory DAMPs subtype, which is distinguished by the enrichment of proinflammatory cytokine signaling. The inflammatory DAMPs subgroup had a better prognosis, while the nuclear DAMPs subgroup exhibited a worse outcome. In validation cohorts, the subtyping system was effectively verified. We further identified the genetic differences between the two DAMPs subtypes. It was projected that the inflammatory DAMPs subtype will respond to immunotherapy more favorably, suggesting that the developed clustering method may be implemented to predict the effectiveness of immunotherapy. CONCLUSION We constructed a subtyping model based on ICD-associated DAMPs in MPM, which might serve as a signature to gauge the outcomes of immune checkpoint blockades. Our research may aid in the development of innovative immunomodulators as well as the advancement of precision immunotherapy for MPM.
Collapse
Affiliation(s)
| | | | | | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Sun R, Yang Y, Lü W, Yang Y, Li Y, Liu Z, Diao D, Wang Y, Chang S, Lu M, Jiang Q, Dai B, Ma X, Zhao C, Lü M, Zhang J, Ding C, Li N, Zhang J, Xiao Z, Zhou D, Huang C. Single-cell transcriptomic analysis of normal and pathological tissues from the same patient uncovers colon cancer progression. Cell Biosci 2023; 13:62. [PMID: 36944972 PMCID: PMC10031920 DOI: 10.1186/s13578-023-01002-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023] Open
Abstract
The aim of the present study was to elucidate the evolutionary trajectory of colon cells from normal colon mucosa, to adenoma, then to carcinoma in the same microenvironment. Normal colon, adenoma and carcinoma tissues from the same patient were analyzed by single-cell sequencing, which perfectly simulated the process of time-dependent colon cancer due to the same microenvironment. A total of 22 cell types were identified. Results suggest the presence of dominant clones of same cells including C2 goblet cell, epithelial cell subtype 1 (Epi1), enterocyte cell subset 0 (Entero0), and Entero5 in carcinoma. Epi1 and Entero0 were Co-enriched in antibacterial and IL-17 signaling, Entero5 was enriched in immune response and mucin-type O-glycan biosynthesis. We discovered new colon cancer related genes including AC007952.4, NEK8, CHRM3, ANO7, B3GNT6, NEURL1, ODC1 and KCNMA1. The function of TBC1D4, LTB, C2CD4A, AND GBP4/5 in T cells needs to be clarified. We used colon samples from the same person, which provide new information for colon cancer therapy.
Collapse
Affiliation(s)
- Ruifang Sun
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Yang Yang
- School of Public Health, Shaanxi University of Chinese Medicine, Middle Section of Century Avenue, Xianyang, Shaanxi, People's Republic of China.
| | - Weidong Lü
- Department of Thoracic Surgery, Shaanxi Provincial Tumor Hospital, Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Yanqi Yang
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Yulong Li
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, 256 Youyi West Road, Xi'an, Shaanxi, People's Republic of China
| | - Zhigang Liu
- Department of Thoracic Surgery, Shaanxi Provincial Tumor Hospital, Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Dongmei Diao
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Yang Wang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Su'e Chang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xi'an, Shaanxi, People's Republic of China
| | - Mengnan Lu
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xi'an, Shaanxi, People's Republic of China
| | - Qiuyu Jiang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Bingling Dai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xi'an, Shaanxi, People's Republic of China
| | - Chang'an Zhao
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Moqi Lü
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Juan Zhang
- Department of Pathology, Shaanxi Provincial Tumor Hospital, Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Caixia Ding
- Department of Pathology, Shaanxi Provincial Tumor Hospital, Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Na Li
- School of Pharmacy, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang, Henan, People's Republic of China
| | - Jian Zhang
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China
| | - Zhengtao Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China.
| | - Dangxia Zhou
- Department of Pathology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China.
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
48
|
Azzi G, Tavallai M, Aushev VN, Koyen Malashevich A, Botta GP, Tejani MA, Hanna D, Krinshpun S, Malhotra M, Jurdi A, Aleshin A, Kasi PM. Using Tumor-Informed Circulating Tumor DNA (ctDNA)-Based Testing for Patients with Anal Squamous Cell Carcinoma. Oncologist 2023; 28:220-229. [PMID: 36562592 PMCID: PMC10020810 DOI: 10.1093/oncolo/oyac249] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/27/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Anal squamous cell carcinoma (SCCA) is an uncommon malignancy with a rising incidence that has a high cure rate in its early stages. There is an unmet need for a reliable method to monitor response to treatment and assist in surveillance. Circulating tumor DNA (ctDNA) testing has shown great promise in other solid tumors for monitoring disease progression and detecting relapse in real time. This study aimed to determine the feasibility and use of personalized and tumor-informed ctDNA testing in SCCA. PATIENTS AND METHODS We analyzed real-world data from 251 patients (817 plasma samples) with stages I-IV SCCA, collected between 11/5/19 and 5/31/22. The tumor genomic landscape and feasibility of ctDNA testing was examined for all patients. The prognostic value of longitudinal ctDNA testing was assessed in patients with clinical follow-up (N = 37). RESULTS Whole-exome sequencing analysis revealed PIK3CA as the most commonly mutated gene, and no associations between mutations and stage. Anytime ctDNA positivity and higher ctDNA levels (MTM/mL) were associated with metastatic disease (P = .004). For 37 patients with clinical follow-up, median follow-up time was 21.0 months (range: 4.1-67.3) post-diagnosis. For patients with stages I-III disease, anytime ctDNA-positivity after definitive treatment was associated with reduced DFS (HR: 28.0; P = .005). CONCLUSIONS Our study demonstrates the feasibility of personalized and tumor-informed ctDNA testing as an adjunctive tool in patients with SCCA as well as potential use for detection of molecular/minuteimal residual disease, and relapse during surveillance. Prospective studies are needed to better evaluate the use of ctDNA testing in this indication.
Collapse
Affiliation(s)
- Georges Azzi
- HolyCross Medical Group, Ft. Lauderdale, FL, USA
| | | | | | | | | | | | - Diana Hanna
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | | | | | | | - Pashtoon M Kasi
- Corresponding author: Pashtoon M. Kasi, MD, MS, Department of Medical Oncology and Hematology, Weill Cornell Medicine, Englander Institute of Precision Medicine, Meyer Cancer Center, NewYork-Presbyterian Hospital, New York, NY 10021, USA. E-mail:
| |
Collapse
|
49
|
Yao Y, Yang F, Chen A, Hua Q, Gao W. Costimulatory molecule-related lncRNA model as a potential prognostic biomarker in non-small cell lung cancer. Cancer Med 2023; 12:6419-6436. [PMID: 36305249 PMCID: PMC10028169 DOI: 10.1002/cam4.5391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/14/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Costimulatory molecules have been demonstrated to exert essential roles in multiple cancers. However, their role in lung cancer remains elusive. Here, we sought to identify costimulatory molecule-related lncRNAs in non-small cell lung cancer (NSCLC) and establish a prognostic signature to predict the prognosis of patients with NSCLC. METHODS A total of 535 lung adenocarcinoma (LUAD) and 502 lung squamous cell carcinoma (LUSC) patients from the cancer genome atlas (TCGA) database were recruited. A novel costimulatory molecule-based lncRNA prognostic model was constructed using the least absolute shrinkage and selection operator (LASSO) algorithm to predict the overall survival. The Homo_sapiens.GRCh38 data set was set as a reference file for probe annotation. RESULTS A total of 593 costimulatory molecule-related lncRNAs were extracted. After analysis, six costimulatory molecule-related lncRNAs (AC084859.1, AC079949.2, HSPC324, LINC01150, LINC01150, and AC090617.5) were screened. A prognostic model based on the six lncRNAs was established using systematic bioinformatics analyses. The prognostic model had a prognostic value in NSCLC patients. Furthermore, a prognostic nomogram was established based on clinical parameters and a risk-score model. Patients with different risk scores had considerably different tumor-infiltrating immune cells, somatic mutational loading, clinical outcomes, signaling pathways, and immunotherapy efficacy. In addition, LINC01137 was associated with unfavorable disease outcomes and fueled tumor progression in NSCLC. CONCLUSION Taken together, our study demonstrated that a costimulatory molecule-related lncRNA model could be a potential prognostic biomarker in NSCLC. Moreover, LINC01137 could facilitate the proliferation and invasion of lung cancer.
Collapse
Affiliation(s)
- Yuanshan Yao
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Department of Thoracic Oncology, Ningbo No. 2 Hospital, Ningbo, China
| | - Fuzhi Yang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Anna Chen
- Ningbo CRRC Times Transducer Technology Co., Ltd., Ningbo, China
| | - Qingwang Hua
- Department of Thoracic Oncology, Ningbo No. 2 Hospital, Ningbo, China
| | - Wen Gao
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
50
|
Wang Q, Zhang W, Guo Y, Shi F, Li Y, Kong Y, Lyu J, Wang S. A mutational signature and significantly mutated driver genes associated with immune checkpoint inhibitor response across multiple cancers. Int Immunopharmacol 2023; 116:109821. [PMID: 36753986 DOI: 10.1016/j.intimp.2023.109821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/19/2023] [Accepted: 01/28/2023] [Indexed: 02/08/2023]
Abstract
Immune checkpoint inhibitor (ICI) treatments dramatically prolong the survival outcomes of several advanced cancers. However, as multiple studies reported, only a subset of patients could benefit from the ICI treatment. In this study, we aim to uncover novel molecular biomarkers predictive of immunotherapy efficacy across multiple cancers. Pre-treatment somatic mutational profiles and immunotherapy clinical information were obtained from 1097 samples of multiple cancers, including melanoma, non-small cell lung cancer (NSCLC), clear cell renal cell carcinoma (ccRCC), bladder carcinoma (BLCA), and head and neck squamous cell carcinoma (HNSCC). Mutational signatures, molecular subtypes, and significantly mutated genes (SMGs) were determined, and their connections with ICI response and outcome were also evaluated. We extracted a total of six mutational signatures across all samples. Among, a mutational signature featured by T > C substitutions was identified to associate with an ICI resistance. A molecular subtype determined based on mutational activities was connected with a significantly improved ICI response rate and outcome. Totaling 50 SMGs were identified, and we observed that patients with COL11A1 or COL4A6 mutations exhibited a superior ICI treatment efficacy than those without such mutations. In this study, we uncovered several novel molecular determinants of cancer immunotherapy response under a multiple-cancer setting, which provides clues for enrolling patients to receive immunotherapy and customizing personalized treatment strategies.
Collapse
Affiliation(s)
- Qinghua Wang
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Wenjing Zhang
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Weifang Medical University, Weifang, Shandong 261053, China
| | - Yuxian Guo
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Weifang Medical University, Weifang, Shandong 261053, China
| | - Fuyan Shi
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Weifang Medical University, Weifang, Shandong 261053, China
| | - Yuting Li
- Tianjin Cancer Institute, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yujia Kong
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Weifang Medical University, Weifang, Shandong 261053, China
| | - Juncheng Lyu
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Weifang Medical University, Weifang, Shandong 261053, China
| | - Suzhen Wang
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Weifang Medical University, Weifang, Shandong 261053, China
| |
Collapse
|