1
|
Hu B, Yin G, Zhu J, Bai Y, Sun X. Continuous prediction for tumor mutation burden based on transcriptional data in gastrointestinal cancers. BMC Med Inform Decis Mak 2024; 24:384. [PMID: 39695561 DOI: 10.1186/s12911-024-02794-8] [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: 08/19/2023] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Tumor mutation burden (TMB) has been considered a biomarker for utilization of immune checkpoint inhibitors(ICIs), but whole exome sequencing(WES) and cancer gene panel(CGP) based on next generation sequencing for TMB detection are costly. Here, we use transcriptome data of TCGA to construct a model for TMB prediction in gastrointestinal tumors. METHODS Transcriptome data, somatic mutation data and clinical data of four gastrointestinal tumors from TCGA, including esophageal cancer (ESCA), stomach adenocarcinoma (STAD), colon adenocarcinoma (COAD) and rectal adenocarcinoma (READ). Using R, we performed visual analysis of somatic mutation data, differentially expressed genes (DEGs) function enrichment analysis, gene set enrichment analysis (GSEA), and estimated TMB value in clinic. Finally, a deep neural network (DNN) model was constructed for TMB prediction. RESULTS Visualization of somatic mutation data summarized the classification of mutation, frequency of each mutation type, and top-mutated genes. GSEA showed the enrichment of CD4+/CD8+ T cells in the high TMB group and the activation of tumor suppressing pathways. Single-sample GSEA (ssGSEA) manifested that the high-TMB group had higher level of multiple immune cells infiltration. In addition, distribution of TMB was related to clinical parameters. Like age, M stage, N stage, AJCC stage, and overall survival(OS). After model optimization using genetic algorithm, in the training set, validation set, and testing set, the Pearson relevance coefficient r between predicted values and actual values reaches 0.98, 0.82, and 0.92, respectively; the coefficient of determination R2 is 0.95, 0.82, and 0.7, respectively. CONCLUSION TMB correlates with clinicopathological parameters in gastrointestinal carcinoma, and patients with high TMB have higher levels of immune infiltration. In addition, the DNN model based on 31 genes predicts TMB of gastrointestinal tumors in a high accuracy.
Collapse
Affiliation(s)
- Beibei Hu
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guohui Yin
- Key Laboratory of Traffic Safety On Track (Central South University), Ministry of Education, School of Traffic and Transportation Engineering, Central South University, Changsha, 410075, China
| | - Jialin Zhu
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yi Bai
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuren Sun
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Tang Y, Shi T, Lin S, Fang T. Current status of research on the mechanisms of tumor-associated macrophages in esophageal cancer progression. Front Oncol 2024; 14:1450603. [PMID: 39678502 PMCID: PMC11638059 DOI: 10.3389/fonc.2024.1450603] [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: 06/17/2024] [Accepted: 09/27/2024] [Indexed: 12/17/2024] Open
Abstract
Esophageal carcinoma (EC) is one of the most common tumors in China and seriously affects patient survival and quality of life. In recent years, increasing studies have shown that the tumor microenvironment is crucial in promoting tumor progression and metastasis. Tumor-associated macrophages (TAM) are key components of the tumor immune microenvironment and promote both tumor growth and antitumor immunity. Much evidence suggests that TAMs are closely associated with esophageal tumors. However, understanding of the clinical value and mechanism of action of TAM in esophageal cancer remains limited. Therefore, we reviewed the status of research on the role and mechanism of action of TAM in EC progression and summarized its potential clinical application value to provide a theoretical basis for the clinical treatment of EC.
Collapse
Affiliation(s)
- Yuchao Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Tingting Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
3
|
Zhang XJ, Yu Y, Zhao HP, Guo L, Dai K, Lv J. Mechanisms of tumor immunosuppressive microenvironment formation in esophageal cancer. World J Gastroenterol 2024; 30:2195-2208. [PMID: 38690024 PMCID: PMC11056912 DOI: 10.3748/wjg.v30.i16.2195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/05/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
As a highly invasive malignancy, esophageal cancer (EC) is a global health issue, and was the eighth most prevalent cancer and the sixth leading cause of cancer-related death worldwide in 2020. Due to its highly immunogenic nature, emer-ging immunotherapy approaches, such as immune checkpoint blockade, have demonstrated promising efficacy in treating EC; however, certain limitations and challenges still exist. In addition, tumors may exhibit primary or acquired resistance to immunotherapy in the tumor immune microenvironment (TIME); thus, understanding the TIME is urgent and crucial, especially given the im-portance of an immunosuppressive microenvironment in tumor progression. The aim of this review was to better elucidate the mechanisms of the suppressive TIME, including cell infiltration, immune cell subsets, cytokines and signaling pathways in the tumor microenvironment of EC patients, as well as the downregulated expression of major histocompatibility complex molecules in tumor cells, to obtain a better understanding of the differences in EC patient responses to immunotherapeutic strategies and accurately predict the efficacy of immunotherapies. Therefore, personalized treatments could be developed to maximize the advantages of immunotherapy.
Collapse
Affiliation(s)
- Xiao-Jun Zhang
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Yan Yu
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - He-Ping Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Lei Guo
- Department of Spinal Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Kun Dai
- Department of Clinical Laboratory, Yanliang Railway Hospital of Xi’an, Xi’an 710089, Shaanxi Province, China
| | - Jing Lv
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| |
Collapse
|
4
|
Yi B, Zeng J, Li L, Zhang J, Chen Y, Gao Y. Prognostic and clinical significance of tumor-associated macrophages in esophageal squamous cell carcinoma after surgery: do biomarkers and distributions matter? Biosci Rep 2024; 44:BSR20231194. [PMID: 38501293 PMCID: PMC10994813 DOI: 10.1042/bsr20231194] [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: 07/03/2023] [Revised: 01/19/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND The role of tumor-associated macrophages (TAMs) in patients with esophageal squamous cell carcinoma (ESCC) following surgery remains controversial. Hence, we performed the present study to systematically analyze the prognostic and clinical significance of distinct TAMs biomarkers and distributions in ESCC patients underwent surgery. METHODS PubMed, Web of Science, and EMBASE databases were searched up to March 31, 2023. The pooled analysis was conducted to evaluate the effects of TAMs on overall survival (OS), disease-free survival (DFS), and clinicopathological characteristics using fixed-effects or random-effect model. RESULTS Involving a total of 2,502 ESCC patients underwent surgery from 15 studies, the results suggested that the total count of CD68+ TAMs was inversely associated with OS and DFS in ESCC patients, which was also noticed in the relationship of CD68+ TAMs in tumor islet (TI) with OS (all P<0.05), although no association between CD68+ TAMs in tumor stroma (TS) and OS (P>0.05). Moreover, either islet or stromal CD163+ TAMs density was a prognostic factor ESCC (all P<0.05). Similarly, an elevated CD204+ TAMs density in TI predicted a poor DFS (P<0.05), although CD204+ TAMs in TI had no relationship with OS (P>0.05). Besides, a high CD68+ TAMs density was significantly associated with lymphatic vessel invasion, vascular invasion, and lymph node metastasis (all P<0.05). CONCLUSION Our results demonstrated the prognostic and clinical significance of TAMs in ESCC patients underwent surgery. TAMs should be considered a target that could improve prognostic stratification and clinical outcomes in ESCC after surgery.
Collapse
Affiliation(s)
- Bin Yi
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008 Hunan, P.R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha,410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, P.R. China
| | - Jun Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008 Hunan, P.R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha,410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, P.R. China
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008 Hunan, P.R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha,410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, P.R. China
| | - Junjie Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008 Hunan, P.R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha,410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, P.R. China
| | - Yufan Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008 Hunan, P.R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha,410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, P.R. China
| | - Yang Gao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008 Hunan, P.R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha,410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, P.R. China
| |
Collapse
|
5
|
Deng Z, Qishan S, Zhang Q, Wang J, Yue Y, Geng L, Wu N. Low molecular weight fucoidan LF2 improves the immunosuppressive tumor microenvironment and enhances the anti-pancreatic cancer activity of oxaliplatin. Biomed Pharmacother 2024; 173:116360. [PMID: 38422657 DOI: 10.1016/j.biopha.2024.116360] [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: 12/04/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
Chemotherapy remains the cornerstone of pancreatic cancer treatment. However, the dense interstitial and immunosuppressive microenvironment frequently render the ineffective anti-tumor activity of chemotherapeutic agents. Macrophages play a key role in the tumor immunomodulation. In this study, we found that low molecular weight of fucoidan (LF2) directly regulated the differentiation of mononuclear macrophages into the CD86+ M1 phenotype. LF2 significantly upregulated the expressions of M1 macrophage-specific cytokines, including iNOS, IL-6, TNFα and IL-12. LF2 modulated macrophage phenotypic transformation through activation of TLR4-NFκB pathway. Furthermore, we observed that LF2 enhanced the pro-apoptotic activity of oxaliplatin (OXA) in vitro by converting macrophages to a tumoricidal M1 phenotype. Meanwhile, LF2 increased intratumoral M1 macrophage infiltration and ameliorated the immunosuppressed tumor microenvironment, which in turn enhanced the anti-pancreatic ductal adenocarcinoma (PDAC) activity of OXA in vivo. Taken together, our results suggested that LF2 could act as a TLR4 agonist targeting macrophages and has a synergistic effect against PDAC when combined with OXA.
Collapse
Affiliation(s)
- Zhenzhen Deng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Suo Qishan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Quanbin Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Yang Yue
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Lihua Geng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Ning Wu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine drugs and biological products, Pilot National Laboratory for Marine Science and Technology (Qingdao), China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Zhang J, Dong Y, Di S, Xie S, Fan B, Gong T. Tumor associated macrophages in esophageal squamous carcinoma: Promising therapeutic implications. Biomed Pharmacother 2023; 167:115610. [PMID: 37783153 DOI: 10.1016/j.biopha.2023.115610] [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: 08/16/2023] [Revised: 09/18/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023] Open
Abstract
Esophageal squamous carcinoma (ESCC) is a prevalent and highly lethal malignant tumor, with a five-year survival rate of approximately 20 %. Tumor-associated macrophages (TAMs) are the most prominent immune cells in the tumor microenvironment (TME), comprising over 50 % of the tumor volume. TAMs can be polarized into two distinct phenotypes, M1-type and M2-type, through interactions with cancer cells. M2-type TAMs are more abundant than M1-type TAMs in the TME, contributing to tumor progression, such as tumor cell survival and the construction of an immunosuppressive environment. This review focuses on the role of TAMs in ESCC, including their polarization, impact on tumor proliferation, angiogenesis, invasion, migration, therapy resistance, and immunosuppression. In addition, we discuss the potential of targeting TAMs for clinical therapy in ESCC. A thorough comprehension of the molecular biology about TAMs is essential for the development of innovative therapeutic strategies to treat ESCC.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China; Department of Thoracic Surgery, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yanxin Dong
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China; Department of Thoracic Surgery, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shouyin Di
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Shun Xie
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Boshi Fan
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China.
| | - Taiqian Gong
- Department of Thoracic Surgery, the Sixth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
7
|
Hiranuma K, Asami Y, Kato MK, Murakami N, Shimada Y, Matsuda M, Yazaki S, Fujii E, Sudo K, Kuno I, Komatsu M, Hamamoto R, Makinoshima H, Matsumoto K, Ishikawa M, Kohno T, Terao Y, Itakura A, Yoshida H, Shiraishi K, Kato T. Rare FGFR fusion genes in cervical cancer and transcriptome-based subgrouping of patients with a poor prognosis. Cancer Med 2023; 12:17835-17848. [PMID: 37537783 PMCID: PMC10524028 DOI: 10.1002/cam4.6415] [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: 03/09/2023] [Revised: 06/25/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Although cervical cancer is often characterized as preventable, its incidence continues to increase in low- and middle-income countries, underscoring the need to develop novel therapeutics for this disease.This study assessed the distribution of fusion genes across cancer types and used an RNA-based classification to divide cervical cancer patients with a poor prognosis into subgroups. MATERIAL AND METHODS RNA sequencing of 116 patients with cervical cancer was conducted. Fusion genes were extracted using StarFusion program. To identify a high-risk group for recurrence, 65 patients who received postoperative adjuvant therapy were subjected to non-negative matrix factorization to identify differentially expressed genes between recurrent and nonrecurrent groups. RESULTS We identified three cases with FGFR3-TACC3 and one with GOPC-ROS1 fusion genes as potential targets. A search of publicly available data from cBioPortal (21,789 cases) and the Center for Cancer Genomics and Advanced Therapeutics (32,608 cases) showed that the FGFR3 fusion is present in 1.5% and 0.6% of patients with cervical cancer, respectively. The frequency of the FGFR3 fusion gene was higher in cervical cancer than in other cancers, regardless of ethnicity. Non-negative matrix factorization identified that the patients were classified into four Basis groups. Pathway enrichment analysis identified more extracellular matrix kinetics dysregulation in Basis 3 and more immune system dysregulation in Basis 4 than in the good prognosis group. CIBERSORT analysis showed that the fraction of M1 macrophages was lower in the poor prognosis group than in the good prognosis group. CONCLUSIONS The distribution of FGFR fusion genes in patients with cervical cancer was determined by RNA-based analysis and used to classify patients into clinically relevant subgroups.
Collapse
Affiliation(s)
- Kengo Hiranuma
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineTokyoJapan
| | - Yuka Asami
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Obstetrics and GynecologyShowa University School of MedicineTokyoJapan
| | - Mayumi Kobayashi Kato
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Naoya Murakami
- Department of Radiation OncologyNational Cancer Center HospitalTokyoJapan
| | - Yoko Shimada
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
| | - Maiko Matsuda
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
| | - Shu Yazaki
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Erisa Fujii
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Kazuki Sudo
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Ikumi Kuno
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Masaaki Komatsu
- Division of Medical AI Research and DevelopmentNational Cancer Center Research InstituteTokyoJapan
- Cancer Translational Research TeamRIKEN Center for Advanced Intelligence ProjectTokyoJapan
| | - Ryuji Hamamoto
- Division of Medical AI Research and DevelopmentNational Cancer Center Research InstituteTokyoJapan
- Cancer Translational Research TeamRIKEN Center for Advanced Intelligence ProjectTokyoJapan
| | | | - Koji Matsumoto
- Department of Obstetrics and GynecologyShowa University School of MedicineTokyoJapan
| | - Mitsuya Ishikawa
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Takashi Kohno
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
| | - Yasuhisa Terao
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineTokyoJapan
| | - Atsuo Itakura
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineTokyoJapan
| | - Hiroshi Yoshida
- Department of Diagnostic PathologyNational Cancer Center HospitalTokyoJapan
| | - Kouya Shiraishi
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Clinical GenomicsNational Cancer Center Research InstituteTokyoJapan
| | - Tomoyasu Kato
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
8
|
Kong P, Yang H, Tong Q, Dong X, Yi MA, Yan D. Expression of tumor-associated macrophages and PD-L1 in patients with hepatocellular carcinoma and construction of a prognostic model. J Cancer Res Clin Oncol 2023; 149:10685-10700. [PMID: 37306737 PMCID: PMC10423131 DOI: 10.1007/s00432-023-04949-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an inflammation-associated tumor involved in immune tolerance and evasion in the immune microenvironment. Immunotherapy can enhance the immune response of the body, break immune tolerance, and then recognize and kill tumor cells. The polarization homeostasis of M1 and M2 macrophages in tumor microenvironment (TME) is involved in the occurrence and development of tumors and has been considered a hot topic in tumor research. Programmed cell death ligand 1 (PD-L1) plays an important role in the polarity of TAM and affects the prognosis of HCC patients as a target of immunotherapy. To this end, efforts were hereby made to further explore the application value of PD-L1, M1 macrophages (CD86), and M2 macrophages (CD206) in the prognosis assessment of HCC, their correlation with immune cell infiltration in HCC tissues, and their bioenrichment function. METHODS The gene expression omnibus (GEO) and the Cancer Genome Atlas (TCGA) database were used to analyze the expression of PD-L1, CD86, and CD206 in different tumor tissues. The correlation between the expression of PD-L1, CD86, and CD206 and the infiltration of immune cells was analyzed using the Tumor Immune Estimation Resource (TIMER). The tissue specimens and clinicopathological data of hepatocellular carcinoma patients having undergone surgical treatment in our hospital were collected. Immunohistochemistry was used to verify the expression of PD-L1, CD86, and CD206, and analyze the relationship with clinicopathological features and prognosis of patients. Besides, nomogram was constructed to predict the overall survival (OS) of patients at 3 and 5 years. Finally, the protein-protein interaction network information was analyzed using STRING database, and GO analysis and KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis were performed to study the biological functions of PD-L1, CD86, and CD206. RESULT Bioinformatics analysis found that PD-L1, CD86, and CD206 were underexpressed in various tumor tissues including liver cancer, while the present immunohistochemical detection found that PD-L1, CD86, and CD206 were overexpressed in liver cancer tissues. Expressions of PD-L1, CD86, and CD206 were positively correlated with the infiltration level of immune cells in liver cancer, while the expression of PD-L1 was positively correlated with the degree of tumor differentiation. Meanwhile, the expression level of CD206 was positively correlated with gender and preoperative hepatitis, and patients with high expression of PD-L1 or low expression of CD86 had poor prognosis. AJCC stage, preoperative hepatitis, and the expression levels of PD-L1 and CD86 in cancer tissues were independent risk factors affecting survival of patients after radical hepatoma surgery. KEGG pathway enrichment analysis showed that PD-L1 was significantly enriched in T cell aggregation and lymphocyte aggregation, and might be involved in the formation of T cell antigen receptor CD3 complex and cell membrane. Besides, CD86 was significantly enriched in positive regulation of cell adhesion, regulation of mononuclear cell proliferation, regulation of leukocyte proliferation, and transduction of T cell receptor signaling pathway, while CD206 was significantly enriched in type 2 immune response, cellular response to LPS, cellular response to LPS, and involvement in cellular response to LPS. CONCLUSION In conclusion, these results suggest that PD-L1, CD86, and CD206 may be involved not only in the occurrence and development of HCC, but also in immune regulation, indicating the potential role of PD-L1 and CD86 as potential biomarkers and new therapeutic targets for prognosis assessment of liver cancer.
Collapse
Affiliation(s)
- Panpan Kong
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Huan Yang
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Qing Tong
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Xiaogang Dong
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Mamumaimaitijiang-Abula Yi
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Dong Yan
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China.
| |
Collapse
|
9
|
Wang S, Xu G, Li M, Zheng J, Wang Y, Feng X, Luo J, Wang S, Liu H, Duan W, Zhang H, Huang D, Zhao F, Nie Y, Yang J. M1 macrophage predicted efficacy of neoadjuvant camrelizumab combined with chemotherapy vs chemotherapy alone for locally advanced ESCC: A pilot study. Front Oncol 2023; 13:1139990. [PMID: 36969032 PMCID: PMC10038194 DOI: 10.3389/fonc.2023.1139990] [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: 01/08/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
Introduction The efficacy and safety of immunotherapy have been widely recognized in gastrointestinal-related cancers. However, the efficacy of neoadjuvant camrelizumab for locally advanced esophageal squamous cell carcinoma (ESCC) has not been firmly established. This study compared the efficacy of camrelizumab in combination with neoadjuvant DCF (docetaxel, cisplatin and fluorouracil), with DCF alone for ESCC, and exploring biomarkers related to immune infiltration of the ESCC immunotherapy response. Methods We enrolled and randomly assigned patients with stage II-IVa ESCC to two study treatments: camrelizumab combined with docetaxel, cisplatin and fluorouracil (DCF) regimen and DCF regimen alone. The tissue for multiplex immunofluorescence (mIF) was obtained before and after neoadjuvant therapy. The Response Evaluation Criteria in Solid Tumors RECIST Version 1.1 (RECIST 1.1) and Tumor Regression Grade (TRG) was used to evaluate efficacy. Results A total of 30 patients were enrolled in the study. Following neoadjuvant camrelizumab, the objective response rate (ORR) and the disease control rate (DCR) were 46.7% (7/15) and 95.7% (14/15), respectively. No patients reported complete remission, while ORR and DCR in the chemotherapy group were 26.7% (4/15) and 86.7% (13/15), respectively. R0 resection after neoadjuvant treatment was achieved in 3 out of 15 patients in the combined group and in all patients (15/15) in the chemotherapy group. In the combined group, M1-type tumor-associated macrophages and CD56dim NK cells were more abundant in responders than in non-responders (p < 0.05). A higher M1/M2 ratio was observed in responders (p < 0.05). With respect to the NGS, among the copy number amplified genes, the 11q13 amplicon (CCND1/FGF19/FGF4/FGF3) showed the highest frequency (47%, 7/15). Conclusions Neoadjuvant camrelizumab combined with chemotherapy improved ORR in locally advanced ESCC. M1-type tumor-associated macrophages and CD56dim NK cells might be utilized to predict camrelizumab efficacy.
Collapse
Affiliation(s)
- Shu Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Guanghui Xu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Mengbin Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jiyang Zheng
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yuhao Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xiangying Feng
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jialin Luo
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Shibo Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Huan Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Weiming Duan
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Hushan Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Depei Huang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Feilong Zhao
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Jianjun Yang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
10
|
Huang Z, Li B, Qin H, Mo X. Invasion characteristics and clinical significance of tumor-associated macrophages in gastrointestinal Krukenberg tumors. Front Oncol 2023; 13:1006183. [PMID: 36910657 PMCID: PMC9999382 DOI: 10.3389/fonc.2023.1006183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/06/2023] [Indexed: 03/14/2023] Open
Abstract
Background Tumor-associated macrophages (TAMs) have been used as potential drug targets in preclinical research and clinical trials of various cancers. However, their distribution in Krukenberg tumors (KTs) remains unclear. We investigated the expression and prognostic value of TAMs in patients with gastrointestinal cancer with KTs. Methods The infiltration of various types of TAMs was detected in surgical tissues of 35 patients with KTs using immunohistochemical staining. The level of infiltration of TAMs in tumor nests (TN), tumor stroma (TS), and invasive margin (IM) areas was evaluated. The Kaplan-Meier method and univariate/multivariate Cox regression risk models were used to analyze the relationship between the degree of TAMs invasion and overall survival (OS) and progression-free survival (PFS). Results The distribution of TAMs exhibited spatial heterogeneity between TN, TS, and IM regions in primary tumor (PT) and KT tissues. TAMs infiltrated in the TN had greater prognostic value and were barely influenced by preoperative neoadjuvant therapy, despite similar grades of invasion in PT and KT tissues. Moreover, the number of CD68+ TAMs in TN of KT tissues was an independent risk factor affecting patient OS, whereas tumor resection scope might be an independent risk factor affecting patient PFS. Conclusions In view of the close relationship between TAMs, the tumor microenvironment and patient prognosis, targeting TAMs combined with chemotherapy is expected to become a new approach for the treatment of patients with KTs.
Collapse
Affiliation(s)
| | | | - Haiquan Qin
- Guangxi Clinical Research Center for Colorectal Cancer, Division of Colorectal & Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xianwei Mo
- *Correspondence: Haiquan Qin, ; Xianwei Mo,
| |
Collapse
|
11
|
Dai Y, Chen W, Huang J, Zheng L, Lin Q, Cui T, Huang C. Multiomics Integrative Analysis Identifying EPC1 as a Prognostic Biomarker in Head and Neck Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1074412. [PMID: 36158885 PMCID: PMC9507713 DOI: 10.1155/2022/1074412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/10/2022] [Indexed: 12/09/2022]
Abstract
Background Biomarker research in head and neck squamous cell carcinoma (HNSCC) is constantly revealing promising findings. An enhancer of polycomb homolog 1 (EPC1) was found to play a procancer role in nasopharyngeal carcinoma (NPC), but its role in HNSCC with strong heterogeneity is still unclear. Herein, we investigated the prognostic significance and related mechanisms of EPC1 in HNSCC. Methods The Kaplan-Meier plotter was used to evaluate the prognostic significance of EPC1. Based on a range of published public databases, the multiomics expression of EPC1 in HNSCC was explored to investigate the mechanisms affecting prognosis. Results According to the clinical data, high EPC1 expression in HNSCC was a predictor of patient prognosis (hazard ratio (HR) = 0.64; 95% confidence interval (CI) 0.49-0.83; P < 0.01). EPC1 expression varied among clinical subtypes and was related to key factors, such as TP53 and human papillomavirus (HPV) (P < 0.05). At the genetic level, EPC1 expression level may be associated with protein phosphorylation, cell adhesion, cancer-related pathways, etc. For the noncoding region, a competing endogenous RNA network was constructed, and 6 microRNAs and 12 long noncoding RNAs were identified. At the protein level, a protein-protein interaction (PPI) network related to EPC1 expression was constructed and found to be involved in HPV infection, endocrine resistance, and multiple cancer pathways. At the immune level, EPC1 expression was correlated with a variety of immune cells and immune molecules, which together constituted the immune microenvironments of tumors. Conclusion High EPC1 expression may predict a better prognosis in HNSCC, as it is more frequently found in HNSCC with HPV infection. EPC1 may participate in the genomics, transcriptomics, proteomics, and immunomics of HNSCC, and the results can provide a reference for the development of targeted drugs and evaluation of patient prognosis.
Collapse
Affiliation(s)
- Yongmei Dai
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University & Fujian Provincial Hospital, Fujian 350001, China
| | - Wenhan Chen
- The Second Clinical Medical College of Fujian Medical University, Fujian 362000, China
- Department of Clinical Medicine, Fujian Medical University, Fujian 350122, China
| | - Junpeng Huang
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University & Fujian Provincial Hospital, Fujian 350001, China
| | - Lijing Zheng
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University & Fujian Provincial Hospital, Fujian 350001, China
| | - Qing Lin
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University & Fujian Provincial Hospital, Fujian 350001, China
| | - Tongjian Cui
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University & Fujian Provincial Hospital, Fujian 350001, China
| | - Chen Huang
- Shengli Clinical Medical College of Fujian Medical University & Fujian Provincial Hospital, Fujian 350001, China
| |
Collapse
|
12
|
Cui MY, Yi X, Cao ZZ, Zhu DX, Wu J. Targeting Strategies for Aberrant Lipid Metabolism Reprogramming and the Immune Microenvironment in Esophageal Cancer: A Review. JOURNAL OF ONCOLOGY 2022; 2022:4257359. [PMID: 36106333 PMCID: PMC9467784 DOI: 10.1155/2022/4257359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 12/24/2022]
Abstract
Esophageal cancer is of high importance to occurrence, development, and treatment resistance. As evidenced by recent studies, pathways (e.g., Wnt/β-catenin, AMPK, and Hippo) are critical to the proliferation, differentiation, and self-renewal of esophageal cancer. In addition, the above pathways play a certain role in regulating esophageal cancer and act as potential therapeutic targets. Over the past few years, the function of lipid metabolism in controlling tumor cells and immune cells has aroused extensive attention. It has been reported that there are intricate interactions between lipid metabolism reprogramming between immune and esophageal cancer cells, whereas molecular mechanisms should be studied in depth. Immune cells have been commonly recognized as a vital player in the esophageal cancer microenvironment, having complex crosstalk with cancer cells. It is increasingly evidenced that the function of immune cells in the tumor microenvironment (TME) is significantly correlated with abnormal lipid metabolism. In this review, the latest findings in lipid metabolism reprogramming in TME are summarized, and the above findings are linked to esophageal cancer progression. Aberrant lipid metabolism and associated signaling pathways are likely to serve as a novel strategy to treat esophageal cancer through lipid metabolism reprogramming.
Collapse
Affiliation(s)
- Meng-Ying Cui
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xing Yi
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Zhen-Zhen Cao
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Dan-Xia Zhu
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jun Wu
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
13
|
Hu J, Ma Y, Ma J, Yang Y, Ning Y, Zhu J, Wang P, Chen G, Liu Y. M2 Macrophage-Based Prognostic Nomogram for Gastric Cancer After Surgical Resection. Front Oncol 2021; 11:690037. [PMID: 34458140 PMCID: PMC8397443 DOI: 10.3389/fonc.2021.690037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
A good prediction model is useful to accurately predict patient prognosis. Tumor-node-metastasis (TNM) staging often cannot accurately predict prognosis when used alone. Some researchers have shown that the infiltration of M2 macrophages in many tumors indicates poor prognosis. This approach has the potential to predict prognosis more accurately when used in combination with TNM staging, but there is less research in gastric cancer. A multivariate analysis demonstrated that CD163 expression, TNM staging, age, and gender were independent risk factors for overall survival. Thus, these parameters were assessed to develop the nomogram in the training data set, which was tested in the validation and whole data sets. The model showed a high degree of discrimination, calibration, and good clinical benefit in the training, validation, and whole data sets. In conclusion, we combined CD163 expression in macrophages, TNM staging, age, and gender to develop a nomogram to predict 3- and 5-year overall survivals after curative resection for gastric cancer. This model has the potential to provide further diagnostic and prognostic value for patients with gastric cancer.
Collapse
Affiliation(s)
- Jianwen Hu
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yongchen Ma
- Endoscopy Center, Peking University First Hospital, Beijing, China
| | - Ju Ma
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yanpeng Yang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yingze Ning
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Jing Zhu
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Pengyuan Wang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Guowei Chen
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yucun Liu
- Department of General Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|